1
|
Huang TX, Wang S, Ran C. Interoceptive processing in the nucleus of the solitary tract. Curr Opin Neurobiol 2025; 93:103021. [PMID: 40239364 DOI: 10.1016/j.conb.2025.103021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2024] [Revised: 03/17/2025] [Accepted: 03/21/2025] [Indexed: 04/18/2025]
Abstract
The interoceptive nervous system continuously monitors the status of visceral organs to synthesize internal perceptions and regulate behavioral and physiological responses. The nucleus of the solitary tract (NTS) in the brainstem serves as a central interoceptive hub and the initial site where sensory information from internal organs is processed in the brain. Here we review the neurobiological underpinnings of interoceptive processing in the NTS, focusing on recent progress enabled by modern genetic and optical tools for neural circuit dissection and neuronal recordings. Sensory information from internal organs is organized into a topographic map within the NTS, computed locally, modulated by descending inputs from higher brain regions, and distributed to downstream targets via projection neurons to control behavior and physiology. We present a sensory processing perspective on interoceptive coding within this brain structure.
Collapse
Affiliation(s)
- Tianxiao X Huang
- Department of Neuroscience, Dorris Neuroscience Center, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Shiqi Wang
- Department of Neuroscience, Dorris Neuroscience Center, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Chen Ran
- Department of Neuroscience, Dorris Neuroscience Center, The Scripps Research Institute, La Jolla, CA 92037, USA.
| |
Collapse
|
2
|
Klausen M, Knudsen G, Vilsbøll T, Fink‐Jensen A. Effects of GLP-1 Receptor Agonists in Alcohol Use Disorder. Basic Clin Pharmacol Toxicol 2025; 136:e70004. [PMID: 39891507 PMCID: PMC11786240 DOI: 10.1111/bcpt.70004] [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: 08/26/2024] [Revised: 12/12/2024] [Accepted: 01/20/2025] [Indexed: 02/03/2025]
Abstract
In the search for novel treatment strategies for alcohol use disorder (AUD), glucagon-like peptide-1 (GLP-1) receptor agonists (GLP-1RAs) approved for treating Type 2 diabetes and obesity have caught much attention. GLP-1 is a naturally occurring peptide produced in the small intestines and the brain, regulating plasma glucose levels and satiety. This focused review will report on the preclinical studies, case stories, register-based cohort studies, brain-imaging data and secondary analysis of clinical data supporting the role of GLP-1RAs as a novel treatment of AUD. Several clinical trials are ongoing, examining the potential effects of the GLP-1RA semaglutide in AUD.
Collapse
Affiliation(s)
- Mette Kruse Klausen
- Psychiatric Centre Copenhagen, Mental Health Services in the Capitol Region of DenmarkCopenhagen University Hospital FrederiksbergFrederiksbergDenmark
| | - Gitte Moos Knudsen
- Department of Clinical Medicine, Faculty of Health and Medical SciencesUniversity of CopenhagenCopenhagenDenmark
- Neurobiology Research UnitCopenhagen University Hospital RigshospitaletCopenhagenDenmark
| | - Tina Vilsbøll
- Department of Clinical Medicine, Faculty of Health and Medical SciencesUniversity of CopenhagenCopenhagenDenmark
- Clinical ResearchSteno Diabetes Center CopenhagenCopenhagenDenmark
| | - Anders Fink‐Jensen
- Psychiatric Centre Copenhagen, Mental Health Services in the Capitol Region of DenmarkCopenhagen University Hospital FrederiksbergFrederiksbergDenmark
- Department of Clinical Medicine, Faculty of Health and Medical SciencesUniversity of CopenhagenCopenhagenDenmark
| |
Collapse
|
3
|
Merkel R, Hernandez NS, Weir V, Zhang Y, Caffrey A, Rich MT, Crist RC, Reiner BC, Schmidt HD. An endogenous GLP-1 circuit engages VTA GABA neurons to regulate mesolimbic dopamine neurons and attenuate cocaine seeking. SCIENCE ADVANCES 2025; 11:eadr5051. [PMID: 40009667 PMCID: PMC11864183 DOI: 10.1126/sciadv.adr5051] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/03/2024] [Accepted: 01/28/2025] [Indexed: 02/28/2025]
Abstract
Recent studies show that systemic administration of a glucagon-like peptide-1 receptor (GLP-1R) agonist is sufficient to attenuate cocaine seeking. However, the neural mechanisms mediating these effects and the role of endogenous central GLP-1 signaling in cocaine seeking remain unknown. Here, we show that voluntary cocaine taking decreased plasma GLP-1 levels in rats and that chemogenetic activation of GLP-1-producing neurons in the nucleus tractus solitarius that project to the ventral tegmental area (VTA) decreased cocaine seeking. Single-nuclei transcriptomics and FISH studies revealed that GLP-1Rs are expressed primarily on GABA neurons in the VTA. Using in vivo fiber photometry, we found that the efficacy of a systemic GLP-1R agonist to attenuate cocaine seeking was associated with increased activity of VTA GABA neurons and decreased activity of VTA dopamine neurons. Together, these findings suggest that targeting central GLP-1 circuits may be an effective strategy toward reducing cocaine relapse and highlight a functional role of GABAergic GLP-1R-expressing midbrain neurons in drug seeking.
Collapse
Affiliation(s)
- Riley Merkel
- Department of Biobehavioral Health Sciences, School of Nursing, University of Pennsylvania, Philadelphia, PA 19104, USA
- Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Nicole S. Hernandez
- Department of Biobehavioral Health Sciences, School of Nursing, University of Pennsylvania, Philadelphia, PA 19104, USA
- Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Vanessa Weir
- Department of Biobehavioral Health Sciences, School of Nursing, University of Pennsylvania, Philadelphia, PA 19104, USA
- Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Vaegelos College of Physicians and Surgeons, Columbia University, New York, NY 10032, USA
| | - Yafang Zhang
- Department of Biobehavioral Health Sciences, School of Nursing, University of Pennsylvania, Philadelphia, PA 19104, USA
- Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Antonia Caffrey
- Department of Biobehavioral Health Sciences, School of Nursing, University of Pennsylvania, Philadelphia, PA 19104, USA
- Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Matthew T. Rich
- Department of Psychiatry, Brain Health Institute, Robert Wood Johnson Medical School, Rutgers University, Piscataway, NJ 08854, USA
| | - Richard C. Crist
- Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Benjamin C. Reiner
- Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Heath D. Schmidt
- Department of Biobehavioral Health Sciences, School of Nursing, University of Pennsylvania, Philadelphia, PA 19104, USA
- Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| |
Collapse
|
4
|
Jerlhag E. GLP-1 Receptor Agonists: Promising Therapeutic Targets for Alcohol Use Disorder. Endocrinology 2025; 166:bqaf028. [PMID: 39980336 PMCID: PMC11879929 DOI: 10.1210/endocr/bqaf028] [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: 08/19/2024] [Revised: 11/07/2024] [Accepted: 02/18/2025] [Indexed: 02/22/2025]
Abstract
Glucagon-like peptide-1 (GLP-1) is abundant in the circulation, and it is well-known to regulate glucose homeostasis, feeding, and body weight. GLP-1 receptor agonists are therefore approved for treating type 2 diabetes and obesity. However, more recent research has demonstrated that GLP-1 acts within the brain to modulate reward responses, thereby highlighting GLP-1 as a potential target for addiction. Specifically, preclinical studies demonstrated that GLP-1 receptor agonists decrease alcohol intake, reduce the motivation to consume alcohol, and prevent relapse drinking by potentially lowering alcohol-induced reward. These preclinical results have been confirmed and extended in human studies in which GLP-1 receptor agonists reduce alcohol intake in patients with alcohol use disorder (AUD) who have a regular weight or comorbidity of obesity or type 2 diabetes. On a similar note, genetic variations in genes encoding for the GLP-1 receptor are associated with AUD and heavy drinking. The central mechanisms by which GLP-1 regulates alcohol-related behaviors are not fully defined, but may involve areas central to reward as well as regions projecting to these reward areas, such as the nucleus tractus solitarius of the brainstem. Together, existing preclinical and clinical data suggest that GLP-1 is involved in the AUD process and implies its role as a tentative treatment for AUD.
Collapse
Affiliation(s)
- Elisabet Jerlhag
- Department of Pharmacology, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, SE-405 30 Gothenburg, Sweden
| |
Collapse
|
5
|
Duran M, Willis JR, Dalvi N, Fokakis Z, Virkus SA, Hardaway JA. Integration of Glucagon-Like Peptide 1 Receptor Actions Through the Central Amygdala. Endocrinology 2025; 166:bqaf019. [PMID: 39888375 PMCID: PMC11850305 DOI: 10.1210/endocr/bqaf019] [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: 10/16/2024] [Revised: 12/31/2024] [Accepted: 01/25/2025] [Indexed: 02/01/2025]
Abstract
Understanding the detailed mechanism of action of glucagon-like peptide 1 receptor (GLP-1R) agonists on distinct topographic and genetically defined brain circuits is critical for improving the efficacy and mitigating adverse side effects of these compounds. In this mini-review, we propose that the central nucleus of the amygdala (CeA) is a critical mediator of GLP-1R agonist-driven hypophagia. Here, we review the extant literature demonstrating CeA activation via GLP-1R agonists across multiple species and through multiple routes of administration. The precise role of GLP-1Rs within the CeA is unclear but the site-specific GLP-1Rs may mediate distinct behavioral and physiological hallmarks of GLP-1R agonists on food intake. Thus, we propose important novel directions and methods to test the role of the CeA in mediating GLP-1R actions.
Collapse
Affiliation(s)
- Miguel Duran
- Department of Psychiatry and Behavioral Neurobiology, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Jennifer R Willis
- Department of Psychiatry and Behavioral Neurobiology, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Nilay Dalvi
- Department of Psychiatry and Behavioral Neurobiology, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Zoe Fokakis
- Department of Psychiatry and Behavioral Neurobiology, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Sonja A Virkus
- Department of Psychiatry and Behavioral Neurobiology, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - J Andrew Hardaway
- Department of Psychiatry and Behavioral Neurobiology, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| |
Collapse
|
6
|
Wang H, Lou R, Wang Y, Hao L, Wang Q, Li R, Su J, Liu S, Zhou X, Gao X, Hao Q, Chen Z, Xu Y, Wu C, Zheng Y, Guo Q, Bai L. Parallel gut-to-brain pathways orchestrate feeding behaviors. Nat Neurosci 2025; 28:320-335. [PMID: 39627537 DOI: 10.1038/s41593-024-01828-8] [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: 04/23/2024] [Accepted: 10/29/2024] [Indexed: 02/08/2025]
Abstract
The caudal nucleus of the solitary tract (cNTS) in the brainstem serves as a hub for integrating interoceptive cues from diverse sensory pathways. However, the mechanisms by which cNTS neurons transform these signals into behaviors remain debated. We analyzed 18 cNTS-Cre mouse lines and cataloged the dynamics of nine cNTS cell types during feeding. We show that Th+ cNTS neurons encode esophageal mechanical distension and transient gulp size via vagal afferent inputs, providing quick feedback regulation of ingestion speed. By contrast, Gcg+ cNTS neurons monitor intestinal nutrients and cumulative ingested calories and have long-term effects on food satiation and preference. These nutritive signals are conveyed through a portal vein-spinal ascending pathway rather than vagal sensory neurons. Our findings underscore distinctions among cNTS subtypes marked by differences in temporal dynamics, sensory modalities, associated visceral organs and ascending sensory pathways, all of which contribute to specific functions in coordinated feeding regulation.
Collapse
Affiliation(s)
- Hongyun Wang
- Chinese Institute for Brain Research, Beijing, China
- Academy for Advanced Interdisciplinary Studies, Peking University, Beijing, China
| | - Runxiang Lou
- Chinese Institute for Brain Research, Beijing, China
| | - Yunfeng Wang
- Chinese Institute for Brain Research, Beijing, China
| | - Liufang Hao
- Chinese Institute for Brain Research, Beijing, China
| | - Qiushi Wang
- Chinese Institute for Brain Research, Beijing, China
| | - Rui Li
- Chinese Institute for Brain Research, Beijing, China
- State Key Laboratory of Cognitive Neuroscience and Leaning, Beijing Normal University, Beijing, China
| | - Jiayi Su
- Chinese Institute for Brain Research, Beijing, China
| | - Shuhan Liu
- Chinese Institute for Brain Research, Beijing, China
- State Key Laboratory of Cognitive Neuroscience and Leaning, Beijing Normal University, Beijing, China
| | - Xiangyu Zhou
- Chinese Institute for Brain Research, Beijing, China
| | - Xinwei Gao
- Chinese Institute for Brain Research, Beijing, China
| | - Qianxi Hao
- Chinese Institute for Brain Research, Beijing, China
| | - Zihe Chen
- Chinese Institute for Brain Research, Beijing, China
| | - Yibo Xu
- Chinese Institute for Brain Research, Beijing, China
| | - Chongwei Wu
- Chinese Institute for Brain Research, Beijing, China
| | - Yang Zheng
- Chinese Institute for Brain Research, Beijing, China
| | - Qingchun Guo
- Chinese Institute for Brain Research, Beijing, China
- School of Biomedical Engineering, Capital Medical University, Beijing, China
| | - Ling Bai
- Chinese Institute for Brain Research, Beijing, China.
| |
Collapse
|
7
|
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
|
8
|
Krieger JP, Daniels D, Lee S, Mastitskaya S, Langhans W. Glucagon-Like Peptide-1 Links Ingestion, Homeostasis, and the Heart. Compr Physiol 2025; 15:e7. [PMID: 39887844 PMCID: PMC11790259 DOI: 10.1002/cph4.7] [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: 11/02/2024] [Revised: 11/02/2024] [Accepted: 11/05/2024] [Indexed: 02/01/2025]
Abstract
Glucagon-like peptide-1 (GLP-1), a hormone released from enteroendocrine cells in the distal small and large intestines in response to nutrients and other stimuli, not only controls eating and insulin release, but is also involved in drinking control as well as renal and cardiovascular functions. Moreover, GLP-1 functions as a central nervous system peptide transmitter, produced by preproglucagon (PPG) neurons in the hindbrain. Intestinal GLP-1 inhibits eating by activating vagal sensory neurons directly, via GLP-1 receptors (GLP-1Rs), but presumably also indirectly, by triggering the release of serotonin from enterochromaffin cells. GLP-1 enhances glucose-dependent insulin release via a vago-vagal reflex and by direct action on beta cells. Finally, intestinal GLP-1 acts on the kidneys to modulate electrolyte and water movements, and on the heart, where it provides numerous benefits, including anti-inflammatory, antiatherogenic, and vasodilatory effects, as well as protection against ischemia/reperfusion injury and arrhythmias. Hindbrain PPG neurons receive multiple inputs and project to many GLP-1R-expressing brain areas involved in reward, autonomic functions, and stress. PPG neuron-derived GLP-1 is involved in the termination of large meals and is implicated in the inhibition of water intake. This review details GLP-1's roles in these interconnected systems, highlighting recent findings and unresolved issues, and integrating them to discuss the physiological and pathological relevance of endogenous GLP-1 in coordinating these functions. As eating poses significant threats to metabolic, fluid, and immune homeostasis, the body needs mechanisms to mitigate these challenges while sustaining essential nutrient intake. Endogenous GLP-1 plays a crucial role in this "ingestive homeostasis."
Collapse
Affiliation(s)
- Jean-Philippe Krieger
- Jean-Philippe Krieger, Institute of Veterinary Pharmacology and Toxicology, Vetsuisse Faculty, University of Zurich, Winterthurerstr. 260, 8057 Zurich
| | - Derek Daniels
- Department of Biological Sciences and the Center for Ingestive Behavior Research, University at Buffalo, the State University of New York, Buffalo NY 14260 USA
| | - Shin Lee
- Shin J. Lee, Neurimmune AG, Wagistrasse 18, 8952 Schlieren, Switzerland
| | - Svetlana Mastitskaya
- Department of Translational Health Sciences, Bristol Medical School, University of Bristol, Bristol, UK
| | - Wolfgang Langhans
- Physiology and Behavior Laboratory, Dept. of Health Sciences and Technology, ETH Zurich, 8603 Schwerzenbach, Switzerland
| |
Collapse
|
9
|
Moiz A, Filion KB, Tsoukas MA, Yu OH, Peters TM, Eisenberg MJ. Mechanisms of GLP-1 Receptor Agonist-Induced Weight Loss: A Review of Central and Peripheral Pathways in Appetite and Energy Regulation. Am J Med 2025:S0002-9343(25)00059-2. [PMID: 39892489 DOI: 10.1016/j.amjmed.2025.01.021] [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: 01/23/2025] [Accepted: 01/24/2025] [Indexed: 02/03/2025]
Abstract
Glucagon-like peptide-1 (GLP-1) receptor agonists (RAs) have become central in managing obesity and type 2 diabetes, primarily through appetite suppression and metabolic regulation. This review explores the mechanisms underlying GLP-1 RA-induced weight loss, focusing on central and peripheral pathways. Centrally, GLP-1 RAs modulate brain regions controlling appetite, influencing neurotransmitter and peptide release to regulate hunger and energy expenditure. Peripherally, GLP-1 RAs improve glycemic control by enhancing insulin secretion, reducing glucagon release, delaying gastric emptying, and regulating gut hormones. They also reduce triglycerides and low-density lipoprotein cholesterol, mitigate adipose tissue inflammation, and minimize ectopic fat deposition, promoting overall metabolic health. Emerging dual and triple co-agonists, targeting GLP-1 alongside glucose-dependent insulinotropic polypeptide, and glucagon pathways, may enhance weight loss and metabolic flexibility. Understanding these mechanisms is crucial as the therapeutic landscape evolves, offering clinicians and researchers insights to optimize the efficacy of current and future obesity treatments.
