1
|
Melnik BC, Weiskirchen R, Stremmel W, John SM, Schmitz G. Risk of Fat Mass- and Obesity-Associated Gene-Dependent Obesogenic Programming by Formula Feeding Compared to Breastfeeding. Nutrients 2024; 16:2451. [PMID: 39125332 PMCID: PMC11314333 DOI: 10.3390/nu16152451] [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] [Received: 06/18/2024] [Revised: 07/21/2024] [Accepted: 07/25/2024] [Indexed: 08/12/2024] Open
Abstract
It is the purpose of this review to compare differences in postnatal epigenetic programming at the level of DNA and RNA methylation and later obesity risk between infants receiving artificial formula feeding (FF) in contrast to natural breastfeeding (BF). FF bears the risk of aberrant epigenetic programming at the level of DNA methylation and enhances the expression of the RNA demethylase fat mass- and obesity-associated gene (FTO), pointing to further deviations in the RNA methylome. Based on a literature search through Web of Science, Google Scholar, and PubMed databases concerning the dietary and epigenetic factors influencing FTO gene and FTO protein expression and FTO activity, FTO's impact on postnatal adipogenic programming was investigated. Accumulated translational evidence underscores that total protein intake as well as tryptophan, kynurenine, branched-chain amino acids, milk exosomal miRNAs, NADP, and NADPH are crucial regulators modifying FTO gene expression and FTO activity. Increased FTO-mTORC1-S6K1 signaling may epigenetically suppress the WNT/β-catenin pathway, enhancing adipocyte precursor cell proliferation and adipogenesis. Formula-induced FTO-dependent alterations of the N6-methyladenosine (m6A) RNA methylome may represent novel unfavorable molecular events in the postnatal development of adipogenesis and obesity, necessitating further investigations. BF provides physiological epigenetic DNA and RNA regulation, a compelling reason to rely on BF.
Collapse
Affiliation(s)
- Bodo C. Melnik
- Department of Dermatology, Environmental Medicine and Health Theory, University of Osnabrück, D-49076 Osnabrück, Germany
| | - Ralf Weiskirchen
- Institute of Molecular Pathobiochemistry, Experimental Gene Therapy and Clinical Chemistry (IFMPEGKC), RWTH University Hospital Aachen, D-52074 Aachen, Germany;
| | - Wolfgang Stremmel
- Praxis for Internal Medicine, Beethovenstrasse 2, D-76530 Baden-Baden, Germany;
| | - Swen Malte John
- Department of Dermatology, Environmental Medicine and Health Theory, University of Osnabrück, D-49076 Osnabrück, Germany
- Institute for Interdisciplinary Dermatological Prevention and Rehabilitation (iDerm), University of Osnabrück, D-49076 Osnabrück, Germany;
| | - Gerd Schmitz
- Institute for Clinical Chemistry and Laboratory Medicine, University Hospital of Regensburg, D-93053 Regensburg, Germany;
| |
Collapse
|
2
|
Zhang L, Liu J, Gao D, Li D. Role of ghrelin in promoting catch-up growth and maintaining metabolic homeostasis in small-for-gestational-age infants. Front Pediatr 2024; 12:1395571. [PMID: 38903769 PMCID: PMC11187245 DOI: 10.3389/fped.2024.1395571] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/04/2024] [Accepted: 05/27/2024] [Indexed: 06/22/2024] Open
Abstract
Small-for-gestational age (SGA) has been a great concern in the perinatal period as it leads to adverse perinatal outcomes and increased neonatal morbidity and mortality, has an impact on long-term health outcomes, and increases the risk of metabolic disorders, cardiovascular, and endocrine diseases in adulthood. As an endogenous ligand of the growth hormone secretagotor (GHS-R), ghrelin may play an important role in regulating growth and energy metabolic homeostasis from fetal to adult life. We reviewed the role of ghrelin in catch-up growth and energy metabolism of SGA in recent years. In addition to promoting SGA catch-up growth, ghrelin may also participate in SGA energy metabolism and maintain metabolic homeostasis. The causes of small gestational age infants are very complex and may be related to a variety of metabolic pathway disorders. The related signaling pathways regulated by ghrelin may help to identify high-risk groups of SGA metabolic disorders and formulate targeted interventions to prevent the occurrence of adult dwarfism, insulin resistance-related metabolic syndrome and other diseases.
Collapse
Affiliation(s)
- Li Zhang
- Department of Pediatrics, The Second Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Jingfei Liu
- Department of Neonatology, Dalian Women and Children’s Medical Group, Dalian, China
| | - Dianyong Gao
- Department of Orthopedics, Lushunkou District People’s Hospital, Dalian, China
| | - Dong Li
- Department of Neonatology, The First Affiliated Hospital of Dalian Medical University, Dalian, China
| |
Collapse
|
3
|
Mora-Maltas B, Baenas I, Etxandi M, Lucas I, Granero R, Fernández-Aranda F, Tovar S, Solé-Morata N, Gómez-Peña M, Moragas L, Del Pino-Gutiérrez A, Tapia J, Diéguez C, Goudriaan AE, Jiménez-Murcia S. Association between endocrine and neuropsychological endophenotypes and gambling disorder severity. Addict Behav 2024; 153:107968. [PMID: 38447412 DOI: 10.1016/j.addbeh.2024.107968] [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] [Received: 09/25/2023] [Revised: 01/07/2024] [Accepted: 01/22/2024] [Indexed: 03/08/2024]
Abstract
BACKGROUND Neurobiological characteristics have been identified regarding the severity of gambling disorder (GD). The aims of this study were: (1) to examine, through a path analysis, whether there was a relationship between neuroendocrine features, potentially mediational GD variables, and GD severity, and (2) to associate neuroendocrine variables, with GD severity-related variables according to gambling preferences. METHODS The sample included 297 outpatients with GD. We analyzed endocrine concentrations of different appetite-related hormones (ghrelin, liver antimicrobial peptide 2 [LEAP-2], leptin, adiponectin), and neuropsychological performance (working memory, cognitive flexibility, inhibition, decision making, premorbid intelligence). Path analysis assessed mechanisms between neuroendocrine features and GD severity, including mediational GD variables (impulsivity traits and gambling-related cognitive distortions). Partial correlations evaluated the associations between neuroendocrine variables, including impulsivity traits, and variables related to GD severity (DSM-5, South Oaks Gambling Screen, illness duration, and gambling-related cognitive distortions). RESULTS Lower adiponectin concentrations predicted greater GD severity, while higher LEAP-2 concentrations predicted more gambling-related cognitive distortions. Likewise, better neuropsychological performance directly predicted GD severity, but worse neuropsychological performance was associated with GD severity through the mediational variables of impulsivity traits and gambling-related cognitive distortions. Also, in non-strategic individuals with GD, poor working memory was associated with gambling expectancies and predictive control. In strategic individuals with GD, poor cognitive flexibility was associated with illusion of control, predictive control, and inability to stop gambling. CONCLUSIONS These results provide updated information about the comprehension of the interaction between neuroendocrine features, clinical variables, and severity of GD. Thus, neurobiological functions seem to be strongly related to GD severity.
