1
|
Thulasinathan B, Suvilesh KN, Maram S, Grossmann E, Ghouri Y, Teixeiro EP, Chan J, Kaif JT, Rachagani S. The impact of gut microbial short-chain fatty acids on colorectal cancer development and prevention. Gut Microbes 2025; 17:2483780. [PMID: 40189834 PMCID: PMC11980463 DOI: 10.1080/19490976.2025.2483780] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/18/2024] [Revised: 02/18/2025] [Accepted: 03/18/2025] [Indexed: 04/11/2025] Open
Abstract
Cancer is a long-term illness that involves an imbalance in cellular and immune functions. It can be caused by a range of factors, including exposure to environmental carcinogens, poor diet, infections, and genetic alterations. Maintaining a healthy gut microbiome is crucial for overall health, and short-chain fatty acids (SCFAs) produced by gut microbiota play a vital role in this process. Recent research has established that alterations in the gut microbiome led to decreased production of SCFA's in lumen of the colon, which associated with changes in the intestinal epithelial barrier function, and immunity, are closely linked to colorectal cancer (CRC) development and its progression. SCFAs influence cancer progression by modifying epigenetic mechanisms such as DNA methylation, histone modifications, and non-coding RNA functions thereby affecting tumor initiation and metastasis. This suggests that restoring SCFA levels in colon through microbiota modulation could serve as an innovative strategy for CRC prevention and treatment. This review highlights the critical relationship between gut microbiota and CRC, emphasizing the potential of targeting SCFAs to enhance gut health and reduce CRC risk.
Collapse
Affiliation(s)
- Boobalan Thulasinathan
- Department of Veterinary Medicine and Surgery, University of Missouri, Columbia, MO, USA
- Roy Blunt NextGen Precision Health Institute, University of Missouri, Columbia, MO, USA
| | - Kanve N. Suvilesh
- Roy Blunt NextGen Precision Health Institute, University of Missouri, Columbia, MO, USA
- Department of Surgery, Ellis Fischel Cancer Centre, University of Missouri, Columbia, MO, USA
- Harry S. Truman Memorial Veterans’ Hospital, Columbia, MO, USA
| | - Sumanas Maram
- Roy Blunt NextGen Precision Health Institute, University of Missouri, Columbia, MO, USA
| | - Erik Grossmann
- Department of Surgery, Ellis Fischel Cancer Centre, University of Missouri, Columbia, MO, USA
- Department of Medicine, Digestive Centre, Ellis Fischel Cancer Centre, University of Missouri, Columbia, MO, USA
| | - Yezaz Ghouri
- Department of Medicine, Digestive Centre, Ellis Fischel Cancer Centre, University of Missouri, Columbia, MO, USA
| | - Emma Pernas Teixeiro
- Roy Blunt NextGen Precision Health Institute, University of Missouri, Columbia, MO, USA
- Department of Molecular Microbiology and Immunology, University of Missouri, Columbia, MO, USA
| | - Joshua Chan
- Chemical and Biological Engineering, Colorado State University, Fort Collins, CO, USA
| | - Jussuf T. Kaif
- Department of Surgery, Ellis Fischel Cancer Centre, University of Missouri, Columbia, MO, USA
- Harry S. Truman Memorial Veterans’ Hospital, Columbia, MO, USA
- Siteman Cancer Centre, Washington University, St. Louis, MO, USA
| | - Satyanarayana Rachagani
- Department of Veterinary Medicine and Surgery, University of Missouri, Columbia, MO, USA
- Roy Blunt NextGen Precision Health Institute, University of Missouri, Columbia, MO, USA
- Department of Surgery, Ellis Fischel Cancer Centre, University of Missouri, Columbia, MO, USA
- Harry S. Truman Memorial Veterans’ Hospital, Columbia, MO, USA
- Siteman Cancer Centre, Washington University, St. Louis, MO, USA
| |
Collapse
|
2
|
Lemche E, Hortobágyi T, Kiecker C, Turkheimer F. Neuropathological links between T2DM and LOAD: systematic review and meta-analysis. Physiol Rev 2025; 105:1429-1486. [PMID: 40062731 DOI: 10.1152/physrev.00040.2024] [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: 10/01/2024] [Revised: 02/01/2025] [Accepted: 02/22/2025] [Indexed: 04/16/2025] Open
Abstract
Recent decades have described parallel neuropathological mechanisms increasing the risk for developing late-onset Alzheimer's dementia (LOAD) in type 2 diabetes mellitus (T2DM); however, still little is known of the role of diabetic encephalopathy and brain atrophy in LOAD. The aim of this systematic review is to provide a comprehensive view on diabetic encephalopathy/cerebral atrophy, taking into account neuroimaging data, neuropathology, metabolic and endocrine mechanisms, amyloid formation, brain perfusion impairments, neuroimmunology, and inflammasome activation. Key switches were identified, to further meta-analyze genomic candidate loci and epigenetic modifications. For the qualitative meta-analysis of genomic bases extracted, human linkage studies were examined; for epigenetic mechanisms, data from both human and animal studies are described. For the systematic review of pathophysiological mechanisms, 1,259 publications were evaluated and 93 gene loci extracted for candidate risk linkages. Sixty-six publications were evaluated for genomic association and descriptions of epigenomic modifications. Overall accumulated results highlight the insulin signaling system, vascular markers, inflammation and inflammasome pathways, amylin interactions, and glycosylation mechanisms. The protocol was registered with PROSPERO (ID: CRD42023440535).
Collapse
Affiliation(s)
- Erwin Lemche
- Section of Cognitive Neuropsychiatry, Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, United Kingdom
- Centre for Neuroimaging Sciences, Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, United Kingdom
| | - Tibor Hortobágyi
- Institute of Neuropathology, University Hospital Zurich, Zurich, Switzerland
- Department of Old Age Psychiatry, Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, United Kingdom
- Department of Neurology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Clemens Kiecker
- Department for Developmental Neurobiology, Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, United Kingdom
| | - Federico Turkheimer
- Centre for Neuroimaging Sciences, Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, United Kingdom
| |
Collapse
|
3
|
Piątkowska-Chmiel I, Wicha-Komsta K, Pawłowski K, Syrytczyk A, Kocki T, Dudka J, Herbet M. Beyond Diabetes: Semaglutide's Role in Modulating Mood Disorders through Neuroinflammation Pathways. Cell Mol Neurobiol 2025; 45:22. [PMID: 40059125 PMCID: PMC11891103 DOI: 10.1007/s10571-025-01534-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2024] [Accepted: 01/25/2025] [Indexed: 05/13/2025]
Abstract
Diabetes and mood disorders are intricately interconnected, with each condition elevating the risk of the other. This bidirectional relationship, further exacerbated by neuroinflammation, fosters an environment conducive to the development of anxiety and depression. Glucagon-like peptide-1 receptor agonists, such as semaglutide, offer promising therapeutic options that not only target type 2 diabetes but also can positively influence mood. Our study's primary goal was to evaluate the effectiveness of semaglutide, in mitigating anxiety and depression within an animal model of diabetes. The neuroprotective properties of semaglutide were evaluated by examining its influence on the kynurenine pathway and neurobiological markers (GFAP, NEFL, NSE, and GAL3) in the perfrontal cortex, selected for its key role in cognitive function and emotional regulation, impaired in diabetes and mood disorders. Additionally, we examined semaglutide's impact on peripheral inflammation and stress parameters to elucidate its role in modulating systemic inflammatory responses linked to mood disorders. Additionally, we conducted behavioral assessments to better understand how semaglutide influences anxiety and depression-related behaviors in diabetic mice. Semaglutide therapy significantly improved behavioral patterns and neurochemical markers in diabetic mice. The frequency of administration significantly influenced the outcomes, whereas the dosage appeared to have a limited impact. Here we show that semaglutide expands its therapeutic potential beyond diabetes, significantly influencing mood disorders through neuroinflammatory pathways. Semaglutide has the potential to be a key element in formulating integrated treatment strategies that address both metabolic health and mental well-being, ultimately enhancing the quality of life for individuals navigating these interrelated challenges.
Collapse
Affiliation(s)
- Iwona Piątkowska-Chmiel
- Department of Toxicology, Faculty of Pharmacy, Medical University of Lublin, 8B Jaczewskiego Street, 20-090, Lublin, Poland.
| | - Katarzyna Wicha-Komsta
- Department of Experimental and Clinical Pharmacology, Faculty of Medicine, Medical University of Lublin, Lublin, Poland
| | - Kamil Pawłowski
- Department of Toxicology, Faculty of Pharmacy, Medical University of Lublin, 8B Jaczewskiego Street, 20-090, Lublin, Poland
| | - Aleksandra Syrytczyk
- Department of Toxicology, Faculty of Pharmacy, Medical University of Lublin, 8B Jaczewskiego Street, 20-090, Lublin, Poland
| | - Tomasz Kocki
- Department of Experimental and Clinical Pharmacology, Faculty of Medicine, Medical University of Lublin, Lublin, Poland
| | - Jarosław Dudka
- Department of Toxicology, Faculty of Pharmacy, Medical University of Lublin, 8B Jaczewskiego Street, 20-090, Lublin, Poland
| | - Mariola Herbet
- Department of Toxicology, Faculty of Pharmacy, Medical University of Lublin, 8B Jaczewskiego Street, 20-090, Lublin, Poland
| |
Collapse
|
4
|
Tokgöz S, Boss M, Jansen TJ, Meijer R, Frielink C, van Bon AC, Tack CJ, de Galan BE, Gotthardt M. Activation of the HPA Axis Does Not Explain Nonresponsiveness to GLP-1R Agonist Treatment in Individuals With Type 2 Diabetes. Diabetes 2025; 74:212-222. [PMID: 39561332 PMCID: PMC11755685 DOI: 10.2337/db24-0463] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/04/2024] [Accepted: 11/13/2024] [Indexed: 11/21/2024]
Abstract
ARTICLE HIGHLIGHTS It is unclear why some individuals with type 2 diabetes are unresponsive to treatment with glucagon-like peptide 1 receptor (GLP-1R) agonists, but hypothalamic-pituitary-adrenal (HPA) axis activation could play a role. We used [68Ga]Ga-NODAGA-exendin-4 positron emission tomography/computed tomography to compare pituitary GLP-1R expression between responders and nonresponders to treatment with GLP-1R agonists. Pituitary GLP-1R expression and HPA axis activation did not differ between responders and nonresponders to GLP-1R agonist treatment. In addition, pituitary radiolabeled exendin uptake was markedly higher in men than in women. Further study is required to explain treatment differences and understand sex differences in pituitary radiolabeled exendin uptake.
Collapse
Affiliation(s)
- Sevilay Tokgöz
- Department of Medical Imaging, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Marti Boss
- Department of Medical Imaging, Radboud University Medical Center, Nijmegen, the Netherlands
- Netherlands Heart Institute, Utrecht, the Netherlands
| | - Theodorus J.P. Jansen
- Department of Medical Imaging, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Rick Meijer
- Department of Internal Medicine, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Cathelijne Frielink
- Department of Medical Imaging, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Arianne C. van Bon
- Department of Internal Medicine, Rijnstate Hospital, Arnhem, the Netherlands
| | - Cees J. Tack
- Department of Internal Medicine, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Bastiaan E. de Galan
- Department of Internal Medicine, Radboud University Medical Center, Nijmegen, the Netherlands
- Department of Internal Medicine, Maastricht University Medical Center, Maastricht, the Netherlands
- Cardiovascular Research Institute Maastricht School for Cardiovascular Disease, Maastricht University, Maastricht, the Netherlands
| | - Martin Gotthardt
- Department of Medical Imaging, Radboud University Medical Center, Nijmegen, the Netherlands
| |
Collapse
|
5
|
Holt MK, Valderrama N, Polanco MJ, Hayter I, Badenoch EG, Trapp S, Rinaman L. Modulation of stress-related behaviour by preproglucagon neurons and hypothalamic projections to the nucleus of the solitary tract. Mol Metab 2025; 91:102076. [PMID: 39603502 PMCID: PMC11667184 DOI: 10.1016/j.molmet.2024.102076] [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: 09/26/2024] [Revised: 11/12/2024] [Accepted: 11/20/2024] [Indexed: 11/29/2024] Open
Abstract
Stress-induced behaviours are driven by complex neural circuits and some neuronal populations concurrently modulate diverse behavioural and physiological responses to stress. Glucagon-like peptide-1 (GLP-1)-producing preproglucagon (PPG) neurons within the lower brainstem caudal nucleus of the solitary tract (cNTS) are particularly sensitive to stressful stimuli and are implicated in multiple physiological and behavioural responses to interoceptive and psychogenic threats. However, the afferent inputs driving stress-induced activation of PPG neurons are largely unknown, and the role of PPG neurons in anxiety-like behaviour is controversial. Through chemogenetic manipulations we reveal that cNTS PPG neurons have the ability to moderately increase anxiety-like behaviours in mice in a sex-dependent manner. Using an intersectional approach, we show that input from the paraventricular nucleus of the hypothalamus (PVN) drives activation of both the cNTS as a whole and PPG neurons in particular in response to acute restraint stress, but that while this input is rich in corticotropin-releasing hormone (CRH), PPG neurons do not express significant levels of receptors for CRH and are not activated following lateral ventricle delivery of CRH. Finally, we demonstrate that cNTS-projecting PVN neurons are necessary for the ability of restraint stress to suppress food intake in male mice. Our findings reveal sex differences in behavioural responses to PPG neural activation and highlight a hypothalamic-brainstem pathway in stress-induced hypophagia.
Collapse
Affiliation(s)
- Marie K Holt
- Florida State University, Department of Psychology and Program in Neuroscience, Tallahassee, FL, USA; Centre for Cardiovascular and Metabolic Neuroscience, Department of Neuroscience, Physiology & Pharmacology, UCL, London, UK; University of Warwick, School of Life Sciences, Coventry, UK.
| | - Natalia Valderrama
- Florida State University, Department of Psychology and Program in Neuroscience, Tallahassee, FL, USA
| | - Maria J Polanco
- Florida State University, Department of Psychology and Program in Neuroscience, Tallahassee, FL, USA
| | - Imogen Hayter
- Centre for Cardiovascular and Metabolic Neuroscience, Department of Neuroscience, Physiology & Pharmacology, UCL, London, UK; GlaxoSmithKline Pharmaceuticals, London, UK
| | | | - Stefan Trapp
- Centre for Cardiovascular and Metabolic Neuroscience, Department of Neuroscience, Physiology & Pharmacology, UCL, London, UK
| | - Linda Rinaman
- Florida State University, Department of Psychology and Program in Neuroscience, Tallahassee, FL, USA
| |
Collapse
|
6
|
Janket SJ, Chatanaka MK, Sohaei D, Tamimi F, Meurman JH, Diamandis EP. Does Incretin Agonism Have Sustainable Efficacy? Cells 2024; 13:1842. [PMID: 39594592 PMCID: PMC11592889 DOI: 10.3390/cells13221842] [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: 09/13/2024] [Revised: 10/23/2024] [Accepted: 10/29/2024] [Indexed: 11/28/2024] Open
Abstract
Recent clinical trials using synthetic incretin hormones, glucagon-like peptide 1 (GLP-1), and glucose-dependent insulinotropic polypeptide (GIP) receptor agonists have demonstrated that these treatments ameliorated many complications related to obesity, emphasizing the significant impact of body weight on overall health. Incretins are enteroendocrine hormones secreted by gut endothelial cells triggered by nutrient ingestion. The phenomenon that oral ingestion of glucose elicits a much higher insulin secretion than intra-venous injection of equimolar glucose is known as the incretin effect. This also alludes to the thesis that food intake is the root cause of insulin resistance. Synthetic GLP-1 and GIP agonists have demonstrated unprecedented glucoregulation and body weight reduction. Also, randomized trials have shown their ability to prevent complications of obesity, including development of diabetes from prediabetes, reducing cardiovascular disease risks and renal complications in diabetic patients. Moreover, the benefits of these agonists persist among the patients who are already on metformin or insulin. The ultimate question is "Are these benefits of incretin agonism sustainable?" Chronic agonism of pancreatic β-cells may decrease the number of receptors and cause β-cell exhaustion, leading to β-cell failure. Unfortunately, the long-term effects of these drugs are unknown at the present because the longest duration in randomized trials is 3 years. Additionally, manipulation of the neurohormonal axis to control satiety and food intake may hinder the long-term sustainability of these treatments. In this review, we will discuss the incretins' mechanism of action, challenges, and future directions. We will briefly review other molecules involved in glucose homeostasis such as amylin and glucagon. Amylin is co-expressed with insulin from the pancreas β-cells but does not have insulinotropic function. Amylin suppresses glucagon secretion, slowing gastric emptying and suppressing the reward center in the central nervous system, leading to weight loss. However, amylin can self-aggregate and cause serious cytotoxicity and may cause β-cell apoptosis. Glucagon is secreted by pancreatic α-cells and participates in glucose homeostasis in a glucose-dependent manner. In hypoglycemia, glucagon increases the blood glucose level by glycogenolysis and gluconeogenesis and inhibits glycogenesis in the liver. Several triple agonists, in combination with dual incretins and glucagon, are being developed.
