1
|
Kuczynski-Noyau L, Karmann S, Alberton P, Martinez-Corral I, Nampoothiri S, Sauvé F, Lhomme T, Quarta C, Apte SS, Bouret S, Aszodi A, Rasika S, Ciofi P, Dam J, Prévot V, Mattot V. A plastic aggrecan barrier modulated by peripheral energy state gates metabolic signal access to arcuate neurons. Nat Commun 2024; 15:6701. [PMID: 39112471 PMCID: PMC11306556 DOI: 10.1038/s41467-024-50798-9] [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: 03/12/2024] [Accepted: 07/17/2024] [Indexed: 08/10/2024] Open
Abstract
The hypothalamic arcuate nucleus (ARH) contains neurons vital for maintaining energy homeostasis that sense and respond to changes in blood-borne metabolic hormones. Despite its juxtaposition to the median eminence (ME), a circumventricular organ lacking a blood-brain barrier and thus exposed to circulating molecules, only a few ventral ARH neurons perceive these extravasating metabolic signals due to a poorly understood ME/ARH diffusion barrier. Here, we show in male mice that aggrecan, a perineural-net proteoglycan deposited by orexigenic ARH neurons, creates a peculiar ventrodorsal diffusion gradient. Fasting enhances aggrecan deposition more dorsally, reinforcing the diffusion barrier, particularly around neurons adjacent to fenestrated capillary loops that enter the ARH. The disruption of aggrecan deposits results in unregulated diffusion of blood-borne molecules into the ARH and impairs food intake. Our findings reveal the molecular nature and plasticity of the ME/ARH diffusion barrier, and indicate its physiological role in hypothalamic metabolic hormone sensing.
Collapse
Affiliation(s)
- Laura Kuczynski-Noyau
- Univ. Lille, INSERM, CHU Lille, Laboratory of Development and Plasticity of the Neuroendocrine Brain, Lille Neuroscience & Cognition, UMR-S1172, EGID, Lille, France
| | - Sixtine Karmann
- Univ. Lille, INSERM, CHU Lille, Laboratory of Development and Plasticity of the Neuroendocrine Brain, Lille Neuroscience & Cognition, UMR-S1172, EGID, Lille, France
| | - Paolo Alberton
- Department for Orthopaedics and Trauma Surgery Musculoskeletal University Center Munich (MUM) University Hospital, LMU, Munich, Germany
- Division of Hand, Plastic and Aesthetic Surgery LMU University Hospital, LMU, Munich, Germany
| | - Ines Martinez-Corral
- Univ. Lille, INSERM, CHU Lille, Laboratory of Development and Plasticity of the Neuroendocrine Brain, Lille Neuroscience & Cognition, UMR-S1172, EGID, Lille, France
| | - Sreekala Nampoothiri
- Univ. Lille, INSERM, CHU Lille, Laboratory of Development and Plasticity of the Neuroendocrine Brain, Lille Neuroscience & Cognition, UMR-S1172, EGID, Lille, France
| | - Florent Sauvé
- Univ. Lille, INSERM, CHU Lille, Laboratory of Development and Plasticity of the Neuroendocrine Brain, Lille Neuroscience & Cognition, UMR-S1172, EGID, Lille, France
| | - Tori Lhomme
- Univ. Lille, INSERM, CHU Lille, Laboratory of Development and Plasticity of the Neuroendocrine Brain, Lille Neuroscience & Cognition, UMR-S1172, EGID, Lille, France
| | - Carmelo Quarta
- Univ. Bordeaux, INSERM, Neurocentre Magendie, U1215, Bordeaux, France
| | - Suneel S Apte
- Department of Biomedical Engineering, Cleveland, OH, USA
| | - Sébastien Bouret
- Univ. Lille, INSERM, CHU Lille, Laboratory of Development and Plasticity of the Neuroendocrine Brain, Lille Neuroscience & Cognition, UMR-S1172, EGID, Lille, France
| | - Attila Aszodi
- Department for Orthopaedics and Trauma Surgery Musculoskeletal University Center Munich (MUM) University Hospital, LMU, Munich, Germany
| | - Sowmyalakshmi Rasika
- Univ. Lille, INSERM, CHU Lille, Laboratory of Development and Plasticity of the Neuroendocrine Brain, Lille Neuroscience & Cognition, UMR-S1172, EGID, Lille, France
| | - Philippe Ciofi
- Univ. Bordeaux, INSERM, Neurocentre Magendie, U1215, Bordeaux, France
| | - Julie Dam
- Institut Cochin, INSERM, CNRS, Université Paris Cité, Paris, France
| | - Vincent Prévot
- Univ. Lille, INSERM, CHU Lille, Laboratory of Development and Plasticity of the Neuroendocrine Brain, Lille Neuroscience & Cognition, UMR-S1172, EGID, Lille, France
| | - Virginie Mattot
- Univ. Lille, INSERM, CHU Lille, Laboratory of Development and Plasticity of the Neuroendocrine Brain, Lille Neuroscience & Cognition, UMR-S1172, EGID, Lille, France.
| |
Collapse
|
2
|
Charrière K, Schneider V, Perrignon-Sommet M, Lizard G, Benani A, Jacquin-Piques A, Vejux A. Exploring the Role of Apigenin in Neuroinflammation: Insights and Implications. Int J Mol Sci 2024; 25:5041. [PMID: 38732259 PMCID: PMC11084463 DOI: 10.3390/ijms25095041] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2024] [Revised: 05/01/2024] [Accepted: 05/01/2024] [Indexed: 05/13/2024] Open
Abstract
Neuroinflammation, a hallmark of various central nervous system disorders, is often associated with oxidative stress and neuronal or oligodendrocyte cell death. It is therefore very interesting to target neuroinflammation pharmacologically. One therapeutic option is the use of nutraceuticals, particularly apigenin. Apigenin is present in plants: vegetables (parsley, celery, onions), fruits (oranges), herbs (chamomile, thyme, oregano, basil), and some beverages (tea, beer, and wine). This review explores the potential of apigenin as an anti-inflammatory agent across diverse neurological conditions (multiple sclerosis, Parkinson's disease, Alzheimer's disease), cancer, cardiovascular diseases, cognitive and memory disorders, and toxicity related to trace metals and other chemicals. Drawing upon major studies, we summarize apigenin's multifaceted effects and underlying mechanisms in neuroinflammation. Our review underscores apigenin's therapeutic promise and calls for further investigation into its clinical applications.
Collapse
Affiliation(s)
- Karine Charrière
- Université de Franche-Comté, CHU Besançon, UMR 1322 LINC, INSERM CIC 1431, 25000 Besançon, France;
| | - Vincent Schneider
- Centre des Sciences du Goût et de l’Alimentation, CNRS, INRAE, Institut Agro, Université de Bourgogne, 21000 Dijon, France; (V.S.); (M.P.-S.); (A.B.); (A.J.-P.)
- Neurology and Clinical Neurophysiology Department, CHU F. Mitterrand, 21000 Dijon, France
| | - Manon Perrignon-Sommet
- Centre des Sciences du Goût et de l’Alimentation, CNRS, INRAE, Institut Agro, Université de Bourgogne, 21000 Dijon, France; (V.S.); (M.P.-S.); (A.B.); (A.J.-P.)
| | - Gérard Lizard
- Bio-PeroxIL Laboratory, EA7270, University of Bourgogne, 21000 Dijon, France;
| | - Alexandre Benani
- Centre des Sciences du Goût et de l’Alimentation, CNRS, INRAE, Institut Agro, Université de Bourgogne, 21000 Dijon, France; (V.S.); (M.P.-S.); (A.B.); (A.J.-P.)
| | - Agnès Jacquin-Piques
- Centre des Sciences du Goût et de l’Alimentation, CNRS, INRAE, Institut Agro, Université de Bourgogne, 21000 Dijon, France; (V.S.); (M.P.-S.); (A.B.); (A.J.-P.)
- Neurology and Clinical Neurophysiology Department, CHU F. Mitterrand, 21000 Dijon, France
- Memory Resource and Research Center (CMRR), CHU F. Mitterrand, 21000 Dijon, France
| | - Anne Vejux
- Centre des Sciences du Goût et de l’Alimentation, CNRS, INRAE, Institut Agro, Université de Bourgogne, 21000 Dijon, France; (V.S.); (M.P.-S.); (A.B.); (A.J.-P.)
- Bio-PeroxIL Laboratory, EA7270, University of Bourgogne, 21000 Dijon, France;
| |
Collapse
|
3
|
Sayar-Atasoy N, Yavuz Y, Laule C, Dong C, Kim H, Rysted J, Flippo K, Davis D, Aklan I, Yilmaz B, Tian L, Atasoy D. Opioidergic signaling contributes to food-mediated suppression of AgRP neurons. Cell Rep 2024; 43:113630. [PMID: 38165803 PMCID: PMC10865729 DOI: 10.1016/j.celrep.2023.113630] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2022] [Revised: 11/22/2023] [Accepted: 12/13/2023] [Indexed: 01/04/2024] Open
Abstract
Opioids are generally known to promote hedonic food consumption. Although much of the existing evidence is primarily based on studies of the mesolimbic pathway, endogenous opioids and their receptors are widely expressed in hypothalamic appetite circuits as well; however, their role in homeostatic feeding remains unclear. Using a fluorescent opioid sensor, deltaLight, here we report that mediobasal hypothalamic opioid levels increase by feeding, which directly and indirectly inhibits agouti-related protein (AgRP)-expressing neurons through the μ-opioid receptor (MOR). AgRP-specific MOR expression increases by energy surfeit and contributes to opioid-induced suppression of appetite. Conversely, its antagonists diminish suppression of AgRP neuron activity by food and satiety hormones. Mice with AgRP neuron-specific ablation of MOR expression have increased fat preference without increased motivation. These results suggest that post-ingestion release of endogenous opioids contributes to AgRP neuron inhibition to shape food choice through MOR signaling.
Collapse
Affiliation(s)
- Nilufer Sayar-Atasoy
- Department of Neuroscience and Pharmacology, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, IA 52242, USA
| | - Yavuz Yavuz
- Department of Neuroscience and Pharmacology, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, IA 52242, USA; Department of Physiology, School of Medicine, Yeditepe University, Istanbul 34755, Turkey
| | - Connor Laule
- Department of Neuroscience and Pharmacology, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, IA 52242, USA
| | - Chunyang Dong
- Department of Biochemistry and Molecular Medicine, School of Medicine, University of California, Davis, Davis, CA 95616, USA
| | - Hyojin Kim
- Department of Neuroscience and Pharmacology, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, IA 52242, USA
| | - Jacob Rysted
- Department of Neuroscience and Pharmacology, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, IA 52242, USA
| | - Kyle Flippo
- Department of Neuroscience and Pharmacology, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, IA 52242, USA
| | - Debbie Davis
- Department of Neuroscience and Pharmacology, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, IA 52242, USA
| | - Iltan Aklan
- Department of Neuroscience and Pharmacology, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, IA 52242, USA
| | - Bayram Yilmaz
- Department of Physiology, School of Medicine, Yeditepe University, Istanbul 34755, Turkey
| | - Lin Tian
- Department of Biochemistry and Molecular Medicine, School of Medicine, University of California, Davis, Davis, CA 95616, USA
| | - Deniz Atasoy
- Department of Neuroscience and Pharmacology, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, IA 52242, USA; Iowa Neuroscience Institute, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, IA 52242, USA; Fraternal Order of Eagles Diabetes Research Center (FOEDRC), Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, IA 52242, USA.
| |
Collapse
|
4
|
Massa MG, Scott RL, Cara AL, Cortes LR, Vander PB, Sandoval NP, Park JW, Ali SL, Velez LM, Wang HB, Ati SS, Tesfaye B, Reue K, van Veen JE, Seldin MM, Correa SM. Feeding neurons integrate metabolic and reproductive states in mice. iScience 2023; 26:107918. [PMID: 37817932 PMCID: PMC10561062 DOI: 10.1016/j.isci.2023.107918] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2023] [Revised: 07/27/2023] [Accepted: 09/12/2023] [Indexed: 10/12/2023] Open
Abstract
Balance between metabolic and reproductive processes is important for survival, particularly in mammals that gestate their young. How the nervous system coordinates this balance is an active area of study. Herein, we demonstrate that somatostatin (SST) neurons of the tuberal hypothalamus alter feeding in a manner sensitive to metabolic and reproductive states in mice. Whereas chemogenetic activation of SST neurons increased food intake across sexes, ablation decreased food intake only in female mice during proestrus. This ablation effect was only apparent in animals with low body mass. Fat transplantation and bioinformatics analysis of SST neuronal transcriptomes revealed white adipose as a key modulator of these effects. These studies indicate that SST hypothalamic neurons integrate metabolic and reproductive cues by responding to varying levels of circulating estrogens to modulate feeding differentially based on energy stores. Thus, gonadal steroid modulation of neuronal circuits can be context dependent and gated by metabolic status.
Collapse
Affiliation(s)
- Megan G. Massa
- Department of Integrative Biology and Physiology, University of California – Los Angeles, Los Angeles, CA 90095, USA
- Neuroscience Interdepartmental Doctoral Program, University of California – Los Angeles, Los Angeles, CA 90095, USA
| | - Rachel L. Scott
- Department of Integrative Biology and Physiology, University of California – Los Angeles, Los Angeles, CA 90095, USA
| | - Alexandra L. Cara
- Department of Integrative Biology and Physiology, University of California – Los Angeles, Los Angeles, CA 90095, USA
| | - Laura R. Cortes
- Department of Integrative Biology and Physiology, University of California – Los Angeles, Los Angeles, CA 90095, USA
| | - Paul B. Vander
- Department of Integrative Biology and Physiology, University of California – Los Angeles, Los Angeles, CA 90095, USA
| | - Norma P. Sandoval
- Department of Integrative Biology and Physiology, University of California – Los Angeles, Los Angeles, CA 90095, USA
| | - Jae W. Park
- Department of Integrative Biology and Physiology, University of California – Los Angeles, Los Angeles, CA 90095, USA
| | - Sahara L. Ali
- Department of Integrative Biology and Physiology, University of California – Los Angeles, Los Angeles, CA 90095, USA
| | - Leandro M. Velez
- Department of Human Genetics, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095, USA
| | - Huei-Bin Wang
- Department of Integrative Biology and Physiology, University of California – Los Angeles, Los Angeles, CA 90095, USA
| | - Shomik S. Ati
- Department of Integrative Biology and Physiology, University of California – Los Angeles, Los Angeles, CA 90095, USA
| | - Bethlehem Tesfaye
- Department of Integrative Biology and Physiology, University of California – Los Angeles, Los Angeles, CA 90095, USA
| | - Karen Reue
- Department of Human Genetics, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095, USA
| | - J. Edward van Veen
- Department of Integrative Biology and Physiology, University of California – Los Angeles, Los Angeles, CA 90095, USA
| | - Marcus M. Seldin
- Department of Biological Chemistry, School of Medicine, University of California – Irvine, Irvine, CA 92697, USA
| | - Stephanie M. Correa
- Department of Integrative Biology and Physiology, University of California – Los Angeles, Los Angeles, CA 90095, USA
| |
Collapse
|
5
|
Adriaenssens A, Broichhagen J, de Bray A, Ast J, Hasib A, Jones B, Tomas A, Burgos NF, Woodward O, Lewis J, O’Flaherty E, El K, Cui C, Harada N, Inagaki N, Campbell J, Brierley D, Hodson DJ, Samms R, Gribble F, Reimann F. Hypothalamic and brainstem glucose-dependent insulinotropic polypeptide receptor neurons employ distinct mechanisms to affect feeding. JCI Insight 2023; 8:e164921. [PMID: 37212283 PMCID: PMC10322681 DOI: 10.1172/jci.insight.164921] [Citation(s) in RCA: 18] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2022] [Accepted: 04/18/2023] [Indexed: 05/23/2023] Open
Abstract
Central glucose-dependent insulinotropic polypeptide (GIP) receptor (GIPR) signaling is critical in GIP-based therapeutics' ability to lower body weight, but pathways leveraged by GIPR pharmacology in the brain remain incompletely understood. We explored the role of Gipr neurons in the hypothalamus and dorsal vagal complex (DVC) - brain regions critical to the control of energy balance. Hypothalamic Gipr expression was not necessary for the synergistic effect of GIPR/GLP-1R coagonism on body weight. While chemogenetic stimulation of both hypothalamic and DVC Gipr neurons suppressed food intake, activation of DVC Gipr neurons reduced ambulatory activity and induced conditioned taste avoidance, while there was no effect of a short-acting GIPR agonist (GIPRA). Within the DVC, Gipr neurons of the nucleus tractus solitarius (NTS), but not the area postrema (AP), projected to distal brain regions and were transcriptomically distinct. Peripherally dosed fluorescent GIPRAs revealed that access was restricted to circumventricular organs in the CNS. These data demonstrate that Gipr neurons in the hypothalamus, AP, and NTS differ in their connectivity, transcriptomic profile, peripheral accessibility, and appetite-controlling mechanisms. These results highlight the heterogeneity of the central GIPR signaling axis and suggest that studies into the effects of GIP pharmacology on feeding behavior should consider the interplay of multiple regulatory pathways.
