1
|
Harvey T, Rios M. The Role of BDNF and TrkB in the Central Control of Energy and Glucose Balance: An Update. Biomolecules 2024; 14:424. [PMID: 38672441 PMCID: PMC11048226 DOI: 10.3390/biom14040424] [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: 02/20/2024] [Revised: 03/27/2024] [Accepted: 03/29/2024] [Indexed: 04/28/2024] Open
Abstract
The global rise in obesity and related health issues, such as type 2 diabetes and cardiovascular disease, is alarming. Gaining a deeper insight into the central neural pathways and mechanisms that regulate energy and glucose homeostasis is crucial for developing effective interventions to combat this debilitating condition. A significant body of evidence from studies in humans and rodents indicates that brain-derived neurotrophic factor (BDNF) signaling plays a key role in regulating feeding, energy expenditure, and glycemic control. BDNF is a highly conserved neurotrophin that signals via the tropomyosin-related kinase B (TrkB) receptor to facilitate neuronal survival, differentiation, and synaptic plasticity and function. Recent studies have shed light on the mechanisms through which BDNF influences energy and glucose balance. This review will cover our current understanding of the brain regions, neural circuits, and cellular and molecular mechanisms underlying the metabolic actions of BDNF and TrkB.
Collapse
Affiliation(s)
- Theresa Harvey
- Graduate Program in Neuroscience, Graduate School of Biomedical Sciences, Tufts University School of Medicine, Boston, MA 02111, USA;
| | - Maribel Rios
- Graduate Program in Neuroscience, Graduate School of Biomedical Sciences, Tufts University School of Medicine, Boston, MA 02111, USA;
- Department of Neuroscience, Tufts University School of Medicine, 136 Harrison Avenue, Boston, MA 02111, USA
| |
Collapse
|
2
|
Stincic TL, Kelly MJ. Estrogenic regulation of reproduction and energy homeostasis by a triumvirate of hypothalamic arcuate neurons. J Neuroendocrinol 2022; 34:e13145. [PMID: 35581942 DOI: 10.1111/jne.13145] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/18/2021] [Revised: 03/31/2022] [Accepted: 04/15/2022] [Indexed: 11/29/2022]
Abstract
Pregnancy is energetically demanding and therefore, by necessity, reproduction and energy balance are inextricably linked. With insufficient or excessive energy stores a female is liable to suffer complications during pregnancy or produce unhealthy offspring. Gonadotropin-releasing hormone neurons are responsible for initiating both the pulsatile and subsequent surge release of luteinizing hormone to control ovulation. Meticulous work has identified two hypothalamic populations of kisspeptin (Kiss1) neurons that are critical for this pattern of release. The involvement of the hypothalamus is unsurprising because its quintessential function is to couple the endocrine and nervous systems, coordinating energy balance and reproduction. Estrogens, more specifically 17β-estradiol (E2 ), orchestrate the activity of a triumvirate of hypothalamic neurons within the arcuate nucleus (ARH) that govern the physiological underpinnings of these behavioral dynamics. Arising from a common progenitor pool, these cells differentiate into ARH kisspeptin, pro-opiomelanocortin (POMC), and agouti related peptide/neuropeptide Y (AgRP) neurons. Although the excitability of all these subpopulations is subject to genomic and rapid estrogenic regulation, Kiss1 neurons are the most sensitive, reflecting their integral function in female fertility. Based on the premise that E2 coordinates autonomic functions around reproduction, we review recent findings on how Kiss1 neurons interact with gonadotropin-releasing hormone, AgRP and POMC neurons, as well as how the rapid membrane-initiated and intracellular signaling cascades activated by E2 in these neurons are critical for control of homeostatic functions supporting reproduction. In particular, we highlight how Kiss1 and POMC neurons conspire to inhibit AgRP neurons and diminish food motivation in service of reproductive success.
Collapse
Affiliation(s)
- Todd L Stincic
- Department of Chemical Physiology and Biochemistry, Oregon Health and Science University, Portland, OR, USA
| | - Martin J Kelly
- Department of Chemical Physiology and Biochemistry, Oregon Health and Science University, Portland, OR, USA
- Division of Neuroscience, Oregon National Primate Research Center, Oregon Health and Science University, Beaverton, OR, USA
| |
Collapse
|
3
|
Naito M, Iwakoshi-Ukena E, Moriwaki S, Narimatsu Y, Kato M, Furumitsu M, Miyamoto Y, Esumi S, Ukena K. Immunohistochemical Analysis of Neurotransmitters in Neurosecretory Protein GL-Producing Neurons of the Mouse Hypothalamus. Biomedicines 2022; 10:biomedicines10020454. [PMID: 35203663 PMCID: PMC8962320 DOI: 10.3390/biomedicines10020454] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2022] [Revised: 02/10/2022] [Accepted: 02/11/2022] [Indexed: 01/03/2023] Open
Abstract
We recently discovered a novel neuropeptide of 80 amino acid residues: neurosecretory protein GL (NPGL), in the hypothalamus of birds and rodents. NPGL is localized in the lateral posterior part of the arcuate nucleus (ArcLP), and it enhances feeding behavior and fat accumulation in mice. Various neurotransmitters, such as catecholamine, glutamate, and γ-aminobutyric acid (GABA), produced in the hypothalamus are also involved in energy metabolism. The colocalization of neurotransmitters and NPGL in neurons of the ArcLP leads to the elucidation of the regulatory mechanism of NPGL neurons. In this study, we performed double immunofluorescence staining to elucidate the relationship between NPGL and neurotransmitters in mice. The present study revealed that NPGL neurons did not co-express tyrosine hydroxylase as a marker of catecholaminergic neurons and vesicular glutamate transporter-2 as a marker of glutamatergic neurons. In contrast, NPGL neurons co-produced glutamate decarboxylase 67, a marker for GABAergic neurons. In addition, approximately 50% of NPGL neurons were identical to GABAergic neurons. These results suggest that some functions of NPGL neurons may be related to those of GABA. This study provides insights into the neural network of NPGL neurons that regulate energy homeostasis, including feeding behavior and fat accumulation.
Collapse
Affiliation(s)
- Mana Naito
- Laboratory of Neurometabolism, Graduate School of Integrated Sciences for Life, Hiroshima University, Hiroshima 739-8521, Japan; (M.N.); (E.I.-U.); (S.M.); (Y.N.); (M.K.); (M.F.)
| | - Eiko Iwakoshi-Ukena
- Laboratory of Neurometabolism, Graduate School of Integrated Sciences for Life, Hiroshima University, Hiroshima 739-8521, Japan; (M.N.); (E.I.-U.); (S.M.); (Y.N.); (M.K.); (M.F.)
| | - Shogo Moriwaki
- Laboratory of Neurometabolism, Graduate School of Integrated Sciences for Life, Hiroshima University, Hiroshima 739-8521, Japan; (M.N.); (E.I.-U.); (S.M.); (Y.N.); (M.K.); (M.F.)
| | - Yuki Narimatsu
- Laboratory of Neurometabolism, Graduate School of Integrated Sciences for Life, Hiroshima University, Hiroshima 739-8521, Japan; (M.N.); (E.I.-U.); (S.M.); (Y.N.); (M.K.); (M.F.)
| | - Masaki Kato
- Laboratory of Neurometabolism, Graduate School of Integrated Sciences for Life, Hiroshima University, Hiroshima 739-8521, Japan; (M.N.); (E.I.-U.); (S.M.); (Y.N.); (M.K.); (M.F.)
| | - Megumi Furumitsu
- Laboratory of Neurometabolism, Graduate School of Integrated Sciences for Life, Hiroshima University, Hiroshima 739-8521, Japan; (M.N.); (E.I.-U.); (S.M.); (Y.N.); (M.K.); (M.F.)
| | - Yuta Miyamoto
- Department of Anatomy and Neurobiology, Graduate School of Medical Sciences, Kumamoto University, Kumamoto 860-8556, Japan; (Y.M.); (S.E.)
| | - Shigeyuki Esumi
- Department of Anatomy and Neurobiology, Graduate School of Medical Sciences, Kumamoto University, Kumamoto 860-8556, Japan; (Y.M.); (S.E.)
| | - Kazuyoshi Ukena
- Laboratory of Neurometabolism, Graduate School of Integrated Sciences for Life, Hiroshima University, Hiroshima 739-8521, Japan; (M.N.); (E.I.-U.); (S.M.); (Y.N.); (M.K.); (M.F.)
- Correspondence:
| |
Collapse
|
4
|
van Galen KA, Ter Horst KW, Serlie MJ. Serotonin, food intake, and obesity. Obes Rev 2021; 22:e13210. [PMID: 33559362 PMCID: PMC8243944 DOI: 10.1111/obr.13210] [Citation(s) in RCA: 39] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/08/2020] [Revised: 12/28/2020] [Accepted: 12/28/2020] [Indexed: 12/16/2022]
Abstract
The role of serotonin in food intake has been studied for decades. Food intake is mainly regulated by two brain circuitries: (i) the homeostatic circuitry, which matches energy intake to energy expenditure, and (ii) the hedonic circuitry, which is involved in rewarding and motivational aspects of energy consumption. In the homeostatic circuitry, serotonergic signaling contributes to the integration of metabolic signals that convey the body's energy status and facilitates the ability to suppress food intake when homeostatic needs have been met. In the hedonic circuitry, serotonergic signaling may reduce reward-related, motivational food consumption. In contrast, peripherally acting serotonin promotes energy absorption and storage. Disturbed serotonergic signaling is associated with obesity, emphasizing the importance to understand the role of serotonergic signaling in food intake. However, unraveling the serotonin-mediated regulation of food intake is complex, as the effects of serotonergic signaling in different brain regions depend on the regional expression of serotonin receptor subtypes and downstream effects via connections to other brain regions. We therefore provide an overview of the effects of serotonergic signaling in brain regions of the homeostatic and hedonic regulatory systems on food intake. Furthermore, we discuss the disturbances in serotonergic signaling in obesity and its potential therapeutic implications.
Collapse
Affiliation(s)
- Katy A van Galen
- Department of Endocrinology and Metabolism, Amsterdam University Medical Centers, Amsterdam, The Netherlands
| | - Kasper W Ter Horst
- Department of Endocrinology and Metabolism, Amsterdam University Medical Centers, Amsterdam, The Netherlands
| | - Mireille J Serlie
- Department of Endocrinology and Metabolism, Amsterdam University Medical Centers, Amsterdam, The Netherlands
| |
Collapse
|
5
|
Gómez-Martínez DG, Ramos M, del Valle-Padilla JL, Rosales JH, Robles F, Ramos F. A bioinspired model of short-term satiety of hunger influenced by food properties in virtual creatures. COGN SYST RES 2021. [DOI: 10.1016/j.cogsys.2020.10.008] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
|
6
|
Samodien E, Chellan N. Hypothalamic neurogenesis and its implications for obesity-induced anxiety disorders. Front Neuroendocrinol 2021; 60:100871. [PMID: 32976907 DOI: 10.1016/j.yfrne.2020.100871] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/27/2020] [Revised: 09/08/2020] [Accepted: 09/13/2020] [Indexed: 01/14/2023]
Abstract
Obesity and anxiety are public health problems that have no effective cure. Obesity-induced anxiety is also the most common behavioural trait exhibited amongst obese patients, with the mechanisms linking these disorders being poorly understood. The hypothalamus and hippocampus are reciprocally connected, important neurogenic brain regions that could be vital to understanding these disorders. Dietary, physical activity and lifestyle interventions have been shown to be able to enhance neurogenesis within the hippocampus, while the effects thereof within the hypothalamus is yet to be ascertained. This review describes hypothalamic neurogenesis, its impairment in obesity as well as the effect of interventional therapies. Obesity is characterized by a neurogenic shift towards neuropeptide Y neurons, promoting appetite and weight gain. While, nutraceuticals and exercise promote proopiomelanocortin neuron proliferation, causing diminished appetite and reduced weight gain. Through the furthered development of multimodal approaches targeting both these brain regions could hold an even greater therapeutic potential.
Collapse
Affiliation(s)
- Ebrahim Samodien
- Biomedical Research and Innovation Platform, South African Medical Research Council, Tygerberg, Cape Town, South Africa.
| | - Nireshni Chellan
- Biomedical Research and Innovation Platform, South African Medical Research Council, Tygerberg, Cape Town, South Africa; Department of Medical Physiology, Stellenbosch University, Tygerberg, Cape Town, South Africa
| |
Collapse
|
7
|
Blanco AM, Bertucci JI, Hatef A, Unniappan S. Feeding and food availability modulate brain-derived neurotrophic factor, an orexigen with metabolic roles in zebrafish. Sci Rep 2020; 10:10727. [PMID: 32612127 PMCID: PMC7329848 DOI: 10.1038/s41598-020-67535-z] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2020] [Accepted: 06/10/2020] [Indexed: 12/13/2022] Open
Abstract
Emerging findings point to a role for brain-derived neurotrophic factor (BDNF) on feeding in mammals. However, its role on energy balance is unclear. Moreover, whether BDNF regulates energy homeostasis in non-mammals remain unknown. This research aimed to determine whether BDNF is a metabolic peptide in zebrafish. Our results demonstrate that BDNF mRNAs and protein, as well as mRNAs encoding its receptors trkb2, p75ntra and p75ntrb, are detectable in the zebrafish brain, foregut and liver. Intraperitoneal injection of BDNF increased food intake at 1, 2 and 6 h post-administration, and caused an upregulation of brain npy, agrp and orexin, foregut ghrelin, and hepatic leptin mRNAs, and a reduction in brain nucb2. Fasting for 7 days increased bdnf and p75ntrb mRNAs in the foregut, while decreased bdnf, trkb2, p75ntra and p75ntrb mRNAs in the brain and liver. Additionally, the expression of bdnf and its receptors increased preprandially, and decreased after a meal in the foregut and liver. Finally, we observed BDNF-induced changes in the expression and/or activity of enzymes involved in glucose and lipid metabolism in the liver. Overall, present results indicate that BDNF is a novel regulator of appetite and metabolism in fish, which is modulated by energy intake and food availability.
