1
|
Lu Z, Ngan MP, Liu JYH, Yang L, Tu L, Chan SW, Giuliano C, Lovati E, Pietra C, Rudd JA. The growth hormone secretagogue receptor 1a agonists, anamorelin and ipamorelin, inhibit cisplatin-induced weight loss in ferrets: Anamorelin also exhibits anti-emetic effects via a central mechanism. Physiol Behav 2024; 284:114644. [PMID: 39043357 DOI: 10.1016/j.physbeh.2024.114644] [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: 05/31/2024] [Revised: 07/17/2024] [Accepted: 07/19/2024] [Indexed: 07/25/2024]
Abstract
This study investigated whether ghrelin mimetics, namely anamorelin and ipamorelin, can alleviate weight loss and inhibition of feeding observed during acute and delayed phases of cisplatin-induced emesis in ferrets. The potential of anamorelin to inhibit electrical field stimulation (EFS)-induced contractions of isolated ferret ileum was compared with ipamorelin. In other experiments, ferrets were administered anamorelin (1-3 mg/kg), ipamorelin (1-3 mg/kg), or vehicle intraperitoneally (i.p.) 30 s before cisplatin (5 mg/kg, i.p.) and then every 24 h, and their behaviour was recorded for up to 72 h. Food and water consumption was measured every 24 h. The effect of anamorelin (10 µg) was also assessed following intracerebroventricular administration. Anamorelin and ipamorelin inhibited EFS-induced contractions of isolated ileum by 94.4 % (half-maximal inhibitory concentration [IC50]=14.0 µM) and 54.4 % (IC50=11.7 µM), respectively. Neither of compounds administered i.p. had any effect on cisplatin-induced acute or delayed emesis, but both inhibited associated cisplatin-induced weight loss on the last day of delayed phase (48-72 h) by approximately 24 %. Anamorelin (10 µg) administered intracerebroventricularly reduced cisplatin-induced acute emesis by 60 % but did not affect delayed emesis. It also improved food and water consumption by approximately 20 %-40 % during acute phase, but not delayed phase, and reduced associated cisplatin-induced weight loss during delayed phase by ∼23 %. In conclusion, anamorelin and ipamorelin administered i.p. had beneficial effects in alleviating cisplatin-induced weight loss during delayed phase, and these effects were seen when centrally administered anamorelin. Anamorelin inhibited cisplatin-induced acute emesis following intracerebroventricular but not intraperitoneal administration, suggesting that brain penetration is important for its anti-emetic mechanism of action.
Collapse
Affiliation(s)
- Zengbing Lu
- Emesis Research Group, School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, New Territories, Hong Kong
| | - Man P Ngan
- Emesis Research Group, School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, New Territories, Hong Kong
| | - Julia Y H Liu
- Emesis Research Group, School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, New Territories, Hong Kong
| | - Lingqing Yang
- Emesis Research Group, School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, New Territories, Hong Kong
| | - Longlong Tu
- Emesis Research Group, School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, New Territories, Hong Kong; USDA/ARS Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, USA
| | - Sze Wa Chan
- School of Health Sciences, Saint Francis University, Tseung Kwan O, New Territories, Hong Kong
| | | | | | | | - John A Rudd
- Emesis Research Group, School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, New Territories, Hong Kong.
| |
Collapse
|
2
|
Montalban E, Giralt A, Taing L, Nakamura Y, Pelosi A, Brown M, de Pins B, Valjent E, Martin M, Nairn AC, Greengard P, Flajolet M, Hervé D, Gambardella N, Roussarie JP, Girault JA. Operant Training for Highly Palatable Food Alters Translating Messenger RNA in Nucleus Accumbens D 2 Neurons and Reveals a Modulatory Role of Ncdn. Biol Psychiatry 2024; 95:926-937. [PMID: 37579933 PMCID: PMC11059129 DOI: 10.1016/j.biopsych.2023.08.006] [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: 03/14/2023] [Revised: 08/04/2023] [Accepted: 08/04/2023] [Indexed: 08/16/2023]
Abstract
BACKGROUND Highly palatable food triggers behavioral responses including strong motivation. These effects involve the reward system and dopamine neurons, which modulate neurons in the nucleus accumbens (NAc). The molecular mechanisms underlying the long-lasting effects of highly palatable food on feeding behavior are poorly understood. METHODS We studied the effects of 2-week operant conditioning of mice with standard or isocaloric highly palatable food. We investigated the behavioral responses and dendritic spine modifications in the NAc. We compared the translating messenger RNA in NAc neurons identified by the type of dopamine receptors they express, depending on the kind of food and training. We tested the consequences of invalidation of an abundant downregulated gene, Ncdn. RESULTS Operant conditioning for highly palatable food increased motivation for food even in well-fed mice. In wild-type mice, free choice between regular and highly palatable food increased weight compared with access to regular food only. Highly palatable food increased spine density in the NAc. In animals trained for highly palatable food, translating messenger RNAs were modified in NAc neurons expressing dopamine D2 receptors, mostly corresponding to striatal projection neurons, but not in neurons expressing D1 receptors. Knockout of Ncdn, an abundant downregulated gene, opposed the conditioning-induced changes in satiety-sensitive feeding behavior and apparent motivation for highly palatable food, suggesting that downregulation may be a compensatory mechanism. CONCLUSIONS Our results emphasize the importance of messenger RNA alterations in D2 striatal projection neurons in the NAc in the behavioral consequences of highly palatable food conditioning and suggest a modulatory contribution of Ncdn downregulation.
Collapse
Affiliation(s)
- Enrica Montalban
- Institut National de la Santé et de la Recherche Médicale Unite Mixte de Recherche-S 1270, Paris, France; Faculty of Sciences and Engineering, Sorbonne Université, Paris, France; Institut du Fer à Moulin, Paris, France.
| | - Albert Giralt
- Institut National de la Santé et de la Recherche Médicale Unite Mixte de Recherche-S 1270, Paris, France; Faculty of Sciences and Engineering, Sorbonne Université, Paris, France; Institut du Fer à Moulin, Paris, France
| | - Lieng Taing
- Institut National de la Santé et de la Recherche Médicale Unite Mixte de Recherche-S 1270, Paris, France; Faculty of Sciences and Engineering, Sorbonne Université, Paris, France; Institut du Fer à Moulin, Paris, France
| | - Yuki Nakamura
- Institut National de la Santé et de la Recherche Médicale Unite Mixte de Recherche-S 1270, Paris, France; Faculty of Sciences and Engineering, Sorbonne Université, Paris, France; Institut du Fer à Moulin, Paris, France
| | - Assunta Pelosi
- Institut National de la Santé et de la Recherche Médicale Unite Mixte de Recherche-S 1270, Paris, France; Faculty of Sciences and Engineering, Sorbonne Université, Paris, France; Institut du Fer à Moulin, Paris, France
| | - Mallory Brown
- Laboratory of Molecular and Cellular Neuroscience, Rockefeller University, New York, New York
| | - Benoit de Pins
- Institut National de la Santé et de la Recherche Médicale Unite Mixte de Recherche-S 1270, Paris, France; Faculty of Sciences and Engineering, Sorbonne Université, Paris, France; Institut du Fer à Moulin, Paris, France
| | - Emmanuel Valjent
- Institut de Génomique Fonctionnelle, University of Montpellier, Centre National de la Recherche Scientifique, Institut National de la Santé et de la Recherche Médicale, Montpellier, France
| | - Miquel Martin
- Eurecat, Centre Tecnològic de Catalunya, Unitat de Nutrició i Salut, Reus, Spain; Instituto de investigaciones médicas Hospital del Mar, Barcelona, Spain
| | - Angus C Nairn
- Department of Psychiatry, Yale School of Medicine, Connecticut Mental Health Center, New Haven, Connecticut
| | - Paul Greengard
- Laboratory of Molecular and Cellular Neuroscience, Rockefeller University, New York, New York
| | - Marc Flajolet
- Laboratory of Molecular and Cellular Neuroscience, Rockefeller University, New York, New York
| | - Denis Hervé
- Institut National de la Santé et de la Recherche Médicale Unite Mixte de Recherche-S 1270, Paris, France; Faculty of Sciences and Engineering, Sorbonne Université, Paris, France; Institut du Fer à Moulin, Paris, France
| | | | - Jean-Pierre Roussarie
- Laboratory of Molecular and Cellular Neuroscience, Rockefeller University, New York, New York
| | - Jean-Antoine Girault
- Institut National de la Santé et de la Recherche Médicale Unite Mixte de Recherche-S 1270, Paris, France; Faculty of Sciences and Engineering, Sorbonne Université, Paris, France; Institut du Fer à Moulin, Paris, France.
| |
Collapse
|
3
|
Zhu S, Li J, Li Z, Wang Z, Wei Q, Shi F. Effects of non-nutritive sweeteners on growth and intestinal health by regulating hypothalamic RNA profile and ileum microbiota in guinea pigs. JOURNAL OF THE SCIENCE OF FOOD AND AGRICULTURE 2024; 104:4342-4353. [PMID: 38328855 DOI: 10.1002/jsfa.13320] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/24/2023] [Revised: 01/08/2024] [Accepted: 01/10/2024] [Indexed: 02/09/2024]
Abstract
BACKGROUND Non-nutritive sweeteners (NNS) are commonly used in sweetened foods and beverages; however their role in metabolic regulation is still not clear. In this experiment, we used guinea pigs as an animal model to study the effect of NNS on body growth and intestinal health by modifying gut microbiota and hypothalamus-related proteins. RESULTS For a 28-day feeding experiment a total of 40 guinea pigs were randomly divided into four groups, one control (CN) group and three treatments, in which three NNS were added to the diet: rebaudioside A (RA, 330 mg kg-1), sodium saccharin (SS, 800 mg kg-1), and sucralose (TGS, 167 mg kg-1), respectively. The TGS group exhibited significantly reduced food consumption in comparison with the CN group (P < 0.05) whereas the RA group showed increased food consumption in comparison with the CN group (P < 0.05). Notably, Taste receptor type 1 subunit 2 (T1R2) expression in the hypothalamus was significantly higher in the RA group than in the CN group (P < 0.05). The mRNA expressions of appetite-stimulated genes arouti-related neuropeptide (AGRP), neuropeptide Y (NPY), and thyroid stimulating hormone (TSHB) were significantly higher than those in the CN group (P < 0.05) but mRNA expressions of appetite-suppressed genes tryptophan hydroxylase 2(THP2) were significantly lower in the TGS group (P < 0.05). Furthermore, NNS in the guinea pig diets (RA, SS, TGS) significantly increased the relative abundance of Muribaculaceae but decreased the relative abundance of Clostridia_vadin BB60 in comparison with the CN group (P < 0.05). We also found that dietary supplementation with RA also significantly altered the relative abundance of Lactobacillus. CONCLUSION Our finding confirmed that dietary supplementation with RA and TGS affected body growth and intestinal health by modulating hypothalamic RNA profiles and ileum microbiota, suggesting that NNS should be included in guinea-pig feeding. © 2024 Society of Chemical Industry.
Collapse
Affiliation(s)
- Shanli Zhu
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, China
- College of Agriculture, Jinhua Polytechnic, Jinhua, China
| | - Junrong Li
- College of Agriculture, Jinhua Polytechnic, Jinhua, China
| | - Ziqing Li
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, China
| | - Zhe Wang
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, China
| | - Quanwei Wei
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, China
| | - Fangxiong Shi
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, China
| |
Collapse
|
4
|
Galmiche M, Déchelotte P. Rôle de l’axe microbiote-intestin-cerveau dans la dérégulation du comportement alimentaire au cours de l’obésité et de l’hyperphagie boulimique : les mécanismes. NUTR CLIN METAB 2023; 37:2S16-2S25. [DOI: 10.1016/s0985-0562(24)00007-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
5
|
Derrigo K, LaFata EM. Examining the proportions of food addiction among women with and without polycystic ovarian syndrome who do and do not take hormonal birth control. Eat Behav 2023; 51:101824. [PMID: 37950975 DOI: 10.1016/j.eatbeh.2023.101824] [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: 04/10/2023] [Revised: 10/31/2023] [Accepted: 10/31/2023] [Indexed: 11/13/2023]
Abstract
Polycystic ovarian syndrome (PCOS) is a reproductive disorder driven by insulin resistance. Insulin resistance may increase weight gain and increase the rewarding intake of ultra-processed foods (UPFs). Individuals with PCOS may be more susceptible to the reinforcing properties of UPFs, increasing the risk to consume UPFs in addictive-like ways, operationalized by food addiction (FA). Additionally, hormonal birth control, commonly prescribed to women with PCOS, are found to increase food cravings and overeating. This study examined the relationships between PCOS status, FA, and hormonal birth control use. The study sample (N = 365, assigned female at birth) was drawn from Amazon Mechanical Turk, with half of the sample (n = 181) reporting having PCOS and the other half not (n = 184). Participants answered questions about women's reproductive health (i.e., PCOS, hormonal birth control use) and completed the modified Yale Food Addiction Scale 2.0 (mYFAS 2.0). A chi-square test found that 51.9 % of participants with PCOS (M = 6.23, SD = 3.82) met diagnostic criteria for FA (assessed by the mYFAS 2.0) compared to 16.8 % of participants without PCOS (M = 2.47, SD = 3.39). A hierarchical linear regression found independent main effects of PCOS status (β = 0.40, t(352) = 8.61, p < .001) and hormonal birth control use (β = 0.16, t(351) = 3.59, p < .001), to be associated with higher mYFAS symptom count scores. No differences were found between the types of hormonal birth control participants reported taking. The finding suggests that FA is an overlooked, understudied psychological condition impacting these individuals in weight loss treatments. Future studies are needed to understand the relationship between FA and PCOS in clinical samples.
Collapse
Affiliation(s)
- Karly Derrigo
- Department of Psychology, Drexel University, Philadelphia, PA, United States.
| | - Erica M LaFata
- Department of Psychology, Drexel University, Philadelphia, PA, United States.
| |
Collapse
|
6
|
Sakazaki M, Yoshikawa Y, Kamemoto K, Tataka Y, Yamada Y, Wu CL, Miyashita M. Effects of pre-exercise high and low glycaemic index meals on substrate metabolism and appetite in middle-aged women. J Nutr Sci 2023; 12:e114. [PMID: 38025305 PMCID: PMC10660074 DOI: 10.1017/jns.2023.96] [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: 09/13/2023] [Accepted: 10/13/2023] [Indexed: 12/01/2023] Open
Abstract
Few studies have examined the influence of pre-exercise meals with different glycaemic indices (GIs) on substrate oxidation and non-homeostatic appetite (i.e. food reward) in adults of various ages and ethnicities. We aimed to examine the effects of pre-exercise high and low GI meals on substrate oxidation and food reward in middle-aged Japanese women. This randomised crossover trial included fifteen middle-aged women (aged 40⋅9 ± 6⋅5 years, mean ± sd). The participants consumed a high or low GI breakfast at 09.00 and rested until 11.00. Thereafter, participants performed a 60-min walk at 50 % of their estimated maximum oxygen uptake (11.00-12.00) and rested until 13.00. Expired gas samples were collected every 30 min prior to walking, and samples were collected continuously throughout the walking and post-walking periods. Blood samples and subjective appetite ratings were collected every 30 min, except during walking. The Leeds Food Preference Questionnaire in Japanese (LFPQ-J) was used to assess food reward at 09.00, 10.00, and 13.00 h. The cumulative fat oxidation during exercise was higher in the low GI trial than in the high GI trial (P = 0⋅03). The cumulative carbohydrate oxidation during walking was lower in the low GI trial than in the high GI trial (P = 0⋅01). Trial-by-time interactions were not found for any food-reward parameters between trials. Low GI meals elicited enhanced fat oxidation during a subsequent 60-min walk in middle-aged women. However, meals with different GIs did not affect food reward evaluated over time in the present study.
Collapse
Affiliation(s)
- Miki Sakazaki
- Graduate School of Sport Sciences, Waseda University, 2-579-15 Mikajima, Tokorozawa, Saitama 359-1192, Japan
| | - Yoshie Yoshikawa
- Graduate School of Sport Sciences, Waseda University, 2-579-15 Mikajima, Tokorozawa, Saitama 359-1192, Japan
| | - Kayoko Kamemoto
- Waseda Institute for Sport Science, Waseda University, 2-579-15 Mikajima, Tokorozawa, Saitama 359-1192, Japan
| | - Yusei Tataka
- Graduate School of Sport Sciences, Waseda University, 2-579-15 Mikajima, Tokorozawa, Saitama 359-1192, Japan
| | - Yoshiki Yamada
- Graduate School of Sport Sciences, Waseda University, 2-579-15 Mikajima, Tokorozawa, Saitama 359-1192, Japan
| | - Ching-Lin Wu
- Graduate Institute of Sports and Health Management, National Chung Hsing University, Taichung 402202, Taiwan
| | - Masashi Miyashita
- Faculty of Sport Sciences, Waseda University, 2-579-15 Mikajima, Tokorozawa, Saitama 359-1192, Japan
- School of Sport, Exercise and Health Sciences, Loughborough University, Loughborough, Leicestershire LE11 3TU, UK
- Department of Sports Science and Physical Education, The Chinese University of Hong Kong, Shatin, Hong Kong
| |
Collapse
|
7
|
Tezenas du Montcel C, Duriez P, Cao J, Lebrun N, Ramoz N, Viltart O, Gorwood P, Tolle V. The role of dysregulated ghrelin/LEAP-2 balance in anorexia nervosa. iScience 2023; 26:107996. [PMID: 37867951 PMCID: PMC10587521 DOI: 10.1016/j.isci.2023.107996] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2023] [Revised: 08/24/2023] [Accepted: 09/18/2023] [Indexed: 10/24/2023] Open
Abstract
LEAP-2 is a ghrelin antagonist with an anorexigenic drive. This study investigates the evolution of plasma ghrelin and LEAP-2 concentrations in 29 patients with anorexia nervosa (AN) before and after refeeding and compares it to physiological adaptations during fasting in healthy controls or to mouse model of chronic food restriction and refeeding. Acute and chronic food restriction decrease LEAP-2 and increase ghrelin concentrations in both humans and mice, while patients with AN displayed higher ghrelin and LEAP-2 concentrations before than after refeeding (p = 0.043). After 6 months follow-up, patients with unstable weight gain (n = 17) had significantly decreased LEAP-2 concentrations after refeeding (p = 0.044), in contrast to patients with stable weight gain (n = 12). We provide evidence that the ghrelin/LEAP-2 system is not regulated according to the nutritional status in AN, in contrast to what is physiologically expected when coping with food restriction.