Collapse
Affiliation(s)
- Alice Adriaenssens
- Institute of Metabolic Science & MRC Metabolic Diseases Unit, University of Cambridge, Cambridge, United Kingdom
- Department of Neuroscience, Physiology, and Pharmacology, University College London, London, United Kingdom
| | | | - Anne de Bray
- Oxford Center for Diabetes, Endocrinology and Metabolism (OCDEM), NIHR Oxford Biomedical Research Center, Churchill Hospital, Radcliffe Department of Medicine, University of Oxford, Oxford, United Kingdom
- Institute of Metabolism and Systems Research (IMSR) and Center of Membrane Proteins and Receptors (COMPARE), University of Birmingham, Birmingham, United Kingdom
| | - Julia Ast
- Oxford Center for Diabetes, Endocrinology and Metabolism (OCDEM), NIHR Oxford Biomedical Research Center, Churchill Hospital, Radcliffe Department of Medicine, University of Oxford, Oxford, United Kingdom
- Institute of Metabolism and Systems Research (IMSR) and Center of Membrane Proteins and Receptors (COMPARE), University of Birmingham, Birmingham, United Kingdom
| | - Annie Hasib
- Institute of Metabolism and Systems Research (IMSR) and Center of Membrane Proteins and Receptors (COMPARE), University of Birmingham, Birmingham, United Kingdom
| | - Ben Jones
- Department of Metabolism, Digestion and Reproduction, Imperial College London, London, United Kingdom
| | - Alejandra Tomas
- Department of Metabolism, Digestion and Reproduction, Imperial College London, London, United Kingdom
| | - Natalie Figueredo Burgos
- Department of Neuroscience, Physiology, and Pharmacology, University College London, London, United Kingdom
| | - Orla Woodward
- Institute of Metabolic Science & MRC Metabolic Diseases Unit, University of Cambridge, Cambridge, United Kingdom
| | - Jo Lewis
- Institute of Metabolic Science & MRC Metabolic Diseases Unit, University of Cambridge, Cambridge, United Kingdom
| | - Elisabeth O’Flaherty
- Institute of Metabolic Science & MRC Metabolic Diseases Unit, University of Cambridge, Cambridge, United Kingdom
| | - Kimberley El
- Department of Medicine, Duke University Hospital, Durham, North Carolina, USA
| | - Canqi Cui
- Department of Medicine, Duke University Hospital, Durham, North Carolina, USA
| | - Norio Harada
- Department of Diabetes, Endocrinology and Nutrition, Kyoto University, Kyoto, Japan
| | - Nobuya Inagaki
- Department of Diabetes, Endocrinology and Nutrition, Kyoto University, Kyoto, Japan
| | - Jonathan Campbell
- Department of Medicine, Duke University Hospital, Durham, North Carolina, USA
| | - Daniel Brierley
- Department of Neuroscience, Physiology, and Pharmacology, University College London, London, United Kingdom
| | - David J. Hodson
- Oxford Center for Diabetes, Endocrinology and Metabolism (OCDEM), NIHR Oxford Biomedical Research Center, Churchill Hospital, Radcliffe Department of Medicine, University of Oxford, Oxford, United Kingdom
- Institute of Metabolism and Systems Research (IMSR) and Center of Membrane Proteins and Receptors (COMPARE), University of Birmingham, Birmingham, United Kingdom
| | - Ricardo Samms
- Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, Indiana, USA
| | - Fiona Gribble
- Institute of Metabolic Science & MRC Metabolic Diseases Unit, University of Cambridge, Cambridge, United Kingdom
| | - Frank Reimann
- Institute of Metabolic Science & MRC Metabolic Diseases Unit, University of Cambridge, Cambridge, United Kingdom
| |
Collapse
|
6
|
Yu Q, Gamayun I, Wartenberg P, Zhang Q, Qiao S, Kusumakshi S, Candlish S, Götz V, Wen S, Das D, Wyatt A, Wahl V, Ectors F, Kattler K, Yildiz D, Prevot V, Schwaninger M, Ternier G, Giacobini P, Ciofi P, Müller TD, Boehm U. Bitter taste cells in the ventricular walls of the murine brain regulate glucose homeostasis. Nat Commun 2023; 14:1588. [PMID: 36949050 PMCID: PMC10033832 DOI: 10.1038/s41467-023-37099-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Accepted: 03/02/2023] [Indexed: 03/24/2023] Open
Abstract
The median eminence (ME) is a circumventricular organ at the base of the brain that controls body homeostasis. Tanycytes are its specialized glial cells that constitute the ventricular walls and regulate different physiological states, however individual signaling pathways in these cells are incompletely understood. Here, we identify a functional tanycyte subpopulation that expresses key taste transduction genes including bitter taste receptors, the G protein gustducin and the gustatory ion channel TRPM5 (M5). M5 tanycytes have access to blood-borne cues via processes extended towards diaphragmed endothelial fenestrations in the ME and mediate bidirectional communication between the cerebrospinal fluid and blood. This subpopulation responds to metabolic signals including leptin and other hormonal cues and is transcriptionally reprogrammed upon fasting. Acute M5 tanycyte activation induces insulin secretion and acute diphtheria toxin-mediated M5 tanycyte depletion results in impaired glucose tolerance in diet-induced obese mice. We provide a cellular and molecular framework that defines how bitter taste cells in the ME integrate chemosensation with metabolism.
Collapse
Affiliation(s)
- Qiang Yu
- Department of Pharmacology, Center for Molecular Signaling (PZMS), Saarland University School of Medicine, Homburg, Germany
| | - Igor Gamayun
- Department of Pharmacology, Center for Molecular Signaling (PZMS), Saarland University School of Medicine, Homburg, Germany
| | - Philipp Wartenberg
- Department of Pharmacology, Center for Molecular Signaling (PZMS), Saarland University School of Medicine, Homburg, Germany
| | - Qian Zhang
- Institute for Diabetes and Obesity, Helmholtz Diabetes Center, Helmholtz Zentrum München, Neuherberg, Germany; German Center for Diabetes Research (DZD), Neuherberg, Germany
| | - Sen Qiao
- Department of Pharmacology, Center for Molecular Signaling (PZMS), Saarland University School of Medicine, Homburg, Germany
| | - Soumya Kusumakshi
- Department of Pharmacology, Center for Molecular Signaling (PZMS), Saarland University School of Medicine, Homburg, Germany
| | - Sarah Candlish
- Department of Pharmacology, Center for Molecular Signaling (PZMS), Saarland University School of Medicine, Homburg, Germany
| | - Viktoria Götz
- Department of Pharmacology, Center for Molecular Signaling (PZMS), Saarland University School of Medicine, Homburg, Germany
| | - Shuping Wen
- Department of Pharmacology, Center for Molecular Signaling (PZMS), Saarland University School of Medicine, Homburg, Germany
| | - Debajyoti Das
- Department of Pharmacology, Center for Molecular Signaling (PZMS), Saarland University School of Medicine, Homburg, Germany
| | - Amanda Wyatt
- Department of Pharmacology, Center for Molecular Signaling (PZMS), Saarland University School of Medicine, Homburg, Germany
| | - Vanessa Wahl
- Department of Pharmacology, Center for Molecular Signaling (PZMS), Saarland University School of Medicine, Homburg, Germany
| | - Fabien Ectors
- FARAH Mammalian Transgenics Platform, Liège University, Liège, Belgium
| | - Kathrin Kattler
- Department of Genetics, Saarland University, Saarbrücken, Germany
| | - Daniela Yildiz
- Department of Pharmacology, Center for Molecular Signaling (PZMS), Saarland University School of Medicine, Homburg, Germany
| | - Vincent Prevot
- Univ. Lille, Inserm, CHU Lille, Laboratory of Development and Plasticity of the Postnatal Brain, Lille Neuroscience & Cognition, UMR-S1172, Lille, France
| | - Markus Schwaninger
- Institute for Experimental and Clinical Pharmacology and Toxicology, Center of Brain, Behavior and Metabolism (CBBM), University of Lübeck, Lübeck, Germany
| | - Gaetan Ternier
- Univ. Lille, Inserm, CHU Lille, Laboratory of Development and Plasticity of the Postnatal Brain, Lille Neuroscience & Cognition, UMR-S1172, Lille, France
| | - Paolo Giacobini
- Univ. Lille, Inserm, CHU Lille, Laboratory of Development and Plasticity of the Postnatal Brain, Lille Neuroscience & Cognition, UMR-S1172, Lille, France
| | - Philippe Ciofi
- Neurocentre Magendie - INSERM Unit 1215, University of Bordeaux, Bordeaux, France
| | - Timo D Müller
- Institute for Diabetes and Obesity, Helmholtz Diabetes Center, Helmholtz Zentrum München, Neuherberg, Germany; German Center for Diabetes Research (DZD), Neuherberg, Germany
| | - Ulrich Boehm
- Department of Pharmacology, Center for Molecular Signaling (PZMS), Saarland University School of Medicine, Homburg, Germany.
| |
Collapse
|
7
|
Massa MG, Scott RL, Cara AL, Cortes LR, Sandoval NP, Park JW, Ali S, Velez LM, Tesfaye B, Reue K, van Veen JE, Seldin M, Correa SM. Feeding Neurons Integrate Metabolic and Reproductive States in Mice. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.01.25.525595. [PMID: 36747631 PMCID: PMC9900829 DOI: 10.1101/2023.01.25.525595] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
Trade-offs between metabolic and reproductive processes are important for survival, particularly in mammals that gestate their young. Puberty and reproduction, as energetically taxing life stages, are often gated by metabolic availability in animals with ovaries. How the nervous system coordinates these trade-offs is an active area of study. We identify somatostatin neurons of the tuberal nucleus (TNSST) as a node of the feeding circuit that alters feeding in a manner sensitive to metabolic and reproductive states in mice. Whereas chemogenetic activation of TNSST neurons increased food intake across sexes, selective ablation decreased food intake only in female mice during proestrus. Interestingly, this ablation effect was only apparent in animals with a low body mass. Fat transplantation and bioinformatics analysis of TNSST neuronal transcriptomes revealed white adipose as a key modulator of the effects of TNSST neurons on food intake. Together, these studies point to a mechanism whereby TNSST hypothalamic neurons modulate feeding by responding to varying levels of circulating estrogens differentially based on energy stores. This research provides insight into how neural circuits integrate reproductive and metabolic signals, and illustrates how gonadal steroid modulation of neuronal circuits can be context-dependent and gated by metabolic status.
Collapse
Affiliation(s)
- Megan G Massa
- Department of Integrative Biology and Physiology, University of California, Los Angeles, CA
| | - Rachel L Scott
- Department of Integrative Biology and Physiology, University of California, Los Angeles, CA
| | - Alexandra L Cara
- Department of Integrative Biology and Physiology, University of California, Los Angeles, CA
| | - Laura R Cortes
- Department of Integrative Biology and Physiology, University of California, Los Angeles, CA
| | - Norma P Sandoval
- Department of Integrative Biology and Physiology, University of California, Los Angeles, CA
| | - Jae W Park
- Department of Integrative Biology and Physiology, University of California, Los Angeles, CA
| | - Sahara Ali
- Department of Integrative Biology and Physiology, University of California, Los Angeles, CA
| | - Leandro M Velez
- Department of Biological Chemistry, School of Medicine, University of California, Irvine, CA
| | - Bethlehem Tesfaye
- Department of Integrative Biology and Physiology, University of California, Los Angeles, CA
| | - Karen Reue
- Department of Human Genetics, David Geffen School of Medicine at University of California, Los Angeles, CA
| | - J Edward van Veen
- Department of Integrative Biology and Physiology, University of California, Los Angeles, CA
| | - Marcus Seldin
- Department of Biological Chemistry, School of Medicine, University of California, Irvine, CA
| | - Stephanie M Correa
- Department of Integrative Biology and Physiology, University of California, Los Angeles, CA
| |
Collapse
|
8
|
Folick A, Cheang RT, Valdearcos M, Koliwad SK. Metabolic factors in the regulation of hypothalamic innate immune responses in obesity. Exp Mol Med 2022; 54:393-402. [PMID: 35474339 PMCID: PMC9076660 DOI: 10.1038/s12276-021-00666-z] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2021] [Accepted: 05/13/2021] [Indexed: 12/14/2022] Open
Abstract
The hypothalamus is a central regulator of body weight and energy homeostasis. There is increasing evidence that innate immune activation in the mediobasal hypothalamus (MBH) is a key element in the pathogenesis of diet-induced obesity. Microglia, the resident immune cells in the brain parenchyma, have been shown to play roles in diverse aspects of brain function, including circuit refinement and synaptic pruning. As such, microglia have also been implicated in the development and progression of neurological diseases. Microglia express receptors for and are responsive to a wide variety of nutritional, hormonal, and immunological signals that modulate their distinct functions across different brain regions. We showed that microglia within the MBH sense and respond to a high-fat diet and regulate the function of hypothalamic neurons to promote food intake and obesity. Neurons, glia, and immune cells within the MBH are positioned to sense and respond to circulating signals that regulate their capacity to coordinate aspects of systemic energy metabolism. Here, we review the current knowledge of how these peripheral signals modulate the innate immune response in the MBH and enable microglia to regulate metabolic control.
Collapse
Affiliation(s)
- Andrew Folick
- Diabetes Center and Division of Endocrinology and Metabolism, Department of Medicine, University of California, San Francisco, CA, USA
| | - Rachel T Cheang
- Diabetes Center and Division of Endocrinology and Metabolism, Department of Medicine, University of California, San Francisco, CA, USA
| | - Martin Valdearcos
- Diabetes Center and Division of Endocrinology and Metabolism, Department of Medicine, University of California, San Francisco, CA, USA.
| | - Suneil K Koliwad
- Diabetes Center and Division of Endocrinology and Metabolism, Department of Medicine, University of California, San Francisco, CA, USA.
| |
Collapse
|
9
|
Rodríguez-Cortés B, Hurtado-Alvarado G, Martínez-Gómez R, León-Mercado LA, Prager-Khoutorsky M, Buijs RM. Suprachiasmatic nucleus-mediated glucose entry into the arcuate nucleus determines the daily rhythm in blood glycemia. Curr Biol 2022; 32:796-805.e4. [PMID: 35030330 DOI: 10.1016/j.cub.2021.12.039] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2021] [Revised: 11/19/2021] [Accepted: 12/16/2021] [Indexed: 12/20/2022]
Abstract
Glycemia is maintained within very narrow boundaries with less than 5% variation at a given time of the day. However, over the circadian cycle, glycemia changes with almost 50% difference. How the suprachiasmatic nucleus, the biological clock, maintains these day-night variations with such tiny disparities remains obscure. We show that via vasopressin release at the beginning of the sleep phase, the suprachiasmatic nucleus increases the glucose transporter GLUT1 in tanycytes. Hereby GLUT1 promotes glucose entrance into the arcuate nucleus, thereby lowering peripheral glycemia. Conversely, blocking vasopressin activity or the GLUT1 transporter at the daily trough of glycemia increases circulating glucose levels usually seen at the peak of the rhythm. Thus, biological clock-controlled mechanisms promoting glucose entry into the arcuate nucleus explain why peripheral blood glucose is low before sleep onset.
