1
|
Lawson EA. Understanding oxytocin in human physiology and pathophysiology: A path towards therapeutics. COMPREHENSIVE PSYCHONEUROENDOCRINOLOGY 2024; 19:100242. [PMID: 38974962 PMCID: PMC11225698 DOI: 10.1016/j.cpnec.2024.100242] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2023] [Revised: 05/06/2024] [Accepted: 05/27/2024] [Indexed: 07/09/2024] Open
Abstract
•Oxytocin is a multifaceted hypothalamic-pituitary hormone involved in energy homeostasis, mental health, and bone metabolism.•Oxytocin deficiency in energy deficit states and in hypopituitarism is associated with worse mental health and bone health.•Oxytocin modulates appetitive neurocircuitry, improves impulse control, and reduces food intake in humans.•Defining the oxytocin system in human physiology and pathophysiology could lead to novel therapeutic strategies.
Collapse
Affiliation(s)
- Elizabeth A. Lawson
- Neuroendocrine Unit, Massachusetts General Hospital, Department of Medicine, Harvard Medical School, 50 Staniford Street, Suite 750B, Boston, MA, 02114, USA
| |
Collapse
|
2
|
Plessow F, Kerem L, Wronski ML, Asanza E, O'Donoghue ML, Stanford FC, Eddy KT, Holmes TM, Misra M, Thomas JJ, Galbiati F, Muhammed M, Sella AC, Hauser K, Smith SE, Holman K, Gydus J, Aulinas A, Vangel M, Healy B, Kheterpal A, Torriani M, Holsen LM, Bredella MA, Lawson EA. Intranasal Oxytocin for Obesity. NEJM EVIDENCE 2024; 3:EVIDoa2300349. [PMID: 38815173 PMCID: PMC11427243 DOI: 10.1056/evidoa2300349] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/01/2024]
Abstract
BACKGROUND Accumulating preclinical and preliminary translational evidence shows that the hypothalamic peptide oxytocin reduces food intake, increases energy expenditure, and promotes weight loss. It is currently unknown whether oxytocin administration is effective in treating human obesity. METHODS In this randomized, double-blind, placebo-controlled trial, we randomly assigned adults with obesity 1:1 (stratified by sex and obesity class) to receive intranasal oxytocin (24 IU) or placebo four times daily for 8 weeks. The primary end point was change in body weight (kg) from baseline to week 8. Key secondary end points included change in body composition (total fat mass [g], abdominal visceral adipose tissue [cm2], and liver fat fraction [proportion; range, 0 to 1; higher values indicate a higher proportion of fat]), and resting energy expenditure (kcal/day; adjusted for lean mass) from baseline to week 8 and caloric intake (kcal) at an experimental test meal from baseline to week 6. RESULTS Sixty-one participants (54% women; mean age ± standard deviation, 33.6 ± 6.2 years; body-mass index [the weight in kilograms divided by the square of the height in meters], 36.9 ± 4.9) were randomly assigned. There was no difference in body weight change from baseline to week 8 between oxytocin and placebo groups (0.20 vs. 0.26 kg; P=0.934). Oxytocin (vs. placebo) was not associated with beneficial effects on body composition or resting energy expenditure from baseline to week 8 (total fat: difference [95% confidence interval], 196.0 g [-1036 to 1428]; visceral fat: 3.1 cm2 [-11.0 to 17.2]; liver fat: -0.01 [-0.03 to 0.01]; resting energy expenditure: -64.0 kcal/day [-129.3 to 1.4]). Oxytocin compared with placebo was associated with reduced caloric intake at the test meal (-31.4 vs. 120.6 kcal; difference [95% confidence interval], -152.0 kcal [-302.3 to -1.7]). There were no serious adverse events. Incidence and severity of adverse events did not differ between groups. CONCLUSIONS In this randomized, placebo-controlled trial in adults with obesity, intranasal oxytocin administered four times daily for 8 weeks did not reduce body weight. (Funded by the National Institute of Diabetes and Digestive and Kidney Diseases and others; ClinicalTrials.gov number, NCT03043053.).
Collapse
Affiliation(s)
- Franziska Plessow
- Neuroendocrine Unit, Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston
| | - Liya Kerem
- Neuroendocrine Unit, Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston
- Division of Pediatric Endocrinology, Department of Pediatrics, Hadassah-Hebrew University Medical Center, Jerusalem
| | - Marie-Louis Wronski
- Neuroendocrine Unit, Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston
- Translational Developmental Neuroscience Section, Division of Psychological and Social Medicine and Developmental Neurosciences, Faculty of Medicine, Technische Universität Dresden, Dresden, Germany
| | - Elisa Asanza
- Neuroendocrine Unit, Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston
| | - Michelle L O'Donoghue
- TIMI Study Group, Division of Cardiovascular Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston
| | - Fatima C Stanford
- Neuroendocrine Unit, Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston
- Division of Pediatric Endocrinology, Department of Pediatrics, Massachusetts General Hospital and Harvard Medical School, Boston
| | - Kamryn T Eddy
- Eating Disorders Clinical and Research Program, Department of Psychiatry, Massachusetts General Hospital and Harvard Medical School, Boston
| | - Tara M Holmes
- Translational and Clinical Research Centers, Massachusetts General Hospital and Harvard Medical School, Boston
| | - Madhusmita Misra
- Neuroendocrine Unit, Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston
- Division of Pediatric Endocrinology, Department of Pediatrics, Massachusetts General Hospital and Harvard Medical School, Boston
| | - Jennifer J Thomas
- Eating Disorders Clinical and Research Program, Department of Psychiatry, Massachusetts General Hospital and Harvard Medical School, Boston
| | - Francesca Galbiati
- Neuroendocrine Unit, Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston
| | - Maged Muhammed
- Neuroendocrine Unit, Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston
| | - Aluma Chovel Sella
- Neuroendocrine Unit, Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston
- The Jesse Z. and Sara Lea Shafer Institute of Endocrinology and Diabetes, National Center for Childhood Diabetes, Schneider Children's Medical Center of Israel, Petah Tikva, Israel
| | - Kristine Hauser
- Neuroendocrine Unit, Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston
| | - Sarah E Smith
- Neuroendocrine Unit, Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston
| | - Katherine Holman
- Neuroendocrine Unit, Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston
| | - Julia Gydus
- Neuroendocrine Unit, Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston
| | - Anna Aulinas
- Neuroendocrine Unit, Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston
- Department of Endocrinology and Nutrition, Hospital de la Santa Creu i Sant Pau, IIB-Sant Pau, Barcelona
| | - Mark Vangel
- Biostatistics Center, Massachusetts General Hospital and Harvard Medical School, Boston
| | - Brian Healy
- Biostatistics Center, Massachusetts General Hospital and Harvard Medical School, Boston
| | - Arvin Kheterpal
- Division of Musculoskeletal Imaging and Intervention, Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston
| | - Martin Torriani
- Division of Musculoskeletal Imaging and Intervention, Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston
| | - Laura M Holsen
- Division of Women's Health, Department of Medicine and Department of Psychiatry, Brigham and Women's Hospital and Harvard Medical School, Boston
| | - Miriam A Bredella
- Division of Musculoskeletal Imaging and Intervention, Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston
| | - Elizabeth A Lawson
- Neuroendocrine Unit, Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston
| |
Collapse
|
3
|
Iovino M, Messana T, Marucci S, Triggiani D, Giagulli VA, Guastamacchia E, Piazzolla G, De Pergola G, Lisco G, Triggiani V. The neurohypophyseal hormone oxytocin and eating behaviors: a narrative review. Hormones (Athens) 2024; 23:15-23. [PMID: 37979096 PMCID: PMC10847364 DOI: 10.1007/s42000-023-00505-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/17/2023] [Accepted: 11/03/2023] [Indexed: 11/19/2023]
Abstract
BACKGROUND The neuropeptide oxytocin (OT) is crucial in several conditions, such as lactation, parturition, mother-infant interaction, and psychosocial function. Moreover, OT may be involved in the regulation of eating behaviors. METHODS This review briefly summarizes data concerning the role of OT in eating behaviors. Appropriate keywords and medical subject headings were identified and searched for in PubMed/MEDLINE. References of original articles and reviews were screened, examined, and selected. RESULTS Hypothalamic OT-secreting neurons project to different cerebral areas controlling eating behaviors, such as the amygdala, area postrema, nucleus of the solitary tract, and dorsal motor nucleus of the vagus nerve. Intracerebral/ventricular OT administration decreases food intake and body weight in wild and genetically obese rats. OT may alter food intake and the quality of meals, especially carbohydrates and sweets, in humans. DISCUSSION OT may play a role in the pathophysiology of eating disorders with potential therapeutic perspectives. In obese patients and those with certain eating disorders, such as bulimia nervosa or binge/compulsive eating, OT may reduce appetite and caloric consumption. Conversely, OT administered to patients with anorexia nervosa may paradoxically stimulate appetite, possibly by lowering anxiety which usually complicates the management of these patients. Nevertheless, OT administration (e.g., intranasal route) is not always associated with clinical benefit, probably because intranasally administered OT fails to achieve therapeutic intracerebral levels of the hormone. CONCLUSION OT administration could play a therapeutic role in managing eating disorders and disordered eating. However, specific studies are needed to clarify this issue with regard to dose-finding and route and administration time.