Collapse
Affiliation(s)
- Sok-Ja Janket
- Retired Research Associate Professor, Boston University Goldman School of Dental Medicine, Boston, MA 02118, USA;
| | - Miyo K. Chatanaka
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON M5S 1A8, Canada;
| | - Dorsa Sohaei
- M.D., C.M. Candidate 2026, Faculty of Medicine and Health Sciences, McGill University, Montreal, QC H4A QT2, Canada;
| | - Faleh Tamimi
- Department of Restorative Dentistry, College of Dental Medicine, Qatar University, Doha P.O. Box 2713, Qatar;
| | - Jukka H. Meurman
- Department of Oral and Maxillofacial Diseases, Helsinki University Hospital and University of Helsinki, FI-00290 Helsinki, Finland;
| | | |
Collapse
|
7
|
Herman RJ, Schmidt HD. Targeting GLP-1 receptors to reduce nicotine use disorder: Preclinical and clinical evidence. Physiol Behav 2024; 281:114565. [PMID: 38663460 PMCID: PMC11128349 DOI: 10.1016/j.physbeh.2024.114565] [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: 01/31/2024] [Revised: 04/18/2024] [Accepted: 04/22/2024] [Indexed: 04/30/2024]
Abstract
Nicotine use disorder (NUD) remains a leading cause of preventable death in the U.S. Unfortunately, current FDA-approved pharmacotherapies for smoking cessation have limited efficacy and are associated with high rates of relapse. One major barrier to long-term smoking abstinence is body weight gain during withdrawal. Nicotine withdrawal-induced body weight gain can also lead to development of chronic disease states like obesity and type II diabetes mellitus. Therefore, it is critical to identify novel pharmacotherapies for NUD that decrease relapse and nicotine withdrawal symptoms including body weight gain. Recent studies demonstrate that glucagon-like peptide-1 receptor (GLP-1R) agonists attenuate voluntary nicotine taking and seeking and prevent withdrawal-induced hyperphagia and body weight gain. Emerging evidence also suggests that GLP-1R agonists improve cognitive deficits, as well as depressive- and anxiety-like behaviors, which contribute to smoking relapse during withdrawal. While further studies are necessary to fully characterize the effects of GLP-1R agonists on NUD and understand the mechanisms by which GLP-1R agonists decrease nicotine withdrawal-mediated behaviors, the current literature supports GLP-1R-based approaches to treating NUD.
Collapse
Affiliation(s)
- Rae J Herman
- Neuroscience Graduate Group, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Heath D Schmidt
- Department of Biobehavioral Health Sciences, School of Nursing, University of Pennsylvania, Philadelphia, PA, United States; Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States.
| |
Collapse
|
8
|
Lemche E, Killick R, Mitchell J, Caton PW, Choudhary P, Howard JK. Molecular mechanisms linking type 2 diabetes mellitus and late-onset Alzheimer's disease: A systematic review and qualitative meta-analysis. Neurobiol Dis 2024; 196:106485. [PMID: 38643861 DOI: 10.1016/j.nbd.2024.106485] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Revised: 03/18/2024] [Accepted: 03/23/2024] [Indexed: 04/23/2024] Open
Abstract
Research evidence indicating common metabolic mechanisms through which type 2 diabetes mellitus (T2DM) increases risk of late-onset Alzheimer's dementia (LOAD) has accumulated over recent decades. The aim of this systematic review is to provide a comprehensive review of common mechanisms, which have hitherto been discussed in separate perspectives, and to assemble and evaluate candidate loci and epigenetic modifications contributing to polygenic risk linkages between T2DM and LOAD. For the systematic review on pathophysiological mechanisms, both human and animal studies up to December 2023 are included. For the qualitative meta-analysis of genomic bases, human association studies were examined; for epigenetic mechanisms, data from human studies and animal models were accepted. Papers describing pathophysiological studies were identified in databases, and further literature gathered from cited work. For genomic and epigenomic studies, literature mining was conducted by formalised search codes using Boolean operators in search engines, and augmented by GeneRif citations in Entrez Gene, and other sources (WikiGenes, etc.). For the systematic review of pathophysiological mechanisms, 923 publications were evaluated, and 138 gene loci extracted for testing candidate risk linkages. 3 57 publications were evaluated for genomic association and descriptions of epigenomic modifications. Overall accumulated results highlight insulin signalling, inflammation and inflammasome pathways, proteolysis, gluconeogenesis and glycolysis, glycosylation, lipoprotein metabolism and oxidation, cell cycle regulation or survival, autophagic-lysosomal pathways, and energy. Documented findings suggest interplay between brain insulin resistance, neuroinflammation, insult compensatory mechanisms, and peripheral metabolic dysregulation in T2DM and LOAD linkage. The results allow for more streamlined longitudinal studies of T2DM-LOAD risk linkages.
Collapse
Affiliation(s)
- Erwin Lemche
- Section of Cognitive Neuropsychiatry and Centre for Neuroimaging Sciences, Institute of Psychiatry, Psychology & Neuroscience, King's College London, De Crespigny Park, London SE5 8AF, United Kingdom.
| | - Richard Killick
- Section of Old Age Psychiatry, Maurice Wohl Clinical Neuroscience Institute, Institute of Psychiatry, Psychology & Neuroscience, King's College London, De Crespigny Park, London SE5 8AF, United Kingdom
| | - Jackie Mitchell
- Department of Basic and Clinical Neurosciences, Maurice Wohl CIinical Neurosciences Institute, Institute of Psychiatry, Psychology & Neuroscience, King's College London, 125 Coldharbour Lane, London SE5 9NU, United Kingdom
| | - Paul W Caton
- Diabetes Research Group, School of Life Course Sciences, King's College London, Hodgkin Building, Guy's Campus, London SE1 1UL, United Kingdom
| | - Pratik Choudhary
- Diabetes Research Group, Weston Education Centre, King's College London, 10 Cutcombe Road, London SE5 9RJ, United Kingdom
| | - Jane K Howard
- School of Cardiovascular and Metabolic Medicine & Sciences, Hodgkin Building, Guy's Campus, King's College London, Great Maze Pond, London SE1 1UL, United Kingdom
| |
Collapse
|
9
|
Shankar K, Ramborger J, Bonnet-Zahedi S, Carrette LLG, George O. Acute nicotine intake increases feeding behavior through decreasing glucagon signaling in dependent male and female rats. Horm Behav 2024; 159:105447. [PMID: 37926623 PMCID: PMC11384237 DOI: 10.1016/j.yhbeh.2023.105447] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/21/2023] [Revised: 09/26/2023] [Accepted: 10/20/2023] [Indexed: 11/07/2023]
Abstract
Chronic use of nicotine is known to dysregulate metabolic signaling through altering circulating levels of feeding-related hormones, contributing to the onset of disorders like type 2 diabetes. However, little is known about the acute effects of nicotine on hormonal signaling. We previously identified an acute increase in food intake following acute nicotine, and we sought to determine whether this behavior was due to a change in hormone levels. We first identified that acute nicotine injection produces an increase in feeding behavior in dependent rats, but not nondependent rats. We confirmed that chronic nicotine use increases circulating levels of insulin, leptin, and ghrelin, and these correlate with rats' body weight and food intake. Acute nicotine injection in dependent animals decreased circulating GLP-1 and glucagon levels, and administration of glucagon prior to acute nicotine injection prevented the acute increase in feeding behavior. Thus, acute nicotine injection increases feeding behavior in dependent rats by decreasing glucagon signaling.
Collapse
Affiliation(s)
- Kokila Shankar
- Department of Psychiatry, University of California San Diego, La Jolla, CA 92093, USA; The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Jarryd Ramborger
- Department of Psychiatry, University of California San Diego, La Jolla, CA 92093, USA
| | - Sélène Bonnet-Zahedi
- Department of Psychiatry, University of California San Diego, La Jolla, CA 92093, USA; Institut de Neurosciences de la Timone, Aix-Marseille Université, Marseille 13005, France
| | - Lieselot L G Carrette
- Department of Psychiatry, University of California San Diego, La Jolla, CA 92093, USA
| | - Olivier George
- Department of Psychiatry, University of California San Diego, La Jolla, CA 92093, USA.
| |
Collapse
|
10
|
Fortin SM, Chen JC, Petticord MC, Ragozzino FJ, Peters JH, Hayes MR. The locus coeruleus contributes to the anorectic, nausea, and autonomic physiological effects of glucagon-like peptide-1. SCIENCE ADVANCES 2023; 9:eadh0980. [PMID: 37729419 PMCID: PMC10511187 DOI: 10.1126/sciadv.adh0980] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/09/2023] [Accepted: 08/21/2023] [Indexed: 09/22/2023]
Abstract
Increasing the therapeutic potential and reducing the side effects of U.S. Food and Drug Administration-approved glucagon-like peptide-1 receptor (GLP-1R) agonists used to treat obesity require complete characterization of the central mechanisms that mediate both the food intake-suppressive and illness-like effects of GLP-1R signaling. Our studies, in the rat, demonstrate that GLP-1Rs in the locus coeruleus (LC) are pharmacologically and physiologically relevant for food intake control. Furthermore, agonism of LC GLP-1Rs induces illness-like behaviors, and antagonism of LC GLP-1Rs can attenuate GLP-1R-mediated nausea. Electrophysiological and behavioral pharmacology data support a role for LC GLP-1Rs expressed on presynaptic glutamatergic terminals in the control of feeding and malaise. Collectively, our work establishes the LC as a site of action for GLP-1 signaling and extends our understanding of the GLP-1 signaling mechanism necessary for the development of improved obesity pharmacotherapies.
Collapse
Affiliation(s)
- Samantha M. Fortin
- Department of Psychiatry, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Jack C. Chen
- Department of Psychiatry, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Marisa C. Petticord
- Department of Psychiatry, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Forrest J. Ragozzino
- Department of Integrative Physiology and Neuroscience, Washington State University, Pullman, WA 99164, USA
| | - James H. Peters
- Department of Integrative Physiology and Neuroscience, Washington State University, Pullman, WA 99164, USA
| | - Matthew R. Hayes
- Department of Psychiatry, University of Pennsylvania, Philadelphia, PA 19104, USA
| |
Collapse
|
11
|
Le TDV, Fathi P, Watters AB, Ellis BJ, Besing GLK, Bozadjieva-Kramer N, Perez MB, Sullivan AI, Rose JP, Baggio LL, Koehler J, Brown JL, Bales MB, Nwaba KG, Campbell JE, Drucker DJ, Potthoff MJ, Seeley RJ, Ayala JE. Fibroblast growth factor-21 is required for weight loss induced by the glucagon-like peptide-1 receptor agonist liraglutide in male mice fed high carbohydrate diets. Mol Metab 2023; 72:101718. [PMID: 37030441 PMCID: PMC10131131 DOI: 10.1016/j.molmet.2023.101718] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/24/2023] [Revised: 03/20/2023] [Accepted: 03/29/2023] [Indexed: 04/10/2023] Open
Abstract
OBJECTIVE Glucagon-like peptide-1 receptor (GLP-1R) agonists (GLP-1RA) and fibroblast growth factor-21 (FGF21) confer similar metabolic benefits. GLP-1RA induce FGF21, leading us to investigate mechanisms engaged by the GLP-1RA liraglutide to increase FGF21 levels and the metabolic relevance of liraglutide-induced FGF21. METHODS Circulating FGF21 levels were measured in fasted male C57BL/6J, neuronal GLP-1R knockout, β-cell GLP-1R knockout, and liver peroxisome proliferator-activated receptor alpha knockout mice treated acutely with liraglutide. To test the metabolic relevance of liver FGF21 in response to liraglutide, chow-fed control and liver Fgf21 knockout (LivFgf21-/-) mice were treated with vehicle or liraglutide in metabolic chambers. Body weight and composition, food intake, and energy expenditure were measured. Since FGF21 reduces carbohydrate intake, we measured body weight in mice fed matched diets with low- (LC) or high-carbohydrate (HC) content and in mice fed a high-fat, high-sugar (HFHS) diet. This was done in control and LivFgf21-/- mice and in mice lacking neuronal β-klotho (Klb) expression to disrupt brain FGF21 signaling. RESULTS Liraglutide increases FGF21 levels independently of decreased food intake via neuronal GLP-1R activation. Lack of liver Fgf21 expression confers resistance to liraglutide-induced weight loss due to attenuated reduction of food intake in chow-fed mice. Liraglutide-induced weight loss was impaired in LivFgf21-/- mice when fed HC and HFHS diets but not when fed a LC diet. Loss of neuronal Klb also attenuated liraglutide-induced weight loss in mice fed HC or HFHS diets. CONCLUSIONS Our findings support a novel role for a GLP-1R-FGF21 axis in regulating body weight in a dietary carbohydrate-dependent manner.
Collapse
Affiliation(s)
- Thao D V Le
- Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, 2215 Garland Avenue, Nashville, TN 37232, USA.
| | - Payam Fathi
- Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, 2215 Garland Avenue, Nashville, TN 37232, USA.
| | - Amanda B Watters
- Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, 2215 Garland Avenue, Nashville, TN 37232, USA.
| | - Blair J Ellis
- Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, 2215 Garland Avenue, Nashville, TN 37232, USA
| | - Gai-Linn K Besing
- Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, 2215 Garland Avenue, Nashville, TN 37232, USA.
| | - Nadejda Bozadjieva-Kramer
- Department of Surgery, University of Michigan, 1500 E. Medical Center Drive, Ann Arbor, MI 48109, USA; Veterans Affairs Ann Arbor Healthcare System, Research Service, 2215 Fuller Road, Ann Arbor, MI 48105, USA.
| | - Misty B Perez
- Department of Neuroscience and Pharmacology, University of Iowa Carver College of Medicine, 375 Newton Road, Iowa City, IA 52242, USA.
| | - Andrew I Sullivan
- Department of Neuroscience and Pharmacology, University of Iowa Carver College of Medicine, 375 Newton Road, Iowa City, IA 52242, USA.
| | - Jesse P Rose
- Department of Neuroscience and Pharmacology, University of Iowa Carver College of Medicine, 375 Newton Road, Iowa City, IA 52242, USA.
| | - Laurie L Baggio
- Lunenfeld Tanenbaum Research Institute, Mount Sinai Hospital, Department of Medicine, University of Toronto, 600 University Avenue, Toronto, Ontario M5G 1X5, Canada.
| | - Jacqueline Koehler
- Lunenfeld Tanenbaum Research Institute, Mount Sinai Hospital, Department of Medicine, University of Toronto, 600 University Avenue, Toronto, Ontario M5G 1X5, Canada
| | - Jennifer L Brown
- Duke Molecular Physiology Institute, Duke University, 300 N. Duke Street, Durham, NC 27701, USA
| | - Michelle B Bales
- Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, 2215 Garland Avenue, Nashville, TN 37232, USA.
| | - Kaitlyn G Nwaba
- Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, 2215 Garland Avenue, Nashville, TN 37232, USA
| | - Jonathan E Campbell
- Duke Molecular Physiology Institute, Duke University, 300 N. Duke Street, Durham, NC 27701, USA.
| | - Daniel J Drucker
- Lunenfeld Tanenbaum Research Institute, Mount Sinai Hospital, Department of Medicine, University of Toronto, 600 University Avenue, Toronto, Ontario M5G 1X5, Canada.
| | - Matthew J Potthoff
- Department of Neuroscience and Pharmacology, University of Iowa Carver College of Medicine, 375 Newton Road, Iowa City, IA 52242, USA.
| | - Randy J Seeley
- Department of Surgery, University of Michigan, 1500 E. Medical Center Drive, Ann Arbor, MI 48109, USA.
| | - Julio E Ayala
- Department of Molecular Physiology and Biophysics, 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; Vanderbilt Center for Addiction Research, Vanderbilt University School of Medicine, 2215 Garland Avenue, Nashville, TN 37232, USA.
| |
Collapse
|
12
|
Le TDV, Fathi P, Watters AB, Ellis BJ, Bozadjieva-Kramer N, Perez MB, Sullivan AI, Rose JP, Baggio LL, Koehler J, Brown JL, Bales MB, Nwaba KG, Campbell JE, Drucker DJ, Potthoff MJ, Seeley RJ, Ayala JE. Liver Fibroblast Growth Factor 21 (FGF21) is Required for the Full Anorectic Effect of the Glucagon-Like Peptide-1 Receptor Agonist Liraglutide in Male Mice fed High Carbohydrate Diets. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.01.03.522509. [PMID: 36711605 PMCID: PMC9881863 DOI: 10.1101/2023.01.03.522509] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/15/2023]
Abstract
Glucagon-like peptide-1 receptor (GLP-1R) agonists and fibroblast growth factor 21 (FGF21) confer similar metabolic benefits. Studies report that GLP-1RA induce FGF21. Here, we investigated the mechanisms engaged by the GLP-1R agonist liraglutide to increase FGF21 levels and the metabolic relevance of liraglutide-induced FGF21. We show that liraglutide increases FGF21 levels via neuronal GLP-1R activation. We also demonstrate that lack of liver Fgf21 expression confers partial resistance to liraglutide-induced weight loss. Since FGF21 reduces carbohydrate intake, we tested whether the contribution of FGF21 to liraglutide-induced weight loss is dependent on dietary carbohydrate content. In control and liver Fgf21 knockout (Liv Fgf21 -/- ) mice fed calorically matched diets with low- (LC) or high-carbohydrate (HC) content, we found that only HC-fed Liv Fgf21 -/- mice were resistant to liraglutide-induced weight loss. Similarly, liraglutide-induced weight loss was partially impaired in Liv Fgf21 -/- mice fed a high-fat, high-sugar (HFHS) diet. Lastly, we show that loss of neuronal β-klotho expression also diminishes liraglutide-induced weight loss in mice fed a HC or HFHS diet, indicating that FGF21 mediates liraglutide-induced weight loss via neuronal FGF21 action. Our findings support a novel role for a GLP-1R-FGF21 axis in regulating body weight in the presence of high dietary carbohydrate content.