Collapse
Affiliation(s)
- Betty Rodríguez-Cortés
- Department of Cellular Biology and Physiology, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Mario de la Cueva Circuit, Mexico City 04510, Mexico
| | - Gabriela Hurtado-Alvarado
- Department of Cellular Biology and Physiology, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Mario de la Cueva Circuit, Mexico City 04510, Mexico
| | - Ricardo Martínez-Gómez
- Department of Cellular Biology and Physiology, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Mario de la Cueva Circuit, Mexico City 04510, Mexico
| | - Luis A León-Mercado
- Department of Internal Medicine, Center for Hypothalamic Research, University of Texas Southwestern Medical Center, 5323 Harry Hines Boulevard, Dallas, TX 75390, USA
| | - Masha Prager-Khoutorsky
- Department of Physiology, McIntyre Medical Sciences Building, McGill University, 3655 Promenade Sir-William-Osler, Montréal, QC H3G 1Y6, Canada
| | - Ruud M Buijs
- Department of Cellular Biology and Physiology, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Mario de la Cueva Circuit, Mexico City 04510, Mexico.
| |
Collapse
|
10
|
Uriarte M, De Francesco PN, Fernández G, Castrogiovanni D, D'Arcangelo M, Imbernon M, Cantel S, Denoyelle S, Fehrentz JA, Praetorius J, Prevot V, Perello M. Circulating ghrelin crosses the blood-cerebrospinal fluid barrier via growth hormone secretagogue receptor dependent and independent mechanisms. Mol Cell Endocrinol 2021; 538:111449. [PMID: 34478806 DOI: 10.1016/j.mce.2021.111449] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/09/2021] [Revised: 08/13/2021] [Accepted: 08/30/2021] [Indexed: 01/17/2023]
Abstract
Ghrelin is a peptide hormone mainly secreted from gastrointestinal tract that acts via the growth hormone secretagogue receptor (GHSR), which is highly expressed in the brain. Strikingly, the accessibility of ghrelin to the brain seems to be limited and restricted to few brain areas. Previous studies in mice have shown that ghrelin can access the brain via the blood-cerebrospinal fluid (CSF) barrier, an interface constituted by the choroid plexus and the hypothalamic tanycytes. Here, we performed a variety of in vivo and in vitro studies to test the hypothesis that the transport of ghrelin across the blood-CSF barrier occurs in a GHSR-dependent manner. In vivo, we found that the uptake of systemically administered fluorescent ghrelin in the choroid plexus epithelial (CPE) cells and in hypothalamic tanycytes depends on the presence of GHSR. Also, we detected lower levels of CSF ghrelin after a systemic ghrelin injection in GHSR-deficient mice, as compared to WT mice. In vitro, the internalization of fluorescent ghrelin was reduced in explants of choroid plexus from GHSR-deficient mice, and unaffected in primary cultures of hypothalamic tanycytes derived from GHSR-deficient mice. Finally, we found that the GHSR mRNA is detected in a pool of CPE cells, but is nearly undetectable in hypothalamic tanycytes with current approaches. Thus, our results suggest that circulating ghrelin crosses the blood-CSF barrier mainly by a mechanism that involves the GHSR, and also possibly via a GHSR-independent mechanism.
Collapse
Affiliation(s)
- Maia Uriarte
- Laboratory of Neurophysiology, [Argentine Research Council (CONICET), Scientific Research Commission of the Province of Buenos Aires (CIC-PBA) and National University of La Plata (UNLP)], La Plata, Argentina
| | - Pablo N De Francesco
- Laboratory of Neurophysiology, [Argentine Research Council (CONICET), Scientific Research Commission of the Province of Buenos Aires (CIC-PBA) and National University of La Plata (UNLP)], La Plata, Argentina
| | - Gimena Fernández
- Laboratory of Neurophysiology, [Argentine Research Council (CONICET), Scientific Research Commission of the Province of Buenos Aires (CIC-PBA) and National University of La Plata (UNLP)], La Plata, Argentina
| | - Daniel Castrogiovanni
- Cell Culture Facility of the Multidisciplinary Institute of Cell Biology [Argentine Research Council (CONICET), Scientific Research Commission of the Province of Buenos Aires (CIC-PBA) and National University of La Plata (UNLP)], La Plata, Argentina
| | - Micaela D'Arcangelo
- Laboratory of Neurophysiology, [Argentine Research Council (CONICET), Scientific Research Commission of the Province of Buenos Aires (CIC-PBA) and National University of La Plata (UNLP)], La Plata, Argentina
| | - Mónica Imbernon
- Univ. Lille, Inserm, CHU Lille, Laboratory of Development and Plasticity of the Neuroendocrine Brain, Lille Neuroscience and Cognition, UMR, S1172, Lille, France
| | - Sonia Cantel
- Institut des Biomolécules Max Mousseron, UMR, 5247, CNRS-Université Montpellier-ENSCM, Faculté de Pharmacie, Montpellier, France
| | - Severine Denoyelle
- Institut des Biomolécules Max Mousseron, UMR, 5247, CNRS-Université Montpellier-ENSCM, Faculté de Pharmacie, Montpellier, France
| | - Jean-Alain Fehrentz
- Institut des Biomolécules Max Mousseron, UMR, 5247, CNRS-Université Montpellier-ENSCM, Faculté de Pharmacie, Montpellier, France
| | | | - Vincent Prevot
- Univ. Lille, Inserm, CHU Lille, Laboratory of Development and Plasticity of the Neuroendocrine Brain, Lille Neuroscience and Cognition, UMR, S1172, Lille, France
| | - Mario Perello
- Laboratory of Neurophysiology, [Argentine Research Council (CONICET), Scientific Research Commission of the Province of Buenos Aires (CIC-PBA) and National University of La Plata (UNLP)], La Plata, Argentina.
| |
Collapse
|
11
|
Salliss ME, Farland LV, Mahnert ND, Herbst-Kralovetz MM. The role of gut and genital microbiota and the estrobolome in endometriosis, infertility and chronic pelvic pain. Hum Reprod Update 2021; 28:92-131. [PMID: 34718567 DOI: 10.1093/humupd/dmab035] [Citation(s) in RCA: 75] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2021] [Revised: 08/25/2021] [Indexed: 12/30/2022] Open
Abstract
BACKGROUND Endometriosis is a chronic, burdensome condition that is historically understudied. Consequently, there is a lack of understanding of the etiology of the disease and its associated symptoms, including infertility and chronic pelvic pain (CPP). Endometriosis development is influenced by estrogen metabolism and inflammation, which are modulated by several factors including the microbiome and the estrobolome (the collection of genes encoding estrogen-metabolizing enzymes in the gut microbiome). Therefore, there is increasing interest in understanding the role of microbiota in endometriosis etiology. OBJECTIVE AND RATIONALE To date, there is no cure for endometriosis and treatment options often are ineffective. This manuscript will review the potential relationship between the microbiome and endometriosis, infertility and CPP and highlight the available data on the microbiome in relation to endometriosis and its related symptoms. The overarching goal of this manuscript is to inform future microbiome research that will lead to a deeper understanding of the etiology of the disease and possible diagnostic modalities and treatments. The potential impact of the microbiome on estrogen regulation modulated by the estrobolome, as well as inflammation and other endometriosis-promoting mechanisms within the genital tract, will be reviewed. The methodological limitations of microbiome-related studies will be critically assessed to provide improved guidelines for future microbiome and clinical studies. SEARCH METHODS PubMed databases were searched using the following keywords: endometriosis AND microbiome, infertility AND microbiome, pelvic pain AND microbiome, IVF (in-vitro fertilization) AND microbiome, endometriosis AND infertility. Clinical and preclinical animal trials that were eligible for review, and related to microbiome and endometriosis, infertility or CPP were included. All available manuscripts were published in 2002-2021. OUTCOMES In total, 28 clinical and 6 animal studies were included in the review. In both human and animal studies, bacteria were enriched in endometriosis groups, although there was no clear consensus on specific microbiota compositions that were associated with endometriosis, and no studies included infertility or CPP with endometriosis. However, bacterial vaginosis-associated bacteria and Lactobacillus depletion in the cervicovaginal microbiome were associated with endometriosis and infertility in the majority (23/28) of studies. Interpretation of endometrial studies is limited owing to a variety of methodological factors, discussed in this review. In addition, metadata outlining antibiotic usage, age, race/ethnicity, menopausal status and timing of sample collection in relation to diagnosis of endometriosis was not consistently reported. Animal studies (6/6) support a bidirectional relationship between the gut microbiota and endometriosis onset and progression. WIDER IMPLICATIONS There is evidence that a dysbiotic gut or genital microbiota is associated with multiple gynecologic conditions, with mounting data supporting an association between the microbiome and endometriosis and infertility. These microbiomes likely play a role in the gut-brain axis, which further supports a putative association with the spectrum of symptoms associated with endometriosis, including infertility and CPP. Collectively, this review highlights the demand for more rigorous and transparent methodology and controls, consistency across the field, and inclusion of key demographic and clinical characteristics of disease and comparison participants. Rigorous study designs will allow for a better understanding of the potential role of the microbiome in endometriosis etiology and the relationship to other disorders of the female reproductive tract.
Collapse
Affiliation(s)
- Mary E Salliss
- Department of Obstetrics and Gynecology, University of Arizona-College of Medicine, Phoenix, AZ, USA.,Department of Biology and Biochemistry, Bath University, Bath, UK
| | - Leslie V Farland
- Department of Epidemiology and Biostatistics, Mel and Enid Zuckerman College of Public Health, University of Arizona, Tucson, AZ, USA.,Department of Obstetrics and Gynecology, University of Arizona-College of Medicine Tucson, Tucson, AZ, USA
| | - Nichole D Mahnert
- Department of Obstetrics and Gynecology, University of Arizona-College of Medicine, Phoenix, AZ, USA.,Department of Obstetrics and Gynecology, Banner-University Medical Center Phoenix, Phoenix, AZ, USA
| | - Melissa M Herbst-Kralovetz
- Department of Obstetrics and Gynecology, University of Arizona-College of Medicine, Phoenix, AZ, USA.,Department of Basic Medical Sciences, University of Arizona-College of Medicine, Phoenix, AZ, USA
| |
Collapse
|
12
|
Olivera GC, Vetter L, Tesoriero C, Del Gallo F, Hedberg G, Basile J, Rottenberg ME. Role of T cells during the cerebral infection with Trypanosoma brucei. PLoS Negl Trop Dis 2021; 15:e0009764. [PMID: 34587172 PMCID: PMC8530334 DOI: 10.1371/journal.pntd.0009764] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2021] [Revised: 10/21/2021] [Accepted: 08/25/2021] [Indexed: 11/18/2022] Open
Abstract
The infection by Trypanosoma brucei brucei (T.b.b.), a protozoan parasite, is characterized by an early-systemic stage followed by a late stage in which parasites invade the brain parenchyma in a T cell-dependent manner. Here we found that early after infection effector-memory T cells were predominant among brain T cells, whereas, during the encephalitic stage T cells acquired a tissue resident memory phenotype (TRM) and expressed PD1. Both CD4 and CD8 T cells were independently redundant for the penetration of T.b.b. and other leukocytes into the brain parenchyma. The role of lymphoid cells during the T.b.b. infection was studied by comparing T- and B-cell deficient rag1-/- and WT mice. Early after infection, parasites located in circumventricular organs, brain structures with increased vascular permeability, particularly in the median eminence (ME), paced closed to the sleep-wake regulatory arcuate nucleus of the hypothalamus (Arc). Whereas parasite levels in the ME were higher in rag1-/- than in WT mice, leukocytes were instead reduced. Rag1-/- infected mice showed increased levels of meca32 mRNA coding for a blood /hypothalamus endothelial molecule absent in the blood-brain-barrier (BBB). Both immune and metabolic transcripts were elevated in the ME/Arc of WT and rag1-/- mice early after infection, except for ifng mRNA, which levels were only increased in WT mice. Finally, using a non-invasive sleep-wake cycle assessment method we proposed a putative role of lymphocytes in mediating sleep alterations during the infection with T.b.b. Thus, the majority of T cells in the brain during the early stage of T.b.b. infection expressed an effector-memory phenotype while TRM cells developed in the late stage of infection. T cells and parasites invade the ME/Arc altering the metabolic and inflammatory responses during the early stage of infection and modulating sleep disturbances. Trypanosoma brucei (T.b.) causes an early systemic and a late encephalitic infection characterized by sleep alterations. In rodent models, brain invasion by T.b. brucei (T.b.b.) is strictly dependent on T cells. However, an in-depth characterization of T cell functions and phenotypes in the outcome of T.b.b. infection is still lacking. Here we found that during the early stage of infection of mice, most brain T cells differentiated into memory cells, and acquired a tissue-resident memory phenotype during the encephalitic stage. CD4 and CD8 T cells were redundant for the invasion of other T cells and parasites into the brain. Early after infection T.b.b. and leukocytes invade different circumventricular organs (brain areas that lack a blood-brain barrier) including the median eminence (ME) located close to sleep-regulating arcuate nucleus (Arc). T.b.b. infection induced the expression of immune and metabolic molecules in this area. Lymphocytes modulated 1) the levels of invading parasites and leukocytes in the ME; 2) the structure of the blood/ hypothalamus interphase and 3) the expression of IFN-γ in the ME/Arc early after infection. Lymphocytes may also be involved in the regulation of sleep alterations observed in African trypanosomiasis.
Collapse
Affiliation(s)
- Gabriela C. Olivera
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden
| | - Leonie Vetter
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden
| | - Chiara Tesoriero
- Department of Neuroscience, Biomedicine and Movement Sciences, University of Verona, Verona, Italy
| | - Federico Del Gallo
- Department of Neuroscience, Biomedicine and Movement Sciences, University of Verona, Verona, Italy
| | - Gustav Hedberg
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden
| | - Juan Basile
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden
| | - Martin E. Rottenberg
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden
- * E-mail:
| |
Collapse
|
13
|
Momoi Y, Tsusaki T, Yamashita H, Takahashi H. The effectiveness of an SNP marker in the cholecystokinin type A receptor gene for improving growth traits in Amakusa Daioh cross chickens. J APPL POULTRY RES 2021. [DOI: 10.1016/j.japr.2021.100166] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022] Open
|
14
|
Jeong JK, Dow SA, Young CN. Sensory Circumventricular Organs, Neuroendocrine Control, and Metabolic Regulation. Metabolites 2021; 11:metabo11080494. [PMID: 34436435 PMCID: PMC8402088 DOI: 10.3390/metabo11080494] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2021] [Revised: 07/13/2021] [Accepted: 07/27/2021] [Indexed: 11/16/2022] Open
Abstract
The central nervous system is critical in metabolic regulation, and accumulating evidence points to a distributed network of brain regions involved in energy homeostasis. This is accomplished, in part, by integrating peripheral and central metabolic information and subsequently modulating neuroendocrine outputs through the paraventricular and supraoptic nucleus of the hypothalamus. However, these hypothalamic nuclei are generally protected by a blood-brain-barrier limiting their ability to directly sense circulating metabolic signals—pointing to possible involvement of upstream brain nuclei. In this regard, sensory circumventricular organs (CVOs), brain sites traditionally recognized in thirst/fluid and cardiovascular regulation, are emerging as potential sites through which circulating metabolic substances influence neuroendocrine control. The sensory CVOs, including the subfornical organ, organum vasculosum of the lamina terminalis, and area postrema, are located outside the blood-brain-barrier, possess cellular machinery to sense the metabolic interior milieu, and establish complex neural networks to hypothalamic neuroendocrine nuclei. Here, evidence for a potential role of sensory CVO-hypothalamic neuroendocrine networks in energy homeostasis is presented.
Collapse
Affiliation(s)
| | | | - Colin N. Young
- Correspondence: ; Tel.: +1-202-994-9575; Fax: +1-202-994-287
| |
Collapse
|
15
|
Butiaeva LI, Slutzki T, Swick HE, Bourguignon C, Robins SC, Liu X, Storch KF, Kokoeva MV. Leptin receptor-expressing pericytes mediate access of hypothalamic feeding centers to circulating leptin. Cell Metab 2021; 33:1433-1448.e5. [PMID: 34129812 DOI: 10.1016/j.cmet.2021.05.017] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/08/2020] [Revised: 02/19/2021] [Accepted: 05/21/2021] [Indexed: 12/18/2022]
Abstract
Knowledge of how leptin receptor (LepR) neurons of the mediobasal hypothalamus (MBH) access circulating leptin is still rudimentary. Employing intravital microscopy, we found that almost half of the blood-vessel-enwrapping pericytes in the MBH express LepR. Selective disruption of pericytic LepR led to increased food intake, increased fat mass, and loss of leptin-dependent signaling in nearby LepR neurons. When delivered intravenously, fluorescently tagged leptin accumulated at hypothalamic LepR pericytes, which was attenuated upon pericyte-specific LepR loss. Because a paracellular tracer was also preferentially retained at LepR pericytes, we pharmacologically targeted regulators of inter-endothelial junction tightness and found that they affect LepR neuronal signaling and food intake. Optical imaging in MBH slices revealed a long-lasting, tonic calcium increase in LepR pericytes in response to leptin, suggesting pericytic contraction and vessel constriction. Together, our data indicate that LepR pericytes facilitate localized, paracellular blood-brain barrier leaks, enabling MBH LepR neurons to access circulating leptin.
