1
|
Pistolesi A, Tuniz S, Luceri C, Molli A, Urru M, La Rocca AI, Tanturli M, De Cesaris F, Buonvicino D, Chiarugi A. Anti-CGRP Monoclonal Antibodies Counteract Establishment of Food Aversive Memories and Chemotherapy-induced Anorexia and Weight Loss. Pharmacol Res 2025:107818. [PMID: 40490094 DOI: 10.1016/j.phrs.2025.107818] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/02/2025] [Revised: 05/29/2025] [Accepted: 06/05/2025] [Indexed: 06/11/2025]
Abstract
Glutamatergic neurons of external lateral parabrachial nucleus co-expressing calcitonin gene related peptide (elPBCGRP) negatively regulate food intake and establish food aversive memories. They also promote malaise-dependent anorexia, sickness behavior and tumor cachexia. In spite of the pathogenetic potential of elPBCGRP neurons in numerous human disorders, whether they can be targeted with drugs inhibiting CGRP-dependent neuromodulation remains unknown. We report that systemically administered anti-CGRP mAbs recently approved for migraine prevention reach the rat brain and counteract CGRP-regulated food aversive memories as well as chemotherapy-induced anorexia and weight loss. Anti-CGRP antibodies also partially reduce fear responses, but are unable to prevent anorexia by the GLP-1 agonist liraglutide. Data disclose the therapeutic potential of anti-CGRP monoclonals to treatment of eating disorders, including tumor treatment-associated syndromes such as chemotherapy induced nausea and vomiting or cachexia.
Collapse
Affiliation(s)
- Alessandra Pistolesi
- Section of Clinical Pharmacology and Oncology, Department of Health Sciences, University of Florence, Florence, Italy
| | - Simone Tuniz
- Section of Clinical Pharmacology and Oncology, Department of Health Sciences, University of Florence, Florence, Italy
| | - Cristina Luceri
- Department of NEUROFARBA, Division of Pharmacology and Toxicology, University of Florence, Italy
| | - Alice Molli
- Section of Clinical Pharmacology and Oncology, Department of Health Sciences, University of Florence, Florence, Italy
| | - Matteo Urru
- Section of Clinical Pharmacology and Oncology, Department of Health Sciences, University of Florence, Florence, Italy
| | - Antonino Iurato La Rocca
- Department of NEUROFARBA, Division of Pharmacology and Toxicology, University of Florence, Italy
| | - Michele Tanturli
- Department of Experimental and Clinical Biomedical Sciences "Mario Serio", University of Florence, Italy
| | - Francesco De Cesaris
- Headache Center and Clinical Pharmacology Unit, Careggi University Hospital, Florence, Italy
| | - Daniela Buonvicino
- Section of Clinical Pharmacology and Oncology, Department of Health Sciences, University of Florence, Florence, Italy
| | - Alberto Chiarugi
- Section of Clinical Pharmacology and Oncology, Department of Health Sciences, University of Florence, Florence, Italy; Headache Center and Clinical Pharmacology Unit, Careggi University Hospital, Florence, Italy.
| |
Collapse
|
2
|
Scarlett JM, Hwang E, Richardson NE, Bryan CL, Redford I, Quah E, Odderson ETR, Choi PP, Hwang MK, Phan BA, Kadlec K, Alonge KM, Morton GJ, Williams KW, Schwartz MW. A key role for parabrachial nucleus CGRP neurons in FGF1-Induced anorexia. Mol Metab 2025; 96:102138. [PMID: 40189100 DOI: 10.1016/j.molmet.2025.102138] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/31/2025] [Revised: 03/26/2025] [Accepted: 04/01/2025] [Indexed: 04/14/2025] Open
Abstract
In addition to sustained glucose lowering, centrally administered fibroblast growth factor 1 (FGF1) induces a potent but transient anorexia in animal models of type 2 diabetes. To investigate the mechanism(s) underlying this anorexic response, the current work focused on a specific neuronal subset located in the external lateral subdivision of the parabrachial nucleus marked by the expression of calcitonin gene-related peptide (elPBNCGRP neurons). These neurons can be activated by withdrawal of upstream GABAergic inhibitory input and are implicated as mediators of the adaptive response (including anorexia) to a wide range of aversive stimuli. To determine if FGF1-induced anorexia is associated with elPBNCGRP neuron activation, we employed adult male CalcaCre:GFP/+ transgenic mice in which GFP is fused to Cre recombinase driven by the CGRP-encoding gene Calca. Here, we show that FGF1 activates elPBNCGRP neurons, both after intracerebroventricular (icv) injection in vivo and when applied ex vivo in a slice preparation, and that the mechanism underlying this effect depends upon reduced GABAergic input from neurons lying upstream. Consistent with this interpretation, we report that the anorexic response to icv FGF1 is reduced by ∼70% when elPBNCGRP neurons are silenced using chemogenetics. Last, we report that effects of icv FGF1 injection on both elPBNCGRP neuron activity and food intake are strongly attenuated by systemic administration of the GABAA receptor agonist Bretazenil. We conclude that in adult male mice, elPBNCGRP neuron activation is a key mediator of FGF1-induced anorexia, and that this activation response is mediated at least in part by withdrawal of GABAergic inhibition.
Collapse
Affiliation(s)
- Jarrad M Scarlett
- University of Washington Medicine Diabetes Institute, Department of Medicine, Seattle, WA, 98109, USA; Department of Pediatric Gastroenterology and Hepatology, Seattle Children's Hospital, Seattle, WA, 98145, USA
| | - Eunsang Hwang
- Center for Hypothalamic Research, Department of Internal Medicine, Peter O'Donnell Jr. Brain Institute, the University of Texas Southwestern Medical Center at Dallas, Dallas, TX, USA
| | - Nicole E Richardson
- University of Washington Medicine Diabetes Institute, Department of Medicine, Seattle, WA, 98109, USA
| | - Caeley L Bryan
- University of Washington Medicine Diabetes Institute, Department of Medicine, Seattle, WA, 98109, USA
| | - Ingrid Redford
- University of Washington Medicine Diabetes Institute, Department of Medicine, Seattle, WA, 98109, USA
| | - Emily Quah
- University of Washington Medicine Diabetes Institute, Department of Medicine, Seattle, WA, 98109, USA
| | - Erik Tyr R Odderson
- University of Washington Medicine Diabetes Institute, Department of Medicine, Seattle, WA, 98109, USA
| | - Pique P Choi
- University of Washington Medicine Diabetes Institute, Department of Medicine, Seattle, WA, 98109, USA
| | - Matthew K Hwang
- University of Washington Medicine Diabetes Institute, Department of Medicine, Seattle, WA, 98109, USA
| | - Bao Anh Phan
- University of Washington Medicine Diabetes Institute, Department of Medicine, Seattle, WA, 98109, USA
| | - Kelly Kadlec
- University of Washington Medicine Diabetes Institute, Department of Medicine, Seattle, WA, 98109, USA
| | - Kimberly M Alonge
- University of Washington Medicine Diabetes Institute, Department of Medicine, Seattle, WA, 98109, USA; Department of Medicinal Chemistry, University of Washington, Seattle, WA, 98115, USA
| | - Gregory J Morton
- University of Washington Medicine Diabetes Institute, Department of Medicine, Seattle, WA, 98109, USA
| | - Kevin W Williams
- Center for Hypothalamic Research, Department of Internal Medicine, Peter O'Donnell Jr. Brain Institute, the University of Texas Southwestern Medical Center at Dallas, Dallas, TX, USA.
| | - Michael W Schwartz
- University of Washington Medicine Diabetes Institute, Department of Medicine, Seattle, WA, 98109, USA
| |
Collapse
|
3
|
Teixidor-Deulofeu J, Blid Sköldheden S, Font-Gironès F, Feješ A, Ruud J, Engström Ruud L. Semaglutide effects on energy balance are mediated by Adcyap1 + neurons in the dorsal vagal complex. Cell Metab 2025:S1550-4131(25)00256-6. [PMID: 40409256 DOI: 10.1016/j.cmet.2025.04.018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/14/2024] [Revised: 02/10/2025] [Accepted: 04/29/2025] [Indexed: 05/25/2025]
Abstract
The use of the GLP-1R agonist semaglutide is revolutionizing the treatment of obesity, yet its mechanistic effects on energy balance remain elusive. Here, we demonstrate that reactivation of semaglutide-responsive dorsal vagal complex neurons mimics the drug's effects of reducing food intake and body weight and promoting fat utilization and conditioned taste aversion. We observe that many of the semaglutide-activated area postrema (AP) and nucleus of the solitary tract (NTS) neurons express Adcyap1 mRNA, and ablation of AP/NTS Adcyap1+ neurons largely reverses semaglutide's effects on energy balance acutely in lean mice and in subchronically treated obese mice. Semaglutide-activated AP/NTS Adcyap1+ neurons promote the loss of fat rather than lean mass, with only a modest effect on conditioned taste aversion. Furthermore, NTS Adcyap1+ neurons are engaged by GLP-1R-expressing AP neurons and are necessary for semaglutide-induced activation of several downstream satiety-related structures. Selective targeting of semaglutide-responsive Adcyap1+ neurons holds potential for improved future anti-obesity treatments.
Collapse
Affiliation(s)
- Júlia Teixidor-Deulofeu
- Department of Physiology, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, 41390 Gothenburg, Sweden
| | - Sebastian Blid Sköldheden
- Department of Physiology, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, 41390 Gothenburg, Sweden
| | - Ferran Font-Gironès
- Department of Physiology, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, 41390 Gothenburg, Sweden
| | - Andrej Feješ
- Department of Physiology, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, 41390 Gothenburg, Sweden; Institute of Molecular Biomedicine, Faculty of Medicine, Comenius University, 83303 Bratislava, Slovakia
| | - Johan Ruud
- Department of Physiology, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, 41390 Gothenburg, Sweden
| | - Linda Engström Ruud
- Department of Physiology, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, 41390 Gothenburg, Sweden.
| |
Collapse
|
4
|
Liu CM, Killion EA, Hammoud R, Lu SC, Komorowski R, Liu T, Kanke M, Thomas VA, Cook K, Sivits GN, Ben AB, Atangan LI, Hussien R, Tang A, Shkumatov A, Li CM, Drucker DJ, Véniant MM. GIPR-Ab/GLP-1 peptide-antibody conjugate requires brain GIPR and GLP-1R for additive weight loss in obese mice. Nat Metab 2025:10.1038/s42255-025-01295-w. [PMID: 40301582 DOI: 10.1038/s42255-025-01295-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/16/2024] [Accepted: 03/28/2025] [Indexed: 05/01/2025]
Abstract
Glucose-dependent insulinotropic polypeptide receptor (GIPR) and glucagon-like peptide 1 receptor (GLP-1R) are expressed in the central nervous system (CNS) and regulate food intake. Here, we demonstrate that a peptide-antibody conjugate that blocks GIPR while simultaneously activating GLP-1R (GIPR-Ab/GLP-1) requires both CNS GIPR and CNS GLP-1R for maximal weight loss in obese, primarily male, mice. Moreover, dulaglutide produces greater weight loss in CNS GIPR knockout (KO) mice, and the weight loss achieved with dulaglutide + GIPR-Ab is attenuated in CNS GIPR KO mice. Wild-type mice treated with GIPR-Ab/GLP-1 and CNS GIPR KO mice exhibit similar changes in gene expression related to tissue remodelling, lipid metabolism and inflammation in white adipose tissue and liver. Moreover, GIPR-Ab/GLP-1 is detected in circumventricular organs in the brain and activates c-FOS in downstream neural substrates involved in appetite regulation. Hence, both CNS GIPR and GLP-1R signalling are required for the full weight loss effect of a GIPR-Ab/GLP-1 peptide-antibody conjugate.
Collapse
Affiliation(s)
- Clarissa M Liu
- Department of Cardiometabolic Disorders, Amgen Research, Thousand Oaks, CA, USA
- Amgen R&D Postdoctoral Fellows Program, Amgen Inc., Thousand Oaks, CA, USA
| | - Elizabeth A Killion
- Department of Cardiometabolic Disorders, Amgen Research, Thousand Oaks, CA, USA
| | - Rola Hammoud
- The Department of Medicine, Lunenfeld-Tanenbaum Research Institute, Mt. Sinai Hospital, University of Toronto, Toronto, Ontario, Canada
| | - Shu-Chen Lu
- Department of Cardiometabolic Disorders, Amgen Research, Thousand Oaks, CA, USA
| | - Renee Komorowski
- Department of Cardiometabolic Disorders, Amgen Research, Thousand Oaks, CA, USA
| | - Tongyu Liu
- Center for Research Acceleration by Digital Innovation, Amgen Research, Thousand Oaks, CA, USA
| | - Matt Kanke
- Department of Research Technologies, Amgen Research, South San Francisco, CA, USA
| | - Veena A Thomas
- Department of Pharmacokinetics and Drug Metabolism, Amgen Research, South San Francisco, CA, USA
| | - Kevin Cook
- Department of Pharmacokinetics and Drug Metabolism, Amgen Research, South San Francisco, CA, USA
| | - Glenn N Sivits
- Department of Cardiometabolic Disorders, Amgen Research, Thousand Oaks, CA, USA
| | - Aerielle B Ben
- Department of Cardiometabolic Disorders, Amgen Research, Thousand Oaks, CA, USA
| | - Larissa I Atangan
- Department of Cardiometabolic Disorders, Amgen Research, Thousand Oaks, CA, USA
| | - Rajaa Hussien
- Department of Translational Safety and Bioanalytical Sciences, Amgen Research, South San Francisco, CA, USA
| | - Amy Tang
- Department of Translational Safety and Bioanalytical Sciences, Amgen Research, South San Francisco, CA, USA
| | - Artem Shkumatov
- Department of Translational Safety and Bioanalytical Sciences, Amgen Research, South San Francisco, CA, USA
| | - Chi-Ming Li
- Department of Research Technologies, Amgen Research, South San Francisco, CA, USA
| | - Daniel J Drucker
- The Department of Medicine, Lunenfeld-Tanenbaum Research Institute, Mt. Sinai Hospital, University of Toronto, Toronto, Ontario, Canada
| | - Murielle M Véniant
- Department of Cardiometabolic Disorders, Amgen Research, Thousand Oaks, CA, USA.
| |
Collapse
|
5
|
Mao L, Wang L, Huang Z, Chen JK, Tucker L, Zhang Q. Comprehensive insights into emerging advances in the Neurobiology of anorexia. J Adv Res 2025:S2090-1232(25)00206-1. [PMID: 40180244 DOI: 10.1016/j.jare.2025.03.046] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2025] [Revised: 03/07/2025] [Accepted: 03/24/2025] [Indexed: 04/05/2025] Open
Abstract
BACKGROUND Anorexia is a complex eating disorder influenced by genetic, environmental, psychological, and socio-cultural factors. Research into its molecular mechanisms and neural circuits has deepened our understanding of its pathogenesis. Recent advances in neuroscience, molecular biology, and genetics have revealed key molecular and neural circuit mechanisms underlying anorexia. AIM OF REVIEW Clarify the peripheral and central molecular mechanisms regulating various types of anorexia, identify key cytokines and neural circuits, and propose new strategies for its treatment. Key scientific concepts of review: Anorexia animal models, including activity-induced, genetic mutation, and inflammation-induced types, are explored for their relevance to studying the disorder. Anorexic behavior is regulated by cytokines, hormones (like GDF15, GLP-1, and leptin), and neural circuits such as AgRP, serotonergic, dopaminergic, and glutamatergic pathways. Disruptions in these pathways, including GABAergic signaling in AgRP neurons and 5-HT2C and D2 receptors, contribute to anorexia. Potential therapies target neurotransmitter receptors, ghrelin receptors, and the GDF15-GFRAL pathway, offering insights for treating anorexia, immune responses, and obesity.