Collapse
|
13
|
Akalestou E, Lopez-Noriega L, Christakis I, Hu M, Miras AD, Leclerc I, Rutter GA. Vertical sleeve gastrectomy normalizes circulating glucocorticoid levels and lowers glucocorticoid action tissue-selectively in mice. Front Endocrinol (Lausanne) 2022; 13:1020576. [PMID: 36246869 PMCID: PMC9556837 DOI: 10.3389/fendo.2022.1020576] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/16/2022] [Accepted: 09/08/2022] [Indexed: 11/13/2022] Open
Abstract
Objectives Glucocorticoids produced by the adrenal cortex are essential for the maintenance of metabolic homeostasis. Glucocorticoid activation is catalysed by 11β-hydroxysteroid dehydrogenase 1 (11β-HSD1). Excess glucocorticoids are associated with insulin resistance and hyperglycaemia. A small number of studies have demonstrated effects on glucocorticoid metabolism of bariatric surgery, a group of gastrointestinal procedures known to improve insulin sensitivity and secretion, which were assumed to result from weight loss. In this study, we hypothesize that a reduction in glucocorticoid action following bariatric surgery contributes to the widely observed euglycemic effects of the treatment. Methods Glucose and insulin tolerance tests were performed at ten weeks post operatively and circulating corticosterone was measured. Liver and adipose tissues were harvested from fed mice and 11β-HSD1 levels were measured by quantitative RT-PCR or Western (immuno-) blotting, respectively. 11β-HSD1 null mice (Hsd11b1 -/-) were generated using CRISPR/Cas9 genome editing. Wild type and littermate Hsd11b1 -/- mice underwent Vertical Sleeve Gastrectomy (VSG) or sham surgery. Results Under the conditions used, no differences in weight loss were observed between VSG treated and sham operated mice. However, both lean and obese WT VSG mice displayed significantly improved glucose clearance and insulin sensitivity. Remarkably, VSG restored physiological corticosterone production in HFD mice and reduced 11β-HSD1 expression in liver and adipose tissue post-surgery. Elimination of the 11β-HSD1/Hsd11b1 gene by CRISPR/Cas9 mimicked the effects of VSG on body weight and tolerance to 1g/kg glucose challenge. However, at higher glucose loads, the euglycemic effect of VSG was superior to Hsd11b1 elimination. Conclusions Bariatric surgery improves insulin sensitivity and reduces glucocorticoid activation at the tissular level, under physiological and pathophysiological (obesity) conditions, irrespective of weight loss. These findings point towards a physiologically relevant gut-glucocorticoid axis, and suggest that lowered glucocorticoid exposure may represent an additional contribution to the health benefits of bariatric surgery.
Collapse
Affiliation(s)
- Elina Akalestou
- Section of Cell Biology and Functional Genomics, Department of Metabolism, Digestion and Reproduction, Imperial College London, London, United Kingdom
| | - Livia Lopez-Noriega
- Section of Cell Biology and Functional Genomics, Department of Metabolism, Digestion and Reproduction, Imperial College London, London, United Kingdom
| | - Ioannis Christakis
- Endocrine and General Surgery, Nottingham University Hospitals NHS Trust, Nottingham, United Kingdom
| | - Ming Hu
- Section of Cell Biology and Functional Genomics, Department of Metabolism, Digestion and Reproduction, Imperial College London, London, United Kingdom
| | - Alexander D. Miras
- Section of Investigative Medicine, Department of Metabolism, Digestion and Reproduction, Imperial College London, London, United Kingdom
| | - Isabelle Leclerc
- Section of Cell Biology and Functional Genomics, Department of Metabolism, Digestion and Reproduction, Imperial College London, London, United Kingdom
- Centre de Recherches du CHUM, University of Montreal, Montreal, QC, Canada
| | - Guy A. Rutter
- Section of Cell Biology and Functional Genomics, Department of Metabolism, Digestion and Reproduction, Imperial College London, London, United Kingdom
- Centre de Recherches du CHUM, University of Montreal, Montreal, QC, Canada
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Singapore
| |
Collapse
|
14
|
Bolus Injection of Liraglutide Raises Plasma Glucose in Normal Rats by Activating Glucagon-like Peptide 1 Receptor in the Brain. Pharmaceuticals (Basel) 2022; 15:ph15070904. [PMID: 35890201 PMCID: PMC9320491 DOI: 10.3390/ph15070904] [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: 06/05/2022] [Revised: 07/14/2022] [Accepted: 07/18/2022] [Indexed: 11/16/2022] Open
Abstract
Diabetes is commonly treated with glucagon-like peptide-1 receptor (GLP-1R) agonists including liraglutide and others. However, liraglutide was found to raise plasma glucose levels in normal rats. The current study aims to determine how liraglutide causes this contentious condition in rats, both normal and diabetic. An adrenalectomy was performed to investigate the relationship between steroid hormone and liraglutide. To investigate the effect of central liraglutide infusion on blood glucose in rats, rats were intracerebroventricularly administrated with liraglutide with or without HPA axis inhibitors such as berberine and dexamethasone. The results showed that a single injection of liraglutide caused a temporary increase in blood glucose in healthy rats. Another GLP-1R agonist, Exendin-4 (Ex-4), increased blood sugar in a manner similar to that of liraglutide. The effects of liraglutide were also blocked by guanethidine pretreatment and vanished in normal rats with adrenalectomy. Additionally, central infusion of liraglutide via intracerebroventricular (icv) injection into normal rats also causes a temporary increase in blood glucose that was blocked by GLP-1R antagonists or the inhibitors such as berberine and dexamethasone. Similarly, central liraglutide treatment causes temporary increases in plasma glucose, adrenocorticotropic hormone (ACTH), and cortisol levels, which were reversed by inhibitors for the hypothalamic-pituitary-adrenal (HPA) axis. In normal rats, the temporary glucose-increasing effect of liraglutide was gradually eliminated during consecutive daily treatments, indicating tolerance formation. Additionally, liraglutide and Ex-4 cross-tolerance was also discovered in normal rats. Liraglutide was more effective in diabetic rats than in normal rats in activating GLP-1R gene expression in the isolated adrenal gland. Interestingly, the effect of liraglutide on glycemic control varied depending on whether the rats were diabetic or not. In normal rats, bolus injection of liraglutide, such as Ex-4, may stimulate the HPA axis, resulting in hyperglycemia. The cross-tolerance of liraglutide and Ex-4 provided a novel perspective on GLP-1R activation.
Collapse
|
15
|
Patel H. The role of the lateral septum in neuropsychiatric disease. J Neurosci Res 2022; 100:1422-1437. [PMID: 35443088 DOI: 10.1002/jnr.25052] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2021] [Revised: 02/22/2022] [Accepted: 03/23/2022] [Indexed: 12/25/2022]
Abstract
The lateral septum (LS) is a structure in the midline of the brain that is interconnected with areas associated with stress and feeding. This review highlights the role of the LS in anxiety, depression, and eating disorders and their comorbidity. There is a prevailing view that the LS is anxiolytic. This review finds that the LS is both anxiolytic and anxiogenic. Furthermore, the LS can promote and inhibit feeding. Given these shared roles, the LS represents a common site for the comorbidity of neuropsychiatric disorders, and therefore a potential pharmacological target. This is crucial since currently available treatments are not always effective. Corticotrophin-releasing factor 2 antagonists are potential drugs for the treatment of anxiety and anorexia and require further research. Furthermore, other drugs currently in trials for binge eating, such as alpha-adrenergic agonists, may in fact promote food intake. It is hoped that the advancements in chemo- and optogenetic techniques will allow future studies to profile the specific neural connections of the LS and their function. This information could facilitate our understanding of the underlying mechanisms, and therefore pharmacological targets, of these psychiatric conditions.
Collapse
|
16
|
O'Riordan KJ, Collins MK, Moloney GM, Knox EG, Aburto MR, Fülling C, Morley SJ, Clarke G, Schellekens H, Cryan JF. Short chain fatty acids: Microbial metabolites for gut-brain axis signalling. Mol Cell Endocrinol 2022; 546:111572. [PMID: 35066114 DOI: 10.1016/j.mce.2022.111572] [Citation(s) in RCA: 220] [Impact Index Per Article: 73.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/14/2021] [Revised: 01/07/2022] [Accepted: 01/11/2022] [Indexed: 02/08/2023]
Abstract
The role of the intestinal microbiota as a regulator of gut-brain axis signalling has risen to prominence in recent years. Understanding the relationship between the gut microbiota, the metabolites it produces, and the brain will be critical for the subsequent development of new therapeutic approaches, including the identification of novel psychobiotics. A key focus in this regard have been the short-chain fatty acids (SCFAs) produced by bacterial fermentation of dietary fibre, which include butyrate, acetate, and propionate. Ongoing research is focused on the entry of SCFAs into systemic circulation from the gut lumen, their migration to cerebral circulation and across the blood brain barrier, and their potential to exert acute and chronic effects on brain structure and function. This review aims to discuss our current mechanistic understanding of the direct and indirect influence that SCFAs have on brain function, behaviour and physiology, which will inform future microbiota-targeted interventions for brain disorders.
Collapse
Affiliation(s)
| | - Michael K Collins
- APC Microbiome Ireland, University College Cork, Ireland; Department of Anatomy & Neuroscience, University College Cork, Ireland
| | - Gerard M Moloney
- APC Microbiome Ireland, University College Cork, Ireland; Department of Anatomy & Neuroscience, University College Cork, Ireland
| | - Emily G Knox
- APC Microbiome Ireland, University College Cork, Ireland; School of Pharmacy, University College Cork, Ireland
| | - María R Aburto
- APC Microbiome Ireland, University College Cork, Ireland
| | | | - Shane J Morley
- APC Microbiome Ireland, University College Cork, Ireland
| | - Gerard Clarke
- APC Microbiome Ireland, University College Cork, Ireland; Department of Psychiatry and Neurobehavioural Science, University College Cork, Cork, Ireland
| | - Harriët Schellekens
- APC Microbiome Ireland, University College Cork, Ireland; Department of Anatomy & Neuroscience, University College Cork, Ireland
| | - John F Cryan
- APC Microbiome Ireland, University College Cork, Ireland; Department of Anatomy & Neuroscience, University College Cork, Ireland.
| |
Collapse
|
17
|
Graham DL, Madkour HS, Noble BL, Schatschneider C, Stanwood GD. Long-term functional alterations following prenatal GLP-1R activation. Neurotoxicol Teratol 2021; 87:106984. [PMID: 33864929 PMCID: PMC8555578 DOI: 10.1016/j.ntt.2021.106984] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2020] [Revised: 03/29/2021] [Accepted: 04/09/2021] [Indexed: 12/11/2022]
Abstract
Evidence supporting the use of glucagon-like peptide-1 (GLP-1) analogues to pharmacologically treat disorders beyond type 2 diabetes and obesity is increasing. However, little is known about how activation of the GLP-1 receptor (GLP-1R) during pregnancy affects maternal and offspring outcomes. We treated female C57Bl/6 J mice prior to conception and throughout gestation with a long-lasting GLP-1R agonist, Exendin-4. While GLP-1R activation has significant effects on food and drug reward, depression, locomotor activity, and cognition in adults, we found few changes in these domains in exendin-4-exposed offspring. Repeated injections of Exendin-4 had minimal effects on the dams and may have enhanced maternal care. Offspring exposed to the drug weighed significantly more than their control counterparts during the preweaning period and demonstrated alterations in anxiety-like outcomes, which indicate a developmental role for GLP-1R modulation in the stress response that may be sex-specific.
Collapse
Affiliation(s)
- Devon L Graham
- Department of Biomedical Sciences and Center for Brain Repair, Florida State University College of Medicine, Tallahassee, FL 32306, United States of America
| | - Haley S Madkour
- Department of Biomedical Sciences and Center for Brain Repair, Florida State University College of Medicine, Tallahassee, FL 32306, United States of America
| | - Brenda L Noble
- Department of Biomedical Sciences and Center for Brain Repair, Florida State University College of Medicine, Tallahassee, FL 32306, United States of America
| | - Chris Schatschneider
- Department of Psychology, Florida State University, Tallahassee, FL 32306, United States of America
| | - Gregg D Stanwood
- Department of Biomedical Sciences and Center for Brain Repair, Florida State University College of Medicine, Tallahassee, FL 32306, United States of America.
| |
Collapse
|
18
|
Heinla K, Vasar E, Sedman T, Volke V. A GLP-1 Receptor Agonist Inhibits Aldosterone Release in Healthy Volunteers. Horm Metab Res 2021; 53:402-407. [PMID: 34154031 DOI: 10.1055/a-1498-7098] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Glucagon-like peptide 1 receptor agonists (GLP-1 RAs) are antidiabetic drugs with effects beyond antihyperglycemic action. The aim of the study was to examine whether a single dose of exenatide could be used as a stimulation test for the pituitary-adrenal axis. We carried out a single-group, open-label pilot clinical trial in an ambulatory setting. Ten healthy volunteers of both sexes with body weight>65 kg and age between 18-50 years were recruited. After fasting for 12 hours the subjects received 10 μg of exenatide solution subcutaneously. Blood samples were taken before the administration of exenatide and up to 150 minutes thereafter. The primary outcome was the maximal level of cortisol after the administration of exenatide. Single administration of exenatide 10 μg resulted in a modest increase in ACTH and cortisol levels, as compared to untreated values, and a decrease in blood glucose levels. Remarkably, a robust suppression of both renin and aldosterone levels occurred. We showed that acute administration of exenatide in a full therapeutic dose modestly stimulates the hypothalamic-pituitary-adrenal axis but inhibits the renin-aldosterone system. Further research is warranted to confirm this finding in the placebo-controlled study.
Collapse
Affiliation(s)
- Keiu Heinla
- Department of Physiology, Institute of Biomedicine and Translational Medicine, University of Tartu, Tartu, Estonia
| | - Eero Vasar
- Department of Physiology, Institute of Biomedicine and Translational Medicine, University of Tartu, Tartu, Estonia
| | - Tuuli Sedman
- Department of Physiology, Institute of Biomedicine and Translational Medicine, University of Tartu, Tartu, Estonia
| | - Vallo Volke
- Department of Physiology, Institute of Biomedicine and Translational Medicine, University of Tartu, Tartu, Estonia
- Endocrinology Unit, Tartu University Hospital, Tartu, Estonia
| |
Collapse
|
19
|
Holt MK, Rinaman L. The role of nucleus of the solitary tract glucagon-like peptide-1 and prolactin-releasing peptide neurons in stress: anatomy, physiology and cellular interactions. Br J Pharmacol 2021; 179:642-658. [PMID: 34050926 PMCID: PMC8820208 DOI: 10.1111/bph.15576] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2021] [Revised: 05/04/2021] [Accepted: 05/17/2021] [Indexed: 02/06/2023] Open
Abstract
Neuroendocrine, behavioural and autonomic responses to stressful stimuli are orchestrated by complex neural circuits. The caudal nucleus of the solitary tract (cNTS) in the dorsomedial hindbrain is uniquely positioned to integrate signals of both interoceptive and psychogenic stress. Within the cNTS, glucagon‐like peptide‐1 (GLP‐1) and prolactin‐releasing peptide (PrRP) neurons play crucial roles in organising neural responses to a broad range of stressors. In this review we discuss the anatomical and functional overlap between PrRP and GLP‐1 neurons. We outline their co‐activation in response to stressful stimuli and their importance as mediators of behavioural and physiological stress responses. Finally, we review evidence that PrRP neurons are downstream of GLP‐1 neurons and outline unexplored areas of the research field. Based on the current state‐of‐knowledge, PrRP and GLP‐1 neurons may be compelling targets in the treatment of stress‐related disorders.