Collapse
Affiliation(s)
- Liliia I Butiaeva
- Division of Endocrinology, Department of Medicine, McGill University Health Center Research Institute, McGill University, Montreal QC H4A 3J1, Canada; Integrated Program in Neuroscience, McGill University, Montreal QC H3A 2B4, Canada
| | - Tal Slutzki
- Division of Endocrinology, Department of Medicine, McGill University Health Center Research Institute, McGill University, Montreal QC H4A 3J1, Canada; Integrated Program in Neuroscience, McGill University, Montreal QC H3A 2B4, Canada
| | - Hannah E Swick
- Division of Endocrinology, Department of Medicine, McGill University Health Center Research Institute, McGill University, Montreal QC H4A 3J1, Canada; Integrated Program in Neuroscience, McGill University, Montreal QC H3A 2B4, Canada
| | - Clément Bourguignon
- Department of Psychiatry, Douglas Mental Health University Institute, McGill University, Montreal QC H4H 1R3, Canada; Integrated Program in Neuroscience, McGill University, Montreal QC H3A 2B4, Canada
| | - Sarah C Robins
- Division of Endocrinology, Department of Medicine, McGill University Health Center Research Institute, McGill University, Montreal QC H4A 3J1, Canada
| | - Xiaohong Liu
- Division of Endocrinology, Department of Medicine, McGill University Health Center Research Institute, McGill University, Montreal QC H4A 3J1, Canada
| | - Kai-Florian Storch
- Department of Psychiatry, Douglas Mental Health University Institute, McGill University, Montreal QC H4H 1R3, Canada
| | - Maia V Kokoeva
- Division of Endocrinology, Department of Medicine, McGill University Health Center Research Institute, McGill University, Montreal QC H4A 3J1, Canada.
| |
Collapse
|
16
|
van Galen KA, Ter Horst KW, Serlie MJ. Serotonin, food intake, and obesity. Obes Rev 2021; 22:e13210. [PMID: 33559362 PMCID: PMC8243944 DOI: 10.1111/obr.13210] [Citation(s) in RCA: 39] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/08/2020] [Revised: 12/28/2020] [Accepted: 12/28/2020] [Indexed: 12/16/2022]
Abstract
The role of serotonin in food intake has been studied for decades. Food intake is mainly regulated by two brain circuitries: (i) the homeostatic circuitry, which matches energy intake to energy expenditure, and (ii) the hedonic circuitry, which is involved in rewarding and motivational aspects of energy consumption. In the homeostatic circuitry, serotonergic signaling contributes to the integration of metabolic signals that convey the body's energy status and facilitates the ability to suppress food intake when homeostatic needs have been met. In the hedonic circuitry, serotonergic signaling may reduce reward-related, motivational food consumption. In contrast, peripherally acting serotonin promotes energy absorption and storage. Disturbed serotonergic signaling is associated with obesity, emphasizing the importance to understand the role of serotonergic signaling in food intake. However, unraveling the serotonin-mediated regulation of food intake is complex, as the effects of serotonergic signaling in different brain regions depend on the regional expression of serotonin receptor subtypes and downstream effects via connections to other brain regions. We therefore provide an overview of the effects of serotonergic signaling in brain regions of the homeostatic and hedonic regulatory systems on food intake. Furthermore, we discuss the disturbances in serotonergic signaling in obesity and its potential therapeutic implications.
Collapse
Affiliation(s)
- Katy A van Galen
- Department of Endocrinology and Metabolism, Amsterdam University Medical Centers, Amsterdam, The Netherlands
| | - Kasper W Ter Horst
- Department of Endocrinology and Metabolism, Amsterdam University Medical Centers, Amsterdam, The Netherlands
| | - Mireille J Serlie
- Department of Endocrinology and Metabolism, Amsterdam University Medical Centers, Amsterdam, The Netherlands
| |
Collapse
|
17
|
Ullman JC, Arguello A, Getz JA, Bhalla A, Mahon CS, Wang J, Giese T, Bedard C, Kim DJ, Blumenfeld JR, Liang N, Ravi R, Nugent AA, Davis SS, Ha C, Duque J, Tran HL, Wells RC, Lianoglou S, Daryani VM, Kwan W, Solanoy H, Nguyen H, Earr T, Dugas JC, Tuck MD, Harvey JL, Reyzer ML, Caprioli RM, Hall S, Poda S, Sanchez PE, Dennis MS, Gunasekaran K, Srivastava A, Sandmann T, Henne KR, Thorne RG, Di Paolo G, Astarita G, Diaz D, Silverman AP, Watts RJ, Sweeney ZK, Kariolis MS, Henry AG. Brain delivery and activity of a lysosomal enzyme using a blood-brain barrier transport vehicle in mice. Sci Transl Med 2021; 12:12/545/eaay1163. [PMID: 32461331 DOI: 10.1126/scitranslmed.aay1163] [Citation(s) in RCA: 120] [Impact Index Per Article: 40.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2019] [Revised: 02/10/2020] [Accepted: 04/02/2020] [Indexed: 12/20/2022]
Abstract
Most lysosomal storage diseases (LSDs) involve progressive central nervous system (CNS) impairment, resulting from deficiency of a lysosomal enzyme. Treatment of neuronopathic LSDs remains a considerable challenge, as approved intravenously administered enzyme therapies are ineffective in modifying CNS disease because they do not effectively cross the blood-brain barrier (BBB). We describe a therapeutic platform for increasing the brain exposure of enzyme replacement therapies. The enzyme transport vehicle (ETV) is a lysosomal enzyme fused to an Fc domain that has been engineered to bind to the transferrin receptor, which facilitates receptor-mediated transcytosis across the BBB. We demonstrate that ETV fusions containing iduronate 2-sulfatase (ETV:IDS), the lysosomal enzyme deficient in mucopolysaccharidosis type II, exhibited high intrinsic activity and degraded accumulated substrates in both IDS-deficient cell and in vivo models. ETV substantially improved brain delivery of IDS in a preclinical model of disease, enabling enhanced cellular distribution to neurons, astrocytes, and microglia throughout the brain. Improved brain exposure for ETV:IDS translated to a reduction in accumulated substrates in these CNS cell types and peripheral tissues and resulted in a complete correction of downstream disease-relevant pathologies in the brain, including secondary accumulation of lysosomal lipids, perturbed gene expression, neuroinflammation, and neuroaxonal damage. These data highlight the therapeutic potential of the ETV platform for LSDs and provide preclinical proof of concept for TV-enabled therapeutics to treat CNS diseases more broadly.
Collapse
Affiliation(s)
- Julie C Ullman
- Denali Therapeutics Inc., 161 Oyster Point Blvd., South San Francisco, CA 94080, USA
| | - Annie Arguello
- Denali Therapeutics Inc., 161 Oyster Point Blvd., South San Francisco, CA 94080, USA
| | - Jennifer A Getz
- Denali Therapeutics Inc., 161 Oyster Point Blvd., South San Francisco, CA 94080, USA
| | - Akhil Bhalla
- Denali Therapeutics Inc., 161 Oyster Point Blvd., South San Francisco, CA 94080, USA
| | - Cathal S Mahon
- Denali Therapeutics Inc., 161 Oyster Point Blvd., South San Francisco, CA 94080, USA
| | - Junhua Wang
- Denali Therapeutics Inc., 161 Oyster Point Blvd., South San Francisco, CA 94080, USA
| | - Tina Giese
- Denali Therapeutics Inc., 161 Oyster Point Blvd., South San Francisco, CA 94080, USA
| | - Catherine Bedard
- Denali Therapeutics Inc., 161 Oyster Point Blvd., South San Francisco, CA 94080, USA
| | - Do Jin Kim
- Denali Therapeutics Inc., 161 Oyster Point Blvd., South San Francisco, CA 94080, USA
| | - Jessica R Blumenfeld
- Denali Therapeutics Inc., 161 Oyster Point Blvd., South San Francisco, CA 94080, USA
| | - Nicholas Liang
- Denali Therapeutics Inc., 161 Oyster Point Blvd., South San Francisco, CA 94080, USA
| | - Ritesh Ravi
- Denali Therapeutics Inc., 161 Oyster Point Blvd., South San Francisco, CA 94080, USA
| | - Alicia A Nugent
- Denali Therapeutics Inc., 161 Oyster Point Blvd., South San Francisco, CA 94080, USA
| | - Sonnet S Davis
- Denali Therapeutics Inc., 161 Oyster Point Blvd., South San Francisco, CA 94080, USA
| | - Connie Ha
- Denali Therapeutics Inc., 161 Oyster Point Blvd., South San Francisco, CA 94080, USA
| | - Joseph Duque
- Denali Therapeutics Inc., 161 Oyster Point Blvd., South San Francisco, CA 94080, USA
| | - Hai L Tran
- Denali Therapeutics Inc., 161 Oyster Point Blvd., South San Francisco, CA 94080, USA
| | - Robert C Wells
- Denali Therapeutics Inc., 161 Oyster Point Blvd., South San Francisco, CA 94080, USA
| | - Steve Lianoglou
- Denali Therapeutics Inc., 161 Oyster Point Blvd., South San Francisco, CA 94080, USA
| | - Vinay M Daryani
- Denali Therapeutics Inc., 161 Oyster Point Blvd., South San Francisco, CA 94080, USA
| | - Wanda Kwan
- Denali Therapeutics Inc., 161 Oyster Point Blvd., South San Francisco, CA 94080, USA
| | - Hilda Solanoy
- Denali Therapeutics Inc., 161 Oyster Point Blvd., South San Francisco, CA 94080, USA
| | - Hoang Nguyen
- Denali Therapeutics Inc., 161 Oyster Point Blvd., South San Francisco, CA 94080, USA
| | - Timothy Earr
- Denali Therapeutics Inc., 161 Oyster Point Blvd., South San Francisco, CA 94080, USA
| | - Jason C Dugas
- Denali Therapeutics Inc., 161 Oyster Point Blvd., South San Francisco, CA 94080, USA
| | - Michael D Tuck
- Mass Spectrometry Research Center, Vanderbilt University, 9160 MRB III, 465 21 Avenue South, Nashville, TN 37240, USA
| | - Jennifer L Harvey
- Mass Spectrometry Research Center, Vanderbilt University, 9160 MRB III, 465 21 Avenue South, Nashville, TN 37240, USA
| | - Michelle L Reyzer
- Mass Spectrometry Research Center, Vanderbilt University, 9160 MRB III, 465 21 Avenue South, Nashville, TN 37240, USA
| | - Richard M Caprioli
- Mass Spectrometry Research Center, Vanderbilt University, 9160 MRB III, 465 21 Avenue South, Nashville, TN 37240, USA
| | - Sejal Hall
- Denali Therapeutics Inc., 161 Oyster Point Blvd., South San Francisco, CA 94080, USA
| | - Suresh Poda
- Denali Therapeutics Inc., 161 Oyster Point Blvd., South San Francisco, CA 94080, USA
| | - Pascal E Sanchez
- Denali Therapeutics Inc., 161 Oyster Point Blvd., South San Francisco, CA 94080, USA
| | - Mark S Dennis
- Denali Therapeutics Inc., 161 Oyster Point Blvd., South San Francisco, CA 94080, USA
| | - Kannan Gunasekaran
- Denali Therapeutics Inc., 161 Oyster Point Blvd., South San Francisco, CA 94080, USA
| | - Ankita Srivastava
- Denali Therapeutics Inc., 161 Oyster Point Blvd., South San Francisco, CA 94080, USA
| | - Thomas Sandmann
- Denali Therapeutics Inc., 161 Oyster Point Blvd., South San Francisco, CA 94080, USA
| | - Kirk R Henne
- Denali Therapeutics Inc., 161 Oyster Point Blvd., South San Francisco, CA 94080, USA
| | - Robert G Thorne
- Denali Therapeutics Inc., 161 Oyster Point Blvd., South San Francisco, CA 94080, USA
| | - Gilbert Di Paolo
- Denali Therapeutics Inc., 161 Oyster Point Blvd., South San Francisco, CA 94080, USA
| | - Giuseppe Astarita
- Denali Therapeutics Inc., 161 Oyster Point Blvd., South San Francisco, CA 94080, USA
| | - Dolores Diaz
- Denali Therapeutics Inc., 161 Oyster Point Blvd., South San Francisco, CA 94080, USA
| | - Adam P Silverman
- Denali Therapeutics Inc., 161 Oyster Point Blvd., South San Francisco, CA 94080, USA
| | - Ryan J Watts
- Denali Therapeutics Inc., 161 Oyster Point Blvd., South San Francisco, CA 94080, USA
| | - Zachary K Sweeney
- Denali Therapeutics Inc., 161 Oyster Point Blvd., South San Francisco, CA 94080, USA
| | - Mihalis S Kariolis
- Denali Therapeutics Inc., 161 Oyster Point Blvd., South San Francisco, CA 94080, USA.
| | - Anastasia G Henry
- Denali Therapeutics Inc., 161 Oyster Point Blvd., South San Francisco, CA 94080, USA.
| |
Collapse
|
18
|
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: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 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
|
19
|
Csaba Z, Vitalis T, Charriaut-Marlangue C, Margaill I, Coqueran B, Leger PL, Parente I, Jacquens A, Titomanlio L, Constans C, Demene C, Santin MD, Lehericy S, Perrière N, Glacial F, Auvin S, Tanter M, Ghersi-Egea JF, Adle-Biassette H, Aubry JF, Gressens P, Dournaud P. A simple novel approach for detecting blood-brain barrier permeability using GPCR internalization. Neuropathol Appl Neurobiol 2020; 47:297-315. [PMID: 32898926 PMCID: PMC7891648 DOI: 10.1111/nan.12665] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2020] [Revised: 07/30/2020] [Accepted: 08/22/2020] [Indexed: 01/01/2023]
Abstract
Aims Impairment of blood–brain barrier (BBB) is involved in numerous neurological diseases from developmental to aging stages. Reliable imaging of increased BBB permeability is therefore crucial for basic research and preclinical studies. Today, the analysis of extravasation of exogenous dyes is the principal method to study BBB leakage. However, these procedures are challenging to apply in pups and embryos and may appear difficult to interpret. Here we introduce a novel approach based on agonist‐induced internalization of a neuronal G protein‐coupled receptor widely distributed in the mammalian brain, the somatostatin receptor type 2 (SST2). Methods The clinically approved SST2 agonist octreotide (1 kDa), when injected intraperitoneally does not cross an intact BBB. At sites of BBB permeability, however, OCT extravasates and induces SST2 internalization from the neuronal membrane into perinuclear compartments. This allows an unambiguous localization of increased BBB permeability by classical immunohistochemical procedures using specific antibodies against the receptor. Results We first validated our approach in sensory circumventricular organs which display permissive vascular permeability. Through SST2 internalization, we next monitored BBB opening induced by magnetic resonance imaging‐guided focused ultrasound in murine cerebral cortex. Finally, we proved that after intraperitoneal agonist injection in pregnant mice, SST2 receptor internalization permits analysis of BBB integrity in embryos during brain development. Conclusions This approach provides an alternative and simple manner to assess BBB dysfunction and development in different physiological and pathological conditions.