Collapse
Affiliation(s)
- Chloé Tezenas du Montcel
- Université Paris Cité, UMR-S 1266 INSERM, Institut de Psychiatrie et Neuroscience de Paris (IPNP), 75014 Paris, France
- Clinique des Maladies Mentales et de l’Encéphale, GHU Paris Psychiatrie et Neurosciences, Hôpital Sainte-Anne, 75014 Paris, France
| | - Philibert Duriez
- Université Paris Cité, UMR-S 1266 INSERM, Institut de Psychiatrie et Neuroscience de Paris (IPNP), 75014 Paris, France
- Clinique des Maladies Mentales et de l’Encéphale, GHU Paris Psychiatrie et Neurosciences, Hôpital Sainte-Anne, 75014 Paris, France
| | - Jingxian Cao
- Université Paris Cité, UMR-S 1266 INSERM, Institut de Psychiatrie et Neuroscience de Paris (IPNP), 75014 Paris, France
| | - Nicolas Lebrun
- Université Paris Cité, UMR-S 1266 INSERM, Institut de Psychiatrie et Neuroscience de Paris (IPNP), 75014 Paris, France
| | - Nicolas Ramoz
- Université Paris Cité, UMR-S 1266 INSERM, Institut de Psychiatrie et Neuroscience de Paris (IPNP), 75014 Paris, France
| | - Odile Viltart
- Université Paris Cité, UMR-S 1266 INSERM, Institut de Psychiatrie et Neuroscience de Paris (IPNP), 75014 Paris, France
- Université de Lille, SCALab - Sciences Cognitives et Sciences Affectives, UMR CNRS 9193, PsySEF département, 59653 Lille, France
| | - Philip Gorwood
- Université Paris Cité, UMR-S 1266 INSERM, Institut de Psychiatrie et Neuroscience de Paris (IPNP), 75014 Paris, France
- Clinique des Maladies Mentales et de l’Encéphale, GHU Paris Psychiatrie et Neurosciences, Hôpital Sainte-Anne, 75014 Paris, France
| | - Virginie Tolle
- Université Paris Cité, UMR-S 1266 INSERM, Institut de Psychiatrie et Neuroscience de Paris (IPNP), 75014 Paris, France
| |
Collapse
|
8
|
Iosif CI, Bashir ZI, Apps R, Pickford J. Cerebellar Prediction and Feeding Behaviour. CEREBELLUM (LONDON, ENGLAND) 2023; 22:1002-1019. [PMID: 36121552 PMCID: PMC10485105 DOI: 10.1007/s12311-022-01476-3] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 09/01/2022] [Indexed: 06/15/2023]
Abstract
Given the importance of the cerebellum in controlling movements, it might be expected that its main role in eating would be the control of motor elements such as chewing and swallowing. Whilst such functions are clearly important, there is more to eating than these actions, and more to the cerebellum than motor control. This review will present evidence that the cerebellum contributes to homeostatic, motor, rewarding and affective aspects of food consumption.Prediction and feedback underlie many elements of eating, as food consumption is influenced by expectation. For example, circadian clocks cause hunger in anticipation of a meal, and food consumption causes feedback signals which induce satiety. Similarly, the sight and smell of food generate an expectation of what that food will taste like, and its actual taste will generate an internal reward value which will be compared to that expectation. Cerebellar learning is widely thought to involve feed-forward predictions to compare expected outcomes to sensory feedback. We therefore propose that the overarching role of the cerebellum in eating is to respond to prediction errors arising across the homeostatic, motor, cognitive, and affective domains.
Collapse
Affiliation(s)
- Cristiana I Iosif
- School of Physiology, Pharmacology and Neuroscience, University of Bristol, Biomedical Sciences Building, University Walk, Bristol, BS8 1TD, UK.
| | - Zafar I Bashir
- School of Physiology, Pharmacology and Neuroscience, University of Bristol, Biomedical Sciences Building, University Walk, Bristol, BS8 1TD, UK
| | - Richard Apps
- School of Physiology, Pharmacology and Neuroscience, University of Bristol, Biomedical Sciences Building, University Walk, Bristol, BS8 1TD, UK
| | - Jasmine Pickford
- School of Physiology, Pharmacology and Neuroscience, University of Bristol, Biomedical Sciences Building, University Walk, Bristol, BS8 1TD, UK
| |
Collapse
|
9
|
Heberden C, Maximin E, Rabot S, Naudon L. Male mice engaging differently in emotional eating present distinct plasmatic and neurological profiles. Nutr Neurosci 2023; 26:1034-1044. [PMID: 36154930 DOI: 10.1080/1028415x.2022.2122137] [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: 10/14/2022]
Abstract
Objective: Stressed individuals tend to turn to calorie-rich food, also known as 'comfort food' for the temporary relief it provides. The emotional eating drive is highly variable among subjects. Using a rodent model, we explored the plasmatic and neurobiological differences between 'high and low emotional eaters' (HEE and LEE).Methods: 40 male mice were exposed for 5 weeks to a protocol of unpredictable chronic mild stress. Every 3 or 4 days, they were submitted to a 1-h restraint stress, immediately followed by a 3-h period during which a choice between chow and chocolate sweet cereals was proposed. The dietary intake was measured by weighing. Plasmatic and neurobiological characteristics were compared in mice displaying high vs low intakes.Results: Out of 40 mice, 8 were considered as HEE because of their high post-stress eating score, and 8 as LEE because of their consistent low intake. LEE displayed higher plasma corticosterone and lower levels of NPY than HEE, but acylated and total ghrelin were similar in both groups. In the brain, the abundance of NPY neurons in the arcuate nucleus of the hypothalamus was similar in both groups, but was higher in the ventral hippocampus and the basal lateral amygdala of LEE. The lateral hypothalamus LEE had also more orexin (OX) positive neurons. Both NPY and OX are orexigenic peptides and mood regulators.Discussion: Emotional eating difference was reflected in plasma and brain structures implicated in emotion and eating regulation. These results concur with the psychological side of food consumption.
Collapse
Affiliation(s)
- Christine Heberden
- INRAE, AgroParisTech, Micalis Institute, Université Paris-Saclay Jouy-en-Josas, France
| | - Elise Maximin
- INRAE, AgroParisTech, Micalis Institute, Université Paris-Saclay Jouy-en-Josas, France
| | - Sylvie Rabot
- INRAE, AgroParisTech, Micalis Institute, Université Paris-Saclay Jouy-en-Josas, France
| | - Laurent Naudon
- INRAE, AgroParisTech, CNRS, Micalis Institute, Université Paris-Saclay Jouy-en-Josas, France
| |
Collapse
|
10
|
López-Méndez I, Maldonado-Rojas ADC, Uribe M, Juárez-Hernández E. Hunger & satiety signals: another key mechanism involved in the NAFLD pathway. Front Endocrinol (Lausanne) 2023; 14:1213372. [PMID: 37753211 PMCID: PMC10518611 DOI: 10.3389/fendo.2023.1213372] [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: 06/09/2023] [Accepted: 08/28/2023] [Indexed: 09/28/2023] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD) is a highly prevalent metabolic disease, although prevalence could change according to region, nowadays is considered a public health problem whose real impact on the health system is unknown. NAFLD has a multifactorial and complex pathophysiology, due to this, developing a unique and effective pharmacological treatment has not been successful in reverting or avoiding the progression of this liver disease. Even though NAFLD pathophysiology is known, all actual treatments are focused on modifying or regulating the metabolic pathways, some of which interplay with obesity. It has been known that impairments in hunger and satiety signals are associated with obesity, however, abnormalities in these signals in patients with NAFLD and obesity are not fully elucidated. To describe these mechanisms opens an additional option as a therapeutic target sharing metabolic pathways with NAFLD, therefore, this review aims to describe the hormones and peptides implicated in both hunger-satiety in NAFLD. It has been established that NAFLD pharmacological treatment cannot be focused on a single purpose; hence, identifying interplays that lead to adding or modifying current treatment options could also have an impact on another related outcome such as hunger or satiety signals.
Collapse
Affiliation(s)
- Iván López-Méndez
- Hepatology and Transplants Unit, Medica Sur Clinic & Foundation, Mexico City, Mexico
| | | | - Misael Uribe
- Gastroenterology and Obesity Unit, Medica Sur Clinic & Foundation, Mexico City, Mexico
| | - Eva Juárez-Hernández
- Translational Research Unit, Medica Sur Clinic & Foundation, Mexico City, Mexico
| |
Collapse
|
11
|
Deschaine SL, Hedegaard MA, Pince CL, Farokhnia M, Moose JE, Stock IA, Adusumalli S, Akhlaghi F, Hougland JL, Sulima A, Rice KC, Koob GF, Vendruscolo LF, Holst B, Leggio L. Initial Pharmacological Characterization of a Major Hydroxy Metabolite of PF-5190457: Inverse Agonist Activity of PF-6870961 at the Ghrelin Receptor. J Pharmacol Exp Ther 2023; 386:117-128. [PMID: 36631279 PMCID: PMC10353127 DOI: 10.1124/jpet.122.001393] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2022] [Revised: 11/21/2022] [Accepted: 12/09/2022] [Indexed: 01/13/2023] Open
Abstract
Preclinical and clinical studies have identified the ghrelin receptor [growth hormone secretagogue receptor (GHSR)1a] as a potential target for treating alcohol use disorder. A recent phase 1a clinical trial of a GHSR1a antagonist/inverse agonist, PF-5190457, in individuals with heavy alcohol drinking identified a previously undetected major hydroxy metabolite of PF-5190457, namely PF-6870961. Here, we further characterized PF-6870961 by screening for off-target interactions in a high-throughput screen and determined its in vitro pharmacodynamic profile at GHSR1a through binding and concentration-response assays. Moreover, we determined whether the metabolite demonstrated an in vivo effect by assessing effects on food intake in male and female rats. We found that PF-6870961 had no off-target interactions and demonstrated both binding affinity and inverse agonist activity at GHSR1a. In comparison with its parent compound, PF-5190457, the metabolite PF-6870961 had lower binding affinity and potency at inhibiting GHSR1a-induced inositol phosphate accumulation. However, PF-6870961 had increased inhibitory potency at GHSR1a-induced β-arrestin recruitment relative to its parent compound. Intraperitoneal injection of PF-6870961 suppressed food intake under conditions of both food restriction and with ad libitum access to food in male and female rats, demonstrating in vivo activity. The effects of PF-6870961 on food intake were abolished in male and female rats knockout for GHSR, thus demonstrating that its effects on food intake are in fact mediated by the GHSR receptor. Our findings indicate that the newly discovered major hydroxy metabolite of PF-5190457 may contribute to the overall activity of PF-5190457 by demonstrating inhibitory activity at GHSR1a. SIGNIFICANCE STATEMENT: Antagonists or inverse agonists of the growth hormone secretagogue receptor (GHSR)1a have demonstrated substantial potential as therapeutics for alcohol use disorder. We here expand understanding of the pharmacology of one such GHSR1a inverse agonist, PF-5190457, by studying the safety and pharmacodynamics of its major hydroxy metabolite, PF-6870961. Our data demonstrate biased inverse agonism of PF-6870961 at GHSR1a and provide new structure-activity relationship insight into GHSR1a inverse agonism.
Collapse
Affiliation(s)
- Sara L Deschaine
- Clinical Psychoneuroendocrinology and Neuropsychopharmacology Section, Translational Addiction Medicine Branch, National Institute on Drug Abuse Intramural Research Program and National Institute on Alcohol Abuse and Alcoholism Division of Intramural Clinical and Biological Research, National Institutes of Health, Baltimore and Bethesda, Maryland (S.L.D., C.L.P., M.F., L.L.); Department of Pharmacology, University of Michigan Medical School, Ann Arbor, Michigan (S.L.D.); Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark (M.A.H., B.H.); Neurobiology of Addiction Section, Integrative Neuroscience Research Branch, National Institute on Drug Abuse (C.L.P., G.F.K., L.F.V.) and Medication Development Program, National Institute on Drug Abuse Intramural Research Program (A.S., K.C.R., L.L.), National Institutes of Health, Baltimore, Maryland; Department of Mental Health, Johns Hopkins Bloomberg School of Public Health (M.F.) and Division of Addiction Medicine, Department of Medicine, School of Medicine (L.L.), Johns Hopkins University, Baltimore, Maryland; Department of Chemistry (J.E.M., J.L.H.), Department of Biology (J.L.H.), and BioInspired Syracuse (J.L.H.), Syracuse University, Syracuse, New York; Pfizer Inc. Medicine Design, Groton, Connecticut (I.A.S.); Clinical Pharmacokinetics Research Laboratory, Department of Biomedical and Pharmaceutical Sciences, University of Rhode Island, Kingston, Rhode Island (S.A., F.A.); Drug Design and Synthesis Section, Molecular Targets and Medications Discovery Branch, Intramural Research Program, National Institute on Drug Abuse and National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, Maryland (A.S., K.C.R.); and Center for Alcohol Addiction Studies, Department of Behavioral and Social Sciences, School of Public Health, Brown University, Providence, Rhode Island (L.L.)
| | - Morten A Hedegaard
- Clinical Psychoneuroendocrinology and Neuropsychopharmacology Section, Translational Addiction Medicine Branch, National Institute on Drug Abuse Intramural Research Program and National Institute on Alcohol Abuse and Alcoholism Division of Intramural Clinical and Biological Research, National Institutes of Health, Baltimore and Bethesda, Maryland (S.L.D., C.L.P., M.F., L.L.); Department of Pharmacology, University of Michigan Medical School, Ann Arbor, Michigan (S.L.D.); Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark (M.A.H., B.H.); Neurobiology of Addiction Section, Integrative Neuroscience Research Branch, National Institute on Drug Abuse (C.L.P., G.F.K., L.F.V.) and Medication Development Program, National Institute on Drug Abuse Intramural Research Program (A.S., K.C.R., L.L.), National Institutes of Health, Baltimore, Maryland; Department of Mental Health, Johns Hopkins Bloomberg School of Public Health (M.F.) and Division of Addiction Medicine, Department of Medicine, School of Medicine (L.L.), Johns Hopkins University, Baltimore, Maryland; Department of Chemistry (J.E.M., J.L.H.), Department of Biology (J.L.H.), and BioInspired Syracuse (J.L.H.), Syracuse University, Syracuse, New York; Pfizer Inc. Medicine Design, Groton, Connecticut (I.A.S.); Clinical Pharmacokinetics Research Laboratory, Department of Biomedical and Pharmaceutical Sciences, University of Rhode Island, Kingston, Rhode Island (S.A., F.A.); Drug Design and Synthesis Section, Molecular Targets and Medications Discovery Branch, Intramural Research Program, National Institute on Drug Abuse and National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, Maryland (A.S., K.C.R.); and Center for Alcohol Addiction Studies, Department of Behavioral and Social Sciences, School of Public Health, Brown University, Providence, Rhode Island (L.L.)
| | - Claire L Pince
- Clinical Psychoneuroendocrinology and Neuropsychopharmacology Section, Translational Addiction Medicine Branch, National Institute on Drug Abuse Intramural Research Program and National Institute on Alcohol Abuse and Alcoholism Division of Intramural Clinical and Biological Research, National Institutes of Health, Baltimore and Bethesda, Maryland (S.L.D., C.L.P., M.F., L.L.); Department of Pharmacology, University of Michigan Medical School, Ann Arbor, Michigan (S.L.D.); Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark (M.A.H., B.H.); Neurobiology of Addiction Section, Integrative Neuroscience Research Branch, National Institute on Drug Abuse (C.L.P., G.F.K., L.F.V.) and Medication Development Program, National Institute on Drug Abuse Intramural Research Program (A.S., K.C.R., L.L.), National Institutes of Health, Baltimore, Maryland; Department of Mental Health, Johns Hopkins Bloomberg School of Public Health (M.F.) and Division of Addiction Medicine, Department of Medicine, School of Medicine (L.L.), Johns Hopkins University, Baltimore, Maryland; Department of Chemistry (J.E.M., J.L.H.), Department of Biology (J.L.H.), and BioInspired Syracuse (J.L.H.), Syracuse University, Syracuse, New York; Pfizer Inc. Medicine Design, Groton, Connecticut (I.A.S.); Clinical Pharmacokinetics Research Laboratory, Department of Biomedical and Pharmaceutical Sciences, University of Rhode Island, Kingston, Rhode Island (S.A., F.A.); Drug Design and Synthesis Section, Molecular Targets and Medications Discovery Branch, Intramural Research Program, National Institute on Drug Abuse and National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, Maryland (A.S., K.C.R.); and Center for Alcohol Addiction Studies, Department of Behavioral and Social Sciences, School of Public Health, Brown University, Providence, Rhode Island (L.L.)
| | - Mehdi Farokhnia
- Clinical Psychoneuroendocrinology and Neuropsychopharmacology Section, Translational Addiction Medicine Branch, National Institute on Drug Abuse Intramural Research Program and National Institute on Alcohol Abuse and Alcoholism Division of Intramural Clinical and Biological Research, National Institutes of Health, Baltimore and Bethesda, Maryland (S.L.D., C.L.P., M.F., L.L.); Department of Pharmacology, University of Michigan Medical School, Ann Arbor, Michigan (S.L.D.); Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark (M.A.H., B.H.); Neurobiology of Addiction Section, Integrative Neuroscience Research Branch, National Institute on Drug Abuse (C.L.P., G.F.K., L.F.V.) and Medication Development Program, National Institute on Drug Abuse Intramural Research Program (A.S., K.C.R., L.L.), National Institutes of Health, Baltimore, Maryland; Department of Mental Health, Johns Hopkins Bloomberg School of Public Health (M.F.) and Division of Addiction Medicine, Department of Medicine, School of Medicine (L.L.), Johns Hopkins University, Baltimore, Maryland; Department of Chemistry (J.E.M., J.L.H.), Department of Biology (J.L.H.), and BioInspired Syracuse (J.L.H.), Syracuse University, Syracuse, New York; Pfizer Inc. Medicine Design, Groton, Connecticut (I.A.S.); Clinical Pharmacokinetics Research Laboratory, Department of Biomedical and Pharmaceutical Sciences, University of Rhode Island, Kingston, Rhode Island (S.A., F.A.); Drug Design and Synthesis Section, Molecular Targets and Medications Discovery Branch, Intramural Research Program, National Institute on Drug Abuse and National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, Maryland (A.S., K.C.R.); and Center for Alcohol Addiction Studies, Department of Behavioral and Social Sciences, School of Public Health, Brown University, Providence, Rhode Island (L.L.)