Collapse
Affiliation(s)
- Ayelén Melisa Blanco
- Laboratory of Integrative Neuroendocrinology, Department of Veterinary Biomedical Sciences, Western College of Veterinary Medicine, University of Saskatchewan, 52 Campus Drive, Saskatoon, SK, S7N 5B4, Canada
- Laboratorio de Fisioloxía Animal, Departamento de Bioloxía Funcional e Ciencias da Saúde, Facultade de Bioloxía and Centro de Investigación Mariña, Universidade de Vigo, Vigo, Pontevedra, Spain
| | - Juan Ignacio Bertucci
- Laboratory of Integrative Neuroendocrinology, Department of Veterinary Biomedical Sciences, Western College of Veterinary Medicine, University of Saskatchewan, 52 Campus Drive, Saskatoon, SK, S7N 5B4, Canada
- Toxicology Centre, University of Saskatchewan, Saskatoon, SK, Canada
| | - Azadeh Hatef
- Laboratory of Integrative Neuroendocrinology, Department of Veterinary Biomedical Sciences, Western College of Veterinary Medicine, University of Saskatchewan, 52 Campus Drive, Saskatoon, SK, S7N 5B4, Canada
- Toxicology Centre, University of Saskatchewan, Saskatoon, SK, Canada
| | - Suraj Unniappan
- Laboratory of Integrative Neuroendocrinology, Department of Veterinary Biomedical Sciences, Western College of Veterinary Medicine, University of Saskatchewan, 52 Campus Drive, Saskatoon, SK, S7N 5B4, Canada.
| |
Collapse
|
8
|
Li N, Cao T, Wu X, Tang M, Xiang D, Cai H. Progress in Genetic Polymorphisms Related to Lipid Disturbances Induced by Atypical Antipsychotic Drugs. Front Pharmacol 2020; 10:1669. [PMID: 32116676 PMCID: PMC7011106 DOI: 10.3389/fphar.2019.01669] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2018] [Accepted: 12/20/2019] [Indexed: 12/11/2022] Open
Abstract
Metabolic side effects such as weight gain and disturbed lipid metabolism are often observed in the treatment of atypical antipsychotic drugs (AAPDs), which contribute to an excessive prevalence of metabolic syndrome among schizophrenic patients. Great individual differences are observed but the underlying mechanisms are still uncertain. Research on pharmacogenomics indicates that gene polymorphisms involved in the pathways controlling food intake and lipid metabolism may play a significant role. In this review, relevant genes (HTR2C, DRD2, LEP, NPY, MC4R, BDNF, MC4R, CNR1, INSIG2, ADRA2A) and genetic polymorphisms related to metabolic side effects of AAPDs especially dyslipidemia were summarized. Apart from clinical studies, in vitro and in vivo evidence is also analyzed to support related theories. The association of central and peripheral mechanisms is emphasized, enabling the possibility of using peripheral gene expression to predict the central status. Novel methodological development of pharmacogenomics is in urgent need, so as to provide references for individualized medication and further to shed some light on the mechanisms underlying AAPD-induced lipid disturbances.
Collapse
Affiliation(s)
- Nana Li
- Department of Pharmacy, The Second Xiangya Hospital of Central South University, Changsha, China.,Institute of Clinical Pharmacy, Central South University, Changsha, China
| | - Ting Cao
- Department of Pharmacy, The Second Xiangya Hospital of Central South University, Changsha, China.,Institute of Clinical Pharmacy, Central South University, Changsha, China
| | - Xiangxin Wu
- Department of Pharmacy, The Second Xiangya Hospital of Central South University, Changsha, China.,Institute of Clinical Pharmacy, Central South University, Changsha, China
| | - Mimi Tang
- Department of Pharmacy, Xiangya Hospital, Central South University, Changsha, China.,Institute of Hospital Pharmacy, Xiangya Hospital, Central South University, Changsha, China
| | - Daxiong Xiang
- Department of Pharmacy, The Second Xiangya Hospital of Central South University, Changsha, China.,Institute of Clinical Pharmacy, Central South University, Changsha, China
| | - Hualin Cai
- Department of Pharmacy, The Second Xiangya Hospital of Central South University, Changsha, China.,Institute of Clinical Pharmacy, Central South University, Changsha, China
| |
Collapse
|
9
|
Bae-Gartz I, Janoschek R, Breuer S, Schmitz L, Hoffmann T, Ferrari N, Branik L, Oberthuer A, Kloppe CS, Appel S, Vohlen C, Dötsch J, Hucklenbruch-Rother E. Maternal Obesity Alters Neurotrophin-Associated MAPK Signaling in the Hypothalamus of Male Mouse Offspring. Front Neurosci 2019; 13:962. [PMID: 31572115 PMCID: PMC6753176 DOI: 10.3389/fnins.2019.00962] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2019] [Accepted: 08/28/2019] [Indexed: 12/26/2022] Open
Abstract
Purpose Maternal obesity has emerged as an important risk factor for the development of metabolic disorders in the offspring. The hypothalamus as the center of energy homeostasis regulation is known to function based on complex neuronal networks that evolve during fetal and early postnatal development and maintain their plasticity into adulthood. Development of hypothalamic feeding networks and their functional plasticity can be modulated by various metabolic cues, especially in early stages of development. Here, we aimed at determining the underlying molecular mechanisms that contribute to disturbed hypothalamic network formation in offspring of obese mouse dams. Methods Female mice were fed either a control diet (CO) or a high-fat diet (HFD) after weaning until mating and during pregnancy and gestation. Male offspring was sacrificed at postnatal day (P) 21. The hypothalamus was subjected to gene array analysis, quantitative PCR and western blot analysis. Results P21 HFD offspring displayed increased body weight, circulating insulin levels, and strongly increased activation of the hypothalamic insulin signaling cascade with a concomitant increase in ionized calcium binding adapter molecule 1 (IBA1) expression. At the same time, the global gene expression profile in CO and HFD offspring differed significantly. More specifically, manifest influences on several key pathways of hypothalamic neurogenesis, axogenesis, and regulation of synaptic transmission and plasticity were detectable. Target gene expression analysis revealed significantly decreased mRNA expression of several neurotrophic factors and co-factors and their receptors, accompanied by decreased activation of their respective intracellular signal transduction. Conclusion Taken together, these results suggest a potential role for disturbed neurotrophin signaling and thus impaired neurogenesis, axogenesis, and synaptic plasticity in the pathogenesis of the offspring’s hypothalamic feeding network dysfunction due to maternal obesity.
Collapse
Affiliation(s)
- Inga Bae-Gartz
- Department of Pediatrics, University Hospital of Cologne, Cologne, Germany
| | - Ruth Janoschek
- Department of Pediatrics, University Hospital of Cologne, Cologne, Germany
| | - Saida Breuer
- Department of Pediatrics, University Hospital of Cologne, Cologne, Germany
| | - Lisa Schmitz
- Department of Pediatrics, University Hospital of Cologne, Cologne, Germany
| | - Thorben Hoffmann
- Department of Pediatrics, University Hospital of Cologne, Cologne, Germany
| | - Nina Ferrari
- Heart Center, Cologne Center for Prevention in Childhood and Youth, University Hospital of Cologne, Cologne, Germany
| | - Lena Branik
- Department of Pediatrics, University Hospital of Cologne, Cologne, Germany
| | - Andre Oberthuer
- Department of Pediatrics, University Hospital of Cologne, Cologne, Germany
| | - Cora-Sophia Kloppe
- Department of Pediatrics, University Hospital of Cologne, Cologne, Germany
| | - Sarah Appel
- Department of Pediatrics, University Hospital of Cologne, Cologne, Germany
| | - Christina Vohlen
- Department of Pediatrics, University Hospital of Cologne, Cologne, Germany
| | - Jörg Dötsch
- Department of Pediatrics, University Hospital of Cologne, Cologne, Germany
| | | |
Collapse
|
10
|
Hurley MM, Anderson EM, Chen C, Maunze B, Hess EM, Block ME, Patel N, Cooper Z, McCoy R, Dabra T, Conley W, Reilly MJ, Hearing M, Choi S. Acute Blockade of PACAP-Dependent Activity in the Ventromedial Nucleus of the Hypothalamus Disrupts Leptin-Induced Behavioral and Molecular Changes in Rats. Neuroendocrinology 2019; 110:271-281. [PMID: 31167202 PMCID: PMC6895395 DOI: 10.1159/000501337] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/09/2019] [Accepted: 06/05/2019] [Indexed: 12/18/2022]
Abstract
Leptin signaling pathways, stemming primarily from the hypothalamus, are necessary for maintaining normal energy homeostasis and body weight. In both rodents and humans, dysregulation of leptin signaling leads to morbid obesity and diabetes. Since leptin resistance is considered a primary factor underlying obesity, understanding the regulation of leptin signaling could lead to therapeutic tools and provide insights into the causality of obesity. While leptin actions in some hypothalamic regions such as the arcuate nuclei have been characterized, less is known about leptin activity in the hypothalamic ventromedial nuclei (VMN). Recently, pituitary adenylate cyclase-activating polypeptide (PACAP) has been shown to reduce feeding behavior and alter metabolism when administered into the VMN in a pattern similar to that of leptin. In the current study, we examined whether leptin and PACAP actions in the VMN share overlapping pathways in the regulation of energy balance. Interestingly, PACAP administration into the VMN increased STAT3 phosphorylation and SOCS3 mRNA expression, both of which are hallmarks of leptin receptor activation. In addition, BDNF mRNA expression in the VMN was increased by both leptin and PACAP administration. Moreover, antagonizing PACAP receptors fully reversed the behavioral and cellular effects of leptin injections into the VMN. Electrophysiological studies further illustrated that leptin-induced effects on VMN neurons were blocked by antagonizing PACAP receptors. We conclude that leptin dependency on PACAP signaling in the VMN suggests a potential common signaling cascade, allowing a tonically and systemically secreted neuropeptide to be more precisely regulated by central neuropeptides.
Collapse
Affiliation(s)
- Matthew M Hurley
- Department of Biomedical Sciences, Marquette University, Milwaukee, Wisconsin, USA
| | - Eden M Anderson
- Department of Biomedical Sciences, Marquette University, Milwaukee, Wisconsin, USA
| | - Christopher Chen
- Department of Biomedical Sciences, Marquette University, Milwaukee, Wisconsin, USA
| | - Brian Maunze
- Department of Biomedical Sciences, Marquette University, Milwaukee, Wisconsin, USA
| | - Evan M Hess
- Department of Biomedical Sciences, Marquette University, Milwaukee, Wisconsin, USA
| | - Megan E Block
- Department of Biomedical Sciences, Marquette University, Milwaukee, Wisconsin, USA
| | - Neerali Patel
- Department of Biomedical Sciences, Marquette University, Milwaukee, Wisconsin, USA
| | - Zane Cooper
- Department of Biomedical Sciences, Marquette University, Milwaukee, Wisconsin, USA
| | - Riley McCoy
- Department of Biomedical Sciences, Marquette University, Milwaukee, Wisconsin, USA
| | - Tanya Dabra
- Department of Biomedical Sciences, Marquette University, Milwaukee, Wisconsin, USA
| | - William Conley
- Department of Biomedical Sciences, Marquette University, Milwaukee, Wisconsin, USA
| | - Michael J Reilly
- Department of Biomedical Sciences, Marquette University, Milwaukee, Wisconsin, USA
| | - Matthew Hearing
- Department of Biomedical Sciences, Marquette University, Milwaukee, Wisconsin, USA
| | - SuJean Choi
- Department of Biomedical Sciences, Marquette University, Milwaukee, Wisconsin, USA,
| |
Collapse
|
11
|
Luo F, Mu Y, Gao C, Xiao Y, Zhou Q, Yang Y, Ni X, Shen WL, Yang J. Whole-brain patterns of the presynaptic inputs and axonal projections of BDNF neurons in the paraventricular nucleus. J Genet Genomics 2019; 46:31-40. [DOI: 10.1016/j.jgg.2018.11.004] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2018] [Revised: 11/21/2018] [Accepted: 11/25/2018] [Indexed: 12/22/2022]
|
12
|
McAllan L, Maynard KR, Kardian AS, Stayton AS, Fox SL, Stephenson EJ, Kinney CE, Alshibli NK, Gomes CK, Pierre JF, Puchowicz MA, Bridges D, Martinowich K, Han JC. Disruption of brain-derived neurotrophic factor production from individual promoters generates distinct body composition phenotypes in mice. Am J Physiol Endocrinol Metab 2018; 315:E1168-E1184. [PMID: 30253111 PMCID: PMC6336959 DOI: 10.1152/ajpendo.00205.2018] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
Brain-derived neurotrophic factor (BDNF) is a key neuropeptide in the central regulation of energy balance. The Bdnf gene contains nine promoters, each producing specific mRNA transcripts that encode a common protein. We sought to assess the phenotypic outcomes of disrupting BDNF production from individual Bdnf promoters. Mice with an intact coding region but selective disruption of BDNF production from Bdnf promoters I, II, IV, or VI (Bdnf-e1-/-, -e2-/-, -e4-/-, and -e6-/-) were created by inserting an enhanced green fluorescent protein-STOP cassette upstream of the targeted promoter splice donor site. Body composition was measured by MRI weekly from age 4 to 22 wk. Energy expenditure was measured by indirect calorimetry at 18 wk. Food intake was measured in Bdnf-e1-/- and Bdnf-e2-/- mice, and pair feeding was conducted. Weight gain, lean mass, fat mass, and percent fat of Bdnf-e1-/- and Bdnf-e2-/- mice (both sexes) were significantly increased compared with wild-type littermates. For Bdnf-e4-/- and Bdnf-e6-/- mice, obesity was not observed with either chow or high-fat diet. Food intake was increased in Bdnf-e1-/- and Bdnf-e2-/- mice, and pair feeding prevented obesity. Mutant and wild-type littermates for each strain (both sexes) had similar total energy expenditure after adjustment for body composition. These findings suggest that the obesity phenotype observed in Bdnf-e1-/- and Bdnf-e2-/- mice is attributable to hyperphagia and not altered energy expenditure. Our findings show that disruption of BDNF from specific promoters leads to distinct body composition effects, with disruption from promoters I or II, but not IV or VI, inducing obesity.