Collapse
Affiliation(s)
- Bernat Mora-Maltas
- Clinical Psychology Department, Bellvitge University Hospital- Bellvitge Biomedical Research Institute (IDIBELL), Barcelona, Spain; Ciber Physiopathology of Obesity and Nutrition (CIBERObn), Instituto de Salud Carlos III, Barcelona, Spain; Doctorate in Medicine and Traslational Research Programme, University of Barcelona (UB), Barcelona, Spain
| | - Isabel Baenas
- Clinical Psychology Department, Bellvitge University Hospital- Bellvitge Biomedical Research Institute (IDIBELL), Barcelona, Spain; Ciber Physiopathology of Obesity and Nutrition (CIBERObn), Instituto de Salud Carlos III, Barcelona, Spain; Doctorate in Medicine and Traslational Research Programme, University of Barcelona (UB), Barcelona, Spain; Psychoneurobiology of Eating and Addictive Behaviors Group, Neurosciences Programme, Bellvitge Biomedical Research Institute (IDIBELL), Barcelona, Spain
| | - Mikel Etxandi
- Clinical Psychology Department, Bellvitge University Hospital- Bellvitge Biomedical Research Institute (IDIBELL), Barcelona, Spain; Doctorate in Medicine and Traslational Research Programme, University of Barcelona (UB), Barcelona, Spain; Department of Psychiatry, Hospital Universitari Germans Trias i Pujol, IGTP Campus Can Ruti, Badalona, Spain
| | - Ignacio Lucas
- Clinical Psychology Department, Bellvitge University Hospital- Bellvitge Biomedical Research Institute (IDIBELL), Barcelona, Spain; Ciber Physiopathology of Obesity and Nutrition (CIBERObn), Instituto de Salud Carlos III, Barcelona, Spain; Psychoneurobiology of Eating and Addictive Behaviors Group, Neurosciences Programme, Bellvitge Biomedical Research Institute (IDIBELL), Barcelona, Spain
| | - Roser Granero
- Ciber Physiopathology of Obesity and Nutrition (CIBERObn), Instituto de Salud Carlos III, Barcelona, Spain; Psychoneurobiology of Eating and Addictive Behaviors Group, Neurosciences Programme, Bellvitge Biomedical Research Institute (IDIBELL), Barcelona, Spain; Department of Psychobiology and Methodology, Autonomous University of Barcelona, Barcelona, Spain
| | - Fernando Fernández-Aranda
- Clinical Psychology Department, Bellvitge University Hospital- Bellvitge Biomedical Research Institute (IDIBELL), Barcelona, Spain; Ciber Physiopathology of Obesity and Nutrition (CIBERObn), Instituto de Salud Carlos III, Barcelona, Spain; Psychoneurobiology of Eating and Addictive Behaviors Group, Neurosciences Programme, Bellvitge Biomedical Research Institute (IDIBELL), Barcelona, Spain; Department of Clinical Sciences, School of Medicine and Health Sciences, University of Barcelona, Barcelona, Spain
| | - Sulay Tovar
- Ciber Physiopathology of Obesity and Nutrition (CIBERObn), Instituto de Salud Carlos III, Barcelona, Spain; Department of Physiology, CIMUS, University of Santiago de Compostela, Instituto de Investigación Sanitaria, Santiago de Compostela, Spain
| | - Neus Solé-Morata
- Clinical Psychology Department, Bellvitge University Hospital- Bellvitge Biomedical Research Institute (IDIBELL), Barcelona, Spain
| | - Mónica Gómez-Peña
- Clinical Psychology Department, Bellvitge University Hospital- Bellvitge Biomedical Research Institute (IDIBELL), Barcelona, Spain; Psychoneurobiology of Eating and Addictive Behaviors Group, Neurosciences Programme, Bellvitge Biomedical Research Institute (IDIBELL), Barcelona, Spain
| | - Laura Moragas
- Clinical Psychology Department, Bellvitge University Hospital- Bellvitge Biomedical Research Institute (IDIBELL), Barcelona, Spain; Psychoneurobiology of Eating and Addictive Behaviors Group, Neurosciences Programme, Bellvitge Biomedical Research Institute (IDIBELL), Barcelona, Spain
| | - Amparo Del Pino-Gutiérrez
- Ciber Physiopathology of Obesity and Nutrition (CIBERObn), Instituto de Salud Carlos III, Barcelona, Spain; Psychoneurobiology of Eating and Addictive Behaviors Group, Neurosciences Programme, Bellvitge Biomedical Research Institute (IDIBELL), Barcelona, Spain; Department of Public Health, Mental Health and Perinatal Nursing, School of Nursing, University of Barcelona, Barcelona, Spain
| | - Javier Tapia
- Doctorate in Medicine and Traslational Research Programme, University of Barcelona (UB), Barcelona, Spain; Psychoneurobiology of Eating and Addictive Behaviors Group, Neurosciences Programme, Bellvitge Biomedical Research Institute (IDIBELL), Barcelona, Spain; Medical Direction of Ambulatory Processes, South Metropolitan Territorial Management, Bellvitge University Hospital, Barcelona, Spain
| | - Carlos Diéguez
- Ciber Physiopathology of Obesity and Nutrition (CIBERObn), Instituto de Salud Carlos III, Barcelona, Spain; Department of Physiology, CIMUS, University of Santiago de Compostela, Instituto de Investigación Sanitaria, Santiago de Compostela, Spain
| | - Anna E Goudriaan
- Arkin Mental Health Care, Jellinek, Amsterdam Institute for Addiction Research, Amsterdam, the Netherlands; Amsterdam UMC, Department of Psychiatry, University of Amsterdam, Amsterdam, the Netherlands; Amsterdam Public Health Research Institute, Amsterdam, the Netherlands
| | - Susana Jiménez-Murcia
- Clinical Psychology Department, Bellvitge University Hospital- Bellvitge Biomedical Research Institute (IDIBELL), Barcelona, Spain; Ciber Physiopathology of Obesity and Nutrition (CIBERObn), Instituto de Salud Carlos III, Barcelona, Spain; Psychoneurobiology of Eating and Addictive Behaviors Group, Neurosciences Programme, Bellvitge Biomedical Research Institute (IDIBELL), Barcelona, Spain; Department of Clinical Sciences, School of Medicine and Health Sciences, University of Barcelona, Barcelona, Spain; Centre for Psychological Services, University of Barcelona, Spain.
| |
Collapse
|
4
|
Mourino N, Zhang Z, Pérez-Ríos M, Yolton K, Lanphear BP, Chen A, Buckley JP, Kalkwarf HJ, Cecil KM, Braun JM. Early life exposure to secondhand tobacco smoke and eating behaviors at age 12 years. Environ Health 2024; 23:37. [PMID: 38609912 PMCID: PMC11015554 DOI: 10.1186/s12940-024-01076-0] [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: 02/06/2024] [Accepted: 03/25/2024] [Indexed: 04/14/2024]
Abstract
BACKGROUND Prenatal or early childhood secondhand tobacco smoke (SHS) exposure increases obesity risk. However, the potential mechanisms underlying this association are unclear, but obesogenic eating behaviors are one pathway that components of SHS could perturb. Our aim was to assess associations of prenatal and early childhood SHS exposure with adolescent eating behaviors. METHODS Data came from a prospective pregnancy and birth cohort (N = 207, Cincinnati, OH). With multiple informant models, we estimated associations of prenatal (mean of 16 and 26 weeks of gestation maternal serum cotinine concentrations) and early childhood cotinine (average concentration across ages 12, 24, 36, and 48 months) with eating behaviors at age 12 years (Child Eating Behaviors Questionnaire). We tested whether associations differed by exposure periods and adolescent's sex. Models adjusted for maternal and child covariates. RESULTS We found no statistically significant associations between cotinine measures and adolescent's eating behaviors. Yet, in females, prenatal cotinine was associated with greater food responsiveness (β: 0.23; 95% CI: 0.08, 0.38) and lower satiety responsiveness (β: -0.14; 95% CI: -0.26, -0.02); in males, prenatal and postnatal cotinine was related to lower food responsiveness (prenatal: β: -0.25; 95% CI: -0.04, -0.06; postnatal: β: -0.36; 95% CI: -0.06, -0.11). No significant effect modification by sex or exposure window was found for other eating behaviors. CONCLUSION Prenatal and early childhood SHS exposures were not related to adolescent's eating behavior in this cohort; however, biological sex may modify these associations.
Collapse
Affiliation(s)
- Nerea Mourino
- Department of Preventive Medicine and Public Health, University of Santiago de Compostela, Santiago de Compostela, Spain
| | - Zhuoya Zhang
- Department of Epidemiology, Dartmouth College, Hanover, NH, USA.
| | - Mónica Pérez-Ríos
- Department of Preventive Medicine and Public Health, University of Santiago de Compostela, Santiago de Compostela, Spain
- Consortium for Biomedical Research in Epidemiology and Public Health (CIBERESP), Madrid, Spain
- Health Research Institute of Santiago de Compostela (IDIS), Santiago de Compostela, Spain
| | - Kimberly Yolton
- Department of Pediatrics, Cincinnati Children's Hospital Medical Center, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Bruce P Lanphear
- Faculty of Health Sciences, Simon Fraser University, Vancouver, BC, Canada
| | - Aimin Chen
- Department of Biostatistics, Epidemiology and Informatics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Jessie P Buckley
- Department of Epidemiology, University of North Carolina at Chapel Hill Gillings School of Global Public Health, Chapel Hill, NC, USA
| | - Heidi J Kalkwarf
- Department of Pediatrics, Cincinnati Children's Hospital Medical Center, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Kim M Cecil
- Department of Pediatrics, Cincinnati Children's Hospital Medical Center, University of Cincinnati College of Medicine, Cincinnati, OH, USA
- Department of Radiology, Cincinnati Children's Hospital Medical Center, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Joseph M Braun
- Department of Epidemiology, Brown University, Providence, RI, USA
| |
Collapse
|
5
|
Saito H, Watanabe H, Ono M. Synthesis and biological evaluation of novel 18F-labeled 2,4-diaminopyrimidine derivatives for detection of ghrelin receptor in the brain. Bioorg Med Chem Lett 2024; 99:129625. [PMID: 38253227 DOI: 10.1016/j.bmcl.2024.129625] [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] [Received: 11/06/2023] [Revised: 12/25/2023] [Accepted: 01/19/2024] [Indexed: 01/24/2024]
Abstract
The ghrelin receptor (GHSR) is known to regulate various physiological processes including appetite, food intake, and growth hormone release. Its expression is mainly observed in the brain, pancreas, stomach, and intestine. However, the functions of the receptor have not been fully elucidated. GHSR imaging with positron emission tomography (PET) is expected to further understanding of the functions and pathologies of the receptor. In this study, we newly designed and synthesized diaminopyrimidine derivatives ([18F]BPP-1 and [18F]BPP-2) and evaluated their utility as novel PET probes targeting GHSR. In in vitro competitive binding assays, the binding affinity of BPP-2 for GHSR (Ki = 274 nM) was comparable to that of the diaminopyimidine lead compound Abb8a (Ki = 109 nM). In a biodistribution study using normal mice, [18F]BPP-2 displayed low uptake in the brain and moderate uptake in the pancreas, but high radioactivity accumulation in bone was observed due to its defluorination in vivo. Taken together, although further improvement of the pharmacokinetics is needed, the diaminopyrimidine scaffold has potential for the development of useful GHSR-targeting PET probes.
