1
|
Shen YR, Cheng L, Zhang DF. TRPV1: A novel target for the therapy of diabetes and diabetic complications. Eur J Pharmacol 2024; 984:177021. [PMID: 39362389 DOI: 10.1016/j.ejphar.2024.177021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2024] [Revised: 09/18/2024] [Accepted: 09/30/2024] [Indexed: 10/05/2024]
Abstract
BACKGROUND Diabetes mellitus is a chronic metabolic disease characterized by abnormally elevated blood glucose levels. Type II diabetes accounts for approximately 90% of all cases. Several drugs are available for hyperglycemia treatment. However, the current therapies for managing high blood glucose do not prevent or reverse the disease progression, which may result in complications and adverse effects, including diabetic neuropathy, retinopathy, and nephropathy. Hence, developing safer and more effective methods for lowering blood glucose levels is imperative. Transient receptor potential vanilloid-1 (TRPV1) is a significant member of the transient receptor potential family. It is present in numerous body tissues and organs and performs vital physiological functions. PURPOSE This review aimed to develop new targeted TRPV1 hypoglycemic drugs by systematically summarizing the mechanism of action of the TRPV1-based signaling pathway in preventing and treating diabetes and its complications. METHODS Literature searches were performed in the PubMed, Web of Science, Google Scholar, Medline, and Scopus databases for 10 years from 2013 to 2023. The search terms included "diabetes," "TRPV1," "diabetic complications," and "capsaicin." RESULTS TRPV1 is an essential potential target for treating diabetes mellitus and its complications. It reduces hepatic glucose production and food intake and promotes thermogenesis, metabolism, and insulin secretion. Activation of TRPV1 ameliorates diabetic nephropathy, retinopathy, myocardial infarction, vascular endothelial dysfunction, gastroparesis, and bladder dysfunction. Suppression of TRPV1 improves diabetes-related osteoporosis. However, the therapeutic effects of activating or suppressing TRPV1 may vary when treating diabetic neuropathy and periodontitis. CONCLUSION This review demonstrates that TRPV1 is a potential therapeutic target for diabetes and its complications. Additionally, it provides a theoretical basis for developing new hypoglycemic drugs that target TRPV1.
Collapse
Affiliation(s)
- Yu-Rong Shen
- Department of Pharmacognosy, School of Pharmacy, China Medical University, Shenyang 110122, China
| | - Long Cheng
- Department of Pharmacognosy, School of Pharmacy, China Medical University, Shenyang 110122, China.
| | - Dong-Fang Zhang
- Department of Pharmacognosy, School of Pharmacy, China Medical University, Shenyang 110122, China.
| |
Collapse
|
2
|
Coverdell TC, Abbott SBG, Campbell JN. Molecular cell types as functional units of the efferent vagus nerve. Semin Cell Dev Biol 2024; 156:210-218. [PMID: 37507330 PMCID: PMC10811285 DOI: 10.1016/j.semcdb.2023.07.007] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2022] [Revised: 07/20/2023] [Accepted: 07/20/2023] [Indexed: 07/30/2023]
Abstract
The vagus nerve vitally connects the brain and body to coordinate digestive, cardiorespiratory, and immune functions. Its efferent neurons, which project their axons from the brainstem to the viscera, are thought to comprise "functional units" - neuron populations dedicated to the control of specific vagal reflexes or organ functions. Previous research indicates that these functional units differ from one another anatomically, neurochemically, and physiologically but have yet to define their identity in an experimentally tractable way. However, recent work with genetic technology and single-cell genomics suggests that genetically distinct subtypes of neurons may be the functional units of the efferent vagus. Here we review how these approaches are revealing the organizational principles of the efferent vagus in unprecedented detail.
Collapse
Affiliation(s)
- Tatiana C Coverdell
- Biomedical Sciences Graduate Program, University of Virginia, Charlottesville, VA 22903, USA
| | - Stephen B G Abbott
- Department of Pharmacology, University of Virginia, Charlottesville, VA 22903, USA
| | - John N Campbell
- Department of Biology, University of Virginia, Charlottesville, VA 22903, USA.
| |
Collapse
|
3
|
Boyle CA, Kola PK, Oraegbuna CS, Lei S. Leptin excites basolateral amygdala principal neurons and reduces food intake by LepRb-JAK2-PI3K-dependent depression of GIRK channels. J Cell Physiol 2024; 239:e31117. [PMID: 37683049 PMCID: PMC10920395 DOI: 10.1002/jcp.31117] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2023] [Revised: 08/08/2023] [Accepted: 08/25/2023] [Indexed: 09/10/2023]
Abstract
Leptin is an adipocyte-derived hormone that modulates food intake, energy balance, neuroendocrine status, thermogenesis, and cognition. Whereas a high density of leptin receptors has been detected in the basolateral amygdala (BLA) neurons, the physiological functions of leptin in the BLA have not been determined yet. We found that application of leptin excited BLA principal neurons by activation of the long form leptin receptor, LepRb. The LepRb-elicited excitation of BLA neurons was mediated by depression of the G protein-activated inwardly rectifying potassium (GIRK) channels. Janus Kinase 2 (JAK2) and phosphoinositide 3-kinase (PI3K) were required for leptin-induced excitation of BLA neurons and depression of GIRK channels. Microinjection of leptin into the BLA reduced food intake via activation of LepRb, JAK2, and PI3K. Our results may provide a cellular and molecular mechanism to explain the physiological roles of leptin in vivo.
Collapse
Affiliation(s)
- Cody A. Boyle
- Department of Biomedical Sciences, School of Medicine and Health Sciences, University of North Dakota, Grand Forks, ND58203, USA
| | - Phani K. Kola
- Department of Biomedical Sciences, School of Medicine and Health Sciences, University of North Dakota, Grand Forks, ND58203, USA
| | - Chidiebele S. Oraegbuna
- Department of Biomedical Sciences, School of Medicine and Health Sciences, University of North Dakota, Grand Forks, ND58203, USA
| | - Saobo Lei
- Department of Biomedical Sciences, School of Medicine and Health Sciences, University of North Dakota, Grand Forks, ND58203, USA
| |
Collapse
|
4
|
Juras JA, Pitra S, Smith BN. Systemic Glucose Regulation by a Hindbrain Inhibitory Circuit in a Mouse Model of Type 1 Diabetes. Neuroendocrinology 2024; 114:302-312. [PMID: 38194945 DOI: 10.1159/000536142] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/14/2023] [Accepted: 01/04/2024] [Indexed: 01/11/2024]
Abstract
INTRODUCTION Previous work showed that increasing the electrical activity of inhibitory neurons in the dorsal vagal complex (DVC) is sufficient to increase whole-body glucose concentration in normoglycemic mice. Here we tested the hypothesis that deactivating GABAergic neurons in the dorsal hindbrain of hyperglycemic mice decreases synaptic inhibition of parasympathetic motor neurons in the dorsal motor nucleus of the vagus (DMV) and reduces systemic glucose levels. METHODS Chemogenetic activation or inactivation of GABAergic neurons in the nucleus tractus solitarius (NTS) was used to assess effects of modulating parasympathetic output on blood glucose concentration in normoglycemic and hyperglycemic mice. Patch-clamp electrophysiology in vitro was used to assess cellular effects of chemogenetic manipulation of NTS GABA neurons. RESULTS Chemogenetic activation of GABAergic NTS neurons in normoglycemic mice increased their action potential firing, resulting in increased inhibitory synaptic input to DMV motor neurons and elevated blood glucose concentration. Deactivation of GABAergic DVC neurons in normoglycemic mice altered their electrical activity but did not alter systemic glucose levels. Conversely, stimulation of GABAergic DVC neurons in mice that were hyperglycemic subsequent to treatment with streptozotocin changed their electrical activity but did not alter whole-body glucose concentration, while deactivation of this inhibitory circuit significantly decreased circulating glucose concentration. Peripheral administration of a brain impermeant muscarinic acetylcholine receptor antagonist abolished these effects. CONCLUSION Disinhibiting vagal motor neurons decreases hyperglycemia in a mouse model of type 1 diabetes. This inhibitory brainstem circuit emerges as a key parasympathetic regulator of whole-body glucose homeostasis that undergoes functional plasticity in hyperglycemic conditions.
Collapse
Affiliation(s)
- J Anna Juras
- Department of Neuroscience, University of Kentucky, Lexington, Kentucky, USA
| | - Soledad Pitra
- Department of Neuroscience, University of Kentucky, Lexington, Kentucky, USA
- Department of Biomedical Sciences, Colorado State University, Fort Collins, Colorado, USA
| | - Bret N Smith
- Department of Neuroscience, University of Kentucky, Lexington, Kentucky, USA
- Department of Biomedical Sciences, Colorado State University, Fort Collins, Colorado, USA
| |
Collapse
|
5
|
Jiang Y, Rezai-Zadeh K, Desmoulins LD, Muenzberg H, Derbenev AV, Zsombok A. GABAergic leptin receptor-expressing neurons in the dorsomedial hypothalamus project to brown adipose tissue-related neurons in the paraventricular nucleus of mice. Auton Neurosci 2023; 245:103058. [PMID: 36538864 PMCID: PMC9899324 DOI: 10.1016/j.autneu.2022.103058] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2022] [Revised: 11/14/2022] [Accepted: 12/08/2022] [Indexed: 12/15/2022]
Abstract
Brown adipose tissue (BAT) contributes to energy homeostasis via nonshivering thermogenesis. The BAT is densely innervated by the sympathetic nervous system (SNS) and activity of pre-autonomic neurons modulates the sympathetic outflow. Leptin, an adipocyte hormone, alters energy homeostasis and thermogenesis of BAT via several neuronal circuits; however, the cellular effects of leptin on interscapular BAT (iBAT)-related neurons in the hypothalamus remain to be determined. In this study, we used pseudorabies virus (PRV) to identify iBAT-related neurons in the paraventricular nucleus (PVN) of the hypothalamus and test the hypothesis that iBAT-related PVN neurons are modulated by leptin. Inoculation of iBAT with PRV in leptin receptor reporter mice (Lepr:EGFP) demonstrated that a population of iBAT-related PVN neurons expresses Lepr receptors. Our electrophysiological findings revealed that leptin application caused hyperpolarization in some of iBAT-related PVN neurons. Bath application of leptin also modulated excitatory and inhibitory neurotransmission to most of iBAT-related PVN neurons. Using channel rhodopsin assisted circuit mapping we found that GABAergic and glutamatergic Lepr-expressing neurons in the dorsomedial hypothalamus/dorsal hypothalamic area (dDMH/DHA) project to PVN neurons; however, connected iBAT-related PVN neurons receive exclusively inhibitory signals from Lepr-expressing dDMH/DHA neurons.
Collapse
Affiliation(s)
- Yanyan Jiang
- Department of Physiology, School of Medicine, Tulane University, New Orleans, LA, United States of America; Neuroscience Program, Tulane Brain Institute, Tulane University, New Orleans, LA, United States of America
| | - Kavon Rezai-Zadeh
- Central Leptin Signaling, Pennington Biomedical Research Center, LSU System, Baton Rouge, LA, United States of America
| | - Lucie D Desmoulins
- Department of Physiology, School of Medicine, Tulane University, New Orleans, LA, United States of America
| | - Heike Muenzberg
- Central Leptin Signaling, Pennington Biomedical Research Center, LSU System, Baton Rouge, LA, United States of America
| | - Andrei V Derbenev
- Department of Physiology, School of Medicine, Tulane University, New Orleans, LA, United States of America; Neuroscience Program, Tulane Brain Institute, Tulane University, New Orleans, LA, United States of America
| | - Andrea Zsombok
- Department of Physiology, School of Medicine, Tulane University, New Orleans, LA, United States of America; Neuroscience Program, Tulane Brain Institute, Tulane University, New Orleans, LA, United States of America.
| |
Collapse
|
6
|
Hyun U, Sohn JW. Autonomic control of energy balance and glucose homeostasis. Exp Mol Med 2022; 54:370-376. [PMID: 35474336 PMCID: PMC9076646 DOI: 10.1038/s12276-021-00705-9] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2021] [Accepted: 10/07/2021] [Indexed: 11/21/2022] Open
Abstract
Neurons in the central nervous system (CNS) communicate with peripheral organs largely via the autonomic nervous system (ANS). Through such communications, the sympathetic and parasympathetic efferent divisions of the ANS may affect thermogenesis and blood glucose levels. In contrast, peripheral organs send feedback to the CNS via hormones and autonomic afferent nerves. These humoral and neural feedbacks, as well as neural commands from higher brain centers directly or indirectly shape the metabolic function of autonomic neurons. Notably, recent developments in mouse genetics have enabled more detailed studies of ANS neurons and circuits, which have helped elucidate autonomic control of metabolism. Here, we will summarize the functional organization of the ANS and discuss recent updates on the roles of neural and humoral factors in the regulation of energy balance and glucose homeostasis by the ANS. Cutting-edge techniques should be harnessed to unravel how metabolism is modulated by a key part of the body’s nervous system. The autonomic nervous system (ANS) regulates many involuntary physiological processes, such as heart rate, breathing, and blood pressure. Scientists now believe that the ANS is involved in regulating metabolism, but its precise roles are unclear. Jong-Woo Sohn and Uisu Hyun at the Korea Advanced Institute of Science and Technology, Daejeon, Korea, reviewed understanding of how the ANS regulates energy balance, appetite, and glucose homeostasis. Recently-developed mouse models have provided insights into how ANS neurons translate neuronal and hormonal signals into commands during feeding, sending instructions to the liver, and mediating blood glucose levels. Several hormones have been identified that may act on a specific part of the ANS to influence appetite and metabolism.
