1
|
Solheim MH, Stroganov S, Chen W, Subagia PS, Bauder CA, Wnuk-Lipinski D, Del Río-Martín A, Sotelo-Hitschfeld T, Beddows CA, Klemm P, Dodd GT, Lundh S, Secher A, Wunderlich FT, Steuernagel L, Brüning JC. Hypothalamic PNOC/NPY neurons constitute mediators of leptin-controlled energy homeostasis. Cell 2025:S0092-8674(25)00403-9. [PMID: 40273910 DOI: 10.1016/j.cell.2025.04.001] [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: 03/27/2024] [Revised: 12/23/2024] [Accepted: 04/01/2025] [Indexed: 04/26/2025]
Abstract
Leptin acts in the brain to suppress appetite, yet the responsible neurocircuitries underlying leptin's anorectic effect are incompletely defined. Prepronociceptin (PNOC)-expressing neurons mediate diet-induced hyperphagia and weight gain in mice. Here, we show that leptin regulates appetite and body weight via PNOC neurons, and that loss of leptin receptor (Lepr) expression in PNOC-expressing neurons in the arcuate nucleus of the hypothalamus (ARC) causes hyperphagia and obesity. Restoring Lepr expression in PNOC neurons on a Lepr-null obese background substantially reduces body weight. Lepr inactivation in PNOC neurons increases neuropeptide Y (Npy) expression in a subset of hypothalamic PNOC neurons that do not express agouti-related peptide (Agrp). Selective chemogenetic activation of PNOC/NPY neurons promotes feeding to the same extent as activating all PNOCARC neurons, and overexpression of Npy in PNOCARC neurons promotes hyperphagia and obesity. Thus, we introduce PNOC/NPYARC neurons as an additional critical mediator of leptin action and as a promising target for obesity therapeutics.
Collapse
Affiliation(s)
- Marie H Solheim
- Department of Neuronal Control of Metabolism, Max Planck Institute for Metabolism Research, Cologne, Germany
| | - Sima Stroganov
- Department of Neuronal Control of Metabolism, Max Planck Institute for Metabolism Research, Cologne, Germany
| | - Weiyi Chen
- Department of Neuronal Control of Metabolism, Max Planck Institute for Metabolism Research, Cologne, Germany
| | - P Sicilia Subagia
- Department of Neuronal Control of Metabolism, Max Planck Institute for Metabolism Research, Cologne, Germany
| | - Corinna A Bauder
- Department of Neuronal Control of Metabolism, Max Planck Institute for Metabolism Research, Cologne, Germany
| | - Daria Wnuk-Lipinski
- Department of Neuronal Control of Metabolism, Max Planck Institute for Metabolism Research, Cologne, Germany
| | - Almudena Del Río-Martín
- Department of Neuronal Control of Metabolism, Max Planck Institute for Metabolism Research, Cologne, Germany
| | - Tamara Sotelo-Hitschfeld
- Department of Neuronal Control of Metabolism, Max Planck Institute for Metabolism Research, Cologne, Germany
| | - Cait A Beddows
- Department of Anatomy and Physiology, the University of Melbourne, Melbourne, VIC, Australia
| | - Paul Klemm
- Department of Neuronal Control of Metabolism, Max Planck Institute for Metabolism Research, Cologne, Germany
| | - Garron T Dodd
- Department of Anatomy and Physiology, the University of Melbourne, Melbourne, VIC, Australia
| | - Sofia Lundh
- Global Drug Discovery, Novo Nordisk A/S, Måløv, Denmark
| | - Anna Secher
- Global Drug Discovery, Novo Nordisk A/S, Måløv, Denmark
| | - F Thomas Wunderlich
- Department of Neuronal Control of Metabolism, Max Planck Institute for Metabolism Research, Cologne, Germany
| | - Lukas Steuernagel
- Department of Neuronal Control of Metabolism, Max Planck Institute for Metabolism Research, Cologne, Germany; Neurogenomics Group, Max Planck Institute for Metabolism Research, Cologne, Germany
| | - Jens C Brüning
- Department of Neuronal Control of Metabolism, Max Planck Institute for Metabolism Research, Cologne, Germany; Policlinic for Endocrinology, Diabetes, and Preventive Medicine (PEDP), University Hospital Cologne, Cologne, Germany; Excellence Cluster on Cellular Stress Responses in Aging Associated Diseases (CECAD) and Center of Molecular Medicine Cologne (CMMC), University of Cologne, Cologne, Germany; National Center for Diabetes Research (DZD), Ingolstädter Landstrasse 1, 85764 Neuherberg, Germany.
| |
Collapse
|
2
|
AlTarrah D, Al-Abdullah L, Alhusayan M, Canha D, Almazeedi S, Al-Serri A, Abulhasan M, Alsomly T, Almutawa N, Al-Onaizi M, Fagherazzi G, Alzaid F. Cross-sectional analysis of eating disorder risk and risk correlates in candidates for bariatric surgery from the BariPredict cohort. Sci Rep 2025; 15:11191. [PMID: 40169756 PMCID: PMC11961600 DOI: 10.1038/s41598-025-95614-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2024] [Accepted: 03/24/2025] [Indexed: 04/03/2025] Open
Abstract
Bariatric surgery is an effective intervention for managing obesity. Persons with obesity are a high-risk population for eating disorders (ED), and these can negatively impact perioperative and long-term outcomes of surgery. We aim to understand prevalence and correlates of ED in preintervention patients, identifying those needing psychological support. Baseline cross-sectional analysis of 275 patients of the BariPredict cohort (NCT06480058), a study to assess predictors of long-term surgery outcomes. Psychological assessments were conducted using SCOFF, KUAS, and BDI tools. Data were analyzed for prevalence of high ED risk and for associations of clinical, biological and demographic factors. Mean age was 38.5 years, mean BMI was 42.3 kg/m², with 62.5% being female. 65.8% of patients had a SCOFF score ≥ 2 indicating high ED risk. Class II obesity (p < 0.05), younger age (p < 0.01), and higher depression (p < 0.01) were associated with ED risk in a logistic regression adjusted for age, obesity class, diabetes, HbA1c, depression and anxiety scores. We report high preintervention prevalence of ED, with a risk profile corresponding to BMI of 35-39.9 Kg/m2 in younger adults with concurrent depression. This patient profile should be prioritized for psychological assessment and support to potentially improve outcomes of bariatric surgery.
Collapse
Affiliation(s)
- Dana AlTarrah
- Department of Social and Behavioral Science, Faculty of Public Health, Kuwait University, Kuwait City, Kuwait
| | - Lulwa Al-Abdullah
- Dasman Diabetes Institute, Gulf Road intersecting Jassim Al Bahar St., Sharq, Block 3, Dasman, P.O. Box 1180, Kuwait City, Kuwait
| | - Mohammed Alhusayan
- Dasman Diabetes Institute, Gulf Road intersecting Jassim Al Bahar St., Sharq, Block 3, Dasman, P.O. Box 1180, Kuwait City, Kuwait
- Department of Cancer Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Dulce Canha
- Deep Digital Phenotyping Research Unit, Department of Precision Health, Luxembourg Institute of Health, 1A-B, rue Thomas Edison, 1445, Strassen, Luxembourg
- Faculty of Science, Technology and Medicine, University of Luxembourg, Esch-sur-Alzette, Luxembourg
| | | | - Ahmad Al-Serri
- Department of Pathology, Faculty of Medicine, Kuwait University, Kuwait City, Kuwait
| | - Maryam Abulhasan
- Kuwait Center for Mental Health, Ministry of Health, Kuwait City, Kuwait
| | - Talia Alsomly
- Department of Surgery, Jaber Al-Ahmed Hospital, Kuwait City, Kuwait
- King Faisal Specialist Hospital & Research Centre, Riyadh, Saudi Arabia
| | - Naif Almutawa
- Department of Community Medicine and Behavioral Science, Faculty of Medicine, Kuwait University, Kuwait City, Kuwait
| | - Mohammed Al-Onaizi
- Dasman Diabetes Institute, Gulf Road intersecting Jassim Al Bahar St., Sharq, Block 3, Dasman, P.O. Box 1180, Kuwait City, Kuwait
- Department of Anatomy, Faculty of Medicine, Kuwait University, Kuwait City, Kuwait
| | - Guy Fagherazzi
- Deep Digital Phenotyping Research Unit, Department of Precision Health, Luxembourg Institute of Health, 1A-B, rue Thomas Edison, 1445, Strassen, Luxembourg
| | - Fawaz Alzaid
- Dasman Diabetes Institute, Gulf Road intersecting Jassim Al Bahar St., Sharq, Block 3, Dasman, P.O. Box 1180, Kuwait City, Kuwait.
- INSERM UMR-S1151, CNRS UMR-S8253, Université Paris Cité, Institut Necker Enfants Malades, Paris, France.
| |
Collapse
|
3
|
Holmes TL, Chabronova A, Denning C, James V, Peffers MJ, Smith JGW. Footprints in the Sno: investigating the cellular and molecular mechanisms of SNORD116. Open Biol 2025; 15:240371. [PMID: 40101781 PMCID: PMC11919532 DOI: 10.1098/rsob.240371] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2024] [Revised: 01/11/2025] [Accepted: 02/04/2025] [Indexed: 03/20/2025] Open
Abstract
The small nucleolar RNA (snoRNA) SNORD116 is a small non-coding RNA of interest across multiple biomedical fields of research. Much of the investigation into SNORD116 has been undertaken in the context of the congenital disease Prader-Willi syndrome, wherein SNORD116 expression is lost. However, emerging evidence indicates wider roles in various disease and tissue contexts such as cellular growth, metabolism and signalling. Nevertheless, a conclusive mechanism of action for SNORD116 remains to be established. Here, we review the key findings from these investigations, with the aim of identifying common elements from which to elucidate potential targets and mechanisms of SNORD116. A key recurring element identified is disruption to the insulin/IGF-1 and PI3K/mTOR signalling pathways, contributing to many of the phenotypes associated with SNORD116 modulation explored in this review.
Collapse
Affiliation(s)
- Terri L. Holmes
- Centre for Metabolic Health, Norwich Medical School, University of East Anglia, Norwich, NorfolkNR4 7UQ, UK
| | - Alzbeta Chabronova
- Department of Musculoskeletal Ageing Science, University of Liverpool, Liverpool, UK
| | - Chris Denning
- Department of Stem Cell Biology, University of Nottingham, Nottingham, UK
| | - Victoria James
- School of Veterinary Medicine and Science, University of Nottingham, Nottingham, UK
| | - Mandy J. Peffers
- Department of Musculoskeletal Ageing Science, University of Liverpool, Liverpool, UK
| | - James G. W. Smith
- Centre for Metabolic Health, Norwich Medical School, University of East Anglia, Norwich, NorfolkNR4 7UQ, UK
| |
Collapse
|
4
|
Esbati R, Yazdani O, Simonetti J. Management of Obesity-Related Genetic Disorders. Endocrinol Metab Clin North Am 2025; 54:17-38. [PMID: 39919873 DOI: 10.1016/j.ecl.2024.11.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/09/2025]
Abstract
Obesity-related genetic disorders are marked by severe, early-onset obesity caused by mutations that disrupt key biological mechanisms regulating hunger, energy balance, and fat storage. These disorders commonly impact systems such as the hypothalamic leptin-melanocortin signaling network, which plays a crucial role in controlling appetite and body weight, mainly through the melanocortin-4 receptor (MC4R) pathway. This review explores current management strategies and emerging therapies for genetic obesity disorders, highlighting the importance of treatment approaches and expanded genetic diagnostics to improve outcomes for affected individuals.
Collapse
Affiliation(s)
- Romina Esbati
- Department of Medicine, Division of Endocrinology, Diabetes and Hypternsion, Brigham and Women's Hospital, Boston, MA 02115, USA
| | - Omid Yazdani
- Department of Medicine, Division of Gastroenterology, Hepatology and Endoscopy, Brigham and Women's Hospital, Harvard Medical University, Boston, MA 02115, USA
| | - Juliana Simonetti
- Department of Medicine, Division of Endocrinology, Diabetes and Metabolism, Obesity Medicine Program, University of Utah, Salt Lake City, UT 84108, USA.
| |
Collapse
|
5
|
Khalil M, Di Ciaula A, Mahdi L, Jaber N, Di Palo DM, Graziani A, Baffy G, Portincasa P. Unraveling the Role of the Human Gut Microbiome in Health and Diseases. Microorganisms 2024; 12:2333. [PMID: 39597722 PMCID: PMC11596745 DOI: 10.3390/microorganisms12112333] [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: 10/28/2024] [Revised: 11/12/2024] [Accepted: 11/14/2024] [Indexed: 11/29/2024] Open
Abstract
The human gut is a complex ecosystem that supports billions of living species, including bacteria, viruses, archaea, phages, fungi, and unicellular eukaryotes. Bacteria give genes and enzymes for microbial and host-produced compounds, establishing a symbiotic link between the external environment and the host at both the gut and systemic levels. The gut microbiome, which is primarily made up of commensal bacteria, is critical for maintaining the healthy host's immune system, aiding digestion, synthesizing essential nutrients, and protecting against pathogenic bacteria, as well as influencing endocrine, neural, humoral, and immunological functions and metabolic pathways. Qualitative, quantitative, and/or topographic shifts can alter the gut microbiome, resulting in dysbiosis and microbial dysfunction, which can contribute to a variety of noncommunicable illnesses, including hypertension, cardiovascular disease, obesity, diabetes, inflammatory bowel disease, cancer, and irritable bowel syndrome. While most evidence to date is observational and does not establish direct causation, ongoing clinical trials and advanced genomic techniques are steadily enhancing our understanding of these intricate interactions. This review will explore key aspects of the relationship between gut microbiota, eubiosis, and dysbiosis in human health and disease, highlighting emerging strategies for microbiome engineering as potential therapeutic approaches for various conditions.
Collapse
Affiliation(s)
- Mohamad Khalil
- Clinica Medica “A. Murri”, Department of Precision and Regenerative Medicine and Ionian Area (DiMePre-J), Medical School, University of Bari Aldo Moro, 70124 Bari, Italy; (M.K.); (A.D.C.); (L.M.); (N.J.)
| | - Agostino Di Ciaula
- Clinica Medica “A. Murri”, Department of Precision and Regenerative Medicine and Ionian Area (DiMePre-J), Medical School, University of Bari Aldo Moro, 70124 Bari, Italy; (M.K.); (A.D.C.); (L.M.); (N.J.)
| | - Laura Mahdi
- Clinica Medica “A. Murri”, Department of Precision and Regenerative Medicine and Ionian Area (DiMePre-J), Medical School, University of Bari Aldo Moro, 70124 Bari, Italy; (M.K.); (A.D.C.); (L.M.); (N.J.)
| | - Nour Jaber
- Clinica Medica “A. Murri”, Department of Precision and Regenerative Medicine and Ionian Area (DiMePre-J), Medical School, University of Bari Aldo Moro, 70124 Bari, Italy; (M.K.); (A.D.C.); (L.M.); (N.J.)
| | - Domenica Maria Di Palo
- Division of Hygiene, Department of Interdisciplinary Medicine, University of Bari Aldo Moro, Piazza Giulio Cesare 11, 70124 Bari, Italy;
| | - Annarita Graziani
- Institut AllergoSan Pharmazeutische Produkte Forschungs- und Vertriebs GmbH, 8055 Graz, Austria;
| | - Gyorgy Baffy
- Division of Gastroenterology, Hepatology and Endoscopy, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02130, USA;
- Section of Gastroenterology, Department of Medicine, VA Boston Healthcare System, Boston, MA 02130, USA
| | - Piero Portincasa
- Clinica Medica “A. Murri”, Department of Precision and Regenerative Medicine and Ionian Area (DiMePre-J), Medical School, University of Bari Aldo Moro, 70124 Bari, Italy; (M.K.); (A.D.C.); (L.M.); (N.J.)
| |
Collapse
|
6
|
Mohammed I, Selvaraj S, Ahmed WS, Al-Barazenji T, Dauleh H, Love DR, Saraiva LR, Hussain K. Functional Evaluation of a Novel Homozygous ADCY3 Variant Causing Childhood Obesity. Int J Mol Sci 2024; 25:11815. [PMID: 39519366 PMCID: PMC11547096 DOI: 10.3390/ijms252111815] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2024] [Revised: 10/24/2024] [Accepted: 11/01/2024] [Indexed: 11/16/2024] Open
Abstract
Adenylate cyclase 3 (ADCY3) is a transmembrane protein predominantly expressed in the primary cilia of neurons. It plays a vital role in converting ATP to cAMP, a secondary messenger that regulates various downstream signaling pathways such as carbohydrates and lipids metabolism. Homozygous loss-of-function variants in the ADCY3 gene lead to severe early-onset obesity and insulin resistance whereas gain-of-function variants protect against obesity. To describe a novel pathogenic ADCY3 variant implicated in early-onset obesity and functionally characterize this variant via in vitro and in silico validation, we identified a novel homozygous nonsense variant c.2520C>G, p.Thr840X in the ADCY3 gene using gene panel sequencing in a four-year-old girl. She was born to first-cousin consanguineous parents. The patient presented with severe obesity, and exhibited hepatomegaly and insulin resistance, with other biochemical and hormonal tests being normal. In vitro and in silico functional analyses showed downregulation and impaired activation of the ADCY3 protein. Our findings contribute to existing research that supports the role of ADCY3 in the genetic pathogenesis of early-onset obesity. In vitro and in silico functional characterization of the novel p.Thr840X variant showed impaired enzymatic activity leading to receptor loss of function, consistent with the patient's phenotype. Genetic testing is essential in severe early-onset obesity and early diagnosis could benefit patients with personalized treatment strategies.