Collapse
Affiliation(s)
- Michele Iovino
- Interdisciplinary Department of Medicine, Section of Internal Medicine, Geriatrics, Endocrinology and Rare Diseases, University of Bari "Aldo Moro", School of Medicine, Bari, Apulia, Italy
| | - Tullio Messana
- Infantile Neuropsychiatry, IRCCS - Institute of Neurological Sciences, Bologna, Italy
| | - Simonetta Marucci
- Università Campus Biomedico, Dip. "Scienze e Tecnologie per l'Uomo e l'ambiente", Via Alvaro del Portillo, 21, Roma, Italy
| | - Domenico Triggiani
- Interdisciplinary Department of Medicine, Section of Internal Medicine, Geriatrics, Endocrinology and Rare Diseases, University of Bari "Aldo Moro", School of Medicine, Bari, Apulia, Italy
| | - Vito Angelo Giagulli
- Interdisciplinary Department of Medicine, Section of Internal Medicine, Geriatrics, Endocrinology and Rare Diseases, University of Bari "Aldo Moro", School of Medicine, Bari, Apulia, Italy
| | - Edoardo Guastamacchia
- Interdisciplinary Department of Medicine, Section of Internal Medicine, Geriatrics, Endocrinology and Rare Diseases, University of Bari "Aldo Moro", School of Medicine, Bari, Apulia, Italy
| | - Giuseppina Piazzolla
- Interdisciplinary Department of Medicine, Section of Internal Medicine, Geriatrics, Endocrinology and Rare Diseases, University of Bari "Aldo Moro", School of Medicine, Bari, Apulia, Italy
| | - Giovanni De Pergola
- National Institute of Gastroenterology IRCCS "Saverio de Bellis", Research Hospital, Castellana Grotte, Bari, Italy
- Department of Biomedical Science and Human Oncology, University of Bari, School of Medicine, Bari, Apulia, Italy
| | - Giuseppe Lisco
- Interdisciplinary Department of Medicine, Section of Internal Medicine, Geriatrics, Endocrinology and Rare Diseases, University of Bari "Aldo Moro", School of Medicine, Bari, Apulia, Italy.
| | - Vincenzo Triggiani
- Interdisciplinary Department of Medicine, Section of Internal Medicine, Geriatrics, Endocrinology and Rare Diseases, University of Bari "Aldo Moro", School of Medicine, Bari, Apulia, Italy
| |
Collapse
|
4
|
Cuesta-Marti C, Uhlig F, Muguerza B, Hyland N, Clarke G, Schellekens H. Microbes, oxytocin and stress: Converging players regulating eating behavior. J Neuroendocrinol 2023; 35:e13243. [PMID: 36872624 DOI: 10.1111/jne.13243] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/21/2022] [Revised: 01/26/2023] [Accepted: 02/02/2023] [Indexed: 02/17/2023]
Abstract
Oxytocin is a peptide-hormone extensively studied for its multifaceted biological functions and has recently gained attention for its role in eating behavior, through its action as an anorexigenic neuropeptide. Moreover, the gut microbiota is involved in oxytocinergic signaling through the brain-gut axis, specifically in the regulation of social behavior. The gut microbiota is also implicated in appetite regulation and is postulated to play a role in central regulation of hedonic eating. In this review, we provide an overview on oxytocin and its individual links with the microbiome, the homeostatic and non-homeostatic regulation of eating behavior as well as social behavior and stress.
Collapse
Affiliation(s)
- Cristina Cuesta-Marti
- Department of Anatomy & Neuroscience, University College Cork, Cork, Ireland
- APC Microbiome Ireland, Cork, Ireland
| | - Friederike Uhlig
- APC Microbiome Ireland, Cork, Ireland
- Department of Physiology, University College Cork, Ireland
| | - Begoña Muguerza
- Department of Anatomy & Neuroscience, University College Cork, Cork, Ireland
- APC Microbiome Ireland, Cork, Ireland
- Universitat Rovira i Virgili, Department of Biochemistry & Biotechnology, Nutrigenomics Research Group, Tarragona, Spain
| | - Niall Hyland
- APC Microbiome Ireland, Cork, Ireland
- Department of Physiology, University College Cork, Ireland
| | - Gerard Clarke
- APC Microbiome Ireland, Cork, Ireland
- Department of Psychiatry & Neurobehavioural Science, University College Cork, Cork, Ireland
| | - Harriët Schellekens
- Department of Anatomy & Neuroscience, University College Cork, Cork, Ireland
- APC Microbiome Ireland, Cork, Ireland
| |
Collapse
|
5
|
Aulinas A, Muhammed M, Becker KR, Asanza E, Hauser K, Stern C, Gydus J, Holmes T, Murray HB, Breithaupt L, Micali N, Misra M, Eddy KT, Thomas JJ, Lawson EA. Oxytocin response to food intake in avoidant/restrictive food intake disorder. Eur J Endocrinol 2023; 189:149-155. [PMID: 37474111 PMCID: PMC10396082 DOI: 10.1093/ejendo/lvad087] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/22/2023] [Revised: 05/16/2023] [Accepted: 06/07/2023] [Indexed: 07/22/2023]
Abstract
OBJECTIVE To investigate the response of anorexigenic oxytocin to food intake among adolescents and young adults with avoidant/restrictive food intake disorder (ARFID), a restrictive eating disorder characterized by lack of interest in food or eating, sensory sensitivity to food, and/or fear of aversive consequences of eating, compared with healthy controls (HC). DESIGN Cross-sectional. METHODS A total of 109 participants (54 with ARFID spectrum and 55 HC) were instructed to eat a ∼400-kcal standardized mixed meal. We sampled serum oxytocin at fasting and at 30-, 60-, and 120-min postmeal. We tested the hypothesis that ARFID would show higher mean oxytocin levels across time points compared with HC using a mixed model ANOVA. We then used multivariate regression analysis to identify the impact of clinical characteristics (sex, age, and body mass index [BMI] percentile) on oxytocin levels in individuals with ARFID. RESULTS Participants with ARFID exhibited greater mean oxytocin levels at all time points compared with HC, and these differences remained significant even after controlling for sex and BMI percentile (P = .004). Clinical variables (sex, age, and BMI percentile) did not show any impact on fasting and postprandial oxytocin levels among individuals with ARFID. CONCLUSIONS Consistently high oxytocin levels might be involved in low appetite and sensory aversions to food, contributing to food avoidance in individuals with ARFID.
Collapse
Affiliation(s)
- Anna Aulinas
- Department of Endocrinology and Nutrition, Hospital de la Santa Creu i Sant Pau, IIB-Sant Pau, 08041 Barcelona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER Unidad 747), ISCIII, 08025 Barcelona, Spain
- Department of Medicine, University of Vic—Central University of Catalonia, 08500 Vic, Barcelona, Spain
| | - Maged Muhammed
- Neuroendocrine Unit, Division of Endocrinology, Department of Medicine, Massachusetts General Hospital, 02114 Boston, MA, United States
| | - Kendra R Becker
- Department of Medicine, Harvard Medical School, 02115 Boston, MA, United States
- Eating Disorders Clinical and Research Program, Department of Psychiatry, Massachusetts General Hospital, 02114 Boston, MA, United States
| | - Elisa Asanza
- Neuroendocrine Unit, Division of Endocrinology, Department of Medicine, Massachusetts General Hospital, 02114 Boston, MA, United States
| | - Kristine Hauser
- Neuroendocrine Unit, Division of Endocrinology, Department of Medicine, Massachusetts General Hospital, 02114 Boston, MA, United States
| | - Casey Stern
- Neuroendocrine Unit, Division of Endocrinology, Department of Medicine, Massachusetts General Hospital, 02114 Boston, MA, United States
- Eating Disorders Clinical and Research Program, Department of Psychiatry, Massachusetts General Hospital, 02114 Boston, MA, United States
| | - Julia Gydus
- Neuroendocrine Unit, Division of Endocrinology, Department of Medicine, Massachusetts General Hospital, 02114 Boston, MA, United States
- Eating Disorders Clinical and Research Program, Department of Psychiatry, Massachusetts General Hospital, 02114 Boston, MA, United States
| | - Tara Holmes
- Translational and Clinical Research Center, Massachusetts General Hospital, 02114 Boston, MA, United States
| | - Helen Burton Murray
- Department of Medicine, Harvard Medical School, 02115 Boston, MA, United States
- Center for Neurointestinal Health, Division of Gastroenterology, Massachusetts General Hospital, 02114 Boston, MA, United States
| | - Lauren Breithaupt
- Department of Medicine, Harvard Medical School, 02115 Boston, MA, United States
- Eating Disorders Clinical and Research Program, Department of Psychiatry, Massachusetts General Hospital, 02114 Boston, MA, United States
| | - Nadia Micali
- Eating Disorders Research Unit, Mental Health Services of the Capital Region of Denmark, Psychiatric Centre Ballerup, 2750 Ballerup, Denmark
- Population, Policy and Practice, Great Ormond Street Institute of Child Health, University College London, WC1N 1EH, London, United Kingdom
| | - Madhusmita Misra
- Neuroendocrine Unit, Division of Endocrinology, Department of Medicine, Massachusetts General Hospital, 02114 Boston, MA, United States
- Department of Medicine, Harvard Medical School, 02115 Boston, MA, United States
- Division of Pediatric Endocrinology, Massachusetts General Hospital, 02114 Boston, MA, United States
| | - Kamryn T Eddy
- Department of Medicine, Harvard Medical School, 02115 Boston, MA, United States
- Eating Disorders Clinical and Research Program, Department of Psychiatry, Massachusetts General Hospital, 02114 Boston, MA, United States
| | - Jennifer J Thomas
- Department of Medicine, Harvard Medical School, 02115 Boston, MA, United States
- Eating Disorders Clinical and Research Program, Department of Psychiatry, Massachusetts General Hospital, 02114 Boston, MA, United States
| | - Elizabeth A Lawson
- Neuroendocrine Unit, Division of Endocrinology, Department of Medicine, Massachusetts General Hospital, 02114 Boston, MA, United States
- Department of Medicine, Harvard Medical School, 02115 Boston, MA, United States
| |
Collapse
|
6
|
Becetti I, Bwenyi EL, de Araujo IE, Ard J, Cryan JF, Farooqi IS, Ferrario CR, Gluck ME, Holsen LM, Kenny PJ, Lawson EA, Lowell BB, Schur EA, Stanley TL, Tavakkoli A, Grinspoon SK, Singhal V. The Neurobiology of Eating Behavior in Obesity: Mechanisms and Therapeutic Targets: A Report from the 23rd Annual Harvard Nutrition Obesity Symposium. Am J Clin Nutr 2023; 118:314-328. [PMID: 37149092 PMCID: PMC10375463 DOI: 10.1016/j.ajcnut.2023.05.003] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2022] [Revised: 04/03/2023] [Accepted: 05/01/2023] [Indexed: 05/08/2023] Open
Abstract
Obesity is increasing at an alarming rate. The effectiveness of currently available strategies for the treatment of obesity (including pharmacologic, surgical, and behavioral interventions) is limited. Understanding the neurobiology of appetite and the important drivers of energy intake (EI) can lead to the development of more effective strategies for the prevention and treatment of obesity. Appetite regulation is complex and is influenced by genetic, social, and environmental factors. It is intricately regulated by a complex interplay of endocrine, gastrointestinal, and neural systems. Hormonal and neural signals generated in response to the energy state of the organism and the quality of food eaten are communicated by paracrine, endocrine, and gastrointestinal signals to the nervous system. The central nervous system integrates homeostatic and hedonic signals to regulate appetite. Although there has been an enormous amount of research over many decades regarding the regulation of EI and body weight, research is only now yielding potentially effective treatment strategies for obesity. The purpose of this article is to summarize the key findings presented in June 2022 at the 23rd annual Harvard Nutrition Obesity Symposium entitled "The Neurobiology of Eating Behavior in Obesity: Mechanisms and Therapeutic Targets." Findings presented at the symposium, sponsored by NIH P30 Nutrition Obesity Research Center at Harvard, enhance our current understanding of appetite biology, including innovative techniques used to assess and systematically manipulate critical hedonic processes, which will shape future research and the development of therapeutics for obesity prevention and treatment.