Collapse
Affiliation(s)
- Uisu Hyun
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology, Daejeon, 34141, South Korea
| | - Jong-Woo Sohn
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology, Daejeon, 34141, South Korea.
| |
Collapse
|
7
|
Powley TL. Brain-gut communication: vagovagal reflexes interconnect the two "brains". Am J Physiol Gastrointest Liver Physiol 2021; 321:G576-G587. [PMID: 34643086 PMCID: PMC8616589 DOI: 10.1152/ajpgi.00214.2021] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/28/2021] [Revised: 09/24/2021] [Accepted: 09/28/2021] [Indexed: 01/31/2023]
Abstract
The gastrointestinal tract has its own "brain," the enteric nervous system or ENS, that executes routine housekeeping functions of digestion. The dorsal vagal complex in the central nervous system (CNS) brainstem, however, organizes vagovagal reflexes and establishes interconnections between the entire neuroaxis of the CNS and the gut. Thus, the dorsal vagal complex links the "CNS brain" to the "ENS brain." This brain-gut connectome provides reflex adjustments that optimize digestion and assimilation of nutrients and fluid. Vagovagal circuitry also generates the plasticity and adaptability needed to maintain homeostasis to coordinate among organs and to react to environmental situations. Arguably, this dynamic flexibility provided by the vagal circuitry may, in some circumstances, lead to or complicate maladaptive disorders.
Collapse
Affiliation(s)
- Terry L Powley
- Department of Psychological Sciences, Purdue University, West Lafayette, Indiana
| |
Collapse
|
8
|
Wean JB, Smith BN. FGF19 in the Hindbrain Lowers Blood Glucose and Alters Excitability of Vagal Motor Neurons in Hyperglycemic Mice. Endocrinology 2021; 162:6127285. [PMID: 33534906 PMCID: PMC7906449 DOI: 10.1210/endocr/bqab021] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/12/2020] [Indexed: 12/11/2022]
Abstract
Fibroblast growth factor 19 (FGF19) is a protein hormone that produces antidiabetic effects when administered intracerebroventricularly in the forebrain. However, no studies have examined how FGF19 affects hindbrain neurons that participate directly in autonomic control of systemic glucose regulation. Within the dorsal hindbrain, parasympathetic motor neurons of the dorsal motor nucleus of the vagus (DMV) express fibroblast growth factor receptors and their activity regulates visceral homeostatic processes, including energy balance. This study tested the hypothesis that FGF19 acts in the hindbrain to alter DMV neuron excitability and lower blood glucose concentration. Fourth ventricle administration of FGF19 produced no effect on blood glucose concentration in control mice, but induced a significant, peripheral muscarinic receptor-dependent decrease in systemic hyperglycemia for up to 12 h in streptozotocin-treated mice, a model of type 1 diabetes. Patch-clamp recordings from DMV neurons in vitro revealed that FGF19 application altered synaptic and intrinsic membrane properties of DMV neurons, with the balance of FGF19 effects being significantly modified by a recent history of systemic hyperglycemia. These findings identify central parasympathetic circuitry as a novel target for FGF19 and suggest that FGF19 acting in the dorsal hindbrain can alter vagal output to produce its beneficial metabolic effects.
Collapse
Affiliation(s)
- Jordan B Wean
- Department of Physiology, College of Medicine, University of Kentucky, Lexington, KY, USA
| | - Bret N Smith
- Department of Physiology, College of Medicine, University of Kentucky, Lexington, KY, USA
- Department of Neuroscience, College of Medicine, University of Kentucky, Lexington, KY, USA
- Correspondence: Bret N Smith, PhD, Department of Neuroscience, 800 Rose Street, Lexington, KY 40536-0298.
| |
Collapse
|
9
|
Browning KN, Carson KE. Central Neurocircuits Regulating Food Intake in Response to Gut Inputs-Preclinical Evidence. Nutrients 2021; 13:nu13030908. [PMID: 33799575 PMCID: PMC7998662 DOI: 10.3390/nu13030908] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2021] [Revised: 03/02/2021] [Accepted: 03/07/2021] [Indexed: 02/07/2023] Open
Abstract
The regulation of energy balance requires the complex integration of homeostatic and hedonic pathways, but sensory inputs from the gastrointestinal (GI) tract are increasingly recognized as playing critical roles. The stomach and small intestine relay sensory information to the central nervous system (CNS) via the sensory afferent vagus nerve. This vast volume of complex sensory information is received by neurons of the nucleus of the tractus solitarius (NTS) and is integrated with responses to circulating factors as well as descending inputs from the brainstem, midbrain, and forebrain nuclei involved in autonomic regulation. The integrated signal is relayed to the adjacent dorsal motor nucleus of the vagus (DMV), which supplies the motor output response via the efferent vagus nerve to regulate and modulate gastric motility, tone, secretion, and emptying, as well as intestinal motility and transit; the precise coordination of these responses is essential for the control of meal size, meal termination, and nutrient absorption. The interconnectivity of the NTS implies that many other CNS areas are capable of modulating vagal efferent output, emphasized by the many CNS disorders associated with dysregulated GI functions including feeding. This review will summarize the role of major CNS centers to gut-related inputs in the regulation of gastric function with specific reference to the regulation of food intake.
Collapse
|
10
|
Pitra S, Smith BN. Musings on the wanderer: What's new in our understanding of vago-vagal reflexes? VI. Central vagal circuits that control glucose metabolism. Am J Physiol Gastrointest Liver Physiol 2021; 320:G175-G182. [PMID: 33205998 PMCID: PMC7938771 DOI: 10.1152/ajpgi.00368.2020] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Neurons in the brain stem dorsal vagal complex (DVC) take part in a continuous bidirectional crosstalk, in which they receive and respond to a vast array of signaling molecules, including glucose. Importantly, chronic dysregulation of blood glucose concentration, a hallmark of high prevalence pathologies, such as diabetes and metabolic syndrome, can induce neuroplasticity in DVC neural networks, which is hypothesized to either contribute to or compensate for the glycemic or insulinemic dysregulation observed in these conditions. Here, we revisit the topic of vagal reflexes to review recent research on the importance of DVC function in regulating systemic glucose homeostasis and the neuroplastic changes in this brain region that are associated with systemic glucose alterations. We also discuss the critical connection between these nuclei and the gut and the role of central vagal circuits in the favorable outcomes associated with bariatric surgical procedures for metabolic disorders.
Collapse
Affiliation(s)
- Soledad Pitra
- 1Department of Neuroscience, University of Kentucky, Lexington, Kentucky
| | - Bret N. Smith
- 1Department of Neuroscience, University of Kentucky, Lexington, Kentucky,2Department of Physiology, College of Medicine, University of Kentucky, Lexington, Kentucky
| |
Collapse
|
11
|
Wean JB, Smith BN. Fibroblast Growth Factor 19 Increases the Excitability of Pre-Motor Glutamatergic Dorsal Vagal Complex Neurons From Hyperglycemic Mice. Front Endocrinol (Lausanne) 2021; 12:765359. [PMID: 34858337 PMCID: PMC8632226 DOI: 10.3389/fendo.2021.765359] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/26/2021] [Accepted: 10/26/2021] [Indexed: 01/14/2023] Open
Abstract
Intracerebroventricular administration of the protein hormone fibroblast growth factor 19 (FGF19) to the hindbrain produces potent antidiabetic effects in hyperglycemic mice that are likely mediated through a vagal parasympathetic mechanism. FGF19 increases the synaptic excitability of parasympathetic motor neurons in the dorsal motor nucleus of the vagus (DMV) from hyperglycemic, but not normoglycemic, mice but the source of this synaptic input is unknown. Neurons in the area postrema (AP) and nucleus tractus solitarius (NTS) express high levels of FGF receptors and exert glutamatergic control over the DMV. This study tested the hypothesis that FGF19 increases glutamate release in the DMV by increasing the activity of glutamatergic AP and NTS neurons in hyperglycemic mice. Glutamate photoactivation experiments confirmed that FGF19 increases synaptic glutamate release from AP and NTS neurons that connect to the DMV in hyperglycemic, but not normoglycemic mice. Contrary to expectations, FGF19 produced a mixed effect on intrinsic membrane properties in the NTS with a trend towards inhibition, suggesting that another mechanism was responsible for the observed effects on glutamate release in the DMV. Consistent with the hypothesis, FGF19 increased action potential-dependent glutamate release in the NTS in hyperglycemic mice only. Finally, glutamate photoactivation experiments confirmed that FGF19 increases the activity of glutamatergic AP neurons that project to the NTS in hyperglycemic mice. Together, these results support the hypothesis that FGF19 increases glutamate release from AP and NTS neurons that project to the DMV in hyperglycemic mice. FGF19 therefore modifies the local vago-vagal reflex circuitry at several points. Additionally, since the AP and NTS communicate with several other metabolic regulatory nuclei in the brain, FGF19 in the hindbrain may alter neuroendocrine and behavioral aspects of metabolism, in addition to changes in parasympathetic output.
Collapse
Affiliation(s)
- Jordan B. Wean
- Department of Physiology, College of Medicine, University of Kentucky, Lexington, KY, United States
| | - Bret N. Smith
- Department of Physiology, College of Medicine, University of Kentucky, Lexington, KY, United States
- Department of Neuroscience, College of Medicine, University of Kentucky, Lexington, KY, United States
- *Correspondence: Bret N. Smith,
| |
Collapse
|
12
|
Espinoza L, Boychuk CR. Diabetes, and its treatment, as an effector of autonomic nervous system circuits and its functions. Curr Opin Pharmacol 2020; 54:18-26. [PMID: 32721846 DOI: 10.1016/j.coph.2020.06.006] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2019] [Revised: 06/18/2020] [Accepted: 06/22/2020] [Indexed: 12/24/2022]
Abstract
Diabetes increases the risk of cardiovascular complications, including heart failure, hypertension, and stroke. There is a strong involvement of autonomic dysfunction in individuals with diabetes that exhibit clinical manifestations of cardiovascular diseases (CVD). Still, the mechanisms by which diabetes and its treatments alter autonomic function and subsequently affect cardiovascular complications remain elusive. For this reason, understanding the brainstem circuits involved in sensing metabolic state(s) and enacting autonomic control of the cardiovascular system are important to develop more comprehensive therapies for individuals with diabetes at increased risk for CVD. We review how autonomic nervous system circuits change during these disease states and discuss their potential role in current pharmacotherapies that target diabetic states. Overall, this review proposes that the brainstem circuits provide an integrative sensorimotor network capable of responding to metabolic cues to regulate cardiovascular function and this network is modified by, and in turn affects, diabetes-induced CVD and its treatment.
Collapse
Affiliation(s)
- Liliana Espinoza
- Department of Cellular and Integrative Physiology, Long School of Medicine, University of Texas Health San Antonio, United States
| | - Carie R Boychuk
- Department of Cellular and Integrative Physiology, Long School of Medicine, University of Texas Health San Antonio, United States.
| |
Collapse
|
13
|
Littlejohn EL, Fedorchak S, Boychuk CR. Sex-steroid-dependent plasticity of brain-stem autonomic circuits. Am J Physiol Regul Integr Comp Physiol 2020; 319:R60-R68. [PMID: 32493037 DOI: 10.1152/ajpregu.00357.2019] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
In the central nervous system (CNS), nuclei of the brain stem play a critical role in the integration of peripheral sensory information and the regulation of autonomic output in mammalian physiology. The nucleus tractus solitarius of the brain stem acts as a relay center that receives peripheral sensory input from vagal afferents of the nodose ganglia, integrates information from within the brain stem and higher central centers, and then transmits autonomic efferent output through downstream premotor nuclei, such as the nucleus ambiguus, the dorsal motor nucleus of the vagus, and the rostral ventral lateral medulla. Although there is mounting evidence that sex and sex hormones modulate autonomic physiology at the level of the CNS, the mechanisms and neurocircuitry involved in producing these functional consequences are poorly understood. Of particular interest in this review is the role of estrogen, progesterone, and 5α-reductase-dependent neurosteroid metabolites of progesterone (e.g., allopregnanolone) in the modulation of neurotransmission within brain-stem autonomic neurocircuits. This review will discuss our understanding of the actions and mechanisms of estrogen, progesterone, and neurosteroids at the cellular level of brain-stem nuclei. Understanding the complex interaction between sex hormones and neural signaling plasticity of the autonomic nervous system is essential to elucidating the role of sex in overall physiology and disease.
Collapse
Affiliation(s)
- Erica L Littlejohn
- Department of Cellular and Integrative Physiology, Long School of Medicine, University of Texas Health San Antonio, San Antonio, Texas
| | - Stephanie Fedorchak
- Department of Cellular and Integrative Physiology, Long School of Medicine, University of Texas Health San Antonio, San Antonio, Texas
| | - Carie R Boychuk
- Department of Cellular and Integrative Physiology, Long School of Medicine, University of Texas Health San Antonio, San Antonio, Texas
| |
Collapse
|
14
|
Cowen N, Bhatnagar A. The Potential Role of Activating the ATP-Sensitive Potassium Channel in the Treatment of Hyperphagic Obesity. Genes (Basel) 2020; 11:genes11040450. [PMID: 32326226 PMCID: PMC7230375 DOI: 10.3390/genes11040450] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2020] [Accepted: 04/16/2020] [Indexed: 02/06/2023] Open
Abstract
To evaluate the potential role of ATP-sensitive potassium (KATP) channel activation in the treatment of hyperphagic obesity, a PubMed search was conducted focused on the expression of genes encoding the KATP channel, the response to activating the KATP channel in tissues regulating appetite and the establishment and maintenance of obesity, the evaluation of KATP activators in obese hyperphagic animal models, and clinical studies on syndromic obesity. KATP channel activation is mechanistically involved in the regulation of appetite in the arcuate nucleus; the regulation of hyperinsulinemia, glycemic control, appetite and satiety in the dorsal motor nucleus of vagus; insulin secretion by β-cells; and the synthesis and β-oxidation of fatty acids in adipocytes. KATP channel activators have been evaluated in hyperphagic obese animal models and were shown to reduce hyperphagia, induce fat loss and weight loss in older animals, reduce the accumulation of excess body fat in growing animals, reduce circulating and hepatic lipids, and improve glycemic control. Recent experience with a KATP channel activator in Prader-Willi syndrome is consistent with the therapeutic responses observed in animal models. KATP channel activation, given the breadth of impact and animal model and clinical results, is a viable target in hyperphagic obesity.