Collapse
Affiliation(s)
- Liwei Mao
- Department of Neurology, Medical College of Georgia, Augusta University, 1120 15th Street, Augusta, GA 30912, USA
| | - Lian Wang
- Department of Neurology, Medical College of Georgia, Augusta University, 1120 15th Street, Augusta, GA 30912, USA
| | - Zhihai Huang
- Department of Neurology, Louisiana State University Health Sciences Center, 1501 Kings Highway, Shreveport, LA 71103, USA
| | - Jian-Kang Chen
- Departments of Cellular Biology & Anatomy and Medicine, Medical College of Georgia, Augusta University, 1120 15th Street, Augusta, GA 30912, USA
| | - Lorelei Tucker
- Department of Neurology, Medical College of Georgia, Augusta University, 1120 15th Street, Augusta, GA 30912, USA
| | - Quanguang Zhang
- Department of Neurology, Medical College of Georgia, Augusta University, 1120 15th Street, Augusta, GA 30912, USA.
| |
Collapse
|
6
|
Fermani F, Chang S, Mastrodicasa Y, Peters C, Gaitanos L, Alcala Morales PL, Ramakrishnan C, Deisseroth K, Klein R. Food and water intake are regulated by distinct central amygdala circuits revealed using intersectional genetics. Nat Commun 2025; 16:3072. [PMID: 40157920 PMCID: PMC11954953 DOI: 10.1038/s41467-025-58144-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2024] [Accepted: 03/10/2025] [Indexed: 04/01/2025] Open
Abstract
The central amygdala (CeA) plays a crucial role in defensive and appetitive behaviours. It contains genetically defined GABAergic neuron subpopulations distributed over three anatomical subregions, capsular (CeC), lateral (CeL), and medial (CeM). The roles that these molecularly- and anatomically-defined CeA neurons play in appetitive behavior remain unclear. Using intersectional genetics in mice, we found that neurons driving food or water consumption are confined to the CeM. Separate CeM subpopulations exist for water only versus water or food consumption. In vivo calcium imaging revealed that CeMHtr2a neurons promoting feeding are responsive towards appetitive cues with little regard for their physical attributes. CeMSst neurons involved in drinking are sensitive to the physical properties of salient stimuli. Both CeM subtypes receive inhibitory input from CeL and send projections to the parabrachial nucleus to promote appetitive behavior. These results suggest that distinct CeM microcircuits evaluate liquid and solid appetitive stimuli to drive the appropriate behavioral responses.
Collapse
Affiliation(s)
- Federica Fermani
- Department of Molecules - Signaling - Development, Max-Planck Institute for Biological Intelligence, Martinsried, Germany
| | - Simon Chang
- Cellular Neurobiology, Department of Behavioral and Molecular Neurobiology, University of Regensburg, Regensburg, Germany
| | - Ylenia Mastrodicasa
- Department of Molecules - Signaling - Development, Max-Planck Institute for Biological Intelligence, Martinsried, Germany
| | - Christian Peters
- Department of Molecules - Signaling - Development, Max-Planck Institute for Biological Intelligence, Martinsried, Germany
| | - Louise Gaitanos
- Department of Molecules - Signaling - Development, Max-Planck Institute for Biological Intelligence, Martinsried, Germany
| | - Pilar L Alcala Morales
- Department of Molecules - Signaling - Development, Max-Planck Institute for Biological Intelligence, Martinsried, Germany
| | | | - Karl Deisseroth
- Department of Bioengineering, Stanford University, Stanford, CA, USA
- Department of Psychiatry & Behavioral Sciences, Stanford University, Stanford, CA, USA
- Howard Hughes Medical Institute, Stanford University, Stanford, CA, USA
| | - Rüdiger Klein
- Department of Molecules - Signaling - Development, Max-Planck Institute for Biological Intelligence, Martinsried, Germany.
| |
Collapse
|
7
|
Zhu Z, Gong R, Rodriguez V, Quach KT, Chen X, Sternson SM. Hedonic eating is controlled by dopamine neurons that oppose GLP-1R satiety. Science 2025; 387:eadt0773. [PMID: 40146831 PMCID: PMC12009138 DOI: 10.1126/science.adt0773] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2024] [Accepted: 01/27/2025] [Indexed: 03/29/2025]
Abstract
Hedonic eating is defined as food consumption driven by palatability without physiological need. However, neural control of palatable food intake is poorly understood. We discovered that hedonic eating is controlled by a neural pathway from the peri-locus ceruleus to the ventral tegmental area (VTA). Using photometry-calibrated optogenetics, we found that VTA dopamine (VTADA) neurons encode palatability to bidirectionally regulate hedonic food consumption. VTADA neuron responsiveness was suppressed during food consumption by semaglutide, a glucagon-like peptide receptor 1 (GLP-1R) agonist used as an antiobesity drug. Mice recovered palatable food appetite and VTADA neuron activity during repeated semaglutide treatment, which was reversed by consumption-triggered VTADA neuron inhibition. Thus, hedonic food intake activates VTADA neurons, which sustain further consumption, a mechanism that opposes appetite reduction by semaglutide.
Collapse
Affiliation(s)
- Zhenggang Zhu
- Department of Neurosciences, University of California, San Diego; La Jolla, CA 92093, USA
| | - Rong Gong
- Janelia Research Campus, Howard Hughes Medical Institute; Ashburn, VA 20147, USA
| | - Vicente Rodriguez
- Howard Hughes Medical Institute; University of California, San Diego; La Jolla, CA 92093, USA
| | - Kathleen T. Quach
- Howard Hughes Medical Institute; University of California, San Diego; La Jolla, CA 92093, USA
| | - Xinyu Chen
- Howard Hughes Medical Institute; University of California, San Diego; La Jolla, CA 92093, USA
| | - Scott M. Sternson
- Department of Neurosciences, University of California, San Diego; La Jolla, CA 92093, USA
- Janelia Research Campus, Howard Hughes Medical Institute; Ashburn, VA 20147, USA
- Howard Hughes Medical Institute; University of California, San Diego; La Jolla, CA 92093, USA
| |
Collapse
|
8
|
Chowdhury S, Kamatkar NG, Wang WX, Akerele CA, Huang J, Wu J, Nwankpa A, Kane CM, Bhave VM, Huang H, Wang X, Nectow AR. Brainstem neuropeptidergic neurons link a neurohumoral axis to satiation. Cell 2025; 188:1563-1579.e18. [PMID: 39914383 DOI: 10.1016/j.cell.2025.01.018] [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: 02/16/2024] [Revised: 12/03/2024] [Accepted: 01/10/2025] [Indexed: 03/23/2025]
Abstract
Hunger is evolutionarily hardwired to ensure that an animal has sufficient energy to survive and reproduce. Just as important as knowing when to start eating is knowing when to stop eating. Here, using spatially resolved single-cell phenotyping, we characterize a population of neuropeptidergic neurons in the brainstem's dorsal raphe nucleus (DRN) and describe how they regulate satiation. These neurons track food from sensory presentation through ingestion, integrate these signals with slower-acting humoral cues, and express cholecystokinin (CCK). These CCK neurons bidirectionally regulate meal size, driving a sustained meal termination signal with a built-in delay. They are also well positioned to sense and respond to ingestion: they express a host of metabolic signaling factors and are integrated into an extended network known to regulate feeding. Together, this work demonstrates how DRN CCK neurons regulate satiation and identifies a likely conserved cellular mechanism that transforms diverse neurohumoral signals into a key behavioral output.
Collapse
Affiliation(s)
| | | | - Wendy Xueyi Wang
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; Department of Chemistry, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; John A. Paulson School of Engineering and Applied Sciences, Harvard University, Boston, MA 02134, USA
| | - Christa A Akerele
- Department of Medicine, Columbia University, New York, NY 10032, USA
| | - Jiahao Huang
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; Department of Chemistry, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Junlin Wu
- Department of Medicine, Columbia University, New York, NY 10032, USA
| | - Amajindi Nwankpa
- Department of Medicine, Columbia University, New York, NY 10032, USA
| | - Charlotte M Kane
- Department of Medicine, Columbia University, New York, NY 10032, USA
| | - Varun M Bhave
- Department of Medicine, Columbia University, New York, NY 10032, USA
| | - Hao Huang
- Princeton Neuroscience Institute, Princeton University, Princeton, NJ 08540, USA
| | - Xiao Wang
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; Department of Chemistry, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Alexander R Nectow
- Department of Medicine, Columbia University, New York, NY 10032, USA; Division of Cardiology, Columbia University, New York, NY 10032, USA.
| |
Collapse
|
9
|
Yu H, Feng N, Zhong W, Han Y, Cheng Y, Zhang Z, Wang Y, Gao P, Huang R, Zhang C, Liu Z, Dong J, He Z, Lai H, Shen Z, Zhai Q. Nmnat2 deficiency in the arcuate nucleus or paraventricular nucleus induces Sarm1-independent neuron loss and liraglutide-reversible obesity. FASEB J 2025; 39:e70400. [PMID: 39964232 DOI: 10.1096/fj.202402546r] [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: 10/22/2024] [Revised: 01/08/2025] [Accepted: 02/06/2025] [Indexed: 05/10/2025]
Abstract
Nicotinamide mononucleotide adenylyltransferase 2 (Nmnat2) plays an important role in maintaining axon integrity, and the arcuate nucleus (ARC), and paraventricular nucleus (PVN) are crucial nuclei in the control of energy balance. However, the effect of Nmnat2 deficiency in ARC and PVN is still unclear. Nmnat2 loxP/loxP or Nmnat2 loxP/loxP , Sarm1 -/- mice were bilaterally injected with AAV-CMV-GFP-Cre once into the ARC, PVN, or lateral parabrachial nucleus (LPBN) to obtain Nmnat2 ARC-/- , Nmnat2 PVN-/- , Nmnat2 LPBN-/- , Nmnat2 ARC-/- , SKO, Nmnat2 PVN-/- , SKO, or Nmnat2 LPBN-/- , SKO mice. Syn1-Cre mice were bilaterally injected with AAV-EF1a-flex-taCasp3-TEVp once into the ARC or PVN to specifically induce neuron loss. Metabolic changes were measured in the mice intraperitoneally injected with or without liraglutide, a glucagon-like peptide-1 (GLP-1) analog. Neuron loss and neuron activation were monitored by immunofluorescence. Deletion of Nmnat2 in ARC or PVN of mice leads to neuron loss, increased food intake, and obesity in a Sarm1-independent manner. Intraperitoneal injection of liraglutide activates neurons in PVN and LPBN, and attenuates hyperphagia and obesity induced by Nmnat2 deletion or apoptosis of Syn1-positive neurons in ARC or PVN, but has no significant effect on neuron loss. Nmnat2 deficiency in LPBN leads to death within 2 weeks, which can be markedly rescued by Sarm1 deficiency. These data show that deletion of Nmnat2 in ARC or PVN in adult mice leads to Sarm1-independent neuron loss, and liraglutide-reversible hyperphagia and obesity. These findings also elucidate the integrated role of ARC or PVN for downregulating food intake, the requirement of LPBN for survival, and the ARC- or PVN-independent effect of GLP-1 on food intake.
Collapse
Affiliation(s)
- Huimin Yu
- CAS Key Laboratory of Nutrition, Metabolism and Food Safety, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Ning Feng
- CAS Key Laboratory of Nutrition, Metabolism and Food Safety, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Wuling Zhong
- CAS Key Laboratory of Nutrition, Metabolism and Food Safety, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Yumo Han
- School of Life Science and Technology, ShanghaiTech University, Shanghai, China
| | - Yalan Cheng
- CAS Key Laboratory of Nutrition, Metabolism and Food Safety, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Zhentong Zhang
- CAS Key Laboratory of Nutrition, Metabolism and Food Safety, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Yingqi Wang
- Department of Neuroscience, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Peidong Gao
- CAS Key Laboratory of Nutrition, Metabolism and Food Safety, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Rui Huang
- CAS Key Laboratory of Nutrition, Metabolism and Food Safety, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Cong Zhang
- CAS Key Laboratory of Nutrition, Metabolism and Food Safety, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Zongyang Liu
- CAS Key Laboratory of Nutrition, Metabolism and Food Safety, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Jieya Dong
- CAS Key Laboratory of Nutrition, Metabolism and Food Safety, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Zhishui He
- CAS Key Laboratory of Nutrition, Metabolism and Food Safety, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Hejin Lai
- CAS Key Laboratory of Nutrition, Metabolism and Food Safety, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Ziru Shen
- CAS Key Laboratory of Nutrition, Metabolism and Food Safety, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Qiwei Zhai
- CAS Key Laboratory of Nutrition, Metabolism and Food Safety, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
- School of Life Science and Technology, ShanghaiTech University, Shanghai, China
| |
Collapse
|
10
|
Douros JD, Flak JN, Knerr PJ. The agony and the efficacy: central mechanisms of GLP-1 induced adverse events and their mitigation by GIP. Front Endocrinol (Lausanne) 2025; 16:1530985. [PMID: 39963285 PMCID: PMC11830610 DOI: 10.3389/fendo.2025.1530985] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/19/2024] [Accepted: 01/02/2025] [Indexed: 02/20/2025] Open
Affiliation(s)
| | - Jonathan N. Flak
- Indiana Biosciences Research Institute, Indianapolis, IN, United States
- Department of Pharmacology and Toxicology, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Patrick J. Knerr
- Indiana Biosciences Research Institute, Indianapolis, IN, United States
| |
Collapse
|
11
|
Zhang C. Neural pathways of nausea and roles in energy balance. Curr Opin Neurobiol 2025; 90:102963. [PMID: 39765206 PMCID: PMC11839311 DOI: 10.1016/j.conb.2024.102963] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2024] [Revised: 12/04/2024] [Accepted: 12/06/2024] [Indexed: 01/18/2025]
Abstract
Our internal sensory systems encode various gut-related sensations, such as hunger, feelings of fullness, and nausea. These internal feelings influence our eating behaviors and play a vital role in regulating energy balance. Among them, the neurological basis for nausea has been the least well characterized, which has hindered comprehension of the connection between these sensations. Single-cell sequencing, along with functional mapping, has brought clarity to the neural pathways of nausea involving the brainstem area postrema. In addition, the newly discovered nausea sensory signals have deepened our understanding of the area postrema in regulating feeding behaviors. Nausea has significant clinical implications, especially in developing drugs for weight loss and metabolism. This review summarizes recent research on the neural pathways of nausea, particularly highlighting their contribution to energy balance.
Collapse
Affiliation(s)
- Chuchu Zhang
- Department of Physiology, David Geffen School of Medicine, University of California, Los Angeles, CA, USA.
| |
Collapse
|
12
|
Hankir MK, Lutz TA. Novel neural pathways targeted by GLP-1R agonists and bariatric surgery. Pflugers Arch 2025; 477:171-185. [PMID: 39644359 PMCID: PMC11761532 DOI: 10.1007/s00424-024-03047-3] [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: 10/17/2024] [Revised: 11/18/2024] [Accepted: 11/19/2024] [Indexed: 12/09/2024]
Abstract
The glucagon-like peptide 1 receptor (GLP-1R) agonist semaglutide has revolutionized the treatment of obesity, with other gut hormone-based drugs lined up that show even greater weight-lowering ability in obese patients. Nevertheless, bariatric surgery remains the mainstay treatment for severe obesity and achieves unparalleled weight loss that generally stands the test of time. While their underlying mechanisms of action remain incompletely understood, it is clear that the common denominator between GLP-1R agonists and bariatric surgery is that they suppress food intake by targeting the brain. In this Review, we highlight recent preclinical studies using contemporary neuroscientific techniques that provide novel concepts in the neural control of food intake and body weight with reference to endogenous GLP-1, GLP-1R agonists, and bariatric surgery. We start in the periphery with vagal, intestinofugal, and spinal sensory nerves and then progress through the brainstem up to the hypothalamus and finish at non-canonical brain feeding centers such as the zona incerta and lateral septum. Further defining the commonalities and differences between GLP-1R agonists and bariatric surgery in terms of how they target the brain may not only help bridge the gap between pharmacological and surgical interventions for weight loss but also provide a neural basis for their combined use when each individually fails.