Collapse
Affiliation(s)
- Marie K Holt
- Department of Neuroscience, Physiology and Pharmacology, University College London, London, UK
| | - Linda Rinaman
- Department of Psychology and Program in Neuroscience, Florida State University, Tallahassee, Florida, USA
| |
Collapse
|
20
|
Detka J, Głombik K. Insights into a possible role of glucagon-like peptide-1 receptor agonists in the treatment of depression. Pharmacol Rep 2021; 73:1020-1032. [PMID: 34003475 PMCID: PMC8413152 DOI: 10.1007/s43440-021-00274-8] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2021] [Revised: 05/04/2021] [Accepted: 05/06/2021] [Indexed: 01/23/2023]
Abstract
Depression is a highly prevalent mood disorder and one of the major health concerns in modern society. Moreover, it is characterized by a high prevalence of coexistence with many other diseases including metabolic disorders such as type 2 diabetes mellitus (T2DM) and obesity. Currently used antidepressant drugs, which mostly target brain monoaminergic neurotransmission, have limited clinical efficacy. Although the etiology of depression has not been fully elucidated, current scientific data emphasize the role of neurotrophic factors deficiencies, disturbed homeostasis between the nervous system and the immune and endocrine systems, as well as disturbances in brain energy metabolism and dysfunctions in the gut-brain axis as important factors in the pathogenesis of this neuropsychiatric disorder. Therefore, therapeutic options that could work in a way other than classic antidepressants are being sought to increase the effectiveness of the treatment. Interestingly, glucagon-like peptide-1 receptor agonists (GLP-1RAs), used in the treatment of T2DM and obesity, are known to show pro-cognitive and neuroprotective properties, and exert modulatory effects on immune, endocrine and metabolic processes in the central nervous system. This review article discusses the potential antidepressant effects of GLP-1RAs, especially in the context of their action on the processes related to neuroprotection, inflammation, stress response, energy metabolism, gut-brain crosstalk and the stability of the gut microbiota.
Collapse
Affiliation(s)
- Jan Detka
- Laboratory of Immunoendocrinology, Department of Experimental Neuroendocrinology, Polish Academy of Sciences, Maj Institute of Pharmacology, 12 Smętna Street, 31-343, Cracow, Poland.
| | - Katarzyna Głombik
- Laboratory of Immunoendocrinology, Department of Experimental Neuroendocrinology, Polish Academy of Sciences, Maj Institute of Pharmacology, 12 Smętna Street, 31-343, Cracow, Poland
| |
Collapse
|
21
|
Leon RM, Borner T, Stein LM, Urrutia NA, De Jonghe BC, Schmidt HD, Hayes MR. Activation of PPG neurons following acute stressors differentially involves hindbrain serotonin in male rats. Neuropharmacology 2021; 187:108477. [PMID: 33581143 DOI: 10.1016/j.neuropharm.2021.108477] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2020] [Revised: 01/19/2021] [Accepted: 01/23/2021] [Indexed: 12/16/2022]
Abstract
Within the hindbrain, serotonin (5-HT) functions as a modulator of the central glucagon-like peptide-1 (GLP-1) system. This interaction between 5-HT and GLP-1 is achieved via 5-HT2C and 5-HT3 receptors and is relevant for GLP-1-mediated feeding behavior. The central GLP-1 system is activated by various stressors, activates the hypothalamic pituitary adrenocortical (HPA) axis, and contributes to stress-related behaviors. Whether 5-HT modulates GLP-1's role in the stress response in unknown. We hypothesized that the serotonergic modulation of GLP-1-producing neurons (i.e., PPG neurons) is stimuli-specific and that stressed-induced PPG activity is one of the modalities in which 5-HT plays a role. In this study, we investigated the roles of 5-HT2C and 5-HT3 receptors in mediating the activation of PPG neurons in the nucleus tractus solitarius (NTS) following exposure to three different acute stressors: lithium chloride (LiCl), noncontingent cocaine (Coc), and novel restraint stress (RES). Results showed that increased c-Fos expression in PPG neurons following LiCl and RES-but not Coc-is dependent on hindbrain 5-HT2C and 5-HT3 receptor signaling. Additionally, stressors that depend on 5-HT signaling to activate PPG neurons (i.e., LiCl and RES) increased c-Fos expression in 5-HT-expressing neurons within the caudal raphe (CR), specifically in the raphe magnus (RMg). Finally, we showed that RMg neurons innervate NTS PPG neurons and that some of these PPG neurons lie in close proximity to 5-HT axons, suggesting RMg 5-HT-expressing neurons are the source of 5-HT input responsible for engaging NTS PPG neurons. Together, these findings identify a direct RMg to NTS pathway responsible for the modulatory effect of 5-HT on the central GLP-1 system-specifically via activation of 5-HT2C and 5-HT3 receptors-in the facilitation of acute stress responses.
Collapse
Affiliation(s)
- Rosa M Leon
- Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Tito Borner
- Department of Biobehavioral Health Sciences, School of Nursing, University of Pennsylvania, Philadelphia, PA, USA
| | - Lauren M Stein
- Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Norma A Urrutia
- Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Bart C De Jonghe
- Department of Biobehavioral Health Sciences, School of Nursing, University of Pennsylvania, Philadelphia, PA, USA
| | - Heath D Schmidt
- Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA; Department of Biobehavioral Health Sciences, School of Nursing, University of Pennsylvania, Philadelphia, PA, USA
| | - Matthew R Hayes
- Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA; Department of Biobehavioral Health Sciences, School of Nursing, University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
22
|
Panagiotou C, Lambadiari V, Maratou E, Geromeriati C, Artemiadis A, Dimitriadis G, Moutsatsou P. Insufficient glucocorticoid receptor signaling and flattened salivary cortisol profile are associated with metabolic and inflammatory indices in type 2 diabetes. J Endocrinol Invest 2021; 44:37-48. [PMID: 32394161 DOI: 10.1007/s40618-020-01260-2] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/01/2020] [Accepted: 04/15/2020] [Indexed: 12/22/2022]
Abstract
PURPOSE Impaired negative feedback and hyperactivation of the hypothalamic-pituitary-adrenal (HPA) axis characterizes type 2 diabetes mellitus (T2DM). The glucocorticoid receptor (GR) is a key mediator of HPA axis negative feedback; however, its role in linking hypercortisolemia and T2DM-associated hyperglycemia, hyperlipidemia and inflammation is not yet known. METHODS In peripheral mononuclear cells (PBMC) from 31 T2DM patients and 24 healthy controls, we measured various GR-signaling parameters such as phosphorylated GR (pGR-S211), GRα/GRβ gene expression and GC-sensitivity [using the basal and dexamethasone (DEX)-induced leucine zipper (GILZ) and FK506 binding-protein (FKBP5) mRNA levels as well as the basal interleukin (IL)-1β protein levels]. Diurnal salivary cortisol curve parameters such as the cortisol awaking response (CAR) and area under the curve (AUCtotal and AUCi) as well as inflammatory and metabolic indices were also determined. RESULTS T2DM patients exhibited diminished pGR-S211 protein content, increased GRβ, decreased basal GILZ and FKBP5 mRNA levels and increased IL-1β levels. Flattened DEX-induced GILZ and FKBP5 response curves and a flattened salivary cortisol profile characterized T2DM patients. Significant associations of GR measures and saliva cortisol curve parameters with biochemical and clinical characteristics were found. CONCLUSION Our novel data implicate an insufficient GR signaling in PBMCs in T2DM patients and HPA axis dysfunction. The significant associations of GR-signaling parameters with inflammatory and metabolic indices implicate that GR may be the critical link between HPA axis dysfunction, hypercortisolemia and diabetes-associated metabolic disturbances. Our findings provide significant insights into the contribution of GR-mediated mechanisms in T2DM aetiopathology and therapy.
Collapse
Affiliation(s)
- C Panagiotou
- Department of Clinical Biochemistry, National and Kapodistrian University of Athens, School of Medicine, University General Hospital Attikon, Rimini 1, Haidari, 12462, Athens, Greece
| | - V Lambadiari
- Second Department of Internal Medicine and Research Institute, University General Hospital Attikon, Haidari, Greece
| | - E Maratou
- Department of Clinical Biochemistry, National and Kapodistrian University of Athens, School of Medicine, University General Hospital Attikon, Rimini 1, Haidari, 12462, Athens, Greece
| | - C Geromeriati
- Department of Clinical Biochemistry, National and Kapodistrian University of Athens, School of Medicine, University General Hospital Attikon, Rimini 1, Haidari, 12462, Athens, Greece
| | - A Artemiadis
- Medical School, University of Cyprus, Nicosia, Cyprus
| | - G Dimitriadis
- Second Department of Internal Medicine and Research Institute, University General Hospital Attikon, Haidari, Greece
| | - P Moutsatsou
- Department of Clinical Biochemistry, National and Kapodistrian University of Athens, School of Medicine, University General Hospital Attikon, Rimini 1, Haidari, 12462, Athens, Greece.
| |
Collapse
|
23
|
Kong Q, Li Y, Yue J, Wu X, Xu M. Reducing alcohol and/or cocaine-induced reward and toxicity via an epidermal stem cell-based gene delivery platform. Mol Psychiatry 2021; 26:5266-5276. [PMID: 33619338 PMCID: PMC8380265 DOI: 10.1038/s41380-021-01043-y] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/19/2020] [Revised: 01/14/2021] [Accepted: 02/01/2021] [Indexed: 01/31/2023]
Abstract
Alcohol use disorder (AUD) is one of the foremost public health problems. Alcohol is also frequently co-abused with cocaine. There is a huge unmet need for the treatment of AUD and/or cocaine co-abuse. We recently demonstrated that skin grafts generated from mouse epidermal stem cells that had been engineered by CRISPR-mediated genome editing could be transplanted onto mice as a gene delivery platform. Here, we show that expression of the glucagon-like peptide-1 (GLP1) gene delivered by epidermal stem cells attenuated development and reinstatement of alcohol-induced drug-taking and seeking as well as voluntary oral alcohol consumption. GLP1 derived from the skin grafts decreased alcohol-induced increase in dopamine levels in the nucleus accumbens. In exploring the potential of this platform in reducing concurrent use of drugs, we developed a novel co-grafting procedure for both modified human butyrylcholinesterase (hBChE)- and GLP1-expressing cells. Epidermal stem cell-derived hBChE and GLP1 reduced acquisition of drug-taking and toxicity induced by alcohol and cocaine co-administration. These results imply that cutaneous gene delivery through skin transplants may add a new option to treat drug abuse and co-abuse.
Collapse
Affiliation(s)
- Qingyao Kong
- grid.170205.10000 0004 1936 7822Department of Anesthesia and Critical Care, The University of Chicago, Chicago, IL USA
| | - Yuanyuan Li
- grid.170205.10000 0004 1936 7822Ben May Department for Cancer Research, The University of Chicago, Chicago, IL USA
| | - Jiping Yue
- grid.170205.10000 0004 1936 7822Ben May Department for Cancer Research, The University of Chicago, Chicago, IL USA
| | - Xiaoyang Wu
- Ben May Department for Cancer Research, The University of Chicago, Chicago, IL, USA.
| | - Ming Xu
- Department of Anesthesia and Critical Care, The University of Chicago, Chicago, IL, USA.
| |
Collapse
|
24
|
Diz-Chaves Y, Herrera-Pérez S, González-Matías LC, Lamas JA, Mallo F. Glucagon-Like Peptide-1 (GLP-1) in the Integration of Neural and Endocrine Responses to Stress. Nutrients 2020; 12:nu12113304. [PMID: 33126672 PMCID: PMC7692797 DOI: 10.3390/nu12113304] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2020] [Revised: 10/14/2020] [Accepted: 10/27/2020] [Indexed: 12/20/2022] Open
Abstract
Glucagon like-peptide 1 (GLP-1) within the brain is produced by a population of preproglucagon neurons located in the caudal nucleus of the solitary tract. These neurons project to the hypothalamus and another forebrain, hindbrain, and mesolimbic brain areas control the autonomic function, feeding, and the motivation to feed or regulate the stress response and the hypothalamic-pituitary-adrenal axis. GLP-1 receptor (GLP-1R) controls both food intake and feeding behavior (hunger-driven feeding, the hedonic value of food, and food motivation). The activation of GLP-1 receptors involves second messenger pathways and ionic events in the autonomic nervous system, which are very relevant to explain the essential central actions of GLP-1 as neuromodulator coordinating food intake in response to a physiological and stress-related stimulus to maintain homeostasis. Alterations in GLP-1 signaling associated with obesity or chronic stress induce the dysregulation of eating behavior. This review summarized the experimental shreds of evidence from studies using GLP-1R agonists to describe the neural and endocrine integration of stress responses and feeding behavior.
Collapse
Affiliation(s)
- Yolanda Diz-Chaves
- CINBIO, Universidade de Vigo, Grupo FB3A, Laboratorio de Endocrinología, 36310 Vigo, Spain;
- Correspondence: (Y.D.-C.); (F.M.); Tel.: +34-(986)-130226 (Y.D.-C.); +34-(986)-812393 (F.M.)
| | - Salvador Herrera-Pérez
- CINBIO, Universidade de Vigo, Grupo FB3B, Laboratorio de Neurociencia, 36310 Vigo, Spain; (S.H.-P.); (J.A.L.)
| | | | - José Antonio Lamas
- CINBIO, Universidade de Vigo, Grupo FB3B, Laboratorio de Neurociencia, 36310 Vigo, Spain; (S.H.-P.); (J.A.L.)
| | - Federico Mallo
- CINBIO, Universidade de Vigo, Grupo FB3A, Laboratorio de Endocrinología, 36310 Vigo, Spain;
- Correspondence: (Y.D.-C.); (F.M.); Tel.: +34-(986)-130226 (Y.D.-C.); +34-(986)-812393 (F.M.)
| |
Collapse
|
25
|
Glucagon-like peptide-1 receptors and sexual behaviors in male mice. Psychoneuroendocrinology 2020; 117:104687. [PMID: 32388229 DOI: 10.1016/j.psyneuen.2020.104687] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/17/2020] [Revised: 03/20/2020] [Accepted: 04/11/2020] [Indexed: 01/02/2023]
Abstract
The gut-brain peptide glucagon-like peptide-1 (GLP-1) reduces reward from palatable food and drugs of abuse. Recent rodent studies show that activation of GLP-1 receptors (GLP-1R) within the nucleus of the solitary tract (NTS) not only suppresses the motivation and intake of palatable food, but also reduces alcohol-related behaviors. As reward induced by addictive drugs and sexual behaviors involve similar neurocircuits, we hypothesized that activation of GLP-1R suppresses sexual behavior in sexually naïve male mice. We initially identified that systemic administration of the GLP-1R agonist, exendin-4 (Ex4), decreased the frequency and duration of mounting behaviors, but did not alter the preference for females or female bedding. Thereafter infusion of Ex4 into the NTS decreased various behaviors of the sexual interaction chain, namely social, mounting and self-grooming behaviors. In male mice tested in the sexual interaction test, NTS-Ex4 increased dopamine turnover and enhanced serotonin levels in the nucleus accumbens (NAc). In addition, these mice displayed higher corticosterone, but not testosterone, levels in plasma. Finally, GLP-1R antagonist, exendin-3 (9-39) amide (Ex9), infused into the NTS differentially altered the ability of systemic-Ex4 to suppress the various behaviors of the sexual interaction chain, indicating that GLP-1R within the NTS is one of many sub-regions contributing to the GLP-1 dependent sexual behavior link. In these mice NTS-Ex9 partly blocked the systemic-Ex4 enhancement of corticosterone levels. Collectively, these data highlight that activation of GLP-1R, specifically those in the NTS, reduces sexual interaction behaviors in sexually naïve male mice and further provide a link between NTS-GLP-1R activation and reward-related behaviors.