Collapse
Affiliation(s)
- Haruka Saito
- Department of Patho-Functional Bioanalysis, Graduate School of Pharmaceutical Sciences, Kyoto University, 46-29 Yoshida Shimoadachi-cho, Sakyo-ku, Kyoto 606-8501, Japan
| | - Hiroyuki Watanabe
- Department of Patho-Functional Bioanalysis, Graduate School of Pharmaceutical Sciences, Kyoto University, 46-29 Yoshida Shimoadachi-cho, Sakyo-ku, Kyoto 606-8501, Japan.
| | - Masahiro Ono
- Department of Patho-Functional Bioanalysis, Graduate School of Pharmaceutical Sciences, Kyoto University, 46-29 Yoshida Shimoadachi-cho, Sakyo-ku, Kyoto 606-8501, Japan.
| |
Collapse
|
6
|
Baenas I, Mora-Maltas B, Etxandi M, Lucas I, Granero R, Fernández-Aranda F, Tovar S, Solé-Morata N, Gómez-Peña M, Moragas L, Del Pino-Gutiérrez A, Tapia J, Diéguez C, Goudriaan AE, Jiménez-Murcia S. Cluster analysis in gambling disorder based on sociodemographic, neuropsychological, and neuroendocrine features regulating energy homeostasis. Compr Psychiatry 2024; 128:152435. [PMID: 37976998 DOI: 10.1016/j.comppsych.2023.152435] [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: 07/26/2023] [Revised: 10/12/2023] [Accepted: 10/26/2023] [Indexed: 11/19/2023] Open
Abstract
BACKGROUND The heterogeneity of gambling disorder (GD) has led to the identification of different subtypes, mostly including phenotypic features, with distinctive implications on the GD severity and treatment outcome. However, clustering analyses based on potential endophenotypic features, such as neuropsychological and neuroendocrine factors, are scarce so far. AIMS This study firstly aimed to identify empirical clusters in individuals with GD based on sociodemographic (i.e., age and sex), neuropsychological (i.e., cognitive flexibility, inhibitory control, decision making, working memory, attention, and set-shifting), and neuroendocrine factors regulating energy homeostasis (i.e., leptin, ghrelin, adiponectin, and liver-expressed antimicrobial peptide 2, LEAP-2). The second objective was to compare the profiles between clusters, considering the variables used for the clustering procedure and other different sociodemographic, clinical, and psychological features. METHODS 297 seeking-treatment adult outpatients with GD (93.6% males, mean age of 39.58 years old) were evaluated through a semi-structured clinical interview, self-reported psychometric assessments, and a protocolized neuropsychological battery. Plasma concentrations of neuroendocrine factors were assessed in peripheral blood after an overnight fast. Agglomerative hierarchical clustering was applied using sociodemographic, neuropsychological, and neuroendocrine variables as indicators for the grouping procedure. Comparisons between the empirical groups were performed using Chi-square tests (χ2) for categorical variables, and analysis of variance (ANOVA) for quantitative measures. RESULTS Three-mutually-exclusive groups were obtained, being neuropsychological features those with the greatest weight in differentiating groups. The largest cluster (Cluster 1, 65.3%) was composed by younger males with strategic and online gambling preferences, scoring higher on self-reported impulsivity traits, but with a lower cognitive impairment. Cluster 2 (18.2%) and 3 (16.5%) were characterized by a significantly higher proportion of females and older patients with non-strategic gambling preferences and a worse neuropsychological performance. Particularly, Cluster 3 had the poorest neuropsychological performance, especially in cognitive flexibility, while Cluster 2 reported the poorest inhibitory control. This latter cluster was also distinguished by a poorer self-reported emotion regulation, the highest prevalence of food addiction, as well as a metabolic profile characterized by the highest mean concentrations of leptin, adiponectin, and LEAP-2. CONCLUSIONS To the best of our knowledge, this is the first study to identify well-differentiated GD clusters using neuropsychological and neuroendocrine features. Our findings reinforce the heterogeneous nature of the disorder and emphasize a role of potential endophenotypic features in GD subtyping. This more comprehensive characterization of GD profiles could contribute to optimize therapeutic interventions based on a medicine of precision.
Collapse
Affiliation(s)
- Isabel Baenas
- Clinical Psychology Department, Bellvitge University Hospital, Barcelona, Spain; Psychoneurobiology of Eating and Addictive Behaviors Group, Neurosciences Program, Bellvitge Biomedical Research Institute (IDIBELL), Barcelona, Spain; Ciber Physiopathology of Obesity and Nutrition (CIBERObn), Instituto de Salud Carlos III, Barcelona, Spain; Doctoral Program in Medicine and Translational Research, University of Barcelona (UB), Barcelona, Spain
| | - Bernat Mora-Maltas
- Clinical Psychology Department, Bellvitge University Hospital, Barcelona, Spain; Ciber Physiopathology of Obesity and Nutrition (CIBERObn), Instituto de Salud Carlos III, Barcelona, Spain; Doctoral Program in Medicine and Translational Research, University of Barcelona (UB), Barcelona, Spain
| | - Mikel Etxandi
- Doctoral Program in Medicine and Translational Research, University of Barcelona (UB), Barcelona, Spain; Department of Psychiatry, Hospital Universitari Germans Trias i Pujol, IGTP Campus Can Ruti, Badalona, Spain
| | - Ignacio Lucas
- Clinical Psychology Department, Bellvitge University Hospital, Barcelona, Spain; Psychoneurobiology of Eating and Addictive Behaviors Group, Neurosciences Program, Bellvitge Biomedical Research Institute (IDIBELL), Barcelona, Spain; Ciber Physiopathology of Obesity and Nutrition (CIBERObn), Instituto de Salud Carlos III, Barcelona, Spain
| | - Roser Granero
- Psychoneurobiology of Eating and Addictive Behaviors Group, Neurosciences Program, Bellvitge Biomedical Research Institute (IDIBELL), Barcelona, Spain; Ciber Physiopathology of Obesity and Nutrition (CIBERObn), Instituto de Salud Carlos III, Barcelona, Spain; Department of Psychobiology and Methodology, Autonomous University of Barcelona, Barcelona, Spain
| | - Fernando Fernández-Aranda
- Clinical Psychology Department, Bellvitge University Hospital, Barcelona, Spain; Psychoneurobiology of Eating and Addictive Behaviors Group, Neurosciences Program, Bellvitge Biomedical Research Institute (IDIBELL), Barcelona, Spain; Ciber Physiopathology of Obesity and Nutrition (CIBERObn), Instituto de Salud Carlos III, Barcelona, Spain; Department of Clinical Sciences, School of Medicine and Health Sciences, University of Barcelona, Barcelona, Spain
| | - Sulay Tovar
- Ciber Physiopathology of Obesity and Nutrition (CIBERObn), Instituto de Salud Carlos III, Barcelona, Spain; Department of Physiology, CIMUS, University of Santiago de Compostela, Instituto de Investigación Sanitaria (IDIS), Santiago de Compostela, Spain
| | - Neus Solé-Morata
- Clinical Psychology Department, Bellvitge University Hospital, Barcelona, Spain
| | - Mónica Gómez-Peña
- Clinical Psychology Department, Bellvitge University Hospital, Barcelona, Spain; Psychoneurobiology of Eating and Addictive Behaviors Group, Neurosciences Program, Bellvitge Biomedical Research Institute (IDIBELL), Barcelona, Spain
| | - Laura Moragas
- Clinical Psychology Department, Bellvitge University Hospital, Barcelona, Spain; Psychoneurobiology of Eating and Addictive Behaviors Group, Neurosciences Program, Bellvitge Biomedical Research Institute (IDIBELL), Barcelona, Spain
| | - Amparo Del Pino-Gutiérrez
- Psychoneurobiology of Eating and Addictive Behaviors Group, Neurosciences Program, Bellvitge Biomedical Research Institute (IDIBELL), Barcelona, Spain; Ciber Physiopathology of Obesity and Nutrition (CIBERObn), Instituto de Salud Carlos III, Barcelona, Spain; Department of Public Health, Mental Health and Perinatal Nursing, School of Nursing, University of Barcelona, Barcelona, Spain
| | - Javier Tapia
- Psychoneurobiology of Eating and Addictive Behaviors Group, Neurosciences Program, Bellvitge Biomedical Research Institute (IDIBELL), Barcelona, Spain; Doctoral Program in Medicine and Translational Research, University of Barcelona (UB), Barcelona, Spain; Medical Direction of Ambulatory Processes, South Metropolitan Territorial Management, Bellvitge University Hospital, Barcelona, Spain
| | - Carlos Diéguez
- Ciber Physiopathology of Obesity and Nutrition (CIBERObn), Instituto de Salud Carlos III, Barcelona, Spain; Department of Physiology, CIMUS, University of Santiago de Compostela, Instituto de Investigación Sanitaria (IDIS), Santiago de Compostela, Spain
| | - Anna E Goudriaan
- Arkin Mental Health Care, Jellinek, Amsterdam Institute for Addiction Research, Amsterdam, The Netherlands; Amsterdam UMC, Department of Psychiatry, University of Amsterdam, Amsterdam, The Netherlands; Amsterdam Public Health Research Institute, Amsterdam, The Netherlands
| | - Susana Jiménez-Murcia
- Clinical Psychology Department, Bellvitge University Hospital, Barcelona, Spain; Psychoneurobiology of Eating and Addictive Behaviors Group, Neurosciences Program, Bellvitge Biomedical Research Institute (IDIBELL), Barcelona, Spain; Ciber Physiopathology of Obesity and Nutrition (CIBERObn), Instituto de Salud Carlos III, Barcelona, Spain; Department of Clinical Sciences, School of Medicine and Health Sciences, University of Barcelona, Barcelona, Spain.