Collapse
Affiliation(s)
- Liam McAllan
- Department of Pediatrics, University of Tennessee Health Science Center , Memphis, Tennessee
- Children's Foundation Research Institute, Le Bonheur Children's Hospital , Memphis, Tennessee
| | - Kristen R Maynard
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, Maryland
| | - Alisha S Kardian
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, Maryland
| | - Amanda S Stayton
- Department of Pediatrics, University of Tennessee Health Science Center , Memphis, Tennessee
- Children's Foundation Research Institute, Le Bonheur Children's Hospital , Memphis, Tennessee
| | - Shelby L Fox
- Department of Pediatrics, University of Tennessee Health Science Center , Memphis, Tennessee
- Children's Foundation Research Institute, Le Bonheur Children's Hospital , Memphis, Tennessee
| | - Erin J Stephenson
- Department of Pediatrics, University of Tennessee Health Science Center , Memphis, Tennessee
- Children's Foundation Research Institute, Le Bonheur Children's Hospital , Memphis, Tennessee
| | - Clint E Kinney
- Department of Pediatrics, University of Tennessee Health Science Center , Memphis, Tennessee
- Children's Foundation Research Institute, Le Bonheur Children's Hospital , Memphis, Tennessee
| | - Noor K Alshibli
- Department of Pediatrics, University of Tennessee Health Science Center , Memphis, Tennessee
| | - Charles K Gomes
- Department of Pediatrics, University of Tennessee Health Science Center , Memphis, Tennessee
- Children's Foundation Research Institute, Le Bonheur Children's Hospital , Memphis, Tennessee
| | - Joseph F Pierre
- Department of Pediatrics, University of Tennessee Health Science Center , Memphis, Tennessee
- Children's Foundation Research Institute, Le Bonheur Children's Hospital , Memphis, Tennessee
| | - Michelle A Puchowicz
- Department of Pediatrics, University of Tennessee Health Science Center , Memphis, Tennessee
- Children's Foundation Research Institute, Le Bonheur Children's Hospital , Memphis, Tennessee
| | - Dave Bridges
- Department of Pediatrics, University of Tennessee Health Science Center , Memphis, Tennessee
- Children's Foundation Research Institute, Le Bonheur Children's Hospital , Memphis, Tennessee
- Department of Physiology, University of Tennessee Health Science Center , Memphis, Tennessee
| | - Keri Martinowich
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, Maryland
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine , Baltimore, Maryland
- Department of Neuroscience, Johns Hopkins University School of Medicine , Baltimore, Maryland
| | - Joan C Han
- Department of Pediatrics, University of Tennessee Health Science Center , Memphis, Tennessee
- Children's Foundation Research Institute, Le Bonheur Children's Hospital , Memphis, Tennessee
- Department of Physiology, University of Tennessee Health Science Center , Memphis, Tennessee
| |
Collapse
|
13
|
Felsted JA, Chien CH, Wang D, Panessiti M, Ameroso D, Greenberg A, Feng G, Kong D, Rios M. Alpha2delta-1 in SF1 + Neurons of the Ventromedial Hypothalamus Is an Essential Regulator of Glucose and Lipid Homeostasis. Cell Rep 2018; 21:2737-2747. [PMID: 29212022 DOI: 10.1016/j.celrep.2017.11.048] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2017] [Revised: 10/06/2017] [Accepted: 11/13/2017] [Indexed: 12/29/2022] Open
Abstract
The central mechanisms controlling glucose and lipid homeostasis are inadequately understood. We show that α2δ-1 is an essential regulator of glucose and lipid balance, acting in steroidogenic factor-1 (SF1) neurons of the ventromedial hypothalamus (VMH). These effects are body weight independent and involve regulation of SF1+ neuronal activity and sympathetic output to metabolic tissues. Accordingly, mice with α2δ-1 deletion in SF1 neurons exhibit glucose intolerance, altered lipolysis, and decreased cholesterol content in adipose tissue despite normal energy balance regulation. Profound reductions in the firing rate of SF1 neurons, decreased sympathetic output, and elevated circulating levels of serotonin are associated with these alterations. Normal calcium currents but reduced excitatory postsynaptic currents in mutant SF1 neurons implicate α2δ-1 in the promotion of excitatory synaptogenesis separate from its canonical role as a calcium channel subunit. Collectively, these findings identify an essential mechanism that regulates VMH neuronal activity and glycemic and lipid control and may be a target for tackling metabolic disease.
Collapse
Affiliation(s)
- Jennifer A Felsted
- Graduate Program in Biochemical and Molecular Nutrition, Friedman School of Nutrition Science and Policy, Tufts University, Boston, MA 02111, USA
| | - Cheng-Hao Chien
- Department of Neuroscience, Tufts University School of Medicine, Boston, MA 02111, USA
| | - Dongqing Wang
- McGovern Institute for Brain Research, Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Micaella Panessiti
- Graduate Program in Neuroscience, Sackler School of Graduate Biomedical Sciences, Tufts University School of Medicine, Boston, MA 02111, USA
| | - Dominique Ameroso
- Graduate Program in Neuroscience, Sackler School of Graduate Biomedical Sciences, Tufts University School of Medicine, Boston, MA 02111, USA
| | - Andrew Greenberg
- Graduate Program in Biochemical and Molecular Nutrition, Friedman School of Nutrition Science and Policy, Tufts University, Boston, MA 02111, USA; USDA Human Nutrition Research Center on Aging at Tufts University, Boston, MA 02111, USA
| | - Guoping Feng
- McGovern Institute for Brain Research, Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Dong Kong
- Department of Neuroscience, Tufts University School of Medicine, Boston, MA 02111, USA; Graduate Program in Neuroscience, Sackler School of Graduate Biomedical Sciences, Tufts University School of Medicine, Boston, MA 02111, USA
| | - Maribel Rios
- Department of Neuroscience, Tufts University School of Medicine, Boston, MA 02111, USA; Graduate Program in Neuroscience, Sackler School of Graduate Biomedical Sciences, Tufts University School of Medicine, Boston, MA 02111, USA.
| |
Collapse
|
14
|
BDNF/TrkB signaling mediates the anorectic action of estradiol in the nucleus tractus solitarius. Oncotarget 2017; 8:84028-84038. [PMID: 29137402 PMCID: PMC5663574 DOI: 10.18632/oncotarget.21062] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2017] [Accepted: 08/31/2017] [Indexed: 01/11/2023] Open
Abstract
Although compelling evidence indicates that estradiol (E2) acts in the nucleus tractus solitarius (NTS) to reduce food intake, the underlying mechanisms are largely unknown. We now report that estrogen's anorectic action occurs through enhancing the strength of brain-derived neurotrophic factor (BDNF) and tropomyosin receptor kinase (TrkB) signaling in the NTS. Intra-4th-ventricular administration of a low dose of BDNF reduced food intake to a greater extent in ovariectomized (OVX) rats cyclically treated with E2 than in vehicle-treated OVX rats, implying that cyclic E2 replacement increases BDNF's satiating potency. OVX significantly decreased bdnf gene expression in the NTS, and this was reversed by cyclic replacement of E2. Treatment of cultured primary neuronal cells from embryonic rat brainstem with E2 or PPT (ERα agonist), but not with DPN (ERβ agonist), significantly increased bdnf mRNA levels, indicating that ERα is the primary receptor mediating E2's stimulatory effect on bdnf gene expression. Administration of the selective TrkB antagonist, ANA-12, directly into the NTS significantly attenuated E2-induced reductions of food intake and body weight gain in OVX rats, indicating that TrkB receptor activation is necessary for E2's anorectic effect. Finally, relative to controls, OVX mice with bdnf gene knockdown specifically in the NTS had a blunted feeding response to E2. These data collectively imply that BDNF/TrkB receptor signaling in the NTS is a downstream mediator of E2 in the control of energy intake.
Collapse
|
15
|
Gilland KE, Fox EA. Effect of food deprivation or short-term Western diet feeding on BDNF protein expression in the hypothalamic arcuate, paraventricular, and ventromedial nuclei. Am J Physiol Regul Integr Comp Physiol 2017; 312:R611-R625. [PMID: 28202438 DOI: 10.1152/ajpregu.00256.2016] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2016] [Revised: 02/01/2017] [Accepted: 02/06/2017] [Indexed: 12/15/2022]
Abstract
Mutations in the brain-derived neurotrophic factor (BDNF) gene are associated with human obesity, and BDNF has potent inhibitory effects on eating and body weight. Little is known about the effects of energy balance manipulations on BDNF protein in the hypothalamus, though this brain region is critical for regulation of feeding and body weight and has high levels of BDNF. Here we investigated the effects of negative and positive energy status on BDNF protein levels in the arcuate (ARC), paraventricular, and ventromedial (VMH) hypothalamic nuclei and the ectorhinal cortex. To achieve this, mice were food deprived for 48 h or fed a Western diet (WD), a restricted amount of WD, or chow for 6 h, 48 h, 1 wk, or 3 wk. BDNF protein levels were estimated as the number of neurons in each brain region that exhibited BDNF-like immunoreactivity. Food deprivation decreased BDNF protein (and mRNA) expression in the ARC compared with fed mice (32%). In contrast, 1 wk of WD consumption increased BDNF protein expression in the VMH compared with chow or restricted WD feeding (40%) and, unexpectedly, increased BDNF protein in the ectorhinal cortex (20%). Furthermore, of the diet conditions and durations tested, only 1 wk of WD consumption was associated with both hyperphagia and excess weight, suggesting that effects of one or both contributed to the changes in BDNF levels. The decrease in ARC BDNF may support increased feeding in food-deprived mice, whereas the increase in the VMH may moderate overeating in WD-fed mice.
Collapse
Affiliation(s)
- Kaitlyn E Gilland
- Behavioral Neurogenetics Laboratory, Department of Psychological Sciences, Purdue University, West Lafayette, Indiana
| | - Edward A Fox
- Behavioral Neurogenetics Laboratory, Department of Psychological Sciences, Purdue University, West Lafayette, Indiana
| |
Collapse
|
16
|
Developmental changes in the hypothalamic mRNA expression levels of brain-derived neurotrophic factor and serum leptin levels: Their responses to fasting in male and female rats. Int J Dev Neurosci 2016; 54:1-5. [PMID: 27521083 DOI: 10.1016/j.ijdevneu.2016.08.001] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2016] [Revised: 08/04/2016] [Accepted: 08/08/2016] [Indexed: 01/19/2023] Open
Abstract
The actions and responses of hypothalamic appetite regulatory factors change markedly during the neonatal to pre-pubertal period in order to maintain appropriate metabolic and nutritional conditions. In this study, we examined the developmental changes in the hypothalamic mRNA levels of brain-derived neurotrophic factor (BDNF), which is a potent anorectic factor and the changes in the sensitivity of the hypothalamic expression of this factor to fasting during the neonatal to pre-pubertal period. Under fed conditions, hypothalamic BDNF mRNA expression decreased during development in both male and female rats. Similarly, the serum levels of leptin, which is a positive regulator of hypothalamic BDNF expression, also tended to fall during the developmental period. The serum leptin level and the hypothalamic BDNF mRNA level were found to be positively correlated in both sexes under the fed conditions. Hypothalamic BDNF mRNA expression was decreased by 24h fasting (separating the rats from their mothers) in the early neonatal period (postnatal day 10) in both males and females, but no such changes were seen at postnatal day 20. Twenty-four hours' fasting (food deprivation) did not affect hypothalamic BDNF mRNA expression in the pre-pubertal period (postnatal day 30). On the other hand, the rats' serum leptin levels were decreased by 24h fasting (separating the rats from their mothers at postnatal day 10 and 20, and food deprivation at postnatal day 30) throughout the early neonatal to pre-pubertal period. The correlation between serum leptin and hypothalamic BDNF mRNA levels was not significant under the fasted conditions. It can be speculated that leptin partially regulates hypothalamic BDNF mRNA levels, but only in fed conditions. Such changes in hypothalamic BDNF expression might play a role in maintaining appropriate metabolic and nutritional conditions and promoting normal physical development. In addition, because maternal separation induces a negative energy balance and short- and long-term stress responses, it is also possible that reductions in hypothalamic BDNF mRNA levels in the early neonatal period (postnatal day 10) may be partially induced by stress responses of the maternal deprivation.
