1
|
Semenova E, Guo A, Liang H, Hernandez CJ, John EB, Thaker VV. The expanding landscape of genetic causes of obesity. Pediatr Res 2024:10.1038/s41390-024-03780-6. [PMID: 39690244 DOI: 10.1038/s41390-024-03780-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/10/2024] [Revised: 10/18/2024] [Accepted: 11/03/2024] [Indexed: 12/19/2024]
Abstract
Obesity and weight regulation disorders are determined by the combined effects of genetics and environment. Polygenic obesity results from the combination of common variants in several genes which predisposes the individual to obesity and its related complications. In contrast, monogenic obesity results from changes in single genes, especially those in leptin-melanocortin pathway, and presents with early onset severe obesity, with or without other syndromic features. Rare variants in melanocortin 4 receptor are the commonest form of monogenic obesity. In addition, structural variation in small or large segments of chromosomes may also present with syndromic forms of obesity. Prader-Willi Syndrome, caused by imprinting errors in chromosome 15q11-13, is the most prevalent genetic cause of severe hyperphagia and obesity. With the advances in technologies, the past decade has witnessed a revolution in the identification of novel genetic causes of obesity, primarily in genes related to the leptin melanocortin pathway. The availability of safe melanocortin analogs holds the potential for targeted therapies for some of these disorders. This review summarizes known and novel rare genetic forms of obesity, along with approaches for the clinical investigation of copy number and sequence variants. The goal is to provide a reference for practicing clinicians to encourage genetic testing in obesity. IMPACT: What does this article add to the existing literature? Genetic obesity is an expanding frontier with potential to change management. Here, we summarize current information on the genetic causes of obesity and provide guidance for genetic testing. Emerging treatments may provide targeted precise treatment and change management practices.
Collapse
Affiliation(s)
- Ekaterina Semenova
- Division of Molecular Genetics, Department of Pediatrics, Columbia University Irving Medical Center, New York, NY, USA
| | - Alex Guo
- Division of Molecular Genetics, Department of Pediatrics, Columbia University Irving Medical Center, New York, NY, USA
| | - Harry Liang
- Vagelos College of Physicians and Surgeons, Columbia University, New York, NY, USA
| | - Cindy J Hernandez
- Division of Molecular Genetics, Department of Pediatrics, Columbia University Irving Medical Center, New York, NY, USA
| | - Ella B John
- Division of Molecular Genetics, Department of Pediatrics, Columbia University Irving Medical Center, New York, NY, USA
| | - Vidhu V Thaker
- Division of Molecular Genetics, Department of Pediatrics, Columbia University Irving Medical Center, New York, NY, USA.
- Vagelos College of Physicians and Surgeons, Columbia University, New York, NY, USA.
- Division of Pediatric Endocrinology, Department of Pediatrics, Columbia University Irving Medical Center, New York, NY, USA.
| |
Collapse
|
2
|
Yilmaz B, Erdogan CS, Sandal S, Kelestimur F, Carpenter DO. Obesogens and Energy Homeostasis: Definition, Mechanisms of Action, Exposure, and Adverse Effects on Human Health. Neuroendocrinology 2024; 115:72-100. [PMID: 39622213 DOI: 10.1159/000542901] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/07/2024] [Accepted: 11/28/2024] [Indexed: 02/26/2025]
Abstract
BACKGROUND Obesity is a major risk factor for noncommunicable diseases and is associated with a reduced life expectancy of up to 20 years, as well as with other consequences such as unemployment and increased economic burden for society. It is a multifactorial disease, and physiopathology of obesity involves dysregulated calorie utilization and energy balance, disrupted homeostasis of appetite and satiety, lifestyle factors including sedentary lifestyle, lower socioeconomic status, genetic predisposition, epigenetics, and environmental factors. Some endocrine-disrupting chemicals (EDCs) have been proposed as "obesogens" that stimulate adipogenesis leading to obesity. In this review, definition of obesogens, their adverse effects, underlying mechanisms, and metabolic implications will be updated and discussed. SUMMARY Disruption of lipid homeostasis by EDCs involves multiple mechanisms including increase in the number and size of adipocytes, disruption of endocrine-regulated adiposity and metabolism, alteration of hypothalamic regulation of appetite, satiety, food preference and energy balance, and modification of insulin sensitivity in the liver, skeletal muscle, pancreas, gastrointestinal system, and the brain. At a cellular level, obesogens can exert their endocrine disruptive effects by interfering with peroxisome proliferator-activated receptors and steroid receptors. Human exposure to chemical obesogens mainly occurs by ingestion and, to some extent, by inhalation and dermal uptake, usually in an unconscious manner. Persistent pollutants are lipophilic features; thus, they bioaccumulate in adipose tissue. KEY MESSAGES Although there are an increasing number of reports studying the effects of obesogens, their mechanisms of action remain to be elucidated. In addition, epidemiological studies are needed in order to evaluate human exposure to obesogens.
Collapse
Affiliation(s)
- Bayram Yilmaz
- Department of Physiology, Faculty of Medicine, Yeditepe University, Istanbul, Turkey
- Izmir Biomedicine and Genome Center, Izmir, Turkey
- Department of Physiology, Faculty of Medicine, Dokuz Eylül University, Izmir, Turkey
| | | | - Suleyman Sandal
- Department of Physiology, Faculty of Medicine, Inonu University, Malatya, Turkey
| | - Fahrettin Kelestimur
- Department of Clinical Endocrinology, Faculty of Medicine, Yeditepe University, Istanbul, Turkey
| | - David O Carpenter
- Institute for Health and the Environment, 5 University Place, University at Albany, Rensselaer, New York, USA
| |
Collapse
|
3
|
Fitch AK, Malhotra S, Conroy R. Differentiating monogenic and syndromic obesities from polygenic obesity: Assessment, diagnosis, and management. OBESITY PILLARS 2024; 11:100110. [PMID: 38766314 PMCID: PMC11101890 DOI: 10.1016/j.obpill.2024.100110] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/07/2024] [Revised: 04/18/2024] [Accepted: 04/18/2024] [Indexed: 05/22/2024]
Abstract
Background Obesity is a multifactorial neurohormonal disease that results from dysfunction within energy regulation pathways and is associated with increased morbidity, mortality, and reduced quality of life. The most common form is polygenic obesity, which results from interactions between multiple gene variants and environmental factors. Highly penetrant monogenic and syndromic obesities result from rare genetic variants with minimal environmental influence and can be differentiated from polygenic obesity depending on key symptoms, including hyperphagia; early-onset, severe obesity; and suboptimal responses to nontargeted therapies. Timely diagnosis of monogenic or syndromic obesity is critical to inform management strategies and reduce disease burden. We outline the physiology of weight regulation, role of genetics in obesity, and differentiating characteristics between polygenic and rare genetic obesity to facilitate diagnosis and transition toward targeted therapies. Methods In this narrative review, we focused on case reports, case studies, and natural history studies of patients with monogenic and syndromic obesities and clinical trials examining the efficacy, safety, and quality of life impact of nontargeted and targeted therapies in these populations. We also provide comprehensive algorithms for diagnosis of patients with suspected rare genetic causes of obesity. Results Patients with monogenic and syndromic obesities commonly present with hyperphagia (ie, pathologic, insatiable hunger) and early-onset, severe obesity, and the presence of hallmark characteristics can inform genetic testing and diagnostic approach. Following diagnosis, specialized care teams can address complex symptoms, and hyperphagia is managed behaviorally. Various pharmacotherapies show promise in these patient populations, including setmelanotide and glucagon-like peptide-1 receptor agonists. Conclusion Understanding the pathophysiology and differentiating characteristics of monogenic and syndromic obesities can facilitate diagnosis and management and has led to development of targeted pharmacotherapies with demonstrated efficacy for reducing body weight and hunger in the affected populations.
Collapse
Affiliation(s)
| | - Sonali Malhotra
- Harvard Medical School, Boston, MA, USA
- Rhythm Pharmaceuticals, Inc., Boston, MA, USA
- Massachussetts General Hospital, Boston, MA, USA
| | | |
Collapse
|
4
|
Vos N, Haghshenas S, van der Laan L, Russel PKM, Rooney K, Levy MA, Relator R, Kerkhof J, McConkey H, Maas SM, Vissers LELM, de Vries BBA, Pfundt R, Elting MW, van Hagen JM, Verbeek NE, Jongmans MCJ, Lakeman P, Rumping L, Bosch DGM, Vitobello A, Thauvin-Robinet C, Faivre L, Nambot S, Garde A, Willems M, Genevieve D, Nicolas G, Busa T, Toutain A, Gérard M, Bizaoui V, Isidor B, Merla G, Accadia M, Schwartz CE, Ounap K, Hoffer MJV, Nezarati MM, van den Boogaard MJH, Tedder ML, Rogers C, Brusco A, Ferrero GB, Spodenkiewicz M, Sidlow R, Mussa A, Trajkova S, McCann E, Mroczkowski HJ, Jansen S, Donker-Kaat L, Duijkers FAM, Stuurman KE, Mannens MMAM, Alders M, Henneman P, White SM, Sadikovic B, van Haelst MM. The detection of a strong episignature for Chung-Jansen syndrome, partially overlapping with Börjeson-Forssman-Lehmann and White-Kernohan syndromes. Hum Genet 2024; 143:761-773. [PMID: 38787418 PMCID: PMC11186873 DOI: 10.1007/s00439-024-02679-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2023] [Accepted: 05/09/2024] [Indexed: 05/25/2024]
Abstract
Chung-Jansen syndrome is a neurodevelopmental disorder characterized by intellectual disability, behavioral problems, obesity and dysmorphic features. It is caused by pathogenic variants in the PHIP gene that encodes for the Pleckstrin homology domain-interacting protein, which is part of an epigenetic modifier protein complex. Therefore, we hypothesized that PHIP haploinsufficiency may impact genome-wide DNA methylation (DNAm). We assessed the DNAm profiles of affected individuals with pathogenic and likely pathogenic PHIP variants with Infinium Methylation EPIC arrays and report a specific and sensitive DNAm episignature biomarker for Chung-Jansen syndrome. In addition, we observed similarities between the methylation profile of Chung-Jansen syndrome and that of functionally related and clinically partially overlapping genetic disorders, White-Kernohan syndrome (caused by variants in DDB1 gene) and Börjeson-Forssman-Lehmann syndrome (caused by variants in PHF6 gene). Based on these observations we also proceeded to develop a common episignature biomarker for these disorders. These newly defined episignatures can be used as part of a multiclass episignature classifier for screening of affected individuals with rare disorders and interpretation of genetic variants of unknown clinical significance, and provide further insights into the common molecular pathophysiology of the clinically-related Chung-Jansen, Börjeson-Forssman-Lehmann and White-Kernohan syndromes.
Collapse
Affiliation(s)
- Niels Vos
- Amsterdam UMC, Department of Human Genetics, University of Amsterdam, Meibergdreef 9, 1105 AZ, Amsterdam, The Netherlands
- Amsterdam Reproduction & Development Research Institute, Amsterdam, The Netherlands
| | - Sadegheh Haghshenas
- Verspeeten Clinical Genome Centre, London Health Sciences Centre, London, ON, N6A 5W9, Canada
| | - Liselot van der Laan
- Amsterdam UMC, Department of Human Genetics, University of Amsterdam, Meibergdreef 9, 1105 AZ, Amsterdam, The Netherlands
- Amsterdam Reproduction & Development Research Institute, Amsterdam, The Netherlands
| | - Perle K M Russel
- Amsterdam UMC, Department of Human Genetics, University of Amsterdam, Meibergdreef 9, 1105 AZ, Amsterdam, The Netherlands
- Amsterdam Reproduction & Development Research Institute, Amsterdam, The Netherlands
| | - Kathleen Rooney
- Verspeeten Clinical Genome Centre, London Health Sciences Centre, London, ON, N6A 5W9, Canada
- Department of Pathology and Laboratory Medicine, Western University, London, ON, N6A 3K7, Canada
| | - Michael A Levy
- Verspeeten Clinical Genome Centre, London Health Sciences Centre, London, ON, N6A 5W9, Canada
| | - Raissa Relator
- Verspeeten Clinical Genome Centre, London Health Sciences Centre, London, ON, N6A 5W9, Canada
| | - Jennifer Kerkhof
- Verspeeten Clinical Genome Centre, London Health Sciences Centre, London, ON, N6A 5W9, Canada
| | - Haley McConkey
- Verspeeten Clinical Genome Centre, London Health Sciences Centre, London, ON, N6A 5W9, Canada
- Department of Pathology and Laboratory Medicine, Western University, London, ON, N6A 3K7, Canada
| | - Saskia M Maas
- Amsterdam UMC, Department of Human Genetics, University of Amsterdam, Meibergdreef 9, 1105 AZ, Amsterdam, The Netherlands
- Amsterdam Reproduction & Development Research Institute, Amsterdam, The Netherlands
| | - Lisenka E L M Vissers
- Department of Human Genetics, Research Institute for Medical Innovation, Radboud University Medical Center, 6525 GA, Nijmegen, The Netherlands
| | - Bert B A de Vries
- Department of Human Genetics, Research Institute for Medical Innovation, Radboud University Medical Center, 6525 GA, Nijmegen, The Netherlands
| | - Rolph Pfundt
- Department of Human Genetics, Research Institute for Medical Innovation, Radboud University Medical Center, 6525 GA, Nijmegen, The Netherlands
| | - Mariet W Elting
- Amsterdam UMC, Department of Human Genetics, University of Amsterdam, Meibergdreef 9, 1105 AZ, Amsterdam, The Netherlands
- Amsterdam Reproduction & Development Research Institute, Amsterdam, The Netherlands
| | - Johanna M van Hagen
- Amsterdam UMC, Department of Human Genetics, University of Amsterdam, Meibergdreef 9, 1105 AZ, Amsterdam, The Netherlands
- Amsterdam Reproduction & Development Research Institute, Amsterdam, The Netherlands
| | - Nienke E Verbeek
- Department of Genetics, University Medical Center Utrecht, 3584 CX, Utrecht, The Netherlands
| | - Marjolijn C J Jongmans
- Department of Genetics, University Medical Center Utrecht, 3584 CX, Utrecht, The Netherlands
| | - Phillis Lakeman
- Amsterdam UMC, Department of Human Genetics, University of Amsterdam, Meibergdreef 9, 1105 AZ, Amsterdam, The Netherlands
- Amsterdam Reproduction & Development Research Institute, Amsterdam, The Netherlands
| | - Lynne Rumping
- Center for Medical Genetics, Antwerp University Hospital, University of Antwerp, Drie Eikenstraat 655, 2650, Edegem, Belgium
| | - Danielle G M Bosch
- Department of Clinical Genetics, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Antonio Vitobello
- Université de Bourgogne, Inserm U1231, Equipe GAD, Dijon, France
- CHU Dijon Bourgogne, FHU-TRANSLAD, Unité Fonctionnelle Innovation en Diagnostic Génomique Des Maladies Rares, 21000, Dijon, France
| | - Christel Thauvin-Robinet
- Université de Bourgogne, Inserm U1231, Equipe GAD, Dijon, France
- CHU Dijon Bourgogne, FHU-TRANSLAD, Unité Fonctionnelle Innovation en Diagnostic Génomique Des Maladies Rares, 21000, Dijon, France
- CHU Dijon Bourgogne, Centre de Génétique, Centre de Référence Maladies Rares «Déficiences Intellectuelles de Causes Rares», FHU-TRANSLAD, Dijon, France
| | - Laurence Faivre
- Université de Bourgogne, Inserm U1231, Equipe GAD, Dijon, France
- CHU Dijon Bourgogne, Centre de Génétique, Centre de Référence Maladies Rares «Anomalies du Développement et Syndromes Malformatifs», FHU-TRANSLAD, Dijon, France
| | - Sophie Nambot
- Université de Bourgogne, Inserm U1231, Equipe GAD, Dijon, France
- CHU Dijon Bourgogne, FHU-TRANSLAD, Unité Fonctionnelle Innovation en Diagnostic Génomique Des Maladies Rares, 21000, Dijon, France
- CHU Dijon Bourgogne, Centre de Génétique, Centre de Référence Maladies Rares «Anomalies du Développement et Syndromes Malformatifs», FHU-TRANSLAD, Dijon, France
| | - Aurore Garde
- Université de Bourgogne, Inserm U1231, Equipe GAD, Dijon, France
