1
|
Sudar FP, Zekerallah SS, Paulzen M, Mathiak K, Gaebler AJ. Unraveling antipsychotic induced weight gain in schizophrenia - A proof-of-concept study exploring the impact of the cumulative historical occupancy of different receptors by antipsychotics. Psychiatry Res 2025; 348:116452. [PMID: 40147087 DOI: 10.1016/j.psychres.2025.116452] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/26/2024] [Revised: 03/16/2025] [Accepted: 03/19/2025] [Indexed: 03/29/2025]
Abstract
Obesity is a common complication in schizophrenia contributing to increased mortality rates. We present a proof-of-concept study displaying a new method to investigate the impact of antipsychotic drugs (APs) on obesity in terms of their cumulative historical receptor occupancy (CHRO) in 150 selected from 174 patients with schizophrenia. Based on a thorough medication history, we estimated CHRO of serotonin 5-HT2C, histamine H1, dopamine D2 and muscarinic M3 receptors and studied their relationship with different metabolic outcome variables utilizing stepwise regression analysis and structural equation modelling (SEM). Stepwise regression analysis revealed a significant positive relationship of Body Mass Index (BMI) with H1-CHRO, but a negative relationship with M3-CHRO. Moreover, H1-CHRO was associated with increased triglyceride concentration, while 5-HT2C-CHRO was associated with increased waist circumference and blood pressure. SEM, while confirming the diverging effects of H1-/5-HT2C- and M3-CHRO on obesity, suggested that their effect on other metabolic variables was indirect, i.e. mediated by obesity. Our results suggest that the metabolic side effects of antipsychotics can be described by their cumulative historical receptor occupancy with unique contributions of the different receptors. In particular, M3 receptor antagonism seems to exert a protective effect, confirming findings from rodent M3 receptor knock out models. These findings may provide a framework for estimating the metabolic burden of future APs, guiding the development of drugs with more favorable metabolic profiles.
Collapse
Affiliation(s)
- Federico Pacheco Sudar
- Department of Psychiatry, Psychotherapy and Psychosomatics, Faculty of Medicine, RWTH Aachen, Germany; JARA - Translational Brain Medicine, Aachen, Germany
| | - Samar Samy Zekerallah
- Department of Psychiatry, Psychotherapy and Psychosomatics, Faculty of Medicine, RWTH Aachen, Germany; JARA - Translational Brain Medicine, Aachen, Germany
| | - Michael Paulzen
- Department of Psychiatry, Psychotherapy and Psychosomatics, Faculty of Medicine, RWTH Aachen, Germany; JARA - Translational Brain Medicine, Aachen, Germany; Alexianer Hospital, Aachen, Germany
| | - Klaus Mathiak
- Department of Psychiatry, Psychotherapy and Psychosomatics, Faculty of Medicine, RWTH Aachen, Germany; JARA - Translational Brain Medicine, Aachen, Germany
| | - Arnim Johannes Gaebler
- Department of Psychiatry, Psychotherapy and Psychosomatics, Faculty of Medicine, RWTH Aachen, Germany; JARA - Translational Brain Medicine, Aachen, Germany; Institute for Neurophysiology, Faculty of Medicine, RWTH Aachen, Germany.
| |
Collapse
|
2
|
Dacaya P, Sarapis K, Moschonis G. The Role and Mechanisms of Probiotic Supplementation on Depressive Symptoms: A Narrative Review. Curr Nutr Rep 2025; 14:53. [PMID: 40153103 PMCID: PMC11953144 DOI: 10.1007/s13668-025-00644-1] [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] [Accepted: 03/14/2025] [Indexed: 03/30/2025]
Abstract
PURPOSE OF REVIEW The microbiota-gut-brain-axis (MGBA) plays a role in the aetiology of mental disorders. Depression, a leading cause of disability worldwide, may be improved by probiotics. The aim of this narrative review is to investigate and synthesize the current evidence linking probiotic food supplementation with depressive symptomology. RECENT FINDINGS The gut and the brain communicate and interact via the MGBA through inflammation and the immune system, short chain fatty acid production, neuronal innervation and activation as well as endocrine and neurotransmitter modulation. Dysregulation of gut-brain pathways are caused by gut dysbiosis and implicated in the onset, persistence and exacerbation of depression related symptoms. Modulation of the gut microbiota via administration of probiotics has shown to reduce depressive symptom severity with Bifidobacterium and Lactobacillus strains being the most reported. Probiotics may produce greater benefits in mild depression rather than in chronic, treatment resistant depression. Probiotic supplementation is a promising and safe approach for the prevention of severe depressive disorders in high-risk individuals such as people with subthreshold depression. However, the mechanistic pathways of the MGBA require further investigation and additional human clinical trials are necessary to evaluate the role of probiotics on depression.
Collapse
Affiliation(s)
- Pauline Dacaya
- Discipline of Food, Nutrition and Dietetics, Department of Sport, Exercise and Nutrition Sciences, School of Allied Health, Human Services and Sport, La Trobe University, Melbourne, VIC, 3086, Australia
| | - Katerina Sarapis
- Discipline of Food, Nutrition and Dietetics, Department of Sport, Exercise and Nutrition Sciences, School of Allied Health, Human Services and Sport, La Trobe University, Melbourne, VIC, 3086, Australia
| | - George Moschonis
- Discipline of Food, Nutrition and Dietetics, Department of Sport, Exercise and Nutrition Sciences, School of Allied Health, Human Services and Sport, La Trobe University, Melbourne, VIC, 3086, Australia.
| |
Collapse
|
3
|
Sun X, Liu B, Yuan Y, Rong Y, Pang R, Li Q. Neural and hormonal mechanisms of appetite regulation during eating. Front Nutr 2025; 12:1484827. [PMID: 40201582 PMCID: PMC11977392 DOI: 10.3389/fnut.2025.1484827] [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: 08/22/2024] [Accepted: 03/07/2025] [Indexed: 04/10/2025] Open
Abstract
Numerous animal and clinical studies have demonstrated that the arcuate nucleus of the hypothalamus, a central regulator of appetite, plays a significant role in modulating feeding behavior. However, current research primarily focuses on long-term dietary changes and their effects on the body, with limited investigation into neuroendocrine dynamics during individual meals across diverse populations. In contrast to long-term dietary adjustments, directives for dietary behavior during a specific meal are more actionable, potentially enhancing patient adherence and achieving better outcomes in dietary behavior interventions. This review aimed to explore the neural pathways and endocrine changes activated by gastrointestinal expansion and variations in blood nutrient levels during a single meal, with the goal of informing dietary behavior guidance.
Collapse
Affiliation(s)
- Xurui Sun
- Key Laboratory of Endocrine Glucose and Lipids Metabolism and Brain Aging, Ministry of Education; Department of Endocrinology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Binghan Liu
- Department of Neurosurgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Yuan Yuan
- Department of Clinical Nutrition, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Ying Rong
- Department of Clinical Nutrition, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Rui Pang
- Key Laboratory of Endocrine Glucose and Lipids Metabolism and Brain Aging, Ministry of Education; Department of Endocrinology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Qiu Li
- Key Laboratory of Endocrine Glucose and Lipids Metabolism and Brain Aging, Ministry of Education; Department of Endocrinology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| |
Collapse
|
4
|
Rapps K, Weller A, Meiri N. Epigenetic regulation is involved in reversal of obesity. Neurosci Biobehav Rev 2024; 167:105906. [PMID: 39343077 DOI: 10.1016/j.neubiorev.2024.105906] [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: 07/01/2024] [Revised: 09/10/2024] [Accepted: 09/23/2024] [Indexed: 10/01/2024]
Abstract
Epigenetic processes play a crucial role in mediating the impact of environmental energetic challenges, from overconsumption to starvation. Over-nutrition of energy-dense foods and sedentary lifestyles contribute to the development of obesity, characterized by excessive fat storage and impaired metabolic signaling, stemming from disrupted brain signaling. Conversely, dieting and physical activity facilitate body weight rebalancing and trigger adaptive neural responses. These adaptations involve the upregulation of neurogenesis, synaptic plasticity and optimized brain function and energy homeostasis, balanced hormone signaling, normal metabolism, and reduced inflammation. The transition of the brain from a maladaptive to an adaptive state is partially guided by epigenetic mechanisms. While epigenetic mechanisms underlying obesity-related brain changes have been described, their role in mediating the reversal of maladaptation/obesity through lifestyle interventions remains less explored. This review focuses on elucidating epigenetic mechanisms involved in hypothalamic adaptations induced by lifestyle interventions. Given that lifestyle interventions are widely prescribed and accessible approaches for weight loss and maintenance, it is our challenge to uncover epigenetic mechanisms moderating these hypothalamic-functional beneficial changes.
Collapse
Affiliation(s)
- Kayla Rapps
- Faculty of Life Sciences, Bar Ilan University, Ramat-Gan, Israel; Institute of Animal Science, Agricultural Research Organization, The Volcani Center, Bet Dagan, Israel; Gonda Multidisciplinary Brain Research Center, Bar Ilan University, Ramat-Gan, Israel
| | - Aron Weller
- Gonda Multidisciplinary Brain Research Center, Bar Ilan University, Ramat-Gan, Israel; Department of Psychology, Bar Ilan University, Ramat-Gan, Israel
| | - Noam Meiri
- Institute of Animal Science, Agricultural Research Organization, The Volcani Center, Bet Dagan, Israel.
| |
Collapse
|
5
|
Davenport JB, Güler AD, Zhang Q. Methodology for Studying Hypothalamic Regulation of Feeding Behaviors. Methods Protoc 2024; 7:86. [PMID: 39584979 PMCID: PMC11586955 DOI: 10.3390/mps7060086] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2024] [Revised: 10/02/2024] [Accepted: 10/16/2024] [Indexed: 11/26/2024] Open
Abstract
Continuous advances in neurological research techniques are enabling researchers to further understand the neural mechanisms that regulate energy balance. In this review, we specifically highlight key tools and techniques and explore how they have been applied to study the role of the hypothalamic arcuate nucleus in feeding behaviors. Additionally, we provide a detailed discussion of the advantages and limitations associated with each methodology.
Collapse
Affiliation(s)
- Julia B. Davenport
- Department of Biology, University of Virginia, Charlottesville, VA 22904, USA; (J.B.D.)
| | - Ali D. Güler
- Department of Biology, University of Virginia, Charlottesville, VA 22904, USA; (J.B.D.)
- Program in Fundamental Neuroscience, Charlottesville, VA 22904, USA
- Department of Neuroscience, School of Medicine, University of Virginia, Charlottesville, VA 22903, USA
| | - Qi Zhang
- Department of Biology, University of Virginia, Charlottesville, VA 22904, USA; (J.B.D.)
| |
Collapse
|
6
|
Svendstrup M, Rasmussen AK, Kistorp C, Klose M, Andreassen M. Semaglutide treatment of hypothalamic obesity - a real-life data study. Pituitary 2024; 27:685-692. [PMID: 39120810 PMCID: PMC11513754 DOI: 10.1007/s11102-024-01429-5] [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] [Accepted: 07/09/2024] [Indexed: 08/10/2024]
Abstract
PURPOSE Patients with tumors involving the hypothalamic region are at high risk of developing morbid obesity due to disturbances in the appetite regulative nuclei in hypothalamus. We evaluated the effect of the Glucagon-like peptide 1 (GLP-1) analogue semaglutide in patients with hypothalamic obesity. METHODS We recorded weight changes from real-time data before and after treatment with semaglutide in patients with hypothalamic obesity from our outpatient clinic at the Department of Endocrinology at Rigshospitalet, from September 2020 to November 2023. RESULTS A total of 26 patients were included in this study (15 females, median age at initiation of semaglutide was 52 (range 18-65) years). Body mass index (BMI) at initial diagnosis was median 25 (range 20-38) kg/m2 while BMI at initiation of semaglutide was median 38 (range 28-58) kg/m2. All but one patient lost weight during semaglutide treatment with a mean weight loss of 13.4 kg (95% CI 10.3-16.5 kg, p = < 0.001) after 12 months corresponding to a loss in BMI of 4.4 kg/m2 (95% CI 3.4-5.4 kg/m2, p = < 0.001) with a median dosage of semaglutide of 1.6 (range 0.5-2.5) mg. Fifteen patients (58%) lost more than 10% and two patients (8%) lost more than 20% of initial body weight, respectively. CONCLUSION Treatment with semaglutide shows promising results in reducing body weight in patients with acquired hypothalamic obesity. Whether the weight reduction remains stable after long time follow-up needs further investigation.
Collapse
Affiliation(s)
- Mathilde Svendstrup
- Department of Endocrinology, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark.
| | - Aase Krogh Rasmussen
- Department of Endocrinology, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark
| | - Caroline Kistorp
- Department of Endocrinology, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark
| | - Marianne Klose
- Department of Endocrinology, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark
| | - Mikkel Andreassen
- Department of Endocrinology, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark
| |
Collapse
|
7
|
González-Flores O, Domínguez-Ordóñez R, Delgado-Macuil RJ, Tlachi-López JL, Luna-Hernández A, Montes-Narváez O, Pfaus JG, García-Juárez M. Participation of kisspeptin, progesterone, and GnRH receptors on lordosis behavior induced by kisspeptin. Physiol Behav 2024; 283:114609. [PMID: 38851441 DOI: 10.1016/j.physbeh.2024.114609] [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: 04/04/2024] [Revised: 05/27/2024] [Accepted: 06/05/2024] [Indexed: 06/10/2024]
Abstract
The neuropeptide kisspeptin (Kiss) is crucial in regulating the hypothalamic-pituitary-gonadal axis. It is produced by two main groups of neurons in the hypothalamus: the rostral periventricular region around the third ventricle and the arcuate nucleus. Kiss is the peptide product of the KiSS-1 gene and serves as the endogenous agonist for the GPR54 receptor. The Kiss/GPR54 system functions as a critical regulator of the reproductive system. Thus, we examined the effect of intracerebroventricular administration of 3 μg of Kiss to the right lateral ventricle of ovariectomized rats primed with a dose of 5 μg subcutaneous (sc) of estradiol benzoate (EB). Kiss treatment increased the lordosis quotient at all times tested. However, the lordosis reflex score was comparatively lower yet still significant compared to the control group. To investigate receptor specificity and downstream mechanisms on lordosis, we infused 10 μg of GPR54 receptor antagonist, Kiss-234, 5 μg of the progestin receptor antagonist, RU486, or 3 μg of antide, a gonadotropin-releasing hormone-1 (GnRH-1) receptor antagonist, to the right lateral ventricle 30 min before an infusion of 3 μg of Kiss. Results demonstrated a significant reduction in the facilitation of lordosis behavior by Kiss at 60 and 120 min when Kiss-234, RU486, or antide were administered. These findings suggest that Kiss stimulates lordosis expression by activating GPR54 receptors on GnRH neurons and that Kiss/GPR54 system is an essential intermediary by which progesterone activates GnRH.