Collapse
|
17
|
Abstract
Energy balance--that is, the relationship between energy intake and energy expenditure--is regulated by a complex interplay of hormones, brain circuits and peripheral tissues. Leptin is an adipocyte-derived cytokine that suppresses appetite and increases energy expenditure. Ironically, obese individuals have high levels of plasma leptin and are resistant to leptin treatment. Neurotrophic factors, particularly ciliary neurotrophic factor (CNTF) and brain-derived neurotrophic factor (BDNF), are also important for the control of body weight. CNTF can overcome leptin resistance in order to reduce body weight, although CNTF and leptin activate similar signalling cascades. Mutations in the gene encoding BDNF lead to insatiable appetite and severe obesity.
Collapse
Affiliation(s)
- Baoji Xu
- Department of Neuroscience, The Scripps Research Institute Florida, 130 Scripps Way, Jupiter, Florida 33458, USA
| | - Xiangyang Xie
- Department of Neuroscience, The Scripps Research Institute Florida, 130 Scripps Way, Jupiter, Florida 33458, USA
| |
Collapse
|
18
|
Yang H, An JJ, Sun C, Xu B. Regulation of Energy Balance via BDNF Expressed in Nonparaventricular Hypothalamic Neurons. Mol Endocrinol 2016; 30:494-503. [PMID: 27003443 DOI: 10.1210/me.2015-1329] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
Abstract
Brain-derived neurotrophic factor (BDNF) expressed in the paraventricular hypothalamus (PVH) has been shown to play a key role in regulating energy intake and energy expenditure. BDNF is also expressed in other hypothalamic nuclei; however, the role in the control of energy balance for BDNF produced in these structures remains largely unknown. We found that deleting the Bdnf gene in the ventromedial hypothalamus (VMH) during embryogenesis using the Sf1-Cre transgene had no effect on body weight in mice. In contrast, deleting the Bdnf gene in the adult VMH using Cre-expressing virus led to significant hyperphagia and obesity. These observations indicate that the lack of a hyperphagia phenotype in the Sf1-Cre/Bdnf mutant mice is likely due to developmental compensation. To investigate the role of BDNF expressed in other hypothalamic areas, we employed the hypothalamus-specific Nkx2.1-Cre transgene to delete the Bdnf gene. We found that the Nkx2.1-Cre transgene could abolish BDNF expression in many hypothalamic nuclei, but not in the PVH, and that the resulting mutant mice developed modest obesity due to reduced energy expenditure. Thus, BDNF produced in the VMH plays a role in regulating energy intake. Furthermore, BDNF expressed in hypothalamic areas other than PVH and VMH is also involved in the control of energy expenditure.
Collapse
Affiliation(s)
- Haili Yang
- College of Animal Science and Technology (H.Y., C.S.), Northwest A&F University, Yangling, Shaanxi 712100, China; and Department of Neuroscience (H.Y., J.J.A., B.X.), The Scripps Research Institute Florida, Jupiter, Florida 33458
| | - Juan Ji An
- College of Animal Science and Technology (H.Y., C.S.), Northwest A&F University, Yangling, Shaanxi 712100, China; and Department of Neuroscience (H.Y., J.J.A., B.X.), The Scripps Research Institute Florida, Jupiter, Florida 33458
| | - Chao Sun
- College of Animal Science and Technology (H.Y., C.S.), Northwest A&F University, Yangling, Shaanxi 712100, China; and Department of Neuroscience (H.Y., J.J.A., B.X.), The Scripps Research Institute Florida, Jupiter, Florida 33458
| | - Baoji Xu
- College of Animal Science and Technology (H.Y., C.S.), Northwest A&F University, Yangling, Shaanxi 712100, China; and Department of Neuroscience (H.Y., J.J.A., B.X.), The Scripps Research Institute Florida, Jupiter, Florida 33458
| |
Collapse
|
19
|
Teske JA, Perez-Leighton CE, Noble EE, Wang C, Billington CJ, Kotz CM. Effect of Housing Types on Growth, Feeding, Physical Activity, and Anxiety-Like Behavior in Male Sprague-Dawley Rats. Front Nutr 2016; 3:4. [PMID: 26870735 PMCID: PMC4740365 DOI: 10.3389/fnut.2016.00004] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2015] [Accepted: 01/15/2016] [Indexed: 01/15/2023] Open
Abstract
Background Animal welfare and accurate data collection are equally important in rodent research. Housing influences study outcomes and can challenge studies that monitor feeding, so housing choice needs to be evidence-based. The goal of these studies was to (1) compare established measures of well-being between rodents housed in wire grid-bottom floors with a resting platform compared to solid-bottom floors with bedding and (2) determine whether presence of a chewable device (Nylabone) affects orexin-A-induced hyperphagia. Methods Rodents were crossed over to the alternate housing twice after 2-week periods. Time required to complete food intake measurements was recorded as an indicator of feasibility. Food intake stimulated by orexin-A was compared with and without the Nylabone. Blood corticosterone and hypothalamic BDNF were assessed. Results Housing had no effect on growth, energy expenditure, corticosterone, hypothalamic BDNF, behavior, and anxiety measures. Food intake was disrupted after housing cross-over. Time required to complete food intake measurements was significantly higher for solid-bottom bedded cages. The Nylabone had no effect on orexin-A-stimulated feeding. Conclusion Well-being is not significantly different between rodents housed on grid-bottom floors and those in solid-bottom-bedded cages based on overall growth and feeding but alternating between housing confounds measures of feeding.
Collapse
Affiliation(s)
- Jennifer A Teske
- Department of Nutritional Sciences, University of Arizona, Tucson, AZ, USA; Minneapolis VA Health Care System, Minneapolis, MN, USA; Minnesota Obesity Center, University of Minnesota, Saint Paul, MN, USA; Department of Food Science and Nutrition, University of Minnesota, Saint Paul, MN, USA
| | - Claudio Esteban Perez-Leighton
- Department of Food Science and Nutrition, University of Minnesota, Saint Paul, MN, USA; Center for Integrative Medicine and Innovative Science, Universidad Andres Bello, Santiago, Chile
| | - Emily E Noble
- Department of Integrative Biology and Physiology, University of California Los Angeles , Los Angeles, CA , USA
| | - Chuanfeng Wang
- Minneapolis VA Health Care System, Minneapolis, MN, USA; Minnesota Obesity Center, University of Minnesota, Saint Paul, MN, USA; Department of Food Science and Nutrition, University of Minnesota, Saint Paul, MN, USA
| | - Charles J Billington
- Minneapolis VA Health Care System, Minneapolis, MN, USA; Minnesota Obesity Center, University of Minnesota, Saint Paul, MN, USA; Department of Food Science and Nutrition, University of Minnesota, Saint Paul, MN, USA; Department of Medicine, University of Minnesota, Minneapolis, MN, USA
| | - Catherine M Kotz
- Minneapolis VA Health Care System, Minneapolis, MN, USA; Minnesota Obesity Center, University of Minnesota, Saint Paul, MN, USA; Department of Food Science and Nutrition, University of Minnesota, Saint Paul, MN, USA; Geriatric Research Education and Clinical Center, Minneapolis VA Health Care System, Minneapolis, MN, USA
| |
Collapse
|
20
|
Han J. Rare Syndromes and Common Variants of the Brain-Derived Neurotrophic Factor Gene in Human Obesity. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2016; 140:75-95. [DOI: 10.1016/bs.pmbts.2015.12.002] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
|
21
|
Ozek C, Zimmer DJ, De Jonghe BC, Kalb RG, Bence KK. Ablation of intact hypothalamic and/or hindbrain TrkB signaling leads to perturbations in energy balance. Mol Metab 2015; 4:867-80. [PMID: 26629410 PMCID: PMC4632115 DOI: 10.1016/j.molmet.2015.08.002] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/29/2015] [Revised: 08/10/2015] [Accepted: 08/11/2015] [Indexed: 12/16/2022] Open
Abstract
OBJECTIVE Brain-derived neurotrophic factor (BDNF) and its receptor, tropomyosin receptor kinase B (TrkB), play a paramount role in the central regulation of energy balance. Despite the substantial body of genetic evidence implicating BDNF- or TrkB-deficiency in human obesity, the critical brain region(s) contributing to the endogenous role of BDNF/TrkB signaling in metabolic control remain unknown. METHODS We assessed the importance of intact hypothalamic or hindbrain TrkB signaling in central regulation of energy balance by generating Nkx2.1-Ntrk2-/- and Phox2b-Ntrk2+/- mice, respectively, and comparing metabolic parameters (body weight, adiposity, food intake, energy expenditure and glucose homeostasis) under high-fat diet or chow fed conditions. RESULTS Our data show that when fed a high-fat diet, male and female Nkx2.1-Ntrk2-/- mice have significantly increased body weight and adiposity that is likely driven by reduced locomotor activity and core body temperature. When maintained on a chow diet, female Nkx2.1-Ntrk2-/- mice exhibit an increased body weight and adiposity phenotype more robust than in males, which is accompanied by hyperphagia that precedes the onset of a body weight difference. In addition, under both diet conditions, Nkx2.1-Ntrk2-/- mice show increased blood glucose, serum insulin and leptin levels. Mice with complete hindbrain TrkB-deficiency (Phox2b-Ntrk2-/-) are perinatal lethal, potentially indicating a vital role for TrkB in visceral motor neurons that control cardiovascular, respiratory, and digestive functions during development. Phox2b-Ntrk2+/- heterozygous mice are similar in body weight, adiposity and glucose homeostasis parameters compared to wild type littermate controls when maintained on a high-fat or chow diet. Interestingly, despite the absence of a body weight difference, Phox2b-Ntrk2+/- heterozygous mice exhibit pronounced hyperphagia. CONCLUSION Taken together, our findings suggest that the hypothalamus is a key brain region involved in endogenous BDNF/TrkB signaling and central metabolic control and that endogenous hindbrain TrkB likely plays a role in modulating food intake and survival of mice. Our findings also show that female mice lacking TrkB in the hypothalamus have a more robust metabolic phenotype.
Collapse
Key Words
- Agrp, agouti-related peptide
- BAT, brown adipose tissue
- BDNF
- BDNF, brain-derived neurotrophic factor
- Cidea, cell death-inducing DFFA-like effector a
- Cre, Cre recombinase
- DVC, dorsal vagal complex
- Elovl3, elongation of very long fatty acids-like 3
- GTT, glucose tolerance test
- HFD, high-fat diet
- HPA axis, hypothalamic-pituitary-adrenal axis
- Hindbrain
- Hypothalamus
- LepR, leptin receptor
- Mc4R, melanocortin 4 receptor
- NTS, nucleus of the solitary tract
- Nkx2.1, Nk2 homeobox 1 protein
- Npy, neuropeptide Y
- Obesity
- PVH, paraventricular nucleus of the hypothalamus
- Pgc1α, peroxisome proliferator-activated receptor gamma coactivator 1 alpha
- Phox2b, paired-like homeobox 2b protein
- Pomc, pro-opiomelanocortin
- Pparγ, peroxisome proliferator-activated receptor gamma
- Prdm16, PR domain containing 16
- TrkB
- TrkB, tropomyosin receptor kinase B
- Ucp1, uncoupling protein 1
- VMH, ventromedial nucleus of the hypothalamus
- eWAT, epididymal white adipose tissue
Collapse
Affiliation(s)
- Ceren Ozek
- Department of Biomedical Sciences, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Derek J Zimmer
- Department of Biomedical Sciences, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Bart C De Jonghe
- Department of Biobehavioral Health Sciences, School of Nursing, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Robert G Kalb
- Department of Neurology, Children's Hospital of Philadelphia, Philadelphia, PA, 19104, USA
| | - Kendra K Bence
- Department of Biomedical Sciences, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| |
Collapse
|
22
|
Notaras M, Hill R, van den Buuse M. The BDNF gene Val66Met polymorphism as a modifier of psychiatric disorder susceptibility: progress and controversy. Mol Psychiatry 2015; 20:916-30. [PMID: 25824305 DOI: 10.1038/mp.2015.27] [Citation(s) in RCA: 187] [Impact Index Per Article: 20.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/16/2014] [Revised: 12/22/2014] [Accepted: 02/09/2015] [Indexed: 02/06/2023]
Abstract
Brain-derived neurotrophic factor (BDNF) has a primary role in neuronal development, differentiation and plasticity in both the developing and adult brain. A single-nucleotide polymorphism in the proregion of BDNF, termed the Val66Met polymorphism, results in deficient subcellular translocation and activity-dependent secretion of BDNF, and has been associated with impaired neurocognitive function in healthy adults and in the incidence and clinical features of several psychiatric disorders. Research investigating the Val66Met polymorphism has increased markedly in the past decade, and a gap in integration exists between and within academic subfields interested in the effects of this variant. Here we comprehensively review the role and relevance of the Val66Met polymorphism in psychiatric disorders, with emphasis on suicidal behavior and anxiety, eating, mood and psychotic disorders. The cognitive and molecular neuroscience of the Val66Met polymorphism is also concisely reviewed to illustrate the effects of this genetic variant in healthy controls, and is complemented by a commentary on the behavioral neuroscience of BDNF and the Val66Met polymorphism where relevant to specific disorders. Lastly, a number of controversies and unresolved issues, including small effect sizes, sampling of allele inheritance but not genotype and putative ethnicity-specific effects of the Val66Met polymorphism, are also discussed to direct future research.