Collapse
Affiliation(s)
- Z Csaba
- NeuroDiderot, Inserm U1141, Université de Paris, Paris, France
| | - T Vitalis
- NeuroDiderot, Inserm U1141, Université de Paris, Paris, France
| | | | - I Margaill
- Research Team "Pharmacology of Cerebral Circulation" EA4475, Faculté de Pharmacie de Paris, Université de Paris, Paris, France
| | - B Coqueran
- Research Team "Pharmacology of Cerebral Circulation" EA4475, Faculté de Pharmacie de Paris, Université de Paris, Paris, France
| | - P-L Leger
- NeuroDiderot, Inserm U1141, Université de Paris, Paris, France
| | - I Parente
- NeuroDiderot, Inserm U1141, Université de Paris, Paris, France
| | - A Jacquens
- NeuroDiderot, Inserm U1141, Université de Paris, Paris, France
| | - L Titomanlio
- NeuroDiderot, Inserm U1141, Université de Paris, Paris, France
| | - C Constans
- Institut Langevin, ESPCI Paris, PSL Research University, CNRS UMR7587, Inserm U979, Inserm Technology Research Accelerator in Biomedical Ultrasound, Université de Paris, Paris, France
| | - C Demene
- Institut Langevin, ESPCI Paris, PSL Research University, CNRS UMR7587, Inserm U979, Inserm Technology Research Accelerator in Biomedical Ultrasound, Université de Paris, Paris, France
| | - M D Santin
- Brain and Spine Institute-ICM, Center for NeuroImaging Research - CENIR, Sorbonne Paris Cité, UPMC Université Paris 06, Inserm U1127, CNRS UMR 7225, Paris, France
| | - S Lehericy
- Brain and Spine Institute-ICM, Center for NeuroImaging Research - CENIR, Sorbonne Paris Cité, UPMC Université Paris 06, Inserm U1127, CNRS UMR 7225, Paris, France
| | - N Perrière
- BrainPlotting, Brain and Spine Institute-ICM, Paris, France
| | - F Glacial
- BrainPlotting, Brain and Spine Institute-ICM, Paris, France
| | - S Auvin
- NeuroDiderot, Inserm U1141, Université de Paris, Paris, France
| | - M Tanter
- Institut Langevin, ESPCI Paris, PSL Research University, CNRS UMR7587, Inserm U979, Inserm Technology Research Accelerator in Biomedical Ultrasound, Université de Paris, Paris, France
| | - J-F Ghersi-Egea
- Fluid Team, Lyon Neurosciences Research Center, Inserm U1028, CNRS, UMR5292, University Lyon-1, Villeurbanne, France
| | - H Adle-Biassette
- NeuroDiderot, Inserm U1141, Université de Paris, Paris, France.,Service d'Anatomie et de Cytologie Pathologiques, Hôpital Lariboisière, APHP, Paris, France
| | - J-F Aubry
- Institut Langevin, ESPCI Paris, PSL Research University, CNRS UMR7587, Inserm U979, Inserm Technology Research Accelerator in Biomedical Ultrasound, Université de Paris, Paris, France
| | - P Gressens
- NeuroDiderot, Inserm U1141, Université de Paris, Paris, France
| | - P Dournaud
- NeuroDiderot, Inserm U1141, Université de Paris, Paris, France
| |
Collapse
|
20
|
Glucose transporters in brain in health and disease. Pflugers Arch 2020; 472:1299-1343. [PMID: 32789766 PMCID: PMC7462931 DOI: 10.1007/s00424-020-02441-x] [Citation(s) in RCA: 219] [Impact Index Per Article: 54.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2020] [Revised: 07/20/2020] [Accepted: 07/24/2020] [Indexed: 12/15/2022]
Abstract
Energy demand of neurons in brain that is covered by glucose supply from the blood is ensured by glucose transporters in capillaries and brain cells. In brain, the facilitative diffusion glucose transporters GLUT1-6 and GLUT8, and the Na+-d-glucose cotransporters SGLT1 are expressed. The glucose transporters mediate uptake of d-glucose across the blood-brain barrier and delivery of d-glucose to astrocytes and neurons. They are critically involved in regulatory adaptations to varying energy demands in response to differing neuronal activities and glucose supply. In this review, a comprehensive overview about verified and proposed roles of cerebral glucose transporters during health and diseases is presented. Our current knowledge is mainly based on experiments performed in rodents. First, the functional properties of human glucose transporters expressed in brain and their cerebral locations are described. Thereafter, proposed physiological functions of GLUT1, GLUT2, GLUT3, GLUT4, and SGLT1 for energy supply to neurons, glucose sensing, central regulation of glucohomeostasis, and feeding behavior are compiled, and their roles in learning and memory formation are discussed. In addition, diseases are described in which functional changes of cerebral glucose transporters are relevant. These are GLUT1 deficiency syndrome (GLUT1-SD), diabetes mellitus, Alzheimer’s disease (AD), stroke, and traumatic brain injury (TBI). GLUT1-SD is caused by defect mutations in GLUT1. Diabetes and AD are associated with changed expression of glucose transporters in brain, and transporter-related energy deficiency of neurons may contribute to pathogenesis of AD. Stroke and TBI are associated with changes of glucose transporter expression that influence clinical outcome.
Collapse
|
21
|
Navarro VM. Metabolic regulation of kisspeptin - the link between energy balance and reproduction. Nat Rev Endocrinol 2020; 16:407-420. [PMID: 32427949 PMCID: PMC8852368 DOI: 10.1038/s41574-020-0363-7] [Citation(s) in RCA: 115] [Impact Index Per Article: 28.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 04/16/2020] [Indexed: 12/17/2022]
Abstract
Hypothalamic kisspeptin neurons serve as the nodal regulatory centre of reproductive function. These neurons are subjected to a plethora of regulatory factors that ultimately affect the release of kisspeptin, which modulates gonadotropin-releasing hormone (GnRH) release from GnRH neurons to control the reproductive axis. The presence of sufficient energy reserves is critical to achieve successful reproduction. Consequently, metabolic factors impose a very tight control over kisspeptin synthesis and release. This Review offers a synoptic overview of the different steps in which kisspeptin neurons are subjected to metabolic regulation, from early developmental stages to adulthood. We cover an ample array of known mechanisms that underlie the metabolic regulation of KISS1 expression and kisspeptin release. Furthermore, the novel role of kisspeptin neurons as active players within the neuronal circuits that govern energy balance is discussed, offering evidence of a bidirectional role of these neurons as a nexus between metabolism and reproduction.
Collapse
Affiliation(s)
- Víctor M Navarro
- Department of Medicine, Division of Endocrinology, Diabetes and Hypertension, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA.
- Harvard Graduate Program in Neuroscience, Boston, MA, USA.
| |
Collapse
|
22
|
Gosztonyi G, Ludwig H, Bode L, Kao M, Sell M, Petrusz P, Halász B. Obesity induced by Borna disease virus in rats: key roles of hypothalamic fast-acting neurotransmitters and inflammatory infiltrates. Brain Struct Funct 2020; 225:1459-1482. [PMID: 32394093 DOI: 10.1007/s00429-020-02063-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2019] [Accepted: 03/21/2020] [Indexed: 12/30/2022]
Abstract
Human obesity epidemic is increasing worldwide with major adverse consequences on health. Among other possible causes, the hypothesis of an infectious contribution is worth it to be considered. Here, we report on an animal model of virus-induced obesity which might help to better understand underlying processes in human obesity. Eighty Wistar rats, between 30 and 60 days of age, were intracerebrally inoculated with Borna disease virus (BDV-1), a neurotropic negative-strand RNA virus infecting an unusually broad host spectrum including humans. Half of the rats developed fatal encephalitis, while the other half, after 3-4 months, continuously gained weight. At tripled weights, rats were sacrificed by trans-cardial fixative perfusion. Neuropathology revealed prevailing inflammatory infiltrates in the median eminence (ME), progressive degeneration of neurons of the paraventricular nucleus, the entorhinal cortex and the amygdala, and a strikingly high-grade involution of the hippocampus with hydrocephalus. Immune histology revealed that major BDV-1 antigens were preferentially present at glutamatergic receptor sites, while GABAergic areas remained free from BDV-1. Virus-induced suppression of the glutamatergic system caused GABAergic predominance. In the hypothalamus, this shifted the energy balance to the anabolic appetite-stimulating side governed by GABA, allowing for excessive fat accumulation in obese rats. Furthermore, inflammatory infiltrates in the ME and ventro-medial arcuate nucleus hindered free access of appetite-suppressing hormones leptin and insulin. The hormone transport system in hypothalamic areas outside the ME became blocked by excessively produced leptin, leading to leptin resistance. The resulting hyperleptinemic milieu combined with suppressed glutamatergic mechanisms was a characteristic feature of the found metabolic pathology. In conclusion, the study provided clear evidence that BDV-1 induced obesity in the rat model is the result of interdependent structural and functional metabolic changes. They can be explained by an immunologically induced hypothalamic microcirculation-defect, combined with a disturbance of neurotransmitter regulatory systems. The proposed mechanism may also have implications for human health. BDV-1 infection has been frequently found in depressive patients. Independently, comorbidity between depression and obesity has been reported, either. Future studies should address the exciting question of whether BDV-1 infection could be a link, whatsoever, between these two conditions.
Collapse
Affiliation(s)
- Georg Gosztonyi
- Institute of Neuropathology, Charité, University Medicine Berlin, 10117, Berlin, Germany.
| | - Hanns Ludwig
- Freelance Bornavirus Workgroup, 14163, Berlin, Germany
| | - Liv Bode
- Freelance Bornavirus Workgroup, 14163, Berlin, Germany
| | - Moujahed Kao
- Landesbetrieb Hessisches Landeslabor, 35392, Giessen, Germany
| | - Manfred Sell
- Division of Pathology, Martin Luther Hospital, 12351, Berlin, Germany
| | - Peter Petrusz
- Department of Cell and Developmental Biology, University of North Carolina At Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Béla Halász
- Neuromorphological and Neuroendocrine Research Laboratory, Semmelweis University, 1094, Budapest, Hungary
| |
Collapse
|
23
|
Gao J, Li X, Wang Y, Cao Y, Yao D, Sun L, Qin L, Qiu H, Zhan X. Adipocyte-derived extracellular vesicles modulate appetite and weight through mTOR signalling in the hypothalamus. Acta Physiol (Oxf) 2020; 228:e13339. [PMID: 31278836 DOI: 10.1111/apha.13339] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2018] [Revised: 06/29/2019] [Accepted: 07/01/2019] [Indexed: 12/13/2022]
Abstract
AIM Type 2 diabetes and obesity are diseases related to surplus energy in the body. Abnormal interaction between the hypothalamus and adipose tissues is a key trigger of energy metabolism dysfunction. Extracellular vesicles (EVs) regulate intercellular communication by transporting intracellular cargo to recipient cells thereby altering the function of recipient cells. This study aimed to evaluate whether adipocyte-derived EVs can act on hypothalamic neurons to modulate energy intake and to identify the EV-associated non-coding RNAs. METHODS Confocal imaging was used to trace the uptake of labelled adipocyte-derived exosomes by hypothalamic anorexigenic POMC neurons. The effects of adipocyte-derived EVs on the mammalian target of rapamycin (mTOR) signalling pathway in POMC neurons were evaluated based on mRNA and protein expression in vitro using quantitative real-time PCR and western blotting. In addition, adipocyte-derived EVs were injected into recipient mice, and changes in mice body weight and daily food intake were monitored. The biological effects of the EV-associated MALAT1 on POMC neurons were explored. RESULTS Adipocyte-derived EVs were successfully transferred into POMC neurons in vitro. Results showed that adipocytes of obese mice secreted MALAT1-containing EVs, which increased appetite and weight when administered to lean mice. Conversely, adipocyte-derived EVs from lean mice decreased food intake and weight when administered to obese mice. CONCLUSION Adipocyte-derived EVs play important roles in mediating the interaction between adipocytes and hypothalamic neurons. Adipocyte-derived EVs can regulate POMC expression through the hypothalamic mTOR signalling in vivo and in vitro, thereby affecting body energy intake.
Collapse
Affiliation(s)
- Jie Gao
- Department of Endocrinology First Affiliated Hospital of Harbin Medical University Harbin China
| | - Xinyu Li
- Department of Endocrinology First Affiliated Hospital of Harbin Medical University Harbin China
| | - You Wang
- National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics Chinese Academy of Sciences Beijing China
| | - Yan Cao
- Department of Endocrinology First Affiliated Hospital of Harbin Medical University Harbin China
| | - Dengju Yao
- Software and Microelectronics School Harbin University of Science and Technology Harbin China
| | - Lijie Sun
- Department of Endocrinology First Affiliated Hospital of Harbin Medical University Harbin China
| | - Lv Qin
- Department of Endocrinology First Affiliated Hospital of Harbin Medical University Harbin China
| | - Hui Qiu
- Department of Endocrinology First Affiliated Hospital of Harbin Medical University Harbin China
| | - Xiaorong Zhan
- Department of Endocrinology First Affiliated Hospital of Harbin Medical University Harbin China
| |
Collapse
|
24
|
Morita-Takemura S, Wanaka A. Blood-to-brain communication in the hypothalamus for energy intake regulation. Neurochem Int 2019; 128:135-142. [DOI: 10.1016/j.neuint.2019.04.007] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2018] [Revised: 04/08/2019] [Accepted: 04/11/2019] [Indexed: 01/03/2023]
|
25
|
Perello M, Cabral A, Cornejo MP, De Francesco PN, Fernandez G, Uriarte M. Brain accessibility delineates the central effects of circulating ghrelin. J Neuroendocrinol 2019; 31:e12677. [PMID: 30582239 DOI: 10.1111/jne.12677] [Citation(s) in RCA: 54] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/17/2018] [Revised: 12/04/2018] [Accepted: 12/04/2018] [Indexed: 12/27/2022]
Abstract
Ghrelin is a hormone produced in the gastrointestinal tract that acts via the growth hormone secretagogue receptor. In the central nervous system, ghrelin signalling is able to recruit different neuronal targets that regulate the behavioural, neuroendocrine, metabolic and autonomic effects of the hormone. Notably, several studies using radioactive or fluorescent variants of ghrelin have found that the accessibility of circulating ghrelin into the mouse brain is both strikingly low and restricted to some specific brain areas. A variety of studies addressing central effects of systemically injected ghrelin in mice have also provided indirect evidence that the accessibility of plasma ghrelin into the brain is limited. Here, we review these previous observations and discuss the putative pathways that would allow plasma ghrelin to gain access into the brain together with their physiological implications. Additionally, we discuss some potential features regarding the accessibility of plasma ghrelin into the human brain based on the observations reported by studies that investigate the consequences of ghrelin administration to humans.
Collapse
Affiliation(s)
- Mario Perello
- Laboratorio de Neurofisiología del Instituto Multidisciplinario de Biología Celular, Consejo Nacional de Investigaciones Científicas y Técnicas de Argentina, Universidad Nacional de La Plata y Comisión de Investigaciones Científicas-Provincia de Buenos Aires, Buenos Aires, Argentina
| | - Agustina Cabral
- Laboratorio de Neurofisiología del Instituto Multidisciplinario de Biología Celular, Consejo Nacional de Investigaciones Científicas y Técnicas de Argentina, Universidad Nacional de La Plata y Comisión de Investigaciones Científicas-Provincia de Buenos Aires, Buenos Aires, Argentina
| | - María P Cornejo
- Laboratorio de Neurofisiología del Instituto Multidisciplinario de Biología Celular, Consejo Nacional de Investigaciones Científicas y Técnicas de Argentina, Universidad Nacional de La Plata y Comisión de Investigaciones Científicas-Provincia de Buenos Aires, Buenos Aires, Argentina
| | - Pablo N De Francesco
- Laboratorio de Neurofisiología del Instituto Multidisciplinario de Biología Celular, Consejo Nacional de Investigaciones Científicas y Técnicas de Argentina, Universidad Nacional de La Plata y Comisión de Investigaciones Científicas-Provincia de Buenos Aires, Buenos Aires, Argentina
| | - Gimena Fernandez
- Laboratorio de Neurofisiología del Instituto Multidisciplinario de Biología Celular, Consejo Nacional de Investigaciones Científicas y Técnicas de Argentina, Universidad Nacional de La Plata y Comisión de Investigaciones Científicas-Provincia de Buenos Aires, Buenos Aires, Argentina
| | - Maia Uriarte
- Laboratorio de Neurofisiología del Instituto Multidisciplinario de Biología Celular, Consejo Nacional de Investigaciones Científicas y Técnicas de Argentina, Universidad Nacional de La Plata y Comisión de Investigaciones Científicas-Provincia de Buenos Aires, Buenos Aires, Argentina
| |
Collapse
|
26
|
Abstract
Animal models are valuable for the study of complex behaviours and physiology such as the control of appetite because genetic, pharmacological and surgical approaches allow the investigation of underlying mechanisms. However, the majority of such studies are carried out in just two species, laboratory mice and rats. These conventional laboratory species have been intensely selected for high growth rate and fecundity, and have a high metabolic rate and short lifespan. These aspects limit their translational relevance for human appetite control. This review will consider the value of studies carried out in a seasonal species, the Siberian hamster, which shows natural photoperiod-regulated annual cycles in appetite, growth and fattening. Such studies reveal that this long-term control is not simply an adjustment of the known hypothalamic neuronal systems that control hunger and satiety in the short term. Long-term cyclicity is probably driven by hypothalamic tanycytes, glial cells that line the ventricular walls of the hypothalamus. These unique cells sense nutrients and metabolic hormones, integrate seasonal signals and effect plasticity of surrounding neural circuits through their function as a stem cell niche in the adult. Studies of glial cell function in the hypothalamus offer new potential for identifying central targets for appetite and body weight control amenable to dietary or pharmacological manipulation.
Collapse
|
27
|
Zorec R, Županc TA, Verkhratsky A. Astrogliopathology in the infectious insults of the brain. Neurosci Lett 2018; 689:56-62. [PMID: 30096375 DOI: 10.1016/j.neulet.2018.08.003] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2018] [Revised: 08/03/2018] [Accepted: 08/04/2018] [Indexed: 12/28/2022]
Abstract
Astroglia, a heterogeneous type of neuroglia, play key homeostatic functions in the central nervous system (CNS) and represent an important defence system. Impaired homeostatic capacity of astrocytes manifests in diseases and this is mirrored in various astrocyte-based pathological features including reactive astrogliosis, astrodegeneration with astroglial atrophy and pathological remodelling of astrocytes. All of these manifestations are most prominently associated with infectious insults, mediated by bacteria, protozoa and viruses. Here we focus onto neurotropic viruses such as tick-borne encephalitis (TBEV) and Zika virus (ZIKV), both belonging to Flaviviridae and both causing severe neurological impairments. We argue that astrocytes provide a route through which neurotropic infectious agents attack the CNS, since they are anatomically associated with the blood-brain barrier and exhibit aerobic glycolysis, a metabolic specialisation of highly morphologically dynamic cells, which may provide a suitable metabolic milieu for proliferation of infectious agents, including viral bodies.
