1
|
Nestor LJ, Ersche KD. Gut Hormones: Possible Mediators of Addictive Disorders? Eur Addict Res 2024:1-8. [PMID: 39389039 DOI: 10.1159/000540743] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/16/2024] [Accepted: 08/02/2024] [Indexed: 10/12/2024]
Abstract
BACKGROUND Alcohol and drug dependence are major health and economic burdens to society. One of the major challenges to reducing this burden will be to develop more effective and better tolerated medications that target alternative mechanisms in the brain. While the dopamine system has been well characterized for mediating the reward value of drugs, there is evidence that the endocrine system also conveys signals to the same neural systems using gut hormones. SUMMARY These gut hormones, produced in the stomach and intestine and that regulate food intake, have also been shown to control the use of other substances, such as alcohol and drugs of abuse. Examples of such hormones are ghrelin and glucagon-like peptide-1, which exert their effects on dopamine transmission in parts of the brain known to be involved in some of the core features of addiction, such as reward sensitivity. KEY MESSAGES This raises the possibility that gut hormone systems may play a pivotal role in addictive disorders. This review will briefly outline emerging evidence that the ghrelin and glucagon-like peptide-1 hormones are contrasting mediators of alcohol and drug use and may present a promising alternative target for treatment intervention in addictive disorders.
Collapse
Affiliation(s)
- Liam J Nestor
- Department of Psychiatry, University of Cambridge, Cambridge, UK
| | - Karen D Ersche
- Department of Psychiatry, University of Cambridge, Cambridge, UK
- Department of Systems Neuroscience, University Medical Centre Hamburg-Eppendorf, Hamburg, Germany
- Department of Addictive Behaviour and Addiction Medicine, Central Institute of Mental Health, University of Heidelberg, Mannheim, Germany
| |
Collapse
|
2
|
Chen B, Yu X, Horvath-Diano C, Ortuño MJ, Tschöp MH, Jastreboff AM, Schneeberger M. GLP-1 programs the neurovascular landscape. Cell Metab 2024; 36:2173-2189. [PMID: 39357509 DOI: 10.1016/j.cmet.2024.09.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/26/2024] [Revised: 09/06/2024] [Accepted: 09/06/2024] [Indexed: 10/04/2024]
Abstract
Readily available nutrient-rich foods exploit our inherent drive to overconsume, creating an environment of overnutrition. This transformative setting has led to persistent health issues, such as obesity and metabolic syndrome. The development of glucagon-like peptide-1 receptor (GLP-1R) agonists reveals our ability to pharmacologically manage weight and address metabolic conditions. Obesity is directly linked to chronic low-grade inflammation, connecting our metabolic environment to neurodegenerative diseases. GLP-1R agonism in curbing obesity, achieved by impacting appetite and addressing associated metabolic defects, is revealing additional benefits extending beyond weight loss. Whether GLP-1R agonism directly impacts brain health or does so indirectly through improved metabolic health remains to be elucidated. In exploring the intricate connection between obesity and neurological conditions, recent literature suggests that GLP-1R agonism may have the capacity to shape the neurovascular landscape. Thus, GLP-1R agonism emerges as a promising strategy for addressing the complex interplay between metabolic health and cognitive well-being.
Collapse
Affiliation(s)
- Bandy Chen
- Laboratory of Neurovascular Control of Homeostasis, Department of Cellular and Molecular Physiology, Yale University School of Medicine, New Haven, CT, USA.
| | - Xiaofei Yu
- State Key Laboratory of Genetic Engineering, School of Life Sciences, Fudan University, Shanghai, China
| | - Claudia Horvath-Diano
- Departments of Medicine (Endocrinology & Metabolism) and Pediatrics (Pediatric Endocrinology), Yale University School of Medicine, New Haven, CT, USA
| | - María José Ortuño
- Department of Genetics, Yale University School of Medicine, New Haven, CT, USA
| | - Matthias H Tschöp
- Helmholtz Zentrum München, Neuherberg, Germany; Division of Metabolic Diseases, Department of Medicine, Technische Universität München, München, Germany
| | - Ania M Jastreboff
- Departments of Medicine (Endocrinology & Metabolism) and Pediatrics (Pediatric Endocrinology), Yale University School of Medicine, New Haven, CT, USA
| | - Marc Schneeberger
- Laboratory of Neurovascular Control of Homeostasis, Department of Cellular and Molecular Physiology, Yale University School of Medicine, New Haven, CT, USA; Wu Tsai Institute for Mind and Brain, Yale University, New Haven, CT, USA.
| |
Collapse
|
3
|
Bruns Vi N, Tressler EH, Vendruscolo LF, Leggio L, Farokhnia M. IUPHAR review - Glucagon-like peptide-1 (GLP-1) and substance use disorders: An emerging pharmacotherapeutic target. Pharmacol Res 2024; 207:107312. [PMID: 39032839 PMCID: PMC11467891 DOI: 10.1016/j.phrs.2024.107312] [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: 06/14/2024] [Revised: 07/10/2024] [Accepted: 07/15/2024] [Indexed: 07/23/2024]
Abstract
Addiction is a chronic relapsing disease with high morbidity and mortality. Treatments for addiction include pharmacological and psychosocial interventions; however, currently available medications are limited in number and efficacy. The glucagon-like-peptide-1 (GLP-1) system is emerging as a potential novel pharmacotherapeutic target for alcohol and other substance use disorders (ASUDs). In this review, we summarize and discuss the wealth of available evidence from testing GLP-1 receptor (GLP-1R) agonist medications in preclinical models and humans with ASUDs, possible mechanisms underlying the impact of GLP-1R agonists on alcohol/substance use, gaps in knowledge, and future directions. Most of the research with GLP-1R agonists has been conducted in relation to alcohol use; psychostimulants, opioids, and nicotine have also been investigated. Preclinical evidence suggests that GLP-1R agonists reduce alcohol/substance use and other related outcomes. The main proposed mechanisms are related to reward processing, stress, and cognitive function, as well as broader mechanisms related to satiety, changes in gastric motility, and glucose homeostasis. More in-depth mechanistic studies are warranted. Clinical studies have been limited and their findings have been less conclusive; however, most support the safety and potential efficacy of GLP-1R agonists in ASUD treatment. Identifying preferred compounds, as well as possible subgroups who are most responsive to GLP-1R agonists are some of the key research questions to translate the promising preclinical data into clinical settings. Several clinical trials are underway to test GLP-1R agonists in people with ASUDs.
Collapse
Affiliation(s)
- Nicolaus Bruns Vi
- Clinical Psychoneuroendocrinology and Neuropsychopharmacology Section, Translational Addiction Medicine Branch, National Institute on Drug Abuse Intramural Research Program and National Institute on Alcohol Abuse and Alcoholism Division of Intramural Clinical and Biological Research, National Institutes of Health, Baltimore, MD, USA; Neurobiology of Addiction Section, Integrative Neuroscience Research Branch, National Institute on Drug Abuse Intramural Research Program, National Institutes of Health, Baltimore, MD, USA; Stress & Addiction Neuroscience Unit, Integrative Neuroscience Research Branch, National Institute on Drug Abuse Intramural Research Program and National Institute on Alcohol Abuse and Alcoholism Division of Intramural Clinical and Biological Research, National Institutes of Health, Baltimore, MD, USA
| | - Elizabeth H Tressler
- Clinical Psychoneuroendocrinology and Neuropsychopharmacology Section, Translational Addiction Medicine Branch, National Institute on Drug Abuse Intramural Research Program and National Institute on Alcohol Abuse and Alcoholism Division of Intramural Clinical and Biological Research, National Institutes of Health, Baltimore, MD, USA; Neurobiology of Addiction Section, Integrative Neuroscience Research Branch, National Institute on Drug Abuse Intramural Research Program, National Institutes of Health, Baltimore, MD, USA; Stress & Addiction Neuroscience Unit, Integrative Neuroscience Research Branch, National Institute on Drug Abuse Intramural Research Program and National Institute on Alcohol Abuse and Alcoholism Division of Intramural Clinical and Biological Research, National Institutes of Health, Baltimore, MD, USA
| | - Leandro F Vendruscolo
- Stress & Addiction Neuroscience Unit, Integrative Neuroscience Research Branch, National Institute on Drug Abuse Intramural Research Program and National Institute on Alcohol Abuse and Alcoholism Division of Intramural Clinical and Biological Research, National Institutes of Health, Baltimore, MD, USA
| | - Lorenzo Leggio
- Clinical Psychoneuroendocrinology and Neuropsychopharmacology Section, Translational Addiction Medicine Branch, National Institute on Drug Abuse Intramural Research Program and National Institute on Alcohol Abuse and Alcoholism Division of Intramural Clinical and Biological Research, National Institutes of Health, Baltimore, MD, USA.
| | - Mehdi Farokhnia
- Clinical Psychoneuroendocrinology and Neuropsychopharmacology Section, Translational Addiction Medicine Branch, National Institute on Drug Abuse Intramural Research Program and National Institute on Alcohol Abuse and Alcoholism Division of Intramural Clinical and Biological Research, National Institutes of Health, Baltimore, MD, USA.
| |
Collapse
|
4
|
Lengsfeld S, Probst L, Emara Y, Werlen L, Vogt DR, Bathelt C, Baur F, Caviezel B, Vukajlovic T, Fischer M, Winzeler B. Effects of the glucagon-like peptide-1 receptor agonist dulaglutide on sexuality in healthy men: a randomised, double-blind, placebo-controlled crossover study. EBioMedicine 2024; 107:105284. [PMID: 39232425 PMCID: PMC11404067 DOI: 10.1016/j.ebiom.2024.105284] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2024] [Revised: 07/31/2024] [Accepted: 07/31/2024] [Indexed: 09/06/2024] Open
Abstract
BACKGROUND The reward-regulatory properties of GLP-1 are attracting increasing interest. Animal studies show that GLP-1 receptor agonists not only reduce consumption of addictive substances, but also influence sexual behaviour. We aimed to investigate the effect of dulaglutide versus placebo on sexual desire in humans. METHODS In this randomised, double-blind, placebo-controlled crossover trial, healthy eugonadal men of normal weight, aged 18-50 years with active and satisfactory sex lifes were (1:1) randomly allocated to dulaglutide or placebo for four weeks. We assessed sexual desire (Massachusetts General Hospital-Sexual Functioning Questionnaire [MGH-SFQ]), hormones of the hypothalamic-pituitary-gonadal axis (total testosterone, follicle-stimulating hormone [FSH], luteinizing hormone [LH]) and sperm parameters. Changes in these parameters were compared under dulaglutide versus placebo using paired t-tests. FINDINGS 24 out of 26 randomised participants completed the study (13 participants randomised to dulaglutide first and 13 to placebo first). No change in the MGH-SFQ was observed after four weeks of dulaglutide versus placebo (estimated difference 0.58 [95% CI -0.83 to 2.00], p-value = 0.402). Hormones of the hypothalamic-pituitary-gonadal axis (estimated differences: total testosterone (nmol/l) 0.9 [95% CI -1.5 to 3.3], FSH (IU/l) -0.2 [95% CI -0.3 to 0.0] and LH (IU/l) -0.8 [95% CI -1.5 to 0.0]) as well as sperm parameters all remained in the normal range without significant differences between the treatments. No severe adverse events occurred. INTERPRETATION In this study of healthy men, we found no evidence of negative impacts of a four-week treatment with the widely used GLP-1 receptor agonist dulaglutide on sexual desire, hypothalamic-pituitary-gonadal axis hormones or sperm parameters. FUNDING Swiss National Science Foundation (PZ00P3_193206), Gottfried and Julia Bangerter-Rhyner Foundation, Goldschmidt-Jacobson Foundation, Swiss Academy of Medical Sciences.