Collapse
Affiliation(s)
- Oscar González-Flores
- Centro de Investigación en Reproducción Animal, Universidad Autónoma de Tlaxcala-CINVESTAV, Tlaxcala, México
| | - Raymundo Domínguez-Ordóñez
- Licenciatura en Ingeniería Agronómica y Zootecnia, CRC, Benemérita Universidad Autónoma de, Puebla, México
| | - Raul Jacobo Delgado-Macuil
- Centro de Investigación en Biotecnología Aplicada, Instituto Politécnico Nacional, Santa Inés, Tecuexcomac, Tlaxcala, México
| | | | - Ailyn Luna-Hernández
- Doctorado en Ciencias Biológicas, Universidad Autónoma de Tlaxcala, Tlaxcala, México
| | - Omar Montes-Narváez
- Doctorado en Ciencias Biológicas, Universidad Autónoma de Tlaxcala, Tlaxcala, México
| | - James G Pfaus
- Center for Sexual Health and Intervention, Czech National Institute of Mental Health, Klecany, Czech Republic; Department of Psychology and Life Sciences, Charles University, Prague, Czech Republic
| | - Marcos García-Juárez
- Centro de Investigación en Reproducción Animal, Universidad Autónoma de Tlaxcala-CINVESTAV, Tlaxcala, México.
| |
Collapse
|
8
|
Lee TH, Cota D, Quarta C. Yin-Yang control of energy balance by lipids in the hypothalamus: The endocannabinoids vs bile acids case. Biochimie 2024; 223:188-195. [PMID: 35863558 DOI: 10.1016/j.biochi.2022.07.006] [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: 05/23/2022] [Revised: 06/22/2022] [Accepted: 07/12/2022] [Indexed: 11/02/2022]
Abstract
Obesity is a chronic and debilitating disorder that originates from alterations in energy-sensing brain circuits controlling body weight gain and food intake. The dysregulated syntheses and actions of lipid mediators in the hypothalamus induce weight gain and overfeeding, but the molecular and cellular underpinnings of these alterations remain elusive. In response to changes in the nutritional status, different lipid sensing pathways in the hypothalamus direct body energy needs in a Yin-Yang model. Endocannabinoids orchestrate the crosstalk between hypothalamic circuits and the sympathetic nervous system to promote food intake and energy accumulation during fasting, whereas bile acids act on the same top-down axis to reduce energy intake and possibly storage after the meal. In obesity, the bioavailability and downstream cellular actions of endocannabinoids and bile acids are altered in hypothalamic neurons involved in body weight and metabolic control. Thus, the onset and progression of this disease might result from an imbalance in hypothalamic sensing of multiple lipid signals, which are possibly integrated by common molecular nodes. In this viewpoint, we discuss a possible model that explains how bile acids and endocannabinoids may exert their effects on energy balance regulation via interconnected mechanisms at the level of the hypothalamic neuronal circuits. Therefore, we propose a new conceptual framework for understanding and treating central mechanisms of maladaptive lipid action in obesity.
Collapse
Affiliation(s)
- Thomas H Lee
- University of Bordeaux, INSERM, Neurocentre Magendie, U1215, F-3300, Bordeaux, France
| | - Daniela Cota
- University of Bordeaux, INSERM, Neurocentre Magendie, U1215, F-3300, Bordeaux, France
| | - Carmelo Quarta
- University of Bordeaux, INSERM, Neurocentre Magendie, U1215, F-3300, Bordeaux, France.
| |
Collapse
|
9
|
Comesaña S, Antomagesh F, Soengas JL, Blanco AM, Vijayan MM. Valine administration in the hypothalamus alters the brain and plasma metabolome in rainbow trout. Am J Physiol Regul Integr Comp Physiol 2024; 327:R261-R273. [PMID: 38881412 DOI: 10.1152/ajpregu.00056.2024] [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: 03/03/2024] [Revised: 05/28/2024] [Accepted: 06/06/2024] [Indexed: 06/18/2024]
Abstract
Central administration of valine has been shown to cause hyperphagia in fish. Although mechanistic target of rapamycin (mTOR) is involved in this response, the contributions to feed intake of central and peripheral metabolite changes due to excess valine are unknown. Here, we investigated whether intracerebroventricular injection of valine modulates central and peripheral metabolite profiles and may provide insights into feeding response in fish. Juvenile rainbow trout (Oncorhynchus mykiss) were administered an intracerebroventricular injection of valine (10 µg·µL-1 at 1 μL·100·g-1 body wt), and the metabolite profile in plasma, hypothalamus, and rest of the brain (composing of telencephalon, optic tectum, cerebellum, and medulla oblongata) was carried out by liquid chromatography-mass spectrometry (LC/MS)-based metabolomics. Valine administration led to a spatially distinct metabolite profile at 1 h postinjection in the brain: enrichment of amino acid metabolism and energy production pathways in the rest of the brain but not in hypothalamus. This suggests a role for extrahypothalamic input in the regulation of feed intake. Also, there was enrichment of several amino acids, including tyrosine, proline, valine, phenylalanine, and methionine, in plasma in response to valine. Changes in liver transcript abundance and protein expression reflect an increased metabolic capacity, including energy production from glucose and fatty acids, and a lower protein kinase B (Akt) phosphorylation in the valine group. Altogether, valine intracerebroventricular administration affects central and peripheral metabolism in rainbow trout, and we propose a role for the altered metabolite profile in modulating the feeding response to this branched-chain amino acid.NEW & NOTEWORTHY Valine causes hyperphagia in fish when it is centrally administered; however, the exact mechanisms are far from clear. We tested how intracerebroventricular injection of valine in rainbow trout affected the brain and plasma metabolome. The metabolite changes in response to valine were more evident in the rest of the brain compared with the hypothalamus. Furthermore, we demonstrated for the first time that central valine administration affects peripheral metabolism in rainbow trout.
Collapse
Affiliation(s)
- Sara Comesaña
- Centro de Investigación Mariña, Laboratorio de Fisioloxía Animal, Departamento de Bioloxía Funcional e Ciencias da Saúde, Facultade de Bioloxía, Universidade de Vigo, Vigo, Spain
| | | | - José L Soengas
- Centro de Investigación Mariña, Laboratorio de Fisioloxía Animal, Departamento de Bioloxía Funcional e Ciencias da Saúde, Facultade de Bioloxía, Universidade de Vigo, Vigo, Spain
| | - Ayelén M Blanco
- Centro de Investigación Mariña, Laboratorio de Fisioloxía Animal, Departamento de Bioloxía Funcional e Ciencias da Saúde, Facultade de Bioloxía, Universidade de Vigo, Vigo, Spain
| | | |
Collapse
|
10
|
Feighan KM, Nesan D, Kurrasch DM. Gestational bisphenol A exposure alters energy homeostasis and adult hypothalamic neurogenesis in female mice. Sci Rep 2024; 14:16082. [PMID: 38992091 PMCID: PMC11239822 DOI: 10.1038/s41598-024-66726-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2023] [Accepted: 07/03/2024] [Indexed: 07/13/2024] Open
Abstract
Regulation of physiological homeostasis, including energy balance, is thought to be modified by low levels of adult neurogenesis in the hypothalamus. Hormones such as oestradiol can influence both embryonic and adult hypothalamic neurogenic programs, demonstrating a sensitivity of hypothalamic neural progenitor cells to endogenous hormones. Previously we showed that gestational exposure to environmental levels of the xenoestrogen bisphenol A (BPA) changed neural progenitor cell behaviors in the embryo; however, we did not examine if these changes were permanent to affect adult neurogenesis. Here we investigated whether adult neuro- and/or gliogenesis were altered in mice prenatally exposed to BPA and placed on a high-fat diet challenge. Gestationally exposed adult female mice on a standard diet gained less weight than non-BPA controls, whereas gestationally exposed BPA females on a high-fat diet gained more weight than controls. Males exposed to gestational BPA showed no differences in weight gain relative to control males. Concomitantly, adult neurogenesis was increased in the VMH, DMH, and PVN of adult female mice exposed to BPA on standard diet, suggesting that disrupted adult neurogenesis might perturb normal energy balance regulation in females. These results add to growing evidence that low-dose BPA exposure in utero causes changes to adult hypothalamic function.
Collapse
Affiliation(s)
- Kira M Feighan
- Department of Medical Genetics, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
- Alberta Children's Hospital Research Institute, University of Calgary, Calgary, AB, Canada
- Hotchkiss Brain Institute, University of Calgary, Calgary, AB, Canada
- Montreal Neurological Institute, McGill University, Montreal, QC, Canada
| | - Dinushan Nesan
- Department of Medical Genetics, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
- Alberta Children's Hospital Research Institute, University of Calgary, Calgary, AB, Canada
- Hotchkiss Brain Institute, University of Calgary, Calgary, AB, Canada
| | - Deborah M Kurrasch
- Department of Medical Genetics, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada.
- Alberta Children's Hospital Research Institute, University of Calgary, Calgary, AB, Canada.
- Hotchkiss Brain Institute, University of Calgary, Calgary, AB, Canada.
| |
Collapse
|
11
|
Oluwagbenga EM, Bergman M, Ajuwon KM, Fraley GS. Sex differences in intestinal morphology and increase in diencephalic neuropeptide Y gene expression in female but not male Pekin ducks exposed to chronic heat stress. J Neuroendocrinol 2024:e13424. [PMID: 38960698 DOI: 10.1111/jne.13424] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/15/2024] [Revised: 06/03/2024] [Accepted: 06/11/2024] [Indexed: 07/05/2024]
Abstract
The impact of heat stress (HS) on production is intricately linked with feed intake. We investigated the effects of HS on intestines and diencephalic genes in Pekin ducks. One hundred and sixty adult ducks were allocated to two treatment rooms. The control room was maintained at 22°C and the HS room at 35°C for the first 10 h of the day then reduced to 29.5°C. After 3 weeks, 10 hens and 5 drakes were euthanized from each room and jejunum and ileum collected for histology. Brains were collected for gene expression analysis using qRT-PCR. Intestinal morphology data were analyzed with two-way ANOVA and diencephalic gene data were analyzed with Kruskal-Wallis test. There was an increase in villi width in the ileum (p = .0136) and jejunum (p = .0019) of HS hens compared to controls. HS drakes showed a higher crypt depth (CD) in the jejunum (p = .0198) compared to controls. There was an increase in crypt goblet cells (GC) count in the ileum (p = .0169) of HS drakes compared to HS hens. There was higher villi GC count (p = .07) in the jejunum of HS drakes compared to controls. There was an increase in the crypt GC density (p = .0054) in the ileum, not jejunum, of HS drakes compared to HS hens. Further, there were no differences in the proopiomelanocortin gene expression in either sex but there was an increase in the expression of neuropeptide Y (NPY) gene in HS hens (p = .031) only and a decrease in the corticotropin releasing hormone gene in the HS drakes (p = .037) compared to controls. These data show that there are sex differences in the effect of HS on gut morphology while the upregulation in NPY gene may suggest a role in mediating response to chronic HS.
Collapse
Affiliation(s)
- E M Oluwagbenga
- Animal Sciences, Purdue University, West Lafayette, Indiana, USA
| | - M Bergman
- Animal Sciences, Purdue University, West Lafayette, Indiana, USA
| | - K M Ajuwon
- Animal Sciences, Purdue University, West Lafayette, Indiana, USA
| | - G S Fraley
- Animal Sciences, Purdue University, West Lafayette, Indiana, USA
| |
Collapse
|
12
|
Strnadová V, Pačesová A, Charvát V, Šmotková Z, Železná B, Kuneš J, Maletínská L. Anorexigenic neuropeptides as anti-obesity and neuroprotective agents: exploring the neuroprotective effects of anorexigenic neuropeptides. Biosci Rep 2024; 44:BSR20231385. [PMID: 38577975 PMCID: PMC11043025 DOI: 10.1042/bsr20231385] [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: 01/31/2024] [Revised: 03/26/2024] [Accepted: 04/05/2024] [Indexed: 04/06/2024] Open
Abstract
Since 1975, the incidence of obesity has increased to epidemic proportions, and the number of patients with obesity has quadrupled. Obesity is a major risk factor for developing other serious diseases, such as type 2 diabetes mellitus, hypertension, and cardiovascular diseases. Recent epidemiologic studies have defined obesity as a risk factor for the development of neurodegenerative diseases, such as Alzheimer's disease (AD) and other types of dementia. Despite all these serious comorbidities associated with obesity, there is still a lack of effective antiobesity treatment. Promising candidates for the treatment of obesity are anorexigenic neuropeptides, which are peptides produced by neurons in brain areas implicated in food intake regulation, such as the hypothalamus or the brainstem. These peptides efficiently reduce food intake and body weight. Moreover, because of the proven interconnection between obesity and the risk of developing AD, the potential neuroprotective effects of these two agents in animal models of neurodegeneration have been examined. The objective of this review was to explore anorexigenic neuropeptides produced and acting within the brain, emphasizing their potential not only for the treatment of obesity but also for the treatment of neurodegenerative disorders.
Collapse
Affiliation(s)
- Veronika Strnadová
- Department of Biochemistry and Molecular Biology, Institute of Organic Chemistry and Biochemistry, Czech Academy of Sciences, Prague, Czech Republic
| | - Andrea Pačesová
- Department of Biochemistry and Molecular Biology, Institute of Organic Chemistry and Biochemistry, Czech Academy of Sciences, Prague, Czech Republic
| | - Vilém Charvát
- Department of Biochemistry and Molecular Biology, Institute of Organic Chemistry and Biochemistry, Czech Academy of Sciences, Prague, Czech Republic
| | - Zuzana Šmotková
- Department of Biochemistry and Molecular Biology, Institute of Organic Chemistry and Biochemistry, Czech Academy of Sciences, Prague, Czech Republic
| | - Blanka Železná
- Department of Biochemistry and Molecular Biology, Institute of Organic Chemistry and Biochemistry, Czech Academy of Sciences, Prague, Czech Republic
| | - Jaroslav Kuneš
- Department of Biochemistry and Molecular Biology, Institute of Organic Chemistry and Biochemistry, Czech Academy of Sciences, Prague, Czech Republic
- Department of Biochemistry and Molecular Biology, Institute of Physiology, Czech Academy of Sciences, Prague, Czech Republic
| | - Lenka Maletínská
- Department of Biochemistry and Molecular Biology, Institute of Organic Chemistry and Biochemistry, Czech Academy of Sciences, Prague, Czech Republic
| |
Collapse
|
13
|
Hamamah S, Hajnal A, Covasa M. Influence of Bariatric Surgery on Gut Microbiota Composition and Its Implication on Brain and Peripheral Targets. Nutrients 2024; 16:1071. [PMID: 38613104 PMCID: PMC11013759 DOI: 10.3390/nu16071071] [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: 03/14/2024] [Revised: 03/29/2024] [Accepted: 04/02/2024] [Indexed: 04/14/2024] Open
Abstract
Obesity remains a significant global health challenge, with bariatric surgery remaining as one of the most effective treatments for severe obesity and its related comorbidities. This review highlights the multifaceted impact of bariatric surgery beyond mere physical restriction or nutrient malabsorption, underscoring the importance of the gut microbiome and neurohormonal signals in mediating the profound effects on weight loss and behavior modification. The various bariatric surgery procedures, such as Roux-en-Y gastric bypass (RYGB) and sleeve gastrectomy (SG), act through distinct mechanisms to alter the gut microbiome, subsequently impacting metabolic health, energy balance, and food reward behaviors. Emerging evidence has shown that bariatric surgery induces profound changes in the composition of the gut microbiome, notably altering the Firmicutes/Bacteroidetes ratio and enhancing populations of beneficial bacteria such as Akkermansia. These microbiota shifts have far-reaching effects beyond gut health, influencing dopamine-mediated reward pathways in the brain and modulating the secretion and action of key gut hormones including ghrelin, leptin, GLP-1, PYY, and CCK. The resultant changes in dopamine signaling and hormone levels contribute to reduced hedonic eating, enhanced satiety, and improved metabolic outcomes. Further, post-bariatric surgical effects on satiation targets are in part mediated by metabolic byproducts of gut microbiota like short-chain fatty acids (SCFAs) and bile acids, which play a pivotal role in modulating metabolism and energy expenditure and reducing obesity-associated inflammation, as well as influencing food reward pathways, potentially contributing to the regulation of body weight and reduction in hedonic eating behaviors. Overall, a better understanding of these mechanisms opens the door to developing non-surgical interventions that replicate the beneficial effects of bariatric surgery on the gut microbiome, dopamine signaling, and gut hormone regulation, offering new avenues for obesity treatment.