- CHU Dijon Bourgogne, Centre de Génétique, Centre de Référence Maladies Rares «Déficiences Intellectuelles de Causes Rares», FHU-TRANSLAD, Dijon, France
| | - Marjolaine Willems
- INserm U1183, Department of Clinical Genetics, Montpellier University, 34090 CHU Montpellier, Montpellier, France
| | - David Genevieve
- INserm U1183, Department of Clinical Genetics, Montpellier University, 34090 CHU Montpellier, Montpellier, France
| | - Gaël Nicolas
- Inserm U1245 and CHU Rouen, Department of Genetics and Reference Center for Developmental Disorders, Univ Rouen Normandie, 76000, Rouen, France
| | - Tiffany Busa
- Department of Medical Genetics, Timone Hospital, Marseille, France
| | - Annick Toutain
- Genetics Department, University Hospital, UMR 1253, iBrain, University of Tours, Inserm, Tours, France
| | - Marion Gérard
- APHP, Department of Genetics, Robert Debré Hospital, 75019, Paris, France
| | - Varoona Bizaoui
- Clinical Genetics and Neurodevelopmental Disorders, Centre Hospitalier de L'Estran, 50170, Pontorson, France
| | - Bertrand Isidor
- Service de Génétique Médicale, CHU de Nantes, 44000, Nantes, France
| | - Giuseppe Merla
- Laboratory of Regulatory and Functional Genomics, Fondazione IRCCS Casa Sollievo Della Sofferenza, San Giovanni Rotondo, Foggia, Italy
- Department of Molecular Medicine and Medical Biotechnology, University of Naples Federico II, Via S. Pansini 5, 80131, Naples, Italy
| | - Maria Accadia
- Servizio di Genetica Medica, Ospedale Cardinale G. Panico, Tricase, LE, Italy
| | - Charles E Schwartz
- Department of Pediatrics and Human Development, College of Human Medicine, Michigan State University, Grand Rapids, MI, 49503, USA
| | - Katrin Ounap
- Department of Clinical Genetics, Genetic and Personalized Medicine Clinic, Tartu University Hospital, Tartu, Estonia
- Department of Clinical Genetics, Institute of Clinical Medicine, University of Tartu, Tartu, Estonia
| | - Mariëtte J V Hoffer
- Department of Clinical Genetics, Leiden University Medical Center, Leiden, The Netherlands
| | - Marjan M Nezarati
- Genetics Program, North York General Hospital, Toronto, ON, M2K 1E1, Canada
| | | | | | | | - Alfredo Brusco
- Department of Medical Sciences, University of Torino, Via Santena 19, 10126, Turin, Italy
- Unit of Medical Genetics, Città Della Salute e Della Scienza Hospital, Turin, Italy
| | - Giovanni B Ferrero
- Department of Clinical and Biological Science, University of Torino, Turin, Italy
| | | | - Richard Sidlow
- Department of Medical Genetics and Metabolism, Valley Children's Hospital, Madera, CA, USA
| | - Alessandro Mussa
- Department of Public Health and Pediatric Sciences, University of Torino, Turin, Italy
- Pediatric Clinical Genetics Unit, Regina Margherita Childrens' Hospital, Turin, Italy
| | - Slavica Trajkova
- Department of Medical Sciences, University of Torino, Via Santena 19, 10126, Turin, Italy
| | - Emma McCann
- Liverpool Center for Genomic Medicine, Liverpool Women's Hospital, Liverpool, UK
| | - Henry J Mroczkowski
- Department of Pediatrics, Le Bonheur Children's Hospital, Memphis, TN, USA
- Division of Genetics, Department of Pediatrics, University of Tennessee Health Science Center, Memphis, TN, USA
| | - Sandra Jansen
- Amsterdam UMC, Department of Human Genetics, University of Amsterdam, Meibergdreef 9, 1105 AZ, Amsterdam, The Netherlands
- Amsterdam Reproduction & Development Research Institute, Amsterdam, The Netherlands
| | - Laura Donker-Kaat
- Department of Clinical Genetics, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Floor A M Duijkers
- Amsterdam UMC, Department of Human Genetics, University of Amsterdam, Meibergdreef 9, 1105 AZ, Amsterdam, The Netherlands
- Amsterdam Reproduction & Development Research Institute, Amsterdam, The Netherlands
| | - Kyra E Stuurman
- Department of Clinical Genetics, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Marcel M A M Mannens
- Amsterdam UMC, Department of Human Genetics, University of Amsterdam, Meibergdreef 9, 1105 AZ, Amsterdam, The Netherlands
- Amsterdam Reproduction & Development Research Institute, Amsterdam, The Netherlands
| | - Mariëlle Alders
- Amsterdam UMC, Department of Human Genetics, University of Amsterdam, Meibergdreef 9, 1105 AZ, Amsterdam, The Netherlands
- Amsterdam Reproduction & Development Research Institute, Amsterdam, The Netherlands
| | - Peter Henneman
- Amsterdam UMC, Department of Human Genetics, University of Amsterdam, Meibergdreef 9, 1105 AZ, Amsterdam, The Netherlands
- Amsterdam Reproduction & Development Research Institute, Amsterdam, The Netherlands
| | - Susan M White
- Victorian Clinical Genetics Services, Murdoch Children's Research Institute, Parkville, VIC, 3052, Australia
- Department of Paediatrics, University of Melbourne, Melbourne, Australia
| | - Bekim Sadikovic
- Verspeeten Clinical Genome Centre, London Health Sciences Centre, London, ON, N6A 5W9, Canada.
- Department of Pathology and Laboratory Medicine, Western University, London, ON, N6A 3K7, Canada.
| | - Mieke M van Haelst
- Amsterdam UMC, Department of Human Genetics, University of Amsterdam, Meibergdreef 9, 1105 AZ, Amsterdam, The Netherlands.
- Amsterdam Reproduction & Development Research Institute, Amsterdam, The Netherlands.
- Amsterdam UMC, Department of Paediatrics, Emma Children's Hospital, University of Amsterdam, Meibergdreef 9, 1105 AZ, Amsterdam, The Netherlands.
- Amsterdam UMC, Emma Center for Personalized Medicine, Amsterdam, The Netherlands.
| |
Collapse
|
5
|
Brown SSG, Westwater ML, Seidlitz J, Ziauddeen H, Fletcher PC. Hypothalamic volume is associated with body mass index. Neuroimage Clin 2023; 39:103478. [PMID: 37558541 PMCID: PMC10509524 DOI: 10.1016/j.nicl.2023.103478] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2023] [Revised: 06/19/2023] [Accepted: 07/18/2023] [Indexed: 08/11/2023]
Abstract
The hypothalamus is an important neuroendocrine hub for the control of appetite and satiety. In animal studies it has been established that hypothalamic lesioning or stimulation causes alteration to feeding behaviour and consequently body mass, and exposure to high calorie diets induces hypothalamic inflammation. These findings suggest that alterations in hypothalamic structure and function are both a cause and a consequence of changes to food intake. However, there is limited in vivo human data relating the hypothalamus to obesity or eating disorders, in part due to technical problems relating to its small size. Here, we used a novel automated segmentation algorithm to exploratorily investigate the relationship between hypothalamic volume, normalised to intracranial volume, and body mass index (BMI). The analysis was applied across four independent datasets comprising of young adults (total n = 1,351 participants) spanning a range of BMIs (13.3 - 47.8 kg/m2). We compared underweight (including individuals with anorexia nervosa), healthy weight, overweight and obese individuals in a series of complementary analyses. We report that overall hypothalamic volume is significantly larger in overweight and obese groups of young adults. This was also observed for a number of hypothalamic sub-regions. In the largest dataset (the HCP-Young Adult dataset (n = 1111)) there was a significant relationship between hypothalamic volume and BMI. We suggest that our findings of a positive relationship between hypothalamic volume and BMI is potentially consistent with hypothalamic inflammation as seen in animal models in response to high fat diet, although more research is needed to establish a causal relationship. Overall, we present novel, in vivo findings that link elevated BMI to altered hypothalamic structure. This has important implications for study of the neural mechanisms of obesity in humans.
Collapse
Affiliation(s)
- Stephanie S G Brown
- Department of Psychiatry, University of Cambridge, Addenbrookes Hospital, Cambridge CB2 0QQ, United Kingdom.
| | - Margaret L Westwater
- Department of Psychiatry, University of Cambridge, Addenbrookes Hospital, Cambridge CB2 0QQ, United Kingdom; Department of Psychiatry, University of Oxford, Warneford Hospital, Oxford OX3 7JX, United Kingdom
| | - Jakob Seidlitz
- Department of Child and Adolescent Psychiatry and Behavioral Science, Children's Hospital of Philadelphia, Philadelphia, PA, USA; Department of Psychiatry, University of Pennsylvania, Philadelphia, PA, USA; Lifespan Brain Institute of Children's Hospital of Philadelphia and University of Pennsylvania, Philadelphia, PA, USA
| | - Hisham Ziauddeen
- Department of Psychiatry, University of Cambridge, Addenbrookes Hospital, Cambridge CB2 0QQ, United Kingdom
| | - Paul C Fletcher
- Department of Psychiatry, University of Cambridge, Addenbrookes Hospital, Cambridge CB2 0QQ, United Kingdom; Wellcome Trust MRC Institute of Metabolic Science, University of Cambridge, Cambridge Biomedical Campus, Cambridge CB2 0QQ, United Kingdom; Cambridgeshire and Peterborough NHS Trust, United Kingdom
| |
Collapse
|
6
|
Xu Y, Jiang Z, Li H, Cai J, Jiang Y, Otiz-Guzman J, Xu Y, Arenkiel BR, Tong Q. Lateral septum as a melanocortin downstream site in obesity development. Cell Rep 2023; 42:112502. [PMID: 37171957 DOI: 10.1016/j.celrep.2023.112502] [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: 11/08/2022] [Revised: 03/27/2023] [Accepted: 04/26/2023] [Indexed: 05/14/2023] Open
Abstract
The melanocortin pathway is well established to be critical for body-weight regulation in both rodents and humans. Despite extensive studies focusing on this pathway, the downstream brain sites that mediate its action are not clear. Here, we found that, among the known paraventricular hypothalamic (PVH) neuron groups, those expressing melanocortin receptors 4 (PVHMc4R) preferably project to the ventral part of the lateral septum (LSv), a brain region known to be involved in emotional behaviors. Photostimulation of PVHMc4R neuron terminals in the LSv reduces feeding and causes aversion, whereas deletion of Mc4Rs or disruption of glutamate release from LSv-projecting PVH neurons causes obesity. In addition, disruption of AMPA receptor function in PVH-projected LSv neurons causes obesity. Importantly, chronic inhibition of PVH- or PVHMc4R-projected LSv neurons causes obesity associated with reduced energy expenditure. Thus, the LSv functions as an important node in mediating melanocortin action on body-weight regulation.
Collapse
Affiliation(s)
- Yuanzhong Xu
- Brown Foundation of Molecular Medicine for the Prevention of Human Diseases of McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX 77030, USA.
| | - Zhiying Jiang
- Brown Foundation of Molecular Medicine for the Prevention of Human Diseases of McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Hongli Li
- Brown Foundation of Molecular Medicine for the Prevention of Human Diseases of McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Jing Cai
- Brown Foundation of Molecular Medicine for the Prevention of Human Diseases of McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX 77030, USA; MD Anderson Cancer Center & UTHealth Houston Graduate School for Biomedical Sciences, University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Yanyan Jiang
- Brown Foundation of Molecular Medicine for the Prevention of Human Diseases of McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Joshua Otiz-Guzman
- Department of Molecular and Human Genetics and Department of Neuroscience, Baylor College of Medicine, and Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, TX, USA
| | - Yong Xu
- Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA
| | - Benjamin R Arenkiel
- Department of Molecular and Human Genetics and Department of Neuroscience, Baylor College of Medicine, and Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, TX, USA
| | - Qingchun Tong
- Brown Foundation of Molecular Medicine for the Prevention of Human Diseases of McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX 77030, USA; MD Anderson Cancer Center & UTHealth Houston Graduate School for Biomedical Sciences, University of Texas Health Science Center at Houston, Houston, TX 77030, USA; Department of Neurobiology and Anatomy of McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX 77030, USA.
| |
Collapse
|
7
|
Li H, Xu Y, Jiang Y, Jiang Z, Otiz-Guzman J, Morrill JC, Cai J, Mao Z, Xu Y, Arenkiel BR, Huang C, Tong Q. The melanocortin action is biased toward protection from weight loss in mice. Nat Commun 2023; 14:2200. [PMID: 37069175 PMCID: PMC10110624 DOI: 10.1038/s41467-023-37912-z] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2022] [Accepted: 04/05/2023] [Indexed: 04/19/2023] Open
Abstract
The melanocortin action is well perceived for its ability to regulate body weight bidirectionally with its gain of function reducing body weight and loss of function promoting obesity. However, this notion cannot explain the difficulty in identifying effective therapeutics toward treating general obesity via activation of the melanocortin action. Here, we provide evidence that altered melanocortin action is only able to cause one-directional obesity development. We demonstrate that chronic inhibition of arcuate neurons expressing proopiomelanocortin (POMC) or paraventricular hypothalamic neurons expressing melanocortin receptor 4 (MC4R) causes massive obesity. However, chronic activation of these neuronal populations failed to reduce body weight. Furthermore, gain of function of the melanocortin action through overexpression of MC4R, POMC or its derived peptides had little effect on obesity prevention or reversal. These results reveal a bias of the melanocortin action towards protection of weight loss and provide a neural basis behind the well-known, but mechanistically ill-defined, predisposition to obesity development.
Collapse
Affiliation(s)
- Hongli Li
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, 1200 Cailun Road, Shanghai, 201203, China
- Brown Foundation of Molecular Medicine for the Prevention of Human Diseases of McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX, 77030, USA
| | - Yuanzhong Xu
- Brown Foundation of Molecular Medicine for the Prevention of Human Diseases of McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX, 77030, USA
| | - Yanyan Jiang
- Brown Foundation of Molecular Medicine for the Prevention of Human Diseases of McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX, 77030, USA
| | - Zhiying Jiang
- Brown Foundation of Molecular Medicine for the Prevention of Human Diseases of McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX, 77030, USA
| | - Joshua Otiz-Guzman
- Department of Molecular and Human Genetics and Department of Neuroscience, Baylor College of Medicine, and Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, TX, USA
| | - Jessie C Morrill
- Brown Foundation of Molecular Medicine for the Prevention of Human Diseases of McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX, 77030, USA
- MD Anderson Cancer Center & UTHealth Graduate School for Biomedical Sciences, University of Texas Health Science at Houston, 77030, Houston, TX, USA
| | - Jing Cai
- Brown Foundation of Molecular Medicine for the Prevention of Human Diseases of McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX, 77030, USA
- MD Anderson Cancer Center & UTHealth Graduate School for Biomedical Sciences, University of Texas Health Science at Houston, 77030, Houston, TX, USA
| | - Zhengmei Mao
- Brown Foundation of Molecular Medicine for the Prevention of Human Diseases of McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX, 77030, USA
| | - Yong Xu
- Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, One Baylor Plaza, Houston, TX, 77030, USA
| | - Benjamin R Arenkiel
- Department of Molecular and Human Genetics and Department of Neuroscience, Baylor College of Medicine, and Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, TX, USA
| | - Cheng Huang
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, 1200 Cailun Road, Shanghai, 201203, China.
| | - Qingchun Tong
- Brown Foundation of Molecular Medicine for the Prevention of Human Diseases of McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX, 77030, USA.
- MD Anderson Cancer Center & UTHealth Graduate School for Biomedical Sciences, University of Texas Health Science at Houston, 77030, Houston, TX, USA.