Collapse
Affiliation(s)
- Sophia Lengsfeld
- Department of Endocrinology, Diabetology and Metabolism, University Hospital Basel, Petersgraben 4, 4031 Basel, Switzerland; Department of Clinical Research, University Hospital Basel, University of Basel, Spitalstrasse 8/12, 4031 Basel, Switzerland
| | - Leila Probst
- Department of Endocrinology, Diabetology and Metabolism, University Hospital Basel, Petersgraben 4, 4031 Basel, Switzerland; Department of Clinical Research, University Hospital Basel, University of Basel, Spitalstrasse 8/12, 4031 Basel, Switzerland
| | - Yara Emara
- Department of Endocrinology, Diabetology and Metabolism, University Hospital Basel, Petersgraben 4, 4031 Basel, Switzerland; Department of Clinical Research, University Hospital Basel, University of Basel, Spitalstrasse 8/12, 4031 Basel, Switzerland
| | - Laura Werlen
- Department of Clinical Research, University Hospital Basel, University of Basel, Spitalstrasse 8/12, 4031 Basel, Switzerland
| | - Deborah R Vogt
- Department of Clinical Research, University Hospital Basel, University of Basel, Spitalstrasse 8/12, 4031 Basel, Switzerland
| | - Cemile Bathelt
- Department of Endocrinology, Diabetology and Metabolism, University Hospital Basel, Petersgraben 4, 4031 Basel, Switzerland; Department of Clinical Research, University Hospital Basel, University of Basel, Spitalstrasse 8/12, 4031 Basel, Switzerland
| | - Fabienne Baur
- Department of Endocrinology, Diabetology and Metabolism, University Hospital Basel, Petersgraben 4, 4031 Basel, Switzerland; Department of Clinical Research, University Hospital Basel, University of Basel, Spitalstrasse 8/12, 4031 Basel, Switzerland
| | - Brida Caviezel
- Department of Endocrinology, Diabetology and Metabolism, University Hospital Basel, Petersgraben 4, 4031 Basel, Switzerland; Department of Clinical Research, University Hospital Basel, University of Basel, Spitalstrasse 8/12, 4031 Basel, Switzerland
| | - Tanja Vukajlovic
- Department of Endocrinology, Diabetology and Metabolism, University Hospital Basel, Petersgraben 4, 4031 Basel, Switzerland; Department of Clinical Research, University Hospital Basel, University of Basel, Spitalstrasse 8/12, 4031 Basel, Switzerland
| | - Manuel Fischer
- Reproductive Medicine and Gynecological Endocrinology (RME), Women's Health Clinic, University Hospital Basel, Vogesenstrasse 134, 4031 Basel, Switzerland
| | - Bettina Winzeler
- Department of Endocrinology, Diabetology and Metabolism, University Hospital Basel, Petersgraben 4, 4031 Basel, Switzerland; Department of Clinical Research, University Hospital Basel, University of Basel, Spitalstrasse 8/12, 4031 Basel, Switzerland.
| |
Collapse
|
5
|
Fink-Jensen A, Wörtwein G, Klausen MK, Holst JJ, Hartmann B, Thomsen M, Ptito M, Beierschmitt A, Palmour RM. Effect of the glucagon-like peptide-1 (GLP-1) receptor agonist semaglutide on alcohol consumption in alcohol-preferring male vervet monkeys. Psychopharmacology (Berl) 2024:10.1007/s00213-024-06637-2. [PMID: 38884652 DOI: 10.1007/s00213-024-06637-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/12/2024] [Accepted: 05/20/2024] [Indexed: 06/18/2024]
Abstract
RATIONALE Glucagon-like peptide-1 (GLP-1) receptor agonists reduce alcohol consumption in rodents and non-human primates. Semaglutide is a new long-acting GLP-1 receptor agonist, widely used in the clinic against type 2 diabetes and obesity. It is also reported to reduce alcohol intake in rodents. OBJECTIVES This study investigates the possible inhibitory effect of semaglutide on alcohol intake in alcohol-preferring African green monkeys. METHODS We performed a vehicle-controlled study on male monkeys that had demonstrated a preference for alcohol. In the monkeys selected for voluntary alcohol drinking, alcohol consumption was measured for ten days at baseline (Monday to Friday for two weeks). During this period, the monkeys had access to alcohol 4 h per day and free access to water 24 h per day. After two weeks of baseline measurements, the monkeys were randomized to semaglutide or vehicle. Each group consisted of ten monkeys, and the two groups were balanced with respect to baseline alcohol intake. Following the baseline period, the monkeys were treated with escalating doses of semaglutide (up to 0.05 mg/kg) or vehicle subcutaneously twice weekly for two weeks during which period alcohol was not available. After uptitration, the monkeys had access to alcohol 4 h daily for 20 days (Monday to Friday for 4 weeks), and alcohol consumption was measured. During this alcohol exposure period, treatment with semaglutide (0.05 mg/kg twice weekly) or vehicle continued for three weeks followed by a one-week washout period. RESULTS Compared to the vehicle, semaglutide significantly reduced alcohol intake. There were no signs of emetic events or changes in water intake. CONCLUSIONS These data demonstrate for the first time the potent effect of semaglutide in reducing voluntary alcohol intake in non-human primates and further substantiate the need for clinical trials investigating the effect of semaglutide in patients with alcohol-use disorder.
Collapse
Affiliation(s)
- Anders Fink-Jensen
- Laboratory of Neuropsychiatry, Mental Health Centre Copenhagen, Mental Health Services in the Capital Region of Denmark, University of Copenhagen, Hovedvejen 17, Frederiksberg, DK-2000, Denmark.
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark.
| | - Gitta Wörtwein
- Laboratory of Neuropsychiatry, Mental Health Centre Copenhagen, Mental Health Services in the Capital Region of Denmark, University of Copenhagen, Hovedvejen 17, Frederiksberg, DK-2000, Denmark
- Department of Public Health, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Mette Kruse Klausen
- Laboratory of Neuropsychiatry, Mental Health Centre Copenhagen, Mental Health Services in the Capital Region of Denmark, University of Copenhagen, Hovedvejen 17, Frederiksberg, DK-2000, Denmark
| | - Jens Juul Holst
- Novo Nordisk Foundation Center for Basic Metabolic Research, Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Bolette Hartmann
- Novo Nordisk Foundation Center for Basic Metabolic Research, Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Morgan Thomsen
- Laboratory of Neuropsychiatry, Mental Health Centre Copenhagen, Mental Health Services in the Capital Region of Denmark, University of Copenhagen, Hovedvejen 17, Frederiksberg, DK-2000, Denmark
- Department of Neuroscience, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Maurice Ptito
- Department of Neuroscience, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- School of Optometry, Université de Montréal, Montréal (Qc), Canada
| | - Amy Beierschmitt
- Behavioural Science Foundation, St. Kitts, Saint Kitts and Nevis
| | - Roberta M Palmour
- Behavioural Science Foundation, St. Kitts, Saint Kitts and Nevis
- Faculty of Medicine and Health Sciences, McGill University, Montréal (Qc), Canada
| |
Collapse
|
6
|
Claro AE, Palanza C, Mazza M, Rizzi A, Corsello A, Tartaglione L, Marano G, Muti Schuenemann GEU, Rigoni M, Pontecorvi A, Janiri L, Muti P, Pitocco D. Reconsidering the role of depression and common psychiatric disorders as partners in the type 2 diabetes epidemic. World J Diabetes 2024; 15:1374-1380. [PMID: 38983820 PMCID: PMC11229977 DOI: 10.4239/wjd.v15.i6.1374] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/27/2024] [Revised: 02/27/2024] [Accepted: 04/18/2024] [Indexed: 06/11/2024] Open
Abstract
Common psychiatric disorders (CPDs) and depression contribute significantly to the global epidemic of type 2 diabetes (T2D). We postulated a possible pathophysiological mechanism that through Bridge-Symptoms present in depression and CPDs, promotes the establishment of emotional eating, activation of the reward system, onset of overweight and obesity and, ultimately the increased risk of developing T2D. The plausibility of the proposed pathophysiological mechanism is supported by the mechanism of action of drugs such as naltrexone-bupropion currently approved for the treatment of both obesity/overweight with T2D and as separate active pharmaceutical ingredients in drug addiction, but also from initial evidence that is emerging regarding glucagon-like peptide 1 receptor agonists that appear to be effective in the treatment of drug addiction. We hope that our hypothesis may be useful in interpreting the higher prevalence of CPDs and depression in patients with T2D compared with the general population and may help refine the integrated psychiatric-diabetic therapy approach to improve the treatment and or remission of T2D.
Collapse
Affiliation(s)
- Angelo Emilio Claro
- Diabetes Care Unit, Department of Medical and Surgical Sciences, Fondazione Policlinico Universitario A Gemelli IRCCS, Rome 00168, Italy
- Department of Biomedical, Surgical and Dental Sciences, University of Milan, Milan 20122, Italy
| | - Clelia Palanza
- Istituto Italiano di Antropologia, ISItA, Rome 00100, Italy
| | - Marianna Mazza
- Unit of Psychiatry, Department of Neurosciences, Fondazione Policlinico Universitario A Gemelli IRCCS, Università Cattolica del Sacro Cuore, Rome 00168, Italy
| | - Alessandro Rizzi
- Diabetes Care Unit, Department of Medical and Surgical Sciences, Fondazione Policlinico Universitario A Gemelli IRCCS, Rome 00168, Italy
| | - Andrea Corsello
- Dipartimento di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, Rome 00168, Italy
| | - Linda Tartaglione
- Diabetes Care Unit, Department of Medical and Surgical Sciences, Fondazione Policlinico Universitario A Gemelli IRCCS, Rome 00168, Italy
| | - Giuseppe Marano
- Unit of Psychiatry, Department of Neurosciences, Fondazione Policlinico Universitario A Gemelli IRCCS, Università Cattolica del Sacro Cuore, Rome 00168, Italy
| | | | - Marta Rigoni
- Department of Biomedical, Surgical and Dental Sciences, University of Milan, Milan 20122, Italy
| | - Alfredo Pontecorvi
- Department of Endocrine-Metabolic and Dermo-Rheumatology, Fondazione Policlinico Universitario A Gemelli IRCCS, Università Cattolica del Sacro Cuore, Rome 00168, Italy
| | - Luigi Janiri
- Unit of Psychiatry, Department of Neurosciences, Fondazione Policlinico Universitario A Gemelli IRCCS, Università Cattolica del Sacro Cuore, Rome 00168, Italy
| | - Paola Muti
- Department of Biomedical, Surgical and Dental Sciences, University of Milan, Milan 20122, Italy
| | - Dario Pitocco
- Diabetes Care Unit, Department of Medical and Surgical Sciences, Fondazione Policlinico Universitario A Gemelli IRCCS, Rome 00168, Italy
| |
Collapse
|
7
|
Heilig M, Witkiewitz K, Ray LA, Leggio L. Novel medications for problematic alcohol use. J Clin Invest 2024; 134:e172889. [PMID: 38828724 PMCID: PMC11142745 DOI: 10.1172/jci172889] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/05/2024] Open
Abstract
Alcohol-related harm, a major cause of disease burden globally, affects people along a spectrum of use. When a harmful pattern of drinking is present in the absence of significant behavioral pathology, low-intensity brief interventions that provide information about health consequences of continued use provide large health benefits. At the other end of the spectrum, profound behavioral pathology, including continued use despite knowledge of potentially fatal consequences, warrants a medical diagnosis, and treatment is strongly indicated. Available behavioral and pharmacological treatments are supported by scientific evidence but are vastly underutilized. Discovery of additional medications, with a favorable balance of efficacy versus safety and tolerability can improve clinical uptake of treatment, allow personalized treatment, and improve outcomes. Here, we delineate the clinical conditions when pharmacotherapy should be considered in relation to the main diagnostic systems in use and discuss clinical endpoints that represent meaningful clinical benefits. We then review specific developments in three categories of targets that show promise for expanding the treatment toolkit. GPCRs remain the largest category of successful drug targets across contemporary medicine, and several GPCR targets are currently pursued for alcohol-related indications. Endocrine systems are another established category, and several promising targets have emerged for alcohol indications. Finally, immune modulators have revolutionized treatment of multiple medical conditions, and they may also hold potential to produce benefits in patients with alcohol problems.