Collapse
Affiliation(s)
- Sevag Hamamah
- Department of Basic Medical Sciences, College of Osteopathic Medicine, Western University of Health Sciences, Pomona, CA 9176, USA;
| | - Andras Hajnal
- Department of Neural and Behavioral Sciences, College of Medicine, The Pennsylvania State University, Hershey, PA 17033, USA;
| | - Mihai Covasa
- Department of Basic Medical Sciences, College of Osteopathic Medicine, Western University of Health Sciences, Pomona, CA 9176, USA;
- Department of Biomedical Sciences, College of Medicine and Biological Science, University of Suceava, 7200229 Suceava, Romania
| |
Collapse
|
14
|
Spezani R, Marinho TS, Reis TS, Aguila MB, Mandarim-de-Lacerda CA. Cotadutide (GLP-1/Glucagon dual receptor agonist) modulates hypothalamic orexigenic and anorexigenic neuropeptides in obese mice. Peptides 2024; 173:171138. [PMID: 38147963 DOI: 10.1016/j.peptides.2023.171138] [Citation(s) in RCA: 4] [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: 09/19/2023] [Revised: 11/15/2023] [Accepted: 12/18/2023] [Indexed: 12/28/2023]
Abstract
The hypothalamic neuropeptides linked to appetite and satiety were investigated in obese mice treated with cotadutide (a dual receptor agonist of glucagon-like peptide 1 (GLP-1R)/Glucagon (GCGR)). Twelve-week-old male C57BL/6 mice were fed a control diet (C group, n = 20) or a high-fat diet (HF group, n = 20) for ten weeks. Each group was further divided, adding cotadutide treatment and forming groups C, CC, HF, and HFC for four additional weeks. The hypothalamic arcuate neurons were labeled by immunofluorescence, and protein expressions (Western blotting) for neuropeptide Y (NPY), proopiomelanocortin (POMC), agouti-related protein (AgRP), and cocaine- and amphetamine-regulated transcript (CART). Cotadutide enhanced POMC and CART neuropeptides and depressed NPY and AGRP neuropeptides. In addition, gene expressions (RT-qPCR) determined that Lepr (leptin receptor) and Calcr (calcitonin receptor) were diminished in HF compared to C but enhanced in CC compared to C and HFC compared to HF. Besides, Socs3 (suppressor of cytokine signaling 3) was decreased in HFC compared to HF, while Sst (somatostatin) was higher in HFC compared to HF; Tac1 (tachykinin 1) and Mc4r (melanocortin-4-receptor) were lower in HF compared to C but increased in HFC compared to HF. Also, Glp1r and Gcgr were higher in HFC compared to HF. In conclusion, the findings are compelling, demonstrating the effects of cotadutide on hypothalamic neuropeptides and hormone receptors of obese mice. Cotadutide modulates energy balance through the gut-brain axis and its associated signaling pathways. The study provides insights into the mechanisms underlying cotadutide's anti-obesity effects and its possible implications for obesity treatment.
Collapse
Affiliation(s)
- Renata Spezani
- Laboratory of Morphometry, Metabolism, and Cardiovascular Disease, Institute of Biology, Biomedical Center, The University of the State of Rio de Janeiro, Brazil
| | - Thatiany Souza Marinho
- Laboratory of Morphometry, Metabolism, and Cardiovascular Disease, Institute of Biology, Biomedical Center, The University of the State of Rio de Janeiro, Brazil
| | - Thiago Santos Reis
- Laboratory of Morphometry, Metabolism, and Cardiovascular Disease, Institute of Biology, Biomedical Center, The University of the State of Rio de Janeiro, Brazil
| | - Marcia Barbosa Aguila
- Laboratory of Morphometry, Metabolism, and Cardiovascular Disease, Institute of Biology, Biomedical Center, The University of the State of Rio de Janeiro, Brazil
| | - Carlos A Mandarim-de-Lacerda
- Laboratory of Morphometry, Metabolism, and Cardiovascular Disease, Institute of Biology, Biomedical Center, The University of the State of Rio de Janeiro, Brazil.
| |
Collapse
|
15
|
Xue J, Brawner AT, Thompson JR, Yelhekar TD, Newmaster KT, Qiu Q, Cooper YA, Yu CR, Ahmed-Braima YH, Kim Y, Lin Y. Spatiotemporal Mapping and Molecular Basis of Whole-brain Circuit Maturation. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.01.03.572456. [PMID: 38260331 PMCID: PMC10802351 DOI: 10.1101/2024.01.03.572456] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/24/2024]
Abstract
Brain development is highly dynamic and asynchronous, marked by the sequential maturation of functional circuits across the brain. The timing and mechanisms driving circuit maturation remain elusive due to an inability to identify and map maturing neuronal populations. Here we create DevATLAS (Developmental Activation Timing-based Longitudinal Acquisition System) to overcome this obstacle. We develop whole-brain mapping methods to construct the first longitudinal, spatiotemporal map of circuit maturation in early postnatal mouse brains. Moreover, we uncover dramatic impairments within the deep cortical layers in a neurodevelopmental disorders (NDDs) model, demonstrating the utility of this resource to pinpoint when and where circuit maturation is disrupted. Using DevATLAS, we reveal that early experiences accelerate the development of hippocampus-dependent learning by increasing the synaptically mature granule cell population in the dentate gyrus. Finally, DevATLAS enables the discovery of molecular mechanisms driving activity-dependent circuit maturation.
Collapse
Affiliation(s)
- Jian Xue
- Department of Psychiatry, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Andrew T. Brawner
- Department of Psychiatry and Behavioral Sciences, SUNY Upstate Medical University, Syracuse, NY 13210, USA
- Neuroscience Graduate Program, SUNY Upstate Medical University, Syracuse, NY 13210, USA
- Equal contribution
| | - Jacqueline R. Thompson
- Department of Psychiatry and Behavioral Sciences, SUNY Upstate Medical University, Syracuse, NY 13210, USA
- Neuroscience Graduate Program, SUNY Upstate Medical University, Syracuse, NY 13210, USA
- Equal contribution
| | - Tushar D. Yelhekar
- Department of Psychiatry, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Kyra T. Newmaster
- Department of Neural and Behavioral Sciences, The Pennsylvania State University, Hershey, PA 17033, USA
| | - Qiang Qiu
- Stowers Institute for Medical Research, Kansas City, MO 64110, USA
- Department of Anatomy and Cell Biology, University of Kansas Medical Center, Kansas City, MO 66160, USA
| | - Yonatan A. Cooper
- Current address: Department of Human Genetics, David Geffen School of Medicine, University of California, Los Angeles, CA 90095, USA
| | - C. Ron Yu
- Stowers Institute for Medical Research, Kansas City, MO 64110, USA
- Department of Anatomy and Cell Biology, University of Kansas Medical Center, Kansas City, MO 66160, USA
| | | | - Yongsoo Kim
- Department of Neural and Behavioral Sciences, The Pennsylvania State University, Hershey, PA 17033, USA
| | - Yingxi Lin
- Department of Psychiatry, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
- Lead contact
| |
Collapse
|
16
|
Song J, Choi SY. Arcuate Nucleus of the Hypothalamus: Anatomy, Physiology, and Diseases. Exp Neurobiol 2023; 32:371-386. [PMID: 38196133 PMCID: PMC10789173 DOI: 10.5607/en23040] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2023] [Revised: 12/24/2023] [Accepted: 12/27/2023] [Indexed: 01/11/2024] Open
Abstract
The hypothalamus is part of the diencephalon and has several nuclei, one of which is the arcuate nucleus. The arcuate nucleus of hypothalamus (ARH) consists of neuroendocrine neurons and centrally-projecting neurons. The ARH is the center where the homeostasis of nutrition/metabolism and reproduction are maintained. As such, dysfunction of the ARH can lead to disorders of nutrition/metabolism and reproduction. Here, we review various types of neurons in the ARH and several genetic disorders caused by mutations in the ARH.
Collapse
Affiliation(s)
- Juhyun Song
- Department of Anatomy, Chonnam National University Medical School, Hwasun 58128, Korea
| | - Seok-Yong Choi
- Department of Biomedical Sciences, Chonnam National University Medical School, Hwasun 58128, Korea
| |
Collapse
|
17
|
Zhang M, Thieux M, Arvis L, Lin JS, Guyon A, Plancoulaine S, Villanueva C, Franco P. Metabolic disturbances in children with narcolepsy: a retrospective study. Sleep 2023; 46:zsad076. [PMID: 36971181 DOI: 10.1093/sleep/zsad076] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2022] [Revised: 09/22/2022] [Indexed: 07/20/2023] Open
Abstract
STUDY OBJECTIVES To determine the prevalence of metabolic syndrome (MS) in children with narcolepsy and to evaluate their clinical and sleep characteristics according to the different components of MS. METHODS This retrospective study consisted of 58 de novo children with narcolepsy (median age: 12.7 years, 48.3% of boys). The recently published MS criteria in a French population of children were used. Clinical and sleep characteristics were compared between groups with different components of MS. RESULTS MS was present in 17.2% of children with narcolepsy, among whom 79.3% presented with high homeostasis model assessment for insulin resistance (HOMA-IR), 25.9% with high body mass index, 24.1% with low high-density lipoprotein cholesterol (HDL-C), and 12.1% with high triglycerides. Patients with at least two MS components had more night eating behaviors and tended to have lower percentage of slow-wave sleep and more fragmented sleep. On multiple sleep latency test, they had shorter mean sleep latencies to rapid eye movement (REM), non-REM sleep and tended to have more sleep onset REM periods (SOREMPs) than those with less than two MS components. CONCLUSIONS Insulin resistance was found to be the core metabolic disturbance in obese as well as in nonobese children with narcolepsy. Children with narcolepsy with at least two MS components presented a more severe daytime sleepiness and a higher prevalence of night-eating behaviors than those with less than two MS components. Such children might benefit from early evaluation and management in order to prevent future complications.
Collapse
Affiliation(s)
- Min Zhang
- Integrative Physiology of the Brain Arousal Systems, CRNL, INSERM U1028, CNRS UMR5292, University of Lyon 1, Lyon, France
| | - Marine Thieux
- Integrative Physiology of the Brain Arousal Systems, CRNL, INSERM U1028, CNRS UMR5292, University of Lyon 1, Lyon, France
- Pediatric Sleep Unit, Hôpital Femme Mère Enfant, Hospices Civils de Lyon and National Reference Center for Narcolepsy, University of Lyon1, Lyon, France
| | - Laura Arvis
- Pediatric endocrinology Unit, Hôpital Femme Mère Enfant, Hospices Civils de Lyon, Université Claude Bernard Lyon 1, Lyon, France
| | - Jian-Sheng Lin
- Integrative Physiology of the Brain Arousal Systems, CRNL, INSERM U1028, CNRS UMR5292, University of Lyon 1, Lyon, France
| | - Aurore Guyon
- Integrative Physiology of the Brain Arousal Systems, CRNL, INSERM U1028, CNRS UMR5292, University of Lyon 1, Lyon, France
- Pediatric Sleep Unit, Hôpital Femme Mère Enfant, Hospices Civils de Lyon and National Reference Center for Narcolepsy, University of Lyon1, Lyon, France
| | | | - Carine Villanueva
- Pediatric endocrinology Unit, Hôpital Femme Mère Enfant, Hospices Civils de Lyon, Université Claude Bernard Lyon 1, Lyon, France
| | - Patricia Franco
- Integrative Physiology of the Brain Arousal Systems, CRNL, INSERM U1028, CNRS UMR5292, University of Lyon 1, Lyon, France
- Pediatric Sleep Unit, Hôpital Femme Mère Enfant, Hospices Civils de Lyon and National Reference Center for Narcolepsy, University of Lyon1, Lyon, France
| |
Collapse
|
18
|
Sharma A, Sur S, Tripathi V, Kumar V. Genetic Control of Avian Migration: Insights from Studies in Latitudinal Passerine Migrants. Genes (Basel) 2023; 14:1191. [PMID: 37372370 DOI: 10.3390/genes14061191] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2023] [Revised: 05/24/2023] [Accepted: 05/25/2023] [Indexed: 06/29/2023] Open
Abstract
Twice-a-year, large-scale movement of billions of birds across latitudinal gradients is one of the most fascinating behavioral phenomena seen among animals. These seasonal voyages in autumn southwards and in spring northwards occur within a discrete time window and, as part of an overall annual itinerary, involve close interaction of the endogenous rhythm at several levels with prevailing photoperiod and temperature. The overall success of seasonal migrations thus depends on their close coupling with the other annual sub-cycles, namely those of the breeding, post-breeding recovery, molt and non-migratory periods. There are striking alterations in the daily behavior and physiology with the onset and end of the migratory period, as shown by the phase inversions in behavioral (a diurnal passerine bird becomes nocturnal and flies at night) and neural activities. Interestingly, there are also differences in the behavior, physiology and regulatory strategies between autumn and spring (vernal) migrations. Concurrent molecular changes occur in regulatory (brain) and metabolic (liver, flight muscle) tissues, as shown in the expression of genes particularly associated with 24 h timekeeping, fat accumulation and the overall metabolism. Here, we present insights into the genetic basis of migratory behavior based on studies using both candidate and global gene expression approaches in passerine migrants, with special reference to Palearctic-Indian migratory blackheaded and redheaded buntings.
Collapse
Affiliation(s)
- Aakansha Sharma
- IndoUS Center in Chronobiology, Department of Zoology, University of Lucknow, Lucknow 226007, India
| | - Sayantan Sur
- IndoUS Center in Chronobiology, Department of Zoology, University of Lucknow, Lucknow 226007, India
| | - Vatsala Tripathi
- Department of Zoology, Dyal Singh College, University of Delhi, Delhi 110003, India
| | - Vinod Kumar
- IndoUS Center in Chronobiology, Department of Zoology, University of Delhi, Delhi 110007, India
| |
Collapse
|
19
|
Li Y, Xiao Y, Wu W, Jin L, Jia Y, Cai K, Qiao N, Cao L, Gui S. Effects of Craniotomy and Endoscopic Endonasal Transsphenoidal Surgery on Bodyweight in Adult-Onset Craniopharyngioma: A Single-Center Retrospective Study. J Clin Med 2023; 12:1578. [PMID: 36836112 PMCID: PMC9962365 DOI: 10.3390/jcm12041578] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2022] [Revised: 02/04/2023] [Accepted: 02/14/2023] [Indexed: 02/18/2023] Open
Abstract
Craniopharyngioma (CP) is a histologically benign tumor with high mortality and morbidity. Although surgical treatment is essential in managing CP, the best surgical approach is debated. A retrospective cohort of 117 patients with adult-onset CP (AOCP) treated between 2018 and 2020 in Beijing Tiantan Hospital was identified and examined. The effects of traditional craniotomy (TC) and endoscopic endonasal transsphenoidal surgery (EETS) on the extent of surgical resection, hypothalamic involvement (HI), postoperative endocrine function, and postoperative weight were compared in the cohort. The cohort comprised 43 males and 74 females, divided into the TC (n = 59) and EETS (n = 58) groups. The EETS group possessed a higher rate of gross total resection (GTR) (adjusted odds ratio (aOR) = 4.08, p = 0.029) and improved HI (aOR = 2.58, p = 0.041) than the TC group. Worse postoperative HI was only observed in the TC group (5 patients). The EETS was associated with fewer adverse hormonal outcomes, including posterior pituitary dysfunction (aOR = 0.386, p = 0.040) and hypopituitarism (aOR = 0.384, p = 0.031). Additionally, multivariate logistic regression analysis confirmed that EETS was related to fewer cases of weight gain >5% (aOR = 0.376, p = 0.034), significant weight change (aOR = 0.379, p = 0.022), and postoperative obesity (aOR = 0.259, p = 0.032). Compared to TC, EETS shows advantages in accomplishing GTR, hypothalamus protection, postoperative endocrine function reservation, and postoperative weight control. These data suggest that the EETS deserves more application in managing patients with AOCP.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Songbai Gui
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing 100070, China
| |
Collapse
|
20
|
Rachmasari KN, Strauss SB, Phillips CD, Lantos JE, An A, Cisse B, Ramakrishna R, Schwartz TH, Dobri GA. Posterior hypothalamic involvement on pre-operative MRI predicts hypothalamic obesity in craniopharyngiomas. Pituitary 2023; 26:105-114. [PMID: 36462067 DOI: 10.1007/s11102-022-01294-0] [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: 11/23/2022] [Indexed: 12/05/2022]
Abstract
PURPOSE Hypothalamic obesity (HO) is a complication associated with craniopharyngioma (CP). Attempts have been made to perioperatively predict the development of this complication, which can be severe and difficult to treat. METHODS Patients who underwent first transsphenoidal surgical resection in a single center between February 2005 and March 2019 were screened; those who have had prior surgery or radiation, were aged below 18 years, or did not have follow up body mass index (BMI) after surgery were excluded. Primary end point was BMI within 2 years post-surgery. Hypothalamic involvement (HI) was graded based on preoperative and postoperative imaging with regards to anterior, posterior, left and right involvement. Data on baseline demographics, pre-operative and post-operative MRI, and endocrine function were collected. RESULTS 45 patients met the inclusion and exclusion criteria. Most patients in our cohort underwent gross total resection (n = 35 patients). 13 patients were from no HI or anterior HI only group and 22 patients were classified as both anterior (ant) and posterior (post) HI group. There was no significant difference between the two groups in the gross total, subtotal or near total resection. Pre-operative BMI and post-operative BMI were significantly higher in patients who had ant and post HI on pre-operative MRI (p < 0.05 and p < 0.01, respectively). Similarly, post-operative BMI at 13-24 months was also significantly higher in the ant and post HI group on post-op MRI (p < 0.01). There was no significant difference between the two groups in terms of baseline adrenal insufficiency, thyroid insufficiency, gonadal insufficiency, IGF-1 levels, hyperprolactinemia, and diabetes insipidus. Diabetes insipidus was more common following surgery among those who had anterior and posterior involvement on pre-operative MRI (p < 0.05). CONCLUSIONS HO appears to be predetermined by tumor involvement in the posterior hypothalamus observed on pre-operative MRI. Posterior HI on pre-operative MRI was also associated with the development of diabetes insipidus after surgery.
