1
|
Torbahn G, Lischka J, Brown T, Ells LJ, Kelly AS, Wabitsch M, Weghuber D. Anti-Obesity Medication in the Management of Children and Adolescents With Obesity: Recent Developments and Research Gaps. Clin Endocrinol (Oxf) 2024. [PMID: 39257303 DOI: 10.1111/cen.15133] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/17/2024] [Revised: 07/20/2024] [Accepted: 08/21/2024] [Indexed: 09/12/2024]
Abstract
BACKGROUND Paediatric obesity is a global public health concern. While in most countries the incidence keeps rising, the need for effective and long-term management for children and adolescents living with this chronic, relapsing disease is pressing. Health behaviour and lifestyle treatment (HBLT) is recommended as first-line treatment. METHODS Narrative review. RESULTS A new generation of recently approved anti-obesity medications (AOM) now has the potential to fill the gap between limited effects on body mass index (BMI) by HBLT alone and large effects by metabolic and bariatric surgery in adolescents with obesity aged 12 years and older. While, for semaglutide and phentermine/topiramate, effectiveness is substantial with relevant, but mostly mild to moderate adverse events, there is a gap in evidence regarding long-term effects and safety, effects on outcomes beyond BMI reduction and data for certain groups of patients, such as children < 12 years and minority groups. When integrating AOM treatment into national healthcare systems it should be offered as part of a comprehensive patient-centred approach. CONCLUSION This article summarizes recent AOM developments, integration into paediatric obesity management, and identifies research gaps.
Collapse
Affiliation(s)
- Gabriel Torbahn
- Department of Pediatrics, Paracelsus Medical University, Klinikum Nürnberg, Universitätsklinik der Paracelsus Medizinischen Privatuniversität Nürnberg, Nuremberg, Germany
- Department of Pediatrics, Paracelsus Medical University, Salzburg, Austria
- Department of Pediatrics, Obesity Research Unit, Paracelsus Medical University, Salzburg, Austria
| | - Julia Lischka
- Department of Pediatrics, Paracelsus Medical University, Salzburg, Austria
- Department of Pediatrics, Obesity Research Unit, Paracelsus Medical University, Salzburg, Austria
| | - Tamara Brown
- School of Health, Obesity Institute, Leeds Beckett University, Leeds, UK
| | - Louisa J Ells
- School of Health, Obesity Institute, Leeds Beckett University, Leeds, UK
| | - Aaron S Kelly
- Department of Pediatrics, Center for Pediatric Obesity Medicine, University of Minnesota Medical School Minneapolis, Minneapolis, Minnesota, USA
| | - Martin Wabitsch
- Department of Pediatrics and Adolescent Medicine, Division of Pediatric Endocrinology and Diabetes, Center for Rare Endocrine Diseases, University of Ulm, Ulm, Germany
| | - Daniel Weghuber
- Department of Pediatrics, Paracelsus Medical University, Salzburg, Austria
- Department of Pediatrics, Obesity Research Unit, Paracelsus Medical University, Salzburg, Austria
| |
Collapse
|
2
|
Manglani K, Anika NN, Patel D, Jhaveri S, Avanthika C, Sudan S, Alimohamed Z, Tiwari K. Correlation of Leptin in Patients With Type 2 Diabetes Mellitus. Cureus 2024; 16:e57667. [PMID: 38707092 PMCID: PMC11070180 DOI: 10.7759/cureus.57667] [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] [Accepted: 04/05/2024] [Indexed: 05/07/2024] Open
Abstract
The exponential increase in diabetes mellitus (DM) poses serious public health concerns. In this review, we focus on the role of leptin in type 2 DM. The peripheral actions of leptin consist of upregulating proinflammatory cytokines which play an important role in the pathogenesis of type 2 DM and insulin resistance. Moreover, leptin is known to inhibit insulin secretion and plays a significant role in insulin resistance in obesity and type 2 DM. A literature search was conducted on Medline, Cochrane, Embase, and Google Scholar for relevant articles published until December 2023. The following search strings and Medical Subject Headings (MeSH terms) were used: "Diabetes Mellitus," "Leptin," "NPY," and "Biomarker." This article aims to discuss the physiology of leptin in type 2 DM, its glucoregulatory actions, its relationship with appetite, the impact that various lifestyle modifications can have on leptin levels, and, finally, explore leptin as a potential target for various treatment strategies.
Collapse
Affiliation(s)
- Kajol Manglani
- Internal Medicine, MedStar Washington Hospital Center, Washington, USA
| | | | - Dhriti Patel
- Medicine and Surgery, B.J. Medical College and Civil Hospital, Ahmedabad, IND
| | - Sharan Jhaveri
- Medicine and Surgery, Smt. Nathiba Hargovandas Lakhmichand Municipal Medical College, Gujarat University, Ahmedabad, IND
| | - Chaithanya Avanthika
- Pediatrics, Icahn School of Medicine at Mount Sinai, Elmhurst Hospital Center, New York, USA
- Medicine and Surgery, Karnataka Institute of Medical Sciences, Hubballi, IND
| | - Sourav Sudan
- Internal Medicine, Government Medical College, Rajouri, Rajouri, IND
| | - Zainab Alimohamed
- Division of Research & Academic Affairs, Larkin Health System, South Miami, USA
| | - Kripa Tiwari
- Internal Medicine, Maimonides Medical Center, New York, USA
| |
Collapse
|
3
|
Mohammed I, Haris B, Al-Barazenji T, Vasudeva D, Tomei S, Al Azwani I, Dauleh H, Shehzad S, Chirayath S, Mohamadsalih G, Petrovski G, Khalifa A, Love DR, Al-Shafai M, Hussain K. Understanding the Genetics of Early-Onset Obesity in a Cohort of Children From Qatar. J Clin Endocrinol Metab 2023; 108:3201-3213. [PMID: 37329217 PMCID: PMC10655519 DOI: 10.1210/clinem/dgad366] [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: 02/16/2023] [Revised: 06/10/2023] [Accepted: 06/14/2023] [Indexed: 06/18/2023]
Abstract
CONTEXT Monogenic obesity is a rare form of obesity due to pathogenic variants in genes implicated in the leptin-melanocortin signaling pathway and accounts for around 5% of severe early-onset obesity. Mutations in the genes encoding the MC4R, leptin, and leptin receptor are commonly reported in various populations to cause monogenic obesity. Determining the genetic cause has important clinical benefits as novel therapeutic interventions are now available for some forms of monogenic obesity. OBJECTIVE To unravel the genetic causes of early-onset obesity in the population of Qatar. METHODS In total, 243 patients with early-onset obesity (above the 95% percentile) and age of onset below 10 years were screened for monogenic obesity variants using a targeted gene panel, consisting of 52 obesity-related genes. RESULTS Thirty rare variants potentially associated with obesity were identified in 36 of 243 (14.8%) probands in 15 candidate genes (LEP, LEPR, POMC, MC3R, MC4R, MRAP2, SH2B1, BDNF, NTRK2, DYRK1B, SIM1, GNAS, ADCY3, RAI1, and BBS2). Twenty-three of the variants identified were novel to this study and the rest, 7 variants, were previously reported in literature. Variants in MC4R were the most common cause of obesity in our cohort (19%) and the c.485C>T p.T162I variant was the most frequent MC4R variant seen in 5 patients. CONCLUSION We identified likely pathogenic/pathogenic variants that seem to explain the phenotype of around 14.8% of our cases. Variants in the MC4R gene are the commonest cause of early-onset obesity in our population. Our study represents the largest monogenic obesity cohort in the Middle East and revealed novel obesity variants in this understudied population. Functional studies will be required to elucidate the molecular mechanism of their pathogenicity.
Collapse
Affiliation(s)
- Idris Mohammed
- College of Health & Life Sciences, Hamad Bin Khalifa University, PO Box 34110, Doha, Qatar
- Division of Endocrinology, Department of Pediatric Medicine, Sidra Medicine, PO Box 26999, Doha, Qatar
| | - Basma Haris
- Division of Endocrinology, Department of Pediatric Medicine, Sidra Medicine, PO Box 26999, Doha, Qatar
| | - Tara Al-Barazenji
- Department of Biomedical Sciences, College of Health Sciences, QU Health, Qatar University, PO Box 2713, Doha, Qatar
| | - Dhanya Vasudeva
- Division of Endocrinology, Department of Pediatric Medicine, Sidra Medicine, PO Box 26999, Doha, Qatar
| | - Sara Tomei
- Omics Core, Integrated Genomic Services, Research Branch, Sidra Medicine, PO Box 26999, Doha, Qatar
| | - Iman Al Azwani
- Omics Core, Integrated Genomic Services, Research Branch, Sidra Medicine, PO Box 26999, Doha, Qatar
| | - Hajar Dauleh
- Division of Endocrinology, Department of Pediatric Medicine, Sidra Medicine, PO Box 26999, Doha, Qatar
| | - Saira Shehzad
- Division of Endocrinology, Department of Pediatric Medicine, Sidra Medicine, PO Box 26999, Doha, Qatar
| | - Shiga Chirayath
- Division of Endocrinology, Department of Pediatric Medicine, Sidra Medicine, PO Box 26999, Doha, Qatar
| | - Ghassan Mohamadsalih
- Division of Endocrinology, Department of Pediatric Medicine, Sidra Medicine, PO Box 26999, Doha, Qatar
| | - Goran Petrovski
- Division of Endocrinology, Department of Pediatric Medicine, Sidra Medicine, PO Box 26999, Doha, Qatar
| | - Amel Khalifa
- Division of Endocrinology, Department of Pediatric Medicine, Sidra Medicine, PO Box 26999, Doha, Qatar
| | - Donald R Love
- Division of Genetic Pathology, Department of Pathology, Sidra Medicine, PO Box 26999, Doha, Qatar
| | - Mashael Al-Shafai
- Department of Biomedical Sciences, College of Health Sciences, QU Health, Qatar University, PO Box 2713, Doha, Qatar
- Biomedical Research Center, Qatar University, PO Box 2713, Doha, Qatar
| | - Khalid Hussain
- Division of Endocrinology, Department of Pediatric Medicine, Sidra Medicine, PO Box 26999, Doha, Qatar
| |
Collapse
|
4
|
Mazen IH, El-Gammal MA, Elaidy AA, Anwar GM, Ashaat EA, Abdel-Ghafar SF, Abdel-Hamid MS. Congenital leptin and leptin receptor deficiencies in nine new families: identification of six novel variants and review of literature. Mol Genet Genomics 2023; 298:919-929. [PMID: 37140700 DOI: 10.1007/s00438-023-02025-1] [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: 12/08/2022] [Accepted: 04/24/2023] [Indexed: 05/05/2023]
Abstract
Early childhood obesity is a real public health problem worldwide. Identifying the etiologies, especially treatable and preventable causes, can direct health professionals toward proper management. Measurement of serum leptin levels is helpful in the diagnosis of congenital leptin and leptin receptor deficiencies which are considered important rare causes of early childhood obesity. The main aim of this study was to investigate the frequency of LEP, LEPR, and MC4R gene variants among a cohort of Egyptian patients with severe early onset obesity. The current cross-sectional study included 30 children who developed obesity during the first year of life with BMI > 2SD (for age and sex). The studied patients were subjected to full medical history taking, anthropometric measurements, serum leptin and insulin assays, and genetic testing of LEP, LEPR and MC4R. Disease causing variants in LEP and LEPR were identified in 10/30 patients with a detection rate of 30%. Eight different homozygous variants (two pathogenic, three likely pathogenic, and three variants of uncertain significant) were identified in the two genes, including six previously unreported LEPR variants. Of them, a new frameshift variant in LEPR gene (c.1045delT, p.S349Lfs*22) was recurrent in two unrelated families and seems to have a founder effect in our population. In conclusion, we reported ten new patients with leptin and leptin receptor deficiencies and identified six novel LEPR variants expanding the mutational spectrum of this rare disorder. Furthermore, the diagnosis of these patients helped us in genetic counseling and patients' managements specially with the availability of drugs for LEP and LEPR deficiencies.
Collapse
Affiliation(s)
- Inas H Mazen
- Clinical Genetics Department, Human Genetics and Genome Research Institute, National Research Centre, Cairo, Egypt
| | - Mona A El-Gammal
- Clinical Genetics Department, Human Genetics and Genome Research Institute, National Research Centre, Cairo, Egypt
| | - Aya A Elaidy
- Clinical Genetics Department, Human Genetics and Genome Research Institute, National Research Centre, Cairo, Egypt
| | - Ghada M Anwar
- Department of Pediatrics, Faculty of Medicine, Cairo University, Cairo, Egypt
| | - Engy A Ashaat
- Clinical Genetics Department, Human Genetics and Genome Research Institute, National Research Centre, Cairo, Egypt
| | - Sherif F Abdel-Ghafar
- Medical Molecular Genetics Department, Human Genetics and Genome Research Institute, National Research Centre, Eltahrir Street, Dokki, Cairo, 12311, Egypt
| | - Mohamed S Abdel-Hamid
- Medical Molecular Genetics Department, Human Genetics and Genome Research Institute, National Research Centre, Eltahrir Street, Dokki, Cairo, 12311, Egypt.
| |
Collapse
|
5
|
Postnatal Leptin Levels Correlate with Breast Milk Leptin Content in Infants Born before 32 Weeks Gestation. Nutrients 2022; 14:nu14245224. [PMID: 36558383 PMCID: PMC9782260 DOI: 10.3390/nu14245224] [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: 10/31/2022] [Revised: 12/01/2022] [Accepted: 12/06/2022] [Indexed: 12/13/2022] Open
Abstract
Perinatal leptin deficiency and reduced intake of mother’s milk may contribute to the development of childhood obesity. Preterm infants have reduced leptin production, and they are at heightened risk of neonatal leptin deficiency. Because fresh human milk contains significantly more leptin than donor milk, we used a cross-over design to determine if blood leptin levels in maternal milk-fed preterm infants fall during conversion to donor human milk. Infants born between 22 0/7 and 31 6/7 weeks gestation on exclusive maternal milk feedings were enrolled into a 21-day cross-over trial. On days 1−7 and 15−21, infants were fed maternal milk, and on days 8−14, infants were fed donor milk. On day 1, study infants had a mean postmenstrual age of 33 weeks. Plasma leptin correlated with milk leptin, and leptin levels in maternal milk far exceed the leptin levels of donor milk. Plasma leptin did not increase during donor milk administration, but it did following resumption of maternal milk (p < 0.05). In this crossover trial, preterm infant blood leptin levels correlated with milk leptin content. This suggests that preterm infants can enterally absorb leptin from human milk, and leptin-rich breast milk may be a targeted therapy for the prevention of obesity.