| |
Collapse
|
7
|
Knowles LG, Armanious AJ, Peng Y, Welsh WJ, James MH. Recent advances in drug discovery efforts targeting the sigma 1 receptor system: Implications for novel medications designed to reduce excessive drug and food seeking. ADDICTION NEUROSCIENCE 2023; 8:100126. [PMID: 37753198 PMCID: PMC10519676 DOI: 10.1016/j.addicn.2023.100126] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/28/2023]
Abstract
Psychiatric disorders characterized by uncontrolled reward seeking, such as substance use disorders (SUDs), alcohol use disorder (AUD) and some eating disorders, impose a significant burden on individuals and society. Despite their high prevalence and substantial morbidity and mortality rates, treatment options for these disorders remain limited. Over the past two decades, there has been a gradual accumulation of evidence pointing to the sigma-1 receptor (S1R) system as a promising target for therapeutic interventions designed to treat these disorders. S1R is a chaperone protein that resides in the endoplasmic reticulum, but under certain conditions translocates to the plasma membrane. In the brain, S1Rs are expressed in several regions important for reward, and following translocation, they physically associate with several reward-related GPCRs, including dopamine receptors 1 and 2 (D1R and D2R). Psychostimulants, alcohol, as well as palatable foods, all alter expression of S1R in regions important for motivated behavior, and S1R antagonists generally decrease behavioral responses to these rewards. Recent advances in structural modeling have permitted the development of highly-selective S1R antagonists with favorable pharmacokinetic profiles, thus providing a therapeutic avenue for S1R-based medications. Here, we provide an up-to-date overview of work linking S1R with motivated behavior for drugs of abuse and food, as well as evidence supporting the clinical utility of S1R antagonists to reduce their excessive consumption. We also highlight potential challenges associated with targeting the S1R system, including the need for a more comprehensive understanding of the underlying neurobiology and careful consideration of the pharmacological properties of S1R-based drugs.
Collapse
Affiliation(s)
- Liam G. Knowles
- Harpur School of Arts and Sciences, Binghamton University, Vestal, NY, USA
| | - Abanoub J. Armanious
- Department of Psychiatry, Robert Wood Johnson Medical School, Rutgers University and Rutgers Biomedical Health Sciences, Piscataway, NJ, USA
- Brain Health Institute, Rutgers University and Rutgers Biomedical and Health Sciences, Piscataway, NJ, USA
| | - Youyi Peng
- Cancer Institute of New Jersey, Rutgers University, New Brunswick, NJ, USA
| | - William J. Welsh
- Department of Pharmacology, Robert Wood Johnson Medical School, Rutgers University and Rutgers Biomedical Health Sciences, Piscataway, NJ, USA
| | - Morgan H. James
- Department of Psychiatry, Robert Wood Johnson Medical School, Rutgers University and Rutgers Biomedical Health Sciences, Piscataway, NJ, USA
- Brain Health Institute, Rutgers University and Rutgers Biomedical and Health Sciences, Piscataway, NJ, USA
| |
Collapse
|
8
|
Bonifazi A, Del Bello F, Giorgioni G, Piergentili A, Saab E, Botticelli L, Cifani C, Micioni Di Bonaventura E, Micioni Di Bonaventura MV, Quaglia W. Targeting orexin receptors: Recent advances in the development of subtype selective or dual ligands for the treatment of neuropsychiatric disorders. Med Res Rev 2023; 43:1607-1667. [PMID: 37036052 DOI: 10.1002/med.21959] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2022] [Revised: 03/08/2023] [Accepted: 03/28/2023] [Indexed: 04/11/2023]
Abstract
Orexin-A and orexin-B, also named hypocretin-1 and hypocretin-2, are two hypothalamic neuropeptides highly conserved across mammalian species. Their effects are mediated by two distinct G protein-coupled receptors, namely orexin receptor type 1 (OX1-R) and type 2 (OX2-R), which share 64% amino acid identity. Given the wide expression of OX-Rs in different central nervous system and peripheral areas and the several pathophysiological functions in which they are involved, including sleep-wake cycle regulation (mainly mediated by OX2-R), emotion, panic-like behaviors, anxiety/stress, food intake, and energy homeostasis (mainly mediated by OX1-R), both subtypes represent targets of interest for many structure-activity relationship (SAR) campaigns carried out by pharmaceutical companies and academies. However, before 2017 the research was predominantly directed towards dual-orexin ligands, and limited chemotypes were investigated. Analytical characterizations, including resolved structures for both OX1-R and OX2-R in complex with agonists and antagonists, have improved the understanding of the molecular basis of receptor recognition and are assets for medicinal chemists in the design of subtype-selective ligands. This review is focused on the medicinal chemistry aspects of small molecules acting as dual or subtype selective OX1-R/OX2-R agonists and antagonists belonging to different chemotypes and developed in the last years, including radiolabeled OX-R ligands for molecular imaging. Moreover, the pharmacological effects of the most studied ligands in different neuropsychiatric diseases, such as sleep, mood, substance use, and eating disorders, as well as pain, have been discussed. Poly-pharmacology applications and multitarget ligands have also been considered.
Collapse
Affiliation(s)
- Alessandro Bonifazi
- Medicinal Chemistry Section, Molecular Targets and Medications Discovery Branch, National Institute on Drug Abuse - Intramural Research Program, National Institutes of Health, Baltimore, Maryland, United States
| | - Fabio Del Bello
- School of Pharmacy, Medicinal Chemistry Unit, University of Camerino, Camerino, Italy
| | - Gianfabio Giorgioni
- School of Pharmacy, Medicinal Chemistry Unit, University of Camerino, Camerino, Italy
| | | | - Elizabeth Saab
- Medicinal Chemistry Section, Molecular Targets and Medications Discovery Branch, National Institute on Drug Abuse - Intramural Research Program, National Institutes of Health, Baltimore, Maryland, United States
| | - Luca Botticelli
- School of Pharmacy, Pharmacology Unit, University of Camerino, Camerino, Italy
| | - Carlo Cifani
- School of Pharmacy, Pharmacology Unit, University of Camerino, Camerino, Italy
| | | | | | - Wilma Quaglia
- School of Pharmacy, Medicinal Chemistry Unit, University of Camerino, Camerino, Italy
| |
Collapse
|
9
|
Botticelli L, Micioni Di Bonaventura E, Del Bello F, Giorgioni G, Piergentili A, Quaglia W, Bonifazi A, Cifani C, Micioni Di Bonaventura MV. The neuromedin U system: Pharmacological implications for the treatment of obesity and binge eating behavior. Pharmacol Res 2023; 195:106875. [PMID: 37517560 DOI: 10.1016/j.phrs.2023.106875] [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: 06/20/2023] [Revised: 07/25/2023] [Accepted: 07/28/2023] [Indexed: 08/01/2023]
Abstract
Neuromedin U (NMU) is a bioactive peptide produced in the gut and in the brain, with a role in multiple physiological processes. NMU acts by binding and activating two G protein coupled receptors (GPCR), the NMU receptor 1 (NMU-R1), which is predominantly expressed in the periphery, and the NMU receptor 2 (NMU-R2), mainly expressed in the central nervous system (CNS). In the brain, NMU and NMU-R2 are consistently present in the hypothalamus, commonly recognized as the main "feeding center". Considering its distribution pattern, NMU revealed to be an important neuropeptide involved in the regulation of food intake, with a powerful anorexigenic ability. This has been observed through direct administration of NMU and by studies using genetically modified animals, which revealed an obesity phenotype when the NMU gene is deleted. Thus, the development of NMU analogs or NMU-R2 agonists might represent a promising pharmacological strategy to treat obese individuals. Furthermore, NMU has been demonstrated to influence the non-homeostatic aspect of food intake, playing a potential role in binge eating behavior. This review aims to discuss and summarize the current literature linking the NMU system with obesity and binge eating behavior, focusing on the influence of NMU on food intake and the neuronal mechanisms underlying its anti-obesity properties. Pharmacological strategies to improve the pharmacokinetic profile of NMU will also be reported.
Collapse
Affiliation(s)
- Luca Botticelli
- School of Pharmacy, Pharmacology Unit, University of Camerino, via Madonna delle Carceri, 9, Camerino 62032, Italy
| | | | - Fabio Del Bello
- School of Pharmacy, Medicinal Chemistry Unit, University of Camerino, via Madonna delle Carceri, Camerino 62032, Italy
| | - Gianfabio Giorgioni
- School of Pharmacy, Medicinal Chemistry Unit, University of Camerino, via Madonna delle Carceri, Camerino 62032, Italy
| | - Alessandro Piergentili
- School of Pharmacy, Medicinal Chemistry Unit, University of Camerino, via Madonna delle Carceri, Camerino 62032, Italy
| | - Wilma Quaglia
- School of Pharmacy, Medicinal Chemistry Unit, University of Camerino, via Madonna delle Carceri, Camerino 62032, Italy
| | - Alessandro Bonifazi
- Medicinal Chemistry Section, Molecular Targets and Medications Discovery Branch, National Institute on Drug Abuse, Intramural Research Program, National Institutes of Health, 333 Cassell Drive, Baltimore, MD 21224, United States
| | - Carlo Cifani
- School of Pharmacy, Pharmacology Unit, University of Camerino, via Madonna delle Carceri, 9, Camerino 62032, Italy.
| | | |
Collapse
|
10
|
Feng B, Harms J, Chen E, Gao P, Xu P, He Y. Current Discoveries and Future Implications of Eating Disorders. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2023; 20:6325. [PMID: 37510558 PMCID: PMC10379623 DOI: 10.3390/ijerph20146325] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Revised: 03/20/2023] [Accepted: 07/05/2023] [Indexed: 07/30/2023]
Abstract
Eating disorders (EDs) are characterized by severe disturbances in eating behaviors and can sometimes be fatal. Eating disorders are also associated with distressing thoughts and emotions. They can be severe conditions affecting physical, psychological, and social functions. Preoccupation with food, body weight, and shape may also play an important role in the regulation of eating disorders. Common eating disorders have three major types: anorexia nervosa (AN), bulimia nervosa (BN), and binge eating disorder (BED). In some cases, EDs can have serious consequences for an individual's physical and mental health. These disorders often develop during adolescence or early adulthood and affect both males and females, although they are more commonly diagnosed in young adult females. Treatment for EDs typically involves a combination of therapy, nutrition counseling, and medical care. In this narrative review, the authors summarized what is known of EDs and discussed the future directions that may be worth exploring in this emerging area.