| | - Jacob E Moose
- Clinical Psychoneuroendocrinology and Neuropsychopharmacology Section, Translational Addiction Medicine Branch, National Institute on Drug Abuse Intramural Research Program and National Institute on Alcohol Abuse and Alcoholism Division of Intramural Clinical and Biological Research, National Institutes of Health, Baltimore and Bethesda, Maryland (S.L.D., C.L.P., M.F., L.L.); Department of Pharmacology, University of Michigan Medical School, Ann Arbor, Michigan (S.L.D.); Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark (M.A.H., B.H.); Neurobiology of Addiction Section, Integrative Neuroscience Research Branch, National Institute on Drug Abuse (C.L.P., G.F.K., L.F.V.) and Medication Development Program, National Institute on Drug Abuse Intramural Research Program (A.S., K.C.R., L.L.), National Institutes of Health, Baltimore, Maryland; Department of Mental Health, Johns Hopkins Bloomberg School of Public Health (M.F.) and Division of Addiction Medicine, Department of Medicine, School of Medicine (L.L.), Johns Hopkins University, Baltimore, Maryland; Department of Chemistry (J.E.M., J.L.H.), Department of Biology (J.L.H.), and BioInspired Syracuse (J.L.H.), Syracuse University, Syracuse, New York; Pfizer Inc. Medicine Design, Groton, Connecticut (I.A.S.); Clinical Pharmacokinetics Research Laboratory, Department of Biomedical and Pharmaceutical Sciences, University of Rhode Island, Kingston, Rhode Island (S.A., F.A.); Drug Design and Synthesis Section, Molecular Targets and Medications Discovery Branch, Intramural Research Program, National Institute on Drug Abuse and National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, Maryland (A.S., K.C.R.); and Center for Alcohol Addiction Studies, Department of Behavioral and Social Sciences, School of Public Health, Brown University, Providence, Rhode Island (L.L.)
| | - Ingrid A Stock
- Clinical Psychoneuroendocrinology and Neuropsychopharmacology Section, Translational Addiction Medicine Branch, National Institute on Drug Abuse Intramural Research Program and National Institute on Alcohol Abuse and Alcoholism Division of Intramural Clinical and Biological Research, National Institutes of Health, Baltimore and Bethesda, Maryland (S.L.D., C.L.P., M.F., L.L.); Department of Pharmacology, University of Michigan Medical School, Ann Arbor, Michigan (S.L.D.); Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark (M.A.H., B.H.); Neurobiology of Addiction Section, Integrative Neuroscience Research Branch, National Institute on Drug Abuse (C.L.P., G.F.K., L.F.V.) and Medication Development Program, National Institute on Drug Abuse Intramural Research Program (A.S., K.C.R., L.L.), National Institutes of Health, Baltimore, Maryland; Department of Mental Health, Johns Hopkins Bloomberg School of Public Health (M.F.) and Division of Addiction Medicine, Department of Medicine, School of Medicine (L.L.), Johns Hopkins University, Baltimore, Maryland; Department of Chemistry (J.E.M., J.L.H.), Department of Biology (J.L.H.), and BioInspired Syracuse (J.L.H.), Syracuse University, Syracuse, New York; Pfizer Inc. Medicine Design, Groton, Connecticut (I.A.S.); Clinical Pharmacokinetics Research Laboratory, Department of Biomedical and Pharmaceutical Sciences, University of Rhode Island, Kingston, Rhode Island (S.A., F.A.); Drug Design and Synthesis Section, Molecular Targets and Medications Discovery Branch, Intramural Research Program, National Institute on Drug Abuse and National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, Maryland (A.S., K.C.R.); and Center for Alcohol Addiction Studies, Department of Behavioral and Social Sciences, School of Public Health, Brown University, Providence, Rhode Island (L.L.)
| | - Sravani Adusumalli
- Clinical Psychoneuroendocrinology and Neuropsychopharmacology Section, Translational Addiction Medicine Branch, National Institute on Drug Abuse Intramural Research Program and National Institute on Alcohol Abuse and Alcoholism Division of Intramural Clinical and Biological Research, National Institutes of Health, Baltimore and Bethesda, Maryland (S.L.D., C.L.P., M.F., L.L.); Department of Pharmacology, University of Michigan Medical School, Ann Arbor, Michigan (S.L.D.); Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark (M.A.H., B.H.); Neurobiology of Addiction Section, Integrative Neuroscience Research Branch, National Institute on Drug Abuse (C.L.P., G.F.K., L.F.V.) and Medication Development Program, National Institute on Drug Abuse Intramural Research Program (A.S., K.C.R., L.L.), National Institutes of Health, Baltimore, Maryland; Department of Mental Health, Johns Hopkins Bloomberg School of Public Health (M.F.) and Division of Addiction Medicine, Department of Medicine, School of Medicine (L.L.), Johns Hopkins University, Baltimore, Maryland; Department of Chemistry (J.E.M., J.L.H.), Department of Biology (J.L.H.), and BioInspired Syracuse (J.L.H.), Syracuse University, Syracuse, New York; Pfizer Inc. Medicine Design, Groton, Connecticut (I.A.S.); Clinical Pharmacokinetics Research Laboratory, Department of Biomedical and Pharmaceutical Sciences, University of Rhode Island, Kingston, Rhode Island (S.A., F.A.); Drug Design and Synthesis Section, Molecular Targets and Medications Discovery Branch, Intramural Research Program, National Institute on Drug Abuse and National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, Maryland (A.S., K.C.R.); and Center for Alcohol Addiction Studies, Department of Behavioral and Social Sciences, School of Public Health, Brown University, Providence, Rhode Island (L.L.)
| | - Fatemeh Akhlaghi
- Clinical Psychoneuroendocrinology and Neuropsychopharmacology Section, Translational Addiction Medicine Branch, National Institute on Drug Abuse Intramural Research Program and National Institute on Alcohol Abuse and Alcoholism Division of Intramural Clinical and Biological Research, National Institutes of Health, Baltimore and Bethesda, Maryland (S.L.D., C.L.P., M.F., L.L.); Department of Pharmacology, University of Michigan Medical School, Ann Arbor, Michigan (S.L.D.); Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark (M.A.H., B.H.); Neurobiology of Addiction Section, Integrative Neuroscience Research Branch, National Institute on Drug Abuse (C.L.P., G.F.K., L.F.V.) and Medication Development Program, National Institute on Drug Abuse Intramural Research Program (A.S., K.C.R., L.L.), National Institutes of Health, Baltimore, Maryland; Department of Mental Health, Johns Hopkins Bloomberg School of Public Health (M.F.) and Division of Addiction Medicine, Department of Medicine, School of Medicine (L.L.), Johns Hopkins University, Baltimore, Maryland; Department of Chemistry (J.E.M., J.L.H.), Department of Biology (J.L.H.), and BioInspired Syracuse (J.L.H.), Syracuse University, Syracuse, New York; Pfizer Inc. Medicine Design, Groton, Connecticut (I.A.S.); Clinical Pharmacokinetics Research Laboratory, Department of Biomedical and Pharmaceutical Sciences, University of Rhode Island, Kingston, Rhode Island (S.A., F.A.); Drug Design and Synthesis Section, Molecular Targets and Medications Discovery Branch, Intramural Research Program, National Institute on Drug Abuse and National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, Maryland (A.S., K.C.R.); and Center for Alcohol Addiction Studies, Department of Behavioral and Social Sciences, School of Public Health, Brown University, Providence, Rhode Island (L.L.)
| | - James L Hougland
- Clinical Psychoneuroendocrinology and Neuropsychopharmacology Section, Translational Addiction Medicine Branch, National Institute on Drug Abuse Intramural Research Program and National Institute on Alcohol Abuse and Alcoholism Division of Intramural Clinical and Biological Research, National Institutes of Health, Baltimore and Bethesda, Maryland (S.L.D., C.L.P., M.F., L.L.); Department of Pharmacology, University of Michigan Medical School, Ann Arbor, Michigan (S.L.D.); Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark (M.A.H., B.H.); Neurobiology of Addiction Section, Integrative Neuroscience Research Branch, National Institute on Drug Abuse (C.L.P., G.F.K., L.F.V.) and Medication Development Program, National Institute on Drug Abuse Intramural Research Program (A.S., K.C.R., L.L.), National Institutes of Health, Baltimore, Maryland; Department of Mental Health, Johns Hopkins Bloomberg School of Public Health (M.F.) and Division of Addiction Medicine, Department of Medicine, School of Medicine (L.L.), Johns Hopkins University, Baltimore, Maryland; Department of Chemistry (J.E.M., J.L.H.), Department of Biology (J.L.H.), and BioInspired Syracuse (J.L.H.), Syracuse University, Syracuse, New York; Pfizer Inc. Medicine Design, Groton, Connecticut (I.A.S.); Clinical Pharmacokinetics Research Laboratory, Department of Biomedical and Pharmaceutical Sciences, University of Rhode Island, Kingston, Rhode Island (S.A., F.A.); Drug Design and Synthesis Section, Molecular Targets and Medications Discovery Branch, Intramural Research Program, National Institute on Drug Abuse and National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, Maryland (A.S., K.C.R.); and Center for Alcohol Addiction Studies, Department of Behavioral and Social Sciences, School of Public Health, Brown University, Providence, Rhode Island (L.L.)
| | - Agnieszka Sulima
- Clinical Psychoneuroendocrinology and Neuropsychopharmacology Section, Translational Addiction Medicine Branch, National Institute on Drug Abuse Intramural Research Program and National Institute on Alcohol Abuse and Alcoholism Division of Intramural Clinical and Biological Research, National Institutes of Health, Baltimore and Bethesda, Maryland (S.L.D., C.L.P., M.F., L.L.); Department of Pharmacology, University of Michigan Medical School, Ann Arbor, Michigan (S.L.D.); Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark (M.A.H., B.H.); Neurobiology of Addiction Section, Integrative Neuroscience Research Branch, National Institute on Drug Abuse (C.L.P., G.F.K., L.F.V.) and Medication Development Program, National Institute on Drug Abuse Intramural Research Program (A.S., K.C.R., L.L.), National Institutes of Health, Baltimore, Maryland; Department of Mental Health, Johns Hopkins Bloomberg School of Public Health (M.F.) and Division of Addiction Medicine, Department of Medicine, School of Medicine (L.L.), Johns Hopkins University, Baltimore, Maryland; Department of Chemistry (J.E.M., J.L.H.), Department of Biology (J.L.H.), and BioInspired Syracuse (J.L.H.), Syracuse University, Syracuse, New York; Pfizer Inc. Medicine Design, Groton, Connecticut (I.A.S.); Clinical Pharmacokinetics Research Laboratory, Department of Biomedical and Pharmaceutical Sciences, University of Rhode Island, Kingston, Rhode Island (S.A., F.A.); Drug Design and Synthesis Section, Molecular Targets and Medications Discovery Branch, Intramural Research Program, National Institute on Drug Abuse and National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, Maryland (A.S., K.C.R.); and Center for Alcohol Addiction Studies, Department of Behavioral and Social Sciences, School of Public Health, Brown University, Providence, Rhode Island (L.L.)
| | - Kenner C Rice
- Clinical Psychoneuroendocrinology and Neuropsychopharmacology Section, Translational Addiction Medicine Branch, National Institute on Drug Abuse Intramural Research Program and National Institute on Alcohol Abuse and Alcoholism Division of Intramural Clinical and Biological Research, National Institutes of Health, Baltimore and Bethesda, Maryland (S.L.D., C.L.P., M.F., L.L.); Department of Pharmacology, University of Michigan Medical School, Ann Arbor, Michigan (S.L.D.); Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark (M.A.H., B.H.); Neurobiology of Addiction Section, Integrative Neuroscience Research Branch, National Institute on Drug Abuse (C.L.P., G.F.K., L.F.V.) and Medication Development Program, National Institute on Drug Abuse Intramural Research Program (A.S., K.C.R., L.L.), National Institutes of Health, Baltimore, Maryland; Department of Mental Health, Johns Hopkins Bloomberg School of Public Health (M.F.) and Division of Addiction Medicine, Department of Medicine, School of Medicine (L.L.), Johns Hopkins University, Baltimore, Maryland; Department of Chemistry (J.E.M., J.L.H.), Department of Biology (J.L.H.), and BioInspired Syracuse (J.L.H.), Syracuse University, Syracuse, New York; Pfizer Inc. Medicine Design, Groton, Connecticut (I.A.S.); Clinical Pharmacokinetics Research Laboratory, Department of Biomedical and Pharmaceutical Sciences, University of Rhode Island, Kingston, Rhode Island (S.A., F.A.); Drug Design and Synthesis Section, Molecular Targets and Medications Discovery Branch, Intramural Research Program, National Institute on Drug Abuse and National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, Maryland (A.S., K.C.R.); and Center for Alcohol Addiction Studies, Department of Behavioral and Social Sciences, School of Public Health, Brown University, Providence, Rhode Island (L.L.)
| | - George F Koob
- Clinical Psychoneuroendocrinology and Neuropsychopharmacology Section, Translational Addiction Medicine Branch, National Institute on Drug Abuse Intramural Research Program and National Institute on Alcohol Abuse and Alcoholism Division of Intramural Clinical and Biological Research, National Institutes of Health, Baltimore and Bethesda, Maryland (S.L.D., C.L.P., M.F., L.L.); Department of Pharmacology, University of Michigan Medical School, Ann Arbor, Michigan (S.L.D.); Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark (M.A.H., B.H.); Neurobiology of Addiction Section, Integrative Neuroscience Research Branch, National Institute on Drug Abuse (C.L.P., G.F.K., L.F.V.) and Medication Development Program, National Institute on Drug Abuse Intramural Research Program (A.S., K.C.R., L.L.), National Institutes of Health, Baltimore, Maryland; Department of Mental Health, Johns Hopkins Bloomberg School of Public Health (M.F.) and Division of Addiction Medicine, Department of Medicine, School of Medicine (L.L.), Johns Hopkins University, Baltimore, Maryland; Department of Chemistry (J.E.M., J.L.H.), Department of Biology (J.L.H.), and BioInspired Syracuse (J.L.H.), Syracuse University, Syracuse, New York; Pfizer Inc. Medicine Design, Groton, Connecticut (I.A.S.); Clinical Pharmacokinetics Research Laboratory, Department of Biomedical and Pharmaceutical Sciences, University of Rhode Island, Kingston, Rhode Island (S.A., F.A.); Drug Design and Synthesis Section, Molecular Targets and Medications Discovery Branch, Intramural Research Program, National Institute on Drug Abuse and National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, Maryland (A.S., K.C.R.); and Center for Alcohol Addiction Studies, Department of Behavioral and Social Sciences, School of Public Health, Brown University, Providence, Rhode Island (L.L.)
| | - Leandro F Vendruscolo
- Clinical Psychoneuroendocrinology and Neuropsychopharmacology Section, Translational Addiction Medicine Branch, National Institute on Drug Abuse Intramural Research Program and National Institute on Alcohol Abuse and Alcoholism Division of Intramural Clinical and Biological Research, National Institutes of Health, Baltimore and Bethesda, Maryland (S.L.D., C.L.P., M.F., L.L.); Department of Pharmacology, University of Michigan Medical School, Ann Arbor, Michigan (S.L.D.); Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark (M.A.H., B.H.); Neurobiology of Addiction Section, Integrative Neuroscience Research Branch, National Institute on Drug Abuse (C.L.P., G.F.K., L.F.V.) and Medication Development Program, National Institute on Drug Abuse Intramural Research Program (A.S., K.C.R., L.L.), National Institutes of Health, Baltimore, Maryland; Department of Mental Health, Johns Hopkins Bloomberg School of Public Health (M.F.) and Division of Addiction Medicine, Department of Medicine, School of Medicine (L.L.), Johns Hopkins University, Baltimore, Maryland; Department of Chemistry (J.E.M., J.L.H.), Department of Biology (J.L.H.), and BioInspired Syracuse (J.L.H.), Syracuse University, Syracuse, New York; Pfizer Inc. Medicine Design, Groton, Connecticut (I.A.S.); Clinical Pharmacokinetics Research Laboratory, Department of Biomedical and Pharmaceutical Sciences, University of Rhode Island, Kingston, Rhode Island (S.A., F.A.); Drug Design and Synthesis Section, Molecular Targets and Medications Discovery Branch, Intramural Research Program, National Institute on Drug Abuse and National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, Maryland (A.S., K.C.R.); and Center for Alcohol Addiction Studies, Department of Behavioral and Social Sciences, School of Public Health, Brown University, Providence, Rhode Island (L.L.)
| | - Birgitte Holst
- Clinical Psychoneuroendocrinology and Neuropsychopharmacology Section, Translational Addiction Medicine Branch, National Institute on Drug Abuse Intramural Research Program and National Institute on Alcohol Abuse and Alcoholism Division of Intramural Clinical and Biological Research, National Institutes of Health, Baltimore and Bethesda, Maryland (S.L.D., C.L.P., M.F., L.L.); Department of Pharmacology, University of Michigan Medical School, Ann Arbor, Michigan (S.L.D.); Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark (M.A.H., B.H.); Neurobiology of Addiction Section, Integrative Neuroscience Research Branch, National Institute on Drug Abuse (C.L.P., G.F.K., L.F.V.) and Medication Development Program, National Institute on Drug Abuse Intramural Research Program (A.S., K.C.R., L.L.), National Institutes of Health, Baltimore, Maryland; Department of Mental Health, Johns Hopkins Bloomberg School of Public Health (M.F.) and Division of Addiction Medicine, Department of Medicine, School of Medicine (L.L.), Johns Hopkins University, Baltimore, Maryland; Department of Chemistry (J.E.M., J.L.H.), Department of Biology (J.L.H.), and BioInspired Syracuse (J.L.H.), Syracuse University, Syracuse, New York; Pfizer Inc. Medicine Design, Groton, Connecticut (I.A.S.); Clinical Pharmacokinetics Research Laboratory, Department of Biomedical and Pharmaceutical Sciences, University of Rhode Island, Kingston, Rhode Island (S.A., F.A.); Drug Design and Synthesis Section, Molecular Targets and Medications Discovery Branch, Intramural Research Program, National Institute on Drug Abuse and National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, Maryland (A.S., K.C.R.); and Center for Alcohol Addiction Studies, Department of Behavioral and Social Sciences, School of Public Health, Brown University, Providence, Rhode Island (L.L.)
| | - Lorenzo Leggio
- Clinical Psychoneuroendocrinology and Neuropsychopharmacology Section, Translational Addiction Medicine Branch, National Institute on Drug Abuse Intramural Research Program and National Institute on Alcohol Abuse and Alcoholism Division of Intramural Clinical and Biological Research, National Institutes of Health, Baltimore and Bethesda, Maryland (S.L.D., C.L.P., M.F., L.L.); Department of Pharmacology, University of Michigan Medical School, Ann Arbor, Michigan (S.L.D.); Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark (M.A.H., B.H.); Neurobiology of Addiction Section, Integrative Neuroscience Research Branch, National Institute on Drug Abuse (C.L.P., G.F.K., L.F.V.) and Medication Development Program, National Institute on Drug Abuse Intramural Research Program (A.S., K.C.R., L.L.), National Institutes of Health, Baltimore, Maryland; Department of Mental Health, Johns Hopkins Bloomberg School of Public Health (M.F.) and Division of Addiction Medicine, Department of Medicine, School of Medicine (L.L.), Johns Hopkins University, Baltimore, Maryland; Department of Chemistry (J.E.M., J.L.H.), Department of Biology (J.L.H.), and BioInspired Syracuse (J.L.H.), Syracuse University, Syracuse, New York; Pfizer Inc. Medicine Design, Groton, Connecticut (I.A.S.); Clinical Pharmacokinetics Research Laboratory, Department of Biomedical and Pharmaceutical Sciences, University of Rhode Island, Kingston, Rhode Island (S.A., F.A.); Drug Design and Synthesis Section, Molecular Targets and Medications Discovery Branch, Intramural Research Program, National Institute on Drug Abuse and National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, Maryland (A.S., K.C.R.); and Center for Alcohol Addiction Studies, Department of Behavioral and Social Sciences, School of Public Health, Brown University, Providence, Rhode Island (L.L.)
| |
Collapse
|
12
|
Chae Y, Lee IS. Central Regulation of Eating Behaviors in Humans: Evidence from Functional Neuroimaging Studies. Nutrients 2023; 15:3010. [PMID: 37447336 PMCID: PMC10347214 DOI: 10.3390/nu15133010] [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: 06/12/2023] [Revised: 06/27/2023] [Accepted: 06/28/2023] [Indexed: 07/15/2023] Open
Abstract
Neuroimaging has great potential to provide insight into the neural response to food stimuli. Remarkable advances have been made in understanding the neural activity underlying food perception, not only in normal eating but also in obesity, eating disorders, and disorders of gut-brain interaction in recent decades. In addition to the abnormal brain function in patients with eating disorders compared to healthy controls, new therapies, such as neurofeedback and neurostimulation techniques, have been developed that target the malfunctioning brain regions in patients with eating disorders based on the results of neuroimaging studies. In this review, we present an overview of early and more recent research on the central processing and regulation of eating behavior in healthy and patient populations. In order to better understand the relationship between the gut and the brain as well as the neural mechanisms underlying abnormal ingestive behaviors, we also provide suggestions for future directions to enhance our current methods used in food-related neuroimaging studies.