Collapse
|
26
|
PPG neurons in the nucleus of the solitary tract modulate heart rate but do not mediate GLP-1 receptor agonist-induced tachycardia in mice. Mol Metab 2020; 39:101024. [PMID: 32446875 PMCID: PMC7317700 DOI: 10.1016/j.molmet.2020.101024] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/24/2020] [Revised: 05/13/2020] [Accepted: 05/13/2020] [Indexed: 01/07/2023] Open
Abstract
Objective Glucagon-like peptide-1 receptor agonists (GLP-1RAs) are used as anti-diabetic drugs and are approved for obesity treatment. However, GLP-1RAs also affect heart rate (HR) and arterial blood pressure (ABP) in rodents and humans. Although the activation of GLP-1 receptors (GLP-1R) is known to increase HR, the circuits recruited are unclear, and in particular, it is unknown whether GLP-1RAs activate preproglucagon (PPG) neurons, the brain source of GLP-1, to elicit these effects. Methods We investigated the effect of GLP-1RAs on heart rate in anaesthetized adult mice. In a separate study, we manipulated the activity of nucleus tractus solitarius (NTS) PPG neurons (PPGNTS) in awake, freely behaving transgenic Glu-Cre mice implanted with biotelemetry probes and injected with AAV-DIO-hM3Dq:mCherry or AAV-mCherry-FLEX-DTA. Results Systemic administration of the GLP-1RA Ex-4 increased resting HR in anaesthetized or conscious mice, but had no effect on ABP in conscious mice. This effect was abolished by β-adrenoceptor blockade with atenolol, but unaffected by the muscarinic antagonist atropine. Furthermore, Ex-4-induced tachycardia persisted when PPGNTS neurons were ablated, and Ex-4 did not induce expression of the neuronal activity marker cFos in PPGNTS neurons. PPGNTS ablation or acute chemogenetic inhibition of these neurons via hM4Di receptors had no effect on resting HR. In contrast, chemogenetic activation of PPGNTS neurons increased resting HR. Furthermore, the application of GLP-1 within the subarachnoid space of the middle thoracic spinal cord, a major projection target of PPG neurons, increased HR. Conclusions These results demonstrate that both systemic application of Ex-4 or GLP-1 and chemogenetic activation of PPGNTS neurons increases HR. Ex-4 increases the activity of cardiac sympathetic preganglionic neurons of the spinal cord without recruitment of PPGNTS neurons, and thus likely recapitulates the physiological effects of PPG neuron activation. These neurons therefore do not play a significant role in controlling resting HR and ABP but are capable of inducing tachycardia and so are likely involved in cardiovascular responses to acute stress. Activation of PPG neurons triggers increases in heart rate in mice. PPG neurons do not provide a tonic sympathetic drive to the heart. The tachycardic effect of systemic Ex-4 is not mediated by PPG neurons. GLP-1 receptor activation has a sympathoexcitatory effect that increases heart rate. Local activation of GLP-1R in the spinal cord is sufficient to elicit tachycardia.
Collapse
|
27
|
Mukherjee A, Hum A, Gustafson TJ, Mietlicki-Baase EG. Binge-like palatable food intake in rats reduces preproglucagon in the nucleus tractus solitarius. Physiol Behav 2020; 219:112830. [PMID: 32061682 DOI: 10.1016/j.physbeh.2020.112830] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2019] [Revised: 02/01/2020] [Accepted: 02/05/2020] [Indexed: 01/08/2023]
Abstract
Binge eating involves eating larger than normal quantities of food within a discrete period of time. The neurohormonal controls governing binge-like palatable food intake are not well understood. Glucagon-like peptide-1 (GLP-1), a hormone produced peripherally in the intestine and centrally in the nucleus tractus solitarius (NTS), reduces food intake. Given that the NTS plays a critical role in integrating peripheral and central signals relevant for food intake, as well as the role of GLP-1 in motivated feeding, we tested the hypothesis that expression of the GLP-1 precursor preproglucagon (PPG) would be reduced in the NTS of rats with a history of binge-like palatable food intake. Adult male rats received access to fat for 1 h shortly before lights off, either every day (Daily, D) or only 3d/week (Intermittent, INT). INT rats ate significantly more fat than did D rats in sessions where all rats had fat access. After ~8.5 weeks of diet maintenance, we measured plasma GLP-1 as well as NTS PPG and GLP-1 receptor expression. INT rats had significantly lower NTS PPG mRNA expression compared to D rats. However, plasma GLP-1 was significantly increased in the INT group versus D rats. No significant differences were observed in NTS GLP-1 receptor expression. We also measured plasma insulin levels, fasted blood glucose, and plasma corticosterone but no differences were detected between groups. These results support the hypothesis that binge-like eating reduces NTS GLP-1 expression, and furthermore, demonstrate divergent impacts of binge-like eating on peripheral (plasma) versus central GLP-1.
Collapse
Affiliation(s)
- Ashmita Mukherjee
- Psychology, University at Buffalo, State University of New York, Buffalo, NY 14260, USA
| | - Avery Hum
- Biological Sciences, University at Buffalo, State University of New York, Buffalo, NY 14260, USA
| | - Tyler J Gustafson
- Exercise and Nutrition Sciences, University at Buffalo, State University of New York, G10G Farber Hall, Buffalo, NY 14214, USA
| | - Elizabeth G Mietlicki-Baase
- Exercise and Nutrition Sciences, University at Buffalo, State University of New York, G10G Farber Hall, Buffalo, NY 14214, USA; Center for Ingestive Behavior Research, University at Buffalo, State University of New York, Buffalo, NY 14260, USA.
| |
Collapse
|
28
|
Akalestou E, Genser L, Rutter GA. Glucocorticoid Metabolism in Obesity and Following Weight Loss. Front Endocrinol (Lausanne) 2020; 11:59. [PMID: 32153504 PMCID: PMC7045057 DOI: 10.3389/fendo.2020.00059] [Citation(s) in RCA: 65] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/01/2019] [Accepted: 01/30/2020] [Indexed: 01/08/2023] Open
Abstract
Glucocorticoids are steroid hormones produced by the adrenal cortex and are essential for the maintenance of various metabolic and homeostatic functions. Their function is regulated at the tissue level by 11β-hydroxysteroid dehydrogenases and they signal through the glucocorticoid receptor, a ligand-dependent transcription factor. Clinical observations have linked excess glucocorticoid levels with profound metabolic disturbances of intermediate metabolism resulting in abdominal obesity, insulin resistance and dyslipidaemia. In this review, we discuss the physiological mechanisms of glucocorticoid secretion, regulation and function, and survey the metabolic consequences of excess glucocorticoid action resulting from elevated release and activation or up-regulated signaling. Finally, we summarize the reported impact of weight loss by diet, exercise, or bariatric surgery on circulating and tissue-specific glucocorticoid levels and examine the therapeutic possibility of reversing glucocorticoid-associated metabolic disorders.
Collapse
Affiliation(s)
- Elina Akalestou
- Section of Cell Biology and Functional Genomics, Division of Diabetes, Endocrinology and Metabolism, Department of Metabolism, Digestion and Reproduction, Imperial Centre for Translational and Experimental Medicine, Imperial College London, London, United Kingdom
| | - Laurent Genser
- Section of Cell Biology and Functional Genomics, Division of Diabetes, Endocrinology and Metabolism, Department of Metabolism, Digestion and Reproduction, Imperial Centre for Translational and Experimental Medicine, Imperial College London, London, United Kingdom
- Department of Digestive and Hepato-Pancreato-Biliary Surgery, Liver Transplantation, Assistance Publique-Hôpitaux de Paris, Pitié-Salpêtrière University Hospital, Institut Hospitalo-Universitaire ICAN, Sorbonne Université, Paris, France
| | - Guy A. Rutter
- Section of Cell Biology and Functional Genomics, Division of Diabetes, Endocrinology and Metabolism, Department of Metabolism, Digestion and Reproduction, Imperial Centre for Translational and Experimental Medicine, Imperial College London, London, United Kingdom
- *Correspondence: Guy A. Rutter
| |
Collapse
|
29
|
Domae C, Nanba F, Maruo T, Suzuki T, Ashida H, Yamashita Y. Black soybean seed coat polyphenols promote nitric oxide production in the aorta through glucagon-like peptide-1 secretion from the intestinal cells. Food Funct 2019; 10:7875-7882. [PMID: 31746899 DOI: 10.1039/c9fo02050k] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2023]
Abstract
Black soybean seed coat polyphenols were reported to possess various bioregulatory functions. However, the effects of black soybean seed coat polyphenols on vascular functions are unknown. Vascular dysfunction caused by aging and vascular stiffness is associated with a risk of cardiovascular disease (CVD), and a reduction in nitric oxide (NO) levels can trigger the onset of CVD. In the present study, we investigated the effect of polyphenol-rich black soybean seed coat extract (BE) on vascular functions and the underlying mechanisms involved. The oral administration of BE at 50 mg per kg body weight to Wistar rats increased NO levels as determined by eNOS phosphorylation. The administration of BE also increased GLP-1 and cAMP levels. Furthermore, the effects of BE were inhibited in the presence of a GLP-1 receptor antagonist. This suggests that GLP-1 is strongly involved in the underlying mechanism of NO production in vivo. In conclusion, BE contributes to the improvement of vascular functions by promoting NO production. Regarding the putative underlying mechanism, GLP-1 secreted from intestinal cells by the polyphenols in BE activates eNOS in vascular endothelial cells.
Collapse
Affiliation(s)
- Chiaki Domae
- Department of Agrobioscience, Graduate School of Agricultural Science, Kobe University, Nada-ku, Kobe, Hyogo 657-8501, Japan.
| | | | | | | | | | | |
Collapse
|
30
|
Müller TD, Finan B, Bloom SR, D'Alessio D, Drucker DJ, Flatt PR, Fritsche A, Gribble F, Grill HJ, Habener JF, Holst JJ, Langhans W, Meier JJ, Nauck MA, Perez-Tilve D, Pocai A, Reimann F, Sandoval DA, Schwartz TW, Seeley RJ, Stemmer K, Tang-Christensen M, Woods SC, DiMarchi RD, Tschöp MH. Glucagon-like peptide 1 (GLP-1). Mol Metab 2019; 30:72-130. [PMID: 31767182 PMCID: PMC6812410 DOI: 10.1016/j.molmet.2019.09.010] [Citation(s) in RCA: 1118] [Impact Index Per Article: 186.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/17/2019] [Revised: 09/10/2019] [Accepted: 09/22/2019] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND The glucagon-like peptide-1 (GLP-1) is a multifaceted hormone with broad pharmacological potential. Among the numerous metabolic effects of GLP-1 are the glucose-dependent stimulation of insulin secretion, decrease of gastric emptying, inhibition of food intake, increase of natriuresis and diuresis, and modulation of rodent β-cell proliferation. GLP-1 also has cardio- and neuroprotective effects, decreases inflammation and apoptosis, and has implications for learning and memory, reward behavior, and palatability. Biochemically modified for enhanced potency and sustained action, GLP-1 receptor agonists are successfully in clinical use for the treatment of type-2 diabetes, and several GLP-1-based pharmacotherapies are in clinical evaluation for the treatment of obesity. SCOPE OF REVIEW In this review, we provide a detailed overview on the multifaceted nature of GLP-1 and its pharmacology and discuss its therapeutic implications on various diseases. MAJOR CONCLUSIONS Since its discovery, GLP-1 has emerged as a pleiotropic hormone with a myriad of metabolic functions that go well beyond its classical identification as an incretin hormone. The numerous beneficial effects of GLP-1 render this hormone an interesting candidate for the development of pharmacotherapies to treat obesity, diabetes, and neurodegenerative disorders.
Collapse
Affiliation(s)
- T D Müller
- Institute for Diabetes and Obesity, Helmholtz Diabetes Center, Helmholtz Zentrum München, German Research Center for Environmental Health (GmbH), Neuherberg, Germany; German Center for Diabetes Research (DZD), Neuherberg, Germany; Department of Pharmacology and Experimental Therapy, Institute of Experimental and Clinical Pharmacology and Toxicology, Eberhard Karls University Hospitals and Clinics, Tübingen, Germany.
| | - B Finan
- Novo Nordisk Research Center Indianapolis, Indianapolis, IN, USA
| | - S R Bloom
- Division of Diabetes, Endocrinology and Metabolism, Imperial College London, London, UK
| | - D D'Alessio
- Division of Endocrinology, Duke University Medical Center, Durham, NC, USA
| | - D J Drucker
- The Department of Medicine, Lunenfeld-Tanenbaum Research Institute, Mt. Sinai Hospital, University of Toronto, Ontario, M5G1X5, Canada
| | - P R Flatt
- SAAD Centre for Pharmacy & Diabetes, Ulster University, Coleraine, Northern Ireland, UK
| | - A Fritsche
- German Center for Diabetes Research (DZD), Neuherberg, Germany; Institute for Diabetes Research and Metabolic Diseases of the Helmholtz Center Munich at the University of Tübingen, Tübingen, Germany; Division of Endocrinology, Diabetology, Vascular Disease, Nephrology and Clinical Chemistry, Department of Internal Medicine, University of Tübingen, Tübingen, Germany
| | - F Gribble
- Metabolic Research Laboratories and Medical Research Council Metabolic Diseases Unit, Wellcome Trust-Medical Research Council, Institute of Metabolic Science, Addenbrooke's Hospital, University of Cambridge, Cambridge, CB2 0QQ, UK
| | - H J Grill
- Institute of Diabetes, Obesity and Metabolism, Department of Psychology, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - J F Habener
- Laboratory of Molecular Endocrinology, Massachusetts General Hospital, Harvard University, Boston, MA, USA
| | - J J Holst
- Novo Nordisk Foundation Center for Basic Metabolic Research, Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - W Langhans
- Physiology and Behavior Laboratory, ETH Zurich, Schwerzenbach, Switzerland
| | - J J Meier
- Diabetes Division, St Josef Hospital, Ruhr-University Bochum, Bochum, Germany
| | - M A Nauck
- Diabetes Center Bochum-Hattingen, St Josef Hospital (Ruhr-Universität Bochum), Bochum, Germany
| | - D Perez-Tilve
- Department of Internal Medicine, University of Cincinnati-College of Medicine, Cincinnati, OH, USA
| | - A Pocai
- Cardiovascular & ImmunoMetabolism, Janssen Research & Development, Welsh and McKean Roads, Spring House, PA, 19477, USA
| | - F Reimann
- Metabolic Research Laboratories and Medical Research Council Metabolic Diseases Unit, Wellcome Trust-Medical Research Council, Institute of Metabolic Science, Addenbrooke's Hospital, University of Cambridge, Cambridge, CB2 0QQ, UK
| | - D A Sandoval
- Department of Surgery, University of Michigan Medical School, Ann Arbor, MI, USA
| | - T W Schwartz
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, DL-2200, Copenhagen, Denmark; Department of Biomedical Sciences, University of Copenhagen, DK-2200, Copenhagen, Denmark
| | - R J Seeley
- Department of Surgery, University of Michigan Medical School, Ann Arbor, MI, USA
| | - K Stemmer
- Institute for Diabetes and Obesity, Helmholtz Diabetes Center, Helmholtz Zentrum München, German Research Center for Environmental Health (GmbH), Neuherberg, Germany; German Center for Diabetes Research (DZD), Neuherberg, Germany
| | - M Tang-Christensen
- Obesity Research, Global Drug Discovery, Novo Nordisk A/S, Måløv, Denmark
| | - S C Woods
- Department of Psychiatry and Behavioral Neuroscience, University of Cincinnati, Cincinnati, OH, USA
| | - R D DiMarchi
- Novo Nordisk Research Center Indianapolis, Indianapolis, IN, USA; Department of Chemistry, Indiana University, Bloomington, IN, USA
| | - M H Tschöp
- German Center for Diabetes Research (DZD), Neuherberg, Germany; Division of Metabolic Diseases, Department of Medicine, Technische Universität München, Munich, Germany; Helmholtz Zentrum München, German Research Center for Environmental Health (GmbH), Neuherberg, Germany
| |
Collapse
|
31
|
Abstract
Short-chain fatty acids (SCFAs), the main metabolites produced by bacterial fermentation of dietary fibre in the gastrointestinal tract, are speculated to have a key role in microbiota-gut-brain crosstalk. However, the pathways through which SCFAs might influence psychological functioning, including affective and cognitive processes and their neural basis, have not been fully elucidated. Furthermore, research directly exploring the role of SCFAs as potential mediators of the effects of microbiota-targeted interventions on affective and cognitive functioning is sparse, especially in humans. This Review summarizes existing knowledge on the potential of SCFAs to directly or indirectly mediate microbiota-gut-brain interactions. The effects of SCFAs on cellular systems and their interaction with gut-brain signalling pathways including immune, endocrine, neural and humoral routes are described. The effects of microbiota-targeted interventions such as prebiotics, probiotics and diet on psychological functioning and the putative mediating role of SCFA signalling will also be discussed, as well as the relationship between SCFAs and psychobiological processes. Finally, future directions to facilitate direct investigation of the effect of SCFAs on psychological functioning are outlined.