Collapse
Affiliation(s)
- Imen Becetti
- Division of Pediatric Endocrinology, Massachusetts General Hospital for Children and Harvard Medical School, Boston, MA, United States.
| | - Esther L Bwenyi
- Metabolism Unit, Massachusetts General Hospital, Boston, MA, United States; Nutrition Obesity Research Center at Harvard Medical School, Massachusetts General Hospital, Boston, MA, United States
| | - Ivan E de Araujo
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York City, NY, United States; Diabetes, Obesity, and Metabolism Institute, Icahn School of Medicine at Mount Sinai, New York City, NY, United States
| | - Jamy Ard
- Epidemiology and Prevention, Wake Forest University School of Medicine, Winston-Salem, NC, United States; Bariatric and Weight Management Center, Wake Forest Baptist Health, Winston-Salem, NC, United States; Center on Diabetes, Obesity, and Metabolism, Wake Forest University School of Medicine, Winston-Salem, NC, United States; Sticht Center for Healthy Aging and Alzheimer's Prevention, Wake Forest University School of Medicine, Winston-Salem, NC, United States; Hypertension and Vascular Research Center, Cardiovascular Sciences Center, Wake Forest University School of Medicine, Winston-Salem, NC, United States; Maya Angelou Center for Healthy Equity, Wake Forest University School of Medicine, Winston-Salem, NC, United States
| | - John F Cryan
- Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland; APC Microbiome Ireland, University College Cork, Cork, Ireland
| | - Ismaa Sadaf Farooqi
- University of Cambridge Metabolic Research Laboratories and National Institute for Health and Care Research (NIHR) Cambridge Biomedical Research Centre, University of Cambridge, Cambridge, United Kingdom; Wellcome-Medical Research Council (MRC) Institute of Metabolic Science, University of Cambridge, Cambridge, United Kingdom; Addenbrooke's Hospital, Cambridge University Hospitals, Cambridge, United Kingdom
| | - Carrie R Ferrario
- Department of Pharmacology, Psychology Department (Biopsychology Area), University of Michigan, Ann Arbor, MI, United States
| | - Marci E Gluck
- National Institutes of Health, Phoenix, AZ, United States; National Institute of Diabetes and Digestive and Kidney Disease, Obesity and Diabetes Clinical Research Section, Phoenix Epidemiology and Clinical Research Branch, Phoenix, AZ, United States
| | - Laura M Holsen
- Harvard Medical School, Boston, MA, United States; Division of Women's Health, Department of Medicine, Brigham and Women's Hospital, Boston, MA, United States; Department of Psychiatry, Brigham and Women's Hospital, Boston, MA, United States
| | - Paul J Kenny
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York City, NY, United States; Diabetes, Obesity, and Metabolism Institute, Icahn School of Medicine at Mount Sinai, New York City, NY, United States
| | - Elizabeth A Lawson
- Nutrition Obesity Research Center at Harvard Medical School, Massachusetts General Hospital, Boston, MA, United States; Department of Medicine, Harvard Medical School, Boston, MA, United States; Neuroendocrine Unit, Massachusetts General Hospital, Boston, MA, United States
| | - Bradford B Lowell
- Department of Medicine, Division of Endocrinology, Diabetes and Metabolism, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA, United States
| | - Ellen A Schur
- Division of General Internal Medicine, University of Washington, Seattle, WA, United States; Univeristy of Washington Medicine Diabetes Institute, University of Washington, Seattle, WA, United States; Univeristy of Washington Nutrition and Obesity Research Center, University of Washington, Seattle, WA, United States; Clinical and Translational Research Services Core, University of Washington, Seattle, WA, United States
| | - Takara L Stanley
- Division of Pediatric Endocrinology, Massachusetts General Hospital for Children and Harvard Medical School, Boston, MA, United States; Metabolism Unit, Massachusetts General Hospital, Boston, MA, United States; Nutrition Obesity Research Center at Harvard Medical School, Massachusetts General Hospital, Boston, MA, United States; Harvard Medical School, Boston, MA, United States
| | - Ali Tavakkoli
- Division of General and Gastrointestinal (GI) Surgery, Center for Weight Management and Wellness, Advanced Minimally Invasive Fellowship, Harvard Medical School, Boston, MA, United States
| | - Steven K Grinspoon
- Metabolism Unit, Massachusetts General Hospital, Boston, MA, United States; Nutrition Obesity Research Center at Harvard Medical School, Massachusetts General Hospital, Boston, MA, United States; Harvard Medical School, Boston, MA, United States; Department of Medicine, Harvard Medical School, Boston, MA, United States
| | - Vibha Singhal
- Division of Pediatric Endocrinology, Massachusetts General Hospital for Children and Harvard Medical School, Boston, MA, United States; Harvard Medical School, Boston, MA, United States; Pediatric Endocrinology and Obesity Medicine, Massachusetts General Hospital, Boston, MA, United States; Pediatric Program MGH Weight Center, Massachusetts General Hospital, Boston, MA, United States
| |
Collapse
|
7
|
Szmygin H, Szmygin M, Cheda M, Kłobuszewski B, Drelich-Zbroja A, Matyjaszek-Matuszek B. Current Insights into the Potential Role of fMRI in Discovering the Mechanisms Underlying Obesity. J Clin Med 2023; 12:4379. [PMID: 37445414 DOI: 10.3390/jcm12134379] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2023] [Revised: 06/19/2023] [Accepted: 06/28/2023] [Indexed: 07/15/2023] Open
Abstract
Obesity is becoming one of the major global health concerns. This chronic disease affects around 650 million people worldwide and is an underlying cause of a number of significant comorbidities. According to the World Health Organization (WHO) report on obesity from 2022, this disorder became the fourth leading cause of deaths in Europe. Thus, understanding the mechanisms underlying obesity is of essential importance to successfully prevent and treat this disease. The aim of this study was to review the current insights into the potential role of fMRI in discovering the mechanisms underlying obesity on the basis of recent scientific literature published up to December 2022 and searches of the PubMed, Google Scholar and Web of Science databases. The literature assessed indicated that a growing body of evidence suggests that obesity leads to changes in both structure and connectivity within the central nervous system. Emerging data from recent functional magnetic resonance imaging (fMRI) studies prove that obese individuals present an increased motivational drive to eat as well as impaired processing in reward- and control-related brain regions. Apart from this, it is clear that fMRI might be a useful tool in detection of obesity-induced changes within the central nervous system.
