1
|
Josselsohn A, Zhao Y, Espinoza D, Hollander E. Oxytocin in neurodevelopmental disorders: Autism spectrum disorder and Prader-Willi syndrome. Pharmacol Ther 2024:108734. [PMID: 39455012 DOI: 10.1016/j.pharmthera.2024.108734] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2023] [Revised: 10/02/2024] [Accepted: 10/22/2024] [Indexed: 10/28/2024]
Abstract
This manuscript reviews recent work on oxytocin and its use in neurodevelopmental disorders including spectrum disorder (ASD) and Prader-Willi syndrome (PWS). Oxytocin is involved in social recognition, bonding, maternal behaviors, anxiety, food motivation, and hyperphagia. While the pathophysiology of ASD and PWS involve abnormalities in the oxytocin system, clinical trials have shown discrepant results in the effectiveness of oxytocin as a treatment for core symptoms associated with these disorders. In this review, we outline oxytocin's clinical pharmacology, safety considerations, and results in recent clinical trials. We propose that oxytocin may be most beneficial in these populations if dosed in a dynamic regimen (PRN) and paired with social interventions.
Collapse
Affiliation(s)
| | - Yin Zhao
- Albert Einstein College of Medicine, Montefiore Medical Center
| | | | - Eric Hollander
- Albert Einstein College of Medicine, Montefiore Medical Center.
| |
Collapse
|
2
|
Petersson M, Höybye C. Is Oxytocin a Contributor to Behavioral and Metabolic Features in Prader-Willi Syndrome? Curr Issues Mol Biol 2024; 46:8767-8779. [PMID: 39194735 DOI: 10.3390/cimb46080518] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2024] [Revised: 08/05/2024] [Accepted: 08/10/2024] [Indexed: 08/29/2024] Open
Abstract
Prader-Willi Syndrome (PWS) is a rare genetic disorder typically characterized by decreased social interaction, hyperphagia, poor behavioral control and temper tantrums, together with a high risk of morbid obesity unless food intake is controlled. The genetic defects that cause PWS include paternal 15q deletion (estimated in 60% of cases), chromosome 15 maternal uniparental disomy (UPD) (estimated in 35% of cases) and imprinting defects and translocations. Several studies indicate an oxytocin deficiency in PWS. Oxytocin is a hypothalamic nonapeptide with receptors located in the brain and in various other tissues in the body. It acts as a neuropeptide in several brain areas of great importance for behavioral and metabolic effects, as well as a neurohypophyseal hormone released into the circulation. Oxytocin in both rats and humans has strong and long-lasting behavioral and metabolic effects. Thus, an oxytocin deficiency might be involved in several of the behavioral and metabolic symptoms characterizing PWS. Treatment with oxytocin has, in some studies, shown improvement in psycho-social behavior and hyperphagia in individuals with PWS. This review focus on the behavioral and metabolic effects of oxytocin, the symptoms of a potential oxytocin deficiency in PWS and the effects of oxytocin treatment.
Collapse
Affiliation(s)
- Maria Petersson
- Department of Endocrinology, Karolinska University Hospital, 171 76 Stockholm, Sweden
- Department of Molecular Medicine and Surgery, Karolinska Institutet, 171 76 Stockholm, Sweden
| | - Charlotte Höybye
- Department of Endocrinology, Karolinska University Hospital, 171 76 Stockholm, Sweden
- Department of Molecular Medicine and Surgery, Karolinska Institutet, 171 76 Stockholm, Sweden
| |
Collapse
|
3
|
Chasseloup F, Tabarin A, Chanson P. Diabetes insipidus: Vasopressin deficiency…. ANNALES D'ENDOCRINOLOGIE 2024; 85:294-299. [PMID: 38316255 DOI: 10.1016/j.ando.2023.11.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/08/2023] [Accepted: 11/20/2023] [Indexed: 02/07/2024]
Abstract
Diabetes insipidus is a disorder characterized by hypo-osmotic polyuria secondary to abnormal synthesis, regulation, or renal action of antidiuretic hormone. Recently, an expert group, with the support of patient associations, proposed that diabetes insipidus be renamed to avoid confusion with diabetes mellitus. The most common form of diabetes insipidus is secondary to a dysfunction of the neurohypophysis (central diabetes insipidus) and would be therefore named 'vasopressin deficiency'. The rarer form, which is linked to renal vasopressin resistance (nephrogenic diabetes insipidus), would then be named 'vasopressin resistance'. The etiology of diabetes insipidus is sometimes clear, in the case of a neurohypophyseal cause (tumoral or infiltrative damage) or a renal origin, but in some cases diabetes insipidus can be difficult to distinguish from primary polydipsia, which is characterized by consumption of excessive quantities of water without any abnormality in regulation or action of antidiuretic hormone. Apart from patients' medical history, physical examination, and imaging of the hypothalamic-pituitary region, functional tests such as water deprivation or stimulation of copeptin by hyperosmolarity (induced by infusion of hypertonic saline) can be proposed in order to distinguish between these different etiologies. The treatment of diabetes insipidus depends on the underlying etiology, and in the case of a central etiology, is based on the administration of desmopressin which improves patient symptoms but does not always result in an optimal quality of life. The cause of this altered quality of life may be oxytocin deficiency, oxytocin being also secreted from the neurohypophysis, though this has not been fully established. The possibility of a new test using stimulation of oxytocin to identify alterations in oxytocin synthesis is of interest and would allow confirmation of a deficiency in those patients presenting with diabetes insipidus linked to neurohypophyseal dysfunction.
Collapse
Affiliation(s)
- Fanny Chasseloup
- Service d'endocrinologie et des maladies de la reproduction, centre de référence des maladies rares de l'hypophyse, université Paris-Saclay, Inserm, physiologie et physiopathologie endocriniennes, AP-HP, hôpital Bicêtre, Le Kremlin-Bicêtre, France.
| | - Antoine Tabarin
- Service d'endocrinologie, diabète et nutrition, hôpital Haut Lévêque, centre hospitalier universitaire de Bordeaux, Pessac, France
| | - Philippe Chanson
- Service d'endocrinologie et des maladies de la reproduction, centre de référence des maladies rares de l'hypophyse, université Paris-Saclay, Inserm, physiologie et physiopathologie endocriniennes, AP-HP, hôpital Bicêtre, Le Kremlin-Bicêtre, France
| |
Collapse
|
4
|
Gan HW, Cerbone M, Dattani MT. Appetite- and Weight-Regulating Neuroendocrine Circuitry in Hypothalamic Obesity. Endocr Rev 2024; 45:309-342. [PMID: 38019584 PMCID: PMC11074800 DOI: 10.1210/endrev/bnad033] [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: 02/04/2023] [Revised: 10/25/2023] [Accepted: 11/27/2023] [Indexed: 11/30/2023]
Abstract
Since hypothalamic obesity (HyOb) was first described over 120 years ago by Joseph Babinski and Alfred Fröhlich, advances in molecular genetic laboratory techniques have allowed us to elucidate various components of the intricate neurocircuitry governing appetite and weight regulation connecting the hypothalamus, pituitary gland, brainstem, adipose tissue, pancreas, and gastrointestinal tract. On a background of an increasing prevalence of population-level common obesity, the number of survivors of congenital (eg, septo-optic dysplasia, Prader-Willi syndrome) and acquired (eg, central nervous system tumors) hypothalamic disorders is increasing, thanks to earlier diagnosis and management as well as better oncological therapies. Although to date the discovery of several appetite-regulating peptides has led to the development of a range of targeted molecular therapies for monogenic obesity syndromes, outside of these disorders these discoveries have not translated into the development of efficacious treatments for other forms of HyOb. This review aims to summarize our current understanding of the neuroendocrine physiology of appetite and weight regulation, and explore our current understanding of the pathophysiology of HyOb.
Collapse
Affiliation(s)
- Hoong-Wei Gan
- Department of Endocrinology, Great Ormond Street Hospital for Children NHS Foundation Trust, Great Ormond Street, London WC1N 3JH, UK
- Genetics & Genomic Medicine Research & Teaching Department, University College London Great Ormond Street Institute for Child Health, 30 Guilford Street, London WC1N 1EH, UK
| | - Manuela Cerbone
- Department of Endocrinology, Great Ormond Street Hospital for Children NHS Foundation Trust, Great Ormond Street, London WC1N 3JH, UK
- Genetics & Genomic Medicine Research & Teaching Department, University College London Great Ormond Street Institute for Child Health, 30 Guilford Street, London WC1N 1EH, UK
| | - Mehul Tulsidas Dattani
- Department of Endocrinology, Great Ormond Street Hospital for Children NHS Foundation Trust, Great Ormond Street, London WC1N 3JH, UK
- Genetics & Genomic Medicine Research & Teaching Department, University College London Great Ormond Street Institute for Child Health, 30 Guilford Street, London WC1N 1EH, UK
| |
Collapse
|
5
|
Bottemanne H, English I, Bottemanne L, Torres P, Beauquier B, Joly L. From love to pain: is oxytocin the key to grief complications? L'ENCEPHALE 2024; 50:85-90. [PMID: 37993287 DOI: 10.1016/j.encep.2023.08.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/29/2021] [Revised: 07/20/2023] [Accepted: 08/05/2023] [Indexed: 11/24/2023]
Abstract
While most adults confronted with the death of a loved one manage to grieve, about 10-20% of individuals develop complicated grief, characterized by persistent distress and impaired social skills, or pathological grief, defined by the onset or decompensation of a psychiatric disorder. Little is known about the biological causes of these grief complications. Recent work suggests that oxytocin, a major neuroendocrine hormone regulating many neurocognitive mechanisms, may be involved in this process. Oxytocin is widely studied and well known for its impact on the mother-child bond and hormonal and brain systems related to attachment and social interactions. In this article, we propose a neurocognitive model of grief complications based on existing data on the role of oxytocin in interpersonal attachment and its impact on brain activity. We suggest that complicated grief is associated with dysfunctional cerebral oxytocinergic signaling and persistent hyperactivation of the nucleus accumbens. This mechanism is involved in limiting the reduction of interpersonal attachment to the deceased during acute phases and in searching for new interpersonal relationships during the recovery phase. We show how the exploration of cerebral oxytocinergic signaling would improve the understanding of physiological grief mechanisms in the general population and could allow the development of new therapeutic perspectives against the complications of grief.
Collapse
Affiliation(s)
- Hugo Bottemanne
- Paris Brain Institute - Institut du Cerveau (ICM), UMR 7225/UMRS 1127, CNRS, Inserm, Paris, France; Department of Psychiatry, Pitié-Salpêtrière Hospital, DMU Neuroscience, AP-HP, Sorbonne University, Paris, France; Department of Philosophy, SND Research Unit, UMR 8011, Sorbonne University, Paris, France.
| | - Isolde English
- Paris Brain Institute - Institut du Cerveau (ICM), UMR 7225/UMRS 1127, CNRS, Inserm, Paris, France
| | - Laure Bottemanne
- Paris Brain Institute - Institut du Cerveau (ICM), UMR 7225/UMRS 1127, CNRS, Inserm, Paris, France
| | - Paloma Torres
- Department of Psychiatry, Pitié-Salpêtrière Hospital, DMU Neuroscience, AP-HP, Sorbonne University, Paris, France
| | | | - Lucie Joly
- Paris Brain Institute - Institut du Cerveau (ICM), UMR 7225/UMRS 1127, CNRS, Inserm, Paris, France; Department of Psychiatry, Saint-Antoine Hospital, DMU Neuroscience, AP-HP, Sorbonne University, Paris, France
| |
Collapse
|
6
|
Han JC, Rasmussen MC, Forte AR, Schrage SB, Zafar SK, Haqq AM. Management of Monogenic and Syndromic Obesity. Gastroenterol Clin North Am 2023; 52:733-750. [PMID: 37919024 DOI: 10.1016/j.gtc.2023.08.005] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2023]
Abstract
Similar to the general population, lifestyle interventions focused on nutrition and physical activity form the foundation for treating obesity caused by rare genetic disorders. Additional therapies, including metreleptin and setmelanotide, that target defects within the leptin signaling pathway can effectively synergize with lifestyle efforts to treat monogenic disorders of leptin, leptin receptor, proopiomelanocortin (POMC), and proprotein convertase subtilisin/kexin type 1 (PCSK1) and syndromic conditions, such as the ciliopathies Bardet-Biedl and Alström syndromes, whose pathophysiological mechanisms also converge on the leptin pathway. Investigational treatments for Prader-Willi syndrome target specific defects caused by reduced expression of paternally derived genes within the chromosome 15q region.