Collapse
Affiliation(s)
- Areesha Moiz
- Centre of Clinical Epidemiology, Lady Davis Institute, Jewish General Hospital, Montreal, Canada; Division of Experimental Medicine, McGill University, Montreal, Canada
| | - Kristian B Filion
- Centre of Clinical Epidemiology, Lady Davis Institute, Jewish General Hospital, Montreal, Canada; Department of Epidemiology, Biostatistics and Occupational Health, McGill University, Montreal, Canada; Department of Medicine, McGill University, Montreal, Canada
| | - Michael A Tsoukas
- Department of Medicine, McGill University, Montreal, Canada; Division of Endocrinology and Metabolism, McGill University, Montreal, Canada
| | - Oriana Hy Yu
- Centre of Clinical Epidemiology, Lady Davis Institute, Jewish General Hospital, Montreal, Canada; Department of Epidemiology, Biostatistics and Occupational Health, McGill University, Montreal, Canada; Department of Medicine, McGill University, Montreal, Canada; Division of Endocrinology and Metabolism, McGill University, Montreal, Canada
| | - Tricia M Peters
- Centre of Clinical Epidemiology, Lady Davis Institute, Jewish General Hospital, Montreal, Canada; Division of Experimental Medicine, McGill University, Montreal, Canada; Department of Epidemiology, Biostatistics and Occupational Health, McGill University, Montreal, Canada; Department of Medicine, McGill University, Montreal, Canada; Division of Endocrinology and Metabolism, McGill University, Montreal, Canada
| | - Mark J Eisenberg
- Centre of Clinical Epidemiology, Lady Davis Institute, Jewish General Hospital, Montreal, Canada; Division of Experimental Medicine, McGill University, Montreal, Canada; Department of Epidemiology, Biostatistics and Occupational Health, McGill University, Montreal, Canada; Department of Medicine, McGill University, Montreal, Canada; Division of Cardiology, Jewish General Hospital/McGill University, Canada.
| |
Collapse
|
10
|
Winzenried ET, Neyens DM, Calkins R, Appleyard SM. CCK-expressing neurons in the NTS are directly activated by CCK-sensitive C-type vagal afferents. Am J Physiol Regul Integr Comp Physiol 2025; 328:R121-R132. [PMID: 39509587 DOI: 10.1152/ajpregu.00280.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2023] [Revised: 08/27/2024] [Accepted: 10/10/2024] [Indexed: 11/15/2024]
Abstract
Vagal sensory afferents carrying information from the gastrointestinal tract (GI) terminate in the nucleus of the solitary tract (NTS). Different subpopulations of NTS neurons then relay this information throughout the brain. Cholecystokinin (CCK) is a satiety peptide that activates vagal afferents in the GI. However, CCK is also expressed by neurons in the NTS, and activation of these neurons decreases food intake. What is less clear is how these NTS CCK neurons are activated by vagal afferents and what type of information they integrate about meal size and content. To address this, we identified NTS-CCK neurons by crossing CCK-IRES-Cre mice with floxed-Rosa-tdtomato mice and made a horizontal brain slice containing vagal afferents in the solitary tract (ST). Voltage clamp recordings of NTS-CCK neurons show that activation of the ST evokes excitatory postsynaptic currents (EPSCs) mediated by both α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA) and N-methyl-d-aspartate (NMDA) receptors. Analysis of these EPSCs revealed that 80% of NTS-CCK neurons receive direct, monosynaptic inputs, with many also receiving indirect, or polysynaptic, inputs. NTS-CCK neurons are sensitive to the transient receptor potential vanilloid type 1 agonist capsaicin, suggesting that they are downstream of C-fibers. In addition, both CCK and a 5 hydroxytryptamine 3 receptor (5-HT3R) agonist increased spontaneous EPSC (sEPSC) frequency in NTS-CCK neurons, with 69% of NTS-CCK neurons sensitive to CCK and 42% to the 5-HT3 receptor agonist, as well as 45% sensitive to both and 10% to neither. Taken together with previous studies, this suggests that NTS-CCK neurons are driven primarily by vagal afferents that are sensitive to CCK and are only weakly driven by those sensitive to serotonin.NEW & NOTEWORTHY Nucleus of the solitary tract (NTS) cholecystokinin (CCK) expressing neurons are directly activated by glutamate released from vagal afferents. They are downstream of primarily C-type CCK-sensitive afferents, with a small proportion also downstream of serotonin-sensitive afferents. These findings suggest that NTS-CCK neurons integrate signals from the gut about ingestion of fats and proteins as well as stretch of the stomach, which they then relay to other brain regions important for the control of food intake.
Collapse
Affiliation(s)
- Eric T Winzenried
- Department of Integrative Physiology and Neuroscience, Washington State University, Pullman, Washington, United States
| | - Drew M Neyens
- Department of Integrative Physiology and Neuroscience, Washington State University, Pullman, Washington, United States
| | - Rowan Calkins
- Department of Integrative Physiology and Neuroscience, Washington State University, Pullman, Washington, United States
| | - Suzanne M Appleyard
- Department of Integrative Physiology and Neuroscience, Washington State University, Pullman, Washington, United States
| |
Collapse
|
11
|
Gasparini S, Almeida‐Pereira G, Munuzuri ASP, Resch JM, Geerling JC. Molecular Ontology of the Nucleus of Solitary Tract. J Comp Neurol 2024; 532:e70004. [PMID: 39629676 PMCID: PMC11615840 DOI: 10.1002/cne.70004] [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: 08/31/2024] [Revised: 11/03/2024] [Accepted: 11/15/2024] [Indexed: 12/08/2024]
Abstract
The nucleus of the solitary tract (NTS) receives visceral information and regulates appetitive, digestive, and cardiorespiratory systems. Within the NTS, diverse processes operate in parallel to sustain life, but our understanding of their cellular composition is incomplete. Here, we integrate histologic and transcriptomic analysis to identify and compare molecular features that distinguish neurons in this brain region. Most glutamatergic neurons in the NTS and area postrema co-express the transcription factors Lmx1b and Phox2b, except for a ventral band of neurons in the far-caudal NTS, which include the Gcg-expressing neurons that produce glucagon-like peptide 1 (GLP-1). GABAergic interneurons intermingle through the Lmx1b+Phox2b macropopulation, and dense clusters of GABAergic neurons surround the NTS. The Lmx1b+Phox2b macropopulation includes subpopulations with distinct distributions expressing Grp, Hsd11b2, Npff, Pdyn, Pou3f1, Sctr, Th, and other markers. These findings highlight Lmx1b-Phox2b co-expression as a common feature of glutamatergic neurons in the NTS and improve our understanding of the organization and distribution of neurons in this critical brain region.
Collapse
Affiliation(s)
| | | | | | - Jon M. Resch
- Department of Neuroscience and PharmacologyUniversity of IowaIowa CityIowaUSA
- Iowa Neuroscience InstituteUniversity of IowaIowa CityIowaUSA
| | - Joel C. Geerling
- Department of NeurologyUniversity of IowaIowa CityIowaUSA
- Iowa Neuroscience InstituteUniversity of IowaIowa CityIowaUSA
| |
Collapse
|
12
|
Aguggia J, Fernandez G, Cassano D, Mustafá ER, Rodríguez SS, Cantel S, Fehrentz JA, Raingo J, Schiöth HB, Habib AM, De Francesco PN, Perello M. Selective Colocalization of GHSR and GLP-1R in a Subset of Hypothalamic Neurons and Their Functional Interaction. Endocrinology 2024; 166:bqae160. [PMID: 39737802 DOI: 10.1210/endocr/bqae160] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/23/2024] [Indexed: 01/01/2025]
Abstract
The GH secretagogue receptor (GHSR) and the glucagon-like peptide-1 receptor (GLP-1R) are G protein-coupled receptors with critical, yet opposite, roles in regulating energy balance. Interestingly, these receptors are expressed in overlapping brain regions. However, the extent to which they target the same neurons and engage in molecular crosstalk remains unclear. To explore the potential colocalization of GHSR and GLP-1R in specific neurons, we performed detailed mapping of cells positive for both receptors using GHSR-eGFP reporter mice or wild-type mice infused with fluorescent ghrelin, alongside an anti-GLP-1R antibody. We found that GHSR+ and GLP-1R+ cells are largely segregated in the mouse brain. The highest overlap was observed in the hypothalamic arcuate nucleus, where 15% to 20% of GHSR+ cells were also GLP-1R+ cells. Additionally, we examined RNA-sequencing datasets from mouse and human brains to assess the fraction and distribution of neurons expressing both receptors, finding that double-positive Ghsr+/Glp1r+ cells are highly segregated, with a small subset of double-positive Ghsr+/Glp1r+ cells representing <10% of all Ghsr+ or Glp1r+ cells, primarily enriched in the hypothalamus. Furthermore, we conducted functional studies using patch-clamp recordings in a heterologous expression system to assess potential crosstalk in regulating presynaptic calcium channels. We provide the first evidence that liraglutide-evoked GLP-1R activity inhibits presynaptic channels, and that the presence of one GPCR attenuates the inhibitory effects of ligand-evoked activity mediated by the other on presynaptic calcium channels. In conclusion, while GHSR and GLP-1R can engage in molecular crosstalk, they are largely segregated across most neuronal types within the brain.
Collapse
Affiliation(s)
- Julieta Aguggia
- Laboratory of Neurophysiology, Multidisciplinary Institute of Cell Biology [IMBICE; Argentine Research Council (CONICET); Scientific Research Commission, Province of Buenos Aires (CIC-PBA); National University of La Plata], B1906APO La Plata, Buenos Aires, Argentina
| | - Gimena Fernandez
- Laboratory of Neurophysiology, Multidisciplinary Institute of Cell Biology [IMBICE; Argentine Research Council (CONICET); Scientific Research Commission, Province of Buenos Aires (CIC-PBA); National University of La Plata], B1906APO La Plata, Buenos Aires, Argentina
| | - Daniela Cassano
- Laboratory of Neurophysiology, Multidisciplinary Institute of Cell Biology [IMBICE; Argentine Research Council (CONICET); Scientific Research Commission, Province of Buenos Aires (CIC-PBA); National University of La Plata], B1906APO La Plata, Buenos Aires, Argentina
| | - Emilio R Mustafá
- Laboratory of Electrophysiology, Multidisciplinary Institute of Cell Biology [IMBICE; Argentine Research Council (CONICET); Scientific Research Commission, Province of Buenos Aires (CIC-PBA), National University of La Plata], B1906APO La Plata, Buenos Aires, Argentina
| | - Silvia S Rodríguez
- Laboratory of Electrophysiology, Multidisciplinary Institute of Cell Biology [IMBICE; Argentine Research Council (CONICET); Scientific Research Commission, Province of Buenos Aires (CIC-PBA), National University of La Plata], B1906APO La Plata, Buenos Aires, Argentina
| | - Sonia Cantel
- Institut des Biomolécules Max Mousseron, University of Montpellier, CNRS, ENSCM, 34293 Montpellier, France
| | - Jean-Alain Fehrentz
- Institut des Biomolécules Max Mousseron, University of Montpellier, CNRS, ENSCM, 34293 Montpellier, France
| | - Jesica Raingo
- Laboratory of Electrophysiology, Multidisciplinary Institute of Cell Biology [IMBICE; Argentine Research Council (CONICET); Scientific Research Commission, Province of Buenos Aires (CIC-PBA), National University of La Plata], B1906APO La Plata, Buenos Aires, Argentina
| | - Helgi B Schiöth
- Department of Surgical Sciences, Functional Pharmacology and Neuroscience, University of Uppsala, 751 24 Uppsala, Sweden
| | - Abdella M Habib
- College of Medicine, QU Health, Qatar University, P.O. Box 2713, Doha, Qatar
| | - Pablo N De Francesco
- Laboratory of Neurophysiology, Multidisciplinary Institute of Cell Biology [IMBICE; Argentine Research Council (CONICET); Scientific Research Commission, Province of Buenos Aires (CIC-PBA); National University of La Plata], B1906APO La Plata, Buenos Aires, Argentina
| | - Mario Perello
- Laboratory of Neurophysiology, Multidisciplinary Institute of Cell Biology [IMBICE; Argentine Research Council (CONICET); Scientific Research Commission, Province of Buenos Aires (CIC-PBA); National University of La Plata], B1906APO La Plata, Buenos Aires, Argentina
- Department of Surgical Sciences, Functional Pharmacology and Neuroscience, University of Uppsala, 751 24 Uppsala, Sweden
| |
Collapse
|
13
|
Martinelli S, Mazzotta A, Longaroni M, Petrucciani N. Potential role of glucagon-like peptide-1 (GLP-1) receptor agonists in substance use disorder: A systematic review of randomized trials. Drug Alcohol Depend 2024; 264:112424. [PMID: 39288591 DOI: 10.1016/j.drugalcdep.2024.112424] [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: 05/10/2024] [Revised: 08/17/2024] [Accepted: 08/18/2024] [Indexed: 09/19/2024]
Abstract
BACKGROUND Increasing evidence suggests that GLP-1 receptor agonists (GLP-1RA) have a potential use in addiction treatment. Few studies have assessed the impact of GLP-1RA on substance use disorder (SUD), particularly in humans. The study aimed to do systematic review of clinical trials to assess GLP-1RA's effect on reducing SUD in patients. METHODS The scientific literature was reviewed using the MEDLINE, Scopus and Cochrane Library databases, following PRISMA guidelines. Studies including patients with a diagnosis of SU who were treated with GLP-1RA were selected. The primary outcome was GLP-1RA's therapeutic effect on SUD, and the secondary outcomes were therapeutic effects of GLP-1RA on weight, BMI and HbA1c. RESULTS 1218 studies were retrieved, resulting in 507 papers after title and abstract screening. Following full-text review, only 5 articles met inclusion criteria. We incorporated a total of 630 participants utilizing Exenatide (n=3) and Dulaglutide (n=2) as GLP-1RAs. Therapeutic effect of GLP-1RA on SUD was assessed in 5 studies, with 3 demonstrating a significant decrease in SUD (alcohol and nicotine). GLP-1RA's impact on body weight, BMI, and HbA1c, was reported in 3 studies. These revealed a notable reduction in these parameters among the GLP-1RA treated group. CONCLUSION This review will give an overview of current new findings in human studies; we suggest that the effects of GLP-1RA in SUD is a possible new option of therapy in addiction medicine.
Collapse
Affiliation(s)
- Silvia Martinelli
- Department of Life Sciences and Public Health, Catholic University of the Sacred Heart, Rome, Italy; Department of Mental Health, Local Health Authority Viterbo, Viterbo, Italy
| | - Alessandro Mazzotta
- Department of Surgery, M.G. General Vannini Hospital, Istituto Figlie Di San Camillo, Rome, Italy
| | - Mattia Longaroni
- Department of Surgery, Santa Maria della Misericordia Hospital, University of Perugia, Italy
| | - Niccolò Petrucciani
- Department of Medical and Surgical Sciences and Translational Medicine, Division of General and Hepatobiliary Surgery, St. Andrea Hospital, Sapienza University of Rome, Italy.
| |
Collapse
|
14
|
Fenech C, Winters BL, Otsu Y, Aubrey KR. Supraspinal glycinergic neurotransmission in pain: A scoping review of current literature. J Neurochem 2024; 168:3663-3684. [PMID: 39075923 DOI: 10.1111/jnc.16191] [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: 02/07/2024] [Revised: 07/16/2024] [Accepted: 07/16/2024] [Indexed: 07/31/2024]
Abstract
The neurotransmitter glycine is an agonist at the strychnine-sensitive glycine receptors. In addition, it has recently been discovered to act at two new receptors, the excitatory glycine receptor and metabotropic glycine receptor. Glycine's neurotransmitter roles have been most extensively investigated in the spinal cord, where it is known to play essential roles in pain, itch, and motor function. In contrast, less is known about supraspinal glycinergic functions, and their contributions to pain circuits are largely unrecognized. As glycinergic neurons are absent from cortical regions, a clearer understanding of how supraspinal glycine modulates pain could reveal new pharmacological targets. This review aims to synthesize the published research on glycine's role in the adult brain, highlighting regions where glycine signaling may modulate pain responses. This was achieved through a scoping review methodology identifying several key regions of supraspinal pain circuitry where glycine signaling is involved. Therefore, this review unveils critical research gaps for supraspinal glycine's potential roles in pain and pain-associated responses, encouraging researchers to consider glycinergic neurotransmission more widely when investigating neural mechanisms of pain.