Collapse
Affiliation(s)
- Mohammed K Hankir
- Institute of Veterinary Physiology, University of Zurich, Zurich, Switzerland
- School of Biochemistry and Immunology, Trinity College Dublin, Dublin, Ireland
| | - Thomas A Lutz
- Institute of Veterinary Physiology, University of Zurich, Zurich, Switzerland.
| |
Collapse
|
13
|
Hankir MK, Le Foll C. Central nervous system pathways targeted by amylin in the regulation of food intake. Biochimie 2025; 229:95-104. [PMID: 39426704 DOI: 10.1016/j.biochi.2024.10.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2024] [Revised: 10/04/2024] [Accepted: 10/17/2024] [Indexed: 10/21/2024]
Abstract
Amylin is a peptide hormone co-released with insulin from pancreatic β-cells during a meal and primarily serves to promote satiation. While the caudal hindbrain was originally implicated as a major site of action in this regard, it is becoming increasingly clear that amylin recruits numerous central nervous system pathways to exert multifaceted effects on food intake. In this Review, we discuss the evidence derived from preclinical studies showing that amylin and the related peptide salmon calcitonin (sCT) directly or indirectly target genetically distinct neurons in the caudal hindbrain (nucleus tractus solitarius and area postrema), rostral hindbrain (lateral parabrachial nucleus), midbrain (lateral dorsal tegmentum and ventral tegmental area) and hypothalamus (arcuate nucleus and parasubthalamic nucleus) via activation of amylin and/or calcitonin receptors. Given that the stable amylin analogue cagrilintide is under clinical development for the treatment of obesity, it is important to determine whether this drug recruits overlapping or distinct central nervous system pathways to that of amylin and sCT with implications for minimising any aversive effects it potentially causes. Such insight will also be important to understand how amylin and sCT analogues synergize with other molecules as part of dual or triple agonist therapies for obesity, especially the glucagon-like peptide 1 receptor (GLP-1R) agonist semaglutide, which has been shown to synergistically lower body weight with cagrilintide (CagriSema) in clinical trials.
Collapse
Affiliation(s)
- Mohammed K Hankir
- Department of Veterinary Physiology, University of Zurich, Zurich, Switzerland; School of Biochemistry and Immunology, Trinity College Dublin, Dublin, Ireland.
| | - Christelle Le Foll
- Department of Veterinary Physiology, University of Zurich, Zurich, Switzerland.
| |
Collapse
|
14
|
Yang K, Wu YT, He Y, Dai JX, Luo YL, Xie JH, Ding WJ. GLP-1 and IL-6 regulates obesity in the gut and brain. Life Sci 2025; 362:123339. [PMID: 39730038 DOI: 10.1016/j.lfs.2024.123339] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2024] [Revised: 11/06/2024] [Accepted: 12/23/2024] [Indexed: 12/29/2024]
Abstract
Obesity is a chronic metabolic disease characterized by excessive nutrient intake leading to increased subcutaneous or visceral fat, resulting in pathological and physiological changes. The incidence rate of obesity, an important form of metabolic syndrome, is increasing worldwide. Excess appetite is a key pathogenesis of obesity, and the inflammatory response induced by obesity has received increasing attention. This review focuses on the role of appetite-regulating factor (Glucogan-like peptide 1) and inflammatory factor (Interleukin-6) in the gut and brain in individuals with obesity and draws insights from the current literature.
Collapse
Affiliation(s)
- Kun Yang
- School of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Yu-Ting Wu
- Chengdu University of Traditional Chinese Medicine, 1166 Luitai Avenue, Chengdu, Sichuan 611137, China
| | - Yan He
- School of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Jin-Xiu Dai
- Chengdu University of Traditional Chinese Medicine, 1166 Luitai Avenue, Chengdu, Sichuan 611137, China
| | - Yu-Lu Luo
- School of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Jing-Hui Xie
- School of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Wei-Jun Ding
- School of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China.
| |
Collapse
|
15
|
Korkutata M, De Luca R, Fitzgerald B, Khanday MA, Arrigoni E, Scammell TE. Afferent Projections to the Calca/CGRP-Expressing Parabrachial Neurons in Mice. J Comp Neurol 2025; 533:e70018. [PMID: 39801453 PMCID: PMC11777123 DOI: 10.1002/cne.70018] [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: 06/12/2024] [Revised: 12/05/2024] [Accepted: 12/30/2024] [Indexed: 01/22/2025]
Abstract
The parabrachial nucleus (PB), located in the dorsolateral pons, contains primarily glutamatergic neurons that regulate responses to a variety of interoceptive and cutaneous sensory signals. One lateral PB subpopulation expresses the Calca gene, which codes for the neuropeptide calcitonin gene-related peptide (CGRP). These PBCalca /CGRP neurons relay signals related to threatening stimuli such as hypercarbia, pain, and nausea, yet their inputs and their neurochemical identity are only partially understood. We mapped the afferent projections to the lateral part of the PB in mice using conventional cholera toxin B subunit (CTb) retrograde tracing and then used conditional rabies virus retrograde tracing to map monosynaptic inputs specifically targeting the PBCalca /CGRP neurons. Using vesicular GABA (vGAT) and glutamate (vGLUT2) transporter reporter mice, we found that lateral PB neurons receive GABAergic afferents from regions such as the lateral part of the central nucleus of the amygdala, lateral dorsal subnucleus of the bed nucleus of the stria terminalis, substantia innominata, and ventrolateral periaqueductal gray. Additionally, they receive glutamatergic afferents from the infralimbic and insular cortex, paraventricular nucleus, parasubthalamic nucleus, trigeminal complex, medullary reticular nucleus, and nucleus of the solitary tract. Using anterograde tracing and confocal microscopy, we then identified close axonal appositions between these afferents and PBCalca /CGRP neurons. Finally, we used channelrhodopsin-assisted circuit mapping and found that GABAergic neurons of the central nucleus of the amygdala directly inhibit the PBCalca /CGRP neurons. These findings provide a comprehensive neuroanatomical framework for understanding the afferent projections regulating the PBCalca /CGRP neurons.
Collapse
Affiliation(s)
- Mustafa Korkutata
- Department of Neurology, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA 02215, USA
| | - Roberto De Luca
- Department of Neurology, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA 02215, USA
| | - Bridget Fitzgerald
- Department of Neurology, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA 02215, USA
| | - Mudasir A. Khanday
- Department of Neurology, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA 02215, USA
| | - Elda Arrigoni
- Department of Neurology, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA 02215, USA
| | - Thomas E. Scammell
- Department of Neurology, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA 02215, USA
| |
Collapse
|
16
|
Asgari R, Caceres-Valdiviezo M, Wu S, Hamel L, Humber BE, Agarwal SM, Fletcher PJ, Fulton S, Hahn MK, Pereira S. Regulation of energy balance by leptin as an adiposity signal and modulator of the reward system. Mol Metab 2025; 91:102078. [PMID: 39615837 PMCID: PMC11696864 DOI: 10.1016/j.molmet.2024.102078] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/13/2024] [Revised: 11/02/2024] [Accepted: 11/26/2024] [Indexed: 12/08/2024] Open
Abstract
BACKGROUND Leptin is an adipose tissue-derived hormone that plays a crucial role in body weight, appetite, and behaviour regulation. Leptin controls energy balance as an indicator of adiposity levels and as a modulator of the reward system, which is associated with liking palatable foods. Obesity is characterized by expanded adipose tissue mass and consequently, elevated concentrations of leptin in blood. Leptin's therapeutic potential for most forms of obesity is hampered by leptin resistance and a narrow dose-response window. SCOPE OF REVIEW This review describes the current knowledge of the brain regions and intracellular pathways through which leptin promotes negative energy balance and restrains neural circuits affecting food reward. We also describe mechanisms that hinder these biological responses in obesity and highlight potential therapeutic interventions. MAJOR CONCLUSIONS Additional research is necessary to understand how pathways engaged by leptin in different brain regions are interconnected in the control of energy balance.
Collapse
Affiliation(s)
| | - Maria Caceres-Valdiviezo
- Centre for Addiction and Mental Health, Toronto, ON, Canada; Laboratory of Omic Sciences, School of Medicine, Universidad de Especialidades Espíritu Santo, Samborondón, Ecuador
| | - Sally Wu
- Centre for Addiction and Mental Health, Toronto, ON, Canada
| | - Laurie Hamel
- Centre for Addiction and Mental Health, Toronto, ON, Canada
| | | | - Sri Mahavir Agarwal
- Centre for Addiction and Mental Health, Toronto, ON, Canada; Department of Psychiatry, University of Toronto, Toronto, ON, Canada; Banting & Best Diabetes Centre, University of Toronto, Toronto, ON, Canada
| | - Paul J Fletcher
- Centre for Addiction and Mental Health, Toronto, ON, Canada; Department of Psychiatry, University of Toronto, Toronto, ON, Canada; Department of Psychology, University of Toronto, Toronto, ON, Canada
| | - Stephanie Fulton
- Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), Montreal Diabetes Research Center, Montréal, QC, Canada; Department of Nutrition, Université de Montréal, QC, Canada
| | - Margaret K Hahn
- Centre for Addiction and Mental Health, Toronto, ON, Canada; Department of Psychiatry, University of Toronto, Toronto, ON, Canada; Banting & Best Diabetes Centre, University of Toronto, Toronto, ON, Canada; Institute of Medical Sciences, University of Toronto, Toronto, ON, Canada; Department of Pharmacology, University of Toronto, Toronto, ON, Canada.
| | - Sandra Pereira
- Centre for Addiction and Mental Health, Toronto, ON, Canada; Department of Physiology, University of Toronto, Toronto, ON, Canada.
| |
Collapse
|
17
|
Münzberg H, Heymsfield SB, Berthoud HR, Morrison CD. History and future of leptin: Discovery, regulation and signaling. Metabolism 2024; 161:156026. [PMID: 39245434 PMCID: PMC11570342 DOI: 10.1016/j.metabol.2024.156026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Revised: 08/27/2024] [Accepted: 09/04/2024] [Indexed: 09/10/2024]
Abstract
The cloning of leptin 30 years ago in 1994 was an important milestone in obesity research. Prior to the discovery of leptin, obesity was stigmatized as a condition caused by lack of character and self-control. Mutations in either leptin or its receptor were the first single gene mutations found to cause severe obesity, and it is now recognized that obesity is caused mostly by a dysregulation of central neuronal circuits. Since the discovery of the leptin-deficient obese mouse (ob/ob) the cloning of leptin (ob aka lep) and leptin receptor (db aka lepr) genes, we have learned much about leptin and its action in the central nervous system. The first hope that leptin would cure obesity was quickly dampened because humans with obesity have increased leptin levels and develop leptin resistance. Nevertheless, leptin target sites in the brain represent an excellent blueprint to understand how neuronal circuits control energy homeostasis. Our expanding understanding of leptin function, interconnection of leptin signaling with other systems and impact on distinct physiological functions continues to guide and improve the development of safe and effective interventions to treat metabolic illnesses. This review highlights past concepts and current emerging concepts of the hormone leptin, leptin receptor signaling pathways and central targets to mediate distinct physiological functions.
Collapse
Affiliation(s)
- Heike Münzberg
- Pennington Biomedical Research Center, LSU System, Baton Rouge, LA, United States of America.
| | - Steven B Heymsfield
- Pennington Biomedical Research Center, LSU System, Baton Rouge, LA, United States of America
| | - Hans-Rudolf Berthoud
- Pennington Biomedical Research Center, LSU System, Baton Rouge, LA, United States of America
| | - Christopher D Morrison
- Pennington Biomedical Research Center, LSU System, Baton Rouge, LA, United States of America
| |
Collapse
|
18
|
Mahishi D, Agrawal N, Jiang W, Yapici N. From Mammals to Insects: Exploring the Genetic and Neural Basis of Eating Behavior. Annu Rev Genet 2024; 58:455-485. [PMID: 39585905 DOI: 10.1146/annurev-genet-111523-102202] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2024]
Abstract
Obesity and anorexia are life-threatening diseases that are still poorly understood at the genetic and neuronal levels. Patients suffering from these conditions experience disrupted regulation of food consumption, leading to extreme weight gain or loss and, in severe situations, death from metabolic dysfunction. Despite the development of various behavioral and pharmacological interventions, current treatments often yield limited and short-lived success. To address this, a deeper understanding of the genetic and neural mechanisms underlying food perception and appetite regulation is essential for identifying new drug targets and developing more effective treatment methods. This review summarizes the progress of past research in understanding the genetic and neural mechanisms controlling food consumption and appetite regulation, focusing on two key model organisms: the fruit fly Drosophila melanogaster and the mouse Mus musculus. These studies investigate how the brain senses energy and nutrient deficiency, how sensory signals trigger appetitive behaviors, and how food intake is regulated through interconnected neural circuits in the brain.
Collapse
Affiliation(s)
- Deepthi Mahishi
- Department of Neurobiology and Behavior, Cornell University, Ithaca, New York, USA;
| | - Naman Agrawal
- Department of Neurobiology and Behavior, Cornell University, Ithaca, New York, USA;
| | - Wenshuai Jiang
- Department of Neurobiology and Behavior, Cornell University, Ithaca, New York, USA;
| | - Nilay Yapici
- Department of Neurobiology and Behavior, Cornell University, Ithaca, New York, USA;
| |
Collapse
|
19
|
Kuralay A, McDonough MC, Resch JM. Control of sodium appetite by hindbrain aldosterone-sensitive neurons. Mol Cell Endocrinol 2024; 592:112323. [PMID: 38936597 PMCID: PMC11381173 DOI: 10.1016/j.mce.2024.112323] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/31/2024] [Accepted: 06/25/2024] [Indexed: 06/29/2024]
Abstract
Mineralocorticoids play a key role in hydromineral balance by regulating sodium retention and potassium wasting. Through favoring sodium, mineralocorticoids can cause hypertension from fluid overload under conditions of hyperaldosteronism, such as aldosterone-secreting tumors. An often-overlooked mechanism by which aldosterone functions to increase sodium is through stimulation of salt appetite. To drive sodium intake, aldosterone targets neurons in the hindbrain which uniquely express 11β-hydroxysteroid dehydrogenase type 2 (HSD2). This enzyme is a necessary precondition for aldosterone-sensing cells as it metabolizes glucocorticoids - preventing their activation of the mineralocorticoid receptor. In this review, we will consider the role of hindbrain HSD2 neurons in regulating sodium appetite by discussing HSD2 expression in the brain, regulation of hindbrain HSD2 neuron activity, and the circuitry mediating the effects of these aldosterone-sensitive neurons. Reducing the activity of hindbrain HSD2 neurons may be a viable strategy to reduce sodium intake and cardiovascular risk, particularly for conditions of hyperaldosteronism.
