1
|
Mirzadeh Z, Faber C. Brain Defense of Glycemia in Health and Diabetes. Diabetes 2024; 73:1952-1966. [PMID: 39401393 PMCID: PMC11579547 DOI: 10.2337/dbi24-0001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/08/2024] [Accepted: 07/03/2024] [Indexed: 11/22/2024]
Abstract
The brain coordinates the homeostatic defense of multiple metabolic variables, including blood glucose levels, in the context of ever-changing external and internal environments. The biologically defended level of glycemia (BDLG) is the net result of brain modulation of insulin-dependent mechanisms in cooperation with the islet, and insulin-independent mechanisms through direct innervation and neuroendocrine control of glucose effector tissues. In this article, we highlight evidence from animal and human studies to develop a framework for the brain's core homeostatic functions-sensory/afferent, integration/processing, and motor/efferent-that contribute to the normal BDLG in health and its elevation in diabetes. ARTICLE HIGHLIGHTS
Collapse
Affiliation(s)
- Zaman Mirzadeh
- Department of Neurosurgery, Barrow Neurological Institute, St. Joseph’s Hospital and Medical Center, Phoenix, AZ
| | - Chelsea Faber
- Department of Neurosurgery, Barrow Neurological Institute, St. Joseph’s Hospital and Medical Center, Phoenix, AZ
| |
Collapse
|
2
|
Myeong J, Stunault MI, Klyachko VA, Ashrafi G. Metabolic regulation of single synaptic vesicle exo- and endocytosis in hippocampal synapses. Cell Rep 2024; 43:114218. [PMID: 38758651 PMCID: PMC11221188 DOI: 10.1016/j.celrep.2024.114218] [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: 11/13/2023] [Revised: 02/26/2024] [Accepted: 04/25/2024] [Indexed: 05/19/2024] Open
Abstract
Glucose has long been considered a primary energy source for synaptic function. However, it remains unclear to what extent alternative fuels, such as lactate/pyruvate, contribute to powering synaptic transmission. By detecting individual release events in hippocampal synapses, we find that mitochondrial ATP production regulates basal vesicle release probability and release location within the active zone (AZ), evoked by single action potentials. Mitochondrial inhibition shifts vesicle release closer to the AZ center and alters the efficiency of vesicle retrieval by increasing the occurrence of ultrafast endocytosis. Furthermore, we uncover that terminals can use oxidative fuels to maintain the vesicle cycle during trains of activity. Mitochondria are sparsely distributed along hippocampal axons, and we find that terminals containing mitochondria display enhanced vesicle release and reuptake during high-frequency trains. Our findings suggest that mitochondria not only regulate several fundamental features of synaptic transmission but may also contribute to modulation of short-term synaptic plasticity.
Collapse
Affiliation(s)
- Jongyun Myeong
- Department of Cell Biology and Physiology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Marion I Stunault
- Department of Cell Biology and Physiology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Vitaly A Klyachko
- Department of Cell Biology and Physiology, Washington University School of Medicine, St. Louis, MO 63110, USA.
| | - Ghazaleh Ashrafi
- Department of Cell Biology and Physiology, Washington University School of Medicine, St. Louis, MO 63110, USA; Needleman Center for Neurometabolism and Axonal Therapeutics, Washington University School of Medicine, St. Louis, MO 63110, USA.
| |
Collapse
|
3
|
Konanur VR, Hurh SJ, Hsu TM, Roitman MF. Dopamine neuron activity evoked by sucrose and sucrose-predictive cues is augmented by peripheral and central manipulations of glucose availability. Eur J Neurosci 2024; 59:2419-2435. [PMID: 38057909 PMCID: PMC11108752 DOI: 10.1111/ejn.16214] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2023] [Revised: 10/23/2023] [Accepted: 11/16/2023] [Indexed: 12/08/2023]
Abstract
Food deprivation drives eating through multiple signals and circuits. Decreased glucose availability (i.e., cytoglucopenia) drives eating and also increases the value of sucrose. Ventral tegmental area (VTA) dopamine neurons (DANs) contribute to the evaluation of taste stimuli, but their role in integrating glucoprivic signals remains unknown. We monitored VTA DAN activity via Cre-dependent expression of a calcium indicator with in vivo fibre photometry. In ad libitum fed rats, intraoral sucrose evoked a phasic increase in DAN activity. To manipulate glucose availability, we administered (intraperitoneal, lateral or fourth ventricular) the antiglycolytic agent 5-thio-D-glucose (5TG), which significantly augmented the phasic DAN activity to sucrose. 5TG failed to alter DAN activity to water or saccharin, suggesting the response was selective for caloric stimuli. 5TG enhancement of sucrose-evoked DAN activity was stronger after fourth ventricular administration, suggesting a critical node of action within the hindbrain. As 5TG also increases blood glucose, in a separate study, we used peripheral insulin, which stimulates eating, to decrease blood glucose-which was associated with increased DAN activity to intraoral sucrose. DAN activity developed to a cue predictive of intraoral sucrose. While 5TG augmented cue-evoked DAN activity, its action was most potent when delivered to the lateral ventricle. Together, the studies point to central glucose availability as a key modulator of phasic DAN activity to food and food-cues. As glucose sensing neurons are known to populate the hypothalamus and brainstem, results suggest differential modulation of cue-evoked and sucrose-evoked DAN activity.
Collapse
Affiliation(s)
- Vaibhav R. Konanur
- Department of Psychology, University of Illinois at Chicago, Chicago, IL
- Current affiliation: Department of Biology, University of Illinois at Chicago, Chicago, IL
| | - Samantha J. Hurh
- Department of Psychology, University of Illinois at Chicago, Chicago, IL
| | - Ted M. Hsu
- Department of Psychology, University of Illinois at Chicago, Chicago, IL
| | | |
Collapse
|
4
|
Qin C, Yuan Q, Liu M, Zhuang L, Xu L, Wang P. Biohybrid tongue based on hypothalamic neuronal network-on-a-chip for real-time blood glucose sensing and assessment. Biosens Bioelectron 2024; 244:115784. [PMID: 37939416 DOI: 10.1016/j.bios.2023.115784] [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: 08/15/2023] [Revised: 10/14/2023] [Accepted: 10/22/2023] [Indexed: 11/10/2023]
Abstract
The expression of sweet receptors in the hypothalamus has been implicated in energy homeostasis control and the pathogenesis of obesity and diabetes. However, the exact mechanism by which hypothalamic glucose-sensing neurons function remains unclear. Conventional detection methods, such as fiber photometry, optogenetics, brain-machine interfaces, patch clamp and calcium imaging, pose limitations for real-time glucose perception due to their complexity, cytotoxicity and so on. Therefore, this study proposes a biohybrid tongue based on hypothalamic neuronal network (HNN)-on-a-chip coupling with microelectrode array (MEA) for real-time glucose perception. Hypothalamic neuronal cultures were cultivated on a two-dimensional "brain-on-chip" device, enabling the formation of neuronal networks and electrophysiological signal detection. Additionally, we investigated the endogenous expression of sweet taste receptors (T1R2/T1R3) in hypothalamic neuronal cells, providing the basis for the biohybrid tongue based on HNN-on-a-chip's sweetness detection capabilities. The spike signal response to sucrose and glucose stimulation was detected, and concentration-dependent responses were explored with glucose concentrations ranging from 0.01 mM to 8 mM. MEAs allow for real-time recordings, enabling the observation of dynamic changes in neuronal responses to glucose fluctuations over time. The biohybrid tongue based on HNN-on-a-chip can measure various parameters, including spike frequency and amplitude, providing insights into neuronal firing patterns and excitability. Moreover, hypothalamic glucoregulatory neurons that sense and respond to changes in blood glucose was identified, including glucose-excited neurons (GE-Neurons) and glucose-inhibited neurons (GI-Neurons). The detection range for GE-Neurons spans from 0.4 to 6 mM, while GI-Neurons demonstrate sensitivity within the range of 1-8 mM. And the glucose detection limit was firmly established at 0.01 mM. Through non-linear regression analysis, the IC50 for GI-Neurons' spike firing was determined to be 4.18 mM. In conclusion, the biohybrid tongue based on HNN-on-a-chip offers a valuable in vitro tool for studying hypothalamic neurons, elucidating glucose sensing mechanisms, and understanding hypothalamic neuronal function.
Collapse
Affiliation(s)
- Chunlian Qin
- College of Biosystems Engineering and Food Science, Zhejiang University, Hangzhou, 310058, China; ZJU-Hangzhou Global Scientific and Technological Innovation Center, Zhejiang University, Hangzhou, 311215, China
| | - Qunchen Yuan
- Biosensor National Special Laboratory, Key Laboratory for Biomedical Engineering of Education Ministry, Department of Biomedical Engineering, Zhejiang University, Hangzhou, 310027, China; Innovation Center for Smart Medical Technologies & Devices, Binjiang Institute of Zhejiang University, Hangzhou, Zhejiang, 310053, China
| | - Mengxue Liu
- Biosensor National Special Laboratory, Key Laboratory for Biomedical Engineering of Education Ministry, Department of Biomedical Engineering, Zhejiang University, Hangzhou, 310027, China
| | - Liujing Zhuang
- Biosensor National Special Laboratory, Key Laboratory for Biomedical Engineering of Education Ministry, Department of Biomedical Engineering, Zhejiang University, Hangzhou, 310027, China
| | - Lizhou Xu
- College of Biosystems Engineering and Food Science, Zhejiang University, Hangzhou, 310058, China; ZJU-Hangzhou Global Scientific and Technological Innovation Center, Zhejiang University, Hangzhou, 311215, China.
| | - Ping Wang
- Biosensor National Special Laboratory, Key Laboratory for Biomedical Engineering of Education Ministry, Department of Biomedical Engineering, Zhejiang University, Hangzhou, 310027, China; Innovation Center for Smart Medical Technologies & Devices, Binjiang Institute of Zhejiang University, Hangzhou, Zhejiang, 310053, China.
| |
Collapse
|
5
|
Tiwari A, Hashemiaghdam A, Laramie MA, Maschi D, Haddad T, Stunault MI, Bergom C, Javaheri A, Klyachko V, Ashrafi G. Sirtuin3 ensures the metabolic plasticity of neurotransmission during glucose deprivation. J Cell Biol 2024; 223:e202305048. [PMID: 37988067 PMCID: PMC10660140 DOI: 10.1083/jcb.202305048] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2023] [Revised: 09/18/2023] [Accepted: 10/31/2023] [Indexed: 11/22/2023] Open
Abstract
Neurotransmission is an energetically expensive process that underlies cognition. During intense electrical activity or dietary restrictions, the glucose level in the brain plummets, forcing neurons to utilize alternative fuels. However, the molecular mechanisms of neuronal metabolic plasticity remain poorly understood. Here, we demonstrate that glucose-deprived neurons activate the CREB and PGC1α transcriptional program, which induces expression of the mitochondrial deacetylase Sirtuin 3 (Sirt3) both in vitro and in vivo. We show that Sirt3 localizes to axonal mitochondria and stimulates mitochondrial oxidative capacity in hippocampal nerve terminals. Sirt3 plays an essential role in sustaining synaptic transmission in the absence of glucose by providing metabolic support for the retrieval of synaptic vesicles after release. These results demonstrate that the transcriptional induction of Sirt3 facilitates the metabolic plasticity of synaptic transmission.
Collapse
Affiliation(s)
- Anupama Tiwari
- Department of Cell Biology and Physiology, Washington University School of Medicine, St. Louis, MO, USA
| | - Arsalan Hashemiaghdam
- Department of Cell Biology and Physiology, Washington University School of Medicine, St. Louis, MO, USA
| | - Marissa A. Laramie
- Department of Cell Biology and Physiology, Washington University School of Medicine, St. Louis, MO, USA
| | - Dario Maschi
- Department of Cell Biology and Physiology, Washington University School of Medicine, St. Louis, MO, USA
| | - Tristaan Haddad
- Department of Cell Biology and Physiology, Washington University School of Medicine, St. Louis, MO, USA
| | - Marion I. Stunault
- Department of Cell Biology and Physiology, Washington University School of Medicine, St. Louis, MO, USA
| | - Carmen Bergom
- Department of Radiation Oncology, Washington University School of Medicine, St. Louis, MO, USA
- Alvin J. Siteman Cancer Center, Washington University School of Medicine, St. Louis, MO, USA
| | - Ali Javaheri
- Division of Cardiology, Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA
- John Cochran VA Hospital, St. Louis, MO, USA
| | - Vitaly Klyachko
- Department of Cell Biology and Physiology, Washington University School of Medicine, St. Louis, MO, USA
| | - Ghazaleh Ashrafi
- Department of Cell Biology and Physiology, Washington University School of Medicine, St. Louis, MO, USA
- Needleman Center for Neurometabolism and Axonal Therapeutics, Washington University School of Medicine, St. Louis, MO, USA
| |
Collapse
|
6
|
Myeong J, Stunault MI, Klyachko VA, Ashrafi G. Metabolic Regulation of Single Synaptic Vesicle Exo- and Endocytosis in Hippocampal Synapses. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.11.08.566236. [PMID: 37986894 PMCID: PMC10659320 DOI: 10.1101/2023.11.08.566236] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/22/2023]
Abstract
Glucose has long been considered a primary source of energy for synaptic function. However, it remains unclear under what conditions alternative fuels, such as lactate/pyruvate, contribute to powering synaptic transmission. By detecting individual release events in cultured hippocampal synapses, we found that mitochondrial ATP production from oxidation of lactate/pyruvate regulates basal vesicle release probability and release location within the active zone (AZ) evoked by single action potentials (APs). Mitochondrial inhibition shifted vesicle release closer to the AZ center, suggesting that the energetic barrier for vesicle release is lower in the AZ center that the periphery. Mitochondrial inhibition also altered the efficiency of single AP evoked vesicle retrieval by increasing occurrence of ultrafast endocytosis, while inhibition of glycolysis had no effect. Mitochondria are sparsely distributed along hippocampal axons and we found that nerve terminals containing mitochondria displayed enhanced vesicle release and reuptake during high-frequency trains, irrespective of whether neurons were supplied with glucose or lactate. Thus, synaptic terminals can entirely bypass glycolysis to robustly maintain the vesicle cycle using oxidative fuels in the absence of glucose. These observations further suggest that mitochondrial metabolic function not only regulates several fundamental features of synaptic transmission but may also contribute to modulation of short-term synaptic plasticity.
