1
|
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
|
2
|
Potegal M. How it ends: A review of behavioral and psychological phenomena, physiological processes and neural circuits in the termination of aggression in other animals and anger in people. Behav Brain Res 2024; 456:114676. [PMID: 37739229 DOI: 10.1016/j.bbr.2023.114676] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2023] [Revised: 08/26/2023] [Accepted: 09/17/2023] [Indexed: 09/24/2023]
Abstract
More is known about aggression initiation and persistence in other animals, and anger in people, than about their cessation. This review summarizes knowledge of relevant factors in aggression, mostly in vertebrates, and anger termination in people. The latency, probability and intensity of offensive aggression in mice is controlled by activity in a neuronal subpopulation in ventromedial hypothalamus [VMH]. This activity instantiates an aggressive state termed angriffsbereitschaft ["attack-readiness"]. Fighting in many species is broken into bouts with interbout breaks due to fatigue and/or signals from dorsal raphe to VMH. Eventually, losers decide durations and outcomes of fighting by transitioning to submission or flight. Factors reducing angriffsbereitschaft and triggering these defeat behaviors could include metabolic costs, e.g., lactate accumulation and glucose depletion detected by the hypothalamus, central fatigue perhaps sensed by the Salience Network [insula and anterior cingulate gyrus] and pain of injuries, the latter insufficiently blunted by opioid and non-opioid stress analgesia and transduced by anterior VMH neurons. Winners' angriffsbereitschaft continue for awhile, as indicated by post-victory attacks and, perhaps, triumph displays of some species, including humans. In longer term situations, sensory and/or response habituation of aggression may explain the "Dear enemy" tolerance of competitive neighbors. Prolonged satiation of predatory behavior could involve habenula-regulated reduction of dopaminergic reward in nucleus accumbens. Termination of human anger involves at least three processes, metaphorically termed decay, quenching and catharsis. Hypothesized neural mechanisms include anger diminution by negative feedback from accumbens to anterior cingulate and/or activity in the Salience Network that controls anger's "accumulation/offset" phase.
Collapse
Affiliation(s)
- M Potegal
- University of Minnesota, United States.
| |
Collapse
|
3
|
Song J. Amygdala activity and amygdala-hippocampus connectivity: Metabolic diseases, dementia, and neuropsychiatric issues. Biomed Pharmacother 2023; 162:114647. [PMID: 37011482 DOI: 10.1016/j.biopha.2023.114647] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2023] [Revised: 03/30/2023] [Accepted: 03/31/2023] [Indexed: 04/04/2023] Open
Abstract
With rapid aging of the population worldwide, the number of people with dementia is dramatically increasing. Some studies have emphasized that metabolic syndrome, which includes obesity and diabetes, leads to increased risks of dementia and cognitive decline. Factors such as insulin resistance, hyperglycemia, high blood pressure, dyslipidemia, and central obesity in metabolic syndrome are associated with synaptic failure, neuroinflammation, and imbalanced neurotransmitter levels, leading to the progression of dementia. Due to the positive correlation between diabetes and dementia, some studies have called it "type 3 diabetes". Recently, the number of patients with cognitive decline due to metabolic imbalances has considerably increased. In addition, recent studies have reported that neuropsychiatric issues such as anxiety, depressive behavior, and impaired attention are common factors in patients with metabolic disease and those with dementia. In the central nervous system (CNS), the amygdala is a central region that regulates emotional memory, mood disorders, anxiety, attention, and cognitive function. The connectivity of the amygdala with other brain regions, such as the hippocampus, and the activity of the amygdala contribute to diverse neuropathological and neuropsychiatric issues. Thus, this review summarizes the significant consequences of the critical roles of amygdala connectivity in both metabolic syndromes and dementia. Further studies on amygdala function in metabolic imbalance-related dementia are needed to treat neuropsychiatric problems in patients with this type of dementia.