Collapse
|
6
|
Hebebrand J, Hildebrandt T, Schlögl H, Seitz J, Denecke S, Vieira D, Gradl-Dietsch G, Peters T, Antel J, Lau D, Fulton S. The role of hypoleptinemia in the psychological and behavioral adaptation to starvation: implications for anorexia nervosa. Neurosci Biobehav Rev 2022; 141:104807. [PMID: 35931221 DOI: 10.1016/j.neubiorev.2022.104807] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2022] [Revised: 06/11/2022] [Accepted: 07/31/2022] [Indexed: 12/17/2022]
Abstract
This narrative review aims to pinpoint mental and behavioral effects of starvation, which may be triggered by hypoleptinemia and as such may be amenable to treatment with leptin receptor agonists. The reduced leptin secretion results from the continuous loss of fat mass, thus initiating a graded triggering of diverse starvation related adaptive functions. In light of leptin receptors located in several peripheral tissues and many brain regions adaptations may extend beyond those of the hypothalamus-pituitary-end organ-axes. We focus on gastrointestinal tract and reward system as relevant examples of peripheral and central effects of leptin. Despite its association with extreme obesity, congenital leptin deficiency with its many parallels to a state of starvation allows the elucidation of mental symptoms amenable to treatment with exogenous leptin in both ob/ob mice and humans with this autosomal recessive disorder. For starvation induced behavioral changes with an intact leptin signaling we particularly focus on rodent models for which proof of concept has been provided for the causative role of hypoleptinemia. For humans, we highlight the major cognitive, emotional and behavioral findings of the Minnesota Starvation Experiment to contrast them with results obtained upon a lesser degree of caloric restriction. Evidence for hypoleptinemia induced mental changes also stems from findings obtained in lipodystrophies. In light of the recently reported beneficial cognitive, emotional and behavioral effects of metreleptin-administration in anorexia nervosa we discuss potential implications for the treatment of this eating disorder. We postulate that leptin has profound psychopharmacological effects in the state of starvation.
Collapse
Affiliation(s)
- Johannes Hebebrand
- Department of Child and Adolescent Psychiatry, Psychosomatics and Psychotherapy, University Hospital Essen, University of Duisburg-Essen, Wickenburgstr. 21, 45134 Essen, Germany
| | - Tom Hildebrandt
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY 10029 USA
| | - Haiko Schlögl
- Department of Endocrinology, Nephrology, Rheumatology, Division of Endocrinology, University Hospital Leipzig, Liebigstr. 20, 04103 Leipzig, Germany; Helmholtz Institute for Metabolic, Obesity and Vascular Research (HI-MAG) of the Helmholtz Zentrum München at the University of Leipzig and University Hospital Leipzig, Philipp-Rosenthal-Str. 27, 04103 Leipzig, Germany
| | - Jochen Seitz
- Department of Child and Adolescent Psychiatry, Psychosomatics and Psychotherapy, RWTH University Hospital Aachen, Germany
| | - Saskia Denecke
- Department of Child and Adolescent Psychiatry, Psychosomatics and Psychotherapy, University Hospital Essen, University of Duisburg-Essen, Wickenburgstr. 21, 45134 Essen, Germany
| | - Diana Vieira
- Department of Child and Adolescent Psychiatry, Psychosomatics and Psychotherapy, University Hospital Essen, University of Duisburg-Essen, Wickenburgstr. 21, 45134 Essen, Germany
| | - Gertraud Gradl-Dietsch
- Department of Child and Adolescent Psychiatry, Psychosomatics and Psychotherapy, University Hospital Essen, University of Duisburg-Essen, Wickenburgstr. 21, 45134 Essen, Germany
| | - Triinu Peters
- Department of Child and Adolescent Psychiatry, Psychosomatics and Psychotherapy, University Hospital Essen, University of Duisburg-Essen, Wickenburgstr. 21, 45134 Essen, Germany
| | - Jochen Antel
- Department of Child and Adolescent Psychiatry, Psychosomatics and Psychotherapy, University Hospital Essen, University of Duisburg-Essen, Wickenburgstr. 21, 45134 Essen, Germany
| | - David Lau
- Department of Nutrition, Neuroscience - University of Montreal & CRCHUM, Montréal QC H3T1J4, Canada
| | - Stephanie Fulton
- Department of Nutrition, Neuroscience - University of Montreal & CRCHUM, Montréal QC H3T1J4, Canada
| |
Collapse
|
7
|
Parvanehvar S, Tehranian N, Kazemnejad A, Mozdarani H, Hedayati M. Maternal serum adiponectin and leptin levels and their gene expression, postpartum body mass index, and breastfeeding duration based on mode of delivery: A prospective study. J Obstet Gynaecol Res 2022; 48:1768-1774. [PMID: 35384157 DOI: 10.1111/jog.15251] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2021] [Revised: 02/19/2022] [Accepted: 03/21/2022] [Indexed: 12/27/2022]
Abstract
BACKGROUND Adipokines are involved in inflammatory responses, associated with body mass index whose concentrations may change in response to inflammatory conditions, including surgery and delivery. We examined adiponectin and leptin levels and their gene expression at birth, body mass index, and breastfeeding duration at 24 months postpartum according to mode of delivery. METHODS In this study, 90 normal pregnant women were investigated. Blood samples were collected after delivery. Serum levels and gene expression of adiponectin and leptin were evaluated. Body mass index and breastfeeding duration were calculated at 24 months postpartum. Data were analyzed using SPSS-16 and p < 0.05 was considered as significant. RESULTS Serum leptin level was significantly higher in vaginal delivery than in cesarean section (p = 0.033). No significant difference was found between two groups regarding adiponectin level and gene expression, while leptin gene expression was significantly higher in cesarean (p = 0.005). Postpartum body mass index did not differ between the two groups (p = 0.14). On the other hand, postpartum body mass index was significantly higher than the equivalent prepregnancy index in both groups (p < 0.001) and was associated with serum leptin and adiponectin in vaginal delivery (r = 0.46, p = 0.001, and r = -0.3, p = 0.04, respectively). The duration of breastfeeding was longer in vaginal delivery (p = 0.008). CONCLUSION Cesarean section was associated with lower maternal leptin levels and shorter breast-feeding duration compared to vaginal delivery. Leptin gene expression was significantly higher in cesarean section than in vaginal delivery. Postpartum body mass index, adiponectin level, and gene expression did not differ between the two groups.
Collapse
Affiliation(s)
- Simin Parvanehvar
- Department of Reproductive Health and Midwifery, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran.,Department of Midwifery, Varamin-Pishva Branch, Islamic Azad University, Varamin, Iran
| | - Najmeh Tehranian
- Department of Reproductive Health and Midwifery, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Anoshirvan Kazemnejad
- Department of Biostatistics, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Hossein Mozdarani
- Department of Medical Genetics -Medical Cytogenetics, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Mehdi Hedayati
- Cellular and Molecular Endocrine Research Center, Research Institute for Endocrine Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
8
|
Apperley LJ, Blackburn J, Erlandson-Parry K, Gait L, Laing P, Senniappan S. Childhood obesity: A review of current and future management options. Clin Endocrinol (Oxf) 2022; 96:288-301. [PMID: 34750858 DOI: 10.1111/cen.14625] [Citation(s) in RCA: 25] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/19/2021] [Revised: 10/12/2021] [Accepted: 10/13/2021] [Indexed: 02/02/2023]
Abstract
Obesity is becoming increasingly prevalent in paediatric populations worldwide. In addition to increasing prevalence, the severity of obesity is also continuing to rise. Taken together, these findings demonstrate a worrying trend and highlight one of the most significant challenges to public health. Childhood obesity affects multiple organs in the body and is associated with both significant morbidity and ultimately premature mortality. The prevalence of complications associated with obesity, including dyslipidaemia, hypertension, fatty liver disease and psychosocial complications are becoming increasingly prevalent within the paediatric populations. Treatment guidelines currently focus on intervention with lifestyle and behavioural modifications, with pharmacotherapy and surgery reserved for patients who are refractory to such treatment. Research into adult obesity has established pharmacological novel therapies, which have been approved and established in clinical practice; however, the research and implementation of such therapies in paediatric populations have been lagging behind. Despite the relative lack of widespread research in comparison to the adult population, newer therapies are being trialled, which should allow a greater availability of treatment options for childhood obesity in the future. This review summarizes the current evidence for the management of obesity in terms of medical and surgical options. Both future therapeutic agents and those which cause weight loss but have an alternative indication are also included and discussed as part of the review. The review summarizes the most recent research for each intervention and demonstrates the potential efficacy and limitations of each treatment option.
Collapse
Affiliation(s)
- Louise J Apperley
- Department of Paediatric Endocrinology, Alder Hey Children's Hospital, Liverpool, UK
| | - James Blackburn
- Centre for Endocrinology, William Harvey Research Institute, Barts and the London School of Medicine, Queen Mary University of London, London, UK
| | | | - Lucy Gait
- Department of Paediatric Clinical Psychology, Alder Hey Children's Hospital, Liverpool, UK
| | - Peter Laing
- Department of Paediatric Endocrinology, Alder Hey Children's Hospital, Liverpool, UK
| | - Senthil Senniappan
- Department of Paediatric Endocrinology, Alder Hey Children's Hospital, Liverpool, UK
| |
Collapse
|
9
|
Fırat SN, Onay H. Early-onset severe obesity due to homozygous p.R105W (c313C> T) mutation in leptin gene in Turkish siblings: Two cases reports. Obes Res Clin Pract 2021; 15:600-603. [PMID: 34802983 DOI: 10.1016/j.orcp.2021.11.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/16/2021] [Revised: 10/23/2021] [Accepted: 11/08/2021] [Indexed: 11/17/2022]
Abstract
Congenital leptin deficiency (CLD) is a rare cause of monogenic form obesity due to homozygous or compound heterozygous mutations in the LEP gene. To date, nine pathogenic mutations have been reported. In this study, we present are; an 18-year-old morbidly obese girl and a 14-year-old obese brother, both with homozygous mutation in the LEP gene [p.R105W (c313C> T)] and their data after three years of recombinant leptin treatment. To date, few cases of CLD have been reported in the literature. The cases reported here were siblings who were not diagnosed despite presentation at the clinic due to obesity in childhood, and diagnosis was delayed until adolescence. Clinicians need to consider CLD, a monogenic form of obesity in children with early severe obesity onset, especially if they are the child of a consanguineous marriage.
Collapse
Affiliation(s)
- Sevde Nur Fırat
- Department of Endocrinology and Metabolism, University of Health Sciences Ankara Training and Research Hospital, Ankara, Turkey.
| | - Hüseyin Onay
- Department of Medical Genetics, MULTIGEN Genetic Diseases Evaluation Center, İzmir, Turkey.
| |
Collapse
|
10
|
Dhurandhar NV, Petersen KS, Webster C. Key Causes and Contributors of Obesity: A Perspective. Nurs Clin North Am 2021; 56:449-464. [PMID: 34749887 DOI: 10.1016/j.cnur.2021.07.007] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Obesity is a disease with several potential causes and contributors. This article provides a focused overview of key known causes of obesity and factors that contribute to obesity. Obesity ultimately results from impaired energy storage mechanisms, such as dysregulation of hunger, satiety, digestion, fat storage, and metabolic rate. In addition, myriad contributors promote its expression, including dietary factors, sleep quality and duration, psychological health and well-being, and tobacco cessation, among others. This article concludes with a discussion of the clinical relevance of causes and contributors in obesity prevention and treatment, which is paramount to providing effective, individualized clinical management.
Collapse
Affiliation(s)
- Nikhil V Dhurandhar
- Department of Nutritional Sciences, Texas Tech University, 1301 Akron Avenue, P.O. Box 41270, Lubbock, TX 79409, USA.
| | - Kristina S Petersen
- Department of Nutritional Sciences, Texas Tech University, 1301 Akron Avenue, P.O. Box 41270, Lubbock, TX 79409, USA
| | - Chelsi Webster
- Department of Nutritional Sciences, Texas Tech University, 1301 Akron Avenue, P.O. Box 41270, Lubbock, TX 79409, USA
| |
Collapse
|
11
|
Abstract
Leptin for over 25 years has been a central theme in the study of appetite, obesity, and starvation. As the major site of leptin production is peripheral, and the site of action of greatest interest is the hypothalamus, how leptin accesses the central nervous system (CNS) and crosses the blood-brain barrier (BBB) has been of great interest. We review here the ongoing research that addresses fundamental questions such as the sites of leptin resistances in obesity and other conditions, the causes of resistances and their relations to one another, the three barrier sites of entry into the CNS, why recent studies using suprapharmacological doses cannot address these questions but give insight into nonsaturable entry of leptin into the CNS, and how that might be useful in using leptin therapeutically. The current status of the controversy of whether the short form of the leptin receptor acts as the BBB leptin transporter and how obesity may transform leptin transport is reviewed. Review of these and other topics summarizes in a new appreciation of what leptin may have actually evolved to do and what physiological role leptin resistance may play. © 2021 American Physiological Society. Compr Physiol 11:1-19, 2021.