Collapse
Affiliation(s)
- Caitlin Fenech
- Pain Management Research Institute, Kolling Institute, Royal North Shore Hospital, St Leonards, New South Wales, Australia
- Faculty of Medicine and Health, University of Sydney, Sydney, New South Wales, Australia
| | - Bryony L Winters
- Pain Management Research Institute, Kolling Institute, Royal North Shore Hospital, St Leonards, New South Wales, Australia
- Discipline of Pharmacology, School of Pharmacy, Faculty of Medicine and Health, University of Sydney, Sydney, New South Wales, Australia
| | - Yo Otsu
- Pain Management Research Institute, Kolling Institute, Royal North Shore Hospital, St Leonards, New South Wales, Australia
- Faculty of Medicine and Health, University of Sydney, Sydney, New South Wales, Australia
| | - Karin R Aubrey
- Pain Management Research Institute, Kolling Institute, Royal North Shore Hospital, St Leonards, New South Wales, Australia
- Faculty of Medicine and Health, University of Sydney, Sydney, New South Wales, Australia
| |
Collapse
|
15
|
Naccarato MC, Oliveira LM, Ferreira CB, Moreira TS, Takakura AC. Nucleus of the solitary tract neuronal degeneration and impaired hypoxia response in a model of Parkinson's disease. Exp Neurol 2024; 380:114924. [PMID: 39147260 DOI: 10.1016/j.expneurol.2024.114924] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2024] [Revised: 07/30/2024] [Accepted: 08/12/2024] [Indexed: 08/17/2024]
Abstract
Parkinson's disease (PD) involves the degeneration of dopaminergic neurons in the substantia nigra (SNpc) and manifests with both classic and non-classic motor symptoms, including respiratory failure. Our study aims to investigate the involvement of the commissural and intermediate nucleus of the solitary tract (cNTS and iNTS) in the attenuated respiratory response to hypoxia in PD. Using a PD rat model induced by bilateral injection of 6-hydroxydopamine (6-OHDA) into the striatum of male Wistar rats, we explored potential alterations in the population of Phox2b neurons or hypoxia-activated neurons in the NTS projecting to the retrotrapezoid nucleus (RTN). Additionally, we explored neuronal connectivity between SNpc and cNTS. Projections pathways were assessed using unilateral injection of the retrograde tracer Fluorogold (FG) in the cNTS and RTN. Neuronal activation was evaluated by analyzing fos expression in rats exposed to hypoxia. In the PD model, the ventilatory response, measured through whole-body plethysmography, was impaired at both baseline and in response to hypoxia. A reduction in Phox2b-expressing neurons or hypoxia-activated neurons projecting to the RTN was observed. Additionally, we identified an indirect pathway linking the SNpc and cNTS, which passes through the periaqueductal gray (PAG). In conclusion, our findings suggest impairment in the SNpc-PAG-cNTS pathway in the PD model, explaining the loss of Phox2b-expressing neurons or hypoxia-activated neurons in the cNTS and subsequent respiratory impairment during hypoxic stimulation. We propose that the reduced population of Phox2b-expressing neurons in the NTS may include the same neurons activated by hypoxia and projecting to the RTN.
Collapse
Affiliation(s)
- Monique C Naccarato
- Department of Pharmacology, Instituto de Ciencias Biomedicas, Universidade de Sao Paulo, SP, 05508 Sao Paulo, SP, Brazil
| | - Luiz M Oliveira
- Department of Pharmacology, Instituto de Ciencias Biomedicas, Universidade de Sao Paulo, SP, 05508 Sao Paulo, SP, Brazil; Center for Integrative Brain Research, Seattle Children's Research Institute, 1900 9th Avenue, Seattle, WA 98101, USA
| | - Caroline B Ferreira
- Department of Pharmacology, Instituto de Ciencias Biomedicas, Universidade de Sao Paulo, SP, 05508 Sao Paulo, SP, Brazil; Department of Neurobiology, University of Pittsburgh School of Medicine, USA
| | - Thiago S Moreira
- Department of Physiology and Biophysics, Instituto de Ciencias Biomedicas, Universidade de Sao Paulo, SP, 05508 Sao Paulo, SP, Brazil
| | - Ana C Takakura
- Department of Pharmacology, Instituto de Ciencias Biomedicas, Universidade de Sao Paulo, SP, 05508 Sao Paulo, SP, Brazil.
| |
Collapse
|
16
|
Ricci A, Rubino E, Serra GP, Wallén-Mackenzie Å. Concerning neuromodulation as treatment of neurological and neuropsychiatric disorder: Insights gained from selective targeting of the subthalamic nucleus, para-subthalamic nucleus and zona incerta in rodents. Neuropharmacology 2024; 256:110003. [PMID: 38789078 DOI: 10.1016/j.neuropharm.2024.110003] [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: 02/06/2024] [Revised: 04/26/2024] [Accepted: 05/14/2024] [Indexed: 05/26/2024]
Abstract
Neuromodulation such as deep brain stimulation (DBS) is advancing as a clinical intervention in several neurological and neuropsychiatric disorders, including Parkinson's disease, dystonia, tremor, and obsessive-compulsive disorder (OCD) for which DBS is already applied to alleviate severely afflicted individuals of symptoms. Tourette syndrome and drug addiction are two additional disorders for which DBS is in trial or proposed as treatment. However, some major remaining obstacles prevent this intervention from reaching its full therapeutic potential. Side-effects have been reported, and not all DBS-treated individuals are relieved of their symptoms. One major target area for DBS electrodes is the subthalamic nucleus (STN) which plays important roles in motor, affective and associative functions, with impact on for example movement, motivation, impulsivity, compulsivity, as well as both reward and aversion. The multifunctionality of the STN is complex. Decoding the anatomical-functional organization of the STN could enhance strategic targeting in human patients. The STN is located in close proximity to zona incerta (ZI) and the para-subthalamic nucleus (pSTN). Together, the STN, pSTN and ZI form a highly heterogeneous and clinically important brain area. Rodent-based experimental studies, including opto- and chemogenetics as well as viral-genetic tract tracings, provide unique insight into complex neuronal circuitries and their impact on behavior with high spatial and temporal precision. This research field has advanced tremendously over the past few years. Here, we provide an inclusive review of current literature in the pre-clinical research fields centered around STN, pSTN and ZI in laboratory mice and rats; the three highly heterogeneous and enigmatic structures brought together in the context of relevance for treatment strategies. Specific emphasis is placed on methods of manipulation and behavioral impact.
Collapse
Affiliation(s)
- Alessia Ricci
- Uppsala University, Department of Organism Biology, 756 32 Uppsala, Sweden; Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD 20815, USA
| | - Eleonora Rubino
- Uppsala University, Department of Organism Biology, 756 32 Uppsala, Sweden; Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD 20815, USA
| | - Gian Pietro Serra
- Uppsala University, Department of Organism Biology, 756 32 Uppsala, Sweden; Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD 20815, USA
| | - Åsa Wallén-Mackenzie
- Uppsala University, Department of Organism Biology, 756 32 Uppsala, Sweden; Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD 20815, USA.
| |
Collapse
|
17
|
Cai H, Schnapp WI, Mann S, Miscevic M, Shcmit MB, Conteras M, Fang C. Neural circuits regulation of satiation. Appetite 2024; 200:107512. [PMID: 38801994 PMCID: PMC11227400 DOI: 10.1016/j.appet.2024.107512] [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: 02/05/2024] [Revised: 05/21/2024] [Accepted: 05/22/2024] [Indexed: 05/29/2024]
Abstract
Terminating a meal after achieving satiation is a critical step in maintaining a healthy energy balance. Despite the extensive collection of information over the last few decades regarding the neural mechanisms controlling overall eating, the mechanism underlying different temporal phases of eating behaviors, especially satiation, remains incompletely understood and is typically embedded in studies that measure the total amount of food intake. In this review, we summarize the neural circuits that detect and integrate satiation signals to suppress appetite, from interoceptive sensory inputs to the final motor outputs. Due to the well-established role of cholecystokinin (CCK) in regulating the satiation, we focus on the neural circuits that are involved in regulating the satiation effect caused by CCK. We also discuss several general principles of how these neural circuits control satiation, as well as the limitations of our current understanding of the circuits function. With the application of new techniques involving sophisticated cell-type-specific manipulation and mapping, as well as real-time recordings, it is now possible to gain a better understanding of the mechanisms specifically underlying satiation.
Collapse
Affiliation(s)
- Haijiang Cai
- Department of Neuroscience, University of Arizona, Tucson, AZ, 85721, USA; Bio 5 Institute and Department of Neurology, University of Arizona, Tucson, AZ, 85721, USA.
| | - Wesley I Schnapp
- Department of Neuroscience, University of Arizona, Tucson, AZ, 85721, USA; Graduate Interdisciplinary Program in Neuroscience, University of Arizona, Tucson, AZ, 85721, USA
| | - Shivani Mann
- Department of Neuroscience, University of Arizona, Tucson, AZ, 85721, USA
| | - Masa Miscevic
- Department of Neuroscience, University of Arizona, Tucson, AZ, 85721, USA; Graduate Interdisciplinary Program in Physiological Sciences, University of Arizona, Tucson, AZ, 85721, USA
| | - Matthew B Shcmit
- Department of Neuroscience, University of Arizona, Tucson, AZ, 85721, USA; Graduate Interdisciplinary Program in Neuroscience, University of Arizona, Tucson, AZ, 85721, USA
| | - Marco Conteras
- Department of Neuroscience, University of Arizona, Tucson, AZ, 85721, USA
| | - Caohui Fang
- Department of Neuroscience, University of Arizona, Tucson, AZ, 85721, USA
| |
Collapse
|
18
|
Jerlhag E. Ghrelin system and GLP-1 as potential treatment targets for alcohol use disorder. INTERNATIONAL REVIEW OF NEUROBIOLOGY 2024; 178:401-432. [PMID: 39523062 DOI: 10.1016/bs.irn.2024.07.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/16/2024]
Abstract
Peptides of the gut-brain axis have gained recent attention as potential treatment targets for addiction. While the number of gut-brain peptides is vast, ghrelin and glucagon-like peptide-1 (GLP-1) have been suggested as important players. Ghrelin is traditionally considered an orexigenic peptide, but recent studies found that it increases alcohol intake in rodents and craving for alcohol in humans. Additionally, suppression of the ghrelin receptor attenuates alcohol-related responses in animal models reflecting alcohol use disorder (AUD). For instance, a lower alcohol intake, suppressed motivation to consume alcohol, and attenuated reward from alcohol is observed after ghrelin receptor antagonism treatment. On a similar note, a partial ghrelin receptor agonist prevents hangover symptoms in humans. When it comes to the anorexigenic peptide GLP-1, agonists of its receptor are approved to treat diabetes type 2 and obesity. Extensive preclinical studies have revealed that these GLP-1 receptor agonists reduce alcohol intake, suppress the motivation to consume alcohol, and prevent relapse drink, with effects tentatively associated with a reduced alcohol-induced reward. These preclinical findings have to some extent been varied in humans, as GLP-1 receptor agonists decrease alcohol intake in overweight patients with AUD. Furthermore, genetic variations in either the genes encoding for pre-pro-ghrelin, GHSR, GLP-1, or its receptor, are associated with AUD and heavy alcohol drinking. While central mechanisms appear to modulate the ability of either ghrelin or GLP-1 to regulate alcohol-related responses the exact mechanisms have not been defined. Taken together these preclinical and clinical data imply that gut-brain peptides participate in the addiction process and should be considered as potential targets for AUD treatment.
Collapse
Affiliation(s)
- Elisabet Jerlhag
- Institute of Neuroscience and Physiology, Department of Pharmacology, The Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden.
| |
Collapse
|
19
|
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: 1] [Impact Index Per Article: 1.0] [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
|
20
|
Randolph AB, Zheng H, Rinaman L. Populations of Hindbrain Glucagon-Like Peptide 1 (GLP1) Neurons That Innervate the Hypothalamic PVH, Thalamic PVT, or Limbic Forebrain BST Have Axon Collaterals That Reach All Central Regions Innervated by GLP1 Neurons. J Neurosci 2024; 44:e2063232024. [PMID: 38811166 PMCID: PMC11293452 DOI: 10.1523/jneurosci.2063-23.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2023] [Revised: 05/20/2024] [Accepted: 05/21/2024] [Indexed: 05/31/2024] Open
Abstract
Neurons in the caudal nucleus of the solitary tract (cNTS) and intermediate reticular nucleus (IRt) that express the glucagon gene (Gcg) give rise to glucagon-like peptide 1 (GLP1)-immunopositive axons in the spinal cord and many subcortical brain regions. Central GLP1 receptor signaling contributes to motivated behavior and stress responses in rats and mice, in which hindbrain GLP1 neurons are activated to express c-Fos in a metabolic state-dependent manner. The present study examined whether GLP1 inputs to distinct brain regions arise from distinct subsets of Gcg-expressing neurons, and mapped the distribution of axon collaterals arising from projection-defined GLP1 neural populations. Using our Gcg-Cre knock-in rat model, Cre-dependent adeno-associated virus (AAV) tracing was conducted in adult male and female rats to compare axonal projections of IRt versus cNTS GLP1 neurons. Overlapping projections were observed in all brain regions that receive GLP1 input, with the caveat that cNTS injections produced Cre-dependent labeling of some IRt neurons, and vice versa. In additional experiments, specific diencephalic or limbic forebrain nuclei were microinjected with Cre-dependent retrograde AAVs (AAVrg) that expressed reporters to fully label the axon collaterals of transduced GLP1 neurons. AAVrg injected into each forebrain site labeled Gcg-expressing neurons in both the cNTS and IRt. The collective axon collaterals of labeled neurons entered the spinal cord and every brain region previously reported to contain GLP1-positive axons. These results indicate that the axons of GLP1 neural populations that innervate the thalamic paraventricular nucleus, paraventricular nucleus of the hypothalamus, and/or bed nucleus of the stria terminalis collectively innervate all central regions that receive GLP1 axonal input.
Collapse
Affiliation(s)
- Abigail B Randolph
- Department of Psychology and Program in Neuroscience, Florida State University, Tallahassee, Florida 32306
| | - Huiyuan Zheng
- Department of Psychology and Program in Neuroscience, Florida State University, Tallahassee, Florida 32306
| | - Linda Rinaman
- Department of Psychology and Program in Neuroscience, Florida State University, Tallahassee, Florida 32306
| |
Collapse
|
21
|
Lu Y, Wang L, Luo F, Savani R, Rossi MA, Pang ZP. Dorsolateral septum GLP-1R neurons regulate feeding via lateral hypothalamic projections. Mol Metab 2024; 85:101960. [PMID: 38763494 PMCID: PMC11153235 DOI: 10.1016/j.molmet.2024.101960] [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: 03/25/2024] [Revised: 05/08/2024] [Accepted: 05/15/2024] [Indexed: 05/21/2024] Open
Abstract
OBJECTIVE Although glucagon-like peptide 1 (GLP-1) is known to regulate feeding, the central mechanisms contributing to this function remain enigmatic. Here, we aim to test the role of neurons expressing GLP-1 receptors (GLP-1R) in the dorsolateral septum (dLS; dLSGLP-1R) that project to the lateral hypothalamic area (LHA) on food intake and determine the relationship with feeding regulation. METHODS Using chemogenetic manipulations, we assessed how activation or inhibition of dLSGLP-1R neurons affected food intake in Glp1r-ires-Cre mice. Then, we used channelrhodopsin-assisted circuit mapping, chemogenetics, and electrophysiological recordings to identify and assess the role of the pathway from dLSGLP-1R →LHA projections in regulating food intake. RESULTS Chemogenetic inhibition of dLSGLP-1R neurons increases food intake. LHA is a major downstream target of dLSGLP-1R neurons. The dLSGLP-1R→LHA projections are GABAergic, and chemogenetic inhibition of this pathway also promotes food intake. While chemogenetic activation of dLSGLP-1R→LHA projections modestly decreases food intake, optogenetic stimulation of the dLSGLP-1R→LHA projection terminals in the LHA rapidly suppresses feeding behavior. Finally, we demonstrate that the GLP-1R agonist, Exendin 4 enhances dLSGLP-1R →LHA GABA release. CONCLUSIONS Together, these results demonstrate that dLS-GLP-1R neurons and the inhibitory pathway to LHA can regulate feeding behavior, which might serve as a potential therapeutic target for the treatment of eating disorders or obesity.