Collapse
Affiliation(s)
- Joan C Han
- Division of Pediatric Endocrinology and Diabetes, Department of Pediatrics, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Diabetes, Obesity, and Metabolism Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA; The Mindich Child Health and Development Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
| | - Marcus C Rasmussen
- Division of Pediatric Endocrinology and Diabetes, Department of Pediatrics, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Alison R Forte
- Division of Pediatric Endocrinology and Diabetes, Department of Pediatrics, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Stephanie B Schrage
- Division of Pediatric Endocrinology and Diabetes, Department of Pediatrics, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Sarah K Zafar
- Division of Pediatric Endocrinology and Diabetes, Department of Pediatrics, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Andrea M Haqq
- Department of Pediatrics, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada; Department of Agricultural, Food and Nutritional Science, University of Alberta, Edmonton, Alberta, Canada
| |
Collapse
|
7
|
Balestrino R, Losa M, Albano L, Barzaghi LR, Mortini P. Intranasal oxytocin as a treatment for obesity: safety and efficacy. Expert Rev Endocrinol Metab 2023; 18:295-306. [PMID: 37232186 DOI: 10.1080/17446651.2023.2216794] [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: 03/14/2023] [Revised: 05/12/2023] [Accepted: 05/18/2023] [Indexed: 05/27/2023]
Abstract
INTRODUCTION Known for its effect on labor and lactation and on emotional and social functions, oxytocin has recently emerged as a key modulator of feeding behavior and indeed suggested as a potential treatment for obesity. The potential positive effect of oxytocin on both metabolic and psychological-behavioral complications of hypothalamic lesions makes it a promising tool in the management of these conditions. AREAS COVERED The aim of the present review article is to provide an overview of the mechanism of action and clinical experience of the use of oxytocin in different forms of obesity. EXPERT OPINION Current evidence suggests a potential role of oxytocin in the treatment of obesity with different causes. Several challenges remain: an improved understanding of the physiological regulation, mechanisms of action of oxytocin, and interplay with other endocrine axes is fundamental to clarify its role. Further clinical trials are needed to determine the safety and efficacy of oxytocin for the treatment of different forms of obesity. Understanding the mechanism(s) of action of oxytocin on body weight regulation might also improve our understanding of obesity and reveal possible new therapeutic targets - as well as promoting advances in other fields in which oxytocin might be used.
Collapse
Affiliation(s)
- Roberta Balestrino
- Department of Neurosurgery and Gamma Knife Radiosurgery, IRCCS San Raffaele, Milano, Italy
- Neuroimaging Research Unit, Division of Neuroscience, IRCCS San Raffaele, Milan, Italy
- Vita-Salute San Raffaele University, Milan, Italy
| | - Marco Losa
- Department of Neurosurgery and Gamma Knife Radiosurgery, IRCCS San Raffaele, Milano, Italy
| | - Luigi Albano
- Department of Neurosurgery and Gamma Knife Radiosurgery, IRCCS San Raffaele, Milano, Italy
- Neuroimaging Research Unit, Division of Neuroscience, IRCCS San Raffaele, Milan, Italy
| | - Lina R Barzaghi
- Department of Neurosurgery and Gamma Knife Radiosurgery, IRCCS San Raffaele, Milano, Italy
| | - Pietro Mortini
- Department of Neurosurgery and Gamma Knife Radiosurgery, IRCCS San Raffaele, Milano, Italy
| |
Collapse
|
8
|
Roof E, Deal CL, McCandless SE, Cowan RL, Miller JL, Hamilton JK, Roeder ER, McCormack SE, Roshan Lal TR, Abdul-Latif HD, Haqq AM, Obrynba KS, Torchen LC, Vidmar AP, Viskochil DH, Chanoine JP, Lam CKL, Pierce MJ, Williams LL, Bird LM, Butler MG, Jensen DE, Myers SE, Oatman OJ, Baskaran C, Chalmers LJ, Fu C, Alos N, McLean SD, Shah A, Whitman BY, Blumenstein BA, Leonard SF, Ernest JP, Cormier JW, Cotter SP, Ryman DC. Intranasal Carbetocin Reduces Hyperphagia, Anxiousness, and Distress in Prader-Willi Syndrome: CARE-PWS Phase 3 Trial. J Clin Endocrinol Metab 2023; 108:1696-1708. [PMID: 36633570 PMCID: PMC10271225 DOI: 10.1210/clinem/dgad015] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/30/2022] [Revised: 01/09/2023] [Accepted: 01/09/2023] [Indexed: 01/13/2023]
Abstract
CONTEXT Prader-Willi syndrome (PWS) is a rare genetic disorder characterized by endocrine and neuropsychiatric problems including hyperphagia, anxiousness, and distress. Intranasal carbetocin, an oxytocin analog, was investigated as a selective oxytocin replacement therapy. OBJECTIVE To evaluate safety and efficacy of intranasal carbetocin in PWS. DESIGN Randomized, double-blind, placebo-controlled phase 3 trial with long-term follow-up. SETTING Twenty-four ambulatory clinics at academic medical centers. PARTICIPANTS A total of 130 participants with PWS aged 7 to 18 years. INTERVENTIONS Participants were randomized to 9.6 mg/dose carbetocin, 3.2 mg/dose carbetocin, or placebo 3 times daily during an 8-week placebo-controlled period (PCP). During a subsequent 56-week long-term follow-up period, placebo participants were randomly assigned to 9.6 mg or 3.2 mg carbetocin, with carbetocin participants continuing at their previous dose. MAIN OUTCOME MEASURES Primary endpoints assessed change in hyperphagia (Hyperphagia Questionnaire for Clinical Trials [HQ-CT]) and obsessive-compulsive symptoms (Children's Yale-Brown Obsessive-Compulsive Scale [CY-BOCS]) during the PCP for 9.6 mg vs placebo, and the first secondary endpoints assessed these same outcomes for 3.2 mg vs placebo. Additional secondary endpoints included assessments of anxiousness and distress behaviors (PWS Anxiousness and Distress Behaviors Questionnaire [PADQ]) and clinical global impression of change (CGI-C). RESULTS Because of onset of the COVID-19 pandemic, enrollment was stopped prematurely. The primary endpoints showed numeric improvements in both HQ-CT and CY-BOCS which were not statistically significant; however, the 3.2-mg arm showed nominally significant improvements in HQ-CT, PADQ, and CGI-C scores vs placebo. Improvements were sustained in the long-term follow-up period. The most common adverse event during the PCP was mild to moderate flushing. CONCLUSIONS Carbetocin was well tolerated, and the 3.2-mg dose was associated with clinically meaningful improvements in hyperphagia and anxiousness and distress behaviors in participants with PWS. CLINICAL TRIALS REGISTRATION NUMBER NCT03649477.
Collapse
Affiliation(s)
| | - Cheri L Deal
- Department of Pediatrics, Centre Hospitalier Universitaire Sainte-Justine Centre de Recherche, Montréal, Québec H3T 1C5, Canada
| | - Shawn E McCandless
- Department of Pediatrics, Section of Genetics and Metabolism, University of Colorado School of Medicine and Children's Hospital Colorado, Aurora, CO 80309, USA
| | - Ronald L Cowan
- Department of Psychiatry, The University of Tennessee Health Science Center College of Medicine, Memphis, TN 37996, USA
| | - Jennifer L Miller
- Department of Pediatrics, University of Florida College of Medicine, Gainesville, FL 32611, USA
| | - Jill K Hamilton
- Division of Endocrinology, The Hospital for Sick Children, Toronto M5G 1X8, Canada
- Department of Pediatrics, University of Toronto, Toronto M5G 1X8, Canada
| | - Elizabeth R Roeder
- Department of Pediatrics, Baylor College of Medicine, San Antonio, TX 78207, USA
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA
| | - Shana E McCormack
- Neuroendocrine Center, The Children's Hospital of Philadelphia Division of Endocrinology and Diabetes, Philadelphia, PA 19104, USA
- Department of Pediatrics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA
| | - Tamanna R Roshan Lal
- Genetics and Metabolism, Children's National Hospital, Washington, DC 20010, USA
| | - Hussein D Abdul-Latif
- Division of Pediatric Endocrinology and Diabetes, Children's of Alabama, Birmingham, AL 35233, USA
| | - Andrea M Haqq
- Department of Pediatrics, University of Alberta, Edmonton, Alberta T6G 2R3, Canada
| | - Kathryn S Obrynba
- Division of Endocrinology and Diabetes, Nationwide Children's Hospital, Columbus, OH 43205, USA
- Department of Pediatrics, The Ohio State University, Columbus, OH 43205, USA
| | - Laura C Torchen
- Division of Endocrinology, Ann and Robert H. Lurie Children's Hospital of Chicago, Feinberg School of Medicine, Northwestern University, Chicago, IL 60208, USA
| | - Alaina P Vidmar
- Diabetes & Obesity Program, Center for Endocrinology, Diabetes and Metabolism, Children's Hospital Los Angeles Department of Pediatrics, Los Angeles, CA 90027, USA
- Keck School of Medicine of USC, Los Angeles, CA 90033, USA
| | - David H Viskochil
- Department of Pediatrics, Division of Medical Genetics, The University of Utah School of Medicine, Salt Lake City, UT 84112, USA
- Shriners Hospital for Children, Salt Lake City, UT 84112, USA
| | - Jean-Pierre Chanoine
- Department of Pediatrics, Endocrinology and Diabetes Unit, The University of British Columbia, Vancouver V6H 3V4, Canada
| | - Carol K L Lam
- Department of Pediatrics, Endocrinology and Diabetes Unit, The University of British Columbia, Vancouver V6H 3V4, Canada
| | - Melinda J Pierce
- Diabetes & Endocrinology, Children's Minnesota—St Paul, St Paul, MN 55404, USA
| | - Laurel L Williams
- Menninger Department of Psychiatry & Behavioral Sciences, Baylor College of Medicine, Houston, TX 77030, USA
| | - Lynne M Bird
- Department of Pediatrics, University of California San Diego, San Diego, CA 92037, USA
- Rady Children's Hospital, San Diego, CA 92123, USA
| | - Merlin G Butler
- Department of Psychiatry & Behavioral Sciences, University of Kansas Medical Center, Kansas City, KS 66160, USA
| | - Diane E Jensen
- Children's Health Queensland Hospital and Health Services, South Brisbane, Queensland 4101, Australia
- Centre for Children's Health Research, University of Queensland, Brisbane, Queensland 4101, Australia
| | - Susan E Myers
- Department of Pediatrics, Saint Louis University School of Medicine, Cardinal Glennon Children's Hospital, Saint Louis, MO 63104, USA
| | - Oliver J Oatman
- Division of Endocrinology and Diabetes, Phoenix Children's Hospital, Phoenix, AZ 85016, USA
| | - Charumathi Baskaran
- Division of Endocrinology, Boston Children's Hospital, Boston, MA 02115, USA
- Department of Pediatrics, Harvard Medical School, Boston, MA 02115, USA
| | - Laura J Chalmers
- Department of Pediatrics, The University of Oklahoma School of Community Medicine, Tulsa, OK 73117, USA
| | - Cary Fu
- Vanderbilt University, Nashville, TN 37240, USA
| | - Nathalie Alos
- Department of Pediatrics, Centre Hospitalier Universitaire Sainte-Justine Centre de Recherche, Montréal, Québec H3T 1C5, Canada
| | - Scott D McLean
- Department of Pediatrics, Baylor College of Medicine, San Antonio, TX 78207, USA
| | - Ajay Shah
- Menninger Department of Psychiatry & Behavioral Sciences, Baylor College of Medicine, Houston, TX 77030, USA
| | - Barbara Y Whitman
- Department of Pediatrics, Saint Louis University School of Medicine, Cardinal Glennon Children's Hospital, Saint Louis, MO 63104, USA
| | | | | | | | | | | | | |
Collapse
|
9
|
Rice LJ, Agu J, Carter CS, Harris JC, Nazarloo HP, Naanai H, Einfeld SL. The relationship between endogenous oxytocin and vasopressin levels and the Prader-Willi syndrome behaviour phenotype. Front Endocrinol (Lausanne) 2023; 14:1183525. [PMID: 37313445 PMCID: PMC10259653 DOI: 10.3389/fendo.2023.1183525] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/10/2023] [Accepted: 05/09/2023] [Indexed: 06/15/2023] Open
Abstract
Background Oxytocin and vasopressin systems are altered in Prader Willi syndrome (PWS). However, investigations into endogenous oxytocin and vasopressin levels as well as clinical trials evaluating the effect of exogenous oxytocin on PWS symptoms have had mixed results. It is also unknown whether endogenous oxytocin and vasopressin levels are associated with certain PWS behaviours. Method We compared plasma oxytocin and vasopressin and saliva oxytocin levels in 30 adolescents and adults with PWS to 30 typically developing age-matched controls. We also compared neuropeptide levels between gender and genetic subtypes within the PWS cohort and examined the relationship between neuropeptide levels and PWS behaviours. Results While we did not measure a group difference in plasma or saliva oxytocin levels, plasma vasopressin was significantly lower in individuals with PWS compared to controls. Within the PWS cohort, saliva oxytocin levels were higher in females compared to males and individuals with the mUPD compared to the deletion genetic subtype. We also found the neuropeptides correlated with different PWS behaviours for males and females and for genetic subtypes. For the deletion group, higher plasma and saliva oxytocin levels were related to fewer behaviour problems. For the mUPD group, higher plasma vasopressin levels were related to more behaviour problems. Conclusion These findings support existing evidence of a vasopressin system defect in PWS and for the first time identify potential differences in the oxytocin and vasopressin systems across PWS genetic subtypes.