- Department of Neurobiology and Anatomy of McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX, 77030, USA.
| |
Collapse
|
8
|
The Association of the Hypothalamic-Pituitary-Adrenal Axis with Appetite Regulation in Children with Fetal Alcohol Spectrum Disorders (FASDs). Nutrients 2023; 15:nu15061366. [PMID: 36986097 PMCID: PMC10053353 DOI: 10.3390/nu15061366] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2023] [Revised: 03/08/2023] [Accepted: 03/09/2023] [Indexed: 03/14/2023] Open
Abstract
Prenatal alcohol exposure causes growth impairment and a wide range of developmental, physical, and cognitive disorders in children, collectively referred to as fetal alcohol spectrum disorders (FASDs). In the course of FASDs, abnormalities can also affect eating behavior and nutritional status, but these problems have received little attention. Therefore, the aim of our study was to determine the levels of hormones involved in the action of the hypothalamic–pituitary–adrenal axis: proopiomelanocortin (POMC), cortisol, and adrenocorticotropic hormone (ACTH), in the serum of patients with FASDs. To our knowledge, none of these hormones studied have yet been evaluated in FASDs to date. We investigated 62 FASD patients and 23 healthy controls by applying an enzyme-linked immunosorbent method (ELISA). Fasting POMC levels were significantly lower in patients with FASDs (10.97 vs. 18,57 ng/mL, p = 0.039) compared to controls. However, there were no differences in cortisol concentrations. Additionally, the sex and subgroup status (fetal alcohol syndrome (FAS), neurobehavioral disorder associated with prenatal alcohol exposure (ND-PAE), and FASD risk) did not affect hormone levels. POMC was positively correlated with some clinical parameters such as age, BMI percentile, carbohydrate biomarkers, and ACTH. A positive correlation was observed between ACTH and cortisol levels, as well as ACTH and cholesterol levels. Data analysis showed no HPA axis abnormalities in the form of elevated serum cortisol and ACTH levels. Differences in POMC concentration may indicate the involvement and/or impairment of central nervous system structures in hormonal alterations in FASD individuals, caused by prenatal alcohol exposure. Hormonal dysregulation in FASDs can contribute to reduced growth and development, as well as many other disturbed processes, including neurological/neurodevelopmental dysfunctions. Further insightful studies involving a larger group of patients are needed to determine the potential impact of the measured hormones.
Collapse
|
9
|
Djafar JV, Johnson AM, Elvidge KL, Farrar MA. Childhood Dementia: A Collective Clinical Approach to Advance Therapeutic Development and Care. Pediatr Neurol 2023; 139:76-85. [PMID: 36571866 DOI: 10.1016/j.pediatrneurol.2022.11.015] [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: 07/29/2022] [Revised: 11/14/2022] [Accepted: 11/26/2022] [Indexed: 12/05/2022]
Abstract
Childhood dementias are a group of over 100 rare and ultra-rare pediatric conditions that are clinically characterized by chronic global neurocognitive decline. This decline is associated with a progressive loss of skills and shortened life expectancy. With an estimated incidence of one in 2800 births and less than 5% of the conditions having disease-modifying therapies, the impact is profound for patients and their families. Traditional research, care, and advocacy efforts have focused on individual disorders, or groups classified by molecular pathogenesis, and this has established robust foundations for further progress and collaboration. This review describes the shared and disease-specific clinical changes contributing to childhood dementia and considers these as potential indicators of underlying pathophysiologic processes. Like adult neurodegenerative syndromes, the heterogeneous phenotypes extend beyond cognitive decline and may involve changes in eating, motor function, pain, sleep, and behavior, mediated by physiological changes in neural networks. Importantly, these physiological phenotypes are associated with significant carer stress, anxiety, and challenges in care. These phenotypes are also pertinent for the development of therapeutics and optimization of best practice management. A collective approach to childhood dementia is anticipated to identify relevant biomarkers of prognosis or therapeutic efficacy, streamline the path from preclinical studies to clinical trials, increase opportunities for the development of multiple therapeutics, and refine clinical care.
Collapse
Affiliation(s)
- Jason V Djafar
- Discipline of Paediatrics and Child Health, School of Clinical Medicine, UNSW Medicine and Health, UNSW Sydney, Sydney, NSW, Australia; Department of Neurology, Sydney Children's Hospital Network, Sydney, NSW, Australia
| | - Alexandra M Johnson
- Discipline of Paediatrics and Child Health, School of Clinical Medicine, UNSW Medicine and Health, UNSW Sydney, Sydney, NSW, Australia; Department of Neurology, Sydney Children's Hospital Network, Sydney, NSW, Australia
| | | | - Michelle A Farrar
- Discipline of Paediatrics and Child Health, School of Clinical Medicine, UNSW Medicine and Health, UNSW Sydney, Sydney, NSW, Australia; Department of Neurology, Sydney Children's Hospital Network, Sydney, NSW, Australia.
| |
Collapse
|
10
|
Martins N, Castro C, Oliva-Teles A, Peres H. The Interplay between Central and Peripheral Systems in Feed Intake Regulation in European Seabass ( Dicentrarchus labrax) Juveniles. Animals (Basel) 2022; 12:ani12233287. [PMID: 36496811 PMCID: PMC9739057 DOI: 10.3390/ani12233287] [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: 11/21/2022] [Accepted: 11/23/2022] [Indexed: 11/29/2022] Open
Abstract
The present study aimed to evaluate the effects of feeding or feed deprivation on the orexigenic and anorexigenic responses at the central (whole brain) and peripheral (anterior and posterior intestine, stomach, and liver) system levels in European seabass. For this purpose, a group of fish (208 g) was fed a single meal daily for 8 days (fed group) and another group was feed-deprived for 8 days (unfed group). Compared to the fed group, in the whole brain, feed deprivation did not induce changes in npy, agrp1, and cart2 expression, but increased agrp2 and pomc1 expression. In the anterior intestine, feed deprivation increased cck expression, while in the posterior intestine, the npy expression increased and pyyb decreased. In the stomach, the ghr expression decreased regardless of the feeding status. The hepatic lep expression increased in the unfed fish. The present results suggest a feed intake regulation mechanism in European seabass similar to that observed in other teleosts.
Collapse
Affiliation(s)
- Nicole Martins
- Departamento de Biologia, Faculdade de Ciências da Universidade do Porto, Rua do Campo Alegre s/n, Edifício FC4, 4169-007 Porto, Portugal
- CIIMAR, Centro Interdisciplinar de Investigação Marinha e Ambiental, Universidade do Porto, Terminal de Cruzeiros do Porto de Leixões, Avenida General Norton de Matos s/n 289, 4450-208 Matosinhos, Portugal
- Correspondence:
| | - Carolina Castro
- FLATLANTIC—Atividades Piscícolas, S.A., Rua do Aceiros s/n, 3070-732 Praia de Mira, Portugal
| | - Aires Oliva-Teles
- Departamento de Biologia, Faculdade de Ciências da Universidade do Porto, Rua do Campo Alegre s/n, Edifício FC4, 4169-007 Porto, Portugal
- CIIMAR, Centro Interdisciplinar de Investigação Marinha e Ambiental, Universidade do Porto, Terminal de Cruzeiros do Porto de Leixões, Avenida General Norton de Matos s/n 289, 4450-208 Matosinhos, Portugal
| | - Helena Peres
- Departamento de Biologia, Faculdade de Ciências da Universidade do Porto, Rua do Campo Alegre s/n, Edifício FC4, 4169-007 Porto, Portugal
- CIIMAR, Centro Interdisciplinar de Investigação Marinha e Ambiental, Universidade do Porto, Terminal de Cruzeiros do Porto de Leixões, Avenida General Norton de Matos s/n 289, 4450-208 Matosinhos, Portugal
| |
Collapse
|
11
|
De Hert E, Verboven K, Wouters K, Jocken JWE, De Meester I. Prolyl Carboxypeptidase Activity Is Present in Human Adipose Tissue and Is Elevated in Serum of Obese Men with Type 2 Diabetes. Int J Mol Sci 2022; 23:13529. [PMID: 36362314 PMCID: PMC9655216 DOI: 10.3390/ijms232113529] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2022] [Revised: 10/31/2022] [Accepted: 11/02/2022] [Indexed: 07/30/2023] Open
Abstract
Prolyl carboxypeptidase (PRCP) is involved in metabolic disorders by hydrolyzing anorexigenic peptides. A link between serum PRCP activity and obesity has been reported, but its origin/source is still unclear. Previously proven correlations between human serum PRCP activity and the amount of adipose tissue may suggest that adipose tissue is an important source of circulating PRCP. We investigated PRCP activity in visceral, subcutaneous adipose tissue (VAT and SCAT), skeletal muscle tissue and serum of lean and obese men with or without type 2 diabetes (T2D). Correlations between PRCP activity, metabolic and biochemical parameters and immune cell populations were assessed. PRCP activity was the highest in VAT, compared to SCAT, and was very low in skeletal muscle tissue in the overall group. Serum PRCP activity was significantly higher in T2-diabetic obese men, compared to lean and obese non-diabetic men, and was positively correlated with glycemic control. A positive correlation was observed between serum PRCP activity and VAT immune cell populations, which might indicate that circulating PRCP activity is deriving rather from the immune fraction than from adipocytes. In conclusion, PRCP activity was observed in human adipose tissue for the first time and serum PRCP activity is correlated with T2D in obese men.
Collapse
Affiliation(s)
- Emilie De Hert
- Laboratory of Medical Biochemistry, Faculty of Pharmaceutical, Biomedical and Veterinary Sciences, University of Antwerp, 2610 Wilrijk, Belgium
| | - Kenneth Verboven
- REVAL—Rehabilitation Research Center, Faculty of Rehabilitation Sciences, Hasselt University, 3590 Diepenbeek, Belgium
- BIOMED—Biomedical Research Center, Faculty of Medicine and Life Sciences, Hasselt University, 3590 Diepenbeek, Belgium
| | - Kristiaan Wouters
- Cardiovascular Research Institute Maastricht (CARIM), Department of Internal Medicine, Maastricht University Medical Centre+, 6229 ER Maastricht, The Netherlands
| | - Johan W. E. Jocken
- Department of Human Biology, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Centre+, 6229 ER Maastricht, The Netherlands
| | - Ingrid De Meester
- Laboratory of Medical Biochemistry, Faculty of Pharmaceutical, Biomedical and Veterinary Sciences, University of Antwerp, 2610 Wilrijk, Belgium
| |
Collapse
|
12
|
Cho JH, Kim K, Cho HC, Lee J, Kim EK. Silencing of hypothalamic FGF11 prevents diet-induced obesity. Mol Brain 2022; 15:75. [PMID: 36064426 PMCID: PMC9447329 DOI: 10.1186/s13041-022-00962-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2022] [Accepted: 08/21/2022] [Indexed: 11/18/2022] Open
Abstract
Fibroblast growth factor 11 (FGF11) is a member of the intracellular fibroblast growth factor family. Here, we report the central role of FGF11 in the regulation of metabolism. Lentiviral injection of Fgf11 shRNA into the arcuate nucleus of the mouse hypothalamus decreased weight gain and fat mass, increased brown adipose tissue thermogenesis, and improved glucose and insulin intolerances under high-fat diet conditions. Fgf11 was expressed in the NPY–expressing neurons, and Fgf11 knockdown considerably decreased Npy expression and projection, leading to increased expression of tyrosine hydroxylase in the paraventricular nucleus. Mechanistically, FGF11 regulated Npy gene expression through the glycogen synthase kinase 3–cAMP response element-binding protein pathway. Our study defines the physiological significance of hypothalamic FGF11 in the regulation of metabolism in response to overnutrition such as high-fat diet.
Collapse
Affiliation(s)
- Jae Hyun Cho
- Department of Brain Sciences, Daegu Gyeongbuk Institute of Science and Technology, 333, Techno Jungang-Daero, Hyeonpung-Myeon, Daegu, Dalseonggun, 42988, South Korea
| | - Kyungchan Kim
- Department of Brain Sciences, Daegu Gyeongbuk Institute of Science and Technology, 333, Techno Jungang-Daero, Hyeonpung-Myeon, Daegu, Dalseonggun, 42988, South Korea
| | - Han Chae Cho
- Department of Brain Sciences, Daegu Gyeongbuk Institute of Science and Technology, 333, Techno Jungang-Daero, Hyeonpung-Myeon, Daegu, Dalseonggun, 42988, South Korea
| | - Jaemeun Lee
- Department of Brain Sciences, Daegu Gyeongbuk Institute of Science and Technology, 333, Techno Jungang-Daero, Hyeonpung-Myeon, Daegu, Dalseonggun, 42988, South Korea
| | - Eun-Kyoung Kim
- Department of Brain Sciences, Daegu Gyeongbuk Institute of Science and Technology, 333, Techno Jungang-Daero, Hyeonpung-Myeon, Daegu, Dalseonggun, 42988, South Korea. .,Neurometabolomics Research Center, Daegu Gyeongbuk Institute of Science and Technology, 333, Techno Jungang-Daero, Hyeonpung-Myeon, Daegu, Dalseonggun, 42988, South Korea.
| |
Collapse
|
13
|
Atractylodin alleviates cancer anorexia-cachexia syndrome by regulating NPY through hypothalamic Sirt1/AMPK axis-induced autophagy. Biochem Biophys Res Commun 2022; 625:154-160. [DOI: 10.1016/j.bbrc.2022.08.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2022] [Accepted: 08/04/2022] [Indexed: 11/23/2022]
|
14
|
Guzmán-Ruiz MA, Jiménez A, Cárdenas-Rivera A, Guerrero-Vargas NN, Organista-Juárez D, Guevara-Guzmán R. Regulation of Metabolic Health by an "Olfactory-Hypothalamic Axis" and Its Possible Implications for the Development of Therapeutic Approaches for Obesity and T2D. Cell Mol Neurobiol 2022; 42:1727-1743. [PMID: 33813677 PMCID: PMC11421737 DOI: 10.1007/s10571-021-01080-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2020] [Accepted: 03/12/2021] [Indexed: 12/12/2022]
Abstract
The olfactory system is responsible for the reception, integration and interpretation of odors. However, in the last years, it has been discovered that the olfactory perception of food can rapidly modulate the activity of hypothalamic neurons involved in the regulation of energy balance. Conversely, the hormonal signals derived from changes in the metabolic status of the body can also change the sensitivity of the olfactory system, suggesting that the bidirectional relationship established between the olfactory and the hypothalamic systems is key for the maintenance of metabolic homeostasis. In the first part of this review, we describe the possible mechanisms and anatomical pathways involved in the modulation of energy balance regulated by the olfactory system. Hence, we propose a model to explain its implication in the maintenance of the metabolic homeostasis of the organism. In the second part, we discuss how the olfactory system could be involved in the development of metabolic diseases such as obesity and type two diabetes and, finally, we propose the use of intranasal therapies aimed to regulate and improve the activity of the olfactory system that in turn will be able to control the neuronal activity of hypothalamic centers to prevent or ameliorate metabolic diseases.
Collapse
Affiliation(s)
- Mara Alaide Guzmán-Ruiz
- Laboratorio Sensorial, Departamento de Fisiología, Facultad de Medicina, Edificio A, 4º piso, Universidad Nacional Autónoma de México (UNAM), 04510, Ciudad de México, México.
| | - Adriana Jiménez
- Laboratorio Sensorial, Departamento de Fisiología, Facultad de Medicina, Edificio A, 4º piso, Universidad Nacional Autónoma de México (UNAM), 04510, Ciudad de México, México
| | - Alfredo Cárdenas-Rivera
- Centro de Investigación en Bioingeniería, Universidad de Ingeniería y Tecnología, Lima, Perú
| | - Natalí N Guerrero-Vargas
- Departamento de Anatomía, Facultad de Medicina, Universidad Nacional Autónoma de México, México City, México
| | - Diana Organista-Juárez
- Laboratorio Sensorial, Departamento de Fisiología, Facultad de Medicina, Edificio A, 4º piso, Universidad Nacional Autónoma de México (UNAM), 04510, Ciudad de México, México
| | - Rosalinda Guevara-Guzmán
- Laboratorio Sensorial, Departamento de Fisiología, Facultad de Medicina, Edificio A, 4º piso, Universidad Nacional Autónoma de México (UNAM), 04510, Ciudad de México, México.
| |
Collapse
|
15
|
Wabitsch M, Farooqi S, Flück CE, Bratina N, Mallya UG, Stewart M, Garrison J, van den Akker E, Kühnen P. Natural History of Obesity Due to POMC, PCSK1, and LEPR Deficiency and the Impact of Setmelanotide. J Endocr Soc 2022; 6:bvac057. [PMID: 35528826 PMCID: PMC9070354 DOI: 10.1210/jendso/bvac057] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/27/2021] [Indexed: 11/19/2022] Open
Abstract
Context Rare homozygous or biallelic variants in POMC, PCSK1, and LEPR can disrupt signaling through the melanocortin-4 receptor (MC4R) pathway, resulting in hyperphagia and severe early-onset obesity. In pivotal Phase 3 clinical trials, treatment with the MC4R agonist setmelanotide reduced hunger and weight in patients with obesity due to proopiomelanocortin (POMC), proprotein convertase subtilisin/kexin type 1 (PCSK1), or leptin receptor (LEPR) deficiency. Objective To characterize the historical weight trajectory in these patients. Methods This analysis included data from 2 pivotal single-arm, open-label, Phase 3 trials (NCT02896192, NCT03287960). These were multicenter trials. Patients had obesity due to POMC/PCSK1 or LEPR deficiency. During the trial, patients were treated with setmelanotide. Historical data on measured weight and height were obtained during screening. Results A total of 17 patients (POMC, n = 8; PCSK1, n = 1; LEPR, n = 8) with historical weight and height data were included in this analysis. Before setmelanotide treatment, patients with obesity due to POMC/PCSK1 or LEPR deficiency were above the 95th percentile for weight throughout childhood, demonstrated continuous weight gain, and did not show long-term weight loss upon interventions (eg, diet, surgery, exercise). Setmelanotide treatment attenuated weight and body mass index trajectories over the observation period of 1 year. Conclusion In patients with POMC, PCSK1, or LEPR deficiency, traditional interventions for weight loss had limited impact on the trajectory of severe early-onset obesity. However, setmelanotide treatment attenuated weight and body mass index trajectories and led to weight loss associated with health benefits in most individuals.