Collapse
Affiliation(s)
- Yi Lu
- Child Health Institute of New Jersey, Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ 08901, USA
| | - Le Wang
- Child Health Institute of New Jersey, Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ 08901, USA
| | - Fang Luo
- Child Health Institute of New Jersey, Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ 08901, USA
| | - Rohan Savani
- Child Health Institute of New Jersey, Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ 08901, USA
| | - Mark A Rossi
- Child Health Institute of New Jersey, Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ 08901, USA; Department of Psychiatry, Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ 08901, USA; Brain Health Institute, Rutgers University, New Brunswick, NJ 08901, USA; Department of Neuroscience and Cell Biology, Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ 08901, USA.
| | - Zhiping P Pang
- Child Health Institute of New Jersey, Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ 08901, USA; Department of Neuroscience and Cell Biology, Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ 08901, USA.
| |
Collapse
|
22
|
Merkel R, Hernandez N, Weir V, Zhang Y, Rich MT, Crist RC, Reiner BC, Schmidt HD. An endogenous GLP-1 circuit engages VTA GABA neurons to regulate mesolimbic dopamine neurons and attenuate cocaine seeking. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.20.599574. [PMID: 38979354 PMCID: PMC11230186 DOI: 10.1101/2024.06.20.599574] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/10/2024]
Abstract
Recent studies show that systemic administration of a glucagon-like peptide-1 receptor (GLP-1R) agonist is sufficient to attenuate the reinstatement of cocaine-seeking behavior, an animal model of relapse. However, the neural mechanisms mediating these effects and the role of endogenous central GLP-1 signaling in cocaine seeking remain unknown. Here, we show that voluntary cocaine taking decreased plasma GLP-1 levels in rats and that chemogenetic activation of GLP-1-producing neurons in the nucleus tractus solitarius (NTS) that project to the ventral tegmental area (VTA) decreased cocaine reinstatement. Single nuclei transcriptomics and FISH studies revealed GLP-1Rs are expressed primarily on GABA neurons in the VTA. Using in vivo fiber photometry, we found that the efficacy of a systemic GLP-1R agonist to attenuate cocaine seeking was associated with increased activity of VTA GABA neurons and decreased activity of VTA dopamine neurons. Together, these findings suggest that targeting central GLP-1 circuits may be an effective strategy toward reducing cocaine relapse and highlight a novel functional role of GABAergic GLP-1R-expressing midbrain neurons in drug seeking.
Collapse
|
23
|
Shao L, Kong F, Tian X, Deng T, Wang Y, Ji Y, Wang X, Yu H, Yuan F, Fu C, Wang S. Whole-brain inputs and outputs of Phox2b and GABAergic neurons in the nucleus tractus solitarii. Front Neurosci 2024; 18:1427384. [PMID: 38948926 PMCID: PMC11211284 DOI: 10.3389/fnins.2024.1427384] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2024] [Accepted: 06/04/2024] [Indexed: 07/02/2024] Open
Abstract
The nucleus tractus solitarii (NTS) plays a critical role in the homeostatic regulation of respiration, blood pressure, sodium consumption and metabolic processes. Despite their significance, the circuitry mechanisms facilitating these diverse physiological functions remain incompletely understood. In this study, we present a whole-brain mapping of both the afferent and efferent connections of Phox2b-expressing and GABAergic neurons within the NTS. Our findings reveal that these neuronal populations not only receive monosynaptic inputs primarily from the medulla oblongata, pons, midbrain, supra-midbrain and cortical areas, but also mutually project their axons to these same locales. Moreover, intense monosynaptic inputs are received from the central amygdala, the paraventricular nucleus of the hypothalamus, the parasubthalamic nucleus and the intermediate reticular nucleus, along with brainstem nuclei explicitly engaged in respiratory regulation. In contrast, both neuronal groups extensively innervate brainstem nuclei associated with respiratory functions, although their projections to regions above the midbrain are comparatively limited. These anatomical findings provide a foundational platform for delineating an anatomical framework essential for dissecting the specific functional mechanisms of these circuits.
Collapse
Affiliation(s)
- Liuqi Shao
- Department of Neurobiology, Hebei Medical University, Shijiazhuang, Hebei, China
| | - Fanrao Kong
- Department of Neurobiology, Hebei Medical University, Shijiazhuang, Hebei, China
| | - Xiaochen Tian
- Department of Neurobiology, Hebei Medical University, Shijiazhuang, Hebei, China
| | - Tianjiao Deng
- Department of Neurobiology, Hebei Medical University, Shijiazhuang, Hebei, China
| | - Yakun Wang
- Department of Sleep Medicine, Third Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Yake Ji
- Department of Neurobiology, Hebei Medical University, Shijiazhuang, Hebei, China
| | - Xiaoyi Wang
- Department of Neurobiology, Hebei Medical University, Shijiazhuang, Hebei, China
| | - Hongxiao Yu
- Department of Neurobiology, Hebei Medical University, Shijiazhuang, Hebei, China
| | - Fang Yuan
- Department of Neurobiology, Hebei Medical University, Shijiazhuang, Hebei, China
| | - Congrui Fu
- Nursing School, Hebei Medical University, Shijiazhuang, Hebei, China
| | - Sheng Wang
- Department of Neurobiology, Hebei Medical University, Shijiazhuang, Hebei, China
- Hebei Key Laboratory of Neurophysiology, Shijiazhuang, Hebei, China
| |
Collapse
|
24
|
Dereli AS, Oh AYS, McMullan S, Kumar NN. Galaninergic and hypercapnia-activated neuronal projections to the ventral respiratory column. Brain Struct Funct 2024; 229:1121-1142. [PMID: 38578351 PMCID: PMC11147908 DOI: 10.1007/s00429-024-02782-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2023] [Accepted: 02/26/2024] [Indexed: 04/06/2024]
Abstract
In mammals, the ventral respiratory column (VRC) plays a pivotal role in integrating neurochemically diverse inputs from brainstem and forebrain regions to generate respiratory motor patterns. VRC microinjection of the neuropeptide galanin has been reported to dampen carbon dioxide (CO2)-mediated chemoreflex responses. Additionally, we previously demonstrated that galaninergic neurons in the retrotrapezoid nucleus (RTN) are implicated in the adaptive response to hypercapnic stimuli, suggesting a link between RTN neuroplasticity and increased neuronal drive to the VRC. VRC neurons express galanin receptor 1, suggesting potential regulatory action by galanin, however, the precise galaninergic chemoreceptor-VRC circuitry remains to be determined. This study aimed to identify sources of galaninergic input to the VRC that contribute to central respiratory chemoreception. We employed a combination of retrograde neuronal tracing, in situ hybridisation and immunohistochemistry to investigate VRC-projecting neurons that synthesise galanin mRNA. In an additional series of experiments, we used acute hypercapnia exposure (10% CO2, 1 h) and c-Fos immunohistochemistry to ascertain which galaninergic nuclei projecting to the VRC are activated. Our findings reveal that a total of 30 brain nuclei and 51 subnuclei project to the VRC, with 12 of these containing galaninergic neurons, including the RTN. Among these galaninergic populations, only a subset of the RTN neurons (approximately 55%) exhibited activation in response to acute hypercapnia. Our findings highlight that the RTN is the likely source of galaninergic transmission to the VRC in response to hypercapnic stimuli.
Collapse
Affiliation(s)
- Ayse S Dereli
- Department of Pharmacology, School of Biomedical Sciences, University of New South Wales, Sydney, Australia
| | - Alice Y S Oh
- Department of Pharmacology, School of Biomedical Sciences, University of New South Wales, Sydney, Australia
| | - Simon McMullan
- Macquarie Medical School, Macquarie University, Sydney, Australia
| | - Natasha N Kumar
- Department of Pharmacology, School of Biomedical Sciences, University of New South Wales, Sydney, Australia.
| |
Collapse
|
25
|
Łupina M, Wąsik A, Baranowska-Bosiacka I, Tarnowski M, Słowik T, Listos P, Kotlińska J, Kosik-Bogacka D, Gutowska I, Listos J. Acute and Chronic Exposure to Linagliptin, a Selective Inhibitor of Dipeptidyl Peptidase-4 (DPP-4), Has an Effect on Dopamine, Serotonin and Noradrenaline Level in the Striatum and Hippocampus of Rats. Int J Mol Sci 2024; 25:3008. [PMID: 38474255 PMCID: PMC10932243 DOI: 10.3390/ijms25053008] [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: 01/22/2024] [Revised: 02/29/2024] [Accepted: 03/01/2024] [Indexed: 03/14/2024] Open
Abstract
Linagliptin is a selective dipeptidyl peptidase-4 (DPP-4) inhibitor that indirectly elevates the glucagon-like peptide-1 (GLP-1) level. The aim of the present study was to check whether linagliptin has an influence on neurotransmission in rat brain. Rats were acutely and chronically exposed to linagliptin (10 and 20 mg/kg, intraperitoneally (i.p.)). Twenty-four hours later, the striatum and hippocampus were selected for further studies. In neurochemical experiments, using high-performance liquid chromatography with electrochemical detection (HPLC-ED), the concentrations of three major neurotransmitters-dopamine, serotonin and noradrenaline-and their metabolites were measured. The analysis of mRNA expression of dopamine (D1 and D2), serotonin (5-HT-1 and 5-HT-2) and noradrenaline (α1 and α2a) receptors was also investigated using real-time quantitative reverse transcription polymerase chain reaction (RQ-PCR) in the same brain areas. Linagliptin has the ability to influence the dopaminergic system. In the striatum, the elevation of dopamine and its metabolites was observed after repeated administration of that linagliptin, and in the hippocampus, a reduction in dopamine metabolism was demonstrated. Acute linagliptin exposure increases the serotonin level in both areas, while after chronic linagliptin administration a tendency for the mRNA expression of serotoninergic receptors (5-HT1A and 5-HT2A) to increase was observed. A single instance of exposure to linagliptin significantly modified the noradrenaline level in the striatum and intensified noradrenaline turnover in the hippocampus. The recognition of the interactions in the brain between DPP-4 inhibitors and neurotransmitters and/or receptors is a crucial step for finding novel discoveries in the pharmacology of DPP-4 inhibitors and raises hope for further applications of DPP-4 inhibitors in clinical practices.
Collapse
Affiliation(s)
- Małgorzata Łupina
- Department of Experimental and Clinical Pharmacology, Medical University of Lublin, Jaczewskiego 8b St., 20-090 Lublin, Poland
| | - Agnieszka Wąsik
- Department of Neurochemistry, Maj Institute of Pharmacology PAS, Smetna St. 12, 31-343 Kraków, Poland;
| | - Irena Baranowska-Bosiacka
- Department of Biochemistry and Medical Chemistry, Pomeranian Medical University, Powstańców Wlkp. 72 Av., 70-111 Szczecin, Poland;
| | - Maciej Tarnowski
- Department of Physiology in Health Sciences, Pomeranian Medical University, Żołnierska 54, 70-210 Szczecin, Poland;
| | - Tymoteusz Słowik
- Experimental Medicine Center, Medical University of Lublin, Jaczewskiego 8d St., 20-090 Lublin, Poland;
| | - Piotr Listos
- Department of Pathological Anatomy, Faculty of Veterinary Medicine, University of Life Sciences, Głęboka 30, 20-612 Lublin, Poland;
| | - Jolanta Kotlińska
- Department of Pharmacology and Pharmacodynamics, Medical University of Lublin, Chodźki 4a St., 20-093 Lublin, Poland; (J.K.); (J.L.)
| | - Danuta Kosik-Bogacka
- Independent Laboratory of Pharmaceutical Botany, Department of Biology and Medical Parasitology, Pomeranian Medical University in Szczecin, Powstańców Wlkp. 72, 70-111 Szczecin, Poland;
| | - Izabela Gutowska
- Department of Biochemistry and Human Nutrition, Pomeranian Medical University, Broniewskiego 24, 71-460 Szczecin, Poland;
| | - Joanna Listos
- Department of Pharmacology and Pharmacodynamics, Medical University of Lublin, Chodźki 4a St., 20-093 Lublin, Poland; (J.K.); (J.L.)
| |
Collapse
|
26
|
Iovino M, Messana T, Marucci S, Triggiani D, Giagulli VA, Guastamacchia E, Piazzolla G, De Pergola G, Lisco G, Triggiani V. The neurohypophyseal hormone oxytocin and eating behaviors: a narrative review. Hormones (Athens) 2024; 23:15-23. [PMID: 37979096 PMCID: PMC10847364 DOI: 10.1007/s42000-023-00505-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/17/2023] [Accepted: 11/03/2023] [Indexed: 11/19/2023]
Abstract
BACKGROUND The neuropeptide oxytocin (OT) is crucial in several conditions, such as lactation, parturition, mother-infant interaction, and psychosocial function. Moreover, OT may be involved in the regulation of eating behaviors. METHODS This review briefly summarizes data concerning the role of OT in eating behaviors. Appropriate keywords and medical subject headings were identified and searched for in PubMed/MEDLINE. References of original articles and reviews were screened, examined, and selected. RESULTS Hypothalamic OT-secreting neurons project to different cerebral areas controlling eating behaviors, such as the amygdala, area postrema, nucleus of the solitary tract, and dorsal motor nucleus of the vagus nerve. Intracerebral/ventricular OT administration decreases food intake and body weight in wild and genetically obese rats. OT may alter food intake and the quality of meals, especially carbohydrates and sweets, in humans. DISCUSSION OT may play a role in the pathophysiology of eating disorders with potential therapeutic perspectives. In obese patients and those with certain eating disorders, such as bulimia nervosa or binge/compulsive eating, OT may reduce appetite and caloric consumption. Conversely, OT administered to patients with anorexia nervosa may paradoxically stimulate appetite, possibly by lowering anxiety which usually complicates the management of these patients. Nevertheless, OT administration (e.g., intranasal route) is not always associated with clinical benefit, probably because intranasally administered OT fails to achieve therapeutic intracerebral levels of the hormone. CONCLUSION OT administration could play a therapeutic role in managing eating disorders and disordered eating. However, specific studies are needed to clarify this issue with regard to dose-finding and route and administration time.
Collapse
Affiliation(s)
- Michele Iovino
- Interdisciplinary Department of Medicine, Section of Internal Medicine, Geriatrics, Endocrinology and Rare Diseases, University of Bari "Aldo Moro", School of Medicine, Bari, Apulia, Italy
| | - Tullio Messana
- Infantile Neuropsychiatry, IRCCS - Institute of Neurological Sciences, Bologna, Italy
| | - Simonetta Marucci
- Università Campus Biomedico, Dip. "Scienze e Tecnologie per l'Uomo e l'ambiente", Via Alvaro del Portillo, 21, Roma, Italy
| | - Domenico Triggiani
- Interdisciplinary Department of Medicine, Section of Internal Medicine, Geriatrics, Endocrinology and Rare Diseases, University of Bari "Aldo Moro", School of Medicine, Bari, Apulia, Italy
| | - Vito Angelo Giagulli
- Interdisciplinary Department of Medicine, Section of Internal Medicine, Geriatrics, Endocrinology and Rare Diseases, University of Bari "Aldo Moro", School of Medicine, Bari, Apulia, Italy
| | - Edoardo Guastamacchia
- Interdisciplinary Department of Medicine, Section of Internal Medicine, Geriatrics, Endocrinology and Rare Diseases, University of Bari "Aldo Moro", School of Medicine, Bari, Apulia, Italy
| | - Giuseppina Piazzolla
- Interdisciplinary Department of Medicine, Section of Internal Medicine, Geriatrics, Endocrinology and Rare Diseases, University of Bari "Aldo Moro", School of Medicine, Bari, Apulia, Italy
| | - Giovanni De Pergola
- National Institute of Gastroenterology IRCCS "Saverio de Bellis", Research Hospital, Castellana Grotte, Bari, Italy
- Department of Biomedical Science and Human Oncology, University of Bari, School of Medicine, Bari, Apulia, Italy
| | - Giuseppe Lisco
- Interdisciplinary Department of Medicine, Section of Internal Medicine, Geriatrics, Endocrinology and Rare Diseases, University of Bari "Aldo Moro", School of Medicine, Bari, Apulia, Italy.
| | - Vincenzo Triggiani
- Interdisciplinary Department of Medicine, Section of Internal Medicine, Geriatrics, Endocrinology and Rare Diseases, University of Bari "Aldo Moro", School of Medicine, Bari, Apulia, Italy
| |
Collapse
|
27
|
Lataro RM, Brognara F, Iturriaga R, Paton JFR. Inflammation of some visceral sensory systems and autonomic dysfunction in cardiovascular disease. Auton Neurosci 2024; 251:103137. [PMID: 38104365 DOI: 10.1016/j.autneu.2023.103137] [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: 07/27/2023] [Revised: 11/15/2023] [Accepted: 12/04/2023] [Indexed: 12/19/2023]
Abstract
The sensitization and hypertonicity of visceral afferents are highly relevant to the development and progression of cardiovascular and respiratory disease states. In this review, we described the evidence that the inflammatory process regulates visceral afferent sensitivity and tonicity, affecting the control of the cardiovascular and respiratory system. Some inflammatory mediators like nitric oxide, angiotensin II, endothelin-1, and arginine vasopressin may inhibit baroreceptor afferents and contribute to the baroreflex impairment observed in cardiovascular diseases. Cytokines may act directly on peripheral afferent terminals that transmit information to the central nervous system (CNS). TLR-4 receptors, which recognize lipopolysaccharide, were identified in the nodose and petrosal ganglion and have been implicated in disrupting the blood-brain barrier, which can potentiate the inflammatory process. For example, cytokines may cross the blood-brain barrier to access the CNS. Additionally, pro-inflammatory cytokines such as IL-1β, IL-6, TNF-α and some of their receptors have been identified in the nodose ganglion and carotid body. These pro-inflammatory cytokines also sensitize the dorsal root ganglion or are released in the nucleus of the solitary tract. In cardiovascular disease, pro-inflammatory mediators increase in the brain, heart, vessels, and plasma and may act locally or systemically to activate/sensitize afferent nervous terminals. Recent evidence demonstrated that the carotid body chemoreceptor cells might sense systemic pro-inflammatory molecules, supporting the novel proposal that the carotid body is part of the afferent pathway in the central anti-inflammatory reflexes. The exact mechanisms of how pro-inflammatory mediators affects visceral afferent signals and contribute to the pathophysiology of cardiovascular diseases awaits future research.