Collapse
Affiliation(s)
- Jongyun Myeong
- Department of Cell Biology and Physiology, Washington University School of Medicine, St. Louis, MO, 63132, United States
| | - Marion I Stunault
- Department of Cell Biology and Physiology, Washington University School of Medicine, St. Louis, MO, 63132, United States
| | - Vitaly A Klyachko
- Department of Cell Biology and Physiology, Washington University School of Medicine, St. Louis, MO, 63132, United States
| | - Ghazaleh Ashrafi
- Department of Cell Biology and Physiology, Washington University School of Medicine, St. Louis, MO, 63132, United States
- Department of Genetics, Washington University School of Medicine, St. Louis, MO, 63132, United States
| |
Collapse
|
7
|
Tiwari A, Hashemiaghdam A, Laramie MA, Maschi D, Haddad T, Stunault MI, Bergom C, Javaheri A, Klyachko V, Ashrafi G. Sirtuin3 ensures the metabolic plasticity of neurotransmission during glucose deprivation. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.03.08.531724. [PMID: 36945567 PMCID: PMC10028948 DOI: 10.1101/2023.03.08.531724] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/12/2023]
Abstract
Neurotransmission is an energetically expensive process that underlies cognition. During intense electrical activity or dietary restrictions, glucose levels in the brain plummet, forcing neurons to utilize alternative fuels. However, the molecular mechanisms of neuronal metabolic plasticity remain poorly understood. Here, we demonstrate that glucose-deprived neurons activate the CREB and PGC1α transcriptional program that induces the expression of the mitochondrial deacetylase Sirtuin 3 (Sirt3) both in vitro and in vivo . We show that Sirt3 localizes to axonal mitochondria and stimulates mitochondrial oxidative capacity in hippocampal nerve terminals. Sirt3 plays an essential role in sustaining synaptic transmission in the absence of glucose by powering the retrieval of synaptic vesicles after release. These results demonstrate that the transcriptional induction of Sirt3 ensures the metabolic plasticity of synaptic transmission. Highlights Glucose deprivation drives transcriptional reprogramming of neuronal metabolism via CREB and PGC1α. Glucose or food deprivation trigger the neuronal expression of mitochondrial deacetylase sirtuin 3 (Sirt3) both in vitro and in vivo . Sirt3 stimulates oxidative ATP synthesis in nerve terminals.Sirt3 sustains the synaptic vesicle cycle in the absence of glucose.
Collapse
|
8
|
Abstract
Pericytes, attached to the surface of capillaries, play an important role in regulating local blood flow. Using optogenetic tools and genetically encoded reporters in conjunction with confocal and multiphoton imaging techniques, the 3D structure, anatomical organization, and physiology of pericytes have recently been the subject of detailed examination. This work has revealed novel functions of pericytes and morphological features such as tunneling nanotubes in brain and tunneling microtubes in heart. Here, we discuss the state of our current understanding of the roles of pericytes in blood flow control in brain and heart, where functions may differ due to the distinct spatiotemporal metabolic requirements of these tissues. We also outline the novel concept of electro-metabolic signaling, a universal mechanistic framework that links tissue metabolic state with blood flow regulation by pericytes and vascular smooth muscle cells, with capillary KATP and Kir2.1 channels as primary sensors. Finally, we present major unresolved questions and outline how they can be addressed.
Collapse
Affiliation(s)
- Thomas A Longden
- Department of Physiology, University of Maryland School of Medicine, Baltimore, Maryland, USA; ,
- Laboratory of Neurovascular Interactions, Center for Biomedical Engineering and Technology, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Guiling Zhao
- Department of Physiology, University of Maryland School of Medicine, Baltimore, Maryland, USA; ,
- Laboratory of Molecular Cardiology, Center for Biomedical Engineering and Technology, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Ashwini Hariharan
- Department of Physiology, University of Maryland School of Medicine, Baltimore, Maryland, USA; ,
- Laboratory of Neurovascular Interactions, Center for Biomedical Engineering and Technology, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - W Jonathan Lederer
- Department of Physiology, University of Maryland School of Medicine, Baltimore, Maryland, USA; ,
- Laboratory of Molecular Cardiology, Center for Biomedical Engineering and Technology, University of Maryland School of Medicine, Baltimore, Maryland, USA
| |
Collapse
|
9
|
Hariharan A, Robertson CD, Garcia DCG, Longden TA. Brain capillary pericytes are metabolic sentinels that control blood flow through a K ATP channel-dependent energy switch. Cell Rep 2022; 41:111872. [PMID: 36577387 PMCID: PMC10187957 DOI: 10.1016/j.celrep.2022.111872] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2022] [Revised: 10/10/2022] [Accepted: 11/30/2022] [Indexed: 12/28/2022] Open
Abstract
Despite the abundance of capillary thin-strand pericytes and their proximity to neurons and glia, little is known of the contributions of these cells to the control of brain hemodynamics. We demonstrate that the pharmacological activation of thin-strand pericyte KATP channels profoundly hyperpolarizes these cells, dilates upstream penetrating arterioles and arteriole-proximate capillaries, and increases capillary blood flow. Focal stimulation of pericytes with a KATP channel agonist is sufficient to evoke this response, mediated via KIR2.1 channel-dependent retrograde propagation of hyperpolarizing signals, whereas genetic inactivation of pericyte KATP channels eliminates these effects. Critically, we show that decreasing extracellular glucose to less than 1 mM or inhibiting glucose uptake by blocking GLUT1 transporters in vivo flips a mechanistic energy switch driving rapid KATP-mediated pericyte hyperpolarization to increase local blood flow. Together, our findings recast capillary pericytes as metabolic sentinels that respond to local energy deficits by increasing blood flow to neurons to prevent energetic shortfalls.
Collapse
Affiliation(s)
- Ashwini Hariharan
- Department of Physiology, School of Medicine, University of Maryland, Baltimore, MD, USA; Laboratory of Neurovascular Interactions, Center for Biomedical Engineering and Technology, School of Medicine, University of Maryland, Baltimore, MD, USA
| | - Colin D Robertson
- Department of Pharmacology, School of Medicine, University of Maryland, Baltimore, MD, USA
| | - Daniela C G Garcia
- Department of Physiology, School of Medicine, University of Maryland, Baltimore, MD, USA; Laboratory of Neurovascular Interactions, Center for Biomedical Engineering and Technology, School of Medicine, University of Maryland, Baltimore, MD, USA
| | - Thomas A Longden
- Department of Physiology, School of Medicine, University of Maryland, Baltimore, MD, USA; Laboratory of Neurovascular Interactions, Center for Biomedical Engineering and Technology, School of Medicine, University of Maryland, Baltimore, MD, USA.
| |
Collapse
|
10
|
Kuebler IRK, Jolton JA, Hermreck C, Hubbard NA, Wakabayashi KT. Contrasting dose-dependent effects of acute intravenous methamphetamine on lateral hypothalamic extracellular glucose dynamics in male and female rats. J Neurophysiol 2022; 128:819-836. [PMID: 36043803 PMCID: PMC9529272 DOI: 10.1152/jn.00257.2022] [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: 06/13/2022] [Revised: 08/22/2022] [Accepted: 08/24/2022] [Indexed: 11/22/2022] Open
Abstract
Glucose is the brain's primary energetic resource. The brain's use of glucose is dynamic, balancing delivery from the neurovasculature with local metabolism. Although glucose metabolism is known to differ in humans with and without methamphetamine use disorder (MUD), it is unknown how central glucose regulation changes with acute methamphetamine experience. Here, we determined how intravenous methamphetamine regulates extracellular glucose levels in a brain region implicated in MUD-like behavior, the lateral hypothalamus (LH). We measured extracellular LH glucose in awake adult male and female drug-naive Wistar rats using enzyme-linked amperometric glucose biosensors. Changes in LH glucose were monitored during a single session after: 1) natural nondrug stimuli (novel object presentation and a tail-touch), 2) increasing cumulative doses of intravenous methamphetamine (0.025, 0.05, 0.1, and 0.2 mg/kg), and 3) an injection of 60 mg of glucose. We found second-scale fluctuations in LH glucose in response to natural stimuli that differed by both stimulus type and sex. Although rapid, second-scale changes in LH glucose during methamphetamine injections were variable, slow, minute-scale changes following most injections were robust and resulted in a reduction in LH glucose levels. Dose and sex differences at this timescale indicated that female rats may be more sensitive to the impact of methamphetamine on central glucose regulation. These findings suggest that the effects of MUD on healthy brain function may be linked to how methamphetamine alters extracellular glucose regulation in the LH and point to possible mechanisms by which methamphetamine influences central glucose metabolism more broadly.NEW & NOTEWORTHY Enzyme-linked glucose biosensors were used to monitor lateral hypothalamic (LH) extracellular fluctuations during nondrug stimuli and intravenous methamphetamine injections in drug-naive awake male and female rats. Second-scale glucose changes occurred after nondrug stimuli, differing by modality and sex. Robust minute-scale decreases followed most methamphetamine injections. Sex differences at the minute-scale indicate female central glucose regulation is more sensitive to methamphetamine effects. We discuss likely mechanisms underlying these fluctuations, and their implications in methamphetamine use disorder.
Collapse
Affiliation(s)
- Isabel R K Kuebler
- Neurocircuitry of Motivated Behavior Laboratory, Department of Psychology, University of Nebraska-Lincoln, Lincoln, Nebraska
| | - Joshua A Jolton
- Neurocircuitry of Motivated Behavior Laboratory, Department of Psychology, University of Nebraska-Lincoln, Lincoln, Nebraska
| | - Chase Hermreck
- Neurocircuitry of Motivated Behavior Laboratory, Department of Psychology, University of Nebraska-Lincoln, Lincoln, Nebraska
| | - Nicholas A Hubbard
- Neurocircuitry of Motivated Behavior Laboratory, Department of Psychology, University of Nebraska-Lincoln, Lincoln, Nebraska
| | - Ken T Wakabayashi
- Neurocircuitry of Motivated Behavior Laboratory, Department of Psychology, University of Nebraska-Lincoln, Lincoln, Nebraska
| |
Collapse
|
11
|
Aldhshan MS, Jhanji G, Poritsanos NJ, Mizuno TM. Glucose Stimulates Glial Cell Line-Derived Neurotrophic Factor Gene Expression in Microglia through a GLUT5-Independent Mechanism. Int J Mol Sci 2022; 23:ijms23137073. [PMID: 35806073 PMCID: PMC9266953 DOI: 10.3390/ijms23137073] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2022] [Revised: 06/20/2022] [Accepted: 06/24/2022] [Indexed: 01/27/2023] Open
Abstract
Feeding-regulating neurotrophic factors are expressed in both neurons and glial cells. However, nutritional regulation of anorexigenic glial cell line-derived neurotrophic factor (GDNF) and orexigenic mesencephalic astrocyte-derived neurotrophic factor (MANF) expression in specific cell types remains poorly understood. Hypothalamic glucose sensing plays a critical role in the regulation of food intake. It has been theorized that local glucose concentration modulates microglial activity partially via glucose transporter 5 (GLUT5). We hypothesized that an increased local glucose concentration stimulates GDNF expression while inhibiting MANF expression in the hypothalamus and microglia via GLUT5. The present study investigated the effect of glucose on Gdnf and Manf mRNA expression in the mouse hypothalamus and murine microglial cell line SIM-A9. Intracerebroventricular glucose treatment significantly increased Gdnf mRNA levels in the hypothalamus without altering Manf mRNA levels. Exposure to high glucose caused a significant increase in Gdnf mRNA expression and a time-dependent change in Manf mRNA expression in SIM-A9 cells. GLUT5 inhibitor treatment did not block glucose-induced Gdnf mRNA expression in these cells. These findings suggest that microglia are responsive to changes in the local glucose concentration and increased local glucose availability stimulates the expression of microglial GNDF through a GLUT5-independent mechanism, contributing to glucose-induced feeding suppression.
Collapse
|
12
|
Halloun R, Galderisi A, Caprio S, Weiss R. Lack of Evidence for a Causal Role of Hyperinsulinemia in the Progression of Obesity in Children and Adolescents: A Longitudinal Study. Diabetes Care 2022; 45:1400-1407. [PMID: 35235641 PMCID: PMC9210872 DOI: 10.2337/dc21-2210] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/24/2021] [Accepted: 01/23/2022] [Indexed: 02/03/2023]
Abstract
OBJECTIVE The carbohydrate-insulin model (CIM) claims that chronic exposure to hyperinsulinemia induced by dietary carbohydrates explains development of obesity via direct effects of insulin and/or low postprandial metabolic fuel levels. We aimed at testing whether indices of hyperinsulinemia and postprandial glucose levels can predict increases in the degree of obesity over time. RESEARCH DESIGN AND METHODS Children and adolescents with obesity attending a pediatric obesity clinic performed oral glucose tolerance tests (OGTTs) and received standard obesity management. Indices of hyperinsulinemia and insulin secretion were derived from the OGTT and evaluated in the face of changes in the degree of obesity over time. RESULTS A total of 591 children (217 males and 374 females) participated, and the mean follow-up was 1.86 ± 1.29 years. OGTT-derived area under the curve of insulin, peak insulin, fasting insulin, the insulinogenic index, or insulin at 30 min were not associated with greater changes in the degree of obesity in univariate or multivariate analyses (adjusted for baseline age, BMI z score, sex, and ethnicity). Low postprandial glucose <75 mg/dL was not associated with greater changes in the degree of obesity in univariate or multivariate analyses. In a subsample of 104 participants with a follow-up >4 years, none of these parameters was associated with greater increases in the degree of obesity. CONCLUSIONS In children and adolescents with obesity, exposure to hyperinsulinemia, greater insulin secretion, or low postprandial glucose is not associated with greater increases in the degree of obesity over 2-4 years. The CIM should be evaluated in children with lower BMI and for longer follow-up periods.