Collapse
Affiliation(s)
- Lauren J. Rice
- Faculty of Medicine and Health, Brain and Mind Centre, The University of Sydney, Sydney, NSW, Australia
- Faculty of Medicine and Health, Specialty of Child and Adolescent Health, The University of Sydney Children’s Hospital Westmead Clinical School, Sydney, NSW, Australia
| | - Josephine Agu
- Faculty of Medicine and Health, Specialty of Child and Adolescent Health, The University of Sydney Children’s Hospital Westmead Clinical School, Sydney, NSW, Australia
| | - C. Sue Carter
- Department of Psychology, University of Virginia, Charlottesville, VA, United States
- Kinsey Institute, Indiana University, Bloomington, IN, United States
| | - James C. Harris
- Department of Psychiatry and Behavioural Sciences and Paediatrics, Johns Hopkins University, Baltimore, MD, United States
| | - Hans P. Nazarloo
- Department of Psychology, University of Virginia, Charlottesville, VA, United States
- Kinsey Institute, Indiana University, Bloomington, IN, United States
| | - Habiba Naanai
- Faculty of Medicine and Health, Specialty of Child and Adolescent Health, The University of Sydney Children’s Hospital Westmead Clinical School, Sydney, NSW, Australia
| | - Stewart L. Einfeld
- Faculty of Medicine and Health, Brain and Mind Centre, The University of Sydney, Sydney, NSW, Australia
| |
Collapse
|
10
|
Shaffer RC, Reisinger DL, Schmitt LM, Lamy M, Dominick KC, Smith EG, Coffman MC, Esbensen AJ. Systematic Review: Emotion Dysregulation in Syndromic Causes of Intellectual and Developmental Disabilities. J Am Acad Child Adolesc Psychiatry 2023; 62:518-557. [PMID: 36007813 DOI: 10.1016/j.jaac.2022.06.020] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Revised: 06/03/2022] [Accepted: 08/15/2022] [Indexed: 11/30/2022]
Abstract
OBJECTIVE To summarize the current state of the literature regarding emotion dysregulation (ED) in syndromic intellectual disabilities (S-IDs) in 6 of the most common forms of S-IDs-Down syndrome, fragile X syndrome (FXS), tuberous sclerosis complex, Williams syndrome, Prader-Willi syndrome, and Angelman syndrome-and to determine future research directions for identification and treatment of ED. METHOD PubMed bibliographic database was searched from date of inception to May 2021. PRISMA 2020 guidelines were followed with the flowchart, table of included studies, list of excluded studies, and checklist provided. Filters applied included human research and English. Only original research articles were included in the final set, but review articles were used to identify secondary citations of primary studies. All articles were reviewed for appropriateness by 2 authors and summarized. Inclusion criteria were met by 145 articles (Down syndrome = 29, FXS = 55, tuberous sclerosis complex = 11, Williams syndrome = 18, Prader-Willi syndrome = 24, Angelman syndrome = 8). RESULTS Each syndrome review was summarized separately and further subdivided into articles related to underlying neurobiology, behaviors associated with ED, assessment, and targeted intervention. FXS had the most thorough research base, followed by Down syndrome and Prader-Willi syndrome, with the other syndromes having more limited available research. Very limited research was available regarding intervention for all disorders except FXS. CONCLUSION Core underlying characteristics of S-IDs appear to place youth at higher risk for ED, but further research is needed to better assess and treat ED in S-IDs. Future studies should have a standard assessment measure of ED, such as the Emotion Dysregulation Inventory, and explore adapting established curricula for ED from the neurotypical and autism spectrum disorder fields.
Collapse
Affiliation(s)
- Rebecca C Shaffer
- Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio; University of Cincinnati School of Medicine, Cincinnati, Ohio.
| | | | - Lauren M Schmitt
- Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio; University of Cincinnati School of Medicine, Cincinnati, Ohio
| | - Martine Lamy
- Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio; University of Cincinnati School of Medicine, Cincinnati, Ohio
| | - Kelli C Dominick
- Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio; University of Cincinnati School of Medicine, Cincinnati, Ohio
| | - Elizabeth G Smith
- Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio; University of Cincinnati School of Medicine, Cincinnati, Ohio
| | | | - Anna J Esbensen
- Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio; University of Cincinnati School of Medicine, Cincinnati, Ohio
| |
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
|
Clinical Trials in Prader-Willi Syndrome: A Review. Int J Mol Sci 2023; 24:ijms24032150. [PMID: 36768472 PMCID: PMC9916985 DOI: 10.3390/ijms24032150] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2022] [Revised: 12/28/2022] [Accepted: 01/16/2023] [Indexed: 01/25/2023] Open
Abstract
Prader-Willi syndrome (PWS) is a complex, genetic, neurodevelopmental disorder. PWS has three molecular genetic classes. The most common defect is due to a paternal 15q11-q13 deletion observed in about 60% of individuals. This is followed by maternal disomy 15 (both 15 s from the mother), found in approximately 35% of cases. the remaining individuals have a defect of the imprinting center that controls the activity of imprinted genes on chromosome 15. Mild cognitive impairment and behavior problems in PWS include self-injury, anxiety, compulsions, and outbursts in childhood, impacted by genetic subtypes. Food seeking and hyperphagia can lead to morbid obesity and contribute to diabetes and cardiovascular or orthopedic problems. The control of hyperphagia and improving food-related behaviors are the most important unmet needs in PWS and could be addressed with the development of a new therapeutic agent, as currently no approved therapeutics exist for PWS treatment. The status of clinical trials with existing results for the management of obesity and hyperphagia in PWS will be discussed in this review, including treatments such as beloranib, setmelanotide, a diazoxide choline controlled-release tablet (DCCR), an unacylated ghrelin analogue, oxytocin and related compounds, glucagon-like peptide 1 receptor agonists, surgical intervention, and transcranial direct-current stimulation.
Collapse
|
13
|
Althammer F, Wimmer MC, Krabichler Q, Küppers S, Schimmer J, Fröhlich H, Dötsch L, Gruber T, Wunsch S, Schubert T, Kirchner MK, Stern JE, Charlet A, Grinevich V, Schaaf CP. Analysis of the hypothalamic oxytocin system and oxytocin receptor-expressing astrocytes in a mouse model of Prader-Willi syndrome. J Neuroendocrinol 2022; 34:e13217. [PMID: 36458331 DOI: 10.1111/jne.13217] [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: 08/11/2022] [Revised: 11/10/2022] [Accepted: 11/10/2022] [Indexed: 11/17/2022]
Abstract
Prader-Willi syndrome (PWS) is a neurodevelopmental disorder characterized by hyperphagia, obesity, developmental delay and intellectual disability. Studies suggest dysfunctional signaling of the neuropeptide oxytocin as one of the key mechanisms in PWS, and administration of oxytocin via intranasal or systemic routes yielded promising results in both humans and mouse models. However, a detailed assessment of the oxytocin system in mouse models of PWS such as the Magel2-deficient Magel2tm1.Stw mouse, is lacking. In the present study, we performed an automated counting of oxytocin cells in the entire paraventricular nucleus of the hypothalamus of Magel2tm1.Stw and wild-type control mice and found a significant reduction in the caudal part, which represents the parvocellular subdivision. In addition, based on the recent discovery that some astrocytes express the oxytocin receptor (OTR), we performed detailed analysis of astrocyte numbers and morphology in various brain regions, and assessed expression levels of the astrocyte marker glial fibrillary acidic protein, which was significantly decreased in the hypothalamus, but not other brain regions in Magel2tm1.Stw mice. Finally, we analyzed the number of OTR-expressing astrocytes in various brain regions and found a significant reduction in the nucleus accumbens of Magel2tm1.Stw mice, as well as a sex-specific difference in the lateral septum. This study suggests a role for caudal paraventricular nucleus oxytocin neurons as well as OTR-expressing astrocytes in a mouse model of PWS, provides novel information about sex-specific expression of astrocytic OTRs, and presents several new brain regions containing OTR-expressing astrocytes in the mouse brain.
Collapse
Affiliation(s)
| | | | - Quirin Krabichler
- Department of Neuropeptide Research in Psychiatry, Central Institute of Mental Health, Heidelberg University, Mannheim, Germany
| | - Stephanie Küppers
- Department of Neuropeptide Research in Psychiatry, Central Institute of Mental Health, Heidelberg University, Mannheim, Germany
| | - Jonas Schimmer
- Department of Neuropeptide Research in Psychiatry, Central Institute of Mental Health, Heidelberg University, Mannheim, Germany
| | - Henning Fröhlich
- Institute of Human Genetics, Heidelberg University, Heidelberg, Germany
| | - Laura Dötsch
- Institute of Human Genetics, Heidelberg University, Heidelberg, Germany
| | - Tim Gruber
- Van Andel Institute, Grand Rapids, MI, USA
| | - Selina Wunsch
- Institute of Human Genetics, Heidelberg University, Heidelberg, Germany
| | - Tim Schubert
- Institute of Human Genetics, Heidelberg University, Heidelberg, Germany
| | - Matthew K Kirchner
- Center for Neuroinflammation and Cardiometabolic Diseases, Georgia State University, Atlanta, GA, USA
| | - Javier E Stern
- Center for Neuroinflammation and Cardiometabolic Diseases, Georgia State University, Atlanta, GA, USA
| | - Alexandre Charlet
- Centre National de la Recherche Scientifique and University of Strasbourg, Institute of Cellular and Integrative Neuroscience, Strasbourg, France
| | - Valery Grinevich
- Department of Neuropeptide Research in Psychiatry, Central Institute of Mental Health, Heidelberg University, Mannheim, Germany
| | | |
Collapse
|
14
|
Oztan O, Zyga O, Stafford DEJ, Parker KJ. Linking oxytocin and arginine vasopressin signaling abnormalities to social behavior impairments in Prader-Willi syndrome. Neurosci Biobehav Rev 2022; 142:104870. [PMID: 36113782 PMCID: PMC11024898 DOI: 10.1016/j.neubiorev.2022.104870] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2022] [Revised: 09/06/2022] [Accepted: 09/12/2022] [Indexed: 11/19/2022]
Abstract
Prader-Willi syndrome (PWS) is a genetic neurodevelopmental disorder. Global hypothalamic dysfunction is a core feature of PWS and has been implicated as a driver of many of PWS's phenotypic characteristics (e.g., hyperphagia-induced obesity, hypogonadism, short stature). Although the two neuropeptides (i.e., oxytocin [OXT] and arginine vasopressin [AVP]) most implicated in mammalian prosocial functioning are of hypothalamic origin, and social functioning is markedly impaired in PWS, there has been little consideration of how dysregulation of these neuropeptide signaling pathways may contribute to PWS's social behavior impairments. The present article addresses this gap in knowledge by providing a comprehensive review of the preclinical and clinical PWS literature-spanning endogenous neuropeptide measurement to exogenous neuropeptide administration studies-to better understand the roles of OXT and AVP signaling in this population. The preponderance of evidence indicates that OXT and AVP signaling are indeed dysregulated in PWS, and that these neuropeptide pathways may provide promising targets for therapeutic intervention in a patient population that currently lacks a pharmacological strategy for its debilitating social behavior symptoms.
Collapse
Affiliation(s)
- Ozge Oztan
- 1201 Welch Road, Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA 94305, USA
| | - Olena Zyga
- 1201 Welch Road, Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA 94305, USA
| | - Diane E J Stafford
- Center for Academic Medicine, 453 Quarry Road, Department of Pediatrics, Division of Pediatric Endocrinology, Stanford University, Palo Alto, CA 94304, USA
| | - Karen J Parker
- 1201 Welch Road, Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA 94305, USA; 300 Pasteur Drive, Department of Comparative Medicine, Stanford University, Stanford, CA 94305, USA.
| |
Collapse
|
15
|
Oxytocin-based therapies for treatment of Prader-Willi and Schaaf-Yang syndromes: evidence, disappointments, and future research strategies. Transl Psychiatry 2022; 12:318. [PMID: 35941105 PMCID: PMC9360032 DOI: 10.1038/s41398-022-02054-1] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/14/2022] [Revised: 06/23/2022] [Accepted: 07/01/2022] [Indexed: 11/09/2022] Open
Abstract
The prosocial neuropeptide oxytocin is being developed as a potential treatment for various neuropsychiatric disorders including autism spectrum disorder (ASD). Early studies using intranasal oxytocin in patients with ASD yielded encouraging results and for some time, scientists and affected families placed high hopes on the use of intranasal oxytocin for behavioral therapy in ASD. However, a recent Phase III trial obtained negative results using intranasal oxytocin for the treatment of behavioral symptoms in children with ASD. Given the frequently observed autism-like behavioral phenotypes in Prader-Willi and Schaaf-Yang syndromes, it is unclear whether oxytocin treatment represents a viable option to treat behavioral symptoms in these diseases. Here we review the latest findings on intranasal OT treatment, Prader-Willi and Schaaf-Yang syndromes, and propose novel research strategies for tailored oxytocin-based therapies for affected individuals. Finally, we propose the critical period theory, which could explain why oxytocin-based treatment seems to be most efficient in infants, but not adolescents.