Collapse
Affiliation(s)
- Markus Heilig
- Center for Social and Affective Neuroscience, Linköping University, and Department of Psychiatry, Linköping University Hospital, Linköping, Sweden
| | - Katie Witkiewitz
- Department of Psychology and Center on Alcohol, Substance Use and Addictions, University of New Mexico, Albuquerque, New Mexico, USA
| | - Lara A. Ray
- Department of Psychology, UCLA, Los Angeles, California, USA
| | - Lorenzo Leggio
- Clinical Psychoneuroendocrinology and Neuropsychopharmacology Section, Translational Addiction Medicine Branch, National Institute on Drug Abuse Intramural Research Program and National Institute on Alcohol Abuse and Alcoholism Division of Intramural Clinical and Biological Research, NIH, Baltimore and Bethesda, Maryland, USA
| |
Collapse
|
8
|
Dumiaty Y, Underwood BM, Phy-Lim J, Chee MJ. Neurocircuitry underlying the actions of glucagon-like peptide 1 and peptide YY 3-36 in the suppression of food, drug-seeking, and anxiogenesis. Neuropeptides 2024; 105:102427. [PMID: 38579490 DOI: 10.1016/j.npep.2024.102427] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/23/2024] [Revised: 03/27/2024] [Accepted: 03/29/2024] [Indexed: 04/07/2024]
Abstract
Obesity is a critical health condition worldwide that increases the risks of comorbid chronic diseases, but it can be managed with weight loss. However, conventional interventions relying on diet and exercise are inadequate for achieving and maintaining weight loss, thus there is significant market interest for pharmaceutical anti-obesity agents. For decades, receptor agonists for the gut peptide glucagon-like peptide 1 (GLP-1) featured prominently in anti-obesity medications by suppressing appetite and food reward to elicit rapid weight loss. As the neurocircuitry underlying food motivation overlaps with that for drugs of abuse, GLP-1 receptor agonism has also been shown to decrease substance use and relapse, thus its therapeutic potential may extend beyond weight management to treat addictions. However, as prolonged use of anti-obesity drugs may increase the risk of mood-related disorders like anxiety and depression, and individuals taking GLP-1-based medication commonly report feeling demotivated, the long-term safety of such drugs is an ongoing concern. Interestingly, current research now focuses on dual agonist approaches that include GLP-1 receptor agonism to enable synergistic effects on weight loss or associated functions. GLP-1 is secreted from the same intestinal cells as the anorectic gut peptide, Peptide YY3-36 (PYY3-36), thus this review assessed the therapeutic potential and underlying neural circuits targeted by PYY3-36 when administered independently or in combination with GLP-1 to curb the appetite for food or drugs of abuse like opiates, alcohol, and nicotine. Additionally, we also reviewed animal and human studies to assess the impact, if any, for GLP-1 and/or PYY3-36 on mood-related behaviors in relation to anxiety and depression. As dual agonists targeting GLP-1 and PYY3-36 may produce synergistic effects, they can be effective at lower doses and offer an alternative approach for therapeutic benefits while mitigating undesirable side effects.
Collapse
Affiliation(s)
- Yasmina Dumiaty
- Department of Neuroscience, Carleton University, 1125 Colonel By Drive, Ottawa, ON K1S 5B6, Canada.
| | - Brett M Underwood
- Department of Neuroscience, Carleton University, 1125 Colonel By Drive, Ottawa, ON K1S 5B6, Canada.
| | - Jenny Phy-Lim
- Department of Neuroscience, Carleton University, 1125 Colonel By Drive, Ottawa, ON K1S 5B6, Canada.
| | - Melissa J Chee
- Department of Neuroscience, Carleton University, 1125 Colonel By Drive, Ottawa, ON K1S 5B6, Canada.
| |
Collapse
|
9
|
Liu W, Wang Z, Wang W, Wang Z, Xing Y, Hölscher C. Liraglutide Reduces Alcohol Consumption, Anxiety, Memory Impairment, and Synapse Loss in Alcohol Dependent Mice. Neurochem Res 2024; 49:1061-1075. [PMID: 38267691 DOI: 10.1007/s11064-023-04093-6] [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: 08/18/2023] [Revised: 12/02/2023] [Accepted: 12/22/2023] [Indexed: 01/26/2024]
Abstract
Glucagon-like peptide 1 (GLP-1) analogues have been commercialized for the management of type 2 diabetes. Recent studies have underscored GLP-1's role as a modulator of alcohol-related behavior. However, the role of the GLP-1 analogue liraglutide on alcohol-withdrawal responses have not been fully elucidated. Liraglutide binds to the G-protein-coupled receptor and activates an adenylyl cyclase and the associated classic growth factor signaling pathway, which acts growth factor-like and neuroprotective properties. The underlying neurobiological mechanisms of liraglutide on alcohol withdrawal remains unknown. This study endeavored to explore the effects of liraglutide on the emotion and memory ability of alcohol-withdrawal mice, and synaptic morphology in the medial prefrontal cortex (mPFC) and the hippocampus (HP), and thus affects the relapse-like drinking of alcohol-withdrawal mice. The alcohol-withdrawal group was reintroduced to a 20% v/v alcohol and water through the two-bottle choice for four consecutive days, a period referred to as alcohol re-drinking. Male C57BL/6J mice were exposed to a regimen of 20% alcohol and water for a duration of 6 weeks. This regimen established the two-bottle choice model of alcohol exposure. Learning capabilities, memory proficiency, and anxiety-like behavior were evaluated using the Morris water maze, open field, and elevated plus maze paradigms. Furthermore, synaptic morphology and the levels of synaptic transport-related proteins were assessed via Golgi staining and Western Blot analysis after a two-week alcohol deprivation period. Alcohol re-drinking of alcohol-withdrawal mice was also evaluated using a two-bottle choice paradigm. Our findings indicate that liraglutide can substantially decrease alcohol consumption and preference (p < 0.05) in the alcohol group and enhance learning and memory performance (p < 0.01), as well as alleviate anxiety-like behavior (p < 0.01) of alcohol-withdrawal mice. Alcohol consumption led to a reduction in dendritic spine density in the mPFC and HP, which was restored to normal levels by liraglutide (p < 0.001). Furthermore, liraglutide was found to augment the levels of synaptic transport-related proteins in mice subjected to alcohol withdrawal (p < 0.01). The study findings corroborate that liraglutide has the potential to mitigate alcohol consumption and ameliorate the memory impairments and anxiety induced by alcohol withdrawal. The therapeutic efficacy of liraglutide might be attributed to its role in counteracting synapse loss in the mPFC and HP regions and thus prevented relapse-like drinking in alcohol-withdrawal mice.
Collapse
Affiliation(s)
- Weizhen Liu
- Department of Physiology and Neurobiology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, 450001, Henan, China
- The Academy of Medical Sciences of Zhengzhou University, Zhengzhou, 450001, Henan, China
| | - Ziliang Wang
- Department of Physiology and Neurobiology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, 450001, Henan, China
- The Academy of Medical Sciences of Zhengzhou University, Zhengzhou, 450001, Henan, China
| | - Wang Wang
- Department of Physiology and Neurobiology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, 450001, Henan, China
- The Academy of Medical Sciences of Zhengzhou University, Zhengzhou, 450001, Henan, China
| | - Zhiju Wang
- Department of Physiology and Neurobiology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, 450001, Henan, China
| | - Ying Xing
- Department of Physiology and Neurobiology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, 450001, Henan, China.
| | - Christian Hölscher
- Henan Academy of Innovation in Medical Science, XinZheng, 451100, Henan, China.
| |
Collapse
|
10
|
White B, Sirohi S. A Complex Interplay between Nutrition and Alcohol use Disorder: Implications for Breaking the Vicious Cycle. Curr Pharm Des 2024; 30:1822-1837. [PMID: 38797900 DOI: 10.2174/0113816128292367240510111746] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2023] [Revised: 04/24/2024] [Accepted: 04/25/2024] [Indexed: 05/29/2024]
Abstract
Approximately 16.5% of the United States population met the diagnostic criteria for substance use disorder (SUD) in 2021, including 29.5 million individuals with alcohol use disorder (AUD). Individuals with AUD are at increased risk for malnutrition, and impairments in nutritional status in chronic alcohol users can be detrimental to physical and emotional well-being. Furthermore, these nutritional deficiencies could contribute to the never-ending cycle of alcoholism and related pathologies, thereby jeopardizing the prospects of recovery and treatment outcomes. Improving nutritional status in AUD patients may not only compensate for general malnutrition but could also reduce adverse symptoms during recovery, thereby promoting abstinence and successful treatment of AUD. In this review, we briefly summarize alterations in the nutritional status of people with addictive disorders, in addition to the underlying neurobiological mechanisms and clinical implications regarding the role of nutritional intervention in recovery from alcohol use disorder.
Collapse
Affiliation(s)
- Brooke White
- Laboratory of Endocrine and Neuropsychiatric Disorders, Division of Basic Pharmaceutical Sciences, College of Pharmacy, Xavier University of Louisiana, New Orleans, LA 70125, USA
| | - Sunil Sirohi
- Laboratory of Endocrine and Neuropsychiatric Disorders, Division of Basic Pharmaceutical Sciences, College of Pharmacy, Xavier University of Louisiana, New Orleans, LA 70125, USA
| |
Collapse
|
11
|
Quddos F, Hubshman Z, Tegge A, Sane D, Marti E, Kablinger AS, Gatchalian KM, Kelly AL, DiFeliceantonio AG, Bickel WK. Semaglutide and Tirzepatide reduce alcohol consumption in individuals with obesity. Sci Rep 2023; 13:20998. [PMID: 38017205 PMCID: PMC10684505 DOI: 10.1038/s41598-023-48267-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2023] [Accepted: 11/24/2023] [Indexed: 11/30/2023] Open
Abstract
Alcohol Use Disorder (AUD) contributes significantly to global mortality. GLP-1 (Glucagon-like peptide-1) and GLP-1/GIP (Glucose-dependent Insulinotropic Polypeptide) agonists, FDA-approved for managing type 2 diabetes and obesity, where the former has shown to effectively reduce the consumption of alcohol in animal models but no reports exist on the latter. In this report, we conducted two studies. In the first study, we conducted an analysis of abundant social media texts. Specifically, a machine-learning based attribution mapping of ~ 68,250 posts related to GLP-1 or GLP-1/GIP agonists on the Reddit platform. Secondly, we recruited participants (n = 153; current alcohol drinkers; BMI ≥ 30) who self-reported either taking Semaglutide (GLP-1 agonist), Tirzepatide (the GLP-1/GIP combination) for ≥ 30 days or, as a control group; no medication to manage diabetes or weight loss for a within and between subject remote study. In the social media study, we report 8 major themes including effects of medications (30%); diabetes (21%); and Weight loss and obesity (19%). Among the alcohol-related posts (n = 1580), 71% were identified as craving reduction, decreased desire to drink, and other negative effects. In the remote study, we observe a significantly lower self-reported intake of alcohol, drinks per drinking episode, binge drinking odds, Alcohol Use Disorders Identification Test (AUDIT) scores, and stimulating, and sedative effects in the Semaglutide or Tirzepatide group when compared to prior to starting medication timepoint (within-subjects) and the control group (between-subjects). In summary, we provide initial real-world evidence of reduced alcohol consumption in people with obesity taking Semaglutide or Tirzepatide medications, suggesting potential efficacy for treatment in AUD comorbid with obesity.
Collapse
Affiliation(s)
- Fatima Quddos
- Fralin Biomedical Research Institute at VTC, Virginia Tech, Roanoke, VA, USA
- Graduate Program in Translational Biology, Medicine, and Health, Blacksburg, VA, USA
| | - Zachary Hubshman
- Fralin Biomedical Research Institute at VTC, Virginia Tech, Roanoke, VA, USA
- Virginia Polytechnic Institute and State University, Blacksburg, VA, USA
| | - Allison Tegge
- Fralin Biomedical Research Institute at VTC, Virginia Tech, Roanoke, VA, USA
| | - Daniel Sane
- Fralin Biomedical Research Institute at VTC, Virginia Tech, Roanoke, VA, USA
- Virginia Polytechnic Institute and State University, Blacksburg, VA, USA
| | - Erin Marti
- Fralin Biomedical Research Institute at VTC, Virginia Tech, Roanoke, VA, USA
| | | | | | - Amber L Kelly
- Fralin Biomedical Research Institute at VTC, Virginia Tech, Roanoke, VA, USA
- Graduate Program in Translational Biology, Medicine, and Health, Blacksburg, VA, USA
| | | | - Warren K Bickel
- Fralin Biomedical Research Institute at VTC, Virginia Tech, Roanoke, VA, USA.
| |
Collapse
|
12
|
Probst L, Monnerat S, Vogt DR, Lengsfeld S, Burkard T, Meienberg A, Bathelt C, Christ-Crain M, Winzeler B. Effects of dulaglutide on alcohol consumption during smoking cessation. JCI Insight 2023; 8:e170419. [PMID: 37991022 PMCID: PMC10721313 DOI: 10.1172/jci.insight.170419] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2023] [Accepted: 10/04/2023] [Indexed: 11/23/2023] Open
Abstract
BACKGROUNDAlcohol use disorder has a detrimental impact on global health and new treatment targets are needed. Preclinical studies show attenuating effects of glucagon-like peptide-1 (GLP-1) agonists on addiction-related behaviors in rodents and nonhuman primates. Some trials have shown an effect of GLP-1 agonism on reward processes in humans; however, results from clinical studies remain inconclusive.METHODSThis is a predefined secondary analysis of a double-blind, randomized, placebo-controlled trial evaluating the GLP-1 agonist dulaglutide as a therapy for smoking cessation. The main objective was to assess differences in alcohol consumption after 12 weeks of treatment with dulaglutide compared to placebo. The effect of dulaglutide on alcohol consumption was analyzed using a multivariable generalized linear model.RESULTSIn the primary analysis, participants out of the cohort (n = 255) who reported drinking alcohol at baseline and who completed 12 weeks of treatment (n = 151; placebo n = 75, dulaglutide n = 76) were included. The median age was 42 (IQR 33-53) with 61% (n = 92) females. At week 12, participants receiving dulaglutide drank 29% less (relative effect = 0.71, 95% CI 0.52-0.97, P = 0.04) than participants receiving placebo. Changes in alcohol consumption were not correlated with smoking status at week 12.CONCLUSIONThese results provide evidence that dulaglutide reduces alcohol intake in humans and contribute to the growing body of literature promoting the use of GLP-1 agonists in treatment of substance use disorders.TRIAL REGISTRATIONClinicalTrials.gov NCT03204396.FUNDINGSwiss National Foundation, Gottfried Julia Bangerter-Rhyner Foundation, Goldschmidt-Jacobson Foundation, Hemmi Foundation, University of Basel, University Hospital Basel, Swiss Academy of Medical Science.