Collapse
Affiliation(s)
- Younbyoung Chae
- College of Korean Medicine, Kyung Hee University, Seoul 02447, Republic of Korea
| | - In-Seon Lee
- College of Korean Medicine, Kyung Hee University, Seoul 02447, Republic of Korea
| |
Collapse
|
13
|
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: 5] [Impact Index Per Article: 2.5] [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
|
14
|
Carrillo-Ruiz JD, Carrillo-Márquez JR, Beltrán JQ, Jiménez-Ponce F, García-Muñoz L, Navarro-Olvera JL, Márquez-Franco R, Velasco F. Innovative perspectives in limbic surgery using deep brain stimulation. Front Neurosci 2023; 17:1167244. [PMID: 37274213 PMCID: PMC10233042 DOI: 10.3389/fnins.2023.1167244] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2023] [Accepted: 04/24/2023] [Indexed: 06/06/2023] Open
Abstract
Limbic surgery is one of the most attractive and retaken fields of functional neurosurgery in the last two decades. Psychiatric surgery emerged from the incipient work of Moniz and Lima lesioning the prefrontal cortex in agitated patients. Since the onset of stereotactic and functional neurosurgery with Spiegel and Wycis, the treatment of mental diseases gave attention to refractory illnesses mainly with the use of thalamotomies. Neurosis and some psychotic symptoms were treated by them. Several indications when lesioning the brain were included: obsessive-compulsive disorder, depression, and aggressiveness among others with a diversity of targets. The indiscriminately use of anatomical sites without enough scientific evidence, and uncertainly defined criteria for selecting patients merged with a deficiency in ethical aspects, brought a lack of procedures for a long time: only select clinics allowed this surgery around the world from 1950 to the 1990s. In 1999, Nuttin et al. began a new chapter in limbic surgery with the use of Deep Brain Stimulation, based on the experience of pain, Parkinson's disease, and epilepsy. The efforts were focused on different targets to treat depression and obsessive-compulsive disorders. Nevertheless, other diseases were added to use neuromodulation. The goal of this article is to show the new opportunities to treat neuropsychiatric diseases.
Collapse
Affiliation(s)
- José Damián Carrillo-Ruiz
- Stereotactic, Functional & Radiosurgery Unit of Neurosurgery Service, Mexico General Hospital, Mexico City, Mexico
- Research Direction, Mexico General Hospital, Mexico City, Mexico
- Neuroscience Coordination, Psychology Faculty, Anahuac University, Mexico City, Mexico
| | - José Rodrigo Carrillo-Márquez
- Faculty of Health Sciences, Anahuac University, Mexico City, Mexico
- Alpha Health Sciences Leadership Program, Anahuac University, Mexico City, Mexico
| | - Jesús Quetzalcóatl Beltrán
- Stereotactic, Functional & Radiosurgery Unit of Neurosurgery Service, Mexico General Hospital, Mexico City, Mexico
| | - Fiacro Jiménez-Ponce
- Stereotactic, Functional & Radiosurgery Unit of Neurosurgery Service, Mexico General Hospital, Mexico City, Mexico
| | - Luis García-Muñoz
- Stereotactic, Functional & Radiosurgery Unit of Neurosurgery Service, Mexico General Hospital, Mexico City, Mexico
| | - José Luis Navarro-Olvera
- Stereotactic, Functional & Radiosurgery Unit of Neurosurgery Service, Mexico General Hospital, Mexico City, Mexico
| | - René Márquez-Franco
- Stereotactic, Functional & Radiosurgery Unit of Neurosurgery Service, Mexico General Hospital, Mexico City, Mexico
| | - Francisco Velasco
- Stereotactic, Functional & Radiosurgery Unit of Neurosurgery Service, Mexico General Hospital, Mexico City, Mexico
| |
Collapse
|
15
|
Montalban E, Giralt A, Taing L, Nakamura Y, Pelosi A, Brown M, de Pins B, Valjent E, Martin M, Nairn AC, Greengard P, Flajolet M, Herv D, Gambardella N, Roussarie JP, Girault JA. Operant training for highly palatable food alters translating mRNA in nucleus accumbens D2 neurons and reveals a modulatory role of Neurochondrin. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.03.07.531496. [PMID: 36945487 PMCID: PMC10028890 DOI: 10.1101/2023.03.07.531496] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/18/2023]
Abstract
BACKGROUND Highly palatable food triggers behavioral alterations reminiscent of those induced by addictive drugs. These effects involve the reward system and dopamine neurons, which modulate neurons in the nucleus accumbens (NAc). The molecular mechanisms underlying the effects of highly palatable food on feeding behavior are poorly understood. METHODS We studied the effects of 2-week operant conditioning of mice with standard or isocaloric highly palatable food. We investigated the behavioral effects and dendritic spine modifications in the NAc. We compared the translating mRNA in NAc neurons identified by the type of dopamine receptors they express, depending on the type of food and training. We tested the consequences of invalidation of an abundant downregulated gene, Ncdn (Neurochondrin). RESULTS Operant conditioning for highly palatable food increases motivation for food even in well-fed mice. In control mice, free access to regular or highly palatable food results in increased weight as compared to regular food only. Highly palatable food increases spine density in the NAc. In animals trained for highly palatable food, translating mRNAs are modified in NAc dopamine D2-receptor-expressing neurons, mostly corresponding to striatal projection neurons, but not in those expressing D1-receptors. Knock-out of Ncdn, an abundant down-regulated gene, opposes the conditioning-induced changes in satiety-sensitive feeding behavior and apparent motivation for highly palatable food, suggesting down-regulation may be a compensatory mechanism. CONCLUSIONS Our results emphasize the importance of mRNA alterations D2 striatal projection neurons in the NAc in the behavioral consequences of highly palatable food conditioning and suggest a modulatory contribution of Ncdn downregulation.
Collapse
|
16
|
Chamorro R, Jouffe C, Oster H, Uhlenhaut NH, Meyhöfer SM. When should I eat: A circadian view on food intake and metabolic regulation. Acta Physiol (Oxf) 2023; 237:e13936. [PMID: 36645134 DOI: 10.1111/apha.13936] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Revised: 12/19/2022] [Accepted: 01/11/2023] [Indexed: 01/17/2023]
Abstract
The circadian clock is a hierarchical timing system regulating most physiological and behavioral functions with a period of approximately 24 h in humans and other mammalian species. The circadian clock drives daily eating rhythms that, in turn, reinforce the circadian clock network itself to anticipate and orchestrate metabolic responses to food intake. Eating is tightly interconnected with the circadian clock and recent evidence shows that the timing of meals is crucial for the control of appetite and metabolic regulation. Obesity results from combined long-term dysregulation in food intake (homeostatic and hedonic circuits), energy expenditure, and energy storage. Increasing evidence supports that the loss of synchrony of daily rhythms significantly impairs metabolic homeostasis and is associated with obesity. This review presents an overview of mechanisms regulating food intake (homeostatic/hedonic) and focuses on the crucial role of the circadian clock on the metabolic response to eating, thus providing a fundamental research axis to maintain a healthy eating behavior.
Collapse
Affiliation(s)
- Rodrigo Chamorro
- Institute for Endocrinology and Diabetes, University of Lübeck, Lübeck, Germany.,Department of Nutrition, Faculty of Medicine, University of Chile, Santiago, Chile
| | - Céline Jouffe
- Institute for Diabetes and Endocrinology, Helmholtz Diabetes Center, Helmholtz Zentrum München, Neuherberg, Germany.,Institute for Diabetes and Cancer, Helmholtz Diabetes Center, Helmholtz Zentrum München, Neuherberg, Germany
| | - Henrik Oster
- Center of Brain, Behavior and Metabolism (CBBM), University of Lübeck, Lübeck, Germany.,Institute of Neurobiology, University of Lübeck, Lübeck, Germany
| | - N Henriette Uhlenhaut
- Institute for Diabetes and Endocrinology, Helmholtz Diabetes Center, Helmholtz Zentrum München, Neuherberg, Germany.,Chair for Metabolic Programming, TUM School of Life Sciences Weihenstephan, & ZIEL-Institute for Food & Health, Freising, Germany
| | - Sebastian M Meyhöfer
- Institute for Endocrinology and Diabetes, University of Lübeck, Lübeck, Germany.,Center of Brain, Behavior and Metabolism (CBBM), University of Lübeck, Lübeck, Germany.,German Center for Diabetes Research (DZD), München-Neuherberg, Germany
| |
Collapse
|
17
|
Role of the Endocannabinoid System in Metabolic Control Processes and in the Pathogenesis of Metabolic Syndrome: An Update. Biomedicines 2023; 11:biomedicines11020306. [PMID: 36830844 PMCID: PMC9952954 DOI: 10.3390/biomedicines11020306] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2022] [Revised: 01/14/2023] [Accepted: 01/18/2023] [Indexed: 01/24/2023] Open
Abstract
Metabolic syndrome is a complex disease state, which appears mostly as a consequence of an unhealthy, sedentary lifestyle. Metabolic complications include insulin resistance (IR), diabetes, dyslipidemia, hypertension, and atherosclerosis, impairing life standards and reducing life expectancy. The endocannabinoid system (ECS) has an important role in signalization processes, not only in the central nervous system, but also in the peripheral tissues. Several physiological functions are affected, and overexpression or downregulation contributes to several diseases. A better understanding of the functions of cannabinoid (CB) receptors may propose potential therapeutic effects by influencing receptor signaling and enzymes involved in downstream pathways. In this review, we summarize recent information regarding the roles of the ECS and the CB1 receptor signaling in the physiology and pathophysiology of energy and metabolic homeostasis, in the development of obesity by enhancing food intake, upregulating energy balance and fat accumulation, increasing lipogenesis and glucose production, and impairing insulin sensitivity and secretion. By analyzing the roles of the ECS in physiological and pathophysiological mechanisms, we introduce some recently identified signaling pathways in the mechanism of the pathogenesis of metabolic syndrome. Our review emphasizes that the presence of such recently identified ECS signaling steps raises new therapeutic potential in the treatment of complex metabolic diseases such as diabetes, insulin resistance, obesity, and hypertension.
Collapse
|
18
|
Marcos JL, Olivares-Barraza R, Ceballo K, Wastavino M, Ortiz V, Riquelme J, Martínez-Pinto J, Muñoz P, Cruz G, Sotomayor-Zárate R. Obesogenic Diet-Induced Neuroinflammation: A Pathological Link between Hedonic and Homeostatic Control of Food Intake. Int J Mol Sci 2023; 24:ijms24021468. [PMID: 36674982 PMCID: PMC9866213 DOI: 10.3390/ijms24021468] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2022] [Revised: 12/22/2022] [Accepted: 12/26/2022] [Indexed: 01/13/2023] Open
Abstract
Obesity-induced neuroinflammation is a chronic aseptic central nervous system inflammation that presents systemic characteristics associated with increased pro-inflammatory cytokines such as interleukin 1 beta (IL-1β) and interleukin 18 (IL-18) and the presence of microglia and reactive astrogliosis as well as the activation of the NLRP3 inflammasome. The obesity pandemic is associated with lifestyle changes, including an excessive intake of obesogenic foods and decreased physical activity. Brain areas such as the lateral hypothalamus (LH), lateral septum (LS), ventral tegmental area (VTA), and nucleus accumbens (NAcc) have been implicated in the homeostatic and hedonic control of feeding in experimental models of diet-induced obesity. In this context, a chronic lipid intake triggers neuroinflammation in several brain regions such as the hypothalamus, hippocampus, and amygdala. This review aims to present the background defining the significant impact of neuroinflammation and how this, when induced by an obesogenic diet, can affect feeding control, triggering metabolic and neurological alterations.
Collapse
Affiliation(s)
- José Luis Marcos
- Centro de Neurobiología y Fisiopatología Integrativa (CENFI), Instituto de Fisiología, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso 2360102, Chile
- Escuela de Ciencias Agrícolas y Veterinarias, Universidad Viña del Mar, Viña del Mar 2572007, Chile
- Programa de Doctorado en Ciencias e Ingeniería para la Salud, Universidad de Valparaíso, Valparaíso 2360102, Chile
| | - Rossy Olivares-Barraza
- Centro de Neurobiología y Fisiopatología Integrativa (CENFI), Instituto de Fisiología, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso 2360102, Chile
- Programa de Doctorado en Ciencias Mención Neurociencias, Universidad de Valparaíso, Valparaíso 2360102, Chile
| | - Karina Ceballo
- Centro de Neurobiología y Fisiopatología Integrativa (CENFI), Instituto de Fisiología, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso 2360102, Chile
- Programa de Doctorado en Ciencias Mención Neurociencias, Universidad de Valparaíso, Valparaíso 2360102, Chile
| | - Melisa Wastavino
- Centro de Neurobiología y Fisiopatología Integrativa (CENFI), Instituto de Fisiología, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso 2360102, Chile
| | - Víctor Ortiz
- Centro de Neurobiología y Fisiopatología Integrativa (CENFI), Instituto de Fisiología, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso 2360102, Chile
| | - Julio Riquelme
- Escuela de Medicina y Centro de Neurología Traslacional (CENTRAS), Facultad de Medicina, Universidad de Valparaíso, Viña del Mar 2540064, Chile
| | - Jonathan Martínez-Pinto
- Centro de Neurobiología y Fisiopatología Integrativa (CENFI), Instituto de Fisiología, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso 2360102, Chile
| | - Pablo Muñoz
- Escuela de Medicina y Centro de Neurología Traslacional (CENTRAS), Facultad de Medicina, Universidad de Valparaíso, Viña del Mar 2540064, Chile
| | - Gonzalo Cruz
- Centro de Neurobiología y Fisiopatología Integrativa (CENFI), Instituto de Fisiología, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso 2360102, Chile
| | - Ramón Sotomayor-Zárate
- Centro de Neurobiología y Fisiopatología Integrativa (CENFI), Instituto de Fisiología, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso 2360102, Chile
- Correspondence: ; Tel.: +56-32-2508050
| |
Collapse
|
19
|
Ralevski E, Horvath TL, Shanabrough M, Newcomb J, Pisani E, Petrakis I. Ghrelin Predicts Stimulant and Sedative Effects of Alcohol in Heavy Drinkers. Alcohol Alcohol 2023; 58:100-106. [PMID: 36382470 PMCID: PMC9830489 DOI: 10.1093/alcalc/agac058] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2022] [Revised: 09/16/2022] [Accepted: 10/10/2022] [Indexed: 11/17/2022] Open
Abstract
AIM The aim of this study was to examine the relationship between ghrelin levels and the subjective effects of alcohol in heavy drinkers, and to compare them to healthy controls. METHODS Ghrelin levels were collected as part of two laboratory studies. Both groups received either IV infusion of saline or high dose of alcohol (100 mg%). In the study of heavy drinkers, ghrelin was gathered on all subjects, but data was analyzed only for participants who received placebo (N=12). Healthy controls (N=20) came from another study that collected data on family history. Ghrelin levels and measures of alcohol effects (BAES, VAS, NDS, YCS [see manuscript for details]) were collected at 4 timepoints: baseline, before infusion, during infusion and after infusion. RESULTS IV alcohol significantly reduced ghrelin levels and higher fasting ghrelin levels were associated with more intense subjective alcohol effects. There were no differences in fasting ghrelin levels or subjective effects between heavy drinkers and controls. However, while both groups showed similar decline in ghrelin levels following alcohol infusion, on the placebo day, ghrelin levels in the healthy subjects increased significantly and exponentially over time while for the heavy drinkers ghrelin levels remained flat. CONCLUSIONS Our findings support the role of ghrelin in reward mechanisms for alcohol. Contrary to others, we found no differences in fasting ghrelin levels or subjective experiences of alcohol between heavy drinkers and healthy controls. However, the group differences on the IV placebo day may be a possible indication of ghrelin abnormalities in heavy drinkers.