Collapse
Affiliation(s)
- Kharisa N Rachmasari
- Department of Medicine, New York-Presbyterian Hospital - Weill Cornell Medicine, New York, NY, USA.
| | - Sara B Strauss
- Department of Neuroradiology, Weill Cornell Medicine, New York, NY, USA
| | | | - Joshua E Lantos
- Department of Neuroradiology, Weill Cornell Medicine, New York, NY, USA
| | - Anjile An
- Division of Biostatistics and Epidemiology, Weill Cornell Medicine, New York, NY, USA
| | - Babacar Cisse
- Department of Neurosurgery, Weill Cornell Medicine, New York, NY, USA
| | - Rohan Ramakrishna
- Department of Neurosurgery, Weill Cornell Medicine, New York, NY, USA
| | | | - Georgiana A Dobri
- Department of Neurosurgery, Weill Cornell Medicine, New York, NY, USA
- Department of Endocrinology, Weill Cornell Medicine, New York, NY, USA
| |
Collapse
|
21
|
Li X, Sun Z. Circadian clock and temporal meal pattern. MEDICAL REVIEW (2021) 2023; 3:85-101. [PMID: 37724110 PMCID: PMC10471112 DOI: 10.1515/mr-2022-0021] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/05/2022] [Accepted: 08/02/2022] [Indexed: 09/20/2023]
Abstract
The central circadian clock in the brain controls the time-of-the-day variations in acute meal responses, with a low glycemic response but a high satiety/thermogenic response to meals consumed at waking compared to other time points. Consistently, studies show that consuming a significant proportion of calories, particularly carbohydrates, in breakfast is beneficial for the chronic management of obesity and its associated metabolic syndrome, compared to consuming identical meals at dinner. Conversely, breakfast skipping or/and late dinner can have unfavorable metabolic outcomes. It remains controversial how meal frequency affects metabolic health. In contrast, irregular meals, especially irregular breakfasts, show consistent adverse metabolic consequences. Time-restricted feeding (TRF), with all calories consumed within less than 12-h per day, can improve metabolism and extend lifespan. A major component of TRF in humans is caloric restriction, which contributes significantly to the beneficial effects of TRF in humans. By comparison, TRF effects in rodents can be independent of caloric restriction and show day/night phase specificity. TRF could alleviate metabolic abnormalities due to circadian disruption, but its effects appear independent of the circadian clock in rodents. Understanding neuroendocrine mechanisms underlying clock-mediated metabolic regulation will shed light on the metabolic effects of temporal meal patterns.
Collapse
Affiliation(s)
- Xin Li
- Department of Medicine – Endocrinology, Diabetes, and Metabolism, Baylor College of Medicine, Houston, TX, USA
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA
| | - Zheng Sun
- Department of Medicine – Endocrinology, Diabetes, and Metabolism, Baylor College of Medicine, Houston, TX, USA
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA
| |
Collapse
|
22
|
Rapps K, Kisliouk T, Marco A, Weller A, Meiri N. Dieting reverses histone methylation and hypothalamic AgRP regulation in obese rats. Front Endocrinol (Lausanne) 2023; 14:1121829. [PMID: 36817590 PMCID: PMC9930686 DOI: 10.3389/fendo.2023.1121829] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/12/2022] [Accepted: 01/17/2023] [Indexed: 02/04/2023] Open
Abstract
INTRODUCTION Although dieting is a key factor in improving physiological functions associated with obesity, the role by which histone methylation modulates satiety/hunger regulation of the hypothalamus through weight loss remains largely elusive. Canonically, H3K9me2 is a transcriptional repressive post-translational epigenetic modification that is involved in obesity, however, its role in the hypothalamic arcuate nucleus (ARC) has not been thoroughly explored. Here we explore the role that KDM4D, a specific demethylase of residue H3K9, plays in energy balance by directly modulating the expression of AgRP, a key neuropeptide that regulates hunger response. METHODS We used a rodent model of diet-induced obesity (DIO) to assess whether histone methylation malprogramming impairs energy balance control and how caloric restriction may reverse this phenotype. Using ChIP-qPCR, we assessed the repressive modification of H3K9me2 at the site of AgRP. To elucidate the functional role of KDM4D in reversing obesity via dieting, a pharmacological agent, JIB-04 was used to inhibit the action of KDM4D in vivo. RESULTS In DIO, downregulation of Kdm4d mRNA results in both enrichment of H3K9me2 on the AgRP promoter and transcriptional repression of AgRP. Because epigenetic modifications are dynamic, it is possible for some of these modifications to be reversed when external cues are altered. The reversal phenomenon was observed in calorically restricted rats, in which upregulation of Kdm4d mRNA resulted in demethylation of H3K9 on the AgRP promoter and transcriptional increase of AgRP. In order to verify that KDM4D is necessary to reverse obesity by dieting, we demonstrated that in vivo inhibition of KDM4D activity by pharmacological agent JIB-04 in naïve rats resulted in transcriptional repression of AgRP, decreasing orexigenic signaling, thus inhibiting hunger. DISCUSSION We propose that the action of KDM4D through the demethylation of H3K9 is critical in maintaining a stable epigenetic landscape of the AgRP promoter, and may offer a target to develop new treatments for obesity.
Collapse
Affiliation(s)
- Kayla Rapps
- Faculty of Life Sciences, Bar Ilan University, Ramat-Gan, Israel
- Institute of Animal Science, Agricultural Research Organization, The Volcani Center, Rishon LeZiyyon, Israel
- Gonda Multidisciplinary Brain Research Center, Bar Ilan University, Ramat-Gan, Israel
| | - Tatiana Kisliouk
- Institute of Animal Science, Agricultural Research Organization, The Volcani Center, Rishon LeZiyyon, Israel
| | - Asaf Marco
- Neuro-Epigenetics Laboratory, Faculty of Agriculture, Food and Environment, The Hebrew University of Jerusalem, Rehovot, Israel
| | - Aron Weller
- Gonda Multidisciplinary Brain Research Center, Bar Ilan University, Ramat-Gan, Israel
- Department of Psychology, Bar Ilan University, Ramat-Gan, Israel
| | - Noam Meiri
- Institute of Animal Science, Agricultural Research Organization, The Volcani Center, Rishon LeZiyyon, Israel
| |
Collapse
|
23
|
Deng Y, Dickey JE, Saito K, Deng G, Singh U, Jiang J, Toth BA, Zhu Z, Zingman LV, Resch JM, Grobe JL, Cui H. Elucidating the role of Rgs2 expression in the PVN for metabolic homeostasis in mice. Mol Metab 2022; 66:101622. [PMID: 36307046 PMCID: PMC9638802 DOI: 10.1016/j.molmet.2022.101622] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/23/2022] [Revised: 10/09/2022] [Accepted: 10/21/2022] [Indexed: 11/06/2022] Open
Abstract
OBJECTIVE RGS2 is a GTPase activating protein that modulates GPCR-Gα signaling and mice lacking RGS2 globally exhibit metabolic alterations. While RGS2 is known to be broadly expressed throughout the body including the brain, the relative contribution of brain RGS2 to metabolic homeostasis remains unknown. The purpose of this study was to characterize RGS2 expression in the paraventricular nucleus of hypothalamus (PVN) and test its role in metabolic homeostasis. METHODS We used a combination of RNAscope in situ hybridization (ISH), immunohistochemistry, and bioinformatic analyses to characterize the pattern of Rgs2 expression in the PVN. We then created mice lacking Rgs2 either prenatally or postnatally in the PVN and evaluated their metabolic consequences. RESULTS RNAscope ISH analysis revealed a broad but regionally enriched Rgs2 mRNA expression throughout the mouse brain, with the highest expression being observed in the PVN along with several other brain regions, such as the arcuate nucleus of hypothalamus and the dorsal raphe nucleus. Within the PVN, we found that Rgs2 is specifically enriched in CRH+ endocrine neurons and is further increased by calorie restriction. Functionally, although Sim1-Cre-mediated prenatal deletion of Rgs2 in PVN neurons had no major effects on metabolic homeostasis, AAV-mediated adult deletion of Rgs2 in the PVN led to significantly increased food intake, body weight (both fat and fat-free masses), body length, and blood glucose levels in both male and female mice. Strikingly, we found that prolonged postnatal loss of Rgs2 leads to neuronal cell death in the PVN, while rapid body weight gain in the early phase of viral-mediated PVN Rgs2 deletion is independent of PVN neuronal loss. CONCLUSIONS Our results provide the first evidence to show that PVN Rgs2 expression is not only sensitive to metabolic challenge but also critically required for PVN endocrine neurons to function and maintain metabolic homeostasis.
Collapse
Affiliation(s)
- Yue Deng
- Department of Neuroscience and Pharmacology, University of Iowa Carver College of Medicine, Iowa City, IA, United States
| | - Jacob E Dickey
- Department of Neuroscience and Pharmacology, University of Iowa Carver College of Medicine, Iowa City, IA, United States
| | - Kenji Saito
- Department of Neuroscience and Pharmacology, University of Iowa Carver College of Medicine, Iowa City, IA, United States
| | - Guorui Deng
- Department of Neuroscience and Pharmacology, University of Iowa Carver College of Medicine, Iowa City, IA, United States
| | - Uday Singh
- Department of Neuroscience and Pharmacology, University of Iowa Carver College of Medicine, Iowa City, IA, United States
| | - Jingwei Jiang
- Department of Neuroscience and Pharmacology, University of Iowa Carver College of Medicine, Iowa City, IA, United States
| | - Brandon A Toth
- Department of Neuroscience and Pharmacology, University of Iowa Carver College of Medicine, Iowa City, IA, United States
| | - Zhiyong Zhu
- Department of Internal Medicine, University of Iowa Carver College of Medicine, Iowa City, IA, United States
| | - Leonid V Zingman
- Department of Internal Medicine, University of Iowa Carver College of Medicine, Iowa City, IA, United States; Iowa Neuroscience Institute, University of Iowa Carver College of Medicine, Iowa City, IA, United States
| | - Jon M Resch
- Department of Neuroscience and Pharmacology, University of Iowa Carver College of Medicine, Iowa City, IA, United States; Iowa Neuroscience Institute, University of Iowa Carver College of Medicine, Iowa City, IA, United States; F.O.E. Diabetes Research Center, University of Iowa Carver College of Medicine, Iowa City, IA, United States
| | - Justin L Grobe
- Department of Physiology, Medical College of Wisconsin, Milwaukee, WI 53226, USA; Comprehensive Rodent Metabolic Phenotyping Core, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Huxing Cui
- Department of Neuroscience and Pharmacology, University of Iowa Carver College of Medicine, Iowa City, IA, United States; Iowa Neuroscience Institute, University of Iowa Carver College of Medicine, Iowa City, IA, United States; F.O.E. Diabetes Research Center, University of Iowa Carver College of Medicine, Iowa City, IA, United States.
| |
Collapse
|
24
|
Zhang Q, Tang Q, Purohit NM, Davenport JB, Brennan C, Patel RK, Godschall E, Zwiefel LS, Spano A, Campbell JN, Güler AD. Food-induced dopamine signaling in AgRP neurons promotes feeding. Cell Rep 2022; 41:111718. [PMID: 36450244 PMCID: PMC9753708 DOI: 10.1016/j.celrep.2022.111718] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2022] [Revised: 09/21/2022] [Accepted: 11/02/2022] [Indexed: 12/02/2022] Open
Abstract
Obesity comorbidities such as diabetes and cardiovascular disease are pressing public health concerns. Overconsumption of calories leads to weight gain; however, neural mechanisms underlying excessive food consumption are poorly understood. Here, we demonstrate that dopamine receptor D1 (Drd1) expressed in the agouti-related peptide/neuropeptide Y (AgRP/NPY) neurons of the arcuate hypothalamus is required for appropriate responses to a high-fat diet (HFD). Stimulation of Drd1 and AgRP/NPY co-expressing arcuate neurons is sufficient to induce voracious feeding. Delivery of a HFD after food deprivation acutely induces dopamine (DA) release in the ARC, whereas animals that lack Drd1 expression in ARCAgRP/NPY neurons (Drd1AgRP-KO) exhibit attenuated foraging and refeeding of HFD. These results define a role for the DA input to the ARC that encodes acute responses to food and position Drd1 signaling in the ARCAgRP/NPY neurons as an integrator of the hedonic and homeostatic neuronal feeding circuits.
Collapse
Affiliation(s)
- Qi Zhang
- Department of Biology, University of Virginia, Charlottesville, VA 22904, USA
| | - Qijun Tang
- Department of Biology, University of Virginia, Charlottesville, VA 22904, USA
| | - Nidhi M. Purohit
- Department of Biology, University of Virginia, Charlottesville, VA 22904, USA
| | - Julia B. Davenport
- Department of Biology, University of Virginia, Charlottesville, VA 22904, USA
| | - Charles Brennan
- Department of Biology, University of Virginia, Charlottesville, VA 22904, USA
| | - Rahul K. Patel
- Department of Biology, University of Virginia, Charlottesville, VA 22904, USA
| | - Elizabeth Godschall
- Department of Biology, University of Virginia, Charlottesville, VA 22904, USA
| | - Larry S. Zwiefel
- Departments of Pharmacology and Psychiatry and Behavioral Sciences, University of Washington, 1959 NE Pacific Street, Seattle, WA 98195, USA
| | - Anthony Spano
- Department of Biology, University of Virginia, Charlottesville, VA 22904, USA
| | - John N. Campbell
- Department of Biology, University of Virginia, Charlottesville, VA 22904, USA,Program in Fundamental Neuroscience, University of Virginia, Charlottesville, VA 22904, USA
| | - Ali D. Güler
- Department of Biology, University of Virginia, Charlottesville, VA 22904, USA,Program in Fundamental Neuroscience, University of Virginia, Charlottesville, VA 22904, USA,Lead contact,Correspondence:
| |
Collapse
|
25
|
Heat stress in pigs and broilers: role of gut dysbiosis in the impairment of the gut-liver axis and restoration of these effects by probiotics, prebiotics and synbiotics. J Anim Sci Biotechnol 2022; 13:126. [PMCID: PMC9673442 DOI: 10.1186/s40104-022-00783-3] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2022] [Accepted: 09/14/2022] [Indexed: 11/19/2022] Open
Abstract
AbstractHeat stress is one of the most challenging stressors for animal production due to high economic losses resulting from impaired animal’s productivity, health and welfare. Despite the fact that all farm animal species are susceptible to heat stress, birds and pigs are particularly sensitive to heat stress due to either lacking or non-functional sweat glands. Convincing evidence in the literature exists that gut dysbiosis, a term used to describe a perturbation of commensal gut microbiota, develops in broilers and pigs under heat stress. Owing to the protective role of commensal bacteria for the gut barrier, gut dysbiosis causes a disruption of the gut barrier leading to endotoxemia, which contributes to the typical characteristics of heat stressed broilers and growing and growing-finishing pigs, such as reduced feed intake, decreased growth and reduced lean carcass weight. A substantial number of studies have shown that feeding of probiotics, prebiotics and synbiotics is an efficacious strategy to protect broilers from heat stress-induced gut barrier disruption through altering the gut microbiota and promoting all decisive structural, biochemical, and immunological elements of the intestinal barrier. In most of the available studies in heat stressed broilers, the alterations of gut microbiota and improvements of gut barrier function induced by feeding of either probiotics, prebiotics or synbiotics were accompanied by an improved productivity, health and/or welfare when compared to non-supplemented broilers exposed to heat stress. These findings indicate that the restoration of gut homeostasis and function is a key target for dietary interventions aiming to provide at least partial protection of broilers from the detrimental impact of heat stress conditions. Despite the fact that the number of studies dealing with the same feeding strategy in heat stressed pigs is limited, the available few studies suggest that feeding of probiotics might also be a suitable approach to enhance productivity, health and welfare in pigs kept under heat stress conditions.