Collapse
Affiliation(s)
- Bing Feng
- Pennington Biomedical Research Center, Louisiana State University, Baton Rouge, LA 70808, USA
| | - Jerney Harms
- Pennington Biomedical Research Center, Louisiana State University, Baton Rouge, LA 70808, USA
- Biology Department, Centenary College of Louisiana, Shreveport, LA 71104, USA
| | - Emily Chen
- Pennington Biomedical Research Center, Louisiana State University, Baton Rouge, LA 70808, USA
| | - Peiyu Gao
- Pennington Biomedical Research Center, Louisiana State University, Baton Rouge, LA 70808, USA
| | - Pingwen Xu
- The Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, The University of Illinois at Chicago, Chicago, IL 60612, USA
| | - Yanlin He
- Pennington Biomedical Research Center, Louisiana State University, Baton Rouge, LA 70808, USA
| |
Collapse
|
11
|
Jin R, Sun S, Hu Y, Zhang H, Sun X. Neuropeptides Modulate Feeding via the Dopamine Reward Pathway. Neurochem Res 2023:10.1007/s11064-023-03954-4. [PMID: 37233918 DOI: 10.1007/s11064-023-03954-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2023] [Revised: 05/13/2023] [Accepted: 05/16/2023] [Indexed: 05/27/2023]
Abstract
Dopamine (DA) is a catecholamine neurotransmitter widely distributed in the central nervous system. It participates in various physiological functions, such as feeding, anxiety, fear, sleeping and arousal. The regulation of feeding is exceptionally complex, involving energy homeostasis and reward motivation. The reward system comprises the ventral tegmental area (VTA), nucleus accumbens (NAc), hypothalamus, and limbic system. This paper illustrates the detailed mechanisms of eight typical orexigenic and anorexic neuropeptides that regulate food intake through the reward system. According to recent literature, neuropeptides released from the hypothalamus and other brain regions regulate reward feeding predominantly through dopaminergic neurons projecting from the VTA to the NAc. In addition, their effect on the dopaminergic system is mediated by the prefrontal cortex, paraventricular thalamus, laterodorsal tegmental area, amygdala, and complex neural circuits. Research on neuropeptides involved in reward feeding can help identify more targets to treat diseases with metabolic disorders, such as obesity.
Collapse
Affiliation(s)
- Ruijie Jin
- Department of Physiology and Pathophysiology, School of Basic Medicine, Qingdao University, Qingdao, Shandong, China
- Department of Clinical Medicine, Medical College, Qingdao University, Qingdao, China
| | - Shanbin Sun
- Department of Physiology and Pathophysiology, School of Basic Medicine, Qingdao University, Qingdao, Shandong, China
- Department of Clinical Medicine, Medical College, Qingdao University, Qingdao, China
| | - Yang Hu
- Department of Physiology and Pathophysiology, School of Basic Medicine, Qingdao University, Qingdao, Shandong, China
- Department of Clinical Medicine, Medical College, Qingdao University, Qingdao, China
| | - Hongfei Zhang
- Department of Physiology and Pathophysiology, School of Basic Medicine, Qingdao University, Qingdao, Shandong, China
- Department of Clinical Medicine, Medical College, Qingdao University, Qingdao, China
| | - Xiangrong Sun
- Department of Physiology and Pathophysiology, School of Basic Medicine, Qingdao University, Qingdao, Shandong, China.
| |
Collapse
|
12
|
Baenas I, Miranda-Olivos R, Solé-Morata N, Jiménez-Murcia S, Fernández-Aranda F. Neuroendocrinological factors in binge eating disorder: A narrative review. Psychoneuroendocrinology 2023; 150:106030. [PMID: 36709632 DOI: 10.1016/j.psyneuen.2023.106030] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Revised: 01/19/2023] [Accepted: 01/20/2023] [Indexed: 01/30/2023]
Abstract
Neuroendocrine mechanisms play a key role in the regulation of eating behavior. In individuals with binge eating disorder (BED), alterations in these mechanisms signaling hunger and satiety have been observed. It has been investigated that these alterations may underlie the development and maintenance of compulsive overeating in BED. The present narrative review examined the current literature related to the neurobiological processes involved in feeding dysregulation in BED with the aim of updating the most relevant aspects with special attention to neuroendocrine signaling. Studies have shown both central and peripheral endocrine dysfunctions in hormones participating in homeostatic and hedonic pathways in BED. Most studies have been especially focused on orexigenic signals, pointing out the existence of a hyperactivated mechanism promoting hunger. Fewer studies have explored anorexigenic pathways, but the findings so far seem to suggest an abnormal satiety threshold. Despite this, to date, it is unable to identify whether these alterations are typical of the BED pathophysiology or are related to an obesogenic pattern due to most studies included patients with BED and obesity. The identification of endophenotypes in BED may provide a new approach to aberrant eating behavior, favoring the implementation of biological therapeutic targets.
Collapse
Affiliation(s)
- Isabel Baenas
- Department of Psychiatry, Bellvitge University Hospital, 08907 Barcelona, Spain; Psychoneurobiology of Eating and Addictive Behaviors Group, Neurosciences Program, Bellvitge Biomedical Research Institute (IDIBELL), 08908 Barcelona, Spain; Ciber Fisiopatología Obesidad y Nutrición (CIBERObn), Instituto de Salud Carlos III, 28029 Madrid, Spain.
| | - Romina Miranda-Olivos
- Department of Psychiatry, Bellvitge University Hospital, 08907 Barcelona, Spain; Psychoneurobiology of Eating and Addictive Behaviors Group, Neurosciences Program, Bellvitge Biomedical Research Institute (IDIBELL), 08908 Barcelona, Spain; Ciber Fisiopatología Obesidad y Nutrición (CIBERObn), Instituto de Salud Carlos III, 28029 Madrid, Spain.
| | - Neus Solé-Morata
- Department of Psychiatry, Bellvitge University Hospital, 08907 Barcelona, Spain.
| | - Susana Jiménez-Murcia
- Department of Psychiatry, Bellvitge University Hospital, 08907 Barcelona, Spain; Psychoneurobiology of Eating and Addictive Behaviors Group, Neurosciences Program, Bellvitge Biomedical Research Institute (IDIBELL), 08908 Barcelona, Spain; Ciber Fisiopatología Obesidad y Nutrición (CIBERObn), Instituto de Salud Carlos III, 28029 Madrid, Spain; Department of Clinical Sciences, School of Medicine and Health Sciences, University of Barcelona, 08907 Barcelona, Spain.
| | - Fernando Fernández-Aranda
- Department of Psychiatry, Bellvitge University Hospital, 08907 Barcelona, Spain; Psychoneurobiology of Eating and Addictive Behaviors Group, Neurosciences Program, Bellvitge Biomedical Research Institute (IDIBELL), 08908 Barcelona, Spain; Ciber Fisiopatología Obesidad y Nutrición (CIBERObn), Instituto de Salud Carlos III, 28029 Madrid, Spain; Department of Clinical Sciences, School of Medicine and Health Sciences, University of Barcelona, 08907 Barcelona, Spain.
| |
Collapse
|
13
|
Jerlhag E. Animal studies reveal that the ghrelin pathway regulates alcohol-mediated responses. Front Psychiatry 2023; 14:1050973. [PMID: 36970276 PMCID: PMC10030715 DOI: 10.3389/fpsyt.2023.1050973] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/22/2022] [Accepted: 02/21/2023] [Indexed: 03/29/2023] Open
Abstract
Alcohol use disorder (AUD) is often described as repeated phases of binge drinking, compulsive alcohol-taking, craving for alcohol during withdrawal, and drinking with an aim to a reduce the negative consequences. Although multifaceted, alcohol-induced reward is one aspect influencing the former three of these. The neurobiological mechanisms regulating AUD processes are complex and one of these systems is the gut-brain peptide ghrelin. The vast physiological properties of ghrelin are mediated via growth hormone secretagogue receptor (GHSR, ghrelin receptor). Ghrelin is well known for its ability to control feeding, hunger, and metabolism. Moreover, ghrelin signaling appears central for alcohol-mediated responses; findings reviewed herein. In male rodents GHSR antagonism reduces alcohol consumption, prevents relapse drinking, and attenuates the motivation to consume alcohol. On the other hand, ghrelin increases the consumption of alcohol. This ghrelin-alcohol interaction is also verified to some extent in humans with high alcohol consumption. In addition, either pharmacological or genetic suppression of GHSR decreases several alcohol-related effects (behavioral or neurochemical). Indeed, this suppression blocks the alcohol-induced hyperlocomotion and dopamine release in nucleus accumbens as well as ablates the alcohol reward in the conditioned place preference model. Although not fully elucidated, this interaction appears to involve areas central for reward, such as the ventral tegmental area (VTA) and brain nodes targeted by VTA projections. As reviewed briefly, the ghrelin pathway does not only modulate alcohol-mediated effects, it regulates reward-related behaviors induced by addictive drugs. Although personality traits like impulsivity and risk-taking behaviors are common in patients with AUD, the role of the ghrelin pathway thereof is unknown and remains to be studied. In summary, the ghrelin pathway regulates addiction processes like AUD and therefore the possibility that GHSR antagonism reduces alcohol or drug-taking should be explored in randomized clinical trials.