Collapse
|
32
|
Turkson S, Kloster A, Hamilton PJ, Neigh GN. Neuroendocrine drivers of risk and resilience: The influence of metabolism & mitochondria. Front Neuroendocrinol 2019; 54:100770. [PMID: 31288042 PMCID: PMC6886586 DOI: 10.1016/j.yfrne.2019.100770] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/02/2019] [Revised: 06/20/2019] [Accepted: 07/03/2019] [Indexed: 02/07/2023]
Abstract
The manifestation of risk versus resilience has been considered from varying perspectives including genetics, epigenetics, early life experiences, and type and intensity of the challenge with which the organism is faced. Although all of these factors are central to determining risk and resilience, the current review focuses on what may be a final common pathway: metabolism. When an organism is faced with a perturbation to the environment, whether internal or external, appropriate energy allocation is essential to resolving the divergence from equilibrium. This review examines the potential role of metabolism in the manifestation of stress-induced neural compromise. In addition, this review details the current state of knowledge on neuroendocrine factors which are poised to set the tone of the metabolic response to a systemic challenge. The goal is to provide an essential framework for understanding stress in a metabolic context and appreciation for key neuroendocrine signals.
Collapse
Affiliation(s)
- Susie Turkson
- Department of Anatomy & Neurobiology, Virginia Commonwealth University, Richmond, VA, United States
| | - Alix Kloster
- Department of Anatomy & Neurobiology, Virginia Commonwealth University, Richmond, VA, United States
| | - Peter J Hamilton
- Department of Anatomy & Neurobiology, Virginia Commonwealth University, Richmond, VA, United States
| | - Gretchen N Neigh
- Department of Anatomy & Neurobiology, Virginia Commonwealth University, Richmond, VA, United States.
| |
Collapse
|
33
|
Winzeler B, da Conceição I, Refardt J, Sailer CO, Dutilh G, Christ-Crain M. Effects of Glucagon-Like Peptide-1 Receptor Agonists on Hypothalamic-Pituitary-Adrenal Axis in Healthy Volunteers. J Clin Endocrinol Metab 2019; 104:202-208. [PMID: 30272170 DOI: 10.1210/jc.2018-01420] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/28/2018] [Accepted: 09/25/2018] [Indexed: 02/13/2023]
Abstract
CONTEXT Recent findings from animal and human studies indicate that glucagon-like peptide-1 (GLP-1) receptor agonists (RAs) modulate stress response by activating the hypothalamic-pituitary-adrenal (HPA) axis, which may have relevant clinical implications. OBJECTIVE To investigate the influence of GLP-1 RA treatment on HPA axis activity compared with placebo in healthy volunteers. DESIGN Double-blind, randomized, crossover study. SETTING University Hospital Basel, Switzerland. PARTICIPANTS Twenty healthy volunteers. INTERVENTION Dulaglutide (Trulicity®) 1.5 mg and placebo (0.9% sodium chloride) were given subcutaneously once weekly for 3 weeks. MAIN OUTCOME MEASURES Twenty-four-hour urinary free cortisol, circadian rhythm of serum and salivary cortisol, cortisol after 1 mg dexamethasone suppression test, and cortisol levels before and after stimulation with ACTH. RESULTS Urinary free cortisol levels were similar under dulaglutide [median (interquartile range) 240 nmol/L (164, 324)] vs placebo [188 nmol/L (133, 338), P = 0.131]. The circadian rhythm of serum and salivary cortisol were comparable in both groups as were cortisol levels after dexamethasone [dulaglutide 28 nmol/L (22, 47.5) vs placebo 26.5 nmol/L (15.8, 45.5), P = 0.4]. Serum cortisol levels in dulaglutide and placebo treated participants were 522 nmol (388, 710) and 530 nmol/L (394, 747), before (P = 0.6), and 658 nmol/L (604, 810) and 636 nmol/L (512, 910) after ACTH stimulation (P = 0.87). CONCLUSION Our results suggest that there is no activation of the HPA axis by long-term GLP-1 RA exposure, particularly dulaglutide, at the medically approved dosage of 1.5 mg once weekly.
Collapse
Affiliation(s)
- Bettina Winzeler
- Department of Endocrinology, Diabetology and Metabolism, University Hospital Basel, Basel, Switzerland
| | - Ismael da Conceição
- Department of Endocrinology, Diabetology and Metabolism, University Hospital Basel, Basel, Switzerland
| | - Julie Refardt
- Department of Endocrinology, Diabetology and Metabolism, University Hospital Basel, Basel, Switzerland
| | - Clara O Sailer
- Department of Endocrinology, Diabetology and Metabolism, University Hospital Basel, Basel, Switzerland
| | - Gilles Dutilh
- Department of Clinical Research, University of Basel, Basel, Switzerland
| | - Mirjam Christ-Crain
- Department of Endocrinology, Diabetology and Metabolism, University Hospital Basel, Basel, Switzerland
| |
Collapse
|
34
|
Fandiño J, Vaz AA, Toba L, Romaní-Pérez M, González-Matías L, Mallo F, Diz-Chaves Y. Liraglutide Enhances the Activity of the ACE-2/Ang(1-7)/Mas Receptor Pathway in Lungs of Male Pups from Food-Restricted Mothers and Prevents the Reduction of SP-A. Int J Endocrinol 2018; 2018:6920620. [PMID: 30627159 PMCID: PMC6304858 DOI: 10.1155/2018/6920620] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/30/2018] [Accepted: 07/30/2018] [Indexed: 02/07/2023] Open
Abstract
In utero growth restriction and being born small for gestational age are risk factors for respiratory morbidity. IUGR (in utero growth retardation) is associated to overall reduction in lung weight, surfactant content and activity, impaired maturation of the alveolar type II cells, and decreased alveolar formation. The renin-angiotensin system (RAS) may be a key target underlying pathophysiological lung alterations. GLP-1 and agonists of its receptor modulate the expression levels of different components of RAS and also are very important for lung maturation and the production of surfactant proteins. The aim of this study was to elucidate the effects of IUGR induced by perinatal food restriction of the mother in the lung function of pups at early stages of life (PD21) and to determine if liraglutide had any effect during gestational period. Sprague-Dawley pregnant rats were randomly assigned to 50% food restriction (MPFR) or ad libitum control (CT) groups at day of pregnancy 12 (GD12). From GD14 to parturition, pregnant MPFR and CT rats were treated with liraglutide or vehicle. At postnatal day 21 and before weaning, 20 CT and 20 FR male pups were sacrificed and lungs were analyzed by RT-PCR. Liraglutide restored surfactant protein A (SP-A) mRNA expression in pup lungs from food-restricted mothers. Surfactant protein B (SP-B) mRNA expression is not affected by neither IUGR nor liraglutide treatment. Moreover, liraglutide modulated different elements of RAS, increasing angiotensin-converting enzyme 2 (ACE2) and MasR mRNA expression only in pups from food-restricted mothers (MPFR), despite food restriction had not any direct effect at this early stage. Liraglutide also increased endothelial nitric oxide synthase (eNOS) expression in MPFR lungs, reflecting the activation of MasR by angiotensin 1-7. In conclusion, liraglutide prevented the alteration in lung function induced by IUGR and promoted the positive effects of ACE2-Ang(1-7)-MasR in restoring lung function.
Collapse
Affiliation(s)
- J. Fandiño
- Laboratory Endocrinology, LabEndo, Centro de Investigaciones Biomédicas (CINBIO), University of Vigo, E36310 Vigo, Spain
| | - A. A. Vaz
- Laboratory Endocrinology, LabEndo, Centro de Investigaciones Biomédicas (CINBIO), University of Vigo, E36310 Vigo, Spain
| | - L. Toba
- Laboratory Endocrinology, LabEndo, Centro de Investigaciones Biomédicas (CINBIO), University of Vigo, E36310 Vigo, Spain
| | - M. Romaní-Pérez
- Laboratory Endocrinology, LabEndo, Centro de Investigaciones Biomédicas (CINBIO), University of Vigo, E36310 Vigo, Spain
| | - L. González-Matías
- Laboratory Endocrinology, LabEndo, Centro de Investigaciones Biomédicas (CINBIO), University of Vigo, E36310 Vigo, Spain
| | - F. Mallo
- Laboratory Endocrinology, LabEndo, Centro de Investigaciones Biomédicas (CINBIO), University of Vigo, E36310 Vigo, Spain
| | - Y. Diz-Chaves
- Laboratory Endocrinology, LabEndo, Centro de Investigaciones Biomédicas (CINBIO), University of Vigo, E36310 Vigo, Spain
| |
Collapse
|
35
|
Stengel A, Taché Y. Gut-Brain Neuroendocrine Signaling Under Conditions of Stress-Focus on Food Intake-Regulatory Mediators. Front Endocrinol (Lausanne) 2018; 9:498. [PMID: 30210455 PMCID: PMC6122076 DOI: 10.3389/fendo.2018.00498] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/31/2018] [Accepted: 08/08/2018] [Indexed: 12/12/2022] Open
Abstract
The gut-brain axis represents a bidirectional communication route between the gut and the central nervous system comprised of neuronal as well as humoral signaling. This system plays an important role in the regulation of gastrointestinal as well as homeostatic functions such as hunger and satiety. Recent years also witnessed an increased knowledge on the modulation of this axis under conditions of exogenous or endogenous stressors. The present review will discuss the alterations of neuroendocrine gut-brain signaling under conditions of stress and the respective implications for the regulation of food intake.
Collapse
Affiliation(s)
- Andreas Stengel
- Department of Psychosomatic Medicine and Psychotherapy, University Hospital Tübingen, Tübingen, Germany
- Charité Center for Internal Medicine and Dermatology, Department for Psychosomatic Medicine, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin and Berlin Institute of Health, Berlin, Germany
| | - Yvette Taché
- CURE/Digestive Diseases Research Center, Vatche and Tamar Manoukian Digestive Diseases Division, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, United States
- VA Greater Los Angeles Health Care System, Los Angeles, CA, United States
| |
Collapse
|
36
|
Terrill SJ, Maske CB, Williams DL. Endogenous GLP-1 in lateral septum contributes to stress-induced hypophagia. Physiol Behav 2018; 192:17-22. [PMID: 29510158 PMCID: PMC6019151 DOI: 10.1016/j.physbeh.2018.03.001] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2017] [Revised: 02/16/2018] [Accepted: 03/02/2018] [Indexed: 01/25/2023]
Abstract
Glucagon-like peptide 1 (GLP-1) neurons of the caudal brainstem project to many brain areas, including the lateral septum (LS), which has a known role in stress responses. Previously, we showed that endogenous GLP-1 in the LS plays a physiologic role in the control of feeding under non-stressed conditions, however, central GLP-1 is also involved in behavioral and endocrine responses to stress. Here, we asked whether LS GLP-1 receptors (GLP-1R) contribute to stress-induced hypophagia. Male rats were implanted with bilateral cannulas targeting the dorsal subregion of the LS (dLS). In a within-subjects design, shortly before the onset of the dark phase, rats received dLS injections of saline or the GLP-1R antagonist Exendin (9-39) (Ex9) prior to 30 min restraint stress. Food intake was measured continuously for the next 20 h. The stress-induced hypophagia observed within the first 30 min of dark was not influenced by Ex9 pretreatment, but Ex9 tended to blunt the effect of stress as early as 1 and 2 h into the dark phase. By 4-6 h, there were significant stress X drug interactions, and Ex9 pretreatment blocked the stress-induced suppression of feeding. These effects were mediated entirely through changes in average meal size; stress suppressed meal size while dLS Ex9 attenuated this effect. Using a similar design, we examined the role of dLS GLP-1R in the neuroendocrine response to acute restraint stress. As expected, stress potently increased serum corticosterone, but blockade of dLS GLP-1Rs did not affect this response. Together, these data show that endogenous GLP-1 action in the dLS plays a role in some but not all of the physiologic responses to acute stress.
Collapse
Affiliation(s)
- Sarah J Terrill
- Department of Psychology & Program in Neuroscience, Florida State University, Tallahassee, FL 32306, United States
| | - Calyn B Maske
- Department of Psychology & Program in Neuroscience, Florida State University, Tallahassee, FL 32306, United States
| | - Diana L Williams
- Department of Psychology & Program in Neuroscience, Florida State University, Tallahassee, FL 32306, United States.
| |
Collapse
|
37
|
Oride A, Kanasaki H, Mijiddorj T, Sukhbaatar U, Hara T, Tumurbaatar T, Kyo S. GLP-1 increases Kiss-1 mRNA expression in kisspeptin-expressing neuronal cells†. Biol Reprod 2017; 97:240-248. [DOI: 10.1093/biolre/iox087] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2017] [Accepted: 08/04/2017] [Indexed: 12/26/2022] Open
|
38
|
Holt MK, Llewellyn-Smith IJ, Reimann F, Gribble FM, Trapp S. Serotonergic modulation of the activity of GLP-1 producing neurons in the nucleus of the solitary tract in mouse. Mol Metab 2017; 6:909-921. [PMID: 28752054 PMCID: PMC5518719 DOI: 10.1016/j.molmet.2017.06.002] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/27/2017] [Revised: 06/01/2017] [Accepted: 06/05/2017] [Indexed: 12/21/2022] Open
Abstract
Objective Glucagon-like peptide-1 (GLP-1) and 5-HT are potent regulators of food intake within the brain. GLP-1 is expressed by preproglucagon (PPG) neurons in the nucleus tractus solitarius (NTS). We have previously shown that PPG neurons innervate 5-HT neurons in the ventral brainstem. Here, we investigate whether PPG neurons receive serotonergic input and respond to 5-HT. Methods We employed immunohistochemistry to reveal serotonergic innervation of PPG neurons. We investigated the responsiveness of PPG neurons to 5-HT using in vitro Ca2+ imaging in brainstem slices from transgenic mice expressing the Ca2+ indicator, GCaMP3, in PPG neurons, and cell-attached patch-clamp recordings. Results Close appositions from 5-HT-immunoreactive axons occurred on many PPG neurons. Application of 20 μM 5-HT produced robust Ca2+ responses in NTS PPG dendrites but little change in somata. Dendritic Ca2+ spikes were concentration-dependent (2, 20, and 200 μM) and unaffected by blockade of glutamatergic transmission, suggesting 5-HT receptors on PPG neurons. Neither activation nor blockade of 5-HT3 receptors affected [Ca2+]i. In contrast, inhibition of 5-HT2 receptors attenuated increases in intracellular Ca2+ and 5-HT2C receptor activation produced Ca2+ spikes. Patch-clamp recordings revealed that 44% of cells decreased their firing rate under 5-HT, an effect blocked by 5-HT1A receptor antagonism. Conclusions PPG neurons respond directly to 5-HT with a 5-HT2C receptor-dependent increase in dendritic [Ca2+]i. Electrical responses to 5-HT revealed additional inhibitory effects due to somatic 5-HT1A receptors. Reciprocal innervation between 5-HT and PPG neurons suggests that the coordinated activity of these brainstem neurons may play a role in the regulation of food intake. Brainstem PPG neurons receive close appositions from 5-HT-containing axons. 5-HT activates NTS PPG dendrites directly via 5-HT2 receptors. 5-HT inhibits a subset of somata via 5-HT1A receptors. Activation of 5-HT3 receptors does not affect PPG cell [Ca2+]i. 5-HT2C receptor activation induces spatially confined Ca2+ spikes in PPG neurons.