Collapse
Affiliation(s)
- R M Lataro
- Department of Physiological Sciences, Center of Biological Sciences, Federal University of Santa Catarina, Florianópolis, Santa Catarina, Brazil.
| | - F Brognara
- Department of Nursing, General and Specialized, Nursing School of Ribeirão Preto, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
| | - R Iturriaga
- Facultad de Ciencias Biológicas, Pontificia Universidad Catolica de Chile, Santiago, Chile; Centro de Investigación en Fisiología y Medicina en Altura - FIMEDALT, Universidad de Antofagasta, Antofagasta, Chile
| | - J F R Paton
- Manaaki Manawa - The Centre for Heart Research, Department of Physiology, Faculty of Medical & Health Sciences, University of Auckland, Grafton, Auckland, New Zealand
| |
Collapse
|
28
|
Aguilar LA, Coker CR, McCullers Z, Evans A, Showemimo O, Melkumyan M, Keller BN, Snyder AE, Bingaman SS, Randall PA, Hajnal A, Browning KN, Arnold AC, Silberman Y. Adolescent alcohol disrupts development of noradrenergic neurons in the nucleus of the tractus solitarius and enhances stress behaviors in adulthood in mice in a sex specific manner. ADDICTION NEUROSCIENCE 2023; 9:100132. [PMID: 38162404 PMCID: PMC10756564 DOI: 10.1016/j.addicn.2023.100132] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/03/2024]
Abstract
Alcohol use disorders (AUDs) are common mental health issues worldwide and can lead to other chronic diseases. Stress is a major factor in the development and continuation of AUDs, and adolescent alcohol exposure can lead to enhanced stress-responsivity and increased risk for AUD development in adulthood. The exact mechanisms behind the interaction between adolescence, stress, and alcohol are not fully understood and require further research. In this regard, the nucleus of the tractus solitarius (NTS) provides dense norepinephrine projections to the extended amygdala, providing a key pathway for stress-related alcohol behaviors. While NTS norepinephrine neurons are known to be alcohol sensitive, whether adolescent alcohol disrupts NTS-norepinephrine neuron development and if this is related to altered stress-sensitivity and alcohol preference in adulthood has not previously been examined. Here, we exposed male and female C57Bl/6J mice to the commonly used adolescent intermittent ethanol (AIE) vapor model during postnatal day 28-42 and examined AIE effects on: 1) tyrosine hydroxylase (TH) mRNA expression in the NTS across various ages (postnatal day 21-90), 2) behavioral responses to acute stress in the light/dark box test in adulthood, 3) NTS TH neuron responses to acute stress and ethanol challenges in adulthood, and 4) ethanol conditioned place preference behavior in adulthood. Overall the findings indicate that AIE alters NTS TH mRNA expression and increases anxiety-like behaviors following acute stress exposure in a sex-dependent manner. These mRNA expression and behavioral changes occur in the absence of AIE-induced changes in NTS TH neuron sensitivity to either acute stress or acute alcohol exposure or changes to ethanol conditioned place preference.
Collapse
Affiliation(s)
- Liz A. Aguilar
- Department of Neural and Behavioral Sciences, Penn State College of Medicine, USA
- Currently at Department of Biology, Indiana University Bloomington, USA
| | - Caitlin R. Coker
- Department of Neural and Behavioral Sciences, Penn State College of Medicine, USA
- Penn State College of Medicine, Graduate Program in Anatomy, USA
- Currently at Georgetown University School of Medicine, USA
| | - Zari McCullers
- Department of Neural and Behavioral Sciences, Penn State College of Medicine, USA
- Penn State College of Medicine, Graduate Program in Biomedical Sciences, USA
| | - Alexandra Evans
- Department of Neural and Behavioral Sciences, Penn State College of Medicine, USA
- Penn State College of Medicine, Graduate Program in Biomedical Sciences, USA
| | - Opeyemi Showemimo
- Department of Neural and Behavioral Sciences, Penn State College of Medicine, USA
- Penn State College of Medicine, Graduate Program in Anatomy, USA
| | - Mariam Melkumyan
- Department of Neural and Behavioral Sciences, Penn State College of Medicine, USA
- Penn State College of Medicine, Graduate Program in Neuroscience, USA
| | - Bailey N. Keller
- Department of Neural and Behavioral Sciences, Penn State College of Medicine, USA
- Penn State College of Medicine, Graduate Program in Neuroscience, USA
| | - Angela E. Snyder
- Department of Neural and Behavioral Sciences, Penn State College of Medicine, USA
- Penn State College of Medicine, Graduate Program in Neuroscience, USA
| | - Sarah S. Bingaman
- Department of Neural and Behavioral Sciences, Penn State College of Medicine, USA
| | | | - Andras Hajnal
- Department of Neural and Behavioral Sciences, Penn State College of Medicine, USA
| | - Kirsteen N. Browning
- Department of Neural and Behavioral Sciences, Penn State College of Medicine, USA
| | - Amy C. Arnold
- Department of Neural and Behavioral Sciences, Penn State College of Medicine, USA
| | - Yuval Silberman
- Department of Neural and Behavioral Sciences, Penn State College of Medicine, USA
| |
Collapse
|
29
|
Sayar-Atasoy N, Laule C, Aklan I, Kim H, Yavuz Y, Ates T, Coban I, Koksalar-Alkan F, Rysted J, Davis D, Singh U, Alp MI, Yilmaz B, Cui H, Atasoy D. Adrenergic modulation of melanocortin pathway by hunger signals. Nat Commun 2023; 14:6602. [PMID: 37857606 PMCID: PMC10587058 DOI: 10.1038/s41467-023-42362-8] [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: 01/10/2023] [Accepted: 10/09/2023] [Indexed: 10/21/2023] Open
Abstract
Norepinephrine (NE) is a well-known appetite regulator, and the nor/adrenergic system is targeted by several anti-obesity drugs. To better understand the circuitry underlying adrenergic appetite control, here we investigated the paraventricular hypothalamic nucleus (PVN), a key brain region that integrates energy signals and receives dense nor/adrenergic input, using a mouse model. We found that PVN NE level increases with signals of energy deficit and decreases with food access. This pattern is recapitulated by the innervating catecholaminergic axon terminals originating from NTSTH-neurons. Optogenetic activation of rostral-NTSTH → PVN projection elicited strong motivation to eat comparable to overnight fasting whereas its inhibition attenuated both fasting-induced & hypoglycemic feeding. We found that NTSTH-axons functionally targeted PVNMC4R-neurons by predominantly inhibiting them, in part, through α1-AR mediated potentiation of GABA release from ARCAgRP presynaptic terminals. Furthermore, glucoprivation suppressed PVNMC4R activity, which was required for hypoglycemic feeding response. These results define an ascending nor/adrenergic circuit, NTSTH → PVNMC4R, that conveys peripheral hunger signals to melanocortin pathway.
Collapse
Affiliation(s)
- Nilufer Sayar-Atasoy
- Department of Pharmacology, Iowa Neuroscience Institute, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, IA, USA
| | - Connor Laule
- Department of Pharmacology, Iowa Neuroscience Institute, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, IA, USA
| | - Iltan Aklan
- Department of Pharmacology, Iowa Neuroscience Institute, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, IA, USA
| | - Hyojin Kim
- Department of Pharmacology, Iowa Neuroscience Institute, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, IA, USA
| | - Yavuz Yavuz
- Department of Physiology, School of Medicine, Yeditepe University, Istanbul, Turkey
| | - Tayfun Ates
- Department of Pharmacology, Iowa Neuroscience Institute, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, IA, USA
| | - Ilknur Coban
- Institute for Anatomy and Cell Biology, Heidelberg University, Heidelberg, Germany
| | | | - Jacob Rysted
- Department of Pharmacology, Iowa Neuroscience Institute, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, IA, USA
| | - Debbie Davis
- Department of Pharmacology, Iowa Neuroscience Institute, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, IA, USA
| | - Uday Singh
- Department of Pharmacology, Iowa Neuroscience Institute, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, IA, USA
| | - Muhammed Ikbal Alp
- Department of Physiology, School of Medicine, Research Institute for Health Sciences and Technologies (SABITA), Istanbul Medipol University, Istanbul, Turkey
| | - Bayram Yilmaz
- Department of Physiology, School of Medicine, Yeditepe University, Istanbul, Turkey
| | - Huxing Cui
- Department of Pharmacology, Iowa Neuroscience Institute, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, IA, USA
| | - Deniz Atasoy
- Department of Pharmacology, Iowa Neuroscience Institute, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, IA, USA.
| |
Collapse
|
30
|
Passeri A, Municchi D, Cavalieri G, Babicola L, Ventura R, Di Segni M. Linking drug and food addiction: an overview of the shared neural circuits and behavioral phenotype. Front Behav Neurosci 2023; 17:1240748. [PMID: 37767338 PMCID: PMC10520727 DOI: 10.3389/fnbeh.2023.1240748] [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: 06/15/2023] [Accepted: 08/21/2023] [Indexed: 09/29/2023] Open
Abstract
Despite a lack of agreement on its definition and inclusion as a specific diagnosable disturbance, the food addiction construct is supported by several neurobiological and behavioral clinical and preclinical findings. Recognizing food addiction is critical to understanding how and why it manifests. In this overview, we focused on those as follows: 1. the hyperpalatable food effects in food addiction development; 2. specific brain regions involved in both food and drug addiction; and 3. animal models highlighting commonalities between substance use disorders and food addiction. Although results collected through animal studies emerged from protocols differing in several ways, they clearly highlight commonalities in behavioral manifestations and neurobiological alterations between substance use disorders and food addiction characteristics. To develop improved food addiction models, this heterogeneity should be acknowledged and embraced so that research can systematically investigate the role of specific variables in the development of the different behavioral features of addiction-like behavior in preclinical models.
Collapse
Affiliation(s)
- Alice Passeri
- IRCCS Fondazione Santa Lucia, Rome, Italy
- Department of Psychology and Center “Daniel Bovet”, Sapienza University, Rome, Italy
| | - Diana Municchi
- IRCCS Fondazione Santa Lucia, Rome, Italy
- Department of Psychology and Center “Daniel Bovet”, Sapienza University, Rome, Italy
| | - Giulia Cavalieri
- Department of Psychology and Center “Daniel Bovet”, Sapienza University, Rome, Italy
| | | | - Rossella Ventura
- Department of Psychology and Center “Daniel Bovet”, Sapienza University, Rome, Italy
- IRCCS San Raffaele, Rome, Italy
| | - Matteo Di Segni
- IRCCS Fondazione Santa Lucia, Rome, Italy
- Department of Psychology and Center “Daniel Bovet”, Sapienza University, Rome, Italy
| |
Collapse
|
31
|
Sun L, Zhu M, Wang M, Hao Y, Hao Y, Jing X, Yu H, Shi Y, Zhang X, Wang S, Yuan F, Yuan XS. Whole-brain monosynaptic inputs and outputs of leptin receptor b neurons of the nucleus tractus solitarii in mice. Brain Res Bull 2023; 201:110693. [PMID: 37348822 DOI: 10.1016/j.brainresbull.2023.110693] [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: 04/16/2023] [Revised: 06/13/2023] [Accepted: 06/19/2023] [Indexed: 06/24/2023]
Abstract
The nucleus tractus solitarii (NTS) is the primary central station that integrates visceral afferent information and regulates respiratory, gastrointestinal, cardiovascular, and other physiological functions. Leptin receptor b (LepRb)-expressing neurons of the NTS (NTSLepRb neurons) are implicated in central respiration regulation, respiratory facilitation, and respiratory drive enhancement. Furthermore, LepRb dysfunction is involved in obesity, insulin resistance, and sleep-disordered breathing. However, the monosynaptic inputs and outputs of NTSLepRb neurons in whole-brain mapping remain to be elucidated. Therefore, the exploration of its whole-brain connection system may provide strong support for comprehensively understanding the physiological and pathological functions of NTSLepRb neurons. In the present study, we used a cell type-specific, modified rabies virus and adeno-associated virus with the Cre-loxp system to map monosynaptic inputs and outputs of NTSLepRb neurons in LepRb-Cre mice. The results showed that NTSLepRb neurons received inputs from 48 nuclei in the whole brain from five brain regions, including especially the medulla. We found that NTSLepRb neurons received inputs from nuclei associated with respiration, such as the pre-Bötzinger complex, ambiguus nucleus, and parabrachial nucleus. Interestingly, some brain areas related to cardiovascular regulation-i.e., the ventrolateral periaqueductal gray and locus coeruleus-also sent a small number of inputs to NTSLepRb neurons. In addition, anterograde tracing results demonstrated that NTSLepRb neurons sent efferent projections to 15 nuclei, including the dorsomedial hypothalamic nucleus and arcuate hypothalamic nucleus, which are involved in regulation of energy metabolism and feeding behaviors. Quantitative statistical analysis revealed that the inputs of the whole brain to NTSLepRb neurons were significantly greater than the outputs. Our study comprehensively revealed neuronal connections of NTSLepRb neurons in the whole brain and provided a neuroanatomical basis for further research on physiological and pathological functions of NTSLepRb neurons.
Collapse
Affiliation(s)
- Lu Sun
- Department of Neurobiology, Hebei Medical University, Shijiazhuang 050017, Hebei Province, China; Department of Anatomy and Histoembryology, School of Basic Medical Sciences, Fudan University, Shanghai 200032, China
| | - Mengchu Zhu
- Department of Neurobiology, Hebei Medical University, Shijiazhuang 050017, Hebei Province, China; Department of Anatomy and Histoembryology, School of Basic Medical Sciences, Fudan University, Shanghai 200032, China
| | - Meng Wang
- Department of Neurobiology, Hebei Medical University, Shijiazhuang 050017, Hebei Province, China
| | - Yinchao Hao
- Department of Neurobiology, Hebei Medical University, Shijiazhuang 050017, Hebei Province, China
| | - Yaxin Hao
- Department of Neurobiology, Hebei Medical University, Shijiazhuang 050017, Hebei Province, China
| | - Xinyi Jing
- Department of Neurobiology, Hebei Medical University, Shijiazhuang 050017, Hebei Province, China
| | - Hongxiao Yu
- Department of Neurobiology, Hebei Medical University, Shijiazhuang 050017, Hebei Province, China; Hebei Key Laboratory of Neurophysiology, Shijiazhuang 050017, Hebei Province, China
| | - Yishuo Shi
- Department of Neurobiology, Hebei Medical University, Shijiazhuang 050017, Hebei Province, China
| | - Xiang Zhang
- Department of Neurobiology, Hebei Medical University, Shijiazhuang 050017, Hebei Province, China
| | - Sheng Wang
- Department of Neurobiology, Hebei Medical University, Shijiazhuang 050017, Hebei Province, China; Hebei Key Laboratory of Neurophysiology, Shijiazhuang 050017, Hebei Province, China
| | - Fang Yuan
- Department of Neurobiology, Hebei Medical University, Shijiazhuang 050017, Hebei Province, China; Hebei Key Laboratory of Neurophysiology, Shijiazhuang 050017, Hebei Province, China.
| | - Xiang Shan Yuan
- Department of Anatomy and Histoembryology, School of Basic Medical Sciences, Fudan University, Shanghai 200032, China.
| |
Collapse
|
32
|
Shaffer C, Barrett LF, Quigley KS. Signal processing in the vagus nerve: Hypotheses based on new genetic and anatomical evidence. Biol Psychol 2023; 182:108626. [PMID: 37419401 PMCID: PMC10563766 DOI: 10.1016/j.biopsycho.2023.108626] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2023] [Revised: 06/25/2023] [Accepted: 07/03/2023] [Indexed: 07/09/2023]
Abstract
Each organism must regulate its internal state in a metabolically efficient way as it interacts in space and time with an ever-changing and only partly predictable world. Success in this endeavor is largely determined by the ongoing communication between brain and body, and the vagus nerve is a crucial structure in that dialogue. In this review, we introduce the novel hypothesis that the afferent vagus nerve is engaged in signal processing rather than just signal relay. New genetic and structural evidence of vagal afferent fiber anatomy motivates two hypotheses: (1) that sensory signals informing on the physiological state of the body compute both spatial and temporal viscerosensory features as they ascend the vagus nerve, following patterns found in other sensory architectures, such as the visual and olfactory systems; and (2) that ascending and descending signals modulate one another, calling into question the strict segregation of sensory and motor signals, respectively. Finally, we discuss several implications of our two hypotheses for understanding the role of viscerosensory signal processing in predictive energy regulation (i.e., allostasis) as well as the role of metabolic signals in memory and in disorders of prediction (e.g., mood disorders).