Collapse
Affiliation(s)
- William A Banks
- Geriatric Research Education and Clinical Center, Veterans Affairs Puget Sound Health Care System, Seattle, Washington, USA.,Division of Gerontology and Geriatric Medicine, Department of Medicine, University of Washington School of Medicine, Seattle, Washington, USA
| |
Collapse
|
12
|
Salum KCR, Rolando JDM, Zembrzuski VM, Carneiro JRI, Mello CB, Maya-Monteiro CM, Bozza PT, Kohlrausch FB, da Fonseca ACP. When Leptin Is Not There: A Review of What Nonsyndromic Monogenic Obesity Cases Tell Us and the Benefits of Exogenous Leptin. Front Endocrinol (Lausanne) 2021; 12:722441. [PMID: 34504472 PMCID: PMC8421737 DOI: 10.3389/fendo.2021.722441] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/08/2021] [Accepted: 07/30/2021] [Indexed: 01/01/2023] Open
Abstract
Obesity is a pandemic condition of complex etiology, resulting from the increasing exposition to obesogenic environmental factors combined with genetic susceptibility. In the past two decades, advances in genetic research identified variants of the leptin-melanocortin pathway coding for genes, which are related to the potentiation of satiety and hunger, immune system, and fertility. Here, we review cases of congenital leptin deficiency and the possible beneficial effects of leptin replacement therapy. In summary, the cases presented here show clinical phenotypes of disrupted bodily energy homeostasis, biochemical and hormonal disorders, and abnormal immune response. Some phenotypes can be partially reversed by exogenous administration of leptin. With this review, we aim to contribute to the understanding of leptin gene mutations as targets for obesity diagnostics and treatment strategies.
Collapse
Affiliation(s)
- Kaio Cezar Rodrigues Salum
- Human Genetic Laboratory, Department of General Biology, Institute of Biology, Federal Fluminense University, Niterói, Brazil
- Human Genetics Laboratory, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro, Brazil
| | - Jônatas de Mendonça Rolando
- Human Genetic Laboratory, Department of General Biology, Institute of Biology, Federal Fluminense University, Niterói, Brazil
| | | | - João Regis Ivar Carneiro
- Clementino Fraga Filho University Hospital, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Cicero Brasileiro Mello
- Human Genetic Laboratory, Department of General Biology, Institute of Biology, Federal Fluminense University, Niterói, Brazil
| | | | - Patrícia Torres Bozza
- Laboratory of Immunopharmacology, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro, Brazil
| | - Fabiana Barzotto Kohlrausch
- Human Genetic Laboratory, Department of General Biology, Institute of Biology, Federal Fluminense University, Niterói, Brazil
| | - Ana Carolina Proença da Fonseca
- Human Genetics Laboratory, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro, Brazil
- Laboratory of Immunopharmacology, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro, Brazil
| |
Collapse
|
13
|
de Leeuw AJM, Oude Luttikhuis MAM, Wellen AC, Müller C, Calkhoven CF. Obesity and its impact on COVID-19. J Mol Med (Berl) 2021; 99:899-915. [PMID: 33824998 PMCID: PMC8023779 DOI: 10.1007/s00109-021-02072-4] [Citation(s) in RCA: 45] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2021] [Revised: 03/22/2021] [Accepted: 03/30/2021] [Indexed: 01/08/2023]
Abstract
The severe acute respiratory syndrome-coronavirus-2 (SARS-CoV-2) pandemic has proven a challenge to healthcare systems since its first appearance in late 2019. The global spread and devastating effects of coronavirus disease 2019 (COVID-19) on patients have resulted in countless studies on risk factors and disease progression. Overweight and obesity emerged as one of the major risk factors for developing severe COVID-19. Here we review the biology of coronavirus infections in relation to obesity. In particular, we review literature about the impact of adiposity-related systemic inflammation on the COVID-19 disease severity, involving cytokine, chemokine, leptin, and growth hormone signaling, and we discuss the involvement of hyperactivation of the renin-angiotensin-aldosterone system (RAAS). Due to the sheer number of publications on COVID-19, we cannot be completed, and therefore, we apologize for all the publications that we do not cite.
Collapse
Affiliation(s)
- Angélica J M de Leeuw
- University Medical Center Groningen (UMCG), University of Groningen, 9700, AD, Groningen, The Netherlands
| | | | - Annemarijn C Wellen
- University Medical Center Groningen (UMCG), University of Groningen, 9700, AD, Groningen, The Netherlands
| | - Christine Müller
- European Research Institute for the Biology of Ageing (ERIBA), University Medical Center Groningen, University of Groningen, 9700, AD, Groningen, The Netherlands
| | - Cornelis F Calkhoven
- European Research Institute for the Biology of Ageing (ERIBA), University Medical Center Groningen, University of Groningen, 9700, AD, Groningen, The Netherlands.
| |
Collapse
|
14
|
Study of LEP, MRAP2 and POMC genes as potential causes of severe obesity in Brazilian patients. Eat Weight Disord 2021; 26:1399-1408. [PMID: 32578125 DOI: 10.1007/s40519-020-00946-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/15/2020] [Accepted: 06/11/2020] [Indexed: 10/24/2022] Open
Abstract
PURPOSE Monogenic forms of obesity are caused by single-gene variants which affect the energy homeostasis by increasing food intake and decreasing energy expenditure. Most of these variants result from disruption of the leptin-melanocortin signaling, which can cause severe early-onset obesity and hyperphagia. These mutation have been identified in genes encoding essential proteins to this pathway, including leptin (LEP), melanocortin 2 receptor accessory proteins 2 (MRAP2) and proopiomelanocortin (POMC). We aimed to investigate the prevalence of LEP, MRAP2 and POMC rare variants in severely obese adults, who developed obesity during childhood. To the best of our knowledge, this is the first study screening rare variants of these genes in patients from Brazil. METHODS A total of 122 Brazilian severely obese patients (BMI ≥ 35 kg/m2) were screened for the coding regions of LEP, MRAP2 and POMC by Sanger sequencing. All patients are candidates to the bariatric surgery. Clinical characteristics were described in patients with novel and/or potentially pathogenic variants. RESULTS Sixteen different variants were identified in these genes, of which two were novel. Among them, one previous variant with potentially deleterious effect in MRAP2 (p.Arg125Cys) was found. In addition, two heterozygous mutations in POMC (p.Phe87Leu and p.Arg90Leu) were predicted to impair protein function. We also observed a POMC homozygous 9 bp insertion (p.Gly99_Ala100insSerSerGly) in three patients. No pathogenic variant was observed in LEP. CONCLUSION Our study described for the first time the prevalence of rare potentially pathogenic MRAP2 and POMC variants in a cohort of Brazilian severely obese adults. LEVEL OF EVIDENCE Level V, cross-sectional descriptive study.
Collapse
|
15
|
Guglielmi V, Colangeli L, D’Adamo M, Sbraccia P. Susceptibility and Severity of Viral Infections in Obesity: Lessons from Influenza to COVID-19. Does Leptin Play a Role? Int J Mol Sci 2021; 22:ijms22063183. [PMID: 33804765 PMCID: PMC8003928 DOI: 10.3390/ijms22063183] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2021] [Revised: 03/05/2021] [Accepted: 03/18/2021] [Indexed: 12/13/2022] Open
Abstract
The recent pandemic Sars-CoV2 infection and studies on previous influenza epidemic have drawn attention to the association between the obesity and infectious diseases susceptibility and worse outcome. Metabolic complications, nutritional aspects, physical inactivity, and a chronic unbalance in the hormonal and adipocytokine microenvironment are major determinants in the severity of viral infections in obesity. By these pleiotropic mechanisms obesity impairs immune surveillance and the higher leptin concentrations produced by adipose tissue and that characterize obesity substantially contribute to such immune response dysregulation. Indeed, leptin not only controls energy balance and body weight, but also plays a regulatory role in the interplay between energy metabolism and immune system. Since leptin receptor is expressed throughout the immune system, leptin may exert effects on cells of both innate and adaptive immune system. Chronic inflammatory states due to metabolic (i.e., obesity) as well as infectious diseases increase leptin concentrations and consequently lead to leptin resistance further fueling inflammation. Multiple factors, including inflammation and ER stress, contribute to leptin resistance. Thus, if leptin is recognized as one of the adipokines responsible for the low grade inflammation found in obesity, on the other hand, impairments of leptin signaling due to leptin resistance appear to blunt the immunologic effects of leptin and possibly contribute to impaired vaccine-induced immune responses. However, many aspects concerning leptin interactions with inflammation and immune system as well as the therapeutical approaches to overcome leptin resistance and reduced vaccine effectiveness in obesity remain a challenge for future research.
Collapse
|
16
|
Abstract
A healthy nutritional state is required for all aspects of reproduction and is signaled by the adipokine leptin. Leptin acts in a relatively narrow concentration range: too much or too little will compromise fertility. The leptin signal timing is important to prepubertal development in both sexes. In the brain, leptin acts on ventral premammillary neurons which signal kisspeptin (Kiss1) neurons to stimulate gonadotropin releasing hormone (GnRH) neurons. Suppression of Kiss1 neurons occurs when agouti-related peptide neurons are activated by reduced leptin, because leptin normally suppresses these orexigenic neurons. In the pituitary, leptin stimulates production of GnRH receptors (GnRHRs) and follicle-stimulating hormone at midcycle, by activating pathways that derepress actions of the messenger ribonucleic acid translational regulatory protein Musashi. In females, rising estrogen stimulates a rise in serum leptin, which peaks at midcycle, synchronizing with nocturnal luteinizing hormone pulses. The normal range of serum leptin levels (10-20 ng/mL) along with gonadotropins and growth factors promote ovarian granulosa and theca cell functions and oocyte maturation. In males, the prepubertal rise in leptin promotes testicular development. However, a decline in leptin levels in prepubertal boys reflects inhibition of leptin secretion by rising androgens. In adult males, leptin levels are 10% to 50% of those in females, and high leptin inhibits testicular function. The obesity epidemic has elucidated leptin resistance pathways, with too much leptin in either sex leading to infertility. Under conditions of balanced nutrition, however, the secretion of leptin is timed and regulated within a narrow level range that optimizes its trophic effects.
Collapse
Affiliation(s)
- Gwen V Childs
- Department of Neurobiology and Developmental Sciences, University of Arkansas for Medical Sciences, Little Rock, Arkansas
- Correspondence: Gwen V. Childs, PhD, University of Arkansas for Medical Sciences, Little Rock, AR, USA. E-mail:
| | - Angela K Odle
- Department of Neurobiology and Developmental Sciences, University of Arkansas for Medical Sciences, Little Rock, Arkansas
| | - Melanie C MacNicol
- Department of Neurobiology and Developmental Sciences, University of Arkansas for Medical Sciences, Little Rock, Arkansas
| | - Angus M MacNicol
- Department of Neurobiology and Developmental Sciences, University of Arkansas for Medical Sciences, Little Rock, Arkansas
| |
Collapse
|
17
|
Lu SC, Akanji AO. Leptin, Obesity, and Hypertension: A Review of Pathogenetic Mechanisms. Metab Syndr Relat Disord 2020; 18:399-405. [PMID: 32876506 DOI: 10.1089/met.2020.0065] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
The adipokine leptin is expressed at higher concentrations in obese subjects, who also incidentally have a higher prevalence of hypertension. The pathogenesis of this obesity-related hypertension is controversial and is believed to be related to many factors including increased sympathetic activity, abnormalities of the renin-angiotensin system, sodium retention, and an endotheliopathy acting independently or in concert with increased circulating leptin. This review discusses the potential mechanisms through which changes in leptin signal transduction pathways in tissues with the leptin receptor, especially the hypothalamus, mediate the pathogenetic relationships between obesity and hypertension. The hypothesis is explored that leptin effects on blood pressure (BP) are meditated by the downstream effects of hypothalamic leptin signaling and ultimately result in activation of specific melanocortin receptors located on sympathetic neurons in the spinal cord. The physiological consequences of this sympathetic activation of the heart and kidney are activation of the renin-angiotensin system, sodium retention and circulatory expansion and finally, elevated BP. This sequence of events has been elegantly demonstrated with leptin infusion and gene knockout studies in animal models but has not been convincingly reproducibly confirmed in humans. Further studies in human subjects on the specific roles of hypothalamic leptin in essential hypertension are indicated as elucidation of the signaling pathways should provide better understanding of the role of weight loss in BP control and afford an additional mechanism for pharmacologic control of BP in adults and children at risk of cardiovascular disease.
Collapse
Affiliation(s)
- Song Chi Lu
- Department of Medical Sciences, Frank H. Netter School of Medicine, NH-MED, Quinnipiac University, Hamden, Connecticut, USA
| | - Abayomi O Akanji
- Department of Medical Sciences, Frank H. Netter School of Medicine, NH-MED, Quinnipiac University, Hamden, Connecticut, USA
| |
Collapse
|
18
|
Short-term metreleptin treatment of patients with anorexia nervosa: rapid on-set of beneficial cognitive, emotional, and behavioral effects. Transl Psychiatry 2020; 10:303. [PMID: 32855384 PMCID: PMC7453199 DOI: 10.1038/s41398-020-00977-1] [Citation(s) in RCA: 58] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/14/2020] [Revised: 07/17/2020] [Accepted: 07/29/2020] [Indexed: 12/17/2022] Open
Abstract
To examine the hypothesis that normalization of low circulating leptin levels in patients with anorexia nervosa ameliorates hyperactivity, three seriously ill females with hyperactivity were treated off-label with metreleptin (recombinant human leptin) for up to 14 days. Drive for activity, repetitive thoughts of food, inner restlessness, and weight phobia decreased in two patients. Surprisingly, depression improved rapidly in all patients. No serious adverse events occurred. Due to obvious limitations of uncontrolled case series, placebo-controlled clinical trials are mandatory to confirm the observed rapid onset of beneficial effects. Our findings suggest an important role of hypoleptinemia in the mental and behavioral phenotype of anorexia nervosa.