Collapse
Affiliation(s)
- Hanna Szmygin
- Department of Endocrinology, Diabetology and Metabolic Diseases, Medical University of Lublin, 20-093 Lublin, Poland
| | - Maciej Szmygin
- Department of Interventional Radiology and Neuroradiology, Medical University of Lublin, 20-093 Lublin, Poland
| | - Mateusz Cheda
- Department of Interventional Radiology and Neuroradiology, Medical University of Lublin, 20-093 Lublin, Poland
| | - Bartosz Kłobuszewski
- Department of Interventional Radiology and Neuroradiology, Medical University of Lublin, 20-093 Lublin, Poland
| | - Anna Drelich-Zbroja
- Department of Interventional Radiology and Neuroradiology, Medical University of Lublin, 20-093 Lublin, Poland
| | - Beata Matyjaszek-Matuszek
- Department of Endocrinology, Diabetology and Metabolic Diseases, Medical University of Lublin, 20-093 Lublin, Poland
| |
Collapse
|
8
|
Kilpatrick LA, An HM, Pawar S, Sood R, Gupta A. Neuroimaging Investigations of Obesity: a Review of the Treatment of Sex from 2010. Curr Obes Rep 2023; 12:163-174. [PMID: 36933153 PMCID: PMC10250271 DOI: 10.1007/s13679-023-00498-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 02/15/2023] [Indexed: 03/19/2023]
Abstract
PURPOSE OF REVIEW To summarize the results of adult obesity neuroimaging studies (structural, resting-state, task-based, diffusion tensor imaging) published from 2010, with a focus on the treatment of sex as an important biological variable in the analysis, and identify gaps in sex difference research. RECENT FINDINGS Neuroimaging studies have shown obesity-related changes in brain structure, function, and connectivity. However, relevant factors such as sex are often not considered. We conducted a systematic review and keyword co-occurrence analysis. Literature searches identified 6281 articles, of which 199 met inclusion criteria. Among these, only 26 (13%) considered sex as an important variable in the analysis, directly comparing the sexes (n = 10; 5%) or providing single-sex/disaggregated data (n = 16, 8%); the remaining studies controlled for sex (n = 120, 60%) or did not consider sex in the analysis (n = 53, 27%). Synthesizing sex-based results, obesity-related parameters (e.g., body mass index, waist circumference, obese status) may be generally associated with more robust morphological alterations in men and more robust structural connectivity alterations in women. Additionally, women with obesity generally expressed increased reactivity in affect-related regions, while men with obesity generally expressed increased reactivity in motor-related regions; this was especially true under a fed state. The keyword co-occurrence analysis indicated that sex difference research was especially lacking in intervention studies. Thus, although sex differences in the brain associated with obesity are known to exist, a large proportion of the literature informing the research and treatment strategies of today has not specifically examined sex effects, which is needed to optimize treatment.
Collapse
Affiliation(s)
- Lisa A Kilpatrick
- Vatche and Tamar Manoukian Division of Digestive Diseases, David Geffen School of Medicine, University of California, Los Angeles, USA
- David Geffen School of Medicine, Goodman-Luskin Microbiome Center, University of California, Los Angeles, USA
- G. Oppenheimer Center for Neurobiology of Stress and Resilience, The Obesity and Ingestive Behavior Program, Vatche and Tamar Manoukian Division of Digestive Diseases, David Geffen School of Medicine, University of California, 10833 Le Conte Avenue, Center for Health Sciences 42-210, Los Angeles, CA, 90095, USA
| | - Hyeon Min An
- Vatche and Tamar Manoukian Division of Digestive Diseases, David Geffen School of Medicine, University of California, Los Angeles, USA
- David Geffen School of Medicine, Goodman-Luskin Microbiome Center, University of California, Los Angeles, USA
- G. Oppenheimer Center for Neurobiology of Stress and Resilience, The Obesity and Ingestive Behavior Program, Vatche and Tamar Manoukian Division of Digestive Diseases, David Geffen School of Medicine, University of California, 10833 Le Conte Avenue, Center for Health Sciences 42-210, Los Angeles, CA, 90095, USA
| | - Shrey Pawar
- G. Oppenheimer Center for Neurobiology of Stress and Resilience, The Obesity and Ingestive Behavior Program, Vatche and Tamar Manoukian Division of Digestive Diseases, David Geffen School of Medicine, University of California, 10833 Le Conte Avenue, Center for Health Sciences 42-210, Los Angeles, CA, 90095, USA
| | - Riya Sood
- G. Oppenheimer Center for Neurobiology of Stress and Resilience, The Obesity and Ingestive Behavior Program, Vatche and Tamar Manoukian Division of Digestive Diseases, David Geffen School of Medicine, University of California, 10833 Le Conte Avenue, Center for Health Sciences 42-210, Los Angeles, CA, 90095, USA
| | - Arpana Gupta
- Vatche and Tamar Manoukian Division of Digestive Diseases, David Geffen School of Medicine, University of California, Los Angeles, USA.
- David Geffen School of Medicine, Goodman-Luskin Microbiome Center, University of California, Los Angeles, USA.
- G. Oppenheimer Center for Neurobiology of Stress and Resilience, The Obesity and Ingestive Behavior Program, Vatche and Tamar Manoukian Division of Digestive Diseases, David Geffen School of Medicine, University of California, 10833 Le Conte Avenue, Center for Health Sciences 42-210, Los Angeles, CA, 90095, USA.
| |
Collapse
|
9
|
Brain functional and structural magnetic resonance imaging of obesity and weight loss interventions. Mol Psychiatry 2023; 28:1466-1479. [PMID: 36918706 DOI: 10.1038/s41380-023-02025-y] [Citation(s) in RCA: 29] [Impact Index Per Article: 29.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/22/2022] [Revised: 02/26/2023] [Accepted: 02/28/2023] [Indexed: 03/16/2023]
Abstract
Obesity has tripled over the past 40 years to become a major public health issue, as it is linked with increased mortality and elevated risk for various physical and neuropsychiatric illnesses. Accumulating evidence from neuroimaging studies suggests that obesity negatively affects brain function and structure, especially within fronto-mesolimbic circuitry. Obese individuals show abnormal neural responses to food cues, taste and smell, resting-state activity and functional connectivity, and cognitive tasks including decision-making, inhibitory-control, learning/memory, and attention. In addition, obesity is associated with altered cortical morphometry, a lowered gray/white matter volume, and impaired white matter integrity. Various interventions and treatments including bariatric surgery, the most effective treatment for obesity in clinical practice, as well as dietary, exercise, pharmacological, and neuromodulation interventions such as transcranial direct current stimulation, transcranial magnetic stimulation and neurofeedback have been employed and achieved promising outcomes. These interventions and treatments appear to normalize hyper- and hypoactivations of brain regions involved with reward processing, food-intake control, and cognitive function, and also promote recovery of brain structural abnormalities. This paper provides a comprehensive literature review of the recent neuroimaging advances on the underlying neural mechanisms of both obesity and interventions, in the hope of guiding development of novel and effective treatments.
Collapse
|
10
|
Asker M, Krieger JP, Liles A, Tinsley IC, Borner T, Maric I, Doebley S, Furst CD, Börchers S, Longo F, Bhat YR, De Jonghe BC, Hayes MR, Doyle RP, Skibicka KP. Peripherally restricted oxytocin is sufficient to reduce food intake and motivation, while CNS entry is required for locomotor and taste avoidance effects. Diabetes Obes Metab 2023; 25:856-877. [PMID: 36495318 PMCID: PMC9987651 DOI: 10.1111/dom.14937] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/17/2022] [Revised: 11/21/2022] [Accepted: 12/02/2022] [Indexed: 12/14/2022]
Abstract
OBJECTIVES Oxytocin (OT) has a well-established role in reproductive behaviours; however, it recently emerged as an important regulator of energy homeostasis. In addition to central nervous system (CNS), OT is found in the plasma and OT receptors (OT-R) are found in peripheral tissues relevant to energy balance regulation. Here, we aim to determine whether peripheral OT-R activation is sufficient to alter energy intake and expenditure. METHODS AND RESULTS We first show that systemic OT potently reduced food intake and food-motivated behaviour for a high-fat reward in male and female rats. As it is plausible that peripherally, intraperitoneally (IP) injected OT crosses the blood-brain barrier (BBB) to produce some of the metabolic effects within the CNS, we screened, with a novel fluorescently labelled-OT (fAF546-OT, Roxy), for the presence of IP-injected Roxy in CNS tissue relevant to feeding control and compared such with BBB-impermeable fluorescent OT-B12 (fCy5-OT-B12; BRoxy). While Roxy did penetrate the CNS, BRoxy did not. To evaluate the behavioural and thermoregulatory impact of exclusive activation of peripheral OT-R, we generated a novel BBB-impermeable OT (OT-B12 ), with equipotent binding at OT-R in vitro. In vivo, IP-injected OT and OT-B12 were equipotent at food intake suppression in rats of both sexes, suggesting that peripheral OT acts on peripheral OT-R to reduce feeding behaviour. Importantly, OT induced a potent conditioned taste avoidance, indistinguishable from that induced by LiCl, when applied peripherally. Remarkably, and in contrast to OT, OT-B12 did not induce any conditioned taste avoidance. Limiting the CNS entry of OT also resulted in a dose-dependent reduction of emesis in male shrews. While both OT and OT-B12 proved to have similar effects on body temperature, only OT resulted in home-cage locomotor depression. CONCLUSIONS Together our data indicate that limiting systemic OT CNS penetrance preserves the anorexic effects of the peptide and reduces the clinically undesired side effects of OT: emesis, taste avoidance and locomotor depression. Thus, therapeutic targeting of peripheral OT-R may be a viable strategy to achieve appetite suppression with better patient outcomes.