Collapse
Affiliation(s)
- M Notaras
- Florey Institute of Neuroscience and Mental Health, Melbourne, VIC, Australia
| | - R Hill
- Florey Institute of Neuroscience and Mental Health, Melbourne, VIC, Australia
| | - M van den Buuse
- 1] Florey Institute of Neuroscience and Mental Health, Melbourne, VIC, Australia [2] School of Psychological Science, La Trobe University, Melbourne, VIC, Australia
| |
Collapse
|
23
|
Jin YJ, Cao PJ, Bian WH, Li ME, Zhou R, Zhang LY, Yang MZ. BDNF levels in adipose tissue and hypothalamus were reduced in mice with MSG-induced obesity. Nutr Neurosci 2015; 18:376-82. [DOI: 10.1179/1476830515y.0000000039] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
|
24
|
Rossetti C, Halfon O, Boutrel B. Controversies about a common etiology for eating and mood disorders. Front Psychol 2014; 5:1205. [PMID: 25386150 PMCID: PMC4209809 DOI: 10.3389/fpsyg.2014.01205] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2014] [Accepted: 10/06/2014] [Indexed: 12/25/2022] Open
Abstract
Obesity and depression represent a growing health concern worldwide. For many years, basic science and medicine have considered obesity as a metabolic illness, while depression was classified a psychiatric disorder. Despite accumulating evidence suggesting that obesity and depression may share commonalities, the causal link between eating and mood disorders remains to be fully understood. This etiology is highly complex, consisting of multiple environmental and genetic risk factors that interact with each other. In this review, we sought to summarize the preclinical and clinical evidence supporting a common etiology for eating and mood disorders, with a particular emphasis on signaling pathways involved in the maintenance of energy balance and mood stability, among which orexigenic and anorexigenic neuropeptides, metabolic factors, stress responsive hormones, cytokines, and neurotrophic factors.
Collapse
Affiliation(s)
- Clara Rossetti
- Center for Psychiatric Neuroscience, Lausanne University Hospital Lausanne, Switzerland
| | - Olivier Halfon
- Division of Child and Adolescent Psychiatry, Department of Psychiatry, Lausanne University Hospital Lausanne, Switzerland
| | - Benjamin Boutrel
- Center for Psychiatric Neuroscience, Lausanne University Hospital Lausanne, Switzerland ; Division of Child and Adolescent Psychiatry, Department of Psychiatry, Lausanne University Hospital Lausanne, Switzerland
| |
Collapse
|
25
|
Ozek C, Kanoski SE, Zhang ZY, Grill HJ, Bence KK. Protein-tyrosine phosphatase 1B (PTP1B) is a novel regulator of central brain-derived neurotrophic factor and tropomyosin receptor kinase B (TrkB) signaling. J Biol Chem 2014; 289:31682-31692. [PMID: 25288805 DOI: 10.1074/jbc.m114.603621] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023] Open
Abstract
Neuronal protein-tyrosine phosphatase 1B (PTP1B) deficiency in mice results in enhanced leptin signaling and protection from diet-induced obesity; however, whether additional signaling pathways in the brain contribute to the metabolic effects of PTP1B deficiency remains unclear. Here, we show that the tropomyosin receptor kinase B (TrkB) receptor is a direct PTP1B substrate and implicate PTP1B in the regulation of the central brain-derived neurotrophic factor (BDNF) signaling. PTP1B interacts with activated TrkB receptor in mouse brain and human SH-SY5Y neuroblastoma cells. PTP1B overexpression reduces TrkB phosphorylation and activation of downstream signaling pathways, whereas PTP1B inhibition augments TrkB signaling. Notably, brains of Ptpn1(-/-) mice exhibit enhanced TrkB phosphorylation, and Ptpn1(-/-) mice are hypersensitive to central BDNF-induced increase in core temperature. Taken together, our findings demonstrate that PTP1B is a novel physiological regulator of TrkB and that enhanced BDNF/TrkB signaling may contribute to the beneficial metabolic effects of PTP1B deficiency.
Collapse
Affiliation(s)
- Ceren Ozek
- Department of Animal Biology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104
| | - Scott E Kanoski
- Department of Biological Sciences, Human and Evolutionary Biology Section, University of Southern California, Los Angeles, California 90089, and
| | - Zhong-Yin Zhang
- Department of Biochemistry and Molecular Biology, School of Medicine, Indiana University, Indianapolis, Indiana 46202
| | - Harvey J Grill
- Department of Psychology, School of Arts and Sciences, University of Pennsylvania, Philadelphia, Pennsylvania 19104
| | - Kendra K Bence
- Department of Animal Biology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104,.
| |
Collapse
|
26
|
Noble EE, Billington CJ, Kotz CM, Wang C. Oxytocin in the ventromedial hypothalamic nucleus reduces feeding and acutely increases energy expenditure. Am J Physiol Regul Integr Comp Physiol 2014; 307:R737-45. [PMID: 24990860 DOI: 10.1152/ajpregu.00118.2014] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Central oxytocin reduces food intake and increases energy expenditure. The ventromedial hypothalamic nucleus (VMN) is associated with energy balance and contains a high density of oxytocin receptors. We hypothesized that oxytocin in the VMN is a negative regulator of energy balance acting to reduce feeding and increase energy expenditure. To test this idea, oxytocin or vehicle was injected directly into the VMN of Sprague-Dawley rats during fasted and nonfasted conditions. Energy expenditure (via indirect calorimetry) and spontaneous physical activity (SPA) were recorded simultaneously. Animals were also exposed to a conditioned taste aversion test, to determine whether oxytocin's effects on food intake were associated with malaise. When food was available during testing, oxytocin-induced elevations in energy expenditure lasted for 1 h, after which overall energy expenditure was reduced. In the absence of food during the testing period, oxytocin similarly increased energy expenditure during the first hour, but differences in 12-h energy expenditure were eliminated, implying that the differences may have been due to the thermic effects of feeding (digestion, absorption, and metabolic processing). Oxytocin acutely elevated SPA and reduced feeding at doses that did not cause a conditioned taste aversion during both the fed and fasted states. Together, these data suggest that oxytocin in the VMN promotes satiety and acutely elevates energy expenditure and SPA and implicates the VMN as a relevant site for the antiobesity effects of oxytocin.
Collapse
Affiliation(s)
- Emily E Noble
- Department of Food Science and Nutrition, University of Minnesota, Saint Paul, Minnesota; Minnesota Obesity Center, University of Minnesota, Saint Paul, Minnesota; and
| | - Charles J Billington
- Department of Food Science and Nutrition, University of Minnesota, Saint Paul, Minnesota; Department of Medicine, University of Minnesota, Saint Paul, Minnesota; Minnesota Obesity Center, University of Minnesota, Saint Paul, Minnesota; and Minneapolis VA Health Care System, Minneapolis, Minnesota
| | - Catherine M Kotz
- Department of Food Science and Nutrition, University of Minnesota, Saint Paul, Minnesota; Department of Neuroscience, University of Minnesota, Saint Paul, Minnesota; Minnesota Obesity Center, University of Minnesota, Saint Paul, Minnesota; and Minneapolis VA Health Care System, Minneapolis, Minnesota
| | - ChuanFeng Wang
- Department of Food Science and Nutrition, University of Minnesota, Saint Paul, Minnesota; Minnesota Obesity Center, University of Minnesota, Saint Paul, Minnesota; and Minneapolis VA Health Care System, Minneapolis, Minnesota
| |
Collapse
|
27
|
Liu X, Zhu Z, Kalyani M, Janik JM, Shi H. Effects of energy status and diet on Bdnf expression in the ventromedial hypothalamus of male and female rats. Physiol Behav 2014; 130:99-107. [PMID: 24709620 DOI: 10.1016/j.physbeh.2014.03.028] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2013] [Revised: 03/04/2014] [Accepted: 03/26/2014] [Indexed: 11/29/2022]
Abstract
Sex differences exist in the regulation of energy homeostasis in response to calorie scarcity or excess. Brain-derived neurotrophic factor (BDNF) is one of the anorexigenic neuropeptides regulating energy homeostasis. Expression of Bdnf mRNA in the ventromedial nucleus of the hypothalamus (VMH) is closely associated with energy and reproductive status. We hypothesized that Bdnf expression in the VMH was differentially regulated by altered energy balance in male and female rats. Using dietary intervention, including fasting-induced negative energy status and high-fat diet (HFD) feeding-induced positive energy status, along with low-fat diet (LFD) feeding and HFD pair-feeding (HFD-PF), effects of diets and changes in energy status on VMH Bdnf expression were compared between male and female rats. Fasted males but not females had lower VMH Bdnf expression than their fed counterparts following 24-hour fasting, suggesting that fasted males reduced Bdnf expression to drive hyperphagia and body weight gain. Male HFD obese and HFD-PF non-obese rats had similarly reduced expression of Bdnf compared with LFD males, indicating that dampened Bdnf expression was associated with feeding a diet high in fat instead of increased adiposity. Decreased BDNF signaling during HFD feeding would increase a drive to eat and may contribute to diet-induced obesity in males. In contrast, VMH Bdnf expression was stably maintained in females when energy homeostasis was disturbed. These results suggest sex-distinct regulation of central Bdnf expression by diet and energy status.
Collapse
Affiliation(s)
- Xian Liu
- Cell, Molecular, and Structural Biology, Miami University, OH, United States
| | - Zheng Zhu
- Physiology and Neuroscience, Department of Biology, Miami University, OH, United States; Department of Statistics, Miami University, OH, United States
| | - Manu Kalyani
- Physiology and Neuroscience, Department of Biology, Miami University, OH, United States
| | - James M Janik
- Cell, Molecular, and Structural Biology, Miami University, OH, United States; Physiology and Neuroscience, Department of Biology, Miami University, OH, United States
| | - Haifei Shi
- Cell, Molecular, and Structural Biology, Miami University, OH, United States; Physiology and Neuroscience, Department of Biology, Miami University, OH, United States.
| |
Collapse
|
28
|
Hypothalamic dysfunction of the thrombospondin receptor α2δ-1 underlies the overeating and obesity triggered by brain-derived neurotrophic factor deficiency. J Neurosci 2014; 34:554-65. [PMID: 24403154 DOI: 10.1523/jneurosci.1572-13.2014] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
Brain-derived neurotrophic factor (BDNF) and its receptor, TrkB, are critical components of the neural circuitry controlling appetite and body weight. Diminished BDNF signaling in mice results in severe hyperphagia and obesity. In humans, BDNF haploinsufficiency and the functional Bdnf Val66Met polymorphism have been linked to elevated food intake and body weight. The mechanisms underlying this dysfunction are poorly defined. We demonstrate a chief role of α2δ-1, a calcium channel subunit and thrombospondin receptor, in triggering overeating in mice with central BDNF depletion. We show reduced α2δ-1 cell-surface expression in the BDNF mutant ventromedial hypothalamus (VMH), an energy balance-regulating center. This deficit contributes to the hyperphagia exhibited by BDNF mutant mice because selective inhibition of α2δ-1 by gabapentin infusion into wild-type VMH significantly increases feeding and body weight gain. Importantly, viral-mediated α2δ-1 rescue in BDNF mutant VMH significantly mitigates their hyperphagia, obesity, and liver steatosis and normalizes deficits in glucose homeostasis. Whole-cell recordings in BDNF mutant VMH neurons revealed normal calcium currents but reduced frequency of EPSCs. These results suggest calcium channel-independent effects of α2δ-1 on feeding and implicate α2δ-1-thrombospondin interactions known to facilitate excitatory synapse assembly. Our findings identify a central mechanism mediating the inhibitory effects of BDNF on feeding. They also demonstrate a novel and critical role for α2δ-1 in appetite control and suggest a mechanism underlying weight gain in humans treated with gabapentinoid drugs.
Collapse
|
29
|
Abstract
Complex interactions between the brain and peripheral tissues mediate the effective control of energy balance and body weight. Hypothalamic and hindbrain neural circuits integrate peripheral signals informing the nutritional status of the animal and in response regulate nutrient intake and energy utilization. Obesity and its many medical complications emerge from the dysregulation of energy homeostasis. Excessive weight gain might also arise from alterations in reward systems of the brain that drive consumption of calorie dense, palatable foods in the absence of an energy requirement. Several neurotrophins, most notably brain-derived neurotrophic factor, have been implicated in the molecular and cellular processes underlying body weight regulation. Here, we review investigations interrogating their roles in energy balance and reward centers of the brain impacting feeding behavior and energy expenditure.
Collapse
Affiliation(s)
- M Rios
- Department of Neuroscience, Tufts University School of Medicine, Boston, MA, 02111, USA,
| |
Collapse
|
30
|
Verge VMK, Andreassen CS, Arnason TG, Andersen H. Mechanisms of disease: role of neurotrophins in diabetes and diabetic neuropathy. HANDBOOK OF CLINICAL NEUROLOGY 2014; 126:443-60. [PMID: 25410238 DOI: 10.1016/b978-0-444-53480-4.00032-1] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Neuropathy is an insidious and devastating consequence of diabetes. Early studies provided a strong rationale for deficient neurotrophin support in the pathogenesis of diabetic neuropathy in a number of critical tissues and organs. It has now been over a decade since the first failed human neurotrophin supplementation clinical trials, but mounting evidence still implicates these trophic factors in diabetic neuropathy. Since then, tremendous advances have been made in our understanding of the complexities of neurotrophin signaling and processing and how the diabetic milieu might impact this. This in turn changes both our perception of how the altered trophic environment contributes to the etiology of diabetic neuropathy and the design of future neurotrophin therapeutic interventions. This chapter summarizes some of these findings and attempts to integrate neurotrophin actions on the nervous system with an increasing appreciation of their role in the regulation of metabolic processes in diabetes that impact the diabetic neuropathic state.