Collapse
|
19
|
Boutari C, Mantzoros CS. Adiponectin and leptin in the diagnosis and therapy of NAFLD. Metabolism 2020; 103:154028. [PMID: 31785257 DOI: 10.1016/j.metabol.2019.154028] [Citation(s) in RCA: 61] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/21/2019] [Accepted: 11/22/2019] [Indexed: 12/11/2022]
Affiliation(s)
- Chrysoula Boutari
- Department of Internal Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, 330 Brookline Avenue, Boston, MA 02215, USA
| | - Christos S Mantzoros
- Department of Internal Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, 330 Brookline Avenue, Boston, MA 02215, USA; Section of Endocrinology, Diabetes and Metabolism, Boston VA Healthcare System, 150 South Huntington Avenue, Boston, MA 02130, USA.
| |
Collapse
|
20
|
ElSaeed G, Mousa N, El-Mougy F, Hafez M, Khodeera S, Alhelbawy M, Fouda E, Elsheikh S, ElKaffas R, Eldeeb S, Elsharkawy M. Monogenic leptin deficiency in early childhood obesity. Pediatr Obes 2020; 15:e12574. [PMID: 31483094 DOI: 10.1111/ijpo.12574] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/09/2019] [Revised: 07/28/2019] [Accepted: 08/05/2019] [Indexed: 12/23/2022]
Abstract
BACKGROUND Early childhood obesity is a public health problem worldwide. It affects different aspects of physical and mental child's health. Identifying the etiologies, especially treatable and preventable causes, can direct health professionals toward proper management. Analysis of serum leptin levels and leptin gene mutations is a rapid and easy step toward the diagnosis of congenital leptin deficiency that is considered an important cause in early childhood obesity. OBJECTIVES The aim of this study was to diagnose monogenic leptin deficiency in Egyptian children presenting with early onset obesity (EOO). METHODS The current cross-sectional study included 80 children who developed obesity during the first year of life with BMI > 2 SD (for age and sex). The studied population was subjected to history taking, auxological assessment, serum leptin assay, and leptin gene sequencing. RESULTS Ten cases had leptin deficiency (12.5%), while 18 cases showed elevated leptin levels (22.5%). Leptin gene variants in the coding region were identified in 30% of the leptin-deficient group: two novel homozygous disease-causing variants (c.104 T > G and c.34 delC) and another previously reported homozygous pathogenic variant (c.313C > T). CONCLUSION Leptin deficiency is considered a significant cause of monogenic obesity in Egyptian children with early-onset obesity as the diagnosis of these patients would be a perfect target for recombinant leptin therapy.
Collapse
Affiliation(s)
- Gehan ElSaeed
- Clinical Pathology Department, Faculty of Medicine, Monoufia University, Al Minufiyah, Egypt
| | - Noha Mousa
- Pediatrics Department, Faculty of Medicine, Cairo University, Cairo, Egypt
| | - Fatma El-Mougy
- Clinical and Chemical Pathology Department, Faculty of Medicine, Cairo University, Cairo, Egypt
| | - Mona Hafez
- Pediatrics Department, Faculty of Medicine, Cairo University, Cairo, Egypt
| | - Seham Khodeera
- Clinical Pathology Department, Faculty of Medicine, Monoufia University, Al Minufiyah, Egypt
| | - Mohamed Alhelbawy
- Clinical Pathology Department, Faculty of Medicine, Monoufia University, Al Minufiyah, Egypt
| | - Engy Fouda
- Pediatrics Department, Faculty of Medicine, Cairo University, Cairo, Egypt
| | - Suzan Elsheikh
- Clinical and Chemical Pathology Department, Faculty of Medicine, Cairo University, Cairo, Egypt
| | - Rasha ElKaffas
- Clinical and Chemical Pathology Department, Faculty of Medicine, Cairo University, Cairo, Egypt
| | - Sara Eldeeb
- Clinical Pathology Department, Faculty of Medicine, Monoufia University, Al Minufiyah, Egypt
| | - Marwa Elsharkawy
- Clinical and Chemical Pathology Department, Faculty of Medicine, Cairo University, Cairo, Egypt
| |
Collapse
|
21
|
Samodien E, Pheiffer C, Erasmus M, Mabasa L, Louw J, Johnson R. Diet-induced DNA methylation within the hypothalamic arcuate nucleus and dysregulated leptin and insulin signaling in the pathophysiology of obesity. Food Sci Nutr 2019; 7:3131-3145. [PMID: 31660128 PMCID: PMC6804761 DOI: 10.1002/fsn3.1169] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2019] [Revised: 07/18/2019] [Accepted: 07/24/2019] [Indexed: 12/24/2022] Open
Abstract
Obesity rates continue to rise in an unprecedented manner in what could be the most rapid population‐scale shift in human phenotype ever to occur. Increased consumption of unhealthy, calorie‐dense foods, coupled with sedentary lifestyles, is the main factor contributing to a positive energy balance and the development of obesity. Leptin and insulin are key hormones implicated in pathogenesis of this disorder and are crucial for controlling whole‐body energy homeostasis. Their respective function is mediated by the counterbalance of anorexigenic and orexigenic neurons located within the hypothalamic arcuate nucleus. Dysregulation of leptin and insulin signaling pathways within this brain region may contribute not only to the development of obesity, but also systemically affect the peripheral organs, thereby manifesting as metabolic diseases. Although the exact mechanisms detailing how these hypothalamic nuclei contribute to disease pathology are still unclear, increasing evidence suggests that altered DNA methylation may be involved. This review evaluates animal studies that have demonstrated diet‐induced DNA methylation changes in genes that regulate energy homeostasis within the arcuate nucleus, and elucidates possible mechanisms causing hypothalamic leptin and insulin resistance leading to the development of obesity and metabolic diseases.
Collapse
Affiliation(s)
- Ebrahim Samodien
- Biomedical Research and Innovation Platform South African Medical Research Council. Tygerberg Cape Town South Africa
| | - Carmen Pheiffer
- Biomedical Research and Innovation Platform South African Medical Research Council. Tygerberg Cape Town South Africa.,Department of Medical Physiology Stellenbosch University Tygerberg South Africa
| | - Melisse Erasmus
- Biomedical Research and Innovation Platform South African Medical Research Council. Tygerberg Cape Town South Africa.,Department of Medical Physiology Stellenbosch University Tygerberg South Africa
| | - Lawrence Mabasa
- Biomedical Research and Innovation Platform South African Medical Research Council. Tygerberg Cape Town South Africa
| | - Johan Louw
- Biomedical Research and Innovation Platform South African Medical Research Council. Tygerberg Cape Town South Africa.,Department of Biochemistry and Microbiology University of Zululand KwaDlangezwa South Africa
| | - Rabia Johnson
- Biomedical Research and Innovation Platform South African Medical Research Council. Tygerberg Cape Town South Africa.,Department of Medical Physiology Stellenbosch University Tygerberg South Africa
| |
Collapse
|
22
|
Liu Y, Yang W, Hua R, Huang Y, Zhang X. Pathogenic mutations in two Chinese patients exhibiting severe early-onset obesity. SCIENCE CHINA-LIFE SCIENCES 2019; 62:1554-1556. [PMID: 31102178 DOI: 10.1007/s11427-018-9443-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/10/2018] [Revised: 01/05/2019] [Indexed: 11/25/2022]
Affiliation(s)
- Yanshan Liu
- McKusick-Zhang Center for Genetic Medicine, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences School of Basic Medicine Peking Union Medical College, Beijing, 100005, China.
| | - Wei Yang
- Capital Institute of Pediatrics, Beijing, 100020, China
| | - Rui Hua
- Department of Obstetrics and Gynecology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Yingzhi Huang
- McKusick-Zhang Center for Genetic Medicine, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences School of Basic Medicine Peking Union Medical College, Beijing, 100005, China
| | - Xue Zhang
- McKusick-Zhang Center for Genetic Medicine, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences School of Basic Medicine Peking Union Medical College, Beijing, 100005, China.
| |
Collapse
|
23
|
Hebebrand J, Milos G, Wabitsch M, Teufel M, Führer D, Bühlmeier J, Libuda L, Ludwig C, Antel J. Clinical Trials Required to Assess Potential Benefits and Side Effects of Treatment of Patients With Anorexia Nervosa With Recombinant Human Leptin. Front Psychol 2019; 10:769. [PMID: 31156489 PMCID: PMC6533856 DOI: 10.3389/fpsyg.2019.00769] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2019] [Accepted: 03/20/2019] [Indexed: 12/16/2022] Open
Abstract
The core phenotype of anorexia nervosa (AN) comprises the age and stage dependent intertwining of both its primary and secondary (i.e., starvation induced) somatic and mental symptoms. Hypoleptinemia acts as a key trigger for the adaptation to starvation by affecting diverse brain regions including the reward system and by induction of alterations of the hypothalamus-pituitary-“target-organ” axes, e.g., resulting in amenorrhea as a characteristic symptom of AN. Particularly, the rat model activity-based anorexia (ABA) convincingly demonstrates the pivotal role of hypoleptinemia in the development of starvation-induced hyperactivity. STAT3 signaling in dopaminergic neurons in the ventral tegmental area (VTA) plays a crucial role in the transmission of the leptin signal in ABA. In patients with AN, an inverted U-shaped relationship has been observed between their serum leptin levels and physical activity. Albeit obese and therewith of a very different phenotype, humans diagnosed with rare congenital leptin deficiency have starvation like symptoms including hypothalamic amenorrhea in females. Over the past 20 years, such patients have been successfully treated with recombinant human (rh) leptin (metreleptin) within a compassionate use program. The extreme hunger of these patients subsides within hours upon initiation of treatment; substantial weight loss and menarche in females ensue after medium term treatment. In contrast, metreleptin had little effect in patients with multifactorial obesity. Small clinical trials have been conducted for hypothalamic amenorrhea and to increase bone mineral density, in which metreleptin proved beneficial. Up to now, metreleptin has not yet been used to treat patients with AN. Metreleptin has been approved by the FDA under strict regulations solely for the treatment of generalized lipodystrophy. The recent approval by the EMA may offer, for the first time, the possibility to treat extremely hyperactive patients with AN off-label. Furthermore, a potential dissection of hypoleptinemia-induced AN symptoms from the primary cognitions and behaviors of these patients could ensue. Accordingly, the aim of this article is to review the current state of the art of leptin in relation to AN to provide the theoretical basis for the initiation of clinical trials for treatment of this eating disorder.
Collapse
Affiliation(s)
- Johannes Hebebrand
- Department of Child and Adolescent Psychiatry, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Gabriella Milos
- Department of Consultation-Liaison Psychiatry and Psychosomatic Medicine, University Hospital of Zürich, Zurich, Switzerland
| | - Martin Wabitsch
- Division of Paediatric Endocrinology and Diabetes, Department of Paediatrics and Adolescent Medicine, Ulm University Hospital, Ulm, Germany
| | - Martin Teufel
- Department of Psychosomatic Medicine, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Dagmar Führer
- Department of Endocrinology and Metabolism, Medical Center and Central Laboratory, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Judith Bühlmeier
- Department of Child and Adolescent Psychiatry, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Lars Libuda
- Department of Child and Adolescent Psychiatry, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Christine Ludwig
- Department of Child and Adolescent Psychiatry, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Jochen Antel
- Department of Child and Adolescent Psychiatry, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| |
Collapse
|
24
|
Yupanqui-Lozno H, Bastarrachea RA, Yupanqui-Velazco ME, Alvarez-Jaramillo M, Medina-Méndez E, Giraldo-Peña AP, Arias-Serrano A, Torres-Forero C, Garcia-Ordoñez AM, Mastronardi CA, Restrepo CM, Rodriguez-Ayala E, Nava-Gonzalez EJ, Arcos-Burgos M, Kent JW, Cole SA, Licinio J, Celis-Regalado LG. Congenital Leptin Deficiency and Leptin Gene Missense Mutation Found in Two Colombian Sisters with Severe Obesity. Genes (Basel) 2019; 10:genes10050342. [PMID: 31067764 PMCID: PMC6562380 DOI: 10.3390/genes10050342] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2019] [Revised: 04/23/2019] [Accepted: 04/30/2019] [Indexed: 12/16/2022] Open
Abstract
Background: Congenital leptin deficiency is a recessive genetic disorder associated with severe early-onset obesity. It is caused by mutations in the leptin (LEP) gene, which encodes the protein product leptin. These mutations may cause nonsense-mediated mRNA decay, defective secretion or the phenomenon of biologically inactive leptin, but typically lead to an absence of circulating leptin, resulting in a rare type of monogenic extreme obesity with intense hyperphagia, and serious metabolic abnormalities. Methods: We present two severely obese sisters from Colombia, members of the same lineal consanguinity. Their serum leptin was measured by MicroELISA. DNA sequencing was performed on MiSeq equipment (Illumina) of a next-generation sequencing (NGS) panel involving genes related to severe obesity, including LEP. Results: Direct sequencing of the coding region of LEP gene in the sisters revealed a novel homozygous missense mutation in exon 3 [NM_002303.3], C350G>T [p.C117F]. Detailed information and clinical measurements of these sisters were also collected. Their serum leptin levels were undetectable despite their markedly elevated fat mass. Conclusions: The mutation of LEP, absence of detectable leptin, and the severe obesity found in these sisters provide the first evidence of monogenic leptin deficiency reported in the continents of North and South America.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | - Claudio A Mastronardi
- NeuRos, Instituto de Medicina Traslacional, Escuela de Medicina y Ciencias de la Salud, Universidad del Rosario, Bogotá 111711, Colombia.
| | - Carlos M Restrepo
- GeniURos, CIGGUR, Instituto de Medicina Traslacional, Escuela de Medicina y Ciencias de la Salud, Universidad del Rosario, Bogotá 111711, Colombia.
| | | | | | - Mauricio Arcos-Burgos
- Grupo de Investigación en Psiquiatría (GIPSI), Instituto de Investigaciones Médicas. Facultad de Medicina, Universidad de Antioquia, Medellín 050010, Colombia.
| | - Jack W Kent
- Texas Biomedical Research Institute, San Antonio, TX 78227, USA.
| | - Shelley A Cole
- Texas Biomedical Research Institute, San Antonio, TX 78227, USA.
| | - Julio Licinio
- SUNY Upstate Medical University, Syracuse, NY 13210, USA.
| | | |
Collapse
|
25
|
Bramante CT, Raatz S, Bomberg EM, Oberle MM, Ryder JR. Cardiovascular Risks and Benefits of Medications Used for Weight Loss. Front Endocrinol (Lausanne) 2019. [PMID: 32010059 DOI: 10.3389/fendo.2019.00883/bibtex] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 05/15/2023] Open
Abstract
Obesity is a complex disease influenced by many neurohormonal pathways which regulate body weight toward homeostasis. Presently, the disease of obesity effects over a billion individuals worldwide with scalable treatment options in dire need. Pharmacologic interventions for obesity have been developed to help promote weight loss in individuals with obesity. This area is rapidly developing and will only exponentially increase to serve the demand for persons with obesity seeking biologically orientated solutions to treat their disease. Therefore, understanding the cardiovascular risks and benefits of these weight loss medications is of particularly importance due to obesities strong association with cardiovascular (CV) disease risk. Moreover, past experiences with pharmacotherapy agents with weight loss properties have demonstrated an association with adverse CV outcomes, leading to market removal, in most cases and concerns over using similar medications. To better understand the CV risks and benefits pharmacotherapy agents used for weight loss, this review will discuss medications which are FDA-approved for weight loss, as well as medications commonly used off-label for this indication. The goal is to provide an overview of the risks and benefits many of these medications can offer to help guide clinical decision making and patient education.