Collapse
Affiliation(s)
- Leila Probst
- Department of Endocrinology, Diabetology and Metabolism, University Hospital Basel, Basel, Switzerland
- Department of Clinical Research, University of Basel and University Hospital Basel, Basel, Switzerland
| | - Sophie Monnerat
- Department of Endocrinology, Diabetology and Metabolism, University Hospital Basel, Basel, Switzerland
- Department of Clinical Research, University of Basel and University Hospital Basel, Basel, Switzerland
| | - Deborah R. Vogt
- Department of Clinical Research, University of Basel and University Hospital Basel, Basel, Switzerland
| | - Sophia Lengsfeld
- Department of Endocrinology, Diabetology and Metabolism, University Hospital Basel, Basel, Switzerland
- Department of Clinical Research, University of Basel and University Hospital Basel, Basel, Switzerland
| | - Thilo Burkard
- Department of Cardiology, and
- Medical Outpatient Department, University Hospital Basel, Basel, Switzerland
| | - Andrea Meienberg
- Department of Cardiology, and
- Medical Outpatient Department, University Hospital Basel, Basel, Switzerland
| | - Cemile Bathelt
- Department of Endocrinology, Diabetology and Metabolism, University Hospital Basel, Basel, Switzerland
- Department of Clinical Research, University of Basel and University Hospital Basel, Basel, Switzerland
| | - Mirjam Christ-Crain
- Department of Endocrinology, Diabetology and Metabolism, University Hospital Basel, Basel, Switzerland
- Department of Clinical Research, University of Basel and University Hospital Basel, Basel, Switzerland
| | - Bettina Winzeler
- Department of Endocrinology, Diabetology and Metabolism, University Hospital Basel, Basel, Switzerland
- Department of Clinical Research, University of Basel and University Hospital Basel, Basel, Switzerland
| |
Collapse
|
13
|
Aranäs C, Blid Sköldheden S, Jerlhag E. Antismoking agents do not contribute synergistically to semaglutide's ability to reduce alcohol intake in rats. Front Pharmacol 2023; 14:1180512. [PMID: 37719854 PMCID: PMC10500129 DOI: 10.3389/fphar.2023.1180512] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2023] [Accepted: 08/17/2023] [Indexed: 09/19/2023] Open
Abstract
Preclinical studies have identified glucagon-like peptide-1 receptor (GLP-1R) agonists, and the antismoking agents varenicline and bupropion as tentative agents for treatment of alcohol use disorder (AUD). Combining different medications is a recent approach that has gained attention regarding heterogenous and difficult-to-treat diseases, like AUD. Successfully, this approach has been tested for the combination of varenicline and bupropion as it prevents relapse to alcohol drinking in rats. However, studies assessing the effects of the combination of semaglutide, an FDA-approved GLP-1R agonist for diabetes type II, and varenicline or bupropion to reduce alcohol intake in male and female rats remains to be conducted. Another approach to influence treatment outcome is to combine a medication with feeding interventions like high fat diet (HFD). While HFD reduces alcohol intake, the ability of the combination of HFD and semaglutide to alter alcohol drinking is unknown and thus the subject for a pilot study. Therefore, three intermittent alcohol drinking experiments were conducted to elucidate the effectiveness of these treatment combinations. We show that semaglutide, bupropion or HFD reduces alcohol intake in male as well as female rats. While various studies reveal beneficial effects of combinatorial pharmacotherapies for the treatment of AUD, we herein do not report any additive effects on alcohol intake by adding either varenicline or bupropion to semaglutide treatment. Neither does HFD exposure alter the ability of semaglutide to reduce alcohol intake. Although no additive effects by the combinatorial treatments are found, these findings collectively provide insight into possible monotherapeutical treatments for AUD.
Collapse
Affiliation(s)
| | | | - Elisabet Jerlhag
- Department of Pharmacology, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
| |
Collapse
|
14
|
Paulson OB, Schousboe A, Hultborn H. The history of Danish neuroscience. Eur J Neurosci 2023; 58:2893-2960. [PMID: 37477973 DOI: 10.1111/ejn.16062] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2022] [Revised: 05/04/2023] [Accepted: 05/29/2023] [Indexed: 07/22/2023]
Abstract
The history of Danish neuroscience starts with an account of impressive contributions made at the 17th century. Thomas Bartholin was the first Danish neuroscientist, and his disciple Nicolaus Steno became internationally one of the most prominent neuroscientists in this period. From the start, Danish neuroscience was linked to clinical disciplines. This continued in the 19th and first half of the 20th centuries with new initiatives linking basic neuroscience to clinical neurology and psychiatry in the same scientific environment. Subsequently, from the middle of the 20th century, basic neuroscience was developing rapidly within the preclinical university sector. Clinical neuroscience continued and was even reinforced during this period with important translational research and a close co-operation between basic and clinical neuroscience. To distinguish 'history' from 'present time' is not easy, as many historical events continue in present time. Therefore, we decided to consider 'History' as new major scientific developments in Denmark, which were launched before the end of the 20th century. With this aim, scientists mentioned will have been born, with a few exceptions, no later than the early 1960s. However, we often refer to more recent publications in documenting the developments of initiatives launched before the end of the last century. In addition, several scientists have moved to Denmark after the beginning of the present century, and they certainly are contributing to the present status of Danish neuroscience-but, again, this is not the History of Danish neuroscience.
Collapse
Affiliation(s)
- Olaf B Paulson
- Neurobiology Research Unit, Department of Neurology, Rigshospitalet, 9 Blegdamsvej, Copenhagen, Denmark
- Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
| | - Arne Schousboe
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Hans Hultborn
- Department of Neuroscience, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
15
|
Chuong V, Farokhnia M, Khom S, Pince CL, Elvig SK, Vlkolinsky R, Marchette RC, Koob GF, Roberto M, Vendruscolo LF, Leggio L. The glucagon-like peptide-1 (GLP-1) analogue semaglutide reduces alcohol drinking and modulates central GABA neurotransmission. JCI Insight 2023; 8:e170671. [PMID: 37192005 PMCID: PMC10371247 DOI: 10.1172/jci.insight.170671] [Citation(s) in RCA: 33] [Impact Index Per Article: 33.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2023] [Accepted: 05/10/2023] [Indexed: 05/17/2023] Open
Abstract
Growing evidence indicates that the glucagon-like peptide-1 (GLP-1) system is involved in the neurobiology of addictive behaviors, and GLP-1 analogues may be used for the treatment of alcohol use disorder (AUD). Here, we examined the effects of semaglutide, a long-acting GLP-1 analogue, on biobehavioral correlates of alcohol use in rodents. A drinking-in-the-dark procedure was used to test the effects of semaglutide on binge-like drinking in male and female mice. We also tested the effects of semaglutide on binge-like and dependence-induced alcohol drinking in male and female rats, as well as acute effects of semaglutide on spontaneous inhibitory postsynaptic currents (sIPSCs) from central amygdala (CeA) and infralimbic cortex (ILC) neurons. Semaglutide dose-dependently reduced binge-like alcohol drinking in mice; a similar effect was observed on the intake of other caloric/noncaloric solutions. Semaglutide also reduced binge-like and dependence-induced alcohol drinking in rats. Semaglutide increased sIPSC frequency in CeA and ILC neurons from alcohol-naive rats, suggesting enhanced GABA release, but had no overall effect on GABA transmission in alcohol-dependent rats. In conclusion, the GLP-1 analogue semaglutide decreased alcohol intake across different drinking models and species and modulated central GABA neurotransmission, providing support for clinical testing of semaglutide as a potentially novel pharmacotherapy for AUD.
Collapse
Affiliation(s)
- Vicky Chuong
- Clinical Psychoneuroendocrinology and Neuropsychopharmacology Section, Translational Addiction Medicine Branch, National Institute on Drug Abuse Intramural Research Program (NIDA IRP) and National Institute on Alcohol Abuse and Alcoholism Division of Intramural Clinical and Biological Research (NIAAA DICBR), NIH, Baltimore and Bethesda, Maryland, USA
- Neurobiology of Addiction Section, NIDA IRP, NIH, Baltimore, Maryland, USA
| | - Mehdi Farokhnia
- Clinical Psychoneuroendocrinology and Neuropsychopharmacology Section, Translational Addiction Medicine Branch, National Institute on Drug Abuse Intramural Research Program (NIDA IRP) and National Institute on Alcohol Abuse and Alcoholism Division of Intramural Clinical and Biological Research (NIAAA DICBR), NIH, Baltimore and Bethesda, Maryland, USA
| | - Sophia Khom
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, California, USA
- Department of Pharmaceutical Sciences, University of Vienna, Vienna, Austria
| | - Claire L. Pince
- Clinical Psychoneuroendocrinology and Neuropsychopharmacology Section, Translational Addiction Medicine Branch, National Institute on Drug Abuse Intramural Research Program (NIDA IRP) and National Institute on Alcohol Abuse and Alcoholism Division of Intramural Clinical and Biological Research (NIAAA DICBR), NIH, Baltimore and Bethesda, Maryland, USA
- Neurobiology of Addiction Section, NIDA IRP, NIH, Baltimore, Maryland, USA
| | - Sophie K. Elvig
- Neurobiology of Addiction Section, NIDA IRP, NIH, Baltimore, Maryland, USA
| | - Roman Vlkolinsky
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, California, USA
| | | | - George F. Koob
- Neurobiology of Addiction Section, NIDA IRP, NIH, Baltimore, Maryland, USA
| | - Marisa Roberto
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, California, USA
| | - Leandro F. Vendruscolo
- Stress and Addiction Neuroscience Unit, NIDA IRP and National Institute on Alcohol Abuse and Alcoholism Division of Intramural Clinical and Biological Research, National Institutes of Health, Baltimore, Maryland, USA
| | - Lorenzo Leggio
- Clinical Psychoneuroendocrinology and Neuropsychopharmacology Section, Translational Addiction Medicine Branch, National Institute on Drug Abuse Intramural Research Program (NIDA IRP) and National Institute on Alcohol Abuse and Alcoholism Division of Intramural Clinical and Biological Research (NIAAA DICBR), NIH, Baltimore and Bethesda, Maryland, USA
| |
Collapse
|
16
|
Aranäs C, Edvardsson CE, Shevchouk OT, Zhang Q, Witley S, Blid Sköldheden S, Zentveld L, Vallöf D, Tufvesson-Alm M, Jerlhag E. Semaglutide reduces alcohol intake and relapse-like drinking in male and female rats. EBioMedicine 2023; 93:104642. [PMID: 37295046 PMCID: PMC10363436 DOI: 10.1016/j.ebiom.2023.104642] [Citation(s) in RCA: 23] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2023] [Revised: 05/17/2023] [Accepted: 05/22/2023] [Indexed: 06/12/2023] Open
Abstract
BACKGROUND Glucagon-like peptide1 receptor (GLP-1R) agonists have been found to reduce alcohol drinking in rodents and overweight patients with alcohol use disorder (AUD). However, the probability of low semaglutide doses, an agonist with higher potency and affinity for GLP-1R, to attenuate alcohol-related responses in rodents and the underlying neuronal mechanisms is unknown. METHODS In the intermittent access model, we examined the ability of semaglutide to decrease alcohol intake and block relapse-like drinking, as well as imaging the binding of fluorescently marked semaglutide to nucleus accumbens (NAc) in both male and female rats. The suppressive effect of semaglutide on alcohol-induced locomotor stimulation and in vivo dopamine release in NAc was tested in male mice. We evaluated effect of semaglutide on the in vivo release of dopamine metabolites (DOPAC and HVA) and gene expression of enzymes metabolising dopamine (MAOA and COMT) in male mice. FINDINGS In male and female rats, acute and repeated semaglutide administration reduced alcohol intake and prevented relapse-like drinking. Moreover, fluorescently labelled semaglutide was detected in NAc of alcohol-drinking male and female rats. Further, semaglutide attenuated the ability of alcohol to cause hyperlocomotion and to elevate dopamine in NAc in male mice. As further shown in male mice, semaglutide enhanced DOPAC and HVA in NAc when alcohol was onboard and increased the gene expression of COMT and MAOA. INTERPRETATION Altogether, this indicates that semaglutide reduces alcohol drinking behaviours, possibly via a reduction in alcohol-induced reward and NAc dependent mechanisms. As semaglutide also decreased body weight of alcohol-drinking rats of both sexes, upcoming clinical studies should test the plausibility that semaglutide reduces alcohol intake and body weight in overweight AUD patients. FUNDING Swedish Research Council (2019-01676), LUA/ALF (723941) from the Sahlgrenska University Hospital and the Swedish brain foundation.