Collapse
Affiliation(s)
- Clare Shaffer
- Department of Psychology, College of Science, Northeastern University, Boston, MA, USA.
| | - Lisa Feldman Barrett
- Department of Psychology, College of Science, Northeastern University, Boston, MA, USA; Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, USA
| | - Karen S Quigley
- Department of Psychology, College of Science, Northeastern University, Boston, MA, USA.
| |
Collapse
|
33
|
Allingbjerg ML, Hansen SN, Secher A, Thomsen M. Glucagon-like peptide-1 receptors in nucleus accumbens, ventral hippocampus, and lateral septum reduce alcohol reinforcement in mice. Exp Clin Psychopharmacol 2023; 31:612-620. [PMID: 36480394 PMCID: PMC10198891 DOI: 10.1037/pha0000620] [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] [Indexed: 12/13/2022]
Abstract
Glucagon-like peptide 1 (GLP-1) receptor agonists can decrease alcohol intake by central mechanisms that are still poorly understood. The lateral septum (LS) and the ventral/caudal part of the hippocampus are enriched in GLP-1 receptors, and activity in these regions was shown to modulate reward-related behaviors. Using microinfusions of the GLP-1 receptor agonist exendin-4 in mice trained to self-administer oral alcohol in an operant assay, we tested whether pharmacological stimulation of GLP-1 receptors in hippocampus and LS decrease alcohol self-administration. We report that infusion of exendin-4 in the ventral hippocampus or LS was sufficient to reduce alcohol self-administration with as large effect sizes as we previously reported with systemic exendin-4 administration. Infusion of exendin-4 into the nucleus accumbens also reduced alcohol self-administration, as anticipated based on earlier reports, while infusion of exendin-4 into the caudate-putamen (dorsal striatum) had little effect, consistent with lack of GLP-1 receptor expression in this region. The distribution of exendin-4 after infusion into the LS or caudate putamen was visualized using a fluorescently labeled ligand. These findings add to our understanding of the circuit-level mechanisms underlying the ability of GLP-1 receptor agonists to reduce alcohol self-administration. (PsycInfo Database Record (c) 2023 APA, all rights reserved).
Collapse
Affiliation(s)
- Marie-Louise Allingbjerg
- Laboratory of Neuropsychiatry, University Hospital of Copenhagen, Mental Health Services, Capital Region of Denmark
| | | | | | - Morgane Thomsen
- Laboratory of Neuropsychiatry, University Hospital of Copenhagen, Mental Health Services, Capital Region of Denmark
| |
Collapse
|
34
|
Wunderlich ALM, Martins AB, de Souza CF, Stopa LRS, Monteiro ÉCAM, Aguiar DD, Guergolette RP, Zaia CTBV, Uchôa ET. Neonatal overnutrition, but not neonatal undernutrition, disrupts CCK-induced hypophagia and neuron activation of the nucleus of the solitary tract and paraventricular nucleus of hypothalamus of male Wistar rats. Brain Res Bull 2023; 195:109-119. [PMID: 36813046 DOI: 10.1016/j.brainresbull.2023.02.012] [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: 09/12/2022] [Revised: 02/16/2023] [Accepted: 02/18/2023] [Indexed: 02/22/2023]
Abstract
Metabolic programming may be induced by reduction or enhancement of litter size, which lead to neonatal over or undernutrition, respectively. Changes in neonatal nutrition can challenge some regulatory processes in adulthood, such as the hypophagic effect of cholecystokinin (CCK). In order to investigate the effects of nutritional programming on the anorexigenic function of CCK in adulthood, pups were raised in small (SL, 3 pups per dam), normal (NL, 10 pups per dam), or large litters (LL, 16 pups per dam), and on postnatal day 60, male rats were treated with vehicle or CCK (10 µg/Kg) for the evaluation of food intake and c-Fos expression in the area postrema (AP), nucleus of solitary tract (NTS), and paraventricular (PVN), arcuate (ARC), ventromedial (VMH), and dorsomedial (DMH) nuclei of the hypothalamus. Overnourished rats showed increased body weight gain that was inversely correlated with neuronal activation of PaPo, VMH, and DMH neurons, whereas undernourished rats had lower body weight gain, inversely correlated with increased neuronal activation of PaPo only. SL rats showed no anorexigenic response and lower neuron activation in the NTS and PVN induced by CCK. LL exhibited preserved hypophagia and neuron activation in the AP, NTS, and PVN in response to CCK. CCK showed no effect in c-Fos immunoreactivity in the ARC, VMH, and DMH in any litter. These results indicate that anorexigenic actions, associated with neuron activation in the NTS and PVN, induced by CCK were impaired by neonatal overnutrition. However, these responses were not disrupted by neonatal undernutrition. Thus, data suggest that an excess or poor supply of nutrients during lactation display divergent effects on programming CCK satiation signaling in male adult rats.
Collapse
Affiliation(s)
| | - Andressa Busetti Martins
- Multicenter Postgraduate Program in Physiological Sciences, State University of Londrina, Londrina, PR, Brazil
| | - Camila Franciele de Souza
- Multicenter Postgraduate Program in Physiological Sciences, State University of Londrina, Londrina, PR, Brazil
| | - Larissa Rugila S Stopa
- Postgraduate Program in Physiological Sciences, State University of Londrina, Londrina, PR, Brazil
| | | | - Danielly D Aguiar
- Postgraduate Program in Physiological Sciences, State University of Londrina, Londrina, PR, Brazil
| | - Rhauany P Guergolette
- Postgraduate Program in Physiological Sciences, State University of Londrina, Londrina, PR, Brazil
| | - Cássia Thaïs B V Zaia
- Postgraduate Program in Physiological Sciences, State University of Londrina, Londrina, PR, Brazil; Multicenter Postgraduate Program in Physiological Sciences, State University of Londrina, Londrina, PR, Brazil; Department of Physiological Sciences, State University of Londrina, Londrina, PR, Brazil
| | - Ernane Torres Uchôa
- Postgraduate Program in Physiological Sciences, State University of Londrina, Londrina, PR, Brazil; Multicenter Postgraduate Program in Physiological Sciences, State University of Londrina, Londrina, PR, Brazil; Department of Physiological Sciences, State University of Londrina, Londrina, PR, Brazil.
| |
Collapse
|
35
|
Jerlhag E. The therapeutic potential of glucagon-like peptide-1 for persons with addictions based on findings from preclinical and clinical studies. Front Pharmacol 2023; 14:1063033. [PMID: 37063267 PMCID: PMC10097922 DOI: 10.3389/fphar.2023.1063033] [Citation(s) in RCA: 30] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2022] [Accepted: 03/17/2023] [Indexed: 04/03/2023] Open
Abstract
Although the multifaceted mechanisms underlying alcohol use disorder (AUD) have been partially defined, the neurobiological complexity of this disorder is yet to be unraveled. One of the systems that have gained attention in recent times is the gut–brain axis. Although numerous peptides participate in this axis, glucagon-like peptide-1 (GLP-1) plays a central role. GLP-1 is a crucial anorexigenic peptide, with potent abilities to reduce food intake and body weight. The physiological complexity of GLP-1 entails glucose homeostasis, gastrointestinal motility, and the release of insulin and glucagon. As reviewed in this study, acute or repeated treatment with GLP-1 receptor (GLP-1R) agonists decreases alcohol consumption in rodents. Moreover, the abilities of alcohol to promote hyperlocomotion, dopamine release in the nucleus accumbens, and reward in the conditioned place preference paradigm are all suppressed by GLP-1R ligands. Moreover, activation of GLP-1R suppresses the motivation to consume alcohol, alcohol-seeking behaviors, and relapse drinking in male rodents. Similarly, abstinence symptoms experienced during alcohol withdrawal are attenuated by activation of the GLP-1 pathway. On a similar note, the activation of GLP-1 receptors within areas of the brain that are processing reward modulates these alcohol-related responses. Another area that is crucial for this ability is the nucleus of the solitary tract, which is where GLP-1 is produced and from which GLP-1-containing neurons project to areas of reward. These findings may have clinical relevance as AUD is associated with polymorphisms in GLP-1-related genes. Although a GLP-1R agonist does not alter alcohol intake in AUD patients, it reduces this consumption in a sub-population of obese AUD individuals. Given the uncertainty of this outcome, additional clinical studies of obese AUD patients should explore the effects of the GLP-1R agonists on alcohol intake and body weight. Furthermore, GLP-1 receptors modulate the behavioral and neurochemical responses to addictive drugs. Taken together, these preclinical and clinical findings imply that the GLP-1 pathway plays a role in the complex mechanisms regulating alcohol and drug consumption patterns, unveiling a novel aspect of addiction medicine.
Collapse
|
36
|
Hamada S, Mikami K, Ueda S, Nagase M, Nagashima T, Yamamoto M, Bito H, Takemoto-Kimura S, Ohtsuka T, Watabe AM. Experience-dependent changes in affective valence of taste in male mice. Mol Brain 2023; 16:28. [PMID: 36906607 PMCID: PMC10007816 DOI: 10.1186/s13041-023-01017-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2022] [Accepted: 03/03/2023] [Indexed: 03/13/2023] Open
Abstract
Taste plays an essential role in the evaluation of food quality by detecting potential harm and benefit in what animals are about to eat and drink. While the affective valence of taste signals is supposed to be innately determined, taste preference can also be drastically modified by previous taste experiences of the animals. However, how the experience-dependent taste preference is developed and the neuronal mechanisms involved in this process are poorly understood. Here, we investigate the effects of prolonged exposure to umami and bitter tastants on taste preference using two-bottle tests in male mice. Prolonged umami exposure significantly enhanced umami preference with no changes in bitter preference, while prolonged bitter exposure significantly decreased bitter avoidance with no changes in umami preference. Because the central amygdala (CeA) is postulated as a critical node for the valence processing of sensory information including taste, we examined the responses of cells in the CeA to sweet, umami, and bitter tastants using in vivo calcium imaging. Interestingly, both protein kinase C delta (Prkcd)-positive and Somatostatin (Sst)-positive neurons in the CeA showed an umami response comparable to the bitter response, and no difference in cell type-specific activity patterns to different tastants was observed. Meanwhile, fluorescence in situ hybridization with c-Fos antisense probe revealed that a single umami experience significantly activates the CeA and several other gustatory-related nuclei, and especially CeA Sst-positive neurons were strongly activated. Intriguingly, after prolonged umami experience, umami tastant also significantly activates the CeA neurons, but the Prkcd-positive neurons instead of Sst-positive neurons were highly activated. These results suggest a relationship between amygdala activity and experience-dependent plasticity developed in taste preference and the involvement of the genetically defined neural populations in this process.
Collapse
Affiliation(s)
- Shun Hamada
- Department of Biochemistry, Faculty of Medicine, University of Yamanashi, Yamanashi, 409-3898, Japan
| | - Kaori Mikami
- Institute of Clinical Medicine and Research, Research Center for Medical Sciences, The Jikei University School of Medicine, 163-1 Kashiwashita, Kashiwa, Chiba, 277-8567, Japan
| | - Shuhei Ueda
- Department of Neuroscience I, Research Institute of Environmental Medicine, Nagoya University, Nagoya, 464-8601, Japan.,Department of Molecular/Cellular Neuroscience, Nagoya University Graduate School of Medicine, Nagoya, 466-8550, Japan
| | - Masashi Nagase
- Institute of Clinical Medicine and Research, Research Center for Medical Sciences, The Jikei University School of Medicine, 163-1 Kashiwashita, Kashiwa, Chiba, 277-8567, Japan
| | - Takashi Nagashima
- Institute of Clinical Medicine and Research, Research Center for Medical Sciences, The Jikei University School of Medicine, 163-1 Kashiwashita, Kashiwa, Chiba, 277-8567, Japan
| | - Mikiyasu Yamamoto
- Department of Neuroscience I, Research Institute of Environmental Medicine, Nagoya University, Nagoya, 464-8601, Japan.,Department of Molecular/Cellular Neuroscience, Nagoya University Graduate School of Medicine, Nagoya, 466-8550, Japan
| | - Haruhiko Bito
- Department of Neurochemistry, Graduate School of Medicine, The University of Tokyo, Tokyo, 113-0033, Japan
| | - Sayaka Takemoto-Kimura
- Department of Neuroscience I, Research Institute of Environmental Medicine, Nagoya University, Nagoya, 464-8601, Japan.,Department of Molecular/Cellular Neuroscience, Nagoya University Graduate School of Medicine, Nagoya, 466-8550, Japan
| | - Toshihisa Ohtsuka
- Department of Biochemistry, Faculty of Medicine, University of Yamanashi, Yamanashi, 409-3898, Japan
| | - Ayako M Watabe
- Institute of Clinical Medicine and Research, Research Center for Medical Sciences, The Jikei University School of Medicine, 163-1 Kashiwashita, Kashiwa, Chiba, 277-8567, Japan.
| |
Collapse
|
37
|
Krohn F, Novello M, van der Giessen RS, De Zeeuw CI, Pel JJM, Bosman LWJ. The integrated brain network that controls respiration. eLife 2023; 12:83654. [PMID: 36884287 PMCID: PMC9995121 DOI: 10.7554/elife.83654] [Citation(s) in RCA: 54] [Impact Index Per Article: 27.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2022] [Accepted: 01/29/2023] [Indexed: 03/09/2023] Open
Abstract
Respiration is a brain function on which our lives essentially depend. Control of respiration ensures that the frequency and depth of breathing adapt continuously to metabolic needs. In addition, the respiratory control network of the brain has to organize muscular synergies that integrate ventilation with posture and body movement. Finally, respiration is coupled to cardiovascular function and emotion. Here, we argue that the brain can handle this all by integrating a brainstem central pattern generator circuit in a larger network that also comprises the cerebellum. Although currently not generally recognized as a respiratory control center, the cerebellum is well known for its coordinating and modulating role in motor behavior, as well as for its role in the autonomic nervous system. In this review, we discuss the role of brain regions involved in the control of respiration, and their anatomical and functional interactions. We discuss how sensory feedback can result in adaptation of respiration, and how these mechanisms can be compromised by various neurological and psychological disorders. Finally, we demonstrate how the respiratory pattern generators are part of a larger and integrated network of respiratory brain regions.
Collapse
Affiliation(s)
- Friedrich Krohn
- Department of Neuroscience, Erasmus MC, Rotterdam, Netherlands
| | - Manuele Novello
- Department of Neuroscience, Erasmus MC, Rotterdam, Netherlands
| | | | - Chris I De Zeeuw
- Department of Neuroscience, Erasmus MC, Rotterdam, Netherlands.,Netherlands Institute for Neuroscience, Royal Academy of Arts and Sciences, Amsterdam, Netherlands
| | - Johan J M Pel
- Department of Neuroscience, Erasmus MC, Rotterdam, Netherlands
| | | |
Collapse
|
38
|
Watts AG. Paraventricular nucleus-Medullary interactions: How they help enable endocrine responses to metabolic stress. CURRENT OPINION IN ENDOCRINE AND METABOLIC RESEARCH 2022; 27:100401. [PMID: 39575062 PMCID: PMC11580161 DOI: 10.1016/j.coemr.2022.100401] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/24/2024]
Abstract
The paraventricular hypothalamic nucleus (PVH) organizes neuroendocrine and autonomic responses to rapidly and slowly developing metabolic stressors that limit their impact on energy balance. The PVH together with the lateral hypothalamic area, and the arcuate and dorsomedial nuclei form a network that is defined by its inputs from medullary catecholamine neurons. These medullary neurons convey important glycemia and glucocorticoid feedback information that is integrated by the PVH and the rest of this network to control a variety of responses to metabolic stressors that have rapid (hypoglycemia) or slow onsets (eating a high calorie diet). This review focuses on how the responses to these two challenges are enabled by these catecholamine neurons, and the integrative nature of the network into which they project.