Collapse
Affiliation(s)
- Ahmet Kuralay
- Department of Neuroscience and Pharmacology, University of Iowa, Iowa City, IA, USA; Interdisciplinary Graduate Program in Neuroscience, University of Iowa, Iowa City, IA, USA
| | - Miriam C McDonough
- Department of Neuroscience and Pharmacology, University of Iowa, Iowa City, IA, USA; Molecular Medicine Graduate Program, University of Iowa, Iowa City, IA, USA
| | - Jon M Resch
- Department of Neuroscience and Pharmacology, University of Iowa, Iowa City, IA, USA; Iowa Neuroscience Institute, University of Iowa, Iowa City, IA, USA; Fraternal Order of Eagles Diabetes Research Center, University of Iowa, Iowa City, IA, USA; Interdisciplinary Graduate Program in Neuroscience, University of Iowa, Iowa City, IA, USA; Molecular Medicine Graduate Program, University of Iowa, Iowa City, IA, USA.
| |
Collapse
|
20
|
D'Ávila M, Hall S, Horvath TL. GLP-1, GIP, and Glucagon Agonists for Obesity Treatment: A Hunger Perspective. Endocrinology 2024; 165:bqae128. [PMID: 39301751 DOI: 10.1210/endocr/bqae128] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Revised: 08/31/2024] [Accepted: 09/19/2024] [Indexed: 09/22/2024]
Abstract
For centuries, increasingly sophisticated methods and approaches have been brought to bear to promote weight loss. Second only to the Holy Grail of research on aging, the idea of finding a single and simple way to lose weight has long preoccupied the minds of laymen and scientists alike. The effects of obesity are far-reaching and not to be minimized; the need for more effective treatments is obvious. Is there a single silver bullet that addresses this issue without effort on the part of the individual? The answer to this question has been one of the most elusive and sought-after in modern history. Now and then, a miraculous discovery propagates the illusion that a simple solution is possible. Now there are designer drugs that seem to accomplish the task: we can lose weight without effort using mono, dual, and triple agonists of receptors for glucagon-like peptide-1 (GLP-1), glucose-dependent insulinotropic peptide (GIP), and glucagon. There are, however, fundamental biological principles that raise intriguing questions about these therapies beyond the currently reported side-effects. This perspective reflects upon these issues from the angle of complex goal-oriented behaviors, and systemic and cellular metabolism associated with satiety and hunger.
Collapse
Affiliation(s)
- Mateus D'Ávila
- Department of Comparative Medicine, Yale School of Medicine, New Haven, CT 06520, USA
- Interdepartmental Neuroscience Program, Yale University, New Haven, CT 06520, USA
| | - Samantha Hall
- Department of Comparative Medicine, Yale School of Medicine, New Haven, CT 06520, USA
| | - Tamas L Horvath
- Department of Comparative Medicine, Yale School of Medicine, New Haven, CT 06520, USA
- Interdepartmental Neuroscience Program, Yale University, New Haven, CT 06520, USA
| |
Collapse
|
21
|
Turcano P, Savica R, Benarroch E. What Is the Role of Glucagon-Like Peptide 1 Signaling in the Nervous System and Its Potential Neuroprotective Effects? Neurology 2024; 103:e209781. [PMID: 39079072 DOI: 10.1212/wnl.0000000000209781] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2024] [Accepted: 06/20/2024] [Indexed: 01/24/2025] Open
|
22
|
Gautier-Stein A, Vily-Petit J, Rajas F, Mithieux G. Intestinal gluconeogenesis: A translator of nutritional information needed for glycemic and emotional balance. Biochimie 2024; 223:206-214. [PMID: 38040189 DOI: 10.1016/j.biochi.2023.11.012] [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: 07/25/2023] [Revised: 11/17/2023] [Accepted: 11/23/2023] [Indexed: 12/03/2023]
Abstract
At the interface between the outside world and the self, the intestine is the first organ receiving nutritional information. One intestinal function, gluconeogenesis, is activated by various nutrients, particularly diets enriched in fiber or protein, and thus results in glucose production in the portal vein in the post-absorptive period. The detection of portal glucose induces a nervous signal controlling the activity of the central nuclei involved in the regulation of metabolism and emotional behavior. Induction of intestinal gluconeogenesis is necessary for the beneficial effects of fiber or protein-enriched diets on metabolism and emotional behavior. Through its ability to translate nutritional information from the diet to the brain's regulatory centers, intestinal gluconeogenesis plays an essential role in maintaining physiological balance.
Collapse
Affiliation(s)
- Amandine Gautier-Stein
- Universite Claude Bernard Lyon 1, NUDICE, UMR_S 1213, Villeurbanne, 69100, France; Institut National de la Sante et de la Recherche Medicale, NUDICE, UMR_S 1213, Lyon, 69372, France.
| | - Justine Vily-Petit
- Universite Claude Bernard Lyon 1, NUDICE, UMR_S 1213, Villeurbanne, 69100, France; Institut National de la Sante et de la Recherche Medicale, NUDICE, UMR_S 1213, Lyon, 69372, France
| | - Fabienne Rajas
- Universite Claude Bernard Lyon 1, NUDICE, UMR_S 1213, Villeurbanne, 69100, France; Institut National de la Sante et de la Recherche Medicale, NUDICE, UMR_S 1213, Lyon, 69372, France
| | - Gilles Mithieux
- Universite Claude Bernard Lyon 1, NUDICE, UMR_S 1213, Villeurbanne, 69100, France; Institut National de la Sante et de la Recherche Medicale, NUDICE, UMR_S 1213, Lyon, 69372, France
| |
Collapse
|
23
|
Catalbas K, Pattnaik T, Congdon S, Nelson C, Villano LC, Sweeney P. Hypothalamic AgRP neurons regulate the hyperphagia of lactation. Mol Metab 2024; 86:101975. [PMID: 38925247 PMCID: PMC11268337 DOI: 10.1016/j.molmet.2024.101975] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/18/2024] [Revised: 06/19/2024] [Accepted: 06/21/2024] [Indexed: 06/28/2024] Open
Abstract
OBJECTIVE The lactational period is associated with profound hyperphagia to accommodate the energy demands of nursing. These changes are important for the long-term metabolic health of the mother and children as altered feeding during lactation increases the risk of mothers and offspring developing metabolic disorders later in life. However, the specific behavioral mechanisms and neural circuitry mediating the hyperphagia of lactation are incompletely understood. METHODS Here, we utilized home cage feeding devices to characterize the dynamics of feeding behavior in lactating mice. A combination of pharmacological and behavioral assays were utilized to determine how lactation alters meal structure, circadian aspects of feeding, hedonic feeding, and sensitivity to hunger and satiety signals in lactating mice. Finally, we utilized chemogenetic, immunohistochemical, and in vivo imaging approaches to characterize the role of hypothalamic agouti-related peptide (AgRP) neurons in lactational-hyperphagia. RESULTS The lactational period is associated with increased meal size, altered circadian patterns of feeding, reduced sensitivity to gut-brain satiety signals, and enhanced sensitivity to negative energy balance. Hypothalamic AgRP neurons display increased sensitivity to negative energy balance and altered in vivo activity during the lactational state. Further, using in vivo imaging approaches we demonstrate that AgRP neurons are directly activated by lactation. Chemogenetic inhibition of AgRP neurons acutely reduces feeding in lactating mice, demonstrating an important role for these neurons in lactational-hyperphagia. CONCLUSIONS Together, these results show that lactation collectively alters multiple components of feeding behavior and position AgRP neurons as an important cellular substrate mediating the hyperphagia of lactation.
Collapse
Affiliation(s)
- Kerem Catalbas
- University of Illinois Urbana-Champaign, Department of Molecular and Integrative Physiology, USA; University of Illinois Urbana-Champaign Neuroscience Program, USA
| | - Tanya Pattnaik
- University of Illinois Urbana-Champaign, Department of Molecular and Integrative Physiology, USA
| | - Samuel Congdon
- University of Illinois Urbana-Champaign, Department of Molecular and Integrative Physiology, USA
| | - Christina Nelson
- University of Illinois Urbana-Champaign, Department of Molecular and Integrative Physiology, USA
| | - Lara C Villano
- University of Illinois Urbana-Champaign, Department of Molecular and Integrative Physiology, USA
| | - Patrick Sweeney
- University of Illinois Urbana-Champaign, Department of Molecular and Integrative Physiology, USA; University of Illinois Urbana-Champaign Neuroscience Program, USA.
| |
Collapse
|
24
|
Huang KP, Acosta AA, Ghidewon MY, McKnight AD, Almeida MS, Nyema NT, Hanchak ND, Patel N, Gbenou YSK, Adriaenssens AE, Bolding KA, Alhadeff AL. Dissociable hindbrain GLP1R circuits for satiety and aversion. Nature 2024; 632:585-593. [PMID: 38987598 DOI: 10.1038/s41586-024-07685-6] [Citation(s) in RCA: 39] [Impact Index Per Article: 39.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2023] [Accepted: 06/06/2024] [Indexed: 07/12/2024]
Abstract
The most successful obesity therapeutics, glucagon-like peptide-1 receptor (GLP1R) agonists, cause aversive responses such as nausea and vomiting1,2, effects that may contribute to their efficacy. Here, we investigated the brain circuits that link satiety to aversion, and unexpectedly discovered that the neural circuits mediating these effects are functionally separable. Systematic investigation across drug-accessible GLP1R populations revealed that only hindbrain neurons are required for the efficacy of GLP1-based obesity drugs. In vivo two-photon imaging of hindbrain GLP1R neurons demonstrated that most neurons are tuned to either nutritive or aversive stimuli, but not both. Furthermore, simultaneous imaging of hindbrain subregions indicated that area postrema (AP) GLP1R neurons are broadly responsive, whereas nucleus of the solitary tract (NTS) GLP1R neurons are biased towards nutritive stimuli. Strikingly, separate manipulation of these populations demonstrated that activation of NTSGLP1R neurons triggers satiety in the absence of aversion, whereas activation of APGLP1R neurons triggers strong aversion with food intake reduction. Anatomical and behavioural analyses revealed that NTSGLP1R and APGLP1R neurons send projections to different downstream brain regions to drive satiety and aversion, respectively. Importantly, GLP1R agonists reduce food intake even when the aversion pathway is inhibited. Overall, these findings highlight NTSGLP1R neurons as a population that could be selectively targeted to promote weight loss while avoiding the adverse side effects that limit treatment adherence.
Collapse
Affiliation(s)
| | | | - Misgana Y Ghidewon
- Monell Chemical Senses Center, Philadelphia, PA, USA
- Department of Neuroscience, University of Pennsylvania, Philadelphia, PA, USA
| | - Aaron D McKnight
- Monell Chemical Senses Center, Philadelphia, PA, USA
- Department of Neuroscience, University of Pennsylvania, Philadelphia, PA, USA
| | | | | | | | - Nisha Patel
- Monell Chemical Senses Center, Philadelphia, PA, USA
| | | | - Alice E Adriaenssens
- Department of Neuroscience, Physiology, and Pharmacology, University College London, London, UK
| | - Kevin A Bolding
- Monell Chemical Senses Center, Philadelphia, PA, USA
- Department of Neuroscience, University of Pennsylvania, Philadelphia, PA, USA
| | - Amber L Alhadeff
- Monell Chemical Senses Center, Philadelphia, PA, USA.
- Department of Neuroscience, University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
25
|
Shankar K, Bonnet-Zahedi S, Milan K, D'argence AR, Sneddon E, Qiao R, Chonwattangul S, Carrette LLG, Kallupi M, George O. Acute nicotine activates orectic and inhibits anorectic brain regions in rats exposed to chronic nicotine. Neuropharmacology 2024; 253:109959. [PMID: 38648925 PMCID: PMC11734747 DOI: 10.1016/j.neuropharm.2024.109959] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2024] [Revised: 04/09/2024] [Accepted: 04/11/2024] [Indexed: 04/25/2024]
Abstract
Nicotine use produces psychoactive effects, and chronic use is associated with physiological and psychological symptoms of addiction. However, chronic nicotine use is known to decrease food intake and body weight gain, suggesting that nicotine also affects central metabolic and appetite regulation. We recently showed that acute nicotine self-administration in nicotine-dependent animals produces a short-term increase in food intake, contrary to its long-term decrease of feeding behavior. As feeding behavior is regulated by complex neural signaling mechanisms, this study aimed to test the hypothesis that nicotine intake in animals exposed to chronic nicotine may increase activation of pro-feeding regions and decrease activation of pro-satiety regions to produce the acute increase in feeding behavior. FOS immunohistochemistry revealed that acute nicotine intake in nicotine self-administering animals increased activation of the pro-feeding arcuate and lateral hypothalamic nuclei and decreased activation of the pro-satiety parabrachial nucleus. Regional correlational analysis also showed that acute nicotine changes the functional connectivity of the hunger/satiety network. Further dissection of the role of the arcuate nucleus using electrophysiology found that putative POMC neurons in animals given chronic nicotine exhibited decreased firing following acute nicotine application. These brain-wide central signaling changes may contribute to the acute increase in feeding behavior we see in rats after acute nicotine and provide new areas of focus for studying both nicotine addiction and metabolic regulation.
Collapse
Affiliation(s)
- Kokila Shankar
- Department of Psychiatry, University of California San Diego School of Medicine, La Jolla, CA, 92093, USA
| | - Sélène Bonnet-Zahedi
- Department of Psychiatry, University of California San Diego School of Medicine, La Jolla, CA, 92093, USA; Institut de Neurosciences de la Timone, Aix-Marseille Université, Marseille, 13005, France
| | - Kristel Milan
- Department of Psychiatry, University of California San Diego School of Medicine, La Jolla, CA, 92093, USA
| | - Andrea Ruiz D'argence
- Department of Psychiatry, University of California San Diego School of Medicine, La Jolla, CA, 92093, USA
| | - Elizabeth Sneddon
- Department of Psychiatry, University of California San Diego School of Medicine, La Jolla, CA, 92093, USA
| | - Ran Qiao
- Department of Psychiatry, University of California San Diego School of Medicine, La Jolla, CA, 92093, USA
| | - Supakorn Chonwattangul
- Department of Psychiatry, University of California San Diego School of Medicine, La Jolla, CA, 92093, USA
| | - Lieselot L G Carrette
- Department of Psychiatry, University of California San Diego School of Medicine, La Jolla, CA, 92093, USA
| | - Marsida Kallupi
- Department of Psychiatry, University of California San Diego School of Medicine, La Jolla, CA, 92093, USA
| | - Olivier George
- Department of Psychiatry, University of California San Diego School of Medicine, La Jolla, CA, 92093, USA.
| |
Collapse
|
26
|
Ye H, Yang X, Feng B, Luo P, Torres Irizarry VC, Carrillo-Sáenz L, Yu M, Yang Y, Eappen BP, Munoz MD, Patel N, Schaul S, Ibrahimi L, Lai P, Qi X, Zhou Y, Kota M, Dixit D, Mun M, Liew CW, Jiang Y, Wang C, He Y, Xu P. 27-Hydroxycholesterol acts on estrogen receptor α expressed by POMC neurons in the arcuate nucleus to modulate feeding behavior. SCIENCE ADVANCES 2024; 10:eadi4746. [PMID: 38996023 PMCID: PMC11244552 DOI: 10.1126/sciadv.adi4746] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/27/2023] [Accepted: 02/05/2024] [Indexed: 07/14/2024]
Abstract
Oxysterols are metabolites of cholesterol that regulate cholesterol homeostasis. Among these, the most abundant oxysterol is 27-hydroxycholesterol (27HC), which can cross the blood-brain barrier. Because 27HC functions as an endogenous selective estrogen receptor modulator, we hypothesize that 27HC binds to the estrogen receptor α (ERα) in the brain to regulate energy balance. Supporting this view, we found that delivering 27HC to the brain reduced food intake and activated proopiomelanocortin (POMC) neurons in the arcuate nucleus of the hypothalamus (POMCARH) in an ERα-dependent manner. In addition, we observed that inhibiting brain ERα, deleting ERα in POMC neurons, or chemogenetic inhibition of POMCARH neurons blocked the anorexigenic effects of 27HC. Mechanistically, we further revealed that 27HC stimulates POMCARH neurons by inhibiting the small conductance of the calcium-activated potassium (SK) channel. Together, our findings suggest that 27HC, through its interaction with ERα and modulation of the SK channel, inhibits food intake as a negative feedback mechanism against a surge in circulating cholesterol.