Collapse
Affiliation(s)
- Rana Halloun
- Department of Pediatrics, Ruth Children’s Hospital, Rambam Medical Center, Haifa, Israel
| | - Alfonso Galderisi
- Department of Woman and Child’s Health, University of Padova, Padova, Italy
- Department of Pediatrics, Yale University, New Haven, CT
| | - Sonia Caprio
- Department of Pediatrics, Yale University, New Haven, CT
| | - Ram Weiss
- Department of Pediatrics, Ruth Children’s Hospital, Rambam Medical Center, Haifa, Israel
| |
Collapse
|
13
|
Tu L, Fukuda M, Tong Q, Xu Y. The ventromedial hypothalamic nucleus: watchdog of whole-body glucose homeostasis. Cell Biosci 2022; 12:71. [PMID: 35619170 PMCID: PMC9134642 DOI: 10.1186/s13578-022-00799-2] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2021] [Accepted: 04/25/2022] [Indexed: 02/06/2023] Open
Abstract
The brain, particularly the ventromedial hypothalamic nucleus (VMH), has been long known for its involvement in glucose sensing and whole-body glucose homeostasis. However, it is still not fully understood how the brain detects and responds to the changes in the circulating glucose levels, as well as brain-body coordinated control of glucose homeostasis. In this review, we address the growing evidence implicating the brain in glucose homeostasis, especially in the contexts of hypoglycemia and diabetes. In addition to neurons, we emphasize the potential roles played by non-neuronal cells, as well as extracellular matrix in the hypothalamus in whole-body glucose homeostasis. Further, we review the ionic mechanisms by which glucose-sensing neurons sense fluctuations of ambient glucose levels. We also introduce the significant implications of heterogeneous neurons in the VMH upon glucose sensing and whole-body glucose homeostasis, in which sex difference is also addressed. Meanwhile, research gaps have also been identified, which necessities further mechanistic studies in future.
Collapse
Affiliation(s)
- Longlong Tu
- USDA/ARS Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, 1100 Bates Street #8066, Houston, TX, 77030, USA
| | - Makoto Fukuda
- USDA/ARS Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, 1100 Bates Street #8066, Houston, TX, 77030, USA
| | - Qingchun Tong
- Brown Foundation Institute of Molecular Medicine, University of Texas Health Science Center at Houston, Houston, TX, 77030, USA
| | - Yong Xu
- USDA/ARS Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, 1100 Bates Street #8066, Houston, TX, 77030, USA.
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, 77030, USA.
| |
Collapse
|
14
|
Ren J, Hu H, Wang S, He Y, Ji Y, Chen Y, Wang K, Zhang H, Zhao Y, Dai F. Prevent Drug Leakage via the Boronic Acid Glucose-Insensitive Micelle for Alzheimer's Disease Combination Treatment. ACS APPLIED MATERIALS & INTERFACES 2022; 14:23182-23193. [PMID: 35544753 DOI: 10.1021/acsami.2c03684] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
Boronic acid (BA) materials have been widely applied to glucose and oxidative stress-sensitive drug delivery for the treatment of cancer, diabetes, and Alzheimer's disease (AD). There are completely various BA-sensitive delivery conditions in different diseases. BA materials in the treatment of diabetes show better performance at a high-glucose environment than normal. In contrast, the concentration of glucose in the brain is much lower than that in the blood of AD patients. Hence, the typical glucose and oxidative stress dual-sensitive BA materials inevitably encounter drug leakage in circulation in AD. Attempts to decrease the glucose-sensitive capacity of BA materials are extremely essential for AD drug delivery. In this study, the epoxy group (electron-donating group) was introduced to increase the pKa values of BA materials by increasing the electron cloud density, and thus, the glucose-insensitive micelle (GIM) was obtained. The treatment effect and the synergism mechanism of the drug-loaded GIM micelle were studied on senescence-accelerated mouse prone 8 mice. This work provided excellent antioxidant drugs (vitamin E succinate, melatonin, and quercetin) and a glucose metabolism drug (insulin) loaded in GIM micelle for AD treatment. The discovery of the combination mechanism is enormously valuable for AD clinical research.
Collapse
Affiliation(s)
- Jian Ren
- State Key Laboratory of Separation Membranes and Membrane Processes, National Center for International Joint Research on Separation Membranes, School of Material Science and Engineering, Tiangong University, Tianjin 300387, China
| | - Haodong Hu
- State Key Laboratory of Separation Membranes and Membrane Processes, National Center for International Joint Research on Separation Membranes, School of Material Science and Engineering, Tiangong University, Tianjin 300387, China
| | - Shaoteng Wang
- State Key Laboratory of Separation Membranes and Membrane Processes, National Center for International Joint Research on Separation Membranes, School of Material Science and Engineering, Tiangong University, Tianjin 300387, China
| | - Yang He
- State Key Laboratory of Separation Membranes and Membrane Processes, National Center for International Joint Research on Separation Membranes, School of Material Science and Engineering, Tiangong University, Tianjin 300387, China
| | - Yanhong Ji
- State Key Laboratory of Separation Membranes and Membrane Processes, National Center for International Joint Research on Separation Membranes, School of Material Science and Engineering, Tiangong University, Tianjin 300387, China
| | - Yiran Chen
- State Key Laboratory of Separation Membranes and Membrane Processes, National Center for International Joint Research on Separation Membranes, School of Material Science and Engineering, Tiangong University, Tianjin 300387, China
| | - Kangna Wang
- State Key Laboratory of Separation Membranes and Membrane Processes, National Center for International Joint Research on Separation Membranes, School of Material Science and Engineering, Tiangong University, Tianjin 300387, China
| | - Haiyan Zhang
- State Key Laboratory of Separation Membranes and Membrane Processes, National Center for International Joint Research on Separation Membranes, School of Material Science and Engineering, Tiangong University, Tianjin 300387, China
| | - Yiping Zhao
- State Key Laboratory of Separation Membranes and Membrane Processes, National Center for International Joint Research on Separation Membranes, School of Material Science and Engineering, Tiangong University, Tianjin 300387, China
| | - Fengying Dai
- State Key Laboratory of Separation Membranes and Membrane Processes, National Center for International Joint Research on Separation Membranes, School of Material Science and Engineering, Tiangong University, Tianjin 300387, China
| |
Collapse
|
15
|
Watts AG, Kanoski SE, Sanchez-Watts G, Langhans W. The physiological control of eating: signals, neurons, and networks. Physiol Rev 2022; 102:689-813. [PMID: 34486393 PMCID: PMC8759974 DOI: 10.1152/physrev.00028.2020] [Citation(s) in RCA: 69] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2020] [Accepted: 08/30/2021] [Indexed: 02/07/2023] Open
Abstract
During the past 30 yr, investigating the physiology of eating behaviors has generated a truly vast literature. This is fueled in part by a dramatic increase in obesity and its comorbidities that has coincided with an ever increasing sophistication of genetically based manipulations. These techniques have produced results with a remarkable degree of cell specificity, particularly at the cell signaling level, and have played a lead role in advancing the field. However, putting these findings into a brain-wide context that connects physiological signals and neurons to behavior and somatic physiology requires a thorough consideration of neuronal connections: a field that has also seen an extraordinary technological revolution. Our goal is to present a comprehensive and balanced assessment of how physiological signals associated with energy homeostasis interact at many brain levels to control eating behaviors. A major theme is that these signals engage sets of interacting neural networks throughout the brain that are defined by specific neural connections. We begin by discussing some fundamental concepts, including ones that still engender vigorous debate, that provide the necessary frameworks for understanding how the brain controls meal initiation and termination. These include key word definitions, ATP availability as the pivotal regulated variable in energy homeostasis, neuropeptide signaling, homeostatic and hedonic eating, and meal structure. Within this context, we discuss network models of how key regions in the endbrain (or telencephalon), hypothalamus, hindbrain, medulla, vagus nerve, and spinal cord work together with the gastrointestinal tract to enable the complex motor events that permit animals to eat in diverse situations.
Collapse
Affiliation(s)
- Alan G Watts
- The Department of Biological Sciences, Dornsife College of Letters, Arts and Sciences, University of Southern California, Los Angeles, California
| | - Scott E Kanoski
- The Department of Biological Sciences, Dornsife College of Letters, Arts and Sciences, University of Southern California, Los Angeles, California
| | - Graciela Sanchez-Watts
- The Department of Biological Sciences, Dornsife College of Letters, Arts and Sciences, University of Southern California, Los Angeles, California
| | - Wolfgang Langhans
- Physiology and Behavior Laboratory, Eidgenössische Technische Hochschule-Zürich, Schwerzenbach, Switzerland
| |
Collapse
|
16
|
Maunze B, Bruckner KW, Desai NN, Chen C, Chen F, Baker D, Choi S. Pituitary adenylate cyclase-activating polypeptide receptor activation in the hypothalamus recruits unique signaling pathways involved in energy homeostasis. Am J Physiol Endocrinol Metab 2022; 322:E199-E210. [PMID: 35001657 PMCID: PMC8897015 DOI: 10.1152/ajpendo.00320.2021] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Pituitary adenylate cyclase activating polypeptide (PACAP) exerts pleiotropic effects on ventromedial nuclei (VMN) of the hypothalamus and its control of feeding and energy expenditure through the type I PAC1 receptor (PAC1R). However, the endogenous role of PAC1Rs in the VMN and the downstream signaling responsible for PACAP's effects on energy balance are unknown. Numerous studies have revealed that PAC1Rs are coupled to both Gαs/adenylyl cyclase/protein kinase A (Gαs/AC/PKA) and Gαq/phospholipase C/protein kinase C (Gαq/PLC/PKC), while also undergoing trafficking following stimulation. To determine the endogenous role of PAC1Rs and downstream signaling that may explain PACAP's pleiotropic effects, we used RNA interference to knockdown VMN PAC1Rs and pharmacologically inhibited PKA, PKC, and PAC1R trafficking. Knocking down PAC1Rs increased meal sizes, reduced total number of meals, and induced body weight gain. Inhibition of either PKA or PKC alone in awake male Sprague-Dawley rats, attenuated PACAP's hypophagic and anorectic effects during the dark phase. However, PKA or PKC inhibition potentiated PACAP's thermogenic effects during the light phase. Analysis of locomotor activity revealed that PKA inhibition augmented PACAP's locomotor effects, whereas PKC inhibition had no effect. Finally, PACAP administration in the VMN induces surface PAC1R trafficking into the cytosol which was blocked by endocytosis inhibitors. Subsequently, inhibition of PAC1R trafficking into the cytosol attenuated PACAP-induced hypophagia. These results revealed that endogenous PAC1Rs uniquely engage PKA, PKC, and receptor trafficking to mediate PACAP's pleiotropic effects in VMN control of feeding and metabolism.NEW & NOTEWORTHY Endogenous PAC1 receptors, integral to VMN management of feeding behavior and body weight regulation, uniquely engage PKA, PKC, and receptor trafficking to mediate the hypothalamic ventromedial nuclei control of feeding and metabolism. PACAP appears to use different signaling mechanisms to regulate feeding behavior from its effects on metabolism.
Collapse
Affiliation(s)
- Brian Maunze
- Department of Biomedical Sciences, Marquette University, Milwaukee, Wisconsin
| | | | - Nikhil Nilesh Desai
- Department of Biomedical Sciences, Marquette University, Milwaukee, Wisconsin
| | - Christopher Chen
- Department of Biomedical Sciences, Marquette University, Milwaukee, Wisconsin
| | - Fanghong Chen
- Department of Biomedical Sciences, Marquette University, Milwaukee, Wisconsin
| | - David Baker
- Department of Biomedical Sciences, Marquette University, Milwaukee, Wisconsin
| | - SuJean Choi
- Department of Biomedical Sciences, Marquette University, Milwaukee, Wisconsin
| |
Collapse
|
17
|
Analysis of the Glucose-Dependent Transcriptome in Murine Hypothalamic Cells. Cells 2022; 11:cells11040639. [PMID: 35203289 PMCID: PMC8870115 DOI: 10.3390/cells11040639] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2022] [Revised: 02/10/2022] [Accepted: 02/10/2022] [Indexed: 11/16/2022] Open
Abstract
Glucose provides vital energy for cells and contributes to gene expression. The hypothalamus is key for metabolic homeostasis, but effects of glucose on hypothalamic gene expression have not yet been investigated in detail. Thus, herein, we monitored the glucose-dependent transcriptome in murine hypothalamic mHypoA-2/10 cells by total RNA-seq analysis. A total of 831 genes were up- and 1390 genes were downregulated by at least 50%. Key genes involved in the cholesterol biosynthesis pathway were upregulated, and total cellular cholesterol levels were significantly increased by glucose. Analysis of single genes involved in fundamental cellular signaling processes also suggested a significant impact of glucose. Thus, we chose ≈100 genes involved in signaling and validated the effects of glucose on mRNA levels by qRT-PCR. We identified Gnai1–3, Adyc6, Irs1, Igfr1, Hras, and Elk3 as new glucose-dependent genes. In line with this, cAMP measurements revealed enhanced noradrenalin-induced cAMP levels, and reporter gene assays elevated activity of the insulin-like growth factor at higher glucose levels. Key data of our studies were confirmed in a second hypothalamic cell line. Thus, our findings link extra cellular glucose levels with hypothalamic lipid synthesis and pivotal intracellular signaling processes, which might be of particular interest in situations of continuously increased glucose levels.
Collapse
|
18
|
Lhomme T, Clasadonte J, Imbernon M, Fernandois D, Sauve F, Caron E, Lima N, Heras V, Martinez-Corral I, Müller-Fielitz H, Rasika S, Schwaninger M, Nogueiras R, Prevot V. Tanycytic networks mediate energy balance by feeding lactate to glucose-insensitive POMC neurons. J Clin Invest 2021; 131:e140521. [PMID: 34324439 DOI: 10.1172/jci140521] [Citation(s) in RCA: 51] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2020] [Accepted: 07/28/2021] [Indexed: 11/17/2022] Open
Abstract
Hypothalamic glucose sensing enables an organism to match energy expenditure and food intake to circulating levels of glucose, the main energy source of the brain. Here, we established that tanycytes of the hypothalamic arcuate nucleus, specialized glia that line the wall of the third ventricle, convert brain glucose supplies into lactate that they transmit through monocarboxylate transporters to arcuate proopiomelanocortin neurons, which integrate this signal to drive their activity and to adapt the metabolic response to meet physiological demands. Furthermore, this transmission required the formation of extensive Connexin-43 gap-junction-mediated metabolic networks by arcuate tanycytes. Selectively suppressing either tanycytic monocarboxylate transporters or gap junctions resulted in altered feeding behavior and energy metabolism. Tanycytic intercellular communication and lactate production are thus integral to the mechanism by which hypothalamic neurons that regulate energy and glucose homeostasis efficiently perceive alterations in systemic glucose levels as a function of the physiological state of the organism.