Collapse
Affiliation(s)
- Idris Mohammed
- College of Health & Life Sciences, Hamad Bin Khalifa University, Doha P.O. Box 34110, Qatar
- Division of Endocrinology, Department of Pediatric Medicine, Sidra Medicine, Doha P.O. Box 26999, Qatar
| | - Senthil Selvaraj
- Division of Translational Medicine, Research Branch, Sidra Medicine, Doha P.O. Box 26999, Qatar
| | - Wesam S. Ahmed
- College of Health & Life Sciences, Hamad Bin Khalifa University, Doha P.O. Box 34110, Qatar
| | - Tara Al-Barazenji
- Division of Endocrinology, Department of Pediatric Medicine, Sidra Medicine, Doha P.O. Box 26999, Qatar
| | - Hajar Dauleh
- Division of Endocrinology, Department of Pediatric Medicine, Sidra Medicine, Doha P.O. Box 26999, Qatar
| | - Donald R. Love
- Division of Genetic Pathology, Department of Pathology, Sidra Medicine, Doha P.O. Box 26999, Qatar;
| | - Luis R. Saraiva
- College of Health & Life Sciences, Hamad Bin Khalifa University, Doha P.O. Box 34110, Qatar
- Division of Translational Medicine, Research Branch, Sidra Medicine, Doha P.O. Box 26999, Qatar
- Department of Comparative Medicine, Yale University School of Medicine, New Haven, CT 06520-8016, USA
| | - Khalid Hussain
- Division of Endocrinology, Department of Pediatric Medicine, Sidra Medicine, Doha P.O. Box 26999, Qatar
| |
Collapse
|
7
|
Abu-Rub LI, Al-Barazenji T, Abiib S, Hammad AS, Abbas A, Hussain K, Al-Shafai M. Identification of KSR2 Variants in Pediatric Patients with Severe Early-Onset Obesity from Qatar. Genes (Basel) 2024; 15:966. [PMID: 39202327 PMCID: PMC11353872 DOI: 10.3390/genes15080966] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2024] [Revised: 07/14/2024] [Accepted: 07/18/2024] [Indexed: 09/03/2024] Open
Abstract
The kinase suppressor of Ras 2 (KSR2) gene is associated with monogenic obesity, and loss-of-function variants in KSR2 have been identified in individuals with severe early-onset obesity. This study investigated KSR2 variants in 9 pediatric patients with severe early-onset obesity in Qatar using whole genome sequencing among a cohort of 240 individuals. We focused on KSR2 variants with a minor allele frequency (MAF) below 1% and a Combined Annotation Dependent Depletion (CADD) score above 13 to identify potential causative variants. Our analysis identified four KSR2 variants: one intronic (c.1765-8G>A) and three missense variants (c.1057G>A, c.1673G>A, and c.923T>C) in nine patients. The intronic variant c.1765-8G>A was the most frequent (seen in six individuals) and had a CADD score of 21.10, suggesting possible pathogenicity. This variant showed a significantly higher allele frequency in the Qatari population compared to the Genome Aggregation Database (gnomAD), indicating a possible founder effect. Molecular modeling of the missense variants revealed structural changes in the protein structure. The study concludes that these four KSR2 variants are associated with monogenic obesity, with an autosomal dominant inheritance pattern. The c.1765-8G>A variant's prevalence in Qatar underscores its importance in genetic screening for severe obesity. This research advances the understanding of genetic factors in severe early-onset obesity and may inform better management strategies.
Collapse
Affiliation(s)
- Lubna I. Abu-Rub
- Department of Biomedical Sciences, College of Health Sciences, QU Health, Qatar University, Doha 2713, Qatar; (L.I.A.-R.); (T.A.-B.); (S.A.); (A.S.H.); (A.A.)
| | - Tara Al-Barazenji
- Department of Biomedical Sciences, College of Health Sciences, QU Health, Qatar University, Doha 2713, Qatar; (L.I.A.-R.); (T.A.-B.); (S.A.); (A.S.H.); (A.A.)
| | - Sumaya Abiib
- Department of Biomedical Sciences, College of Health Sciences, QU Health, Qatar University, Doha 2713, Qatar; (L.I.A.-R.); (T.A.-B.); (S.A.); (A.S.H.); (A.A.)
| | - Ayat S Hammad
- Department of Biomedical Sciences, College of Health Sciences, QU Health, Qatar University, Doha 2713, Qatar; (L.I.A.-R.); (T.A.-B.); (S.A.); (A.S.H.); (A.A.)
| | - Alaa Abbas
- Department of Biomedical Sciences, College of Health Sciences, QU Health, Qatar University, Doha 2713, Qatar; (L.I.A.-R.); (T.A.-B.); (S.A.); (A.S.H.); (A.A.)
| | - Khalid Hussain
- Division of Endocrinology, Department of Pediatric Medicine, Sidra Medicine, Doha P.O. Box 26999, Qatar
| | - Mashael Al-Shafai
- Department of Biomedical Sciences, College of Health Sciences, QU Health, Qatar University, Doha 2713, Qatar; (L.I.A.-R.); (T.A.-B.); (S.A.); (A.S.H.); (A.A.)
- Biomedical Research Center, Qatar University, Doha P.O. Box 2713, Qatar
| |
Collapse
|
8
|
Guglielmi V, Dalle Grave R, Leonetti F, Solini A. Female obesity: clinical and psychological assessment toward the best treatment. Front Endocrinol (Lausanne) 2024; 15:1349794. [PMID: 38765954 PMCID: PMC11099266 DOI: 10.3389/fendo.2024.1349794] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/05/2023] [Accepted: 04/19/2024] [Indexed: 05/22/2024] Open
Abstract
Obesity is a heterogeneous condition which results from complex interactions among sex/gender, sociocultural, environmental, and biological factors. Obesity is more prevalent in women in most developed countries, and several clinical and psychological obesity complications show sex-specific patterns. Females differ regarding fat distribution, with males tending to store more visceral fat, which is highly correlated to increased cardiovascular risk. Although women are more likely to be diagnosed with obesity and appear more motivated to lose weight, as confirmed by their greater representation in clinical trials, males show better outcomes in terms of body weight and intra-abdominal fat loss and improvements in the metabolic risk profile. However, only a few relatively recent studies have investigated gender differences in obesity, and sex/gender is rarely considered in the assessment and management of the disease. This review summarizes the evidence of gender differences in obesity prevalence, contributing factors, clinical complications, and psychological challenges. In addition, we explored gender differences in response to obesity treatments in the specific context of new anti-obesity drugs.
Collapse
Affiliation(s)
- Valeria Guglielmi
- Unit of Internal Medicine and Obesity Center, Department of Systems Medicine, Policlinico Tor Vergata, University of Rome Tor Vergata, Rome, Italy
| | - Riccardo Dalle Grave
- Department of Eating and Weight Disorders, Villa Garda Hospital, Garda, VR, Italy
| | - Frida Leonetti
- Department of Medical-Surgical Sciences and Biotechnologies, Sapienza University of Rome, Rome, Italy
| | - Anna Solini
- Department of Surgical, Medical, Molecular and Critical Area Pathology, University of Pisa, Pisa, Italy
| |
Collapse
|
9
|
Cifuentes L, Ghusn W, Campos A, Bublitz JT, Hurtado MD, Olson J, Acosta A. Leptin-Melanocortin pathway variants and gastric emptying in patients with obesity. Neurogastroenterol Motil 2024; 36:e14764. [PMID: 38361111 PMCID: PMC11042991 DOI: 10.1111/nmo.14764] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/11/2023] [Revised: 01/16/2024] [Accepted: 02/02/2024] [Indexed: 02/17/2024]
Abstract
BACKGROUND Accelerated gastric emptying (GE) is a trait seen in obesity. Mutations in the hypothalamic leptin-melanocortin 4 receptor (Leptin-MC4R) pathway have been associated with obesity. We sought to investigate the association of leptin-MC4R pathway variants and GE in patients with obesity. METHODS This is a cross-sectional study of patients with a history of severe obesity that were genotyped and completed a GE test by scintigraphy. We evaluated the percentage of GE (GE %) at 2 and 4 h between both groups using ANCOVA with weight and sex as covariates. We subdivide patients into carriers based on the location of the identified variants (i.e., upstream or downstream of the Leptin-MC4R pathway) and compared them with noncarriers using ANOVA. Results are presented as mean and standard deviation (± SD). KEY RESULTS A total of 95 patients; nine carriers (67% females; 39.78 ± 12.33 years; BMI: 49.14 ± 12.96 kg/m2) and 86 noncarriers (87% female; 49.98 ± 13.74 years; BMI: 40.75 ± 6.29 kg/m2) were included. At 2 and 4 h, carriers had a delayed GE when compared noncarriers (p = 0.03 and p = 0.005, respectively). In carriers, when compared upstream carriers vs. downstream carriers vs. noncarriers by location there was a significant difference in GE among groups at 2 h and at 4 h (p = 0.02 and p = 0.01, respectively). CONCLUSIONS & INFERENCES Carriers of heterozygous variants in the Leptin-MC4R pathway had a delayed GE compared to noncarriers. These findings point the important relationship between the Leptin-MC4R pathway and gastric motility.
Collapse
Affiliation(s)
- Lizeth Cifuentes
- Precision Medicine for Obesity Program, Division of Gastroenterology and Hepatology, Department of Medicine, Mayo Clinic, Rochester, MN
| | - Wissam Ghusn
- Precision Medicine for Obesity Program, Division of Gastroenterology and Hepatology, Department of Medicine, Mayo Clinic, Rochester, MN
| | - Alejandro Campos
- Precision Medicine for Obesity Program, Division of Gastroenterology and Hepatology, Department of Medicine, Mayo Clinic, Rochester, MN
| | - Joshua T. Bublitz
- Division of Epidemiology, Department of Quantitative Health Sciences, Mayo Clinic, Rochester, MN
| | - Maria Daniela Hurtado
- Precision Medicine for Obesity Program, Division of Gastroenterology and Hepatology, Department of Medicine, Mayo Clinic, Rochester, MN
| | - Janet Olson
- Division of Epidemiology, Department of Quantitative Health Sciences, Mayo Clinic, Rochester, MN
| | - Andres Acosta
- Precision Medicine for Obesity Program, Division of Gastroenterology and Hepatology, Department of Medicine, Mayo Clinic, Rochester, MN
| |
Collapse
|
10
|
Purnell JQ, le Roux CW. Hypothalamic control of body fat mass by food intake: The key to understanding why obesity should be treated as a disease. Diabetes Obes Metab 2024; 26 Suppl 2:3-12. [PMID: 38351898 DOI: 10.1111/dom.15478] [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: 11/08/2023] [Revised: 01/06/2024] [Accepted: 01/18/2024] [Indexed: 03/27/2024]
Abstract
BACKGROUND Hypothalamic centres have been recognized to play a central role in body weight regulation for nearly 70 years. AIMS In this review, we will explore the current undersanding of the role the hypothalamus plays in controlling food intake behaviours. MATERIALS AND METHODS Review of relevant literature from PubMed searches and review article citations. RESULTS Beginning with autopsy studies showing destructive hypothalamic lesions in patients manifesting hyperphagia and rapid weight gain, followed by animal lesioning studies pinpointing adjacent hypothalamic sites as the 'satiety' centre and the 'feeding' centre of the brain, the neurocircuitry that governs our body weight is now understood to consist of a complex, interconnected network, including the hypothalamus and extending to cortical sites, reward centres and brainstem. Neurons in these sites receive afferent signals from the gastrointestinal tract and adipose tissue indicating food availability, calorie content, as well as body fat mass. DISCUSSION Integration of these complex signals leads to modulation of the two prime effector systems that defend a body fat mass set point: food intake and energy expenditure. CONCLUSION Understanding the hypothalamic control of food intake forms the foundation for understanding and managing obesity as a chronic disease.
Collapse
Affiliation(s)
- Jonathan Q Purnell
- Department of Medicine, Oregon Health & Science University, Portland, Oregon, USA
| | - Carel W le Roux
- School of Medicine, University College Dublin, Dublin, Ireland
| |
Collapse
|
11
|
Supti DA, Akter F, Rahman MI, Munim MA, Tonmoy MIQ, Tarin RJ, Afroz S, Reza HA, Yeasmin R, Alam MR, Hossain MS. Meta-analysis investigating the impact of the LEPR rs1137101 (A>G) polymorphism on obesity risk in Asian and Caucasian ethnicities. Heliyon 2024; 10:e27213. [PMID: 38496879 PMCID: PMC10944198 DOI: 10.1016/j.heliyon.2024.e27213] [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: 07/03/2023] [Revised: 12/12/2023] [Accepted: 02/26/2024] [Indexed: 03/19/2024] Open
Abstract
Obesity is a chronic condition which is identified by the buildup of excess body fat caused by a combination of various factors, including genetic predisposition and lifestyle choices. rs1137101 (A > G) polymorphism in the CHR1 domain of LEPR protein linked to different diseases including obesity. Nevertheless, the connection between this polymorphism and the likelihood of developing obesity has not been determined definitively. Therefore, a meta-analysis was conducted to assess the relationship between rs1137101 and the risk of obesity. The meta-analysis included all studies meeting pre-defined criteria, found through searching databases up until February 2023. A combined odds ratio with a 95% confidence interval was estimated as overall and in continent subgroups for homozygous, heterozygous, recessive, dominant and allelic models using the fixed or the random-effects model. The meta-analysis identified 39 eligible studies with cases and controls (6099 cases/6711 controls) in 38 articles under different ethnic backgrounds. The results indicated a significant relationship between rs1137101 and the likelihood of developing obesity in each of the genetic models [the homozygous model (GG vs. AA: 95% Confidence Interval = 1.12-1.73, Odds Ratio = 1.39, P value = 0.003); the heterozygous model (AG vs. AA: 95% Confidence Interval = 1.07-1.42, Odds Ratio = 1.23, P value = 0.005); the dominant model (AG/GG vs AA: 95% Confidence Interval = 1.10-1.49, Odds Ratio = 1.28, P value = 0.001); the recessive model (GG vs AA/AG: 95% Confidence Interval = 1.02-1.45, Odds Ratio = 1.21, P value = 0.03); and the allelic model (G vs A; 95% Confidence Interval = 1.07-1.33, Odds Ratio = 1.19, P value = 0.002)] tested. Additionally, with an FDR <0.05, all genotypic models demonstrated statistical significance. The association remained significant among subgroups of Asian and Caucasian populations, although analysis in some genetic models did not show a significant association. Begg's and Egger's tests did not show publication biases. In sensitivity analysis, one particular study was found to have an impact on the Recessive model's significance, but other models remained unaffected. The current meta-analysis found significant indications supporting the association between rs1137101 and obesity. To avail a deeper understanding of this association, future research should include large-scale studies conducted in diverse ethnic populations.