Collapse
Affiliation(s)
- Hui Ye
- School of Chemistry, Chemical Engineering and Biotechnology, Nanyang Technological University, Singapore 639798, Singapore
- Division of Endocrinology, Department of Medicine, The University of Illinois at Chicago, Chicago, IL 60612, USA
| | - Xiaohua Yang
- Division of Endocrinology, Department of Medicine, The University of Illinois at Chicago, Chicago, IL 60612, USA
- Guangdong Laboratory of Lingnan Modern Agriculture and Guangdong Province Key Laboratory of Animal Nutritional Regulation, National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, 483 Wushan Road, Tianhe District, Guangzhou, Guangdong 510642, China
| | - Bing Feng
- Pennington Biomedical Research Center, Louisiana State University, Baton Rouge, LA 70808, USA
| | - Pei Luo
- Division of Endocrinology, Department of Medicine, The University of Illinois at Chicago, Chicago, IL 60612, USA
- Guangdong Laboratory of Lingnan Modern Agriculture and Guangdong Province Key Laboratory of Animal Nutritional Regulation, National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, 483 Wushan Road, Tianhe District, Guangzhou, Guangdong 510642, China
| | - Valeria C. Torres Irizarry
- Division of Endocrinology, Department of Medicine, The University of Illinois at Chicago, Chicago, IL 60612, USA
- Department of Physiology and Biophysics, The University of Illinois at Chicago, Chicago, IL 60612, USA
| | - Leslie Carrillo-Sáenz
- Division of Endocrinology, Department of Medicine, The University of Illinois at Chicago, Chicago, IL 60612, USA
- Department of Physiology and Biophysics, The University of Illinois at Chicago, Chicago, IL 60612, USA
| | - Meng Yu
- Children’s Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA
| | - Yongjie Yang
- Children’s Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA
| | - Benjamin P. Eappen
- Children’s Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA
| | - Marcos David Munoz
- Division of Endocrinology, Department of Medicine, The University of Illinois at Chicago, Chicago, IL 60612, USA
| | - Nirali Patel
- Division of Endocrinology, Department of Medicine, The University of Illinois at Chicago, Chicago, IL 60612, USA
| | - Sarah Schaul
- Division of Endocrinology, Department of Medicine, The University of Illinois at Chicago, Chicago, IL 60612, USA
| | - Lucas Ibrahimi
- Division of Endocrinology, Department of Medicine, The University of Illinois at Chicago, Chicago, IL 60612, USA
| | - Penghua Lai
- Division of Endocrinology, Department of Medicine, The University of Illinois at Chicago, Chicago, IL 60612, USA
| | - Xinyue Qi
- School of Chemistry, Chemical Engineering and Biotechnology, Nanyang Technological University, Singapore 639798, Singapore
| | - Yuliang Zhou
- School of Chemistry, Chemical Engineering and Biotechnology, Nanyang Technological University, Singapore 639798, Singapore
| | - Maya Kota
- Division of Endocrinology, Department of Medicine, The University of Illinois at Chicago, Chicago, IL 60612, USA
| | - Devin Dixit
- Division of Endocrinology, Department of Medicine, The University of Illinois at Chicago, Chicago, IL 60612, USA
| | - Madeline Mun
- Division of Endocrinology, Department of Medicine, The University of Illinois at Chicago, Chicago, IL 60612, USA
| | - Chong Wee Liew
- Division of Endocrinology, Department of Medicine, The University of Illinois at Chicago, Chicago, IL 60612, USA
- Department of Physiology and Biophysics, The University of Illinois at Chicago, Chicago, IL 60612, USA
| | - Yuwei Jiang
- Division of Endocrinology, Department of Medicine, The University of Illinois at Chicago, Chicago, IL 60612, USA
- Department of Physiology and Biophysics, The University of Illinois at Chicago, Chicago, IL 60612, USA
| | - Chunmei Wang
- Children’s Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA
| | - Yanlin He
- Pennington Biomedical Research Center, Louisiana State University, Baton Rouge, LA 70808, USA
| | - Pingwen Xu
- Division of Endocrinology, Department of Medicine, The University of Illinois at Chicago, Chicago, IL 60612, USA
- Department of Physiology and Biophysics, The University of Illinois at Chicago, Chicago, IL 60612, USA
| |
Collapse
|
27
|
Li J, Hu X, Xie Z, Li J, Huang C, Huang Y. Overview of growth differentiation factor 15 (GDF15) in metabolic diseases. Biomed Pharmacother 2024; 176:116809. [PMID: 38810400 DOI: 10.1016/j.biopha.2024.116809] [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: 03/18/2024] [Revised: 05/18/2024] [Accepted: 05/20/2024] [Indexed: 05/31/2024] Open
Abstract
GDF15 is a stress response cytokine and a distant member of the transforming growth factor beta (TGFβ) superfamily, its levels increase in response to cell stress and certain diseases in the serum. To exert its effects, GDF15 binds to glial-derived neurotrophic factor (GDNF) receptor alpha-like (GFRAL), which was firstly identified in 2017 and highly expressed in the brain stem. Many studies have demonstrated that elevated serum GDF15 is associated with anorexia and weight loss. Herein, we focus on the biology of GDF15, specifically how this circulating protein regulates appetite and metabolism in influencing energy homeostasis through its actions on hindbrain neurons to shed light on its impact on diseases such as obesity and anorexia/cachexia syndromes. It works as an endocrine factor and transmits metabolic signals leading to weight reduction effects by directly reducing appetite and indirectly affecting food intake through complex mechanisms, which could be a promising target for the treatment of energy-intake disorders.
Collapse
Affiliation(s)
- Jian Li
- Department of Nephrology, Institute of Kidney Diseases, West China Hospital of Sichuan University, China
| | - Xiangjun Hu
- West China School of Medicine, Sichuan University, Chengdu, China
| | - Zichuan Xie
- West China School of Medicine, Sichuan University, Chengdu, China
| | - Jiajin Li
- West China School of Medicine, Sichuan University, Chengdu, China
| | - Chen Huang
- Health Management Center, General Practice Medical Center, West China Hospital, Sichuan University, Chengdu, China; Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Yan Huang
- Health Management Center, General Practice Medical Center, West China Hospital, Sichuan University, Chengdu, China.
| |
Collapse
|
28
|
Wulff BS, Kuhre RE, Selvaraj M, Rehfeld JF, Niss K, Fels JJ, Anna S, Raun K, Gerstenberg MK. Improved leptin sensitivity and increased soluble leptin receptor concentrations may underlie the additive effects of combining PYY [, , , , , , , , , , , , , , , , , , , , , , , , , , , , , , , , , ] and exendin-4 on body weight lowering in diet-induced obese mice. Heliyon 2024; 10:e32009. [PMID: 39183855 PMCID: PMC11341243 DOI: 10.1016/j.heliyon.2024.e32009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2023] [Revised: 05/27/2024] [Accepted: 05/27/2024] [Indexed: 08/27/2024] Open
Abstract
Objective Co-treatment with long acting PYY and the GLP-1 receptor agonists has potential as an efficient obesity treatment. This study investigates whether the mechanisms behind additive reduction of food intake and weight loss depends on complementary effects in brain areas regulating food intake and if restoration of leptin sensitivity is involved. Methods Diet-induced obese (DIO) mice were co-treated with PYY(3-36) and exendin-4 (Ex4, GLP-1R agonist) for 14 days using minipumps. Leptin responsiveness was evaluated by measuring food intake and body weight after leptin injection, and gene expression profile was investigated in various of brain regions and liver. Results We show that weight loss associated with co-treatment of PYY(3-36) and Ex4 and Ex4 mono-treatment in DIO mice increased expression of several genes in area postrema (AP) known to be involved in appetite regulation and Cart, Pdyn, Bdnf and Klb were synergistically upregulated by the co-treatment. The upregulations were independent of weight loss, as shown by inclusion of a weight matched control. Moreover, PYY(3-36) and Ex4 co-treatment resulted in synergistically upregulated plasma concentrations of soluble leptin receptor (SLR) and improved sensitivity to exogenous leptin demonstrated by food intake lowering. Conclusion The study results suggest that synergistic upregulation of appetite-regulating genes in AP and improved leptin sensitivity are important mediators for the additive weight loss resulting from PYY and Ex4 co-treatment.
Collapse
Affiliation(s)
| | | | - Madhan Selvaraj
- Translational Research, Global Translation, Novo Nordisk A/S, 2760 Måløv, Denmark
| | - Jens F. Rehfeld
- Department of Clinical Biochemistry, Rigshospitalet, University of Copenhagen, DK-2100 Copenhagen, Denmark
| | - Kristoffer Niss
- Biomarker Discovery, R&ED Digital Science and Innovation, Novo Nordisk A/S, 2760 Måløv, Denmark
| | - Johannes J. Fels
- Research Bioanalysis, Global Research Technologies, Novo Nordisk A/S, 2760 Måløv, Denmark
| | - Secher Anna
- Global Drug Discovery, Novo Nordisk A/S, 2760, Måløv, Denmark
| | - Kirsten Raun
- Global Drug Discovery, Novo Nordisk A/S, 2760, Måløv, Denmark
| | | |
Collapse
|
29
|
Dumiaty Y, Underwood BM, Phy-Lim J, Chee MJ. Neurocircuitry underlying the actions of glucagon-like peptide 1 and peptide YY 3-36 in the suppression of food, drug-seeking, and anxiogenesis. Neuropeptides 2024; 105:102427. [PMID: 38579490 DOI: 10.1016/j.npep.2024.102427] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/23/2024] [Revised: 03/27/2024] [Accepted: 03/29/2024] [Indexed: 04/07/2024]
Abstract
Obesity is a critical health condition worldwide that increases the risks of comorbid chronic diseases, but it can be managed with weight loss. However, conventional interventions relying on diet and exercise are inadequate for achieving and maintaining weight loss, thus there is significant market interest for pharmaceutical anti-obesity agents. For decades, receptor agonists for the gut peptide glucagon-like peptide 1 (GLP-1) featured prominently in anti-obesity medications by suppressing appetite and food reward to elicit rapid weight loss. As the neurocircuitry underlying food motivation overlaps with that for drugs of abuse, GLP-1 receptor agonism has also been shown to decrease substance use and relapse, thus its therapeutic potential may extend beyond weight management to treat addictions. However, as prolonged use of anti-obesity drugs may increase the risk of mood-related disorders like anxiety and depression, and individuals taking GLP-1-based medication commonly report feeling demotivated, the long-term safety of such drugs is an ongoing concern. Interestingly, current research now focuses on dual agonist approaches that include GLP-1 receptor agonism to enable synergistic effects on weight loss or associated functions. GLP-1 is secreted from the same intestinal cells as the anorectic gut peptide, Peptide YY3-36 (PYY3-36), thus this review assessed the therapeutic potential and underlying neural circuits targeted by PYY3-36 when administered independently or in combination with GLP-1 to curb the appetite for food or drugs of abuse like opiates, alcohol, and nicotine. Additionally, we also reviewed animal and human studies to assess the impact, if any, for GLP-1 and/or PYY3-36 on mood-related behaviors in relation to anxiety and depression. As dual agonists targeting GLP-1 and PYY3-36 may produce synergistic effects, they can be effective at lower doses and offer an alternative approach for therapeutic benefits while mitigating undesirable side effects.
Collapse
Affiliation(s)
- Yasmina Dumiaty
- Department of Neuroscience, Carleton University, 1125 Colonel By Drive, Ottawa, ON K1S 5B6, Canada.
| | - Brett M Underwood
- Department of Neuroscience, Carleton University, 1125 Colonel By Drive, Ottawa, ON K1S 5B6, Canada.
| | - Jenny Phy-Lim
- Department of Neuroscience, Carleton University, 1125 Colonel By Drive, Ottawa, ON K1S 5B6, Canada.
| | - Melissa J Chee
- Department of Neuroscience, Carleton University, 1125 Colonel By Drive, Ottawa, ON K1S 5B6, Canada.
| |
Collapse
|
30
|
Korkutata M, De Luca R, Fitzgerald B, Arrigoni E, Scammell TE. Afferent projections to the Calca /CGRP-expressing parabrachial neurons in mice. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.07.593004. [PMID: 38766214 PMCID: PMC11100666 DOI: 10.1101/2024.05.07.593004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/22/2024]
Abstract
The parabrachial nucleus (PB), located in the dorsolateral pons, contains primarily glutamatergic neurons which regulate responses to a variety of interoceptive and cutaneous sensory signals. The lateral PB subpopulation expressing the Calca gene which produces the neuropeptide calcitonin gene-related peptide (CGRP) relays signals related to threatening stimuli such as hypercarbia, pain, and nausea, yet the afferents to these neurons are only partially understood. We mapped the afferent projections to the lateral part of the PB in mice using conventional cholera toxin B subunit (CTb) retrograde tracing, and then used conditional rabies virus retrograde tracing to map monosynaptic inputs specifically targeting the PB Calca /CGRP neurons. Using vesicular GABA (vGAT) and glutamate (vGLUT2) transporter reporter mice, we found that lateral PB neurons receive GABAergic afferents from regions such as the lateral part of the central nucleus of the amygdala, lateral dorsal subnucleus of the bed nucleus of the stria terminalis, substantia innominata, and the ventrolateral periaqueductal gray. Additionally, they receive glutamatergic afferents from the infralimbic and insular cortex, paraventricular nucleus, parasubthalamic nucleus, trigeminal complex, medullary reticular nucleus, and nucleus of the solitary tract. Using anterograde tracing and confocal microscopy, we then identified close axonal appositions between these afferents and PB Calca /CGRP neurons. Finally, we used channelrhodopsin-assisted circuit mapping to test whether some of these inputs directly synapse upon the PB Calca /CGRP neurons. These findings provide a comprehensive neuroanatomical framework for understanding the afferent projections regulating the PB Calca /CGRP neurons.
Collapse
|
31
|
Singh Alvarado J, Lutas A, Madara JC, Isaac J, Lommer C, Massengill C, Andermann ML. Transient cAMP production drives rapid and sustained spiking in brainstem parabrachial neurons to suppress feeding. Neuron 2024; 112:1416-1425.e5. [PMID: 38417435 PMCID: PMC11065603 DOI: 10.1016/j.neuron.2024.02.002] [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: 03/01/2023] [Revised: 12/01/2023] [Accepted: 02/01/2024] [Indexed: 03/01/2024]
Abstract
Brief stimuli can trigger longer-lasting brain states. G-protein-coupled receptors (GPCRs) could help sustain such states by coupling slow-timescale molecular signals to neuronal excitability. Brainstem parabrachial nucleus glutamatergic (PBNGlut) neurons regulate sustained brain states such as pain and express Gs-coupled GPCRs that increase cAMP signaling. We asked whether cAMP in PBNGlut neurons directly influences their excitability and effects on behavior. Both brief tail shocks and brief optogenetic stimulation of cAMP production in PBNGlut neurons drove minutes-long suppression of feeding. This suppression matched the duration of prolonged elevations in cAMP, protein kinase A (PKA) activity, and calcium activity in vivo and ex vivo, as well as sustained, PKA-dependent increases in action potential firing ex vivo. Shortening this elevation in cAMP reduced the duration of feeding suppression following tail shocks. Thus, molecular signaling in PBNGlut neurons helps prolong neural activity and behavioral states evoked by brief, salient bodily stimuli.