Collapse
Affiliation(s)
- Juhyun Song
- Department of Anatomy, Chonnam National University Medical School, Hwasun 58128, Jeollanam-do, Republic of Korea.
| |
Collapse
|
4
|
Palani G, Stortz E, Moheet A. Clinical Presentation and Diagnostic Approach to Hypoglycemia in Adults Without Diabetes Mellitus. Endocr Pract 2023; 29:286-294. [PMID: 36464132 DOI: 10.1016/j.eprac.2022.11.010] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/06/2022] [Revised: 10/24/2022] [Accepted: 11/28/2022] [Indexed: 12/03/2022]
Abstract
OBJECTIVE To review the clinical presentation, causes, and diagnostic approach to spontaneous hypoglycemia in adults without diabetes mellitus. METHODS A literature review was performed using the PubMed and Google Scholar databases. RESULTS Hypoglycemia is uncommon in people who are not on glucose-lowering medications. Under normal physiologic conditions, multiple neural and hormonal counterregulatory mechanisms prevent the development of abnormally low levels of plasma glucose. If spontaneous hypoglycemia is suspected, the Whipple triad should be used to confirm hypoglycemia before pursuing further diagnostic workup. The Whipple criteria include the following: (1) low levels of plasma glucose, (2) signs or symptoms that would be expected with low levels of plasma glucose, and (3) improvement in those signs or symptoms when the level of plasma glucose increases. Spontaneous hypoglycemia can be caused by conditions that cause endogenous hyperinsulinism, including insulinoma, postbariatric hypoglycemia, and noninsulinoma pancreatogenous hypoglycemia. Spontaneous hypoglycemia can also be seen with critical illness, hepatic or renal dysfunction, hormonal deficiency, non-diabetes-related medications, and non-islet cell tumors. The initial diagnostic approach should begin by obtaining a detailed history of the nature and timing of the patient's symptoms, medications, underlying comorbid conditions, and any acute illness. A laboratory evaluation should be conducted at the time of the spontaneous symptomatic episode. Supervised tests such as a 72-hour fast or mixed-meal test may be needed to recreate the situation under which the patient is likely to experience symptoms. CONCLUSION We provide an overview of the physiology of counterregulatory response to hypoglycemia, its causes, and diagnostic approaches to spontaneous hypoglycemia in adults.
Collapse
Affiliation(s)
- Gurunanthan Palani
- Division of Endocrinology and Diabetes, Department of Medicine, University of Minnesota, Minneapolis, Minnesota
| | - Ethan Stortz
- Division of Endocrinology and Diabetes, Department of Medicine, University of Minnesota, Minneapolis, Minnesota
| | - Amir Moheet
- Division of Endocrinology and Diabetes, Department of Medicine, University of Minnesota, Minneapolis, Minnesota.
| |
Collapse
|
5
|
Wang Y, Sui X, Luo J, Yang G, Fan P, Lu B, Li M, Xu Z, Qu L, Song Y, Li Y, Cai X. A Microelectrode Array Modified by PtNPs/PB Nanocomposites Used for the Detection and Analysis of Glucose-Sensitive Neurons under Different Blood Glucose States. ACS APPLIED BIO MATERIALS 2023; 6:1260-1271. [PMID: 36884222 DOI: 10.1021/acsabm.3c00006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/09/2023]
Abstract
Hypoglycemia state damages the organism, and glucose-excited and glucose-inhibited neurons from the ventral medial hypothalamus can regulate this state. Therefore, it is crucial to understand the functional mechanism between blood glucose and electrophysiology of glucose-excited and glucose-inhibited neurons. To better detect and analyze this mechanism, a PtNPs/PB nanomaterials modified 32-channel microelectrode array with low impedance (21.91 ± 6.80 kΩ), slight phase delay (-12.7° ± 2.7°), high double layer capacitance (0.606 μF), and biocompatibility was developed to realize in vivo real-time detection of the electrophysiology activities of glucose-excited and glucose-inhibited neurons. The phase-locking level of some glucose-inhibited neurons elevated during fasting (low blood glucose state) and showed theta rhythms after glucose injection (high blood glucose state). With an independent oscillating ability, glucose-inhibited neurons can provide an essential indicator to prevent severe hypoglycemia. The results reveal a mechanism for glucose-sensitive neurons to respond to blood glucose. Some glucose-inhibited neurons can integrate glucose information input and convert it into theta oscillating or phase lock output. It helps in enhancing the interaction between neurons and glucose. Therefore, the research can provide a basis for further controlling blood glucose by modulating the characteristics of neuronal electrophysiology. This helps reduce the damage of organisms under energy-limiting conditions, such as prolonged manned spaceflight or metabolic disorders.