Collapse
|
26
|
Dhafar HO, BaHammam AS. Body Weight and Metabolic Rate Changes in Narcolepsy: Current Knowledge and Future Directions. Metabolites 2022; 12:1120. [PMID: 36422261 PMCID: PMC9693066 DOI: 10.3390/metabo12111120] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2022] [Revised: 11/13/2022] [Accepted: 11/15/2022] [Indexed: 08/26/2023] Open
Abstract
Narcolepsy is a known auto-immune disease that presents mainly in the teenage years with irresistible sleep attacks. Patients with narcolepsy, especially NT1, have been found to have a high prevalence of obesity and other metabolic derangements. This narrative review aimed to address the relationship between narcolepsy and changes in weight and metabolic rate, and discuss potential mechanisms for weight gain and metabolic changes and future research agendas on this topic. This article will provide a balanced, up-to-date critical review of the current literature, and delineate areas for future research, in order to understand the pathophysiological metabolic changes in narcolepsy. Articles using predefined keywords were searched for in PubMed and Google Scholar databases, with predefined inclusion and exclusion criteria. Compared to controls, patients with narcolepsy are more likely to be obese and have higher BMIs and waist circumferences. According to recent research, weight gain in narcolepsy patients may be higher during the disease's outset. The precise mechanisms causing this weight gain remains unknown. The available information, albeit limited, does not support differences in basal or resting metabolic rates between patients with narcolepsy and controls, other than during the time of disease onset. The evidence supporting the role of orexin in weight gain in humans with narcolepsy is still controversial, in the literature. Furthermore, the available data did not show any appreciable alterations in the levels of CSF melanin-concentrating hormone, plasma and CSF leptin, or serum growth hormone, in relation to weight gain. Other mechanisms have been proposed, including a reduction in sympathetic tone, hormonal changes, changes in eating behavior and physical activity, and genetic predisposition. The association between increased body mass index and narcolepsy is well-recognized; however, the relationship between narcolepsy and other metabolic measures, such as body fat/muscle distribution and metabolic rate independent of BMI, is not well documented, and the available evidence is inconsistent. Future longitudinal studies with larger sample sizes are needed to assess BMR in patients with narcolepsy under a standard protocol at the outset of narcolepsy, with regular follow-up.
Collapse
Affiliation(s)
- Hamza O. Dhafar
- The University Sleep Disorders Center, Department of Medicine, College of Medicine, King Saud University, Riyadh 11451, Saudi Arabia
- Department of Family Medicine, Prince Mansour Military Hospital, Taif 26526, Saudi Arabia
| | - Ahmed S. BaHammam
- The University Sleep Disorders Center, Department of Medicine, College of Medicine, King Saud University, Riyadh 11451, Saudi Arabia
- The Strategic Technologies Program of the National Plan for Sciences and Technology and Innovation in the Kingdom of Saudi Arabia, P.O. Box 2454, Riyadh 11324, Saudi Arabia
| |
Collapse
|
27
|
Olejniczak I, Campbell B, Tsai YC, Tyagarajan SK, Albrecht U, Ripperger JA. Suprachiasmatic to paraventricular nuclei interaction generates normal food searching rhythms in mice. Front Physiol 2022; 13:909795. [PMID: 36277219 PMCID: PMC9582613 DOI: 10.3389/fphys.2022.909795] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Accepted: 09/23/2022] [Indexed: 11/29/2022] Open
Abstract
Searching for food follows a well-organized decision process in mammals to take up food only if necessary. Moreover, scavenging is preferred during their activity phase. Various time-dependent regulatory processes have been identified originating from the suprachiasmatic nuclei (SCN), which convert external light information into synchronizing output signals. However, a direct impact of the SCN on the timing of normal food searching has not yet been found. Here, we revisited the function of the SCN to affect when mice look for food. We found that this process was independent of light but modified by the palatability of the food source. Surprisingly, reducing the output from the SCN, in particular from the vasopressin releasing neurons, reduced the amount of scavenging during the early activity phase. The SCN appeared to transmit a signal to the paraventricular nuclei (PVN) via GABA receptor A1. Finally, the interaction of SCN and PVN was verified by retrograde transport-mediated complementation. None of the genetic manipulations affected the uptake of more palatable food. The data indicate that the PVN are sufficient to produce blunted food searching rhythms and are responsive to hedonistic feeding. Nevertheless, the search for normal food during the early activity phase is significantly enhanced by the SCN.
Collapse
Affiliation(s)
- Iwona Olejniczak
- Department of Biology, Faculty of Science and Medicine, University of Fribourg, Fribourg, Switzerland
| | - Benjamin Campbell
- Institute of Pharmacology and Toxicology, University of Zurich, Zurich, Switzerland
| | - Yuan-Chen Tsai
- Institute of Pharmacology and Toxicology, University of Zurich, Zurich, Switzerland
| | - Shiva K. Tyagarajan
- Institute of Pharmacology and Toxicology, University of Zurich, Zurich, Switzerland
| | - Urs Albrecht
- Department of Biology, Faculty of Science and Medicine, University of Fribourg, Fribourg, Switzerland
| | - Jürgen A. Ripperger
- Department of Biology, Faculty of Science and Medicine, University of Fribourg, Fribourg, Switzerland
- *Correspondence: Jürgen A. Ripperger,
| |
Collapse
|
28
|
Cho JH, Kim K, Cho HC, Lee J, Kim EK. Silencing of hypothalamic FGF11 prevents diet-induced obesity. Mol Brain 2022; 15:75. [PMID: 36064426 PMCID: PMC9447329 DOI: 10.1186/s13041-022-00962-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2022] [Accepted: 08/21/2022] [Indexed: 11/18/2022] Open
Abstract
Fibroblast growth factor 11 (FGF11) is a member of the intracellular fibroblast growth factor family. Here, we report the central role of FGF11 in the regulation of metabolism. Lentiviral injection of Fgf11 shRNA into the arcuate nucleus of the mouse hypothalamus decreased weight gain and fat mass, increased brown adipose tissue thermogenesis, and improved glucose and insulin intolerances under high-fat diet conditions. Fgf11 was expressed in the NPY–expressing neurons, and Fgf11 knockdown considerably decreased Npy expression and projection, leading to increased expression of tyrosine hydroxylase in the paraventricular nucleus. Mechanistically, FGF11 regulated Npy gene expression through the glycogen synthase kinase 3–cAMP response element-binding protein pathway. Our study defines the physiological significance of hypothalamic FGF11 in the regulation of metabolism in response to overnutrition such as high-fat diet.
Collapse
Affiliation(s)
- Jae Hyun Cho
- Department of Brain Sciences, Daegu Gyeongbuk Institute of Science and Technology, 333, Techno Jungang-Daero, Hyeonpung-Myeon, Daegu, Dalseonggun, 42988, South Korea
| | - Kyungchan Kim
- Department of Brain Sciences, Daegu Gyeongbuk Institute of Science and Technology, 333, Techno Jungang-Daero, Hyeonpung-Myeon, Daegu, Dalseonggun, 42988, South Korea
| | - Han Chae Cho
- Department of Brain Sciences, Daegu Gyeongbuk Institute of Science and Technology, 333, Techno Jungang-Daero, Hyeonpung-Myeon, Daegu, Dalseonggun, 42988, South Korea
| | - Jaemeun Lee
- Department of Brain Sciences, Daegu Gyeongbuk Institute of Science and Technology, 333, Techno Jungang-Daero, Hyeonpung-Myeon, Daegu, Dalseonggun, 42988, South Korea
| | - Eun-Kyoung Kim
- Department of Brain Sciences, Daegu Gyeongbuk Institute of Science and Technology, 333, Techno Jungang-Daero, Hyeonpung-Myeon, Daegu, Dalseonggun, 42988, South Korea. .,Neurometabolomics Research Center, Daegu Gyeongbuk Institute of Science and Technology, 333, Techno Jungang-Daero, Hyeonpung-Myeon, Daegu, Dalseonggun, 42988, South Korea.
| |
Collapse
|
29
|
Wang J, Beecher K, Chehrehasa F, Moody H. The limitations of investigating appetite through circuit manipulations: are we biting off more than we can chew? Rev Neurosci 2022; 34:295-311. [PMID: 36054842 DOI: 10.1515/revneuro-2022-0072] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2022] [Accepted: 07/09/2022] [Indexed: 11/15/2022]
Abstract
Disordered eating can underpin a number of debilitating and prevalent chronic diseases, such as obesity. Broader advances in psychopharmacology and biology have motivated some neuroscientists to address diet-induced obesity through reductionist, pre-clinical eating investigations on the rodent brain. Specifically, chemogenetic and optogenetic methods developed in the 21st century allow neuroscientists to perform in vivo, region-specific/projection-specific/promoter-specific circuit manipulations and immediately assess the impact of these manipulations on rodent feeding. These studies are able to rigorously conclude whether a specific neuronal population regulates feeding behaviour in the hope of eventually developing a mechanistic neuroanatomical map of appetite regulation. However, an artificially stimulated/inhibited rodent neuronal population that changes feeding behaviour does not necessarily represent a pharmacological target for treating eating disorders in humans. Chemogenetic/optogenetic findings must therefore be triangulated with the array of theories that contribute to our understanding of appetite. The objective of this review is to provide a wide-ranging discussion of the limitations of chemogenetic/optogenetic circuit manipulation experiments in rodents that are used to investigate appetite. Stepping into and outside of medical science epistemologies, this paper draws on philosophy of science, nutrition, addiction biology and neurophilosophy to prompt more integrative, transdisciplinary interpretations of chemogenetic/optogenetic appetite data. Through discussing the various technical and epistemological limitations of these data, we provide both an overview of chemogenetics and optogenetics accessible to non-neuroscientist obesity researchers, as well as a resource for neuroscientists to expand the number of lenses through which they interpret their circuit manipulation findings.
Collapse
Affiliation(s)
- Joshua Wang
- School of Clinical Sciences, Faculty of Health, Queensland University of Technology, 2 George Street, Brisbane 4000, QLD, Australia
| | - Kate Beecher
- UQ Centre for Clinical Research, Faculty of Medicine, University of Queensland, Building 71/918 Royal Brisbane and Women's Hospital Campus, Herston 4029, QLD, Australia
| | - Fatemeh Chehrehasa
- School of Biomedical Sciences, Faculty of Health, Queensland University of Technology, 2 George Street, Brisbane 4000, QLD, Australia
| | - Hayley Moody
- Queensland University of Technology, 2 George Street, Brisbane 4000, QLD, Australia
| |
Collapse
|
30
|
de Wouters d’Oplinter A, Huwart SJP, Cani PD, Everard A. Gut microbes and food reward: From the gut to the brain. Front Neurosci 2022; 16:947240. [PMID: 35958993 PMCID: PMC9358980 DOI: 10.3389/fnins.2022.947240] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2022] [Accepted: 07/04/2022] [Indexed: 11/13/2022] Open
Abstract
Inappropriate food intake behavior is one of the main drivers for fat mass development leading to obesity. Importantly the gut microbiota-mediated signals have emerged as key actors regulating food intake acting mainly on the hypothalamus, and thereby controlling hunger or satiety/satiation feelings. However, food intake is also controlled by the hedonic and reward systems leading to food intake based on pleasure (i.e., non-homeostatic control of food intake). This review focus on both the homeostatic and the non-homeostatic controls of food intake and the implication of the gut microbiota on the control of these systems. The gut-brain axis is involved in the communications between the gut microbes and the brain to modulate host food intake behaviors through systemic and nervous pathways. Therefore, here we describe several mediators of the gut-brain axis including gastrointestinal hormones, neurotransmitters, bioactive lipids as well as bacterial metabolites and compounds. The modulation of gut-brain axis by gut microbes is deeply addressed in the context of host food intake with a specific focus on hedonic feeding. Finally, we also discuss possible gut microbiota-based therapeutic approaches that could lead to potential clinical applications to restore food reward alterations. Therapeutic applications to tackle these dysregulations is of utmost importance since most of the available solutions to treat obesity present low success rate.
Collapse
|
31
|
Chen L, Wu H, Li Y, Feng X, Zhu S, Xie K, Wu X, Sun Z, Shu G, Wang S, Gao P, Zhu X, Zhu C, Jiang Q, Wang L. Corticotropin-releasing factor receptor type 2 in the midbrain critically contributes to the hedonic feeding behavior of mice under heat stress. Biochem Biophys Res Commun 2022; 602:77-83. [DOI: 10.1016/j.bbrc.2022.02.109] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2021] [Revised: 01/23/2022] [Accepted: 02/26/2022] [Indexed: 11/26/2022]
|
32
|
Watts AG, Kanoski SE, Sanchez-Watts G, Langhans W. The physiological control of eating: signals, neurons, and networks. Physiol Rev 2022; 102:689-813. [PMID: 34486393 PMCID: PMC8759974 DOI: 10.1152/physrev.00028.2020] [Citation(s) in RCA: 69] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2020] [Accepted: 08/30/2021] [Indexed: 02/07/2023] Open
Abstract
During the past 30 yr, investigating the physiology of eating behaviors has generated a truly vast literature. This is fueled in part by a dramatic increase in obesity and its comorbidities that has coincided with an ever increasing sophistication of genetically based manipulations. These techniques have produced results with a remarkable degree of cell specificity, particularly at the cell signaling level, and have played a lead role in advancing the field. However, putting these findings into a brain-wide context that connects physiological signals and neurons to behavior and somatic physiology requires a thorough consideration of neuronal connections: a field that has also seen an extraordinary technological revolution. Our goal is to present a comprehensive and balanced assessment of how physiological signals associated with energy homeostasis interact at many brain levels to control eating behaviors. A major theme is that these signals engage sets of interacting neural networks throughout the brain that are defined by specific neural connections. We begin by discussing some fundamental concepts, including ones that still engender vigorous debate, that provide the necessary frameworks for understanding how the brain controls meal initiation and termination. These include key word definitions, ATP availability as the pivotal regulated variable in energy homeostasis, neuropeptide signaling, homeostatic and hedonic eating, and meal structure. Within this context, we discuss network models of how key regions in the endbrain (or telencephalon), hypothalamus, hindbrain, medulla, vagus nerve, and spinal cord work together with the gastrointestinal tract to enable the complex motor events that permit animals to eat in diverse situations.
Collapse
Affiliation(s)
- Alan G Watts
- The Department of Biological Sciences, Dornsife College of Letters, Arts and Sciences, University of Southern California, Los Angeles, California
| | - Scott E Kanoski
- The Department of Biological Sciences, Dornsife College of Letters, Arts and Sciences, University of Southern California, Los Angeles, California
| | - Graciela Sanchez-Watts
- The Department of Biological Sciences, Dornsife College of Letters, Arts and Sciences, University of Southern California, Los Angeles, California
| | - Wolfgang Langhans
- Physiology and Behavior Laboratory, Eidgenössische Technische Hochschule-Zürich, Schwerzenbach, Switzerland
| |
Collapse
|
33
|
Martins AB, Brownlow ML, Araújo BB, Garnica-Siqueira MC, Zaia DAM, Leite CM, Zaia CTBV, Uchoa ET. Arcuate nucleus of the hypothalamus contributes to the hypophagic effect and plasma metabolic changes induced by vasoactive intestinal peptide and pituitary adenylate cyclase-activating polypeptide. Neurochem Int 2022; 155:105300. [DOI: 10.1016/j.neuint.2022.105300] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2021] [Revised: 01/20/2022] [Accepted: 01/31/2022] [Indexed: 11/30/2022]
|
34
|
Vohra MS, Benchoula K, Serpell CJ, Hwa WE. AgRP/NPY and POMC neurons in the arcuate nucleus and their potential role in treatment of obesity. Eur J Pharmacol 2022; 915:174611. [PMID: 34798121 DOI: 10.1016/j.ejphar.2021.174611] [Citation(s) in RCA: 84] [Impact Index Per Article: 28.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2021] [Revised: 10/27/2021] [Accepted: 10/27/2021] [Indexed: 02/08/2023]
Abstract
Obesity is a major health crisis affecting over a third of the global population. This multifactorial disease is regulated via interoceptive neural circuits in the brain, whose alteration results in excessive body weight. Certain central neuronal populations in the brain are recognised as crucial nodes in energy homeostasis; in particular, the hypothalamic arcuate nucleus (ARC) region contains two peptide microcircuits that control energy balance with antagonistic functions: agouti-related peptide/neuropeptide-Y (AgRP/NPY) signals hunger and stimulates food intake; and pro-opiomelanocortin (POMC) signals satiety and reduces food intake. These neuronal peptides levels react to energy status and integrate signals from peripheral ghrelin, leptin, and insulin to regulate feeding and energy expenditure. To manage obesity comprehensively, it is crucial to understand cellular and molecular mechanisms of information processing in ARC neurons, since these regulate energy homeostasis. Importantly, a specific strategy focusing on ARC circuits needs to be devised to assist in treating obese patients and maintaining weight loss with minimal or no side effects. The aim of this review is to elucidate the recent developments in the study of AgRP-, NPY- and POMC-producing neurons, specific to their role in controlling metabolism. The impact of ghrelin, leptin, and insulin signalling via action of these neurons is also surveyed, since they also impact energy balance through this route. Lastly, we present key proteins, targeted genes, compounds, drugs, and therapies that actively work via these neurons and could potentially be used as therapeutic targets for treating obesity conditions.