Collapse
Affiliation(s)
- Tori Lhomme
- Laboratory of Development and Plasticity of the Neuroendocrine Brain, INSERM U1172, Lille, France
| | - Jerome Clasadonte
- Laboratory of Development and Plasticity of the Neuroendocrine Brain, INSERM U1172, Lille, France
| | - Monica Imbernon
- Laboratory of Development and Plasticity of the Neuroendocrine Brain, INSERM U1172, Lille, France
| | - Daniela Fernandois
- Laboratory of Development and Plasticity of the Neuroendocrine Brain, INSERM U1172, Lille, France
| | - Florent Sauve
- Laboratory of Development and Plasticity of the Neuroendocrine Brain, INSERM U1172, Lille, France
| | - Emilie Caron
- Laboratory of Development and Plasticity of the Neuroendocrine Brain, INSERM U1172, Lille, France
| | - Natalia Lima
- CIMUS, University of Santiago de Compostela, Santiago de Compostela, Spain
| | - Violeta Heras
- CIMUS, University of Santiago de Compostela, Santiago de Compostela, Spain
| | - Ines Martinez-Corral
- Laboratory of Development and Plasticity of the Neuroendocrine Brain, INSERM U1172, Lille, France
| | - Helge Müller-Fielitz
- Institute for Experimental and Clinical Pharmacology and Toxicology, University of Lübeck, Lübeck, Germany
| | - S Rasika
- Laboratory of Development and Plasticity of the Neuroendocrine Brain, INSERM U1172, Lille, France
| | - Markus Schwaninger
- Institute for Experimental and Clinical Pharmacology and Toxicology, University of Lübeck, Lübeck, Germany
| | - Ruben Nogueiras
- CIMUS, University of Santiago de Compostela, Santiago de Compostela, Spain
| | - Vincent Prevot
- Laboratory of Development and Plasticity of the Neuroendocrine Brain, INSERM U1172, Lille, France
| |
Collapse
|
19
|
Myers MG, Affinati AH, Richardson N, Schwartz MW. Central nervous system regulation of organismal energy and glucose homeostasis. Nat Metab 2021; 3:737-750. [PMID: 34158655 DOI: 10.1038/s42255-021-00408-5] [Citation(s) in RCA: 82] [Impact Index Per Article: 20.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/17/2021] [Accepted: 05/12/2021] [Indexed: 02/05/2023]
Abstract
Growing evidence implicates the brain in the regulation of both immediate fuel availability (for example, circulating glucose) and long-term energy stores (that is, adipose tissue mass). Rather than viewing the adipose tissue and glucose control systems separately, we suggest that the brain systems that control them are components of a larger, highly integrated, 'fuel homeostasis' control system. This conceptual framework, along with new insights into the organization and function of distinct neuronal systems, provides a context within which to understand how metabolic homeostasis is achieved in both basal and postprandial states. We also review evidence that dysfunction of the central fuel homeostasis system contributes to the close association between obesity and type 2 diabetes, with the goal of identifying more effective treatment options for these common metabolic disorders.
Collapse
Affiliation(s)
- Martin G Myers
- Departments of Medicine and Molecular & Integrative Physiology, University of Michigan, Ann Arbor, MI, USA
| | - Alison H Affinati
- Departments of Medicine and Molecular & Integrative Physiology, University of Michigan, Ann Arbor, MI, USA
| | - Nicole Richardson
- UW Medicine Diabetes Institute, Department of Medicine, University of Washington, Seattle, WA, USA
| | - Michael W Schwartz
- UW Medicine Diabetes Institute, Department of Medicine, University of Washington, Seattle, WA, USA.
| |
Collapse
|
20
|
Mannal N, Kleiner K, Fauler M, Dougalis A, Poetschke C, Liss B. Multi-Electrode Array Analysis Identifies Complex Dopamine Responses and Glucose Sensing Properties of Substantia Nigra Neurons in Mouse Brain Slices. Front Synaptic Neurosci 2021; 13:635050. [PMID: 33716704 PMCID: PMC7952765 DOI: 10.3389/fnsyn.2021.635050] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2020] [Accepted: 01/08/2021] [Indexed: 12/16/2022] Open
Abstract
Dopaminergic (DA) midbrain neurons within the substantia nigra (SN) display an autonomous pacemaker activity that is crucial for dopamine release and voluntary movement control. Their progressive degeneration is a hallmark of Parkinson's disease. Their metabolically demanding activity-mode affects Ca2+ homeostasis, elevates metabolic stress, and renders SN DA neurons particularly vulnerable to degenerative stressors. Accordingly, their activity is regulated by complex mechanisms, notably by dopamine itself, via inhibitory D2-autoreceptors and the neuroprotective neuronal Ca2+ sensor NCS-1. Analyzing regulation of SN DA neuron activity-pattern is complicated by their high vulnerability. We studied this activity and its control by dopamine, NCS-1, and glucose with extracellular multi-electrode array (MEA) recordings from midbrain slices of juvenile and adult mice. Our tailored MEA- and spike sorting-protocols allowed high throughput and long recording times. According to individual dopamine-responses, we identified two distinct SN cell-types, in similar frequency: dopamine-inhibited and dopamine-excited neurons. Dopamine-excited neurons were either silent in the absence of dopamine, or they displayed pacemaker-activities, similar to that of dopamine-inhibited neurons. Inhibition of pacemaker-activity by dopamine is typical for SN DA neurons, and it can undergo prominent desensitization. We show for adult mice, that the number of SN DA neurons with desensitized dopamine-inhibition was increased (~60–100%) by a knockout of NCS-1, or by prevention of NCS-1 binding to D2-autoreceptors, while time-course and degrees of desensitization were not altered. The number of neurons with desensitized D2-responses was also higher (~65%) at high glucose-levels (25 mM), compared to lower glucose (2.5 mM), while again desensitization-kinetics were unaltered. However, spontaneous firing-rates were significantly higher at high glucose-levels (~20%). Moreover, transient glucose-deprivation (1 mM) induced a fast and fully-reversible pacemaker frequency reduction. To directly address and quantify glucose-sensing properties of SN DA neurons, we continuously monitored their electrical activity, while altering extracellular glucose concentrations stepwise from 0.5 mM up to 25 mM. SN DA neurons were excited by glucose, with EC50 values ranging from 0.35 to 2.3 mM. In conclusion, we identified a novel, common subtype of dopamine-excited SN neurons, and a complex, joint regulation of dopamine-inhibited neurons by dopamine and glucose, within the range of physiological brain glucose-levels.
Collapse
Affiliation(s)
- Nadja Mannal
- Institute of Applied Physiology, University of Ulm, Ulm, Germany
| | | | - Michael Fauler
- Institute of Applied Physiology, University of Ulm, Ulm, Germany
| | | | | | - Birgit Liss
- Institute of Applied Physiology, University of Ulm, Ulm, Germany.,Linacre and New College, University of Oxford, Oxford, United Kingdom
| |
Collapse
|
21
|
Yang S, Qin C, Hu ZW, Zhou LQ, Yu HH, Chen M, Bosco DB, Wang W, Wu LJ, Tian DS. Microglia reprogram metabolic profiles for phenotype and function changes in central nervous system. Neurobiol Dis 2021; 152:105290. [PMID: 33556540 DOI: 10.1016/j.nbd.2021.105290] [Citation(s) in RCA: 125] [Impact Index Per Article: 31.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2020] [Revised: 12/31/2020] [Accepted: 02/03/2021] [Indexed: 12/13/2022] Open
Abstract
In response to various types of environmental and cellular stress, microglia rapidly activate and exhibit either pro- or anti-inflammatory phenotypes to maintain tissue homeostasis. Activation of microglia can result in changes in morphology, phagocytosis capacity, and secretion of cytokines. Furthermore, microglial activation also induces changes to cellular energy demand, which is dependent on the metabolism of various metabolic substrates including glucose, fatty acids, and amino acids. Accumulating evidence demonstrates metabolic reprogramming acts as a key driver of microglial immune response. For instance, microglia in pro-inflammatory states preferentially use glycolysis for energy production, whereas, cells in anti-inflammatory states are mainly powered by oxidative phosphorylation and fatty acid oxidation. In this review, we summarize recent findings regarding microglial metabolic pathways under physiological and pathological circumtances. We will then discuss how metabolic reprogramming can orchestrate microglial response to a variety of central nervous system pathologies. Finally, we highlight how manipulating metabolic pathways can reprogram microglia towards beneficial functions, and illustrate the therapeutic potential for inflammation-related neurological diseases.
Collapse
Affiliation(s)
- Sheng Yang
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Chuan Qin
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Zi-Wei Hu
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Luo-Qi Zhou
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Hai-Han Yu
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Man Chen
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Dale B Bosco
- Department of Neurology, Mayo Clinic, Rochester, MN 55905, United States of America
| | - Wei Wang
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Long-Jun Wu
- Department of Neurology, Mayo Clinic, Rochester, MN 55905, United States of America.
| | - Dai-Shi Tian
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China.
| |
Collapse
|
22
|
Yu K, He Y, Hyseni I, Pei Z, Yang Y, Xu P, Cai X, Liu H, Qu N, Liu H, He Y, Yu M, Liang C, Yang T, Wang J, Gourdy P, Arnal JF, Lenfant F, Xu Y, Wang C. 17β-estradiol promotes acute refeeding in hungry mice via membrane-initiated ERα signaling. Mol Metab 2020; 42:101053. [PMID: 32712433 PMCID: PMC7484552 DOI: 10.1016/j.molmet.2020.101053] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/09/2020] [Revised: 07/17/2020] [Accepted: 07/19/2020] [Indexed: 12/20/2022] Open
Abstract
Objective Estrogen protects animals from obesity through estrogen receptor α (ERα), partially by inhibiting overeating in animals fed ad libitum. However, the effects of estrogen on feeding behavior in hungry animals remain unclear. In this study, we examined the roles of 17β-estradiol (E2) and ERα in the regulation of feeding in hungry female animals and explored the underlying mechanisms. Methods Wild-type female mice with surgical depletion of endogenous estrogens were used to examine the effects of E2 supplementation on acute refeeding behavior after starvation. ERα-C451A mutant mice deficient in membrane-bound ERα activity and ERα-AF20 mutant mice lacking ERα transcriptional activity were used to further examine mechanisms underlying acute feeding triggered by either fasting or central glucopenia (induced by intracerebroventricular injections of 2-deoxy-D-glucose). We also used electrophysiology to explore the impact of these ERα mutations on the neural activities of ERα neurons in the hypothalamus. Results In the wild-type female mice, ovariectomy reduced fasting-induced refeeding, which was restored by E2 supplementation. The ERα-C451A mutation, but not the ERα-AF20 mutation, attenuated acute feeding induced by either fasting or central glucopenia. The ERα-C451A mutation consistently impaired the neural responses of hypothalamic ERα neurons to hypoglycemia. Conclusion In addition to previous evidence that estrogen reduces deviations in energy balance by inhibiting eating at a satiated state, our findings demonstrate the unexpected role of E2 that promotes eating in hungry mice, also contributing to the stability of energy homeostasis. This latter effect specifically requires membrane-bound ERα activity. Endogenous E2 is required to maintain acute refeeding in hungry female mice after starvation. Membrane-bound ERα activity in female mice is required for efficient refeeding after starvation. Membrane-bound ERα activity is required for hypothalamic ERα neurons to respond to hypoglycemia.
Collapse
Affiliation(s)
- Kaifan Yu
- USDA/ARS Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, One Baylor Plaza, Houston, TX, 77030, USA; College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, Jiangsu, 210095, China
| | - Yanlin He
- USDA/ARS Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, One Baylor Plaza, Houston, TX, 77030, USA; Pennington Biomedical Research Center, Brain Glycemic and Metabolism Control Department, Louisiana State University, Baton Rouge, LA, 70808, USA
| | - Ilirjana Hyseni
- USDA/ARS Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, One Baylor Plaza, Houston, TX, 77030, USA
| | - Zhou Pei
- USDA/ARS Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, One Baylor Plaza, Houston, TX, 77030, USA
| | - Yongjie Yang
- USDA/ARS Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, One Baylor Plaza, Houston, TX, 77030, USA
| | - Pingwen Xu
- USDA/ARS Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, One Baylor Plaza, Houston, TX, 77030, USA
| | - Xing Cai
- USDA/ARS Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, One Baylor Plaza, Houston, TX, 77030, USA
| | - Hesong Liu
- USDA/ARS Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, One Baylor Plaza, Houston, TX, 77030, USA
| | - Na Qu
- USDA/ARS Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, One Baylor Plaza, Houston, TX, 77030, USA
| | - Hailan Liu
- USDA/ARS Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, One Baylor Plaza, Houston, TX, 77030, USA
| | - Yang He
- USDA/ARS Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, One Baylor Plaza, Houston, TX, 77030, USA
| | - Meng Yu
- USDA/ARS Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, One Baylor Plaza, Houston, TX, 77030, USA
| | - Chen Liang
- USDA/ARS Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, One Baylor Plaza, Houston, TX, 77030, USA
| | - Tingting Yang
- USDA/ARS Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, One Baylor Plaza, Houston, TX, 77030, USA
| | - Julia Wang
- USDA/ARS Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, One Baylor Plaza, Houston, TX, 77030, USA
| | - Pierre Gourdy
- I2MC, Inserm U1048, CHU de Toulouse and Université de Toulouse III, Toulouse, France
| | - Jean-Francois Arnal
- I2MC, Inserm U1048, CHU de Toulouse and Université de Toulouse III, Toulouse, France
| | - Francoise Lenfant
- I2MC, Inserm U1048, CHU de Toulouse and Université de Toulouse III, Toulouse, France
| | - Yong Xu
- USDA/ARS Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, One Baylor Plaza, Houston, TX, 77030, USA; Department of Molecular and Cellular Biology, Baylor College of Medicine, One Baylor Plaza, Houston, TX, 77030, USA.
| | - Chunmei Wang
- USDA/ARS Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, One Baylor Plaza, Houston, TX, 77030, USA.
| |
Collapse
|
23
|
Hanna L, Kawalek TJ, Beall C, Ellacott KLJ. Changes in neuronal activity across the mouse ventromedial nucleus of the hypothalamus in response to low glucose: Evaluation using an extracellular multi-electrode array approach. J Neuroendocrinol 2020; 32:e12824. [PMID: 31880369 PMCID: PMC7064989 DOI: 10.1111/jne.12824] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/17/2018] [Revised: 12/04/2019] [Accepted: 12/23/2019] [Indexed: 01/01/2023]
Abstract
The hypothalamic ventromedial nucleus (VMN) is involved in maintaining systemic glucose homeostasis. Neurophysiological studies in rodent brain slices have identified populations of VMN glucose-sensing neurones: glucose-excited (GE) neurones, cells which increased their firing rate in response to increases in glucose concentration, and glucose-inhibited (GI) neurones, which show a reduced firing frequency in response to increasing glucose concentrations. To date, most slice electrophysiological studies characterising VMN glucose-sensing neurones in rodents have utilised the patch clamp technique. Multi-electrode arrays (MEAs) are a state-of-the-art electrophysiological tool enabling the electrical activity of many cells to be recorded across multiple electrode sites (channels) simultaneously. We used a perforated MEA (pMEA) system to evaluate electrical activity changes across the dorsal-ventral extent of the mouse VMN region in response to alterations in glucose concentration. Because intrinsic (ie, direct postsynaptic sensing) and extrinsic (ie, presynaptically modulated) glucosensation were not discriminated, we use the terminology 'GE/presynaptically excited by an increase (PER)' and 'GI/presynaptically excited by a decrease (PED)' in the present study to describe responsiveness to changes in extracellular glucose across the mouse VMN. We observed that 15%-60% of channels were GE/PER, whereas 2%-7% were GI/PED channels. Within the dorsomedial portion of the VMN (DM-VMN), significantly more channels were GE/PER compared to the ventrolateral portion of the VMN (VL-VMN). However, GE/PER channels within the VL-VMN showed a significantly higher basal firing rate in 2.5 mmol l-1 glucose than DM-VMN GE/PER channels. No significant difference in the distribution of GI/PED channels was observed between the VMN subregions. The results of the present study demonstrate the utility of the pMEA approach for evaluating glucose responsivity across the mouse VMN. pMEA studies could be used to refine our understanding of other neuroendocrine systems by examining population level changes in electrical activity across brain nuclei, thus providing key functional neuroanatomical information to complement and inform the design of single-cell neurophysiological studies.