Collapse
Affiliation(s)
- Dilara Akhter Supti
- Department of Food Technology and Nutrition Science, Noakhali Science and Technology University, Noakhali, Bangladesh
| | - Farzana Akter
- Department of Biotechnology and Genetic Engineering, Noakhali Science and Technology University, Noakhali, Bangladesh
| | - Md Imranur Rahman
- Department of Biotechnology and Genetic Engineering, Noakhali Science and Technology University, Noakhali, Bangladesh
| | - Md Adnan Munim
- Department of Biotechnology and Genetic Engineering, Noakhali Science and Technology University, Noakhali, Bangladesh
| | | | - Rabia Jahan Tarin
- Department of Biotechnology and Genetic Engineering, Noakhali Science and Technology University, Noakhali, Bangladesh
| | - Sumaiya Afroz
- Department of Biotechnology and Genetic Engineering, Noakhali Science and Technology University, Noakhali, Bangladesh
| | - Hasan Al Reza
- Department of Genetic Engineering and Biotechnology, University of Dhaka, Dhaka, Bangladesh
| | - Roksana Yeasmin
- Department of Biochemistry, Ibrahim Medical College, Dhaka, Bangladesh
| | - Mohammad Rahanur Alam
- Department of Food Technology and Nutrition Science, Noakhali Science and Technology University, Noakhali, Bangladesh
| | - Md Shahadat Hossain
- Department of Biotechnology and Genetic Engineering, Noakhali Science and Technology University, Noakhali, Bangladesh
| |
Collapse
|
12
|
Ferreira SRG, Macotela Y, Velloso LA, Mori MA. Determinants of obesity in Latin America. Nat Metab 2024; 6:409-432. [PMID: 38438626 DOI: 10.1038/s42255-024-00977-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/24/2023] [Accepted: 01/04/2024] [Indexed: 03/06/2024]
Abstract
Obesity rates are increasing almost everywhere in the world, although the pace and timing for this increase differ when populations from developed and developing countries are compared. The sharp and more recent increase in obesity rates in many Latin American countries is an example of that and results from regional characteristics that emerge from interactions between multiple factors. Aware of the complexity of enumerating these factors, we highlight eight main determinants (the physical environment, food exposure, economic and political interest, social inequity, limited access to scientific knowledge, culture, contextual behaviour and genetics) and discuss how they impact obesity rates in Latin American countries. We propose that initiatives aimed at understanding obesity and hampering obesity growth in Latin America should involve multidisciplinary, global approaches that consider these determinants to build more effective public policy and strategies, accounting for regional differences and disease complexity at the individual and systemic levels.
Collapse
Affiliation(s)
| | - Yazmín Macotela
- Instituto de Neurobiología, Universidad Nacional Autónoma de México, UNAM Campus-Juriquilla, Querétaro, Mexico
| | - Licio A Velloso
- Obesity and Comorbidities Research Center, Faculty of Medical Sciences, Universidade Estadual de Campinas, Campinas, Brazil
| | - Marcelo A Mori
- Institute of Biology, Universidade Estadual de Campinas, Campinas, Brazil.
| |
Collapse
|
13
|
Kokkorakis M, Katsarou A, Katsiki N, Mantzoros CS. Milestones in the journey towards addressing obesity; Past trials and triumphs, recent breakthroughs, and an exciting future in the era of emerging effective medical therapies and integration of effective medical therapies with metabolic surgery. Metabolism 2023; 148:155689. [PMID: 37689110 DOI: 10.1016/j.metabol.2023.155689] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/29/2023] [Revised: 08/28/2023] [Accepted: 08/28/2023] [Indexed: 09/11/2023]
Abstract
The 21st century is characterized by an increasing incidence and prevalence of obesity and the burden of its associated comorbidities, especially cardiometabolic diseases, which are reaching pandemic proportions. In the late '90s, the "black box" of adipose tissue and energy homeostasis was opened with the discovery of leptin, transforming the adipose tissue from an "inert fat-storage organ" to the largest human endocrine organ and creating the basis on which more intensified research efforts to elucidate the pathogenesis of obesity and develop novel treatments were based upon. Even though leptin was eventually not proven to be the "standalone magic bullet" for the treatment of common/polygenic obesity, it has been successful in the treatment of monogenic obesity syndromes. Additionally, it shifted the paradigm of treating obesity from a condition due to "lack of willpower" to a disease due to distinct underlying biological mechanisms for which specific pharmacotherapies would be needed in addition to lifestyle modification. Subsequently, the melanocortin pathway proved to be an equally valuable pathway for the pharmacotherapy of obesity. Melanocortin receptor agonists have recently been approved for treating certain types of syndromic obesity. Other molecules- such as incretins, implicated in energy and glucose homeostasis- are secreted by the gastrointestinal tract. Glucagon-like peptide 1 (GLP-1) is the most prominent one, with GLP-1 analogs approved for common/polygenic obesity. Unimolecular combinations with other incretins, e.g., GLP-1 with gastric inhibitory polypeptide and/or glucagon, are expected to be approved soon as more effective pharmacotherapies for obesity and its comorbidities. Unimolecular combinations with other compounds and small molecules activating the receptors of these molecules are currently under investigation as promising future pharmacotherapies. Moreover, metabolic and bariatric surgery has also demonstrated impressive results, especially in the case of morbid obesity. Consequently, this broadening therapeutic armamentarium calls for a well-thought-after and well-coordinated multidisciplinary approach, for instance, through cardiometabolic expertise centers, that would ideally address effectively and cost-effectively obesity and its comorbidities, providing tangible benefits to large segments of the population.
Collapse
Affiliation(s)
- Michail Kokkorakis
- Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Angeliki Katsarou
- Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Niki Katsiki
- Department of Nutritional Sciences and Dietetics, International Hellenic University, 57400 Thessaloniki, Greece
| | - Christos S Mantzoros
- Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA; Section of Endocrinology, VA Boston Healthcare System, Harvard Medical School, Boston, MA 02115, USA.
| |
Collapse
|
14
|
Horwitz A, Birk R. Adipose Tissue Hyperplasia and Hypertrophy in Common and Syndromic Obesity-The Case of BBS Obesity. Nutrients 2023; 15:3445. [PMID: 37571382 PMCID: PMC10421039 DOI: 10.3390/nu15153445] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2023] [Revised: 07/16/2023] [Accepted: 07/31/2023] [Indexed: 08/13/2023] Open
Abstract
Obesity is a metabolic state generated by the expansion of adipose tissue. Adipose tissue expansion depends on the interplay between hyperplasia and hypertrophy, and is mainly regulated by a complex interaction between genetics and excess energy intake. However, the genetic regulation of adipose tissue expansion is yet to be fully understood. Obesity can be divided into common multifactorial/polygenic obesity and monogenic obesity, non-syndromic and syndromic. Several genes related to obesity were found through studies of monogenic non-syndromic obesity models. However, syndromic obesity, characterized by additional features other than obesity, suggesting a more global role of the mutant genes related to the syndrome and, thus, an additional peripheral influence on the development of obesity, were hardly studied to date in this regard. This review summarizes present knowledge regarding the hyperplasia and hypertrophy of adipocytes in common obesity. Additionally, we highlight the scarce research on syndromic obesity as a model for studying adipocyte hyperplasia and hypertrophy, focusing on Bardet-Biedl syndrome (BBS). BBS obesity involves central and peripheral mechanisms, with molecular and mechanistic alternation in adipocyte hyperplasia and hypertrophy. Thus, we argue that using syndromic obesity models, such as BBS, can further advance our knowledge regarding peripheral adipocyte regulation in obesity.
Collapse
Affiliation(s)
| | - Ruth Birk
- Department of Nutrition, Faculty of Health Sciences, Ariel University, Ariel 40700, Israel;
| |
Collapse
|
15
|
Nogueiras R, Nauck MA, Tschöp MH. Gut hormone co-agonists for the treatment of obesity: from bench to bedside. Nat Metab 2023:10.1038/s42255-023-00812-z. [PMID: 37308724 DOI: 10.1038/s42255-023-00812-z] [Citation(s) in RCA: 54] [Impact Index Per Article: 27.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/23/2022] [Accepted: 04/24/2023] [Indexed: 06/14/2023]
Abstract
The discovery and development of so-called gut hormone co-agonists as a new class of drugs for the treatment of diabetes and obesity is considered a transformative breakthrough in the field. Combining action profiles of multiple gastrointestinal hormones within a single molecule, these novel therapeutics achieve synergistic metabolic benefits. The first such compound, reported in 2009, was based on balanced co-agonism at glucagon and glucagon-like peptide-1 (GLP-1) receptors. Today, several classes of gut hormone co-agonists are in development and advancing through clinical trials, including dual GLP-1-glucose-dependent insulinotropic polypeptide (GIP) co-agonists (first described in 2013), and triple GIP-GLP-1-glucagon co-agonists (initially designed in 2015). The GLP-1-GIP co-agonist tirzepatide was approved in 2022 by the US Food and Drug Administration for the treatment of type 2 diabetes, providing superior HbA1c reductions compared to basal insulin or selective GLP-1 receptor agonists. Tirzepatide also achieved unprecedented weight loss of up to 22.5%-similar to results achieved with some types of bariatric surgery-in non-diabetic individuals with obesity. In this Perspective, we summarize the discovery, development, mechanisms of action and clinical efficacy of the different types of gut hormone co-agonists, and discuss potential challenges, limitations and future developments.
Collapse
Affiliation(s)
- Ruben Nogueiras
- CIMUS, University of Santiago de Compostela, Santiago de Compostela, Spain
- Galicia Agency of Innovation, Xunta de Galicia, Santiago de Compostela, Spain
| | - Michael A Nauck
- Diabetes, Endocrinology and Metabolism Section, Medical Department I, St. Josef-Hospital, Katholisches Klinikum Bochum, Ruhr University of Bochum, Bochum, Germany
| | - Matthias H Tschöp
- Helmholtz Zentrum München, Neuherberg, Germany.
- Division of Metabolic Diseases, Department of Medicine, Technische Universität München, München, Germany.
| |
Collapse
|
16
|
Xie X, Houtz J, Liao GY, Chen Y, Xu B. Genetic Val66Met BDNF Variant Increases Hyperphagia on Fat-rich Diets in Mice. Endocrinology 2023; 164:6984997. [PMID: 36631165 DOI: 10.1210/endocr/bqad008] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/12/2022] [Revised: 01/09/2023] [Accepted: 01/10/2023] [Indexed: 01/13/2023]
Abstract
High prevalence of obesity is attributable in part to consumption of highly palatable, fat-rich foods. However, the mechanism controlling dietary fat intake is largely unknown. In this study we investigated the role of brain-derived neurotrophic factor (BDNF) in the control of dietary fat intake in a mouse model that mimics the common human Val-to-Met (Val66Met) polymorphism that impairs BDNF release via the regulated secretory pathway. BdnfMet/Met mice gained weight much faster than wild-type (WT) mice and developed severe obesity due to marked hyperphagia when they were fed HFD. Hyperphagia in these mice worsened when the fat content in their diet was increased. Conversely, mice lacking leptin exhibited similar hyperphagia on chow and HFD. When 2 diets were provided simultaneously, WT and BdnfMet/Met mice showed a comparable preference for the more palatable diet rich in either fat or sucrose, indicating that increased hyperphagia on fat-rich diets in BdnfMet/Met mice is not due to enhanced hedonic drive. In support of this interpretation, WT and BdnfMet/Met mice increased calorie intake to a similar extent during the first day after chow was switched to HFD; however, WT mice decreased HFD intake faster than BdnfMet/Met mice in subsequent days. Furthermore, we found that refeeding after fasting or nocturnal feeding with HFD activated TrkB more strongly than with chow in the hypothalamus of WT mice, whereas TrkB activation under these 2 conditions was greatly attenuated in BdnfMet/Met mice. These results indicate that satiety factors generated during HFD feeding induce BDNF release to suppress excess dietary fat intake.
Collapse
Affiliation(s)
- Xiangyang Xie
- Department of Neuroscience, UF Scripps Biomedical Research, University of Florida, Jupiter, Florida 33458, USA
| | - Jessica Houtz
- Department of Neuroscience, UF Scripps Biomedical Research, University of Florida, Jupiter, Florida 33458, USA
| | - Guey-Ying Liao
- Department of Neuroscience, UF Scripps Biomedical Research, University of Florida, Jupiter, Florida 33458, USA
| | - Yuting Chen
- Department of Neuroscience, UF Scripps Biomedical Research, University of Florida, Jupiter, Florida 33458, USA
- Skaggs Graduate School of Chemical and Biological Sciences, The Scripps Research Institute, Jupiter, Florida 33458, USA
| | - Baoji Xu
- Department of Neuroscience, UF Scripps Biomedical Research, University of Florida, Jupiter, Florida 33458, USA
- Skaggs Graduate School of Chemical and Biological Sciences, The Scripps Research Institute, Jupiter, Florida 33458, USA
| |
Collapse
|
17
|
Fernández E, McCarthy CI, Cerviño RH, Rodríguez SS, Yaneff A, Hernández J, Garrido V, Di Rocco F, Raingo J. Functional alterations of two novel MC4R mutations found in Argentinian pediatric patients with early onset obesity. Mol Cell Endocrinol 2023; 559:111777. [PMID: 36210601 DOI: 10.1016/j.mce.2022.111777] [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: 02/09/2022] [Revised: 09/08/2022] [Accepted: 09/09/2022] [Indexed: 02/03/2023]
Abstract
Loss-of-function mutations in melanocortin-4 receptor (MC4R) are the most common cause of monogenic obesity, a severe type of early-onset obesity. Our aim was to determine the prevalence of MC4R mutations in a cohort of 97 Argentinian children with early-onset obesity. We found two novel mutations (p.V52E and p.G233S) and estimated a prevalence of 2.1%. We investigated the pathogenicity of mutations in HEK293T cells expressing wild-type or mutant MC4R and found that both mutants exhibited reduced plasma membrane expression and altered agonist-induced cAMP responses, with no changes in basal activity. Besides, MC4R G233S mutant demonstrated an altered agonist-dependent inhibition of voltage-gated calcium channels type 2.2. Results using a Gαs protein inhibitor suggest that the G233S mutation could be recruiting a different G-protein signaling pathway. The identification of new mutations in MC4R and characterization of their functional impact provide tools for the diagnosis and treatment of monogenic obesity.
Collapse
Affiliation(s)
- Estefanía Fernández
- Laboratorio de Genética Molecular, Instituto Multidisciplinario de Biología Celular (IMBICE), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Universidad Nacional de La Plata (UNLP) y Comisión de Investigaciones Científicas de la Provincia de Buenos Aires (CIC-PBA), La Plata, Buenos Aires, Argentina.
| | - Clara Inés McCarthy
- Laboratorio de Electrofisiología, IMBICE, CONICET, UNLP y CIC-PBA, La Plata, Buenos Aires, Argentina.
| | - Ramiro Hector Cerviño
- Instituto de Investigaciones Farmacológicas (ININFA, UBA, CONICET), Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, Buenos Aires, Argentina.
| | - Silvia Susana Rodríguez
- Laboratorio de Electrofisiología, IMBICE, CONICET, UNLP y CIC-PBA, La Plata, Buenos Aires, Argentina.
| | - Agustín Yaneff
- Instituto de Investigaciones Farmacológicas (ININFA, UBA, CONICET), Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, Buenos Aires, Argentina.
| | - Julieta Hernández
- Servicio de Nutrición del Hospital de Niños "Sor María Ludovica" de La Plata, La Plata, Buenos Aires, Argentina.
| | - Verónica Garrido
- Servicio de Nutrición del Hospital de Niños "Sor María Ludovica" de La Plata, La Plata, Buenos Aires, Argentina.
| | - Florencia Di Rocco
- Laboratorio de Genética Molecular, Instituto Multidisciplinario de Biología Celular (IMBICE), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Universidad Nacional de La Plata (UNLP) y Comisión de Investigaciones Científicas de la Provincia de Buenos Aires (CIC-PBA), La Plata, Buenos Aires, Argentina.
| | - Jesica Raingo
- Laboratorio de Electrofisiología, IMBICE, CONICET, UNLP y CIC-PBA, La Plata, Buenos Aires, Argentina.
| |
Collapse
|
18
|
Sbraccia P, Dicker D. Obesity is a chronic progressive relapsing disease of particular interest for internal medicine. Intern Emerg Med 2023; 18:1-5. [PMID: 36273046 DOI: 10.1007/s11739-022-03129-z] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Affiliation(s)
- Paolo Sbraccia
- Department of Systems Medicine, University of Rome Tor Vergata, Via Montpellier 1, 00133, Rome, Italy.