Collapse
|
16
|
Erhardt É, Molnár D. Prader–Willi Syndrome: Possibilities of Weight Gain Prevention and Treatment. Nutrients 2022; 14:nu14091950. [PMID: 35565916 PMCID: PMC9103725 DOI: 10.3390/nu14091950] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2022] [Revised: 04/27/2022] [Accepted: 05/02/2022] [Indexed: 11/16/2022] Open
Abstract
Prader–Willi syndrome (PWS) is a complex genetic disorder which involves the endocrine and neurologic systems, metabolism, and behavior. The aim of this paper is to summarize current knowledge on dietary management and treatment of PWS and, in particular, to prevent excessive weight gain. Growth hormone (GH) therapy is the recommended standard treatment for PWS children, because it improves body composition (by changing the proportion of body fat and lean body mass specifically by increasing muscle mass and energy expenditure), linear growth, and in infants, it promotes psychomotor and IQ development. In early childhood, the predominant symptom is hyperphagia which can lead to early onset, severe obesity with different obesity-related comorbidities. There are several studies on anti-obesity medications (metformin, topiramate, liraglutide, setmelanotide). However, these are still limited, and no widely accepted consensus guideline exists concerning these drugs in children with PWS. Until there is a specific treatment for hyperphagia and weight gain, weight must be controlled with the help of diet and exercise. Below the age of one year, children with PWS have no desire to eat and will often fail to thrive, despite adequate calories. After the age of two years, weight begins to increase without a change in calorie intake. Appetite increases later, gradually, and becomes insatiable. Managing the progression of different nutritional phases (0–4) is really important and can delay the early onset of severe obesity. Multidisciplinary approaches are crucial in the diagnosis and lifelong follow-up, which will determine the quality of life of these patients.
Collapse
Affiliation(s)
- Éva Erhardt
- Department of Paediatrics, Medical School, University of Pécs, H-7623 Pécs, Hungary;
- Correspondence: ; Tel.: +36-72-536-000 (ext. 38711); Fax: +36-72-535-971
| | - Dénes Molnár
- Department of Paediatrics, Medical School, University of Pécs, H-7623 Pécs, Hungary;
- National Laboratory for Human Reproduction, Szentágothai Research Centre, University of Pécs, H-7623 Pécs, Hungary
| |
Collapse
|
17
|
Müller HL, Tauber M, Lawson EA, Özyurt J, Bison B, Martinez-Barbera JP, Puget S, Merchant TE, van Santen HM. Hypothalamic syndrome. Nat Rev Dis Primers 2022; 8:24. [PMID: 35449162 DOI: 10.1038/s41572-022-00351-z] [Citation(s) in RCA: 43] [Impact Index Per Article: 21.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 03/08/2022] [Indexed: 12/11/2022]
Abstract
Hypothalamic syndrome (HS) is a rare disorder caused by disease-related and/or treatment-related injury to the hypothalamus, most commonly associated with rare, non-cancerous parasellar masses, such as craniopharyngiomas, germ cell tumours, gliomas, cysts of Rathke's pouch and Langerhans cell histiocytosis, as well as with genetic neurodevelopmental syndromes, such as Prader-Willi syndrome and septo-optic dysplasia. HS is characterized by intractable weight gain associated with severe morbid obesity, multiple endocrine abnormalities and memory impairment, attention deficit and reduced impulse control as well as increased risk of cardiovascular and metabolic disorders. Currently, there is no cure for this condition but treatments for general obesity are often used in patients with HS, including surgery, medication and counselling. However, these are mostly ineffective and no medications that are specifically approved for the treatment of HS are available. Specific challenges in HS are because the syndrome represents an adverse effect of different diseases, and that diagnostic criteria, aetiology, pathogenesis and management of HS are not completely defined.
Collapse
Affiliation(s)
- Hermann L Müller
- Department of Paediatrics and Paediatric Hematology/Oncology, University Children's Hospital, Klinikum Oldenburg AöR, Carl von Ossietzky University, Oldenburg, Germany.
| | - Maithé Tauber
- Centre de Référence du Syndrome de Prader-Willi et autres syndromes avec troubles du comportement alimentaire, Hôpital des Enfants, CHU-Toulouse, Toulouse, France
- Institut Toulousain des Maladies Infectieuses et Inflammatoires (Infinity) INSERM UMR1291 - CNRS UMR5051 - Université Toulouse III, Toulouse, France
| | - Elizabeth A Lawson
- Neuroendocrine Unit, Massachusetts General Hospital, and Department of Medicine, Harvard Medical School, Boston, MA, USA
| | - Jale Özyurt
- Biological Psychology Laboratory, Department of Psychology, School of Medicine and Health Sciences, Carl von Ossietzky University, Oldenburg, Germany
- Research Center Neurosensory Science, Carl von Ossietzky University, Oldenburg, Germany
| | - Brigitte Bison
- Department of Neuroradiology, University Hospital Augsburg, Augsburg, Germany
| | - Juan-Pedro Martinez-Barbera
- Developmental Biology and Cancer Programme, Birth Defects Research Centre, UCL Great Ormond Street Institute of Child Health, University College London, London, UK
| | - Stephanie Puget
- Service de Neurochirurgie, Hôpital Necker-Enfants Malades, Sorbonne Paris Cité, Paris, France
- Service de Neurochirurgie, Hopital Pierre Zobda Quitman, Martinique, France
| | - Thomas E Merchant
- Department of Radiation Oncology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Hanneke M van Santen
- Department of Paediatric Endocrinology, Wilhelmina Children's Hospital, University Medical Center, Utrecht, Netherlands
- Princess Máxima Center for Paediatric Oncology, Utrecht, Netherlands
| |
Collapse
|
18
|
Kim SJ, Cho SY, Jin DK. Prader-Willi syndrome: an update on obesity and endocrine problems. Ann Pediatr Endocrinol Metab 2021; 26:227-236. [PMID: 34991300 PMCID: PMC8749024 DOI: 10.6065/apem.2142164.082] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/16/2021] [Accepted: 08/30/2021] [Indexed: 11/24/2022] Open
Abstract
Prader-Willi syndrome (PWS) is a rare complex genetic disorder that results from a lack of expression of the paternally inherited chromosome 15q11-q13. PWS is characterized by hypotonia and feeding difficulty in early infancy and development of morbid obesity aggravated by uncontrolled hyperphagia after childhood and adolescent. Dysmorphic facial features, delayed motor and language development, various degrees of cognitive impairment, and behavioral problems are common in PWS. Without early, intensive nutritional therapy along with behavioral modification, PWS patients develop severe obesity associated with type 2 diabetes, obstructive sleep apnea, right-side heart failure, and other obesity-related metabolic complications. Hypothalamic dysfunction in PWS can lead to several endocrine disorders, including short stature with growth hormone deficiency, hypothyroidism, central adrenal insufficiency, and hypogonadism. In this review, we discuss the natural history of PWS and the mechanisms of hyperphagia and obesity. We also provide an update on obesity treatments and recommendations for screening and monitoring of various endocrine problems that can occur in PWS.
Collapse
Affiliation(s)
- Su Jin Kim
- Department of Pediatrics, Inha University Hospital, Inha University College of Medicine, Incheon, Korea,Northwest Gyeonggi Regional Center for Rare Disease, Inha University Hospital, Incheon, Korea
| | - Sung Yoon Cho
- Department of Pediatrics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Dong-Kyu Jin
- Department of Pediatrics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea,Address for correspondence: Dong-Kyu Jin Department of Pediatrics, Samsung Medical Center, Sungkyunkwan University School of Medicine, 81 Irwon-ro, Gangnam-gu, Seoul, 06351, Korea
| |
Collapse
|
19
|
Oxytocin and Food Intake Control: Neural, Behavioral, and Signaling Mechanisms. Int J Mol Sci 2021; 22:ijms221910859. [PMID: 34639199 PMCID: PMC8509519 DOI: 10.3390/ijms221910859] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2021] [Revised: 09/29/2021] [Accepted: 10/03/2021] [Indexed: 11/16/2022] Open
Abstract
The neuropeptide oxytocin is produced in the paraventricular hypothalamic nucleus and the supraoptic nucleus of the hypothalamus. In addition to its extensively studied influence on social behavior and reproductive function, central oxytocin signaling potently reduces food intake in both humans and animal models and has potential therapeutic use for obesity treatment. In this review, we highlight rodent model research that illuminates various neural, behavioral, and signaling mechanisms through which oxytocin’s anorexigenic effects occur. The research supports a framework through which oxytocin reduces food intake via amplification of within-meal physiological satiation signals rather than by altering between-meal interoceptive hunger and satiety states. We also emphasize the distributed neural sites of action for oxytocin’s effects on food intake and review evidence supporting the notion that central oxytocin is communicated throughout the brain, at least in part, through humoral-like volume transmission. Finally, we highlight mechanisms through which oxytocin interacts with various energy balance-associated neuropeptide and endocrine systems (e.g., agouti-related peptide, melanin-concentrating hormone, leptin), as well as the behavioral mechanisms through which oxytocin inhibits food intake, including effects on nutrient-specific ingestion, meal size control, food reward-motivated responses, and competing motivations.
Collapse
|
20
|
Anti-Obesity Medication Use in Children and Adolescents with Prader-Willi Syndrome: Case Review and Literature Search. J Clin Med 2021; 10:jcm10194540. [PMID: 34640558 PMCID: PMC8509766 DOI: 10.3390/jcm10194540] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2021] [Revised: 09/24/2021] [Accepted: 09/24/2021] [Indexed: 01/26/2023] Open
Abstract
(1) Background: children with Prader-Willi syndrome (PWS) have high obesity rates due to hyperphagia and decreased metabolic rates. Although anti-obesity medications (AOMs) are prescribed to this population, there are no consensus guidelines on acceptability, safety, and efficacy. We present literature review and case series on AOMs in youth with PWS. (2) Methods: we performed PubMed review from January 2000 to April 2021 utilizing keywords: "Prader-Willi syndrome" or "PWS" and "medication" including: topiramate, metformin, phentermine, liraglutide, orlistat, oxytocin, semaglutide, naltrexone-bupropion. For our case series, patients were identified through retrospective chart reviews from a multi-disciplinary PWS clinic. Eligibility criteria: age ≤ 18 years, genetically confirmed PWS, AOM use for at least 16 weeks, and recent anthropometric data. (3) Results: a literature search yielded 14 articles (3 topiramate, 1 metformin, 4 liraglutide, 5 oxytocin, 1 naltrexone-bupropion). All studies reported improved hyperphagia with variable BMI effects. Ten adolescents met case series eligibility (mean age 13.2 ± 2.6 years, 40% female; AOMs: 6 metformin, 5 topiramate, 2 semaglutide, 3 liraglutide). After AOM course, 60% had decreased or stable BMI z-score. No significant side effects. (4) Conclusions: results suggest AOMs may be useful for weight management in youth with PWS. Additional studies are required to validate findings and support AOM treatment guidelines.
Collapse
|
21
|
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
|
22
|
Tauber M, Diene G. Prader-Willi syndrome: Hormone therapies. HANDBOOK OF CLINICAL NEUROLOGY 2021; 181:351-367. [PMID: 34238470 DOI: 10.1016/b978-0-12-820683-6.00026-9] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Prader-Willi syndrome (PWS) is a rare genetic neurodevelopmental disorder linked to the lack of expression of specific maternally imprinted genes located in the chromosomal region 15q11-q13. Impaired hypothalamic development and function explain most of the phenotype that is characterized by a specific trajectory from anorexia at birth to excessive weight gain at later ages, which is accompanied by hyperphagia and early severe obesity, as well as by other hormonal deficiencies, behavioral deficits, and dysautonomia. In almost all patients, their endocrine dysfunction involves growth hormone deficiency and hypogonadism, which originate from a combination of both peripheral and hypothalamic origin, central hypothyroidism in 40%, precocious adrenarche in 30% of the cases, and in rare cases, also adrenocorticotropin deficiency and precocious puberty. In addition, the oxytocin (OXT) and ghrelin systems are impaired in most patients and involved in a poor suckling response at birth, and hyperphagia with food addiction, poor social skills, and emotional dysregulation. Current hormonal replacement treatments are the same as used in classical hormonal deficiencies, and recombinant human GH treatment is registered since 2000 and has dramatically changed the phenotype of these children. OXT and OXT analogue treatments are currently investigated as well as new molecules targeting the ghrelin system. The severe condition of PWS can be seen as a model to improve the fine description and treatments of hypothalamic dysfunction.
Collapse
Affiliation(s)
- Maithé Tauber
- Centre de Référence du Syndrome de Prader-Willi, Hôpital des Enfants, CHU Toulouse, Toulouse, France.
| | - Gwenaelle Diene
- Centre de Référence du Syndrome de Prader-Willi, Hôpital des Enfants, CHU Toulouse, Toulouse, France
| |
Collapse
|
23
|
Correa‐da‐Silva F, Fliers E, Swaab DF, Yi C. Hypothalamic neuropeptides and neurocircuitries in Prader Willi syndrome. J Neuroendocrinol 2021; 33:e12994. [PMID: 34156126 PMCID: PMC8365683 DOI: 10.1111/jne.12994] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/14/2020] [Revised: 04/19/2021] [Accepted: 05/04/2021] [Indexed: 02/06/2023]
Abstract
Prader-Willi Syndrome (PWS) is a rare and incurable congenital neurodevelopmental disorder, resulting from the absence of expression of a group of genes on the paternally acquired chromosome 15q11-q13. Phenotypical characteristics of PWS include infantile hypotonia, short stature, incomplete pubertal development, hyperphagia and morbid obesity. Hypothalamic dysfunction in controlling body weight and food intake is a hallmark of PWS. Neuroimaging studies have demonstrated that PWS subjects have abnormal neurocircuitry engaged in the hedonic and physiological control of feeding behavior. This is translated into diminished production of hypothalamic effector peptides which are responsible for the coordination of energy homeostasis and satiety. So far, studies with animal models for PWS and with human post-mortem hypothalamic specimens demonstrated changes particularly in the infundibular and the paraventricular nuclei of the hypothalamus, both in orexigenic and anorexigenic neural populations. Moreover, many PWS patients have a severe endocrine dysfunction, e.g. central hypogonadism and/or growth hormone deficiency, which may contribute to the development of increased fat mass, especially if left untreated. Additionally, the role of non-neuronal cells, such as astrocytes and microglia in the hypothalamic dysregulation in PWS is yet to be determined. Notably, microglial activation is persistently present in non-genetic obesity. To what extent microglia, and other glial cells, are affected in PWS is poorly understood. The elucidation of the hypothalamic dysfunction in PWS could prove to be a key feature of rational therapeutic management in this syndrome. This review aims to examine the evidence for hypothalamic dysfunction, both at the neuropeptidergic and circuitry levels, and its correlation with the pathophysiology of PWS.