Collapse
Affiliation(s)
- Lydia Hanna
- Reading School of PharmacyUniversity of ReadingReadingUK
- Institute of Biomedical & Clinical SciencesUniversity of Exeter Medical SchoolExeterUK
- Present address:
Department of Biological SciencesCentre for Biomedical SciencesRoyal Holloway University of LondonEghamUK
| | - Tristan J. Kawalek
- Institute of Biomedical & Clinical SciencesUniversity of Exeter Medical SchoolExeterUK
| | - Craig Beall
- Institute of Biomedical & Clinical SciencesUniversity of Exeter Medical SchoolExeterUK
| | - Kate L. J. Ellacott
- Institute of Biomedical & Clinical SciencesUniversity of Exeter Medical SchoolExeterUK
| |
Collapse
|
24
|
Kiyatkin EA. Brain temperature and its role in physiology and pathophysiology: Lessons from 20 years of thermorecording. Temperature (Austin) 2019; 6:271-333. [PMID: 31934603 PMCID: PMC6949027 DOI: 10.1080/23328940.2019.1691896] [Citation(s) in RCA: 53] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2019] [Revised: 10/29/2019] [Accepted: 10/30/2019] [Indexed: 12/11/2022] Open
Abstract
It is well known that temperature affects the dynamics of all physicochemical processes governing neural activity. It is also known that the brain has high levels of metabolic activity, and all energy used for brain metabolism is finally transformed into heat. However, the issue of brain temperature as a factor reflecting neural activity and affecting various neural functions remains in the shadow and is usually ignored by most physiologists and neuroscientists. Data presented in this review demonstrate that brain temperature is not stable, showing relatively large fluctuations (2-4°C) within the normal physiological and behavioral continuum. I consider the mechanisms underlying these fluctuations and discuss brain thermorecording as an important tool to assess basic changes in neural activity associated with different natural (sexual, drinking, eating) and drug-induced motivated behaviors. I also consider how naturally occurring changes in brain temperature affect neural activity, various homeostatic parameters, and the structural integrity of brain cells as well as the results of neurochemical evaluations conducted in awake animals. While physiological hyperthermia appears to be adaptive, enhancing the efficiency of neural functions, under specific environmental conditions and following exposure to certain psychoactive drugs, brain temperature could exceed its upper limits, resulting in multiple brain abnormalities and life-threatening health complications.
Collapse
Affiliation(s)
- Eugene A Kiyatkin
- Behavioral Neuroscience Branch, National Institute on Drug Abuse - Intramural Research Program, National Institutes of Health, Baltimore, MD, USA
| |
Collapse
|
25
|
Garduño J, Hernández-López S, Rolón DC, de la Cruz L, Hernández-Vázquez F, Reyes-Vaca A, Arenas I, Bravo-Martínez J, Garcia DE. Electrophysiological characterization of glucose sensing neurons in the hypothalamic arcuate nucleus of male rats. Neurosci Lett 2019; 703:168-176. [PMID: 30926375 DOI: 10.1016/j.neulet.2019.03.041] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2018] [Revised: 03/14/2019] [Accepted: 03/25/2019] [Indexed: 12/01/2022]
Abstract
The arcuate nucleus (ARC), located at the base of hypothalamus, contains two main populations of neurons involved in the regulation of food intake and energy expenditure. The NPY neurons are orexigenic and their activation stimulates food intake while the activation of POMC neurons promote the opposite effect. Several works have tried to identify these neurons based on their electrophysiological and pharmacological characteristics. However, the classification of ARC neurons is still inconclusive. In this work, glucose concentrations were changed within at physiological range, and the response of rat ARC neurons to this stimulus was used to identify them. Subsequently, the cells were classified on the basis of their passive and active electrophysiological properties. Finally, calcium imaging experiments were done to study the response of ARC neurons populations changing glucose concentrations. We found that NPY and putative POMC neurons can be distinguished based on their electrophysiological properties such as input resistance and firing pattern. Calcium imaging experiments confirmed the diversity of ARC neurons.
Collapse
Affiliation(s)
- Julieta Garduño
- Departamento de Fisiología, Facultad de Medicina, Universidad Nacional Autónoma de México (UNAM), PO Box 70250, Ciudad de México, 04510, Mexico.
| | - Salvador Hernández-López
- Departamento de Fisiología, Facultad de Medicina, Universidad Nacional Autónoma de México (UNAM), PO Box 70250, Ciudad de México, 04510, Mexico
| | - Diego Castillo Rolón
- Departamento de Fisiología, Facultad de Medicina, Universidad Nacional Autónoma de México (UNAM), PO Box 70250, Ciudad de México, 04510, Mexico
| | - Lizbeth de la Cruz
- Departamento de Fisiología, Facultad de Medicina, Universidad Nacional Autónoma de México (UNAM), PO Box 70250, Ciudad de México, 04510, Mexico
| | - Fabiola Hernández-Vázquez
- División de Neurociencias, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México (UNAM), Ciudad de México, 04510, Mexico
| | - Arturo Reyes-Vaca
- Departamento de Fisiología, Facultad de Medicina, Universidad Nacional Autónoma de México (UNAM), PO Box 70250, Ciudad de México, 04510, Mexico
| | - Isabel Arenas
- Departamento de Fisiología, Facultad de Medicina, Universidad Nacional Autónoma de México (UNAM), PO Box 70250, Ciudad de México, 04510, Mexico
| | - Jorge Bravo-Martínez
- Departamento de Fisiología, Facultad de Medicina, Universidad Nacional Autónoma de México (UNAM), PO Box 70250, Ciudad de México, 04510, Mexico
| | - David E Garcia
- Departamento de Fisiología, Facultad de Medicina, Universidad Nacional Autónoma de México (UNAM), PO Box 70250, Ciudad de México, 04510, Mexico
| |
Collapse
|
26
|
Abstract
Glucose-sensitive neurons have long been implicated in glucose homeostasis, but how glucose-sensing information is used by the brain in this process remains uncertain. Here, we propose a model in which (1) information relevant to the circulating glucose level is essential to the proper function of this regulatory system, (2) this input is provided by neurons located outside the blood-brain barrier (BBB) (since neurons situated behind the BBB are exposed to glucose in brain interstitial fluid, rather than that in the circulation), and (3) while the efferent limb of this system is comprised of neurons situated behind the BBB, many of these neurons are also glucose sensitive. Precedent for such an organizational scheme is found in the thermoregulatory system, which we draw upon in this framework for understanding the role played by brain glucose sensing in glucose homeostasis.
Collapse
Affiliation(s)
- Marie Aare Bentsen
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Science, University of Copenhagen, Blegdamsvej 3B, Building 7 (Maersk Tower), Copenhagen N 2200, Denmark; University of Washington Medicine Diabetes Institute, Department of Medicine, University of Washington at South Lake Union, 750 Republican St, F704, Box 358062, Seattle, WA 98109, USA
| | - Zaman Mirzadeh
- Department of Neurological Surgery, Barrow Neurological Institute, 350 West Thomas Road, Phoenix, AZ 85013, USA
| | - Michael W Schwartz
- University of Washington Medicine Diabetes Institute, Department of Medicine, University of Washington at South Lake Union, 750 Republican St, F704, Box 358062, Seattle, WA 98109, USA.
| |
Collapse
|
27
|
Frankot M, Treesukosol Y. Alternate day fasting decreases preference for a calorically dense diet by increasing chow intake and altering meal pattern parameters. Physiol Behav 2018; 201:12-21. [PMID: 30521879 DOI: 10.1016/j.physbeh.2018.11.039] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2018] [Revised: 10/19/2018] [Accepted: 11/29/2018] [Indexed: 12/19/2022]
Abstract
Alternate day fasting (ADF) is an effective dietary strategy for weight loss in both humans and rats. However, fasting can elicit hyperphagia in rats, particularly upon access to a calorically dense, high-energy (HE) diet. To examine the effects of ADF on HE diet preference, male and female Sprague-Dawley rats were randomly assigned to receive either ad-libitum or alternate day access to both chow and HE food. Meal pattern analysis was conducted to provide a more detailed explanation of changes in HE preference. ADF rats had a decreased preference for the HE diet compared to controls. Both male and female ADF rats increased in overall intake of chow. However, for male ADF rats, the decrease in HE preference was driven by an increase in both size and number of chow meals; for females, it was driven only by an increase in number of chow meals. Meal size is controlled by both positive feedback (e.g., from the oral cavity) and negative feedback (e.g., from postoral inhibitory signals). Thus, for males, fasting appeared to increase orosensory stimulation and/or decrease sensitivity to inhibitory cues towards chow. For females, fasting appeared to decrease sensitivity to inhibitory cues towards chow. The decrease in HE preference observed in the current study may contribute to the effectiveness of ADF as a dietary strategy for weight loss.
Collapse
Affiliation(s)
- Michelle Frankot
- Department of Psychology, California State University Long Beach, Long Beach, CA 90840, USA
| | - Yada Treesukosol
- Department of Psychology, California State University Long Beach, Long Beach, CA 90840, USA.
| |
Collapse
|
28
|
Ugrankar R, Theodoropoulos P, Akdemir F, Henne WM, Graff JM. Circulating glucose levels inversely correlate with Drosophila larval feeding through insulin signaling and SLC5A11. Commun Biol 2018; 1:110. [PMID: 30271990 PMCID: PMC6123810 DOI: 10.1038/s42003-018-0109-4] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2018] [Accepted: 07/03/2018] [Indexed: 12/27/2022] Open
Abstract
In mammals, blood glucose levels likely play a role in appetite regulation yet the mechanisms underlying this phenomenon remain opaque. Mechanisms can often be explored from Drosophila genetic approaches. To determine if circulating sugars might be involved in Drosophila feeding behaviors, we scored hemolymph glucose and trehalose, and food ingestion in larvae subjected to various diets, genetic mutations, or RNAi. We found that larvae with glucose elevations, hyperglycemia, have an aversion to feeding; however, trehalose levels do not track with feeding behavior. We further discovered that insulins and SLC5A11 may participate in glucose-regulated feeding. To see if food aversion might be an appropriate screening method for hyperglycemia candidates, we developed a food aversion screen to score larvae with abnormal feeding for glucose. We found that many feeding defective larvae have glucose elevations. These findings highlight intriguing roles for glucose in fly biology as a potential cue and regulator of appetite. Rupali Ugrankar et al. show that Drosophila larvae with high levels of circulating glucose, but not trehalose, don’t eat much. This study suggests that circulating glucose communicates with insulin signaling and the sodium/solute co-transporter SLC5A11 in the brain to suppress larval appetite.
Collapse
Affiliation(s)
- Rupali Ugrankar
- Department of Developmental Biology, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd., Dallas, TX, 75390, USA. .,Department of Cell Biology, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd., Dallas, TX, 75390, USA.
| | - Pano Theodoropoulos
- Department of Developmental Biology, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd., Dallas, TX, 75390, USA
| | - Fatih Akdemir
- Department of Developmental Biology, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd., Dallas, TX, 75390, USA.,Department of Basic Sciences, Medical Biology, Ataturk University, 25240 Erzurum, Turkey
| | - W Mike Henne
- Department of Cell Biology, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd., Dallas, TX, 75390, USA
| | - Jonathan M Graff
- Department of Developmental Biology, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd., Dallas, TX, 75390, USA. .,Department of Molecular Biology, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd., Dallas, TX, 75390, USA. .,Department of Internal Medicine, Division of Endocrinology, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd., Dallas, TX, 75390, USA.
| |
Collapse
|
29
|
Lee S, Lee J, Kang GM, Kim MS. Leptin directly regulate intrinsic neuronal excitability in hypothalamic POMC neurons but not in AgRP neurons in food restricted mice. Neurosci Lett 2018; 681:105-109. [PMID: 29857041 DOI: 10.1016/j.neulet.2018.05.041] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2015] [Revised: 05/09/2018] [Accepted: 05/28/2018] [Indexed: 10/16/2022]
Abstract
Leptin plays a pivotal role in the central control of energy balance through leptin receptors expressed on specific hypothalamic nuclei. Leptin suppresses food intake and body weight and ameliorates hyperglycemia by acting on the AgRP and POMC neurons of the arcuate nucleus. Leptin action on POMC neurons are essential for control of body weight and blood glucose levels and are known to be mediated by JAK-STAT3 and PI3K signalling pathway thus increase POMC mRNA and intrinsic excitability. The effects of leptin on AgRP neurons are not as clear although it has been reported to hyperpolarize AgRP neurons through change of K+ conductance. Using cell-attached patch and whole cell patch configuration, we directly assessed neuronal response to leptin in GFP labelled AgRP or POMC neurons in mice after 18 h of food deprivation. We found leptin has a direct effect on POMC neuron through increased intrinsic excitability and decreased inhibitory synaptic inputs. However, leptin does not have any effect on intrinsic excitability of AgRP neurons in fasted condition although food deprivation induced increase of firing frequency of AgRP neurons. In conclusion, leptin probably has a direct and acute effect on POMC neurons but not on AgRP neurons to control their excitability within feeding-regulatory neuronal circuitry.