Collapse
Affiliation(s)
- Jonnathan Singh Alvarado
- Division of Endocrinology, Metabolism, and Diabetes, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02115, USA
| | - Andrew Lutas
- Division of Endocrinology, Metabolism, and Diabetes, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02115, USA; Diabetes, Endocrinology, and Obesity Branch, National Institutes of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD 20892, USA.
| | - Joseph C Madara
- Division of Endocrinology, Metabolism, and Diabetes, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02115, USA
| | - Jeremiah Isaac
- Diabetes, Endocrinology, and Obesity Branch, National Institutes of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Caroline Lommer
- Division of Endocrinology, Metabolism, and Diabetes, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02115, USA
| | | | - Mark L Andermann
- Division of Endocrinology, Metabolism, and Diabetes, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02115, USA.
| |
Collapse
|
32
|
Han Y, Xia G, Harris L, Liu P, Guan D, Wu Q. Reversal of Obesity by Enhancing Slow-wave Sleep via a Prokineticin Receptor Neural Circuit. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.04.30.591948. [PMID: 38746393 PMCID: PMC11092673 DOI: 10.1101/2024.04.30.591948] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/16/2024]
Abstract
Obese subjects often exhibit hypersomnia accompanied by severe sleep fragmentation, while emerging evidence suggests that poor sleep quality promotes overeating and exacerbates diet-induced obesity (DIO). However, the neural circuit and signaling mechanism underlying the reciprocal control of appetite and sleep is yet not elucidated. Here, we report a neural circuit where prokineticin receptor 2 (PROKR2)-expressing neurons within the parabrachial nucleus (PBN) of the brainstem received direct projections from neuropeptide Y receptor Y2 (NPY2R)-expressing neurons within the lateral preoptic area (LPO) of the hypothalamus. The RNA-Seq results revealed Prokr2 in the PBN is the most regulated GPCR signaling gene that is responsible for comorbidity of obesity and sleep dysfunction. Furthermore, those NPY2R LPO neurons are minimally active during NREM sleep and maximally active during wakefulness and REM sleep. Activation of the NPY2R LPO →PBN circuit or the postsynaptic PROKR2 PBN neurons suppressed feeding of a high-fat diet and abrogated morbid sleep patterns in DIO mice. Further studies showed that genetic ablation of the PROKR2 signaling within PROKR2 PBN neurons alleviated the hyperphagia and weight gain, and restored sleep dysfunction in DIO mice. We further discovered pterostilbene, a plant-derived stilbenoid, is a powerful anti-obesity and sleep-improving agent, robustly suppressed hyperphagia and promoted reconstruction of a healthier sleep architecture, thereby leading to significant weight loss. Collectively, our results unveil a neural mechanism for the reciprocal control of appetite and sleep, through which pterostilbene, along with a class of similarly structured compounds, may be developed as effective therapeutics for tackling obesity and sleep disorders.
Collapse
|
33
|
Ding W, Weltzien H, Peters C, Klein R. Nausea-induced suppression of feeding is mediated by central amygdala Dlk1-expressing neurons. Cell Rep 2024; 43:113990. [PMID: 38551964 DOI: 10.1016/j.celrep.2024.113990] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2023] [Revised: 01/23/2024] [Accepted: 03/07/2024] [Indexed: 04/28/2024] Open
Abstract
The motivation to eat is suppressed by satiety and aversive stimuli such as nausea. The neural circuit mechanisms of appetite suppression by nausea are not well understood. Pkcδ neurons in the lateral subdivision of the central amygdala (CeA) suppress feeding in response to satiety signals and nausea. Here, we characterized neurons enriched in the medial subdivision (CeM) of the CeA marked by expression of Dlk1. CeADlk1 neurons are activated by nausea, but not satiety, and specifically suppress feeding induced by nausea. Artificial activation of CeADlk1 neurons suppresses drinking and social interactions, suggesting a broader function in attenuating motivational behavior. CeADlk1 neurons form projections to many brain regions and exert their anorexigenic activity by inhibition of neurons of the parabrachial nucleus. CeADlk1 neurons are inhibited by appetitive CeA neurons, but also receive long-range monosynaptic inputs from multiple brain regions. Our results illustrate a CeA circuit that regulates nausea-induced feeding suppression.
Collapse
Affiliation(s)
- Wenyu Ding
- Max Planck Institute for Biological Intelligence, Am Klopferspitz 18, 82152 Martinsried, Germany
| | - Helena Weltzien
- Max Planck Institute for Biological Intelligence, Am Klopferspitz 18, 82152 Martinsried, Germany
| | - Christian Peters
- Max Planck Institute for Biological Intelligence, Am Klopferspitz 18, 82152 Martinsried, Germany
| | - Rüdiger Klein
- Max Planck Institute for Biological Intelligence, Am Klopferspitz 18, 82152 Martinsried, Germany.
| |
Collapse
|
34
|
Jaramillo JCM, Aitken CM, Lawrence AJ, Ryan PJ. Oxytocin-receptor-expressing neurons in the lateral parabrachial nucleus activate widespread brain regions predominantly involved in fluid satiation. J Chem Neuroanat 2024; 137:102403. [PMID: 38452468 DOI: 10.1016/j.jchemneu.2024.102403] [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: 01/22/2024] [Revised: 02/21/2024] [Accepted: 02/23/2024] [Indexed: 03/09/2024]
Abstract
Fluid satiation is an important signal and aspect of body fluid homeostasis. Oxytocin-receptor-expressing neurons (OxtrPBN) in the dorsolateral subdivision of the lateral parabrachial nucleus (dl LPBN) are key neurons which regulate fluid satiation. In the present study, we investigated brain regions activated by stimulation of OxtrPBN neurons in order to better characterise the fluid satiation neurocircuitry in mice. Chemogenetic activation of OxtrPBN neurons increased Fos expression (a proxy marker for neuronal activation) in known fluid-regulating brain nuclei, as well as other regions that have unclear links to fluid regulation and which are likely involved in regulating other functions such as arousal and stress relief. In addition, we analysed and compared Fos expression patterns between chemogenetically-activated fluid satiation and physiological-induced fluid satiation. Both models of fluid satiation activated similar brain regions, suggesting that the chemogenetic model of stimulating OxtrPBN neurons is a relevant model of physiological fluid satiation. A deeper understanding of this neural circuit may lead to novel molecular targets and creation of therapeutic agents to treat fluid-related disorders.
Collapse
Affiliation(s)
- Janine C M Jaramillo
- The Florey Institute of Neuroscience and Mental Health, Parkville, Victoria, Australia; The Florey Department of Neuroscience and Mental Health, University of Melbourne, Parkville, Victoria, Australia
| | - Connor M Aitken
- The Florey Institute of Neuroscience and Mental Health, Parkville, Victoria, Australia; The Florey Department of Neuroscience and Mental Health, University of Melbourne, Parkville, Victoria, Australia
| | - Andrew J Lawrence
- The Florey Institute of Neuroscience and Mental Health, Parkville, Victoria, Australia; The Florey Department of Neuroscience and Mental Health, University of Melbourne, Parkville, Victoria, Australia
| | - Philip J Ryan
- The Florey Institute of Neuroscience and Mental Health, Parkville, Victoria, Australia; The Florey Department of Neuroscience and Mental Health, University of Melbourne, Parkville, Victoria, Australia.
| |
Collapse
|
35
|
Ispoglou T, McCullough D, Windle A, Nair S, Cox N, White H, Burke D, Kanatas A, Prokopidis K. Addressing cancer anorexia-cachexia in older patients: Potential therapeutic strategies and molecular pathways. Clin Nutr 2024; 43:552-566. [PMID: 38237369 DOI: 10.1016/j.clnu.2024.01.009] [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: 11/03/2022] [Revised: 01/04/2024] [Accepted: 01/10/2024] [Indexed: 02/03/2024]
Abstract
Cancer cachexia (CC) syndrome, a feature of cancer-associated muscle wasting, is particularly pronounced in older patients, and is characterised by decreased energy intake and upregulated skeletal muscle catabolic pathways. To address CC, appetite stimulants, anabolic drugs, cytokine mediators, essential amino acid supplementation, nutritional counselling, cognitive behavioural therapy, and enteral nutrition have been utilised. However, pharmacological treatments that have also shown promising results, such as megestrol acetate, anamorelin, thalidomide, and delta-9-tetrahydrocannabinol, have been associated with gastrointestinal and cardiovascular complications. Emerging evidence on the efficacy of probiotics in modulating gut microbiota also presents a promising adjunct to traditional therapies, potentially enhancing nutritional absorption and systemic inflammation control. Additionally, low-dose olanzapine has demonstrated improved appetite and weight management in older patients undergoing chemotherapy, offering a potential refinement to current therapeutic approaches. This review aims to elucidate the molecular mechanisms underpinning CC, with a particular focus on the role of anorexia in exacerbating muscle wasting, and to propose pharmacological and non-pharmacological strategies to mitigate this syndrome, particularly emphasising the needs of an older demographic. Future research targeting CC should focus on refining appetite-stimulating drugs with fewer side-effects, specifically catering to the needs of older patients, and investigating nutritional factors that can either enhance appetite or minimise suppression of appetite in individuals with CC, especially within this vulnerable group.
Collapse
Affiliation(s)
| | | | - Angela Windle
- Department of Nursing and Midwifery, School of Human and Health Sciences, University of Huddersfield, Huddersfield, UK; School of Medicine, University of Leeds, Leeds, UK
| | | | - Natalie Cox
- Academic Geriatric Medicine, Faculty of Medicine, University of Southampton, Southampton, UK
| | - Helen White
- School of Health, Leeds Beckett University, Leeds, UK
| | - Dermot Burke
- School of Medicine, University of Leeds, Leeds, UK
| | | | - Konstantinos Prokopidis
- Department of Musculoskeletal and Ageing Science, Institute of Life Course and Medical Sciences, University of Liverpool, Liverpool, UK; Liverpool Centre for Cardiovascular Science, University of Liverpool, Liverpool, UK
| |
Collapse
|
36
|
Albrechet‐Souza L, Kasten CR, Bertagna NB, Wills TA. Sex-specific negative affect-like behaviour and parabrachial nucleus activation induced by BNST stimulation in adult mice with adolescent alcohol history. Addict Biol 2024; 29:e13366. [PMID: 38380710 PMCID: PMC10883599 DOI: 10.1111/adb.13366] [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: 09/07/2023] [Revised: 11/23/2023] [Accepted: 11/26/2023] [Indexed: 02/22/2024]
Abstract
Adolescent alcohol use is a strong predictor for the subsequent development of alcohol use disorders later in life. Additionally, adolescence is a critical period for the onset of affective disorders, which can contribute to problematic drinking behaviours and relapse, particularly in females. Previous studies from our laboratory have shown that exposure to adolescent intermittent ethanol (AIE) vapour alters glutamatergic transmission in the bed nucleus of the stria terminalis (BNST) and, when combined with adult stress, elicits sex-specific changes in glutamatergic plasticity and negative affect-like behaviours in mice. Building on these findings, the current work investigated whether BNST stimulation could substitute for stress exposure to increase the latency to consume a palatable food in a novel context (hyponeophagia) and promote social avoidance in adult mice with AIE history. Given the dense connections between the BNST and the parabrachial nucleus (PBN), a region involved in mediating threat assessment and feeding behaviours, we hypothesized that increased negative affect-like behaviours would be associated with PBN activation. Our results revealed that the chemogenetic stimulation of the dorsolateral BNST induced hyponeophagia in females with AIE history, but not in female controls or males of either group. Social interaction remained unaffected in both sexes. Notably, this behavioural phenotype was associated with higher activation of calcitonin gene-related peptide and dynorphin cells in the PBN. These findings provide new insights into the neurobiological mechanisms underlying the development of negative affect in females and highlight the potential involvement of the BNST-PBN circuitry in regulating emotional responses to alcohol-related stimuli.
Collapse
Affiliation(s)
- Lucas Albrechet‐Souza
- Department of Cell Biology & Anatomy, School of MedicineLouisiana State University Health Sciences CenterNew OrleansLouisianaUSA
- Alcohol & Drug Center of Excellence, School of MedicineLouisiana State University Health Sciences CenterNew OrleansLouisianaUSA
| | - Chelsea R. Kasten
- Department of Cell Biology & Anatomy, School of MedicineLouisiana State University Health Sciences CenterNew OrleansLouisianaUSA
| | - Natalia B. Bertagna
- Department of Cell Biology & Anatomy, School of MedicineLouisiana State University Health Sciences CenterNew OrleansLouisianaUSA
- Department of PharmacologyFederal University of São PauloSão PauloSPBrazil
| | - Tiffany A. Wills
- Department of Cell Biology & Anatomy, School of MedicineLouisiana State University Health Sciences CenterNew OrleansLouisianaUSA
- Alcohol & Drug Center of Excellence, School of MedicineLouisiana State University Health Sciences CenterNew OrleansLouisianaUSA
- Neuroscience Center of Excellence, School of MedicineLouisiana State University Health Sciences CenterNew OrleansLouisianaUSA
| |
Collapse
|
37
|
Coizet V, Al Tannir R, Pautrat A, Overton PG. Separation of Channels Subserving Approach and Avoidance/Escape at the Level of the Basal Ganglia and Related Brainstem Structures. Curr Neuropharmacol 2024; 22:1473-1490. [PMID: 37594168 PMCID: PMC11097992 DOI: 10.2174/1570159x21666230818154903] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2022] [Revised: 03/23/2023] [Accepted: 03/29/2023] [Indexed: 08/19/2023] Open
Abstract
The basal ganglia have the key function of directing our behavior in the context of events from our environment and/or our internal state. This function relies on afferents targeting the main input structures of the basal ganglia, entering bids for action selection at the level of the striatum or signals for behavioral interruption at the level of the subthalamic nucleus, with behavioral reselection facilitated by dopamine signaling. Numerous experiments have studied action selection in relation to inputs from the cerebral cortex. However, less is known about the anatomical and functional link between the basal ganglia and the brainstem. In this review, we describe how brainstem structures also project to the main input structures of the basal ganglia, namely the striatum, the subthalamic nucleus and midbrain dopaminergic neurons, in the context of approach and avoidance (including escape from threat), two fundamental, mutually exclusive behavioral choices in an animal's repertoire in which the brainstem is strongly involved. We focus on three particularly well-described loci involved in approach and avoidance, namely the superior colliculus, the parabrachial nucleus and the periaqueductal grey nucleus. We consider what is known about how these structures are related to the basal ganglia, focusing on their projections toward the striatum, dopaminergic neurons and subthalamic nucleus, and explore the functional consequences of those interactions.
Collapse
Affiliation(s)
- Véronique Coizet
- Grenoble Institute of Neuroscience, University Grenoble Alpes, Bâtiment E.J. Safra - Chemin Fortuné Ferrini - 38700 La Tronche France;
| | - Racha Al Tannir
- Grenoble Institute of Neuroscience, University Grenoble Alpes, Bâtiment E.J. Safra - Chemin Fortuné Ferrini - 38700 La Tronche France;
| | - Arnaud Pautrat
- Grenoble Institute of Neuroscience, University Grenoble Alpes, Bâtiment E.J. Safra - Chemin Fortuné Ferrini - 38700 La Tronche France;
| | - Paul G. Overton
- Department of Psychology, University of Sheffield, Sheffield, United Kingdom
| |
Collapse
|
38
|
Song J, Choi SY. Arcuate Nucleus of the Hypothalamus: Anatomy, Physiology, and Diseases. Exp Neurobiol 2023; 32:371-386. [PMID: 38196133 PMCID: PMC10789173 DOI: 10.5607/en23040] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2023] [Revised: 12/24/2023] [Accepted: 12/27/2023] [Indexed: 01/11/2024] Open
Abstract
The hypothalamus is part of the diencephalon and has several nuclei, one of which is the arcuate nucleus. The arcuate nucleus of hypothalamus (ARH) consists of neuroendocrine neurons and centrally-projecting neurons. The ARH is the center where the homeostasis of nutrition/metabolism and reproduction are maintained. As such, dysfunction of the ARH can lead to disorders of nutrition/metabolism and reproduction. Here, we review various types of neurons in the ARH and several genetic disorders caused by mutations in the ARH.