Collapse
Affiliation(s)
- Elizabeth Ralevski
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT 06520, USA
- Department of Veteran Affairs, VA Connecticut Healthcare System, West Haven, CT, USA
- Mental Illness Research and Clinical Center, VA Connecticut Healthcare System, West Haven, CT, USA
| | - Tamas L Horvath
- Program of Integrative Cell Signaling and Neurobiology of Metabolism, Section of Comparative Medicine, Yale University School of Medicine, New Haven 06520, CT, USA
- Department of Obstetrics/Gynecology and Reproductive Sciences, Yale University School of Medicine, New Haven 06520, CT, USA
| | - Marya Shanabrough
- Program of Integrative Cell Signaling and Neurobiology of Metabolism, Section of Comparative Medicine, Yale University School of Medicine, New Haven 06520, CT, USA
| | - Jenelle Newcomb
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT 06520, USA
- Department of Veteran Affairs, VA Connecticut Healthcare System, West Haven, CT, USA
- Mental Illness Research and Clinical Center, VA Connecticut Healthcare System, West Haven, CT, USA
| | - Emily Pisani
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT 06520, USA
- Department of Veteran Affairs, VA Connecticut Healthcare System, West Haven, CT, USA
- Mental Illness Research and Clinical Center, VA Connecticut Healthcare System, West Haven, CT, USA
| | - Ismene Petrakis
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT 06520, USA
- Department of Veteran Affairs, VA Connecticut Healthcare System, West Haven, CT, USA
- Mental Illness Research and Clinical Center, VA Connecticut Healthcare System, West Haven, CT, USA
| |
Collapse
|
20
|
Kharbanda KK, Farokhnia M, Deschaine SL, Bhargava R, Rodriguez-Flores M, Casey CA, Goldstone AP, Jerlhag E, Leggio L, Rasineni K. Role of the ghrelin system in alcohol use disorder and alcohol-associated liver disease: A narrative review. Alcohol Clin Exp Res 2022; 46:2149-2159. [PMID: 36316764 PMCID: PMC9772086 DOI: 10.1111/acer.14967] [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: 08/19/2022] [Revised: 10/21/2022] [Accepted: 10/28/2022] [Indexed: 11/07/2022]
Abstract
Unhealthy alcohol consumption is a global health problem. Adverse individual, public health, and socioeconomic consequences are attributable to harmful alcohol use. Epidemiological studies have shown that alcohol use disorder (AUD) and alcohol-associated liver disease (ALD) are the top two pathologies among alcohol-related diseases. Consistent with the major role that the liver plays in alcohol metabolism, uncontrolled drinking may cause significant damage to the liver. This damage is initiated by excessive fat accumulation in the liver, which can further progress to advanced liver disease. The only effective therapeutic strategies currently available for ALD are alcohol abstinence or liver transplantation. Any molecule with dual-pronged effects at the central and peripheral organs controlling addictive behaviors and associated metabolic pathways are a potentially important therapeutic target for treating AUD and ALD. Ghrelin, a hormone primarily derived from the stomach, has such properties, and regulates both behavioral and metabolic functions. In this review, we highlight recent advances in understanding the peripheral and central functions of the ghrelin system and its role in AUD and ALD pathogenesis. We first discuss the correlation between blood ghrelin concentrations and alcohol use or abstinence. Next, we discuss the role of ghrelin in alcohol-seeking behaviors and finally its role in the development of fatty liver by metabolic regulations and organ crosstalk. We propose that a better understanding of the ghrelin system could open an innovative avenue for improved treatments for AUD and associated medical consequences, including ALD.
Collapse
Affiliation(s)
- Kusum K. Kharbanda
- Research Service, Veterans Affairs Nebraska-Western Iowa Health Care System, Omaha, Nebraska, USA
- Department of Internal Medicine, University of Nebraska Medical Center, Omaha, Nebraska, USA
- Department of Biochemistry & Molecular Biology, University of Nebraska Medical Center, Omaha, Nebraska, USA
| | - Mehdi Farokhnia
- Clinical Psychoneuroendocrinology and Neuropsychopharmacology Section, Translational Addiction Medicine Branch, National Institute on Drug Abuse, Intramural Research Program and National Institute on Alcohol Abuse and Alcoholism, Division of Intramural Clinical and Biological Research, National Institutes of Health, Baltimore and Bethesda, Maryland, USA
- Johns Hopkins Bloomberg School of Public Health, Johns Hopkins University, Baltimore, Maryland, USA
| | - Sara L. Deschaine
- Clinical Psychoneuroendocrinology and Neuropsychopharmacology Section, Translational Addiction Medicine Branch, National Institute on Drug Abuse, Intramural Research Program and National Institute on Alcohol Abuse and Alcoholism, Division of Intramural Clinical and Biological Research, National Institutes of Health, Baltimore and Bethesda, Maryland, USA
| | - Raghav Bhargava
- PsychoNeuroEndocrinology Research Group, Division of Psychiatry, Department of Brain Sciences, Imperial College London, Hammersmith Hospital, London, UK
| | - Marcela Rodriguez-Flores
- PsychoNeuroEndocrinology Research Group, Division of Psychiatry, Department of Brain Sciences, Imperial College London, Hammersmith Hospital, London, UK
| | - Carol A. Casey
- Research Service, Veterans Affairs Nebraska-Western Iowa Health Care System, Omaha, Nebraska, USA
- Department of Internal Medicine, University of Nebraska Medical Center, Omaha, Nebraska, USA
- Department of Biochemistry & Molecular Biology, University of Nebraska Medical Center, Omaha, Nebraska, USA
| | - Anthony P. Goldstone
- PsychoNeuroEndocrinology Research Group, Division of Psychiatry, Department of Brain Sciences, Imperial College London, Hammersmith Hospital, London, UK
| | - Elisabet Jerlhag
- Department of Pharmacology, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
| | - Lorenzo Leggio
- Clinical Psychoneuroendocrinology and Neuropsychopharmacology Section, Translational Addiction Medicine Branch, National Institute on Drug Abuse, Intramural Research Program and National Institute on Alcohol Abuse and Alcoholism, Division of Intramural Clinical and Biological Research, National Institutes of Health, Baltimore and Bethesda, Maryland, USA
- Medication Development Program, National Institute on Drug Abuse Intramural Research Program, National Institutes of Health, Baltimore, Maryland, USA
- Center for Alcohol and Addiction Studies, Department of Behavioral and Social Sciences, Brown University, Providence, Rhode Island, USA
- Division of Addiction Medicine, Department of Medicine, School of Medicine, Johns Hopkins University, Baltimore, Maryland, USA
- Department of Neuroscience, Georgetown University Medical Center, Washington DC, USA
| | - Karuna Rasineni
- Research Service, Veterans Affairs Nebraska-Western Iowa Health Care System, Omaha, Nebraska, USA
- Department of Internal Medicine, University of Nebraska Medical Center, Omaha, Nebraska, USA
| |
Collapse
|
21
|
Hanßen R, Schiweck C, Aichholzer M, Reif A, Edwin Thanarajah S. Food reward and its aberrations in obesity. Curr Opin Behav Sci 2022. [DOI: 10.1016/j.cobeha.2022.101224] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
|
22
|
Prida E, Álvarez-Delgado S, Pérez-Lois R, Soto-Tielas M, Estany-Gestal A, Fernø J, Seoane LM, Quiñones M, Al-Massadi O. Liver Brain Interactions: Focus on FGF21 a Systematic Review. Int J Mol Sci 2022; 23:ijms232113318. [PMID: 36362103 PMCID: PMC9658462 DOI: 10.3390/ijms232113318] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2022] [Revised: 10/21/2022] [Accepted: 10/29/2022] [Indexed: 11/06/2022] Open
Abstract
Fibroblast growth factor 21 is a pleiotropic hormone secreted mainly by the liver in response to metabolic and nutritional challenges. Physiologically, fibroblast growth factor 21 plays a key role in mediating the metabolic responses to fasting or starvation and acts as an important regulator of energy homeostasis, glucose and lipid metabolism, and insulin sensitivity, in part by its direct action on the central nervous system. Accordingly, pharmacological recombinant fibroblast growth factor 21 therapies have been shown to counteract obesity and its related metabolic disorders in both rodents and nonhuman primates. In this systematic review, we discuss how fibroblast growth factor 21 regulates metabolism and its interactions with the central nervous system. In addition, we also state our vision for possible therapeutic uses of this hepatic-brain axis.
Collapse
Affiliation(s)
- Eva Prida
- Instituto de Investigación Sanitaria de Santiago de Compostela, Complexo Hospitalario Universitario de Santiago (CHUS/SERGAS), Travesía da Choupana s/n, 15706 Santiago de Compostela, Spain
| | - Sara Álvarez-Delgado
- Instituto de Investigación Sanitaria de Santiago de Compostela, Complexo Hospitalario Universitario de Santiago (CHUS/SERGAS), Travesía da Choupana s/n, 15706 Santiago de Compostela, Spain
| | - Raquel Pérez-Lois
- Instituto de Investigación Sanitaria de Santiago de Compostela, Complexo Hospitalario Universitario de Santiago (CHUS/SERGAS), Travesía da Choupana s/n, 15706 Santiago de Compostela, Spain
- CIBER de Fisiopatología de la Obesidad y la Nutrición, Instituto de Salud Carlos III, 15706 Santiago de Compostela, Spain
| | - Mateo Soto-Tielas
- Instituto de Investigación Sanitaria de Santiago de Compostela, Complexo Hospitalario Universitario de Santiago (CHUS/SERGAS), Travesía da Choupana s/n, 15706 Santiago de Compostela, Spain
| | - Ana Estany-Gestal
- Unidad de Metodología de la Investigación, Fundación Instituto de Investigación de Santiago (FIDIS), 15706 Santiago de Compostela, Spain
| | - Johan Fernø
- Hormone Laboratory, Department of Biochemistry and Pharmacology, Haukeland University Hospital, 5201 Bergen, Norway
| | - Luisa María Seoane
- Instituto de Investigación Sanitaria de Santiago de Compostela, Complexo Hospitalario Universitario de Santiago (CHUS/SERGAS), Travesía da Choupana s/n, 15706 Santiago de Compostela, Spain
- CIBER de Fisiopatología de la Obesidad y la Nutrición, Instituto de Salud Carlos III, 15706 Santiago de Compostela, Spain
| | - Mar Quiñones
- Instituto de Investigación Sanitaria de Santiago de Compostela, Complexo Hospitalario Universitario de Santiago (CHUS/SERGAS), Travesía da Choupana s/n, 15706 Santiago de Compostela, Spain
- CIBER de Fisiopatología de la Obesidad y la Nutrición, Instituto de Salud Carlos III, 15706 Santiago de Compostela, Spain
- Correspondence: (M.Q.); (O.A.-M.); Tel.: +34-981955708 (M.Q.); +34-981955522 (O.A.-M.)
| | - Omar Al-Massadi
- Instituto de Investigación Sanitaria de Santiago de Compostela, Complexo Hospitalario Universitario de Santiago (CHUS/SERGAS), Travesía da Choupana s/n, 15706 Santiago de Compostela, Spain
- CIBER de Fisiopatología de la Obesidad y la Nutrición, Instituto de Salud Carlos III, 15706 Santiago de Compostela, Spain
- Correspondence: (M.Q.); (O.A.-M.); Tel.: +34-981955708 (M.Q.); +34-981955522 (O.A.-M.)
| |
Collapse
|
23
|
Al-Massadi O, Parini P, Fernø J, Luquet S, Quiñones M. Metabolic actions of the growth hormone-insulin growth factor-1 axis and its interaction with the central nervous system. Rev Endocr Metab Disord 2022; 23:919-930. [PMID: 35687272 DOI: 10.1007/s11154-022-09732-x] [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] [Accepted: 05/02/2022] [Indexed: 10/18/2022]
Abstract
The growth hormone/insulin growth factor-1 axis is a key endocrine system that exerts profound effects on metabolism by its actions on different peripheral tissues but also in the brain. Growth hormone together with insulin growth factor-1 perform metabolic adjustments, including regulation of food intake, energy expenditure, and glycemia. The dysregulation of this hepatic axis leads to different metabolic disorders including obesity, type 2 diabetes or liver disease. In this review, we discuss how the growth hormone/insulin growth factor-1 axis regulates metabolism and its interactions with the central nervous system. Finally, we state our vision for possible therapeutic uses of compounds based in the components of this hepatic axis.
Collapse
Affiliation(s)
- Omar Al-Massadi
- Instituto de Investigación Sanitaria de Santiago de Compostela, Complexo Hospitalario Universitario de Santiago (CHUS/SERGAS), Travesía da Choupana s/n, 15706, Santiago de Compostela, Spain.
- CIBER de Fisiopatología de la Obesidad y la Nutrición, Instituto de Salud Carlos III, 15706, Santiago de Compostela, Spain.
| | - Paolo Parini
- Department of Laboratory Medicine, Division of Clinical Chemistry, Karolinska Institute, Stockholm, Sweden
- Department of Medicine, Metabolism Unit, Karolinska Institute at Karolinska University Hospital Huddinge, Stockholm, Sweden
- Patient Area Nephrology and Endocrinology, Inflammation and Infection Theme, Karolinska University Hospital, Stockholm, Sweden
| | - Johan Fernø
- Hormone Laboratory, Haukeland University Hospital, Bergen, Norway
| | - Serge Luquet
- Unité de Biologie Fonctionnelle et Adaptative, Univ Paris Diderot, Sorbonne Paris Cité, CNRS UMR 8251, F-75205, Paris, France
| | - Mar Quiñones
- Instituto de Investigación Sanitaria de Santiago de Compostela, Complexo Hospitalario Universitario de Santiago (CHUS/SERGAS), Travesía da Choupana s/n, 15706, Santiago de Compostela, Spain.
- CIBER de Fisiopatología de la Obesidad y la Nutrición, Instituto de Salud Carlos III, 15706, Santiago de Compostela, Spain.
- Unité de Biologie Fonctionnelle et Adaptative, Univ Paris Diderot, Sorbonne Paris Cité, CNRS UMR 8251, F-75205, Paris, France.
| |
Collapse
|
24
|
Ma Y, Zhang H, Guo W, Yu L. Potential role of ghrelin in the regulation of inflammation. FASEB J 2022; 36:e22508. [PMID: 35983825 DOI: 10.1096/fj.202200634r] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2022] [Revised: 07/27/2022] [Accepted: 08/08/2022] [Indexed: 11/11/2022]
Abstract
Several diseases are caused or progress due to inflammation. In the past few years, accumulating evidence suggests that ghrelin, a gastric hormone of 28-amino acid residue length, exerts protective effects against inflammation by modulating the related pathways. This review focuses on ghrelin's anti-inflammatory and potential therapeutic effects in neurological, cardiovascular, respiratory, hepatic, gastrointestinal, and kidney disorders. Ghrelin significantly alleviates excessive inflammation and reduces damage to different target organs mainly by reducing the secretion of inflammatory cytokines, including interleukin-6 (IL-6), interleukin-1β (IL-1β), and tumor necrosis factor-α (TNF-α), and inhibiting the nuclear factor kappa-B (NF-κB) and NLRP3 inflammasome signaling pathways. Ghrelin also regulates inflammation and apoptosis through the p38 MAPK/c-Jun N-terminal kinase (JNK) signaling pathway; restores cerebral microvascular integrity, and attenuates vascular leakage. Ghrelin activates the phosphoInositide-3 kinase (PI3K)/protein kinase B (Akt) pathway and inhibits inflammatory responses in cardiovascular diseases and acute kidney injury. Some studies show that ghrelin exacerbates colonic and intestinal manifestations of colitis. Interestingly, some inflammatory states, such as non-alcoholic steatohepatitis, inflammatory bowel diseases, and chronic kidney disease, are often associated with high ghrelin levels. Thus, ghrelin may be a potential new therapeutic target for inflammation-related diseases.
Collapse
Affiliation(s)
- Yunxiao Ma
- Department of Endocrinology and Department of Interventional Therapy of First Hospital of Jilin University, State Key Laboratory for Zoonotic Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, Institute of Zoonosis, and College of Veterinary Medicine, Jilin University, Changchun, China
| | - Haifeng Zhang
- Department of Endocrinology and Department of Interventional Therapy of First Hospital of Jilin University, State Key Laboratory for Zoonotic Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, Institute of Zoonosis, and College of Veterinary Medicine, Jilin University, Changchun, China
| | - Weiying Guo
- Department of Endocrinology and Department of Interventional Therapy of First Hospital of Jilin University, State Key Laboratory for Zoonotic Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, Institute of Zoonosis, and College of Veterinary Medicine, Jilin University, Changchun, China
| | - Lu Yu
- Department of Endocrinology and Department of Interventional Therapy of First Hospital of Jilin University, State Key Laboratory for Zoonotic Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, Institute of Zoonosis, and College of Veterinary Medicine, Jilin University, Changchun, China
| |
Collapse
|
25
|
Ceccarini MR, Precone V, Manara E, Paolacci S, Maltese PE, Benfatti V, Dhuli K, Donato K, Guerri G, Marceddu G, Chiurazzi P, Dalla Ragione L, Beccari T, Bertelli M. A next generation sequencing gene panel for use in the diagnosis of anorexia nervosa. Eat Weight Disord 2022; 27:1869-1880. [PMID: 34822136 DOI: 10.1007/s40519-021-01331-0] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/07/2021] [Accepted: 11/07/2021] [Indexed: 12/12/2022] Open
Abstract
PURPOSE The aim of this study was to increase knowledge of genes associated with anorexia nervosa (AN) and their diagnostic offer, using a next generation sequencing (NGS) panel for the identification of genetic variants. The rationale underlying this test is that we first analyze the genes associated with syndromic forms of AN, then genes that were found to carry rare variants in AN patients who had undergone segregation analysis, and finally candidate genes intervening in the same molecular pathways or identified by GWAS or in mouse models. METHODS We developed an NGS gene panel and used it to screen 68 Italian AN patients (63 females, 5 males). The panel included 162 genes. Family segregation study was conducted on available relatives of probands who reported significant genetic variants. RESULTS In our analysis, we found potentially deleterious variants in 2 genes (PDE11A and SLC25A13) associated with syndromic forms of anorexia and predicted deleterious variants in the following 12 genes: CD36, CACNA1C, DRD4, EPHX2, ESR1, GRIN2A, GRIN3B, LRP2, NPY4R, PTGS2, PTPN22 and SGPP2. Furthermore, by Sanger sequencing of the promoter region of NNAT, we confirmed the involvement of this gene in the pathogenesis of AN. Family segregation studies further strengthened the possible causative role of CACNA1C, DRD4, GRIN2A, PTGS2, SGPP2, SLC25A13 and NNAT genes in AN etiology. CONCLUSION The major finding of our study is the confirmation of the involvement of the NNAT gene in the pathogenesis of AN; furthermore, this study suggests that NGS-based testing can play an important role in the diagnostic evaluation of AN, excluding syndromic forms and increasing knowledge of the genetic etiology of AN. LEVEL OF EVIDENCE Level I, experimental study.
Collapse
Affiliation(s)
- Maria Rachele Ceccarini
- Department of Pharmaceutical Sciences, University of Perugia, Perugia, Italy.