Collapse
Affiliation(s)
- Soojung Lee
- Department of Oral Physiology, Faculty of Dentistry, Kyung Hee University, Seoul, Republic of Korea.
| | - Jooyoung Lee
- Department of Oral Physiology, Faculty of Dentistry, Kyung Hee University, Seoul, Republic of Korea
| | - Gil Myoung Kang
- Asan Institute for Life Science, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Min-Seon Kim
- Asan Institute for Life Science, University of Ulsan College of Medicine, Seoul, Republic of Korea
| |
Collapse
|
30
|
Belfort-DeAguiar R, Seo D, Lacadie C, Naik S, Schmidt C, Lam W, Hwang J, Constable T, Sinha R, Sherwin RS. Humans with obesity have disordered brain responses to food images during physiological hyperglycemia. Am J Physiol Endocrinol Metab 2018; 314:E522-E529. [PMID: 29381374 PMCID: PMC6048387 DOI: 10.1152/ajpendo.00335.2017] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Blood glucose levels influence brain regulation of food intake. This study assessed the effect of mild physiological hyperglycemia on brain response to food cues in individuals with obesity (OB) versus normal weight individuals (NW). Brain responses in 10 OB and 10 NW nondiabetic healthy adults [body mass index: 34 (3) vs. 23 (2) kg/m2, means (SD), P < 0.0001] were measured with functional MRI (blood oxygen level-dependent contrast) in combination with a two-step normoglycemic-hyperglycemic clamp. Participants were shown food and nonfood images during normoglycemia (~95 mg/dl) and hyperglycemia (~130 mg/dl). Plasma glucose levels were comparable in both groups during the two-step clamp ( P = not significant). Insulin and leptin levels were higher in the OB group compared with NW, whereas ghrelin levels were lower (all P < 0.05). During hyperglycemia, insula activity showed a group-by-glucose level effect. When compared with normoglycemia, hyperglycemia resulted in decreased activity in the hypothalamus and putamen in response to food images ( P < 0.001) in the NW group, whereas the OB group exhibited increased activity in insula, putamen, and anterior and dorsolateral prefrontal cortex (aPFC/dlPFC; P < 0.001). These data suggest that OB, compared with NW, appears to have disruption of brain responses to food cues during hyperglycemia, with reduced insula response in NW but increased insula response in OB, an area involved in food perception and interoception. In a post hoc analysis, brain activity in obesity appears to be associated with dysregulated motivation (striatum) and inappropriate self-control (aPFC/dlPFC) to food cues during hyperglycemia. Hyperstimulation for food and insensitivity to internal homeostatic signals may favor food consumption to possibly play a role in the pathogenesis of obesity.
Collapse
Affiliation(s)
- Renata Belfort-DeAguiar
- Section of Endocrinology, Department of Internal Medicine, Yale University School of Medicine , New Haven, Connecticut
| | - Dongju Seo
- Department of Psychiatry, Yale University School of Medicine , New Haven, Connecticut
| | - Cheryl Lacadie
- Department of Radiology, Yale University School of Medicine , New Haven, Connecticut
| | - Sarita Naik
- Section of Endocrinology, Department of Internal Medicine, Yale University School of Medicine , New Haven, Connecticut
| | - Christian Schmidt
- Section of Endocrinology, Department of Internal Medicine, Yale University School of Medicine , New Haven, Connecticut
| | - Wai Lam
- Section of Endocrinology, Department of Internal Medicine, Yale University School of Medicine , New Haven, Connecticut
| | - Janice Hwang
- Section of Endocrinology, Department of Internal Medicine, Yale University School of Medicine , New Haven, Connecticut
| | - Todd Constable
- Department of Radiology, Yale University School of Medicine , New Haven, Connecticut
| | - Rajita Sinha
- Department of Psychiatry, Yale University School of Medicine , New Haven, Connecticut
| | - Robert S Sherwin
- Section of Endocrinology, Department of Internal Medicine, Yale University School of Medicine , New Haven, Connecticut
| |
Collapse
|
31
|
Zhou C, Teegala SB, Khan BA, Gonzalez C, Routh VH. Hypoglycemia: Role of Hypothalamic Glucose-Inhibited (GI) Neurons in Detection and Correction. Front Physiol 2018; 9:192. [PMID: 29593556 PMCID: PMC5854653 DOI: 10.3389/fphys.2018.00192] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2018] [Accepted: 02/23/2018] [Indexed: 01/08/2023] Open
Abstract
Hypoglycemia is a profound threat to the brain since glucose is its primary fuel. As a result, glucose sensors are widely located in the central nervous system and periphery. In this perspective we will focus on the role of hypothalamic glucose-inhibited (GI) neurons in sensing and correcting hypoglycemia. In particular, we will discuss GI neurons in the ventromedial hypothalamus (VMH) which express neuronal nitric oxide synthase (nNOS) and in the perifornical hypothalamus (PFH) which express orexin. The ability of VMH nNOS-GI neurons to depolarize in low glucose closely parallels the hormonal response to hypoglycemia which stimulates gluconeogenesis. We have found that nitric oxide (NO) production in low glucose is dependent on oxidative status. In this perspective we will discuss the potential relevance of our work showing that enhancing the glutathione antioxidant system prevents hypoglycemia associated autonomic failure (HAAF) in non-diabetic rats whereas VMH overexpression of the thioredoxin antioxidant system restores hypoglycemia counterregulation in rats with type 1 diabetes.We will also address the potential role of the orexin-GI neurons in the arousal response needed for hypoglycemia awareness which leads to behavioral correction (e.g., food intake, glucose administration). The potential relationship between the hypothalamic sensors and the neurocircuitry in the hindbrain and portal mesenteric vein which is critical for hypoglycemia correction will then be discussed.
Collapse
Affiliation(s)
| | | | | | | | - Vanessa H. Routh
- Department of Pharmacology, Physiology and Neurosciences, New Jersey Medical School, Rutgers, The State University of New Jersey, Newark, NJ, United States
| |
Collapse
|
32
|
Role of microglia-neuron interactions in diabetic encephalopathy. Ageing Res Rev 2018; 42:28-39. [PMID: 29247713 DOI: 10.1016/j.arr.2017.12.005] [Citation(s) in RCA: 60] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2017] [Revised: 11/07/2017] [Accepted: 12/08/2017] [Indexed: 12/11/2022]
Abstract
In the central nervous system, the primary immune cells, the microglia, prevent pathogenic invasion as the first line of defense. Microglial energy consumption is dependent on their degree of activity. Microglia express transporters for the three primary energy substrates (glucose, fatty acids, glutamine) and regulate diabetic encephalopathy via microglia-neuron interactions. Microglia may play a sentry role for rapid protection or even ablation of impaired neurons. Neurons exhibit hyperactivity in response to hyperglycemia, hyperlipidemia, and neurotoxic factors and release potential microglial activators. Microglial activation is also regulated by proinflammatory factors, caspase-3 activity, P2X7 receptor, interferon regulatory factor-8, and glucocorticoids. Modulation of microglia in diabetic encephalopathy may involve CX3CL1, p38 MAPK, purinergic, and CD200/CD200R signaling pathways, and pattern recognition receptors. The microglia-neuron interactions play an important role in diabetic encephalopathy, and modulation of microglial activation may be a therapeutic target for diabetic encephalopathy.
Collapse
|
33
|
Fioramonti X, Chrétien C, Leloup C, Pénicaud L. Recent Advances in the Cellular and Molecular Mechanisms of Hypothalamic Neuronal Glucose Detection. Front Physiol 2017; 8:875. [PMID: 29184506 PMCID: PMC5694446 DOI: 10.3389/fphys.2017.00875] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2017] [Accepted: 10/18/2017] [Indexed: 11/18/2022] Open
Abstract
The hypothalamus have been recognized for decades as one of the major brain centers for the control of energy homeostasis. This area contains specialized neurons able to detect changes in nutrients level. Among them, glucose-sensing neurons use glucose as a signaling molecule in addition to its fueling role. In this review we will describe the different sub-populations of glucose-sensing neurons present in the hypothalamus and highlight their nature in terms of neurotransmitter/neuropeptide expression. This review will particularly discuss whether pro-opiomelanocortin (POMC) neurons from the arcuate nucleus are directly glucose-sensing. In addition, recent observations in glucose-sensing suggest a subtle system with different mechanisms involved in the detection of changes in glucose level and their involvement in specific physiological functions. Several data point out the critical role of reactive oxygen species (ROS) and mitochondria dynamics in the detection of increased glucose. This review will also highlight that ATP-dependent potassium (KATP) channels are not the only channels mediating glucose-sensing and discuss the new role of transient receptor potential canonical channels (TRPC). We will discuss the recent advances in the determination of glucose-sensing machinery and propose potential line of research needed to further understand the regulation of brain glucose detection.
Collapse
Affiliation(s)
- Xavier Fioramonti
- NutriNeuro, Institut National de la Recherche Agronomique, Université de Bordeaux, Bordeaux, France.,Centre des Sciences du Goût et de l'Alimentation, Centre National de la Recherche Scientifique, Institut National de la Recherche Agronomique, Université Bourgogne Franche-Comté, Dijon, France
| | - Chloé Chrétien
- Centre des Sciences du Goût et de l'Alimentation, Centre National de la Recherche Scientifique, Institut National de la Recherche Agronomique, Université Bourgogne Franche-Comté, Dijon, France
| | - Corinne Leloup
- Centre des Sciences du Goût et de l'Alimentation, Centre National de la Recherche Scientifique, Institut National de la Recherche Agronomique, Université Bourgogne Franche-Comté, Dijon, France
| | - Luc Pénicaud
- Centre des Sciences du Goût et de l'Alimentation, Centre National de la Recherche Scientifique, Institut National de la Recherche Agronomique, Université Bourgogne Franche-Comté, Dijon, France.,Stromalab, Centre National de la Recherche Scientifique, Institut National de la Santé et de la Recherche Médicale, Université de Toulouse, Toulouse, France
| |
Collapse
|
34
|
Levin BE. 10 lessons learned by a misguided physician. Physiol Behav 2017; 176:217-222. [PMID: 28034577 DOI: 10.1016/j.physbeh.2016.12.033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2016] [Revised: 12/23/2016] [Accepted: 12/23/2016] [Indexed: 10/20/2022]
Abstract
It was a great and humbling honor to receive the 2016 Distinguished Career Award from my SSIB colleagues. This paper summarizes the major points of my DCA talk at the 2016 annual meeting. It is a reflection on my 50year medical and research career and 10 lessons I have learned over those years which might be of help to young investigators near the beginning of their own research careers. These lessons include: the value of being receptive to the opportunities provided you; how clinician-scientists can serve as critical role models for young investigators like me and a history of how my career developed as a result of their influence; the importance of carefully examining your own data, particularly when it doesn't agree with your preconceived ideas; the critical role that students, postdocs and PhD (and even veterinarian) colleagues can play in developing one's career; the likelihood that your career path will have many interesting twists and turns determined by changes in your own scientific interests and how rewarding various areas of research focus are to you; the importance of building a close-knit laboratory staff family; the fact that science and romance can mix. Finally, I offer 3 somewhat self-evident free pieces of advice for building and maintaining a rewarding career.
Collapse
Affiliation(s)
- Barry E Levin
- Department of Neurology, Rutgers New Jersey Medical School, 185 South Orange Avenue, MSB H506, Newark, NJ 07107, USA.
| |
Collapse
|
35
|
Varela L, Suyama S, Huang Y, Shanabrough M, Tschöp MH, Gao XB, Giordano FJ, Horvath TL. Endothelial HIF-1α Enables Hypothalamic Glucose Uptake to Drive POMC Neurons. Diabetes 2017; 66:1511-1520. [PMID: 28292966 PMCID: PMC5440016 DOI: 10.2337/db16-1106] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/10/2016] [Accepted: 03/08/2017] [Indexed: 12/12/2022]
Abstract
Glucose is the primary driver of hypothalamic proopiomelanocortin (POMC) neurons. We show that endothelial hypoxia-inducible factor 1α (HIF-1α) controls glucose uptake in the hypothalamus and that it is upregulated in conditions of undernourishment, during which POMC neuronal activity is decreased. Endothelium-specific knockdown of HIF-1α impairs the ability of POMC neurons to adapt to the changing metabolic environment in vivo, resulting in overeating after food deprivation in mice. The impaired functioning of POMC neurons was reversed ex vivo or by parenchymal glucose administration. These observations indicate an active role for endothelial cells in the central control of metabolism and suggest that central vascular impairments may cause metabolic disorders.
Collapse
Affiliation(s)
- Luis Varela
- Program in Integrative Cell Signaling and Neurobiology of Metabolism, Section of Comparative Medicine, Yale University School of Medicine, New Haven, CT
| | - Shigetomo Suyama
- Program in Integrative Cell Signaling and Neurobiology of Metabolism, Section of Comparative Medicine, Yale University School of Medicine, New Haven, CT
| | - Yan Huang
- Department of Medicine, Section of Cardiovascular Medicine, Yale University School of Medicine, New Haven, CT
| | - Marya Shanabrough
- Program in Integrative Cell Signaling and Neurobiology of Metabolism, Section of Comparative Medicine, Yale University School of Medicine, New Haven, CT
| | - Matthias H Tschöp
- Helmholtz Diabetes Center, Helmholtz Zentrum München and Division of Metabolic Diseases, Technische Universität München, Neuherberg, Germany
| | - Xiao-Bing Gao
- Program in Integrative Cell Signaling and Neurobiology of Metabolism, Section of Comparative Medicine, Yale University School of Medicine, New Haven, CT
| | - Frank J Giordano
- Department of Medicine, Section of Cardiovascular Medicine, Yale University School of Medicine, New Haven, CT
| | - Tamas L Horvath
- Program in Integrative Cell Signaling and Neurobiology of Metabolism, Section of Comparative Medicine, Yale University School of Medicine, New Haven, CT
- Helmholtz Diabetes Center, Helmholtz Zentrum München and Division of Metabolic Diseases, Technische Universität München, Neuherberg, Germany
- Department of Anatomy and Histology, University of Veterinary Medicine, Budapest, Hungary
| |
Collapse
|
36
|
Clarifying the Ghrelin System's Ability to Regulate Feeding Behaviours Despite Enigmatic Spatial Separation of the GHSR and Its Endogenous Ligand. Int J Mol Sci 2017; 18:ijms18040859. [PMID: 28422060 PMCID: PMC5412441 DOI: 10.3390/ijms18040859] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2017] [Revised: 04/04/2017] [Accepted: 04/11/2017] [Indexed: 12/23/2022] Open
Abstract
Ghrelin is a hormone predominantly produced in and secreted from the stomach. Ghrelin is involved in many physiological processes including feeding, the stress response, and in modulating learning, memory and motivational processes. Ghrelin does this by binding to its receptor, the growth hormone secretagogue receptor (GHSR), a receptor found in relatively high concentrations in hypothalamic and mesolimbic brain regions. While the feeding and metabolic effects of ghrelin can be explained by the effects of this hormone on regions of the brain that have a more permeable blood brain barrier (BBB), ghrelin produced within the periphery demonstrates a limited ability to reach extrahypothalamic regions where GHSRs are expressed. Therefore, one of the most pressing unanswered questions plaguing ghrelin research is how GHSRs, distributed in brain regions protected by the BBB, are activated despite ghrelin’s predominant peripheral production and poor ability to transverse the BBB. This manuscript will describe how peripheral ghrelin activates central GHSRs to encourage feeding, and how central ghrelin synthesis and ghrelin independent activation of GHSRs may also contribute to the modulation of feeding behaviours.