Collapse
|
14
|
Etxandi M, Baenas I, Mora-Maltas B, Granero R, Fernández-Aranda F, Tovar S, Solé-Morata N, Lucas I, Casado S, Gómez-Peña M, Moragas L, del Pino-Gutiérrez A, Codina E, Valenciano-Mendoza E, Potenza MN, Diéguez C, Jiménez-Murcia S. Are Signals Regulating Energy Homeostasis Related to Neuropsychological and Clinical Features of Gambling Disorder? A Case-Control Study. Nutrients 2022; 14:nu14235084. [PMID: 36501114 PMCID: PMC9736671 DOI: 10.3390/nu14235084] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2022] [Revised: 11/04/2022] [Accepted: 11/24/2022] [Indexed: 12/04/2022] Open
Abstract
Gambling disorder (GD) is a modestly prevalent and severe condition for which neurobiology is not yet fully understood. Although alterations in signals involved in energy homeostasis have been studied in substance use disorders, they have yet to be examined in detail in GD. The aims of the present study were to compare different endocrine and neuropsychological factors between individuals with GD and healthy controls (HC) and to explore endocrine interactions with neuropsychological and clinical variables. A case−control design was performed in 297 individuals with GD and 41 individuals without (healthy controls; HCs), assessed through a semi-structured clinical interview and a psychometric battery. For the evaluation of endocrine and anthropometric variables, 38 HCs were added to the 41 HCs initially evaluated. Individuals with GD presented higher fasting plasma ghrelin (p < 0.001) and lower LEAP2 and adiponectin concentrations (p < 0.001) than HCs, after adjusting for body mass index (BMI). The GD group reported higher cognitive impairment regarding cognitive flexibility and decision-making strategies, a worse psychological state, higher impulsivity levels, and a more dysfunctional personality profile. Despite failing to find significant associations between endocrine factors and either neuropsychological or clinical aspects in the GD group, some impaired cognitive dimensions (i.e., WAIS Vocabulary test and WCST Perseverative errors) and lower LEAP2 concentrations statistically predicted GD presence. The findings from the present study suggest that distinctive neuropsychological and endocrine dysfunctions may operate in individuals with GD and predict GD presence. Further exploration of endophenotypic vulnerability pathways in GD appear warranted, especially with respect to etiological and therapeutic potentials.
Collapse
Affiliation(s)
- Mikel Etxandi
- Department of Psychiatry, Bellvitge University Hospital-Bellvitge Institute for Biomedical Research (IDIBELL), 08907 Barcelona, Spain
- Department of Psychiatry, Hospital Universitari Germans Trias i Pujol, IGTP Campus Can Ruti, 08916 Badalona, Spain
| | - Isabel Baenas
- Department of Psychiatry, Bellvitge University Hospital-Bellvitge Institute for Biomedical Research (IDIBELL), 08907 Barcelona, Spain
- Ciber Fisiopatología Obesidad y Nutrición (CIBERObn), Instituto de Salud Carlos III, 28029 Madrid, Spain
- Psychoneurobiology of Eating and Addictive Behaviors Group, Neurosciences Programme, Bellvitge Institute for Biomedical Research (IDIBELL), 08908 Barcelona, Spain
| | - Bernat Mora-Maltas
- Department of Psychiatry, Bellvitge University Hospital-Bellvitge Institute for Biomedical Research (IDIBELL), 08907 Barcelona, Spain
- Psychoneurobiology of Eating and Addictive Behaviors Group, Neurosciences Programme, Bellvitge Institute for Biomedical Research (IDIBELL), 08908 Barcelona, Spain
| | - Roser Granero
- Ciber Fisiopatología Obesidad y Nutrición (CIBERObn), Instituto de Salud Carlos III, 28029 Madrid, Spain
- Psychoneurobiology of Eating and Addictive Behaviors Group, Neurosciences Programme, Bellvitge Institute for Biomedical Research (IDIBELL), 08908 Barcelona, Spain
- Department of Psychobiology and Methodology, Autonomous University of Barcelona, 08193 Barcelona, Spain
| | - Fernando Fernández-Aranda
- Department of Psychiatry, Bellvitge University Hospital-Bellvitge Institute for Biomedical Research (IDIBELL), 08907 Barcelona, Spain
- Ciber Fisiopatología Obesidad y Nutrición (CIBERObn), Instituto de Salud Carlos III, 28029 Madrid, Spain
- Psychoneurobiology of Eating and Addictive Behaviors Group, Neurosciences Programme, Bellvitge Institute for Biomedical Research (IDIBELL), 08908 Barcelona, Spain
- Department of Clinical Sciences, School of Medicine and Health Sciences, University of Barcelona, 08907 Barcelona, Spain
| | - Sulay Tovar
- Ciber Fisiopatología Obesidad y Nutrición (CIBERObn), Instituto de Salud Carlos III, 28029 Madrid, Spain
- Department of Physiology, CIMUS, Instituto de Investigación Sanitaria, University of Santiago de Compostela, 15782 Santiago de Compostela, Spain
| | - Neus Solé-Morata
- Department of Psychiatry, Bellvitge University Hospital-Bellvitge Institute for Biomedical Research (IDIBELL), 08907 Barcelona, Spain
- Psychoneurobiology of Eating and Addictive Behaviors Group, Neurosciences Programme, Bellvitge Institute for Biomedical Research (IDIBELL), 08908 Barcelona, Spain
| | - Ignacio Lucas
- Department of Psychiatry, Bellvitge University Hospital-Bellvitge Institute for Biomedical Research (IDIBELL), 08907 Barcelona, Spain
- Psychoneurobiology of Eating and Addictive Behaviors Group, Neurosciences Programme, Bellvitge Institute for Biomedical Research (IDIBELL), 08908 Barcelona, Spain
| | - Sabela Casado
- Ciber Fisiopatología Obesidad y Nutrición (CIBERObn), Instituto de Salud Carlos III, 28029 Madrid, Spain
- Department of Physiology, CIMUS, Instituto de Investigación Sanitaria, University of Santiago de Compostela, 15782 Santiago de Compostela, Spain
| | - Mónica Gómez-Peña
- Department of Psychiatry, Bellvitge University Hospital-Bellvitge Institute for Biomedical Research (IDIBELL), 08907 Barcelona, Spain
- Psychoneurobiology of Eating and Addictive Behaviors Group, Neurosciences Programme, Bellvitge Institute for Biomedical Research (IDIBELL), 08908 Barcelona, Spain
| | - Laura Moragas
- Department of Psychiatry, Bellvitge University Hospital-Bellvitge Institute for Biomedical Research (IDIBELL), 08907 Barcelona, Spain
- Psychoneurobiology of Eating and Addictive Behaviors Group, Neurosciences Programme, Bellvitge Institute for Biomedical Research (IDIBELL), 08908 Barcelona, Spain
| | - Amparo del Pino-Gutiérrez
- Ciber Fisiopatología Obesidad y Nutrición (CIBERObn), Instituto de Salud Carlos III, 28029 Madrid, Spain
- Psychoneurobiology of Eating and Addictive Behaviors Group, Neurosciences Programme, Bellvitge Institute for Biomedical Research (IDIBELL), 08908 Barcelona, Spain
- Department of Public Health, Mental Health and Perinatal Nursing, School of Nursing, University of Barcelona, 08907 Barcelona, Spain
| | - Ester Codina
- Department of Psychiatry, Bellvitge University Hospital-Bellvitge Institute for Biomedical Research (IDIBELL), 08907 Barcelona, Spain
| | - Eduardo Valenciano-Mendoza
- Department of Psychiatry, Bellvitge University Hospital-Bellvitge Institute for Biomedical Research (IDIBELL), 08907 Barcelona, Spain
| | - Marc N. Potenza
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT 06510, USA
- Child Study Center, Yale University School of Medicine, New Haven, CT 06510, USA
- Connecticut Mental Health Center, New Haven, CT 06519, USA
- Connecticut Council on Problem Gambling, Wethersfield, CT 06106, USA
- Department of Neuroscience, Yale University, New Haven, CT 06520, USA
| | - Carlos Diéguez
- Ciber Fisiopatología Obesidad y Nutrición (CIBERObn), Instituto de Salud Carlos III, 28029 Madrid, Spain
- Department of Physiology, CIMUS, Instituto de Investigación Sanitaria, University of Santiago de Compostela, 15782 Santiago de Compostela, Spain
| | - Susana Jiménez-Murcia
- Department of Psychiatry, Bellvitge University Hospital-Bellvitge Institute for Biomedical Research (IDIBELL), 08907 Barcelona, Spain
- Ciber Fisiopatología Obesidad y Nutrición (CIBERObn), Instituto de Salud Carlos III, 28029 Madrid, Spain
- Psychoneurobiology of Eating and Addictive Behaviors Group, Neurosciences Programme, Bellvitge Institute for Biomedical Research (IDIBELL), 08908 Barcelona, Spain
- Department of Clinical Sciences, School of Medicine and Health Sciences, University of Barcelona, 08907 Barcelona, Spain
- Correspondence:
| |
Collapse
|
15
|
Micioni Di Bonaventura E, Botticelli L, Del Bello F, Giorgioni G, Piergentili A, Quaglia W, Romano A, Gaetani S, Micioni Di Bonaventura MV, Cifani C. Investigating the role of the central melanocortin system in stress and stress-related disorders. Pharmacol Res 2022; 185:106521. [DOI: 10.1016/j.phrs.2022.106521] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/26/2022] [Revised: 10/13/2022] [Accepted: 10/18/2022] [Indexed: 11/30/2022]