Collapse
Key Words
- 5-HT, 5-hydroxytryptamine
- CCK-8, cholecystokinin-8
- CNS, central nervous system
- Dendritic calcium
- GCaMP
- GLP-1, glucagon-like peptide-1
- HPA, hypothalamic–pituitary–adrenal
- IRT, intermediate reticular nucleus
- NTS
- NTS, nucleus tractus solitarius
- PBN, parabrachial nucleus
- PPG, preproglucagon
- Preproglucagon
- Serotonin
- YFP, yellow fluorescent protein
Collapse
Affiliation(s)
- Marie K Holt
- Centre for Cardiovascular and Metabolic Neuroscience, Department of Neuroscience, Physiology & Pharmacology, University College London, London, WC1E 6BT, UK
| | - Ida J Llewellyn-Smith
- Cardiovascular Medicine, Human Physiology and Centre for Neuroscience, Flinders University, Bedford Park, SA 5042, Australia
| | - Frank Reimann
- Cambridge Institute for Medical Research, University of Cambridge, Addenbrooke's Hospital, Hills Road, Cambridge, CB2 0QQ, UK
| | - Fiona M Gribble
- Cambridge Institute for Medical Research, University of Cambridge, Addenbrooke's Hospital, Hills Road, Cambridge, CB2 0QQ, UK
| | - Stefan Trapp
- Centre for Cardiovascular and Metabolic Neuroscience, Department of Neuroscience, Physiology & Pharmacology, University College London, London, WC1E 6BT, UK.
| |
Collapse
|
39
|
Kamei S, Kaneto H, Tanabe A, Kinoshita T, Obata A, Kimura T, Hirukawa H, Tatsumi F, Shimoda M, Kohara K, Anno T, Nakanishi S, Mune T, Kaku K. Increase in cortisol/ACTH ratio after chronic treatment with liraglutide in patients with type 2 diabetes. DIABETES & METABOLISM 2017; 43:398-399. [PMID: 28283288 DOI: 10.1016/j.diabet.2017.01.008] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/26/2017] [Accepted: 01/29/2017] [Indexed: 10/20/2022]
Affiliation(s)
- S Kamei
- Department of Diabetes, Endocrinology and Metabolism, Kawasaki Medical School, 577 Matsushima, 701-0192 Kurashiki, Japan.
| | - H Kaneto
- Department of Diabetes, Endocrinology and Metabolism, Kawasaki Medical School, 577 Matsushima, 701-0192 Kurashiki, Japan
| | - A Tanabe
- Department of Diabetes, Endocrinology and Metabolism, Kawasaki Medical School, 577 Matsushima, 701-0192 Kurashiki, Japan
| | - T Kinoshita
- Department of Diabetes, Endocrinology and Metabolism, Kawasaki Medical School, 577 Matsushima, 701-0192 Kurashiki, Japan
| | - A Obata
- Department of Diabetes, Endocrinology and Metabolism, Kawasaki Medical School, 577 Matsushima, 701-0192 Kurashiki, Japan
| | - T Kimura
- Department of Diabetes, Endocrinology and Metabolism, Kawasaki Medical School, 577 Matsushima, 701-0192 Kurashiki, Japan
| | - H Hirukawa
- Department of Diabetes, Endocrinology and Metabolism, Kawasaki Medical School, 577 Matsushima, 701-0192 Kurashiki, Japan
| | - F Tatsumi
- Department of Diabetes, Endocrinology and Metabolism, Kawasaki Medical School, 577 Matsushima, 701-0192 Kurashiki, Japan
| | - M Shimoda
- Department of Diabetes, Endocrinology and Metabolism, Kawasaki Medical School, 577 Matsushima, 701-0192 Kurashiki, Japan
| | - K Kohara
- Department of Diabetes, Endocrinology and Metabolism, Kawasaki Medical School, 577 Matsushima, 701-0192 Kurashiki, Japan
| | - Takatoshi Anno
- Department of Diabetes, Endocrinology and Metabolism, Kawasaki Medical School, 577 Matsushima, 701-0192 Kurashiki, Japan
| | - S Nakanishi
- Department of Diabetes, Endocrinology and Metabolism, Kawasaki Medical School, 577 Matsushima, 701-0192 Kurashiki, Japan
| | - T Mune
- Department of Diabetes, Endocrinology and Metabolism, Kawasaki Medical School, 577 Matsushima, 701-0192 Kurashiki, Japan
| | - K Kaku
- Department of Diabetes, Endocrinology and Metabolism, Kawasaki Medical School, 577 Matsushima, 701-0192 Kurashiki, Japan
| |
Collapse
|
40
|
Affiliation(s)
- Jian Yang
- Department of Nutrition, Daping Hospital, The Third Military Medical University, Chongqing, China.,Department of Cardiology, Daping Hospital, The Third Military Medical University, Chongqing, China
| | - Pedro A Jose
- Division of Renal Disease & Hypertension, The George Washington University School of Medicine and Health Sciences, Washington, DC
| | - Chunyu Zeng
- Department of Cardiology, Daping Hospital, The Third Military Medical University, Chongqing, China
| |
Collapse
|
41
|
Gaykema RP, Newmyer BA, Ottolini M, Raje V, Warthen DM, Lambeth PS, Niccum M, Yao T, Huang Y, Schulman IG, Harris TE, Patel MK, Williams KW, Scott MM. Activation of murine pre-proglucagon-producing neurons reduces food intake and body weight. J Clin Invest 2017; 127:1031-1045. [PMID: 28218622 DOI: 10.1172/jci81335] [Citation(s) in RCA: 80] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2015] [Accepted: 12/30/2016] [Indexed: 12/14/2022] Open
Abstract
Peptides derived from pre-proglucagon (GCG peptides) act in both the periphery and the CNS to change food intake, glucose homeostasis, and metabolic rate while playing a role in anxiety behaviors and physiological responses to stress. Although the actions of GCG peptides produced in the gut and pancreas are well described, the role of glutamatergic GGC peptide-secreting hindbrain neurons in regulating metabolic homeostasis has not been investigated. Here, we have shown that chemogenetic stimulation of GCG-producing neurons reduces metabolic rate and food intake in fed and fasted states and suppresses glucose production without an effect on glucose uptake. Stimulation of GCG neurons had no effect on corticosterone secretion, body weight, or conditioned taste aversion. In the diet-induced obese state, the effects of GCG neuronal stimulation on gluconeogenesis were lost, while the food intake-lowering effects remained, resulting in reductions in body weight and adiposity. Our work suggests that GCG peptide-expressing neurons can alter feeding, metabolic rate, and glucose production independent of their effects on hypothalamic pituitary-adrenal (HPA) axis activation, aversive conditioning, or insulin secretion. We conclude that GCG neurons likely stimulate separate populations of downstream cells to produce a change in food intake and glucose homeostasis and that these effects depend on the metabolic state of the animal.
Collapse
|
42
|
Holt MK, Trapp S. The physiological role of the brain GLP-1 system in stress. ACTA ACUST UNITED AC 2016; 2:1229086. [PMID: 27722184 PMCID: PMC5043148 DOI: 10.1080/23312025.2016.1229086] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2016] [Accepted: 08/23/2016] [Indexed: 01/08/2023]
Abstract
Glucagon-like peptide-1 (GLP-1) within the brain is a potent regulator of food intake and most studies have investigated the anorexic effects of central GLP-1. A range of brain regions have now been found to be involved in GLP-1 mediated anorexia, including some which are not traditionally associated with appetite regulation. However, a change in food intake can be indicative of not only reduced energy demand, but also changes in the organism’s motivation to eat following stressful stimuli. In fact, acute stress is well-known to reduce food intake. Recently, more research has focused on the role of GLP-1 in stress and the central GLP-1 system has been found to be activated in response to stressful stimuli. The source of GLP-1 within the brain, the preproglucagon (PPG) neurons, are ideally situated in the brainstem to receive and relay signals of stress and our recent data on the projection pattern of the PPG neurons to the spinal cord suggest a potential strong link with the sympathetic nervous system. We review here the role of central GLP-1 in the regulation of stress responses and discuss the potential involvement of the endogenous source of GLP-1 within the brain, the PPG neurons.
Collapse
Affiliation(s)
- Marie K Holt
- Centre for Cardiovascular and Metabolic Neuroscience, Department of Neuroscience, Physiology & Pharmacology, University College London , WC1E 6BT London , UK
| | - Stefan Trapp
- Centre for Cardiovascular and Metabolic Neuroscience, Department of Neuroscience, Physiology & Pharmacology, University College London , WC1E 6BT London , UK
| |
Collapse
|
43
|
Diz-Chaves Y, Gil-Lozano M, Toba L, Fandiño J, Ogando H, González-Matías LC, Mallo F. Stressing diabetes? The hidden links between insulinotropic peptides and the HPA axis. J Endocrinol 2016; 230:R77-94. [PMID: 27325244 DOI: 10.1530/joe-16-0118] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/26/2016] [Accepted: 06/20/2016] [Indexed: 12/25/2022]
Abstract
Diabetes mellitus exerts metabolic stress on cells and it provokes a chronic increase in the long-term activity of the hypothalamus-pituitary-adrenocortical (HPA) axis, perhaps thereby contributing to insulin resistance. GLP-1 receptor (GLP-1R) agonists are pleiotropic hormones that not only affect glycaemic and metabolic control, but they also produce many other effects including activation of the HPA axis. In fact, several of the most relevant effects of GLP-1 might involve, at least in part, the modulation of the HPA axis. Thus, the anorectic activity of GLP-1 could be mediated by increasing CRF at the hypothalamic level, while its lipolytic effects could imply a local increase in glucocorticoids and glucocorticoid receptor (GC-R) expression in adipose tissue. Indeed, the potent activation of the HPA axis by GLP-1R agonists occurs within the range of therapeutic doses and with a short latency. Interestingly, the interactions of GLP-1 with the HPA axis may underlie most of the effects of GLP-1 on food intake control, glycaemic metabolism, adipose tissue biology and the responses to stress. Moreover, such activity has been observed in animal models (mice and rats), as well as in normal humans and in type I or type II diabetic patients. Accordingly, better understanding of how GLP-1R agonists modulate the activity of the HPA axis in diabetic subjects, especially obese individuals, will be crucial to design new and more efficient therapies for these patients.
Collapse
Affiliation(s)
- Yolanda Diz-Chaves
- Laboratory of EndocrinologyCenter for Biomedical Research - CINBIO, University of Vigo, Vigo, Spain Instituto de Investigación Sanitaria Galicia Sur - IISGSVigo, Spain
| | - Manuel Gil-Lozano
- Laboratory of EndocrinologyCenter for Biomedical Research - CINBIO, University of Vigo, Vigo, Spain Instituto de Investigación Sanitaria Galicia Sur - IISGSVigo, Spain
| | - Laura Toba
- Laboratory of EndocrinologyCenter for Biomedical Research - CINBIO, University of Vigo, Vigo, Spain Instituto de Investigación Sanitaria Galicia Sur - IISGSVigo, Spain
| | - Juan Fandiño
- Laboratory of EndocrinologyCenter for Biomedical Research - CINBIO, University of Vigo, Vigo, Spain Instituto de Investigación Sanitaria Galicia Sur - IISGSVigo, Spain
| | - Hugo Ogando
- Laboratory of EndocrinologyCenter for Biomedical Research - CINBIO, University of Vigo, Vigo, Spain Instituto de Investigación Sanitaria Galicia Sur - IISGSVigo, Spain
| | - Lucas C González-Matías
- Laboratory of EndocrinologyCenter for Biomedical Research - CINBIO, University of Vigo, Vigo, Spain Instituto de Investigación Sanitaria Galicia Sur - IISGSVigo, Spain
| | - Federico Mallo
- Laboratory of EndocrinologyCenter for Biomedical Research - CINBIO, University of Vigo, Vigo, Spain Instituto de Investigación Sanitaria Galicia Sur - IISGSVigo, Spain
| |
Collapse
|
44
|
Quarta C, Clemmensen C. Unforeseen role for glucocorticoids in combinatorial anti-obesity pharmacology. Mol Metab 2016; 5:435-436. [PMID: 27408769 PMCID: PMC4921788 DOI: 10.1016/j.molmet.2016.05.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/04/2016] [Accepted: 05/04/2016] [Indexed: 12/04/2022] Open
|
45
|
Lee SJ, Diener K, Kaufman S, Krieger JP, Pettersen KG, Jejelava N, Arnold M, Watts AG, Langhans W. Limiting glucocorticoid secretion increases the anorexigenic property of Exendin-4. Mol Metab 2016; 5:552-565. [PMID: 27408779 PMCID: PMC4921942 DOI: 10.1016/j.molmet.2016.04.008] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/30/2016] [Revised: 04/14/2016] [Accepted: 04/27/2016] [Indexed: 12/25/2022] Open
Abstract
Objective Glucagon-like peptide-1 (GLP-1) analogs are attractive options for the treatment of type II diabetes and obesity because of their incretin and anorexigenic effects. Peripheral administration of the GLP-1R agonist Exendin-4 (Ex-4) also increases glucocorticoid secretion in rodents and humans, but whether the released glucocorticoids interact with Ex-4's anorexigenic effect remains unclear. Methods To test this, we used two experimental approaches that suppress corticosterone secretion and then assessed Ex-4 effects on eating in adult male rats. First, we combined acute and chronic low dose dexamethasone treatment with Ex-4. Second, we ablated hindbrain catecholamine neurons projecting to the hypothalamus with anti-dopamine-β-hydroxylase-saporin (DSAP) to block Ex-4-induced corticosterone secretion. Results Combining dexamethasone and Ex-4 produced a larger acute anorexigenic effect than Ex-4 alone. Likewise, chronic dexamethasone and Ex-4 co-treatment produced a synergistic effect on eating and greater body weight loss in diet-induced obese rats than Ex-4 alone. DSAP lesions not only blunted Ex-4's ability to increase corticosterone secretion, but potentiated the anorexigenic effect of Ex-4, indicating that Ex-4-dependent corticosterone secretion opposes Ex-4's actions. Consistent with the enhancement of Ex-4's anorexigenic effect, DSAP lesion altered Ex-4-dependent changes in neuropeptide Y, preproglucagon, and corticotropin releasing hormone gene expression involved in glucocorticoid feedback. Conclusions Our findings demonstrate that limiting glucocorticoid secretion and actions with low dose dexamethasone or DSAP lesion increases Ex-4's ability to reduce food intake and body weight. Novel glucocorticoid receptor based mechanisms, therefore, may help enhance GLP-1-based obesity therapies. Blocking HPA axis by low dose dexamethasone increased the anorexigenic property of Ex-4. Dexamethasone/Ex-4 co-treatment reduced food intake and body weight in diet-induced obese rats more than Ex-4 alone. A brain lesion model identified a potential central interaction between glucocorticoids and GLP-1 in food intake control.
Collapse
Affiliation(s)
- Shin J Lee
- Physiology and Behavior Laboratory, ETH Zürich, 8603 Schwerzenbach, Switzerland.
| | - Katharina Diener
- Physiology and Behavior Laboratory, ETH Zürich, 8603 Schwerzenbach, Switzerland
| | - Sharon Kaufman
- Physiology and Behavior Laboratory, ETH Zürich, 8603 Schwerzenbach, Switzerland
| | | | - Klaus G Pettersen
- Physiology and Behavior Laboratory, ETH Zürich, 8603 Schwerzenbach, Switzerland
| | - Nino Jejelava
- Physiology and Behavior Laboratory, ETH Zürich, 8603 Schwerzenbach, Switzerland
| | - Myrtha Arnold
- Physiology and Behavior Laboratory, ETH Zürich, 8603 Schwerzenbach, Switzerland
| | - Alan G Watts
- Department of Biological Sciences, University of Southern California, Los Angeles, CA 90089, USA
| | - Wolfgang Langhans
- Physiology and Behavior Laboratory, ETH Zürich, 8603 Schwerzenbach, Switzerland
| |
Collapse
|
46
|
Pais R, Rievaj J, Meek C, De Costa G, Jayamaha S, Alexander RT, Reimann F, Gribble F. Role of enteroendocrine L-cells in arginine vasopressin-mediated inhibition of colonic anion secretion. J Physiol 2016; 594:4865-78. [PMID: 27037495 PMCID: PMC5009784 DOI: 10.1113/jp272053] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2015] [Accepted: 03/20/2016] [Indexed: 12/17/2022] Open
Abstract
Key points Arginine vasopressin (AVP) stimulates the release of enteroendocrine L‐cell derived hormones glucagon‐like peptide‐1 (GLP‐1) and peptide YY (PYY) in vitro from mouse and human colons. This is mediated by the AVP receptor 1B, which is highly enriched in colonic L‐cells and linked to the elevation of L‐cell calcium and cAMP concentrations. By means of Ussing chambers, we show that AVP reduced colonic anion secretion, although this was blocked by a specific neuropeptide Y receptor Y1 receptor antagonist, suggesting that L‐cell‐released PYY acts locally on the epithelium to modulate fluid balance. In human serum samples, PYY concentrations were higher in samples with raised osmolality and copeptin (surrogate marker for AVP). These findings describe, for the first time, the role of L‐cells in AVP regulated intestinal fluid secretion, potentially linking together hormonal control of blood volume and blood glucose levels, and thus adding to our understanding of the complex pathways involved in the gut hormonal response to different stimuli.