- Unit of Internal Medicine, Obesity Center, Policlinico Tor Vergata, Rome, Italy.
| | - Dror Dicker
- Department of Internal Medicine D, Hasharon Hospital Rabin Medical Center, Petah Tikva, Israel
- Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel
| |
Collapse
|
19
|
Banik S. Genetic, Epigenetic, and Molecular Biology of Obesity: From Pathology to Therapeutics the Way Forward. JOURNAL OF THE ASSOCIATION OF PHYSICIANS OF INDIA 2022; 70:11-12. [DOI: 10.5005/japi-11001-0080] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
|
20
|
Rodríguez-López R, Gimeno-Ferrer F, do Santos DA, Ferrer-Bolufer I, Luján CG, Alcalá OZ, García-Banacloy A, Cogollos VB, Juan CS. Reviewed and updated Algorithm for Genetic Characterization of Syndromic Obesity Phenotypes. Curr Genomics 2022; 23:147-162. [PMID: 36777005 PMCID: PMC9878830 DOI: 10.2174/1389202923666220426093436] [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: 10/26/2021] [Revised: 11/30/2021] [Accepted: 02/01/2022] [Indexed: 11/22/2022] Open
Abstract
Background: Individuals with a phenotype of early-onset severe obesity associated with intellectual disability can have molecular diagnoses ranging from monogenic to complex genetic traits. Severe overweight is the major sign of a syndromic physical appearance and predicting the influence of a single gene and/or polygenic risk profile is extremely complicated among the majority of the cases. At present, considering rare monogenic bases as the principal etiology for the majority of obesity cases associated with intellectual disability is scientifically poor. The diversity of the molecular bases responsible for the two entities makes the appliance of the current routinely powerful genomics diagnostic tools essential. Objective: Clinical investigation of these difficult-to-diagnose patients requires pediatricians and neurologists to use optimized descriptions of signs and symptoms to improve genotype correlations. Methods: The use of modern integrated bioinformatics strategies which are conducted by experienced multidisciplinary clinical teams. Evaluation of the phenotype of the patient's family is also of importance. Results: The next step involves discarding the monogenic canonical obesity syndromes and considering infrequent unique molecular cases, and/or then polygenic bases. Adequate management of the application of the new technique and its diagnostic phases is essential for achieving good cost/efficiency balances. Conclusion: With the current clinical management, it is necessary to consider the potential coincidence of risk mutations for obesity in patients with genetic alterations that induce intellectual disability. In this review, we describe an updated algorithm for the molecular characterization and diagnosis of patients with a syndromic obesity phenotype.
Collapse
Affiliation(s)
- Raquel Rodríguez-López
- Laboratory of Molecular Genetics, Clinical Analysis Service, General Hospital Consortium of Valencia, Valencia, Spain;,Address correspondence to this author at the Laboratory of Molecular Genetics, Clinical Analysis Service, General Hospital Consortium of Valencia, Avenida de las Tres Cruces no. 2 46014, Valencia, Spain; Tel: 0034 963 131 800 – 437317; Fax: 0034 963 131 979; E-mail:
| | - Fátima Gimeno-Ferrer
- Laboratory of Molecular Genetics, Clinical Analysis Service, General Hospital Consortium of Valencia, Valencia, Spain
| | - David Albuquerque do Santos
- Laboratory of Molecular Genetics, Clinical Analysis Service, General Hospital Consortium of Valencia, Valencia, Spain
| | - Irene Ferrer-Bolufer
- Laboratory of Molecular Genetics, Clinical Analysis Service, General Hospital Consortium of Valencia, Valencia, Spain
| | - Carola Guzmán Luján
- Laboratory of Molecular Genetics, Clinical Analysis Service, General Hospital Consortium of Valencia, Valencia, Spain
| | - Otilia Zomeño Alcalá
- Laboratory of Molecular Genetics, Clinical Analysis Service, General Hospital Consortium of Valencia, Valencia, Spain
| | - Amor García-Banacloy
- Laboratory of Molecular Genetics, Clinical Analysis Service, General Hospital Consortium of Valencia, Valencia, Spain
| | | | - Carlos Sánchez Juan
- Endocrinology Service, General Hospital Consortium of Valencia, Valencia, Spain
| |
Collapse
|
21
|
Shalitin S, Gat-Yablonski G. Associations of Obesity with Linear Growth and Puberty. Horm Res Paediatr 2022; 95:120-136. [PMID: 34130293 DOI: 10.1159/000516171] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/19/2021] [Accepted: 03/27/2021] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND The prevalence of obesity in childhood has increased dramatically in recent decades with increased risk of developing cardiometabolic and other comorbidities. Childhood adiposity may also influence processes of growth and puberty. SUMMARY Growth patterns of obesity during childhood have been shown to be associated with increased linear growth in early childhood, leading to accelerated epiphyseal growth plate (EGP) maturation. Several hormones secreted by the adipose tissue may affect linear growth in the context of obesity, both via the growth hormone IGF-1 axis and via a direct effect on the EGP. The observation that children with obesity tend to mature earlier than lean children has led to the assumption that the degree of body fatness may trigger the neuroendocrine events that lead to pubertal onset. The most probable link between obesity and puberty is leptin and its interaction with the kisspeptin system, which is an important regulator of puberty. However, peripheral action of adipose tissue could also be involved in changes in the onset of puberty. In addition, nutritional factors, epigenetics, and endocrine-disrupting chemicals are potential mediators linking pubertal onset to obesity. In this review, we focused on interactions of obesity with linear growth and pubertal processes, based on basic research and clinical data in humans. KEY MESSAGE Children with obesity are subject to accelerated linear growth with risk of impaired adult height and early puberty, with its psychological consequences. The data highlight another important objective in combatting childhood obesity, for the prevention of abnormal growth and pubertal patterns.
Collapse
Affiliation(s)
- Shlomit Shalitin
- National Center for Childhood Diabetes, Schneider Children's Medical Center of Israel, The Jesse Z and Sara Lea Shafer Institute for Endocrinology and Diabetes, Petach Tikva, Israel.,Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Galia Gat-Yablonski
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel.,Felsenstein Medical Research Center, Petach Tikva, Israel
| |
Collapse
|
22
|
Investigation of the association between HLA-G polymorphisms and obesity. JOURNAL OF SURGERY AND MEDICINE 2022. [DOI: 10.28982/josam.930690] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
|
23
|
Müller TD, Blüher M, Tschöp MH, DiMarchi RD. Anti-obesity drug discovery: advances and challenges. Nat Rev Drug Discov 2022; 21:201-223. [PMID: 34815532 PMCID: PMC8609996 DOI: 10.1038/s41573-021-00337-8] [Citation(s) in RCA: 520] [Impact Index Per Article: 173.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/06/2021] [Indexed: 12/27/2022]
Abstract
Enormous progress has been made in the last half-century in the management of diseases closely integrated with excess body weight, such as hypertension, adult-onset diabetes and elevated cholesterol. However, the treatment of obesity itself has proven largely resistant to therapy, with anti-obesity medications (AOMs) often delivering insufficient efficacy and dubious safety. Here, we provide an overview of the history of AOM development, focusing on lessons learned and ongoing obstacles. Recent advances, including increased understanding of the molecular gut-brain communication, are inspiring the pursuit of next-generation AOMs that appear capable of safely achieving sizeable and sustained body weight loss.
Collapse
Affiliation(s)
- Timo D Müller
- Institute for Diabetes and Obesity, Helmholtz Diabetes Center, Helmholtz Zentrum München, Neuherberg, Germany.
- German Center for Diabetes Research (DZD), Neuherberg, Germany.
| | - Matthias Blüher
- Helmholtz Institute for Metabolic, Obesity and Vascular Research (HI-MAG) of the Helmholtz Zentrum München at the University of Leipzig and University Hospital Leipzig, Leipzig, Germany
| | - Matthias H Tschöp
- Helmholtz Zentrum München, Neuherberg, Germany
- Division of Metabolic Diseases, Department of Medicine, Technische Universität München, München, Germany
| | | |
Collapse
|
24
|
Hulme KD, Noye EC, Short KR, Labzin LI. Dysregulated Inflammation During Obesity: Driving Disease Severity in Influenza Virus and SARS-CoV-2 Infections. Front Immunol 2021; 12:770066. [PMID: 34777390 PMCID: PMC8581451 DOI: 10.3389/fimmu.2021.770066] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2021] [Accepted: 09/30/2021] [Indexed: 12/15/2022] Open
Abstract
Acute inflammation is a critical host defense response during viral infection. When dysregulated, inflammation drives immunopathology and tissue damage. Excessive, damaging inflammation is a hallmark of both pandemic influenza A virus (IAV) infections and Severe Acute Respiratory Syndrome-Coronavirus-2 (SARS-CoV-2) infections. Chronic, low-grade inflammation is also a feature of obesity. In recent years, obesity has been recognized as a growing pandemic with significant mortality and associated costs. Obesity is also an independent risk factor for increased disease severity and death during both IAV and SARS-CoV-2 infection. This review focuses on the effect of obesity on the inflammatory response in the context of viral respiratory infections and how this leads to increased viral pathology. Here, we will review the fundamentals of inflammation, how it is initiated in IAV and SARS-CoV-2 infection and its link to disease severity. We will examine how obesity drives chronic inflammation and trained immunity and how these impact the immune response to IAV and SARS-CoV-2. Finally, we review both medical and non-medical interventions for obesity, how they impact on the inflammatory response and how they could be used to prevent disease severity in obese patients. As projections of global obesity numbers show no sign of slowing down, future pandemic preparedness will require us to consider the metabolic health of the population. Furthermore, if weight-loss alone is insufficient to reduce the risk of increased respiratory virus-related mortality, closer attention must be paid to a patient’s history of health, and new therapeutic options identified.
Collapse
Affiliation(s)
- Katina D Hulme
- School of Chemistry and Molecular Biosciences, The University of Queensland, Brisbane, QLD, Australia
| | - Ellesandra C Noye
- School of Chemistry and Molecular Biosciences, The University of Queensland, Brisbane, QLD, Australia
| | - Kirsty R Short
- School of Chemistry and Molecular Biosciences, The University of Queensland, Brisbane, QLD, Australia.,Australian Infectious Diseases Research Centre, The University of Queensland, Brisbane, QLD, Australia
| | - Larisa I Labzin
- Australian Infectious Diseases Research Centre, The University of Queensland, Brisbane, QLD, Australia.,Institute for Molecular Bioscience, The University of Queensland, Brisbane, QLD, Australia
| |
Collapse
|
25
|
Adamska-Patruno E, Bauer W, Bielska D, Fiedorczuk J, Moroz M, Krasowska U, Czajkowski P, Wielogorska M, Maliszewska K, Puckowska S, Szczerbinski L, Lipinska D, Gorska M, Kretowski A. An Association between Diet and MC4R Genetic Polymorphism, in Relation to Obesity and Metabolic Parameters-A Cross Sectional Population-Based Study. Int J Mol Sci 2021; 22:ijms222112044. [PMID: 34769477 PMCID: PMC8584592 DOI: 10.3390/ijms222112044] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2021] [Revised: 10/27/2021] [Accepted: 11/05/2021] [Indexed: 11/30/2022] Open
Abstract
The melanocortin-4 receptor (MC4R) gene harbours one of the strongest susceptibility loci for obesity and obesity-related metabolic consequences. We analysed whether dietary factors may attenuate the associations between MC4R genotypes and obesity and metabolic parameters. In 819 participants genotyped for common MC4R polymorphisms (rs17782313, rs12970134, rs633265, and rs135034), the anthropometric measurements, body fat content and distribution (visceral and subcutaneous adipose tissue, VAT and SAT, respectively), and blood glucose, insulin, total-, LDL-, HDL-cholesterol, triglycerides concentrations, and daily macronutrient intake were assessed. ANOVA or Kruskal–Wallis tests were used, and multivariate linear regression models were developed. We observed that the CC genotype carriers (rs17782313) presented higher VAT, VAT/SAT ratio, fasting blood glucose and triglyceride concentrations when they were stratified to the upper quantiles of protein intake. An increase in energy derived from proteins was associated with higher BMI (Est. 5.74, R2 = 0.12), body fat content (Est. 8.44, R2 = 0.82), VAT (Est. 32.59, R2 = 0.06), and VAT/SAT ratio (Est. 0.96, R2 = 0.05). The AA genotype carriers (rs12970134) presented higher BMI, body fat, SAT and VAT, fasting blood glucose, triglycerides and total cholesterol concentrations. An increase in energy derived from proteins by AA carriers was associated with higher VAT (Est.19.95, R2 = 0.06) and VAT/SAT ratio (Est. 0.64, R2 = 0.05). Our findings suggest that associations of the common MC4R SNPs with obesity and its metabolic complications may be dependent on the daily dietary intake, which may open new areas for developing personalised diets for preventing and treating obesity and obesity-related comorbidities.
Collapse
Affiliation(s)
- Edyta Adamska-Patruno
- Clinical Research Centre, Medical University of Bialystok, Marii Sklodowskiej-Curie 24A, 15-276 Bialystok, Poland; (W.B.); (U.K.); (P.C.); (S.P.); (L.S.); (A.K.)
- Correspondence:
| | - Witold Bauer
- Clinical Research Centre, Medical University of Bialystok, Marii Sklodowskiej-Curie 24A, 15-276 Bialystok, Poland; (W.B.); (U.K.); (P.C.); (S.P.); (L.S.); (A.K.)
| | - Dorota Bielska
- Department of Family Medicine, Medical University of Bialystok, Mieszka I 4b, 15-054 Bialystok, Poland;
| | - Joanna Fiedorczuk
- Clinical Research Centre, Medical University of Bialystok Clinical Hospital, Marii Sklodowskiej-Curie 24A, 15-276 Bialystok, Poland; (J.F.); (M.M.)
| | - Monika Moroz
- Clinical Research Centre, Medical University of Bialystok Clinical Hospital, Marii Sklodowskiej-Curie 24A, 15-276 Bialystok, Poland; (J.F.); (M.M.)
| | - Urszula Krasowska
- Clinical Research Centre, Medical University of Bialystok, Marii Sklodowskiej-Curie 24A, 15-276 Bialystok, Poland; (W.B.); (U.K.); (P.C.); (S.P.); (L.S.); (A.K.)
| | - Przemyslaw Czajkowski
- Clinical Research Centre, Medical University of Bialystok, Marii Sklodowskiej-Curie 24A, 15-276 Bialystok, Poland; (W.B.); (U.K.); (P.C.); (S.P.); (L.S.); (A.K.)
| | - Marta Wielogorska
- Department of Endocrinology, Diabetology and Internal Medicine, Medical University of Bialystok, Marii Sklodowskiej-Curie 24A, 15-276 Bialystok, Poland; (M.W.); (K.M.); (D.L.); (M.G.)
| | - Katarzyna Maliszewska
- Department of Endocrinology, Diabetology and Internal Medicine, Medical University of Bialystok, Marii Sklodowskiej-Curie 24A, 15-276 Bialystok, Poland; (M.W.); (K.M.); (D.L.); (M.G.)
| | - Sylwia Puckowska
- Clinical Research Centre, Medical University of Bialystok, Marii Sklodowskiej-Curie 24A, 15-276 Bialystok, Poland; (W.B.); (U.K.); (P.C.); (S.P.); (L.S.); (A.K.)
| | - Lukasz Szczerbinski
- Clinical Research Centre, Medical University of Bialystok, Marii Sklodowskiej-Curie 24A, 15-276 Bialystok, Poland; (W.B.); (U.K.); (P.C.); (S.P.); (L.S.); (A.K.)
| | - Danuta Lipinska
- Department of Endocrinology, Diabetology and Internal Medicine, Medical University of Bialystok, Marii Sklodowskiej-Curie 24A, 15-276 Bialystok, Poland; (M.W.); (K.M.); (D.L.); (M.G.)
| | - Maria Gorska
- Department of Endocrinology, Diabetology and Internal Medicine, Medical University of Bialystok, Marii Sklodowskiej-Curie 24A, 15-276 Bialystok, Poland; (M.W.); (K.M.); (D.L.); (M.G.)
| | - Adam Kretowski
- Clinical Research Centre, Medical University of Bialystok, Marii Sklodowskiej-Curie 24A, 15-276 Bialystok, Poland; (W.B.); (U.K.); (P.C.); (S.P.); (L.S.); (A.K.)