Collapse
|
15
|
Georgescu T, Lyons D, Doslikova B, Garcia AP, Marston O, Burke LK, Chianese R, Lam BYH, Yeo GSH, Rochford JJ, Garfield AS, Heisler LK. Neurochemical Characterization of Brainstem Pro-Opiomelanocortin Cells. Endocrinology 2020; 161:bqaa032. [PMID: 32166324 PMCID: PMC7102873 DOI: 10.1210/endocr/bqaa032] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/08/2019] [Accepted: 03/10/2020] [Indexed: 02/08/2023]
Abstract
Genetic research has revealed pro-opiomelanocortin (POMC) to be a fundamental regulator of energy balance and body weight in mammals. Within the brain, POMC is primarily expressed in the arcuate nucleus of the hypothalamus (ARC), while a smaller population exists in the brainstem nucleus of the solitary tract (POMCNTS). We performed a neurochemical characterization of this understudied population of POMC cells using transgenic mice expressing green fluorescent protein (eGFP) under the control of a POMC promoter/enhancer (PomceGFP). Expression of endogenous Pomc mRNA in the nucleus of the solitary tract (NTS) PomceGFP cells was confirmed using fluorescence-activating cell sorting (FACS) followed by quantitative PCR. In situ hybridization histochemistry of endogenous Pomc mRNA and immunohistochemical analysis of eGFP revealed that POMC is primarily localized within the caudal NTS. Neurochemical analysis indicated that POMCNTS is not co-expressed with tyrosine hydroxylase (TH), glucagon-like peptide 1 (GLP-1), cholecystokinin (CCK), brain-derived neurotrophic factor (BDNF), nesfatin, nitric oxide synthase 1 (nNOS), seipin, or choline acetyltransferase (ChAT) cells, whereas 100% of POMCNTS is co-expressed with transcription factor paired-like homeobox2b (Phox2b). We observed that 20% of POMCNTS cells express receptors for adipocyte hormone leptin (LepRbs) using a PomceGFP:LepRbCre:tdTOM double-reporter line. Elevations in endogenous or exogenous leptin levels increased the in vivo activity (c-FOS) of a small subset of POMCNTS cells. Using ex vivo slice electrophysiology, we observed that this effect of leptin on POMCNTS cell activity is postsynaptic. These findings reveal that a subset of POMCNTS cells are responsive to both changes in energy status and the adipocyte hormone leptin, findings of relevance to the neurobiology of obesity.
Collapse
Affiliation(s)
- Teodora Georgescu
- Rowett Institute, University of Aberdeen, Foresterhill, Aberdeen, UK
- Department of Pharmacology, University of Cambridge, Cambridge, UK
- Centre for Neuroendocrinology & Department of Anatomy, University of Otago, Dunedin, New Zealand
| | - David Lyons
- Rowett Institute, University of Aberdeen, Foresterhill, Aberdeen, UK
| | | | - Ana Paula Garcia
- Department of Pharmacology, University of Cambridge, Cambridge, UK
| | - Oliver Marston
- Department of Pharmacology, University of Cambridge, Cambridge, UK
| | - Luke K Burke
- Department of Pharmacology, University of Cambridge, Cambridge, UK
| | | | - Brian Y H Lam
- MRC Metabolic Diseases Unit, University of Cambridge Metabolic Research Laboratories, Wellcome Trust-MRC Institute of Metabolic Science, Addenbrooke’s Hospital, Cambridge, UK
| | - Giles S H Yeo
- MRC Metabolic Diseases Unit, University of Cambridge Metabolic Research Laboratories, Wellcome Trust-MRC Institute of Metabolic Science, Addenbrooke’s Hospital, Cambridge, UK
| | | | | | - Lora K Heisler
- Rowett Institute, University of Aberdeen, Foresterhill, Aberdeen, UK
- Department of Pharmacology, University of Cambridge, Cambridge, UK
| |
Collapse
|
16
|
Abstract
The discovery of leptin changed the view of adipose tissue from that of a passive vessel that stores fat to that of a dynamic endocrine organ that actively regulates behaviour and metabolism. Secreted by adipose tissue, leptin functions as an afferent signal in a negative feedback loop, acting primarily on neurons in the hypothalamus and regulating feeding and many other functions. The leptin endocrine system serves a critical evolutionary function by maintaining the relative constancy of adipose tissue mass, thereby protecting individuals from the risks associated with being too thin (starvation and infertility) or too obese (predation). In this Review, the biology of leptin is summarized, and a conceptual framework is established for studying the pathogenesis of obesity, which, analogously to diabetes, can result from either leptin hyposecretion or leptin resistance. Herein, these two states are distinguished with the terms 'type 1 obesity' and 'type 2 obesity': type 1 obesity describes a subset of obese individuals with low endogenous plasma leptin levels who respond to leptin therapy, whereas type 2 obesity describes most obese individuals, who are leptin resistant but might respond to leptin therapy in combination with other drugs, such as leptin sensitizers.
Collapse
Affiliation(s)
- Jeffrey M Friedman
- Howard Hughes Medical Institute, The Rockefeller University, New York, NY, USA.
| |
Collapse
|
17
|
Leon Mercado L, Caron A, Wang Y, Burton M, Gautron L. Identification of Leptin Receptor-Expressing Cells in the Nodose Ganglion of Male Mice. Endocrinology 2019; 160:1307-1322. [PMID: 30907928 PMCID: PMC6482037 DOI: 10.1210/en.2019-00021] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/08/2019] [Accepted: 03/18/2019] [Indexed: 12/29/2022]
Abstract
Leptin has been proposed to modulate viscerosensory information directly at the level of vagal afferents. In support of this view, broad expression for the leptin receptor (Lepr) has previously been reported in vagal afferents. However, the exact identity and distribution of leptin-sensitive vagal afferents has not been elucidated. Using quantitative PCR, we found that the whole mouse nodose ganglion was predominantly enriched in the short form of Lepr, rather than its long form. Consistent with this observation, the acute administration of leptin did not stimulate JAK-STAT signaling in the nodose ganglion. Using chromogenic in situ hybridization in wild-type mice and several reporter mouse models, we demonstrated that Lepr mRNA was restricted to nonneuronal cells in the epineurium and parenchyma of the nodose ganglion and a subset of vagal afferents, which accounted for only 3% of all neuronal profiles. Double labeling studies further established that Lepr-expressing vagal afferents were Nav1.8-negative fibers that did not supply the peritoneal cavity. Finally, double chromogenic in situ hybridization revealed that many Lepr-expressing neurons coexpressed the angiotensin 1a receptor (At1ar), which is a gene expressed in baroreceptors. Taken together, our data challenge the commonly held view that Lepr is broadly expressed in vagal afferents. Instead, our data suggest that leptin may exert a previously unrecognized role, mainly via its short form, as a direct modulator of a very small group of At1ar-positive vagal fibers.
Collapse
Affiliation(s)
- Luis Leon Mercado
- Division of Hypothalamic Research, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Alexandre Caron
- Division of Hypothalamic Research, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Yibing Wang
- Department of Biochemistry, Utah Southwestern Medical Center at Dallas, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Michael Burton
- School of Behavioral and Brain Sciences, University of Texas at Dallas, Richardson, Texas
| | - Laurent Gautron
- Division of Hypothalamic Research, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas
- Correspondence: Laurent Gautron, PhD, Division of Hypothalamic Research, Department of Internal Medicine, University of Texas Southwestern Medical Center, 5323 Harry Hines Boulevard, Dallas, Texas 75390. E-mail:
| |
Collapse
|
18
|
A hindbrain inhibitory microcircuit mediates vagally-coordinated glucose regulation. Sci Rep 2019; 9:2722. [PMID: 30804396 PMCID: PMC6389891 DOI: 10.1038/s41598-019-39490-x] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2018] [Accepted: 12/14/2018] [Indexed: 02/07/2023] Open
Abstract
Neurons in the brainstem dorsal vagal complex integrate neural and humoral signals to coordinate autonomic output to viscera that regulate a variety of physiological functions, but how this circuitry regulates metabolism is murky. We tested the hypothesis that premotor, GABAergic neurons in the nucleus tractus solitarius (NTS) form a hindbrain micro-circuit with preganglionic parasympathetic motorneurons of the dorsal motor nucleus of the vagus (DMV) that is capable of modulating systemic blood glucose concentration. In vitro, neuronal activation or inhibition using either excitatory or inhibitory designer receptor exclusively activated by designer drugs (DREADDs) constructs expressed in GABAergic NTS neurons increased or decreased, respectively, action potential firing of GABAergic NTS neurons and downstream synaptic inhibition of the DMV. In vivo, DREADD-mediated activation of GABAergic NTS neurons increased systemic blood glucose concentration, whereas DREADD-mediated silencing of these neurons was without effect. The DREADD-induced hyperglycemia was abolished by blocking peripheral muscarinic receptors, consistent with the hypothesis that altered parasympathetic drive mediated the response. This effect was paralleled by elevated serum glucagon and hepatic phosphoenolpyruvate carboxykinase 1 (PEPCK1) expression, without affecting insulin levels or muscle metabolism. Activity in a hindbrain inhibitory microcircuit is sufficient to modulate systemic glucose concentration, independent of insulin secretion or utilization.
Collapse
|
19
|
Murayama S, Yamamoto K, Fujita S, Takei H, Inui T, Ogiso B, Kobayashi M. Extracellular glucose-dependent IPSC enhancement by leptin in fast-spiking to pyramidal neuron connections via JAK2-PI3K pathway in the rat insular cortex. Neuropharmacology 2019; 149:133-148. [PMID: 30772375 DOI: 10.1016/j.neuropharm.2019.02.021] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2019] [Revised: 02/07/2019] [Accepted: 02/13/2019] [Indexed: 01/29/2023]
Abstract
Leptin is produced in the adipocytes and plays a pivotal role in regulation of energy balance by controlling appetite and metabolism. Leptin receptors are widely distributed in the brain, especially in the hypothalamus, hippocampus, and neocortex. The insular cortex (IC) processes gustatory and visceral information, which functionally correlate to feeding behavior. However, it is still an open issue whether and how leptin modulates IC neural activities. Our paired whole-cell patch-clamp recordings using IC slice preparations demonstrated that unitary inhibitory postsynaptic currents (uIPSCs) but not uEPSCs were potentiated by leptin in the connections between pyramidal (PNs) and fast-spiking neurons (FSNs). The leptin-induced increase in uIPSC amplitude was accompanied by a decrease in paired-pulse ratio. Under application of inhibitors of JAK2-PI3K but not MAPK pathway, leptin did not change uIPSC amplitude. Variance-mean analysis revealed that leptin increased the release probability but not the quantal size and the number of release site. These electrophysiological findings suggest that the leptin-induced uIPSC increase is mediated by activation of JAK2-PI3K pathway in presynaptic FSNs. An in vivo optical imaging revealed that leptin application decreased excitatory propagation in IC induced by electrical stimulation of IC. These leptin-induced effects were not observed under the low energy states: low glucose concentration (2.5 mM) in vitro and one-day-fasting condition in vivo. However, leptin enhanced uIPSCs under application of low glucose with an AMPK inhibitor. These results suggest that leptin suppresses IC excitation by facilitating GABA release in FSN→PN connections, which may not occur under a hunger state.
Collapse
Affiliation(s)
- Shota Murayama
- Department of Pharmacology, Nihon University School of Dentistry, 1-8-13 Kanda-Surugadai, Chiyoda-ku, Tokyo, 101-8310, Japan; Department of Endodontics, Nihon University School of Dentistry, 1-8-13 Kanda-Surugadai, Chiyoda-ku, Tokyo, 101-8310, Japan
| | - Kiyofumi Yamamoto
- Department of Pharmacology, Nihon University School of Dentistry, 1-8-13 Kanda-Surugadai, Chiyoda-ku, Tokyo, 101-8310, Japan; Division of Oral and Craniomaxillofacial Research, Dental Research Center, Nihon University School of Dentistry, 1-8-13 Kanda-Surugadai, Chiyoda-ku, Tokyo, 101-8310, Japan
| | - Satoshi Fujita
- Department of Pharmacology, Nihon University School of Dentistry, 1-8-13 Kanda-Surugadai, Chiyoda-ku, Tokyo, 101-8310, Japan; Division of Oral and Craniomaxillofacial Research, Dental Research Center, Nihon University School of Dentistry, 1-8-13 Kanda-Surugadai, Chiyoda-ku, Tokyo, 101-8310, Japan
| | - Hiroki Takei
- Department of Pharmacology, Nihon University School of Dentistry, 1-8-13 Kanda-Surugadai, Chiyoda-ku, Tokyo, 101-8310, Japan; Department of Pedodontics, Nihon University School of Dentistry, 1-8-13 Kanda-Surugadai, Chiyoda-ku, Tokyo, 101-8310, Japan
| | - Tadashi Inui
- Department of Psychology and Program in Neuroscience, Florida State University, Tallahassee, FL, USA
| | - Bunnai Ogiso
- Department of Endodontics, Nihon University School of Dentistry, 1-8-13 Kanda-Surugadai, Chiyoda-ku, Tokyo, 101-8310, Japan
| | - Masayuki Kobayashi
- Department of Pharmacology, Nihon University School of Dentistry, 1-8-13 Kanda-Surugadai, Chiyoda-ku, Tokyo, 101-8310, Japan; Division of Oral and Craniomaxillofacial Research, Dental Research Center, Nihon University School of Dentistry, 1-8-13 Kanda-Surugadai, Chiyoda-ku, Tokyo, 101-8310, Japan; Molecular Dynamics Imaging Unit, RIKEN Centre for Life Science Technologies, 6-7-3 Minatojima-minamimachi, Chuo-ku, Kobe, 650-0047, Japan.
| |
Collapse
|
20
|
Clyburn C, Travagli RA, Browning KN. Acute high-fat diet upregulates glutamatergic signaling in the dorsal motor nucleus of the vagus. Am J Physiol Gastrointest Liver Physiol 2018; 314:G623-G634. [PMID: 29368945 PMCID: PMC6008060 DOI: 10.1152/ajpgi.00395.2017] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Obesity is associated with dysregulation of vagal neurocircuits controlling gastric functions, including food intake and energy balance. In the short term, however, caloric intake is regulated homeostatically although the precise mechanisms responsible are unknown. The present study examined the effects of acute high-fat diet (HFD) on glutamatergic neurotransmission within central vagal neurocircuits and its effects on gastric motility. Sprague-Dawley rats were fed a control or HFD diet (14% or 60% kcal from fat, respectively) for 3-5 days. Whole cell patch-clamp recordings and brainstem application of antagonists were used to assess the effects of acute HFD on glutamatergic transmission to dorsal motor nucleus of the vagus (DMV) neurons and subsequent alterations in gastric tone and motility. After becoming hyperphagic initially, caloric balance was restored after 3 days following HFD exposure. In control rats, the non- N-methyl-d-aspartate (NMDA) receptor antagonist, 6,7-dinitroquinoxaline-2,3-dione (DNQX), but not the NMDA receptor antagonist, amino-5-phosphonopentanoate (AP5), significantly decreased excitatory synaptic currents and action potential firing rate in gastric-projecting DMV neurons. In contrast, both AP5 and DNQX decreased excitatory synaptic transmission and action potential firing in acute HFD neurons. When microinjected into the brainstem, AP5, but not DNQX, decreased gastric motility and tone in acute HFD rats only. These results suggest that acute HFD upregulates NMDA receptor-mediated currents, increasing DMV neuronal excitability and activating the vagal efferent cholinergic pathway, thus increasing gastric tone and motility. Although such neuroplasticity may be a persistent adaptation to the initial exposure to HFD, it may also be an important mechanism in homeostatic regulation of energy balance. NEW & NOTEWORTHY Vagal neurocircuits are critical to the regulation of gastric functions, including satiation and food intake. Acute high-fat diet upregulates glutamatergic signaling within central vagal neurocircuits via activation of N-methyl-d-aspartate receptors, increasing vagal efferent drive to the stomach. Although it is possible that such neuroplasticity is a persistent adaptation to initial exposure to the high-fat diet, it may also play a role in the homeostatic control of feeding.