- C.I.B., Consorzio Interuniversitario per le Biotecnologie, Trieste, Italy.
| | | | | | | | | | - Valentina Benfatti
- Department of Eating Disorder, Palazzo Francisci Todi, USL 1 Umbria, Todi, PG, Italy
| | | | | | | | | | - Pietro Chiurazzi
- Dipartimento Universitario Scienze della Vita e Sanità Pubblica, Sezione di Medicina Genomica, Università Cattolica del Sacro Cuore, 00168, Rome, Italy
- Fondazione Policlinico Universitario "A. Gemelli" IRCCS, UOC Genetica Medica, 00168, Roma, Italy
| | - Laura Dalla Ragione
- Department of Eating Disorder, Palazzo Francisci Todi, USL 1 Umbria, Todi, PG, Italy
- Food Science and Human Nutrition Unit, University Campus Biomedico of Rome, Rome, Italy
| | - Tommaso Beccari
- Department of Pharmaceutical Sciences, University of Perugia, Perugia, Italy
- C.I.B., Consorzio Interuniversitario per le Biotecnologie, Trieste, Italy
| | | |
Collapse
|
26
|
Lu Y, Zhang R, Lei H, Hang Y, Xue H, Cai X, Lu Y. Supplementation with Fermented Feedstuff Enhances Orexin Expression and Secretion Associated with Increased Feed Intake and Weight Gain in Weaned Pigs. Animals (Basel) 2022; 12:1329. [PMID: 35625175 PMCID: PMC9138051 DOI: 10.3390/ani12101329] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2022] [Revised: 05/17/2022] [Accepted: 05/18/2022] [Indexed: 11/23/2022] Open
Abstract
The health status of weaned pigs is crucial for their subsequent growth performance. Supplementation with fermented feedstuff is able to improve the feed intake and growth of weaned pigs; however, the exact mechanism behind this is not clear. Hence, in the present study a total of 320 Duroc × Landrace × Yorkshire weaned pigs were selected and allocated to the following two groups: unfermented diet group (UFD) and fermented diet group (FD). The experimental period lasted 21 days. At the end of the experiment, feces, blood, and gastrointestinal tissue samples (including the stomach, jejunum, and ileum) were collected and used for further analysis. The results of growth performance suggested that the FD group had significantly increased (p < 0.05) average daily feed intake (ADFI) and average daily gain (ADG) during the first week, during the last two weeks, and over the entire three-week period compared with the UFD group. The results of the apparent nutrient digestibility of pigs showed that, compared with the UFD group, the FD group showed increased phosphorus (p < 0.05) and CP (p < 0.1) digestibility. There were no significant differences in the serum biochemical parameters between the UFD and FD groups. Moreover, our results showed that the FD group showed significantly increased gene expression of SGLT1 and PepT1 in the jejunum (p < 0.05). Compared with the UFD group, the FD group showed an increased (p < 0.05) serum orexin level and prepro-orexin (PPOX) expression in the gastric fundus, jejunum, and ileum mucosa and increased IGF-1 and IGFR expression in the jejunum. Collectively, these results indicated that supplementation with fermented feedstuff in the diet effectively enhanced the feed intake and growth of weaned pigs and that this may have been caused by the increased orexin, IGF-1, and IGFR serum levels.
Collapse
Affiliation(s)
- Yang Lu
- Institute of Animal Husbandry and Veterinary Science, Shanghai Academy of Agricultural Sciences, Shanghai 201106, China; (H.L.); (Y.H.); (H.X.); (X.C.)
| | - Ruiyang Zhang
- College of Animal Science and Veterinary Medicine, Shenyang Agricultural University, Shenyang 110866, China;
| | - Hulong Lei
- Institute of Animal Husbandry and Veterinary Science, Shanghai Academy of Agricultural Sciences, Shanghai 201106, China; (H.L.); (Y.H.); (H.X.); (X.C.)
| | - Yiqiong Hang
- Institute of Animal Husbandry and Veterinary Science, Shanghai Academy of Agricultural Sciences, Shanghai 201106, China; (H.L.); (Y.H.); (H.X.); (X.C.)
| | - Huiqin Xue
- Institute of Animal Husbandry and Veterinary Science, Shanghai Academy of Agricultural Sciences, Shanghai 201106, China; (H.L.); (Y.H.); (H.X.); (X.C.)
| | - Xuan Cai
- Institute of Animal Husbandry and Veterinary Science, Shanghai Academy of Agricultural Sciences, Shanghai 201106, China; (H.L.); (Y.H.); (H.X.); (X.C.)
| | - Yonghong Lu
- Institute of Animal Husbandry and Veterinary Science, Shanghai Academy of Agricultural Sciences, Shanghai 201106, China; (H.L.); (Y.H.); (H.X.); (X.C.)
| |
Collapse
|
27
|
Chen X, Dong J, Jiao Q, Du X, Bi M, Jiang H. "Sibling" battle or harmony: crosstalk between nesfatin-1 and ghrelin. Cell Mol Life Sci 2022; 79:169. [PMID: 35239020 PMCID: PMC11072372 DOI: 10.1007/s00018-022-04193-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2021] [Revised: 02/02/2022] [Accepted: 02/04/2022] [Indexed: 12/17/2022]
Abstract
Ghrelin was first identified as an endogenous ligand of the growth hormone secretagogue receptor (GHSR) in 1999, with the function of stimulating the release of growth hormone (GH), while nesfatin-1 was identified in 2006. Both peptides are secreted by the same kind of endocrine cells, X/A-like cells in the stomach. Compared with ghrelin, nesfatin-1 exerts opposite effects on energy metabolism, glucose metabolism, gastrointestinal functions and regulation of blood pressure, but exerts similar effects on anti-inflammation and neuroprotection. Up to now, nesfatin-1 remains as an orphan ligand because its receptor has not been identified. Several studies have shown the effects of nesfatin-1 are dependent on the receptor of ghrelin. We herein compare the effects of nesfatin-1 and ghrelin in several aspects and explore the possibility of their interactions.
Collapse
Affiliation(s)
- Xi Chen
- Department of Physiology, Shandong Provincial Key Laboratory of Pathogenesis and Prevention of Neurological Disorders and State Key Disciplines: Physiology, School of Basic Medicine, Qingdao University, Qingdao, 266071, People's Republic of China
| | - Jing Dong
- Department of Physiology, Shandong Provincial Key Laboratory of Pathogenesis and Prevention of Neurological Disorders and State Key Disciplines: Physiology, School of Basic Medicine, Qingdao University, Qingdao, 266071, People's Republic of China
| | - Qian Jiao
- Department of Physiology, Shandong Provincial Key Laboratory of Pathogenesis and Prevention of Neurological Disorders and State Key Disciplines: Physiology, School of Basic Medicine, Qingdao University, Qingdao, 266071, People's Republic of China
| | - Xixun Du
- Department of Physiology, Shandong Provincial Key Laboratory of Pathogenesis and Prevention of Neurological Disorders and State Key Disciplines: Physiology, School of Basic Medicine, Qingdao University, Qingdao, 266071, People's Republic of China
| | - Mingxia Bi
- Department of Physiology, Shandong Provincial Key Laboratory of Pathogenesis and Prevention of Neurological Disorders and State Key Disciplines: Physiology, School of Basic Medicine, Qingdao University, Qingdao, 266071, People's Republic of China
| | - Hong Jiang
- Department of Physiology, Shandong Provincial Key Laboratory of Pathogenesis and Prevention of Neurological Disorders and State Key Disciplines: Physiology, School of Basic Medicine, Qingdao University, Qingdao, 266071, People's Republic of China.
| |
Collapse
|
28
|
Hanssen R, Thanarajah SE, Tittgemeyer M, Brüning JC. Obesity - A Matter of Motivation? Exp Clin Endocrinol Diabetes 2022; 130:290-295. [PMID: 35181879 PMCID: PMC9286865 DOI: 10.1055/a-1749-4852] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Excessive food intake and reduced physical activity have long been established as
primary causes of obesity. However, the underlying mechanisms causing this
unhealthy behavior characterized by heightened motivation for food but not for
physical effort are unclear. Despite the common unjustified stigmatization that
obesity is a result of laziness and lack of discipline, it is becoming
increasingly clear that high-fat diet feeding and obesity cause alterations in
brain circuits that are critical for the control of motivational behavior. In this mini-review, we provide a comprehensive overview of incentive motivation,
its neural encoding in the dopaminergic mesolimbic system as well as its
metabolic modulation with a focus on derangements of incentive motivation in
obesity. We further discuss the emerging field of metabolic interventions to
counteract motivational deficits and their potential clinical implications.
Collapse
Affiliation(s)
- Ruth Hanssen
- Max Planck Institute for Metabolism Research, Cologne, Germany.,Policlinic for Endocrinology, Diabetology and Preventive Medicine (PEPD), University of Cologne, Faculty of Medicine and University Hospital Cologne, Cologne, Germany
| | - Sharmili E Thanarajah
- Max Planck Institute for Metabolism Research, Cologne, Germany.,Department of Psychiatry, Psychosomatic Medicine and Psychotherapy, University Hospital Frankfurt, Frankfurt am Main, Germany
| | - Marc Tittgemeyer
- Max Planck Institute for Metabolism Research, Cologne, Germany.,Cluster of Excellence in Cellular Stress Responses in Aging-associated Diseases (CECAD), Cologne, Germany
| | - Jens C Brüning
- Max Planck Institute for Metabolism Research, Cologne, Germany.,Policlinic for Endocrinology, Diabetology and Preventive Medicine (PEPD), University of Cologne, Faculty of Medicine and University Hospital Cologne, Cologne, Germany.,Cluster of Excellence in Cellular Stress Responses in Aging-associated Diseases (CECAD), Cologne, Germany
| |
Collapse
|
29
|
Deschaine SL, Farokhnia M, Gregory-Flores A, Zallar LJ, You ZB, Sun H, Harvey DM, Marchette RCN, Tunstall BJ, Mani BK, Moose JE, Lee MR, Gardner E, Akhlaghi F, Roberto M, Hougland JL, Zigman JM, Koob GF, Vendruscolo LF, Leggio L. A closer look at alcohol-induced changes in the ghrelin system: novel insights from preclinical and clinical data. Addict Biol 2022; 27:e13033. [PMID: 33908131 PMCID: PMC8548413 DOI: 10.1111/adb.13033] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2020] [Revised: 01/16/2021] [Accepted: 03/06/2021] [Indexed: 02/06/2023]
Abstract
Ghrelin is a gastric-derived peptide hormone with demonstrated impact on alcohol intake and craving, but the reverse side of this bidirectional link, that is, the effects of alcohol on the ghrelin system, remains to be fully established. To further characterize this relationship, we examined (1) ghrelin levels via secondary analysis of human laboratory alcohol administration experiments with heavy-drinking participants; (2) expression of ghrelin, ghrelin receptor, and ghrelin-O-acyltransferase (GOAT) genes (GHRL, GHSR, and MBOAT4, respectively) in post-mortem brain tissue from individuals with alcohol use disorder (AUD) versus controls; (3) ghrelin levels in Ghsr knockout and wild-type rats following intraperitoneal (i.p.) alcohol administration; (4) effect of alcohol on ghrelin secretion from gastric mucosa cells ex vivo and GOAT enzymatic activity in vitro; and (5) ghrelin levels in rats following i.p. alcohol administration versus a calorically equivalent non-alcoholic sucrose solution. Acyl- and total-ghrelin levels decreased following acute alcohol administration in humans, but AUD was not associated with changes in central expression of ghrelin system genes in post-mortem tissue. In rats, alcohol decreased acyl-ghrelin, but not des-acyl-ghrelin, in both Ghsr knockout and wild-type rats. No dose-dependent effects of alcohol were observed on acyl-ghrelin secretion from gastric mucosa cells or on GOAT acylation activity. Lastly, alcohol and sucrose produced distinct effects on ghrelin in rats despite equivalent caloric value. Our findings suggest that alcohol acutely decreases peripheral ghrelin concentrations in vivo, but not in proportion to alcohol's caloric value or through direct interaction with ghrelin-secreting gastric mucosal cells, the ghrelin receptor, or the GOAT enzyme.
Collapse
Affiliation(s)
- Sara L. Deschaine
- Clinical Psychoneuroendocrinology and Neuropsychopharmacology Section, Translational Addiction Medicine Branch, National Institute on Drug Abuse and National, Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Baltimore, Maryland, USA
| | - Mehdi Farokhnia
- Clinical Psychoneuroendocrinology and Neuropsychopharmacology Section, Translational Addiction Medicine Branch, National Institute on Drug Abuse and National, Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Baltimore, Maryland, USA,Center on Compulsive Behaviors, National Institutes of Health, Bethesda, Maryland, USA,Johns Hopkins Bloomberg School of Public Health, Johns Hopkins University, Baltimore, Maryland, USA
| | - Adriana Gregory-Flores
- Clinical Psychoneuroendocrinology and Neuropsychopharmacology Section, Translational Addiction Medicine Branch, National Institute on Drug Abuse and National, Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Baltimore, Maryland, USA,Neurobiology of Addiction Section, Intramural Research Program, National Institute on Drug Abuse, National Institutes of Health, Baltimore, Maryland, USA
| | - Lia J. Zallar
- Clinical Psychoneuroendocrinology and Neuropsychopharmacology Section, Translational Addiction Medicine Branch, National Institute on Drug Abuse and National, Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Baltimore, Maryland, USA,Neurobiology of Addiction Section, Intramural Research Program, National Institute on Drug Abuse, National Institutes of Health, Baltimore, Maryland, USA
| | - Zhi-Bing You
- Neuropsychopharmacology Section, Molecular Targets and Medications Discovery Branch, National Institute on Drug Abuse, National Institutes of Health, Baltimore, Maryland, USA
| | - Hui Sun
- Clinical Core Laboratory, Office of the Clinical Director, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, Maryland, USA
| | - Deon M. Harvey
- Office of the Scientific Director, National Institute on Drug Abuse, Baltimore, Maryland, USA
| | - Renata C. N. Marchette
- Center on Compulsive Behaviors, National Institutes of Health, Bethesda, Maryland, USA,Neurobiology of Addiction Section, Intramural Research Program, National Institute on Drug Abuse, National Institutes of Health, Baltimore, Maryland, USA
| | - Brendan J. Tunstall
- Neurobiology of Addiction Section, Intramural Research Program, National Institute on Drug Abuse, National Institutes of Health, Baltimore, Maryland, USA
| | - Bharath K. Mani
- Center for Hypothalamic Research, Department of Internal Medicine, UT Southwestern Medical Center, Dallas, Texas, USA
| | - Jacob E. Moose
- Syracuse Biomaterials Institute, Syracuse University, Syracuse, New York, USA,Department of Chemistry, Syracuse University, Syracuse, New York, USA
| | - Mary R. Lee
- Clinical Psychoneuroendocrinology and Neuropsychopharmacology Section, Translational Addiction Medicine Branch, National Institute on Drug Abuse and National, Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Baltimore, Maryland, USA
| | - Eliot Gardner
- Neuropsychopharmacology Section, Molecular Targets and Medications Discovery Branch, National Institute on Drug Abuse, National Institutes of Health, Baltimore, Maryland, USA
| | - Fatemeh Akhlaghi
- Clinical Pharmacokinetics Research Laboratory, Department of Biomedical and Pharmaceutical Sciences, University of Rhode Island, Kingston, Rhode Island, USA
| | - Marisa Roberto
- Department of Neuroscience, The Scripps Research Institute, La Jolla, California, USA
| | - James L. Hougland
- Syracuse Biomaterials Institute, Syracuse University, Syracuse, New York, USA,Department of Chemistry, Syracuse University, Syracuse, New York, USA,BioInspired Syracuse, Syracuse University, Syracuse, New York, USA
| | - Jeffrey M. Zigman
- Center for Hypothalamic Research, Department of Internal Medicine, UT Southwestern Medical Center, Dallas, Texas, USA,Division of Endocrinology, Department of Internal Medicine, UT Southwestern Medical Center, Dallas, Texas, USA,Department of Psychiatry, UT Southwestern Medical Center, Dallas, Texas, USA
| | - George F. Koob
- Neurobiology of Addiction Section, Intramural Research Program, National Institute on Drug Abuse, National Institutes of Health, Baltimore, Maryland, USA
| | - Leandro F. Vendruscolo
- Neurobiology of Addiction Section, Intramural Research Program, National Institute on Drug Abuse, National Institutes of Health, Baltimore, Maryland, USA
| | - Lorenzo Leggio
- Clinical Psychoneuroendocrinology and Neuropsychopharmacology Section, Translational Addiction Medicine Branch, National Institute on Drug Abuse and National, Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Baltimore, Maryland, USA,Center on Compulsive Behaviors, National Institutes of Health, Bethesda, Maryland, USA,Medication Development Program, National Institute on Drug Abuse Intramural Research Program, National Institutes of Health, Baltimore, Maryland, USA,Center for Alcohol and Addiction Studies, Department of Behavioral and Social Sciences, School of Public Health, Brown University, Providence, Rhode Island, USA,Division of Addiction Medicine, Department of Medicine, School of Medicine, Johns Hopkins University, Baltimore, Maryland, USA,Department of Neuroscience, Georgetown University Medical Center, Washington, District of Columbia, USA
| |
Collapse
|
30
|
Deschaine SL, Leggio L. From "Hunger Hormone" to "It's Complicated": Ghrelin Beyond Feeding Control. Physiology (Bethesda) 2022; 37:5-15. [PMID: 34964687 PMCID: PMC8742734 DOI: 10.1152/physiol.00024.2021] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
Discovered as a peptide involved in releasing growth hormone, ghrelin was initially characterized as the "hunger hormone." However, emerging research indicates that ghrelin appears to play an important part in relaying information regarding nutrient availability and value and adjusting physiological and motivational processes accordingly. These functions make ghrelin an interesting therapeutic candidate for metabolic and neuropsychiatric diseases involving disrupted nutrition that can further potentiate the rewarding effect of maladaptive behaviors.