Collapse
Affiliation(s)
- Cajsa Aranäs
- Department of Pharmacology, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
| | - Christian E Edvardsson
- Department of Pharmacology, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
| | - Olesya T Shevchouk
- Department of Pharmacology, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
| | - Qian Zhang
- Department of Pharmacology, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
| | - Sarah Witley
- Department of Pharmacology, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
| | - Sebastian Blid Sköldheden
- Department of Pharmacology, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
| | - Lindsay Zentveld
- Department of Pharmacology, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
| | - Daniel Vallöf
- Department of Pharmacology, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
| | - Maximilian Tufvesson-Alm
- Department of Pharmacology, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
| | - Elisabet Jerlhag
- Department of Pharmacology, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden.
| |
Collapse
|
17
|
Cawthon CR, Blonde GD, Nisi AV, Bloomston HM, Krubitski B, le Roux CW, Spector AC. Chronic Semaglutide Treatment in Rats Leads to Daily Excessive Concentration-Dependent Sucrose Intake. J Endocr Soc 2023; 7:bvad074. [PMID: 37388574 PMCID: PMC10306276 DOI: 10.1210/jendso/bvad074] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/19/2023] [Indexed: 07/01/2023] Open
Abstract
Context The glucagon-like peptide-1 receptor (GLP-1R) agonist semaglutide (SEMA) produces 15% weight loss when chronically administered to humans with obesity. Methods In 2 separate experiments, rats received daily injections of either vehicle (VEH) or SEMA starting at 7 µg/kg body weight (BW) and increasing over 10 days to the maintenance dose (70 µg/kg-BW), emulating clinical dose escalation strategies. Results During dose escalation and maintenance, SEMA rats reduced chow intake and bodyweight. Experiment 2 meal pattern analysis revealed that meal size, not number, mediated these SEMA-induced changes in chow intake. This suggests SEMA affects neural processes controlling meal termination and not meal initiation. Two-bottle preference tests (vs water) began after 10 to 16 days of maintenance dosing. Rats received either an ascending sucrose concentration series (0.03-1.0 M) and 1 fat solution (Experiment 1) or a 4% and 24% sucrose solution in a crossover design (Experiment 2). At lower sucrose concentrations, SEMA-treated rats in both experiments drank sometimes >2× the volume consumed by VEH controls; at higher sucrose concentrations (and 10% fat), intake was similar between treatment groups. Energy intake of SEMA rats became similar to VEH rats. This was unexpected because GLP-1R agonism is thought to decrease the reward and/or increase the satiating potency of palatable foods. Despite sucrose-driven increases in both groups, a significant bodyweight difference between SEMA- and VEH-treated rats remained. Conclusion The basis of the SEMA-induced overconsumption of sucrose at lower concentrations relative to VEH controls remains unclear, but the effects of chronic SEMA treatment on energy intake and BW appear to depend on the caloric sources available.
Collapse
Affiliation(s)
- Carolina R Cawthon
- Department of Psychology and Program in Neuroscience, Florida State University, Tallahassee, FL 32306, USA
| | - Ginger D Blonde
- Department of Psychology and Program in Neuroscience, Florida State University, Tallahassee, FL 32306, USA
| | - A Valentina Nisi
- Department of Psychology and Program in Neuroscience, Florida State University, Tallahassee, FL 32306, USA
| | - Haley M Bloomston
- Department of Psychology and Program in Neuroscience, Florida State University, Tallahassee, FL 32306, USA
| | - Belle Krubitski
- Department of Psychology and Program in Neuroscience, Florida State University, Tallahassee, FL 32306, USA
| | - Carel W le Roux
- Diabetes Complications Research Center, Conway Institute, School of Medicine, University College Dublin, Dublin, D04 C1P1, Ireland
| | - Alan C Spector
- Department of Psychology and Program in Neuroscience, Florida State University, Tallahassee, FL 32306, USA
| |
Collapse
|
18
|
Allingbjerg ML, Hansen SN, Secher A, Thomsen M. Glucagon-like peptide-1 receptors in nucleus accumbens, ventral hippocampus, and lateral septum reduce alcohol reinforcement in mice. Exp Clin Psychopharmacol 2023; 31:612-620. [PMID: 36480394 PMCID: PMC10198891 DOI: 10.1037/pha0000620] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Glucagon-like peptide 1 (GLP-1) receptor agonists can decrease alcohol intake by central mechanisms that are still poorly understood. The lateral septum (LS) and the ventral/caudal part of the hippocampus are enriched in GLP-1 receptors, and activity in these regions was shown to modulate reward-related behaviors. Using microinfusions of the GLP-1 receptor agonist exendin-4 in mice trained to self-administer oral alcohol in an operant assay, we tested whether pharmacological stimulation of GLP-1 receptors in hippocampus and LS decrease alcohol self-administration. We report that infusion of exendin-4 in the ventral hippocampus or LS was sufficient to reduce alcohol self-administration with as large effect sizes as we previously reported with systemic exendin-4 administration. Infusion of exendin-4 into the nucleus accumbens also reduced alcohol self-administration, as anticipated based on earlier reports, while infusion of exendin-4 into the caudate-putamen (dorsal striatum) had little effect, consistent with lack of GLP-1 receptor expression in this region. The distribution of exendin-4 after infusion into the LS or caudate putamen was visualized using a fluorescently labeled ligand. These findings add to our understanding of the circuit-level mechanisms underlying the ability of GLP-1 receptor agonists to reduce alcohol self-administration. (PsycInfo Database Record (c) 2023 APA, all rights reserved).
Collapse
Affiliation(s)
- Marie-Louise Allingbjerg
- Laboratory of Neuropsychiatry, University Hospital of Copenhagen, Mental Health Services, Capital Region of Denmark
| | | | | | - Morgane Thomsen
- Laboratory of Neuropsychiatry, University Hospital of Copenhagen, Mental Health Services, Capital Region of Denmark
| |
Collapse
|
19
|
Jerlhag E. The therapeutic potential of glucagon-like peptide-1 for persons with addictions based on findings from preclinical and clinical studies. Front Pharmacol 2023; 14:1063033. [PMID: 37063267 PMCID: PMC10097922 DOI: 10.3389/fphar.2023.1063033] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2022] [Accepted: 03/17/2023] [Indexed: 04/03/2023] Open
Abstract
Although the multifaceted mechanisms underlying alcohol use disorder (AUD) have been partially defined, the neurobiological complexity of this disorder is yet to be unraveled. One of the systems that have gained attention in recent times is the gut–brain axis. Although numerous peptides participate in this axis, glucagon-like peptide-1 (GLP-1) plays a central role. GLP-1 is a crucial anorexigenic peptide, with potent abilities to reduce food intake and body weight. The physiological complexity of GLP-1 entails glucose homeostasis, gastrointestinal motility, and the release of insulin and glucagon. As reviewed in this study, acute or repeated treatment with GLP-1 receptor (GLP-1R) agonists decreases alcohol consumption in rodents. Moreover, the abilities of alcohol to promote hyperlocomotion, dopamine release in the nucleus accumbens, and reward in the conditioned place preference paradigm are all suppressed by GLP-1R ligands. Moreover, activation of GLP-1R suppresses the motivation to consume alcohol, alcohol-seeking behaviors, and relapse drinking in male rodents. Similarly, abstinence symptoms experienced during alcohol withdrawal are attenuated by activation of the GLP-1 pathway. On a similar note, the activation of GLP-1 receptors within areas of the brain that are processing reward modulates these alcohol-related responses. Another area that is crucial for this ability is the nucleus of the solitary tract, which is where GLP-1 is produced and from which GLP-1-containing neurons project to areas of reward. These findings may have clinical relevance as AUD is associated with polymorphisms in GLP-1-related genes. Although a GLP-1R agonist does not alter alcohol intake in AUD patients, it reduces this consumption in a sub-population of obese AUD individuals. Given the uncertainty of this outcome, additional clinical studies of obese AUD patients should explore the effects of the GLP-1R agonists on alcohol intake and body weight. Furthermore, GLP-1 receptors modulate the behavioral and neurochemical responses to addictive drugs. Taken together, these preclinical and clinical findings imply that the GLP-1 pathway plays a role in the complex mechanisms regulating alcohol and drug consumption patterns, unveiling a novel aspect of addiction medicine.
Collapse
|
20
|
Tufvesson-Alm M, Shevchouk OT, Jerlhag E. Insight into the role of the gut-brain axis in alcohol-related responses: Emphasis on GLP-1, amylin, and ghrelin. Front Psychiatry 2022; 13:1092828. [PMID: 36699502 PMCID: PMC9868418 DOI: 10.3389/fpsyt.2022.1092828] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/09/2022] [Accepted: 12/15/2022] [Indexed: 01/11/2023] Open
Abstract
Alcohol use disorder (AUD) contributes substantially to global morbidity and mortality. Given the heterogenicity of this brain disease, available pharmacological treatments only display efficacy in sub-set of individuals. The need for additional treatment options is thus substantial and is the goal of preclinical studies unraveling neurobiological mechanisms underlying AUD. Although these neurobiological processes are complex and numerous, one system gaining recent attention is the gut-brain axis. Peptides of the gut-brain axis include anorexigenic peptide like glucagon-like peptide-1 (GLP-1) and amylin as well as the orexigenic peptide ghrelin. In animal models, agonists of the GLP-1 or amylin receptor and ghrelin receptor (GHSR) antagonists reduce alcohol drinking, relapse drinking, and alcohol-seeking. Moreover, these three gut-brain peptides modulate alcohol-related responses (behavioral and neurochemical) in rodents, suggesting that the alcohol reduction may involve a suppression of alcohol's rewarding properties. Brain areas participating in the ability of these gut-brain peptides to reduce alcohol-mediated behaviors/neurochemistry involve those important for reward. Human studies support these preclinical studies as polymorphisms of the genes encoding for GLP-1 receptor or the ghrelin pathway are associated with AUD. Moreover, a GLP-1 receptor agonist decreases alcohol drinking in overweight patients with AUD and an inverse GHSR agonist reduces alcohol craving. Although preclinical and clinical studies reveal an interaction between the gut-brain axis and AUD, additional studies should explore this in more detail.