Collapse
Affiliation(s)
- Courtney Clyburn
- Department of Neural and Behavioral Sciences, Penn State College of Medicine, Hershey, Pennsylvania
| | - R. Alberto Travagli
- Department of Neural and Behavioral Sciences, Penn State College of Medicine, Hershey, Pennsylvania
| | - Kirsteen N. Browning
- Department of Neural and Behavioral Sciences, Penn State College of Medicine, Hershey, Pennsylvania
| |
Collapse
|
21
|
Boychuk CR, Smith KC, Smith BN. Functional and molecular plasticity of γ and α1 GABA A receptor subunits in the dorsal motor nucleus of the vagus after experimentally induced diabetes. J Neurophysiol 2017; 118:2833-2841. [PMID: 28835522 DOI: 10.1152/jn.00085.2017] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2017] [Revised: 08/17/2017] [Accepted: 08/17/2017] [Indexed: 12/13/2022] Open
Abstract
Chronic experimentally induced hyperglycemia augments subunit-specific γ-aminobutyric acid A (GABAA) receptor-mediated inhibition of parasympathetic preganglionic motor neurons in the dorsal motor nucleus of the vagus (DMV). However, the contribution of α1 or γ GABAA receptor subunits, which are ubiquitously expressed on central nervous system neurons, to this elevation in inhibitory tone have not been determined. This study investigated the effect of chronic hyperglycemia/hypoinsulinemia on α1- and γ-subunit-specific GABAA receptor-mediated inhibition using electrophysiological recordings in vitro and quantitative RT-PCR. DMV neurons from streptozotocin-treated mice demonstrated enhancement of both phasic and tonic inhibitory currents in response to application of the α1-subunit-selective GABAA receptor-positive allosteric modulator zolpidem. Responses to low concentrations of the GABAA receptor antagonist gabazine suggested an additional increased contribution of γ-subunit-containing receptors to tonic currents in DMV neurons. Consistent with the functional elevation in α1- and γ-subunit-dependent activity, transcription of both the α1- and γ2-subunits was increased in the dorsal vagal complex of streptozotocin-treated mice. Overall, these findings suggest an increased sensitivity to both zolpidem and gabazine after several days of hyperglycemia/hypoinsulinemia, which could contribute to altered parasympathetic output from DMV neurons in diabetes.NEW & NOTEWORTHY Glutamate and GABA signaling in the dorsal vagal complex is elevated after several days of chronic hyperglycemia in a mouse model of type 1 diabetes. We report persistently enhanced GABAA receptor-mediated responses to the somnolescent zolpidem in preganglionic vagal motor neurons. These results imply a broader impact of chronic hyperglycemia on central vagal function than previously appreciated and reinforce the hypothesis that diabetes effects in the brain can impact regulation of metabolic homeostasis.
Collapse
Affiliation(s)
- Carie R Boychuk
- Department of Physiology, University of Kentucky College of Medicine, Lexington, Kentucky
| | - Katalin C Smith
- Department of Physiology, University of Kentucky College of Medicine, Lexington, Kentucky
| | - Bret N Smith
- Department of Physiology, University of Kentucky College of Medicine, Lexington, Kentucky
| |
Collapse
|
22
|
Bruce KD, Zsombok A, Eckel RH. Lipid Processing in the Brain: A Key Regulator of Systemic Metabolism. Front Endocrinol (Lausanne) 2017; 8:60. [PMID: 28421037 PMCID: PMC5378716 DOI: 10.3389/fendo.2017.00060] [Citation(s) in RCA: 169] [Impact Index Per Article: 21.1] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/02/2017] [Accepted: 03/17/2017] [Indexed: 12/25/2022] Open
Abstract
Metabolic disorders, particularly aberrations in lipid homeostasis, such as obesity, type 2 diabetes mellitus, and hypertriglyceridemia often manifest together as the metabolic syndrome (MetS). Despite major advances in our understanding of the pathogenesis of these disorders, the prevalence of the MetS continues to rise. It is becoming increasingly apparent that intermediary metabolism within the central nervous system is a major contributor to the regulation of systemic metabolism. In particular, lipid metabolism within the brain is tightly regulated to maintain neuronal structure and function and may signal nutrient status to modulate metabolism in key peripheral tissues such as the liver. There is now a growing body of evidence to suggest that fatty acid (FA) sensing in hypothalamic neurons via accumulation of FAs or FA metabolites may signal nutritional sufficiency and may decrease hepatic glucose production, lipogenesis, and VLDL-TG secretion. In addition, recent studies have highlighted the existence of liver-related neurons that have the potential to direct such signals through parasympathetic and sympathetic nervous system activity. However, to date whether these liver-related neurons are FA sensitive remain to be determined. The findings discussed in this review underscore the importance of the autonomic nervous system in the regulation of systemic metabolism and highlight the need for further research to determine the key features of FA neurons, which may serve as novel therapeutic targets for the treatment of metabolic disorders.
Collapse
Affiliation(s)
- Kimberley D. Bruce
- University of Colorado School of Medicine, Division of Endocrinology, Metabolism and Diabetes, Aurora, CO, USA
- *Correspondence: Kimberley D. Bruce,
| | - Andrea Zsombok
- Department of Physiology, School of Medicine, Tulane University, New Orleans, LA, USA
| | - Robert H. Eckel
- University of Colorado School of Medicine, Division of Endocrinology, Metabolism and Diabetes, Aurora, CO, USA
| |
Collapse
|
23
|
Sun J, Gao Y, Yao T, Huang Y, He Z, Kong X, Yu KJ, Wang RT, Guo H, Yan J, Chang Y, Chen H, Scherer PE, Liu T, Williams KW. Adiponectin potentiates the acute effects of leptin in arcuate Pomc neurons. Mol Metab 2016; 5:882-891. [PMID: 27689001 PMCID: PMC5034606 DOI: 10.1016/j.molmet.2016.08.007] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/19/2016] [Revised: 08/08/2016] [Accepted: 08/10/2016] [Indexed: 01/06/2023] Open
Abstract
Objective Adiponectin receptors (AdipoRs) are located on neurons of the hypothalamus involved in metabolic regulation – including arcuate proopiomelanocortin (Pomc) and Neuropeptide Y/Agouti-related peptide (NPY/AgRP) neurons. AdipoRs play a critical role in regulating glucose and fatty acid metabolism by initiating several signaling cascades overlapping with Leptin receptors (LepRs). However, the mechanism by which adiponectin regulates cellular activity in the brain remains undefined. Methods In order to resolve this issue, we utilized neuron-specific transgenic mouse models to identify Pomc and NPY/AgRP neurons which express LepRs for patch-clamp electrophysiology experiments. Results We found that leptin and adiponectin synergistically activated melanocortin neurons in the arcuate nucleus. Conversely, NPY/AgRP neurons were inhibited in response to adiponectin. The adiponectin-induced depolarization of arcuate Pomc neurons occurred via activation of Phosphoinositide-3-kinase (PI3K) signaling, independent of 5′ AMP-activated protein kinase (AMPK) activity. Adiponectin also activated melanocortin neurons at various physiological glucose levels. Conclusions Our results demonstrate a requirement for PI3K signaling in the acute adiponectin-induced effects on the cellular activity of arcuate melanocortin neurons. Moreover, these data provide evidence for PI3K as a substrate for both leptin and adiponectin to regulate energy balance and glucose metabolism via melanocortin activity. Adiponectin activates arcuate Pomc neurons. Adiponectin-induced activation of Pomc neurons requires PI3K (independent of AMPK). Adiponectin inhibits adjacent NPY/AgRP neurons (disinhibiting arcuate Pomc neurons). Leptin potentiates the effects of adiponectin arcuate Pomc neurons.
Collapse
Affiliation(s)
- Jia Sun
- Department of Endocrinology, Zhujiang Hospital, Southern Medical University, Guangzhou, China; Division of Hypothalamic Research, The University of Texas Southwestern Medical Center at Dallas, Dallas, TX, USA
| | - Yong Gao
- National Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China; Division of Hypothalamic Research, The University of Texas Southwestern Medical Center at Dallas, Dallas, TX, USA
| | - Ting Yao
- Department of Physiology and Pathophysiology, Xi'an Jiaotong University School of Medicine, Xi'an, Shaanxi, China; Division of Hypothalamic Research, The University of Texas Southwestern Medical Center at Dallas, Dallas, TX, USA
| | - Yiru Huang
- Department of Neurosurgery, Zhujiang Hospital, Southern Medical University, Guangzhou, China; Division of Hypothalamic Research, The University of Texas Southwestern Medical Center at Dallas, Dallas, TX, USA
| | - Zhenyan He
- Department of Neurosurgery, Zhujiang Hospital, Southern Medical University, Guangzhou, China; Division of Hypothalamic Research, The University of Texas Southwestern Medical Center at Dallas, Dallas, TX, USA
| | - Xingxing Kong
- Division of Endocrinology, Beth Israel Deaconess Medical Center and Harvard Medical School, Harvard University, Boston, MA, 02115, USA
| | - Kai-Jiang Yu
- Department of Intensive Care Unit, The Third Affiliated Hospital, Harbin Medical University, No. 150 Haping St, Nangang District, Harbin, 150081, China
| | - Rui-Tao Wang
- Department of Intensive Care Unit, The Third Affiliated Hospital, Harbin Medical University, No. 150 Haping St, Nangang District, Harbin, 150081, China
| | - Hongbo Guo
- Department of Neurosurgery, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Jianqun Yan
- Department of Physiology and Pathophysiology, Xi'an Jiaotong University School of Medicine, Xi'an, Shaanxi, China
| | - Yongsheng Chang
- National Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Hong Chen
- Department of Endocrinology, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Philipp E Scherer
- Touchstone Diabetes Center, Department of Internal Medicine, The University of Texas Southwestern Medical Center at Dallas, Dallas, TX, 75390, USA
| | - Tiemin Liu
- Department of Intensive Care Unit, The Third Affiliated Hospital, Harbin Medical University, No. 150 Haping St, Nangang District, Harbin, 150081, China; Division of Hypothalamic Research, The University of Texas Southwestern Medical Center at Dallas, Dallas, TX, USA
| | - Kevin W Williams
- Division of Hypothalamic Research, The University of Texas Southwestern Medical Center at Dallas, Dallas, TX, USA.
| |
Collapse
|
24
|
Boychuk CR, Smith BN. Glutamatergic drive facilitates synaptic inhibition of dorsal vagal motor neurons after experimentally induced diabetes in mice. J Neurophysiol 2016; 116:1498-506. [PMID: 27385796 DOI: 10.1152/jn.00325.2016] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2016] [Accepted: 07/01/2016] [Indexed: 12/31/2022] Open
Abstract
The role of central regulatory circuits in modulating diabetes-associated glucose dysregulation has only recently been under rigorous investigation. One brain region of interest is the dorsal motor nucleus of the vagus (DMV), which contains preganglionic parasympathetic motor neurons that regulate subdiaphragmatic visceral function. Previous research has demonstrated that glutamatergic and GABAergic neurotransmission are independently remodeled after chronic hyperglycemia/hypoinsulinemia. However, glutamatergic circuitry within the dorsal brain stem impinges on GABAergic regulation of the DMV. The present study investigated the role of glutamatergic neurotransmission in synaptic GABAergic control of DMV neurons after streptozotocin (STZ)-induced hyperglycemia/hypoinsulinemia by using electrophysiological recordings in vitro. The frequency of spontaneous inhibitory postsynaptic currents (sIPSCs) was elevated in DMV neurons from STZ-treated mice. The effect was abolished in the presence of the ionotropic glutamate receptor blocker kynurenic acid or the sodium channel blocker tetrodotoxin, suggesting that after STZ-induced hyperglycemia/hypoinsulinemia, increased glutamatergic receptor activity occurs at a soma-dendritic location on local GABA neurons projecting to the DMV. Although sIPSCs in DMV neurons normally demonstrated considerable amplitude variability, this variability was significantly increased after STZ-induced hyperglycemia/hypoinsulinemia. The elevated amplitude variability was not related to changes in quantal release, but rather correlated with significantly elevated frequency of sIPSCs in these mice. Taken together, these findings suggest that GABAergic regulation of central vagal circuitry responsible for the regulation of energy homeostasis undergoes complex functional reorganization after several days of hyperglycemia/hypoinsulinemia, including both glutamate-dependent and -independent forms of plasticity.