Collapse
Affiliation(s)
- Muhammad Sufyan Vohra
- School of Medicine, Faculty of Health and Medical Sciences, Taylor's University Lakeside Campus, 47500, Subang Jaya, Selangor Darul Ehsan, Malaysia
| | - Khaled Benchoula
- School of Medicine, Faculty of Health and Medical Sciences, Taylor's University Lakeside Campus, 47500, Subang Jaya, Selangor Darul Ehsan, Malaysia
| | - Christopher J Serpell
- School of Physical Sciences, Ingram Building, University of Kent, Canterbury, Kent, CT2 7NH, United Kingdom
| | - Wong Eng Hwa
- School of Medicine, Faculty of Health and Medical Sciences, Taylor's University Lakeside Campus, 47500, Subang Jaya, Selangor Darul Ehsan, Malaysia.
| |
Collapse
|
35
|
Astaxanthin Exerts Anabolic Effects via Pleiotropic Modulation of the Excitable Tissue. Int J Mol Sci 2022; 23:ijms23020917. [PMID: 35055102 PMCID: PMC8778848 DOI: 10.3390/ijms23020917] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2021] [Revised: 01/04/2022] [Accepted: 01/11/2022] [Indexed: 11/27/2022] Open
Abstract
Astaxanthin is a lipid-soluble carotenoid influencing lipid metabolism, body weight, and insulin sensitivity. We provide a systematic analysis of acute and chronic effects of astaxanthin on different organs. Changes by chronic astaxanthin feeding were analyzed on general metabolism, expression of regulatory proteins in the skeletal muscle, as well as changes of excitation and synaptic activity in the hypothalamic arcuate nucleus of mice. Acute responses were also tested on canine cardiac muscle and different neuronal populations of the hypothalamic arcuate nucleus in mice. Dietary astaxanthin significantly increased food intake. It also increased protein levels affecting glucose metabolism and fatty acid biosynthesis in skeletal muscle. Inhibitory inputs innervating neurons of the arcuate nucleus regulating metabolism and food intake were strengthened by both acute and chronic astaxanthin treatment. Astaxanthin moderately shortened cardiac action potentials, depressed their plateau potential, and reduced the maximal rate of depolarization. Based on its complex actions on metabolism and food intake, our data support the previous findings that astaxanthin is suitable for supplementing the diet of patients with disturbances in energy homeostasis.
Collapse
|
36
|
McIlwraith EK, Lieu CV, Belsham DD. Bisphenol A induces miR-708-5p through an ER stress-mediated mechanism altering neuronatin and neuropeptide Y expression in hypothalamic neuronal models. Mol Cell Endocrinol 2022; 539:111480. [PMID: 34624438 DOI: 10.1016/j.mce.2021.111480] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/10/2021] [Revised: 09/26/2021] [Accepted: 10/01/2021] [Indexed: 01/15/2023]
Abstract
Bisphenol A (BPA) is an endocrine disrupting chemical that promotes obesity. It acts on the hypothalamus by increasing expression of the orexigenic neuropeptides, Npy and Agrp. Exactly how BPA dysregulates energy homeostasis is not completely clear. Since microRNAs (miRNA) have emerged as crucial weight regulators, the question of whether BPA could alter hypothalamic miRNA profiles was examined. Treatment of the mHypoA-59 cell line with 100 μM BPA altered a specific subset of miRNAs, and the most upregulated was miR-708-5p. BPA was found to increase the levels of miR-708-5p, and its parent gene Odz4, through the ER stress-related protein Chop. Overexpression of an miR-708-5p mimic resulted in a reduction of neuronatin, a proteolipid whose loss of expression is associated with obesity, and an increase in orexigenic Npy expression, thus potentially increasing feeding through converging regulatory pathways. Therefore, hypothalamic exposure to BPA can increase miR-708-5p that controls neuropeptides directly linked to obesity.
Collapse
Affiliation(s)
- Emma K McIlwraith
- Departments of Physiology, University of Toronto, Ontario, M5S 1A8, Canada
| | - Calvin V Lieu
- Departments of Physiology, University of Toronto, Ontario, M5S 1A8, Canada
| | - Denise D Belsham
- Departments of Physiology, University of Toronto, Ontario, M5S 1A8, Canada; Departments of Medicine, University of Toronto, Ontario, M5S 1A8, Canada; Departments of Obstetrics and Gynaecology, University of Toronto, Ontario, M5S 1A8, Canada.
| |
Collapse
|
37
|
Shapiro NL, Todd EG, Billot B, Cash DM, Iglesias JE, Warren JD, Rohrer JD, Bocchetta M. In vivo hypothalamic regional volumetry across the frontotemporal dementia spectrum. Neuroimage Clin 2022; 35:103084. [PMID: 35717886 PMCID: PMC9218583 DOI: 10.1016/j.nicl.2022.103084] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2022] [Revised: 06/07/2022] [Accepted: 06/11/2022] [Indexed: 11/04/2022]
Abstract
BACKGROUND Frontotemporal dementia (FTD) is a spectrum of diseases characterised by language, behavioural and motor symptoms. Among the different subcortical regions implicated in the FTD symptomatology, the hypothalamus regulates various bodily functions, including eating behaviours which are commonly present across the FTD spectrum. The pattern of specific hypothalamic involvement across the clinical, pathological, and genetic forms of FTD has yet to be fully investigated, and its possible associations with abnormal eating behaviours have yet to be fully explored. METHODS Using an automated segmentation tool for volumetric T1-weighted MR images, we measured hypothalamic regional volumes in a cohort of 439 patients with FTD (197 behavioural variant FTD [bvFTD]; 7 FTD with associated motor neurone disease [FTD-MND]; 99 semantic variant primary progressive aphasia [svPPA]; 117 non-fluent variant PPA [nfvPPA]; 19 PPA not otherwise specified [PPA-NOS]) and 118 age-matched controls. We compared volumes across the clinical, genetic (29 MAPT, 32 C9orf72, 23 GRN), and pathological diagnoses (61 tauopathy, 40 TDP-43opathy, 4 FUSopathy). We correlated the volumes with presence of abnormal eating behaviours assessed with the revised version of the Cambridge Behavioural Inventory (CBI-R). RESULTS On average, FTD patients showed 14% smaller hypothalamic volumes than controls. The groups with the smallest hypothalamic regions were FTD-MND (20%), MAPT (25%) and FUS (33%), with differences mainly localised in the anterior and posterior regions. The inferior tuberal region was only significantly smaller in tauopathies (MAPT and Pick's disease) and in TDP-43 type C compared to controls and was the only regions that did not correlate with eating symptoms. PPA-NOS and nfvPPA were the groups with the least frequent eating behaviours and the least hypothalamic involvement. CONCLUSIONS Abnormal hypothalamic volumes are present in all the FTD forms, but different hypothalamic regions might play a different role in the development of abnormal eating behavioural and metabolic symptoms. These findings might therefore help in the identification of different underlying pathological mechanisms, suggesting the potential use of hypothalamic imaging biomarkers and the research of potential therapeutic targets within the hypothalamic neuropeptides.
Collapse
Affiliation(s)
- Noah L Shapiro
- Dementia Research Centre, Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, University College London, UK
| | - Emily G Todd
- Dementia Research Centre, Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, University College London, UK
| | - Benjamin Billot
- Centre for Medical Image Computing, Department of Medical Physics and Biomedical Engineering, University College London, UK
| | - David M Cash
- Dementia Research Centre, Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, University College London, UK; Centre for Medical Image Computing, Department of Medical Physics and Biomedical Engineering, University College London, UK; UK Dementia Research Institute at UCL, UCL, London, UK
| | - Juan Eugenio Iglesias
- Centre for Medical Image Computing, Department of Medical Physics and Biomedical Engineering, University College London, UK; Martinos Center for Biomedical Imaging, Massachusetts General Hospital and Harvard Medical School, Boston, USA; Computer Science and Artificial Intelligence Laboratory, Massachusetts Institute of Technology, Boston, USA
| | - Jason D Warren
- Dementia Research Centre, Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, University College London, UK
| | - Jonathan D Rohrer
- Dementia Research Centre, Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, University College London, UK
| | - Martina Bocchetta
- Dementia Research Centre, Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, University College London, UK.
| |
Collapse
|
38
|
van Schaik J, Welling MS, de Groot CJ, van Eck JP, Juriaans A, Burghard M, Oude Ophuis SBJ, Bakker B, Tissing WJE, Schouten-van Meeteren AYN, van den Akker ELT, van Santen HM. Dextroamphetamine Treatment in Children With Hypothalamic Obesity. Front Endocrinol (Lausanne) 2022; 13:845937. [PMID: 35355559 PMCID: PMC8959487 DOI: 10.3389/fendo.2022.845937] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/30/2021] [Accepted: 02/09/2022] [Indexed: 12/14/2022] Open
Abstract
INTRODUCTION Hypothalamic obesity (HO) in children has severe health consequences. Lifestyle interventions are mostly insufficient and currently no drug treatment is approved for children with HO. Amphetamines are known for their stimulant side-effect on resting energy expenditure (REE) and suppressing of appetite. Earlier case series have shown positive effects of amphetamines on weight in children with acquired HO. We present our experiences with dextroamphetamine treatment in the, up to now, largest cohort of children with HO. METHODS A retrospective cohort evaluation was performed of children with HO treated with dextroamphetamine at two academic endocrine pediatric clinics. Off-label use of dextroamphetamine was initiated in patients with progressive, therapy-resistant acquired or congenital HO. Anthropometrics, REE, self-reported (hyperphagic) behavior and energy level, and side effects were assessed at start and during treatment. RESULTS Nineteen patients with a mean age of 12.3 ± 4.0 years had been treated with dextroamphetamine. In two patients, ΔBMI SDS could not be evaluated due to short treatment duration or the simultaneous start of extensive lifestyle treatment. Mean treatment duration of the 17 evaluated patients was 23.7 ± 12.7 months. Fourteen patients (n = 10 with acquired HO, n = 4 with congenital HO) responded by BMI decline or BMI stabilization (mean ΔBMI SDS of -0.6 ± 0.8, after a mean period of 22.4 ± 10.5 months). In three patients, BMI SDS increased (mean ΔBMI SDS of +0.5 ± 0.1, after a mean period of 29.7 ± 22.6 months). In 11 responders, measured REE divided by predicted REE increased with +8.9%. Thirteen patients (68.4%) reported decreased hyperphagia, improvement of energy level and/or behavior during treatment. Two patients developed hypertension during treatment, which resulted in dosage adjustment or discontinuation of treatment. Twelve children continued treatment at last moment of follow-up. CONCLUSION In addition to supportive lifestyle interventions, dextroamphetamine treatment may improve BMI in children with HO. Furthermore, dextroamphetamines have the potential to decrease hyperphagia and improve resting energy expenditure, behavior, and energy level. In patients with acquired HO, these effects seem to be more pronounced when compared to patients with congenital HO. Future studies are needed to support these results.
Collapse
Affiliation(s)
- Jiska van Schaik
- Division of Pediatric Endocrinology, Wilhelmina Children’s Hospital, University Medical Center Utrecht, Utrecht, Netherlands
- Department of Pediatric Oncology, Princess Máxima Center for Pediatric Oncology, Utrecht, Netherlands
- *Correspondence: Jiska van Schaik,
| | - Mila S. Welling
- Obesity Centre Centrum Gezond Gewicht (CGG), Erasmus Medical Center (MC) Sophia Children’s Hospital, University Medical Center Rotterdam, Rotterdam, Netherlands
- Division of Pediatric Endocrinology, Erasmus Medical Center (MC) Sophia Children’s Hospital, University Medical Center Rotterdam, Rotterdam, Netherlands
| | - Corjan J. de Groot
- Obesity Centre Centrum Gezond Gewicht (CGG), Erasmus Medical Center (MC) Sophia Children’s Hospital, University Medical Center Rotterdam, Rotterdam, Netherlands
- Division of Pediatric Endocrinology, Erasmus Medical Center (MC) Sophia Children’s Hospital, University Medical Center Rotterdam, Rotterdam, Netherlands
| | - Judith P. van Eck
- Obesity Centre Centrum Gezond Gewicht (CGG), Erasmus Medical Center (MC) Sophia Children’s Hospital, University Medical Center Rotterdam, Rotterdam, Netherlands
- Division of Pediatric Endocrinology, Erasmus Medical Center (MC) Sophia Children’s Hospital, University Medical Center Rotterdam, Rotterdam, Netherlands
| | - Alicia Juriaans
- Division of Pediatric Endocrinology, Erasmus Medical Center (MC) Sophia Children’s Hospital, University Medical Center Rotterdam, Rotterdam, Netherlands
| | - Marcella Burghard
- Department of Pediatric Oncology, Princess Máxima Center for Pediatric Oncology, Utrecht, Netherlands
- Department of Exercise Physiology, Child Development & Exercise Center, Wilhelmina Children’s Hospital, University Medical Center Utrecht, Utrecht, Netherlands
| | - Sebastianus B. J. Oude Ophuis
- Department of Pediatric Oncology, Princess Máxima Center for Pediatric Oncology, Utrecht, Netherlands
- Department of Pediatric Psychiatry, Wilhelmina Children’s Hospital, University Medical Center Utrecht, Utrecht, Netherlands
| | - Boudewijn Bakker
- Division of Pediatric Endocrinology, Wilhelmina Children’s Hospital, University Medical Center Utrecht, Utrecht, Netherlands
- Department of Pediatric Oncology, Princess Máxima Center for Pediatric Oncology, Utrecht, Netherlands
| | - Wim J. E. Tissing
- Department of Pediatric Oncology, Princess Máxima Center for Pediatric Oncology, Utrecht, Netherlands
- Department of Pediatric Oncology/ Hematology, University of Groningen, University Medical Center Groningen, Groningen, Netherlands
| | | | - Erica L. T. van den Akker
- Obesity Centre Centrum Gezond Gewicht (CGG), Erasmus Medical Center (MC) Sophia Children’s Hospital, University Medical Center Rotterdam, Rotterdam, Netherlands
- Division of Pediatric Endocrinology, Erasmus Medical Center (MC) Sophia Children’s Hospital, University Medical Center Rotterdam, Rotterdam, Netherlands
| | - Hanneke M. van Santen
- Division of Pediatric Endocrinology, Wilhelmina Children’s Hospital, University Medical Center Utrecht, Utrecht, Netherlands
- Department of Pediatric Oncology, Princess Máxima Center for Pediatric Oncology, Utrecht, Netherlands
| |
Collapse
|
39
|
Wang H, Fang X, Ren Q, Zeng Y, Tan X, Ye T, Fan J, Xu Y. Association Between Circulating Regulator of Calcineurin 2 Concentrations With Overweight and Obesity. Front Endocrinol (Lausanne) 2022; 13:857841. [PMID: 35733783 PMCID: PMC9208054 DOI: 10.3389/fendo.2022.857841] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/19/2022] [Accepted: 04/28/2022] [Indexed: 12/01/2022] Open
Abstract
BACKGROUND Regulator of calcineurin 2 (RCAN2) has been reported to promote food intake and weight gain in animal studies. However, its effect on body weight in humans is unclear. OBJECTIVE This study aimed to investigate the relationship between serum RCAN2 concentrations and participants with overweight/obesity. METHODS A cross-sectional study was performed in 872 Chinese adults, including 348 participants with normal weight (NW), 397 participants with overweight (OW), and 127 participants with obesity (OB). All participants were divided into NW, OW and OB groups according to their body mass index (BMI). Serum RCAN2 concentrations were determined by enzyme-linked immunosorbent assay. RESULTS Serum RCAN2 concentrations gradually increased with the increase of BMI (p < 0.001). The percentages of OW/OB gradually increased in tandem with increasing tertiles of RCAN2 (p < 0.001). Additionally, serum RCAN2 concentrations were significantly correlated with a series of anthropometric and metabolic parameters, predominantly including body weight, BMI, SBP, DBP, total cholesterol, triglycerides, HDL-C, LDL-C (all p < 0.05). Furthermore, logistic regression analysis showed that the risk of OW/OB was significantly increased with the increase of serum RCAN2 concentrations. Receiver operation characteristic (ROC) curve analysis revealed that serum RCAN2, especially serum RCAN2/(AST/ALT) ratio, might serve as a candidate biomarker for obesity. CONCLUSION Serum RCAN2 concentrations were increased in subjects with OW/OB. The increased serum RCAN2 concentrations were associated with the increased risks of OW/OB.