Collapse
Affiliation(s)
- Maximilian Tufvesson-Alm
- Department of Pharmacology, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Olesya T Shevchouk
- Department of Pharmacology, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Elisabet Jerlhag
- Department of Pharmacology, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| |
Collapse
|
21
|
Shevchouk OT, Tufvesson-Alm M, Jerlhag E. An Overview of Appetite-Regulatory Peptides in Addiction Processes; From Bench to Bed Side. Front Neurosci 2021; 15:774050. [PMID: 34955726 PMCID: PMC8695496 DOI: 10.3389/fnins.2021.774050] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2021] [Accepted: 11/08/2021] [Indexed: 12/17/2022] Open
Abstract
There is a substantial need for new pharmacological treatments of addiction, and appetite-regulatory peptides are implied as possible candidates. Appetite regulation is complex and involves anorexigenic hormones such as glucagon-like peptide-1 (GLP-1) and amylin, and orexigenic peptides like ghrelin and all are well-known for their effects on feeding behaviors. This overview will summarize more recent physiological aspects of these peptides, demonstrating that they modulate various aspects of addiction processes. Findings from preclinical, genetic, and experimental clinical studies exploring the association between appetite-regulatory peptides and the acute or chronic effects of addictive drugs will be introduced. Short or long-acting GLP-1 receptor agonists independently attenuate the acute rewarding properties of addictive drugs or reduce the chronic aspects of drugs. Genetic variation of the GLP-1 system is associated with alcohol use disorder. Also, the amylin pathway modulates the acute and chronic behavioral responses to addictive drugs. Ghrelin has been shown to activate reward-related behaviors. Moreover, ghrelin enhances, whereas pharmacological or genetic suppression of the ghrelin receptor attenuates the responses to various addictive drugs. Genetic studies and experimental clinical studies further support the associations between ghrelin and addiction processes. Further studies should explore the mechanisms modulating the ability of appetite-regulatory peptides to reduce addiction, and the effects of combination therapies or different diets on substance use are warranted. In summary, these studies provide evidence that appetite-regulatory peptides modulate reward and addiction processes, and deserve to be investigated as potential treatment target for addiction.
Collapse
Affiliation(s)
- Olesya T Shevchouk
- Department of Pharmacology, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
| | - Maximilian Tufvesson-Alm
- Department of Pharmacology, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
| | - Elisabet Jerlhag
- Department of Pharmacology, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
| |
Collapse
|
22
|
Klausen MK, Thomsen M, Wortwein G, Fink-Jensen A. The role of glucagon-like peptide 1 (GLP-1) in addictive disorders. Br J Pharmacol 2021; 179:625-641. [PMID: 34532853 DOI: 10.1111/bph.15677] [Citation(s) in RCA: 55] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2021] [Revised: 05/21/2021] [Accepted: 06/19/2021] [Indexed: 11/29/2022] Open
Abstract
Drug-, alcohol- and tobacco use disorders are a global burden affecting millions of people. Despite decades of research, treatment options are sparse or missing, and relapse rates are high. Glucagon-like peptide-1 (GLP-1) is released in the small intestines, promotes blood glucose homeostasis, slows gastric emptying, and reduces appetite. GLP-1 receptor agonists approved for treating type 2 diabetes mellitus and obesity, have received attention as a potential anti-addiction treatment. Studies in rodents and non-human primates have demonstrated a reduction in intake of alcohol and drugs of abuse, and clinical trials have been initiated to investigate whether the preclinical findings can be translated to patients. This review will give an overview of current findings and discuss the possible mechanisms of action. We suggest that effects of GLP-1 in alcohol- and substance use disorder is mediated centrally, at least partly through dopamine signalling, but precise mechanisms are still to be uncovered.
Collapse
Affiliation(s)
- Mette Kruse Klausen
- Psychiatric Centre Copenhagen and University Hospital of Copenhagen, Copenhagen, Denmark
| | - Morgane Thomsen
- Laboratory of Neuropsychiatry, Psychiatric Centre Copenhagen and University Hospital of Copenhagen, Copenhagen, Denmark
| | - Gitta Wortwein
- Laboratory of Neuropsychiatry, Psychiatric Centre Copenhagen and University Hospital of Copenhagen, Copenhagen, Denmark
| | - Anders Fink-Jensen
- Laboratory of Neuropsychiatry, Psychiatric Centre Copenhagen and University Hospital of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
23
|
Bramming M, Becker U, Jørgensen MB, Neermark S, Bisgaard T, Tolstrup JS. Bariatric surgery and risk of alcohol use disorder: a register-based cohort study. Int J Epidemiol 2021; 49:1826-1835. [PMID: 33085738 DOI: 10.1093/ije/dyaa147] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/17/2020] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND Bariatric surgery has been associated with altered alcohol metabolism. We examined whether patients undergoing bariatric surgery have a higher risk of developing alcohol use disorder (AUD) compared with individuals with obesity who have not received bariatric surgery. METHODS In this prospective cohort study, we followed 13 430 patients undergoing bariatric surgery (95% gastric bypass) between 2005 and 2013 and a reference group of 21 021 individuals with obesity for a median of 6.9 years (5th-95th percentile: 4.0-9.8). Four different approaches were used to account for baseline differences between the two groups: (i) adjustment; (ii) inverse probability of treatment weighting (IPTW); (iii) 1:1 matching based on propensity scores; and (iv) before-and-after analysis comparing the bariatric surgery group with itself 5 years before and after surgery. Cox proportional hazard modelling was used to estimate hazard ratios of AUD defined from national registers. RESULTS When applying the IPTW approach, the hazard ratio (HR) of AUD for bariatric surgery patients was 7.29 [95% confidence interval (CI): 5.06-9.48] compared with individuals without surgery. When employing different approaches (adjustment for baseline variables, matching on propensity scores, before-and-after analyses), results were of similar magnitude. Analysis stratified by time after surgery revealed a higher risk of AUD already within the first year following surgery [HR: 2.77 (95% CI: 1.39-5.53)]. CONCLUSIONS Patients undergoing bariatric surgery have a higher risk of developing AUD compared with individuals without bariatric surgery. The higher risk observed in this group of patients cannot be explained by differences in baseline characteristics such as socioeconomic factors. Despite the higher risk of AUD, only few individuals developed AUD. Individuals with disabling obesity should therefore not rule out surgery based on these results but rather be aware of negative implications.
Collapse
Affiliation(s)
- Maja Bramming
- National Institute of Public Health, University of Southern Denmark, Copenhagen, Denmark
| | - Ulrik Becker
- National Institute of Public Health, University of Southern Denmark, Copenhagen, Denmark.,Gastrounit Medical Division, Copenhagen University Hospital Hvidovre, Hvidovre, Denmark
| | - Maja B Jørgensen
- National Institute of Public Health, University of Southern Denmark, Copenhagen, Denmark
| | - Søren Neermark
- Gastrounit Medical Division, Copenhagen University Hospital Hvidovre, Hvidovre, Denmark.,Center of Planning, Danish Board of Health, Copenhagen, Denmark
| | - Thue Bisgaard
- Surgical Department, Zealand University Hospital, Køge, Denmark
| | - Janne S Tolstrup
- National Institute of Public Health, University of Southern Denmark, Copenhagen, Denmark
| |
Collapse
|
24
|
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: 18] [Impact Index Per Article: 6.0] [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
|
25
|
Kalafateli AL, Vestlund J, Raun K, Egecioglu E, Jerlhag E. Effects of a selective long-acting amylin receptor agonist on alcohol consumption, food intake and body weight in male and female rats. Addict Biol 2021; 26:e12910. [PMID: 32383257 DOI: 10.1111/adb.12910] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2019] [Revised: 04/02/2020] [Accepted: 04/08/2020] [Indexed: 12/15/2022]
Abstract
Alcohol use disorder is a complex neuropsychiatric disorder affecting both males and females worldwide; however, the efficacy of current pharmacotherapies varies. Recent advances show that gut-brain peptides, like amylin, regulate alcohol behavioural responses by acting on brain areas involved in alcohol reward processes. Thus, the activation of amylin receptors (AMYRs) by salmon calcitonin (sCT) decreases alcohol behaviours in male rodents. Given that sCT also activates the sole calcitonin receptor (CTR), studies of more selective AMYR agonists in both male and female rodents are needed to explore amylinergic modulation of alcohol behaviours. Therefore, we investigated the effects of repeated administration of a selective long-acting AMYR agonist, NNC0174-1213 (AM1213), on alcohol, water and food intake, as well as body weight in male and female rats chronically exposed to alcohol. We confirm our previous studies with sCT in male rats, as repeated AM1213 administration for 2 weeks initially decreased alcohol intake in both male and female rats. However, this reduction ceases in both sexes on later sessions, accompanied by an increase in males. AM1213 reduced food intake and body weight in both male and female rats, with sustained body weight loss in males after discontinuation of the treatment. Moreover, AM1213 administration for 3 or 7 days, differentially altered dopamine, serotonin and their metabolites in the reward-related areas in males and females, providing tentative, but different, downstream mechanism through which selective activation of AMYR may alter alcohol intake. Our data provide clarified insight into the importance of AMYRs for alcohol intake regulation in both sexes.
Collapse
Affiliation(s)
- Aimilia Lydia Kalafateli
- Department of Pharmacology, Institute of Neuroscience and Physiology The Sahlgrenska Academy at the University of Gothenburg Gothenburg Sweden
| | - Jesper Vestlund
- Department of Pharmacology, Institute of Neuroscience and Physiology The Sahlgrenska Academy at the University of Gothenburg Gothenburg Sweden
| | | | | | - Elisabet Jerlhag
- Department of Pharmacology, Institute of Neuroscience and Physiology The Sahlgrenska Academy at the University of Gothenburg Gothenburg Sweden
| |
Collapse
|
26
|
Klockars A, Levine AS, Head MA, Perez-Leighton CE, Kotz CM, Olszewski PK. Impact of Gut and Metabolic Hormones on Feeding Reward. Compr Physiol 2021; 11:1425-1447. [PMID: 33577129 DOI: 10.1002/cphy.c190042] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Ingestion of food activates a cascade of endocrine responses (thereby reflecting a contemporaneous feeding status) that include the release of hormones from the gastrointestinal (GI) tract, such as cholecystokinin (CCK), glucagonlike peptide YY (PYY), peptide PP, and oleoylethanolamide, as well as suppression of ghrelin secretion. The pancreas and adipose tissue, on the other hand, release hormones that serve as a measure of the current metabolic state or the long-term energy stores, that is, insulin, leptin, and adiponectin. It is well known and intuitively understandable that these hormones target either directly (by crossing the blood-brain barrier) or indirectly (e.g., via vagal input) the "homeostatic" brainstem-hypothalamic pathways involved in the regulation of appetite. The current article focuses on yet another target of the metabolic and GI hormones that is critical in inducing changes in food intake, namely, the reward system. We discuss the physiological basis of this functional interaction, its importance in the control of appetite, and the impact that disruption of this crosstalk has on energy intake in select physiological and pathophysiological states. We conclude that metabolic and GI hormones have a capacity to strengthen or weaken a response of the reward system to a given food, and thus, they are fundamental in ensuring that feeding reward is plastic and dependent on the energy status of the organism. © 2021 American Physiological Society. Compr Physiol 11:1425-1447, 2021.
Collapse
Affiliation(s)
- Anica Klockars
- Faculty of Science and Engineering, University of Waikato, Hamilton, New Zealand
| | - Allen S Levine
- Department of Food Science and Nutrition, University of Minnesota, St. Paul, Minnesota, USA
| | - Mitchell A Head
- Faculty of Science and Engineering, University of Waikato, Hamilton, New Zealand
| | | | - Catherine M Kotz
- Department of Food Science and Nutrition, University of Minnesota, St. Paul, Minnesota, USA.,Department of Integrative Biology and Physiology, Medical School, University of Minnesota, Minneapolis, Minnesota, USA
| | - Pawel K Olszewski
- Faculty of Science and Engineering, University of Waikato, Hamilton, New Zealand.,Department of Food Science and Nutrition, University of Minnesota, St. Paul, Minnesota, USA.,Department of Integrative Biology and Physiology, Medical School, University of Minnesota, Minneapolis, Minnesota, USA
| |
Collapse
|
27
|
Eren-Yazicioglu CY, Yigit A, Dogruoz RE, Yapici-Eser H. Can GLP-1 Be a Target for Reward System Related Disorders? A Qualitative Synthesis and Systematic Review Analysis of Studies on Palatable Food, Drugs of Abuse, and Alcohol. Front Behav Neurosci 2021; 14:614884. [PMID: 33536884 PMCID: PMC7848227 DOI: 10.3389/fnbeh.2020.614884] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2020] [Accepted: 12/15/2020] [Indexed: 11/15/2022] Open
Abstract
The role of glucagon-like peptide 1 (GLP-1) in insulin-dependent signaling is well-known; GLP-1 enhances glucose-dependent insulin secretion and lowers blood glucose in diabetes. GLP-1 receptors (GLP-1R) are also widely expressed in the brain, and in addition to its role in neuroprotection, it affects reward pathways. This systematic review aimed to analyze the studies on GLP-1 and reward pathways and its currently identified mechanisms. Methods: “Web of Science” and “Pubmed” were searched to identify relevant studies using GLP-1 as the keyword. Among the identified 26,539 studies, 30 clinical, and 71 preclinical studies were included. Data is presented by grouping rodent studies on palatable food intake, drugs of abuse, and studies on humans focusing on GLP-1 and reward systems. Results: GLP-1Rs are located in reward-related areas, and GLP-1, its agonists, and DPP-IV inhibitors are effective in decreasing palatable food intake, along with reducing cocaine, amphetamine, alcohol, and nicotine use in animals. GLP-1 modulates dopamine levels and glutamatergic neurotransmission, which results in observed behavioral changes. In humans, GLP-1 alters palatable food intake and improves activity deficits in the insula, hypothalamus, and orbitofrontal cortex (OFC). GLP-1 reduces food cravings partially by decreasing activity to the anticipation of food in the left insula of obese patients with diabetes and may inhibit overeating by increasing activity to the consumption of food in the right OFC of obese and left insula of obese with diabetes. Conclusion: Current preclinical studies support the view that GLP-1 can be a target for reward system related disorders. More translational research is needed to evaluate its efficacy on human reward system related disorders.