- Clinical Research Centre, Medical University of Bialystok Clinical Hospital, Marii Sklodowskiej-Curie 24A, 15-276 Bialystok, Poland; (J.F.); (M.M.)
- Department of Endocrinology, Diabetology and Internal Medicine, Medical University of Bialystok, Marii Sklodowskiej-Curie 24A, 15-276 Bialystok, Poland; (M.W.); (K.M.); (D.L.); (M.G.)
| |
Collapse
|
26
|
Grannell A, le Roux CW, McGillicuddy D. "You Are Always at War With Yourself" The Perceptions and Beliefs of People With Obesity Regarding Obesity as a Disease. QUALITATIVE HEALTH RESEARCH 2021; 31:2470-2485. [PMID: 34581642 PMCID: PMC11951387 DOI: 10.1177/10497323211040767] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
Obesity as a disease remains poorly understood by key stakeholders. Here, in people living with severe obesity, perceptions and beliefs relating to obesity as a disease and obesity causality were examined. Semi-structured interviews were conducted in a tertiary care obesity clinic. 23 people with obesity (10 males, 13 females) volunteered. An overall agreement that obesity is a disease was present. Perceptions related to why obesity is and is not a disease were diverse: Lack of control and addiction, biological determinism, and personal responsibility. For weight loss maintenance, the perceptions and beliefs were heterogeneous with biological factors not considered a determinant of success. Instead, exercise, support, and willpower were described as associated with success. Barriers related to remaining in a weight-reduced state included the following: Emotional eating, sustainability of diet, occupational impact, and defeatism due to misaligned expectation and outcome. In conclusion, people living with obesity tend to agree obesity is a disease yet an incomplete understanding of the disease is present.
Collapse
|
27
|
Kabahizi A, Wallace B, Lieu L, Chau D, Dong Y, Hwang ES, Williams KW. Glucagon-like peptide-1 (GLP-1) signalling in the brain: From neural circuits and metabolism to therapeutics. Br J Pharmacol 2021; 179:600-624. [PMID: 34519026 PMCID: PMC8820188 DOI: 10.1111/bph.15682] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2021] [Revised: 08/23/2021] [Accepted: 08/27/2021] [Indexed: 12/18/2022] Open
Abstract
Glucagon‐like‐peptide‐1 (GLP‐1) derived from gut enteroendocrine cells and a discrete population of neurons in the caudal medulla acts through humoral and neural pathways to regulate satiety, gastric motility and pancreatic endocrine function. These physiological attributes contribute to GLP‐1 having a potent therapeutic action in glycaemic regulation and chronic weight management. In this review, we provide an overview of the neural circuits targeted by endogenous versus exogenous GLP‐1 and related drugs. We also highlight candidate subpopulations of neurons and cellular mechanisms responsible for the acute and chronic effects of GLP‐1 and GLP‐1 receptor agonists on energy balance and glucose metabolism. Finally, we present potential future directions to translate these findings towards the development of effective therapies for treatment of metabolic disease.
Collapse
Affiliation(s)
- Anita Kabahizi
- Department of Internal Medicine, Center for Hypothalamic Research, University of Texas Southwestern Medical Center at Dallas, Dallas, Texas, USA
| | - Briana Wallace
- Department of Internal Medicine, Center for Hypothalamic Research, University of Texas Southwestern Medical Center at Dallas, Dallas, Texas, USA
| | - Linh Lieu
- Department of Internal Medicine, Center for Hypothalamic Research, University of Texas Southwestern Medical Center at Dallas, Dallas, Texas, USA
| | - Dominic Chau
- Department of Internal Medicine, Center for Hypothalamic Research, University of Texas Southwestern Medical Center at Dallas, Dallas, Texas, USA
| | - Yanbin Dong
- Department of Internal Medicine, Center for Hypothalamic Research, University of Texas Southwestern Medical Center at Dallas, Dallas, Texas, USA
| | - Eun-Sang Hwang
- Department of Internal Medicine, Center for Hypothalamic Research, University of Texas Southwestern Medical Center at Dallas, Dallas, Texas, USA
| | - Kevin W Williams
- Department of Internal Medicine, Center for Hypothalamic Research, University of Texas Southwestern Medical Center at Dallas, Dallas, Texas, USA
| |
Collapse
|
28
|
|
29
|
Nogueiras R. MECHANISMS IN ENDOCRINOLOGY: The gut-brain axis: regulating energy balance independent of food intake. Eur J Endocrinol 2021; 185:R75-R91. [PMID: 34260412 PMCID: PMC8345901 DOI: 10.1530/eje-21-0277] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/18/2021] [Accepted: 07/14/2021] [Indexed: 12/15/2022]
Abstract
Obesity is a global pandemic with a large health and economic burden worldwide. Bodyweight is regulated by the ability of the CNS, and especially the hypothalamus, to orchestrate the function of peripheral organs that play a key role in metabolism. Gut hormones play a fundamental role in the regulation of energy balance, as they modulate not only feeding behavior but also energy expenditure and nutrient partitioning. This review examines the recent discoveries about hormones produced in the stomach and gut, which have been reported to regulate food intake and energy expenditure in preclinical models. Some of these hormones act on the hypothalamus to modulate thermogenesis and adiposity in a food intake-independent fashion. Finally, the association of these gut hormones to eating, energy expenditure, and weight loss after bariatric surgery in humans is discussed.
Collapse
Affiliation(s)
- Ruben Nogueiras
- Department of Physiology, CIMUS, USC, CIBER Fisiopatología Obesidad y Nutrición (CiberOBN), Instituto Salud Carlos III, Galician Agency of Innovation, Xunta de Galicia, Santiago de Compostela, Spain
| |
Collapse
|
30
|
Areola ED, Sabinari IW, Usman TO, Abayomi FI, Onyezia O, Onaolapo B, Adetokunbo PO, Adebanjo OO, Oladipupo FR, Olatunji LA. Sildenafil ameliorates leptin resistance and normalizes lipid handling in the hypothalamic and adipose tissues of testosterone-exposed pregnant rats. Heliyon 2021; 7:e07574. [PMID: 34337184 PMCID: PMC8313495 DOI: 10.1016/j.heliyon.2021.e07574] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2021] [Revised: 06/01/2021] [Accepted: 07/12/2021] [Indexed: 12/22/2022] Open
Abstract
Leptin and hypothalamic-adipose lipid handling are relevant in determining the shift of metabolic activities. There are scanty findings connecting glucose dysregulation as a result of hyperandrogenism during gestation to hypothalamic-adipose axis and leptin resistance. Sildenafil has recently gained attention in the prevention of intra-uterine growth restriction. The present study aimed at demonstrating the effect of sildenafil on leptin resistance and hypothalamic-adipose lipid handling in testosterone-exposed pregnant rats. Three groups of pregnant Wistar rats (n = 5/group) received olive oil (Ctr, S.C.) or testosterone propionate (Tes, 3.0 mg/kg; sc)or testosterone propionate (3.0 mg/kg; sc) and sildenafil (Tes + PDE5, 50 mg/kg; po)from gestational day 14-19. Blood samples, hypothalamus and adipose tissue were excised for biochemical analysis on day 20. Adipose and body weights, plasma leptin and adiponectin, adipose and hypothalamic leptin and triglyceride, adipose uric acid, hypothalamic Nrf2, catalase and nitric oxide were reduced following gestational testosterone exposure. Also, fasting insulin, plasma triglyceride, uric acid, leptin-adiponectin ratio, hypothalamic free fatty acid, total cholesterol, uric acid, aspartate transaminase and cyclic guanine monophosphate were elevated by testosterone exposure to pregnant animals. Sildenafil ameliorated leptin resistance and normalized hypothalamic-adipose lipid handling. Therefore, sildenafil protects against testosterone-induced leptin resistance and adverse hypothalamic-adipose lipid handling in pregnant rats.
Collapse
Affiliation(s)
- Emmanuel Damilare Areola
- HOPE Cardiometabolic Research Team and Department of Physiology, College of Health Sciences, University of Ilorin, Nigeria
| | - Isaiah Woru Sabinari
- HOPE Cardiometabolic Research Team and Department of Physiology, College of Health Sciences, University of Ilorin, Nigeria
| | - Taofeek Olumayowa Usman
- HOPE Cardiometabolic Research Team and Department of Physiology, College of Health Sciences, University of Ilorin, Nigeria.,Cardiovascular Unit, Department of Physiology, College of Health Sciences, Osun State University, Osogbo, Nigeria
| | - Faith Ifeoluwa Abayomi
- HOPE Cardiometabolic Research Team and Department of Physiology, College of Health Sciences, University of Ilorin, Nigeria
| | - Onyeka Onyezia
- HOPE Cardiometabolic Research Team and Department of Physiology, College of Health Sciences, University of Ilorin, Nigeria
| | - Bisola Onaolapo
- HOPE Cardiometabolic Research Team and Department of Physiology, College of Health Sciences, University of Ilorin, Nigeria
| | - Phebe Oluwaseun Adetokunbo
- HOPE Cardiometabolic Research Team and Department of Physiology, College of Health Sciences, University of Ilorin, Nigeria
| | - Olympus Oyewole Adebanjo
- HOPE Cardiometabolic Research Team and Department of Physiology, College of Health Sciences, University of Ilorin, Nigeria
| | - Funmilayo Rebecca Oladipupo
- HOPE Cardiometabolic Research Team and Department of Physiology, College of Health Sciences, University of Ilorin, Nigeria
| | - Lawrence Aderemi Olatunji
- HOPE Cardiometabolic Research Team and Department of Physiology, College of Health Sciences, University of Ilorin, Nigeria
| |
Collapse
|
31
|
Gastric Sensory and Motor Functions and Energy Intake in Health and Obesity-Therapeutic Implications. Nutrients 2021; 13:nu13041158. [PMID: 33915747 PMCID: PMC8065811 DOI: 10.3390/nu13041158] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2021] [Revised: 03/18/2021] [Accepted: 03/26/2021] [Indexed: 01/19/2023] Open
Abstract
Sensory and motor functions of the stomach, including gastric emptying and accommodation, have significant effects on energy consumption and appetite. Obesity is characterized by energy imbalance; altered gastric functions, such as rapid gastric emptying and large fasting gastric volume in obesity, may result in increased food intake prior to reaching usual fullness and increased appetite. Thus, many different interventions for obesity, including different diets, anti-obesity medications, bariatric endoscopy, and surgery, alter gastric functions and gastrointestinal motility. In this review, we focus on the role of the gastric and intestinal functions in food intake, pathophysiology of obesity, and obesity management.
Collapse
|
32
|
The Potential Role of Exosomes in Child and Adolescent Obesity. CHILDREN-BASEL 2021; 8:children8030196. [PMID: 33800718 PMCID: PMC7999028 DOI: 10.3390/children8030196] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/30/2020] [Revised: 02/17/2021] [Accepted: 03/02/2021] [Indexed: 02/07/2023]
Abstract
Child and adolescent obesity constitute one of the greatest contemporary public health menaces. The enduring disproportion between calorie intake and energy consumption, determined by a complex interaction of genetic, epigenetic, and environmental factors, finally leads to the development of overweight and obesity. Child and adolescent overweight/obesity promotes smoldering systemic inflammation (“para-inflammation”) and increases the likelihood of later metabolic and cardiovascular complications, including metabolic syndrome and its components, which progressively deteriorate during adulthood. Exosomes are endosome-derived extracellular vesicles that are secreted by a variety of cells, are naturally taken-up by target cells, and may be involved in many physiological and pathological processes. Over the last decade, intensive research has been conducted regarding the special role of exosomes and the non-coding (nc) RNAs they contain (primarily micro (mi) RNAs, long (l) non-coding RNAs, messenger (m) RNAs and other molecules) in inter-cellular communications. Through their action as communication mediators, exosomes may contribute to the pathogenesis of obesity and associated disorders. There is increasing evidence that exosomal miRNAs and lncRNAs are involved in pivotal processes of adipocyte biology and that, possibly, play important roles in gene regulation linked to human obesity. This review aims to improve our understanding of the roles of exosomes and their cargo in the development of obesity and related metabolic and inflammatory disorders. We examined their potential roles in adipose tissue physiology and reviewed the scarce data regarding the altered patterns of circulating miRNAs and lncRNAs observed in obese children and adolescents, compared them to the equivalent, more abundant existing findings of adult studies, and speculated on their proposed mechanisms of action. Exosomal miRNAs and lncRNAs could be applied as cardiometabolic risk biomarkers, useful in the early diagnosis and prevention of obesity. Furthermore, the targeting of crucial circulating exosomal cargo to tissues involved in the pathogenesis and maintenance of obesity could provide a novel therapeutic approach to this devastating and management-resistant pandemic.
Collapse
|
33
|
Blauw LL, Noordam R, van der Laan SW, Trompet S, Kooijman S, van Heemst D, Jukema JW, van Setten J, de Borst GJ, Tybjærg-Hansen A, Pasterkamp G, Berbée JFP, Rensen PCN. Common Genetic Variation in MC4R Does Not Affect Atherosclerotic Plaque Phenotypes and Cardiovascular Disease Outcomes. J Clin Med 2021; 10:jcm10050932. [PMID: 33804309 PMCID: PMC7957774 DOI: 10.3390/jcm10050932] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2021] [Revised: 02/11/2021] [Accepted: 02/23/2021] [Indexed: 12/01/2022] Open
Abstract
We analyzed the effects of the common BMI-increasing melanocortin 4 receptor (MC4R) rs17782313-C allele with a minor allele frequency of 0.22–0.25 on (1) cardiovascular disease outcomes in two large population-based cohorts (Copenhagen City Heart Study and Copenhagen General Population Study, n = 106,018; and UK Biobank, n = 357,426) and additionally in an elderly population at risk for cardiovascular disease (n = 5241), and on (2) atherosclerotic plaque phenotypes in samples of patients who underwent endarterectomy (n = 1439). Using regression models, we additionally analyzed whether potential associations were modified by sex or explained by changes in body mass index. We confirmed the BMI-increasing effects of +0.22 kg/m2 per additional copy of the C allele (p < 0.001). However, we found no evidence for an association of common MC4R genetic variation with coronary artery disease (HR 1.03; 95% CI 0.99, 1.07), ischemic vascular disease (HR 1.00; 95% CI 0.98, 1.03), myocardial infarction (HR 1.01; 95% CI 0.94, 1.08 and 1.02; 0.98, 1.07) or stroke (HR 0.93; 95% CI 0.85, 1.01), nor with any atherosclerotic plaque phenotype. Thus, common MC4R genetic variation, despite increasing BMI, does not affect cardiovascular disease risk in the general population or in populations at risk for cardiovascular disease.
Collapse
Affiliation(s)
- Lisanne L. Blauw
- Department Medicine, Division Endocrinology, Leiden University Medical Center, P.O. Box 9600, 2300 RC Leiden, The Netherlands; (L.L.B.); (S.K.); (J.F.P.B.); (P.C.N.R.)
- Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, P.O. Box 9600, 2300 RC Leiden, The Netherlands
| | - Raymond Noordam
- Department Medicine, Division Gerontology and Geriatrics, Leiden University Medical Center, P.O. Box 9600, 2300 RC Leiden, The Netherlands; (S.T.); (D.v.H.)
- Correspondence: ; Tel.: +31-71-52-66640
| | - Sander W. van der Laan
- Central Diagnostics Laboratory, Division Laboratory, Pharmacy and Biomedical Genetics, University Medical Center Utrecht, Utrecht University, 3584 CX Utrecht, The Netherlands; (S.W.v.d.L.); (G.P.)
| | - Stella Trompet
- Department Medicine, Division Gerontology and Geriatrics, Leiden University Medical Center, P.O. Box 9600, 2300 RC Leiden, The Netherlands; (S.T.); (D.v.H.)
| | - Sander Kooijman
- Department Medicine, Division Endocrinology, Leiden University Medical Center, P.O. Box 9600, 2300 RC Leiden, The Netherlands; (L.L.B.); (S.K.); (J.F.P.B.); (P.C.N.R.)
- Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, P.O. Box 9600, 2300 RC Leiden, The Netherlands
| | - Diana van Heemst
- Department Medicine, Division Gerontology and Geriatrics, Leiden University Medical Center, P.O. Box 9600, 2300 RC Leiden, The Netherlands; (S.T.); (D.v.H.)
| | - Johan Wouter Jukema
- Department Cardiology, Leiden University Medical Center, P.O. Box 9600, 2300 RC Leiden, The Netherlands;
| | - Jessica van Setten
- Surgery Specialties, University Medical Center Utrecht, Utrecht University, 3584 CX Utrecht, The Netherlands;
| | - Gert J. de Borst
- Department Cardiology, University Medical Center Utrecht, Utrecht University, 3584 CX Utrecht, The Netherlands;
| | - Anne Tybjærg-Hansen
- Department Clinical Biochemistry, Rigshospitalet, Blegdamsvej 9, DK-2100 Copenhagen, Denmark;
- The Copenhagen City Heart Study, Frederiksberg Hospital, Nordre Fasanvej 57, DK-2000 Frederiksberg, Denmark
- The Copenhagen General Population Study and Gentofte Hospital, Herlev Ringvej 75, DK-2730 Herlev, Denmark
- Copenhagen University Hospitals and Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, DK-2100 Copenhagen, Denmark
| | - Gerard Pasterkamp
- Central Diagnostics Laboratory, Division Laboratory, Pharmacy and Biomedical Genetics, University Medical Center Utrecht, Utrecht University, 3584 CX Utrecht, The Netherlands; (S.W.v.d.L.); (G.P.)
| | - Jimmy F. P. Berbée
- Department Medicine, Division Endocrinology, Leiden University Medical Center, P.O. Box 9600, 2300 RC Leiden, The Netherlands; (L.L.B.); (S.K.); (J.F.P.B.); (P.C.N.R.)
- Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, P.O. Box 9600, 2300 RC Leiden, The Netherlands
| | - Patrick C. N. Rensen
- Department Medicine, Division Endocrinology, Leiden University Medical Center, P.O. Box 9600, 2300 RC Leiden, The Netherlands; (L.L.B.); (S.K.); (J.F.P.B.); (P.C.N.R.)
- Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, P.O. Box 9600, 2300 RC Leiden, The Netherlands
| |
Collapse
|
34
|
Dodd GT, Kim SJ, Méquinion M, Xirouchaki CE, Brüning JC, Andrews ZB, Tiganis T. Insulin signaling in AgRP neurons regulates meal size to limit glucose excursions and insulin resistance. SCIENCE ADVANCES 2021; 7:7/9/eabf4100. [PMID: 33637536 PMCID: PMC7909880 DOI: 10.1126/sciadv.abf4100] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/24/2020] [Accepted: 01/14/2021] [Indexed: 05/17/2023]
Abstract
The importance of hypothalamic insulin signaling on feeding and glucose metabolism remains unclear. We report that insulin acts on AgRP neurons to acutely decrease meal size and thereby limit postprandial glucose and insulin excursions. The promotion of insulin signaling in AgRP neurons decreased meal size without altering total caloric intake, whereas the genetic ablation of the insulin receptor had the opposite effect. The promotion of insulin signaling also decreased the intake of sucrose-sweetened water or high-fat food over standard chow, without influencing food-seeking and hedonic behaviors. The ability of heightened insulin signaling to override the hedonistic consumption of highly palatable high-fat food attenuated the development of systemic insulin resistance, without affecting body weight. Our findings define an unprecedented mechanism by which insulin acutely influences glucose metabolism. Approaches that enhance insulin signaling in AgRP neurons may provide a means for altering feeding behavior in a nutrient-dense environment to combat the metabolic syndrome.
Collapse
Affiliation(s)
- Garron T Dodd
- Metabolism, Diabetes and Obesity Program, Monash Biomedicine Discovery Institute, Monash University, Clayton, VIC 3800, Australia.
- Department of Biochemistry and Molecular Biology, Monash University, Clayton, VIC 3800, Australia
| | - Seung Jae Kim
- Metabolism, Diabetes and Obesity Program, Monash Biomedicine Discovery Institute, Monash University, Clayton, VIC 3800, Australia
- Department of Biochemistry and Molecular Biology, Monash University, Clayton, VIC 3800, Australia
| | - Mathieu Méquinion
- Metabolism, Diabetes and Obesity Program, Monash Biomedicine Discovery Institute, Monash University, Clayton, VIC 3800, Australia
- Department of Physiology, Monash University, VIC 3800, Australia
| | - Chrysovalantou E Xirouchaki
- Metabolism, Diabetes and Obesity Program, Monash Biomedicine Discovery Institute, Monash University, Clayton, VIC 3800, Australia
- Department of Biochemistry and Molecular Biology, Monash University, Clayton, VIC 3800, Australia
| | - Jens C Brüning
- Max Planck Institute for Metabolism Research, Department of Neuronal Control of Metabolism, Gleueler Str. 50, 50931 Cologne, Germany
- Center for Endocrinology, Diabetes, and Preventive Medicine (CEDP), University Hospital Cologne, Kerpener Str. 26, 50924 Cologne, Germany
- Excellence Cluster on Cellular Stress Responses in Aging Associated Diseases (CECAD) and Center of Molecular Medicine Cologne (CMMC), University of Cologne, Joseph-Stelzmann-Str. 26, 50931 Cologne, Germany
- National Center for Diabetes Research (DZD), Ingolstädter Land Str. 1, 85764 Neuherberg, Germany
| | - Zane B Andrews
- Metabolism, Diabetes and Obesity Program, Monash Biomedicine Discovery Institute, Monash University, Clayton, VIC 3800, Australia
- Department of Physiology, Monash University, VIC 3800, Australia
| | - Tony Tiganis
- Metabolism, Diabetes and Obesity Program, Monash Biomedicine Discovery Institute, Monash University, Clayton, VIC 3800, Australia.
- Department of Biochemistry and Molecular Biology, Monash University, Clayton, VIC 3800, Australia
- Monash Metabolic Phenotyping Facility, Monash University, VIC, Australia
- Peter MacCallum Cancer Centre, Melbourne, VIC 3000, Australia
| |
Collapse
|
35
|
O'Rahilly S. "Treasure Your Exceptions"-Studying Human Extreme Phenotypes to Illuminate Metabolic Health and Disease: The 2019 Banting Medal for Scientific Achievement Lecture. Diabetes 2021; 70:29-38. [PMID: 33355307 PMCID: PMC7881844 DOI: 10.2337/dbi19-0037] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
The study of humans with genetic mutations which lead to a substantial disturbance of physiological processes has made a contribution to biomedical science that is disproportionate to the rarity of affected individuals. In this lecture, I discuss examples of where such studies have helped to illuminate two areas of human metabolism. First, the control of insulin sensitivity and its disruption in states of insulin resistance and second, the regulation of energy balance and its disturbances in obesity.
Collapse
Affiliation(s)
- Stephen O'Rahilly
- MRC Metabolic Diseases Unit, Wellcome-MRC Institute of Metabolic Science, Addenbrooke's Treatment Centre, University of Cambridge, Cambridge, U.K.
| |
Collapse
|
36
|
Abstract
PURPOSE OF REVIEW Eating behaviours are hypothesised to be the behavioural expression of genetic risk of obesity. In this review, we summarise findings from behavioural genetic research on the association between genetic risk for obesity and validated psychometrics measures of eating behaviours in children and adults (published in the past 10 years). RECENT FINDINGS Twin studies have produced some evidence for a shared genetic aetiology underlying body mass index and eating behaviours. Studies using measured genetic susceptibility to obesity have suggested that increased genetic liability for obesity is associated with variation in obesogenic eating behaviours such as emotional and uncontrolled eating. More research on this topic is needed. Especially longitudinal studies using genetically sensitive designs to investigate the direction of genetic pathways between genetic liability of eating behaviours to weight and vice versa, as well as the potential subsequent link to eating disorders.
Collapse
Affiliation(s)
- Moritz Herle
- Department of Biostatistics & Health Informatics, Institute of Psychiatry, Psychology & Neuroscience, Kings College London, 16 De Crespigny Park, London, SE5 8AF, UK.
| | - Andrea D Smith
- Research Department of Behavioural Science and Health, University College London, London, UK
| | | | - Clare Llewellyn
- Research Department of Behavioural Science and Health, University College London, London, UK
| |
Collapse
|
37
|
Emílio-Silva MT, Rodrigues VP, Bueno G, Ohara R, Martins MG, Horta-Júnior JAC, Branco LGS, Rocha LRM, Hiruma-Lima CA. Hypothermic Effect of Acute Citral Treatment during LPS-induced Systemic Inflammation in Obese Mice: Reduction of Serum TNF-α and Leptin Levels. Biomolecules 2020; 10:E1454. [PMID: 33080865 PMCID: PMC7603063 DOI: 10.3390/biom10101454] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2020] [Revised: 09/16/2020] [Accepted: 10/14/2020] [Indexed: 12/12/2022] Open
Abstract
Citral is a mixture of monoterpenes present in the essential oil of several plants, such as Cymbopogon citratus and Zingiber officinale, possessing anti-inflammatory, anti-ulcerogenic, and antipyretic actions. We investigated the action of citral on body temperature (Tb) and inflammatory signaling in eutrophic and obese mice during Systemic Inflammation (SI) induced by Lipopolysaccharide (LPS). Thus, we assessed the effect of citral (25, 100, and 300 mg/kg) and ibuprofen in LPS-induced SI in Swiss male mice fed a standard diet (SD) or high-fat diet (HFD) for 12 weeks. Following SI induction, we measured Tb and collected the serum, hypothalamus, and gastric mucosa for biochemical measurements. Acute treatment with citral decreased the Tb of both SD and HFD-fed animals. Citral (300 mg/kg) treatment caused a significantly lower Tb variation in HFD-fed animals than in those fed the SD. Citral reduced peripheral levels of tumor necrosis factor (TNF)-α in SD and HFD mice and decreased serum leptin concentration in HFD mice 90 min after the LPS challenge. Furthermore, citral also reduced interleukin (IL)-6 levels in the hypothalamus of obese mice. In summary, citral effectively reduced Tb during SI by reducing inflammatory mediators with a distinct action profile in HFD mice when compared with SD.
Collapse
Affiliation(s)
- Maycon T. Emílio-Silva
- Department of Structural and Functional Biology (Physiology), Institute of Biosciences, São Paulo State University (UNESP), Botucatu, São Paulo 18618-970, Brazil; (M.T.E.-S.); (V.P.R.); (G.B.); (R.O.); (L.R.M.R.)
| | - Vinicius P. Rodrigues
- Department of Structural and Functional Biology (Physiology), Institute of Biosciences, São Paulo State University (UNESP), Botucatu, São Paulo 18618-970, Brazil; (M.T.E.-S.); (V.P.R.); (G.B.); (R.O.); (L.R.M.R.)
| | - Gabriela Bueno
- Department of Structural and Functional Biology (Physiology), Institute of Biosciences, São Paulo State University (UNESP), Botucatu, São Paulo 18618-970, Brazil; (M.T.E.-S.); (V.P.R.); (G.B.); (R.O.); (L.R.M.R.)
| | - Rie Ohara
- Department of Structural and Functional Biology (Physiology), Institute of Biosciences, São Paulo State University (UNESP), Botucatu, São Paulo 18618-970, Brazil; (M.T.E.-S.); (V.P.R.); (G.B.); (R.O.); (L.R.M.R.)
| | - Marina G. Martins
- Department of Physiology, Institute of Biosciences, University of São Paulo (USP), São Paulo 05508-090, Brazil;
| | - José A. C. Horta-Júnior
- Department of Structural and Functional Biology (Anatomy), Institute of Biosciences, São Paulo State University (UNESP), Botucatu, São Paulo 18618-689, Brazil;
| | - Luiz G. S. Branco
- Department of Basic and Oral Biology, Dental School of Ribeirão Preto, University of São Paulo (USP), Ribeirão Preto, São Paulo 14040-904, Brazil;
| | - Lúcia R. M. Rocha
- Department of Structural and Functional Biology (Physiology), Institute of Biosciences, São Paulo State University (UNESP), Botucatu, São Paulo 18618-970, Brazil; (M.T.E.-S.); (V.P.R.); (G.B.); (R.O.); (L.R.M.R.)
| | - Clélia A. Hiruma-Lima
- Department of Structural and Functional Biology (Physiology), Institute of Biosciences, São Paulo State University (UNESP), Botucatu, São Paulo 18618-970, Brazil; (M.T.E.-S.); (V.P.R.); (G.B.); (R.O.); (L.R.M.R.)
| |
Collapse
|
38
|
Al-Nbaheen MS. Impact of weight loss predictors in severe-morbid obesity patients in the Saudi population. Saudi J Biol Sci 2020; 27:2509-2513. [PMID: 32994706 PMCID: PMC7499111 DOI: 10.1016/j.sjbs.2020.03.015] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2020] [Revised: 03/02/2020] [Accepted: 03/08/2020] [Indexed: 12/26/2022] Open
Abstract
Universally, obesity has been affected more than 650 million and converts as global health problem. Obesity is equally affecting starting from children to elder population. Obese subjects are converting into severe obese and then into morbid obesity. Body mass index is proning from 30 to 50 kg/m2 in the adult population. Obesity is connected with the future complications of hypertension, type 2 diabetes mellitus, cardiovascular, stroke, osteoarthritis, obstructive sleep apnea and liver diseases. Loosing of body fat is the only option to avoid obesity and this could be achieved with routine physical activity and diet modifications. Obesity subjects may fail to achieve the daily routine activities or insufficient activity may be involved and finally fail to lose the body fat after the medical course. Then these severe or morbidity obese can be lose with the existing surgery. Currently, Bariatric Surgery (BS) has become the active treatment for long-term weight loss. Various types (Roux-en-Y gastric bypass, sleeve gastrectomy and duodenal switch and the jejunoileal bypass) of BS are performed on the gastrointestinal tract. Throughout the world population, BS has found to be safe in losing the weight and avoiding the future and long-term complications. The prevalence of overweight and obesity in Saudi Arabia is an issue in terms of incidence and health consequences. Maximum obesity studies involved in Saudi Arabia has proven to be develop the long-term complications in the future involving from child to morbid obesity. Limited bariatric studies carried out in the Saudi subjects confirmed as effective tool in lowering the body fat and avoiding the life-threatened complications of human diseases. So, this review recommends BS as effective and safe surgical treatment to lose body fat in the Saudi population. However, post-operative monitoring is mandatory to follow-up.
Collapse
|
39
|
Visweswaraiah N, Nathan K. Adolescent Obesity and Eating Disorders: Can Calorie Restriction have a Positive Impact. CURRENT NUTRITION & FOOD SCIENCE 2020. [DOI: 10.2174/1573401315666190114153400] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Background:
The current obesogenic environment with relatively increased affordability
and availability of high calorie food and beverages, has led to an alarming increase in the prevalence
of obesity and related lifestyle disorders in children and adolescents, predisposing them to accelerated
aging. The increased prevalence may be due to the eating behavior of adolescents, their genetic
and molecular etiology and/or due to the impact of psychological stress and their wrong lifestyle
choices. Calorie restriction has been extensively researched for reducing the obesity in adolescents
and adults but is yet to be successfully implemented.
Objective:
The present review paper focuses on the types of calorie restriction diets, the role of its
mimics and the nutrigenomic mechanisms that may be helpful in reducing obesity and related disorders
in the adolescents. The role of behavioral therapeutic techniques and physical activity has also
been highlighted in addition to the calorie restricted diet for bringing about an overall lifestyle modification
in the management of obesity.
Conclusion:
Food preferences are acquired in childhood and sound nutritional practices should be
established in childhood to prevent lifestyle disorders and premature aging. Though CR is a known
and preferred non-pharmacological intervention in the management of obesity, its implemention has
not been explored and evaluated extensively. This is a vital area that needs scientific research as the
goals of obesity managements are no longer just weight loss through dietary restrictions. An interdisciplinary
method to lifestyle modification in the management of adolescent obesity addressing all
physiological and psychosocial aspects is recommended.