Collapse
Affiliation(s)
- Felipe Correa‐da‐Silva
- Department of Endocrinology and MetabolismAmsterdam Gastroenterology Endocrinology and MetabolismAmsterdam University Medical Center (UMC)University of AmsterdamAmsterdamThe Netherlands
- Laboratory of EndocrinologyAmsterdam University Medical Center (UMC)University of AmsterdamAmsterdamThe Netherlands
- Department of Neuropsychiatric DisordersNetherlands Institute for NeuroscienceAn Institute of the Royal Netherlands Academy of Arts and SciencesAmsterdamThe Netherlands
| | - Eric Fliers
- Department of Endocrinology and MetabolismAmsterdam Gastroenterology Endocrinology and MetabolismAmsterdam University Medical Center (UMC)University of AmsterdamAmsterdamThe Netherlands
| | - Dick F. Swaab
- Department of Neuropsychiatric DisordersNetherlands Institute for NeuroscienceAn Institute of the Royal Netherlands Academy of Arts and SciencesAmsterdamThe Netherlands
| | - Chun‐Xia Yi
- Department of Endocrinology and MetabolismAmsterdam Gastroenterology Endocrinology and MetabolismAmsterdam University Medical Center (UMC)University of AmsterdamAmsterdamThe Netherlands
- Laboratory of EndocrinologyAmsterdam University Medical Center (UMC)University of AmsterdamAmsterdamThe Netherlands
- Department of Neuropsychiatric DisordersNetherlands Institute for NeuroscienceAn Institute of the Royal Netherlands Academy of Arts and SciencesAmsterdamThe Netherlands
| |
Collapse
|
24
|
Damen L, Grootjen LN, Juriaans AF, Donze SH, Huisman TM, Visser JA, Delhanty PJ, Hokken‐Koelega AC. Oxytocin in young children with Prader-Willi syndrome: Results of a randomized, double-blind, placebo-controlled, crossover trial investigating 3 months of oxytocin. Clin Endocrinol (Oxf) 2021; 94:774-785. [PMID: 33296519 PMCID: PMC8246775 DOI: 10.1111/cen.14387] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/28/2020] [Revised: 11/20/2020] [Accepted: 11/29/2020] [Indexed: 12/24/2022]
Abstract
CONTEXT Prader-Willi syndrome (PWS) is characterized by hypothalamic dysfunction, hyperphagia and a typical behavioural phenotype, with characteristics of autism spectrum disorder (ASD) like stubbornness, temper tantrums and compulsivity. It has been suggested that the oxytocin system in patients with PWS is dysfunctional. In ASD, intranasal oxytocin treatment has favourable effects on behaviour. OBJECTIVE To evaluate the effects of 3 months of twice daily intranasal oxytocin (dose range 16-40 IU/day), compared to placebo, on behaviour and hyperphagia in children with PWS. DESIGN Randomized, double-blind, placebo-controlled, crossover study in the Dutch PWS Reference Center. PATIENTS Twenty-six children with PWS aged 3-11 years. MAIN OUTCOME MEASURES (Change in) behaviour and hyperphagia measured by Oxytocin Questionnaire and Dykens hyperphagia questionnaire. RESULTS In the total group, no significant effects of oxytocin on social behaviour or hyperphagia were found. However, in boys, the Oxytocin Questionnaire scores improved significantly during oxytocin treatment, compared to a deterioration during placebo (4.5 (-0.8 to 15.3) vs. -4.0 (-11.3 to 0.8), P = .025). The Dykens hyperphagia questionnaire scores remained similar during oxytocin treatment, while there was a deterioration during placebo (0.0 (-0.8 to 4.3) vs. -3.5 (-6.0 to 0.0), P = .046). Patients with a deletion had significant improvements in both questionnaire scores during oxytocin treatment, but deteriorations during placebo. Oxytocin treatment was well tolerated, and there were no serious adverse events. CONCLUSIONS Intranasal oxytocin treatment has positive effects on social and eating behaviour in 3-11 years aged boys with PWS and in children with a deletion without safety concerns. Intranasal oxytocin in children with PWS might be considered, but individual effects should be carefully evaluated and treatment discontinued if no effects are found.
Collapse
Affiliation(s)
- Layla Damen
- Dutch Growth Research FoundationRotterdamthe Netherlands
- Department of PediatricsSubdivision of EndocrinologyErasmus University Medical CenterSophia Children’s HospitalRotterdamthe Netherlands
- Dutch Reference Center for Prader‐Willi Syndromethe Netherlands
| | - Lionne N. Grootjen
- Dutch Growth Research FoundationRotterdamthe Netherlands
- Department of PediatricsSubdivision of EndocrinologyErasmus University Medical CenterSophia Children’s HospitalRotterdamthe Netherlands
- Dutch Reference Center for Prader‐Willi Syndromethe Netherlands
| | - Alicia F. Juriaans
- Dutch Growth Research FoundationRotterdamthe Netherlands
- Department of PediatricsSubdivision of EndocrinologyErasmus University Medical CenterSophia Children’s HospitalRotterdamthe Netherlands
- Dutch Reference Center for Prader‐Willi Syndromethe Netherlands
| | - Stephany H. Donze
- Dutch Growth Research FoundationRotterdamthe Netherlands
- Department of PediatricsSubdivision of EndocrinologyErasmus University Medical CenterSophia Children’s HospitalRotterdamthe Netherlands
- Dutch Reference Center for Prader‐Willi Syndromethe Netherlands
| | - T. Martin Huisman
- Department of Internal MedicineErasmus University Medical CenterRotterdamthe Netherlands
| | - Jenny A. Visser
- Department of Internal MedicineErasmus University Medical CenterRotterdamthe Netherlands
| | - Patric J.D. Delhanty
- Department of Internal MedicineErasmus University Medical CenterRotterdamthe Netherlands
| | - Anita C.S. Hokken‐Koelega
- Dutch Growth Research FoundationRotterdamthe Netherlands
- Department of PediatricsSubdivision of EndocrinologyErasmus University Medical CenterSophia Children’s HospitalRotterdamthe Netherlands
- Dutch Reference Center for Prader‐Willi Syndromethe Netherlands
| |
Collapse
|
25
|
Abstract
PURPOSE OF REVIEW Childhood obesity is escalating globally. Lifestyle and behavioral changes, which are the frequently used interventions in clinical practice, lead to only modest improvements in children with established obesity. Bariatric surgery is currently the most effective obesity treatment but has very limited utilization in pediatric obesity and is preferentially used for children with worsening comorbidities. There exists a massive treatment gap for children suffering with obesity especially after the failure of lifestyle modifications. Pharmacotherapy that is an established management tool in adults is very infrequently used in children. Only two medications, Phentermine and Orlistat are approved by the Food and Drug Administration (FDA) for use in adolescent obesity. Herein, we discuss the current landscape and available literature on the use of antiobesity pharmacotherapy in children. RECENT FINDINGS There are emerging pediatric data about the efficacy of the many weight loss medications that are FDA approved in adults. Moreover, more clinical trials are underway on the rarer, intractable forms of obesity such as monogenic, syndromic, and hypothalamic obesity. SUMMARY Weight loss medications in children, like adults, have variable efficacy and similar side effect profiles. Rigorous research and improved education of providers about weight loss medications may address the huge treatment gap in severe pediatric obesity.
Collapse
Affiliation(s)
- Vibha Singhal
- Division of Pediatric Endocrinology, Massachusetts General Hospital, Harvard Medical School, Boston, MA
- MGH Weight Center, Harvard Medical School, Boston, MA
| | - Aluma Chovel Sella
- Division of Pediatric Endocrinology, Massachusetts General Hospital, Harvard Medical School, Boston, MA
| | - Sonali Malhotra
- Division of Pediatric Endocrinology, Massachusetts General Hospital, Harvard Medical School, Boston, MA
- MGH Weight Center, Harvard Medical School, Boston, MA
| |
Collapse
|
26
|
The promiscuity of the oxytocin-vasopressin systems and their involvement in autism spectrum disorder. HANDBOOK OF CLINICAL NEUROLOGY 2021; 182:121-140. [PMID: 34266588 DOI: 10.1016/b978-0-12-819973-2.00009-5] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Oxytocin and vasopressin systems have been studied separately in autism spectrum disorder (ASD). Here, we provide evidence from an evolutionary and neuroscience perspective about the shared mechanisms and the common roles in regulating social behaviors. We first discuss findings on the evolutionary history of oxytocin and vasopressin ligands and receptors that highlight their common origin and clarify the evolutionary background of the crosstalk between them. Second, we conducted a comprehensive review of the increasing evidence for the role of both neuropeptides in regulating social behaviors. Third, we reviewed the growing evidence on the associations between the oxytocin/vasopressin systems and ASD, which includes oxytocin and vasopressin dysfunction in animal models of autism and in human patients, and the impact of treatments targeting the oxytocin or the vasopressin systems in children and in adults. Here, we highlight the potential of targeting the oxytocin/vasopressin systems to improve social deficits observed in ASD and the need for further investigations on how to transfer these research innovations into clinical applications.
Collapse
|
27
|
Whittington JE, Holland AJ. Disorders of hypothalamic function: Insights from Prader-Willi syndrome and the effects of craniopharyngioma. HANDBOOK OF CLINICAL NEUROLOGY 2021; 181:381-389. [PMID: 34238472 DOI: 10.1016/b978-0-12-820683-6.00028-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Either physical damage or being born with a specific genetic abnormality can impact on the functioning of the hypothalamus, resulting in diverse physical manifestations and/or specific behavior disorders. The impact of physical damage due to craniopharyngioma (CP) and/or surgery to remove a craniopharyngioma is compared and contrasted with the impact resulting from the genetic abnormalities associated with Prader-Willi syndrome (PWS). Similarities between PWS and CP posttreatment include hyperphagia and weight gain, low growth hormone levels, low bone density in adults, hypogonadism, disturbed temperature regulation, disturbed sleep and daytime sleepiness, memory difficulties, and problems with behavior and with peer relationships. These disturbances are an indication of the hypothalamus's central role in homeostasis. Most of the abnormalities appear to be more severe postoperatively in people with CP. Differences include higher ghrelin levels in PWS, complete absence of pituitary hormones in many cases of CP, higher incidence of thyroid dysfunction in CP, "growth without growth hormone" in obese children with CP, different types of diabetes (diabetes insipidus in CP and diabetes mellitus in PWS), and evidence of developmental delay and low IQ in people with PWS.
Collapse
Affiliation(s)
- Joyce E Whittington
- Department of Psychiatry, University of Cambridge, Cambridge, United Kingdom.
| | - Anthony J Holland
- Department of Psychiatry, University of Cambridge, Cambridge, United Kingdom
| |
Collapse
|
28
|
Abstract
The hypothalamic peptide oxytocin has been increasingly recognized as a hormone and neurotransmitter with important effects on energy intake, metabolism, and body weight and is under investigation as a potential novel therapeutic agent for obesity. The main neurons producing oxytocin and expressing the oxytocin receptor are strategically located in brain areas known to be critically involved in homeostatic energy balance as well as hedonic and motivational aspects of eating behavior. In this chapter, we will review the central and peripheral physiology of oxytocin and the interaction of oxytocin with key hormones and neural circuitries that affect food intake and metabolism. Next, we will synthesize the available data on endogenous oxytocin levels related to caloric intake, body weight, and metabolic status. We will then review the effects of exogenous oxytocin administration on eating behavior, body weight, and metabolism in humans, including in healthy individuals as well as specific populations with suspected perturbations involving oxytocin pathways. Finally, we will address the promise and fundamental challenges of 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, United States; Division of Pediatric Endocrinology, Massachusetts General Hospital for Children, Boston, MA, United States
| | - Elizabeth A Lawson
- Neuroendocrine Unit, Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA, United States.
| |
Collapse
|
29
|
Horta M, Pehlivanoglu D, Ebner NC. The Role of Intranasal Oxytocin on Social Cognition: An Integrative Human Lifespan Approach. Curr Behav Neurosci Rep 2020; 7:175-192. [PMID: 33717829 PMCID: PMC7951958 DOI: 10.1007/s40473-020-00214-5] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 08/31/2020] [Indexed: 12/26/2022]
Abstract
PURPOSE OF REVIEW This narrative review synthesizes research from the last two decades on the modulatory role of intranasal OT administration (IN-OT) on social cognition in early life, young/middle adulthood, and older adulthood. Advances and knowledge gaps are identified, and future research directions are discussed within an integrative human lifespan framework to guide novel research on IN-OT and social cognition. RECENT FINDINGS Current evidence regarding IN-OT modulation of social-cognitive processes, behavior, and related neurocircuitry is mixed, with some studies suggesting benefits (e.g., improved social perception/interactions, emotion processing) depending on contextual (e.g., social stimuli) and interindividual factors (e.g., age, sex, clinical status). Current research, however, is limited by a focus on isolated life phases, males, and select clinical populations as well as a lack of standardized protocols. SUMMARY This literature-based reflection proposes that greater generalizability of findings and scientific advancement on social-cognitive modulation via IN-OT require standardized, multi-method, longitudinal, and cross-sequential assessments in well-powered, well-controlled, and representative samples in line with an integrative lifespan approach, which considers development as a lifelong dynamic process involving both change and stability characterized by the interplay between genetic, neurobiological, and socio-behavioral factors.