Collapse
Affiliation(s)
- Robert Zorec
- University of Ljubljana, Institute of Pathophysiology, Laboratory of Neuroendocrinology and Molecular Cell Physiology, Zaloska cesta 4, SI-1000, Ljubljana, Slovenia; Celica, BIOMEDICAL, Technology Park 24, 1000 Ljubljana, Slovenia
| | - Tatjana Avšič Županc
- Institute of Microbiology and Immunology, Faculty of Medicine, University of Ljubljana, Zaloška 4, 1000 Ljubljana, Slovenia
| | - Alexei Verkhratsky
- University of Ljubljana, Institute of Pathophysiology, Laboratory of Neuroendocrinology and Molecular Cell Physiology, Zaloska cesta 4, SI-1000, Ljubljana, Slovenia; Celica, BIOMEDICAL, Technology Park 24, 1000 Ljubljana, Slovenia; Center for Basic and Translational Neuroscience, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen 2200, Denmark; Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, M13 9PT, UK; Achucarro Center for Neuroscience, IKERBASQUE, Basque Foundation for Science, 48011 Bilbao, Spain.
| |
Collapse
|
28
|
Prevot V, Dehouck B, Sharif A, Ciofi P, Giacobini P, Clasadonte J. The Versatile Tanycyte: A Hypothalamic Integrator of Reproduction and Energy Metabolism. Endocr Rev 2018; 39:333-368. [PMID: 29351662 DOI: 10.1210/er.2017-00235] [Citation(s) in RCA: 147] [Impact Index Per Article: 24.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/01/2017] [Accepted: 01/12/2018] [Indexed: 12/16/2022]
Abstract
The fertility and survival of an individual rely on the ability of the periphery to promptly, effectively, and reproducibly communicate with brain neural networks that control reproduction, food intake, and energy homeostasis. Tanycytes, a specialized glial cell type lining the wall of the third ventricle in the median eminence of the hypothalamus, appear to act as the linchpin of these processes by dynamically controlling the secretion of neuropeptides into the portal vasculature by hypothalamic neurons and regulating blood-brain and blood-cerebrospinal fluid exchanges, both processes that depend on the ability of these cells to adapt their morphology to the physiological state of the individual. In addition to their barrier properties, tanycytes possess the ability to sense blood glucose levels, and play a fundamental and active role in shuttling circulating metabolic signals to hypothalamic neurons that control food intake. Moreover, accumulating data suggest that, in keeping with their putative descent from radial glial cells, tanycytes are endowed with neural stem cell properties and may respond to dietary or reproductive cues by modulating hypothalamic neurogenesis. Tanycytes could thus constitute the missing link in the loop connecting behavior, hormonal changes, signal transduction, central neuronal activation and, finally, behavior again. In this article, we will examine these recent advances in the understanding of tanycytic plasticity and function in the hypothalamus and the underlying molecular mechanisms. We will also discuss the putative involvement and therapeutic potential of hypothalamic tanycytes in metabolic and fertility disorders.
Collapse
Affiliation(s)
- Vincent Prevot
- Inserm, Laboratory of Development and Plasticity of the Neuroendocrine Brain, Jean-Pierre Aubert Research Center, Lille, France.,University of Lille, FHU 1000 Days for Health, School of Medicine, Lille, France
| | - Bénédicte Dehouck
- Inserm, Laboratory of Development and Plasticity of the Neuroendocrine Brain, Jean-Pierre Aubert Research Center, Lille, France.,University of Lille, FHU 1000 Days for Health, School of Medicine, Lille, France
| | - Ariane Sharif
- Inserm, Laboratory of Development and Plasticity of the Neuroendocrine Brain, Jean-Pierre Aubert Research Center, Lille, France.,University of Lille, FHU 1000 Days for Health, School of Medicine, Lille, France
| | - Philippe Ciofi
- Inserm, Neurocentre Magendie, Bordeaux, France.,Université de Bordeaux, Bordeaux, France
| | - Paolo Giacobini
- Inserm, Laboratory of Development and Plasticity of the Neuroendocrine Brain, Jean-Pierre Aubert Research Center, Lille, France.,University of Lille, FHU 1000 Days for Health, School of Medicine, Lille, France
| | - Jerome Clasadonte
- Inserm, Laboratory of Development and Plasticity of the Neuroendocrine Brain, Jean-Pierre Aubert Research Center, Lille, France.,University of Lille, FHU 1000 Days for Health, School of Medicine, Lille, France
| |
Collapse
|
29
|
Abstract
While there is a growing consensus that insulin has diverse and important regulatory actions on the brain, seemingly important aspects of brain insulin physiology are poorly understood. Examples include: what is the insulin concentration within brain interstitial fluid under normal physiologic conditions; whether insulin is made in the brain and acts locally; does insulin from the circulation cross the blood-brain barrier or the blood-CSF barrier in a fashion that facilitates its signaling in brain; is insulin degraded within the brain; do privileged areas with a "leaky" blood-brain barrier serve as signaling nodes for transmitting peripheral insulin signaling; does insulin action in the brain include regulation of amyloid peptides; whether insulin resistance is a cause or consequence of processes involved in cognitive decline. Heretofore, nearly all of the studies examining brain insulin physiology have employed techniques and methodologies that do not appreciate the complex fluid compartmentation and flow throughout the brain. This review attempts to provide a status report on historical and recent work that begins to address some of these issues. It is undertaken in an effort to suggest a framework for studies going forward. Such studies are inevitably influenced by recent physiologic and genetic studies of insulin accessing and acting in brain, discoveries relating to brain fluid dynamics and the interplay of cerebrospinal fluid, brain interstitial fluid, and brain lymphatics, and advances in clinical neuroimaging that underscore the dynamic role of neurovascular coupling.
Collapse
Affiliation(s)
- Sarah M Gray
- Department of Pharmacology, Department of Medicine, University of Virginia, School of Medicine , Charlottesville, Virginia
| | - Eugene J Barrett
- Department of Pharmacology, Department of Medicine, University of Virginia, School of Medicine , Charlottesville, Virginia.,Division of Endocrinology, Department of Medicine, University of Virginia, School of Medicine , Charlottesville, Virginia
| |
Collapse
|
30
|
Rivera HM, Stincic TL. Estradiol and the control of feeding behavior. Steroids 2018; 133:44-52. [PMID: 29180290 PMCID: PMC5864536 DOI: 10.1016/j.steroids.2017.11.011] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/02/2017] [Revised: 11/20/2017] [Accepted: 11/21/2017] [Indexed: 12/21/2022]
Abstract
This review lays out the evidence for the role of E2 in homeostatic and hedonic feeding across several species. While significant effort has been expended on homeostatic feeding research, more studies for hedonic feeding need to be conducted (i.e. are there increases in meal size and enhanced motivation to natural food rewards). By identifying the underlying neural circuitry involved, one can better delineate the mechanisms by which E2 influences feeding behavior. By utilizing more selective neural targeting techniques, such as optogenetics, significant progress can be made toward this goal. Together, behavioral and physiological techniques will help us to better understand neural deficits that can increase the risk for obesity in the absence of E2 (menopause) and aid in developing therapeutic strategies.
Collapse
Affiliation(s)
- H M Rivera
- Department of Physiology and Pharmacology, Oregon Health and Science University, Portland, OR 97239, USA
| | - T L Stincic
- Department of Physiology and Pharmacology, Oregon Health and Science University, Portland, OR 97239, USA.
| |
Collapse
|
31
|
Fioramonti X, Chrétien C, Leloup C, Pénicaud L. Recent Advances in the Cellular and Molecular Mechanisms of Hypothalamic Neuronal Glucose Detection. Front Physiol 2017; 8:875. [PMID: 29184506 PMCID: PMC5694446 DOI: 10.3389/fphys.2017.00875] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2017] [Accepted: 10/18/2017] [Indexed: 11/18/2022] Open
Abstract
The hypothalamus have been recognized for decades as one of the major brain centers for the control of energy homeostasis. This area contains specialized neurons able to detect changes in nutrients level. Among them, glucose-sensing neurons use glucose as a signaling molecule in addition to its fueling role. In this review we will describe the different sub-populations of glucose-sensing neurons present in the hypothalamus and highlight their nature in terms of neurotransmitter/neuropeptide expression. This review will particularly discuss whether pro-opiomelanocortin (POMC) neurons from the arcuate nucleus are directly glucose-sensing. In addition, recent observations in glucose-sensing suggest a subtle system with different mechanisms involved in the detection of changes in glucose level and their involvement in specific physiological functions. Several data point out the critical role of reactive oxygen species (ROS) and mitochondria dynamics in the detection of increased glucose. This review will also highlight that ATP-dependent potassium (KATP) channels are not the only channels mediating glucose-sensing and discuss the new role of transient receptor potential canonical channels (TRPC). We will discuss the recent advances in the determination of glucose-sensing machinery and propose potential line of research needed to further understand the regulation of brain glucose detection.
Collapse
Affiliation(s)
- Xavier Fioramonti
- NutriNeuro, Institut National de la Recherche Agronomique, Université de Bordeaux, Bordeaux, France.,Centre des Sciences du Goût et de l'Alimentation, Centre National de la Recherche Scientifique, Institut National de la Recherche Agronomique, Université Bourgogne Franche-Comté, Dijon, France
| | - Chloé Chrétien
- Centre des Sciences du Goût et de l'Alimentation, Centre National de la Recherche Scientifique, Institut National de la Recherche Agronomique, Université Bourgogne Franche-Comté, Dijon, France
| | - Corinne Leloup
- Centre des Sciences du Goût et de l'Alimentation, Centre National de la Recherche Scientifique, Institut National de la Recherche Agronomique, Université Bourgogne Franche-Comté, Dijon, France
| | - Luc Pénicaud
- Centre des Sciences du Goût et de l'Alimentation, Centre National de la Recherche Scientifique, Institut National de la Recherche Agronomique, Université Bourgogne Franche-Comté, Dijon, France.,Stromalab, Centre National de la Recherche Scientifique, Institut National de la Santé et de la Recherche Médicale, Université de Toulouse, Toulouse, France
| |
Collapse
|
32
|
NamKoong C, Kim MS, Jang BT, Lee YH, Cho YM, Choi HJ. Central administration of GLP-1 and GIP decreases feeding in mice. Biochem Biophys Res Commun 2017; 490:247-252. [DOI: 10.1016/j.bbrc.2017.06.031] [Citation(s) in RCA: 50] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2017] [Accepted: 06/09/2017] [Indexed: 12/18/2022]
|
33
|
Kohno D. Sweet taste receptor in the hypothalamus: a potential new player in glucose sensing in the hypothalamus. J Physiol Sci 2017; 67:459-465. [PMID: 28378265 PMCID: PMC10717116 DOI: 10.1007/s12576-017-0535-y] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2017] [Accepted: 03/24/2017] [Indexed: 01/06/2023]
Abstract
The hypothalamic feeding center plays an important role in energy homeostasis. The feeding center senses the systemic energy status by detecting hormone and nutrient levels for homeostatic regulation, resulting in the control of food intake, heat production, and glucose production and uptake. The concentration of glucose is sensed by two types of glucose-sensing neurons in the feeding center: glucose-excited neurons and glucose-inhibited neurons. Previous studies have mainly focused on glucose metabolism as the mechanism underlying glucose sensing. Recent studies have indicated that receptor-mediated pathways also play a role in glucose sensing. This review describes sweet taste receptors in the hypothalamus and explores the role of sweet taste receptors in energy homeostasis.
Collapse
Affiliation(s)
- Daisuke Kohno
- Advanced Scientific Research Leaders Development Unit, Gunma University, 3-39-15 Showa-machi, Maebashi, Gunma, 371-8512, Japan.
- Metabolic Signal Research Center, Institute for Molecular and Cellular Regulation, Gunma University, Maebashi, 371-8512, Japan.
| |
Collapse
|
34
|
Miller GD. Appetite Regulation: Hormones, Peptides, and Neurotransmitters and Their Role in Obesity. Am J Lifestyle Med 2017; 13:586-601. [PMID: 31662725 DOI: 10.1177/1559827617716376] [Citation(s) in RCA: 51] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2017] [Revised: 05/18/2017] [Accepted: 05/31/2017] [Indexed: 12/29/2022] Open
Abstract
Understanding body weight regulation will aid in the development of new strategies to combat obesity. This review examines energy homeostasis and food intake behaviors, specifically with regards to hormones, peptides, and neurotransmitters in the periphery and central nervous system, and their potential role in obesity. Dysfunction in feeding signals by the brain is a factor in obesity. The hypothalamic (arcuate nucleus) and brainstem (nucleus tractus solitaris) areas integrate behavioral, endocrine, and autonomic responses via afferent and efferent pathways from and to the brainstem and peripheral organs. Neurons present in the arcuate nucleus express pro-opiomelanocortin, Neuropeptide Y, and Agouti Related Peptide, with the former involved in lowering food intake, and the latter two acutely increasing feeding behaviors. Action of peripheral hormones from the gut, pancreas, adipose, and liver are also involved in energy homeostasis. Vagal afferent neurons are also important in regulating energy homeostasis. Peripheral signals respond to the level of stored and currently available fuel. By studying their actions, new agents maybe developed that disable orexigenic responses and enhance anorexigenic signals. Although there are relatively few medications currently available for obesity treatment, a number of agents are in development that work through these pathways.
Collapse
Affiliation(s)
- Gary D Miller
- Department of Health and Exercise Science, Wake Forest University, Winston-Salem, North Carolina
| |
Collapse
|
35
|
Chrétien C, Fenech C, Liénard F, Grall S, Chevalier C, Chaudy S, Brenachot X, Berges R, Louche K, Stark R, Nédélec E, Laderrière A, Andrews ZB, Benani A, Flockerzi V, Gascuel J, Hartmann J, Moro C, Birnbaumer L, Leloup C, Pénicaud L, Fioramonti X. Transient Receptor Potential Canonical 3 (TRPC3) Channels Are Required for Hypothalamic Glucose Detection and Energy Homeostasis. Diabetes 2017; 66:314-324. [PMID: 27899482 DOI: 10.2337/db16-1114] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/14/2016] [Accepted: 11/17/2016] [Indexed: 11/13/2022]
Abstract
The mediobasal hypothalamus (MBH) contains neurons capable of directly detecting metabolic signals such as glucose to control energy homeostasis. Among them, glucose-excited (GE) neurons increase their electrical activity when glucose rises. In view of previous work, we hypothesized that transient receptor potential canonical type 3 (TRPC3) channels are involved in hypothalamic glucose detection and the control of energy homeostasis. To investigate the role of TRPC3, we used constitutive and conditional TRPC3-deficient mouse models. Hypothalamic glucose detection was studied in vivo by measuring food intake and insulin secretion in response to increased brain glucose level. The role of TRPC3 in GE neuron response to glucose was studied by using in vitro calcium imaging on freshly dissociated MBH neurons. We found that whole-body and MBH TRPC3-deficient mice have increased body weight and food intake. The anorectic effect of intracerebroventricular glucose and the insulin secretory response to intracarotid glucose injection are blunted in TRPC3-deficient mice. TRPC3 loss of function or pharmacological inhibition blunts calcium responses to glucose in MBH neurons in vitro. Together, the results demonstrate that TRPC3 channels are required for the response to glucose of MBH GE neurons and the central effect of glucose on insulin secretion and food intake.