Collapse
Affiliation(s)
- Martin Wabitsch
- Division of Pediatric Endocrinology and Diabetes, Center for Rare Endocrine Diseases, Department of Pediatrics and Adolescent Medicine, University of Ulm, Ulm, Germany
| | - Sadaf Farooqi
- Wellcome-MRC Institute of Metabolic Science and NIHR Cambridge Biomedical Research Centre, University of Cambridge, Cambridge, United Kingdom
| | - Christa E Flück
- Paediatric Endocrinology, Diabetology and Metabolism, Department of Paediatrics and Department of BioMedical Research, Bern University Hospital Inselspital and University of Bern, Bern, Switzerland
| | - Natasa Bratina
- Department of Endocrinology, Diabetes and Metabolic Diseases, University Children's Hospital, University Medical Centre Ljubljana, Ljubljana, Slovenia
| | | | | | | | - Erica van den Akker
- Division of Pediatric Endocrinology, Department of Pediatrics, Sophia Children’s Hospital and Obesity Center CGG, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Peter Kühnen
- Institute for Experimental Pediatric Endocrinology, Charité Universitätsmedizin Berlin, Berlin, Germany
| |
Collapse
|
16
|
Relationship between the double burden of malnutrition and mental health in overweight and obese adult women. J Nutr Sci 2022; 11:e12. [PMID: 35291277 PMCID: PMC8889085 DOI: 10.1017/jns.2022.7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2021] [Revised: 12/22/2021] [Accepted: 01/17/2022] [Indexed: 11/23/2022] Open
Abstract
The co-existence of overweight or obesity with concurrent deficiency of one or more nutrients is referred to as double burden of malnutrition (DBM), and numerous mental health impairments have been associated with a variety of nutrient deficiencies. Although DBM is relevant for several health outcomes, the ubiquitous involvement of vitamin D across multiple systems and tissues suggests D insufficiency as a viable target for nutritional modification. The present study aimed to evaluate the contribution of DBM and mental health among adult women. Study participants included 300 women, aged 18–59 years, who presented to one of the 25 health centres in Tehran. Participants with a body mass index (BMI) of greater than 25 kg/m2 and a plasma concentration of 25-hydroxy vitamin D [25(OH)D] of >20 ng/ml were considered to have DBM. The 147-item food frequency questionnaire was used to estimate their dietary intake. Mental health status was assessed using the depression, anxiety and stress scales-21 (DASS-21). The mean ± standard deviation age, weight and BMI of the participants were 36⋅49 ± 8⋅38, 80⋅89 ± 12⋅45 kg and 31⋅04 ± 4⋅31 kg/m2, respectively. DBM was significantly associated with stress, after adjusting for potential confounders, including age, energy and marital status in model 1 (OR = 1⋅28, 95 % confidence interval (CI) 1⋅00, 1⋅65, P < 0⋅04) v. the crude model (OR = 1⋅22; 95 % CI 0⋅96, 1⋅55, P = 0⋅09). No significant association was seen among DBM and DASS-21 outcomes. In this cross-sectional study, stress and DBM were significantly associated. While vitamin D insufficiency was associated with mental health and obesity in opposing directions. Elucidation of whether vitamin D supplementation can improve mental health impairments requires further evaluation.
Collapse
|
17
|
Vohra MS, Benchoula K, Serpell CJ, Hwa WE. AgRP/NPY and POMC neurons in the arcuate nucleus and their potential role in treatment of obesity. Eur J Pharmacol 2022; 915:174611. [PMID: 34798121 DOI: 10.1016/j.ejphar.2021.174611] [Citation(s) in RCA: 84] [Impact Index Per Article: 28.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2021] [Revised: 10/27/2021] [Accepted: 10/27/2021] [Indexed: 02/08/2023]
Abstract
Obesity is a major health crisis affecting over a third of the global population. This multifactorial disease is regulated via interoceptive neural circuits in the brain, whose alteration results in excessive body weight. Certain central neuronal populations in the brain are recognised as crucial nodes in energy homeostasis; in particular, the hypothalamic arcuate nucleus (ARC) region contains two peptide microcircuits that control energy balance with antagonistic functions: agouti-related peptide/neuropeptide-Y (AgRP/NPY) signals hunger and stimulates food intake; and pro-opiomelanocortin (POMC) signals satiety and reduces food intake. These neuronal peptides levels react to energy status and integrate signals from peripheral ghrelin, leptin, and insulin to regulate feeding and energy expenditure. To manage obesity comprehensively, it is crucial to understand cellular and molecular mechanisms of information processing in ARC neurons, since these regulate energy homeostasis. Importantly, a specific strategy focusing on ARC circuits needs to be devised to assist in treating obese patients and maintaining weight loss with minimal or no side effects. The aim of this review is to elucidate the recent developments in the study of AgRP-, NPY- and POMC-producing neurons, specific to their role in controlling metabolism. The impact of ghrelin, leptin, and insulin signalling via action of these neurons is also surveyed, since they also impact energy balance through this route. Lastly, we present key proteins, targeted genes, compounds, drugs, and therapies that actively work via these neurons and could potentially be used as therapeutic targets for treating obesity conditions.
Collapse
Affiliation(s)
- Muhammad Sufyan Vohra
- School of Medicine, Faculty of Health and Medical Sciences, Taylor's University Lakeside Campus, 47500, Subang Jaya, Selangor Darul Ehsan, Malaysia
| | - Khaled Benchoula
- School of Medicine, Faculty of Health and Medical Sciences, Taylor's University Lakeside Campus, 47500, Subang Jaya, Selangor Darul Ehsan, Malaysia
| | - Christopher J Serpell
- School of Physical Sciences, Ingram Building, University of Kent, Canterbury, Kent, CT2 7NH, United Kingdom
| | - Wong Eng Hwa
- School of Medicine, Faculty of Health and Medical Sciences, Taylor's University Lakeside Campus, 47500, Subang Jaya, Selangor Darul Ehsan, Malaysia.
| |
Collapse
|
18
|
Andrews ZB. Neurobiology: How to ask a mouse if it's hungry. Curr Biol 2021; 31:R1056-R1058. [PMID: 34520719 DOI: 10.1016/j.cub.2021.07.065] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Neural circuits influence food intake by responding to interoceptive hunger cues and/or hedonic cues. A new study utilizes a hunger discrimination behavioural task combined with opto- and chemo-genetic manipulation to identify hunger and non-hunger sensing neural circuits driving food intake.
Collapse
Affiliation(s)
- Zane B Andrews
- Department of Physiology, Monash University, Clayton, VIC 3800, Australia.
| |
Collapse
|
19
|
Shakya M, White A, Verchere CB, Low MJ, Lindberg I. Mice lacking PC1/3 expression in POMC-expressing cells do not develop obesity. Endocrinology 2021; 162:6167813. [PMID: 33693631 PMCID: PMC8253230 DOI: 10.1210/endocr/bqab055] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/07/2020] [Indexed: 02/06/2023]
Abstract
Pro-opiomelanocortin (POMC) neurons form an integral part of the central melanocortin system regulating food intake and energy expenditure. Genetic and pharmacological studies have revealed that defects in POMC synthesis, processing, and receptor signaling lead to obesity. It is well established that POMC is extensively processed by a series of enzymes, including prohormone convertases PC1/3 and PC2, and that genetic insufficiency of both PC1/3 and POMC is strongly associated with obesity risk. However, whether PC1/3-mediated POMC processing is absolutely tied to body weight regulation is not known. To investigate this question, we generated a Pomc-CreER T2; Pcsk1 lox/lox mouse model in which Pcsk1 is specifically and temporally knocked out in POMC-expressing cells of adult mice by injecting tamoxifen at eight weeks of age. We then measured the impact of Pcsk1 deletion on POMC cleavage to ACTH and α-MSH, and on body weight. In whole pituitary, POMC cleavage was significantly impacted by the loss of Pcsk1, while hypothalamic POMC-derived peptide levels remained similar in all genotypes. However, intact POMC levels were greatly elevated in Pomc-CreER T2; Pcsk1 lox/lox mice. Males expressed two-fold greater levels of pituitary PC1/3 protein than females, consistent with their increased POMC cleavage. Past studies show that mice with germline removal of PC1/3 do not develop obesity, while mice expressing mutant PC1/3 forms do develop obesity. We conclude that obesity pathways are not disrupted by PC1/3 loss solely in POMC-expressing cells, further disfavoring the idea that alterations in POMC processing underlie obesity in PCSK1 deficiency.
Collapse
Affiliation(s)
- Manita Shakya
- Department of Anatomy and Neurobiology, University of
Maryland-Baltimore, Baltimore, MD 21201,
USA
| | - Surbhi
- Department of Molecular & Integrative Physiology,
University of Michigan, Ann Arbor, MI
481091, USA
| | - Anne White
- Division of Diabetes, Endocrinology and Gastroenterology,
University of Manchester, Manchester, M13
9PT, United Kingdom
| | - C Bruce Verchere
- Departments of Pathology & Laboratory Medicine and
Surgery, University of British Columbia, British
Columbia, V5Z 4H4, Canada
| | - Malcolm J Low
- Department of Molecular & Integrative Physiology,
University of Michigan, Ann Arbor, MI
481091, USA
| | - Iris Lindberg
- Department of Anatomy and Neurobiology, University of
Maryland-Baltimore, Baltimore, MD 21201,
USA
- Correspondence: Iris Lindberg, PhD,
Department of Anatomy and Neurobiology, 20 Penn St., HSF2, S267, University of
Maryland-Baltimore, Baltimore, MD 21201, USA. E-mail:
| |
Collapse
|
20
|
Li L, Xu Y, Zheng J, Kuang Z, Zhang C, Li N, Lin G, Zhang C. Pharmacological modulation of dual melanocortin-4 receptor signaling by melanocortin receptor accessory proteins in the Xenopus laevis. J Cell Physiol 2021; 236:5980-5993. [PMID: 33501674 DOI: 10.1002/jcp.30280] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2020] [Revised: 12/29/2020] [Accepted: 01/04/2021] [Indexed: 01/09/2023]
Abstract
Physiological modulation of melanocortin-4 receptor (MC4R) signaling by MRAP2 proteins plays an indispensable role in appetite control and energy homeostasis in the central nervous system. Great interspecies differences of the interaction and regulation of melanocortin receptors by MRAP protein family have been reported in several diploid vertebrates but never been investigated in the tetrapod amphibian Xenopus laevis. Here, we performed phylogenetic and synteny-based analyses to explore the evolutionary aspects of dual copies of X. laevis MC4R (xlMC4R) and MRAP2 (xlMRAP2) proteins. Our data showed that xlMRAPs directly interacted with xlMC4Rs on the cell surface as a functional antiparallel dimeric topology and pharmacological studies suggested a homology specific regulatory pattern of the melanocortin system in X. laevis.
Collapse
Affiliation(s)
- Lei Li
- Translational Medical Center for Stem Cell Therapy and Institute for Regenerative Medicine, Shanghai East Hospital, Shanghai Key Laboratory of Signaling and Disease Research, Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Tongji University, Shanghai, China
| | - Ying Xu
- Translational Medical Center for Stem Cell Therapy and Institute for Regenerative Medicine, Shanghai East Hospital, Shanghai Key Laboratory of Signaling and Disease Research, Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Tongji University, Shanghai, China
| | - Jihong Zheng
- Translational Medical Center for Stem Cell Therapy and Institute for Regenerative Medicine, Shanghai East Hospital, Shanghai Key Laboratory of Signaling and Disease Research, Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Tongji University, Shanghai, China
| | - Zhe Kuang
- Translational Medical Center for Stem Cell Therapy and Institute for Regenerative Medicine, Shanghai East Hospital, Shanghai Key Laboratory of Signaling and Disease Research, Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Tongji University, Shanghai, China
| | - Cong Zhang
- Translational Medical Center for Stem Cell Therapy and Institute for Regenerative Medicine, Shanghai East Hospital, Shanghai Key Laboratory of Signaling and Disease Research, Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Tongji University, Shanghai, China
| | - Na Li
- Yantai Derui Bio-Tech Co., Ltd., Yantai, Shandong, China
| | - Gufa Lin
- Key Laboratory of Spine and Spinal Cord Injury Repair and Regeneration of Ministry of Education, Orthopaedic Department of Tongji Hospital, School of Life Sciences and Technology, Tongji University, Shanghai, China
| | - Chao Zhang
- Translational Medical Center for Stem Cell Therapy and Institute for Regenerative Medicine, Shanghai East Hospital, Shanghai Key Laboratory of Signaling and Disease Research, Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Tongji University, Shanghai, China
| |
Collapse
|
21
|
Yardman-Frank JM, Fisher DE. Skin pigmentation and its control: From ultraviolet radiation to stem cells. Exp Dermatol 2020; 30:560-571. [PMID: 33320376 DOI: 10.1111/exd.14260] [Citation(s) in RCA: 93] [Impact Index Per Article: 18.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
In the light of substantial discoveries in epithelial and hair pigmentation pathophysiology, this review summarizes the current understanding of skin pigmentation mechanisms. Melanocytes are pigment-producing cells, and their key regulating transcription factor is the melanocyte-specific microphthalmia-associated transcription factor (m-MITF). Ultraviolet (UV) radiation is a unique modulator of skin pigmentation influencing tanning pathways. The delayed tanning pathway occurs as UVB produces keratinocyte DNA damage, causing p53-mediated expression of the pro-opiomelanocortin (POMC) gene that is processed to release α-melanocyte-stimulating hormone (α-MSH). α-MSH stimulates the melanocortin 1 receptor (MC1R) on melanocytes, leading to m-MITF expression and melanogenesis. POMC cleavage also releases β-endorphin, which creates a neuroendocrine pathway that promotes UV-seeking behaviours. Mutations along the tanning pathway can affect pigmentation and increase the risk of skin malignancies. MC1R variants have received considerable attention, yet the allele is highly polymorphic with varied phenotypes. Vitiligo presents with depigmented skin lesions due to autoimmune destruction of melanocytes. UVB phototherapy stimulates melanocyte stem cells in the hair bulge to undergo differentiation and upwards migration resulting in perifollicular repigmentation of vitiliginous lesions, which is under sophisticated signalling control. Melanocyte stem cells, normally quiescent, undergo cyclic activation/differentiation and downward migration with the hair cycle, providing pigment to hair follicles. Physiological hair greying results from progressive loss of melanocyte stem cells and can be accelerated by acute stress-induced, sympathetic driven hyperproliferation of the melanocyte stem cells. Ultimately, by reviewing the pathways governing epithelial and follicular pigmentation, numerous areas of future research and potential points of intervention are highlighted.