Collapse
Affiliation(s)
- Valerie M K Verge
- Department of Anatomy and Cell Biology, University of Saskatchewan, Saskatoon, Canada; Cameco MS Neuroscience Research Center, University of Saskatchewan, Saskatoon City Hospital, Saskatoon, Canada.
| | - Christer S Andreassen
- Department of Otorhinolaryngology and Head and Neck Surgery, Aarhus University Hospital, Aarhus, Denmark
| | - Terra G Arnason
- Department of Anatomy and Cell Biology, University of Saskatchewan, Saskatoon, Canada; Department of Medicine, Division of Endocrinology and Metabolism, University of Saskatchewan, Saskatoon, Canada
| | - Henning Andersen
- Department of Neurology, Aarhus University Hospital, Aarhus, Denmark
| |
Collapse
|
31
|
Abstract
Body weight is determined by a balance between food intake and energy expenditure. Multiple neural circuits in the brain have evolved to process information about food, food-related cues and food consumption to control feeding behavior. Numerous gastrointestinal endocrine cells produce and secrete satiety hormones in response to food consumption and digestion. These hormones suppress hunger and promote satiation and satiety mainly through hindbrain circuits, thus governing meal-by-meal eating behavior. In contrast, the hypothalamus integrates adiposity signals to regulate long-term energy balance and body weight. Distinct hypothalamic areas and various orexigenic and anorexigenic neurons have been identified to homeostatically regulate food intake. The hypothalamic circuits regulate food intake in part by modulating the sensitivity of the hindbrain to short-term satiety hormones. The hedonic and incentive properties of foods and food-related cues are processed by the corticolimbic reward circuits. The mesolimbic dopamine system encodes subjective "liking" and "wanting" of palatable foods, which is subjected to modulation by the hindbrain and the hypothalamic homeostatic circuits and by satiety and adiposity hormones. Satiety and adiposity hormones also promote energy expenditure by stimulating brown adipose tissue (BAT) activity. They stimulate BAT thermogenesis mainly by increasing the sympathetic outflow to BAT. Many defects in satiety and/or adiposity hormone signaling and in the hindbrain and the hypothalamic circuits have been described and are believed to contribute to the pathogenesis of energy imbalance and obesity.
Collapse
Affiliation(s)
- Liangyou Rui
- Department of Molecular & Integrative Physiology, University of Michigan Medical School, Ann Arbor, MI, 48109-0622, USA,
| |
Collapse
|
32
|
Abstract
During critical periods of development early in life, excessive or scarce nutritional environments can disrupt the development of central feeding and metabolic neural circuitry, leading to obesity and metabolic disorders in adulthood. A better understanding of the genetic networks that control the development of feeding and metabolic neural circuits, along with knowledge of how and where dietary signals disrupt this process, can serve as the basis for future therapies aimed at reversing the public health crisis that is now building as a result of the global obesity epidemic. This review of animal and human studies highlights recent insights into the molecular mechanisms that regulate the development of central feeding circuitries, the mechanisms by which gestational and early postnatal nutritional status affects this process, and approaches aimed at counteracting the deleterious effects of early over- and underfeeding.
Collapse
Affiliation(s)
- Daniel A Lee
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, California 91125
| | | |
Collapse
|
33
|
Sun B, Liang NC, Ewald ER, Purcell RH, Boersma GJ, Yan J, Moran TH, Tamashiro KLK. Early postweaning exercise improves central leptin sensitivity in offspring of rat dams fed high-fat diet during pregnancy and lactation. Am J Physiol Regul Integr Comp Physiol 2013; 305:R1076-84. [PMID: 24026073 PMCID: PMC3840316 DOI: 10.1152/ajpregu.00566.2012] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2012] [Accepted: 09/04/2013] [Indexed: 12/16/2022]
Abstract
Maternal high-fat (HF) diet has long-term consequences on the metabolic phenotype of the offspring. Here, we determined the effects of postweaning exercise in offspring of rat dams fed HF diet during gestation and lactation. Pregnant Sprague-Dawley rats were maintained on chow or HF diet throughout gestation and lactation. All pups were weaned onto chow diet on postnatal day (PND) 21. At 4 wk of age, male pups were given free access to running wheels (RW) or remained sedentary (SED) for 3 wk, after which all rats remained sedentary, resulting in four groups: CHOW-SED, CHOW-RW, HF-SED, and HF-RW. Male HF offspring gained more body weight by PND7 compared with CHOW pups and maintained this weight difference through the entire experiment. Three weeks of postweaning exercise did not affect body weight gain in either CHOW or HF offspring, but reduced adiposity in HF offspring. Plasma leptin was decreased at the end of the 3-wk running period in HF-RW rats but was not different from HF-SED 9 wk after the exercise period ended. At 14 wk of age, intracerebroventricular injection of leptin suppressed food intake in CHOW-SED, CHOW-RW, and HF-RW, while it did not affect food intake in HF-SED group. At death, HF-RW rats also had higher leptin-induced phospho-STAT3 level in the arcuate nucleus than HF-SED rats. Both maternal HF diet and postweaning exercise had effects on hypothalamic neuropeptide and receptor mRNA expression in adult offspring. Our data suggest that postweaning exercise improves central leptin sensitivity and signaling in this model.
Collapse
Affiliation(s)
- Bo Sun
- Department of Physiology and Pathophysiology, Xi'an Jiaotong University School of Medicine, Xi'an, Shaanxi, People's Republic of China; and
| | | | | | | | | | | | | | | |
Collapse
|
34
|
Individual differences in novelty seeking predict subsequent vulnerability to social defeat through a differential epigenetic regulation of brain-derived neurotrophic factor expression. J Neurosci 2013; 33:11048-60. [PMID: 23825410 DOI: 10.1523/jneurosci.0199-13.2013] [Citation(s) in RCA: 74] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Some personality traits, including novelty seeking, are good predictors of vulnerability to stress-related mood disorders in both humans and rodents. While high-novelty-seeking rats [high responders (HRs)] are vulnerable to the induction of depressive-like symptoms by social defeat stress, low-novelty-seeking rats [low responders (LRs)] are not. Here, we show that such individual differences are critically regulated by hippocampal BDNF. While LR animals exhibited an increase in BDNF levels following social defeat, HR individuals did not. This difference in hippocampal BDNF expression promoted the vulnerability of HR and the resilience of LR rats. Indeed, preventing activation of BDNF signaling by infusing the BDNF scavenger TrkB-Fc into the dentate gyrus of the hippocampus of LR rats led to social defeat-induced social avoidance, whereas its activation in HR rats by the TrkB agonist 7,8-dihydroxyflavone promoted social approach. Along with the changes in BDNF expression following defeat, we report in LR animals a downregulation of the inactive BDNF receptor TrkB.T1, associated with an activation of CREB through Akt-mediated signaling, but not MSK1-mediated signaling. In HR animals, none of these molecules were affected by social defeat. Importantly, the BDNF upregulation involved an epigenetically controlled transcription of bdnf exon VI, associated with a coherent regulation of relevant epigenetic factors. Altogether, our data support the importance of hippocampal BDNF regulation in response to stressful events. Moreover, we identify a specific and adaptive regulation of bdnf exon VI in the hippocampus as a critical regulator of stress resilience, and strengthen the importance of epigenetic factors in mediating stress-induced adaptive and maladaptive responses in different individuals.
Collapse
|
35
|
Liao GY, Li Y, Xu B. Ablation of TrkB expression in RGS9-2 cells leads to hyperphagic obesity. Mol Metab 2013; 2:491-7. [PMID: 24327964 DOI: 10.1016/j.molmet.2013.08.002] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/24/2013] [Revised: 07/31/2013] [Accepted: 08/03/2013] [Indexed: 01/05/2023] Open
Abstract
Brain-derived neurotrophic factor (BDNF) and its cognate receptor, TrkB (tropomyosin receptor kinase B), are widely expressed in the brain where they regulate a wide variety of biological processes, including energy homeostasis. However, the specific population(s) of TrkB-expressing neurons through which BDNF governs energy homeostasis remain(s) to be determined. Using the Cre-loxP recombination system, we deleted the mouse TrkB gene in RGS9-2-expressing cells. In this mouse mutant, TrkB expression was abolished in several hypothalamic nuclei, including arcuate nucleus, dorsomedial hypothalamus, and lateral hypothalamus. TrkB expression was also abolished in a small number of cells in other brain regions, including the cerebral cortex and striatum. The mutant animals developed hyperphagic obesity with normal energy expenditure. Despite hyperglycemia under fed conditions, these animals exhibited normal fasting blood glucose levels and normal glucose tolerance. These results suggest that BDNF regulates energy homeostasis in part through TrkB-expressing neurons in the hypothalamus.
Collapse
Key Words
- 3V, third ventricle
- ARC, arcuate nucleus
- BDNF
- BS, brainstem
- Cb, cerebellum
- Ctx, cerebral cortex
- DMH, dorsomedial hypothalamus
- Hp, hippocampus
- Hy, hypothalamus
- Hyperphagia
- Hypothalamus
- LH, lateral hypothalamus
- NTS, nucleus of the solitary tract
- Obesity
- PMV, ventral premammillary nucleus
- PVH, paraventricular hypothalamus
- Rgs9-Cre
- SN, substantia nigra
- Stm, striatum
- TrkB
- Tu, olfactory tubercle
- VMH, ventromedial hypothalamus
Collapse
Affiliation(s)
- Guey-Ying Liao
- Department of Pharmacology and Physiology, Georgetown University Medical Center, Washington, DC 20057, USA ; Department of Neuroscience, The Scripps Research Institute Florida, Jupiter, FL 33458, USA
| | | | | |
Collapse
|
36
|
Gao S, Serra D, Keung W, Hegardt FG, Lopaschuk GD. Important role of ventromedial hypothalamic carnitine palmitoyltransferase-1a in the control of food intake. Am J Physiol Endocrinol Metab 2013; 305:E336-47. [PMID: 23736540 DOI: 10.1152/ajpendo.00168.2013] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Carnitine palmitoyltransferase-1 (CPT-1) liver isoform, or CPT-1a, is implicated in CNS control of food intake. However, the exact brain nucleus site(s) in mediating this action of CPT-1a has not been identified. In this report, we assess the role of CPT-1a in hypothalamic ventromedial nucleus (VMN). We stereotaxically injected an adenoviral vector containing CPT-1a coding sequence into the VMN of rats to induce overexpression and activation of CPT-1a. The VMN-selective activation of CPT-1a induced an orexigenic effect, suggesting CPT-1a in the VMN is involved in the central control of feeding. Intracerebroventricular administration of etomoxir, a CPT-1 inhibitor, decreases food intake. Importantly, in the animals with VMN overexpression of a CPT-1a mutant that antagonizes the CPT-1 inhibition by etomoxir, the anorectic response to etomoxir was attenuated. This suggests that VMN is involved in mediating the anorectic effect of central inhibition of CPT-1a. In contrast, arcuate nucleus (Arc) overexpression of the mutant did not alter etomoxir-induced inhibition of food intake, suggesting that Arc CPT-1a does not play significant roles in this anorectic action. Furthermore, in the VMN, CPT-1a appears to act downstream of hypothalamic malonyl-CoA action of feeding. Finally, we show that in the VMN CPT-1 activity was altered in concert with fasting and refeeding states, supporting a physiological role of CPT-1a in mediating the control of feeding. All together, CPT-1a in the hypothalamic VMN appears to play an important role in central control of food intake. VMN-selective modulation of CPT-1a activity may therefore be a promising strategy in controlling food intake and maintaining normal body weight.
Collapse
Affiliation(s)
- Su Gao
- Department of Pediatrics, Mazankowski Alberta Heart Institute, University of Alberta, Edmonton, Alberta, Canada
| | | | | | | | | |
Collapse
|
37
|
Byerly MS, Swanson RD, Semsarzadeh NN, McCulloh PS, Kwon K, Aja S, Moran TH, Wong GW, Blackshaw S. Identification of hypothalamic neuron-derived neurotrophic factor as a novel factor modulating appetite. Am J Physiol Regul Integr Comp Physiol 2013; 304:R1085-95. [PMID: 23576617 DOI: 10.1152/ajpregu.00368.2012] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Disruption of finely coordinated neuropeptide signals in the hypothalamus can result in altered food intake and body weight. We identified neuron-derived neurotrophic factor (NENF) as a novel secreted protein through a large-scale screen aimed at identifying novel secreted hypothalamic proteins that regulate food intake. We observed robust Nenf expression in hypothalamic nuclei known to regulate food intake, and its expression was altered under the diet-induced obese (DIO) condition relative to the fed state. Hypothalamic Nenf mRNA was regulated by brain-derived neurotrophic factor (BDNF) signaling, itself an important regulator of appetite. Delivery of purified recombinant BDNF into the lateral cerebral ventricle decreased hypothalamic Nenf expression, while pharmacological inhibition of trkB signaling increased Nenf mRNA expression. Furthermore, recombinant NENF administered via an intracerebroventricular cannula decreased food intake and body weight and increased hypothalamic Pomc and Mc4r mRNA expression. Importantly, the appetite-suppressing effect of NENF was abrogated in obese mice fed a high-fat diet, demonstrating a diet-dependent modulation of NENF function. We propose the existence of a regulatory circuit involving BDNF, NENF, and melanocortin signaling. Our study validates the power of using an integrated experimental and bioinformatic approach to identify novel CNS-derived proteins with appetite-modulating function and reveals NENF as an important central modulator of food intake.