Collapse
Affiliation(s)
- Yiding Wang
- State Key Laboratory of Transducer Technology, Aerospace Information Research Institute, Chinese Academy of Sciences, Beijing 100190, PR China
- School of Electronic, Electrical and Communication Engineering, University of Chinese Academy of Sciences, Beijing 100049, PR China
| | - Xiukun Sui
- State Key Laboratory of Space Medicine Fundamentals and Application, China Astronaut Research and Training Center, Beijing 100094, PR China
| | - Jinping Luo
- State Key Laboratory of Transducer Technology, Aerospace Information Research Institute, Chinese Academy of Sciences, Beijing 100190, PR China
- School of Electronic, Electrical and Communication Engineering, University of Chinese Academy of Sciences, Beijing 100049, PR China
| | - Gucheng Yang
- State Key Laboratory of Transducer Technology, Aerospace Information Research Institute, Chinese Academy of Sciences, Beijing 100190, PR China
- School of Electronic, Electrical and Communication Engineering, University of Chinese Academy of Sciences, Beijing 100049, PR China
| | - Penghui Fan
- State Key Laboratory of Transducer Technology, Aerospace Information Research Institute, Chinese Academy of Sciences, Beijing 100190, PR China
- School of Electronic, Electrical and Communication Engineering, University of Chinese Academy of Sciences, Beijing 100049, PR China
| | - Botao Lu
- State Key Laboratory of Transducer Technology, Aerospace Information Research Institute, Chinese Academy of Sciences, Beijing 100190, PR China
- School of Electronic, Electrical and Communication Engineering, University of Chinese Academy of Sciences, Beijing 100049, PR China
| | - Ming Li
- State Key Laboratory of Transducer Technology, Aerospace Information Research Institute, Chinese Academy of Sciences, Beijing 100190, PR China
- School of Electronic, Electrical and Communication Engineering, University of Chinese Academy of Sciences, Beijing 100049, PR China
| | - Zhaojie Xu
- State Key Laboratory of Transducer Technology, Aerospace Information Research Institute, Chinese Academy of Sciences, Beijing 100190, PR China
- School of Electronic, Electrical and Communication Engineering, University of Chinese Academy of Sciences, Beijing 100049, PR China
| | - Lina Qu
- State Key Laboratory of Space Medicine Fundamentals and Application, China Astronaut Research and Training Center, Beijing 100094, PR China
| | - Yilin Song
- State Key Laboratory of Transducer Technology, Aerospace Information Research Institute, Chinese Academy of Sciences, Beijing 100190, PR China
- School of Electronic, Electrical and Communication Engineering, University of Chinese Academy of Sciences, Beijing 100049, PR China
| | - Yinghui Li
- State Key Laboratory of Space Medicine Fundamentals and Application, China Astronaut Research and Training Center, Beijing 100094, PR China
| | - Xinxia Cai
- State Key Laboratory of Transducer Technology, Aerospace Information Research Institute, Chinese Academy of Sciences, Beijing 100190, PR China
- School of Electronic, Electrical and Communication Engineering, University of Chinese Academy of Sciences, Beijing 100049, PR China
| |
Collapse
|
6
|
McCrimmon RJ. Remembrance of things past: The consequences of recurrent hypoglycaemia in diabetes. Diabet Med 2022; 39:e14973. [PMID: 36251572 PMCID: PMC10015985 DOI: 10.1111/dme.14973] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/08/2022] [Revised: 10/03/2022] [Accepted: 10/13/2022] [Indexed: 01/18/2023]
Abstract