Collapse
Affiliation(s)
| | - David E Fisher
- Department of Dermatology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
22
|
Female mice exposed to low doses of dioxin during pregnancy and lactation have increased susceptibility to diet-induced obesity and diabetes. Mol Metab 2020; 42:101104. [PMID: 33075544 PMCID: PMC7683344 DOI: 10.1016/j.molmet.2020.101104] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/13/2020] [Revised: 10/05/2020] [Accepted: 10/13/2020] [Indexed: 12/30/2022] Open
Abstract
Objective Exposure to persistent organic pollutants is consistently associated with increased diabetes risk in humans. We investigated the short- and long-term impact of transient low-dose dioxin (2,3,7,8-tetrachlorodibenzo-p-dioxin, TCDD) exposure during pregnancy and lactation on glucose homeostasis and beta cell function in female mice, including their response to a metabolic stressor later in life. Methods Female mice were injected with either corn oil (CO; vehicle control) or 20 ng/kg/d TCDD 2x/week throughout mating, pregnancy and lactation, and then tracked for 6–10 weeks after chemical exposure stopped. A subset of CO- and TCDD-exposed dams was then transferred to a 45% high-fat diet (HFD) or remained on a standard chow diet for an additional 11 weeks to assess the long-term effects of TCDD on adaptability to a metabolic stressor. To summarize, female mice were transiently exposed to TCDD and then subsequently tracked beyond when TCDD had been excreted to identify lasting metabolic effects of TCDD exposure. Results TCDD-exposed dams were hypoglycemic at birth but otherwise had normal glucose homeostasis during and post-TCDD exposure. However, TCDD-exposed dams on a chow diet were modestly heavier than controls starting 5 weeks after the last TCDD injection, and their weight gain accelerated after transitioning to a HFD. TCDD-exposed dams also had an accelerated onset of hyperglycemia, impaired glucose-induced plasma insulin levels, reduced islet size, increased MAFA-ve beta cells, and increased proinsulin accumulation following HFD feeding compared to controls. Overall, our study demonstrates that low-dose TCDD exposure during pregnancy has minimal effects on metabolism during the period of active exposure, but has detrimental long-term effects on metabolic adaptability to HFD feeding. Conclusions Our study suggests that transient low-dose TCDD exposure in female mice impairs metabolic adaptability to HFD feeding, demonstrating that dioxin exposure may be a contributing factor to obesity and diabetes pathogenesis in females. Female mice exposed to TCDD during pregnancy are hypoglycemic at birth. TCDD exposure promotes weight gain long after exposure ceases. TCDD-exposed dams fed a high-fat diet have accelerated onset of glucose intolerance. TCDDHFD dams have defects in islet morphology and beta cell function.
Collapse
|
23
|
Prezotto LD, Thorson JF, Prevot V, Redmer DA, Grazul-Bilska AT. Nutritionally induced tanycytic plasticity in the hypothalamus of adult ewes. Domest Anim Endocrinol 2020; 72:106438. [PMID: 32388344 DOI: 10.1016/j.domaniend.2020.106438] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/31/2019] [Revised: 12/27/2019] [Accepted: 01/16/2020] [Indexed: 02/07/2023]
Abstract
The blood-brain barrier regulates the transport of molecules that convey global energetic status to the feeding circuitry within the hypothalamus. Capillaries within the median eminence (ME) and tight junctions between tanycytes lining the third ventricle (3V) are critical components of this barrier. Herein, we tested the hypothesis that altering the plane of nutrition results in the structural reorganization of tanycytes, tight junctions, and capillary structure within the medial basal hypothalamus. Proopiomelanocortin (POMC) neuronal content within the arcuate nucleus of the hypothalamus (ARC) was also assessed to test whether reduced nutritional status improved access of nutrients to the ARC, while decreasing the access of nutrients of overfed animals. Multiparous, nongestating ewes were stratified by weight and randomly assigned to dietary treatments offered for 75 d: 200% of dietary recommendations (overfed), 100% of dietary recommendations (control), or 60% of dietary recommendations (underfed). The number of POMC-expressing neurons within the ARC was increased (P ≤ 0.002) in underfed ewes. Overfeeding increased (P ≤ 0.01) tanycyte cellular process penetration and density compared with control and underfeeding as assessed using vimentin immunostaining. Immunostaining of tight junctions along the wall of the 3V did not differ (P = 0.32) between treatments. No differences were observed in capillary density (P = 0.21) or classification (P ≥ 0.47) within the ME. These results implicate that changes within the satiety center and morphology of tanycytes within the ARC occur as an adaptation to nutrient availability.
Collapse
Affiliation(s)
- L D Prezotto
- Nutritional & Reproductive Physiology Laboratory, Northern Agricultural Research Center, Montana State University, 3710 Assinniboine Road, Havre, MT 59501, USA.
| | - J F Thorson
- Nutritional & Reproductive Physiology Laboratory, Northern Agricultural Research Center, Montana State University, 3710 Assinniboine Road, Havre, MT 59501, USA
| | - V Prevot
- INSERM, Laboratory of Development and Plasticity of the Neuroendocrine Brain, Jean-Pierre Aubert Research Centre, U1172, Lille, France
| | - D A Redmer
- Department of Animal Sciences, North Dakota State University, Fargo, ND 58108, USA
| | - A T Grazul-Bilska
- Department of Animal Sciences, North Dakota State University, Fargo, ND 58108, USA
| |
Collapse
|
24
|
Georgescu T, Lyons D, Doslikova B, Garcia AP, Marston O, Burke LK, Chianese R, Lam BYH, Yeo GSH, Rochford JJ, Garfield AS, Heisler LK. Neurochemical Characterization of Brainstem Pro-Opiomelanocortin Cells. Endocrinology 2020; 161:bqaa032. [PMID: 32166324 PMCID: PMC7102873 DOI: 10.1210/endocr/bqaa032] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/08/2019] [Accepted: 03/10/2020] [Indexed: 02/08/2023]
Abstract
Genetic research has revealed pro-opiomelanocortin (POMC) to be a fundamental regulator of energy balance and body weight in mammals. Within the brain, POMC is primarily expressed in the arcuate nucleus of the hypothalamus (ARC), while a smaller population exists in the brainstem nucleus of the solitary tract (POMCNTS). We performed a neurochemical characterization of this understudied population of POMC cells using transgenic mice expressing green fluorescent protein (eGFP) under the control of a POMC promoter/enhancer (PomceGFP). Expression of endogenous Pomc mRNA in the nucleus of the solitary tract (NTS) PomceGFP cells was confirmed using fluorescence-activating cell sorting (FACS) followed by quantitative PCR. In situ hybridization histochemistry of endogenous Pomc mRNA and immunohistochemical analysis of eGFP revealed that POMC is primarily localized within the caudal NTS. Neurochemical analysis indicated that POMCNTS is not co-expressed with tyrosine hydroxylase (TH), glucagon-like peptide 1 (GLP-1), cholecystokinin (CCK), brain-derived neurotrophic factor (BDNF), nesfatin, nitric oxide synthase 1 (nNOS), seipin, or choline acetyltransferase (ChAT) cells, whereas 100% of POMCNTS is co-expressed with transcription factor paired-like homeobox2b (Phox2b). We observed that 20% of POMCNTS cells express receptors for adipocyte hormone leptin (LepRbs) using a PomceGFP:LepRbCre:tdTOM double-reporter line. Elevations in endogenous or exogenous leptin levels increased the in vivo activity (c-FOS) of a small subset of POMCNTS cells. Using ex vivo slice electrophysiology, we observed that this effect of leptin on POMCNTS cell activity is postsynaptic. These findings reveal that a subset of POMCNTS cells are responsive to both changes in energy status and the adipocyte hormone leptin, findings of relevance to the neurobiology of obesity.
Collapse
Affiliation(s)
- Teodora Georgescu
- Rowett Institute, University of Aberdeen, Foresterhill, Aberdeen, UK
- Department of Pharmacology, University of Cambridge, Cambridge, UK
- Centre for Neuroendocrinology & Department of Anatomy, University of Otago, Dunedin, New Zealand
| | - David Lyons
- Rowett Institute, University of Aberdeen, Foresterhill, Aberdeen, UK
| | | | - Ana Paula Garcia
- Department of Pharmacology, University of Cambridge, Cambridge, UK
| | - Oliver Marston
- Department of Pharmacology, University of Cambridge, Cambridge, UK
| | - Luke K Burke
- Department of Pharmacology, University of Cambridge, Cambridge, UK
| | | | - Brian Y H Lam
- MRC Metabolic Diseases Unit, University of Cambridge Metabolic Research Laboratories, Wellcome Trust-MRC Institute of Metabolic Science, Addenbrooke’s Hospital, Cambridge, UK
| | - Giles S H Yeo
- MRC Metabolic Diseases Unit, University of Cambridge Metabolic Research Laboratories, Wellcome Trust-MRC Institute of Metabolic Science, Addenbrooke’s Hospital, Cambridge, UK
| | | | | | - Lora K Heisler
- Rowett Institute, University of Aberdeen, Foresterhill, Aberdeen, UK
- Department of Pharmacology, University of Cambridge, Cambridge, UK
| |
Collapse
|
25
|
Goudarzi M, Nahavandi A, Mehrabi S, Eslami M, Shahbazi A, Barati M. Valproic acid administration exerts protective effects against stress-related anhedonia in rats. J Chem Neuroanat 2020; 105:101768. [DOI: 10.1016/j.jchemneu.2020.101768] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2019] [Revised: 02/12/2020] [Accepted: 02/12/2020] [Indexed: 12/22/2022]
|
26
|
Emamgholipour S, Ebrahimi R, Bahiraee A, Niazpour F, Meshkani R. Acetylation and insulin resistance: a focus on metabolic and mitogenic cascades of insulin signaling. Crit Rev Clin Lab Sci 2020:1-19. [DOI: 10.1080/10408363.2019.1699498] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Affiliation(s)
- Solaleh Emamgholipour
- Department of Clinical Biochemistry, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Reyhane Ebrahimi
- Department of Clinical Biochemistry, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
- Students’ Scientific Research Center (SSRC), Tehran University of Medical Sciences, Tehran, Iran
| | - Alireza Bahiraee
- Department of Medical Genetics, Faculty of Medicine, Hormozgan University of Medical Sciences, Bandar Abbas, Iran
| | - Farshad Niazpour
- Department of Clinical Biochemistry, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Reza Meshkani
- Department of Clinical Biochemistry, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
27
|
Erfurth EM. Diagnosis, Background, and Treatment of Hypothalamic Damage in Craniopharyngioma. Neuroendocrinology 2020; 110:767-779. [PMID: 32580186 PMCID: PMC7490511 DOI: 10.1159/000509616] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/11/2020] [Accepted: 06/22/2020] [Indexed: 12/24/2022]
Abstract
Craniopharyngiomas (CP) are rare brain tumors managed primarily with surgery and radiotherapy. There are 2 phenotypes of CP, i.e., one with a rather good outcome without hypothalamic damage and another with hypothalamic damage. With hypothalamic damage, progressive disease with recurrent operations and additional cranial radiotherapy often result in hypothalamic obesity, an affected psychosocial life, and cognitive dysfunction. The morbidity and mortality are increased for particularly cerebrovascular diseases. Preoperative hypothalamic involvement to predict hypothalamic damage is important for decision making for hypothalamus-sparing surgery. Also a postoperative hypothalamic damage evaluation with the use of hypothalamus volume measurement can predict hypothalamic obesity, which is important for early treatment options. The morbidity of CP includes cognitive dysfunction with attention deficits and impaired episodic memory and processing speed. Again patients with hypothalamic damage are more affected. Treatment options of hypothalamic obesity in the chronic phase are scarce and not convincingly successful. The most optimal situation is to try to hinder or stop the evolution of hypothalamic obesity. Prevention of hypothalamic damage is recommended, with special regard to hypothalamus-sparing therapeutic approaches that respect the integrity of essential nuclei located in both the medial and the posterior hypothalamic areas.
Collapse
Affiliation(s)
- Eva-Marie Erfurth
- Department of Endocrinology, Skåne University Hospital, Lund, Sweden,
- Clinical Sciences, Lund University, Lund, Sweden,
| |
Collapse
|
28
|
Caron A, Briscoe DM, Richard D, Laplante M. DEPTOR at the Nexus of Cancer, Metabolism, and Immunity. Physiol Rev 2018; 98:1765-1803. [PMID: 29897294 DOI: 10.1152/physrev.00064.2017] [Citation(s) in RCA: 56] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
DEP domain-containing mechanistic target of rapamycin (mTOR)-interacting protein (DEPTOR) is an important modulator of mTOR, a kinase at the center of two important protein complexes named mTORC1 and mTORC2. These highly studied complexes play essential roles in regulating growth, metabolism, and immunity in response to mitogens, nutrients, and cytokines. Defects in mTOR signaling have been associated with the development of many diseases, including cancer and diabetes, and approaches aiming at modulating mTOR activity are envisioned as an attractive strategy to improve human health. DEPTOR interaction with mTOR represses its kinase activity and rewires the mTOR signaling pathway. Over the last years, several studies have revealed key roles for DEPTOR in numerous biological and pathological processes. Here, we provide the current state of the knowledge regarding the cellular and physiological functions of DEPTOR by focusing on its impact on the mTOR pathway and its role in promoting health and disease.
Collapse
Affiliation(s)
- Alexandre Caron
- Department of Internal Medicine, Division of Hypothalamic Research, The University of Texas Southwestern Medical Center , Dallas, Texas ; Transplant Research Program, Boston Children's Hospital , Boston, Massachusetts ; Department of Pediatrics, Harvard Medical School , Boston, Massachusetts ; Centre de Recherche de l'Institut Universitaire de Cardiologie et de Pneumologie de Québec (CRIUCPQ), Faculté de Médecine, Université Laval , Québec , Canada ; and Centre de Recherche sur le Cancer de l'Université Laval, Université Laval , Québec , Canada
| | - David M Briscoe
- Department of Internal Medicine, Division of Hypothalamic Research, The University of Texas Southwestern Medical Center , Dallas, Texas ; Transplant Research Program, Boston Children's Hospital , Boston, Massachusetts ; Department of Pediatrics, Harvard Medical School , Boston, Massachusetts ; Centre de Recherche de l'Institut Universitaire de Cardiologie et de Pneumologie de Québec (CRIUCPQ), Faculté de Médecine, Université Laval , Québec , Canada ; and Centre de Recherche sur le Cancer de l'Université Laval, Université Laval , Québec , Canada
| | - Denis Richard
- Department of Internal Medicine, Division of Hypothalamic Research, The University of Texas Southwestern Medical Center , Dallas, Texas ; Transplant Research Program, Boston Children's Hospital , Boston, Massachusetts ; Department of Pediatrics, Harvard Medical School , Boston, Massachusetts ; Centre de Recherche de l'Institut Universitaire de Cardiologie et de Pneumologie de Québec (CRIUCPQ), Faculté de Médecine, Université Laval , Québec , Canada ; and Centre de Recherche sur le Cancer de l'Université Laval, Université Laval , Québec , Canada
| | - Mathieu Laplante
- Department of Internal Medicine, Division of Hypothalamic Research, The University of Texas Southwestern Medical Center , Dallas, Texas ; Transplant Research Program, Boston Children's Hospital , Boston, Massachusetts ; Department of Pediatrics, Harvard Medical School , Boston, Massachusetts ; Centre de Recherche de l'Institut Universitaire de Cardiologie et de Pneumologie de Québec (CRIUCPQ), Faculté de Médecine, Université Laval , Québec , Canada ; and Centre de Recherche sur le Cancer de l'Université Laval, Université Laval , Québec , Canada
| |
Collapse
|
29
|
Physiological changes in neurodegeneration - mechanistic insights and clinical utility. Nat Rev Neurol 2018; 14:259-271. [PMID: 29569624 DOI: 10.1038/nrneurol.2018.23] [Citation(s) in RCA: 64] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The effects of neurodegenerative syndromes extend beyond cognitive function to involve key physiological processes, including eating and metabolism, autonomic nervous system function, sleep, and motor function. Changes in these physiological processes are present in several conditions, including frontotemporal dementia, amyotrophic lateral sclerosis, Alzheimer disease and the parkinsonian plus conditions. Key neural structures that mediate physiological changes across these conditions include neuroendocrine and hypothalamic pathways, reward pathways, motor systems and the autonomic nervous system. In this Review, we highlight the key changes in physiological processing in neurodegenerative syndromes and the similarities in these changes between different progressive neurodegenerative brain conditions. The changes and similarities between disorders might provide novel insights into the human neural correlates of physiological functioning. Given the evidence that physiological changes can arise early in the neurodegenerative process, these changes could provide biomarkers to aid in the early diagnosis of neurodegenerative diseases and in treatment trials.