Collapse
Affiliation(s)
- Sara L. Deschaine
- 1Clinical Psychoneuroendocrinology and Neuropsychopharmacology Section, Translational Addiction Medicine Branch, National Institute on Drug Abuse and National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Baltimore and Bethesda, Maryland
| | - Lorenzo Leggio
- 1Clinical Psychoneuroendocrinology and Neuropsychopharmacology Section, Translational Addiction Medicine Branch, National Institute on Drug Abuse and National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Baltimore and Bethesda, Maryland,2Medication Development Program, National Institute on Drug Abuse Intramural Research Program, National Institutes of Health, Baltimore, Maryland,3Center for Alcohol and Addiction Studies, Department of Behavioral and Social Sciences, School of Public Health, Brown University, Providence, Rhode Island,4Division of Addiction Medicine, Department of Medicine, School of Medicine, Johns Hopkins University, Baltimore, Maryland,5Department of Neuroscience, Georgetown University Medical Center, Washington, District of Columbia
| |
Collapse
|
31
|
Al-Massadi O, Dieguez C, Schneeberger M, López M, Schwaninger M, Prevot V, Nogueiras R. Multifaceted actions of melanin-concentrating hormone on mammalian energy homeostasis. Nat Rev Endocrinol 2021; 17:745-755. [PMID: 34608277 DOI: 10.1038/s41574-021-00559-1] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 08/16/2021] [Indexed: 12/12/2022]
Abstract
Melanin-concentrating hormone (MCH) is a small cyclic peptide expressed in all mammals, mainly in the hypothalamus. MCH acts as a robust integrator of several physiological functions and has crucial roles in the regulation of sleep-wake rhythms, feeding behaviour and metabolism. MCH signalling has a very broad endocrine context and is involved in physiological functions and emotional states associated with metabolism, such as reproduction, anxiety, depression, sleep and circadian rhythms. MCH mediates its functions through two receptors (MCHR1 and MCHR2), of which only MCHR1 is common to all mammals. Owing to the wide variety of MCH downstream signalling pathways, MCHR1 agonists and antagonists have great potential as tools for the directed management of energy balance disorders and associated metabolic complications, and translational strategies using these compounds hold promise for the development of novel treatments for obesity. This Review provides an overview of the numerous roles of MCH in energy and glucose homeostasis, as well as in regulation of the mesolimbic dopaminergic circuits that encode the hedonic component of food intake.
Collapse
Affiliation(s)
- Omar Al-Massadi
- Instituto de Investigación Sanitaria de Santiago de Compostela, Complexo Hospitalario Universitario de Santiago (CHUS/SERGAS), Santiago de Compostela, Spain.
- CIBER Fisiopatologia de la Obesidad y Nutrición (CIBERobn), Santiago de Compostela, Spain.
| | - Carlos Dieguez
- CIBER Fisiopatologia de la Obesidad y Nutrición (CIBERobn), Santiago de Compostela, Spain
- Department of Physiology, CIMUS, University of Santiago de Compostela-Instituto de Investigación Sanitaria, Santiago de Compostela, Spain
| | - Marc Schneeberger
- Laboratory of Molecular Genetics, Howard Hughes Medical Institute, The Rockefeller University, New York, NY, USA
| | - Miguel López
- CIBER Fisiopatologia de la Obesidad y Nutrición (CIBERobn), Santiago de Compostela, Spain
- Department of Physiology, CIMUS, University of Santiago de Compostela-Instituto de Investigación Sanitaria, Santiago de Compostela, Spain
| | - Markus Schwaninger
- Institute for Experimental and Clinical Pharmacology and Toxicology, University of Lübeck, Lübeck, Germany
| | - Vincent Prevot
- Univ. Lille, Inserm, CHU Lille, Lille Neuroscience and Cognition, Laboratory of Development and Plasticity of the Neuroendocrine Brain, UMR-S1172, EGID, Lille, France
| | - Ruben Nogueiras
- CIBER Fisiopatologia de la Obesidad y Nutrición (CIBERobn), Santiago de Compostela, Spain.
- Department of Physiology, CIMUS, University of Santiago de Compostela-Instituto de Investigación Sanitaria, Santiago de Compostela, Spain.
- Galician Agency of Innovation (GAIN), Xunta de Galicia, Santiago de Compostela, Spain.
| |
Collapse
|
32
|
Gallegos-Gonzalez G, Pineda-García G, Serrano-Medina A, Martinez AL, Ochoa-Ruiz E. Association between Stress and Metabolic Syndrome and its Mediating Factors in University Students. Am J Health Behav 2021; 45:1091-1102. [PMID: 34969419 DOI: 10.5993/ajhb.45.6.12] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
OBJECTIVES In this study, we evaluated the association between perceived stress and indicators of metabolic syndrome and how this association is mediated by sleep problems, unhealthy eating habits, and night eating syndrome, in addition to serum levels of ghrelin and cortisol in university students. METHODS We recruited 192 students from a public university in Mexico. Weight, height, waist circumference and blood pressure were taken in accordance with standard protocols. Validated questionnaires were used to assess perceived stress, sleep quality and eating habits. Fasting blood samples were taken to measure ghrelin, cortisol, triglycerides, glucose and HDL-C. RESULTS Path Analysis indicated direct positive effects of stress over PSQI (β = 0.341) and NES (β = 0.443); PSQI over NES (β = 0.233) and NES over glucose (β = 0.170), triglycerides over LDL-C (β = 0.215), waist circumference over SBP (β = 0.259). Likewise, standardized negative regression weights of PSQI over Diet Quality Index (β = -0.239) and ghrelin concentrations (β = -0.132), ghrelin over Diet Quality Index (β = -0.188) and waist circumference (β = -0.147). Diet Quality Index over triglycerides (β = -0.184); sleep duration over systolic blood pressure (β = -0.242); waist circumference over HDL-C (β = -0.256). CONCLUSION Psychological stress leads to increased indicators of MetS via decreased sleep quality, inadequate eating habits and eating behavior in university students.
Collapse
Affiliation(s)
| | - Gisela Pineda-García
- Faculty of Medicine and Psychology at the Autonomous University of Baja California, Tijuana, BC, México
| | - Aracely Serrano-Medina
- Faculty of Medicine and Psychology at the Autonomous University of Baja California, Tijuana, BC, México
| | - Ana Laura Martinez
- Faculty of Medicine and Psychology at the Autonomous University of Baja California, Tijuana, BC, México
| | - Estefanía Ochoa-Ruiz
- Faculty of Medicine and Psychology at the Autonomous University of Baja California, Tijuana, BC, México;,
| |
Collapse
|
33
|
The Beneficial Effects of Essential Oils in Anti-Obesity Treatment. Int J Mol Sci 2021; 22:ijms222111832. [PMID: 34769261 PMCID: PMC8584325 DOI: 10.3390/ijms222111832] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2021] [Revised: 10/29/2021] [Accepted: 10/29/2021] [Indexed: 12/22/2022] Open
Abstract
Obesity is a complex disease caused by an excessive amount of body fat. Obesity is a medical problem and represents an important risk factor for the development of serious diseases such as insulin resistance, type 2 diabetes, cardiovascular disease, and some types of cancer. Not to be overlooked are the psychological issues that, in obese subjects, turn into very serious pathologies, such as depression, phobias, anxiety, and lack of self-esteem. In addition to modifying one’s lifestyle, the reduction of body mass can be promoted by different natural compounds such as essential oils (EOs). EOs are mixtures of aromatic substances produced by many plants, particularly in medicinal and aromatic ones. They are odorous and volatile and contain a mixture of terpenes, alcohols, aldehydes, ketones, and esters. Thanks to the characteristics of the various chemical components present in them, EOs are used in the food, cosmetic, and pharmaceutical fields. Indeed, it has been shown that EOs possess great antibiotic, anti-inflammatory, and antitumor powers. Emerging results also demonstrate the anti-obesity effects of EOs. We have examined the main data obtained in experimental studies and, in this review, we summarize the effect of EOs in obesity and obesity-related metabolic diseases.
Collapse
|
34
|
A population-based investigation of the association between alcohol intake and serum total ghrelin concentrations among cigarette-smoking, non-alcohol-dependent male individuals. Drug Alcohol Depend 2021; 226:108835. [PMID: 34214881 PMCID: PMC8355123 DOI: 10.1016/j.drugalcdep.2021.108835] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/25/2021] [Revised: 05/10/2021] [Accepted: 05/11/2021] [Indexed: 11/21/2022]
Abstract
BACKGROUND Ghrelin plays significant roles in regulating appetite, food intake, and metabolism. Furthermore, the ghrelin system is increasingly being studied in relation to alcohol seeking behaviors. To this end, it is important to understand the possible effects of alcohol intake on the ghrelin system. The aim of the present study was to investigate the association between alcohol drinking and circulating ghrelin levels in a large sample of cigarette-smoking, non-alcohol-dependent male individuals. METHODS We utilized data from two nested case-control studies (study A, n = 807; study B, n = 976) based within the Alpha-Tocopherol, Beta-Carotene Cancer Prevention (ATBC) trial. Data on alcohol consumption (grams of pure alcohol consumed per day) were obtained via a food frequency questionnaire. Blood samples were also collected (after 12 h of fasting), and serum concentrations of total ghrelin were measured by radioimmunoassay. RESULTS Dichotomous comparison between alcohol drinkers (>0 g/day of alcohol intake) and non-drinkers (0 g/day of alcohol intake) found higher total ghrelin levels among individuals who drank alcohol than those who did not, with statistically significant results in study A [F (1, 798) = 4.32, P = 0.03] and less robust results in study B [F (1, 966) = 2.62, P = 0.10], controlling for a list of factors that may influence ghrelin levels and/or differ between drinkers and non-drinkers. Bivariate correlational analysis among drinkers found no association between the quantity of daily alcohol intake and blood total ghrelin concentrations. CONCLUSION These results indicate elevated ghrelin levels among alcohol drinkers and provide additional/relevant information on the complex interaction between alcohol use and the ghrelin system.
Collapse
|
35
|
Brassard SL, Balodis IM. A review of effort-based decision-making in eating and weight disorders. Prog Neuropsychopharmacol Biol Psychiatry 2021; 110:110333. [PMID: 33905755 DOI: 10.1016/j.pnpbp.2021.110333] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/01/2020] [Revised: 03/19/2021] [Accepted: 04/21/2021] [Indexed: 12/26/2022]
Abstract
Effort-based decision-making provides a framework to understand the mental computations estimating the amount of work ("effort") required to obtain a reward. The aim of the current review is to systematically synthesize the available literature on effort-based decision-making across the spectrum of eating and weight disorders. More specifically, the current review summarises the literature examining whether 1) individuals with eating disorders and overweight/obesity are willing to expend more effort for rewards compared to healthy controls, 2) if particular components of effort-based decision-making (i.e. risk, discounting) relate to specific binge eating conditions, and 3) how individual differences in effort and reward -processing measures relate to eating pathology and treatment measures. A total of 96 studies were included in our review, following PRISMA guidelines. The review suggests that individuals with binge eating behaviours 1) are more likely to expend greater effort for food rewards, but not monetary rewards, 2) demonstrate greater decision-making impairments under risk and uncertainty, 3) prefer sooner rather than delayed rewards for both food and money, and 4) demonstrate increased implicit 'wanting' for high fat sweet foods. Finally, individual differences in effort and reward -processing measures relating to eating pathology and treatment measures are also discussed.
Collapse
Affiliation(s)
- Sarah L Brassard
- Department of Neuroscience, McMaster University, Canada; Peter Boris Center for Addictions Research, St. Joseph's Healthcare Hamilton, Canada
| | - Iris M Balodis
- Department of Neuroscience, McMaster University, Canada; Peter Boris Center for Addictions Research, St. Joseph's Healthcare Hamilton, Canada; Department of Psychiatry and Behavioural Neuroscience, McMaster University, Canada.
| |
Collapse
|
36
|
Micioni Di Bonaventura E, Botticelli L, Del Bello F, Giorgioni G, Piergentili A, Quaglia W, Cifani C, Micioni Di Bonaventura MV. Assessing the role of ghrelin and the enzyme ghrelin O-acyltransferase (GOAT) system in food reward, food motivation, and binge eating behavior. Pharmacol Res 2021; 172:105847. [PMID: 34438062 DOI: 10.1016/j.phrs.2021.105847] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/09/2021] [Revised: 08/19/2021] [Accepted: 08/20/2021] [Indexed: 02/06/2023]
Abstract
The peripheral peptide hormone ghrelin is a powerful stimulator of food intake, which leads to body weight gain and adiposity in both rodents and humans. The hormone, thus, increases the vulnerability to obesity and binge eating behavior. Several studies have revealed that ghrelin's functions are due to its interaction with the growth hormone secretagogue receptor type 1a (GHSR1a) in the hypothalamic area; besides, ghrelin also promotes the reinforcing properties of hedonic food, acting at extra-hypothalamic sites and interacting with dopaminergic, cannabinoid, opioid, and orexin signaling. The hormone is primarily present in two forms in the plasma and the enzyme ghrelin O-acyltransferase (GOAT) allows the acylation reaction which causes the transformation of des-acyl-ghrelin (DAG) to the active form acyl-ghrelin (AG). DAG has been demonstrated to show antagonist properties; it is metabolically active, and counteracts the effects of AG on glucose metabolism and lipolysis, and reduces food consumption, body weight, and hedonic feeding response. Both peptides seem to influence the hypothalamic-pituitary-adrenal (HPA) axis and the corticosterone/cortisol level that drive the urge to eat under stressful conditions. These findings suggest that DAG and inhibition of GOAT may be targets for obesity and bingeing-related eating disorders and that AG/DAG ratio may be an important potential biomarker to assess the risk of developing maladaptive eating behaviors.
Collapse
Affiliation(s)
| | - Luca Botticelli
- School of Pharmacy, Pharmacology Unit, University of Camerino, via Madonna delle Carceri, 9, 62032 Camerino, Italy
| | - Fabio Del Bello
- School of Pharmacy, Medicinal Chemistry Unit, University of Camerino, via S. Agostino, 1, 62032 Camerino, Italy
| | - Gianfabio Giorgioni
- School of Pharmacy, Medicinal Chemistry Unit, University of Camerino, via S. Agostino, 1, 62032 Camerino, Italy
| | - Alessandro Piergentili
- School of Pharmacy, Medicinal Chemistry Unit, University of Camerino, via S. Agostino, 1, 62032 Camerino, Italy
| | - Wilma Quaglia
- School of Pharmacy, Medicinal Chemistry Unit, University of Camerino, via S. Agostino, 1, 62032 Camerino, Italy
| | - Carlo Cifani
- School of Pharmacy, Pharmacology Unit, University of Camerino, via Madonna delle Carceri, 9, 62032 Camerino, Italy.
| | | |
Collapse
|
37
|
Decoding the Role of Gut-Microbiome in the Food Addiction Paradigm. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2021; 18:ijerph18136825. [PMID: 34202073 PMCID: PMC8297196 DOI: 10.3390/ijerph18136825] [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: 05/11/2021] [Revised: 06/22/2021] [Accepted: 06/23/2021] [Indexed: 12/12/2022]
Abstract
Eating behaviour is characterised by a solid balance between homeostatic and hedonic regulatory mechanisms at the central level and highly influenced by peripheral signals. Among these signals, those generated by the gut microbiota have achieved relevance in recent years. Despite this complex regulation, under certain circumstances eating behaviour can be deregulated becoming addictive. Although there is still an ongoing debate about the food addiction concept, studies agree that patients with eating addictive behaviour present similar symptoms to those experienced by drug addicts, by affecting central areas involved in the control of motivated behaviour. In this context, this review tries to summarise the main data regarding the role of the gut microbiome in eating behaviour and how a gut dysbiosis can be responsible for a maladaptive behaviour such as “food addiction”.
Collapse
|
38
|
Li J, Zhu S, Lv Z, Dai H, Wang Z, Wei Q, Hamdard E, Mustafa S, Shi F, Fu Y. Drinking Water with Saccharin Sodium Alters the Microbiota-Gut-Hypothalamus Axis in Guinea Pig. Animals (Basel) 2021; 11:1875. [PMID: 34201842 PMCID: PMC8300211 DOI: 10.3390/ani11071875] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2021] [Revised: 06/20/2021] [Accepted: 06/21/2021] [Indexed: 01/22/2023] Open
Abstract
The effects of saccharin, as a type of sweetener additive, on the metabolism and development of mammals are still controversial. Our previous research revealed that saccharin sodium (SS) promoted the feed intake and growth of guinea pigs. In this experiment, we used the guinea pig model to study the physiological effect of SS in the microbiota-gut-hypothalamus axis. Adding 1.5 mM SS to drinking water increased the serum level of glucose, followed by the improvement in the morphology and barrier function of the ileal villus, such as SS supplementation which increased the villus height and villus height/crypt depth ratio. Saccharin sodium (SS) treatment activated the sweet receptor signaling in the ileum and altered GHRP hormone secretion. In the hypothalamus of SS and control (CN) group, RNA-seq identified 1370 differently expressed genes (796 upregulated, 574 downregulated), enriching into the taste signaling transduction, and neuroactive ligand-receptor interaction. LEfSe analysis suggested that Lactobacillaceae-Lactobacillus was the microbe with significantly increased abundance of ileum microorganisms in the SS-treated group, while Brevinema-Andersonii and Erysipelotrichaceae-Ilebacterium were the microbes with significantly increased abundance of the control. Furthermore, SS treatment significantly enhanced the functions of chemoheterotrophy and fermentation of ileal microflora compared to the CN group. Accordingly, SS treatment increased levels of lactic acid and short-chain fatty acids (acetic acid, propionic acid and N-valeric acid) in the ileal digesta. In summary, drinking water with 1.5 mM SS activated sweet receptor signaling in the gut and altered GHRP hormone secretion, followed by the taste signaling transduction in the hypothalamus.
Collapse
Affiliation(s)
- Junrong Li
- College of Animal Science, Zhejiang University, Hangzhou 310058, China;
- College of Agriculture, Jinhua Polytechnic, Jinhua 321000, China;
| | - Shanli Zhu
- College of Agriculture, Jinhua Polytechnic, Jinhua 321000, China;
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China; (Z.L.); (H.D.); (Z.W.); (Q.W.); (E.H.); (S.M.)
| | - Zengpeng Lv
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China; (Z.L.); (H.D.); (Z.W.); (Q.W.); (E.H.); (S.M.)
| | - Hongjian Dai
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China; (Z.L.); (H.D.); (Z.W.); (Q.W.); (E.H.); (S.M.)
| | - Zhe Wang
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China; (Z.L.); (H.D.); (Z.W.); (Q.W.); (E.H.); (S.M.)
| | - Quanwei Wei
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China; (Z.L.); (H.D.); (Z.W.); (Q.W.); (E.H.); (S.M.)
| | - Enayatullah Hamdard
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China; (Z.L.); (H.D.); (Z.W.); (Q.W.); (E.H.); (S.M.)
| | - Sheeraz Mustafa
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China; (Z.L.); (H.D.); (Z.W.); (Q.W.); (E.H.); (S.M.)
| | - Fangxiong Shi
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China; (Z.L.); (H.D.); (Z.W.); (Q.W.); (E.H.); (S.M.)
| | - Yan Fu
- College of Animal Science, Zhejiang University, Hangzhou 310058, China;
| |
Collapse
|
39
|
Hypothalamic Actions of SIRT1 and SIRT6 on Energy Balance. Int J Mol Sci 2021; 22:ijms22031430. [PMID: 33572672 PMCID: PMC7866978 DOI: 10.3390/ijms22031430] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2020] [Revised: 01/14/2021] [Accepted: 01/28/2021] [Indexed: 02/07/2023] Open
Abstract
Sirtuins are NAD+ dependent deacetylases that regulate a large number of physiological processes. These enzymes are highly conserved and act as energy sensors to coordinate different metabolic responses in a controlled manner. At present, seven mammalian sirtuins (SIRT 1-7) have been identified, with SIRT1 and SIRT6 shown to exert their metabolic actions in the hypothalamus, both with crucial roles in eliciting responses to dampen metabolic complications associated with obesity. Therefore, our aim is to compile the current understanding on the role of SIRT1 and SIRT6 in the hypothalamus, especially highlighting their actions on the control of energy balance.