Collapse
Affiliation(s)
- Naveen Visweswaraiah
- Foundation for Assessment and Integration of Traditional Health Systems, Bengaluru, Karnataka, 560027, India
| | - Kousalya Nathan
- Department of Lifestyle Management, Apollo Spectra Hospitals, MRC Nagar RA Puram, Chennai, 600028, India
| |
Collapse
|
40
|
Zhang C, He X, Sheng Y, Yang C, Xu J, Zheng S, Liu J, Xu W, Luo Y, Huang K. Allicin-induced host-gut microbe interactions improves energy homeostasis. FASEB J 2020; 34:10682-10698. [PMID: 32619085 DOI: 10.1096/fj.202001007r] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2020] [Revised: 05/25/2020] [Accepted: 06/01/2020] [Indexed: 12/12/2022]
Abstract
Allicin (diallylthiosulfinate) is a natural food compound with multiple biological and pharmacological functions. However, the mechanism of beneficial role of Allicin on energy homeostasis is not well studied. Gut microbiota (GM) profoundly affects host metabolism via microbiota-host interactions and coevolution. Here, we investigated the interventions of beneficial microbiome induced by Allicin on energy homeostasis, particularly obesity, and related complications. Interestingly, Allicin treatment significantly improved GM composition and induced the most significant alteration enrichment of Bifidobacterium and Lactobacillus. Importantly, transplantation of the Allicin-induced GM to HFD mice (AGMT) played a remarkable role in decreasing adiposity, maintaining glucose homeostasis, and ameliorating hepatic steatosis. Furthermore, AGMT was effective in modulating lipid metabolism, activated brown adipose tissues (BATs), induced browning in sWAT, reduced inflammation, and inhibited the degradation of intestinal villi. Mechanically, AGMT significantly increased Blautia [short-chain fatty acids (SCFAs)-producing microbiota] and Bifidobacterium in HFD mice, also increased the SCFAs in the cecum, which has been proved many beneficial effects on energy homeostasis. Our study highlights that Allicin-induced host-gut microbe interactions plays an important role in regulating energy homeostasis, which provides a promising potential therapy for obesity and metabolic disorders based on host-microbe interactions.
Collapse
Affiliation(s)
- Chuanhai Zhang
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, China.,Key Laboratory of Safety Assessment of Genetically Modified Organism (Food Safety), Ministry of Agriculture, Beijing, China
| | - Xiaoyun He
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, China.,Key Laboratory of Safety Assessment of Genetically Modified Organism (Food Safety), Ministry of Agriculture, Beijing, China
| | - Yao Sheng
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, China.,Key Laboratory of Safety Assessment of Genetically Modified Organism (Food Safety), Ministry of Agriculture, Beijing, China
| | - Cui Yang
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, China.,Key Laboratory of Safety Assessment of Genetically Modified Organism (Food Safety), Ministry of Agriculture, Beijing, China
| | - Jia Xu
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, China.,Key Laboratory of Safety Assessment of Genetically Modified Organism (Food Safety), Ministry of Agriculture, Beijing, China
| | - Shujuan Zheng
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, China.,Key Laboratory of Safety Assessment of Genetically Modified Organism (Food Safety), Ministry of Agriculture, Beijing, China
| | - Junyu Liu
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, China.,Key Laboratory of Safety Assessment of Genetically Modified Organism (Food Safety), Ministry of Agriculture, Beijing, China
| | - Wentao Xu
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, China.,Key Laboratory of Safety Assessment of Genetically Modified Organism (Food Safety), Ministry of Agriculture, Beijing, China
| | - Yunbo Luo
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, China.,Key Laboratory of Safety Assessment of Genetically Modified Organism (Food Safety), Ministry of Agriculture, Beijing, China
| | - Kunlun Huang
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, China.,Key Laboratory of Safety Assessment of Genetically Modified Organism (Food Safety), Ministry of Agriculture, Beijing, China
| |
Collapse
|
41
|
Olsson LM, Poitou C, Tremaroli V, Coupaye M, Aron-Wisnewsky J, Bäckhed F, Clément K, Caesar R. Gut microbiota of obese subjects with Prader-Willi syndrome is linked to metabolic health. Gut 2020; 69:1229-1238. [PMID: 31611297 PMCID: PMC7306984 DOI: 10.1136/gutjnl-2019-319322] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/20/2019] [Revised: 09/20/2019] [Accepted: 09/23/2019] [Indexed: 12/11/2022]
Abstract
OBJECTIVE The gut microbiota has been implicated in the aetiology of obesity and associated comorbidities. Patients with Prader-Willi syndrome (PWS) are obese but partly protected against insulin resistance. We hypothesised that the gut microbiota of PWS patients differs from that of non-genetically obese controls and correlate to metabolic health. Therefore, here we used PWS as a model to study the role of gut microbiota in the prevention of metabolic complications linked to obesity. DESIGN We conducted a case-control study with 17 adult PWS patients and 17 obese subjects matched for body fat mass index, gender and age. The subjects were metabolically characterised and faecal microbiota was profiled by 16S ribosomal RNA gene sequencing. The patients' parents were used as a non-obese control group. Stool samples from two PWS patients and two obese controls were used for faecal microbiota transplantations in germ-free mice to examine the impact of the microbiota on glucose metabolism. RESULTS The composition of the faecal microbiota in patients with PWS differed from that of obese controls, and was characterised by higher phylogenetic diversity and increased abundance of several taxa such as Akkermansia, Desulfovibrio and Archaea, and decreased abundance of Dorea. Microbial taxa prevalent in the PWS microbiota were associated with markers of insulin sensitivity. Improved insulin resistance of PWS was partly transmitted by faecal microbiota transplantations into germ-free mice. CONCLUSION The gut microbiota of PWS patients is similar to that of their non-obese parents and might play a role for the protection of PWS patients from metabolic complications.
Collapse
Affiliation(s)
- Lisa M Olsson
- The Wallenberg Laboratory, Department of Molecular and Clinical Medicine, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Christine Poitou
- INSERM, Nutrition and obesities; systemic approaches (NutriOmics) research Unit, Sorbonne Universite, Paris, Île-de-France, France
- Assistance Publique-Hôpitaux de Paris, Reference Center for Rare Diseases (Prader-Willi Syndrome), Nutrition Department, University Hospital Pitié Salpêtrière, Paris, Île-de-France, France
| | - Valentina Tremaroli
- The Wallenberg Laboratory, Department of Molecular and Clinical Medicine, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Muriel Coupaye
- Assistance Publique-Hôpitaux de Paris, Reference Center for Rare Diseases (Prader-Willi Syndrome), Nutrition Department, University Hospital Pitié Salpêtrière, Paris, Île-de-France, France
| | - Judith Aron-Wisnewsky
- INSERM, Nutrition and obesities; systemic approaches (NutriOmics) research Unit, Sorbonne Universite, Paris, Île-de-France, France
- Assistance Publique-Hôpitaux de Paris, Reference Center for Rare Diseases (Prader-Willi Syndrome), Nutrition Department, University Hospital Pitié Salpêtrière, Paris, Île-de-France, France
| | - Fredrik Bäckhed
- The Wallenberg Laboratory, Department of Molecular and Clinical Medicine, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Novo Nordisk Foundation Center for Basic Metabolic Research, Section for Metabolic Receptology and Enteroendocrinology, Faculty of Health Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Karine Clément
- INSERM, Nutrition and obesities; systemic approaches (NutriOmics) research Unit, Sorbonne Universite, Paris, Île-de-France, France
- Assistance Publique-Hôpitaux de Paris, Reference Center for Rare Diseases (Prader-Willi Syndrome), Nutrition Department, University Hospital Pitié Salpêtrière, Paris, Île-de-France, France
| | - Robert Caesar
- The Wallenberg Laboratory, Department of Molecular and Clinical Medicine, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| |
Collapse
|
42
|
Effects of metabolic state on the regulation of melanocortin circuits. Physiol Behav 2020; 224:113039. [PMID: 32610101 PMCID: PMC7387173 DOI: 10.1016/j.physbeh.2020.113039] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2019] [Revised: 06/01/2020] [Accepted: 06/26/2020] [Indexed: 12/14/2022]
Abstract
Dysfunction in neurophysiological systems that regulate food intake and metabolism are at least partly responsible for obesity and related comorbidities. An important component of this process is the hypothalamic melanocortin system, where an imbalance can result in severe obesity and deficits in glucose metabolism. Exercise offers many health benefits related to cardiovascular improvements, hunger control, and blood glucose homeostasis. However, the molecular mechanism underlying the exercise-induced improvements to the melanocortin system remain undefined. Here, we review the role of the melanocortin system to sense hormonal, nutrient, and neuronal signals of energy status. This information is then relayed onto secondary neurons in order to regulate physiological parameters, which promote proper energy and glucose balance. We also provide an overview on the effects of physical exercise to induce biophysical changes in the melanocortin circuit which may regulate food intake, glucose metabolism and improve overall metabolic health.
Collapse
|
43
|
Mason KA, Schoelwer MJ, Rogol AD. Androgens During Infancy, Childhood, and Adolescence: Physiology and Use in Clinical Practice. Endocr Rev 2020; 41:5770947. [PMID: 32115641 DOI: 10.1210/endrev/bnaa003] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/23/2019] [Accepted: 02/28/2020] [Indexed: 12/29/2022]
Abstract
We provide an in-depth review of the role of androgens in male maturation and development, from the fetal stage through adolescence into emerging adulthood, and discuss the treatment of disorders of androgen production throughout these time periods. Testosterone, the primary androgen produced by males, has both anabolic and androgenic effects. Androgen exposure induces virilization and anabolic body composition changes during fetal development, influences growth and virilization during infancy, and stimulates development of secondary sexual characteristics, growth acceleration, bone mass accrual, and alterations of body composition during puberty. Disorders of androgen production may be subdivided into hypo- or hypergonadotropic hypogonadism. Hypogonadotropic hypogonadism may be either congenital or acquired (resulting from cranial radiation, trauma, or less common causes). Hypergonadotropic hypogonadism occurs in males with Klinefelter syndrome and may occur in response to pelvic radiation, certain chemotherapeutic agents, and less common causes. These disorders all require testosterone replacement therapy during pubertal maturation and many require lifelong replacement. Androgen (or gonadotropin) therapy is clearly beneficial in those with persistent hypogonadism and self-limited delayed puberty and is now widely used in transgender male adolescents. With more widespread use and newer formulations approved for adults, data from long-term randomized placebo-controlled trials are needed to enable pediatricians to identify the optimal age of initiation, route of administration, and dosing frequency to address the unique needs of their patients.
Collapse
Affiliation(s)
- Kelly A Mason
- Department of Pediatrics, University of Virginia, Charlottesville, Virginia
| | | | - Alan D Rogol
- Department of Pediatrics, University of Virginia, Charlottesville, Virginia
| |
Collapse
|
44
|
Zhang C, He X, Sheng Y, Xu J, Yang C, Zheng S, Liu J, Li H, Ge J, Yang M, Zhai B, Xu W, Luo Y, Huang K. Allicin Regulates Energy Homeostasis through Brown Adipose Tissue. iScience 2020; 23:101113. [PMID: 32413611 PMCID: PMC7226876 DOI: 10.1016/j.isci.2020.101113] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2019] [Revised: 01/15/2020] [Accepted: 04/24/2020] [Indexed: 12/19/2022] Open
Abstract
Brown adipose tissue (BAT) is a promising potential therapeutic target for the treatment of obesity and related metabolic diseases. Allicin, a natural product in garlic, has multiple biological and pharmacological functions. However, the role of allicin in the regulation of metabolic organs, particularly BAT activation, has not been well studied. Here, we show that allicin imparts a significant effect by inhibiting body weight gain, decreasing adiposity, maintaining glucose homeostasis, improving insulin resistance, and ameliorating hepatic steatosis in obese mice. These observations strongly correlate with the activation of BAT. Notably, allicin plays a role in BAT activation, which may partly contribute to the Sirt1-PGC1α-Tfam pathway. In addition, allicin can significantly increase the succinylation levels of UCP1 in BAT by inhibiting sirt5, whereas excess allicin induces autophagy/mitophagy and mitochondrial dysfunction. Thus, our findings point to allicin as a promising therapeutic approach for the treatment of obesity and metabolic disorders. Allicin reduces adiposity and maintains glucose homeostasis Allicin activates the brown adipocytes and increases the energy expenditure Allicin enhances BAT activity partly through SIRT1-PGC1a-Tfam signaling pathway Allicin regulates mitophagy via suppressed sirt5-mediated succinylation accumulation
Collapse
Affiliation(s)
- Chuanhai Zhang
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing 100083, China; Key Laboratory of Safety Assessment of Genetically Modified Organism (Food Safety), Ministry of Agriculture, Beijing 100083, China
| | - Xiaoyun He
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing 100083, China; Key Laboratory of Safety Assessment of Genetically Modified Organism (Food Safety), Ministry of Agriculture, Beijing 100083, China
| | - Yao Sheng
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing 100083, China; Key Laboratory of Safety Assessment of Genetically Modified Organism (Food Safety), Ministry of Agriculture, Beijing 100083, China
| | - Jia Xu
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing 100083, China; Key Laboratory of Safety Assessment of Genetically Modified Organism (Food Safety), Ministry of Agriculture, Beijing 100083, China
| | - Cui Yang
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing 100083, China; Key Laboratory of Safety Assessment of Genetically Modified Organism (Food Safety), Ministry of Agriculture, Beijing 100083, China
| | - Shujuan Zheng
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing 100083, China; Key Laboratory of Safety Assessment of Genetically Modified Organism (Food Safety), Ministry of Agriculture, Beijing 100083, China
| | - Junyu Liu
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing 100083, China; Key Laboratory of Safety Assessment of Genetically Modified Organism (Food Safety), Ministry of Agriculture, Beijing 100083, China
| | - Haoyu Li
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing 100083, China; Key Laboratory of Safety Assessment of Genetically Modified Organism (Food Safety), Ministry of Agriculture, Beijing 100083, China
| | - Jianbing Ge
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing 100083, China; Key Laboratory of Safety Assessment of Genetically Modified Organism (Food Safety), Ministry of Agriculture, Beijing 100083, China
| | - Minglan Yang
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing 100083, China; Key Laboratory of Safety Assessment of Genetically Modified Organism (Food Safety), Ministry of Agriculture, Beijing 100083, China
| | - Baiqiang Zhai
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing 100083, China; Key Laboratory of Safety Assessment of Genetically Modified Organism (Food Safety), Ministry of Agriculture, Beijing 100083, China
| | - Wentao Xu
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing 100083, China; Key Laboratory of Safety Assessment of Genetically Modified Organism (Food Safety), Ministry of Agriculture, Beijing 100083, China
| | - Yunbo Luo
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing 100083, China; Key Laboratory of Safety Assessment of Genetically Modified Organism (Food Safety), Ministry of Agriculture, Beijing 100083, China.
| | - Kunlun Huang
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing 100083, China; Key Laboratory of Safety Assessment of Genetically Modified Organism (Food Safety), Ministry of Agriculture, Beijing 100083, China.
| |
Collapse
|
45
|
Scotti L, Monteiro AFM, de Oliveira Viana J, Mendonça Junior FJB, Ishiki HM, Tchouboun EN, Santos R, Scotti MT. Multi-Target Drugs Against Metabolic Disorders. Endocr Metab Immune Disord Drug Targets 2020; 19:402-418. [PMID: 30556507 DOI: 10.2174/1871530319666181217123357] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/24/2018] [Revised: 06/18/2018] [Accepted: 06/27/2018] [Indexed: 01/13/2023]
Abstract
BACKGROUND Metabolic disorders are a major cause of illness and death worldwide. Metabolism is the process by which the body makes energy from proteins, carbohydrates, and fats; chemically breaking these down in the digestive system towards sugars and acids which constitute the human body's fuel for immediate use, or to store in body tissues, such as the liver, muscles, and body fat. OBJECTIVE The efficiency of treatments for multifactor diseases has not been proved. It is accepted that to manage multifactor diseases, simultaneous modulation of multiple targets is required leading to the development of new strategies for discovery and development of drugs against metabolic disorders. METHODS In silico studies are increasingly being applied by researchers due to reductions in time and costs for new prototype synthesis; obtaining substances that present better therapeutic profiles. DISCUSSION In the present work, in addition to discussing multi-target drug discovery and the contributions of in silico studies to rational bioactive planning against metabolic disorders such as diabetes and obesity, we review various in silico study contributions to the fight against human metabolic pathologies. CONCLUSION In this review, we have presented various studies involved in the treatment of metabolic disorders; attempting to obtain hybrid molecules with pharmacological activity against various targets and expanding biological activity by using different mechanisms of action to treat a single pathology.