Collapse
Affiliation(s)
- Mardi S Byerly
- Department of Physiology and Center for Metabolism and Obesity Research, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|
38
|
FTO predicts weight regain in the Look AHEAD clinical trial. Int J Obes (Lond) 2013; 37:1545-52. [PMID: 23628854 PMCID: PMC3750057 DOI: 10.1038/ijo.2013.54] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/06/2012] [Revised: 03/18/2013] [Accepted: 03/23/2013] [Indexed: 12/20/2022]
Abstract
Background Genome-wide association studies have provided new insights into the genetic factors that contribute to the development of obesity. We hypothesized that these genetic markers would also predict magnitude of weight loss and weight regain after initial weight loss. Methods Established obesity risk alleles available on the Illumina CARe iSelect (IBC) chip were characterized in 3,899 overweight or obese participants with type 2 diabetes from the Look AHEAD (Action for Health in Diabetes), a randomized trial to determine the effects of intensive lifestyle intervention (ILI) and Diabetes Support and Education (DSE) on cardiovascular morbidity and mortality. Primary analyses examined the interaction between 13 obesity-risk polymorphisms in 8 genes and randomized treatment arm in predicting weight change at year 1, and weight regain at year 4 among individuals who lost 3% or more of their baseline weight by year 1. Results No SNPs were significantly associated with magnitude of weight loss or interacted with treatment arm at year 1. However, FTO rs3751812 predicted weight regain within DSE (1.56 kg per risk allele, p = 0.005), but not ILI (p = 0.761), resulting in SNP×treatment arm interaction (p = 0.009). In a partial replication of prior research, the obesity risk (G) allele at BDNF rs6265 was associated with greater weight regain across treatment arms (0.773 kg per risk allele), although results were of borderline statistical significance (p=0.051). Conclusions Variations in the FTO and BDNF loci may contribute risk of weight regain after weight loss.
Collapse
|
39
|
Vanevski F, Xu B. Molecular and neural bases underlying roles of BDNF in the control of body weight. Front Neurosci 2013; 7:37. [PMID: 23519010 PMCID: PMC3604627 DOI: 10.3389/fnins.2013.00037] [Citation(s) in RCA: 59] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2012] [Accepted: 03/03/2013] [Indexed: 01/05/2023] Open
Abstract
Brain-derived neurotrophic factor (BDNF) is a potent regulator of neuronal development and synaptic plasticity that is fundamental to neural circuit formation and cognition. It is also involved in the control of appetite and body weight, with mutations in the genes for BDNF and its receptor, TrkB, resulting in remarkable hyperphagia and severe obesity in humans and mice. Recent studies have made significant progress in elucidating the source, action sites, and regulatory pathways of BDNF with regard to its role in the control of energy homeostasis, and have shed light on the relationships between BDNF and other molecules involved in the control of body weight. Here we provide a comprehensive review of evidence from pharmacological, genetic, and mechanistic studies, linking BDNF to the control of body weight. This review also aims to organize the main findings on this subject into a more refined framework and to discuss the future research directions necessary to advance the field.
Collapse
Affiliation(s)
- Filip Vanevski
- Department of Pharmacology and Physiology, Georgetown University Medical Center Washington, DC, USA
| | | |
Collapse
|
40
|
Gotoh K, Masaki T, Chiba S, Ando H, Fujiwara K, Shimasaki T, Mitsutomi K, Katsuragi I, Kakuma T, Sakata T, Yoshimatsu H. Brain-derived neurotrophic factor, corticotropin-releasing factor, and hypothalamic neuronal histamine interact to regulate feeding behavior. J Neurochem 2013; 125:588-98. [DOI: 10.1111/jnc.12213] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2012] [Revised: 02/19/2013] [Accepted: 02/19/2013] [Indexed: 11/28/2022]
Affiliation(s)
- Koro Gotoh
- Department of Internal Medicine 1; Faculty of Medicine; Oita University; Yufu Japan
| | - Takayuki Masaki
- Department of Internal Medicine 1; Faculty of Medicine; Oita University; Yufu Japan
| | - Seiichi Chiba
- Department of Internal Medicine 1; Faculty of Medicine; Oita University; Yufu Japan
| | - Hisae Ando
- Department of Internal Medicine 1; Faculty of Medicine; Oita University; Yufu Japan
| | - Kansuke Fujiwara
- Department of Internal Medicine 1; Faculty of Medicine; Oita University; Yufu Japan
| | - Takanobu Shimasaki
- Department of Internal Medicine 1; Faculty of Medicine; Oita University; Yufu Japan
| | - Kimihiko Mitsutomi
- Department of Internal Medicine 1; Faculty of Medicine; Oita University; Yufu Japan
| | - Isao Katsuragi
- Department of Internal Medicine 1; Faculty of Medicine; Oita University; Yufu Japan
| | - Tetsuya Kakuma
- Department of Internal Medicine 1; Faculty of Medicine; Oita University; Yufu Japan
| | - Toshiie Sakata
- Department of Internal Medicine 1; Faculty of Medicine; Oita University; Yufu Japan
| | - Hironobu Yoshimatsu
- Department of Internal Medicine 1; Faculty of Medicine; Oita University; Yufu Japan
| |
Collapse
|
41
|
Zhu Z, Liu X, Kumar SPDS, Zhang J, Shi H. Central expression and anorectic effect of brain-derived neurotrophic factor are regulated by circulating estradiol levels. Horm Behav 2013; 63:533-42. [PMID: 23376487 PMCID: PMC3624754 DOI: 10.1016/j.yhbeh.2013.01.009] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/22/2012] [Revised: 01/15/2013] [Accepted: 01/21/2013] [Indexed: 10/27/2022]
Abstract
Estrogens potently suppress food intake. Compelling evidence suggests that estradiol, the primary form of estrogens, reduces food intake by facilitating other anorectic signals. Brain-derived neurotrophic factor (BDNF), like estradiol, appears to suppress food intake by affecting meal size. We hypothesized that estradiol modulates Bdnf expression and the anorectic effect of BDNF. The first goal was to determine whether Bdnf expression was regulated by endogenous estradiol of cycling rats and by cyclic estradiol treatment using ovariectomized rats. Bdnf expression within the ventromedial nucleus of hypothalamus (VMH) was temporally elevated at estrus following the estradiol peak, which coincided with the decline in feeding at this phase of the ovarian cycle. Additionally, food intake and body weight were increased following ovariectomy with a parallel decrease in Bdnf expression in the VMH. All of these alterations were reversed by cyclic estradiol treatment, suggesting that Bdnf expression within the VMH was regulated in an estradiol-dependent manner. The second goal was to determine whether estradiol modulates the anorectic effect of BDNF. Sham-operated estrous rats and ovariectomized rats cyclically treated with estradiol responded to a lower dose of central administration of BDNF to decrease food intake than male rats and oil-treated ovariectomized rats, implying that endogenous estradiol or cyclic estradiol replacement increased the sensitivity to anorectic effect of BDNF. These data indicate that Bdnf expression within the VMH and the anorectic effect of BDNF varied depending on plasma estradiol levels, suggesting that estradiol may regulate BDNF signaling to regulate feeding.
Collapse
Affiliation(s)
- Zheng Zhu
- Center for Physiology and Neuroscience, Department of Biology, Miami University, Oxford, Ohio, United States
| | - Xian Liu
- Cell, Molecular and Structural Biology, Miami University, Oxford, Ohio, United States
| | | | - Jing Zhang
- Department of Statistics, Miami University, Oxford, Ohio, United States
| | - Haifei Shi
- Center for Physiology and Neuroscience, Department of Biology, Miami University, Oxford, Ohio, United States
- Cell, Molecular and Structural Biology, Miami University, Oxford, Ohio, United States
| |
Collapse
|
42
|
Rios M. BDNF and the central control of feeding: accidental bystander or essential player? Trends Neurosci 2013; 36:83-90. [PMID: 23333344 DOI: 10.1016/j.tins.2012.12.009] [Citation(s) in RCA: 97] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2012] [Revised: 12/19/2012] [Accepted: 12/20/2012] [Indexed: 01/15/2023]
Abstract
A considerable body of evidence links diminished brain-derived neurotrophic factor (BDNF) signaling to energy balance dysregulation and severe obesity in humans and rodents. Because BDNF exhibits broad neurotrophic properties, the underpinnings of these effects and its true role in the central regulation of food intake remain topics of debate in the field. Here, I discuss recent evidence supporting a critical role for this neurotrophin in physiological mechanisms regulating nutrient intake and body weight in the mature brain. They include reports of functional interactions of BDNF with central anorexigenic and orexigenic signaling pathways and evidence of recognized appetite hormones exerting neurotrophic effects similar to those of BDNF.
Collapse
Affiliation(s)
- Maribel Rios
- Tufts University School of Medicine, Department of Neuroscience, Boston, MA 02111, USA.
| |
Collapse
|
43
|
Fargali S, Sadahiro M, Jiang C, Frick AL, Indall T, Cogliani V, Welagen J, Lin WJ, Salton SR. Role of neurotrophins in the development and function of neural circuits that regulate energy homeostasis. J Mol Neurosci 2012; 48:654-9. [PMID: 22581449 DOI: 10.1007/s12031-012-9790-9] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2012] [Accepted: 04/26/2012] [Indexed: 12/21/2022]
Abstract
Members of the neurotrophin family, including nerve growth factor, brain-derived neurotrophic factor, neurotrophin-3, and neurotrophin-4/5, and other neurotrophic growth factors such as ciliary neurotrophic factor and artemin, regulate peripheral and central nervous system development and function. A subset of the neurotrophin-dependent pathways in the hypothalamus, brainstem, and spinal cord, and those that project via the sympathetic nervous system to peripheral metabolic tissues including brown and white adipose tissue, muscle and liver, regulate feeding, energy storage, and energy expenditure. We briefly review the role that neurotrophic growth factors play in energy balance, as regulators of neuronal survival and differentiation, neurogenesis, and circuit formation and function, and as inducers of critical gene products that control energy homeostasis.
Collapse
Affiliation(s)
- Samira Fargali
- Fishberg Department of Neuroscience, Mount Sinai School of Medicine, New York, NY 10029, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Spaeth AM, Kanoski SE, Hayes MR, Grill HJ. TrkB receptor signaling in the nucleus tractus solitarius mediates the food intake-suppressive effects of hindbrain BDNF and leptin. Am J Physiol Endocrinol Metab 2012; 302:E1252-60. [PMID: 22374757 PMCID: PMC3361983 DOI: 10.1152/ajpendo.00025.2012] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Brain-derived neurotrophic factor (BDNF) and TrkB receptor signaling contribute to the central nervous system (CNS) control of energy balance. The role of hindbrain BDNF/TrkB receptor signaling in energy balance regulation is examined here. Hindbrain ventricular BDNF suppressed body weight through reductions in overall food intake and meal size and by increasing core temperature. To localize the neurons mediating the energy balance effects of hindbrain ventricle-delivered BDNF, ventricle subthreshold doses were delivered directly to medial nucleus tractus solitarius (mNTS). mNTS BDNF administration reduced food intake significantly, and this effect was blocked by preadministration of a highly selective TrkB receptor antagonist {[N2-2-2-Oxoazepan-3-yl amino]carbonyl phenyl benzo (b)thiophene-2-carboxamide (ANA-12)}, suggesting that TrkB receptor activation mediates hindbrain BDNF's effect on food intake. Because both BDNF and leptin interact with melanocortin signaling to reduce food intake, we also examined whether the intake inhibitory effects of hindbrain leptin involve hindbrain-specific BDNF/TrkB activation. BDNF protein content within the dorsal vagal complex of the hindbrain was increased significantly by hindbrain leptin delivery. To assess if BDNF/TrkB receptor signaling acts downstream of leptin signaling in the control of energy balance, leptin and ANA-12 were coadministered into the mNTS. Administration of the TrkB receptor antagonist attenuated the intake-suppressive effects of leptin, suggesting that mNTS TrkB receptor activation contributes to the mediation of the anorexigenic effects of hindbrain leptin. Collectively, these results indicate that TrkB-mediated signaling in the mNTS negatively regulates food intake and, in part, the intake inhibitory effects of leptin administered into the NTS.
Collapse
Affiliation(s)
- Andrea M Spaeth
- Dept. of Psychology, Univ. of Pennsylvania, 3720 Walnut St., Philadelphia, PA 19104, USA
| | | | | | | |
Collapse
|
45
|
Novak CM, Burghardt PR, Levine JA. The use of a running wheel to measure activity in rodents: relationship to energy balance, general activity, and reward. Neurosci Biobehav Rev 2012; 36:1001-1014. [PMID: 22230703 DOI: 10.1016/j.neubiorev.2011.12.012] [Citation(s) in RCA: 219] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2011] [Revised: 12/07/2011] [Accepted: 12/22/2011] [Indexed: 12/21/2022]
Abstract
Running wheels are commonly employed to measure rodent physical activity in a variety of contexts, including studies of energy balance and obesity. There is no consensus on the nature of wheel-running activity or its underlying causes, however. Here, we will begin by systematically reviewing how running wheel availability affects physical activity and other aspects of energy balance in laboratory rodents. While wheel running and physical activity in the absence of a wheel commonly correlate in a general sense, in many specific aspects the two do not correspond. In fact, the presence of running wheels alters several aspects of energy balance, including body weight and composition, food intake, and energy expenditure of activity. We contend that wheel-running activity should be considered a behavior in and of itself, reflecting several underlying behavioral processes in addition to a rodent's general, spontaneous activity. These behavioral processes include defensive behavior, predatory aggression, and depression- and anxiety-like behaviors. As it relates to energy balance, wheel running engages several brain systems-including those related to the stress response, mood, and reward, and those responsive to growth factors-that influence energy balance indirectly. We contend that wheel-running behavior represents factors in addition to rodents' tendency to be physically active, engaging additional neural and physiological mechanisms which can then independently alter energy balance and behavior. Given the impact of wheel-running behavior on numerous overlapping systems that influence behavior and physiology, this review outlines the need for careful design and interpretation of studies that utilize running wheels as a means for exercise or as a measurement of general physical activity.