Collapse
|
40
|
Khelifa MS, Skov LJ, Holst B. Biased Ghrelin Receptor Signaling and the Dopaminergic System as Potential Targets for Metabolic and Psychological Symptoms of Anorexia Nervosa. Front Endocrinol (Lausanne) 2021; 12:734547. [PMID: 34646236 PMCID: PMC8503187 DOI: 10.3389/fendo.2021.734547] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/01/2021] [Accepted: 08/16/2021] [Indexed: 12/15/2022] Open
Abstract
Anorexia Nervosa (AN) is a complex disease that impairs the metabolic, mental and physiological health of affected individuals in a severe and sometimes lethal way. Many of the common symptoms in AN patients, such as reduced food intake, anxiety, impaired gut motility or overexercising are connected to both the orexigenic gut hormone ghrelin and the dopaminergic system. Targeting the ghrelin receptor (GhrR) to treat AN seems a promising possibility in current research. However, GhrR signaling is highly complex. First, the GhrR can activate four known intracellular pathways Gαq, Gαi/o, Gα12/13 and the recruitment of β-arrestin. Biased signaling provides the possibility to activate or inhibit only one or a subset of the intracellular pathways of a pleiotropic receptor. This allows specific targeting of physiological functions without adverse effects. Currently little is known on how biased signaling could specifically modulate GhrR effects. Second, GhrR signaling has been shown to be interconnected with the dopaminergic system, particularly in the context of AN symptoms. This review highlights that a biased agonist for the GhrR may be a promising target for the treatment of AN, however extensive and systematic translational studies are still needed and the connection to the dopaminergic system has to be taken into account.
Collapse
|
41
|
"A LEAP 2 conclusions? Targeting the ghrelin system to treat obesity and diabetes". Mol Metab 2020; 46:101128. [PMID: 33246141 PMCID: PMC8085568 DOI: 10.1016/j.molmet.2020.101128] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/30/2020] [Revised: 11/15/2020] [Accepted: 11/20/2020] [Indexed: 02/08/2023] Open
Abstract
BACKGROUND The hormone ghrelin stimulates food intake, promotes adiposity, increases body weight, and elevates blood glucose. Consequently, alterations in plasma ghrelin levels and the functioning of other components of the broader ghrelin system have been proposed as potential contributors to obesity and diabetes. Furthermore, targeting the ghrelin system has been proposed as a novel therapeutic strategy for obesity and diabetes. SCOPE OF REVIEW The current review focuses on the potential for targeting ghrelin and other proteins comprising the ghrelin system as a treatment for obesity and diabetes. The main components of the ghrelin system are introduced. Data supporting a role for the endogenous ghrelin system in the development of obesity and diabetes along with data that seemingly refute such a role are outlined. An argument for further research into the development of ghrelin system-targeted therapeutic agents is delineated. Also, an evidence-based discussion of potential factors and contexts that might influence the efficacy of this class of therapeutics is provided. MAJOR CONCLUSIONS It would not be a "leap to" conclusions to suggest that agents which target the ghrelin system - including those that lower acyl-ghrelin levels, raise LEAP2 levels, block GHSR activity, and/or raise desacyl-ghrelin signaling - could represent efficacious novel treatments for obesity and diabetes.
Collapse
|
42
|
Fritz EM, Singewald N, De Bundel D. The Good, the Bad and the Unknown Aspects of Ghrelin in Stress Coping and Stress-Related Psychiatric Disorders. Front Synaptic Neurosci 2020; 12:594484. [PMID: 33192444 PMCID: PMC7652849 DOI: 10.3389/fnsyn.2020.594484] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2020] [Accepted: 09/14/2020] [Indexed: 12/16/2022] Open
Abstract
Ghrelin is a peptide hormone released by specialized X/A cells in the stomach and activated by acylation. Following its secretion, it binds to ghrelin receptors in the periphery to regulate energy balance, but it also acts on the central nervous system where it induces a potent orexigenic effect. Several types of stressors have been shown to stimulate ghrelin release in rodents, including nutritional stressors like food deprivation, but also physical and psychological stressors such as foot shocks, social defeat, forced immobilization or chronic unpredictable mild stress. The mechanism through which these stressors drive ghrelin release from the stomach lining remains unknown and, to date, the resulting consequences of ghrelin release for stress coping remain poorly understood. Indeed, ghrelin has been proposed to act as a stress hormone that reduces fear, anxiety- and depression-like behaviors in rodents but some studies suggest that ghrelin may - in contrast - promote such behaviors. In this review, we aim to provide a comprehensive overview of the literature on the role of the ghrelin system in stress coping. We discuss whether ghrelin release is more than a byproduct of disrupted energy homeostasis following stress exposure. Furthermore, we explore the notion that ghrelin receptor signaling in the brain may have effects independent of circulating ghrelin and in what way this might influence stress coping in rodents. Finally, we examine how the ghrelin system could be utilized as a therapeutic avenue in stress-related psychiatric disorders (with a focus on anxiety- and trauma-related disorders), for example to develop novel biomarkers for a better diagnosis or new interventions to tackle relapse or treatment resistance in patients.
Collapse
Affiliation(s)
- Eva Maria Fritz
- Department of Pharmacology and Toxicology, Institute of Pharmacy and CMBI, University of Innsbruck, Innsbruck, Austria
| | - Nicolas Singewald
- Department of Pharmacology and Toxicology, Institute of Pharmacy and CMBI, University of Innsbruck, Innsbruck, Austria
| | - Dimitri De Bundel
- Department of Pharmaceutical Sciences, Research Group Experimental Pharmacology, Center for Neurosciences (C4N), Vrije Universiteit Brussel, Brussels, Belgium
| |
Collapse
|
43
|
Kramer J, Dick DM, King A, Ray LA, Sher KJ, Vena A, Vendruscolo LF, Acion L. Mechanisms of Alcohol Addiction: Bridging Human and Animal Studies. Alcohol Alcohol 2020; 55:603-607. [PMID: 32781467 DOI: 10.1093/alcalc/agaa068] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2019] [Revised: 06/18/2020] [Accepted: 06/19/2020] [Indexed: 12/27/2022] Open
Abstract
AIM The purpose of this brief narrative review is to address the complexities and benefits of extending animal alcohol addiction research to the human domain, emphasizing Allostasis and Incentive Sensitization, two models that inform many pre-clinical and clinical studies. METHODS The work reviewed includes a range of approaches, including: a) animal and human studies that target the biology of craving and compulsive consumption; b) human investigations that utilize alcohol self-administration and alcohol challenge paradigms, in some cases across 10 years; c) questionnaires that document changes in the positive and negative reinforcing effects of alcohol with increasing severity of addiction; and d) genomic structural equation modeling based on data from animal and human studies. RESULTS Several general themes emerge from specific study findings. First, positive reinforcement is characteristic of early stage addiction and sometimes diminishes with increasing severity, consistent with both Allostasis and Incentive Sensitization. Second, evidence is less consistent for the predominance of negative reinforcement in later stages of addiction, a key tenant of Allostasis. Finally, there are important individual differences in motivation to drink at a given point in time as well as person-specific change patterns across time. CONCLUSIONS Key constructs of addiction, like stage and reinforcement, are by necessity operationalized differently in animal and human studies. Similarly, testing the validity of addiction models requires different strategies by the two research domains. Although such differences are challenging, they are not insurmountable, and there is much to be gained in understanding and treating addiction by combining pre-clinical and clinical approaches.
Collapse
Affiliation(s)
- John Kramer
- Department of Psychiatry, University of Iowa Carver College of Medicine, 200 Hawkins Dr, 1882JPP, Iowa City, IA 52242-1009, USA
| | - Danielle M Dick
- Department of Psychology, Virginia Commonwealth University, 612 N. Lombardy St., Richmond, VA 23284, USA.,Department Human and Molecular Genetics, Virginia Commonwealth University, 806 West Franklin Street, Box 842018, Richmond, VA 23284, USA
| | - Andrea King
- Department of Psychiatry and Behavioral Neuroscience, University of Chicago, 5841 S. Maryland Ave., Room L470, Chicago, IL 60637, USA
| | - Lara A Ray
- Department of Psychology, UCLA, 1285 Franz Hall, Los Angeles, CA 90095, USA
| | - Kenneth J Sher
- Department of Psychological Sciences, University of Missouri, 210 McAlester Hall, Columbia, MO 65211, USA
| | - Ashley Vena
- Department of Psychiatry and Behavioral Neuroscience, University of Chicago, 5841 S. Maryland Ave., Room L470, Chicago, IL 60637, USA
| | - Leandro F Vendruscolo
- Neurobiology of Addiction Section, National Institute on Drug Abuse Intramural Research Program, National Institutes of Health, 251 Bayview Boulevard, Baltimore, MD 21224, USA
| | - Laura Acion
- Department of Psychiatry, University of Iowa Carver College of Medicine, 200 Hawkins Dr, 1882JPP, Iowa City, IA 52242-1009, USA.,Instituto de Cálculo, Universidad de Buenos Aires-CONICET, Intendente Güiraldes 2160, C1428EGA, Buenos Aires, Argentina
| |
Collapse
|
44
|
Pierre A, Van Schuerbeek A, Allaoui W, Van Laere S, Singewald N, Van Eeckhaut A, Smolders I, De Bundel D. Effects of ghrelin receptor activation on forebrain dopamine release, conditioned fear and fear extinction in C57BL/6J mice. J Neurochem 2020; 154:389-403. [DOI: 10.1111/jnc.14996] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2019] [Revised: 02/25/2020] [Accepted: 02/26/2020] [Indexed: 12/14/2022]
Affiliation(s)
- Anouk Pierre
- Department of Pharmaceutical Sciences Research Group Experimental Pharmacology Center for Neurosciences (C4N) Vrije Universiteit Brussel Brussels Belgium
| | - Andries Van Schuerbeek
- Department of Pharmaceutical Sciences Research Group Experimental Pharmacology Center for Neurosciences (C4N) Vrije Universiteit Brussel Brussels Belgium
| | - Wissal Allaoui
- Department of Pharmaceutical Sciences Research Group Experimental Pharmacology Center for Neurosciences (C4N) Vrije Universiteit Brussel Brussels Belgium
| | - Sven Van Laere
- Interfaculty Center Data Processing & Statistics Vrije Universiteit Brussel Brussels Belgium
| | - Nicolas Singewald
- Department of Pharmacology and Toxicology Institute of Pharmacy and CMBI University of Innsbruck Innsbruck Austria
| | - Ann Van Eeckhaut
- Department of Pharmaceutical Sciences Research Group Experimental Pharmacology Center for Neurosciences (C4N) Vrije Universiteit Brussel Brussels Belgium
| | - Ilse Smolders
- Department of Pharmaceutical Sciences Research Group Experimental Pharmacology Center for Neurosciences (C4N) Vrije Universiteit Brussel Brussels Belgium
| | - Dimitri De Bundel
- Department of Pharmaceutical Sciences Research Group Experimental Pharmacology Center for Neurosciences (C4N) Vrije Universiteit Brussel Brussels Belgium
| |
Collapse
|
45
|
Gut and Metabolic Hormones Changes After Endoscopic Sleeve Gastroplasty (ESG) Vs. Laparoscopic Sleeve Gastrectomy (LSG). Obes Surg 2020; 30:2642-2651. [DOI: 10.1007/s11695-020-04541-0] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
|
46
|
Abstract
Non-alcoholic fatty liver disease (NAFLD) and non-alcoholic steatohepatitis (NASH) are two of the most common liver diseases associated with obesity, type 2 diabetes and metabolic syndrome. The prevalence of these conditions are increasingly rising and presently there is not a pharmacological option available in the market. Elucidation of the mechanism of action and the molecular underpinnings behind liver disease could help to better understand the pathophysiology of these illnesses. In this sense, in the last years modulation of the ghrelin system in preclinical animal models emerge as a promising therapeutic tool. In this review, we compile the latest knowledge of the modulation of ghrelin system and its intracellular pathways that regulates lipid metabolism, hepatic inflammation and liver fibrosis. We also describe novel processes implicated in the regulation of liver disease by ghrelin, such as autophagy or dysregulated circadian rhythms. In conclusion, the information displayed in this review support that the ghrelin system could be an appealing strategy for the treatment of liver disease.
Collapse
Affiliation(s)
- Mar Quiñones
- Department of Physiology, CIMUS, University of Santiago de Compostela-Instituto de Investigación Sanitaria, 15782, Santiago de Compostela, Spain
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), 15706, Santiago de Compostela, Spain
| | - Johan Fernø
- Hormone Laboratory, Haukeland University Hospital, Bergen, Norway
| | - Omar Al-Massadi
- Inserm UMR-S1270, 75005, Paris, France.
- Faculté des Sciences et d'Ingénierie, Sorbonne Université, 75005, Paris, France.
- Institut du Fer a Moulin, Inserm, 17 rue du Fer à Moulin, 75005, Paris, France.
| |
Collapse
|
47
|
Neasta J, Darcq E, Jeanblanc J, Carnicella S, Ben Hamida S. GPCR and Alcohol-Related Behaviors in Genetically Modified Mice. Neurotherapeutics 2020; 17:17-42. [PMID: 31919661 PMCID: PMC7007453 DOI: 10.1007/s13311-019-00828-y] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
G protein-coupled receptors (GPCRs) constitute the largest class of cell surface signaling receptors and regulate major neurobiological processes. Accordingly, GPCRs represent primary targets for the treatment of brain disorders. Several human genetic polymorphisms affecting GPCRs have been associated to different components of alcohol use disorder (AUD). Moreover, GPCRs have been reported to contribute to several features of alcohol-related behaviors in animal models. Besides traditional pharmacological tools, genetic-based approaches mostly aimed at deleting GPCR genes provided substantial information on how key GPCRs drive alcohol-related behaviors. In this review, we summarize the alcohol phenotypes that ensue from genetic manipulation, in particular gene deletion, of key GPCRs in rodents. We focused on GPCRs that belong to fundamental neuronal systems that have been shown as potential targets for the development of AUD treatment. Data are reviewed with particular emphasis on alcohol reward, seeking, and consumption which are behaviors that capture essential aspects of AUD. Literature survey indicates that in most cases, there is still a gap in defining the intracellular transducers and the functional crosstalk of GPCRs as well as the neuronal populations in which their signaling regulates alcohol actions. Further, the implication of only a few orphan GPCRs has been so far investigated in animal models. Combining advanced pharmacological technologies with more specific genetically modified animals and behavioral preclinical models is likely necessary to deepen our understanding in how GPCR signaling contributes to AUD and for drug discovery.
Collapse
Affiliation(s)
- Jérémie Neasta
- Laboratoire de Pharmacologie, Faculté de Pharmacie, University of Montpellier, 34093, Montpellier, France
| | - Emmanuel Darcq
- Douglas Hospital Research Center, Department of Psychiatry, McGill University, 6875 Boulevard LaSalle, Montreal, Quebec, H4H 1R3, Canada
| | - Jérôme Jeanblanc
- Research Group on Alcohol and Pharmacodependences-INSERM U1247, University of Picardie Jules Verne, 80025, Amiens, France
| | - Sebastien Carnicella
- INSERM U1216, Grenoble Institut des Neurosciences (GIN), University of Grenoble Alpes, 38000, Grenoble, France
| | - Sami Ben Hamida
- Douglas Hospital Research Center, Department of Psychiatry, McGill University, 6875 Boulevard LaSalle, Montreal, Quebec, H4H 1R3, Canada.
| |
Collapse
|
48
|
Duriez P, Ramoz N, Gorwood P, Viltart O, Tolle V. A Metabolic Perspective on Reward Abnormalities in Anorexia Nervosa. Trends Endocrinol Metab 2019; 30:915-928. [PMID: 31648936 DOI: 10.1016/j.tem.2019.08.004] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/16/2019] [Revised: 08/04/2019] [Accepted: 08/08/2019] [Indexed: 12/17/2022]
Abstract
Anorexia nervosa (AN) is the psychiatric disorder with the highest mortality rate; however, the mechanisms responsible for its pathogenesis remain largely unknown. Large-scale genome-wide association studies (GWAS) have identified genetic loci associated with metabolic features in AN. Metabolic alterations that occur in AN have been mostly considered as consequences of the chronic undernutrition state but until recently have not been linked to the etiology of the disorder. We review the molecular basis of AN based on human genetics, with an emphasis on the molecular components controlling energy homeostasis, highlight the main metabolic and endocrine alterations occurring in AN, and decipher the possible connection between metabolic factors and abnormalities of reward processes that are central in AN.
Collapse
Affiliation(s)
- Philibert Duriez
- Institute of Psychiatry and Neurosciences of Paris, Unité Mixte de Recherche en Santé (UMRS) 1266 Institut National de la Santé et de la Recherche Médicale (INSERM), University Paris Descartes, Paris, France; Clinique des Maladies Mentales et de l'Encéphale, Groupement Hospitalier Universitaire (GHU) Paris Psychiatry and Neuroscience, Sainte-Anne Hospital, Paris, France
| | - Nicolas Ramoz
- Institute of Psychiatry and Neurosciences of Paris, Unité Mixte de Recherche en Santé (UMRS) 1266 Institut National de la Santé et de la Recherche Médicale (INSERM), University Paris Descartes, Paris, France
| | - Philip Gorwood
- Institute of Psychiatry and Neurosciences of Paris, Unité Mixte de Recherche en Santé (UMRS) 1266 Institut National de la Santé et de la Recherche Médicale (INSERM), University Paris Descartes, Paris, France; Clinique des Maladies Mentales et de l'Encéphale, Groupement Hospitalier Universitaire (GHU) Paris Psychiatry and Neuroscience, Sainte-Anne Hospital, Paris, France
| | - Odile Viltart
- Institute of Psychiatry and Neurosciences of Paris, Unité Mixte de Recherche en Santé (UMRS) 1266 Institut National de la Santé et de la Recherche Médicale (INSERM), University Paris Descartes, Paris, France; University of Lille, Lille, France
| | - Virginie Tolle
- Institute of Psychiatry and Neurosciences of Paris, Unité Mixte de Recherche en Santé (UMRS) 1266 Institut National de la Santé et de la Recherche Médicale (INSERM), University Paris Descartes, Paris, France.
| |
Collapse
|