Collapse
Affiliation(s)
- Hongya Wang
- Department of Endocrinology and Metabolism, The Affiliated Hospital of Southwest Medical University, Luzhou, China
- Cardiovascular and Metabolic Diseases Key Laboratory of Luzhou, Luzhou, China
- Sichuan Clinical Research Center for Nephropathy, Luzhou, China
- Metabolic Vascular Disease Key Laboratory of Sichuan Province, Luzhou, China
| | - Xia Fang
- Department of Endocrinology and Metabolism, The Affiliated Hospital of Southwest Medical University, Luzhou, China
- Cardiovascular and Metabolic Diseases Key Laboratory of Luzhou, Luzhou, China
- Sichuan Clinical Research Center for Nephropathy, Luzhou, China
- Metabolic Vascular Disease Key Laboratory of Sichuan Province, Luzhou, China
| | - Qian Ren
- Department of Endocrinology and Metabolism, The Affiliated Hospital of Southwest Medical University, Luzhou, China
- Cardiovascular and Metabolic Diseases Key Laboratory of Luzhou, Luzhou, China
- Sichuan Clinical Research Center for Nephropathy, Luzhou, China
- Metabolic Vascular Disease Key Laboratory of Sichuan Province, Luzhou, China
| | - Yan Zeng
- Department of Endocrinology and Metabolism, The Affiliated Hospital of Southwest Medical University, Luzhou, China
- Cardiovascular and Metabolic Diseases Key Laboratory of Luzhou, Luzhou, China
- Sichuan Clinical Research Center for Nephropathy, Luzhou, China
- Metabolic Vascular Disease Key Laboratory of Sichuan Province, Luzhou, China
| | - Xiaozhen Tan
- Department of Endocrinology and Metabolism, The Affiliated Hospital of Southwest Medical University, Luzhou, China
- Cardiovascular and Metabolic Diseases Key Laboratory of Luzhou, Luzhou, China
- Sichuan Clinical Research Center for Nephropathy, Luzhou, China
- Metabolic Vascular Disease Key Laboratory of Sichuan Province, Luzhou, China
| | - Ting Ye
- Department of Laboratory Medicine, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Jiahao Fan
- Department of Gastroenterology, The Affiliated Hospital of Southwest Medical University, Luzhou, China
- *Correspondence: Jiahao Fan, ; Yong Xu,
| | - Yong Xu
- Department of Endocrinology and Metabolism, The Affiliated Hospital of Southwest Medical University, Luzhou, China
- Cardiovascular and Metabolic Diseases Key Laboratory of Luzhou, Luzhou, China
- Sichuan Clinical Research Center for Nephropathy, Luzhou, China
- Metabolic Vascular Disease Key Laboratory of Sichuan Province, Luzhou, China
- *Correspondence: Jiahao Fan, ; Yong Xu,
| |
Collapse
|
40
|
Extrahypothalamic Control of Energy Balance and Its Connection with Reproduction: Roles of the Amygdala. Metabolites 2021; 11:metabo11120837. [PMID: 34940594 PMCID: PMC8708157 DOI: 10.3390/metabo11120837] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2021] [Revised: 12/02/2021] [Accepted: 12/02/2021] [Indexed: 11/24/2022] Open
Abstract
Body energy and metabolic homeostasis are exquisitely controlled by multiple, often overlapping regulatory mechanisms, which permit the tight adjustment between fuel reserves, internal needs, and environmental (e.g., nutritional) conditions. As such, this function is sensitive to and closely connected with other relevant bodily systems, including reproduction and gonadal function. The aim of this mini-review article is to summarize the most salient experimental data supporting a role of the amygdala as a key brain region for emotional learning and behavior, including reward processing, in the physiological control of feeding and energy balance. In particular, a major focus will be placed on the putative interplay between reproductive signals and amygdala pathways, as it pertains to the control of metabolism, as complementary, extrahypothalamic circuit for the integral control of energy balance and gonadal function.
Collapse
|
41
|
Bonneville JF, Tshibanda L, Beckers A. An Infundibular Unidentified Object (IUO): a new pituitary stalk marker? Pituitary 2021; 24:964-969. [PMID: 34254253 DOI: 10.1007/s11102-021-01169-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 06/29/2021] [Indexed: 11/24/2022]
Abstract
PURPOSE Measurement of the pituitary stalk (PS) diameter does not always solve the issue of minimal PS thickening. A previously undescribed image is found at the infundibular level on high resolution thin section T2W MRI in a large number of normal individuals. We speculate that this image-whose exact origin is still unknown-may serve as a marker of the normal infundibulum. METHODS In the last 6 months, 350 consecutive adult patients suspected of sellar pathology or controlled after medical or surgical treatment prospectively underwent a pituitary MRI including a sagittal T2W high resolution sequence. One hundred twelwe patients presenting a pituitary mass with suprasellar extension or those whose PS was not entirely visible were excluded. RESULTS A short focal annular T2 hypointense thickening of the wall of the infundibular recess of the third ventricle, more pronounced anteriorly was found in 151/238 patients. Additionally, a more or less tiny ventral extension was demonstrated on sagittal T2W sequence in 105/151 patients. These images were not identified on T1W or on T1W gadolinium enhanced sequences. The ring-like infundibular thickening and/or its ventral extension were not identified in 87/238 patients; in 43/87 of these patients the PS was found severely stretched mainly in case of primary or secondary empty sella. If patients with empty sella were excluded, our finding was observed in 194/238 cases, i.e. in 82%. CONCLUSIONS A detailed appearance of the PS on T2W MRI is described for the first time. A previously unreported T2W hypointense annular focal image prolonged by a tiny spicular or nodular ventral bud is found at the lower part of the infundibulum in a majority of normal patients, but not if the PS is stretched such as in empty sella. This image has to be recognized as a normal anatomical landmark. The possible origin of this image is discussed but not totally elucidated. An ongoing research will demonstrate or not if this image may serve as a marker to improve the early diagnosis of PS lesions.
Collapse
Affiliation(s)
- J-F Bonneville
- Centre Hospitalier Universitaire de Liege, Liège, Belgium.
| | - L Tshibanda
- Centre Hospitalier Universitaire de Liege, Liège, Belgium
| | - A Beckers
- Centre Hospitalier Universitaire de Liege, Liège, Belgium
| |
Collapse
|
42
|
Proteome Analysis of the Hypothalamic Arcuate Nucleus in Chronic High-Fat Diet-Induced Obesity. BIOMED RESEARCH INTERNATIONAL 2021; 2021:3501770. [PMID: 34840970 PMCID: PMC8617565 DOI: 10.1155/2021/3501770] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/28/2021] [Accepted: 10/25/2021] [Indexed: 12/18/2022]
Abstract
The hypothalamus plays a central role in the integrated regulation of feeding and energy homeostasis. The hypothalamic arcuate nucleus (ARC) contains a population of neurons that express orexigenic and anorexigenic factors and is thought to control feeding behavior via several neuronal circuits. In this study, a comparative proteomic analysis of low-fat control diet- (LFD-) and high-fat diet- (HFD-) induced hypothalamic ARC was performed to identify differentially expressed proteins (DEPs) related to changes in body weight. In the ARC in the hypothalamus, 6621 proteins (FDR < 0.01) were detected, and 178 proteins were categorized as DEPs (89 upregulated and 89 downregulated in the HFD group). Among the Gene Ontology molecular function terms associated with the DEPs, protein binding was the most significant. Fibroblast growth factor receptor substrate 2 (Frs2) and SHC adaptor protein 3 (Shc3) were related to protein binding and involved in the neurotrophin signaling pathway according to Kyoto Encyclopedia of Genes and Genomes analysis. Furthermore, high-precision quantitative proteomic analysis revealed that the protein profile of the ARC in mice with HFD-induced obesity differed from that in LFD mice, thereby offering insight into the molecular basis of feeding regulation and suggesting Frs2 and Shc3 as novel treatment targets for central anorexigenic signal induction.
Collapse
|
43
|
Mahmoudinezhad M, Abbasalizad Farhangi M. Association between Ag-RP, alpha-MSH and cardiovascular risk factors regarding adherence to diet quality index-international (DQI-I) among obese individuals. J Cardiovasc Thorac Res 2021; 13:320-329. [PMID: 35047137 PMCID: PMC8749370 DOI: 10.34172/jcvtr.2021.48] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2021] [Revised: 10/12/2021] [Accepted: 11/04/2021] [Indexed: 11/15/2022] Open
Abstract
Introduction: Obesity is a strong promoter of cardiometabolic risk factors and is associated with several chronic comorbidities. Recently, the role of α-melanocyte stimulating hormone (α-MSH) and agouti related peptide (Ag-RP) in regulation of energy balance has attracted much attention. In current study, we evaluated the association between α-MSH and Ag-RP with cardiometabolic factors among obese individuals with different adherence to Diet Quality Index-International (DQI-I) values.
Methods: In this research, 188 obese adults aged between 20 and 50 years old and body mass index (BMI) between 30 and 40 kg/m2 were recruited. Dietary intakes of participants and DQI-I calculation was performed using a semi-quantitative food frequency questionnaire (FFQ) with 132 food items. Serum glucose, lipids, insulin, and plasma α-MSH and Ag-RP levels were measured using ELISA kits. Homeostasis model assessment for insulin resistance index (HOMA-IR) and quantitative insulin sensitivity check index (QUICKI) were also calculated.
Results: Among those with the lowest adherence to DQI-I, Ag-RP was positively associated with systolic blood pressure (SBP) (P = 0.03) among males, which was associated with waist circumference (WC) (P = 0.01) and diastolic blood pressure (DBP) (P = 0.01). Moreover, among males with low and moderate adherence to DQI-I, α-MSH was positively associated with insulin (P = 0.04), weight (P = 0.03), WC (P < 0.01), SDP (P = 0.02) and DBP (P = 0.01). Also, Ag-RP showed a positive association with BMI values (R2 = 0.03; P = 0.03).
Conclusion: According to our findings, in obese subjects with poor to moderate adherence to DQI-I, Ag-RP and α-MSH were in positive correlation with cardiometabolic risk factors. These findings further clarify the clinical importance of these parameters as prognostic factors of cardiometabolic abnormalities.
Collapse
Affiliation(s)
- Mahsa Mahmoudinezhad
- Drug Applied Research Center, Department of Community Nutrition, Faculty of Nutrition, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mahdieh Abbasalizad Farhangi
- Drug Applied Research Center, Department of Community Nutrition, Faculty of Nutrition, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
44
|
Lieu CV, Loganathan N, Belsham DD. Mechanisms Driving Palmitate-Mediated Neuronal Dysregulation in the Hypothalamus. Cells 2021; 10:3120. [PMID: 34831343 PMCID: PMC8617942 DOI: 10.3390/cells10113120] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2021] [Revised: 11/03/2021] [Accepted: 11/04/2021] [Indexed: 12/17/2022] Open
Abstract
The hypothalamus maintains whole-body homeostasis by integrating information from circulating hormones, nutrients and signaling molecules. Distinct neuronal subpopulations that express and secrete unique neuropeptides execute the individual functions of the hypothalamus, including, but not limited to, the regulation of energy homeostasis, reproduction and circadian rhythms. Alterations at the hypothalamic level can lead to a myriad of diseases, such as type 2 diabetes mellitus, obesity, and infertility. The excessive consumption of saturated fatty acids can induce neuroinflammation, endoplasmic reticulum stress, and resistance to peripheral signals, ultimately leading to hyperphagia, obesity, impaired reproductive function and disturbed circadian rhythms. This review focuses on the how the changes in the underlying molecular mechanisms caused by palmitate exposure, the most commonly consumed saturated fatty acid, and the potential involvement of microRNAs, a class of non-coding RNA molecules that regulate gene expression post-transcriptionally, can result in detrimental alterations in protein expression and content. Studying the involvement of microRNAs in hypothalamic function holds immense potential, as these molecular markers are quickly proving to be valuable tools in the diagnosis and treatment of metabolic disease.
Collapse
Affiliation(s)
- Calvin V. Lieu
- Department of Physiology, University of Toronto, Medical Sciences Building 3247A, 1 King’s College Circle, Toronto, ON M5S 1A8, Canada; (C.V.L.); (N.L.)
| | - Neruja Loganathan
- Department of Physiology, University of Toronto, Medical Sciences Building 3247A, 1 King’s College Circle, Toronto, ON M5S 1A8, Canada; (C.V.L.); (N.L.)
| | - Denise D. Belsham
- Department of Physiology, University of Toronto, Medical Sciences Building 3247A, 1 King’s College Circle, Toronto, ON M5S 1A8, Canada; (C.V.L.); (N.L.)
- Departments of Obstetrics/Gynecology and Medicine, University of Toronto, Toronto, ON M5S 1A8, Canada
| |
Collapse
|
45
|
Tran A, He W, Chen JTC, Wellhauser L, Hopperton KE, Bazinet RP, Belsham DD. Palmitate-mediated induction of neuropeptide Y expression occurs through intracellular metabolites and not direct exposure to proinflammatory cytokines. J Neurochem 2021; 159:574-589. [PMID: 34482548 DOI: 10.1111/jnc.15504] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2021] [Revised: 08/19/2021] [Accepted: 08/28/2021] [Indexed: 12/11/2022]
Abstract
A contributing factor to the development of obesity is the consumption of a diet high in saturated fatty acids, such as palmitate. These fats induce hypothalamic neuroinflammation, which dysregulates neuronal function and induces orexigenic neuropeptide Y (Npy) to promote food intake. An inflammatory cytokine array identified multiple candidates that could mediate palmitate-induced up-regulation of Npy mRNA levels. Of these, visfatin or nicotinamide phosphoribosyltransferase (NAMPT), macrophage migratory inhibitory factor (MIF), and IL-17F were chosen for further study. Direct treatment of the neuropeptide Y/agouti-related peptide (NPY/AgRP)-expressing mHypoE-46 neuronal cell line with the aforementioned cytokines demonstrated that visfatin could directly induce Npy mRNA expression. Preventing the intracellular metabolism of palmitate through long-chain acyl-CoA synthetase (ACSL) inhibition was sufficient to block the palmitate-mediated increase in Npy gene expression. Furthermore, thin-layer chromatography revealed that in neurons, palmitate is readily incorporated into ceramides and defined species of phospholipids. Exogenous C16 ceramide, dipalmitoyl-phosphatidylcholine, and dipalmitoyl-phosphatidylethanolamine were sufficient to significantly induce Npy expression. This study suggests that the intracellular metabolism of palmitate and elevation of metabolites, including ceramide and phospholipids, are responsible for the palmitate-mediated induction of the potent orexigen Npy. Furthermore, this suggests that the regulation of Npy expression is less reliant on inflammatory cytokines per se than palmitate metabolites in a model of NPY/AgRP neurons. These lipid species likely induce detrimental downstream cellular signaling events ultimately causing an increase in feeding, resulting in an overweight phenotype and/or obesity.