Collapse
Affiliation(s)
- Carolyn T Bramante
- Center for Pediatric Obesity Medicine, University of Minnesota Medical School, Minneapolis, MN, United States
- Division of General Internal Medicine, University of Minnesota Medical School, Minneapolis, MN, United States
| | - Sarah Raatz
- Center for Pediatric Obesity Medicine, University of Minnesota Medical School, Minneapolis, MN, United States
| | - Eric M Bomberg
- Center for Pediatric Obesity Medicine, University of Minnesota Medical School, Minneapolis, MN, United States
- Division of Pediatric Endocrinology, University of Minnesota Medical School, Minneapolis, MN, United States
| | - Megan M Oberle
- Center for Pediatric Obesity Medicine, University of Minnesota Medical School, Minneapolis, MN, United States
- Division of Pediatric Endocrinology, University of Minnesota Medical School, Minneapolis, MN, United States
| | - Justin R Ryder
- Center for Pediatric Obesity Medicine, University of Minnesota Medical School, Minneapolis, MN, United States
- Division of Pediatric Epidemiology and Clinical Research, University of Minnesota Medical School, Minneapolis, MN, United States
| |
Collapse
|
26
|
|
27
|
Bramante CT, Raatz S, Bomberg EM, Oberle MM, Ryder JR. Cardiovascular Risks and Benefits of Medications Used for Weight Loss. Front Endocrinol (Lausanne) 2019; 10:883. [PMID: 32010059 PMCID: PMC6974445 DOI: 10.3389/fendo.2019.00883] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/16/2019] [Accepted: 12/03/2019] [Indexed: 12/12/2022] Open
Abstract
Obesity is a complex disease influenced by many neurohormonal pathways which regulate body weight toward homeostasis. Presently, the disease of obesity effects over a billion individuals worldwide with scalable treatment options in dire need. Pharmacologic interventions for obesity have been developed to help promote weight loss in individuals with obesity. This area is rapidly developing and will only exponentially increase to serve the demand for persons with obesity seeking biologically orientated solutions to treat their disease. Therefore, understanding the cardiovascular risks and benefits of these weight loss medications is of particularly importance due to obesities strong association with cardiovascular (CV) disease risk. Moreover, past experiences with pharmacotherapy agents with weight loss properties have demonstrated an association with adverse CV outcomes, leading to market removal, in most cases and concerns over using similar medications. To better understand the CV risks and benefits pharmacotherapy agents used for weight loss, this review will discuss medications which are FDA-approved for weight loss, as well as medications commonly used off-label for this indication. The goal is to provide an overview of the risks and benefits many of these medications can offer to help guide clinical decision making and patient education.
Collapse
Affiliation(s)
- Carolyn T. Bramante
- Center for Pediatric Obesity Medicine, University of Minnesota Medical School, Minneapolis, MN, United States
- Division of General Internal Medicine, University of Minnesota Medical School, Minneapolis, MN, United States
- *Correspondence: Carolyn T. Bramante
| | - Sarah Raatz
- Center for Pediatric Obesity Medicine, University of Minnesota Medical School, Minneapolis, MN, United States
| | - Eric M. Bomberg
- Center for Pediatric Obesity Medicine, University of Minnesota Medical School, Minneapolis, MN, United States
- Division of Pediatric Endocrinology, University of Minnesota Medical School, Minneapolis, MN, United States
| | - Megan M. Oberle
- Center for Pediatric Obesity Medicine, University of Minnesota Medical School, Minneapolis, MN, United States
- Division of Pediatric Endocrinology, University of Minnesota Medical School, Minneapolis, MN, United States
| | - Justin R. Ryder
- Center for Pediatric Obesity Medicine, University of Minnesota Medical School, Minneapolis, MN, United States
- Division of Pediatric Epidemiology and Clinical Research, University of Minnesota Medical School, Minneapolis, MN, United States
| |
Collapse
|
28
|
Fedewa MV, Hathaway ED, Ward-Ritacco CL, Williams TD, Dobbs WC. The Effect of Chronic Exercise Training on Leptin: A Systematic Review and Meta-Analysis of Randomized Controlled Trials. Sports Med 2018; 48:1437-1450. [PMID: 29582381 DOI: 10.1007/s40279-018-0897-1] [Citation(s) in RCA: 66] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
BACKGROUND Leptin is a hormone associated with satiety, lipid oxidation, energy expenditure, and energy homeostasis. To date, the current body of research examining the effect of chronic exercise training on leptin has yielded inconsistent results. OBJECTIVE The purpose of this meta-analysis was to provide a quantitative estimate of the magnitude of change in leptin levels following participation in exercise interventions lasting ≥ 2 weeks. METHODS All studies included were peer-reviewed and published in English. To be included, studies randomized human participants to an exercise training group or non-exercise comparison group for an exercise training intervention. Leptin levels were measured at baseline, during, and/or after completion of the exercise training program. Random-effects models were used to aggregate a mean effect size (ES) and 95% confidence intervals (CIs), and identify potential moderators. RESULTS Seventy-two randomized controlled trials met the inclusion criteria and resulted in 107 effects (n = 3826). The mean ES of 0.24 (95% CI 0.16-0.32, p < 0.0001) indicated a decrease in leptin following an exercise training program. A decrease in %Fat (β = - 0.07, p < 0.01) was associated with a decrease in leptin after accounting for the type of control group (β = - 0.38, p < 0.0001) used in each study. CONCLUSION These results suggest that engaging in chronic exercise training (≥ 2 weeks) is associated with a decrease in leptin levels for individuals regardless of age and sex. However, a greater decrease in leptin occurred with a decreased percentage of body fat.
Collapse
Affiliation(s)
- Michael V Fedewa
- Department of Kinesiology, The University of Alabama, 2003 Moore Hall, Box 870312, Tuscaloosa, AL, 35487-0231, USA.
| | - Elizabeth D Hathaway
- Department of Health and Human Performance, The University of Tennessee at Chattanooga, Chattanooga, TN, USA
| | | | - Tyler D Williams
- Department of Kinesiology, Samford University, Birmingham, AL, USA
| | - Ward C Dobbs
- Department of Kinesiology, The University of Alabama, 2003 Moore Hall, Box 870312, Tuscaloosa, AL, 35487-0231, USA
| |
Collapse
|
29
|
Nunziata A, Funcke JB, Borck G, von Schnurbein J, Brandt S, Lennerz B, Moepps B, Gierschik P, Fischer-Posovszky P, Wabitsch M. Functional and Phenotypic Characteristics of Human Leptin Receptor Mutations. J Endocr Soc 2018; 3:27-41. [PMID: 30560226 PMCID: PMC6293235 DOI: 10.1210/js.2018-00123] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/26/2018] [Accepted: 09/12/2018] [Indexed: 11/25/2022] Open
Abstract
Several case series of extreme early-onset obesity due to mutations in the human leptin receptor (LEPR) gene have been reported. In this review we summarize published functional and phenotypic data on mutations in the human LEPR gene causing severe early-onset obesity. Additionally, we included data on six new cases from our obesity center. Literature research was performed using PubMed and OMIM. Functional relevance of mutations was estimated based on reported functional analysis, mutation size, and location, as well as phenotypic characteristics of affected patients. We identified 57 cases with 38 distinct LEPR mutations. We found severe early-onset obesity, hyperphagia, and hypogonadotropic hypogonadism as cardinal features of a complete loss of LEPR function. Other features, for example, metabolic disorders and recurring infections, were variable in manifestation. Obesity degree or other manifestations did not aggregate by genotype. Few patients underwent bariatric surgery with variable success. Most mutations occurred in the fibronectin III and cytokine receptor homology II domains, whereas none was found in cytoplasmic domain. In silico data were available for 25 mutations and in vitro data were available for four mutations, revealing residual activity in one case. By assessing provided information on the clinical phenotype, functional analysis, and character of the 38 mutations, we assume residual LEPR activity for five additional mutations. Functional in vitro analysis is necessary to confirm this assumption.
Collapse
Affiliation(s)
- Adriana Nunziata
- Division of Pediatric Endocrinology and Diabetes, Department of Pediatrics and Adolescent Medicine, University Medical Center Ulm, Ulm, Germany
| | - Jan-Bernd Funcke
- Division of Pediatric Endocrinology and Diabetes, Department of Pediatrics and Adolescent Medicine, University Medical Center Ulm, Ulm, Germany
| | - Guntram Borck
- Institute of Human Genetics, University of Ulm, Ulm, Germany
| | - Julia von Schnurbein
- Division of Pediatric Endocrinology and Diabetes, Department of Pediatrics and Adolescent Medicine, University Medical Center Ulm, Ulm, Germany
| | - Stephanie Brandt
- Division of Pediatric Endocrinology and Diabetes, Department of Pediatrics and Adolescent Medicine, University Medical Center Ulm, Ulm, Germany
| | - Belinda Lennerz
- Division of Endocrinology, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Barbara Moepps
- Institute of Pharmacology and Toxicology, University of Ulm, Ulm, Germany
| | - Peter Gierschik
- Institute of Pharmacology and Toxicology, University of Ulm, Ulm, Germany
| | - Pamela Fischer-Posovszky
- Division of Pediatric Endocrinology and Diabetes, Department of Pediatrics and Adolescent Medicine, University Medical Center Ulm, Ulm, Germany
| | - Martin Wabitsch
- Division of Pediatric Endocrinology and Diabetes, Department of Pediatrics and Adolescent Medicine, University Medical Center Ulm, Ulm, Germany
| |
Collapse
|
30
|
Saeed S, Arslan M, Froguel P. Genetics of Obesity in Consanguineous Populations: Toward Precision Medicine and the Discovery of Novel Obesity Genes. Obesity (Silver Spring) 2018; 26:474-484. [PMID: 29464904 DOI: 10.1002/oby.22064] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/24/2016] [Revised: 09/05/2017] [Accepted: 09/30/2017] [Indexed: 12/31/2022]
Abstract
OBJECTIVE Consanguinity has been instrumental in the elucidation of many Mendelian genetic diseases. Here, the unique advantage of consanguineous populations was considered in the quest for genes causing obesity. METHODS PubMed was searched for articles relevant to consanguinity and obesity published between 1995 and 2016. Some earlier articles of interest were also consulted. RESULTS Although obesity is the most heritable disorder, even in outbred populations, only 2% to 5% of severe obesity cases have so far been proven to be caused by single gene mutations. In some highly consanguineous populations, a remarkably higher proportion of obesity cases because of known and novel monogenic variants has been identified (up to 30%). CONCLUSIONS Combining the power conferred by consanguinity with current large-capacity sequencing techniques should bring new genetic factors and molecular mechanisms to the fore, unveiling a large part of the yet-elusive neurohumoral circuitry involved in the regulation of energy homeostasis and appetite. Importantly, the undertaking of such initiatives is destined to unfold novel targets for the development of precision medicine relevant to different forms of obesity.
Collapse
Affiliation(s)
- Sadia Saeed
- Department of Genomics of Common Disease, Imperial College London, London, UK
- CNRS, Pasteur Institute of Lille, University of Lille, Lille, France
| | - Muhammad Arslan
- Centre for Research in Molecular Medicine, The University of Lahore, Lahore, Pakistan
- Department of Biological Sciences, Forman Christian College, Lahore, Pakistan
| | - Philippe Froguel
- Department of Genomics of Common Disease, Imperial College London, London, UK
- CNRS, Pasteur Institute of Lille, University of Lille, Lille, France
| |
Collapse
|
31
|
da Fonseca ACP, Mastronardi C, Johar A, Arcos-Burgos M, Paz-Filho G. Genetics of non-syndromic childhood obesity and the use of high-throughput DNA sequencing technologies. J Diabetes Complications 2017; 31:1549-1561. [PMID: 28735903 DOI: 10.1016/j.jdiacomp.2017.04.026] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/23/2017] [Revised: 04/18/2017] [Accepted: 04/19/2017] [Indexed: 12/12/2022]
Abstract
BACKGROUND Childhood obesity is a serious public health problem associated with the development of several chronic diseases, such as type 2 diabetes mellitus, dyslipidemia, and hypertension. The elevated prevalence of obesity is mostly due to inadequate diet and lifestyle, but it is also influenced by genetic factors. OBJECTIVES To review recent advances in the field of the genetics of obesity. We summarize the list of genes associated with the rare non-syndromic forms of obesity, and explain their function. Furthermore, we discuss the technologies that are available for the genetic diagnosis of obesity. RESULTS Several studies reported that single gene variants cause Mendelian forms of obesity, determined by mutations of major effect in single genes. Rare, non-syndromic forms of obesity are a result of loss-of-function mutations in genes that act on the development and function of the hypothalamus or the leptin-melanocortin pathway. These variants disrupt enzymes and receptors that play a role in energy homeostasis, resulting in severe early-onset obesity and endocrine dysfunctions. Different approaches and technologies have been used to understand the genetic background of obesity. Currently, whole genome and whole exome sequencing are important diagnostic tools to identify new genes and variants associated with severe obesity, but other approaches are also useful at individual or population levels, such as linkage analysis, candidate gene sequencing, chromosomal microarray analysis, and genome-wide association studies. CONCLUSIONS The understanding of the genetic causes of obesity and the usefulness and limitations of the genetic diagnostic approaches can contribute to the development of new personalized therapeutic targets against obesity.