Collapse
Affiliation(s)
- Mohammed Asker
- Department of Physiology/Metabolic Physiology, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
- Wallenberg Centre for molecular and translational medicine, University of Gothenburg, Gothenburg, Sweden
| | - Jean-Philippe Krieger
- Department of Physiology/Metabolic Physiology, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
| | - Amber Liles
- Department of Chemistry, Syracuse University, Syracuse, New York, USA
| | - Ian C Tinsley
- Department of Chemistry, Syracuse University, Syracuse, New York, USA
| | - Tito Borner
- Department of Biobehavioral Health Sciences, University of Pennsylvania, School of Nursing, Philadelphia, Pennsylvania, USA
- Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Ivana Maric
- Department of Physiology/Metabolic Physiology, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
- Department of Chemistry, Syracuse University, Syracuse, New York, USA
- Department of Nutritional Sciences, Pennsylvania State University, University Park, Pennsylvania, USA
| | - Sarah Doebley
- Department of Biobehavioral Health Sciences, University of Pennsylvania, School of Nursing, Philadelphia, Pennsylvania, USA
| | - C Daniel Furst
- Department of Biobehavioral Health Sciences, University of Pennsylvania, School of Nursing, Philadelphia, Pennsylvania, USA
| | - Stina Börchers
- Department of Physiology/Metabolic Physiology, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
- Wallenberg Centre for molecular and translational medicine, University of Gothenburg, Gothenburg, Sweden
| | - Francesco Longo
- Department of Physiology/Metabolic Physiology, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
| | - Yashaswini R Bhat
- Department of Nutritional Sciences, Pennsylvania State University, University Park, Pennsylvania, USA
| | - Bart C De Jonghe
- Department of Biobehavioral Health Sciences, University of Pennsylvania, School of Nursing, Philadelphia, Pennsylvania, USA
- Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Matthew R Hayes
- Department of Biobehavioral Health Sciences, University of Pennsylvania, School of Nursing, Philadelphia, Pennsylvania, USA
- Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Robert P Doyle
- Department of Chemistry, Syracuse University, Syracuse, New York, USA
- Departments of Medicine and Pharmacology, State University of New York, Upstate Medical University, Syracuse, New York, USA
| | - Karolina P Skibicka
- Department of Physiology/Metabolic Physiology, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
- Wallenberg Centre for molecular and translational medicine, University of Gothenburg, Gothenburg, Sweden
- Department of Nutritional Sciences, Pennsylvania State University, University Park, Pennsylvania, USA
| |
Collapse
|
11
|
Kosmalski M, Deska K, Bąk B, Różycka-Kosmalska M, Pietras T. Pharmacological Support for the Treatment of Obesity-Present and Future. Healthcare (Basel) 2023; 11:433. [PMID: 36767008 PMCID: PMC9914730 DOI: 10.3390/healthcare11030433] [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: 11/15/2022] [Revised: 01/25/2023] [Accepted: 02/01/2023] [Indexed: 02/05/2023] Open
Abstract
Obesity is a growing civilization problem, associated with a number of negative health consequences affecting almost all tissues and organs. Currently, obesity treatment includes lifestyle modifications (including diet and exercise), pharmacologic therapies, and in some clinical situations, bariatric surgery. These treatments seem to be the most effective method supporting the treatment of obesity. However, they are many limitations to the options, both for the practitioners and patients. Often the comorbidities, cost, age of the patient, and even geographic locations may influence the choices. The pharmacotherapy of obesity is a fast-growing market. Currently, we have at our disposal drugs with various mechanisms of action (directly reducing the absorption of calories-orlistat, acting centrally-bupropion with naltrexone, phentermine with topiramate, or multidirectional-liraglutide, dulaglutide, semaglutide). The drugs whose weight-reducing effect is used in the course of the pharmacotherapy of other diseases (e.g., glucose-sodium cotransporter inhibitors, exenatide) are also worth mentioning. The obesity pharmacotherapy is focusing on novel therapeutic agents with improved safety and efficacy profiles. These trends also include an assessment of the usefulness of the weight-reducing properties of the drugs previously used for other diseases. The presented paper is an overview of the studies related to both drugs currently used in the pharmacotherapy of obesity and those undergoing clinical trials, taking into account the individual approach to the patient.
Collapse
Affiliation(s)
- Marcin Kosmalski
- Department of Clinical Pharmacology, Medical University of Lodz, 90-153 Łódź, Poland
| | - Kacper Deska
- Students’ Scientific Association Clinical Pharmacology, Medical University of Lodz, 90-153 Łódź, Poland
| | - Bartłomiej Bąk
- 2nd Department of Psychiatry, Institute of Psychiatry and Neurology in Warsaw, 02-957 Warszawa, Poland
| | | | - Tadeusz Pietras
- Department of Clinical Pharmacology, Medical University of Lodz, 90-153 Łódź, Poland
- 2nd Department of Psychiatry, Institute of Psychiatry and Neurology in Warsaw, 02-957 Warszawa, Poland
| |
Collapse
|
12
|
Camerino C. The Long Way of Oxytocin from the Uterus to the Heart in 70 Years from Its Discovery. Int J Mol Sci 2023; 24:ijms24032556. [PMID: 36768879 PMCID: PMC9916674 DOI: 10.3390/ijms24032556] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2022] [Revised: 01/13/2023] [Accepted: 01/19/2023] [Indexed: 01/31/2023] Open
Abstract
The research program on oxytocin started in 1895, when Oliver and Schafer reported that a substance extracted from the pituitary gland elevates blood pressure when injected intravenously into dogs. Dale later reported that a neurohypophysial substance triggers uterine contraction, lactation, and antidiuresis. Purification of this pituitary gland extracts revealed that the vasopressor and antidiuretic activity could be attributed to vasopressin, while uterotonic and lactation activity could be attributed to oxytocin. In 1950, the amino-acid sequences of vasopressin and oxytocin were determined and chemically synthesized. Vasopressin (CYFQNCPRG-NH2) and oxytocin (CYIQNCPLG-NH2) differ by two amino acids and have a disulfide bridge between the cysteine residues at position one and six conserved in all vasopressin/oxytocin-type peptides. This characterization of oxytocin led to the Nobel Prize awarded in 1955 to Vincent du Vigneaud. Nevertheless, it was only 50 years later when the evidence that mice depleted of oxytocin or its receptor develop late-onset obesity and metabolic syndrome established that oxytocin regulates energy and metabolism. Oxytocin is anorexigenic and regulates the lean/fat mass composition in skeletal muscle. Oxytocin's effect on muscle is mediated by thermogenesis via a pathway initiated in the myocardium. Oxytocin involvement in thermogenesis and muscle contraction is linked to Prader-Willi syndrome in humans, opening exciting therapeutic avenues.
Collapse
Affiliation(s)
- Claudia Camerino
- Department of Biomedical Sciences and Human Oncology, Section of Pharmacology, School of Medicine, University of Bari “Aldo Moro”, P.za G. Cesare 11, 70100 Bari, Italy;
- Department of Physiology and Pharmacology “V. Erspamer”, Sapienza University of Rome, P.le Aldo Moro 5, 00185 Rome, Italy
| |
Collapse
|
13
|
Wronski ML, Plessow F, Kerem L, Asanza E, O'Donoghue ML, Stanford FC, Bredella MA, Torriani M, Soukas AA, Kheterpal A, Eddy KT, Holmes TM, Deckersbach T, Vangel M, Holsen LM, Lawson EA. A randomized, double-blind, placebo-controlled clinical trial of 8-week intranasal oxytocin administration in adults with obesity: Rationale, study design, and methods. Contemp Clin Trials 2022; 122:106909. [PMID: 36087842 PMCID: PMC10329413 DOI: 10.1016/j.cct.2022.106909] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2022] [Revised: 08/28/2022] [Accepted: 09/02/2022] [Indexed: 01/27/2023]
Abstract
BACKGROUND Obesity affects more than one-third of adults in the U.S., and effective treatment options are urgently needed. Oxytocin administration induces weight loss in animal models of obesity via effects on caloric intake, energy expenditure, and fat metabolism. We study intranasal oxytocin, an investigational drug shown to reduce caloric intake in humans, as a potential novel treatment for obesity. METHODS We report the rationale, design, methods, and biostatistical analysis plan of a randomized, double-blind, placebo-controlled clinical trial of intranasal oxytocin for weight loss (primary endpoint) in adults with obesity. Participants (aged 18-45 years) were randomly allocated (1:1) to oxytocin (four times daily over eight weeks) versus placebo. Randomization was stratified by biological sex and BMI (30 to <35, 35 to <40, ≥40 kg/m2). We investigate the efficacy, safety, and mechanisms of oxytocin administration in reducing body weight. Secondary endpoints include changes in resting energy expenditure, body composition, caloric intake, metabolic profile, and brain activation via functional magnetic resonance imaging in response to food images and during an impulse control task. Safety and tolerability (e.g., review of adverse events, vital signs, electrocardiogram, comprehensive metabolic panel) are assessed throughout the study and six weeks after treatment completion. RESULTS Sixty-one male and female participants aged 18-45 years were randomized (mean age 34 years, mean BMI 37 kg/m2). The study sample is diverse with 38% identifying as non-White and 20% Hispanic. CONCLUSION Investigating intranasal oxytocin's efficacy, safety, and mechanisms as an anti-obesity medication will advance the search for optimal treatment strategies for obesity and its associated severe sequelae.