Collapse
|
30
|
Kim GH, Shi G, Somlo DR, Haataja L, Song S, Long Q, Nillni EA, Low MJ, Arvan P, Myers MG, Qi L. Hypothalamic ER-associated degradation regulates POMC maturation, feeding, and age-associated obesity. J Clin Invest 2018; 128:1125-1140. [PMID: 29457782 PMCID: PMC5824855 DOI: 10.1172/jci96420] [Citation(s) in RCA: 55] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2017] [Accepted: 11/14/2017] [Indexed: 12/18/2022] Open
Abstract
Pro-opiomelanocortin (POMC) neurons function as key regulators of metabolism and physiology by releasing prohormone-derived neuropeptides with distinct biological activities. However, our understanding of early events in prohormone maturation in the ER remains incomplete. Highlighting the significance of this gap in knowledge, a single POMC cysteine-to-phenylalanine mutation at position 28 (POMC-C28F) is defective for ER processing and causes early onset obesity in a dominant-negative manner in humans through an unclear mechanism. Here, we report a pathologically important role of Sel1L-Hrd1, the protein complex of ER-associated degradation (ERAD), within POMC neurons. Mice with POMC neuron–specific Sel1L deficiency developed age-associated obesity due, at least in part, to the ER retention of POMC that led to hyperphagia. The Sel1L-Hrd1 complex targets a fraction of nascent POMC molecules for ubiquitination and proteasomal degradation, preventing accumulation of misfolded and aggregated POMC, thereby ensuring that another fraction of POMC can undergo normal posttranslational processing and trafficking for secretion. Moreover, we found that the disease-associated POMC-C28F mutant evades ERAD and becomes aggregated due to the presence of a highly reactive unpaired cysteine thiol at position 50. Thus, this study not only identifies ERAD as an important mechanism regulating POMC maturation within the ER, but also provides insights into the pathogenesis of monogenic obesity associated with defective prohormone folding.
Collapse
Affiliation(s)
- Geun Hyang Kim
- Department of Molecular & Integrative Physiology, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - Guojun Shi
- Department of Molecular & Integrative Physiology, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - Diane Rm Somlo
- Division of Nutritional Sciences, Cornell University, Ithaca, New York, USA
| | - Leena Haataja
- Division of Metabolism, Endocrinology & Diabetes, Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - Soobin Song
- Division of Nutritional Sciences, Cornell University, Ithaca, New York, USA
| | - Qiaoming Long
- Cam-Su Genomic Resource Center, Soochow University, Suzhou, Jiangsu, China
| | - Eduardo A Nillni
- The Warren Alpert Medical School, Department of Medicine, Molecular Biology, Cell Biology and Biochemistry, Brown University, Providence, Rhode Island, USA
| | - Malcolm J Low
- Department of Molecular & Integrative Physiology, University of Michigan Medical School, Ann Arbor, Michigan, USA.,Division of Metabolism, Endocrinology & Diabetes, Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - Peter Arvan
- Department of Molecular & Integrative Physiology, University of Michigan Medical School, Ann Arbor, Michigan, USA.,Division of Metabolism, Endocrinology & Diabetes, Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - Martin G Myers
- Department of Molecular & Integrative Physiology, University of Michigan Medical School, Ann Arbor, Michigan, USA.,Division of Metabolism, Endocrinology & Diabetes, Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - Ling Qi
- Department of Molecular & Integrative Physiology, University of Michigan Medical School, Ann Arbor, Michigan, USA.,Division of Metabolism, Endocrinology & Diabetes, Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, Michigan, USA
| |
Collapse
|
31
|
Al-Najim W, le Roux CW, Docherty NG. Integrated insights into the role of alpha-melanocyte stimulatory hormone in the control of food intake and glycaemia. Peptides 2018; 100:243-248. [PMID: 29412826 DOI: 10.1016/j.peptides.2017.12.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/01/2017] [Revised: 12/01/2017] [Accepted: 12/04/2017] [Indexed: 10/18/2022]
Abstract
Identifying peptide hormones with multipotent actions on both weight and glycaemia can have a significant impact on therapeutic options in the treatment of obesity and diabetes. This has been exemplified by recent advances involving pharmacological exploitation of glucagon-like peptide 1 biology. Herein, we summarise evidence supporting the potential candidacy in this light of alpha-melanocyte stimulatory hormone, an endogenous peptide hormone and a breakdown product of the neuropeptide pro-opiomelanocortin. We reference its well described central actions in the control of food intake and moreover highlight new data pointing to an important role for this peptide hormone in the periphery, in relation to glycaemic control.
Collapse
Affiliation(s)
- Werd Al-Najim
- Diabetes Complications Research Centre, Conway Institute, School of Medicine and Medical Sciences, University College Dublin, Ireland; Investigative Science, Imperial College London, UK
| | - Carel W le Roux
- Diabetes Complications Research Centre, Conway Institute, School of Medicine and Medical Sciences, University College Dublin, Ireland; Department of Gastrosurgical Research and Education, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Sweden; Investigative Science, Imperial College London, UK
| | - Neil G Docherty
- Diabetes Complications Research Centre, Conway Institute, School of Medicine and Medical Sciences, University College Dublin, Ireland; Department of Gastrosurgical Research and Education, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Sweden.
| |
Collapse
|
32
|
Holmberg E, Sjöstedt J, Malinina E, Johansson M, Turkmen S, Ragagnin G, Lundqvist A, Löfgren M, Jaukkuri L, Bixo M, Bäckström T. Allopregnanolone involvement in feeding regulation, overeating and obesity. Front Neuroendocrinol 2018; 48:70-77. [PMID: 28694181 DOI: 10.1016/j.yfrne.2017.07.002] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/30/2017] [Revised: 07/04/2017] [Accepted: 07/05/2017] [Indexed: 01/19/2023]
Abstract
Obesity is strongly associated with ill health, primarily caused by consumption of excessive calories, and promoted (inter alia) by gamma-amino-butyric-acid (GABA) stimulating food intake by activating GABAA receptors (primarily with α3 and α2 subunits) in the hypothalamic arcuate nucleus and paraventricular nucleus. Allopregnanolone is a potent positive GABAA receptor modulating steroid (GAMS). As reviewed here, elevated allopregnanolone levels are associated with increases in food intake, preferences for energy-rich food, and obesity in humans and other mammals. In women with polycystic ovarian disease, high serum allopregnanolone concentrations are linked to uncontrolled eating, and perturbed sensitivity to allopregnanolone. Increases in weight during pregnancy also correlate with increases in allopregnanolone levels. Moreover, Prader-Willis syndrome is associated with massive overeating, absence of a GABAA receptor (with compensatory >12-, >5- and >1.5-fold increases in α4, γ2, and α1, α3 subunits), and increases in the α4, βx, δ receptor subtype, which is highly sensitive to allopregnanolone. GABA and positive GABA-A receptor modulating steroids like allopregnanolone stimulates food intake and weight gain.
Collapse
Affiliation(s)
- E Holmberg
- Umeå Neurosteroid Research Center, Department of Clinical Sciences, Obstetrics and Gynecology, Umeå University, SE-901 85 Umeå, Sweden
| | - J Sjöstedt
- Umeå Neurosteroid Research Center, Department of Clinical Sciences, Obstetrics and Gynecology, Umeå University, SE-901 85 Umeå, Sweden
| | - E Malinina
- Umeå Neurosteroid Research Center, Department of Clinical Sciences, Obstetrics and Gynecology, Umeå University, SE-901 85 Umeå, Sweden
| | - M Johansson
- Umeå Neurosteroid Research Center, Department of Clinical Sciences, Obstetrics and Gynecology, Umeå University, SE-901 85 Umeå, Sweden
| | - S Turkmen
- Umeå Neurosteroid Research Center, Department of Clinical Sciences, Obstetrics and Gynecology, Umeå University, SE-901 85 Umeå, Sweden
| | - G Ragagnin
- Umeå Neurosteroid Research Center, Department of Clinical Sciences, Obstetrics and Gynecology, Umeå University, SE-901 85 Umeå, Sweden
| | - A Lundqvist
- Umeå Neurosteroid Research Center, Department of Clinical Sciences, Obstetrics and Gynecology, Umeå University, SE-901 85 Umeå, Sweden
| | - M Löfgren
- Umeå Neurosteroid Research Center, Department of Clinical Sciences, Obstetrics and Gynecology, Umeå University, SE-901 85 Umeå, Sweden
| | - L Jaukkuri
- Umeå Neurosteroid Research Center, Department of Clinical Sciences, Obstetrics and Gynecology, Umeå University, SE-901 85 Umeå, Sweden
| | - M Bixo
- Umeå Neurosteroid Research Center, Department of Clinical Sciences, Obstetrics and Gynecology, Umeå University, SE-901 85 Umeå, Sweden
| | - T Bäckström
- Umeå Neurosteroid Research Center, Department of Clinical Sciences, Obstetrics and Gynecology, Umeå University, SE-901 85 Umeå, Sweden.
| |
Collapse
|
33
|
Central IGF1 improves glucose tolerance and insulin sensitivity in mice. Nutr Diabetes 2017; 7:2. [PMID: 29259155 PMCID: PMC5865549 DOI: 10.1038/s41387-017-0002-0] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/15/2016] [Revised: 07/26/2017] [Accepted: 08/01/2017] [Indexed: 02/05/2023] Open
Abstract
Insulin-like growth factor 1 (IGF1) is a key factor for tissue growth and fuel metabolism. The potential function of central IGF1 remains unclear. We previously observed that IGF1 expression is increased in the hypothalamus of obese mice lacking STAT5 in the central nervous system (CNS). In this study, we explored the potential metabolic function of central IGF1 by intracerebroventricular (ICV) injection of IGF1, over-expression of central IGF1 by administering an adeno-associated virus (AAV), and ICV injection of an anti-IGF1 antibody. Mice that over-expressed central IGF1 displayed increased appetite, improved glucose tolerance and insulin sensitivity, decreased Pomc levels in the hypothalamus, and increased UCP1 expression in brown fat tissue. This is the first study demonstrating that central IGF1 regulates several important metabolic functions.
Collapse
|
34
|
Fu SP, Hong H, Lu SF, Hu CJ, Xu HX, Li Q, Yu ML, Ou C, Meng JZ, Wang TL, Hennighausen L, Zhu BM. Genome-wide regulation of electro-acupuncture on the neural Stat5-loss-induced obese mice. PLoS One 2017; 12:e0181948. [PMID: 28806763 PMCID: PMC5555711 DOI: 10.1371/journal.pone.0181948] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2016] [Accepted: 07/10/2017] [Indexed: 02/05/2023] Open
Abstract
Acupuncture is reported to be effective in treating obesity related illnesses, but its mechanism is still unclear. To investigate this mechanism we applied electro-acupuncture (EA) in a mouse model of obesity and used RNA-seq to identify molecular consequences. Deletion of the transcription factor STAT5 from neurons (Stat5NKO) led to obesity. Acupuncture, in turn, reduced body weight and the ratio of epididymal white adipose tissue (Epi-WAT) to body weight, and it also decreased plasma concentrations of glucose, triglyceride, and cholesterol. In addition, EA increased cold endurance of Stat5NKO obese mice. EA reversed altered gene expressions in the hypothalamus and Epi-WAT, especially in the hypothalamus in Stat5NKO obese mice. This study provides, for the first time, insight into genomic networks of obesity and their modulation by electro-acupuncture, which in turn reveals potential mechanisms that explain acupuncture-induced weight-loss.
Collapse
Affiliation(s)
- Shu-Ping Fu
- Key Laboratory of Acupuncture and Medicine Research of Ministry of Education, Nanjing University of Chinese Medicine, Nanjing, China
| | - Hao Hong
- Key Laboratory of Acupuncture and Medicine Research of Ministry of Education, Nanjing University of Chinese Medicine, Nanjing, China
| | - Sheng-Feng Lu
- Key Laboratory of Acupuncture and Medicine Research of Ministry of Education, Nanjing University of Chinese Medicine, Nanjing, China
| | - Chen-Jun Hu
- School of Information Technology, Nanjing University of Chinese Medicine, Nanjing, China
| | - Hou-Xi Xu
- Key Laboratory of Acupuncture and Medicine Research of Ministry of Education, Nanjing University of Chinese Medicine, Nanjing, China
| | - Qian Li
- Key Laboratory of Acupuncture and Medicine Research of Ministry of Education, Nanjing University of Chinese Medicine, Nanjing, China
| | - Mei-Ling Yu
- Key Laboratory of Acupuncture and Medicine Research of Ministry of Education, Nanjing University of Chinese Medicine, Nanjing, China
| | - Chen Ou
- Key Laboratory of Acupuncture and Medicine Research of Ministry of Education, Nanjing University of Chinese Medicine, Nanjing, China
| | - Jian-Zhong Meng
- Key Laboratory of Acupuncture and Medicine Research of Ministry of Education, Nanjing University of Chinese Medicine, Nanjing, China
| | - Tian-Lin Wang
- Key Laboratory of Acupuncture and Medicine Research of Ministry of Education, Nanjing University of Chinese Medicine, Nanjing, China
| | - Lothar Hennighausen
- Laboratory of Genetics and Physiology, National Institute of Diabetes, Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD, United States of America
| | - Bing-Mei Zhu
- Regenerative Medicine Research Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
- * E-mail:
| |
Collapse
|
35
|
Lanfray D, Richard D. Emerging Signaling Pathway in Arcuate Feeding-Related Neurons: Role of the Acbd7. Front Neurosci 2017; 11:328. [PMID: 28690493 PMCID: PMC5481368 DOI: 10.3389/fnins.2017.00328] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2017] [Accepted: 05/24/2017] [Indexed: 01/28/2023] Open
Abstract
The understanding of the mechanisms whereby energy balance is regulated is essential to the unraveling of the pathophysiology of obesity. In the last three decades, focus was put on the metabolic role played by the hypothalamic neurons expressing proopiomelanocortin (POMC) and cocaine and amphetamine regulated transcript (CART) and the neurons co-localizing agouti-related peptide (AgRP), neuropeptide Y (NPY), and gamma-aminobutyric acid (GABA). These neurons are part of the leptin-melanocortin pathway, whose role is key in energy balance regulation. More recently, the metabolic involvement of further hypothalamic uncharacterized neuron populations has been suggested. In this review, we discuss the potential homeostatic implication of hypothalamic GABAergic neurons that produce Acyl-Coa-binding domain containing protein 7 (ACBD7), precursor of the nonadecaneuropeptide (NDN), which has recently been characterized as a potent anorexigenic neuropeptide capable of relaying the leptin anorectic/thermogenic effect via the melanocortin system.
Collapse
Affiliation(s)
- Damien Lanfray
- Centre de Recherche de l'Institut Universitaire de Cardiologie et de Pneumologie de Québec, Université LavalQuébec, QC, Canada
| | - Denis Richard
- Centre de Recherche de l'Institut Universitaire de Cardiologie et de Pneumologie de Québec, Université LavalQuébec, QC, Canada
| |
Collapse
|
36
|
Cyclocarya paliurus (Batal.) Ijinskaja Aqueous Extract (CPAE) Ameliorates Obesity by Improving Insulin Signaling in the Hypothalamus of a Metabolic Syndrome Rat Model. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2017; 2017:4602153. [PMID: 28684967 PMCID: PMC5480046 DOI: 10.1155/2017/4602153] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/08/2017] [Revised: 04/28/2017] [Accepted: 05/15/2017] [Indexed: 12/19/2022]
Abstract
Background Antiobesity drugs may not be optimal for treating obesity. However novel antiobesity agents, especially those derived from natural products, may be suitable. Therefore, we investigated the effects and mechanisms of Cyclocarya paliurus (CP) aqueous extract (CPAE) on obesity. Methods SHR.Cg-Leprcp/NDmcr (SHR/cp) rats were used as a model of obesity and metabolic syndrome. Experimental animals were allocated into two groups—control and CPAE (0.5 g/kg)—for a 7-week treatment period. Examinations were performed, including general physiological characteristics, obesity-related biochemical parameters, and insulin-signaling pathway-related proteins in the hypothalamus. Results Treatment with CPAE reduced food intake, body weight, organ weight, fat mass, and body mass index (BMI) in SHR/cp rats. Meanwhile, CPAE also decreased the levels of fasting serum glucose, fasting serum insulin, HOMA-IR, serum free fatty acids, serum malondialdehyde, serum superoxide dismutase, and serum total-glutathione. The levels of phosphorylation of target proteins—including InsR, IRS1, PI3Kp85, Akt, and FoXO1 as well as protein expression of POMC—were significantly upregulated in the hypothalamus, but NPY expression remarkably decreased. Conclusions CPAE has antiobesity, antihypoglycemic, antihypolipidemic, and antioxidant properties. The mechanism responsible for the antiobesity effect of CPAE may be related to suppression of energy intake via regulation of insulin-signaling pathway in the hypothalamus.