Collapse
Affiliation(s)
- Andy Tran
- Department of Physiology, University of Toronto, Ontario, Canada
| | - Wenyuan He
- Department of Physiology, University of Toronto, Ontario, Canada
| | - Jim T C Chen
- Department of Physiology, University of Toronto, Ontario, Canada
| | - Leigh Wellhauser
- Department of Physiology, University of Toronto, Ontario, Canada
| | | | | | - Denise D Belsham
- Department of Physiology, University of Toronto, Ontario, Canada.,Medicine, University of Toronto, Ontario, Canada.,Obstetrics and Gynaecology, University of Toronto, Ontario, Canada
| |
Collapse
|
46
|
Extra-forebrain impact of antipsychotics indicated by c-Fos or FosB/ΔFosB expression: A minireview. Endocr Regul 2021; 55:120-130. [PMID: 34020528 DOI: 10.2478/enr-2021-0013] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/27/2023] Open
Abstract
It is apparent that the c-Fos and FosB/ΔFosB immunohistochemistry has generally become a useful tool for determining the different antipsychotic (AP) drugs activities in the brain. It is also noteworthy that there are no spatial limits, while to the extent of their identification over the whole brain axis. In addition, they can be in a parallel manner utilized in the unmasking of the brain cell phenotype character activated by APs and by this way also to identify the possible brain circuits underwent to the APs action. However, up to date, the number of APs involved in the extra-striatal studies is still limited, what prevents the possibility to fully understand their extra-striatal effects as a complex as well as differentiate their extra-striatal impact in qualitative and quantitative dimensions. Actually, it is very believable that more and more anatomical/functional knowledge might bring new insights into the APs extra-striatal actions by identifying new region-specific activities of APs as well as novel cellular targets affected by APs, which might reveal more details of their possible side effects of the extra-striatal origin.
Collapse
|
47
|
Quiñones M, Hernández-Bautista R, Beiroa D, Heras V, Torres-Leal FL, Lam BYH, Senra A, Fernø J, Gómez-Valadés AG, Schwaninger M, Prevot V, Yeo G, Claret M, López M, Diéguez C, Al-Massadi O, Nogueiras R. Sirt3 in POMC neurons controls energy balance in a sex- and diet-dependent manner. Redox Biol 2021; 41:101945. [PMID: 33744652 PMCID: PMC8005845 DOI: 10.1016/j.redox.2021.101945] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2021] [Revised: 03/09/2021] [Accepted: 03/10/2021] [Indexed: 02/06/2023] Open
Abstract
Sirtuin 3 (SIRT3) is one of the seven mammalian sirtuin homologs of the yeast Sir2 gene that has emerged as an important player in the regulation of energy metabolism in peripheral tissues. However, its role in the hypothalamus has not been explored. Herein, we show that the genetic inhibition of SIRT3 in the hypothalamic arcuate nucleus (ARC) induced a negative energy balance and improvement of several metabolic parameters. These effects are specific for POMC neurons, because ablation of SIRT3 in POMC, but not in AgRP neurons, decreased body weight and adiposity, increased energy expenditure and brown adipose tissue (BAT) activity, and induced browning in white adipose tissue (WAT). Notably, the depletion of SIRT3 in POMC neurons caused these effects in male mice fed a chow diet but failed to affect energy balance in males fed a high fat diet and females under both type of diets. Overall, we provide the first evidence pointing for a key role of SIRT3 in POMC neurons in the regulation of energy balance.
Collapse
Affiliation(s)
- Mar Quiñones
- Department of Physiology, CIMUS, University of Santiago de Compostela-Instituto de Investigación Sanitaria, Santiago de Compostela, 15782, Spain; CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), 15706, Spain.
| | - René Hernández-Bautista
- Department of Physiology, CIMUS, University of Santiago de Compostela-Instituto de Investigación Sanitaria, Santiago de Compostela, 15782, Spain; CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), 15706, Spain
| | - Daniel Beiroa
- Department of Physiology, CIMUS, University of Santiago de Compostela-Instituto de Investigación Sanitaria, Santiago de Compostela, 15782, Spain; CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), 15706, Spain
| | - Violeta Heras
- Department of Physiology, CIMUS, University of Santiago de Compostela-Instituto de Investigación Sanitaria, Santiago de Compostela, 15782, Spain; CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), 15706, Spain
| | - Francisco L Torres-Leal
- Department of Physiology, CIMUS, University of Santiago de Compostela-Instituto de Investigación Sanitaria, Santiago de Compostela, 15782, Spain; Metabolic Diseases, Exercise and Nutrition (DOMEN) Research Group, Federal University of Piauí, Teresina, Brazil
| | - Brian Y H Lam
- MRC Metabolic Diseases Unit, University of Cambridge Metabolic Research Laboratories, Wellcome Trust-MRC Institute of Metabolic Science, Addenbrooke's Hospital, Cambridge, CB2 0QQ, UK
| | - Ana Senra
- Department of Physiology, CIMUS, University of Santiago de Compostela-Instituto de Investigación Sanitaria, Santiago de Compostela, 15782, Spain
| | - Johan Fernø
- Hormone Laboratory, Haukeland University Hospital, Bergen, Norway
| | - Alicia García Gómez-Valadés
- Neuronal Control of Metabolism (NeuCoMe) Laboratory, Institut D'Investigacions Biomèdiques August Pi I Sunyer (IDIBAPS), 08036, Barcelona, Spain
| | - Markus Schwaninger
- Institute for Experimental and Clinical Pharmacology and Toxicology, University of Lübeck, Lübeck, Germany
| | - Vincent Prevot
- Univ. Lille, Inserm, CHU Lille, Laboratory of Development and Plasticity of the Neuroendocrine Brain, Lille Neuroscience & Cognition, UMR-S 1172, European Genomic Institute for Diabetes (EGID), F-59000, Lille, France
| | - Giles Yeo
- MRC Metabolic Diseases Unit, University of Cambridge Metabolic Research Laboratories, Wellcome Trust-MRC Institute of Metabolic Science, Addenbrooke's Hospital, Cambridge, CB2 0QQ, UK
| | - Marc Claret
- Neuronal Control of Metabolism (NeuCoMe) Laboratory, Institut D'Investigacions Biomèdiques August Pi I Sunyer (IDIBAPS), 08036, Barcelona, Spain; CIBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), 08036, Barcelona, Spain; School of Medicine, Universitat de Barcelona, Barcelona, Spain
| | - Miguel López
- Department of Physiology, CIMUS, University of Santiago de Compostela-Instituto de Investigación Sanitaria, Santiago de Compostela, 15782, Spain; CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), 15706, Spain
| | - Carlos Diéguez
- Department of Physiology, CIMUS, University of Santiago de Compostela-Instituto de Investigación Sanitaria, Santiago de Compostela, 15782, Spain; CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), 15706, Spain
| | - Omar Al-Massadi
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), 15706, Spain; 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.
| | - Ruben Nogueiras
- Department of Physiology, CIMUS, University of Santiago de Compostela-Instituto de Investigación Sanitaria, Santiago de Compostela, 15782, Spain; CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), 15706, Spain; Galician Agency of Innovation (GAIN), Xunta de Galicia, Santiago de Compostela, Spain.
| |
Collapse
|
48
|
Mohammadi S, Moosaie F, Saghazadeh A, Mahmoudi M, Rezaei N. Metabolic profile in patients with narcolepsy: a systematic review and meta-analysis. Sleep Med 2021; 81:268-284. [PMID: 33740593 DOI: 10.1016/j.sleep.2021.02.040] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/17/2020] [Revised: 02/10/2021] [Accepted: 02/16/2021] [Indexed: 11/25/2022]
Abstract
Narcolepsy, a sleep disorder characterized by loss of hypocretin neurons, has been associated with metabolic disturbances. Although the metabolic alterations in narcolepsy patients are widely investigated in the literature, the results are controversial. We performed a systematic search of literature to identify metabolic profiling studies in narcolepsy patients. A total of 48 studies were included in the meta-analysis. Narcolepsy patients exhibited higher prevalence of obesity (log OR = 0.93 [0.73-1.13], P < 0.001), diabetes mellitus (log OR = 0.64 [0.34, 0.94], P < 0.001), hypertension (log OR = 0.33 [0.11, 0.55], P < 0.001), and dyslipidemia (log OR = 1.19 [0.60, 1.77], P < 0.001) compared with non-narcoleptic controls. Narcolepsy was associated with higher BMI (SMD = 0.50 [0.32-0.68], P < 0.001), waist circumference (MD = 8.61 [2.03-15.19], P = 0.01), and plasma insulin (SMD = 0.61 [0.14-1.09], P = 0.01). Levels of fasting blood glucose (SMD = -0.25 [-0.61,0.10], P = 0.15), BMR-RMR (SMD = -0.17 [-0.52-0.18], P = 0.34), systolic blood pressure (SMD = 0.29 [-0.39-0.97], P = 0.40), diastolic blood pressure (SMD = 0.39 [-0.62, 1.40], P = 0.45), CSF melanin-concentrating hormone (MD = 5.56 [-30.79-41.91], P = 0.76), serum growth hormone (SMD = 7.84 [-7.90-23.57], P = 0.33), as well as plasma and CSF leptin (SMD = 0.10 [-1.32-1.51], P = 0.89 and MD = 0.01 [-0.02-0.04], P = 0.56, respectively) did not significantly differ between narcolepsy patients and controls. These findings necessitate early screening of metabolic alterations and cardiovascular risk factors in narcolepsy patients to reduce the morbidity and mortality rates.
Collapse
Affiliation(s)
- Soheil Mohammadi
- School of Medicine, Tehran University of Medical Sciences, Tehran, Iran; NeuroImaging Network (NIN), Universal Scientific Education and Research Network (USERN), Tehran University of Medical Sciences, Tehran, Iran
| | - Fatemeh Moosaie
- School of Medicine, Tehran University of Medical Sciences, Tehran, Iran; NeuroImaging Network (NIN), Universal Scientific Education and Research Network (USERN), Tehran University of Medical Sciences, Tehran, Iran
| | - Amene Saghazadeh
- Research Center for Immunodeficiencies, Children's Medical Center, Tehran University of Medical Sciences, Tehran, Iran; MetaCognition Interest Group (MCIG), Universal Scientific Education and Research Network (USERN), Tehran, Iran
| | - Maryam Mahmoudi
- Department of Cellular Molecular Nutrition, School of Nutritional Sciences and Dietetics, Tehran University of Medical Sciences, Tehran, Iran; Dietitians and Nutrition Experts Team (DiNET), Universal Scientific Education and Research Network (USERN), Tehran, Iran
| | - Nima Rezaei
- Research Center for Immunodeficiencies, Children's Medical Center, Tehran University of Medical Sciences, Tehran, Iran; Department of Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran; Systematic Review and Meta-analysis Expert Group (SRMEG), Universal Scientific Education and Research Network (USERN), Tehran, Iran.
| |
Collapse
|
49
|
Caparros-Gonzalez RA, Torre-Luque ADL, Romero-Gonzalez B, Quesada-Soto JM, Alderdice F, Peralta-Ramírez MI. Stress During Pregnancy and the Development of Diseases in the offspring: A Systematic-Review and Meta-Analysis. Midwifery 2021; 97:102939. [PMID: 33647755 DOI: 10.1016/j.midw.2021.102939] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2020] [Revised: 01/21/2021] [Accepted: 02/04/2021] [Indexed: 12/19/2022]
Abstract
OBJECTIVE The goal of this systematic-review and meta-analysis was to assess whether high maternal stress during pregnancy is associated with the development of pediatric pathology. DESIGN Epidemiological peer-reviewed studies published in English or Spanish assessing associations between maternal stress during pregnancy and psychiatric and medical diseases were selected. PARTICIPANTS We retrieved 73,024 citations; 42 studies meeting inclusion criteria were assessed. Overall sample included 65,814,076 women. FINDINGS Overall odds ratio for the development of a medical disease was OR=1.24 (CI95=1.11, 1.39), Z=3.85, p<.01. Overall odds ratio for psychiatric disorders was OR=1.28 (CI95=1.06, 1.56), Z=2.54, p<.02. Multivariate meta-analysis showed a significant coefficient for autism spectrum disorder studies, B=0.42, SE=0.16, Z=2.67, p<.01. We found a significant overall effect size for autism spectrum disorder (OR=1.45 [CI95=1.24, 1.70], Z=4.69, p<.01). In terms of medical diseases, studies including obesity and infantile colic presented a significant overall effect size, as OR=1.20 (CI95=1.03, 1.39), Z=2.41, p<.02. The highest effect size was found regarding the first trimester (B=1.62, SE=0.16, Z=9.90, p<.01). KEY CONCLUSIONS We concluded that exposure to high levels of stress during pregnancy are associated with autism spectrum disorder, obesity, and infantile colic in offspring. IMPLICATIONS FOR PRACTICE Maternal stress during pregnancy should be addressed to tackle its potential impact in health across the life span.
Collapse
Affiliation(s)
| | | | - Borja Romero-Gonzalez
- Mind, Brain, and Behavior Research Center (CIMCYC), University of Granada, Granada, Spain
| | | | - Fiona Alderdice
- National Perinatal Epidemiology Unit, Nuffield Department of Population Health, Medical Sciences Division, University of Oxford, Oxford, United Kingdom
| | | |
Collapse
|
50
|
Kravchychyn ACP, Campos RMDS, Oliveira E Silva L, Ferreira YAM, Corgosinho FC, Masquio DCL, Vicente SEDCF, Oyama LM, Tock L, de Mello MT, Tufik S, Thivel D, Dâmaso AR. Adipocytokine and appetite-regulating hormone response to weight loss in adolescents with obesity: Impact of weight loss magnitude. Nutrition 2021; 87-88:111188. [PMID: 33773406 DOI: 10.1016/j.nut.2021.111188] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2020] [Revised: 01/30/2021] [Accepted: 02/01/2021] [Indexed: 12/20/2022]
Abstract
OBJECTIVE The aim of this study was to investigate the association between the magnitude of weight loss (WL) and serum concentrations of the main adipocytokines and appetite-regulating hormones in adolescents with obesity. METHODS After completion of informed consent,108 adolescents with obesity (14-19 y of age; postpubertal) were submitted to clinical, nutritional, psychological, physical exercise, and physiotherapy support for 1 y. Body composition (BC) and plasma levels of neuropeptides (neuropeptide Y [NPY], agouti-related peptide [AgRP], and α-melanocyte-stimulating hormone [α-MSH]) and leptin were measured at baseline and post-intervention. RESULTS After therapy, adolescents who lost <10% body weight and <10% body weight (were compared. Both groups presented improvements in BC and reduced leptin. The Δα-MSH, Δα-MSH/AgRP ratio, and Δα-MSH/NPY ratio were lower and AgRP and NPY variations were higher in the low weight loss group. The leptin concentration was close to normal in the high weight loss only. The ΔWeight, Δα-MSH and Δleptin were associated with body fat loss by multiple linear regressions for all samples. CONCLUSION Weight loss >10% seems to reverse obesity-induced hyperleptinemia while stabilizing the neuropeptides that control appetite in adolescents with obesity. We were able to produce a prognostic mathematical model to predict body fat loss using weight, leptin, and α-MSH variations.
Collapse
Affiliation(s)
| | | | - Luciana Oliveira E Silva
- Postgraduate Program of Nutrition, Federal University of Sao Paulo; Paulista Medicine School, Sao Paulo, Brazil; Department of Psychobiology, Universidade Federal de São Paulo(UNIFESP), Sao Paulo, Brazil
| | | | | | | | | | - Lila Missae Oyama
- Postgraduate Program of Nutrition, Federal University of Sao Paulo; Paulista Medicine School, Sao Paulo, Brazil
| | - Lian Tock
- Postgraduate Program of Nutrition, Federal University of Sao Paulo; Paulista Medicine School, Sao Paulo, Brazil
| | - Marco Túlio de Mello
- School of Physical Education, Physiotherapy and Occupational Therapy, Federal University of Minas Gerais, Belo Horizonte, Brazil
| | - Sérgio Tufik
- Department of Psychobiology, Universidade Federal de São Paulo(UNIFESP), Sao Paulo, Brazil
| | - David Thivel
- Clermont Auvergne University, CRNH Auvergne, France
| | - Ana Raimunda Dâmaso
- Postgraduate Program of Nutrition, Federal University of Sao Paulo; Paulista Medicine School, Sao Paulo, Brazil
| |
Collapse
|