Collapse
Affiliation(s)
- Marilyn Horta
- Department of Psychology, University of Florida, Gainesville, FL, USA
- Department of Epidemiology, University of Florida, Gainesville, FL, USA
| | | | - Natalie C. Ebner
- Department of Psychology, University of Florida, Gainesville, FL, USA
- Institute on Aging, Department of Aging & Geriatric Research, University of Florida, Gainesville, FL, USA
| |
Collapse
|
30
|
Exogenous effects of oxytocin in five psychiatric disorders: a systematic review, meta-analyses and a personalized approach through the lens of the social salience hypothesis. Neurosci Biobehav Rev 2020; 114:70-95. [DOI: 10.1016/j.neubiorev.2020.04.023] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2019] [Revised: 04/19/2020] [Accepted: 04/21/2020] [Indexed: 02/08/2023]
|
31
|
López-Tobón A, Trattaro S, Testa G. The sociability spectrum: evidence from reciprocal genetic copy number variations. Mol Autism 2020; 11:50. [PMID: 32546261 PMCID: PMC7298749 DOI: 10.1186/s13229-020-00347-0] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2020] [Accepted: 05/11/2020] [Indexed: 02/14/2023] Open
Abstract
Sociability entails some of the most complex behaviors processed by the central nervous system. It includes the detection, integration, and interpretation of social cues and elaboration of context-specific responses that are quintessentially species-specific. There is an ever-growing accumulation of molecular associations to autism spectrum disorders (ASD), from causative genes to endophenotypes across multiple functional layers; these however, have rarely been put in context with the opposite manifestation featured in hypersociability syndromes. Genetic copy number variations (CNVs) allow to investigate the relationships between gene dosage and its corresponding phenotypes. In particular, CNVs of the 7q11.23 locus, which manifest diametrically opposite social behaviors, offer a privileged window to look into the molecular substrates underlying the developmental trajectories of the social brain. As by definition sociability is studied in humans postnatally, the developmental fluctuations causing social impairments have thus far remained a black box. Here, we review key evidence of molecular players involved at both ends of the sociability spectrum, focusing on genetic and functional associations of neuroendocrine regulators and synaptic transmission pathways. We then proceed to propose the existence of a molecular axis centered around the paradigmatic dosage imbalances at the 7q11.23 locus, regulating networks responsible for the development of social behavior in humans and highlight the key role that neurodevelopmental models from reprogrammed pluripotent cells will play for its understanding.
Collapse
Affiliation(s)
- Alejandro López-Tobón
- Laboratory of Stem Cell Epigenetics, IEO, European Institute of Oncology, IRCCS, Milan, Italy.
- Department of Oncology and Hemato-oncology, Università degli studi di Milano, Milan, Italy.
| | - Sebastiano Trattaro
- Laboratory of Stem Cell Epigenetics, IEO, European Institute of Oncology, IRCCS, Milan, Italy.
- Department of Oncology and Hemato-oncology, Università degli studi di Milano, Milan, Italy.
| | - Giuseppe Testa
- Laboratory of Stem Cell Epigenetics, IEO, European Institute of Oncology, IRCCS, Milan, Italy.
- Department of Oncology and Hemato-oncology, Università degli studi di Milano, Milan, Italy.
- Human Technopole, Via Cristina Belgioioso 171, Milan, Italy.
| |
Collapse
|
32
|
Tan Q, Orsso CE, Deehan EC, Triador L, Field CJ, Tun HM, Han JC, Müller TD, Haqq AM. Current and emerging therapies for managing hyperphagia and obesity in Prader-Willi syndrome: A narrative review. Obes Rev 2020; 21:e12992. [PMID: 31889409 DOI: 10.1111/obr.12992] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/16/2019] [Revised: 12/07/2019] [Accepted: 12/09/2019] [Indexed: 12/16/2022]
Abstract
In early childhood, individuals with Prader-Willi syndrome (PWS) experience excess weight gain and severe hyperphagia with food compulsivity, which often leads to early onset morbid obesity. Effective treatments for appetite suppression and weight control are currently unavailable for PWS. Our aim to further understand the pathogenesis of PWS led us to carry out a comprehensive search of the current and emerging therapies for managing hyperphagia and extreme weight gain in PWS. A literature search was performed using PubMed and the following keywords: "PWS" AND "therapy" OR "[drug name]"; reference lists, pharmaceutical websites, and the ClinicalTrials.gov registry were also reviewed. Articles presenting data from current standard treatments in PWS and also clinical trials of pharmacological agents in the pipeline were selected. Current standard treatments include dietary restriction/modifications, exercise, and growth hormone replacement, which appear to have limited efficacy for appetite and weight control in patients with PWS. The long-term safety and effectiveness of bariatric surgery in PWS remains unknown. However, many promising pharmacotherapies are in development and, if approved, will bring much needed choices into the PWS pharmacological armamentarium. With the progress that is currently being made in our understanding of PWS, an effective treatment may not be far off.
Collapse
Affiliation(s)
- Qiming Tan
- Department of Pediatrics, University of Alberta, Edmonton, Alberta, Canada
| | - Camila E Orsso
- Department of Agricultural, Food and Nutritional Science, University of Alberta, Edmonton, Alberta, Canada
| | - Edward C Deehan
- Department of Agricultural, Food and Nutritional Science, University of Alberta, Edmonton, Alberta, Canada
| | - Lucila Triador
- Department of Pediatrics, University of Alberta, Edmonton, Alberta, Canada
| | - Catherine J Field
- Department of Agricultural, Food and Nutritional Science, University of Alberta, Edmonton, Alberta, Canada
| | - Hein Min Tun
- HKU-Pasteur Research Pole, School of Public Health, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, SAR, China
| | - Joan C Han
- Departments of Pediatrics and Physiology, College of Medicine, University of Tennessee Health Science Center and Children's Foundation Research Institute, Le Bonheur Children's Hospital, Memphis, Tennessee, USA
| | - Timo D Müller
- Institute for Diabetes and Obesity, Helmholtz Diabetes Center at Helmholtz Zentrum München, Neuherberg, Germany.,Department of Pharmacology and Experimental Therapy, Institute of Experimental and Clinical Pharmacology and Toxicology, Eberhard Karls University Hospitals and Clinics, Tübingen, Germany
| | - Andrea M Haqq
- Department of Pediatrics, University of Alberta, Edmonton, Alberta, Canada.,Department of Agricultural, Food and Nutritional Science, University of Alberta, Edmonton, Alberta, Canada
| |
Collapse
|
33
|
Gowda S, Seibert T, Uli N, Farrell R. Pediatric Obesity: Endocrinologic and Genetic Etiologies and Management. CURRENT CARDIOVASCULAR RISK REPORTS 2019. [DOI: 10.1007/s12170-019-0632-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
|
34
|
Horta M, Kaylor K, Feifel D, Ebner NC. Chronic oxytocin administration as a tool for investigation and treatment: A cross-disciplinary systematic review. Neurosci Biobehav Rev 2019; 108:1-23. [PMID: 31647964 DOI: 10.1016/j.neubiorev.2019.10.012] [Citation(s) in RCA: 47] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2019] [Revised: 09/10/2019] [Accepted: 10/18/2019] [Indexed: 12/18/2022]
Abstract
Oxytocin (OT) subserves various physiological, behavioral, and cognitive processes. This paired with the ability to administer OT with minimal and inconsistent side effects has spurred research to explore its therapeutic potential. Findings from single-dose studies indicate that OT administration may be beneficial, at least under certain circumstances. The state of the field, however, is less clear regarding effects from chronic OT administration, which more closely resembles long-term treatment. To address this gap, this review synthesizes existing findings on the use of chronic OT administration in animal and human work. In addition to detailing the effects of chronic OT administration across different functional domains, this review highlights factors that have contributed to mixed findings. Based on this review, a basic framework of interrelated regulatory functions sensitive to chronic OT administration is offered. The paper also identifies future research directions across different contexts, populations, and outcomes, specifically calling for more systematic and standardized research on chronic OT administration in humans to supplement and expand what is currently known from preclinical work.
Collapse
Affiliation(s)
- Marilyn Horta
- Department of Psychology, University of Florida, Gainesville, FL, USA.
| | - Kathryn Kaylor
- Department of Psychology, University of Florida, Gainesville, FL, USA
| | - David Feifel
- Department of Psychiatry, University of California, San Diego, CA, USA
| | - Natalie C Ebner
- Department of Psychology, University of Florida, Gainesville, FL, USA; Institute on Aging, Department of Aging & Geriatric Research, University of Florida, Gainesville, FL, USA
| |
Collapse
|
35
|
Bohonowych J, Miller J, McCandless SE, Strong TV. The Global Prader-Willi Syndrome Registry: Development, Launch, and Early Demographics. Genes (Basel) 2019; 10:genes10090713. [PMID: 31540108 PMCID: PMC6770999 DOI: 10.3390/genes10090713] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2019] [Revised: 09/05/2019] [Accepted: 09/09/2019] [Indexed: 12/13/2022] Open
Abstract
Advances in technologies offer new opportunities to collect and integrate data from a broad range of sources to advance the understanding of rare diseases and support the development of new treatments. Prader–Willi syndrome (PWS) is a rare, complex neurodevelopmental disorder, which has a variable and incompletely understood natural history. PWS is characterized by early failure to thrive, followed by the onset of excessive appetite (hyperphagia). Additional characteristics include multiple endocrine abnormalities, hypotonia, hypogonadism, sleep disturbances, a challenging neurobehavioral phenotype, and cognitive disability. The Foundation for Prader–Willi Research’s Global PWS Registry is one of more than twenty-five registries developed to date through the National Organization of Rare Disorders (NORD) IAMRARE Registry Program. The Registry consists of surveys covering general medical history, system-specific clinical complications, diet, medication and supplement use, as well as behavior, mental health, and social information. Information is primarily parent/caregiver entered. The platform is flexible and allows addition of new surveys, including updatable and longitudinal surveys. Launched in 2015, the PWS Registry has enrolled 1696 participants from 37 countries, with 23,550 surveys completed. This resource can improve the understanding of PWS natural history and support medical product development for PWS.
Collapse
Affiliation(s)
| | - Jennifer Miller
- Department of Pediatrics, University of Florida School of Medicine, Gainesville, FL 32611, USA.
| | - Shawn E McCandless
- Section of Genetics and Metabolism, Department of Pediatrics, University of Colorado School of Medicine and Children's Hospital Colorado, Aurora, CO, USA.
| | | |
Collapse
|
36
|
Passone CBG, Pasqualucci PL, Franco RR, Ito SS, Mattar LBF, Koiffmann CP, Soster LA, Carneiro JDA, Cabral Menezes-Filho H, Damiani D. PRADER-WILLI SYNDROME: WHAT IS THE GENERAL PEDIATRICIAN SUPPOSED TO DO? - A REVIEW. ACTA ACUST UNITED AC 2019; 36:345-352. [PMID: 30365815 PMCID: PMC6202899 DOI: 10.1590/1984-0462/;2018;36;3;00003] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2017] [Accepted: 05/12/2017] [Indexed: 11/21/2022]
Abstract
OBJECTIVE To carry out a review about Prader-Willi Syndrome based on the most recent data about the subject and to give recommendation for the general pediatricians for early diagnoses and follow-up. DATA SOURCES Scientific articles in the PubMed and SciELO databases. The research was not limited to a specific time period and included all articles in such databases. DATA SYNTHESIS The Prader-Willi Syndrome (PWS) is a rare genetic disorder resulting from the loss of imprinted gene expression within the paternal chromosome 15q11-q13. PWS is characterized by endocrine abnormalities, such as growth hormone (GH) deficiency, obesity, central adrenal insufficiency, hypothyroidism, hypogonadism and complex behavioral and intellectual difficulties. PWS individuals also may present other comorbidities, such as sleep disorders, scoliosis, constipation, dental issues and coagulation disorders. The follow-up protocol of the Children's Institute at Universidade de São Paulo is based on four main pillars: diet, exercise, recombinant human growth hormone (rhGH) therapy and behavioral and cognitive issues. The diet must include a caloric restriction of 900 kcal/day, according to the Prader-Willi Eating Pyramid and exercise plan is focused on daily aerobic exercises and postural therapy. The rhGH therapy is highly recommended by the international scientific literature and must be started as soon as the diagnostic is made. The management of behavioral issues is based on strategies to establish routine and rules. CONCLUSIONS If the general pediatrician becomes more familiar with PWS, the diagnosis and treatment will start earlier, which is essential to improve the quality of life and care for these individuals.