Collapse
|
37
|
Nebigil CG. Prokineticin Is a New Linker between Obesity and Cardiovascular Diseases. Front Cardiovasc Med 2017; 4:20. [PMID: 28447033 PMCID: PMC5388695 DOI: 10.3389/fcvm.2017.00020] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2017] [Accepted: 03/27/2017] [Indexed: 11/30/2022] Open
Abstract
Obesity is a fast growing epidemic event worldwide. Fatness is associated with a number of comorbidities, including cardiovascular diseases (CVDs). Although obesity can be heredity in 30–70% cases, the environmental contributions also play an important role in the increasing prevalence of obesity. The relationship between development of obesity and CVD is poorly characterized. Obesity and CVD can also be resulted from a common mechanism such as metabolic, inflammatory, and neurohormonal changes. Prokineticins are defined as cytokines (immunoregulatory proteins), adipokines (adipocyte-secreted hormone), angiogenic (increasing vessel formation), or aneroxic (lowering food intake) hormones. Prokineticin-mediated signaling plays a key role in the development of obesity and CVD. Two forms of prokineticins exist in circulation and in various tissues including the brain, heart, kidney, and adipose. Prokineticins act on the two G protein-coupled receptors, namely, PKR1 and PKR2. Prokineticin-2 (PK2) via PKR1 receptor controls food intake and prevents adipose tissue expansion. The anti-adipocyte effect of PKR1 signaling is due to suppression of preadipocyte proliferation and differentiation capacity into adipocytes. PK2/PKR1 signaling promotes transcapillary passages of insulin and increases insulin sensitivity. It also plays an important role in the heart and kidney development and functions. Here, we discuss PK2 as a new adipocytokine in the association between obesity and CVD. We also highlight targeting PKR1 can be a new approach to treat obesity and CVD.
Collapse
|
38
|
Derghal A, Djelloul M, Trouslard J, Mounien L. The Role of MicroRNA in the Modulation of the Melanocortinergic System. Front Neurosci 2017; 11:181. [PMID: 28424580 PMCID: PMC5380727 DOI: 10.3389/fnins.2017.00181] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2016] [Accepted: 03/20/2017] [Indexed: 11/13/2022] Open
Abstract
The central control of energy balance involves a highly regulated neuronal network within the hypothalamus and the dorsal vagal complex. In these structures, pro-opiomelanocortin (POMC) neurons are known to reduce meal size and to increase energy expenditure. In addition, leptin, a peripheral signal that relays information regarding body fat content, modulates the activity of melanocortin pathway neurons including POMC-, Agouti-related peptide (AgRP)/Neuropeptide Y (NPY)-, melanocortin receptors (MC3R and MC4R)-expressing neurons. MicroRNAs (miRNAs) are short non-coding RNAs of 22–26 nucleotides that post-transcriptionally interfere with target gene expression by binding to their mRNAs. Evidence has demonstrated that miRNAs play important roles in the central regulation of energy balance. In this context, different studies identified miRNAs including miR-200 family, miR-103, or miR-488 that could target the genes of melanocortin pathway. More precisely, these different miRNAs can modulate energy homeostasis by affecting leptin transduction pathway in the POMC, or AgRP/NPY neurons. This article reviews the role of identified miRNAs in the modulation of melanocortin pathway in the context of energy homeostasis.
Collapse
Affiliation(s)
- Adel Derghal
- Physiologie et Physiopathologie du Système Nerveux Somatomoteur et Neurovégétatif (PPSN), Aix Marseille UniversityMarseille, France
| | - Mehdi Djelloul
- Physiologie et Physiopathologie du Système Nerveux Somatomoteur et Neurovégétatif (PPSN), Aix Marseille UniversityMarseille, France.,Department of Cell and Molecular Biology, Karolinska InstituteStockholm, Sweden
| | - Jérôme Trouslard
- Physiologie et Physiopathologie du Système Nerveux Somatomoteur et Neurovégétatif (PPSN), Aix Marseille UniversityMarseille, France
| | - Lourdes Mounien
- Physiologie et Physiopathologie du Système Nerveux Somatomoteur et Neurovégétatif (PPSN), Aix Marseille UniversityMarseille, France
| |
Collapse
|
39
|
Mosialou I, Shikhel S, Liu JM, Maurizi A, Luo N, He Z, Huang Y, Zong H, Friedman RA, Barasch J, Lanzano P, Deng L, Leibel RL, Rubin M, Nickolas T, Chung W, Zeltser LM, Williams KW, Pessin JE, Kousteni S. MC4R-dependent suppression of appetite by bone-derived lipocalin 2. Nature 2017; 543:385-390. [PMID: 28273060 PMCID: PMC5975642 DOI: 10.1038/nature21697] [Citation(s) in RCA: 307] [Impact Index Per Article: 38.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2016] [Accepted: 02/09/2017] [Indexed: 12/12/2022]
Abstract
Bone has recently emerged as a pleiotropic endocrine organ that secretes at least two hormones, FGF23 and osteocalcin, which regulate kidney function and glucose homeostasis, respectively. These findings have raised the question of whether other bone-derived hormones exist and what their potential functions are. Here we identify, through molecular and genetic analyses in mice, lipocalin 2 (LCN2) as an osteoblast-enriched, secreted protein. Loss- and gain-of-function experiments in mice demonstrate that osteoblast-derived LCN2 maintains glucose homeostasis by inducing insulin secretion and improves glucose tolerance and insulin sensitivity. In addition, osteoblast-derived LCN2 inhibits food intake. LCN2 crosses the blood-brain barrier, binds to the melanocortin 4 receptor (MC4R) in the paraventricular and ventromedial neurons of the hypothalamus and activates an MC4R-dependent anorexigenic (appetite-suppressing) pathway. These results identify LCN2 as a bone-derived hormone with metabolic regulatory effects, which suppresses appetite in a MC4R-dependent manner, and show that the control of appetite is an endocrine function of bone.
Collapse
Affiliation(s)
- Ioanna Mosialou
- Department of Physiology and Cellular Biophysics, College of Physicians and Surgeons, Columbia University, New York, New York 10032, USA
| | - Steven Shikhel
- Department of Physiology and Cellular Biophysics, College of Physicians and Surgeons, Columbia University, New York, New York 10032, USA
| | - Jian-Min Liu
- Department of Physiology and Cellular Biophysics, College of Physicians and Surgeons, Columbia University, New York, New York 10032, USA
| | - Antonio Maurizi
- Department of Physiology and Cellular Biophysics, College of Physicians and Surgeons, Columbia University, New York, New York 10032, USA
| | - Na Luo
- Department of Physiology and Cellular Biophysics, College of Physicians and Surgeons, Columbia University, New York, New York 10032, USA
| | - Zhenyan He
- Department of Neurosurgery, Zhujiang Hospital, Southern Medical University, Guangzhou 510515, China
- Division of Hypothalamic Research, the University of Texas Southwestern Medical Center at Dallas, Dallas, Texas 75390-9077, USA
| | - Yiru Huang
- Department of Neurosurgery, Zhujiang Hospital, Southern Medical University, Guangzhou 510515, China
- Division of Hypothalamic Research, the University of Texas Southwestern Medical Center at Dallas, Dallas, Texas 75390-9077, USA
| | - Haihong Zong
- Department of Medicine and Molecular Pharmacology, The Albert Einstein College of Medicine, Bronx, New York, New York 10461, USA
| | - Richard A Friedman
- Biomedical Informatics Shared Resource, Department of Biomedical Informatics, Herbert Irving Comprehensive Cancer Center, College of Physicians and Surgeons, Columbia University, New York, New York 10032, USA
| | - Jonathan Barasch
- Department of Medicine, College of Physicians and Surgeons, Columbia University, New York, New York 10032, USA
| | - Patricia Lanzano
- Naomi Berrie Diabetes Center and Division of Molecular Genetics, Department of Pediatrics, College of Physicians and Surgeons, Columbia University, New York, New York 10032, USA
| | - Liyong Deng
- Naomi Berrie Diabetes Center and Division of Molecular Genetics, Department of Pediatrics, College of Physicians and Surgeons, Columbia University, New York, New York 10032, USA
| | - Rudolph L Leibel
- Naomi Berrie Diabetes Center and Division of Molecular Genetics, Department of Pediatrics, College of Physicians and Surgeons, Columbia University, New York, New York 10032, USA
| | - Mishaela Rubin
- Metabolic Bone Disease Unit, Department of Medicine, College of Physicians and Surgeons, Columbia University, New York, New York 10032, USA
| | - Thomas Nickolas
- Department of Medicine Nephrology, College of Physicians and Surgeons, Columbia University, New York, New York 10032, USA
| | - Wendy Chung
- Naomi Berrie Diabetes Center and Division of Molecular Genetics, Department of Pediatrics, College of Physicians and Surgeons, Columbia University, New York, New York 10032, USA
| | - Lori M Zeltser
- Naomi Berrie Diabetes Center and Department of Pathology and Cell Biology, Columbia University, New York, New York 10032, USA
| | - Kevin W Williams
- Division of Hypothalamic Research, the University of Texas Southwestern Medical Center at Dallas, Dallas, Texas 75390-9077, USA
| | - Jeffrey E Pessin
- Department of Medicine and Molecular Pharmacology, The Albert Einstein College of Medicine, Bronx, New York, New York 10461, USA
| | - Stavroula Kousteni
- Department of Physiology and Cellular Biophysics, College of Physicians and Surgeons, Columbia University, New York, New York 10032, USA
| |
Collapse
|
40
|
Jeong JH, Woo YJ, Chua S, Jo YH. Single-Cell Gene Expression Analysis of Cholinergic Neurons in the Arcuate Nucleus of the Hypothalamus. PLoS One 2016; 11:e0162839. [PMID: 27611685 PMCID: PMC5017726 DOI: 10.1371/journal.pone.0162839] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2016] [Accepted: 08/29/2016] [Indexed: 01/18/2023] Open
Abstract
The cholinoceptive system in the hypothalamus, in particular in the arcuate nucleus (ARC), plays a role in regulating food intake. Neurons in the ARC contain multiple neuropeptides, amines, and neurotransmitters. To study molecular and neurochemical heterogeneity of ARC neurons, we combine single-cell qRT-PCR and single-cell whole transcriptome amplification methods to analyze expression patterns of our hand-picked 60 genes in individual neurons in the ARC. Immunohistochemical and single-cell qRT-PCR analyses show choline acetyltransferase (ChAT)-expressing neurons in the ARC. Gene expression patterns are remarkably distinct in each individual cholinergic neuron. Two-thirds of cholinergic neurons express tyrosine hydroxylase (Th) mRNA. A large subset of these Th-positive cholinergic neurons is GABAergic as they express the GABA synthesizing enzyme glutamate decarboxylase and vesicular GABA transporter transcripts. Some cholinergic neurons also express the vesicular glutamate transporter transcript gene. POMC and POMC-processing enzyme transcripts are found in a subpopulation of cholinergic neurons. Despite this heterogeneity, gene expression patterns in individual cholinergic cells appear to be highly regulated in a cell-specific manner. In fact, membrane receptor transcripts are clustered with their respective intracellular signaling and downstream targets. This novel population of cholinergic neurons may be part of the neural circuitries that detect homeostatic need for food and control the drive to eat.
Collapse
Affiliation(s)
- Jae Hoon Jeong
- Division of Endocrinology, Department of Medicine, Albert Einstein College of Medicine, Bronx, United States of America
| | - Young Jae Woo
- Department of Genetics, Albert Einstein College of Medicine, Bronx, United States of America
| | - Streamson Chua
- Division of Endocrinology, Department of Medicine, Albert Einstein College of Medicine, Bronx, United States of America
| | - Young-Hwan Jo
- Division of Endocrinology, Department of Medicine, Albert Einstein College of Medicine, Bronx, United States of America
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, United States of America
- * E-mail:
| |
Collapse
|
41
|
Oh TS, Cho H, Cho JH, Yu SW, Kim EK. Hypothalamic AMPK-induced autophagy increases food intake by regulating NPY and POMC expression. Autophagy 2016; 12:2009-2025. [PMID: 27533078 DOI: 10.1080/15548627.2016.1215382] [Citation(s) in RCA: 86] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Hypothalamic AMP-activated protein kinase (AMPK) plays important roles in the regulation of food intake by altering the expression of orexigenic or anorexigenic neuropeptides. However, little is known about the mechanisms of this regulation. Here, we report that hypothalamic AMPK modulates the expression of NPY (neuropeptide Y), an orexigenic neuropeptide, and POMC (pro-opiomelanocortin-α), an anorexigenic neuropeptide, by regulating autophagic activity in vitro and in vivo. In hypothalamic cell lines subjected to low glucose availability such as 2-deoxy-d-glucose (2DG)-induced glucoprivation or glucose deprivation, autophagy was induced via the activation of AMPK, which regulates ULK1 and MTOR complex 1 followed by increased Npy and decreased Pomc expression. Pharmacological or genetic inhibition of autophagy diminished the effect of AMPK on neuropeptide expression in hypothalamic cell lines. Moreover, AMPK knockdown in the arcuate nucleus of the hypothalamus decreased autophagic activity and changed Npy and Pomc expression, leading to a reduction in food intake and body weight. AMPK knockdown abolished the orexigenic effects of intraperitoneal 2DG injection by decreasing autophagy and changing Npy and Pomc expression in mice fed a high-fat diet. We suggest that the induction of autophagy is a possible mechanism of AMPK-mediated regulation of neuropeptide expression and control of feeding in response to low glucose availability.
Collapse
Affiliation(s)
- Tae Seok Oh
- a Department of Brain & Cognitive Sciences , Daegu Gyeongbuk Institute of Science & Technology , Dalseong-gun , Daegu , Korea
| | - Hanchae Cho
- a Department of Brain & Cognitive Sciences , Daegu Gyeongbuk Institute of Science & Technology , Dalseong-gun , Daegu , Korea
| | - Jae Hyun Cho
- a Department of Brain & Cognitive Sciences , Daegu Gyeongbuk Institute of Science & Technology , Dalseong-gun , Daegu , Korea
| | - Seong-Woon Yu
- a Department of Brain & Cognitive Sciences , Daegu Gyeongbuk Institute of Science & Technology , Dalseong-gun , Daegu , Korea
| | - Eun-Kyoung Kim
- a Department of Brain & Cognitive Sciences , Daegu Gyeongbuk Institute of Science & Technology , Dalseong-gun , Daegu , Korea.,b Neurometabolomics Research Center , Daegu Gyeongbuk Institute of Science & Technology , Dalseong-gun , Daegu , Korea
| |
Collapse
|
42
|
Zhang ZY, Dodd GT, Tiganis T. Protein Tyrosine Phosphatases in Hypothalamic Insulin and Leptin Signaling. Trends Pharmacol Sci 2016; 36:661-674. [PMID: 26435211 DOI: 10.1016/j.tips.2015.07.003] [Citation(s) in RCA: 143] [Impact Index Per Article: 15.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2015] [Revised: 07/01/2015] [Accepted: 07/10/2015] [Indexed: 12/22/2022]
Abstract
The hypothalamus is critical to the coordination of energy balance and glucose homeostasis. It responds to peripheral factors, such as insulin and leptin, that convey to the brain the degree of adiposity and the metabolic status of the organism. The development of leptin and insulin resistance in hypothalamic neurons appears to have a key role in the exacerbation of diet-induced obesity. In rodents, this has been attributed partly to the increased expression of the tyrosine phosphatases Protein Tyrosine Phosphatase 1B (PTP1B) and T cell protein tyrosine phosphatase (TCPTP), which attenuate leptin and insulin signaling. Deficiencies in PTP1B and TCPTP in the brain, or specific neurons, promote insulin and leptin signaling and prevent diet-induced obesity, type 2 diabetes mellitus (T2DM), and fatty liver disease. Although targeting phosphatases and hypothalamic circuits remains challenging, recent advances indicate that such hurdles might be overcome. Here, we focus on the roles of PTP1B and TCPTP in insulin and leptin signaling and explore their potential as therapeutic targets.