|
16
|
Rahati S, Qorbani M, Naghavi A, Pishva H. Association and interaction of the MC4R rs17782313 polymorphism with plasma ghrelin, GLP-1, cortisol, food intake and eating behaviors in overweight/obese Iranian adults. BMC Endocr Disord 2022; 22:234. [PMID: 36123585 PMCID: PMC9487018 DOI: 10.1186/s12902-022-01129-w] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/18/2021] [Accepted: 08/16/2022] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Recent studies have shown that obesity is largely influenced by heredity and created by the interactions between several genes and environmental and behavioral factors. This study aimed to examine association between variant rs17782313 near melanocortin-4 receptor (MC4R) gene and behavioral and hormonal factors then evaluated interactions between variant MC4R rs17782313 with behavioral and hormonal factors on obesity. METHODS This cross-sectional study included 403 subjects, overweight and/or obesity, aged 20-50 years from Iran. The MC4R rs17782313 data were measured by the PCR-RFLP method. Dietary intake, physical activity, stress, anxiety, depression, appetite and emotional eating were assessed by using validated questionnaires. Ghrelin, glucagon-like peptide-1 and cortisol were measured by radioimmunoassay in plasma samples. Participants were also divided into three groups based on rs17782313 genotype and BMI. RESULTS After adjustment for age, gender, energy intake and PA, significant associations were observed between food intake, appetite, emotional eating, stress and physical activity with MC4R rs17782313 (p ˂0.05). Also, significant interactions were observed between fat intake (p-interaction = 0.002), protein intake (p-interaction = 0.01), energy intake (p-interaction = 0.01), emotional eating (p-interaction = 0.02), appetite (p-interaction = 0.04), stress (p-interaction = 0.04), ghrelin (p-interaction = 0.03), cortisol (p-interaction = 0.04) and physical activity (p-interaction = 0.04) and MC4R rs17782313 in terms of BMI. CONCLUSION Interactions between the CC genotype and high intakes of fat and energy, emotional eating, high appetite, and too much stress with high levels of cortisol and ghrelin probably can have an effect on BMI in overweight/obese subjects.
Collapse
Affiliation(s)
- Sara Rahati
- Department of Cellular - Molecular Nutrition, School of Nutrition Sciences and Dietetics, Tehran University of Medical Sciences, PO Box: 14155-6447, Tehran, Iran
| | - Mostafa Qorbani
- Non-Communicable Diseases Research Center, Alborz University of Medical Sciences, Karaj, Iran
| | - Anoosh Naghavi
- Cellular and Molecular Research Center, Resistant Tuberculosis Institute and Department of Genetics, Zahedan University of Medical Sciences, Zahedan, Iran
| | - Hamideh Pishva
- Department of Cellular - Molecular Nutrition, School of Nutrition Sciences and Dietetics, Tehran University of Medical Sciences, PO Box: 14155-6447, Tehran, Iran.
| |
Collapse
|
17
|
Adipokines in Non-Alcoholic Fatty Liver Disease: Are We on the Road toward New Biomarkers and Therapeutic Targets? BIOLOGY 2022; 11:biology11081237. [PMID: 36009862 PMCID: PMC9405285 DOI: 10.3390/biology11081237] [Citation(s) in RCA: 31] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/19/2022] [Revised: 08/16/2022] [Accepted: 08/17/2022] [Indexed: 12/04/2022]
Abstract
Simple Summary Non-alcoholic fatty liver disease (NAFLD) is an unmet medical need due to its increasingly high incidence, severe clinical consequences, and the absence of feasible diagnostic tools and effective drugs. This review summarizes the preclinical and clinical data on adipokines, cytokine-like hormones secreted by adipose tissue, and NAFLD. The aim is to establish the potential of adipokines as diagnostic and prognostic biomarkers, as well as their potential as therapeutic targets for NAFLD. The limitations of current research are also discussed, and future perspectives are outlined. Abstract Non-alcoholic fatty liver disease (NAFLD) has become the major cause of chronic hepatic illness and the leading indication for liver transplantation in the future decades. NAFLD is also commonly associated with other high-incident non-communicable diseases, such as cardiovascular complications, type 2 diabetes, and chronic kidney disease. Aggravating the socio-economic impact of this complex pathology, routinely feasible diagnostic methodologies and effective drugs for NAFLD management are unavailable. The pathophysiology of NAFLD, recently defined as metabolic associated fatty liver disease (MAFLD), is correlated with abnormal adipose tissue–liver axis communication because obesity-associated white adipose tissue (WAT) inflammation and metabolic dysfunction prompt hepatic insulin resistance (IR), lipid accumulation (steatosis), non-alcoholic steatohepatitis (NASH), and fibrosis. Accumulating evidence links adipokines, cytokine-like hormones secreted by adipose tissue that have immunometabolic activity, with NAFLD pathogenesis and progression; however, much uncertainty still exists. Here, the current knowledge on the roles of leptin, adiponectin, ghrelin, resistin, retinol-binding protein 4 (RBP4), visfatin, chemerin, and adipocyte fatty-acid-binding protein (AFABP) in NAFLD, taken from preclinical to clinical studies, is overviewed. The effect of therapeutic interventions on adipokines’ circulating levels are also covered. Finally, future directions to address the potential of adipokines as therapeutic targets and disease biomarkers for NAFLD are discussed.
Collapse
|
18
|
Food Reward Alterations during Obesity Are Associated with Inflammation in the Striatum in Mice: Beneficial Effects of Akkermansia muciniphila. Cells 2022; 11:cells11162534. [PMID: 36010611 PMCID: PMC9406832 DOI: 10.3390/cells11162534] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2022] [Revised: 08/06/2022] [Accepted: 08/09/2022] [Indexed: 11/17/2022] Open
Abstract
The reward system involved in hedonic food intake presents neuronal and behavioral dysregulations during obesity. Moreover, gut microbiota dysbiosis during obesity promotes low-grade inflammation in peripheral organs and in the brain contributing to metabolic alterations. The mechanisms underlying reward dysregulations during obesity remain unclear. We investigated if inflammation affects the striatum during obesity using a cohort of control-fed or diet-induced obese (DIO) male mice. We tested the potential effects of specific gut bacteria on the reward system during obesity by administrating Akkermansia muciniphila daily or a placebo to DIO male mice. We showed that dysregulations of the food reward are associated with inflammation and alterations in the blood–brain barrier in the striatum of obese mice. We identified Akkermansia muciniphila as a novel actor able to improve the dysregulated reward behaviors associated with obesity, potentially through a decreased activation of inflammatory pathways and lipid-sensing ability in the striatum. These results open a new field of research and suggest that gut microbes can be considered as an innovative therapeutic approach to attenuate reward alterations in obesity. This study provides substance for further investigations of Akkermansia muciniphila-mediated behavioral improvements in other inflammatory neuropsychiatric disorders.
Collapse
|
19
|
Bukhari SNA. An insight into the multifunctional role of ghrelin and structure activity relationship studies of ghrelin receptor ligands with clinical trials. Eur J Med Chem 2022; 235:114308. [PMID: 35344905 DOI: 10.1016/j.ejmech.2022.114308] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2021] [Revised: 02/06/2022] [Accepted: 03/18/2022] [Indexed: 11/30/2022]
Abstract
Ghrelin is a multifunctional gastrointestinal acylated peptide, primarily synthesized in the stomach and regulates the secretion of growth hormone and energy homeostasis. It plays a central role in modulating the diverse biological, physiological and pathological functions in vertebrates. The synthesis of ghrelin receptor ligands after the finding of growth hormone secretagogue developed from Met-enkephalin led to reveal the endogenous ligand ghrelin and the receptors. Subsequently, many peptides, small molecules and peptidomimetics focusing on the ghrelin receptor, GHS-R1a, were derived. In this review, the key features of ghrelin's structure, forms, its bio-physiological functions, pathological roles and therapeutic potential have been highlighted. A few peptidomimetics and pseudo peptide synthetic perspectives have also been discussed to make ghrelin receptor ligands, clinical trials and their success.