Collapse
Affiliation(s)
- Carie R Boychuk
- Department of Physiology, University of Kentucky College of Medicine, Lexington, Kentucky
| | - Bret N Smith
- Department of Physiology, University of Kentucky College of Medicine, Lexington, Kentucky
| |
Collapse
|
25
|
Sohn JW, Oh Y, Kim KW, Lee S, Williams KW, Elmquist JK. Leptin and insulin engage specific PI3K subunits in hypothalamic SF1 neurons. Mol Metab 2016; 5:669-679. [PMID: 27656404 PMCID: PMC5021675 DOI: 10.1016/j.molmet.2016.06.004] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/22/2016] [Revised: 06/07/2016] [Accepted: 06/08/2016] [Indexed: 11/21/2022] Open
Abstract
Objective The ventromedial hypothalamic nucleus (VMH) regulates energy balance and glucose homeostasis. Leptin and insulin exert metabolic effects via their cognate receptors expressed by the steroidogenic factor 1 (SF1) neurons within the VMH. However, detailed cellular mechanisms involved in the regulation of these neurons by leptin and insulin remain to be identified. Methods We utilized genetically-modified mouse models and performed patch-clamp electrophysiology experiments to resolve this issue. Results We identified distinct populations of leptin-activated and leptin-inhibited SF1 neurons. In contrast, insulin uniformly inhibited SF1 neurons. Notably, we found that leptin-activated, leptin-inhibited, and insulin-inhibited SF1 neurons are distinct subpopulations within the VMH. Leptin depolarization of SF1 neuron also required the PI3K p110β catalytic subunit. This effect was mediated by the putative transient receptor potential C (TRPC) channel. On the other hand, hyperpolarizing responses of SF1 neurons by leptin and insulin required either of the p110α or p110β catalytic subunits, and were mediated by the putative ATP-sensitive K+ (KATP) channel. Conclusions Our results demonstrate that specific PI3K catalytic subunits are responsible for the acute effects of leptin and insulin on VMH SF1 neurons, and provide insights into the cellular mechanisms of leptin and insulin action on VMH SF1 neurons that regulate energy balance and glucose homeostasis. Leptin recruits p110β/TRPC channels to depolarize/activate SF1 neurons. Leptin recruits p110α/p110β/KATP channels to hyperpolarize/inhibit SF1 neurons. Insulin recruits p110α/p110β/KATP channels to hyperpolarize/inhibit SF1 neurons. Acute leptin and insulin responses are segregated to distinct subsets of VMH SF1 neurons.
Collapse
Affiliation(s)
- Jong-Woo Sohn
- Division of Hypothalamic Research, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA; Department of Biological Sciences, Korea Advanced Institute of Science and Technology, Daejeon, 34141, South Korea.
| | - Youjin Oh
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology, Daejeon, 34141, South Korea
| | - Ki Woo Kim
- Division of Hypothalamic Research, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA; Department of Pharmacology, Yonsei University Wonju College of Medicine, Wonju, 26426, South Korea
| | - Syann Lee
- Division of Hypothalamic Research, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Kevin W Williams
- Division of Hypothalamic Research, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA.
| | - Joel K Elmquist
- Division of Hypothalamic Research, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA; Department of Pharmacology, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| |
Collapse
|
26
|
Anwar IJ, Miyata K, Zsombok A. Brain stem as a target site for the metabolic side effects of olanzapine. J Neurophysiol 2015; 115:1389-98. [PMID: 26719086 DOI: 10.1152/jn.00387.2015] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2015] [Accepted: 12/30/2015] [Indexed: 01/27/2023] Open
Abstract
Olanzapine, an atypical antipsychotic, is widely prescribed for the treatment of schizophrenia and bipolar disorder despite causing undesirable metabolic side effects. A variety of mechanisms and brain sites have been proposed as contributors to the side effects; however, the role of the dorsal motor nucleus of the vagus nerve (DMV), which plays a crucial role in the regulation of subdiaphragmatic organs and thus governs energy and glucose homeostasis, is largely unknown. Identifying the effect of olanzapine on the excitability of DMV neurons in both sexes is thus crucial to understanding possible underlying mechanisms. Whole cell patch-clamp electrophysiological recordings were conducted in stomach- and liver-related DMV neurons identified with retrograde viral tracers and in random DMV neurons. The effect of olanzapine on the neuronal excitability of DMV neurons both in male and female mice was established. Our data demonstrate that olanzapine hyperpolarizes the DMV neurons in both sexes and this effect is reversible. The hyperpolarization is associated with decreased firing rate and input resistance. Olanzapine also decreases the excitability of a subset of stomach- and liver-related DMV neurons. Our study demonstrates that olanzapine has a powerful effect on DMV neurons in both sexes, indicating its ability to reduce vagal output to the subdiaphragmatic organs, which likely contributes to the metabolic side effects observed in both humans and experimental models. These findings suggest that the metabolic side effects of olanzapine may partially originate in the DMV.
Collapse
Affiliation(s)
- Imran J Anwar
- Department of Physiology, School of Medicine, Tulane University, New Orleans, Louisiana
| | - Kayoko Miyata
- Department of Physiology, School of Medicine, Tulane University, New Orleans, Louisiana
| | - Andrea Zsombok
- Department of Physiology, School of Medicine, Tulane University, New Orleans, Louisiana
| |
Collapse
|
27
|
Wilson JL, Enriori PJ. A talk between fat tissue, gut, pancreas and brain to control body weight. Mol Cell Endocrinol 2015; 418 Pt 2:108-19. [PMID: 26316427 DOI: 10.1016/j.mce.2015.08.022] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/20/2015] [Revised: 08/19/2015] [Accepted: 08/20/2015] [Indexed: 12/18/2022]
Abstract
The incidence of obesity and its related disorders are increasing at a rate of pandemic proportions. Understanding the mechanisms behind the maintenance of energy balance is fundamental in developing treatments for clinical syndromes including obesity and diabetes. A neural network located in the nucleus of the solitary tract-area postrema complex in the hindbrain and the hypothalamus in the forebrain has long been implicated in the control of energy balance. In the hypothalamus this central neuronal network consists of small populations of nuclei with distinct functions such as the arcuate nucleus (ARH), the paraventricular nuclei of the hypothalamus (PVH), the dorsomedial (DMH), the ventromedial (VMH) and the lateral hypothalamus (LH). These hypothalamic areas form interconnected neuronal circuits that respond to fluctuations in energy status by altering the expression of neuropeptides, leading to changes in energy intake and expenditure. Regulation of these hypothalamic nuclei involves the actions of orexigenic peptides (ie ghrelin), which act to stimulate energy intake and decrease energy expenditure, and anorexigenic peptides (ie. leptin and insulin), which act to reduce energy intake and stimulate energy expenditure. Here we review the role of the ARH, DMH and PVH in the control of energy homeostasis and how recent advances in research technologies (Cre-loxP technology, optogenetics and pharmacogenetics) have shed light on the role of these hypothalamic nuclei in the control of energy balance. Such novel findings include the implication of ARH POMC and AgRP neurons in the browning of white adipose tissue to regulate energy expenditure as well as the likely existence of divergent hypothalamic pathways in the DMH and PVH in the control of food intake and energy expenditure.
Collapse
Affiliation(s)
- Jenny L Wilson
- Department of Physiology, Monash Obesity & Diabetes Institute, Monash University, Clayton, Victoria 3800, Australia
| | - Pablo J Enriori
- Department of Physiology, Monash Obesity & Diabetes Institute, Monash University, Clayton, Victoria 3800, Australia.
| |
Collapse
|
28
|
Enhanced NMDA receptor-mediated modulation of excitatory neurotransmission in the dorsal vagal complex of streptozotocin-treated, chronically hyperglycemic mice. PLoS One 2015; 10:e0121022. [PMID: 25799386 PMCID: PMC4370733 DOI: 10.1371/journal.pone.0121022] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2014] [Accepted: 02/09/2015] [Indexed: 11/19/2022] Open
Abstract
A variety of metabolic disorders, including complications experienced by diabetic patients, have been linked to altered neural activity in the dorsal vagal complex. This study tested the hypothesis that augmentation of N-Methyl-D-Aspartate (NMDA) receptor-mediated responses in the vagal complex contributes to increased glutamate release in the dorsal motor nucleus of the vagus nerve (DMV) in mice with streptozotocin-induced chronic hyperglycemia (i.e., hyperglycemic mice), a model of type 1 diabetes. Antagonism of NMDA receptors with AP-5 (100 μM) suppressed sEPSC frequency in vagal motor neurons recorded in vitro, confirming that constitutively active NMDA receptors regulate glutamate release in the DMV. There was a greater relative effect of NMDA receptor antagonism in hyperglycemic mice, suggesting that augmented NMDA effects occur in neurons presynaptic to the DMV. Effects of NMDA receptor blockade on mEPSC frequency were equivalent in control and diabetic mice, suggesting that differential effects on glutamate release were due to altered NMDA function in the soma-dendritic membrane of intact afferent neurons. Application of NMDA (300 μM) resulted in greater inward current and current density in NTS neurons recorded from hyperglycemic than control mice, particularly in glutamatergic NTS neurons identified by single-cell RT-PCR for VGLUT2. Overall expression of NR1 protein and message in the dorsal vagal complex were not different between the two groups. Enhanced postsynaptic NMDA responsiveness of glutamatergic NTS neurons is consistent with tonically-increased glutamate release in the DMV in mice with chronic hyperglycemia. Functional augmentation of NMDA-mediated responses may serve as a physiological counter-regulatory mechanism to control pathological disturbances of homeostatic autonomic function in type 1 diabetes.
Collapse
|
29
|
Xu H, Boychuk JA, Boychuk CR, Uteshev VV, Smith BN. Nicotine enhances inhibition of mouse vagal motor neurons by modulating excitability of premotor GABAergic neurons in the nucleus tractus solitarii. J Neurophysiol 2014; 113:1165-74. [PMID: 25429117 DOI: 10.1152/jn.00614.2014] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
The caudal nucleus of the solitary tract (NTS) serves as the site of the first synapse for visceral sensory inputs to the central nervous system. The NTS sends functional projections to multiple brain nuclei, with gastric-related projections primarily targeting the dorsal motor nucleus of the vagus (DMV). Previous studies have demonstrated that the majority of caudal NTS neurons that project to the DMV respond robustly to nicotine and express nicotinic acetylcholine receptors (nAChRs). However, the cytochemical identity and relationship with specific viscera of DMV-projecting, nicotine-responsive caudal NTS neurons have not been determined. The present study used transgenic mice that express enhanced green fluorescent protein (EGFP) under a GAD67 promoter in a subset of GABAergic neurons, in vivo retrograde pseudorabies viral labeling to identify gastric-related vagal complex neurons, and patch-clamp electrophysiology in acute brain stem slices to test the hypothesis that gastric-related and GABAergic inhibitory synaptic input to the DMV from the caudal NTS is under a robust modulatory control by nAChRs. Our results suggest that activation of nAChRs in the caudal NTS, but not DMV, potentiates GABAergic, but not glutamatergic, input to the DMV. Gastric-related caudal NTS and DMV neurons are directly involved in this nicotine-sensitive circuitry. Understanding the central patterns of nicotinic modulation of visceral sensory-motor circuitry may help develop therapeutic interventions to restore autonomic homeostasis in patients with autonomic impairments.
Collapse
Affiliation(s)
- Hong Xu
- Department of Physiology, College of Medicine, University of Kentucky, Lexington, Kentucky; and
| | - Jeffery A Boychuk
- Department of Physiology, College of Medicine, University of Kentucky, Lexington, Kentucky; and
| | - Carie R Boychuk
- Department of Physiology, College of Medicine, University of Kentucky, Lexington, Kentucky; and
| | - Victor V Uteshev
- Department of Pharmacology and Neuroscience, University of North Texas Health Science Center, Fort Worth, Texas
| | - Bret N Smith
- Department of Physiology, College of Medicine, University of Kentucky, Lexington, Kentucky; and
| |
Collapse
|
30
|
Zsombok A, Jiang Y, Gao H, Anwar IJ, Rezai-Zadeh K, Enix CL, Münzberg H, Derbenev AV. Regulation of leptin receptor-expressing neurons in the brainstem by TRPV1. Physiol Rep 2014; 2:2/9/e12160. [PMID: 25263209 PMCID: PMC4270226 DOI: 10.14814/phy2.12160] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
The central nervous system plays a critical role in the regulation of feeding behavior and whole‐body metabolism via controlling the autonomic output to the visceral organs. Activity of the parasympathetic neurons in the dorsal motor nucleus of the vagus (DMV) determines the vagal tone and thereby modulates the function of the subdiaphragmatic organs. Leptin is highly involved in the regulation of food intake and alters neuronal excitability of brainstem neurons. Transient receptor potential vanilloid type 1 (TRPV1) has also been shown to increase neurotransmission in the brainstem and we tested the hypothesis that TRPV1 regulates presynaptic neurotransmitter release to leptin receptor‐expressing (LepRbEGFP) DMV neurons. Whole‐cell patch‐clamp recordings were performed to determine the effect of TRPV1 activation on excitatory and inhibitory postsynaptic currents (EPSC, IPSC) of LepRbEGFP neurons in the DMV. Capsaicin, a TRPV1 agonist increased the frequency of miniature EPSCs in 50% of LepRbEGFP neurons without altering the frequency of miniature IPSCs in the DMV. Stomach‐projecting LepRbEGFP neurons were identified in the DMV using the transsynaptic retrograde viral tracer PRV‐614. Activation of TRPV1 increased the frequency of mEPSC in ~50% of stomach‐related LepRbEGFP DMV neurons. These data demonstrate that TRPV1 increases excitatory neurotransmission to a subpopulation of LepRbEGFP DMV neurons via presynaptic mechanisms and suggest a potential interaction between TRPV1 and leptin signaling in the DMV. e12160 Our data demonstrate that TRPV1 is involved in the regulation of a subpopulation of leptin receptor‐expressing neurons in the dorsal motor nucleus of the vagus via presynaptic mechanisms.