Collapse
Affiliation(s)
- Marie-Louis Wronski
- Neuroendocrine Unit, Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA; Translational Developmental Neuroscience Section, Division of Psychological and Social Medicine and Developmental Neurosciences, Faculty of Medicine, TU Dresden, Dresden, Germany
| | - Franziska Plessow
- Neuroendocrine Unit, Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Liya Kerem
- Division of Pediatric Endocrinology, Department of Pediatrics, Hadassah-Hebrew University Medical Center, Jerusalem, Israel
| | - Elisa Asanza
- Neuroendocrine Unit, Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Michelle L O'Donoghue
- TIMI Study Group, Division of Cardiovascular Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Fatima C Stanford
- Neuroendocrine Unit, Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA; Division of Pediatric Endocrinology, Department of Pediatrics, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Miriam A Bredella
- Division of Musculoskeletal Imaging and Intervention, Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Martin Torriani
- Division of Musculoskeletal Imaging and Intervention, Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Alexander A Soukas
- Center for Genomic Medicine, Diabetes Unit, Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Arvin Kheterpal
- Division of Musculoskeletal Imaging and Intervention, Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Kamryn T Eddy
- Eating Disorders Clinical and Research Program, Department of Psychiatry, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Tara M Holmes
- Translational and Clinical Research Centers, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Thilo Deckersbach
- Diploma Hochschule/University of Applied Sciences, Bad Sooden-Allendorf, Germany
| | - Mark Vangel
- Biostatistics Center, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Laura M Holsen
- Division of Women's Health, Department of Medicine and Department of Psychiatry, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Elizabeth A Lawson
- Neuroendocrine Unit, Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
14
|
Olszewski PK, Noble EE, Paiva L, Ueta Y, Blevins JE. Oxytocin as a potential pharmacological tool to combat obesity. J Neuroendocrinol 2022; 34:e13106. [PMID: 35192207 PMCID: PMC9372234 DOI: 10.1111/jne.13106] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/04/2022] [Revised: 02/06/2022] [Accepted: 02/08/2022] [Indexed: 11/30/2022]
Abstract
The neuropeptide oxytocin (OT) has emerged as an important anorexigen in the regulation of food intake and energy balance. It has been shown that the release of OT and activation of hypothalamic OT neurons coincide with food ingestion. Its effects on feeding have largely been attributed to limiting meal size through interactions in key regulatory brain regions governing the homeostatic control of food intake such as the hypothalamus and hindbrain in addition to key feeding reward areas such as the nucleus accumbens and ventral tegmental area. Furthermore, the magnitude of an anorexigenic response to OT and feeding-related activation of the brain OT circuit are modified by the composition and flavor of a diet, as well as by a social context in which a meal is consumed. OT is particularly effective in reducing consumption of carbohydrates and sweet tastants. Pharmacologic, genetic, and pair-feeding studies indicate that OT-elicited weight loss cannot be fully explained by reductions of food intake and that the overall impact of OT on energy balance is also partly a result of OT-elicited changes in lipolysis, energy expenditure, and glucose regulation. Peripheral administration of OT mimics many of its effects when it is given into the central nervous system, raising the questions of whether and to what extent circulating OT acts through peripheral OT receptors to regulate energy balance. Although OT has been found to elicit weight loss in female mice, recent studies have indicated that sex and estrous cycle may impact oxytocinergic modulation of food intake. Despite the overall promising basic research data, attempts to use OT in the clinical setting to combat obesity and overeating have generated somewhat mixed results. The focus of this mini-review is to briefly summarize the role of OT in feeding and metabolism, address gaps and inconsistencies in our knowledge, and discuss some of the limitations to the potential use of chronic OT that should help guide future research on OT as a tailor-made anti-obesity therapeutic.
Collapse
Affiliation(s)
- Pawel K Olszewski
- Faculty of Science and Engineering, University of Waikato, Waikato, New Zealand
- Department of Food Science and Nutrition, College of Food, Agricultural and Natural Resource Sciences, University of Minnesota, St Paul, Minnesota, USA
- Department of Integrative Biology and Physiology, Medical School, University of Minnesota, Minneapolis, Minnesota, USA
| | - Emily E Noble
- Department of Nutritional Sciences, University of Georgia, Athens, Georgia, USA
| | - Luis Paiva
- Instituto de Ciencia Animal, Facultad de Ciencias Veterinarias, Universidad Austral de Chile, Valdivia, Chile
| | - Yoichi Ueta
- Department of Physiology, School of Medicine, University of Occupational and Environmental Health, Kitakyushu, Fukuoka, Japan
| | - James E Blevins
- Department of Veterans Affairs Medical Center, VA Puget Sound Health Care System, Office of Research and Development Medical Research Service, Seattle, Washington, USA
- Division of Metabolism, Endocrinology and Nutrition, Department of Medicine, University of Washington School of Medicine, Seattle, Washington, USA
| |
Collapse
|
15
|
Goh KK, Chen CYA, Wu TH, Chen CH, Lu ML. Crosstalk between Schizophrenia and Metabolic Syndrome: The Role of Oxytocinergic Dysfunction. Int J Mol Sci 2022; 23:ijms23137092. [PMID: 35806096 PMCID: PMC9266532 DOI: 10.3390/ijms23137092] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Revised: 06/23/2022] [Accepted: 06/23/2022] [Indexed: 02/01/2023] Open
Abstract
The high prevalence of metabolic syndrome in persons with schizophrenia has spurred investigational efforts to study the mechanism beneath its pathophysiology. Early psychosis dysfunction is present across multiple organ systems. On this account, schizophrenia may be a multisystem disorder in which one organ system is predominantly affected and where other organ systems are also concurrently involved. Growing evidence of the overlapping neurobiological profiles of metabolic risk factors and psychiatric symptoms, such as an association with cognitive dysfunction, altered autonomic nervous system regulation, desynchrony in the resting-state default mode network, and shared genetic liability, suggest that metabolic syndrome and schizophrenia are connected via common pathways that are central to schizophrenia pathogenesis, which may be underpinned by oxytocin system dysfunction. Oxytocin, a hormone that involves in the mechanisms of food intake and metabolic homeostasis, may partly explain this piece of the puzzle in the mechanism underlying this association. Given its prosocial and anorexigenic properties, oxytocin has been administered intranasally to investigate its therapeutic potential in schizophrenia and obesity. Although the pathophysiology and mechanisms of oxytocinergic dysfunction in metabolic syndrome and schizophrenia are both complex and it is still too early to draw a conclusion upon, oxytocinergic dysfunction may yield a new mechanistic insight into schizophrenia pathogenesis and treatment.
Collapse
Affiliation(s)
- Kah Kheng Goh
- Department of Psychiatry, Wan-Fang Hospital, Taipei Medical University, Taipei 116, Taiwan; (K.K.G.); (C.Y.-A.C.); (C.-H.C.)
- Psychiatric Research Center, Wan-Fang Hospital, Taipei Medical University, Taipei 116, Taiwan;
- Department of Psychiatry, School of Medicine, College of Medicine, Taipei Medical University, Taipei 110, Taiwan
| | - Cynthia Yi-An Chen
- Department of Psychiatry, Wan-Fang Hospital, Taipei Medical University, Taipei 116, Taiwan; (K.K.G.); (C.Y.-A.C.); (C.-H.C.)
- Psychiatric Research Center, Wan-Fang Hospital, Taipei Medical University, Taipei 116, Taiwan;
| | - Tzu-Hua Wu
- Psychiatric Research Center, Wan-Fang Hospital, Taipei Medical University, Taipei 116, Taiwan;
- Department of Clinical Pharmacy, School of Pharmacy, College of Pharmacy, Taipei Medical University, Taipei 110, Taiwan
| | - Chun-Hsin Chen
- Department of Psychiatry, Wan-Fang Hospital, Taipei Medical University, Taipei 116, Taiwan; (K.K.G.); (C.Y.-A.C.); (C.-H.C.)
- Psychiatric Research Center, Wan-Fang Hospital, Taipei Medical University, Taipei 116, Taiwan;
- Department of Psychiatry, School of Medicine, College of Medicine, Taipei Medical University, Taipei 110, Taiwan
| | - Mong-Liang Lu
- Department of Psychiatry, Wan-Fang Hospital, Taipei Medical University, Taipei 116, Taiwan; (K.K.G.); (C.Y.-A.C.); (C.-H.C.)
- Psychiatric Research Center, Wan-Fang Hospital, Taipei Medical University, Taipei 116, Taiwan;
- Department of Psychiatry, School of Medicine, College of Medicine, Taipei Medical University, Taipei 110, Taiwan
- Correspondence:
| |
Collapse
|
16
|
Ghobadi-Azbari P, Mahdavifar Khayati R, Sangchooli A, Ekhtiari H. Task-Dependent Effective Connectivity of the Reward Network During Food Cue-Reactivity: A Dynamic Causal Modeling Investigation. Front Behav Neurosci 2022; 16:899605. [PMID: 35813594 PMCID: PMC9263922 DOI: 10.3389/fnbeh.2022.899605] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2022] [Accepted: 06/08/2022] [Indexed: 11/13/2022] Open
Abstract
Neural reactivity to food cues may play a central role in overeating and excess weight gain. Functional magnetic resonance imaging (fMRI) studies have implicated regions of the reward network in dysfunctional food cue-reactivity, but neural interactions underlying observed patterns of signal change remain poorly understood. Fifty overweight and obese participants with self-reported cue-induced food craving viewed food and neutral cues during fMRI scanning. Regions of the reward network with significantly greater food versus neutral cue-reactivity were used to specify plausible models of task-related neural interactions underlying the observed blood oxygenation level-dependent (BOLD) signal, and a bi-hemispheric winning model was identified in a dynamic causal modeling (DCM) framework. Neuro-behavioral correlations are investigated with group factor analysis (GFA) and Pearson's correlation tests. The ventral tegmental area (VTA), amygdalae, and orbitofrontal cortices (OFC) showed significant food cue-reactivity. DCM suggests these activations are produced by largely reciprocal dynamic signaling between these regions, with food cues causing regional disinhibition and an apparent shifting of activity to the right amygdala. Intrinsic self-inhibition in the VTA and right amygdala is negatively correlated with measures of food craving and hunger and right-amygdalar disinhibition by food cues is associated with the intensity of cue-induced food craving, but no robust cross-unit latent factors were identified between the neural group and behavioral or demographic variable groups. Our results suggest a rich array of dynamic signals drive reward network cue-reactivity, with the amygdalae mediating much of the dynamic signaling between the VTA and OFCs. Neuro-behavioral correlations suggest particularly crucial roles for the VTA, right amygdala, and the right OFC-amygdala connection but the more robust GFA identified no cross-unit factors, so these correlations should be interpreted with caution. This investigation provides novel insights into dynamic circuit mechanisms with etiologic relevance to obesity, suggesting pathways in biomarker development and intervention.