Collapse
Affiliation(s)
- Juhyun Song
- Department of Anatomy, Chonnam National University Medical School, Hwasun 58128, Korea
| | - Seok-Yong Choi
- Department of Biomedical Sciences, Chonnam National University Medical School, Hwasun 58128, Korea
| |
Collapse
|
39
|
Fila M, Chojnacki J, Pawlowska E, Sobczuk P, Chojnacki C, Blasiak J. The Ketogenic Diet in the Prevention of Migraines in the Elderly. Nutrients 2023; 15:4998. [PMID: 38068855 PMCID: PMC10707997 DOI: 10.3390/nu15234998] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2023] [Revised: 11/23/2023] [Accepted: 11/30/2023] [Indexed: 12/18/2023] Open
Abstract
Migraines display atypical age dependence, as the peak of their prevalence occurs between the ages of 20-40 years. With age, headache attacks occur less frequently and are characterized by a lower amplitude. However, both diagnosis and therapy of migraines in the elderly are challenging due to multiple comorbidities and polypharmacy. Dietary components and eating habits are migraine triggers; therefore, nutrition is a main target in migraine prevention. Several kinds of diets were proposed to prevent migraines, but none are commonly accepted due to inconsistent results obtained in different studies. The ketogenic diet is featured by very low-carbohydrate and high-fat contents. It may replace glucose with ketone bodies as the primary source of energy production. The ketogenic diet and the actions of ketone bodies are considered beneficial in several aspects of health, including migraine prevention, but studies on the ketogenic diet in migraines are not standardized and poorly evidenced. Apart from papers claiming beneficial effects of the ketogenic diet in migraines, several studies have reported that increased levels of ketone bodies may be associated with all-cause and incident heart failure mortality in older adults and are supported by research on mice showing that the ketogenic diets and diet supplementation with a human ketone body precursor may cause life span shortening. Therefore, despite reports showing a beneficial effect of the ketogenic diet in migraines, such a diet requires further studies, including clinical trials, to verify whether it should be recommended in older adults with migraines.
Collapse
Affiliation(s)
- Michal Fila
- Department of Developmental Neurology and Epileptology, Polish Mother’s Memorial Hospital—Research Institute, 93-338 Lodz, Poland;
| | - Jan Chojnacki
- Department of Clinical Nutrition and Gastroenterological Diagnostics, Medical University of Lodz, 90-647 Lodz, Poland; (J.C.), (C.C.)
| | - Elzbieta Pawlowska
- Department of Pediatric Dentistry, Medical University of Lodz, 92-217 Lodz, Poland;
| | - Piotr Sobczuk
- Emergency Medicine and Disaster Medicine Department, Medical University of Lodz, 92-209 Lodz, Poland;
- Department of Orthopaedics and Traumatology, Polish Mother’s Memorial Hospital—Research Institute, Rzgowska 281, 93-338 Lodz, Poland
| | - Cezary Chojnacki
- Department of Clinical Nutrition and Gastroenterological Diagnostics, Medical University of Lodz, 90-647 Lodz, Poland; (J.C.), (C.C.)
| | - Janusz Blasiak
- Faculty of Medicine, Collegium Medicum, The Mazovian Academy in Plock, 09-402 Plock, Poland
| |
Collapse
|
40
|
Aitken CM, Jaramillo JCM, Davis W, Brennan‐Xie L, McDougall SJ, Lawrence AJ, Ryan PJ. Feeding signals inhibit fluid-satiation signals in the mouse lateral parabrachial nucleus to increase intake of highly palatable, caloric solutions. J Neurochem 2023; 167:648-667. [PMID: 37855271 PMCID: PMC10952698 DOI: 10.1111/jnc.15991] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2023] [Revised: 10/01/2023] [Accepted: 10/03/2023] [Indexed: 10/20/2023]
Abstract
Chemogenetic activation of oxytocin receptor-expressing neurons in the parabrachial nucleus (OxtrPBN neurons) acts as a satiation signal for water. In this research, we investigated the effect of activating OxtrPBN neurons on satiation for different types of fluids. Chemogenetic activation of OxtrPBN neurons in male and female transgenic OxtrCre mice robustly suppressed the rapid, initial (15-min) intake of several solutions after dehydration: water, sucrose, ethanol and saccharin, but only slightly decreased intake of Ensure®, a highly caloric solution (1 kcal/mL; containing 3.72 g protein, 3.27 g fat, 13.42 g carbohydrates, and 1.01 g dietary fibre per 100 mL). OxtrPBN neuron activation also suppressed cumulative, longer-term (2-h) intake of lower caloric, less palatable solutions, but not highly caloric, palatable solutions. These results suggest that OxtrPBN neurons predominantly control initial fluid-satiation responses after rehydration, but not longer-term intake of highly caloric, palatable solutions. The suppression of fluid intake was not because of anxiogenesis, but because OxtrPBN neuron activation decreased anxiety-like behaviour. To investigate the role of different PBN subdivisions on the intake of different solutions, we examined FOS as a proxy marker of PBN neuron activation. Different PBN subdivisions were activated by different solutions: the dorsolateral PBN similarly by all fluids; the external lateral PBN by caloric but not non-caloric solutions; and the central lateral PBN primarily by highly palatable solutions, suggesting PBN subdivisions regulate different aspects of fluid intake. To explore the possible mechanisms underlying the minimal suppression of Ensure® after OxtrPBN neuron activation, we demonstrated in in vitro slice recordings that the feeding-associated agouti-related peptide (AgRP) inhibited OxtrPBN neuron firing in a concentration-related manner, suggesting possible inhibition by feeding-related neurocircuitry of fluid satiation neurocircuitry. Overall, this research suggests that although palatable beverages like sucrose- and ethanol-containing beverages activate fluid satiation signals encoded by OxtrPBN neurons, these neurons can be inhibited by hunger-related signals (agouti-related peptide, AgRP), which may explain why these fluids are often consumed in excess of what is required for fluid satiation.
Collapse
Affiliation(s)
- Connor M. Aitken
- Florey Institute of Neuroscience & Mental HealthUniversity of MelbourneParkvilleVictoriaAustralia
- Florey Department of Neuroscience & Mental HealthUniversity of MelbourneParkvilleVictoriaAustralia
| | - Janine C. M. Jaramillo
- Florey Institute of Neuroscience & Mental HealthUniversity of MelbourneParkvilleVictoriaAustralia
- Florey Department of Neuroscience & Mental HealthUniversity of MelbourneParkvilleVictoriaAustralia
| | - Warren Davis
- Florey Institute of Neuroscience & Mental HealthUniversity of MelbourneParkvilleVictoriaAustralia
- Florey Department of Neuroscience & Mental HealthUniversity of MelbourneParkvilleVictoriaAustralia
| | - Liam Brennan‐Xie
- Florey Institute of Neuroscience & Mental HealthUniversity of MelbourneParkvilleVictoriaAustralia
| | - Stuart J. McDougall
- Florey Institute of Neuroscience & Mental HealthUniversity of MelbourneParkvilleVictoriaAustralia
| | - Andrew J. Lawrence
- Florey Institute of Neuroscience & Mental HealthUniversity of MelbourneParkvilleVictoriaAustralia
- Florey Department of Neuroscience & Mental HealthUniversity of MelbourneParkvilleVictoriaAustralia
| | - Philip J. Ryan
- Florey Institute of Neuroscience & Mental HealthUniversity of MelbourneParkvilleVictoriaAustralia
- Florey Department of Neuroscience & Mental HealthUniversity of MelbourneParkvilleVictoriaAustralia
| |
Collapse
|
41
|
Zlatkovic J, Dalmau Gasull A, Hägg D, Font-Gironès F, Bellman J, Meister B, Palsdottir V, Ruud J, Ohlsson C, Dickson SL, Anesten F, Jansson JO. Reduction of body weight by increased loading is associated with activation of norepinephrine neurones in the medial nucleus of the solitary tract. J Neuroendocrinol 2023; 35:e13352. [PMID: 37885347 DOI: 10.1111/jne.13352] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/03/2023] [Revised: 10/10/2023] [Accepted: 10/11/2023] [Indexed: 10/28/2023]
Abstract
We previously provided evidence supporting the existence of a novel leptin-independent body weight homeostat ("the gravitostat") that senses body weight and then initiates a homeostatic feed-back regulation of body weight. We, herein, hypothesize that this feed-back regulation involves a CNS mechanism. To identify populations of neurones of importance for the putative feed-back signal induced by increased loading, high-fat diet-fed rats or mice were implanted intraperitoneally or subcutaneously with capsules weighing ∼15% (Load) or ∼2.5% (Control) of body weight. At 3-5 days after implantation, neuronal activation was assessed in different parts of the brain/brainstem by immunohistochemical detection of FosB. Implantation of weighted capsules, both subcutaneous and intraperitoneal, induced FosB in specific neurones in the medial nucleus of the solitary tract (mNTS), known to integrate information about the metabolic status of the body. These neurones also expressed tyrosine hydroxylase (TH) and dopamine-beta-hydroxylase (DbH), a pattern typical of norepinephrine neurones. In functional studies, we specifically ablated norepinephrine neurones in mNTS, which attenuated the feed-back regulation of increased load on body weight and food intake. In conclusion, increased load appears to reduce body weight and food intake via activation of norepinephrine neurones in the mNTS.
Collapse
Affiliation(s)
- Jovana Zlatkovic
- Departement of Neuroscience and Physiology, University of Gothenburg, Gothenburg, Sweden
| | | | - Daniel Hägg
- Departement of Neuroscience and Physiology, University of Gothenburg, Gothenburg, Sweden
| | - Ferran Font-Gironès
- Departement of Neuroscience and Physiology, University of Gothenburg, Gothenburg, Sweden
| | - Jakob Bellman
- Departement of Neuroscience and Physiology, University of Gothenburg, Gothenburg, Sweden
| | - Björn Meister
- Institute of Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | | | - Johan Ruud
- Departement of Neuroscience and Physiology, University of Gothenburg, Gothenburg, Sweden
| | - Claes Ohlsson
- Center for Bone and Arthritis Research, University of Gothenburg, Gothenburg, Sweden
| | - Suzanne L Dickson
- Departement of Neuroscience and Physiology, University of Gothenburg, Gothenburg, Sweden
| | - Fredrik Anesten
- Departement of Neuroscience and Physiology, University of Gothenburg, Gothenburg, Sweden
| | - John-Olov Jansson
- Departement of Neuroscience and Physiology, University of Gothenburg, Gothenburg, Sweden
| |
Collapse
|
42
|
Ye Q, Nunez J, Zhang X. Zona incerta dopamine neurons encode motivational vigor in food seeking. SCIENCE ADVANCES 2023; 9:eadi5326. [PMID: 37976360 PMCID: PMC10656063 DOI: 10.1126/sciadv.adi5326] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/02/2023] [Accepted: 10/18/2023] [Indexed: 11/19/2023]
Abstract
Energy deprivation triggers food seeking to ensure homeostatic consumption, but the neural coding of motivational vigor in food seeking during physical hunger remains unknown. Here, we report that ablation of dopamine (DA) neurons in zona incerta (ZI) but not ventral tegmental area potently impaired food seeking after fasting. ZI DA neurons and their projections to paraventricular thalamus (PVT) were quickly activated for food approach but inhibited during food consumption. Chemogenetic manipulation of ZI DA neurons bidirectionally regulated feeding motivation to control meal frequency but not meal size for food intake. Activation of ZI DA neurons promoted, but silencing of these neurons blocked, contextual memory associate with food reward. In addition, selective activation of ZI DA projections to PVT promoted food seeking for food consumption and transited positive-valence signals. Together, these findings reveal that ZI DA neurons encode motivational vigor in food seeking for food consumption through their projections to PVT.
Collapse
|
43
|
Condon LF, Yu Y, Park S, Cao F, Pauli JL, Nelson TS, Palmiter RD. Parabrachial Calca neurons drive nociplasticity. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.10.26.564223. [PMID: 37961621 PMCID: PMC10634894 DOI: 10.1101/2023.10.26.564223] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/15/2023]
Abstract
Pain that persists beyond the time required for tissue healing and pain that arises in the absence of tissue injury are poorly understood phenomena mediated by plasticity within the central nervous system. The parabrachial nucleus (PBN) is a hub that relays aversive sensory information and appears to play a role in nociplasticity. Here, by preventing PBN Calca neurons from releasing neurotransmitter or directly stimulating them we demonstrate that activation of Calca neurons is both necessary for the manifestation of chronic pain after nerve ligation and is sufficient to drive nociplasticity in wild-type mice. Aversive stimuli such as exposure to nitroglycerin, cisplatin, or LiCl can drive nociplasticity in a Calca-neuron-dependent manner. Calcium fluorescence imaging reveals that nitroglycerin activates PBN Calca neurons and potentiates their responses to mechanical stimulation. The activity and excitability of Calca neurons increased for several days after aversive events, but prolonged nociplasticity likely occurs in downstream circuitry.
Collapse
Affiliation(s)
- Logan F Condon
- Howard Hughes Medical Institute, University of Washington, Seattle, WA 98195, USA
- Departments of Biochemistry and Genome Sciences, University of Washington, Seattle, WA 98195, USA
- Graduate Program in Neuroscience, University of Washington, Seattle, WA 98195, USA
- Medical Scientist Training Program, University of Washington, Seattle, WA 98195, USA
| | - Ying Yu
- Howard Hughes Medical Institute, University of Washington, Seattle, WA 98195, USA
- Departments of Biochemistry and Genome Sciences, University of Washington, Seattle, WA 98195, USA
| | - Sekun Park
- Howard Hughes Medical Institute, University of Washington, Seattle, WA 98195, USA
- Departments of Biochemistry and Genome Sciences, University of Washington, Seattle, WA 98195, USA
| | - Feng Cao
- Howard Hughes Medical Institute, University of Washington, Seattle, WA 98195, USA
- Departments of Biochemistry and Genome Sciences, University of Washington, Seattle, WA 98195, USA
| | - Jordan L Pauli
- Howard Hughes Medical Institute, University of Washington, Seattle, WA 98195, USA
- Departments of Biochemistry and Genome Sciences, University of Washington, Seattle, WA 98195, USA
| | - Tyler S Nelson
- Department of Molecular Pathobiology, College of Dentistry, New York University, NY 10010, USA
| | - Richard D Palmiter
- Howard Hughes Medical Institute, University of Washington, Seattle, WA 98195, USA
- Departments of Biochemistry and Genome Sciences, University of Washington, Seattle, WA 98195, USA
- Graduate Program in Neuroscience, University of Washington, Seattle, WA 98195, USA
- Lead Contact
| |
Collapse
|
44
|
Jiang SZ, Zhang HY, Eiden LE. PACAP Controls Endocrine and Behavioral Stress Responses via Separate Brain Circuits. BIOLOGICAL PSYCHIATRY GLOBAL OPEN SCIENCE 2023; 3:673-685. [PMID: 37881538 PMCID: PMC10593940 DOI: 10.1016/j.bpsgos.2023.04.001] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2023] [Revised: 04/04/2023] [Accepted: 04/06/2023] [Indexed: 10/27/2023] Open
Abstract
Background The neuropeptide PACAP (pituitary adenylate cyclase-activating polypeptide) is a master regulator of central and peripheral stress responses, yet it is not clear how PACAP projections throughout the brain execute endocrine and behavioral stress responses. Methods We used AAV (adeno-associated virus) neuronal tracing, an acute restraint stress (ARS) paradigm, and intersectional genetics, in C57BL/6 mice, to identify PACAP-containing circuits controlling stress-induced behavior and endocrine activation. Results PACAP deletion from forebrain excitatory neurons, including a projection directly from medial prefrontal cortex to hypothalamus, impairs c-fos activation and corticotropin-releasing hormone (CRH) messenger RNA elevation in the paraventricular nucleus after 2 hours of restraint, without affecting ARS-induced hypophagia, or c-fos elevation in nonhypothalamic brain. Elimination of PACAP within projections from lateral parabrachial nucleus to extended amygdala, on the other hand, attenuates ARS-induced hypophagia, along with extended amygdala fos induction, without affecting ARS-induced CRH messenger RNA elevation in the paraventricular nucleus. PACAP projections to extended amygdala terminate at protein kinase C delta type (PKCδ) neurons in both the central amygdala and the oval bed nucleus of the stria terminalis. Silencing of PKCδ neurons in the central amygdala, but not in the oval bed nucleus of the stria terminalis, attenuates ARS-induced hypophagia. Experiments were carried out in mice of both sexes with n ≥ 3 per group. Conclusions A frontocortical descending PACAP projection controls paraventricular nucleus CRH messenger RNA production to maintain hypothalamic-pituitary-adrenal axis activation and regulate the endocrine response to stress. An ascending PACAPergic projection from the external lateral parabrachial nucleus to PKCδ neurons in the central amygdala regulates behavioral responses to stress. Defining two separate limbs of the acute stress response provides broader insight into the specific brain circuitry engaged by the psychogenic stress response.