Collapse
Affiliation(s)
- Andrea Zsombok
- Department of Physiology, School of Medicine, Tulane University, New Orleans, Louisiana Neuroscience Program, School of Science and Engineering, Tulane University, New Orleans, Louisiana
| | - Yanyan Jiang
- Department of Physiology, School of Medicine, Tulane University, New Orleans, Louisiana Neuroscience Program, School of Science and Engineering, Tulane University, New Orleans, Louisiana
| | - Hong Gao
- Department of Physiology, School of Medicine, Tulane University, New Orleans, Louisiana
| | - Imran J Anwar
- Department of Physiology, School of Medicine, Tulane University, New Orleans, Louisiana
| | - Kavon Rezai-Zadeh
- Central Leptin Signaling, Pennington Biomedical Research Center, LSU System, Baton Rouge, Louisiana
| | - Courtney L Enix
- Department of Physiology, School of Medicine, Tulane University, New Orleans, Louisiana
| | - Heike Münzberg
- Central Leptin Signaling, Pennington Biomedical Research Center, LSU System, Baton Rouge, Louisiana
| | - Andrei V Derbenev
- Department of Physiology, School of Medicine, Tulane University, New Orleans, Louisiana Neuroscience Program, School of Science and Engineering, Tulane University, New Orleans, Louisiana
| |
Collapse
|
31
|
Blake CB, Smith BN. cAMP-dependent insulin modulation of synaptic inhibition in neurons of the dorsal motor nucleus of the vagus is altered in diabetic mice. Am J Physiol Regul Integr Comp Physiol 2014; 307:R711-20. [PMID: 24990858 DOI: 10.1152/ajpregu.00138.2014] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
Pathologies in which insulin is dysregulated, including diabetes, can disrupt central vagal circuitry, leading to gastrointestinal and other autonomic dysfunction. Insulin affects whole body metabolism through central mechanisms and is transported into the brain stem dorsal motor nucleus of the vagus (DMV) and nucleus tractus solitarius (NTS), which mediate parasympathetic visceral regulation. The NTS receives viscerosensory vagal input and projects heavily to the DMV, which supplies parasympathetic vagal motor output. Normally, insulin inhibits synaptic excitation of DMV neurons, with no effect on synaptic inhibition. Modulation of synaptic inhibition in DMV, however, is often sensitive to cAMP-dependent mechanisms. We hypothesized that an effect of insulin on GABAergic synaptic transmission may be uncovered by elevating resting cAMP levels in GABAergic terminals. We used whole cell patch-clamp recordings in brain stem slices from control and diabetic mice to identify insulin effects on inhibitory neurotransmission in the DMV in the presence of forskolin to elevate cAMP levels. In the presence of forskolin, insulin decreased the frequency of inhibitory postsynaptic currents (IPSCs) and the paired-pulse ratio of evoked IPSCs in DMV neurons from control mice. This effect was blocked by brefeldin-A, a Golgi-disrupting agent, or indinavir, a GLUT4 blocker, indicating that protein trafficking and glucose transport were involved. In streptozotocin-treated, diabetic mice, insulin did not affect IPSCs in DMV neurons in the presence of forskolin. Results suggest an impairment of cAMP-induced insulin effects on GABA release in the DMV, which likely involves disrupted protein trafficking in diabetic mice. These findings provide insight into mechanisms underlying vagal dysregulation associated with diabetes.
Collapse
Affiliation(s)
- Camille B Blake
- Department of Physiology, University of Kentucky College of Medicine, Lexington, Kentucky
| | - Bret N Smith
- Department of Physiology, University of Kentucky College of Medicine, Lexington, Kentucky
| |
Collapse
|
32
|
Bassi M, Furuya WI, Menani JV, Colombari DSA, do Carmo JM, da Silva AA, Hall JE, Moreira TS, Wenker IC, Mulkey DK, Colombari E. Leptin into the ventrolateral medulla facilitates chemorespiratory response in leptin-deficient (ob/ob) mice. Acta Physiol (Oxf) 2014; 211:240-8. [PMID: 24521430 PMCID: PMC4365783 DOI: 10.1111/apha.12257] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2013] [Revised: 01/18/2014] [Accepted: 02/07/2014] [Indexed: 12/27/2022]
Abstract
AIM Leptin, an adipocyte-derived hormone, is suggested to participate in the central control of breathing. We hypothesized that leptin may facilitate ventilatory responses to chemoreflex activation by acting on respiratory nuclei of the ventrolateral medulla. The baseline ventilation and the ventilatory responses to CO2 were evaluated before and after daily injections of leptin into the retrotrapezoid nucleus/parafacial respiratory group (RTN/pFRG) for 3 days in obese leptin-deficient (ob/ob) mice. METHODS Male ob/ob mice (40-45 g, n = 7 per group) received daily microinjections of vehicle or leptin (1 μg per 100 nL) for 3 days into the RTN/pFRG. Respiratory responses to CO2 were measured by whole-body plethysmography. RESULTS Unilateral microinjection of leptin into the RTN/pFRG in ob/ob mice increased baseline ventilation (VE ) from 1447 ± 96 to 2405 ± 174 mL min(-1) kg(-1) by increasing tidal volume (VT ) from 6.4 ± 0.4 to 9.1 ± 0.8 mL kg(-1) (P < 0.05). Leptin also enhanced ventilatory responses to 7% CO2 (Δ = 2172 ± 218 mL min(-1) kg(-1) , vs. control: Δ = 1255 ± 105 mL min(-1) kg(-1) ), which was also due to increased VT (Δ = 4.71 ± 0.51 mL kg(-1) , vs. control: Δ = 2.27 ± 0.20 mL kg(-1) ), without changes in respiratory frequency. Leptin treatment into the RTN/pFRG or into the surrounding areas decreased food intake (83 and 70%, respectively), without significantly changing body weight. CONCLUSION The present results suggest that leptin acting in the respiratory nuclei of the ventrolateral medulla improves baseline VE and VT and facilitates respiratory responses to hypercapnia in ob/ob mice.
Collapse
Affiliation(s)
- M. Bassi
- Department of Physiology and Pathology, School of Dentistry, São Paulo State University (UNESP), Araraquara, SP, Brazil
| | - W. I. Furuya
- Department of Physiology and Pathology, School of Dentistry, São Paulo State University (UNESP), Araraquara, SP, Brazil
| | - J. V. Menani
- Department of Physiology and Pathology, School of Dentistry, São Paulo State University (UNESP), Araraquara, SP, Brazil
| | - D. S. A. Colombari
- Department of Physiology and Pathology, School of Dentistry, São Paulo State University (UNESP), Araraquara, SP, Brazil
| | - J. M. do Carmo
- Department of Physiology and Biophysics, University of Mississippi Medical Center, Jackson, MS, USA
| | - A. A. da Silva
- Department of Physiology and Biophysics, University of Mississippi Medical Center, Jackson, MS, USA
| | - J. E. Hall
- Department of Physiology and Biophysics, University of Mississippi Medical Center, Jackson, MS, USA
| | - T. S. Moreira
- Department of Physiology and Biophysics, Institute of Biomedical Science, University of São Paulo (USP), São Paulo, SP, Brazil
| | - I. C. Wenker
- Department of Physiology and Neurobiology, University of Connecticut, Storrs, CT, USA
| | - D. K. Mulkey
- Department of Physiology and Neurobiology, University of Connecticut, Storrs, CT, USA
| | - E. Colombari
- Department of Physiology and Pathology, School of Dentistry, São Paulo State University (UNESP), Araraquara, SP, Brazil
| |
Collapse
|
33
|
Jiang Y, Zsombok A. Regulation of neurons in the dorsal motor nucleus of the vagus by SIRT1. Front Neurosci 2014; 7:270. [PMID: 24454277 PMCID: PMC3887315 DOI: 10.3389/fnins.2013.00270] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2013] [Accepted: 12/20/2013] [Indexed: 12/29/2022] Open
Abstract
Neurons in the dorsal motor nucleus of the vagus (DMV) play a critical role in the regulation of autonomic functions. Previous studies indicated that central activation of sirtuin 1 (SIRT1) has beneficial effects on homeostasis, most likely via modulation of the autonomic output. Sirtuins are NAD+-dependent deacetylases and have been associated with longevity. SIRT1 is one of the best-characterized sirtuins expressed in mammals, and may be involved in the regulation of metabolism. Resveratrol, a SIRT1 activator reduced hyperglycemia likely through activation of vagal output; however, the cellular mechanisms of action have not been determined. In this study, whole-cell patch-clamp electrophysiology on acute brainstem slices was used to test the hypothesis that activation of SIRT1 with resveratrol enhances neurotransmission in DMV neurons. Application of resveratrol increased the frequency of spontaneous excitatory postsynaptic currents (sEPSC). This effect was KATP channel-dependent and was prevented with pre-application of SIRT1 inhibitor, EX527. Resveratrol also increased miniature EPSC (mEPSC) frequency without change in amplitude. Furthermore, our data demonstrated that resveratrol regulates excitatory neurotransmission in a PI3 kinase-dependent manner, since wortmannin, a PI3K inhibitor prevented the increase of mEPSC frequency caused by resveratrol. In conclusion, our data demonstrate that resveratrol via SIRT1 increases excitatory neurotransmission to DMV neurons. These observations suggest that activation of SIRT1 may regulate the function of subdiaphragmatic organs through controlling the activity of parasympathetic DMV neurons.
Collapse
Affiliation(s)
- Yanyan Jiang
- Neuroscience Program, School of Science and Engineering, Tulane University New Orleans, LA, USA ; Department of Physiology, School of Medicine, Tulane University New Orleans, LA, USA
| | - Andrea Zsombok
- Neuroscience Program, School of Science and Engineering, Tulane University New Orleans, LA, USA ; Department of Physiology, School of Medicine, Tulane University New Orleans, LA, USA
| |
Collapse
|
34
|
Derbenev AV, Smith BN. Dexamethasone rapidly increases GABA release in the dorsal motor nucleus of the vagus via retrograde messenger-mediated enhancement of TRPV1 activity. PLoS One 2013; 8:e70505. [PMID: 23936221 PMCID: PMC3728308 DOI: 10.1371/journal.pone.0070505] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2013] [Accepted: 06/23/2013] [Indexed: 11/18/2022] Open
Abstract
Glucocorticoids influence vagal parasympathetic output to the viscera via mechanisms that include modulation of neural circuitry in the dorsal vagal complex, a principal autonomic regulatory center. Glucocorticoids can modulate synaptic neurotransmitter release elsewhere in the brain by inducing release of retrograde signalling molecules. We tested the hypothesis that the glucocorticoid agonist dexamethasone (DEX) modulates GABA release in the rat dorsal motor nucleus of the vagus (DMV). Whole-cell patch-clamp recordings revealed that DEX (1-10 µM) rapidly (i.e. within three minutes) increased the frequency of tetrodotoxin-resistant, miniature IPSCs (mIPSCs) in 67% of DMV neurons recorded in acutely prepared slices. Glutamate-mediated mEPSCs were also enhanced by DEX (10 µM), and blockade of ionotropic glutamate receptors reduced the DEX effect on mIPSC frequency. Antagonists of type I or II corticosteroid receptors blocked the effect of DEX on mIPSCs. The effect was mimicked by application of the membrane-impermeant BSA-conjugated DEX, and intracellular blockade of G protein function with GDP βS in the recorded cell prevented the effect of DEX. The enhancement of GABA release was blocked by the TRPV1 antagonists, 5’-iodoresiniferatoxin or capsazepine, but was not altered by the cannabinoid type 1 receptor antagonist AM251. The DEX effect was prevented by blocking fatty acid amide hydrolysis or by inhibiting anandamide transport, implicating involvement of the endocannabinoid system in the response. These findings indicate that DEX induces an enhancement of GABA release in the DMV, which is mediated by activation of TRPV1 receptors on afferent terminals. The effect is likely induced by anandamide or other ‘endovanilloid’, suggesting activation of a local retrograde signal originating from DMV neurons to enhance synaptic inhibition locally in response to glucocorticoids.
Collapse
Affiliation(s)
- Andrei V. Derbenev
- Department of Physiology, University of Kentucky College of Medicine, Lexington, Kentucky, United States of America
- Department of Cell and Molecular Biology, Tulane University, New Orleans, Louisiana, United States of America
- Department of Physiology, Tulane University School of Medicine, New Orleans, Louisiana, United States of America
| | - Bret N. Smith
- Department of Physiology, University of Kentucky College of Medicine, Lexington, Kentucky, United States of America
- Department of Cell and Molecular Biology, Tulane University, New Orleans, Louisiana, United States of America
- * E-mail:
| |
Collapse
|
35
|
Sohn JW, Elmquist JK, Williams KW. Neuronal circuits that regulate feeding behavior and metabolism. Trends Neurosci 2013; 36:504-12. [PMID: 23790727 DOI: 10.1016/j.tins.2013.05.003] [Citation(s) in RCA: 200] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2013] [Revised: 05/06/2013] [Accepted: 05/20/2013] [Indexed: 12/24/2022]
Abstract
Neurons within the central nervous system receive humoral and central (neurotransmitter or neuropeptide) signals that ultimately regulate ingestive behavior and metabolism. Recent advances in mouse genetics combined with neuroanatomical and electrophysiological techniques have contributed to a better understanding of these central mechanisms. This review integrates recently defined cellular mechanisms and neural circuits relevant to the regulation of feeding behavior, energy expenditure, and glucose homeostasis by metabolic signals.
Collapse
Affiliation(s)
- Jong-Woo Sohn
- Division of Hypothalamic Research, Department of Internal Medicine, The University of Texas Southwestern Medical Center at Dallas, Dallas, TX, USA
| | | | | |
Collapse
|
36
|
Sohn JW, Harris LE, Berglund ED, Liu T, Vong L, Lowell BB, Balthasar N, Williams KW, Elmquist JK. Melanocortin 4 receptors reciprocally regulate sympathetic and parasympathetic preganglionic neurons. Cell 2013; 152:612-9. [PMID: 23374353 DOI: 10.1016/j.cell.2012.12.022] [Citation(s) in RCA: 172] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2012] [Revised: 11/20/2012] [Accepted: 12/18/2012] [Indexed: 10/27/2022]
Abstract
Melanocortin 4 receptors (MC4Rs) in the central nervous system are key regulators of energy and glucose homeostasis. Notably, obese patients with MC4R mutations are hyperinsulinemic and resistant to obesity-induced hypertension. Although these effects are probably dependent upon the activity of the autonomic nervous system, the cellular effects of MC4Rs on parasympathetic and sympathetic neurons remain undefined. Here, we show that MC4R agonists inhibit parasympathetic preganglionic neurons in the brainstem. In contrast, MC4R agonists activate sympathetic preganglionic neurons in the spinal cord. Deletion of MC4Rs in cholinergic neurons resulted in elevated levels of insulin. Furthermore, re-expression of MC4Rs specifically in cholinergic neurons (including sympathetic preganglionic neurons) restores obesity-associated hypertension in MC4R null mice. These findings provide a cellular correlate of the autonomic side effects associated with MC4R agonists and demonstrate a role for MC4Rs expressed in cholinergic neurons in the regulation of insulin levels and in the development of obesity-induced hypertension.