Collapse
Affiliation(s)
- Luciana Scotti
- Teaching and Research Management - University Hospital, Federal University of Paraíba, João Pessoa, PB, Brazil.,Postgraduate Program in Natural and Synthetic Bioactive Products, Federal University of Paraiba, Joao Pessoa, PB, Brazil
| | - Alex France Messias Monteiro
- Postgraduate Program in Natural and Synthetic Bioactive Products, Federal University of Paraiba, Joao Pessoa, PB, Brazil
| | - Jéssika de Oliveira Viana
- Postgraduate Program in Natural and Synthetic Bioactive Products, Federal University of Paraiba, Joao Pessoa, PB, Brazil
| | - Francisco Jaime Bezerra Mendonça Junior
- Postgraduate Program in Natural and Synthetic Bioactive Products, Federal University of Paraiba, Joao Pessoa, PB, Brazil.,Laboratory of Synthesis and Drug Delivery, Department of Biological Science, State University of Paraiba, Joao Pessoa, PB, Brazil
| | - Hamilton M Ishiki
- University of Western Sao Paulo (Unoeste), Presidente Prudente, SP, Brazil
| | | | - Rodrigo Santos
- Laboratory of Synthesis and Drug Delivery, Department of Biological Science, State University of Paraiba, Joao Pessoa, PB, Brazil
| | - Marcus Tullius Scotti
- Postgraduate Program in Natural and Synthetic Bioactive Products, Federal University of Paraiba, Joao Pessoa, PB, Brazil
| |
Collapse
|
46
|
Stern SA, Doerig KR, Azevedo EP, Stoffel E, Friedman JM. Control of non-homeostatic feeding in sated mice using associative learning of contextual food cues. Mol Psychiatry 2020; 25:666-679. [PMID: 29875477 PMCID: PMC6281813 DOI: 10.1038/s41380-018-0072-y] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/26/2017] [Revised: 01/31/2018] [Accepted: 03/13/2018] [Indexed: 02/07/2023]
Abstract
Feeding is a complex motivated behavior controlled by a distributed neural network that processes sensory information to generate adaptive behavioral responses. Accordingly, studies using appetitive Pavlovian conditioning confirm that environmental cues that are associated with food availability can induce feeding even in satiated subjects. However, in mice, appetitive conditioning generally requires intensive training and thus can impede molecular studies that often require large numbers of animals. To address this, we developed and validated a simple and rapid context-induced feeding (Ctx-IF) task in which cues associated with food availability can later lead to increased food consumption in sated mice. We show that the associated increase in food consumption is driven by both positive and negative reinforcement and that spaced training is more effective than massed training. Ctx-IF can be completed in ~1 week and provides an opportunity to study the molecular mechanisms and circuitry underlying non-homeostatic eating. We have used this paradigm to map brain regions that are activated during Ctx-IF with cFos immunohistochemistry and found that the insular cortex, and other regions, are activated following exposure to cues denoting the availability of food. Finally, we show that inhibition of the insular cortex using GABA agonists impairs performance of the task. Our findings provide a novel assay in mice for defining the functional neuroanatomy of appetitive conditioning and identify specific brain regions that are activated during the development of learned behaviors that impact food consumption.
Collapse
Affiliation(s)
- Sarah A. Stern
- 0000 0001 2166 1519grid.134907.8Laboratory of Molecular Genetics, Rockefeller University, New York, NY 10065 USA
| | - Katherine R. Doerig
- 0000 0001 2166 1519grid.134907.8Laboratory of Molecular Genetics, Rockefeller University, New York, NY 10065 USA
| | - Estefania P. Azevedo
- 0000 0001 2166 1519grid.134907.8Laboratory of Molecular Genetics, Rockefeller University, New York, NY 10065 USA
| | - Elina Stoffel
- 0000 0001 2166 1519grid.134907.8Laboratory of Molecular Genetics, Rockefeller University, New York, NY 10065 USA
| | - Jeffrey M. Friedman
- 0000 0001 2166 1519grid.134907.8Laboratory of Molecular Genetics, Rockefeller University, New York, NY 10065 USA ,0000 0001 2167 1581grid.413575.1Howard Hughes Medical Institute, Chevy Chase, MD USA
| |
Collapse
|
47
|
Doulberis M, Papaefthymiou A, Polyzos SA, Katsinelos P, Grigoriadis N, Srivastava DS, Kountouras J. Rodent models of obesity. MINERVA ENDOCRINOL 2019; 45:243-263. [PMID: 31738033 DOI: 10.23736/s0391-1977.19.03058-x] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Obese or overweight people exceed one-third of the global population and obesity along with diabetes mellitus consist basic components of metabolic syndrome, both of which are known cardio-cerebrovascular risk factors with detrimental consequences. These data signify the pandemic character of obesity and the necessity for effective treatments. Substantial advances have been accomplished in preclinical research of obesity by using animal models, which mimic the human disease. In particular, rodent models have been widely used for many decades with success for the elucidation of the pathophysiology of obesity, since they share physiological and genetic components with humans and appear advantageous in their husbandry. The most representative rodents include the laboratory mouse and rat. Within this review, we attempted to consolidate the most widely used mice and rat models of obesity and highlight their strengths as well as weaknesses in a critical way. Our aim was to bridge the gap between laboratory facilities and patient's bed and help the researcher find the appropriate animal model for his/her obesity research. This tactful selection of the appropriate model of obesity may offer more translational derived results. In this regard, we included, the main diet induced models, the chemical/mechanical ones, as well as a selection of monogenic or polygenic models.
Collapse
Affiliation(s)
- Michael Doulberis
- Department of Gastroenterology and Hepatology, University of Zurich, Zurich, Switzerland - .,Department of Internal Medicine, Second Medical Clinic, Ippokration Hospital, Aristotle University of Thessaloniki, Thessaloniki, Greece -
| | | | | | - Panagiotis Katsinelos
- Department of Internal Medicine, Second Medical Clinic, Ippokration Hospital, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Nikolaos Grigoriadis
- First Department of Pharmacology, Faculty of Medicine, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - David S Srivastava
- Second Department of Neurology, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Jannis Kountouras
- Department of Internal Medicine, Second Medical Clinic, Ippokration Hospital, Aristotle University of Thessaloniki, Thessaloniki, Greece
| |
Collapse
|
48
|
Gimeno-Ferrer F, Albuquerque D, García Banacloy A, Guzmán Luján C, Vidal Garcia C, Marcaida Benito G, Sánchez Juan C, Bruna Esteban M, Rodríguez-López R. Genetic screening for MC4R gene identifies three novel mutations associated with severe familiar obesity in a cohort of Spanish individuals. Gene 2019; 704:74-79. [PMID: 30981838 DOI: 10.1016/j.gene.2019.04.018] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2019] [Revised: 03/20/2019] [Accepted: 04/05/2019] [Indexed: 11/19/2022]
Abstract
MC4R gene is a hypothalamic satiety control mediator in which mutations cause a monogenic form of obesity. The aim of this study was to perform a genetic screening to identify variations in the entire region of MC4R gene. A total of 236 unrelated and severely obese patients (BMI ≥ 40 kg/m2) with Spanish ancestry and severe overweight familiar history have been enrolled into the study. Seven MC4R gene variants were identified in the heterozygous state in 21 patients. Coding variants p.Thr101Ile and p.Ala259Asp are new and variants p.Ser30Phe, p.Val103Ile and p.Ile251Leu were previously described. Two variants have been also observed in the promoter region of the MC4R gene; the c.-24G>A mutation, described for the first time, and the known c.-178A>C variant. Both in silico and family segregation analysis confirm the correlation between novel identified mutations in MC4R gene and obesity development. The correlation between the four variants (c.-24G>A, p.Thr101Ile, p.Ala259Asp and p.Ser30Phe) and the obesity phenotype, therefore, allows the conclusion that all of the four mutations cause a monogenic form of obesity.
Collapse
Affiliation(s)
- Fátima Gimeno-Ferrer
- Genomics Group, Fundación Investigación Hospital General Universitario de Valencia, Valencia, Spain
| | - David Albuquerque
- Genomics Group, Fundación Investigación Hospital General Universitario de Valencia, Valencia, Spain; Research Center for Anthropology and Health (CIAS), Department of Life Sciences, University of Coimbra, Coimbra, Portugal
| | - Amor García Banacloy
- Genomics Group, Fundación Investigación Hospital General Universitario de Valencia, Valencia, Spain; Department of Surgery, University of Valencia, Valencia, Spain
| | - Carola Guzmán Luján
- Genomics Group, Fundación Investigación Hospital General Universitario de Valencia, Valencia, Spain; Laboratory of Molecular Genetics, Clinical Analysis Service, Consorcio Hospital General Universitario de Valencia, Valencia, Spain
| | - Clara Vidal Garcia
- Genomics Group, Fundación Investigación Hospital General Universitario de Valencia, Valencia, Spain
| | - Goitzane Marcaida Benito
- Genomics Group, Fundación Investigación Hospital General Universitario de Valencia, Valencia, Spain; Laboratory of Molecular Genetics, Clinical Analysis Service, Consorcio Hospital General Universitario de Valencia, Valencia, Spain
| | - Carlos Sánchez Juan
- Endocrinology and Nutrition Unit, Consorcio Hospital General Universitario de Valencia, Valencia, Spain
| | - Marcos Bruna Esteban
- General and Digestive Surgery Unit, Consorcio Hospital General Universitario de Valencia, Valencia, Spain
| | - Raquel Rodríguez-López
- Genomics Group, Fundación Investigación Hospital General Universitario de Valencia, Valencia, Spain; Laboratory of Molecular Genetics, Clinical Analysis Service, Consorcio Hospital General Universitario de Valencia, Valencia, Spain.
| |
Collapse
|
49
|
Adamska-Patruno E, Goscik J, Czajkowski P, Maliszewska K, Ciborowski M, Golonko A, Wawrusiewicz-Kurylonek N, Citko A, Waszczeniuk M, Kretowski A, Gorska M. The MC4R genetic variants are associated with lower visceral fat accumulation and higher postprandial relative increase in carbohydrate utilization in humans. Eur J Nutr 2019; 58:2929-2941. [PMID: 30945034 PMCID: PMC6768895 DOI: 10.1007/s00394-019-01955-0] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2018] [Accepted: 03/24/2019] [Indexed: 12/14/2022]
Abstract
Purpose The interactions between lifestyle and genetic factors play an important role in obesity development. Mutations in melanocortin-4-receptor (MC4R) gene are one of the most common cause of monogenic obesity, however, the functional effects of polymorphic variants near MC4R gene in general populations remain uncertain. The aim of our study was to analyze whether the common single nucleotide polymorphisms (SNPs) of MC4R gene influence the food preferences, physical activity, body fat content and distribution, as well as fasting and postprandial energy expenditure and substrates utilization. Methods We genotyped previously identified MC4R SNPs: rs17782313, rs633265, rs1350341, rs12970134 in 927 subjects, who underwent anthropometric, total body fat content, visceral (VAT) and subcutaneous adipose tissue (SAT) measurements, and daily physical activity and dietary intake analysis. In randomly selected 47 subjects the energy expenditure, carbohydrate and lipid utilizations were evaluated in fasting state and after high-carbohydrate and control meals intake. Results We found the significant associations between studied SNPs of MC4R gene and VAT and VAT/SAT ratio. Moreover, the GG genotype carriers of rs1350341, who had the lowest VAT accumulation (p = 0.012), presented higher relative increase in postprandial carbohydrate utilization (p = 0.013, p = 0.024). Conclusions We have observed that common SNPs of the MC4R gene influence the body fat content and distribution, as well as relative increase in postprandial carbohydrate utilization. We believe that our study may help to understand better the impact of MC4R gene on obesity development, and to help to provide personalized prevention/treatment strategies to fight against obesity and its metabolic consequences.
Collapse
Affiliation(s)
- Edyta Adamska-Patruno
- Clinical Research Centre, Medical University of Bialystok, M.C. Sklodowskiej-Curie 24A, 15-276, Bialystok, Poland.
| | - Joanna Goscik
- Centre for Experimental Medicine, Medical University of Bialystok, M.C. Sklodowskiej-Curie 24A, 15-276, Bialystok, Poland
| | - Przemyslaw Czajkowski
- Clinical Research Centre, Medical University of Bialystok, M.C. Sklodowskiej-Curie 24A, 15-276, Bialystok, Poland
| | - Katarzyna Maliszewska
- Department of Endocrinology, Diabetology and Internal Medicine, Medical University of Bialystok, M.C. Sklodowskiej-Curie 24A, 15-276, Bialystok, Poland
| | - Michał Ciborowski
- Clinical Research Centre, Medical University of Bialystok, M.C. Sklodowskiej-Curie 24A, 15-276, Bialystok, Poland
| | - Anna Golonko
- Department of Dietetics and Clinical Nutrition, Medical University of Bialystok, Mieszka I-go 4B, 15-054, Bialystok, Poland
| | - Natalia Wawrusiewicz-Kurylonek
- Department of Endocrinology, Diabetology and Internal Medicine, Medical University of Bialystok, M.C. Sklodowskiej-Curie 24A, 15-276, Bialystok, Poland
| | - Anna Citko
- Clinical Research Centre, Medical University of Bialystok, M.C. Sklodowskiej-Curie 24A, 15-276, Bialystok, Poland
| | - Magdalena Waszczeniuk
- Clinical Research Centre, Medical University of Bialystok, M.C. Sklodowskiej-Curie 24A, 15-276, Bialystok, Poland.,Department of Endocrinology, Diabetology and Internal Medicine, Medical University of Bialystok, M.C. Sklodowskiej-Curie 24A, 15-276, Bialystok, Poland
| | - Adam Kretowski
- Clinical Research Centre, Medical University of Bialystok, M.C. Sklodowskiej-Curie 24A, 15-276, Bialystok, Poland.,Department of Endocrinology, Diabetology and Internal Medicine, Medical University of Bialystok, M.C. Sklodowskiej-Curie 24A, 15-276, Bialystok, Poland
| | - Maria Gorska
- Department of Endocrinology, Diabetology and Internal Medicine, Medical University of Bialystok, M.C. Sklodowskiej-Curie 24A, 15-276, Bialystok, Poland
| |
Collapse
|
50
|
TrkB-expressing neurons in the dorsomedial hypothalamus are necessary and sufficient to suppress homeostatic feeding. Proc Natl Acad Sci U S A 2019; 116:3256-3261. [PMID: 30718415 DOI: 10.1073/pnas.1815744116] [Citation(s) in RCA: 50] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Genetic evidence indicates that brain-derived neurotrophic factor (BDNF) signaling through the TrkB receptor plays a critical role in the control of energy balance. Mutations in the BDNF or the TrkB-encoding NTRK2 gene have been found to cause severe obesity in humans and mice. However, it remains unknown which brain neurons express TrkB to control body weight. Here, we report that TrkB-expressing neurons in the dorsomedial hypothalamus (DMH) regulate food intake. We found that the DMH contains both glutamatergic and GABAergic TrkB-expressing neurons, some of which also express the leptin receptor (LepR). As revealed by Fos immunohistochemistry, a significant number of TrkB-expressing DMH (DMHTrkB) neurons were activated upon either overnight fasting or after refeeding. Chemogenetic activation of DMHTrkB neurons strongly suppressed feeding in the dark cycle when mice are physiologically hungry, whereas chemogenetic inhibition of DMHTrkB neurons greatly promoted feeding in the light cycle when mice are physiologically satiated, without affecting feeding in the dark cycle. Neuronal tracing revealed that DMHTrkB neurons do not innervate neurons expressing agouti-related protein in the arcuate nucleus, indicating that DMHTrkB neurons are distinct from previously identified LepR-expressing GABAergic DMH neurons that suppress feeding. Furthermore, selective Ntrk2 deletion in the DMH of adult mice led to hyperphagia, reduced energy expenditure, and obesity. Thus, our data show that DMHTrkB neurons are a population of neurons that are necessary and sufficient to suppress appetite and maintain physiological satiation. Pharmacological activation of these neurons could be a therapeutic intervention for the treatment of obesity.
Collapse
|