Collapse
Affiliation(s)
- Syed Nasir Abbas Bukhari
- Department of Pharmaceutical Chemistry, College of Pharmacy, Jouf University, Sakaka, Aljouf, 2014, Saudi Arabia.
| |
Collapse
|
20
|
Giorgioni G, Del Bello F, Quaglia W, Botticelli L, Cifani C, Micioni Di Bonaventura E, Micioni Di Bonaventura MV, Piergentili A. Advances in the Development of Nonpeptide Small Molecules Targeting Ghrelin Receptor. J Med Chem 2022; 65:3098-3118. [PMID: 35157454 PMCID: PMC8883476 DOI: 10.1021/acs.jmedchem.1c02191] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Ghrelin is an octanoylated peptide acting by the activation of the growth hormone secretagogue receptor, namely, GHS-R1a. The involvement of ghrelin in several physiological processes, including stimulation of food intake, gastric emptying, body energy balance, glucose homeostasis, reduction of insulin secretion, and lipogenesis validates the considerable interest in GHS-R1a as a promising target for the treatment of numerous disorders. Over the years, several GHS-R1a ligands have been identified and some of them have been extensively studied in clinical trials. The recently resolved structures of GHS-R1a bound to ghrelin or potent ligands have provided useful information for the design of new GHS-R1a drugs. This perspective is focused on the development of recent nonpeptide small molecules acting as GHS-R1a agonists, antagonists, and inverse agonists, bearing classical or new molecular scaffolds, as well as on radiolabeled GHS-R1a ligands developed for imaging. Moreover, the pharmacological effects of the most studied ligands have been discussed.
Collapse
Affiliation(s)
- Gianfabio Giorgioni
- School of Pharmacy, Medicinal Chemistry Unit, University of Camerino, Via Madonna delle Carceri, 62032 Camerino, Italy
| | - Fabio Del Bello
- School of Pharmacy, Medicinal Chemistry Unit, University of Camerino, Via Madonna delle Carceri, 62032 Camerino, Italy
| | - Wilma Quaglia
- School of Pharmacy, Medicinal Chemistry Unit, University of Camerino, Via Madonna delle Carceri, 62032 Camerino, Italy
| | - Luca Botticelli
- School of Pharmacy, Pharmacology Unit, University of Camerino, Via Madonna delle Carceri 9, 62032 Camerino, Italy
| | - Carlo Cifani
- School of Pharmacy, Pharmacology Unit, University of Camerino, Via Madonna delle Carceri 9, 62032 Camerino, Italy
| | - E Micioni Di Bonaventura
- School of Pharmacy, Pharmacology Unit, University of Camerino, Via Madonna delle Carceri 9, 62032 Camerino, Italy
| | - M V Micioni Di Bonaventura
- School of Pharmacy, Pharmacology Unit, University of Camerino, Via Madonna delle Carceri 9, 62032 Camerino, Italy
| | - Alessandro Piergentili
- School of Pharmacy, Medicinal Chemistry Unit, University of Camerino, Via Madonna delle Carceri, 62032 Camerino, Italy
| |
Collapse
|
21
|
Tufvesson-Alm M, Shevchouk OT, Jerlhag E. Insight into the role of the gut-brain axis in alcohol-related responses: Emphasis on GLP-1, amylin, and ghrelin. Front Psychiatry 2022; 13:1092828. [PMID: 36699502 PMCID: PMC9868418 DOI: 10.3389/fpsyt.2022.1092828] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/09/2022] [Accepted: 12/15/2022] [Indexed: 01/11/2023] Open
Abstract
Alcohol use disorder (AUD) contributes substantially to global morbidity and mortality. Given the heterogenicity of this brain disease, available pharmacological treatments only display efficacy in sub-set of individuals. The need for additional treatment options is thus substantial and is the goal of preclinical studies unraveling neurobiological mechanisms underlying AUD. Although these neurobiological processes are complex and numerous, one system gaining recent attention is the gut-brain axis. Peptides of the gut-brain axis include anorexigenic peptide like glucagon-like peptide-1 (GLP-1) and amylin as well as the orexigenic peptide ghrelin. In animal models, agonists of the GLP-1 or amylin receptor and ghrelin receptor (GHSR) antagonists reduce alcohol drinking, relapse drinking, and alcohol-seeking. Moreover, these three gut-brain peptides modulate alcohol-related responses (behavioral and neurochemical) in rodents, suggesting that the alcohol reduction may involve a suppression of alcohol's rewarding properties. Brain areas participating in the ability of these gut-brain peptides to reduce alcohol-mediated behaviors/neurochemistry involve those important for reward. Human studies support these preclinical studies as polymorphisms of the genes encoding for GLP-1 receptor or the ghrelin pathway are associated with AUD. Moreover, a GLP-1 receptor agonist decreases alcohol drinking in overweight patients with AUD and an inverse GHSR agonist reduces alcohol craving. Although preclinical and clinical studies reveal an interaction between the gut-brain axis and AUD, additional studies should explore this in more detail.
Collapse
Affiliation(s)
- Maximilian Tufvesson-Alm
- Department of Pharmacology, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Olesya T Shevchouk
- Department of Pharmacology, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Elisabet Jerlhag
- Department of Pharmacology, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| |
Collapse
|
22
|
Micioni Di Bonaventura MV, Coman MM, Tomassoni D, Micioni Di Bonaventura E, Botticelli L, Gabrielli MG, Rossolini GM, Di Pilato V, Cecchini C, Amedei A, Silvi S, Verdenelli MC, Cifani C. Supplementation with Lactiplantibacillus plantarum IMC 510 Modifies Microbiota Composition and Prevents Body Weight Gain Induced by Cafeteria Diet in Rats. Int J Mol Sci 2021; 22:ijms222011171. [PMID: 34681831 PMCID: PMC8540549 DOI: 10.3390/ijms222011171] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Revised: 10/11/2021] [Accepted: 10/12/2021] [Indexed: 12/19/2022] Open
Abstract
Changes in functionality and composition of gut microbiota (GM) have been associated and may contribute to the development and maintenance of obesity and related diseases. The aim of our study was to investigate for the first time the impact of Lactiplantibacillus (L.) plantarum IMC 510 in a rat model of diet-induced obesity, specifically in the cafeteria (CAF) diet. This diet provides a strong motivation to voluntary overeat, due to the palatability and variety of selected energy-dense foods. The oral administration for 84 days of this probiotic strain, added to the CAF diet, decreased food intake and body weight gain. Accordingly, it ameliorated body mass index, liver and white adipose tissue weight, hepatic lipid accumulation, adipocyte size, serum parameters, including glycemia and low-density lipoprotein levels, in CAF fed rats, potentially through leptin control. In this scenario, L. plantarum IMC 510 showed also beneficial effects on GM, limiting the microbial imbalance established by long exposure to CAF diet and preserving the proportion of different bacterial taxa. Further research is necessary to better elucidate the relationship between GM and overweight and then the mechanism of action by which L. plantarum IMC 510 modifies weight. However, these promising results prompt a clear advantage of probiotic supplementation and identify a new potential probiotic as a novel and safe therapeutic approach in obesity prevention and management.
Collapse
Affiliation(s)
| | - Maria Magdalena Coman
- Synbiotec S.r.l., Spin-off of UNICAM, Via Gentile III Da Varano, 62032 Camerino, Italy; (M.M.C.); (C.C.); (M.C.V.)
| | - Daniele Tomassoni
- School of Biosciences and Veterinary Medicine, University of Camerino, 62032 Camerino, Italy; (D.T.); (M.G.G.)
| | - Emanuela Micioni Di Bonaventura
- Pharmacology Unit, School of Pharmacy, University of Camerino, 62032 Camerino, Italy; (M.V.M.D.B.); (E.M.D.B.); (L.B.); (C.C.)
| | - Luca Botticelli
- Pharmacology Unit, School of Pharmacy, University of Camerino, 62032 Camerino, Italy; (M.V.M.D.B.); (E.M.D.B.); (L.B.); (C.C.)
| | - Maria Gabriella Gabrielli
- School of Biosciences and Veterinary Medicine, University of Camerino, 62032 Camerino, Italy; (D.T.); (M.G.G.)
| | - Gian Maria Rossolini
- Department of Experimental and Clinical Medicine, University of Florence, 50134 Florence, Italy; (G.M.R.); (A.A.)
- Microbiology and Virology Unit, Florence Careggi University Hospital, 50134 Florence, Italy
| | - Vincenzo Di Pilato
- Department of Surgical Sciences and Integrated Diagnostics, University of Genoa, 16132 Genova, Italy;
| | - Cinzia Cecchini
- Synbiotec S.r.l., Spin-off of UNICAM, Via Gentile III Da Varano, 62032 Camerino, Italy; (M.M.C.); (C.C.); (M.C.V.)
| | - Amedeo Amedei
- Department of Experimental and Clinical Medicine, University of Florence, 50134 Florence, Italy; (G.M.R.); (A.A.)
| | - Stefania Silvi
- School of Biosciences and Veterinary Medicine, University of Camerino, 62032 Camerino, Italy; (D.T.); (M.G.G.)
- Correspondence:
| | - Maria Cristina Verdenelli
- Synbiotec S.r.l., Spin-off of UNICAM, Via Gentile III Da Varano, 62032 Camerino, Italy; (M.M.C.); (C.C.); (M.C.V.)
| | - Carlo Cifani
- Pharmacology Unit, School of Pharmacy, University of Camerino, 62032 Camerino, Italy; (M.V.M.D.B.); (E.M.D.B.); (L.B.); (C.C.)
| |
Collapse
|