Collapse
Affiliation(s)
| | - Arya Yigit
- School of Medicine, Koç University, Istanbul, Turkey
| | - Ramazan Efe Dogruoz
- Department of Neuroscience, University of Chicago, Chicago, IL, United States
| | - Hale Yapici-Eser
- Koç University, Research Center for Translational Medicine (KUTTAM), Istanbul, Turkey.,Department of Psychiatry, School of Medicine, Koç University, Istanbul, Turkey
| |
Collapse
|
28
|
Mahajan VR, Elvig SK, Vendruscolo LF, Koob GF, Darcey VL, King MT, Kranzler HR, Volkow ND, Wiers CE. Nutritional Ketosis as a Potential Treatment for Alcohol Use Disorder. Front Psychiatry 2021; 12:781668. [PMID: 34916977 PMCID: PMC8670944 DOI: 10.3389/fpsyt.2021.781668] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/23/2021] [Accepted: 11/08/2021] [Indexed: 12/28/2022] Open
Abstract
Alcohol use disorder (AUD) is a chronic, relapsing brain disorder, characterized by compulsive alcohol seeking and disrupted brain function. In individuals with AUD, abstinence from alcohol often precipitates withdrawal symptoms than can be life threatening. Here, we review evidence for nutritional ketosis as a potential means to reduce withdrawal and alcohol craving. We also review the underlying mechanisms of action of ketosis. Several findings suggest that during alcohol intoxication there is a shift from glucose to acetate metabolism that is enhanced in individuals with AUD. During withdrawal, there is a decline in acetate levels that can result in an energy deficit and could contribute to neurotoxicity. A ketogenic diet or ingestion of a ketone ester elevates ketone bodies (acetoacetate, β-hydroxybutyrate and acetone) in plasma and brain, resulting in nutritional ketosis. These effects have been shown to reduce alcohol withdrawal symptoms, alcohol craving, and alcohol consumption in both preclinical and clinical studies. Thus, nutritional ketosis may represent a unique treatment option for AUD: namely, a nutritional intervention that could be used alone or to augment the effects of medications.
Collapse
Affiliation(s)
- Vikrant R Mahajan
- Department of Pharmacology, Vanderbilt University, Nashville, TN, United States
| | - Sophie K Elvig
- Integrative Neuroscience Research Branch, National Institute on Drug Abuse, Baltimore, MD, United States
| | - Leandro F Vendruscolo
- Integrative Neuroscience Research Branch, National Institute on Drug Abuse, Baltimore, MD, United States
| | - George F Koob
- Integrative Neuroscience Research Branch, National Institute on Drug Abuse, Baltimore, MD, United States
| | - Valerie L Darcey
- National Institute of Diabetes and Digestive and Kidney Diseases, Bethesda, MD, United States
| | - M Todd King
- National Institute on Alcohol Abuse and Alcoholism, Rockville, MD, United States
| | - Henry R Kranzler
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Nora D Volkow
- National Institute on Alcohol Abuse and Alcoholism, Rockville, MD, United States
| | - Corinde E Wiers
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| |
Collapse
|
29
|
Clasen MM, Riley AL, Davidson TL. Hippocampal-Dependent Inhibitory Learning and Memory Processes in the Control of Eating and Drug Taking. Curr Pharm Des 2020; 26:2334-2352. [PMID: 32026771 DOI: 10.2174/1381612826666200206091447] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2019] [Accepted: 12/11/2019] [Indexed: 12/11/2022]
Abstract
As manifestations of excessive and uncontrolled intake, obesity and drug addiction have generated much research aimed at identifying common neuroadaptations that could underlie both disorders. Much work has focused on changes in brain reward and motivational circuitry that can overexcite eating and drug-taking behaviors. We suggest that the regulation of both behaviors depends on balancing excitation produced by stimuli associated with food and drug rewards with the behavioral inhibition produced by physiological "satiety" and other stimuli that signal when those rewards are unavailable. Our main hypothesis is that dysregulated eating and drug use are consequences of diet- and drug-induced degradations in this inhibitory power. We first outline a learning and memory mechanism that could underlie the inhibition of both food and drug-intake, and we describe data that identifies the hippocampus as a brain substrate for this mechanism. We then present evidence that obesitypromoting western diets (WD) impair the operation of this process and generate pathophysiologies that disrupt hippocampal functioning. Next, we present parallel evidence that drugs of abuse also impair this same learning and memory process and generate similar hippocampal pathophysiologies. We also describe recent findings that prior WD intake elevates drug self-administration, and the implications of using drugs (i.e., glucagon-like peptide- 1 agonists) that enhance hippocampal functioning to treat both obesity and addiction are also considered. We conclude with a description of how both WD and drugs of abuse could initiate a "vicious-cycle" of hippocampal pathophysiology and impaired hippocampal-dependent behavioral inhibition.
Collapse
Affiliation(s)
- Matthew M Clasen
- Department of Psychology, Program in Neuroscience, Williams College, Williamstown, MA 01267, United States
| | - Anthony L Riley
- Department of Neuroscience, Center for Behavioral Neuroscience, American University, Washington, DC 20016, United States
| | - Terry L Davidson
- Department of Neuroscience, Center for Behavioral Neuroscience, American University, Washington, DC 20016, United States
| |
Collapse
|
30
|
Colvin KJ, Killen HS, Kanter MJ, Halperin MC, Engel L, Currie PJ. Brain Site-Specific Inhibitory Effects of the GLP-1 Analogue Exendin-4 on Alcohol Intake and Operant Responding for Palatable Food. Int J Mol Sci 2020; 21:ijms21249710. [PMID: 33352692 PMCID: PMC7766977 DOI: 10.3390/ijms21249710] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2020] [Revised: 12/14/2020] [Accepted: 12/15/2020] [Indexed: 12/11/2022] Open
Abstract
Approximately 14.4 million Americans are experiencing alcohol use disorder (AUD) and about two-thirds of people who experience drug addiction will relapse, highlighting the need to develop novel and effective treatments. Glucagon-like peptide-1 (GLP-1) is a peptide hormone implicated in the mesocorticolimbic reward system and has become a peptide of interest with respect to its putative inhibitory effects on drug reward. In order to further develop treatments for those diagnosed with AUD, the interplay between GLP-1 receptor signaling and ethanol consumption must be elucidated. In the present study, we investigated the ability of the GLP-1 analogue, exendin-4 (Ex-4), to alter alcohol intake and operant responding for sucrose pellets in order to further understand the role of this compound in mediating reward. We selected multiple sites throughout the prosencephalic and mesencephalic regions of the brain, where we directly administered various doses of Ex-4 to male Sprague Dawley rats. In alcohol investigations, we utilized a two-bottle choice intermittent access protocol. In separate groups of rats, we adopted an operant paradigm in order to examine the effect of Ex-4 on motivated responding for palatable food. Results indicated that GLP-1 receptor signaling effectively suppressed voluntary alcohol intake when injected into the ventral tegmental area (VTA), the accumbens core (NAcC) and shell (NAcS), the dorsomedial hippocampus (DMHipp), and the lateral hypothalamus (LH), which are all structures linked to brain reward mechanisms. The arcuate nucleus (ARcN) and the paraventricular nucleus (PVN) of the hypothalamus were unresponsive, as was the basolateral amygdala (BLA). However, Ex-4 treatment into the ArcN and PVN suppressed operant responding for sucrose pellets. In fact, the VTA, NAcC, NAcS, LH, and the DMHipp all showed comparable suppression of sucrose responding. Overall, our findings suggest that these central structures are implicated in brain reward circuitry, including alcohol and appetitive motivation, which may be mediated by GLP-1 receptor mechanisms. GLP-1, therefore, may play a critical role in modifying addictive behaviors via activation of multiple GLP-1 systems throughout the brain.
Collapse
|
31
|
Nishiya Y, Daimon M, Mizushiri S, Murakami H, Tanabe J, Matsuhashi Y, Yanagimachi M, Tokuda I, Sawada K, Ihara K. Nutrient consumption-dependent association of a glucagon-like peptide-1 receptor gene polymorphism with insulin secretion. Sci Rep 2020; 10:16382. [PMID: 33009421 PMCID: PMC7532183 DOI: 10.1038/s41598-020-71853-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2020] [Accepted: 08/17/2020] [Indexed: 01/07/2023] Open
Abstract
Since type 2 diabetes (DM) is a life-style related disease, life-style should be considered when association between genetic factors and DM are examined. However, most studies did not examine genetic associations in consideration with lifestyle. Glucagon-like peptide-1 (GLP-1) receptor (GLP1R) mediates the insulinotropic action of GLP-1 in β-cells. We here examined the association while taking into consideration of interactions between the gene polymorphism and various nutrient factors. Participants from the population-based Iwaki study of Japanese subjects held in 2014–2017 with information on nutritional intake evaluated by self-administered dietary history questionnaire, and GLP1R genotype (rs3765467: A/G), were included (n = 1,560). Although not significant, insulin secretion indices assessed by homeostasis model assessment of β-cell function (HOMA-β) in subjects with the GG genotype tended to be lower than in those with the AA+AG genotypes in most groups stratified into tertiles based on daily nutrient consumptions (high, middle, and low). Stratification also showed that the GG genotype was a significant risk for decreased insulin secretion (HOMA-β ≤ 30) even after adjustment for multiple factors (age, body mass index, alcohol consumption), but only in the highest tertiles of energy, protein and carbohydrate consumption in men [odds ratios (95% confidence interval) 3.95 (1.03–15.1), 15.83 (1.58–158.9), and 4.23 (1.10–11.2), respectively]. A polymorphism of the GLP1R gene was associated with decreased insulin secretion in a nutrient consumption-dependent manner in Japanese men, indicating an interaction between GLP1R and nutritional factors in the pathophysiology of DM.
Collapse
Affiliation(s)
- Yuki Nishiya
- Department of Endocrinology and Metabolism, Hirosaki University Graduate School of Medicine, Hirosaki, Aomori, Japan
| | - Makoto Daimon
- Department of Endocrinology and Metabolism, Hirosaki University Graduate School of Medicine, Hirosaki, Aomori, Japan.
| | - Satoru Mizushiri
- Department of Endocrinology and Metabolism, Hirosaki University Graduate School of Medicine, Hirosaki, Aomori, Japan
| | - Hiroshi Murakami
- Department of Endocrinology and Metabolism, Hirosaki University Graduate School of Medicine, Hirosaki, Aomori, Japan
| | - Jutaro Tanabe
- Department of Endocrinology and Metabolism, Hirosaki University Graduate School of Medicine, Hirosaki, Aomori, Japan
| | - Yuki Matsuhashi
- Department of Endocrinology and Metabolism, Hirosaki University Graduate School of Medicine, Hirosaki, Aomori, Japan
| | - Miyuki Yanagimachi
- Department of Endocrinology and Metabolism, Hirosaki University Graduate School of Medicine, Hirosaki, Aomori, Japan
| | - Itoyo Tokuda
- Department of Oral Healthcare Science, Hirosaki University Graduate School of Medicine, Hirosaki, Aomori, Japan
| | - Kaori Sawada
- Department of Social Medicine, Hirosaki University Graduate School of Medicine, Hirosaki, Aomori, Japan
| | - Kazushige Ihara
- Department of Social Medicine, Hirosaki University Graduate School of Medicine, Hirosaki, Aomori, Japan
| |
Collapse
|
32
|
Chung W, Promrat K, Wands J. Clinical implications, diagnosis, and management of diabetes in patients with chronic liver diseases. World J Hepatol 2020; 12:533-557. [PMID: 33033564 PMCID: PMC7522556 DOI: 10.4254/wjh.v12.i9.533] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/08/2020] [Revised: 08/03/2020] [Accepted: 08/15/2020] [Indexed: 02/06/2023] Open
Abstract
Diabetes mellitus (DM) negatively affects the development and progression of chronic liver diseases (CLD) of various etiologies. Concurrent DM and CLD are also associated with worse clinical outcomes with respect to mortality, the occurrence of hepatic decompensation, and the development of hepatocellular carcinoma (HCC). Unfortunately, early diagnosis and optimal treatment of DM can be challenging, due to the lack of established clinical guidelines as well as the medical complexity of this patient population. We conducted an exploratory review of relevant literature to provide an up-to-date review for internists and hepatologists caring for this patient population. We reviewed the epidemiological and pathophysiological associations between DM and CLD, the impact of insulin resistance on the progression and manifestations of CLD, the pathogenesis of hepatogenic diabetes, as well as the practical challenges in diagnosis and monitoring of DM in this patient population. We also reviewed the latest clinical evidence on various pharmacological antihyperglycemic therapies with an emphasis on liver disease-related clinical outcomes. Finally, we proposed an algorithm for managing DM in patients with CLD and discussed the clinical and research questions that remain to be addressed.