Collapse
|
37
|
Tups A, Benzler J, Sergi D, Ladyman SR, Williams LM. Central Regulation of Glucose Homeostasis. Compr Physiol 2017; 7:741-764. [PMID: 28333388 DOI: 10.1002/cphy.c160015] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
|
38
|
Solis E, Bola RA, Fasulo BJ, Kiyatkin EA. Brain Hyperglycemia Induced by Heroin: Association with Metabolic Neural Activation. ACS Chem Neurosci 2017; 8:265-271. [PMID: 27736094 DOI: 10.1021/acschemneuro.6b00246] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
Glucose enters the brain extracellular space from arterial blood, and its proper delivery is essential for metabolic activity of brain cells. By using enzyme-based biosensors coupled with high-speed amperometry in freely moving rats, we previously showed that glucose levels in the nucleus accumbens (NAc) display high variability, increasing rapidly following exposure to various arousing stimuli. In this study, the same technology was used to assess NAc glucose fluctuations induced by intravenous heroin. Heroin passively injected at a low dose optimal for maintaining self-administration behavior (100 μg/kg) induces a rapid but moderate glucose rise (∼150-200 μM or ∼15-25% over resting baseline). When the heroin dose was doubled and tripled, the increase became progressively larger in magnitude and longer in duration. Heroin-induced glucose increases also occurred in other brain structures (medial thalamus, lateral striatum, hippocampus), suggesting that brain hyperglycemia is a whole-brain phenomenon but changes were notably distinct in each structure. While local vasodilation appears to be the possible mechanism underlying the rapid rise in extracellular glucose levels, the driving factor for this vasodilation (central vs peripheral) remains to be clarified. The heroin-induced NAc glucose increases positively correlated with increases in intracerebral heat production determined in separate experiments using multisite temperature recordings (NAc, temporal muscle and skin). However, glucose levels rise very rapidly, preceding much slower increases in brain heat production, a measure of metabolic activation associated with glucose consumption.
Collapse
Affiliation(s)
- Ernesto Solis
- In-Vivo Electrophysiology
Unit, Behavioral Neuroscience Branch, National Institute on Drug Abuse
− Intramural Research Program, National Institutes of Health, DHHS, 333 Cassell Drive, Baltimore, Maryland 21224, United States
| | - R. Aaron Bola
- In-Vivo Electrophysiology
Unit, Behavioral Neuroscience Branch, National Institute on Drug Abuse
− Intramural Research Program, National Institutes of Health, DHHS, 333 Cassell Drive, Baltimore, Maryland 21224, United States
| | - Bradley J. Fasulo
- In-Vivo Electrophysiology
Unit, Behavioral Neuroscience Branch, National Institute on Drug Abuse
− Intramural Research Program, National Institutes of Health, DHHS, 333 Cassell Drive, Baltimore, Maryland 21224, United States
| | - Eugene A. Kiyatkin
- In-Vivo Electrophysiology
Unit, Behavioral Neuroscience Branch, National Institute on Drug Abuse
− Intramural Research Program, National Institutes of Health, DHHS, 333 Cassell Drive, Baltimore, Maryland 21224, United States
| |
Collapse
|
39
|
Béland-Millar A, Larcher J, Courtemanche J, Yuan T, Messier C. Effects of Systemic Metabolic Fuels on Glucose and Lactate Levels in the Brain Extracellular Compartment of the Mouse. Front Neurosci 2017; 11:7. [PMID: 28154523 PMCID: PMC5243849 DOI: 10.3389/fnins.2017.00007] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2016] [Accepted: 01/04/2017] [Indexed: 01/11/2023] Open
Abstract
Classic neuroenergetic research has emphasized the role of glucose, its transport and its metabolism in sustaining normal neural function leading to the textbook statement that it is the necessary and sole metabolic fuel of the mammalian brain. New evidence, including the Astrocyte-to-Neuron Lactate Shuttle hypothesis, suggests that the brain can use other metabolic substrates. To further study that possibility, we examined the effect of intraperitoneally administered metabolic fuels (glucose, fructose, lactate, pyruvate, ß-hydroxybutyrate, and galactose), and insulin, on blood, and extracellular brain levels of glucose and lactate in the adult male CD1 mouse. Primary motor cortex extracellular levels of glucose and lactate were monitored in freely moving mice with the use of electrochemical electrodes. Blood concentration of these same metabolites were obtained by tail vein sampling and measured with glucose and lactate meters. Blood and extracellular fluctuations of glucose and lactate were monitored for a 2-h period. We found that the systemic injections of glucose, fructose, lactate, pyruvate, and ß-hydroxybutyrate increased blood lactate levels. Apart for a small transitory rise in brain extracellular lactate levels, the main effect of the systemic injection of glucose, fructose, lactate, pyruvate, and ß-hydroxybutyrate was an increase in brain extracellular glucose levels. Systemic galactose injections produced a small rise in blood glucose and lactate but almost no change in brain extracellular lactate and glucose. Systemic insulin injections led to a decrease in blood glucose and a small rise in blood lactate; however brain extracellular glucose and lactate monotonically decreased at the same rate. Our results support the concept that the brain is able to use alternative fuels and the current experiments suggest some of the mechanisms involved.
Collapse
Affiliation(s)
| | - Jeremy Larcher
- School of Psychology, University of Ottawa Ottawa, ON, Canada
| | | | - Tina Yuan
- School of Psychology, University of Ottawa Ottawa, ON, Canada
| | - Claude Messier
- School of Psychology, University of Ottawa Ottawa, ON, Canada
| |
Collapse
|
40
|
Dadak S, Beall C, Vlachaki Walker JM, Soutar MPM, McCrimmon RJ, Ashford MLJ. Oleate induces K ATP channel-dependent hyperpolarization in mouse hypothalamic glucose-excited neurons without altering cellular energy charge. Neuroscience 2017; 346:29-42. [PMID: 28087336 PMCID: PMC5346158 DOI: 10.1016/j.neuroscience.2016.12.053] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2016] [Accepted: 12/29/2016] [Indexed: 11/29/2022]
Abstract
Oleate and low glucose hyperpolarize and inhibit GT1-7 and mouse GE neurons by activation of KATP. Oleate inhibition of GT1-7 neuron activity is not mediated by AMPK or fatty acid oxidation. Activation of KATP by oleate requires ATP hydrolysis but does not reduce the levels ATP or the ATP:ADP ratio. GT1-7 hyperpolarization by oleate is not dependent on UCP2. Oleate and low glucose depolarize a subpopulation of hypothalamic GI neurons.
The unsaturated fatty acid, oleate exhibits anorexigenic properties reducing food intake and hepatic glucose output. However, its mechanism of action in the hypothalamus has not been fully determined. This study investigated the effects of oleate and glucose on GT1-7 mouse hypothalamic cells (a model of glucose-excited (GE) neurons) and mouse arcuate nucleus (ARC) neurons. Whole-cell and perforated patch-clamp recordings, immunoblotting and cell energy status measures were used to investigate oleate- and glucose-sensing properties of mouse hypothalamic neurons. Oleate or lowered glucose concentration caused hyperpolarization and inhibition of firing of GT1-7 cells by the activation of ATP-sensitive K+ channels (KATP). This effect of oleate was not dependent on fatty acid oxidation or raised AMP-activated protein kinase activity or prevented by the presence of the UCP2 inhibitor genipin. Oleate did not alter intracellular calcium, indicating that CD36/fatty acid translocase may not play a role. However, oleate activation of KATP may require ATP metabolism. The short-chain fatty acid octanoate was unable to replicate the actions of oleate on GT1-7 cells. Although oleate decreased GT1-7 cell mitochondrial membrane potential there was no change in total cellular ATP or ATP/ADP ratios. Perforated patch and whole-cell recordings from mouse hypothalamic slices demonstrated that oleate hyperpolarized a subpopulation of ARC GE neurons by KATP activation. Additionally, in a separate small population of ARC neurons, oleate application or lowered glucose concentration caused membrane depolarization. In conclusion, oleate induces KATP-dependent hyperpolarization and inhibition of firing of a subgroup of GE hypothalamic neurons without altering cellular energy charge.
Collapse
Affiliation(s)
- Selma Dadak
- Division of Molecular and Clinical Medicine, School of Medicine, Ninewells Hospital and Medical School, University of Dundee, Dundee DD1 9SY, UK
| | - Craig Beall
- Division of Molecular and Clinical Medicine, School of Medicine, Ninewells Hospital and Medical School, University of Dundee, Dundee DD1 9SY, UK; Institute of Biomedical and Clinical Sciences, University of Exeter Medical School, RILD Building, Barrack Road, Exeter EX2 5DW, UK
| | - Julia M Vlachaki Walker
- Institute of Biomedical and Clinical Sciences, University of Exeter Medical School, RILD Building, Barrack Road, Exeter EX2 5DW, UK
| | - Marc P M Soutar
- Division of Molecular and Clinical Medicine, School of Medicine, Ninewells Hospital and Medical School, University of Dundee, Dundee DD1 9SY, UK
| | - Rory J McCrimmon
- Division of Molecular and Clinical Medicine, School of Medicine, Ninewells Hospital and Medical School, University of Dundee, Dundee DD1 9SY, UK
| | - Michael L J Ashford
- Division of Molecular and Clinical Medicine, School of Medicine, Ninewells Hospital and Medical School, University of Dundee, Dundee DD1 9SY, UK.
| |
Collapse
|
41
|
Kalsbeek MJT, Mulder L, Yi CX. Microglia energy metabolism in metabolic disorder. Mol Cell Endocrinol 2016; 438:27-35. [PMID: 27687525 DOI: 10.1016/j.mce.2016.09.028] [Citation(s) in RCA: 63] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/20/2016] [Revised: 09/23/2016] [Accepted: 09/26/2016] [Indexed: 12/22/2022]
Abstract
Microglia are the resident macrophages of the CNS, and are in charge of maintaining a healthy microenvironment to ensure neuronal survival. Microglia carry out a non-stop patrol of the CNS, make contact with neurons and look for abnormalities, all of which requires a vast amount of energy. This non-signaling energy demand increases after activation by pathogens, neuronal damage or other kinds of stimulation. Of the three major energy substrates - glucose, fatty acids and glutamine - glucose is crucial for microglia survival and several glucose transporters are expressed to supply sufficient glucose influx. Fatty acids are another source of energy for microglia and have also been shown to strongly influence microglial immune activity. Glutamine, although possibly suitable for use as an energy substrate by microglia, has been shown to have neurotoxic effects when overloaded. Microglial fuel metabolism might be associated with microglial reactivity under different pathophysiological conditions and a microglial fuel switch may thus be the underlying cause of hypothalamic dysregulation, which is associated with obesity.
Collapse
Affiliation(s)
- Martin J T Kalsbeek
- Department of Endocrinology and Metabolism, Academic Medical Center (AMC), University of Amsterdam (UvA), Meibergdreef 9, 1105 AZ, Amsterdam, The Netherlands.
| | - Laurie Mulder
- Department of Endocrinology and Metabolism, Academic Medical Center (AMC), University of Amsterdam (UvA), Meibergdreef 9, 1105 AZ, Amsterdam, The Netherlands
| | - Chun-Xia Yi
- Department of Endocrinology and Metabolism, Academic Medical Center (AMC), University of Amsterdam (UvA), Meibergdreef 9, 1105 AZ, Amsterdam, The Netherlands
| |
Collapse
|
42
|
Abstract
Multiple physiologic and neural systems contribute to the controls over what and how much we eat. These systems include signaling involved in the detection and signaling of nutrient availability, signals arising from consumed nutrients that provide feedback information during a meal to induce satiation, and signals related to the rewarding properties of eating. Each of these has a separate neural representation, but important interactions among these systems are critical to the overall controls of food intake.
Collapse
Affiliation(s)
- Timothy H Moran
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, 720 Rutland Avenue, Baltimore, MD 21205, USA; Global Obesity Prevention Center at Johns Hopkins, Johns Hopkins Bloomberg School of Public Health, 615 N. Wolfe Street, Baltimore, MD 21205, USA.
| | - Ellen E Ladenheim
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, 720 Rutland Avenue, Baltimore, MD 21205, USA
| |
Collapse
|
43
|
Glycemic increase induced by intravenous glucose infusion fails to affect hunger, appetite, or satiety following breakfast in healthy men. Appetite 2016; 105:562-6. [DOI: 10.1016/j.appet.2016.06.032] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2015] [Revised: 06/24/2016] [Accepted: 06/25/2016] [Indexed: 11/20/2022]
|
44
|
Steinbusch LKM, Picard A, Bonnet MS, Basco D, Labouèbe G, Thorens B. Sex-Specific Control of Fat Mass and Counterregulation by Hypothalamic Glucokinase. Diabetes 2016; 65:2920-31. [PMID: 27422385 DOI: 10.2337/db15-1514] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/05/2015] [Accepted: 07/07/2016] [Indexed: 11/13/2022]
Abstract
Glucokinase (Gck) is a critical regulator of glucose-induced insulin secretion by pancreatic β-cells. It has been suggested to also play an important role in glucose signaling in neurons of the ventromedial hypothalamic nucleus (VMN), a brain nucleus involved in the control of glucose homeostasis and feeding. To test the role of Gck in VMN glucose sensing and physiological regulation, we studied mice with genetic inactivation of the Gck gene in Sf1 neurons of the VMN (Sf1Gck(-/-) mice). Compared with control littermates, Sf1Gck(-/-) mice displayed increased white fat mass and adipocyte size, reduced lean mass, impaired hypoglycemia-induced glucagon secretion, and a lack of parasympathetic and sympathetic nerve activation by neuroglucopenia. However, these phenotypes were observed only in female mice. To determine whether Gck was required for glucose sensing by Sf1 neurons, we performed whole-cell patch clamp analysis of brain slices from control and Sf1Gck(-/-) mice. Absence of Gck expression did not prevent the glucose responsiveness of glucose-excited or glucose-inhibited Sf1 neurons in either sex. Thus Gck in the VMN plays a sex-specific role in the glucose-dependent control of autonomic nervous activity; this is, however, unrelated to the control of the firing activity of classical glucose-responsive neurons.