Collapse
|
37
|
Singh D, Wakimoto Y, Filangieri C, Pinkhasov A, Angulo M. Guanfacine Extended Release for the Reduction of Aggression, Attention-Deficit/Hyperactivity Disorder Symptoms, and Self-Injurious Behavior in Prader-Willi Syndrome-A Retrospective Cohort Study. J Child Adolesc Psychopharmacol 2019; 29:313-317. [PMID: 30724590 DOI: 10.1089/cap.2018.0102] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
Objective: To examine the role of Guanfacine Extended Release (GXR) in the management of behavioral disturbances in patients with Prader-Willi Syndrome (PWS). Methods: Twenty from a total of 27 individuals with genetically confirmed PWS, 6-26 years of age, with the following symptoms were identified: significant aggression/agitation, skin picking, and/or symptoms of attention-deficit/hyperactivity disorder (ADHD). Response to GXR for the above noted symptoms was categorized as improved, worsened, or unchanged, while assessing for side effects and tolerability. Results: Eleven of the 20 individuals reported skin-picking, 17 reported aggression/agitation, and 16 reported symptoms of ADHD. Nine (81.8%), 14 (82.3%), and 15 (93.7%) individuals showed an improvement in skin-picking, aggression/agitation, and ADHD, respectively, while on GXR treatment. Two patients with prior complaints of psychotic symptoms did not respond to GXR. Of note, no abnormal weight gain or significant adverse reaction was observed in this group, while on GXR. Conclusions: In this study, GXR demonstrated improvement in symptoms of skin picking, aggression/agitation, and ADHD in patients with PWS. GXR was not effective in reducing psychosis or agitation related to psychotic symptoms. Future studies are warranted to further establish the utility of GXR in PWS patients.
Collapse
Affiliation(s)
- Deepan Singh
- 1 Department of Behavioral Health, NYU Winthrop Hospital, Mineola, New York
| | - Yuji Wakimoto
- 2 Stony Brook University School of Medicine, Stony Brook, New York
| | - Carole Filangieri
- 1 Department of Behavioral Health, NYU Winthrop Hospital, Mineola, New York
| | - Aaron Pinkhasov
- 1 Department of Behavioral Health, NYU Winthrop Hospital, Mineola, New York
| | - Moris Angulo
- 3 Genetics-Endocrine Center, Pediatrics, NYU-Winthrop University Hospital, Mineola, New York
| |
Collapse
|
38
|
Carias KV, Wevrick R. Preclinical Testing in Translational Animal Models of Prader-Willi Syndrome: Overview and Gap Analysis. MOLECULAR THERAPY-METHODS & CLINICAL DEVELOPMENT 2019; 13:344-358. [PMID: 30989085 PMCID: PMC6447752 DOI: 10.1016/j.omtm.2019.03.001] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Prader-Willi syndrome (PWS) is a rare neurodevelopmental disorder causing endocrine, musculoskeletal, and neurological dysfunction. PWS is caused by the inactivation of contiguous genes, complicating the development of targeted therapeutics. Clinical trials are now underway in PWS, with more trials to be implemented in the next few years. PWS-like endophenotypes are recapitulated in gene-targeted mice in which the function of one or more PWS genes is disrupted. These animal models can guide priorities for clinical trials or provide information about efficacy of a compound within the context of the specific disease. We now review the current status of preclinical studies that measure the effect of therapeutics on PWS-like endophenotypes. Seven categories of therapeutics (oxytocin and related compounds, K+-ATP channel agonists, melanocortin 4 receptor agonists, incretin mimetics and/or GLP-1 receptor agonists, cannabinoids, ghrelin agents, and Caralluma fimbriata [cactus] extract) have been tested for their effect on endophenotypes in both PWS animal models and clinical trials. Many other therapeutics have been tested in clinical trials, but not preclinical models of PWS or vice versa. Fostering dialogs among investigators performing preclinical validation of animal models and those implementing clinical studies will accelerate the discovery and translation of therapies into clinical practice in PWS.
Collapse
Affiliation(s)
- K Vanessa Carias
- Department of Medical Genetics, University of Alberta, Edmonton, AB, Canada
| | - Rachel Wevrick
- Department of Medical Genetics, University of Alberta, Edmonton, AB, Canada
| |
Collapse
|
39
|
Bhargava R, Daughters KL, Rees A. Oxytocin therapy in hypopituitarism: Challenges and opportunities. Clin Endocrinol (Oxf) 2019; 90:257-264. [PMID: 30506703 DOI: 10.1111/cen.13909] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/20/2018] [Revised: 11/06/2018] [Accepted: 11/27/2018] [Indexed: 12/19/2022]
Abstract
Patients with hypopituitarism display impaired quality of life and excess morbidity and mortality, despite apparently optimal pituitary hormone replacement. Oxytocin is a neuropeptide synthesized in the anterior hypothalamus which plays an important role in controlling social and emotional behaviour, body weight and metabolism. Recent studies have suggested that a deficiency of oxytocin may be evident in patients with hypopituitarism and craniopharyngioma, and that this may be associated with deficits in cognitive empathy. Preliminary data hint at potential benefits of oxytocin therapy in improving these deficits and the accompanying metabolic disturbances that are common in these conditions. However, several challenges remain, including an incomplete understanding of the regulation and mechanisms of action of oxytocin, difficulties in accurately measuring oxytocin levels and in establishing a diagnosis of oxytocin deficiency, and a need to determine both the optimal mode of administration for oxytocin therapy and an acceptable safety profile with long-term use. This review considers the data linking oxytocin to the neuropsychological and metabolic disturbances evident in patients with craniopharyngioma and hypopituitarism, and describes the challenges that need to be overcome before replacement therapy can be considered as a therapeutic option in clinical practice.
Collapse
Affiliation(s)
- Raghav Bhargava
- Schools of Medicine (RB, DAR) and Psychology (KLD), Neuroscience and Mental Health Research Institute, Cardiff University, Cardiff, UK
| | - Katie L Daughters
- Schools of Medicine (RB, DAR) and Psychology (KLD), Neuroscience and Mental Health Research Institute, Cardiff University, Cardiff, UK
| | - Aled Rees
- Schools of Medicine (RB, DAR) and Psychology (KLD), Neuroscience and Mental Health Research Institute, Cardiff University, Cardiff, UK
| |
Collapse
|
40
|
van Iersel L, Brokke KE, Adan RAH, Bulthuis LCM, van den Akker ELT, van Santen HM. Pathophysiology and Individualized Treatment of Hypothalamic Obesity Following Craniopharyngioma and Other Suprasellar Tumors: A Systematic Review. Endocr Rev 2019; 40:193-235. [PMID: 30247642 DOI: 10.1210/er.2018-00017] [Citation(s) in RCA: 70] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/21/2018] [Accepted: 06/25/2018] [Indexed: 12/26/2022]
Abstract
The development of hypothalamic obesity (HO) following craniopharyngioma (CP) and other suprasellar tumors leads to reduced patient quality of life. No treatment algorithms are currently available for management of HO. Depending on which hypothalamic nuclei are destroyed, the pathophysiologic mechanisms and clinical symptoms that contribute to HO differ among patients. Herein, we review the contribution of the hypothalamus to the pathophysiologic mechanisms and symptoms underlying CP-associated HO. Additionally, we performed a systematic search of MEDLINE and Embase to identify all intervention studies for weight management in patients with CP or other suprasellar tumors published until September 2017. The search yielded 1866 publications, of which 40 were included. Of these 40 studies, we identified four modalities for intervention (i.e., lifestyle, dietary, pharmacotherapeutic, or surgical) within six clinical domains (i.e., psychosocial disorders, hyperphagia, sleep disturbances, decreased energy expenditure, hyperinsulinemia, and hypopituitarism). We used the findings from our systematic review, in addition to current knowledge on the pathophysiology of HO, to develop an evidence-based treatment algorithm for patients with HO caused by CP or other suprasellar tumors. Although the individual effects of the HO interventions were modest, beneficial individual effects may be achieved when the pathophysiologic background and correct clinical domain are considered. These two aspects can be combined in an individualized treatment algorithm with a stepwise approach for each clinical domain. Recently elucidated targets for HO intervention were also explored to improve future management of HO for patients with CP and other suprasellar tumors.
Collapse
Affiliation(s)
- Laura van Iersel
- Department of Pediatric Endocrinology, Wilhelmina Children's Hospital, University Medical Center Utrecht, Utrecht, Netherlands
| | - Karen E Brokke
- Medical Sciences, University Medical Center Utrecht, University of Utrecht, Utrecht, Netherlands
| | - Roger A H Adan
- Department of Translational Neuroscience, University Medical Center Utrecht, Utrecht, Netherlands.,Institute for Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Lauren C M Bulthuis
- Medical Sciences, University Medical Center Utrecht, University of Utrecht, Utrecht, Netherlands
| | - Erica L T van den Akker
- Department of Pediatric Endocrinology, Erasmus MC-Sophia Children's Hospital, Rotterdam, Netherlands
| | - Hanneke M van Santen
- Department of Pediatric Endocrinology, Wilhelmina Children's Hospital, University Medical Center Utrecht, Utrecht, Netherlands
| |
Collapse
|
41
|
Kabasakalian A, Ferretti CJ, Hollander E. Oxytocin and Prader-Willi Syndrome. Curr Top Behav Neurosci 2018; 35:529-557. [PMID: 28956320 DOI: 10.1007/7854_2017_28] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
In the chapter, we explore the relationship between the peptide hormone, oxytocin (OT), and behavioral and metabolic disturbances observed in the genetic disorder Prader-Willi Syndrome (PWS). Phenotypic and genotypic characteristics of PWS are described, as are the potential implications of an abnormal OT system with respect to neural development including the possible effects of OT dysfunction on interactions with other regulatory mediators, including neurotransmitters, neuromodulators, and hormones. The major behavioral characteristics are explored in the context of OT dysfunction, including hyperphagia, impulsivity, anxiety and emotion dysregulation, sensory processing and interoception, repetitive and restrictive behaviors, and dysfunctional social cognition. Behavioral overlaps with autistic spectrum disorders are discussed. The implications of OT dysfunction on the mechanisms of reward and satiety and their possible role in informing behavioral characteristics are also discussed. Treatment implications and future directions for investigation are considered.
Collapse
Affiliation(s)
- Anahid Kabasakalian
- Albert Einstein College of Medicine, Montefiore Medical Center, Bronx, NY, USA
| | - Casara J Ferretti
- Ferkauf Graduate School of Psychology, Yeshiva University, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Eric Hollander
- Albert Einstein College of Medicine, Montefiore Medical Center, Bronx, NY, USA.
| |
Collapse
|
42
|
Grandone A, Di Sessa A, Umano GR, Toraldo R, Miraglia Del Giudice E. New treatment modalities for obesity. Best Pract Res Clin Endocrinol Metab 2018; 32:535-549. [PMID: 30086873 DOI: 10.1016/j.beem.2018.06.007] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
The treatment of childhood obesity represents a greater challenge for pediatricians. To date, it is multidisciplinary, including behavioral, dietary, pharmacological, and surgical options. Given the limited efficacy of available treatments, scientific research on finding new solutions is very active. Several drugs comprising Metformin, Glucagon-like peptide- 1 receptor agonists, Naltrexone-bupropion, Phentermine-Topiramate, and Lorcaserin have been studied as pediatric antiobesity agents. Findings from clinical trials showed a modest but significant effect of these drugs on weight loss, but long-term studies are needed to better define their exact role. Bariatric surgery is also promising for extremely obese adolescents. Moreover, a novel approach to treat obesity might be represented by compounds inducing browning of white adipose tissue, a complex process involved in body energy homeostasis, but at present evidence in humans is lacking. We aimed to review the current knowledge regarding the available new options for pediatric obesity treatment.