Collapse
Affiliation(s)
- Sunny Zhihong Jiang
- Section on Molecular Neuroscience, National Institute of Mental Health Intramural Research Program, Bethesda, Maryland
| | - Hai-Ying Zhang
- Section on Molecular Neuroscience, National Institute of Mental Health Intramural Research Program, Bethesda, Maryland
| | - Lee E. Eiden
- Section on Molecular Neuroscience, National Institute of Mental Health Intramural Research Program, Bethesda, Maryland
| |
Collapse
|
45
|
Behrens R, Dutschmann M, Trewella M, Mazzone SB, Moe AAK. Regulation of vagally-evoked respiratory responses by the lateral parabrachial nucleus in the mouse. Respir Physiol Neurobiol 2023; 316:104141. [PMID: 37597796 DOI: 10.1016/j.resp.2023.104141] [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: 06/28/2023] [Revised: 08/04/2023] [Accepted: 08/14/2023] [Indexed: 08/21/2023]
Abstract
Vagal sensory inputs to the brainstem can alter breathing through the modulation of pontomedullary respiratory circuits. In this study, we set out to investigate the localised effects of modulating lateral parabrachial nucleus (LPB) activity on vagally-evoked changes in breathing pattern. In isoflurane-anaesthetised and instrumented mice, electrical stimulation of the vagus nerve (eVNS) produced stimulation frequency-dependent changes in diaphragm electromyograph (dEMG) activity with an evoked tachypnoea and apnoea at low and high stimulation frequencies, respectively. Muscimol microinjections into the LPB significantly attenuated eVNS-evoked respiratory rate responses. Notably, muscimol injections reaching the caudal LPB, previously unrecognised for respiratory modulation, potently modulated eVNS-evoked apnoea, whilst muscimol injections reaching the intermediate LPB selectively modulated the eVNS-evoked tachypnoea. The effects of muscimol on eVNS-evoked breathing rate changes occurred without altering basal eupneic breathing. These results highlight novel roles for the LPB in regulating vagally-evoked respiratory reflexes.
Collapse
Affiliation(s)
- Robert Behrens
- Department of Anatomy and Physiology, University of Melbourne, VIC, Australia
| | - Mathias Dutschmann
- Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville, VIC, Australia
| | - Matthew Trewella
- Department of Anatomy and Physiology, University of Melbourne, VIC, Australia
| | - Stuart B Mazzone
- Department of Anatomy and Physiology, University of Melbourne, VIC, Australia.
| | - Aung Aung Kywe Moe
- Department of Anatomy and Physiology, University of Melbourne, VIC, Australia; Department of Medical Imaging and Radiation Sciences, Monash University, Clayton, Australia
| |
Collapse
|
46
|
Sweeney P, Gimenez LE, Hernandez CC, Cone RD. Targeting the central melanocortin system for the treatment of metabolic disorders. Nat Rev Endocrinol 2023; 19:507-519. [PMID: 37365323 DOI: 10.1038/s41574-023-00855-y] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 05/18/2023] [Indexed: 06/28/2023]
Abstract
A large body of preclinical and clinical data shows that the central melanocortin system is a promising therapeutic target for treating various metabolic disorders such as obesity and cachexia, as well as anorexia nervosa. Setmelanotide, which functions by engaging the central melanocortin circuitry, was approved by the FDA in 2020 for use in certain forms of syndromic obesity. Furthermore, the FDA approvals in 2019 of two peptide drugs targeting melanocortin receptors for the treatment of generalized hypoactive sexual desire disorder (bremelanotide) and erythropoietic protoporphyria-associated phototoxicity (afamelanotide) demonstrate the safety of this class of peptides. These approvals have also renewed excitement in the development of therapeutics targeting the melanocortin system. Here, we review the anatomy and function of the melanocortin system, discuss progress and challenges in developing melanocortin receptor-based therapeutics, and outline potential metabolic and behavioural disorders that could be addressed using pharmacological agents targeting these receptors.
Collapse
Affiliation(s)
- Patrick Sweeney
- School of Molecular and Cellular Biology, College of Liberal Arts and Sciences, University of Illinois Urbana-Champaign, Champaign, IL, USA
| | - Luis E Gimenez
- Life Sciences Institute, University of Michigan, Ann Arbor, MI, USA
| | | | - Roger D Cone
- Life Sciences Institute, University of Michigan, Ann Arbor, MI, USA.
- Department of Molecular and Integrative Physiology, School of Medicine, University of Michigan, Ann Arbor, MI, USA.
- Department of Molecular, Cellular, and Developmental Biology, College of Literature Science and the Arts, University of Michigan, Ann Arbor, MI, USA.
| |
Collapse
|
47
|
Sandoval-Caballero C, Luarte L, Jiménez Y, Jaque C, Cifuentes F, Arenas GA, Figueroa M, Jara J, Olszewski PK, Teske JA, Pérez-Leighton CE. Meta-analysis of pre-clinical studies on the effects of opioid receptor ligands on food intake, motivation, and choice. Neurosci Biobehav Rev 2023; 152:105288. [PMID: 37331611 DOI: 10.1016/j.neubiorev.2023.105288] [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: 04/28/2023] [Revised: 06/12/2023] [Accepted: 06/15/2023] [Indexed: 06/20/2023]
Abstract
The opioid receptors (OR) regulate food intake. Still, despite extensive pre-clinical research, the overall effects and individual contribution of the mu (MOR), kappa (KOR), and delta (DOR) OR subtypes to feeding behaviors and food intake remain unclear. To address this, we conducted a pre-registered systematic search and meta-analysis of rodent dose-response studies to evaluate the impact of central and peripheral administration of non-selective and selective OR ligands on intake, motivation, and choice of food. All studies had a high bias risk. Still, the meta-analysis confirmed the overall orexigenic and anorexigenic effects of OR agonists and antagonists, respectively. Our results support a larger orexigenic role for central MOR agonists among OR subtypes and that peripheral OR antagonists reduce motivation for and intake of preferred foods. In binary food choice studies, peripheral OR agonists selectively increase the intake of fat-preferred foods; in contrast, they did not increase the intake of sweet carbohydrate-preferred foods. Overall, these data support that OR regulation of intake, motivation, and choice is influenced by food macronutrient composition.
Collapse
Affiliation(s)
- C Sandoval-Caballero
- Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Avenida Libertador Bernardo O'Higgins 340, Santiago 8331150, Region Metropolitana, Chile
| | - L Luarte
- Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Avenida Libertador Bernardo O'Higgins 340, Santiago 8331150, Region Metropolitana, Chile
| | - Y Jiménez
- Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Avenida Libertador Bernardo O'Higgins 340, Santiago 8331150, Region Metropolitana, Chile
| | - C Jaque
- Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Avenida Libertador Bernardo O'Higgins 340, Santiago 8331150, Region Metropolitana, Chile
| | - F Cifuentes
- Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Avenida Libertador Bernardo O'Higgins 340, Santiago 8331150, Region Metropolitana, Chile
| | - G A Arenas
- Instituto de Ciencias de la Ingeniería, Universidad de O'Higgins, Libertador Bernardo O'Higgins #611, Rancagua 2841959, Region del Libertador Bernardo O'Higgins, Chile
| | - M Figueroa
- Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Avenida Libertador Bernardo O'Higgins 340, Santiago 8331150, Region Metropolitana, Chile
| | - J Jara
- Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Avenida Libertador Bernardo O'Higgins 340, Santiago 8331150, Region Metropolitana, Chile
| | - P K Olszewski
- Faculty of Science and Engineering, University of Waikato, Hamilton, Private Bag 3105, Hamilton 3240, New Zealand
| | - J A Teske
- School of Nutritional Sciences and Wellness and the Graduate Interdisciplinary Programs in Physiological Sciences and Neuroscience at the University of Arizona, 1177 E 4th Street Shantz 332, Tucson, AZ 85721, USA
| | - C E Pérez-Leighton
- Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Avenida Libertador Bernardo O'Higgins 340, Santiago 8331150, Region Metropolitana, Chile.
| |
Collapse
|
48
|
Peters C, He S, Fermani F, Lim H, Ding W, Mayer C, Klein R. Transcriptomics reveals amygdala neuron regulation by fasting and ghrelin thereby promoting feeding. SCIENCE ADVANCES 2023; 9:eadf6521. [PMID: 37224253 DOI: 10.1126/sciadv.adf6521] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/04/2022] [Accepted: 04/19/2023] [Indexed: 05/26/2023]
Abstract
The central amygdala (CeA) consists of numerous genetically defined inhibitory neurons that control defensive and appetitive behaviors including feeding. Transcriptomic signatures of cell types and their links to function remain poorly understood. Using single-nucleus RNA sequencing, we describe nine CeA cell clusters, of which four are mostly associated with appetitive and two with aversive behaviors. To analyze the activation mechanism of appetitive CeA neurons, we characterized serotonin receptor 2a (Htr2a)-expressing neurons (CeAHtr2a) that comprise three appetitive clusters and were previously shown to promote feeding. In vivo calcium imaging revealed that CeAHtr2a neurons are activated by fasting, the hormone ghrelin, and the presence of food. Moreover, these neurons are required for the orexigenic effects of ghrelin. Appetitive CeA neurons responsive to fasting and ghrelin project to the parabrachial nucleus (PBN) causing inhibition of target PBN neurons. These results illustrate how the transcriptomic diversification of CeA neurons relates to fasting and hormone-regulated feeding behavior.
Collapse
Affiliation(s)
- Christian Peters
- Department of Molecules-Signaling-Development, Max-Planck Institute for Biological Intelligence, 82152 Martinsried, Germany
| | - Songwei He
- Department of Molecules-Signaling-Development, Max-Planck Institute for Biological Intelligence, 82152 Martinsried, Germany
| | - Federica Fermani
- Department of Molecules-Signaling-Development, Max-Planck Institute for Biological Intelligence, 82152 Martinsried, Germany
| | - Hansol Lim
- Department of Molecules-Signaling-Development, Max-Planck Institute for Biological Intelligence, 82152 Martinsried, Germany
| | - Wenyu Ding
- Department of Molecules-Signaling-Development, Max-Planck Institute for Biological Intelligence, 82152 Martinsried, Germany
| | - Christian Mayer
- Laboratory of Neurogenomics, Max-Planck Institute for Biological Intelligence, 82152 Martinsried, Germany
| | - Rüdiger Klein
- Department of Molecules-Signaling-Development, Max-Planck Institute for Biological Intelligence, 82152 Martinsried, Germany
| |
Collapse
|
49
|
Jamaluddin A, Gorvin CM. RISING STARS: Targeting G protein-coupled receptors to regulate energy homeostasis. J Mol Endocrinol 2023; 70:e230014. [PMID: 36943057 PMCID: PMC10160555 DOI: 10.1530/jme-23-0014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/06/2023] [Accepted: 03/21/2023] [Indexed: 03/23/2023]
Abstract
G protein-coupled receptors (GPCRs) have a critical role in energy homeostasis, contributing to food intake, energy expenditure and glycaemic control. Dysregulation of energy expenditure can lead to metabolic syndrome (abdominal obesity, elevated plasma triglyceride, LDL cholesterol and glucose, and high blood pressure), which is associated with an increased risk of developing obesity, diabetes mellitus, non-alcoholic fatty liver disease and cardiovascular complications. As the prevalence of these chronic diseases continues to rise worldwide, there is an increased need to understand the molecular mechanisms by which energy expenditure is regulated to facilitate the development of effective therapeutic strategies to treat and prevent these conditions. In recent years, drugs targeting GPCRs have been the focus of efforts to improve treatments for type-2 diabetes and obesity, with GLP-1R agonists a particular success. In this review, we focus on nine GPCRs with roles in energy homeostasis that are current and emerging targets to treat obesity and diabetes. We discuss findings from pre-clinical models and clinical trials of drugs targeting these receptors and challenges that must be overcome before these drugs can be routinely used in clinics. We also describe new insights into how these receptors signal, including how accessory proteins, biased signalling, and complex spatial signalling could provide unique opportunities to develop more efficacious therapies with fewer side effects. Finally, we describe how combined therapies, in which multiple GPCRs are targeted, may improve clinical outcomes and reduce off-target effects.
Collapse
Affiliation(s)
- Aqfan Jamaluddin
- Institute of Metabolism and Systems Research (IMSR) and Centre for Diabetes, Endocrinology and Metabolism (CEDAM), University of Birmingham, Birmingham, UK
- Centre of Membrane Proteins and Receptors (COMPARE), Universities of Birmingham and Nottingham, Birmingham, UK
| | - Caroline M Gorvin
- Institute of Metabolism and Systems Research (IMSR) and Centre for Diabetes, Endocrinology and Metabolism (CEDAM), University of Birmingham, Birmingham, UK
- Centre of Membrane Proteins and Receptors (COMPARE), Universities of Birmingham and Nottingham, Birmingham, UK
| |
Collapse
|
50
|
Russo AF, Hay DL. CGRP physiology, pharmacology, and therapeutic targets: migraine and beyond. Physiol Rev 2023; 103:1565-1644. [PMID: 36454715 PMCID: PMC9988538 DOI: 10.1152/physrev.00059.2021] [Citation(s) in RCA: 137] [Impact Index Per Article: 68.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2021] [Revised: 11/23/2022] [Accepted: 11/27/2022] [Indexed: 12/03/2022] Open
Abstract
Calcitonin gene-related peptide (CGRP) is a neuropeptide with diverse physiological functions. Its two isoforms (α and β) are widely expressed throughout the body in sensory neurons as well as in other cell types, such as motor neurons and neuroendocrine cells. CGRP acts via at least two G protein-coupled receptors that form unusual complexes with receptor activity-modifying proteins. These are the CGRP receptor and the AMY1 receptor; in rodents, additional receptors come into play. Although CGRP is known to produce many effects, the precise molecular identity of the receptor(s) that mediates CGRP effects is seldom clear. Despite the many enigmas still in CGRP biology, therapeutics that target the CGRP axis to treat or prevent migraine are a bench-to-bedside success story. This review provides a contextual background on the regulation and sites of CGRP expression and CGRP receptor pharmacology. The physiological actions of CGRP in the nervous system are discussed, along with updates on CGRP actions in the cardiovascular, pulmonary, gastrointestinal, immune, hematopoietic, and reproductive systems and metabolic effects of CGRP in muscle and adipose tissues. We cover how CGRP in these systems is associated with disease states, most notably migraine. In this context, we discuss how CGRP actions in both the peripheral and central nervous systems provide a basis for therapeutic targeting of CGRP in migraine. Finally, we highlight potentially fertile ground for the development of additional therapeutics and combinatorial strategies that could be designed to modulate CGRP signaling for migraine and other diseases.
Collapse
Affiliation(s)
- Andrew F Russo
- Department of Molecular Physiology and Biophysics, University of Iowa, Iowa City, Iowa
- Department of Neurology, University of Iowa, Iowa City, Iowa
- Center for the Prevention and Treatment of Visual Loss, Department of Veterans Affairs Health Center, Iowa City, Iowa
| | - Debbie L Hay
- Department of Pharmacology and Toxicology, University of Otago, Dunedin, New Zealand
- Maurice Wilkins Centre for Molecular Biodiscovery, School of Biological Sciences, The University of Auckland, Auckland, New Zealand
| |
Collapse
|