Collapse
Affiliation(s)
- Waihong Chung
- Division of Gastroenterology, Department of Medicine, Rhode Island Hospital, Providence, RI 02905, United States.
| | - Kittichai Promrat
- Division of Gastroenterology and Hepatology, Providence VA Medical Center, Providence, RI 02908, United States
| | - Jack Wands
- Liver Research Center, The Warren Alpert Medical School of Brown University, Providence, RI 02903, United States
| |
Collapse
|
33
|
Dixon TN, McNally GP, Ong ZY. Glucagon-Like Peptide-1 Receptor Signaling in the Ventral Tegmental Area Reduces Alcohol Self-Administration in Male Rats. Alcohol Clin Exp Res 2020; 44:2118-2129. [PMID: 33043520 DOI: 10.1111/acer.14437] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2020] [Accepted: 08/05/2020] [Indexed: 12/11/2022]
Abstract
BACKGROUND The misuse and abuse of alcohol is a major public health issue. However, available treatments are limited with variable efficacy. Recently, preclinical studies show that glucagon-like-peptide-1 (GLP-1) and its analogue Exendin-4 (Ex4) potently reduce a range of alcohol intake behaviors, thus highlighting its potential as a treatment for alcohol use disorders. However, the neural mechanisms and sites of action mediating the effects of Ex4 on alcohol intake behaviors remain to be characterized. This study examined the ventral tegmental area (VTA) as a site of action for the effects of GLP-1 on alcohol intake. METHODS Male Long-Evans rats were given intermittent access to 20% alcohol and trained to nose poke for 20% alcohol. Rats received intra-VTA injections of Ex4 (vehicle, 0.01, 0.05 μg), and the effects of VTA Ex4 on alcohol self-administration, motivation, and relapse were assessed. RESULTS When compared to vehicle treatment, intra-VTA Ex4 (0.01, 0.05 μg) delivery significantly reduced alcohol self-administration, an effect that was particularly prominent in high alcohol drinkers. However, VTA Ex4 did not reduce reacquisition of alcohol self-administration after extinction nor the motivation to obtain alcohol. Importantly, the lower dose of Ex4 (0.01 μg) used had no effect on food intake or locomotor activity, suggesting that the reduction in alcohol self-administration observed was not secondary to caloric intake or motor deficits. CONCLUSIONS Together, these findings provide support for the VTA as a key site of action for GLP-1 on alcohol self-administration but not the reacquisition of alcohol self-administration or motivation to work for alcohol.
Collapse
Affiliation(s)
- Tiarani N Dixon
- From the, School of Psychology, UNSW Sydney, Sydney, NSW, Australia
| | - Gavan P McNally
- From the, School of Psychology, UNSW Sydney, Sydney, NSW, Australia
| | - Zhi Yi Ong
- From the, School of Psychology, UNSW Sydney, Sydney, NSW, Australia
| |
Collapse
|
34
|
Vallöf D, Kalafateli AL, Jerlhag E. Long-term treatment with a glucagon-like peptide-1 receptor agonist reduces ethanol intake in male and female rats. Transl Psychiatry 2020; 10:238. [PMID: 32678077 PMCID: PMC7367312 DOI: 10.1038/s41398-020-00923-1] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/16/2020] [Revised: 06/23/2020] [Accepted: 07/03/2020] [Indexed: 01/29/2023] Open
Abstract
Given the limited efficacy of available pharmacotherapies for treatment of alcohol use disorder (AUD), the need for new medications is substantial. Preclinical studies have shown that acute administration of glucagon-like peptide-1 receptor (GLP-1R) agonists inhibits various ethanol-related behaviours, indicating this system as a potential target for AUD. However, the effects of long-term systemic treatment of GLP-1R agonists on ethanol intake in male and female rodents are to date unknown. Therefore, we investigated the effects of 9 or 5 weeks of once weekly administration of dulaglutide, a long-acting GLP-1R agonist, on ethanol intake in male and female rats. The ethanol intake during treatment discontinuation was also monitored. In an initial attempt to identify preliminary underlying mechanisms, the effects of 9 weeks of once weekly dulaglutide treatment on monoaminergic signalling in reward-related areas were explored in both sexes. We found that 9 or 5 weeks of once weekly dulaglutide treatment reduced ethanol intake and preference in male and female rats. Following discontinuation of dulaglutide treatment, the decrease in ethanol consumption was prolonged in males, but not females. We demonstrated that 9 weeks of dulaglutide treatment differentially influenced monoaminergic signalling in reward-related areas of male and female rats. Collectively, these data imply that the GLP-1R attracts interest as a potential molecular target in the medical treatment of AUD in humans: more specifically, dulaglutide should be evaluated as a potential medication for treatment thereof.
Collapse
Affiliation(s)
- Daniel Vallöf
- Department of Pharmacology, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
| | - Aimilia Lydia Kalafateli
- Department of Pharmacology, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
| | - Elisabet Jerlhag
- Department of Pharmacology, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden.
| |
Collapse
|
35
|
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: 4] [Impact Index Per Article: 1.0] [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
|
36
|
Jensen ME, Galli A, Thomsen M, Jensen KL, Thomsen GK, Klausen MK, Vilsbøll T, Christensen MB, Holst JJ, Owens A, Robertson S, Daws L, Zanella D, Gether U, Knudsen GM, Fink-Jensen A. Glucagon-like peptide-1 receptor regulation of basal dopamine transporter activity is species-dependent. Neurochem Int 2020; 138:104772. [PMID: 32464226 DOI: 10.1016/j.neuint.2020.104772] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2019] [Revised: 05/17/2020] [Accepted: 05/20/2020] [Indexed: 12/27/2022]
Abstract
INTRODUCTION A solid body of preclinical evidence shows that glucagon-like peptide-1 receptor (GLP-1R) agonists attenuate the effects of substance use disorder related behaviors. The mechanisms underlying these effects remain elusive. In the present study, we hypothesized that GLP-1R activation modulates dopaminetransporter (DAT) and thus dopamine (DA) homeostasis in striatum. This was evaluated in three different experiments: two preclinical and one clinical. METHODS Rat striatal DA uptake, DA clearance and DAT cell surface expression was assessed following GLP-1 (7-36)-amide exposure in vitro. DA uptake in mice was assesed ex vivo following systemic treatment with the GLP-1R agonist exenatide. In addition, DA uptake was measured in GLP-1R knockout mice and compared with DA-uptake in wild type mice. In healthy humans, changes in DAT availability was assessed during infusion of exenatide measured by single-photon emission computed tomography imaging. RESULTS In rats, GLP-1 (7-36)-amide increased DA uptake, DA clearance and DAT cell surface expression in striatum. In mice, exenatide did not change striatal DA uptake. In GLP-1R knockout mice, DA uptake was similar to what was measured in wildtype mice. In humans, systemic infusion of exenatide did not result in acute changes in striatal DAT availability. CONCLUSIONS The GLP-1R agonist-induced modulation of striatal DAT activity in vitro in rats could not be replicated ex vivo in mice and in vivo in humans. Therefore, the underlying mechanisms of action for the GLP-1R agonists-induced efficacy in varios addiction-like behavioural models still remain.
Collapse
Affiliation(s)
- Mathias E Jensen
- Psychiatric Centre Copenhagen, University Hospital of Copenhagen, Denmark.
| | - Aurelio Galli
- Department of Surgery, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Morgane Thomsen
- Psychiatric Centre Copenhagen, University Hospital of Copenhagen, Denmark
| | - Kathrine L Jensen
- Molecular Neuropharmacology and Genetics Laboratory, Department of Neuroscience, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Gerda K Thomsen
- Neurobiology Research Unit, Neuroscience Centre, Copenhagen University Hospital Rigshospitalet, Copenhagen, Denmark
| | - Mette K Klausen
- Psychiatric Centre Copenhagen, University Hospital of Copenhagen, Denmark
| | - Tina Vilsbøll
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark; Steno Diabetes Center Copenhagen, Gentofte Hospital, Denmark
| | - Mikkel B Christensen
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark; Department of Clinical Pharmacology, Bispebjerg Hospital, University of Copenhagen, Denmark
| | - Jens J Holst
- Novo Nordisk Foundation Center for Basic Metabolic Research Endocrinology and Metabolism, Copenhagen, Denmark
| | - Anthony Owens
- Department of Cellular and Integrative Physiology, University of Texas Health Science Center at San Antonio, USA
| | - Sabrina Robertson
- Department of Psychology and Neuroscience, University of North Carolina at Chapel Hill, USA
| | - Lynette Daws
- Department of Cellular and Integrative Physiology, University of Texas Health Science Center at San Antonio, USA
| | - Daniele Zanella
- Department of Surgery, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Ulrik Gether
- Molecular Neuropharmacology and Genetics Laboratory, Department of Neuroscience, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Gitte M Knudsen
- Neurobiology Research Unit, Neuroscience Centre, Copenhagen University Hospital Rigshospitalet, Copenhagen, Denmark; Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Anders Fink-Jensen
- Psychiatric Centre Copenhagen, University Hospital of Copenhagen, Denmark; Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
37
|
Jerlhag E. Alcohol-mediated behaviours and the gut-brain axis; with focus on glucagon-like peptide-1. Brain Res 2020; 1727:146562. [DOI: 10.1016/j.brainres.2019.146562] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2019] [Revised: 10/28/2019] [Accepted: 11/19/2019] [Indexed: 12/15/2022]
|
38
|
Glucagon-Like Peptide-1 Receptor Agonist Treatment Does Not Reduce Abuse-Related Effects of Opioid Drugs. eNeuro 2019; 6:eN-NRS-0443-18. [PMID: 31058214 PMCID: PMC6498420 DOI: 10.1523/eneuro.0443-18.2019] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2018] [Revised: 02/19/2019] [Accepted: 02/28/2019] [Indexed: 12/29/2022] Open
Abstract
Dependence on opioids and the number of opioid overdose deaths are serious and escalating public health problems, but medication-assisted treatments for opioid addiction remain inadequate for many patients. Glucagon-like pepide-1 (GLP-1) is a gut hormone and neuropeptide with actions in peripheral tissues and in the brain, including regulation of blood glucose and food intake. GLP-1 analogs, which are approved diabetes medications, can reduce the reinforcing and rewarding effects of alcohol, cocaine, amphetamine, and nicotine in rodents. Investigations on effects of GLP-1 analogs on opioid reward and reinforcement have not been reported. We assessed the effects of the GLP-1 receptor agonist Exendin-4 (Ex4) on opioid-related behaviors in male mice, i.e., morphine-conditioned place preference (CPP), intravenous self-administration (IVSA) of the short-acting synthetic opioid remifentanil, naltrexone-precipitated morphine withdrawal, morphine analgesia (male and female mice), and locomotor activity. Ex4 treatment had no effect on morphine-induced CPP, withdrawal, or hyperlocomotion. Ex4 failed to decrease remifentanil self-administration, if anything reinforcing effects of remifentanil appeared increased in Ex4-treated mice relative to saline. Ex4 did not significantly affect analgesia. In contrast, Ex4 dose dependently decreased oral alcohol self-administration, and suppressed spontaneous locomotor activity. Taken together, Ex4 did not attenuate the addiction-related behavioral effects of opioids, indicating that GLP-1 analogs would not be useful medications in the treatment of opioid addiction. This difference between opioids and other drug classes investigated to date may shed light on the mechanism of action of GLP-1 receptor treatment in the addictive effects of alcohol, central stimulants, and nicotine.
Collapse
|