Collapse
Affiliation(s)
- Jong-Woo Sohn
- Department of Internal Medicine, Division of Hypothalamic Research, University of Texas Southwestern Medical Center at Dallas, Dallas, TX 75390, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
37
|
Boychuk CR, Zsombok A, Tasker JG, Smith BN. Rapid Glucocorticoid-Induced Activation of TRP and CB1 Receptors Causes Biphasic Modulation of Glutamate Release in Gastric-Related Hypothalamic Preautonomic Neurons. Front Neurosci 2013; 7:3. [PMID: 23386808 PMCID: PMC3560102 DOI: 10.3389/fnins.2013.00003] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2012] [Accepted: 01/07/2013] [Indexed: 02/02/2023] Open
Abstract
Glucocorticoids rapidly regulate synaptic input to neuroendocrine cells in the hypothalamic paraventricular nucleus (PVN) by inducing the retrograde release of endogenous messengers. Here we investigated the rapid effects of dexamethasone (DEX) on excitatory synaptic input to feeding-related, preautonomic PVN neurons using whole-cell patch-clamp recordings. In ∼50% of identified gastric-related preautonomic PVN neurons, DEX elicited a biphasic synaptic response characterized by an initial rapid and transient increase in the frequency of miniature excitatory postsynaptic currents (mEPSCs), followed by a decrease in mEPSC frequency within 9 min; remaining cells displayed only a decrease in mEPSC frequency. The late-phase decrease in mEPSC frequency was mimicked by the cannabinoid receptor agonists anandamide (AEA) and WIN 55,212-2, and it was blocked by the CB1 receptor antagonist AM251. The biphasic DEX effect was mimicked by AEA. The early increase in mEPSCs was mimicked by activation of transient receptor potential vanilloid type 1 (TRPV1) receptors with capsaicin and by activation of TRPV4 receptors with 4-α-PDD. The increase was reduced, but not blocked, by selective TRPV1 antagonists and in TRPV1 knockout mice; it was blocked completely by the broad-spectrum TRPV antagonist ruthenium red and by combined application of selective TRPV1 and TRPV4 antagonists. The DEX effects were prevented entirely by intracellular infusion of the G-protein inhibitor, GDPβS. Thus, DEX biphasically modulates synaptic glutamate onto a subset of gastric-related PVN neurons, which is likely mediated by induction of a retrograde messenger. The effect includes a TRPV1/4 receptor-mediated transient increase and subsequent CB1 receptor-mediated suppression of glutamate release. Multiphasic modulation of glutamate input to PVN neurons represents a previously unappreciated complexity of control of autonomic output by glucocorticoids and endogenous cannabinoids.
Collapse
Affiliation(s)
- Carie R Boychuk
- Department of Physiology, University of Kentucky College of Medicine Lexington, KY, USA
| | | | | | | |
Collapse
|
38
|
Kentish SJ, O'Donnell TA, Isaacs NJ, Young RL, Li H, Harrington AM, Brierley SM, Wittert GA, Blackshaw LA, Page AJ. Gastric vagal afferent modulation by leptin is influenced by food intake status. J Physiol 2012; 591:1921-34. [PMID: 23266933 DOI: 10.1113/jphysiol.2012.247577] [Citation(s) in RCA: 70] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Energy intake is strongly influenced by vagal afferent signals from the stomach, and is also modulated by leptin. Leptin may be secreted from gastric epithelial cells, so we aimed to determine the direct effect of leptin on gastric vagal afferents under different feeding conditions. Female C57BL/6 mice were fed standard laboratory diet, high-fat diet or were food restricted. The expression of leptin receptor (Lep-R) and its signal transduction molecules in vagal afferents was determined by retrograde tracing and reverse-transcription polymerase chain reaction, and the relationship between leptin-immunopositive cells and gastric vagal afferent endings determined by anterograde tracing and leptin immunohistochemistry. An in vitro preparation was used to determine the functional effects of leptin on gastric vagal afferents and the second messenger pathways involved. Leptin potentiated vagal mucosal afferent responses to tactile stimuli, and epithelial cells expressing leptin were found close to vagal mucosal endings. After fasting or diet-induced obesity, potentiation of mucosal afferents by leptin was lost and Lep-R expression reduced in the cell bodies of gastric mucosal afferents. These effects in diet-induced obese mice were accompanied by a reduction in anatomical vagal innervation of the gastric mucosa. In striking contrast, after fasting or diet-induced obesity, leptin actually inhibited responses to distension in tension receptors. The inhibitory effect on gastric tension receptors was mediated through phosphatidylinositol 3-kinase-dependent activation of large-conductance calcium-activated potassium channels. The excitatory effect of leptin on gastric mucosal vagal afferents was mediated by phospholipase C-dependent activation of canonical transient receptor potential channels. These data suggest the effect of leptin on gastric vagal afferent excitability is dynamic and related to the feeding state. Paradoxically, in obesity, leptin may reduce responses to gastric distension following food intake.
Collapse
Affiliation(s)
- Stephen J Kentish
- Nerve-Gut Research Laboratory, Room 1-216-H, Level 1, Hanson Institute, Royal Adelaide Hospital, Adelaide, SA 5000, Australia
| | | | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Gelegen C, Chandarana K, Choudhury AI, Al-Qassab H, Evans IM, Irvine EE, Hyde CB, Claret M, Andreelli F, Sloan SE, Leiter AB, Withers DJ, Batterham RL. Regulation of hindbrain Pyy expression by acute food deprivation, prolonged caloric restriction, and weight loss surgery in mice. Am J Physiol Endocrinol Metab 2012; 303:E659-68. [PMID: 22761162 PMCID: PMC3468511 DOI: 10.1152/ajpendo.00033.2012] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
PYY is a gut-derived putative satiety signal released in response to nutrient ingestion and is implicated in the regulation of energy homeostasis. Pyy-expressing neurons have been identified in the hindbrain of river lamprey, rodents, and primates. Despite this high evolutionary conservation, little is known about central PYY neurons. Using in situ hybridization, PYY-Cre;ROSA-EYFP mice, and immunohistochemistry, we identified PYY cell bodies in the gigantocellular reticular nucleus region of the hindbrain. PYY projections were present in the dorsal vagal complex and hypoglossal nucleus. In the hindbrain, Pyy mRNA was present at E9.5, and expression peaked at P2 and then decreased significantly by 70% at adulthood. We found that, in contrast to the circulation, PYY-(1-36) is the predominant isoform in mouse brainstem extracts in the ad libitum-fed state. However, following a 24-h fast, the relative amounts of PYY-(1-36) and PYY-(3-36) isoforms were similar. Interestingly, central Pyy expression showed nutritional regulation and decreased significantly by acute starvation, prolonged caloric restriction, and bariatric surgery (enterogastroanastomosis). Central Pyy expression correlated with body weight loss and circulating leptin and PYY concentrations. Central regulation of energy metabolism is not limited to the hypothalamus but also includes the midbrain and the brainstem. Our findings suggest a role for hindbrain PYY in the regulation of energy homeostasis and provide a starting point for further research on gigantocellular reticular nucleus PYY neurons, which will increase our understanding of the brain stem pathways in the integrated control of appetite and energy metabolism.
Collapse
Affiliation(s)
- C. Gelegen
- 1Centre for Obesity Research, Department of Medicine, University College London, Rayne Institute, London, United Kingdom;
| | - K. Chandarana
- 1Centre for Obesity Research, Department of Medicine, University College London, Rayne Institute, London, United Kingdom;
| | - A. I. Choudhury
- 2Metabolic Signalling Group, Medical Research Council Clinical Sciences Centre, Imperial College, London, United Kingdom;
| | - H. Al-Qassab
- 1Centre for Obesity Research, Department of Medicine, University College London, Rayne Institute, London, United Kingdom;
| | - I. M. Evans
- 3Centre for Cardiovascular Biology and Medicine, Department of Medicine, Rayne Institute, London, United Kingdom;
| | - E. E. Irvine
- 1Centre for Obesity Research, Department of Medicine, University College London, Rayne Institute, London, United Kingdom;
| | - C. B. Hyde
- 4Scientific Support Services, Wolfson Institute for Biomedical Research and the University College London Cancer Institute, London, United Kingdom;
| | - M. Claret
- 5Diabetes and Obesity Laboratory, Institut d′Investigacions Biomèdiques August Pi i Sunyer, Centre Esther Koplowitz, Barcelona, Spain;
| | - F. Andreelli
- 6Institut Cochin, Université Paris Descartes, Paris, France;
- 7Department of Diabetology, Pitié-Salpêtrière Hospital, University Pierre et Marie Curie-Paris 6, Paris, France;
| | - S. E. Sloan
- 8University of Massachusetts Medical School, Department of Medicine, Worcester, Massachusetts
| | - A. B. Leiter
- 8University of Massachusetts Medical School, Department of Medicine, Worcester, Massachusetts
| | - D. J. Withers
- 2Metabolic Signalling Group, Medical Research Council Clinical Sciences Centre, Imperial College, London, United Kingdom;
| | - R. L. Batterham
- 1Centre for Obesity Research, Department of Medicine, University College London, Rayne Institute, London, United Kingdom;
| |
Collapse
|
40
|
Yoshida R, Niki M, Jyotaki M, Sanematsu K, Shigemura N, Ninomiya Y. Modulation of sweet responses of taste receptor cells. Semin Cell Dev Biol 2012; 24:226-31. [PMID: 22947916 DOI: 10.1016/j.semcdb.2012.08.004] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2012] [Accepted: 08/20/2012] [Indexed: 11/17/2022]
Abstract
Taste receptor cells play a major role in detection of chemical compounds in the oral cavity. Information derived from taste receptor cells, such as sweet, bitter, salty, sour and umami is important for evaluating the quality of food components. Among five basic taste qualities, sweet taste is very attractive for animals and influences food intake. Recent studies have demonstrated that sweet taste sensitivity in taste receptor cells would be affected by leptin and endocannabinoids. Leptin is an anorexigenic mediator that reduces food intake by acting on leptin receptor Ob-Rb in the hypothalamus. Endocannabinoids such as anandamide [N-arachidonoylethanolamine (AEA)] and 2-arachidonoyl glycerol (2-AG) are known as orexigenic mediators that act via cannabinoid receptor 1 (CB1) in the hypothalamus and limbic forebrain to induce appetite and stimulate food intake. At the peripheral gustatory organs, leptin selectively suppresses and endocannabinoids selectively enhance sweet taste sensitivity via Ob-Rb and CB1 expressed in sweet sensitive taste cells. Thus leptin and endocannabinoids not only regulate food intake via central nervous systems but also modulate palatability of foods by altering peripheral sweet taste responses. Such reciprocal modulation of leptin and endocannabinoids on peripheral sweet sensitivity may play an important role in regulating energy homeostasis.
Collapse
Affiliation(s)
- Ryusuke Yoshida
- Section of Oral Neuroscience, Graduate School of Dental Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, Japan
| | | | | | | | | | | |
Collapse
|
41
|
Blake CB, Smith BN. Insulin reduces excitation in gastric-related neurons of the dorsal motor nucleus of the vagus. Am J Physiol Regul Integr Comp Physiol 2012; 303:R807-14. [PMID: 22914748 DOI: 10.1152/ajpregu.00276.2012] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
The dorsal motor nucleus of the vagus (DMV) in the caudal brain stem is composed mainly of preganglionic parasympathetic neurons that control the subdiaphragmatic viscera and thus participates in energy homeostasis regulation. Metabolic pathologies, including diabetes, can disrupt vagal circuitry and lead to gastric dysfunction. Insulin receptors (IRs) are expressed in the DMV, and insulin crosses the blood-brain barrier and is transported into the brain stem. Despite growing evidence that insulin action in the brain is critical for energy homeostasis, little is known about insulin's action in the DMV. We used whole cell patch-clamp recordings in brain stem slices to identify effects of insulin on membrane and synaptic input properties of DMV neurons, including a subset of gastric-related cells identified subsequent to injection of a retrograde label into the gastric wall. Insulin application significantly reduced the frequency of spontaneous and miniature excitatory, but not inhibitory postsynaptic currents, with no change in amplitude (P < 0.05). Insulin also directly hyperpolarized the membrane potential (-4.2 ± 1.3 mV; P < 0.05) and reduced action potential firing (P < 0.05). Insulin effects were eliminated in the presence of a ATP-dependent K+ (K(ATP)) channel antagonist tolbutamide (200 μM), or the phosphatidylinositol 3-kinase (PI3K) inhibitor wortmannin (100 nM), suggesting that insulin inhibition of excitatory input to gastric-related DMV neurons was mediated by K(ATP) channels and depended on PI3K activity. Insulin regulation of synaptic input in the DMV may influence autonomic visceral regulation and thus systemic glucose metabolism.
Collapse
Affiliation(s)
- Camille B Blake
- Dept. of Physiology, Univ. of Kentucky College of Medicine, MS508 Chandler Medical Center, 800 Rose St., Lexington, KY 40536, USA
| | | |
Collapse
|
42
|
Bach EC, Smith BN. Presynaptic NMDA receptor-mediated modulation of excitatory neurotransmission in the mouse dorsal motor nucleus of the vagus. J Neurophysiol 2012; 108:1484-91. [PMID: 22696534 DOI: 10.1152/jn.01036.2011] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Activity of neurons in the dorsal motor nucleus of the vagus nerve (DMV) is closely regulated by synaptic input, and regulation of that input by glutamate receptors on presynaptic terminals has been proposed. Presynaptic N-methyl-d-aspartic acid (NMDA) receptors have been identified in a number of brain regions and act to modulate neurotransmitter release, but functional presynaptic NMDA receptors have not been adequately studied in the DMV. This study identified the presence and physiological function of presynaptic NMDA receptors on synaptic input to DMV neurons. Whole-cell patch-clamp recordings from DMV neurons in acute slices from mice revealed prevalent miniature excitatory postsynaptic currents, which were significantly increased in frequency, but not amplitude, by application of NMDA. Antagonism of NMDA receptors with dl-2-amino-5-phosphonopentanoic acid (100 μM) resulted in a decrease in miniature excitatory postsynaptic current frequency and an increase in the paired pulse ratio of responses following afferent stimulation. No consistent effects of presynaptic NMDA receptor modulation were observed on GABAergic inputs. These results suggest that presynaptic NMDA receptors are present in the dorsal vagal complex and function to facilitate the release of glutamate, preferentially onto DMV neurons tonically, with little effect on GABA release. This type of presynaptic modulation represents a potentially novel form of glutamate regulation in the DMV, which may function to regulate glutamate-induced activity of central parasympathetic circuits.