Collapse
Affiliation(s)
- Zhong-Yin Zhang
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, IN, 46202-5126, USA
| | - Garron T Dodd
- Department of Biochemistry and Molecular Biology, Monash University, VIC 3800, Australia
| | - Tony Tiganis
- Department of Biochemistry and Molecular Biology, Monash University, VIC 3800, Australia.
| |
Collapse
|
43
|
Abstract
PURPOSE OF REVIEW Because of its increasing prevalence and morbi-mortality, obesity is a major health problem. Obesity etiology includes a combination of excess dietary calories and decreased physical activity, coupled with either predisposing genetic factors or metabolic disorders such as insulin resistance. Adipose tissue secretes several metabolically important proteins known as 'adipokines' that play a major role in obesity and insulin resistance. High levels of a newly identified group of adipokines, called prokineticins, have been found in obese adipose tissues. Prokineticins are peptide hormones released principally from macrophages and reproductive organs. They act on the G protein-coupled receptors PKR1 and PKR2. This review aims to provide an overview of current knowledge of the role of prokineticins and their receptors in the development of obesity and insulin resistance. RECENT FINDINGS The principal biological effect of prokineticins in the central nervous system is the control of food intake. Nevertheless, peripheral biological effects of prokineticin are associated with increasing insulin sensitivity and suppressing the adipose tissue expansion. SUMMARY We outline the biological significance of the central and peripheral effects of prokineticins, and the potential of their receptors as targets for the treatment of obesity and insulin resistance.
Collapse
|
44
|
Shukla C, Koch LG, Britton SL, Cai M, Hruby VJ, Bednarek M, Novak CM. Contribution of regional brain melanocortin receptor subtypes to elevated activity energy expenditure in lean, active rats. Neuroscience 2015; 310:252-67. [PMID: 26404873 DOI: 10.1016/j.neuroscience.2015.09.035] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2015] [Revised: 09/10/2015] [Accepted: 09/11/2015] [Indexed: 12/13/2022]
Abstract
Physical activity and non-exercise activity thermogenesis (NEAT) are crucial factors accounting for individual differences in body weight, interacting with genetic predisposition. In the brain, a number of neuroendocrine intermediates regulate food intake and energy expenditure (EE); this includes the brain melanocortin (MC) system, consisting of MC peptides as well as their receptors (MCR). MC3R and MC4R have emerged as critical modulators of EE and food intake. To determine how variance in MC signaling may underlie individual differences in physical activity levels, we examined behavioral response to MC receptor agonists and antagonists in rats that show high and low levels of physical activity and NEAT, that is, high- and low-capacity runners (HCR, LCR), developed by artificial selection for differential intrinsic aerobic running capacity. Focusing on the hypothalamus, we identified brain region-specific elevations in expression of MCR 3, 4, and also MC5R, in the highly active, lean HCR relative to the less active and obesity-prone LCR. Further, the differences in activity and associated EE as a result of MCR activation or suppression using specific agonists and antagonists were similarly region-specific and directly corresponded to the differential MCR expression patterns. The agonists and antagonists investigated here did not significantly impact food intake at the doses used, suggesting that the differential pattern of receptor expression may by more meaningful to physical activity than to other aspects of energy balance regulation. Thus, MCR-mediated physical activity may be a key neural mechanism in distinguishing the lean phenotype and a target for enhancing physical activity and NEAT.
Collapse
Affiliation(s)
- C Shukla
- Department of Biological Sciences, Kent State University, Kent, OH, United States; Harvard Medical School - VA Boston Healthcare System, Boston, MA, United States.
| | - L G Koch
- Department of Anesthesiology, University of Michigan, Ann Arbor, MI, United States; Department of Molecular & Integrative Physiology, University of Michigan, Ann Arbor, MI, United States
| | - S L Britton
- Department of Anesthesiology, University of Michigan, Ann Arbor, MI, United States; Department of Molecular & Integrative Physiology, University of Michigan, Ann Arbor, MI, United States
| | - M Cai
- Department of Chemistry and Biochemistry, University of Arizona, Tucson, AZ, United States
| | - V J Hruby
- Department of Chemistry and Biochemistry, University of Arizona, Tucson, AZ, United States
| | - M Bednarek
- MedImmune Limited, Cambridge, United Kingdom
| | - C M Novak
- Department of Biological Sciences, Kent State University, Kent, OH, United States
| |
Collapse
|
45
|
Abstract
Obesity ensues from an imbalance between energy intake and expenditure that results from gene-environment interactions, which favour a positive energy balance. A society that promotes unhealthy food and encourages sedentary lifestyle (that is, an obesogenic environment) has become a major contributory factor in excess fat deposition in individuals predisposed to obesity. Energy homeostasis relies upon control of energy intake as well as expenditure, which is in part determined by the themogenesis of brown adipose tissue and mediated by the sympathetic nervous system. Several areas of the brain that constitute cognitive and autonomic brain systems, which in turn form networks involved in the control of appetite and thermogenesis, also contribute to energy homeostasis. These networks include the dopamine mesolimbic circuit, as well as the opioid, endocannabinoid and melanocortin systems. The activity of these networks is modulated by peripheral factors such as hormones derived from adipose tissue and the gut, which access the brain via the circulation and neuronal signalling pathways to inform the central nervous system about energy balance and nutritional status. In this Review, I focus on the determinants of energy homeostasis that have emerged as prominent factors relevant to obesity.
Collapse
Affiliation(s)
- Denis Richard
- Institut Universitaire de Cardiologie et de Pneumologie de Québec, 2725 Chemin Sainte-Foy, Québec, QC G1V 4G5, Canada
| |
Collapse
|
46
|
Dodd GT, Decherf S, Loh K, Simonds SE, Wiede F, Balland E, Merry TL, Münzberg H, Zhang ZY, Kahn BB, Neel BG, Bence KK, Andrews ZB, Cowley MA, Tiganis T. Leptin and insulin act on POMC neurons to promote the browning of white fat. Cell 2015; 160:88-104. [PMID: 25594176 DOI: 10.1016/j.cell.2014.12.022] [Citation(s) in RCA: 300] [Impact Index Per Article: 30.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2014] [Revised: 10/14/2014] [Accepted: 12/10/2014] [Indexed: 01/06/2023]
Abstract
The primary task of white adipose tissue (WAT) is the storage of lipids. However, "beige" adipocytes also exist in WAT. Beige adipocytes burn fat and dissipate the energy as heat, but their abundance is diminished in obesity. Stimulating beige adipocyte development, or WAT browning, increases energy expenditure and holds potential for combating metabolic disease and obesity. Here, we report that insulin and leptin act together on hypothalamic neurons to promote WAT browning and weight loss. Deletion of the phosphatases PTP1B and TCPTP enhanced insulin and leptin signaling in proopiomelanocortin neurons and prevented diet-induced obesity by increasing WAT browning and energy expenditure. The coinfusion of insulin plus leptin into the CNS or the activation of proopiomelanocortin neurons also increased WAT browning and decreased adiposity. Our findings identify a homeostatic mechanism for coordinating the status of energy stores, as relayed by insulin and leptin, with the central control of WAT browning.
Collapse
Affiliation(s)
- Garron T Dodd
- Department of Biochemistry and Molecular Biology, Monash University, Victoria 3800, Australia
| | - Stephanie Decherf
- Department of Biochemistry and Molecular Biology, Monash University, Victoria 3800, Australia
| | - Kim Loh
- Department of Biochemistry and Molecular Biology, Monash University, Victoria 3800, Australia
| | | | - Florian Wiede
- Department of Biochemistry and Molecular Biology, Monash University, Victoria 3800, Australia
| | - Eglantine Balland
- Department of Physiology, Monash University, Victoria 3800, Australia
| | - Troy L Merry
- Department of Biochemistry and Molecular Biology, Monash University, Victoria 3800, Australia
| | - Heike Münzberg
- Pennington Biomedical Research Center, LSU Systems, Baton Rouge, LA 70808, USA
| | - Zhong-Yin Zhang
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Barbara B Kahn
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA 02215, USA
| | - Benjamin G Neel
- Campbell Family Cancer Research Institute, Ontario Cancer Institute, Princess Margaret Hospital and Department of Medical Biophysics, University of Toronto, Toronto, ON M5G 2M9, Canada
| | - Kendra K Bence
- Department of Animal Biology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Zane B Andrews
- Department of Physiology, Monash University, Victoria 3800, Australia
| | - Michael A Cowley
- Department of Physiology, Monash University, Victoria 3800, Australia
| | - Tony Tiganis
- Department of Biochemistry and Molecular Biology, Monash University, Victoria 3800, Australia.
| |
Collapse
|
47
|
An R, Li B, You LS, Wang XH. Improvement of Kidney yang syndrome by icariin through regulating hypothalamus-pituitary-adrenal axis. Chin J Integr Med 2015; 21:765-71. [DOI: 10.1007/s11655-015-2063-z] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2012] [Indexed: 10/23/2022]
|
48
|
Wang D, He X, Zhao Z, Feng Q, Lin R, Sun Y, Ding T, Xu F, Luo M, Zhan C. Whole-brain mapping of the direct inputs and axonal projections of POMC and AgRP neurons. Front Neuroanat 2015; 9:40. [PMID: 25870542 PMCID: PMC4375998 DOI: 10.3389/fnana.2015.00040] [Citation(s) in RCA: 188] [Impact Index Per Article: 18.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2015] [Accepted: 03/12/2015] [Indexed: 01/21/2023] Open
Abstract
Pro-opiomelanocortin (POMC) neurons in the arcuate nucleus (ARC) of the hypothalamus and nucleus tractus solitarius (NTS) of the brainstem play important roles in suppressing food intake and maintaining energy homeostasis. Previous tract-tracing studies have revealed the axonal connection patterns of these two brain areas, but the intermingling of POMC neurons with other neuron types has made it challenging to precisely identify the inputs and outputs of POMC neurons. In this study, we used the modified rabies virus to map the brain areas that provide direct inputs to the POMC neurons in the ARC and NTS as well as the inputs to the ARC AgRP neurons for comparison. ARC POMC neurons receive inputs from dozens of discrete structures throughout the forebrain and brainstem. The brain areas containing the presynaptic partners of ARC POMC neurons largely overlap with those of ARC AgRP neurons, although POMC neurons receive relatively broader, denser inputs. Furthermore, POMC neurons in the NTS receive direct inputs predominantly from the brainstem and show very different innervation patterns for POMC neurons in the ARC. By selectively expressing fluorescent markers in the ARC and NTS POMC neurons, we found that almost all of their major presynaptic partners are innervated by POMC neurons in the two areas, suggesting that there are strong reciprocal projections among the major POMC neural pathways. By comprehensively chartering the whole-brain connections of the central melanocortin system in a cell-type-specific manner, this study lays the foundation for dissecting the roles and underlying circuit mechanisms of specific neural pathways in regulating energy homeostasis.
Collapse
Affiliation(s)
- Daqing Wang
- School of Life Sciences, Tsinghua University Beijing China ; National Institute of Biological Sciences Beijing, China
| | - Xiaobing He
- Key Laboratory of Magnetic Resonance in Biological Systems and State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics, Wuhan Institute of Physics and Mathematics, Chinese Academy of Sciences Wuhan, China ; University of Chinese Academy of Sciences Beijing, China
| | - Zhe Zhao
- National Institute of Biological Sciences Beijing, China
| | - Qiru Feng
- National Institute of Biological Sciences Beijing, China
| | - Rui Lin
- National Institute of Biological Sciences Beijing, China
| | - Yue Sun
- National Institute of Biological Sciences Beijing, China
| | - Ting Ding
- Key Laboratory of Magnetic Resonance in Biological Systems and State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics, Wuhan Institute of Physics and Mathematics, Chinese Academy of Sciences Wuhan, China
| | - Fuqiang Xu
- Key Laboratory of Magnetic Resonance in Biological Systems and State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics, Wuhan Institute of Physics and Mathematics, Chinese Academy of Sciences Wuhan, China ; University of Chinese Academy of Sciences Beijing, China ; Wuhan National Laboratory for Optoelectronics Wuhan, China
| | - Minmin Luo
- School of Life Sciences, Tsinghua University Beijing China ; National Institute of Biological Sciences Beijing, China
| | - Cheng Zhan
- National Institute of Biological Sciences Beijing, China
| |
Collapse
|
49
|
Abstract
An increase in the consumption of highly palatable foods coupled with a reduction in the amount of voluntary exercise undertaken has contributed to the rising prevalence of obesity. However, despite the obvious environmental influences, there is considerable evidence to support a genetic component to weight gain. In some people, particularly those who are severely obese, genetic factors play a major role in the development of their obesity and associated complications. Studies into the genetic basis of obesity have yielded insights into the mechanisms involved in the regulation of weight. We now understand that weight is regulated by neural mechanisms that regulate appetite and energy expenditure and that disruption of these pathways can result in severe obesity in some patients. These studies provide a starting point for investigating patients with severe obesity and may ultimately guide the development of more rational targeted therapies.
Collapse
Affiliation(s)
- I Sadaf Farooqi
- Wellcome Trust - MRC Institute of Metabolic ScienceAddenbrooke's Hospital, University of Cambridge Metabolic Research Laboratories, Cambridge, UK
| |
Collapse
|
50
|
Susanti VY, Sasaki T, Yokota-Hashimoto H, Matsui S, Lee YS, Kikuchi O, Shimpuku M, Kim HJ, Kobayashi M, Kitamura T. Sirt1 rescues the obesity induced by insulin-resistant constitutively-nuclear FoxO1 in POMC neurons of male mice. Obesity (Silver Spring) 2014; 22:2115-9. [PMID: 25044690 PMCID: PMC4265245 DOI: 10.1002/oby.20838] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/19/2014] [Accepted: 06/27/2014] [Indexed: 12/11/2022]
Abstract
OBJECTIVE The hypothalamus is the brain center that controls the energy balance. Anorexigenic proopiomelanocortin (POMC) neurons and orexigenic AgRP neurons in the arcuate nucleus of the hypothalamus plays critical roles in energy balance regulation. FoxO1 is a transcription factor regulated by insulin signaling that is deacetylated by Sirt1, a nicotinamide adenine dinucleotide- (NAD(+) -) dependent deacetylase. Overexpression of insulin-resistant constitutively-nuclear FoxO1 (CN-FoxO1) in POMC neurons leads to obesity, whereas Sirt1 overexpression in POMC neurons leads to leanness. Whether overexpression of Sirt1 in POMC neurons could rescue the obesity caused by insulin-resistant CN-FoxO1 was tested here. METHODS POMC neuron-specific CN-FoxO1/Sirt1 double-KI (DKI) mice were analyzed. RESULTS The obese phenotype of CN-FoxO1 KI mice was rescued in male DKI mice. Reduced O2 consumption, increased adiposity, and fewer POMC neurons observed in CN-FoxO1 mice were rescued in male DKI mice without affecting food intake and locomotor activity. Sirt1 overexpression decreased FoxO1 acetylation and protein levels without affecting its nuclear localization in mouse embryonic fibroblasts and hypothalamic N41 cells. CONCLUSIONS Sirt1 rescues the obesity induced by insulin-resistant CN-FoxO1 in POMC neurons of male mice by decreasing FoxO1 protein through deacetylation. Sirt1 ameliorates obesity caused by a genetic model of central insulin resistance.
Collapse
Affiliation(s)
- Vina Yanti Susanti
- Metabolic Signal Research Center, Institute for Molecular and Cellular Regulation, Gunma UniversityMaebashi, Gunma, Japan
| | - Tsutomu Sasaki
- Metabolic Signal Research Center, Institute for Molecular and Cellular Regulation, Gunma UniversityMaebashi, Gunma, Japan
| | - Hiromi Yokota-Hashimoto
- Metabolic Signal Research Center, Institute for Molecular and Cellular Regulation, Gunma UniversityMaebashi, Gunma, Japan
| | - Sho Matsui
- Metabolic Signal Research Center, Institute for Molecular and Cellular Regulation, Gunma UniversityMaebashi, Gunma, Japan
| | - Yong-Soo Lee
- Metabolic Signal Research Center, Institute for Molecular and Cellular Regulation, Gunma UniversityMaebashi, Gunma, Japan
| | - Osamu Kikuchi
- Metabolic Signal Research Center, Institute for Molecular and Cellular Regulation, Gunma UniversityMaebashi, Gunma, Japan
| | - Mayumi Shimpuku
- Metabolic Signal Research Center, Institute for Molecular and Cellular Regulation, Gunma UniversityMaebashi, Gunma, Japan
| | - Hye-Jin Kim
- Metabolic Signal Research Center, Institute for Molecular and Cellular Regulation, Gunma UniversityMaebashi, Gunma, Japan
| | - Masaki Kobayashi
- Metabolic Signal Research Center, Institute for Molecular and Cellular Regulation, Gunma UniversityMaebashi, Gunma, Japan
| | - Tadahiro Kitamura
- Metabolic Signal Research Center, Institute for Molecular and Cellular Regulation, Gunma UniversityMaebashi, Gunma, Japan
| |
Collapse
|