Collapse
Affiliation(s)
| | - Claudio Mastronardi
- Institute of Translational Medicine, Universidad del Rosario, Bogota, Colombia
| | - Angad Johar
- Department of Genome Sciences, John Curtin School of Medical Research, The Australian National University, Australia.
| | | | - Gilberto Paz-Filho
- Department of Genome Sciences, John Curtin School of Medical Research, The Australian National University, Australia.
| |
Collapse
|
32
|
The importance of gene-environment interactions in human obesity. Clin Sci (Lond) 2017; 130:1571-97. [PMID: 27503943 DOI: 10.1042/cs20160221] [Citation(s) in RCA: 108] [Impact Index Per Article: 15.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2016] [Accepted: 05/23/2016] [Indexed: 12/16/2022]
Abstract
The worldwide obesity epidemic has been mainly attributed to lifestyle changes. However, who becomes obese in an obesity-prone environment is largely determined by genetic factors. In the last 20 years, important progress has been made in the elucidation of the genetic architecture of obesity. In parallel with successful gene identifications, the number of gene-environment interaction (GEI) studies has grown rapidly. This paper reviews the growing body of evidence supporting gene-environment interactions in the field of obesity. Heritability, monogenic and polygenic obesity studies provide converging evidence that obesity-predisposing genes interact with a variety of environmental, lifestyle and treatment exposures. However, some skepticism remains regarding the validity of these studies based on several issues, which include statistical modelling, confounding, low replication rate, underpowered analyses, biological assumptions and measurement precision. What follows in this review includes (1) an introduction to the study of GEI, (2) the evidence of GEI in the field of obesity, (3) an outline of the biological mechanisms that may explain these interaction effects, (4) methodological challenges associated with GEI studies and potential solutions, and (5) future directions of GEI research. Thus far, this growing body of evidence has provided a deeper understanding of GEI influencing obesity and may have tremendous applications in the emerging field of personalized medicine and individualized lifestyle recommendations.
Collapse
|
33
|
Styne DM, Arslanian SA, Connor EL, Farooqi IS, Murad MH, Silverstein JH, Yanovski JA. Pediatric Obesity-Assessment, Treatment, and Prevention: An Endocrine Society Clinical Practice Guideline. J Clin Endocrinol Metab 2017; 102:709-757. [PMID: 28359099 PMCID: PMC6283429 DOI: 10.1210/jc.2016-2573] [Citation(s) in RCA: 644] [Impact Index Per Article: 92.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/30/2016] [Accepted: 11/10/2016] [Indexed: 02/06/2023]
Abstract
COSPONSORING ASSOCIATIONS The European Society of Endocrinology and the Pediatric Endocrine Society. This guideline was funded by the Endocrine Society. OBJECTIVE To formulate clinical practice guidelines for the assessment, treatment, and prevention of pediatric obesity. PARTICIPANTS The participants include an Endocrine Society-appointed Task Force of 6 experts, a methodologist, and a medical writer. EVIDENCE This evidence-based guideline was developed using the Grading of Recommendations, Assessment, Development, and Evaluation approach to describe the strength of recommendations and the quality of evidence. The Task Force commissioned 2 systematic reviews and used the best available evidence from other published systematic reviews and individual studies. CONSENSUS PROCESS One group meeting, several conference calls, and e-mail communications enabled consensus. Endocrine Society committees and members and co-sponsoring organizations reviewed and commented on preliminary drafts of this guideline. CONCLUSION Pediatric obesity remains an ongoing serious international health concern affecting ∼17% of US children and adolescents, threatening their adult health and longevity. Pediatric obesity has its basis in genetic susceptibilities influenced by a permissive environment starting in utero and extending through childhood and adolescence. Endocrine etiologies for obesity are rare and usually are accompanied by attenuated growth patterns. Pediatric comorbidities are common and long-term health complications often result; screening for comorbidities of obesity should be applied in a hierarchal, logical manner for early identification before more serious complications result. Genetic screening for rare syndromes is indicated only in the presence of specific historical or physical features. The psychological toll of pediatric obesity on the individual and family necessitates screening for mental health issues and counseling as indicated. The prevention of pediatric obesity by promoting healthful diet, activity, and environment should be a primary goal, as achieving effective, long-lasting results with lifestyle modification once obesity occurs is difficult. Although some behavioral and pharmacotherapy studies report modest success, additional research into accessible and effective methods for preventing and treating pediatric obesity is needed. The use of weight loss medications during childhood and adolescence should be restricted to clinical trials. Increasing evidence demonstrates the effectiveness of bariatric surgery in the most seriously affected mature teenagers who have failed lifestyle modification, but the use of surgery requires experienced teams with resources for long-term follow-up. Adolescents undergoing lifestyle therapy, medication regimens, or bariatric surgery for obesity will need cohesive planning to help them effectively transition to adult care, with continued necessary monitoring, support, and intervention. Transition programs for obesity are an uncharted area requiring further research for efficacy. Despite a significant increase in research on pediatric obesity since the initial publication of these guidelines 8 years ago, further study is needed of the genetic and biological factors that increase the risk of weight gain and influence the response to therapeutic interventions. Also needed are more studies to better understand the genetic and biological factors that cause an obese individual to manifest one comorbidity vs another or to be free of comorbidities. Furthermore, continued investigation into the most effective methods of preventing and treating obesity and into methods for changing environmental and economic factors that will lead to worldwide cultural changes in diet and activity should be priorities. Particular attention to determining ways to effect systemic changes in food environments and total daily mobility, as well as methods for sustaining healthy body mass index changes, is of importance.
Collapse
Affiliation(s)
- Dennis M Styne
- University of California Davis, Sacramento, California 95817
| | | | | | | | | | | | | |
Collapse
|
34
|
Altawil AS, Mawlawi HA, Alghamdi KA, Almijmaj FF. A Novel Homozygous Frameshift Mutation in Exon 2 of LEP Gene Associated with Severe Obesity: A Case Report. CLINICAL MEDICINE INSIGHTS-PEDIATRICS 2016; 10:115-118. [PMID: 27980447 PMCID: PMC5153319 DOI: 10.4137/cmped.s40432] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/22/2016] [Revised: 09/27/2016] [Accepted: 10/06/2016] [Indexed: 02/07/2023]
Abstract
BACKGROUND Monogenic obesity is a rare type of obesity caused by a mutation in a single gene. Patients with monogenic obesity may develop early onset of obesity and severe metabolic abnormalities. CASE PRESENTATION A two-and-half-year-old girl was presented to our clinic because of excessive weight gain and hyperphagia. She was born at full term, by normal vaginal delivery with birth weight of 2.82 kg and no complications during pregnancy. The patient was the second child of two healthy, non-obese Saudis with known consanguinity. She gained weight rapidly leading to obesity at the age of three months. METHODS The demographic data and clinical features were recorded. Blood samples were collected and tested for endocrine and metabolic characteristics and genetic studies. Mutations of the LEP gene were screened. The coding exons 2 and 3 and the corresponding exon–intron boundaries were amplified by polymerase chain reaction using specific primers, analyzed by direct sequencing using an ABI sequencer 3500 xL GA (Applied Biosystems), and evaluated using the JSI SeqPilot software. The resulting sequence data were compared with the reference MM_0002302. CONCLUSION We report a novel homozygous frameshift mutation c.144delin TAC (G1n49Thrfs*23) in exon 2 of the LEP gene associated with extreme obesity.
Collapse
Affiliation(s)
- Ashwaq Shukri Altawil
- Department of Pediatrics, Prince Sultan Military Medical City, Riyadh, Kingdom of Saudi Arabia
| | - Horia Ahmad Mawlawi
- Department of Pediatrics, Prince Sultan Military Medical City, Riyadh, Kingdom of Saudi Arabia
| | - Khalid Ateeq Alghamdi
- Department of Pediatrics, Prince Sultan Military Medical City, Riyadh, Kingdom of Saudi Arabia
| | - Faten Fohaid Almijmaj
- Department of Pediatrics, Prince Sultan Military Medical City, Riyadh, Kingdom of Saudi Arabia
| |
Collapse
|
35
|
Abstract
Although diet and exercise have been the cornerstone of therapy for obesity, efficacy is suboptimal and short lived. Surgical procedures are durable but invasive therapy for obesity. Supplemental therapies for obesity that are minimally invasive, low risk, and effective are needed. Several therapeutic options are being developed that offer obese patients and their health care providers alternatives to what is currently available.
Collapse
Affiliation(s)
- Octavia Pickett-Blakely
- GI Nutrition, Obesity and Celiac Disease Program, Division of Gastroenterology, University of Pennsylvania Perelman School of Medicine, 3400 Convention Avenue, 4 South, Philadelphia, PA 19104, USA.
| | - Carolyn Newberry
- Division of Gastroenterology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA
| |
Collapse
|
36
|
Affiliation(s)
- Stephen O'Rahilly
- Wellcome Trust-MRC Institute of Metabolic Science, University of Cambridge, Cambridge, UK
| |
Collapse
|
37
|
Barger SW. Gene regulation and genetics in neurochemistry, past to future. J Neurochem 2016; 139 Suppl 2:24-57. [PMID: 27747882 DOI: 10.1111/jnc.13629] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2015] [Revised: 03/01/2016] [Accepted: 03/30/2016] [Indexed: 12/14/2022]
Abstract
Ask any neuroscientist to name the most profound discoveries in the field in the past 60 years, and at or near the top of the list will be a phenomenon or technique related to genes and their expression. Indeed, our understanding of genetics and gene regulation has ushered in whole new systems of knowledge and new empirical approaches, many of which could not have even been imagined prior to the molecular biology boon of recent decades. Neurochemistry, in the classic sense, intersects with these concepts in the manifestation of neuropeptides, obviously dependent upon the central dogma (the established rules by which DNA sequence is eventually converted into protein primary structure) not only for their conformation but also for their levels and locales of expression. But, expanding these considerations to non-peptide neurotransmitters illustrates how gene regulatory events impact neurochemistry in a much broader sense, extending beyond the neurochemicals that translate electrical signals into chemical ones in the synapse, to also include every aspect of neural development, structure, function, and pathology. From the beginning, the mutability - yet relative stability - of genes and their expression patterns were recognized as potential substrates for some of the most intriguing phenomena in neurobiology - those instances of plasticity required for learning and memory. Near-heretical speculation was offered in the idea that perhaps the very sequence of the genome was altered to encode memories. A fascinating component of the intervening progress includes evidence that the central dogma is not nearly as rigid and consistent as we once thought. And this mutability extends to the potential to manipulate that code for both experimental and clinical purposes. Astonishing progress has been made in the molecular biology of neurochemistry during the 60 years since this journal debuted. Many of the gains in conceptual understanding have been driven by methodological progress, from automated high-throughput sequencing instruments to recombinant-DNA vectors that can convey color-coded genetic modifications in the chromosomes of live adult animals. This review covers the highlights of these advances, both theoretical and technological, along with a brief window into the promising science ahead. This article is part of the 60th Anniversary special issue.
Collapse
Affiliation(s)
- Steven W Barger
- Department of Geriatrics, Department of Neurobiology and Developmental Sciences, University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA. .,Geriatric Research Education and Clinical Center, Central Arkansas Veterans Healthcare System, Little Rock, Arkansas, USA.
| |
Collapse
|
38
|
Bhaskar V, Goldfine ID, Gerstner R, Michelson K, Tran C, Nonet G, Bohmann D, Pongo E, Zhao J, Horwitz AH, Takeuchi T, White M, Corbin JA. An allosteric antibody to the leptin receptor reduces body weight and reverses the diabetic phenotype in the Lep(ob) /Lep(ob) mouse. Obesity (Silver Spring) 2016; 24:1687-94. [PMID: 27330016 DOI: 10.1002/oby.21539] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/04/2016] [Accepted: 04/08/2016] [Indexed: 12/19/2022]
Abstract
OBJECTIVE Leptin (LEP) deficiency results in major metabolic perturbations, including obesity, dyslipidemia, and diabetes. Although LEP deficiency can be treated with daily injections of a recombinant LEP, generation of an antibody activating the LEP receptor (LEPR) that has both an intrinsically long half-life and low immunogenicity could be useful in the treatment of this condition. METHODS Phage display technology coupled with flow cytometry and cell-based in vitro assays were employed to identify an allosteric agonist of the mouse LEPR. LEP-deficient Lep(ob) /Lep(ob) mice were used to compare in vivo effects of LEP to antibody administration. To evaluate hypothalamic effects of treatment, changes in mRNA levels of neuropeptide Y and proopiomelanocortin were measured. RESULTS XPA.80.037 is a monoclonal antibody that demonstrates allosteric agonism of the mouse LEPR. Treatment of Lep(ob) /Lep(ob) mice with XPA.80.037 markedly reduced hyperphagia and body weight, normalized blood glucose and plasma insulin levels, and corrected dyslipidemia. These metabolic alterations correlated with changes in mRNA levels of neuropeptide Y and proopiomelanocortin, suggesting that XPA.80.037 had hypothalamic effects. CONCLUSIONS Agonist allosteric monoclonal antibodies to the LEPR can correct metabolic effects associated with LEP deficiency in vivo and thereby have the potential to treat conditions of LEP deficiency.