Collapse
Affiliation(s)
| | - Alexandre Picard
- Center for Integrative Genomics, University of Lausanne, Lausanne, Switzerland
| | - Marion S Bonnet
- Center for Integrative Genomics, University of Lausanne, Lausanne, Switzerland
| | - Davide Basco
- Center for Integrative Genomics, University of Lausanne, Lausanne, Switzerland
| | - Gwenaël Labouèbe
- Center for Integrative Genomics, University of Lausanne, Lausanne, Switzerland
| | - Bernard Thorens
- Center for Integrative Genomics, University of Lausanne, Lausanne, Switzerland
| |
Collapse
|
45
|
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
|
46
|
De Backer I, Hussain SS, Bloom SR, Gardiner JV. Insights into the role of neuronal glucokinase. Am J Physiol Endocrinol Metab 2016; 311:E42-55. [PMID: 27189932 PMCID: PMC4967152 DOI: 10.1152/ajpendo.00034.2016] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/26/2016] [Accepted: 05/13/2016] [Indexed: 12/17/2022]
Abstract
Glucokinase is a key component of the neuronal glucose-sensing mechanism and is expressed in brain regions that control a range of homeostatic processes. In this review, we detail recently identified roles for neuronal glucokinase in glucose homeostasis and counterregulatory responses to hypoglycemia and in regulating appetite. We describe clinical implications from these advances in our knowledge, especially for developing novel treatments for diabetes and obesity. Further research required to extend our knowledge and help our efforts to tackle the diabetes and obesity epidemics is suggested.
Collapse
Affiliation(s)
- Ivan De Backer
- Section of Investigative Medicine, Division of Diabetes, Endocrinology and Metabolism, Imperial College London, London, United Kingdom
| | - Sufyan S Hussain
- Section of Investigative Medicine, Division of Diabetes, Endocrinology and Metabolism, Imperial College London, London, United Kingdom
| | - Stephen R Bloom
- Section of Investigative Medicine, Division of Diabetes, Endocrinology and Metabolism, Imperial College London, London, United Kingdom
| | - James V Gardiner
- Section of Investigative Medicine, Division of Diabetes, Endocrinology and Metabolism, Imperial College London, London, United Kingdom
| |
Collapse
|
47
|
Le Foll C, Levin BE. Fatty acid-induced astrocyte ketone production and the control of food intake. Am J Physiol Regul Integr Comp Physiol 2016; 310:R1186-92. [PMID: 27122369 PMCID: PMC4935491 DOI: 10.1152/ajpregu.00113.2016] [Citation(s) in RCA: 62] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2016] [Accepted: 04/22/2016] [Indexed: 11/22/2022]
Abstract
Obesity and Type 2 diabetes are major worldwide public health issues today. A relationship between total fat intake and obesity has been found. In addition, the mechanisms of long-term and excessive high-fat diet (HFD) intake in the development of obesity still need to be elucidated. The ventromedial hypothalamus (VMH) is a major site involved in the regulation of glucose and energy homeostasis where "metabolic sensing neurons" integrate metabolic signals from the periphery. Among these signals, fatty acids (FA) modulate the activity of VMH neurons using the FA translocator/CD36, which plays a critical role in the regulation of energy and glucose homeostasis. During low-fat diet (LFD) intake, FA are oxidized by VMH astrocytes to fuel their ongoing metabolic needs. However, HFD intake causes VMH astrocytes to use FA to generate ketone bodies. We postulate that these astrocyte-derived ketone bodies are exported to neurons where they produce excess ATP and reactive oxygen species, which override CD36-mediated FA sensing and act as a signal to decrease short-term food intake. On a HFD, VMH astrocyte-produced ketones reduce elevated caloric intake to LFD levels after 3 days in rats genetically predisposed to resist (DR) diet-induced obesity (DIO), but not leptin-resistant DIO rats. This suggests that, while VMH ketone production on a HFD can contribute to protection from obesity, the inherent leptin resistance overrides this inhibitory action of ketone bodies on food intake. Thus, astrocytes and neurons form a tight metabolic unit that is able to monitor circulating nutrients to alter food intake and energy homeostasis.
Collapse
Affiliation(s)
- Christelle Le Foll
- Institute of Veterinary Physiology, University of Zurich, Zurich, Switzerland; and
| | - Barry E Levin
- Department of Neurology, Rutgers, New Jersey Medical School, Newark, New Jersey
| |
Collapse
|
48
|
Breit A, Wicht K, Boekhoff I, Glas E, Lauffer L, Mückter H, Gudermann T. Glucose Enhances Basal or Melanocortin-Induced cAMP-Response Element Activity in Hypothalamic Cells. Mol Endocrinol 2016; 30:748-62. [PMID: 27144291 DOI: 10.1210/me.2016-1001] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
Melanocyte-stimulating hormone (MSH)-induced activation of the cAMP-response element (CRE) via the CRE-binding protein in hypothalamic cells promotes expression of TRH and thereby restricts food intake and increases energy expenditure. Glucose also induces central anorexigenic effects by acting on hypothalamic neurons, but the underlying mechanisms are not completely understood. It has been proposed that glucose activates the CRE-binding protein-regulated transcriptional coactivator 2 (CRTC-2) in hypothalamic neurons by inhibition of AMP-activated protein kinases (AMPKs), but whether glucose directly affects hypothalamic CRE activity has not yet been shown. Hence, we dissected effects of glucose on basal and MSH-induced CRE activation in terms of kinetics, affinity, and desensitization in murine, hypothalamic mHypoA-2/10-CRE cells that stably express a CRE-dependent reporter gene construct. Physiologically relevant increases in extracellular glucose enhanced basal or MSH-induced CRE-dependent gene transcription, whereas prolonged elevated glucose concentrations reduced the sensitivity of mHypoA-2/10-CRE cells towards glucose. Glucose also induced CRCT-2 translocation into the nucleus and the AMPK activator metformin decreased basal and glucose-induced CRE activity, suggesting a role for AMPK/CRTC-2 in glucose-induced CRE activation. Accordingly, small interfering RNA-induced down-regulation of CRTC-2 expression decreased glucose-induced CRE-dependent reporter activation. Of note, glucose also induced expression of TRH, suggesting that glucose might affect the hypothalamic-pituitary-thyroid axis via the regulation of hypothalamic CRE activity. These findings significantly advance our knowledge about the impact of glucose on hypothalamic signaling and suggest that TRH release might account for the central anorexigenic effects of glucose and could represent a new molecular link between hyperglycaemia and thyroid dysfunction.
Collapse
Affiliation(s)
- Andreas Breit
- Walther-Straub-Institut für Pharmakologie und Toxikologie, Ludwig-Maximilians-Universität München, 80336 Munich, Germany
| | - Kristina Wicht
- Walther-Straub-Institut für Pharmakologie und Toxikologie, Ludwig-Maximilians-Universität München, 80336 Munich, Germany
| | - Ingrid Boekhoff
- Walther-Straub-Institut für Pharmakologie und Toxikologie, Ludwig-Maximilians-Universität München, 80336 Munich, Germany
| | - Evi Glas
- Walther-Straub-Institut für Pharmakologie und Toxikologie, Ludwig-Maximilians-Universität München, 80336 Munich, Germany
| | - Lisa Lauffer
- Walther-Straub-Institut für Pharmakologie und Toxikologie, Ludwig-Maximilians-Universität München, 80336 Munich, Germany
| | - Harald Mückter
- Walther-Straub-Institut für Pharmakologie und Toxikologie, Ludwig-Maximilians-Universität München, 80336 Munich, Germany
| | - Thomas Gudermann
- Walther-Straub-Institut für Pharmakologie und Toxikologie, Ludwig-Maximilians-Universität München, 80336 Munich, Germany
| |
Collapse
|
49
|
Bola RA, Kiyatkin EA. Robust Brain Hyperglycemia during General Anesthesia: Relationships with Metabolic Brain Inhibition and Vasodilation. Front Physiol 2016; 7:39. [PMID: 26913008 PMCID: PMC4753326 DOI: 10.3389/fphys.2016.00039] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2015] [Accepted: 01/28/2016] [Indexed: 12/13/2022] Open
Abstract
Glucose is the main energetic substrate for the metabolic activity of brain cells and its proper delivery into the extracellular space is essential for maintaining normal neural functions. Under physiological conditions, glucose continuously enters the extracellular space from arterial blood via gradient-dependent facilitated diffusion governed by the GLUT-1 transporters. Due to this gradient-dependent mechanism, glucose levels rise in the brain after consumption of glucose-containing foods and drinks. Glucose entry is also accelerated due to local neuronal activation and neuro-vascular coupling, resulting in transient hyperglycemia to prevent any metabolic deficit. Here, we explored another mechanism that is activated during general anesthesia and results in significant brain hyperglycemia. By using enzyme-based glucose biosensors we demonstrate that glucose levels in the nucleus accumbens (NAc) strongly increase after iv injection of Equthesin, a mixture of chloral hydrate and sodium pentobarbital, which is often used for general anesthesia in rats. By combining electrochemical recordings with brain, muscle, and skin temperature monitoring, we show that the gradual increase in brain glucose occurring during the development of general anesthesia tightly correlate with decreases in brain-muscle temperature differentials, suggesting that this rise in glucose is related to metabolic inhibition. While the decreased consumption of glucose by brain cells could contribute to the development of hyperglycemia, an exceptionally strong positive correlation (r = 0.99) between glucose rise and increases in skin-muscle temperature differentials was also found, suggesting the strong vasodilation of cerebral vessels as the primary mechanism for accelerated entry of glucose into brain tissue. Our present data could explain drastic differences in basal glucose levels found in awake and anesthetized animal preparations. They also suggest that glucose entry into brain tissue could be strongly modulated by pharmacological drugs via drug-induced changes in metabolic activity and the tone of cerebral vessels.
Collapse
Affiliation(s)
- R Aaron Bola
- In-Vivo Electrophysiology Unit, Behavioral Neuroscience Branch, National Institute on Drug Abuse - Intramural Research Program, National Institutes of Health Baltimore, MD, USA
| | - Eugene A Kiyatkin
- In-Vivo Electrophysiology Unit, Behavioral Neuroscience Branch, National Institute on Drug Abuse - Intramural Research Program, National Institutes of Health Baltimore, MD, USA
| |
Collapse
|
50
|
Ranzenhofer LM, Engel SG, Crosby RD, Haigney M, Anderson M, McCaffery JM, Tanofsky-Kraff M. Real-time assessment of heart rate variability and loss of control eating in adolescent girls: A pilot study. Int J Eat Disord 2016; 49:197-201. [PMID: 26401652 PMCID: PMC4733414 DOI: 10.1002/eat.22464] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 08/30/2015] [Indexed: 11/06/2022]
Abstract
OBJECTIVE Studying physiologic underpinnings of loss-of-control (LOC) eating may inform its etiology and contribute to intervention efforts. We therefore examined temporal relationships between autonomic indices [heart rate (HR), heart rate variability (HRV)] and LOC-eating in the natural environment. METHOD For two days, adolescents (n = 17, 14.77 ± 1.55 years, BMI-Z 2.17 ± 0.48) with LOC-eating reported on LOC using an electronic device while HR and HRV were assessed continuously using Holter monitoring. RESULTS Higher HR and lower HRV in the 30-minutes before eating were significantly associated with LOC-eating overall (p's < 0.001) and at the within-participants level (p's < 0.001), but not at the between-participants level (p's > 0.44). Examined categorically, HR was significantly higher, and HRV significantly lower, prior to high-LOC compared to low-LOC episodes (p's < 0.001). DISCUSSION This pilot study suggests that LOC-eating may involve physiologic underpinnings. Additional research with larger samples is needed to further investigate this phenomenon.
Collapse
Affiliation(s)
- Lisa M. Ranzenhofer
- Department of Medical and Clinical Psychology, Uniformed Services University of the Health Sciences (USUHS), DoD, 4301 Jones Bridge Road, Bethesda, MD, 20814, USA,Weight Control and Diabetes Research Center, The Alpert Medical School of Brown University/The Miriam Hospital, 196 Richmond Street, Providence, RI, 02903, USA
| | - Scott G. Engel
- Neuropsychiatric Research Institute (NRI), 120 South 8 Street, Fargo, ND, 58107-1415, USA,Department of Psychiatry and Behavioral Science, University of North Dakota School of Medicine and Health Sciences, 1919 Elm St. North, Fargo, ND, 58102, USA
| | - Ross D. Crosby
- Neuropsychiatric Research Institute (NRI), 120 South 8 Street, Fargo, ND, 58107-1415, USA,Department of Psychiatry and Behavioral Science, University of North Dakota School of Medicine and Health Sciences, 1919 Elm St. North, Fargo, ND, 58102, USA
| | | | - Micheline Anderson
- Department of Medical and Clinical Psychology, Uniformed Services University of the Health Sciences (USUHS), DoD, 4301 Jones Bridge Road, Bethesda, MD, 20814, USA
| | - Jeanne M. McCaffery
- Weight Control and Diabetes Research Center, The Alpert Medical School of Brown University/The Miriam Hospital, 196 Richmond Street, Providence, RI, 02903, USA
| | - Marian Tanofsky-Kraff
- Department of Medical and Clinical Psychology, Uniformed Services University of the Health Sciences (USUHS), DoD, 4301 Jones Bridge Road, Bethesda, MD, 20814, USA
| |
Collapse
|