Collapse
Affiliation(s)
- Chloé Chrétien
- Centre des Sciences du Goût et de l'Alimentation, CNRS, Institut National de la Recherche Agronomique, University of Bourgogne Franche-Comté, Dijon, France
| | - Claire Fenech
- Centre des Sciences du Goût et de l'Alimentation, CNRS, Institut National de la Recherche Agronomique, University of Bourgogne Franche-Comté, Dijon, France
| | - Fabienne Liénard
- Centre des Sciences du Goût et de l'Alimentation, CNRS, Institut National de la Recherche Agronomique, University of Bourgogne Franche-Comté, Dijon, France
| | - Sylvie Grall
- Centre des Sciences du Goût et de l'Alimentation, CNRS, Institut National de la Recherche Agronomique, University of Bourgogne Franche-Comté, Dijon, France
| | - Charlène Chevalier
- Centre des Sciences du Goût et de l'Alimentation, CNRS, Institut National de la Recherche Agronomique, University of Bourgogne Franche-Comté, Dijon, France
| | - Sylvie Chaudy
- Centre des Sciences du Goût et de l'Alimentation, CNRS, Institut National de la Recherche Agronomique, University of Bourgogne Franche-Comté, Dijon, France
| | - Xavier Brenachot
- Centre des Sciences du Goût et de l'Alimentation, CNRS, Institut National de la Recherche Agronomique, University of Bourgogne Franche-Comté, Dijon, France
| | - Raymond Berges
- Centre des Sciences du Goût et de l'Alimentation, CNRS, Institut National de la Recherche Agronomique, University of Bourgogne Franche-Comté, Dijon, France
| | - Katie Louche
- INSERM UMR1048, Institute of Metabolic and Cardiovascular Diseases, Obesity Research Laboratory, University of Toulouse, Toulouse, France
| | - Romana Stark
- Biomedicine Discovery Institute, Metabolic Disease and Obesity Program, Department of Physiology, Monash University, Clayton, VIC, Australia
| | - Emmanuelle Nédélec
- Centre des Sciences du Goût et de l'Alimentation, CNRS, Institut National de la Recherche Agronomique, University of Bourgogne Franche-Comté, Dijon, France
| | - Amélie Laderrière
- Centre des Sciences du Goût et de l'Alimentation, CNRS, Institut National de la Recherche Agronomique, University of Bourgogne Franche-Comté, Dijon, France
| | - Zane B Andrews
- Biomedicine Discovery Institute, Metabolic Disease and Obesity Program, Department of Physiology, Monash University, Clayton, VIC, Australia
| | - Alexandre Benani
- Centre des Sciences du Goût et de l'Alimentation, CNRS, Institut National de la Recherche Agronomique, University of Bourgogne Franche-Comté, Dijon, France
| | - Veit Flockerzi
- Experimental and Clinical Pharmacology and Toxicology, Saarland University School of Medicine, Homburg, Germany
| | - Jean Gascuel
- Centre des Sciences du Goût et de l'Alimentation, CNRS, Institut National de la Recherche Agronomique, University of Bourgogne Franche-Comté, Dijon, France
| | - Jana Hartmann
- Institute of Neuroscience and Center for Integrated Protein Science, Technical University Munich, Munich, Germany
| | - Cédric Moro
- INSERM UMR1048, Institute of Metabolic and Cardiovascular Diseases, Obesity Research Laboratory, University of Toulouse, Toulouse, France
| | - Lutz Birnbaumer
- Neurobiology Laboratory, National Institute of Environmental Health Sciences, Research Triangle Park, NC
- Institute of Biomedical Research, Catholic University of Argentina, Buenos Aires, Argentina
| | - Corinne Leloup
- Centre des Sciences du Goût et de l'Alimentation, CNRS, Institut National de la Recherche Agronomique, University of Bourgogne Franche-Comté, Dijon, France
| | - Luc Pénicaud
- Centre des Sciences du Goût et de l'Alimentation, CNRS, Institut National de la Recherche Agronomique, University of Bourgogne Franche-Comté, Dijon, France
| | - Xavier Fioramonti
- Centre des Sciences du Goût et de l'Alimentation, CNRS, Institut National de la Recherche Agronomique, University of Bourgogne Franche-Comté, Dijon, France
| |
Collapse
|
36
|
Tran DQ, Tse EK, Kim MH, Belsham DD. Diet-induced cellular neuroinflammation in the hypothalamus: Mechanistic insights from investigation of neurons and microglia. Mol Cell Endocrinol 2016; 438:18-26. [PMID: 27208620 DOI: 10.1016/j.mce.2016.05.015] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/28/2016] [Accepted: 05/17/2016] [Indexed: 12/31/2022]
Abstract
Diet-induced obesity can lead to detrimental chronic disorders. The severity of this global epidemic has encouraged ongoing research to characterize the mechanisms underlying obesity and its comorbidities. Recent evidence suggests that saturated fatty acids (SFA) in high-fat diets rapidly generate inflammation in the arcuate nucleus of the hypothalamus (ARC), which centrally regulates whole-body energy homeostasis. Herein, we will review the roles of hypothalamic neurons and resident microglia in the initiation of SFA-induced hypothalamic inflammation. Particularly, we focus on neuronal and microglial free fatty acid-sensing and capacity to produce inflammatory signaling. We also outline a potential role of peripherally-derived monocytes in this inflammation. And finally, we explore synaptic plasticity as a mechanism through which hypothalamic inflammation can modulate ARC circuitry, and thus disrupt energy homeostasis.
Collapse
Affiliation(s)
- Dean Q Tran
- Department of Physiology, University of Toronto, Toronto, ON, Canada
| | - Erika K Tse
- Department of Physiology, University of Toronto, Toronto, ON, Canada
| | - Mun Heui Kim
- Department of Physiology, University of Toronto, Toronto, ON, Canada
| | - Denise D Belsham
- Department of Physiology, University of Toronto, Toronto, ON, Canada; Department of Obstetrics and Gynaecology, University of Toronto, Toronto, ON, Canada; Department of Medicine, University of Toronto, Toronto, ON, Canada.
| |
Collapse
|
37
|
Kohno D, Koike M, Ninomiya Y, Kojima I, Kitamura T, Yada T. Sweet Taste Receptor Serves to Activate Glucose- and Leptin-Responsive Neurons in the Hypothalamic Arcuate Nucleus and Participates in Glucose Responsiveness. Front Neurosci 2016; 10:502. [PMID: 27877104 PMCID: PMC5099526 DOI: 10.3389/fnins.2016.00502] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2016] [Accepted: 10/21/2016] [Indexed: 01/04/2023] Open
Abstract
The hypothalamic feeding center plays an important role in energy homeostasis. In the feeding center, whole-body energy signals including hormones and nutrients are sensed, processed, and integrated. As a result, food intake and energy expenditure are regulated. Two types of glucose-sensing neurons exist in the hypothalamic arcuate nucleus (ARC): glucose-excited neurons and glucose-inhibited neurons. While some molecules are known to be related to glucose sensing in the hypothalamus, the mechanisms underlying glucose sensing in the hypothalamus are not fully understood. The sweet taste receptor is a heterodimer of taste type 1 receptor 2 (T1R2) and taste type 1 receptor 3 (T1R3) and senses sweet tastes. T1R2 and T1R3 are distributed in multiple organs including the tongue, pancreas, adipose tissue, and hypothalamus. However, the role of sweet taste receptors in the ARC remains to be clarified. To examine the role of sweet taste receptors in the ARC, cytosolic Ca2+ concentration ([Ca2+]i) in isolated single ARC neurons were measured using Fura-2 fluorescent imaging. An artificial sweetener, sucralose at 10−5–10−2 M dose dependently increased [Ca2+]i in 12–16% of ARC neurons. The sucralose-induced [Ca2+]i increase was suppressed by a sweet taste receptor inhibitor, gurmarin. The sucralose-induced [Ca2+]i increase was inhibited under an extracellular Ca2+-free condition and in the presence of an L-type Ca2+ channel blocker, nitrendipine. Sucralose-responding neurons were activated by high-concentration of glucose. This response to glucose was markedly suppressed by gurmarin. More than half of sucralose-responding neurons were activated by leptin but not ghrelin. Percentages of proopiomelanocortin (POMC) neurons among sucralose-responding neurons and sweet taste receptor expressing neurons were low, suggesting that majority of sucralose-responding neurons are non-POMC neurons. These data suggest that sweet taste receptor-mediated cellular activation mainly occurs on non-POMC leptin-responding neurons and contributes to glucose responding. Endogenous sweet molecules including glucose may regulate energy homeostasis through sweet taste receptors on glucose-and leptin-responsive neurons in the ARC.
Collapse
Affiliation(s)
- Daisuke Kohno
- Advanced Scientific Research Leaders Development Unit, Gunma UniversityMaebashi, Japan; Metabolic Signal Research Center, Institute for Molecular and Cellular Regulation, Gunma UniversityMaebashi, Japan
| | - Miho Koike
- Advanced Scientific Research Leaders Development Unit, Gunma University Maebashi, Japan
| | - Yuzo Ninomiya
- Division of Sensory Physiology, Research and Development Center for Taste and Odor Sensing, Kyushu UniversityFukuoka, Japan; Monell Chemical Senses CenterPhiladelphia, PA, USA
| | - Itaru Kojima
- Department of Cell Biology, Institute for Molecular and Cellular Regulation, Gunma University Maebashi, Japan
| | - Tadahiro Kitamura
- Metabolic Signal Research Center, Institute for Molecular and Cellular Regulation, Gunma University Maebashi, Japan
| | - Toshihiko Yada
- Division of Integrative Physiology, Department of Physiology, School of Medicine, Jichi Medical University Shimotsuke, Japan
| |
Collapse
|
38
|
Oakley AE, Steiner RA, Chavkin C, Clifton DK, Ferrara LK, Reed SD. κ Agonists as a novel therapy for menopausal hot flashes. Menopause 2016; 22:1328-34. [PMID: 25988798 DOI: 10.1097/gme.0000000000000476] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
OBJECTIVE The etiology of postmenopausal hot flashes is poorly understood, making it difficult to develop and target ideal therapies. A network of hypothalamic estrogen-sensitive neurons producing kisspeptin, neurokinin B and dynorphin-called KNDy neurons-are located adjacent to the thermoregulatory center. KNDy neurons regulate pulsatile secretion of gonadotropin-releasing hormone (GnRH) and luteinizing hormone (LH). Dynorphin may inhibit this system by binding κ opioid receptors within the vicinity of KNDy neurons. We hypothesize that hot flashes are reduced by KNDy neuron manipulation. METHODS A double-blind, cross-over, placebo-controlled pilot study evaluated the effects of a κ agonist. Hot flash frequency was the primary outcome. Twelve healthy postmenopausal women with moderate to severe hot flashes (aged 48-60 y) were randomized. Eight women with sufficient baseline hot flashes for statistical analysis completed all three interventions: placebo, standard-dose pentazocine/naloxone (50/0.5 mg), or low-dose pentazocine/naloxone (25/0.25 mg). In an inpatient research setting, each participant received the three interventions, in randomized order, on three separate days. On each day, an intravenous catheter was inserted for LH blood sampling, and skin conductance and Holter monitors were placed. Subjective hot flash frequency and severity were recorded. RESULTS The mean (SEM) hot flash frequency 2 to 7 hours after therapy initiation was lower than that for placebo (standard-dose κ agonist, 4.75 [0.67] hot flashes per 5 h; low-dose κ agonist, 4.50 [0.57] hot flashes per 5 h; placebo, 5.94 [0.78] hot flashes per 5 h; P = 0.025). Hot flash intensity did not vary between interventions. LH pulsatility mirrored objective hot flashes in some--but not all--women. CONCLUSIONS This pilot study suggests that κ agonists may affect menopausal vasomotor symptoms.
Collapse
Affiliation(s)
- Amy E Oakley
- 1Department of Physiology and Biophysics, University of Washington, Seattle, WA 2Department of Obstetrics and Gynecology, University of Washington, Seattle, WA 3Department of Pharmacology, University of Washington, Seattle, WA
| | | | | | | | | | | |
Collapse
|
39
|
Gumbs MC, van den Heuvel JK, la Fleur SE. The effect of obesogenic diets on brain Neuropeptide Y. Physiol Behav 2016; 162:161-73. [DOI: 10.1016/j.physbeh.2016.04.049] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2015] [Revised: 03/18/2016] [Accepted: 04/26/2016] [Indexed: 12/18/2022]
|
40
|
Kim DW, Glendining KA, Grattan DR, Jasoni CL. Maternal Obesity in the Mouse Compromises the Blood-Brain Barrier in the Arcuate Nucleus of Offspring. Endocrinology 2016; 157:2229-42. [PMID: 27054554 DOI: 10.1210/en.2016-1014] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
The arcuate nucleus (ARC) regulates body weight in response to blood-borne signals of energy balance. Blood-brain barrier (BBB) permeability in the ARC is determined by capillary endothelial cells (ECs) and tanycytes. Tight junctions between ECs limit paracellular entry of blood-borne molecules into the brain, whereas EC transporters and fenestrations regulate transcellular entry. Tanycytes appear to form a barrier that prevents free diffusion of blood-borne molecules. Here we tested the hypothesis that gestation in an obese mother alters BBB permeability in the ARC of offspring. A maternal high-fat diet model was used to generate offspring from normal-weight (control) and obese dams (OffOb). Evans Blue diffusion into the ARC was higher in OffOb compared with controls, indicating that ARC BBB permeability was altered. Vessels investing the ARC in OffOb had more fenestrations than controls, although the total number of vessels was not changed. A reduced number of tanycytic processes in the ARC of OffOb was also observed. The putative transporters, Lrp1 and dysferlin, were up-regulated and tight junction components were differentially expressed in OffOb compared with controls. These data suggest that maternal obesity during pregnancy can compromise BBB formation in the fetus, leading to altered BBB function in the ARC after birth.
Collapse
Affiliation(s)
- Dong Won Kim
- Centre for Neuroendocrinology, Department of Anatomy, University of Otago School of Medical Sciences, Dunedin 9054, New Zealand
| | - Kelly A Glendining
- Centre for Neuroendocrinology, Department of Anatomy, University of Otago School of Medical Sciences, Dunedin 9054, New Zealand
| | - David R Grattan
- Centre for Neuroendocrinology, Department of Anatomy, University of Otago School of Medical Sciences, Dunedin 9054, New Zealand
| | - Christine L Jasoni
- Centre for Neuroendocrinology, Department of Anatomy, University of Otago School of Medical Sciences, Dunedin 9054, New Zealand
| |
Collapse
|
41
|
Knudsen LB, Secher A, Hecksher-Sørensen J, Pyke C. Long-acting glucagon-like peptide-1 receptor agonists have direct access to and effects on pro-opiomelanocortin/cocaine- and amphetamine-stimulated transcript neurons in the mouse hypothalamus. J Diabetes Investig 2016; 7 Suppl 1:56-63. [PMID: 27186357 PMCID: PMC4854506 DOI: 10.1111/jdi.12463] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/14/2015] [Accepted: 12/21/2015] [Indexed: 12/17/2022] Open
Abstract
Liraglutide is a glucagon‐like peptide‐1 receptor (GLP‐1R) agonist marketed for the treatment of type 2 diabetes. Besides lowering blood glucose, liraglutide reduces bodyweight, and has recently also been approved for the obesity indication. Acutely, GLP‐1 markedly reduces gastric emptying, and this effect was previously believed to at least partly explain the effect on bodyweight loss. However, recent studies in both humans and animals have shown that GLP‐1R agonists, such as liraglutide, that lead to pharmacological concentrations for 24 h/day only have a minor effect on gastric emptying; such an effect is unlikely to have lasting effects on appetite reduction. Liraglutide has been shown to have direct effects in the arcuate nucleus of the rodent brain, activating pro‐opiomelanocortin neurons and increasing levels of the cocaine‐ and amphetamine‐stimulated transcript neuropeptide messenger ribonucleic acid, which correlate nicely to clinical studies where liraglutide was shown to increase feelings of satiety. However, despite the lack of a GLP‐1R on agouti‐related peptide/neuropeptide Y neurons, liraglutide also was able to prevent a hunger associated increase in agouti‐related peptide and neuropeptide Y neuropeptide messenger ribonucleic acid, again with a strong correlation to clinical studies that document reduced hunger feelings in patients while taking liraglutide. Studies using fluorescent labeled liraglutide, as well as other GLP‐1R agonists, and analysis using single‐plane illumination microscopy show that such medium‐sized peptide‐based compounds can directly access not only circumventricular organs of the brain, but also directly access discrete regions in the hypothalamus. The direct effects of long‐acting GLP‐1R agonists in the hypothalamus are likely to be an important new pathway in understanding GLP‐1R agonist mediated weight loss.
Collapse
Affiliation(s)
| | - Anna Secher
- Novo Nordisk A/S Novo Nordisk Park Maaloev Denmark
| | | | - Charles Pyke
- Novo Nordisk A/S Novo Nordisk Park Maaloev Denmark
| |
Collapse
|
42
|
Wilson JL, Enriori PJ. A talk between fat tissue, gut, pancreas and brain to control body weight. Mol Cell Endocrinol 2015; 418 Pt 2:108-19. [PMID: 26316427 DOI: 10.1016/j.mce.2015.08.022] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/20/2015] [Revised: 08/19/2015] [Accepted: 08/20/2015] [Indexed: 12/18/2022]
Abstract
The incidence of obesity and its related disorders are increasing at a rate of pandemic proportions. Understanding the mechanisms behind the maintenance of energy balance is fundamental in developing treatments for clinical syndromes including obesity and diabetes. A neural network located in the nucleus of the solitary tract-area postrema complex in the hindbrain and the hypothalamus in the forebrain has long been implicated in the control of energy balance. In the hypothalamus this central neuronal network consists of small populations of nuclei with distinct functions such as the arcuate nucleus (ARH), the paraventricular nuclei of the hypothalamus (PVH), the dorsomedial (DMH), the ventromedial (VMH) and the lateral hypothalamus (LH). These hypothalamic areas form interconnected neuronal circuits that respond to fluctuations in energy status by altering the expression of neuropeptides, leading to changes in energy intake and expenditure. Regulation of these hypothalamic nuclei involves the actions of orexigenic peptides (ie ghrelin), which act to stimulate energy intake and decrease energy expenditure, and anorexigenic peptides (ie. leptin and insulin), which act to reduce energy intake and stimulate energy expenditure. Here we review the role of the ARH, DMH and PVH in the control of energy homeostasis and how recent advances in research technologies (Cre-loxP technology, optogenetics and pharmacogenetics) have shed light on the role of these hypothalamic nuclei in the control of energy balance. Such novel findings include the implication of ARH POMC and AgRP neurons in the browning of white adipose tissue to regulate energy expenditure as well as the likely existence of divergent hypothalamic pathways in the DMH and PVH in the control of food intake and energy expenditure.