Collapse
Affiliation(s)
- Vinay Bhaskar
- Preclinical Development, XOMA Corporation, Berkeley, California, USA
| | - Ira D Goldfine
- Preclinical Development, XOMA Corporation, Berkeley, California, USA
| | - Resi Gerstner
- Discovery Research, XOMA Corporation, Berkeley, California, USA
| | | | - Catarina Tran
- Discovery Research, XOMA Corporation, Berkeley, California, USA
| | - Genevieve Nonet
- Discovery Research, XOMA Corporation, Berkeley, California, USA
| | - David Bohmann
- Discovery Research, XOMA Corporation, Berkeley, California, USA
| | - Elizabeth Pongo
- Discovery Research, XOMA Corporation, Berkeley, California, USA
| | - Jingsong Zhao
- Preclinical Development, XOMA Corporation, Berkeley, California, USA
| | | | | | - Mark White
- Discovery Research, XOMA Corporation, Berkeley, California, USA
| | - John A Corbin
- Discovery Research, XOMA Corporation, Berkeley, California, USA
| |
Collapse
|
39
|
Wasim M, Awan FR, Najam SS, Khan AR, Khan HN. Role of Leptin Deficiency, Inefficiency, and Leptin Receptors in Obesity. Biochem Genet 2016; 54:565-72. [PMID: 27313173 DOI: 10.1007/s10528-016-9751-z] [Citation(s) in RCA: 85] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2015] [Accepted: 05/27/2016] [Indexed: 11/25/2022]
Abstract
Leptin protein consists of 167 amino acids, which is mainly secreted from the white adipose tissue. This protein acts on the hypothalamic regions of the brain which control eating behavior, thus playing a significant role in maintaining body's metabolism. Leptin receptors belong to glycoprotein 130 (gp130) family of cytokine receptors and exist in six isoforms (LEPR a-f), and all the isoforms are encoded by LEPR gene; out of these isoforms, the LEPR-b receptor is the 'longest form,' and in most of the cases, mutations in this isoform cause severe obesity. Also, mutations in the leptin gene (LEP) or its receptors gene can lead to obesity. Some biochemical pathways affect the bioactivity of leptin and/or its receptors. To date, eleven pathogenic mutations have been reported in the LEP which are p.L72S, p.N103K, p.R105W, p.H118L, p.S141C, p.W121X c.104_106delTCA, c.135del3bp, c.398delG, c.481_482delCT, and c.163C>T. Different mutations in the LEPR have also been reported as c.2396-1 G>T, c.1675 G>A, p.P316T, etc. In some studies, where leptin was deficient, leptin replacement therapy has shown positive impact by preventing weight gain and obesity.
Collapse
Affiliation(s)
- Muhammad Wasim
- Diabetes and Cardio-Metabolic Disorders Laboratory, Health Biotechnology Division, National Institute for Biotechnology and Genetic Engineering (NIBGE), Faisalabad, Pakistan.
| | - Fazli Rabbi Awan
- Diabetes and Cardio-Metabolic Disorders Laboratory, Health Biotechnology Division, National Institute for Biotechnology and Genetic Engineering (NIBGE), Faisalabad, Pakistan
| | - Syeda Sadia Najam
- Diabetes and Cardio-Metabolic Disorders Laboratory, Health Biotechnology Division, National Institute for Biotechnology and Genetic Engineering (NIBGE), Faisalabad, Pakistan
| | - Abdul Rehman Khan
- Obesity and Diabetes Research Laboratory, Department of Chemistry, University of Azad Jammu and Kashmir, Muzaffarabad, Pakistan
| | - Haq Nawaz Khan
- Diabetes and Cardio-Metabolic Disorders Laboratory, Health Biotechnology Division, National Institute for Biotechnology and Genetic Engineering (NIBGE), Faisalabad, Pakistan
| |
Collapse
|
40
|
Gonzalez-Carter D, Goode AE, Fiammengo R, Dunlop IE, Dexter DT, Porter AE. Inhibition of Leptin-ObR Interaction Does not Prevent Leptin Translocation Across a Human Blood-Brain Barrier Model. J Neuroendocrinol 2016; 28. [PMID: 27037668 DOI: 10.1111/jne.12392] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/28/2015] [Revised: 03/31/2016] [Accepted: 03/31/2016] [Indexed: 01/01/2023]
Abstract
The adipocyte-derived hormone leptin regulates appetite and energy homeostasis through the activation of leptin receptors (ObR) on hypothalamic neurones; hence, leptin must be transported through the blood-brain barrier (BBB) to reach its target sites in the central nervous system. During obesity, however, leptin BBB transport is decreased, in part precluding leptin as a viable clinical therapy against obesity. Although the short isoform of the ObR (ObRa) has been implicated in the transport of leptin across the BBB as a result of its elevated expression in cerebral microvessels, accumulating evidence indicates that leptin BBB transport is independent of ObRa. In the present study, we employed an ObR-neutralising antibody (9F8) to directly examine the involvement of endothelial ObR in leptin transport across an in vitro human BBB model composed of the human endothelial cell line hCMEC/D3. Our results indicate that, although leptin transport across the endothelial monolayer was nonparacellular, and energy- and endocytosis-dependent, it was not inhibited by pre-treatment with 9F8, despite the ability of the latter to recognise hCMEC/D3-expressed ObR, prevent leptin-ObR binding and inhibit leptin-induced signal transducer and activator of transcription 3 (STAT-3) phosphorylation in hCMEC/D3 cells. Furthermore, hCMEC/D3 cells expressed the transporter protein low-density lipoprotein receptor-related protein-2 (LRP-2), which is capable of binding and endocytosing leptin. In conclusion, our results demonstrate that leptin binding to and signalling through ObR is not required for efficient transport across human endothelial monolayers, indicating that ObR is not the primary leptin transporter at the human BBB, a role which may fall upon LRP-2. A deeper understanding of leptin BBB transport will help clarify the exact causes for leptin resistance seen in obesity and aid in the development of more efficient BBB-penetrating leptin analogues.
Collapse
Affiliation(s)
- D Gonzalez-Carter
- Centre for Neuroinflammation and Neurodegeneration, Division of Brain Sciences, Department of Medicine, Imperial College London, London, UK
- Department of Materials, Faculty of Engineering, Imperial College London, London, UK
| | - A E Goode
- Department of Materials, Faculty of Engineering, Imperial College London, London, UK
| | - R Fiammengo
- Centre for Biomolecular Nanotechnologies @ UniLe, Istituto Italiano di Tecnologia (ITT), Arnesano, Lecce, Italy
| | - I E Dunlop
- Department of Materials, Faculty of Engineering, Imperial College London, London, UK
| | - D T Dexter
- Centre for Neuroinflammation and Neurodegeneration, Division of Brain Sciences, Department of Medicine, Imperial College London, London, UK
| | - A E Porter
- Department of Materials, Faculty of Engineering, Imperial College London, London, UK
| |
Collapse
|
41
|
Barbetti F, Colombo C, Haataja L, Cras-Méneur C, Bernardini S, Arvan P. Hyperglucagonemia in an animal model of insulin- deficient diabetes: what therapy can improve it? Clin Diabetes Endocrinol 2016; 2:11. [PMID: 28702245 PMCID: PMC5471666 DOI: 10.1186/s40842-016-0029-5] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/19/2016] [Accepted: 04/11/2016] [Indexed: 02/06/2023] Open
Abstract
Background Intra-islet insulin contributes to alpha-cell suppression. Akita mice carry a toxic-gain-of- function Ins2 gene mutation encoding proinsulin-C(A7)Y, similar to that described in human Mutant Ins-gene induced Diabetes of Youth, which decreases intra-islet insulin. Herein, we examined Akita mice for examination of circulating insulin and circulating glucagon levels. The possibility that loss of intra-islet suppression of alpha-cells, with increased circulating glucagon, contributes to diabetes under conditions of intra-islet insulin deficiency, raises questions about effective treatments that may be available. Methods Blood glucose, plasma insulin, C-peptide I, C-peptide II, and glucagon were measured at various times during development of diabetes in Akita mice. We also used Akita- like hProC(A7)Y-CpepGFP transgenic mice in Ins2+/+, Ins2+/− and Ins2−/− genetic backgrounds (providing animals with greater or lesser defects in islet insulin production, respectively) in order to examine the relative abundance of immunostainable intra-islet glucagon-positive and insulin-positive cells. Similar measurements were made in Akita mice. Finally, the effects of treatment with insulin, exendin-4, and leptin on blood glucose were then compared in Akita mice. Results Interestingly, total insulin levels in the circulation were not frankly low in Akita mice, although they did not rise appropriately with the onset of hyperglycemia. By contrast, in severely diabetic Akita mice at 6 weeks of age, circulating glucagon levels were significantly elevated. Additionally, in Ins2+/− and Ins2−/− mice bearing the Akita-like hProC(A7)Y-CpepGFP transgene, development of diabetes correlated with an increase in the relative intra-islet abundance of immunostainable glucagon-positive cells, and a similar observation was made in Akita islets. In Akita mice, whereas a brief treatment with exendin-4 resulted in no apparent improvement in hyperglycemia, leptin treatment resulted in restoration of normoglycemia. Curiously, leptin treatment also suppressed circulating glucagon levels. Conclusions Loss of insulin-mediated intra-islet suppression of glucagon production may be a contributor to hyperglycemia in Akita mice, and leptin treatment appears beneficial in such a circumstance. This treatment might also be considered in some human diabetes patients with diminished insulin reserve.
Collapse
Affiliation(s)
- Fabrizio Barbetti
- Department of Experimental Medicine and Surgery, University of Tor Vergata, Rome, Italy.,Bambino Gesù Children's Hospital, IRCCS, Rome, Italy.,Department of Experimental Medicine and Surgery, University of Tor Vergata. Tor Vergata University Hospital, first floor, section D, room 118, Viale Oxford 81, 00133 Rome, Italy
| | - Carlo Colombo
- Department of Experimental Medicine and Surgery, University of Tor Vergata, Rome, Italy
| | - Leena Haataja
- Division of Metabolism, Endocrinology & Diabetes, University of Michigan Medical Center, Ann Arbor, MI USA
| | - Corentin Cras-Méneur
- Division of Metabolism, Endocrinology & Diabetes, University of Michigan Medical Center, Ann Arbor, MI USA
| | - Sergio Bernardini
- Department of Experimental Medicine and Surgery, University of Tor Vergata, Rome, Italy
| | - Peter Arvan
- Division of Metabolism, Endocrinology & Diabetes, University of Michigan Medical Center, Ann Arbor, MI USA.,University of Michigan Medical Center, Brehm Tower room 5112, 1000 Wall St., Ann Arbor, MI 48105 USA
| |
Collapse
|
42
|
Gavello D, Carbone E, Carabelli V. Leptin-mediated ion channel regulation: PI3K pathways, physiological role, and therapeutic potential. Channels (Austin) 2016; 10:282-96. [PMID: 27018500 DOI: 10.1080/19336950.2016.1164373] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
Leptin is produced by adipose tissue and identified as a "satiety signal," informing the brain when the body has consumed enough food. Specific areas of the hypothalamus express leptin receptors (LEPRs) and are the primary site of leptin action for body weight regulation. In response to leptin, appetite is suppressed and energy expenditure allowed. Beside this hypothalamic action, leptin targets other brain areas in addition to neuroendocrine cells. LEPRs are expressed also in the hippocampus, neocortex, cerebellum, substantia nigra, pancreatic β-cells, and chromaffin cells of the adrenal gland. It is intriguing how leptin is able to activate different ionic conductances, thus affecting excitability, synaptic plasticity and neurotransmitter release, depending on the target cell. Most of the intracellular pathways activated by leptin and directed to ion channels involve PI3K, which in turn phosphorylates different downstream substrates, although parallel pathways involve AMPK and MAPK. In this review we will describe the effects of leptin on BK, KATP, KV, CaV, TRPC, NMDAR and AMPAR channels and clarify the landscape of pathways involved. Given the ability of leptin to influence neuronal excitability and synaptic plasticity by modulating ion channels activity, we also provide a short overview of the growing potentiality of leptin as therapeutic agent for treating neurological disorders.
Collapse
Affiliation(s)
- Daniela Gavello
- a Department of Drug Science , Lab of Cellular Physiology and Molecular Neuroscience, NIS Center of Excellence, University of Torino , Torino , Italy
| | - Emilio Carbone
- a Department of Drug Science , Lab of Cellular Physiology and Molecular Neuroscience, NIS Center of Excellence, University of Torino , Torino , Italy
| | - Valentina Carabelli
- a Department of Drug Science , Lab of Cellular Physiology and Molecular Neuroscience, NIS Center of Excellence, University of Torino , Torino , Italy
| |
Collapse
|
43
|
Page AJ. Mimecan: A Newly Identified Adipokine and Regulator of Appetite. EBioMedicine 2016; 2:1584-5. [PMID: 26870776 PMCID: PMC4740335 DOI: 10.1016/j.ebiom.2015.11.008] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2015] [Revised: 11/03/2015] [Accepted: 11/03/2015] [Indexed: 11/29/2022] Open
Affiliation(s)
- Amanda J Page
- Centre for Nutrition and Gastrointestinal Disease, Discipline of Medicine, University of Adelaide, Frome Road, Adelaide, SA 5005, Australia
| |
Collapse
|
44
|
Tchang BG, Shukla AP, Aronne LJ. Metreleptin and generalized lipodystrophy and evolving therapeutic perspectives. Expert Opin Biol Ther 2016; 15:1061-75. [PMID: 26063386 DOI: 10.1517/14712598.2015.1052789] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
INTRODUCTION Metreleptin was recently approved by the Food and Drug Administration for the treatment of generalized lipodystrophy, a condition characterized by leptin deficiency. Its efficacy as hormone replacement therapy suggests broader applications in diseases also characterized by leptin abnormalities, such as familial partial lipodystrophy (FPLD), non-alcoholic fatty liver disease (NAFLD), and common obesity. Metreleptin, in conjunction with other pharmacologic interventions, has the potential to address one of the most widespread epidemics of our time, obesity. AREAS COVERED This review covers the physiology of leptin, the pharmacologic properties of recombinant methionyl human leptin (R-metHu-Leptin, metreleptin), evidence for metreleptin's efficacy in the treatment of generalized lipodystrophy from both completed and ongoing clinical trials, safety concerns, and future directions in metreleptin research. EXPERT OPINION Metreleptin's approval for generalized lipodystrophy is the first step in defining and expanding its role to other metabolic diseases. Clinical trials are underway to delineate its efficacy in FPLD, human immunodeficiency virus/highly active anti-retroviral therapy-associated acquired lipodystrophy (HAL), and NAFLD. Additionally, there is growing data that support a therapeutic role in obesity. One of the barriers to development, however, is metreleptin's safety and immunogenicity. Further advances in biologic compatibility are required before metreleptin can be approved for additional indications.