Collapse
Affiliation(s)
| | | | - Arshiya Sangchooli
- Iranian National Center for Addiction Studies, Tehran University of Medical Sciences, Tehran, Iran
| | - Hamed Ekhtiari
- Department of Psychiatry, University of Minnesota, Minnesota, MN, United States
| |
Collapse
|
17
|
Dunigan AI, Roseberry AG. Actions of feeding-related peptides on the mesolimbic dopamine system in regulation of natural and drug rewards. ADDICTION NEUROSCIENCE 2022; 2:100011. [PMID: 37220637 PMCID: PMC10201992 DOI: 10.1016/j.addicn.2022.100011] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 05/25/2023]
Abstract
The mesolimbic dopamine system is the primary neural circuit mediating motivation, reinforcement, and reward-related behavior. The activity of this system and multiple behaviors controlled by it are affected by changes in feeding and body weight, such as fasting, food restriction, or the development of obesity. Multiple different peptides and hormones that have been implicated in the control of feeding and body weight interact with the mesolimbic dopamine system to regulate many different dopamine-dependent, reward-related behaviors. In this review, we summarize the effects of a selected set of feeding-related peptides and hormones acting within the ventral tegmental area and nucleus accumbens to alter feeding, as well as food, drug, and social reward.
Collapse
Affiliation(s)
- Anna I. Dunigan
- Department of Biology and Neuroscience Institute, Georgia State University, Atlanta, GA 30303, USA
| | - Aaron G. Roseberry
- Department of Biology and Neuroscience Institute, Georgia State University, Atlanta, GA 30303, USA
- Neuroscience Institute, Georgia State University, Atlanta, GA 30303, USA
| |
Collapse
|
18
|
Elfers CT, Blevins JE, Lawson EA, Pittner R, Silva D, Kiselyov A, Roth CL. Robust Reductions of Body Weight and Food Intake by an Oxytocin Analog in Rats. Front Physiol 2021; 12:726411. [PMID: 34646154 PMCID: PMC8502973 DOI: 10.3389/fphys.2021.726411] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2021] [Accepted: 09/02/2021] [Indexed: 01/22/2023] Open
Abstract
Background: Oxytocin is a hypothalamic neuropeptide that participates in the network of appetite regulation. Recently the oxytocin signaling pathway has emerged as an attractive target for treating obesity. However, the short half-life limits its development as a clinical therapeutic. Here we provide results from testing a long-lasting, potent and selective oxytocin analog ASK1476 on its efficacy to reduce food intake and body weight in comparison to the native oxytocin peptide. Methods: ASK1476 features two specific amino acid substitutions in positions 7 and 8 combined with a short polyethylene glycol spacer. Short time dose escalation experiments testing increasing doses of 3 days each were performed in diet-induced overweight (DIO) male rats assessing effects on body weight as well as changes in food intake. Furthermore, DIO rats were tested for changes in body weight, food intake, temperature, and locomotor activity over 28 days of treatment (oxytocin, ASK1476, or vehicle). Results: In dose escalation experiments, significant reductions in food intake relative to baseline were detected beginning with doses of 15 nmol/kg ASK1476 (-15.2 ± 2.3 kcal/d, p = 0.0017) and 20 nmol/kg oxytocin (-11.2.9 ± 2.4 kcal/d, p = 0.0106) with corresponding significant changes in body weight (ASK1476: -5.2 ± 0.8 g, p = 0.0016; oxytocin: -2.6 ± 0.7 g, p = 0.0326). In long-term experiments, there was no difference on body weight change between 120 nmol/kg/d ASK1476 (-71.4 ± 34.2 g, p = 0.039) and 600 nmol/kg/d oxytocin (-91.8 ± 32.2 g, p = 0.035) relative to vehicle (706.9 ± 28.3 g), indicating a stronger dose response for ASK1476. Likewise, both ASK1476 and oxytocin at these doses resulted in similar reductions in 28-day cumulative food intake (ASK1476: -562.7 ± 115.0 kcal, p = 0.0001; oxytocin: -557.1 ± 101.3 kcal, p = 0.0001) relative to vehicle treatment (2716 ± 75.4 kcal), while no effects were detected on locomotor activity or body temperature. Conclusion: This study provides proof-of-concept data demonstrating an oxytocin analog with extended in vivo stability and improved potency to reduce food intake and body weight in DIO animals which could mark a new avenue in anti-obesity drug interventions.
Collapse
Affiliation(s)
- Clinton T. Elfers
- Center for Integrative Brain Research, Seattle Children’s Research Institute, Seattle, WA, United States
| | - James E. Blevins
- VA Puget Sound Health Care System, Office of Research and Development Medical Research Service, Department of Veterans Affairs Medical Center, Seattle, WA, United States
- Division of Metabolism, Endocrinology and Nutrition, Department of Medicine, University of Washington School of Medicine, Seattle, WA, United States
| | - Elizabeth A. Lawson
- Neuroendocrine Unit, Massachusetts General Hospital and Department of Medicine, Harvard Medical School, Boston, MA, United States
| | | | - David Silva
- OXT Therapeutics, Saint Louis, MO, United States
| | | | - Christian L. Roth
- Center for Integrative Brain Research, Seattle Children’s Research Institute, Seattle, WA, United States
- Division of Endocrinology, Department of Pediatrics, University of Washington, Seattle, WA, United States
| |
Collapse
|
19
|
Mitra S, Basu S, Singh O, Lechan RM, Singru PS. Cocaine- and amphetamine-regulated transcript peptide- and dopamine-containing systems interact in the ventral tegmental area of the zebra finch, Taeniopygia guttata, during dynamic changes in energy status. Brain Struct Funct 2021; 226:2537-2559. [PMID: 34392422 DOI: 10.1007/s00429-021-02348-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2020] [Accepted: 07/21/2021] [Indexed: 12/21/2022]
Abstract
The mesolimbic dopamine (DA)-pathway regulates food-reward, feeding-related behaviour and energy balance. Evidence underscores the importance of feeding-related neuropeptides in modulating activity of these DA neurons. The neuropeptide, CART, a crucial regulator of energy balance, modulates DA-release, and influences the activity of ventral tegmental area (VTA) DAergic neurons in the mammalian brain. Whether CART- and DA-containing systems interact at the level of VTA to regulate energy balance, however, is poorly understood. We explored the interaction between CART- and DA-containing systems in midbrain of the zebra finch, Taeniopygia guttata, an interesting model to study dynamic changes in energy balance due to higher BMR/daytime body temperature, and rapid responsiveness of the feeding-related neuropeptides to changes in energy state. Further, its midbrain DA-neurons share similarities with those in mammals. In the midbrain, tyrosine hydroxylase-immunoreactive (TH-i) neurons were seen in the substantia nigra (SN) and VTA [anterior (VTAa), mid (VTAm) and caudal (VTAc)]; those in VTA were smaller. In the VTA, CART-immunoreactive (CART-i)-fibers densely innervated TH-i neurons, and both CART-immunoreactivity (CART-ir) and TH-immunoreactivity (TH-ir) responded to energy status-dependent changes. Compared to fed and fasted birds, refeeding dramatically enhanced TH-ir and the percentage of TH-i neurons co-expressing FOS in the VTA. Increased prepro-CART-mRNA, CART-ir and a transient appearance of CART-i neurons was observed in VTAa of fasted, but not fed birds. To test the functional interaction between CART- and DA-containing systems, ex-vivo superfused midbrain-slices were treated with CART-peptide and changes in TH-ir analysed. Compared to control tissues, CART-treatment increased TH-ir in VTA but not SN. We propose that CART is a potential regulator of VTA DA-neurons and energy balance in T. guttata.