Collapse
Affiliation(s)
- Jenny L Wilson
- Department of Physiology, Monash Obesity & Diabetes Institute, Monash University, Clayton, Victoria 3800, Australia
| | - Pablo J Enriori
- Department of Physiology, Monash Obesity & Diabetes Institute, Monash University, Clayton, Victoria 3800, Australia.
| |
Collapse
|
43
|
Abstract
The endocrine hypothalamus constitutes those cells which project to the median eminence and secrete neurohormones into the hypophysial portal blood to act on cells of the anterior pituitary gland. The entire endocrine system is controlled by these peptides. In turn, the hypothalamic neuroendocrine cells are regulated by feedback signals from the endocrine glands and other circulating factors. The neuroendocrine cells are found in specific regions of the hypothalamus and are regulated by afferents from higher brain centers. Integrated function is clearly complex and the networks between and amongst the neuroendocrine cells allows fine control to achieve homeostasis. The entry of hormones and other factors into the brain, either via the cerebrospinal fluid or through fenestrated capillaries (in the basal hypothalamus) is important because it influences the extent to which feedback regulation may be imposed. Recent evidence of the passage of factors from the pars tuberalis and the median eminence casts a new layer in our understanding of neuroendocrine regulation. The function of neuroendocrine cells and the means by which pulsatile secretion is achieved is best understood for the close relationship between gonadotropin releasing hormone and luteinizing hormone, which is reviewed in detail. The secretion of other neurohormones is less rigid, so the relationship between hypothalamic secretion and the relevant pituitary hormones is more complex.
Collapse
Affiliation(s)
- I J Clarke
- Monash University, Department of Physiology, Clayton, Australia
| |
Collapse
|
44
|
Hoveyda HR, Fraser GL, Roy MO, Dutheuil G, Batt F, El Bousmaqui M, Korac J, Lenoir F, Lapin A, Noël S, Blanc S. Discovery and optimization of novel antagonists to the human neurokinin-3 receptor for the treatment of sex-hormone disorders (Part I). J Med Chem 2015; 58:3060-82. [PMID: 25738882 DOI: 10.1021/jm5017413] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Neurokinin-3 receptor (NK3R) has recently emerged as important in modulating the tonic pulsatile gonadotropin-releasing hormone (GnRH) release. We therefore decided to explore NK3R antagonists as therapeutics for sex-hormone disorders that can potentially benefit from lowering GnRH pulsatility with consequent diminished levels of plasma luteinizing hormone (LH) and correspondingly attenuated levels of circulating androgens and estrogens. The discovery and lead optimization of a novel N-acyl-triazolopiperazine NK3R antagonist chemotype achieved through bioisosteric lead change from the high-throughput screening (HTS) hit is described. A concomitant improvement in the antagonist bioactivity and ligand lipophilic efficiency (LLE) parameter were the principal guidelines in the lead optimization efforts. Examples of advanced lead analogues to demonstrate the amenability of this chemotype to achieving a suitable pharmacokinetic (PK) profile are provided as well as pharmacokinetic-pharmacodynamic (PKPD) correlations to analyze the trends observed for LH inhibition in castrated rats and monkeys that served as preliminary in vivo efficacy models.
Collapse
Affiliation(s)
- Hamid R Hoveyda
- Euroscreen SA, 47 Rue Adrienne Bolland, 6041 Gosselies, Belgium
| | - Graeme L Fraser
- Euroscreen SA, 47 Rue Adrienne Bolland, 6041 Gosselies, Belgium
| | - Marie-Odile Roy
- Euroscreen SA, 47 Rue Adrienne Bolland, 6041 Gosselies, Belgium
| | | | - Frédéric Batt
- Euroscreen SA, 47 Rue Adrienne Bolland, 6041 Gosselies, Belgium
| | | | - Julien Korac
- Euroscreen SA, 47 Rue Adrienne Bolland, 6041 Gosselies, Belgium
| | - François Lenoir
- Euroscreen SA, 47 Rue Adrienne Bolland, 6041 Gosselies, Belgium
| | - Alexey Lapin
- Euroscreen SA, 47 Rue Adrienne Bolland, 6041 Gosselies, Belgium
| | - Sophie Noël
- Euroscreen SA, 47 Rue Adrienne Bolland, 6041 Gosselies, Belgium
| | - Sébastien Blanc
- Euroscreen SA, 47 Rue Adrienne Bolland, 6041 Gosselies, Belgium
| |
Collapse
|
45
|
Marić G, Gazibara T, Zaletel I, Labudović Borović M, Tomanović N, Ćirić M, Puškaš N. The role of gut hormones in appetite regulation (review). ACTA ACUST UNITED AC 2015; 101:395-407. [PMID: 25532952 DOI: 10.1556/aphysiol.101.2014.4.1] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Eating process is an aggregate of complex and different forms of behavior. Its regulation is based on energy homeostasis and appetite control which includes two components: the homeostatic and the hedonistic control. Important signals in appetite regulation are gut-derived hormones. They are produced by enteroendocrine cells in response to nutrient and energy intake, and achieve their effects by influencing brain structures involved in food intake regulation. The key brain structure involved in this process is the hypothalamus. Gut hormones reach the hypothalamus from the circulation or by the vagal nerve via the nucleus of the solitary tract. Among gut peptides, ghrelin is the only orexigenic hormone, leading to an increase in food intake and body weight. All others, such as cholecystokinin, glucagon like peptide-1, oxyntomodulin, peptide tyrosine tyrosine or pancreatic polypeptide, are anorexigenic, leading to decrease in food intake. Also, gut-derived endocannabinoids exert orexigenic effect on appetite. Keeping in mind the growing problem of obesity, the crucial issue when considering gut derived peptides is to understand their mechanisms of acting because of potential role in clinical therapy, and discovering long-lasting gut peptides or their analogues, with no or minimal side effects.
Collapse
Affiliation(s)
- G Marić
- University of Belgrade Institute of Epidemiology, Faculty of Medicine Belgrade Serbia
| | - T Gazibara
- University of Belgrade Institute of Epidemiology, Faculty of Medicine Belgrade Serbia
| | - I Zaletel
- University of Belgrade Institute of Histology and Embryology, Faculty of Medicine Višegradska 26 11000 Belgrade Serbia
| | - M Labudović Borović
- University of Belgrade Institute of Histology and Embryology, Faculty of Medicine Višegradska 26 11000 Belgrade Serbia
| | - N Tomanović
- University of Belgrade Institute of Pathology, Faculty of Medicine Belgrade Serbia
| | - M Ćirić
- University of Nis Institute of Physiology, Faculty of Medicine Nis Serbia
| | - Nela Puškaš
- University of Belgrade Institute of Histology and Embryology, Faculty of Medicine Višegradska 26 11000 Belgrade Serbia
| |
Collapse
|
46
|
Rysz J, Gluba-Brzózka A, Mikhailidis DP, Banach M. Fibroblast growth factor 19-targeted therapies for the treatment of metabolic disease. Expert Opin Investig Drugs 2015; 24:603-10. [PMID: 25604607 DOI: 10.1517/13543784.2015.1006357] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
INTRODUCTION Fibroblast growth factors (FGFs) belong to the FGF superfamily with diverse biological functions, including proliferation, cellular differentiation, wound repair, angiogenesis and tumorigenesis. The ability to reduce liver fat content and concentrations of triglycerides, total cholesterol and plasma glucose, and to improve sensitivity and limit pro-lipogenic properties of insulin, makes FGF19 a promising therapeutic target for the treatment of metabolic syndrome. FGF19 regulates bile acid biosynthesis in the bile duct, glucose metabolism and vitamin D and phosphate homeostasis, raises the metabolic rate, reduces body weight, and ameliorates diabetes in mice. The therapeutic potential of FGF19 to treat metabolic disorders has been widely studied in animal models, but currently there are no reports concerning its use in humans. AREAS COVERED The following article highlights the metabolic effects and mechanism of action of FGF19. It also discusses the potential therapies that target FGF19. EXPERT OPINION FGF19 is emerging as a new target for the therapy of metabolic disorders, including diabetes. The results obtained from animal models are promising. However, there is still much to be done before the translation of these effects into practice will be possible.
Collapse
Affiliation(s)
- Jacek Rysz
- WAM University Hospital of Lodz, Department of Nephrology, Hypertension and Family Medicine , Zeromskiego 113, 90-549 Lodz , Poland +48 42 639 37 50 ; +48 42 639 37 50 ;
| | | | | | | |
Collapse
|
47
|
Bouret S, Levin BE, Ozanne SE. Gene-environment interactions controlling energy and glucose homeostasis and the developmental origins of obesity. Physiol Rev 2015; 95:47-82. [PMID: 25540138 PMCID: PMC4281588 DOI: 10.1152/physrev.00007.2014] [Citation(s) in RCA: 97] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Obesity and type 2 diabetes mellitus (T2DM) often occur together and affect a growing number of individuals in both the developed and developing worlds. Both are associated with a number of other serious illnesses that lead to increased rates of mortality. There is likely a polygenic mode of inheritance underlying both disorders, but it has become increasingly clear that the pre- and postnatal environments play critical roles in pushing predisposed individuals over the edge into a disease state. This review focuses on the many genetic and environmental variables that interact to cause predisposed individuals to become obese and diabetic. The brain and its interactions with the external and internal environment are a major focus given the prominent role these interactions play in the regulation of energy and glucose homeostasis in health and disease.
Collapse
Affiliation(s)
- Sebastien Bouret
- The Saban Research Institute, Neuroscience Program, Childrens Hospital Los Angeles, University of Southern California, Los Angeles, California; Inserm U837, Jean-Pierre Aubert Research Center, University Lille 2, Lille, France; Neurology Service, Veterans Administration Medical Center, East Orange, New Jersey; Department of Neurology and Neurosciences, Rutgers, New Jersey Medical School, Newark, New Jersey; and University of Cambridge Institute of Metabolic Science and MRC Metabolic Diseases Unit, Cambridge, United Kingdom
| | - Barry E Levin
- The Saban Research Institute, Neuroscience Program, Childrens Hospital Los Angeles, University of Southern California, Los Angeles, California; Inserm U837, Jean-Pierre Aubert Research Center, University Lille 2, Lille, France; Neurology Service, Veterans Administration Medical Center, East Orange, New Jersey; Department of Neurology and Neurosciences, Rutgers, New Jersey Medical School, Newark, New Jersey; and University of Cambridge Institute of Metabolic Science and MRC Metabolic Diseases Unit, Cambridge, United Kingdom
| | - Susan E Ozanne
- The Saban Research Institute, Neuroscience Program, Childrens Hospital Los Angeles, University of Southern California, Los Angeles, California; Inserm U837, Jean-Pierre Aubert Research Center, University Lille 2, Lille, France; Neurology Service, Veterans Administration Medical Center, East Orange, New Jersey; Department of Neurology and Neurosciences, Rutgers, New Jersey Medical School, Newark, New Jersey; and University of Cambridge Institute of Metabolic Science and MRC Metabolic Diseases Unit, Cambridge, United Kingdom
| |
Collapse
|
48
|
Langlet F. [Role of tanycytes within the blood-hypothalamus interface]. Biol Aujourdhui 2014; 208:225-235. [PMID: 25474004 DOI: 10.1051/jbio/2014025] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2014] [Indexed: 06/04/2023]
Abstract
Information exchanges between the brain and the periphery are key stages in the regulation of various physiological functions. The mediobasal hypothalamus, which ensures a large part of these functions, must be permanently informed about the physiological state of the body to guarantee the maintaining of homeostasis. For that purpose, it possesses a peculiar blood-brain interface due to the presence of specialized glial cells called tanycytes. This review describes the organization of the blood-hypothalamus interface and characterizes the peculiar place of tanycytes within it, as well as their striking capacity to remodel their own interface in order to ensure the regulation of various physiological functions.
Collapse
Affiliation(s)
- Fanny Langlet
- Inserm, Centre de recherche Jean-Pierre Aubert, U837, Développement et plasticité du cerveau post-natal, 59000 Lille, France - Université de Lille, Faculté de médecine, Institut de Médecine Prédictive et de Recherche Thérapeutique, 59000 Lille, France
| |
Collapse
|
49
|
Langlet F. Tanycytes: a gateway to the metabolic hypothalamus. J Neuroendocrinol 2014; 26:753-60. [PMID: 25131689 DOI: 10.1111/jne.12191] [Citation(s) in RCA: 64] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/12/2014] [Revised: 08/01/2014] [Accepted: 08/01/2014] [Indexed: 01/07/2023]
Abstract
The central regulation of energy balance relies on the ability of the brain to promptly and efficiently sense variations of metabolic state. To achieve this, circulating hormonal and metabolic signals have to cross the blood-brain interface, where unusual glial cells named tanycytes have been described to play a key role in this process. Tanycytes are specialised polarised ependymoglial cells that line the floor of the third ventricle and send a single process to contact hypothalamic neurones and blood vessels. Although their role in the regulation of energy balance via the modulation of neuronal activity or their chemosensitivity has been already described, recent studies ascribe a new function to tanycytes in the regulation of energy homeostasis as a result of their capacity to regulate the access of metabolic signals to the hypothalamus. This review discusses the peculiar place of tanycytes within the blood-hypothalamus interface, as well as a striking capacity to remodel their own interface to ensure an adaptive metabolic response to energy imbalances.
Collapse
Affiliation(s)
- F Langlet
- Inserm, Jean-Pierre Aubert Research Centre, U837, Development and Plasticity of the Postnatal Brain, Lille, France; UDSL, School of Medicine, Lille, France; Université de Lille, Institut de Médecine Prédictive et de Recherche Thérapeutique, Lille, France
| |
Collapse
|
50
|
Secher A, Jelsing J, Baquero AF, Hecksher-Sørensen J, Cowley MA, Dalbøge LS, Hansen G, Grove KL, Pyke C, Raun K, Schäffer L, Tang-Christensen M, Verma S, Witgen BM, Vrang N, Bjerre Knudsen L. The arcuate nucleus mediates GLP-1 receptor agonist liraglutide-dependent weight loss. J Clin Invest 2014; 124:4473-88. [PMID: 25202980 DOI: 10.1172/jci75276] [Citation(s) in RCA: 586] [Impact Index Per Article: 58.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2014] [Accepted: 07/31/2014] [Indexed: 12/13/2022] Open
Abstract
Liraglutide is a glucagon-like peptide-1 (GLP-1) analog marketed for the treatment of type 2 diabetes. Besides lowering blood glucose, liraglutide also reduces body weight. It is not fully understood how liraglutide induces weight loss or to what degree liraglutide acts directly in the brain. Here, we determined that liraglutide does not activate GLP-1-producing neurons in the hindbrain, and liraglutide-dependent body weight reduction in rats was independent of GLP-1 receptors (GLP-1Rs) in the vagus nerve, area postrema, and paraventricular nucleus. Peripheral injection of fluorescently labeled liraglutide in mice revealed the presence of the drug in the circumventricular organs. Moreover, labeled liraglutide bound neurons within the arcuate nucleus (ARC) and other discrete sites in the hypothalamus. GLP-1R was necessary for liraglutide uptake in the brain, as liraglutide binding was not seen in Glp1r(-/-) mice. In the ARC, liraglutide was internalized in neurons expressing proopiomelanocortin (POMC) and cocaine- and amphetamine-regulated transcript (CART). Electrophysiological measurements of murine brain slices revealed that GLP-1 directly stimulates POMC/CART neurons and indirectly inhibits neurotransmission in neurons expressing neuropeptide Y (NPY) and agouti-related peptide (AgRP) via GABA-dependent signaling. Collectively, our findings indicate that the GLP-1R on POMC/CART-expressing ARC neurons likely mediates liraglutide-induced weight loss.
Collapse
|