Collapse
Affiliation(s)
- Colleen M Novak
- Department of Biological Sciences, Kent State University, PO Box 5190, 222 Cunningham Hall, Kent, OH 44242, United States
| | | | - James A Levine
- Mayo Clinic, Endocrine Research Unit, Rochester, MN 55905, United States
| |
Collapse
|
46
|
Cordeira J, Rios M. Weighing in the role of BDNF in the central control of eating behavior. Mol Neurobiol 2011; 44:441-8. [PMID: 22012072 PMCID: PMC3235948 DOI: 10.1007/s12035-011-8212-2] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2011] [Accepted: 10/07/2011] [Indexed: 01/24/2023]
Abstract
The prevalence of obesity and its associated medical complications, including type 2 diabetes and cardiovascular disease, continues to rise globally. Lifestyle changes in the last decades have greatly contributed to the current obesity trends. However, inheritable biological factors that disrupt the tightly regulated equilibrium between caloric intake and energy expenditure also appear to play a critical part. Mounting evidence obtained from human and rodent studies suggests that perturbed brain-derived neurotrophic factor (BDNF) signaling in appetite-regulating centers in the brain might be a culprit. Here, we review findings that inform the critical roles of BDNF and its receptor TrkB in energy balance and reward centers of the brain impacting feeding behavior and body weight.
Collapse
Affiliation(s)
- Joshua Cordeira
- Department of Neuroscience and Program in Neuroscience, Sackler School of Graduate, Biomedical Sciences, Tufts University School of Medicine, Boston, MA 02111 USA
| | - Maribel Rios
- Department of Neuroscience and Program in Neuroscience, Sackler School of Graduate, Biomedical Sciences, Tufts University School of Medicine, Boston, MA 02111 USA
| |
Collapse
|
47
|
Rask-Andersen M, Almén MS, Olausen HR, Olszewski PK, Eriksson J, Chavan RA, Levine AS, Fredriksson R, Schiöth HB. Functional coupling analysis suggests link between the obesity gene FTO and the BDNF-NTRK2 signaling pathway. BMC Neurosci 2011; 12:117. [PMID: 22087873 PMCID: PMC3248879 DOI: 10.1186/1471-2202-12-117] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2010] [Accepted: 11/16/2011] [Indexed: 11/24/2022] Open
Abstract
Background The Fat mass and obesity gene (FTO) has been identified through genome wide association studies as an important genetic factor contributing to a higher body mass index (BMI). However, the molecular context in which this effect is mediated has yet to be determined. We investigated the potential molecular network for FTO by analyzing co-expression and protein-protein interaction databases, Coxpresdb and IntAct, as well as the functional coupling predicting multi-source database, FunCoup. Hypothalamic expression of FTO-linked genes defined with this bioinformatics approach was subsequently studied using quantitative real time-PCR in mouse feeding models known to affect FTO expression. Results We identified several candidate genes for functional coupling to FTO through database studies and selected nine for further study in animal models. We observed hypothalamic expression of Profilin 2 (Pfn2), cAMP-dependent protein kinase catalytic subunit beta (Prkacb), Brain derived neurotrophic factor (Bdnf), neurotrophic tyrosine kinase, receptor, type 2 (Ntrk2), Signal transducer and activator of transcription 3 (Stat3), and Btbd12 to be co-regulated in concert with Fto. Pfn2 and Prkacb have previously not been linked to feeding regulation. Conclusions Gene expression studies validate several candidates generated through database studies of possible FTO-interactors. We speculate about a wider functional role for FTO in the context of current and recent findings, such as in extracellular ligand-induced neuronal plasticity via NTRK2/BDNF, possibly via interaction with the transcription factor CCAAT/enhancer binding protein β (C/EBPβ).
Collapse
Affiliation(s)
- Mathias Rask-Andersen
- Department of Neuroscience, Functional Pharmacology, Uppsala University, BMC, Uppsala SE 75124, Sweden
| | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Godar R, Dai Y, Bainter H, Billington C, Kotz CM, Wang C. Reduction of high-fat diet-induced obesity after chronic administration of brain-derived neurotrophic factor in the hypothalamic ventromedial nucleus. Neuroscience 2011; 194:36-52. [PMID: 21856381 PMCID: PMC3190117 DOI: 10.1016/j.neuroscience.2011.07.079] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2011] [Revised: 06/24/2011] [Accepted: 07/13/2011] [Indexed: 12/16/2022]
Abstract
An acute injection of brain-derived neurotrophic factor (BDNF) in the hypothalamic ventromedial nucleus (VMN) decreases body weight by reducing feeding and increasing energy expenditure (EE) in animals on standard laboratory chow. Animals have divergent responses to high-fat diet (HFD) exposure, with some developing obesity and others remaining lean. In the current study, we tested the hypothesis that BDNF in the VMN reduces HFD-induced obesity. Seventy-two 10-week old rats were allowed HFD ad libitum for 8 weeks and then prepared with bilateral VMN cannulae. Animals were then divided into tertiles based on their fat mass rank: high, intermediate, and low (H, I, and L). Each group was further divided into two subgroups: BDNF (1 μg) or control (artificial cerebrospinal fluid, aCSF); they were then injected every other day for 20 days according to subgroup. Energy intake, body weight, and body composition were measured. Other metabolic indexes were measured before and after treatment. In parallel, another 12 rats were fed control diet (CD), VMN-cannulated, and injected with aCSF. HFD exposure induced obesity in the H group, with a significant increase in energy intake, body weight, fat mass, liver size, and serum glucose, insulin, and leptin. BDNF significantly reduced body weight and fat mass in all phenotypes, while it reduced energy intake only in the I group. However, BDNF increased EE, spontaneous physical activity, and fat oxidation in the H group, suggesting that BDNF-induced EE elevation contributed to reduction of body weight and fat mass. Chronic VMN BDNF reduced insulin elevation and/or reversed hyperleptinemia. These data suggest that the VMN is an important site of action for BDNF reduction of HFD-induced obesity.
Collapse
Affiliation(s)
- Rebecca Godar
- Veterans Affairs Medical Center, One Veterans Drive, Research Route 151, Minneapolis, MN 55417, USA
| | - Yuqiao Dai
- Department of Food Science and Nutrition, University of Minnesota, 1334 Eckles Avenue, Saint Paul, MN 55108, USA
| | - Heather Bainter
- Department of Food Science and Nutrition, University of Minnesota, 1334 Eckles Avenue, Saint Paul, MN 55108, USA
| | - Charles Billington
- Veterans Affairs Medical Center, One Veterans Drive, Research Route 151, Minneapolis, MN 55417, USA
- Minnesota Obesity Center, One Veterans Drive, Research Route 151, Minneapolis, MN 55417, USA
- Department of Food Science and Nutrition, University of Minnesota, 1334 Eckles Avenue, Saint Paul, MN 55108, USA
| | - Catherine M. Kotz
- Veterans Affairs Medical Center, One Veterans Drive, Research Route 151, Minneapolis, MN 55417, USA
- Minnesota Obesity Center, One Veterans Drive, Research Route 151, Minneapolis, MN 55417, USA
- Department of Food Science and Nutrition, University of Minnesota, 1334 Eckles Avenue, Saint Paul, MN 55108, USA
- Graduate Program in Neuroscience, University of Minnesota, 1334 Eckles Avenue, Saint Paul, MN 55108, USA
| | - ChuanFeng Wang
- Veterans Affairs Medical Center, One Veterans Drive, Research Route 151, Minneapolis, MN 55417, USA
- Minnesota Obesity Center, One Veterans Drive, Research Route 151, Minneapolis, MN 55417, USA
- Department of Food Science and Nutrition, University of Minnesota, 1334 Eckles Avenue, Saint Paul, MN 55108, USA
| |
Collapse
|
49
|
Carreño FR, Walch JD, Dutta M, Nedungadi TP, Cunningham JT. Brain-derived neurotrophic factor-tyrosine kinase B pathway mediates NMDA receptor NR2B subunit phosphorylation in the supraoptic nuclei following progressive dehydration. J Neuroendocrinol 2011; 23:894-905. [PMID: 21848649 PMCID: PMC3183156 DOI: 10.1111/j.1365-2826.2011.02209.x] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
We studied the effects of water deprivation (WD) on the phosphorylation of tyrosine kinase B (TrkB) and NMDA receptor subunits in the supraoptic nucleus (SON) of the rat. Laser capture microdissection and quantitative reverse transcriptase polymerase chain reaction was used to demonstrate brain-derived neurotrophic factor (BDNF) and TrkB gene expression in vasopressin SON neurones. Immunohistochemistry confirmed BDNF staining in vasopressin neurones, whereas staining for phosphorylated TrkB was increased following WD. Western blot analysis of brain punches containing the SON revealed that tyrosine phosphorylation of TrkB (pTrkBY(515)), serine phosphorylation of NR1 (pNR1S(866) or pNR1) and tyrosine phosphorylation of NR2B subunits (pNR2BY(1472) or pNR2B) were significantly increased in WD animals compared to controls. Access to water for 2 h reduced pTrkBY(515) content to control levels without affecting pNR1 or pNR2B. Four hours of rehydration was needed to reduce pNR1 and pNR2B to control levels. To test whether increased phosphorylation of TrkB in the present study is mediated by BDNF, a group of animals were instrumented with right SON cannula coupled to mini-osmotic pumps filled with vehicle or TrkB-Fc fusion protein, which prevents BDNF binding to TrkB. In the left SON contralateral to the cannula, TrkB phosphorylation was significantly enhanced following WD. Separate analysis of the right SON, which received TrkB-Fc, showed that the TrkB receptor phosphorylation following WD was significantly attenuated. Although increased pNR1S(866) following WD was not affected by local infusion of TrkB-Fc, pNR2BY(1472) was significantly reduced. Co-immunoprecipitation revealed an increased physical interaction between Fyn kinase and NR2B and TrkB in the SON following WD. Thus, activation of TrkB in the SON following WD may affect cellular excitability through the phosphorylation of NR2B subunits.
Collapse
Affiliation(s)
- Flávia Regina Carreño
- Department of Integrative Physiology, and Cardiovascular Research Institute, University of North Texas Health Science Center at Fort Worth, 3500 Camp Bowie Blvd, Fort Worth, TX 76107
| | - Joseph D Walch
- Department of Integrative Physiology, and Cardiovascular Research Institute, University of North Texas Health Science Center at Fort Worth, 3500 Camp Bowie Blvd, Fort Worth, TX 76107
- Department of Pharmacology and Neuroscience, University of Texas Health Science Center at San Antonio, 7703 Floyd Curl Drive, San Antonio, TX 78229
| | - Mayurika Dutta
- Department of Integrative Physiology, and Cardiovascular Research Institute, University of North Texas Health Science Center at Fort Worth, 3500 Camp Bowie Blvd, Fort Worth, TX 76107
| | - Thekkethil P. Nedungadi
- Department of Integrative Physiology, and Cardiovascular Research Institute, University of North Texas Health Science Center at Fort Worth, 3500 Camp Bowie Blvd, Fort Worth, TX 76107
| | - J. Thomas Cunningham
- Department of Integrative Physiology, and Cardiovascular Research Institute, University of North Texas Health Science Center at Fort Worth, 3500 Camp Bowie Blvd, Fort Worth, TX 76107
| |
Collapse
|
50
|
Brain-derived neurotrophic factor, food intake regulation, and obesity. Arch Med Res 2011; 42:482-94. [PMID: 21945389 DOI: 10.1016/j.arcmed.2011.09.005] [Citation(s) in RCA: 114] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2011] [Accepted: 08/10/2011] [Indexed: 12/14/2022]
Abstract
Brain-derived neurotrophic factor (BDNF) is a neurotrophin that plays a fundamental role in development and plasticity of the central nervous system (CNS). It is currently recognized as a major participant in the regulation of food intake. Multiple studies have shown that different regulators of appetite such as leptin, insulin and pancreatic polypeptide (PP) potentially exert anorexigenic effects through BDNF. Low circulating levels of BDNF are associated with a higher risk of eating disorders such as anorexia nervosa (AN) and bulimia nervosa (BN). Strict food restriction reduces BDNF and may trigger binge-eating episodes and weight gain. The existence of mutations that cause haploinsufficiency of BDNF as well as some genetic variants, notably the BDNF p.Val66Met polymorphism, are also associated with the development of obese phenotypes and hyperphagia. However, association of the Met allele with AN and BN, which have different phenotypic characteristics, shows clearly the existence of other relevant factors that regulate eating behavior. This may, in part, be explained by the epigenetic regulation of BDNF through mechanisms like DNA methylation and histone acetylation. Environmental factors, primarily during early development, are crucial to the establishment of these stable but reversible changes that alter the transcriptional expression and are transgenerationally heritable, with potential concomitant effects on the development of eating disorders and body weight control.
Collapse
|