Collapse
Affiliation(s)
- A Grandone
- Department of Woman, Child and of General and Specialized Surgery, University of Campania "Luigi Vanvitelli", Naples, Italy; Via L. De Crecchio, 4, 80138, Naples, Italy.
| | - A Di Sessa
- Department of Woman, Child and of General and Specialized Surgery, University of Campania "Luigi Vanvitelli", Naples, Italy; Via L. De Crecchio, 4, 80138, Naples, Italy.
| | - G R Umano
- Department of Woman, Child and of General and Specialized Surgery, University of Campania "Luigi Vanvitelli", Naples, Italy; Via L. De Crecchio, 4, 80138, Naples, Italy.
| | - R Toraldo
- Department of Woman, Child and of General and Specialized Surgery, University of Campania "Luigi Vanvitelli", Naples, Italy; Via L. De Crecchio, 4, 80138, Naples, Italy.
| | - E Miraglia Del Giudice
- Department of Woman, Child and of General and Specialized Surgery, University of Campania "Luigi Vanvitelli", Naples, Italy; Via L. De Crecchio, 4, 80138, Naples, Italy.
| |
Collapse
|
43
|
Freeman SM, Ngo J, Singh B, Masnaghetti M, Bales KL, Blevins JE. Effects of Chronic Oxytocin Administration and Diet Composition on Oxytocin and Vasopressin 1a Receptor Binding in the Rat Brain. Neuroscience 2018; 392:241-251. [PMID: 30071278 DOI: 10.1016/j.neuroscience.2018.07.037] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2018] [Revised: 07/19/2018] [Accepted: 07/20/2018] [Indexed: 12/19/2022]
Abstract
Oxytocin (OT) elicits weight loss in diet-induced obese (DIO) rodents, nonhuman primates, and humans, in part, by reducing food intake. Chronic OT administration produces more sustained weight loss in high-fat diet (HFD)-fed DIO rodents relative to chow-fed controls, but the reasons for this effect remain unclear. We hypothesized that HFD-induced obesity is associated with elevated OT receptor (OXTR) binding in brain regions where OT is known to cause decreased food intake and that this sensitized neural system is one mechanism by which OT preferentially elicits weight loss in DIO rodents. We therefore determined the impact of diet (HFD vs chow) and drug treatment (chronic OT infusion vs vehicle) on (1) OXTR binding in hindbrain and forebrain sites where OT suppresses food intake relative to control sites that express OXTR and (2) forebrain vasopressin 1a receptor (AVPR1a) density to evaluate the specificity of any OT effects. Using quantitative receptor autoradiography, we found that (1) diet composition failed to alter OXTR or AVPR1a binding; (2) chronic OT treatment produced largely global reductions in forebrain OXTR and AVPR1a binding without significantly altering hindbrain OXTR binding. These findings suggest that forebrain OXTR and AVPR1a are down-regulated in response to chronic OT treatment. Given that chronic intranasal OT may be used as a therapeutic strategy to treat obesity, future studies should consider the potential downregulatory effect that chronic treatment can have across forebrain and hindbrain nonapeptide receptors and assess the potential contribution of both receptor subtypes to the outcome measures.
Collapse
Affiliation(s)
- Sara M Freeman
- Department of Psychology, University of California, Davis, CA, USA
| | - Julie Ngo
- Department of Psychology, University of California, Davis, CA, USA
| | - Bhavdeep Singh
- Department of Psychology, University of California, Davis, CA, USA
| | | | - Karen L Bales
- Department of Psychology, University of California, Davis, CA, USA
| | - 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 98108, USA; Division of Metabolism, Endocrinology and Nutrition, Department of Medicine, University of Washington School of Medicine, Seattle, WA, USA.
| |
Collapse
|
44
|
Geets E, Meuwissen MEC, Van Hul W. Clinical, molecular genetics and therapeutic aspects of syndromic obesity. Clin Genet 2018; 95:23-40. [PMID: 29700824 DOI: 10.1111/cge.13367] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2018] [Revised: 04/05/2018] [Accepted: 04/16/2018] [Indexed: 12/13/2022]
Abstract
Obesity has become a major health problem worldwide. To date, more than 25 different syndromic forms of obesity are known in which one (monogenic) or multiple (polygenic) genes are involved. This review gives an overview of these forms and focuses more in detail on 6 syndromes: Prader Willi Syndrome and Prader Willi like phenotype, Bardet Biedl Syndrome, Alström Syndrome, Wilms tumor, Aniridia, Genitourinary malformations and mental Retardation syndrome and 16p11.2 (micro)deletions. Years of research provided plenty of information on the molecular genetics of these disorders and the obesity phenotype leading to a more individualized treatment of the symptoms, however, many questions still remain unanswered. As these obesity syndromes have different signs and symptoms in common, it makes it difficult to accurately diagnose patients which may result in inappropriate treatment of the disease. Therefore, the big challenge for clinicians and scientists is to more clearly differentiate all syndromic forms of obesity to provide conclusive genetic explanations and eventually deliver accurate genetic counseling and treatment. In addition, further delineation of the (functions of the) underlying genes with the use of array- or next-generation sequencing-based technology will be helpful to unravel the mechanisms of energy metabolism in the general population.
Collapse
Affiliation(s)
- E Geets
- Department of Medical Genetics, University of Antwerp and Antwerp University Hospital, Antwerp, Belgium
| | - M E C Meuwissen
- Department of Medical Genetics, University of Antwerp and Antwerp University Hospital, Antwerp, Belgium
| | - W Van Hul
- Department of Medical Genetics, University of Antwerp and Antwerp University Hospital, Antwerp, Belgium
| |
Collapse
|
45
|
Miller J. The potential of oxytocin for the treatment of hyperphagia in Prader-Willi Syndrome. Expert Opin Orphan Drugs 2018. [DOI: 10.1080/21678707.2018.1451326] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/17/2022]
Affiliation(s)
- Jennifer Miller
- Department of Pediatrics, Division of Endocrinology, University of Florida, Gainesville, FL, USA
| |
Collapse
|
46
|
Abstract
PURPOSE OF REVIEW PWS is a severe developmental disability for which there is no known treatment. The oxytocin system is currently a primary target for intervention. The aim of this article is to review the evidence for the efficacy of intranasal oxytocin in PWS. RECENT FINDINGS To date, there have been five clinical trials of oxytocin in PWS. Four of these studies reported that oxytocin improved behaviors. However, each of these studies suffered important limitations that likely influenced the findings. For example, one study did not include a control group. Another study did not statistically analyze the effects of oxytocin on behavior. The final two studies used study-specific measures for which psychometric properties have not been assessed. SUMMARY Because of these limitations, the most appropriate conclusion to draw from the existing studies is that there is currently no convincing evidence that intranasal oxytocin improves symptoms of PWS. However, this does not mean that oxytocin is not involved in PWS. Rather, it suggests that further work is needed to understand the nature of the PWS oxytocin abnormality.
Collapse
|
47
|
Crinò A, Fintini D, Bocchini S, Grugni G. Obesity management in Prader-Willi syndrome: current perspectives. Diabetes Metab Syndr Obes 2018; 11:579-593. [PMID: 30323638 PMCID: PMC6175547 DOI: 10.2147/dmso.s141352] [Citation(s) in RCA: 76] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Prader-Willi syndrome (PWS) is a complex multisystem disorder due to the absent expression of the paternally active genes in the PWS critical region on chromosome 15 (15q11.2-q13). The syndrome is considered the most common genetic cause of obesity, occurring in 1:10,000-1:30,000 live births. Its main characteristics include neonatal hypotonia, poor feeding, and lack of appetite in infancy, followed by weight gain, lack of satiety, and uncontrolled appetite, frequently after the age of 2-3 years. The clinical picture includes short stature, multiple endocrine abnormalities (hypogonadism, growth hormone/insulin-like growth factor-I axis dysfunction, hypothyroidism, central adrenal insufficiency), dysmorphic features, scoliosis, osteoporosis, mental retardation, and behavioral and psychiatric problems. Subjects with PWS will become severely obese unless their food intake is strictly controlled. Constant and obsessive food seeking behavior can make life very difficult for both the family and caretakers. Prevention of obesity is mandatory in these patients from the first years of life, because once obesity develops it is difficult to maintain the control of food intake. In fact, PWS subjects die prematurely from complications conventionally related to obesity, including diabetes mellitus, metabolic syndrome, sleep apnea, respiratory insufficiency, and cardiovascular disease. The mechanisms underlying hyperphagia in PWS are not completely known, and to date no drugs have proven their efficacy in controlling appetite. Consequently, dietary restriction, physical activity, and behavior management are fundamental in the prevention and management of obesity in PWS. In spite of all available therapeutic tools, however, successful weight loss and maintenance are hardly accomplished. In this context, clinical trials with new drugs have been initiated in order to find new possibilities of a therapy for obesity in these patients. The preliminary results of these studies seem to be encouraging. On the other hand, until well-proven medical treatments are available, bariatric surgery can be taken into consideration, especially in PWS patients with life-threatening comorbidities.
Collapse
Affiliation(s)
| | - Danilo Fintini
- Endocrinology Unit, Bambino Gesù Children's Hospital, Research Institute, Palidoro, Rome
| | | | - Graziano Grugni
- Division of Auxology, Italian Auxological Institute, Research Institute, Piancavallo, Verbania, Italy
| |
Collapse
|
48
|
Giel K, Zipfel S, Hallschmid M. Oxytocin and Eating Disorders: A Narrative Review on Emerging Findings and Perspectives. Curr Neuropharmacol 2018; 16:1111-1121. [PMID: 29189166 PMCID: PMC6187754 DOI: 10.2174/1570159x15666171128143158] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2017] [Revised: 08/27/2017] [Accepted: 11/23/2017] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND The hypothalamic neuropeptide oxytocin regulates reproductive behavior and mother-infant interaction, and conclusive studies in humans indicate that oxytocin is also a potent modulator of psychosocial function. Pilot experiments have yielded first evidence that this neuropeptide moreover influences eating behavior. METHODS We briefly summarize currently available studies on the involvement of the oxytocin system in the pathophysiology of eating disorders, as well as on the effects of oxytocin administration in patients with these disorders. RESULTS Brain administration of oxytocin in animals with normal weight, but also with diet-induced or genetically induced obesity, attenuates food intake and reduces body weight. In normal-weight and obese individuals, acute intranasal oxytocin delivery curbs calorie intake from main dishes and snacks. Such effects might converge with the poignant social and cognitive impact of oxytocin to also improve dysfunctional eating behavior in the therapeutic context. This assumption has received support in first studies showing that oxytocin might play a role in the disease process of anorexia nervosa. In contrast, respective experiments in patients with bulimia nervosa and binge eating disorder are still scarce. CONCLUSIONS We propose a framework of oxytocin's role and its therapeutic potential in eating disorders that aims at integrating social and metabolic aspects of its pharmacological profile, and ponder perspectives and limitations of oxytocin use in the clinical setting.
Collapse
Affiliation(s)
| | | | - Manfred Hallschmid
- Address correspondence to this author at the Department of Medical
Psychology and Behavioral Neurobiology, Faculty of Medicine, University of Tuebingen, 72076 Tuebingen, Germany; Tel/Fax: ++49-7071-29-88925, +49-7071-29-25016; E-mail:
| |
Collapse
|
49
|
Abstract
Oxytocin, a hypothalamic hormone that is secreted directly into the brain and enters the peripheral circulation through the posterior pituitary gland, regulates a range of physiologic processes, including eating behaviour and metabolism. In rodents and nonhuman primates, chronic oxytocin administration leads to sustained weight reduction by reducing food intake, increasing energy expenditure and inducing lipolysis. Oxytocin might improve glucose homeostasis, independently of its effects on weight. Clinical studies are beginning to translate these important preclinical findings to humans. This Review describes key data linking oxytocin to eating behaviour and metabolism in humans. For example, a single intranasal dose of oxytocin can reduce caloric intake, increase fat oxidation and improve insulin sensitivity in men. Furthermore, a pilot study of 8 weeks of oxytocin treatment in adults with obesity or overweight led to substantial weight loss. Together, these data support further investigation of interventions that target pathways involving oxytocin as potential therapeutics in metabolic disorders, including obesity and diabetes mellitus. Therapeutic considerations and areas for further research are also discussed.
Collapse
Affiliation(s)
- Elizabeth A Lawson
- Neuroendocrine Unit, Massachusetts General Hospital, 55 Fruit Street, Bulfinch 457 D, Boston, Massachusetts 02114, USA
| |
Collapse
|
50
|
A Review of the Safety, Efficacy and Mechanisms of Delivery of Nasal Oxytocin in Children: Therapeutic Potential for Autism and Prader-Willi Syndrome, and Recommendations for Future Research. Paediatr Drugs 2017; 19:391-410. [PMID: 28721467 DOI: 10.1007/s40272-017-0248-y] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
In this article, we conduct a comprehensive review of existing evidence for the safety and therapeutic potential of intranasal oxytocin in pediatric populations. Unique considerations for dosing and delivery of oxytocin to the nasal passageway in pediatric populations and methods to promote adherence are reviewed. Intranasal oxytocin has been administered to 261 children in three open-label studies and eight randomized controlled trials. To date, the only published results in pediatric populations have focused on autism spectrum disorder (ASD) and Prader-Willi syndrome (PWS). Results regarding efficacy for improving social impairment in ASD are equivocal, partially due to mixed methodological designs, dosing regimens, and outcome measures. At present, there is no randomized controlled evidence that oxytocin provides benefit to individuals with PWS. There is no clear evidence of a link between oxytocin administration and any specific adverse event. Adverse events have been assessed using medical interviews, open reports, checklists, and physiological assessments. Adverse events reports have been largely classified as mild (n = 93), with few moderate (n = 9) or severe (n = 3) events reported. There were 35 additional adverse events reported, without severity ratings. Severe events, hyperactivity and irritability, occurred at first administration in both placebo and oxytocin groups, and subsided subsequent to discontinuation. We note that adverse event monitoring is inconsistent and often lacking, and reporting of its relationship to the study drug is poor. Only one study reported adherence data to suggest high adherence. Recommendations are then provided for the delivery of nasal sprays to the nasal passageway, monitoring, and reporting of efficacy, safety, and adherence for oxytocin nasal spray trials in pediatric populations.
Collapse
|