Collapse
Affiliation(s)
- Beverly G Tchang
- Weill Cornell Medical College, Comprehensive Weight Control Center, Division of Endocrinology, Diabetes & Metabolism , 1165 York Avenue, New York, NY, 10065 , USA
| | | | | |
Collapse
|
45
|
Obesity Reduces Cognitive and Motor Functions across the Lifespan. Neural Plast 2016; 2016:2473081. [PMID: 26881095 PMCID: PMC4737453 DOI: 10.1155/2016/2473081] [Citation(s) in RCA: 93] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2015] [Accepted: 10/15/2015] [Indexed: 12/11/2022] Open
Abstract
Due to a sedentary lifestyle, more and more people are becoming obese nowadays. In addition to health-related problems, obesity can also impair cognition and motor performance. Previous results have shown that obesity mainly affects cognition and motor behaviors through altering brain functions and musculoskeletal system, respectively. Many factors, such as insulin/leptin dysregulation and inflammation, mediate the effect of obesity and cognition and motor behaviors. Substantial evidence has suggested exercise to be an effective way to improve obesity and related cognitive and motor dysfunctions. This paper aims to discuss the association of obesity with cognition and motor behaviors and its underlying mechanisms. Following this, mechanisms of exercise to improve obesity-related dysfunctions are described. Finally, implications and future research direction are raised.
Collapse
|
46
|
The Effects of Leptin Replacement on Neural Plasticity. Neural Plast 2016; 2016:8528934. [PMID: 26881138 PMCID: PMC4735938 DOI: 10.1155/2016/8528934] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2015] [Revised: 10/09/2015] [Accepted: 10/19/2015] [Indexed: 12/21/2022] Open
Abstract
Leptin, an adipokine synthesized and secreted mainly by the adipose tissue, has multiple effects on the regulation of food intake, energy expenditure, and metabolism. Its recently-approved analogue, metreleptin, has been evaluated in clinical trials for the treatment of patients with leptin deficiency due to mutations in the leptin gene, lipodystrophy syndromes, and hypothalamic amenorrhea. In such patients, leptin replacement therapy has led to changes in brain structure and function in intra- and extrahypothalamic areas, including the hippocampus. Furthermore, in one of those patients, improvements in neurocognitive development have been observed. In addition to this evidence linking leptin to neural plasticity and function, observational studies evaluating leptin-sufficient humans have also demonstrated direct correlation between blood leptin levels and brain volume and inverse associations between circulating leptin and risk for the development of dementia. This review summarizes the evidence in the literature on the role of leptin in neural plasticity (in leptin-deficient and in leptin-sufficient individuals) and its effects on synaptic activity, glutamate receptor trafficking, neuronal morphology, neuronal development and survival, and microglial function.
Collapse
|
47
|
Reno CM, Ding Y, Sherwin R. Leptin acts in the brain to influence hypoglycemic counterregulation: disparate effects of acute and recurrent hypoglycemia on glucagon release. Am J Physiol Endocrinol Metab 2015; 309:E960-7. [PMID: 26506851 PMCID: PMC4816199 DOI: 10.1152/ajpendo.00361.2015] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/04/2015] [Accepted: 10/21/2015] [Indexed: 12/24/2022]
Abstract
Leptin has been shown to diminish hyperglycemia via reduced glucagon secretion, although it can also enhance sympathoadrenal responses. However, whether leptin can also inhibit glucagon secretion during insulin-induced hypoglycemia or increase epinephrine during acute or recurrent hypoglycemia has not been examined. To test whether leptin acts in the brain to influence counterregulation, hyperinsulinemic hypoglycemic (∼45 mg/dl) clamps were performed on rats exposed to or not exposed to recurrent hypoglycemia (3 days, ∼40 mg/dl). Intracerebroventricular artificial cerebral spinal fluid or leptin was infused during the clamp. During acute hypoglycemia, leptin decreased glucagon responses by 51% but increased epinephrine and norepinephrine by 24 and 48%, respectively. After recurrent hypoglycemia, basal plasma leptin levels were undetectable. Subsequent brain leptin infusion during hypoglycemia paradoxically increased glucagon by 45% as well as epinephrine by 19%. In conclusion, leptin acts within the brain to diminish glucagon secretion during acute hypoglycemia but increases epinephrine, potentially limiting its detrimental effects during hypoglycemia. Exposure to recurrent hypoglycemia markedly suppresses plasma leptin, whereas exogenous brain leptin delivery enhances both glucagon and epinephrine release to subsequent hypoglycemia. These data suggest that recurrent hypoglycemia may diminish counterregulatory responses in part by reducing brain leptin action.
Collapse
Affiliation(s)
- Candace M Reno
- Yale University School of Medicine, Department of Internal Medicine-Section of Endocrinology, New Haven, Connecticut
| | - Yuyan Ding
- Yale University School of Medicine, Department of Internal Medicine-Section of Endocrinology, New Haven, Connecticut
| | - Robert Sherwin
- Yale University School of Medicine, Department of Internal Medicine-Section of Endocrinology, New Haven, Connecticut
| |
Collapse
|
48
|
Rodriguez AJ, Mastronardi CA, Paz-Filho GJ. New advances in the treatment of generalized lipodystrophy: role of metreleptin. Ther Clin Risk Manag 2015; 11:1391-400. [PMID: 26396524 PMCID: PMC4577254 DOI: 10.2147/tcrm.s66521] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
Recombinant methionyl human leptin or metreleptin is a synthetic leptin analog that has been trialed in patients with leptin-deficient conditions, such as leptin deficiency due to mutations in the leptin gene, hypothalamic amenorrhea, and lipodystrophy syndromes. These syndromes are characterized by partial or complete absence of adipose tissue and hormones derived from adipose tissue, most importantly leptin. Patients deficient in leptin exhibit a number of severe metabolic abnormalities such as hyperglycemia, hypertriglyceridemia, and hepatic steatosis, which can progress to diabetes mellitus, acute pancreatitis, and hepatic cirrhosis, respectively. For the management of these abnormalities, multiple therapies are usually required, and advanced stages may be progressively difficult to treat. Following many successful trials, the US Food and Drug Administration approved metreleptin for the treatment of non-HIV-related forms of generalized lipodystrophy. Leptin replacement therapy with metreleptin has, in many cases, reversed these metabolic complications, with improvements in glucose-insulin-lipid homeostasis, and regression of fatty liver disease. Besides being effective, a daily subcutaneous administration of metreleptin is generally safe, but the causal association between metreleptin and immune complications (such as lymphoma) is still unclear. Moreover, further investigation is needed to elucidate mechanisms by which metreleptin leads to the development of anti-leptin antibodies. Herein, we review clinical aspects of generalized lipodystrophy and the pharmacological profile of metreleptin. Further, we examine studies that assessed the safety and efficacy of metreleptin, and outline some clinical perspectives on the drug.
Collapse
Affiliation(s)
| | - Claudio A Mastronardi
- Department of Genome Sciences, The John Curtin School of Medical Research, The Australian National University, Canberra, ACT, Australia
| | - Gilberto J Paz-Filho
- Department of Genome Sciences, The John Curtin School of Medical Research, The Australian National University, Canberra, ACT, Australia
| |
Collapse
|
49
|
Saeed S, Bonnefond A, Manzoor J, Shabbir F, Ayesha H, Philippe J, Durand E, Crouch H, Sand O, Ali M, Butt T, Rathore AW, Falchi M, Arslan M, Froguel P. Genetic variants in LEP, LEPR, and MC4R explain 30% of severe obesity in children from a consanguineous population. Obesity (Silver Spring) 2015; 23:1687-95. [PMID: 26179253 DOI: 10.1002/oby.21142] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/20/2015] [Revised: 03/24/2015] [Accepted: 04/10/2015] [Indexed: 01/17/2023]
Abstract
OBJECTIVE Single gene mutations leading to severe obesity have so far been identified in 3-5% cases in European populations. However, prevalence of these pathogenic mutations has not systematically been examined in specific consanguineous populations. Here we describe the incidence of obesity-associated mutations through a step-wise sequence analysis, in a cohort of 73 Pakistani children with severe obesity from consanguineous families. METHODS Initially, all subjects were screened for mutations in coding regions of leptin (LEP) and melanocortin 4 receptor (MC4R) genes by direct sequencing. Subjects negative for mutation in these genes were screened using microdroplet PCR enrichment and NGS. Genomic structural variation was assessed by genotyping. Serum leptin, insulin, and cortisol were determined by ELISA. RESULTS Among 73 children with severe obesity (BMI SDS > 3.0), we identified 22 probands and 5 relatives, carrying 10 different loss-of-function homozygous mutations in LEP, leptin receptor (LEPR), and MC4R genes, including 4 novel variants. Hypercortisolemia was significantly emphasized in LEP mutation carriers. CONCLUSIONS The prevalence of pathogenic mutations in genes known to directly influence leptin-melanocortin signaling is 30% in our cohort. The results of this study emphasize the desirability of undertaking systematic and in-depth genetic analysis of cases with severe obesity in specific consanguineous populations.
Collapse
Affiliation(s)
- Sadia Saeed
- Department of Genomics of Common Disease, Imperial College London, London, UK
| | - Amélie Bonnefond
- European Genomic Institute for Diabetes (EGID), Lille, France
- CNRS-Umr8199, Lille Pasteur Institute, Lille, France
- Lille University, Lille, France
| | - Jaida Manzoor
- Department of Paediatric Endocrinology, Children's Hospital, Lahore, Pakistan
| | - Faiza Shabbir
- Department of Biological Sciences, Forman Christian College, Lahore, Pakistan
| | - Hina Ayesha
- Department of Paediatrics, Punjab Medical College, Faisalabad, Pakistan
| | - Julien Philippe
- European Genomic Institute for Diabetes (EGID), Lille, France
- CNRS-Umr8199, Lille Pasteur Institute, Lille, France
- Lille University, Lille, France
| | - Emmanuelle Durand
- European Genomic Institute for Diabetes (EGID), Lille, France
- CNRS-Umr8199, Lille Pasteur Institute, Lille, France
- Lille University, Lille, France
| | - Hutokshi Crouch
- Department of Genomics of Common Disease, Imperial College London, London, UK
| | - Olivier Sand
- European Genomic Institute for Diabetes (EGID), Lille, France
- CNRS-Umr8199, Lille Pasteur Institute, Lille, France
- Lille University, Lille, France
| | - Muhammad Ali
- Department of Paediatrics, Mayo Hospital, King Edward Medical University, Lahore, Pakistan
| | - Taeed Butt
- Department of Paediatrics, Fatima Memorial Hospital, Lahore, Pakistan
| | - Ahsan W Rathore
- Department of Paediatric Endocrinology, Children's Hospital, Lahore, Pakistan
| | - Mario Falchi
- Department of Genomics of Common Disease, Imperial College London, London, UK
| | - Muhammad Arslan
- Department of Biological Sciences, Forman Christian College, Lahore, Pakistan
- Centre for Research in Molecular Medicine, the University of Lahore, Lahore, Pakistan
| | - Philippe Froguel
- Department of Genomics of Common Disease, Imperial College London, London, UK
- European Genomic Institute for Diabetes (EGID), Lille, France
- CNRS-Umr8199, Lille Pasteur Institute, Lille, France
- Lille University, Lille, France
| |
Collapse
|
50
|
Insensitivity of well-conditioned mature sheep to central administration of a leptin receptor antagonist. Animal 2015. [PMID: 26220331 DOI: 10.1017/s1751731115001159] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
Abstract
Ruminants remain productive during the energy insufficiency of late pregnancy or early lactation by evoking metabolic adaptations sparing available energy and nutrients (e.g. higher metabolic efficiency and induction of insulin resistance). A deficit in central leptin signaling triggers these adaptations in rodents but whether it does in ruminants remains unclear. To address this issue, five mature ewes were implanted with intracerebroventricular (ICV) cannula in the third ventricle. They were used in two experiments with an ovine leptin antagonist (OLA) when well-conditioned (average body condition score of 3.7 on a 5 point scale). The first experiment tested the ability of OLA to antagonize leptin under in vivo conditions. Ewes received continuous ICV infusion of artificial cerebrospinal fluid (aCSF), ovine leptin (4 µg/h) or the combination of ovine leptin (4 µg/h) and its mutant version OLA (40 µg/h) for 48 h. Dry matter intake (DMI) was measured every day and blood samples were collected on the last day of infusion. ICV infusion of leptin reduced DMI by 24% (P < 0.05), and this effect was completely abolished by OLA co-infusion. A second experiment tested whether a reduction in endogenous leptin signaling in the brain triggers metabolic adaptations. This involved continuous ICV infusions of aCSF or OLA alone (40 µg/h) for 4 consecutive days. The infusion of OLA did not alter voluntary DMI over the treatment period or on any individual day. OLA did not affect plasma variables indicative of insulin action (glucose, non-esterified fatty acids, insulin and the disposition of plasma glucose during an insulin tolerance test) or plasma cortisol, but tended to reduce plasma triiodothyronine and thyroxine (P < 0.07). Overall, these data show that a reduction of central leptin signaling has little impact on insulin action in well-conditioned mature sheep. They also raise the possibility that reduced central leptin signaling plays a role in controlling thyroid hormone production.
Collapse
|