Abstract Arginine vasopressin (AVP) regulates fluid balance and blood pressure via AVP receptor (AVPR)2 in the kidney and AVP receptor 1A in vascular smooth muscle. Its role in intestinal function has received less attention. We hypothesized that enteroendocrine L‐cells producing glucagon‐like peptide 1 (GLP‐1) and peptide YY (PYY) may be a target of AVP and contribute to the control of fluid balance. Avpr1b expression was assessed by quantitative RT‐PCR on flourescence‐activated cell sorting‐isolated L‐ and control cells and was enriched in colonic L‐cells. AVP stimulated GLP‐1 and PYY release from primary cultured murine and human colonic cells and was associated with elevated calcium and cAMP concentrations in L‐cells as measured in cultures from GLU‐Cre/ROSA26‐GCaMP3 and GLU‐Epac2camps mice. An antagonist of AVPR1B reduced AVP‐triggered hormone secretion from murine and human cells. In Ussing chambers, basolaterally applied AVP reduced colonic anion secretion and this effect was blocked by a specific neuropeptide Y receptor Y1 (NPY1R) antagonist. In human serum, PYY concentrations were higher in samples with raised osmolality or copeptin (a surrogate marker for AVP). In conclusion, we propose that AVP activates L‐cell AVPR1B, causing GLP‐1 and PYY secretion. PYY in turn reduces colonic anion secretion via epithelial NPY1R. Our data suggest L‐cells are active players in the hypothalamic control of intestinal fluid homeostasis, providing a potential link between the regulation of blood volume/pressure/osmolality and blood glucose. Arginine vasopressin (AVP) stimulates the release of enteroendocrine L‐cell derived hormones glucagon‐like peptide‐1 (GLP‐1) and peptide YY (PYY) in vitro from mouse and human colons. This is mediated by the AVP receptor 1B, which is highly enriched in colonic L‐cells and linked to the elevation of L‐cell calcium and cAMP concentrations. By means of Ussing chambers, we show that AVP reduced colonic anion secretion, although this was blocked by a specific neuropeptide Y receptor Y1 receptor antagonist, suggesting that L‐cell‐released PYY acts locally on the epithelium to modulate fluid balance. In human serum samples, PYY concentrations were higher in samples with raised osmolality and copeptin (surrogate marker for AVP). These findings describe, for the first time, the role of L‐cells in AVP regulated intestinal fluid secretion, potentially linking together hormonal control of blood volume and blood glucose levels, and thus adding to our understanding of the complex pathways involved in the gut hormonal response to different stimuli.
Collapse
Affiliation(s)
- Ramona Pais
- The Wellcome Trust - MRC Institute of Metabolic Science, Metabolic Research Laboratories, University of Cambridge, Addenbrooke's Hospital, Cambridge, UK
| | - Juraj Rievaj
- The Wellcome Trust - MRC Institute of Metabolic Science, Metabolic Research Laboratories, University of Cambridge, Addenbrooke's Hospital, Cambridge, UK
| | - Claire Meek
- The Wellcome Trust - MRC Institute of Metabolic Science, Metabolic Research Laboratories, University of Cambridge, Addenbrooke's Hospital, Cambridge, UK
| | - Gayan De Costa
- Department of Clinical Biochemistry, Cambridge University Hospitals, Addenbrooke's Hospital, Cambridge, UK
| | - Samanthie Jayamaha
- Department of Clinical Biochemistry, Cambridge University Hospitals, Addenbrooke's Hospital, Cambridge, UK
| | - R Todd Alexander
- Departments of Paediatrics & Physiology, Faculty of Medicine & Dentistry, University of Alberta, Edmonton, Alberta, Canada
| | - Frank Reimann
- The Wellcome Trust - MRC Institute of Metabolic Science, Metabolic Research Laboratories, University of Cambridge, Addenbrooke's Hospital, Cambridge, UK
| | - Fiona Gribble
- The Wellcome Trust - MRC Institute of Metabolic Science, Metabolic Research Laboratories, University of Cambridge, Addenbrooke's Hospital, Cambridge, UK
| |
Collapse
|
47
|
Poudyal H. Mechanisms for the cardiovascular effects of glucagon-like peptide-1. Acta Physiol (Oxf) 2016; 216:277-313. [PMID: 26384481 DOI: 10.1111/apha.12604] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2015] [Revised: 07/25/2015] [Accepted: 09/10/2015] [Indexed: 12/16/2022]
Abstract
Over the past three decades, at least 10 hormones secreted by the enteroendocrine cells have been discovered, which directly affect the cardiovascular system through their innate receptors expressed in the heart and blood vessels or through a neural mechanism. Glucagon-like peptide-1 (GLP-1), an important incretin, is perhaps best studied of these gut-derived hormones with important cardiovascular effects. In this review, I have discussed the mechanism of GLP-1 release from the enteroendocrine L-cells and its physiological effects on the cardiovascular system. Current evidence suggests that GLP-1 has positive inotropic and chronotropic effects on the heart and may be important in preserving left ventricular structure and function by direct and indirect mechanisms. The direct effects of GLP-1 in the heart may be mediated through GLP-1R expressed in atria as well as arteries and arterioles in the left ventricle and mainly involve in the activation of multiple pro-survival kinases and enhanced energy utilization. There is also good evidence to support the involvement of a second, yet to be identified, GLP-1 receptor. Further, GLP-1(9-36)amide, which was previously thought to be the inactive metabolite of the active GLP-1(7-36)amide, may also have direct cardioprotective effects. GLP-1's action on GLP-1R expressed in the central nervous system, kidney, vasculature and the pancreas may indirectly contribute to its cardioprotective effects.
Collapse
Affiliation(s)
- H. Poudyal
- Department of Diabetes, Endocrinology and Nutrition; Graduate School of Medicine and Hakubi Centre for Advanced Research; Kyoto University; Kyoto Japan
| |
Collapse
|
48
|
Sandoval D, Sisley SR. Brain GLP-1 and insulin sensitivity. Mol Cell Endocrinol 2015; 418 Pt 1:27-32. [PMID: 25724479 PMCID: PMC4547906 DOI: 10.1016/j.mce.2015.02.017] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/04/2014] [Revised: 02/02/2015] [Accepted: 02/17/2015] [Indexed: 12/25/2022]
Abstract
Type 2 diabetes is often treated with a class of drugs referred to as glucagon-like peptide-1 (GLP-1) analogs. GLP-1 is a peptide secreted by the gut that acts through only one known receptor, the GLP-1 receptor. The primary function of GLP-1 is thought to be lowering of postprandial glucose levels. Indeed, medications utilizing this system, including the long-acting GLP-1 analogs liraglutide and exenatide, are beneficial in reducing both blood sugars and body weight. GLP-1 analogs were long presumed to affect glucose control through their ability to increase insulin levels through peripheral action on beta cells. However, multiple lines of data point to the ability of GLP-1 to act within the brain to alter glucose regulation. In this review we will discuss the evidence for a central GLP-1 system and the effects of GLP-1 in the brain on regulating multiple facets of glucose homeostasis including glucose tolerance, insulin production, insulin sensitivity, hepatic glucose production, muscle glucose uptake, and connections of the central GLP-1 system to the gut. Although the evidence indicates that GLP-1 receptors in the brain are not necessary for physiologic control of glucose regulation, we discuss the research showing a strong effect of acute manipulation of the central GLP-1 system on glucose control and how it is relevant to type 2 diabetic patients.
Collapse
Affiliation(s)
- Darleen Sandoval
- Department of Surgery, University of Michigan, Ann Arbor, MI, United States
| | - Stephanie R Sisley
- Department of Pediatrics, Division of Nutrition, Baylor College of Medicine, Houston, TX, United States.
| |
Collapse
|
49
|
Outeiriño-Iglesias V, Romaní-Pérez M, González-Matías LC, Vigo E, Mallo F. GLP-1 Increases Preovulatory LH Source and the Number of Mature Follicles, As Well As Synchronizing the Onset of Puberty in Female Rats. Endocrinology 2015; 156:4226-37. [PMID: 26252058 DOI: 10.1210/en.2014-1978] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
Control of estrous cycle and reproductive capacity involves a large number of central and peripheral factors, integrating numerous nutritional and metabolic signals. Here we show that glucagon-like peptide-1 (GLP-1), a peptide with anorexigenic and insulinotropic actions, and the GLP-1 receptor agonist Exendin-4 (Ex4) exert a regulatory influence on the gonadal axis, in both adult and prepubertal female rats. In adult rats, Glp-1 receptor expression varies during the estrous cycle at the hypothalamus, pituitary, and ovary. Furthermore, acute treatment with GLP-1 in the morning proestrus doubled the amplitude of the preovulatory LH surge, as well as influencing estradiol and progesterone levels along the estrous cycle. These changes provoked an important increase in the number of Graafian follicles and corpora lutea, as well as in the litter size. Conversely, Ex4 diminished the levels of LH, later producing a partial blockade at the preovulatory surge, yet not affecting either the number of mature follicles or corpora lutea. Chronic administration of low doses of GLP-1 to prepubertal rats synchronized vaginal opening and increased LH levels on the 35th day of life, yet at these doses it did not modify their body weight, food intake, or ovarian and uterine weight. By contrast, chronic exposure to Ex4 produced a significant reduction in ovarian and uterine weight, and serum LH, and the animals treated chronically with Ex4 showed no vaginal opening in the period studied. Overall, our results demonstrate that GLP-1 and Ex4 act on the gonadal axis, involving the hypothalamic kisspeptin system, to influence reproductive efficiency in female rats.
Collapse
Affiliation(s)
- Verónica Outeiriño-Iglesias
- Laboratory of Endocrinology, Biomedical Research Centre (Centro Investigaciones Biomédicas), University of Vigo, Campus As Lagoas-Marcosende, E-36310 Vigo, Spain; and Institute for Biomedical Research of Vigo, E-36310 Vigo, Spain
| | - Marina Romaní-Pérez
- Laboratory of Endocrinology, Biomedical Research Centre (Centro Investigaciones Biomédicas), University of Vigo, Campus As Lagoas-Marcosende, E-36310 Vigo, Spain; and Institute for Biomedical Research of Vigo, E-36310 Vigo, Spain
| | - Lucas C González-Matías
- Laboratory of Endocrinology, Biomedical Research Centre (Centro Investigaciones Biomédicas), University of Vigo, Campus As Lagoas-Marcosende, E-36310 Vigo, Spain; and Institute for Biomedical Research of Vigo, E-36310 Vigo, Spain
| | - Eva Vigo
- Laboratory of Endocrinology, Biomedical Research Centre (Centro Investigaciones Biomédicas), University of Vigo, Campus As Lagoas-Marcosende, E-36310 Vigo, Spain; and Institute for Biomedical Research of Vigo, E-36310 Vigo, Spain
| | - Federico Mallo
- Laboratory of Endocrinology, Biomedical Research Centre (Centro Investigaciones Biomédicas), University of Vigo, Campus As Lagoas-Marcosende, E-36310 Vigo, Spain; and Institute for Biomedical Research of Vigo, E-36310 Vigo, Spain
| |
Collapse
|
50
|
Romaní-Pérez M, Outeiriño-Iglesias V, Moya CM, Santisteban P, González-Matías LC, Vigo E, Mallo F. Activation of the GLP-1 Receptor by Liraglutide Increases ACE2 Expression, Reversing Right Ventricle Hypertrophy, and Improving the Production of SP-A and SP-B in the Lungs of Type 1 Diabetes Rats. Endocrinology 2015. [PMID: 26196539 DOI: 10.1210/en.2014-1685] [Citation(s) in RCA: 136] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Diabetes alters microvascular function in the vascular beds of organs, including the lungs. Cardiovascular complications of pulmonary vascular affectation may be a consequence of the overactivation of the vasoconstrictive and proliferative components of the renin-angiotensin system. We previously reported that pulmonary physiology and surfactant production is improved by the glucagon-like peptide 1 receptor (GLP-1R) agonist liraglutide (LIR) in a rat model of lung hypoplasia. Because we hypothesized that streptozotocin-induced diabetes rats would show deficiencies in lung function, including surfactant proteins, and develop an imbalance of the renin-angiotensin system in the lungs. This effect would in turn be prevented by long-acting agonists of the GLP-1R, such as LIR. The induction of diabetes reduced the surfactant protein A and B in the lungs and caused the vasoconstrictor component of the renin-angiotensin system to predominate, which in turn increased angiotensin II levels, and ultimately being associated with right ventricle hypertrophy. LIR restored surfactant protein levels and reversed the imbalance in the renin-angiotensin system in this type 1 diabetes mellitus rat model. Moreover, LIR provoked a strong increase in angiotensin-converting enzyme 2 expression in the lungs of both diabetic and control rats, and in the circulating angiotensin(1-7) in diabetic animals. These effects prompted complete reversion of right ventricle hypertrophy. The consequences of LIR administration were independent of glycemic control and of glucocorticoids, and they involved NK2 homeobox 1 signaling. This study demonstrates by first time that GLP-1R agonists, such as LIR, might improve the cardiopulmonary complications associated with diabetes.
Collapse
Affiliation(s)
- Marina Romaní-Pérez
- Laboratory of Endocrinology (M.R.-P., V.O.-I., L.C.G.-M., E.V., F.M.), Centre for Biomedical Research (CINBIO), University of Vigo, Faculty of Biology, E-36310 Vigo, Spain; Institute for Biomedical Research of Vigo (IBIV) (M.R.-P., V.O.-I., L.C.G.-M., E.V., F.M.), University of Vigo/Sergas, E36310, Vigo, Spain; and Institute Biomedical Research Alberto Sols (C.M.M., P.S.), Spanish Council of Research, Universidad Autónoma de Madrid, Madrid E28029, Spain
| | - Verónica Outeiriño-Iglesias
- Laboratory of Endocrinology (M.R.-P., V.O.-I., L.C.G.-M., E.V., F.M.), Centre for Biomedical Research (CINBIO), University of Vigo, Faculty of Biology, E-36310 Vigo, Spain; Institute for Biomedical Research of Vigo (IBIV) (M.R.-P., V.O.-I., L.C.G.-M., E.V., F.M.), University of Vigo/Sergas, E36310, Vigo, Spain; and Institute Biomedical Research Alberto Sols (C.M.M., P.S.), Spanish Council of Research, Universidad Autónoma de Madrid, Madrid E28029, Spain
| | - Christian M Moya
- Laboratory of Endocrinology (M.R.-P., V.O.-I., L.C.G.-M., E.V., F.M.), Centre for Biomedical Research (CINBIO), University of Vigo, Faculty of Biology, E-36310 Vigo, Spain; Institute for Biomedical Research of Vigo (IBIV) (M.R.-P., V.O.-I., L.C.G.-M., E.V., F.M.), University of Vigo/Sergas, E36310, Vigo, Spain; and Institute Biomedical Research Alberto Sols (C.M.M., P.S.), Spanish Council of Research, Universidad Autónoma de Madrid, Madrid E28029, Spain
| | - Pilar Santisteban
- Laboratory of Endocrinology (M.R.-P., V.O.-I., L.C.G.-M., E.V., F.M.), Centre for Biomedical Research (CINBIO), University of Vigo, Faculty of Biology, E-36310 Vigo, Spain; Institute for Biomedical Research of Vigo (IBIV) (M.R.-P., V.O.-I., L.C.G.-M., E.V., F.M.), University of Vigo/Sergas, E36310, Vigo, Spain; and Institute Biomedical Research Alberto Sols (C.M.M., P.S.), Spanish Council of Research, Universidad Autónoma de Madrid, Madrid E28029, Spain
| | - Lucas C González-Matías
- Laboratory of Endocrinology (M.R.-P., V.O.-I., L.C.G.-M., E.V., F.M.), Centre for Biomedical Research (CINBIO), University of Vigo, Faculty of Biology, E-36310 Vigo, Spain; Institute for Biomedical Research of Vigo (IBIV) (M.R.-P., V.O.-I., L.C.G.-M., E.V., F.M.), University of Vigo/Sergas, E36310, Vigo, Spain; and Institute Biomedical Research Alberto Sols (C.M.M., P.S.), Spanish Council of Research, Universidad Autónoma de Madrid, Madrid E28029, Spain
| | - Eva Vigo
- Laboratory of Endocrinology (M.R.-P., V.O.-I., L.C.G.-M., E.V., F.M.), Centre for Biomedical Research (CINBIO), University of Vigo, Faculty of Biology, E-36310 Vigo, Spain; Institute for Biomedical Research of Vigo (IBIV) (M.R.-P., V.O.-I., L.C.G.-M., E.V., F.M.), University of Vigo/Sergas, E36310, Vigo, Spain; and Institute Biomedical Research Alberto Sols (C.M.M., P.S.), Spanish Council of Research, Universidad Autónoma de Madrid, Madrid E28029, Spain
| | - Federico Mallo
- Laboratory of Endocrinology (M.R.-P., V.O.-I., L.C.G.-M., E.V., F.M.), Centre for Biomedical Research (CINBIO), University of Vigo, Faculty of Biology, E-36310 Vigo, Spain; Institute for Biomedical Research of Vigo (IBIV) (M.R.-P., V.O.-I., L.C.G.-M., E.V., F.M.), University of Vigo/Sergas, E36310, Vigo, Spain; and Institute Biomedical Research Alberto Sols (C.M.M., P.S.), Spanish Council of Research, Universidad Autónoma de Madrid, Madrid E28029, Spain
| |
Collapse
|