Collapse
Affiliation(s)
- Saptarsi Mitra
- School of Biological Sciences, National Institute of Science Education and Research (NISER)-Bhubaneswar, P.O. Jatni, Khurda, Odisha, 752050, India.,Homi Bhabha National Institute, Mumbai, 400094, India
| | - Sumela Basu
- School of Biological Sciences, National Institute of Science Education and Research (NISER)-Bhubaneswar, P.O. Jatni, Khurda, Odisha, 752050, India.,Homi Bhabha National Institute, Mumbai, 400094, India
| | - Omprakash Singh
- School of Biological Sciences, National Institute of Science Education and Research (NISER)-Bhubaneswar, P.O. Jatni, Khurda, Odisha, 752050, India.,Homi Bhabha National Institute, Mumbai, 400094, India
| | - Ronald M Lechan
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, Tupper Research Institute, Tufts Medical Center, Boston, MA, USA.,Department of Neuroscience, Tufts University School of Medicine, Boston, USA
| | - Praful S Singru
- School of Biological Sciences, National Institute of Science Education and Research (NISER)-Bhubaneswar, P.O. Jatni, Khurda, Odisha, 752050, India. .,Homi Bhabha National Institute, Mumbai, 400094, India.
| |
Collapse
|
20
|
Kerem L, Lawson EA. The Effects of Oxytocin on Appetite Regulation, Food Intake and Metabolism in Humans. Int J Mol Sci 2021; 22:7737. [PMID: 34299356 PMCID: PMC8306733 DOI: 10.3390/ijms22147737] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2021] [Revised: 07/13/2021] [Accepted: 07/16/2021] [Indexed: 12/18/2022] Open
Abstract
The hypothalamic peptide oxytocin and its receptor are involved in a range of physiological processes, including parturition, lactation, cell growth, wound healing, and social behavior. More recently, increasing evidence has established the effects of oxytocin on food intake, energy expenditure, and peripheral metabolism. In this review, we provide a comprehensive description of the central oxytocinergic system in which oxytocin acts to shape eating behavior and metabolism. Next, we discuss the peripheral beneficial effects oxytocin exerts on key metabolic organs, including suppression of visceral adipose tissue inflammation, skeletal muscle regeneration, and bone tissue mineralization. A brief summary of oxytocin actions learned from animal models is presented, showing that weight loss induced by chronic oxytocin treatment is related not only to its anorexigenic effects, but also to the resulting increase in energy expenditure and lipolysis. Following an in-depth discussion on the technical challenges related to endogenous oxytocin measurements in humans, we synthesize data related to the association between endogenous oxytocin levels, weight status, metabolic syndrome, and bone health. We then review clinical trials showing that in humans, acute oxytocin administration reduces food intake, attenuates fMRI activation of food motivation brain areas, and increases activation of self-control brain regions. Further strengthening the role of oxytocin in appetite regulation, we review conditions of hypothalamic insult and certain genetic pathologies associated with oxytocin depletion that present with hyperphagia, extreme weight gain, and poor metabolic profile. Intranasal oxytocin is currently being evaluated in human clinical trials to learn whether oxytocin-based therapeutics can be used to treat obesity and its associated sequela. At the end of this review, we address the fundamental challenges that remain in translating this line of research to clinical care.
Collapse
Affiliation(s)
- Liya Kerem
- Neuroendocrine Unit, Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA;
- Division of Pediatric Endocrinology, Massachusetts General Hospital for Children, Boston, MA 02114, USA
| | - Elizabeth A. Lawson
- Neuroendocrine Unit, Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA;
| |
Collapse
|
21
|
Bach P, Grosshans M, Koopmann A, Kienle P, Vassilev G, Otto M, Bumb JM, Kiefer F. Reliability of neural food cue-reactivity in participants with obesity undergoing bariatric surgery: a 26-week longitudinal fMRI study. Eur Arch Psychiatry Clin Neurosci 2021; 271:951-962. [PMID: 33331960 PMCID: PMC8236041 DOI: 10.1007/s00406-020-01218-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/12/2020] [Accepted: 11/24/2020] [Indexed: 12/11/2022]
Abstract
Obesity is highly prevalent worldwide and results in a high disease burden. The efforts to monitor and predict treatment outcome in participants with obesity using functional magnetic resonance imaging (fMRI) depends on the reliability of the investigated task-fMRI brain activation. To date, no study has investigated whole-brain reliability of neural food cue-reactivity. To close this gap, we analyzed the longitudinal reliability of an established food cue-reactivity task. Longitudinal reliability of neural food-cue-induced brain activation and subjective food craving ratings over three fMRI sessions (T0: 2 weeks before surgery, T1: 8 weeks and T2: 24 weeks after surgery) were investigated in N = 11 participants with obesity. We computed an array of established reliability estimates, including the intraclass correlation (ICC), the Dice and Jaccard coefficients and similarity of brain activation maps. The data indicated good reliability (ICC > 0.6) of subjective food craving ratings over 26 weeks and excellent reliability (ICC > 0.75) of brain activation signals for the contrast of interest (food > neutral) in the caudate, putamen, thalamus, middle cingulum, inferior, middle and superior occipital gyri, and middle and superior temporal gyri and cunei. Using similarity estimates, it was possible to re-identify individuals based on their neural activation maps (73%) with a fading degree of accuracy, when comparing fMRI sessions further apart. The results show excellent reliability of task-fMRI neural brain activation in several brain regions. Current data suggest that fMRI-based measures might indeed be suitable to monitor and predict treatment outcome in participants with obesity undergoing bariatric surgery.
Collapse
Affiliation(s)
- Patrick Bach
- Department of Addictive Behavior and Addiction Medicine, Medical Faculty Mannheim, Central Institute of Mental Health, Heidelberg University, J5/68159, Mannheim, Germany. .,Feuerlein Center on Translational Addiction Medicine (FCTS), University of Heidelberg, Heidelberg, Germany.
| | - Martin Grosshans
- grid.413757.30000 0004 0477 2235Department of Addictive Behavior and Addiction Medicine, Medical Faculty Mannheim, Central Institute of Mental Health, Heidelberg University, J5/68159 Mannheim, Germany
| | - Anne Koopmann
- grid.413757.30000 0004 0477 2235Department of Addictive Behavior and Addiction Medicine, Medical Faculty Mannheim, Central Institute of Mental Health, Heidelberg University, J5/68159 Mannheim, Germany ,grid.7700.00000 0001 2190 4373Feuerlein Center on Translational Addiction Medicine (FCTS), University of Heidelberg, Heidelberg, Germany
| | - Peter Kienle
- Department of Surgery, Theresienkrankenhaus, Mannheim, Germany
| | - Georgi Vassilev
- grid.411778.c0000 0001 2162 1728Department of Surgery, University Medical Center Mannheim, Heidelberg University, Mannheim, Germany
| | - Mirko Otto
- grid.411778.c0000 0001 2162 1728Department of Surgery, University Medical Center Mannheim, Heidelberg University, Mannheim, Germany
| | - J. Malte Bumb
- grid.413757.30000 0004 0477 2235Department of Addictive Behavior and Addiction Medicine, Medical Faculty Mannheim, Central Institute of Mental Health, Heidelberg University, J5/68159 Mannheim, Germany ,grid.7700.00000 0001 2190 4373Feuerlein Center on Translational Addiction Medicine (FCTS), University of Heidelberg, Heidelberg, Germany
| | - Falk Kiefer
- grid.413757.30000 0004 0477 2235Department of Addictive Behavior and Addiction Medicine, Medical Faculty Mannheim, Central Institute of Mental Health, Heidelberg University, J5/68159 Mannheim, Germany ,grid.7700.00000 0001 2190 4373Feuerlein Center on Translational Addiction Medicine (FCTS), University of Heidelberg, Heidelberg, Germany
| |
Collapse
|
22
|
Investigating resting brain perfusion abnormalities and disease target-engagement by intranasal oxytocin in women with bulimia nervosa and binge-eating disorder and healthy controls. Transl Psychiatry 2020; 10:180. [PMID: 32513936 PMCID: PMC7280271 DOI: 10.1038/s41398-020-00871-w] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/24/2020] [Revised: 05/19/2020] [Accepted: 05/26/2020] [Indexed: 12/25/2022] Open
Abstract
Advances in the treatment of bulimia nervosa and binge-eating disorder (BN/BED) have been marred by our limited understanding of the underpinning neurobiology. Here we measured regional cerebral blood flow (rCBF) to map resting perfusion abnormalities in women with BN/BED compared with healthy controls and investigate whether intranasal oxytocin (OT), proposed as a potential treatment, can restore perfusion in disorder-related brain circuits. Twenty-four women with BN/BED and 23 healthy women participated in a randomized, double-blind, crossover, placebo-controlled study. We used arterial spin labelling MRI to measure rCBF and the effects of an acute dose of intranasal OT (40 IU) or placebo over 18-26 min post dosing, as we have previously shown robust OT-induced changes in resting rCBF in men in a similar time-window (15-36 min post dosing). We tested for effects of treatment, diagnosis and their interaction on extracted rCBF values in anatomical regions-of-interest previously implicated in BN/BED by other neuroimaging modalities, and conducted exploratory whole-brain analyses to investigate previously unidentified brain regions. We demonstrated that women with BN/BED presented increased resting rCBF in the medial prefrontal and orbitofrontal cortices, anterior cingulate gyrus, posterior insula and middle/inferior temporal gyri bilaterally. Hyperperfusion in these areas specifically correlated with eating symptoms severity in patients. Our data did not support a normalizing effect of intranasal OT on perfusion abnormalities in these patients, at least for the specific dose (40 IU) and post-dosing interval (18-26 min) examined. Our findings enhance our understanding of resting brain abnormalities in BN/BED and identify resting rCBF as a non-invasive potential biomarker for disease-related changes and treatment monitoring. They also highlight the need for a comprehensive investigation of intranasal OT pharmacodynamics in women before we can fully ascertain its therapeutic value in disorders affecting predominantly this gender, such as BN/BED.
Collapse
|