Collapse
Affiliation(s)
- Alan G Watts
- The Department of Biological Sciences, USC Dornsife College of Letters, Arts & Sciences, University of Southern California, Los Angeles, USA
| |
Collapse
|
39
|
Zheng H, López-Ferreras L, Krieger JP, Fasul S, Cea Salazar V, Valderrama Pena N, Skibicka KP, Rinaman L. A Cre-driver rat model for anatomical and functional analysis of glucagon (Gcg)-expressing cells in the brain and periphery. Mol Metab 2022; 66:101631. [PMID: 36368622 PMCID: PMC9677222 DOI: 10.1016/j.molmet.2022.101631] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/05/2022] [Revised: 11/02/2022] [Accepted: 11/03/2022] [Indexed: 11/09/2022] Open
Abstract
OBJECTIVE The glucagon gene (Gcg) encodes preproglucagon, which is cleaved to form glucagon-like peptide 1 (GLP1) and other mature signaling molecules implicated in metabolic functions. To date there are no transgenic rat models available for precise manipulation of GLP1-expressing cells in the brain and periphery. METHODS To visualize and manipulate Gcg-expressing cells in rats, CRISPR/Cas9 was used to express iCre under control of the Gcg promoter. Gcg-Cre rats were bred with tdTomato reporter rats to tag Gcg-expressing cells. Cre-dependent AAVs and RNAscope in situ hybridization were used to evaluate the specificity of iCre expression by GLP1 neurons in the caudal nucleus of the solitary tract (cNTS) and intermediate reticular nucleus (IRt), and by intestinal and pancreatic secretory cells. Food intake was assessed in heterozygous (Het) Gcg-Cre rats after chemogenetic stimulation of cNTS GLP1 neurons expressing an excitatory DREADD. RESULTS While genotype has minimal effect on body weight or composition in chow-fed Gcg-Cre rats, homozygous (Homo) rats have lower plasma glucose levels. In neonatal and adult Gcg-Cre/tdTom rats, reporter-labeled cells are present in the cNTS and IRt, and in additional brain regions (e.g., basolateral amygdala, piriform cortex) that lack detectable Gcg mRNA in adults but display transient developmental or persistently low Gcg expression. Compared to wildtype (WT) rats, hindbrain Gcg mRNA and GLP1 protein in brain and plasma are markedly reduced in Homo Gcg-Cre rats. Chemogenetic stimulation of cNTS GLP1 neurons reduced overnight chow intake in males but not females, the effect in males was blocked by antagonism of central GLP1 receptors, and hypophagia was enhanced when combined with a subthreshold dose of cholecystokinin-8 to stimulate gastrointestinal vagal afferents. CONCLUSIONS Gcg-Cre rats are a novel and valuable experimental tool for analyzing the development, anatomy, and function of Gcg-expressing cells in the brain and periphery. In addition, Homo Gcg-Cre rats are a unique model for assessing the role of Gcg-encoded proteins in glucose homeostasis and energy metabolism.
Collapse
Affiliation(s)
- Huiyuan Zheng
- Department of Psychology and Program in Neuroscience, Florida State University, Tallahassee, FL 32306, USA
| | - Lorena López-Ferreras
- Institute of Neuroscience and Physiology, Department of Physiology/Metabolic Physiology, The Sahlgrenska Academy at University of Gothenburg, Sweden
| | - Jean-Phillipe Krieger
- Institute of Neuroscience and Physiology, Department of Physiology/Metabolic Physiology, The Sahlgrenska Academy at University of Gothenburg, Sweden
| | - Stephen Fasul
- Department of Psychology and Program in Neuroscience, Florida State University, Tallahassee, FL 32306, USA
| | - Valentina Cea Salazar
- Department of Psychology and Program in Neuroscience, Florida State University, Tallahassee, FL 32306, USA
| | - Natalia Valderrama Pena
- Department of Psychology and Program in Neuroscience, Florida State University, Tallahassee, FL 32306, USA
| | - Karolina P. Skibicka
- Department of Nutritional Sciences, College of Health and Human Development, Huck Institute, The Pennsylvania State University, University Park, PA, USA,Institute of Neuroscience and Physiology, Department of Physiology/Metabolic Physiology, The Sahlgrenska Academy at University of Gothenburg, Sweden,Corresponding author. Department of Nutritional Sciences, Pennsylvania State University, 204 Chandlee Lab, University Park, PA 16802, USA
| | - Linda Rinaman
- Department of Psychology and Program in Neuroscience, Florida State University, Tallahassee, FL 32306, USA,Corresponding author. Department of Psychology, Program in Neuroscience, Florida State University, 1107 W. Call Street, Tallahassee, FL 32306, USA.
| |
Collapse
|
40
|
Evans B, Stoltzfus B, Acharya N, Nyland JE, Arnold AC, Freet CS, Bunce SC, Grigson PS. Dose titration with the glucagon-like peptide-1 agonist, liraglutide, reduces cue- and drug-induced heroin seeking in high drug-taking rats. Brain Res Bull 2022; 189:163-173. [PMID: 36038016 PMCID: PMC10757750 DOI: 10.1016/j.brainresbull.2022.08.022] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2022] [Revised: 08/17/2022] [Accepted: 08/24/2022] [Indexed: 01/13/2023]
Abstract
Opioid use disorder (OUD), like other substance use disorders (SUDs), is widely understood to be a disorder of persistent relapse. Despite the use of three FDA-approved medications for OUD, typically in conjunction with behavioral treatments, relapse rates remain unacceptably high. Whereas medication assisted therapy (MAT) reduces the risk of opioid overdose mortality, the benefits of MAT are negated when people discontinue the medications. Currently approved medications present barriers to efficient use, including daily visits to a treatment center or work restrictions. With spiking increases in opioid relapse and death, it is imperative to identify new treatments that can reduce the risk of relapse. Recent evidence suggests that glucagon-like peptide-1 receptor agonists (GLP-1RAs), currently FDA-approved to treat obesity and type two diabetes, may be promising candidates to reduce relapse. GLP-1RAs have been shown to reduce relapse in rats, whether elicited by cues, drug, and/or stress. However, GLP-1RAs also can cause gastrointestinal malaise, and therefore, in humans, the medication typically is titrated up to full dose when initiating treatment. Here, we used a rodent model to test whether cue- and drug-induced heroin seeking can be reduced by the GLP-1RA, liraglutide, when the dose is titrated across the abstinence period and prior to test. The results show this titration regimen is effective in reducing both cue-induced heroin seeking and drug-induced reinstatement of heroin seeking, particularly in rats with a history of high drug-taking. Importantly, this treatment regimen had no effect on either circulating glucose or insulin. GLP-1RAs, then, appear strong candidates for the non-opioid prevention of relapse to opioids.
Collapse
Affiliation(s)
- Brianna Evans
- Department of Neural and Behavioral Sciences, The Pennsylvania State University College of Medicine, Hershey, PA 17033, USA; Penn State Addiction Center for Translation, The Pennsylvania State University College of Medicine, Hershey, PA 17033, USA
| | - Brooke Stoltzfus
- Department of Neural and Behavioral Sciences, The Pennsylvania State University College of Medicine, Hershey, PA 17033, USA
| | - Nikhil Acharya
- Department of Neural and Behavioral Sciences, The Pennsylvania State University College of Medicine, Hershey, PA 17033, USA; Penn State Addiction Center for Translation, The Pennsylvania State University College of Medicine, Hershey, PA 17033, USA
| | - Jennifer E Nyland
- Department of Neural and Behavioral Sciences, The Pennsylvania State University College of Medicine, Hershey, PA 17033, USA; Penn State Addiction Center for Translation, The Pennsylvania State University College of Medicine, Hershey, PA 17033, USA
| | - Amy C Arnold
- Department of Neural and Behavioral Sciences, The Pennsylvania State University College of Medicine, Hershey, PA 17033, USA; Penn State Addiction Center for Translation, The Pennsylvania State University College of Medicine, Hershey, PA 17033, USA
| | - Christopher S Freet
- Department of Psychiatry and Behavioral Health, The Pennsylvania State University College of Medicine, Hershey, PA 17033, USA; Penn State Addiction Center for Translation, The Pennsylvania State University College of Medicine, Hershey, PA 17033, USA
| | - Scott C Bunce
- Department of Psychiatry and Behavioral Health, The Pennsylvania State University College of Medicine, Hershey, PA 17033, USA; Penn State Addiction Center for Translation, The Pennsylvania State University College of Medicine, Hershey, PA 17033, USA
| | - Patricia S Grigson
- Department of Neural and Behavioral Sciences, The Pennsylvania State University College of Medicine, Hershey, PA 17033, USA; Penn State Addiction Center for Translation, The Pennsylvania State University College of Medicine, Hershey, PA 17033, USA.
| |
Collapse
|
41
|
Klausen MK, Jensen ME, Møller M, Le Dous N, Jensen AMØ, Zeeman VA, Johannsen CF, Lee A, Thomsen GK, Macoveanu J, Fisher PM, Gillum MP, Jørgensen NR, Bergmann ML, Enghusen Poulsen H, Becker U, Holst JJ, Benveniste H, Volkow ND, Vollstädt-Klein S, Miskowiak KW, Ekstrøm CT, Knudsen GM, Vilsbøll T, Fink-Jensen A. Exenatide once weekly for alcohol use disorder investigated in a randomized, placebo-controlled clinical trial. JCI Insight 2022; 7:e159863. [PMID: 36066977 PMCID: PMC9675448 DOI: 10.1172/jci.insight.159863] [Citation(s) in RCA: 85] [Impact Index Per Article: 28.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2022] [Accepted: 08/31/2022] [Indexed: 11/17/2022] Open
Abstract
BackgroundAlcohol use disorder (AUD) is a chronic, relapsing brain disorder that accounts for 5% of deaths annually, and there is an urgent need to develop new targets for therapeutic intervention. The glucagon-like peptide-1 (GLP-1) receptor agonist exenatide reduces alcohol consumption in rodents and nonhuman primates, but its efficacy in patients with AUD is unknown.MethodsIn a randomized, double-blinded, placebo-controlled clinical trial, treatment-seeking AUD patients were assigned to receive exenatide (2 mg subcutaneously) or placebo once weekly for 26 weeks, in addition to standard cognitive-behavioral therapy. The primary outcome was reduction in number of heavy drinking days. A subgroup also completed functional MRI (fMRI) and single-photon emission CT (SPECT) brain scans.ResultsA total of 127 patients were enrolled. Our data revealed that although exenatide did not significantly reduce the number of heavy drinking days compared with placebo, it significantly attenuated fMRI alcohol cue reactivity in the ventral striatum and septal area, which are crucial brain areas for drug reward and addiction. In addition, dopamine transporter availability was lower in the exenatide group compared with the placebo group. Exploratory analyses revealed that exenatide significantly reduced heavy drinking days and total alcohol intake in a subgroup of obese patients (BMI > 30 kg/m2). Adverse events were mainly gastrointestinal.ConclusionThis randomized controlled trial on the effects of a GLP-1 receptor agonist in AUD patients provides new important knowledge on the effects of GLP-1 receptor agonists as a novel treatment target in addiction.Trial registrationEudraCT: 2016-003343-11. ClinicalTrials.gov (NCT03232112).FundingNovavi Foundation; Research Foundation, Mental Health Services, Capital Region of Denmark; Research Foundation, Capital Region of Denmark; Ivan Nielsen Foundation; A.P. Moeller Foundation; Augustinus Foundation; Woerzner Foundation; Grosserer L.F. Foghts Foundation; Hartmann Foundation; Aase and Ejnar Danielsen Foundation; P.A. Messerschmidt and Wife Foundation; and Lundbeck Foundation.
Collapse
Affiliation(s)
- Mette Kruse Klausen
- Psychiatric Centre Copenhagen, Rigshospitalet, Copenhagen, Denmark
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Mathias Ebbesen Jensen
- Psychiatric Centre Copenhagen, Rigshospitalet, Copenhagen, Denmark
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Marco Møller
- Psychiatric Centre Copenhagen, Rigshospitalet, Copenhagen, Denmark
| | - Nina Le Dous
- Psychiatric Centre Copenhagen, Rigshospitalet, Copenhagen, Denmark
| | | | | | | | - Alycia Lee
- Department of Addictive Behavior and Addiction Medicine, Central Institute of Mental Health, and
- Mannheim Center for Translational Neurosciences, Medical Faculty of Mannheim, University of Heidelberg, Mannheim, Germany
| | - Gerda Krog Thomsen
- Neurobiology Research Unit, Copenhagen University Hospital Rigshospitalet, Copenhagen, Denmark
| | - Julian Macoveanu
- Psychiatric Centre Copenhagen, Rigshospitalet, Copenhagen, Denmark
| | | | - Matthew Paul Gillum
- Novo Nordisk Foundation Center for Basic Metabolic Research and Department of Biomedical Sciences and
| | - Niklas Rye Jørgensen
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- Department of Clinical Biochemistry, Centre of Diagnostic Investigation, University of Copenhagen, Copenhagen, Denmark
| | - Marianne Lerbæk Bergmann
- Department of Biochemistry and Immunology, University Hospital of Southern Denmark, Vejle, Denmark
| | - Henrik Enghusen Poulsen
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- Department of Clinical Pharmacology, Bispebjerg/Frederiksberg Hospital, University Hospital Copenhagen, Copenhagen, Denmark
| | - Ulrik Becker
- National Institute of Public Health, University of Southern Denmark and University of Copenhagen, Copenhagen, Denmark
| | - Jens Juul Holst
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- Novo Nordisk Foundation Center for Basic Metabolic Research and Department of Biomedical Sciences and
| | - Helene Benveniste
- Department of Anesthesiology and Pediatric Anesthesiology, Yale University, New Haven, Connecticut, USA
| | - Nora D. Volkow
- National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, Maryland, USA
| | - Sabine Vollstädt-Klein
- Department of Addictive Behavior and Addiction Medicine, Central Institute of Mental Health, and
- Mannheim Center for Translational Neurosciences, Medical Faculty of Mannheim, University of Heidelberg, Mannheim, Germany
| | - Kamilla Woznica Miskowiak
- Psychiatric Centre Copenhagen, Rigshospitalet, Copenhagen, Denmark
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- Department of Psychology
| | | | - Gitte Moos Knudsen
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- Neurobiology Research Unit, Copenhagen University Hospital Rigshospitalet, Copenhagen, Denmark
| | - Tina Vilsbøll
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- Steno Diabetes Center Copenhagen, University of Copenhagen, Copenhagen, Denmark
| | - Anders Fink-Jensen
- Psychiatric Centre Copenhagen, Rigshospitalet, Copenhagen, Denmark
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
42
|
Bales MB, Centanni SW, Luchsinger JR, Fathi P, Biddinger JE, Le TDV, Nwaba KG, Paldrmic IM, Winder DG, Ayala JE. High fat diet blunts stress-induced hypophagia and activation of Glp1r dorsal lateral septum neurons in male but not in female mice. Mol Metab 2022; 64:101571. [PMID: 35953023 PMCID: PMC9418981 DOI: 10.1016/j.molmet.2022.101571] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/10/2022] [Revised: 07/19/2022] [Accepted: 08/02/2022] [Indexed: 12/17/2022] Open
Abstract
OBJECTIVE While stress typically reduces caloric intake (hypophagia) in chow-fed rodents, presentation of palatable, high calorie substances during stress can increase caloric consumption (i.e. "comfort feeding") and promote obesity. However, little is known about how obesity itself affects feeding behavior in response to stress and the mechanisms that can influence stress-associated feeding in the context of obesity. METHODS We assessed food intake and other metabolic parameters in lean and obese male and female mice following acute restraint stress. We also measured real-time activity of glucagon-like peptide-1 (Glp1) receptor (Glp1r)-expressing neurons in the dorsal lateral septum (dLS) during stress in lean and obese mice using fiber photometry. Glp1r activation in various brain regions, including the dLS, promotes hypophagia in response to stress. Finally, we used inhibitory Designer Receptors Activated Exclusively by Designer Drugs (DREADDs) to test whether activation of Glp1r-expressing neurons in the LS is required for stress-induced hypophagia. RESULTS Lean male mice display the expected hypophagic response following acute restraint stress, but obese male mice are resistant to this acute stress-induced hypophagia. Glp1r-positive neurons in the dLS are robustly activated during acute restraint stress in lean but not in obese male mice. This raises the possibility that activation of dLS Glp1r neurons during restraint stress contributes to subsequent hypophagia. Supporting this, we show that chemogenetic inhibition of LS Glp1r neurons attenuates acute restraint stress hypophagia in male mice. Surprisingly, we show that both lean and obese female mice are resistant to acute restraint stress-induced hypophagia and activation of dLS Glp1r neurons. CONCLUSIONS These results suggest that dLS Glp1r neurons contribute to the hypophagic response to acute restraint stress in male mice, but not in female mice, and that obesity disrupts this response in male mice. Broadly, these findings show sexually dimorphic mechanisms and feeding behaviors in lean vs. obese mice in response to acute stress.