Collapse
Affiliation(s)
- Eva C Bach
- Department of Physiology, University of Kentucky College of Medicine, Lexington, KY 40536, USA
| | | |
Collapse
|
43
|
Gao H, Miyata K, Bhaskaran MD, Derbenev AV, Zsombok A. Transient receptor potential vanilloid type 1-dependent regulation of liver-related neurons in the paraventricular nucleus of the hypothalamus diminished in the type 1 diabetic mouse. Diabetes 2012; 61:1381-90. [PMID: 22492526 PMCID: PMC3357291 DOI: 10.2337/db11-0820] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
The paraventricular nucleus (PVN) of the hypothalamus controls the autonomic neural output to the liver, thereby participating in the regulation of hepatic glucose production (HGP); nevertheless, mechanisms controlling the activity of liver-related PVN neurons are not known. Transient receptor potential vanilloid type 1 (TRPV1) is involved in glucose homeostasis and colocalizes with liver-related PVN neurons; however, the functional role of TRPV1 regarding liver-related PVN neurons has to be elucidated. A retrograde viral tracer was used to identify liver-related neurons within the brain-liver circuit in control, type 1 diabetic, and insulin-treated mice. Our data indicate that TRPV1 regulates liver-related PVN neurons. This TRPV1-dependent excitation diminished in type 1 diabetic mice. In vivo and in vitro insulin restored TRPV1 activity in a phosphatidylinositol 3-kinase/protein kinase C-dependent manner and stimulated TRPV1 receptor trafficking to the plasma membrane. There was no difference in total TRPV1 protein expression; however, increased phosphorylation of TRPV1 receptors was observed in type 1 diabetic mice. Our data demonstrate that TRPV1 plays a pivotal role in the regulation of liver-related PVN neurons. Moreover, TRPV1-dependent excitation of liver-related PVN neurons diminishes in type 1 diabetes, thus indicating that the brain-liver autonomic circuitry is altered in type 1 diabetes and may contribute to the autonomic dysfunction of HGP.
Collapse
Affiliation(s)
- Hong Gao
- Department of Physiology, Tulane University, School of Medicine, New Orleans, Louisiana, USA.
| | | | | | | | | |
Collapse
|
44
|
Pénicaud L, Meillon S, Brondel L. Leptin and the central control of feeding behavior. Biochimie 2012; 94:2069-74. [PMID: 22546505 DOI: 10.1016/j.biochi.2012.04.013] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2011] [Accepted: 04/06/2012] [Indexed: 12/25/2022]
Abstract
The discovery of leptin by Friedman and coll. in 1995 was a major step forward in our comprehensive view of energy homeostasis. Since the original paper, a tremendous amount of work has been performed in laboratories all over the world. Many recent reviews have described this work in details. In the present review, we focus on the role of leptin on food intake. It is accepted by most authors working in this field that the control of food intake can be divided in two closely-related system: the homeostatic system and the hedonic system. Leptin has been shown to act on both systems.
Collapse
Affiliation(s)
- Luc Pénicaud
- CNRS, UMR6265 Centre des Sciences du Goût et de l'Alimentation, F-2100 Dijon, France.
| | | | | |
Collapse
|
45
|
The acute effects of leptin require PI3K signaling in the hypothalamic ventral premammillary nucleus. J Neurosci 2011; 31:13147-56. [PMID: 21917798 DOI: 10.1523/jneurosci.2602-11.2011] [Citation(s) in RCA: 59] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023] Open
Abstract
Evidence suggests that the role played by the adipocyte-derived hormone leptin in female reproductive physiology is mediated in part by neurons located within the ventral premammillary nucleus (PMV). Leptin activates PMV neurons; however, the intracellular signaling pathway and channel(s) involved remain undefined. Notably, leptin's excitatory and inhibitory effects within hypothalamic and brainstem nuclei share the intracellular signaling cascade phosphoinositide 3 kinase (PI3K). Therefore, we assessed whether PI3K signaling is required for the acute effect of leptin to alter cellular activity of PMV neurons that express leptin receptors (LepR PMV neurons). Leptin caused a rapid depolarization in the majority of LepR PMV neurons in patch-clamp recordings of hypothalamic slices, while a subset of LepR PMV neurons were hyperpolarized in response to leptin. Data were obtained from both male and female mice and results demonstrate that the acute effect of leptin on LepR PMV neurons was identical for both sexes. Pharmacological inhibition of PI3K prevented the acute leptin-induced change in neuronal activity of LepR PMV neurons, indicating a PI3K-dependent mechanism of leptin action. Similarly, mice with genetically disrupted PI3K signaling in LepR PMV neurons failed to alter cellular activity in response to leptin. Moreover, the leptin-induced depolarization was dependent on a putative TRPC channel. In contrast, the leptin-induced-hyperpolarization required the activation of a putative Katp channel. Collectively, these results suggest that PI3K signaling in LepR PMV neurons is essential for leptin-induced alteration in cellular activity, and these data may suggest a cellular correlate in which leptin contributes to the initiation of reproductive development.
Collapse
|
46
|
Scott MM, Williams KW, Rossi J, Lee CE, Elmquist JK. Leptin receptor expression in hindbrain Glp-1 neurons regulates food intake and energy balance in mice. J Clin Invest 2011; 121:2413-21. [PMID: 21606595 DOI: 10.1172/jci43703] [Citation(s) in RCA: 135] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2010] [Accepted: 03/09/2011] [Indexed: 11/17/2022] Open
Abstract
Leptin is an adipose-derived hormone that signals to inform the brain of nutrient status; loss of leptin signaling results in marked hyperphagia and obesity. Recent work has identified several groups of neurons that contribute to the effects of leptin to regulate energy balance, but leptin receptors are distributed throughout the brain, and the function of leptin signaling in discrete neuronal populations outside of the hypothalamus has not been defined. In the current study, we produced mice in which the long form of the leptin receptor (Lepr) was selectively ablated using Cre-recombinase selectively expressed in the hindbrain under control of the paired-like homeobox 2b (Phox2b) promoter (Phox2b Cre Lepr(flox/flox) mice). In these mice, Lepr was deleted from glucagon-like 1 peptide-expressing neurons resident in the nucleus of the solitary tract. Phox2b Cre Lepr(flox/flox) mice were hyperphagic, displayed increased food intake after fasting, and gained weight at a faster rate than wild-type controls. Paradoxically, Phox2b Cre Lepr(flox/flox) mice also exhibited an increased metabolic rate independent of a change in locomotor activity that was dependent on food intake, and glucose homeostasis was normal. Together, these data support a physiologically important role of direct leptin action in the hindbrain.
Collapse
Affiliation(s)
- Michael M Scott
- Division of Hypothalamic Research, Department of Internal Medicine, The University of Texas Southwestern Medical Center, Dallas, Texas 75390-9077, USA
| | | | | | | | | |
Collapse
|
47
|
Zsombok A, Gao H, Miyata K, Issa A, Derbenev AV. Immunohistochemical localization of transient receptor potential vanilloid type 1 and insulin receptor substrate 2 and their co-localization with liver-related neurons in the hypothalamus and brainstem. Brain Res 2011; 1398:30-9. [PMID: 21620379 DOI: 10.1016/j.brainres.2011.04.048] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2011] [Revised: 04/29/2011] [Accepted: 04/29/2011] [Indexed: 02/05/2023]
Abstract
The central nervous system plays an important role in the regulation of energy balance and glucose homeostasis mainly via controlling the autonomic output to the visceral organs. The autonomic output is regulated by hormones and nutrients to maintain adequate energy and glucose homeostasis. Insulin action is mediated via insulin receptors (IR) resulting in phosphorylation of insulin receptor substrates (IRS) inducing activation of downstream pathways. Furthermore, insulin enhances transient receptor potential vanilloid type 1 (TRPV1) mediated currents. Activation of the TRPV1 receptor increases excitatory neurotransmitter release in autonomic centers of the brain, thereby impacting energy and glucose homeostasis. The aim of this study is to determine co-expression of IRS2 and TRPV1 receptors in the paraventricular nucleus of the hypothalamus (PVN) and dorsal motor nucleus of the vagus (DMV) in the mouse brain as well as expression of IRS2 and TRPV1 receptors at liver-related preautonomic neurons pre-labeled with a trans-neural, viral tracer (PRV-152). The data indicate that IRS2 and TRPV1 receptors are present and co-express in the PVN and the DMV. A large portion (over 50%) of the liver-related preautonomic DMV and PVN neurons expresses IRS2. Moreover, the majority of liver-related DMV and PVN neurons also express TRPV1 receptors, suggesting that insulin and TRPV1 actions may affect liver-related preautonomic neurons.
Collapse
Affiliation(s)
- Andrea Zsombok
- Department of Physiology, Tulane University Health Science Center, New Orleans, LA 70112, USA.
| | | | | | | | | |
Collapse
|
48
|
Xu L, Scheenen WJJM, Leshan RL, Patterson CM, Elias CF, Bouwhuis S, Roubos EW, Myers MG, Kozicz T. Leptin signaling modulates the activity of urocortin 1 neurons in the mouse nonpreganglionic Edinger-Westphal nucleus. Endocrinology 2011; 152:979-88. [PMID: 21209012 PMCID: PMC3040051 DOI: 10.1210/en.2010-1143] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
A recent study systematically characterized the distribution of the long form of the leptin receptor (LepRb) in the mouse brain and showed substantial LepRb mRNA expression in the nonpreganglionic Edinger-Westphal nucleus (npEW) in the rostroventral part of the midbrain. This nucleus hosts the majority of urocortin 1 (Ucn1) neurons in the rodent brain, and because Ucn1 is a potent satiety hormone and electrical lesioning of the npEW strongly decreases food intake, we have hypothesized a role of npEW-Ucn1 neurons in leptin-controlled food intake. Here, we show by immunohistochemistry that npEW-Ucn1 neurons in the mouse contain LepRb and respond to leptin administration with induction of the Janus kinase 2-signal transducer and activator of transcription 3 pathway, both in vivo and in vitro. Furthermore, systemic leptin administration increases the Ucn1 content of the npEW significantly, whereas in mice that lack LepRb (db/db mice), the npEW contains considerably reduced amount of Ucn1. Finally, we reveal by patch clamping of midbrain Ucn1 neurons that leptin administration reduces the electrical firing activity of the Ucn1 neurons. In conclusion, we provide ample evidence for leptin actions that go beyond leptin's well-known targets in the hypothalamus and propose that leptin can directly influence the activity of the midbrain Ucn1 neurons.
Collapse
Affiliation(s)
- Lu Xu
- Department of Cellular Animal Physiology, Donders Institute for Brain, Cognition and Behaviour, Radboud University Nijmegen, 6500 GL Nijmegen, The Netherlands
| | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Rogers RC, McDougal DH, Hermann GE. Leptin amplifies the action of thyrotropin-releasing hormone in the solitary nucleus: an in vitro calcium imaging study. Brain Res 2011; 1385:47-55. [PMID: 21334313 DOI: 10.1016/j.brainres.2011.02.029] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2010] [Revised: 02/11/2011] [Accepted: 02/11/2011] [Indexed: 12/28/2022]
Abstract
Leptin exerts a powerful permissive influence on neurogenic thermogenesis. During starvation and an absence of leptin, animals cannot produce thermogenic reactions to cold stress. However, thermogenesis is rescued by restoring leptin. We have previously observed a highly cooperative interaction between leptin and thyrotropin-releasing hormone [TRH] to activate hindbrain-generated thermogenic responses (Hermann et al., 2006). In vivo physiological studies (Rogers et al., 2009) suggested that the thermogenic impact of TRH in the hindbrain is amplified by the action of leptin through a leptin receptor-mediated production of phosphoinositol-trisphosphate [PIP3]. In turn, PIP3 can activate a tyrosine kinase whose target is the Src-SH2 regulatory site on the phospholipase C [PLC] complex. The TRH receptor signals through the PLC complex. Our immunohistochemical studies (Barnes et al., 2010) suggest that this transduction interaction between leptin and TRH occurs within neurons of the solitary nucleus [NST], though this interaction had not been verified. The present in vitro live cell calcium imaging study shows that while medial NST neurons are rarely activated by leptin alone, leptin pre-treatment significantly augments NST neurons' responsiveness to TRH. This leptin-mediated priming of NST neurons was uncoupled by pre-treatment with the phosphoinositide 3-kinase [PI3K] inhibitor [wortmannin], the phospholipase C inhibitor [U73122] and the Src-SH2 antagonist [PP2]. TTX did not eliminate the synergistic response of the agonists, thus the sensitization cannot be attributed to pre-synaptic mechanisms. It seems likely that NST neurons are involved in the leptin-mediated increase in BAT temperature by sensitizing the TRH-PLC-IP3-calcium release mechanism.
Collapse
Affiliation(s)
- Richard C Rogers
- Laboratory of Autonomic Neurosciences, Pennington Biomedical Research Center, Baton Rouge, LA 70808, USA
| | | | | |
Collapse
|
50
|
Williams KW, Scott MM, Elmquist JK. Modulation of the central melanocortin system by leptin, insulin, and serotonin: co-ordinated actions in a dispersed neuronal network. Eur J Pharmacol 2011; 660:2-12. [PMID: 21211525 DOI: 10.1016/j.ejphar.2010.11.042] [Citation(s) in RCA: 63] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2010] [Revised: 10/08/2010] [Accepted: 11/02/2010] [Indexed: 10/18/2022]
Abstract
Over the past century, prevalent models of energy and glucose homeostasis have been developed from a better understanding of the neural circuits underlying obesity and diabetes. From the early hypothalamic lesion reports to the more recent pharmacological and molecular/genetic studies, the hypothalamic melanocortin system has been shown to play a critical role in the regulation of metabolism. This review attempts to highlight contributions to our current understanding of how numerous neuromodulators (leptin, insulin, and serotonin) integrate with the central melanocortin system to coordinate alterations in energy and glucose balance.
Collapse
Affiliation(s)
- Kevin W Williams
- Division of Hypothalamic Research, Department of Internal Medicine, The University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | | | | |
Collapse
|