Collapse
Affiliation(s)
- Michelle B Bales
- Department of Molecular Physiology & Biophysics, Vanderbilt University School of Medicine, 2215 Garland Avenue, Nashville, TN 37232, USA
| | - Samuel W Centanni
- Department of Molecular Physiology & Biophysics, Vanderbilt University School of Medicine, 2215 Garland Avenue, Nashville, TN 37232, USA; Vanderbilt Center for Addiction Research, Vanderbilt University School of Medicine, 2215 Garland Avenue, Nashville, TN 37232, USA
| | - Joseph R Luchsinger
- Department of Molecular Physiology & Biophysics, Vanderbilt University School of Medicine, 2215 Garland Avenue, Nashville, TN 37232, USA
| | - Payam Fathi
- Department of Molecular Physiology & Biophysics, Vanderbilt University School of Medicine, 2215 Garland Avenue, Nashville, TN 37232, USA
| | - Jessica E Biddinger
- Department of Molecular Physiology & Biophysics, Vanderbilt University School of Medicine, 2215 Garland Avenue, Nashville, TN 37232, USA
| | - Thao D V Le
- Department of Molecular Physiology & Biophysics, Vanderbilt University School of Medicine, 2215 Garland Avenue, Nashville, TN 37232, USA
| | - Kaitlyn Ginika Nwaba
- Department of Molecular Physiology & Biophysics, Vanderbilt University School of Medicine, 2215 Garland Avenue, Nashville, TN 37232, USA
| | - Isabella M Paldrmic
- Department of Molecular Physiology & Biophysics, Vanderbilt University School of Medicine, 2215 Garland Avenue, Nashville, TN 37232, USA
| | - Danny G Winder
- Department of Molecular Physiology & Biophysics, Vanderbilt University School of Medicine, 2215 Garland Avenue, Nashville, TN 37232, USA; Vanderbilt Center for Addiction Research, Vanderbilt University School of Medicine, 2215 Garland Avenue, Nashville, TN 37232, USA
| | - Julio E Ayala
- Department of Molecular Physiology & Biophysics, Vanderbilt University School of Medicine, 2215 Garland Avenue, Nashville, TN 37232, USA; Vanderbilt Center for Addiction Research, Vanderbilt University School of Medicine, 2215 Garland Avenue, Nashville, TN 37232, USA; Vanderbilt Mouse Metabolic Phenotyping Center, Vanderbilt University School of Medicine, 2215 Garland Avenue, Nashville, TN 37232, USA.
| |
Collapse
|
43
|
Neuhuber WL, Berthoud HR. Functional anatomy of the vagus system: How does the polyvagal theory comply? Biol Psychol 2022; 174:108425. [PMID: 36100134 DOI: 10.1016/j.biopsycho.2022.108425] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2022] [Revised: 09/07/2022] [Accepted: 09/07/2022] [Indexed: 11/16/2022]
Abstract
Due to its pivotal role in autonomic networks and interoception, the vagus attracts continued interest from both basic scientists and therapists of various clinical disciplines. In particular, the widespread use of heart rate variability as an index of autonomic cardiac control and a proposed central role of the vagus in biopsychological concepts, e.g., the polyvagal theory, provide a good opportunity to recall basic features of vagal anatomy. In addition to the "classical" vagal brainstem nuclei, i.e., dorsal motor nucleus, nucleus ambiguus and nucleus tractus solitarii, the spinal trigeminal and paratrigeminal nuclei come into play as targets of vagal afferents. On the other hand, the nucleus of the solitary tract receives and integrates not only visceral but also somatic afferents.
Collapse
Affiliation(s)
- Winfried L Neuhuber
- Institute of Anatomy and Cell Biology, Friedrich-Alexander-Universität, Krankenhausstrasse 9, Erlangen, Germany.
| | - Hans-Rudolf Berthoud
- Neurobiology of Nutrition & Metabolism Department, Pennington Biomedical Research Center, Louisiana State University System, 6400 Perkins Road, Baton Rouge, LA 70808, USA.
| |
Collapse
|
44
|
de Wouters d’Oplinter A, Huwart SJP, Cani PD, Everard A. Gut microbes and food reward: From the gut to the brain. Front Neurosci 2022; 16:947240. [PMID: 35958993 PMCID: PMC9358980 DOI: 10.3389/fnins.2022.947240] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2022] [Accepted: 07/04/2022] [Indexed: 11/13/2022] Open
Abstract
Inappropriate food intake behavior is one of the main drivers for fat mass development leading to obesity. Importantly the gut microbiota-mediated signals have emerged as key actors regulating food intake acting mainly on the hypothalamus, and thereby controlling hunger or satiety/satiation feelings. However, food intake is also controlled by the hedonic and reward systems leading to food intake based on pleasure (i.e., non-homeostatic control of food intake). This review focus on both the homeostatic and the non-homeostatic controls of food intake and the implication of the gut microbiota on the control of these systems. The gut-brain axis is involved in the communications between the gut microbes and the brain to modulate host food intake behaviors through systemic and nervous pathways. Therefore, here we describe several mediators of the gut-brain axis including gastrointestinal hormones, neurotransmitters, bioactive lipids as well as bacterial metabolites and compounds. The modulation of gut-brain axis by gut microbes is deeply addressed in the context of host food intake with a specific focus on hedonic feeding. Finally, we also discuss possible gut microbiota-based therapeutic approaches that could lead to potential clinical applications to restore food reward alterations. Therapeutic applications to tackle these dysregulations is of utmost importance since most of the available solutions to treat obesity present low success rate.
Collapse
|
45
|
Rei D, Saha S, Haddad M, Haider Rubio A, Perlaza BL, Berard M, Ungeheuer MN, Sokol H, Lledo PM. Age-associated gut microbiota impairs hippocampus-dependent memory in a vagus-dependent manner. JCI Insight 2022; 7:147700. [PMID: 35737457 PMCID: PMC9462480 DOI: 10.1172/jci.insight.147700] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2021] [Accepted: 06/20/2022] [Indexed: 12/03/2022] Open
Abstract
Aging is known to be associated with hippocampus-dependent memory decline, but the underlying causes of this age-related memory impairment remain highly debated. Here, we show that fecal microbiota transplantation (FMT) from aged, but not young, animal donors into young mice is sufficient to trigger profound hippocampal alterations, including astrogliosis, decreased adult neurogenesis, decreased novelty-induced neuronal activation, and impairment in hippocampus-dependent memory. Furthermore, similar alterations were reported when mice were subjected to an FMT from aged human donors. To decipher the mechanisms involved in mediating these microbiota-induced effects on brain function, we mapped the vagus nerve–related (VN-related) neuronal activity patterns and report that aged FMT animals showed a reduction in neuronal activity in the ascending-VN output brain structure, whether under basal condition or after VN stimulation. Targeted pharmacogenetic manipulation of VN-ascending neurons demonstrated that the decrease in vagal activity is detrimental to hippocampal functions. In contrast, increasing vagal ascending activity alleviated the adverse effects of aged mouse FMT on hippocampal functions and had a promnesic effect in aged mice. Thus, pharmacogenetic VN stimulation is a potential therapeutic strategy to lessen microbiota-dependent age-associated impairments in hippocampal functions.
Collapse
Affiliation(s)
- Damien Rei
- Neurosciences, Institut Pasteur de Paris, Paris, France
| | - Soham Saha
- Neurosciences, Institut Pasteur de Paris, Paris, France
| | | | | | | | - Marion Berard
- Animalerie Centrale, Institut Pasteur de Paris, Paris, France
| | | | - Harry Sokol
- Centre de Recherche Saint-Antoine, Sorbonne Université, Paris, France
| | | |
Collapse
|
46
|
Tsermpini EE, Goričar K, Kores Plesničar B, Plemenitaš Ilješ A, Dolžan V. Genetic Variability of Incretin Receptors and Alcohol Dependence: A Pilot Study. Front Mol Neurosci 2022; 15:908948. [PMID: 35754710 PMCID: PMC9218814 DOI: 10.3389/fnmol.2022.908948] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Accepted: 05/09/2022] [Indexed: 11/13/2022] Open
Abstract
Alcohol dependence is a chronic mental disorder that leads to decreased quality of life for patients and their relatives and presents a considerable burden to society. Incretin hormones, such as glucose-dependent insulinotropic polypeptide (GIP) and glucagon-like peptide 1 (GLP-1) are endogenous gut-brain peptides, which can travel across the blood-brain barrier and access the nervous system. Their respective receptors, GIPR and GLP-1R, are expressed in the reward-related brain areas and are involved in memory formation and neurogenesis, which results in behavioral changes in rodent models. The current study investigated the potential association of genetic variability of incretin receptors with alcohol dependence and alcohol-related psychosymptomatology. Alcohol dependence and comorbid psychosymptomatology were assessed in a cohort of Slovenian male participants, comprised of 89 hospitalized alcohol-dependent patients, 98 abstinent alcohol-dependent patients, and 93 healthy blood donors. All participants were genotyped for GIPR rs1800437 and GLP1R rs10305420 and rs6923761 polymorphisms. For the statistical analysis Kruskal-Wall and Mann-Whitney tests were used in additive and dominant genetic models. Our findings indicated that GIPR rs1800437 genotypes were associated with an increased risk of alcohol dependence. Statistically significant association between GIPR rs1800437 GG genotype and Brief Social Phobia Scale scores were observed in the abstinent alcohol-dependent patients, while GLP1R rs6923761 GG genotype was associated with Zung anxiety scores in healthy controls. Our pilot study indicates that GIPR rs1800437 may play some role in susceptibility to alcohol dependence, as well as in alcohol-related psychosymptomatology symptoms. To our knowledge, this is the first study that indicates the involvement of GIPR in alcohol dependence. However, studies with larger cohorts are needed to confirm these preliminary findings.
Collapse
Affiliation(s)
- Evangelia Eirini Tsermpini
- Pharmacogenetics Laboratory, Institute of Biochemistry and Molecular Genetics, Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
| | - Katja Goričar
- Pharmacogenetics Laboratory, Institute of Biochemistry and Molecular Genetics, Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
| | - Blanka Kores Plesničar
- University Psychiatric Clinic, Ljubljana, Slovenia
- Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
| | | | - Vita Dolžan
- Pharmacogenetics Laboratory, Institute of Biochemistry and Molecular Genetics, Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
| |
Collapse
|
47
|
Holt MK. The ins and outs of the caudal nucleus of the solitary tract: An overview of cellular populations and anatomical connections. J Neuroendocrinol 2022; 34:e13132. [PMID: 35509189 PMCID: PMC9286632 DOI: 10.1111/jne.13132] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/07/2021] [Revised: 02/25/2022] [Accepted: 03/22/2022] [Indexed: 11/29/2022]
Abstract
The body and brain are in constant two-way communication. Driving this communication is a region in the lower brainstem: the dorsal vagal complex. Within the dorsal vagal complex, the caudal nucleus of the solitary tract (cNTS) is a major first stop for incoming information from the body to the brain carried by the vagus nerve. The anatomy of this region makes it ideally positioned to respond to signals of change in both emotional and bodily states. In turn, the cNTS controls the activity of regions throughout the brain that are involved in the control of both behaviour and physiology. This review is intended to help anyone with an interest in the cNTS. First, I provide an overview of the architecture of the cNTS and outline the wide range of neurotransmitters expressed in subsets of neurons in the cNTS. Next, in detail, I discuss the known inputs and outputs of the cNTS and briefly highlight what is known regarding the neurochemical makeup and function of those connections. Then, I discuss one group of cNTS neurons: glucagon-like peptide-1 (GLP-1)-expressing neurons. GLP-1 neurons serve as a good example of a group of cNTS neurons, which receive input from varied sources and have the ability to modulate both behaviour and physiology. Finally, I consider what we might learn about other cNTS neurons from our study of GLP-1 neurons and why it is important to remember that the manipulation of molecularly defined subsets of cNTS neurons is likely to affect physiology and behaviours beyond those monitored in individual experiments.
Collapse
Affiliation(s)
- Marie K. Holt
- Centre for Cardiovascular and Metabolic Neuroscience, Department of Neuroscience, Physiology and PharmacologyUniversity College LondonLondonUK
| |
Collapse
|
48
|
Dunigan AI, Roseberry AG. Actions of feeding-related peptides on the mesolimbic dopamine system in regulation of natural and drug rewards. ADDICTION NEUROSCIENCE 2022; 2:100011. [PMID: 37220637 PMCID: PMC10201992 DOI: 10.1016/j.addicn.2022.100011] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 05/25/2023]
Abstract
The mesolimbic dopamine system is the primary neural circuit mediating motivation, reinforcement, and reward-related behavior. The activity of this system and multiple behaviors controlled by it are affected by changes in feeding and body weight, such as fasting, food restriction, or the development of obesity. Multiple different peptides and hormones that have been implicated in the control of feeding and body weight interact with the mesolimbic dopamine system to regulate many different dopamine-dependent, reward-related behaviors. In this review, we summarize the effects of a selected set of feeding-related peptides and hormones acting within the ventral tegmental area and nucleus accumbens to alter feeding, as well as food, drug, and social reward.
Collapse
Affiliation(s)
- Anna I. Dunigan
- Department of Biology and Neuroscience Institute, Georgia State University, Atlanta, GA 30303, USA
| | - Aaron G. Roseberry
- Department of Biology and Neuroscience Institute, Georgia State University, Atlanta, GA 30303, USA
- Neuroscience Institute, Georgia State University, Atlanta, GA 30303, USA
| |
Collapse
|
49
|
Li H, Page AJ. Altered Vagal Signaling and Its Pathophysiological Roles in Functional Dyspepsia. Front Neurosci 2022; 16:858612. [PMID: 35527812 PMCID: PMC9072791 DOI: 10.3389/fnins.2022.858612] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2022] [Accepted: 03/28/2022] [Indexed: 11/20/2022] Open
Abstract
The vagus nerve is crucial in the bidirectional communication between the gut and the brain. It is involved in the modulation of a variety of gut and brain functions. Human studies indicate that the descending vagal signaling from the brain is impaired in functional dyspepsia. Growing evidence indicate that the vagal signaling from gut to brain may also be altered, due to the alteration of a variety of gut signals identified in this disorder. The pathophysiological roles of vagal signaling in functional dyspepsia is still largely unknown, although some studies suggested it may contribute to reduced food intake and gastric motility, increased psychological disorders and pain sensation, nausea and vomiting. Understanding the alteration in vagal signaling and its pathophysiological roles in functional dyspepsia may provide information for new potential therapeutic treatments of this disorder. In this review, we summarize and speculate possible alterations in vagal gut-to-brain and brain-to-gut signaling and the potential pathophysiological roles in functional dyspepsia.
Collapse
Affiliation(s)
- Hui Li
- Vagal Afferent Research Group, Adelaide Medical School, The University of Adelaide, Adelaide, SA, Australia
- Nutrition, Diabetes and Gut Health, Lifelong Health Theme, South Australian Health and Medical Research Institute, Adelaide, SA, Australia
- *Correspondence: Hui Li,
| | - Amanda J. Page
- Vagal Afferent Research Group, Adelaide Medical School, The University of Adelaide, Adelaide, SA, Australia
- Nutrition, Diabetes and Gut Health, Lifelong Health Theme, South Australian Health and Medical Research Institute, Adelaide, SA, Australia
| |
Collapse
|
50
|
Linagliptin, a Selective Dipeptidyl Peptidase-4 Inhibitor, Reduces Physical and Behavioral Effects of Morphine Withdrawal. Molecules 2022; 27:molecules27082478. [PMID: 35458676 PMCID: PMC9028142 DOI: 10.3390/molecules27082478] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Revised: 03/18/2022] [Accepted: 04/11/2022] [Indexed: 11/17/2022] Open
Abstract
(1) Background: Recent data indicate that receptors for GLP-1 peptide are involved in the activity of the mesolimbic system. Thus, the purpose of the present study was to examine the effect of the selective dipeptidyl peptidase-4 (DPP-4) inhibitor, linagliptin, on morphine dependence in mice. (2) Methods: Morphine dependence in mice was obtained by administration of increasing doses of morphine for eight consecutive days, twice a day. On the 9th day of the experiment, the naloxone-induced (2 mg/kg, ip) morphine withdrawal signs (jumping) were assessed. Moreover, behavioral effects of short-term (60 h after morphine discontinuation) and long-term (14 days after morphine discontinuation) morphine withdrawal were observed. In terms of behavioral effects, the depressive effect in the forced swim test and anxiety in the elevated plus maze test were investigated. Locomotor activity of mice was also studied. (3) Results: The administration of linagliptin (10 and 20 mg/kg, ip) for 8 consecutive days before morphine injections significantly diminished the number of naloxone-induced morphine withdrawal signs (jumping) in mice. In addition, the cessation of morphine administration induced depressive behavior in mice which were observed during short- and long-term morphine withdrawal. Linagliptin administered during morphine withdrawal significantly reduced the depressive behavior in studied mice. Furthermore, the short-term morphine withdrawal evoked anxiety which also was reduced by linagliptin in mice. (4) Conclusions: The present study reveals that GLP-1 receptors are involved in morphine dependence. What is more, linagliptin might be a valuable drug in attenuating the physical symptoms of morphine dependence. It might be also a useful drug in reducing emotional disturbances which may develop during the morphine withdrawal period.
Collapse
|