AIMS People with type 1 and type 2 diabetes still frequently experience hypoglycaemia, which can be severe, leading to loss of consciousness. This review will examine the cellular consequences of recurrent hypoglycaemia. METHODS This review, based on the Dorothy Hodgkin Lecture given at the Diabetes UK 2022 annual symposium by the author, will discuss our current understanding of the mechanisms by which hypoglycaemia is detected and the consequences of recurrent exposure to hypoglycaemia. RESULTS Glucose-responsive cells found in the periphery as well as multiple areas of the brain are organised in a classical sensori-motor integrative network encompassing peripheral, hindbrain and hypothalamic components. The mechanism used by glucose-responsive neurons to detect hypoglycaemia parallel those of the classical glucose sensor the pancreatic ß-cell, namely in their use of glucokinase, KATP channels and AMP-activated protein kinase. Recurrent exposure to hypoglycaemia results in a series of cellular adaptations that may be designed to increase the resilience of cells to future hypoglycaemia. This review also highlights how hypoglycaemia, as an oxidative stressor, may also exacerbate chronic hyperglycaemia-induced increases in oxidative stress and inflammation, leading to damage to vulnerable brain regions. CONCLUSIONS Impaired awareness of hypoglycaemia follows the adaptation of central glucose-responsive neurons to repeated hypoglycaemia and may represent a form of memory called habituation. In diabetes, recurrent hypoglycaemia may have tissue consequences as a result of a profound disruption in the cellular response to a hypoglycaemic challenge that increases vulnerability to oxidative damage.
Collapse
Affiliation(s)
- Rory J. McCrimmon
- Systems Medicine, School of MedicineUniversity of Dundee, Ninewells Hospital and Medical SchoolDundeeUK
| |
Collapse
|
7
|
Peters A, Sprengell M, Kubera B. The principle of 'brain energy on demand' and its predictive power for stress, sleep, stroke, obesity and diabetes. Neurosci Biobehav Rev 2022; 141:104847. [PMID: 36067964 DOI: 10.1016/j.neubiorev.2022.104847] [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: 01/08/2022] [Revised: 08/10/2022] [Accepted: 08/26/2022] [Indexed: 12/01/2022]
Abstract
Does the brain actively draw energy from the body when needed? There are different schools of thought regarding energy metabolism. In this study, the various theoretical models are classified into one of two categories: (1) conceptualizations of the brain as being purely passively supplied, which we call 'P-models,' and (2) models understanding the brain as not only passively receiving energy but also actively procuring energy for itself on demand, which we call 'A-models.' One prominent example of such theories making use of an A-model is the selfish-brain theory. The ability to make predictions was compared between the A- and P-models. A-models were able to predict and coherently explain all data examined, which included stress, sleep, caloric restriction, stroke, type-1-diabetes mellitus, obesity, and type-2-diabetes, whereas the predictions of P-models failed in most cases. The strength of the evidence supporting A-models is based on the coherence of accurate predictions across a spectrum of metabolic states. The theory test conducted here speaks to a brain that pulls its energy from the body on-demand.
Collapse
Affiliation(s)
- Achim Peters
- Medical Clinic 1, Center of Brain, Behavior and Metabolism, University of Lübeck, Ratzeburger Allee 160, D-23538 Lübeck, Germany.
| | - Marie Sprengell
- Medical Clinic 1, Center of Brain, Behavior and Metabolism, University of Lübeck, Ratzeburger Allee 160, D-23538 Lübeck, Germany
| | - Britta Kubera
- Medical Clinic 1, Center of Brain, Behavior and Metabolism, University of Lübeck, Ratzeburger Allee 160, D-23538 Lübeck, Germany
| |
Collapse
|
8
|
Rawlinson S, Reichenbach A, Clarke RE, Nuñez-Iglesias J, Dempsey H, Lockie SH, Andrews ZB. In Vivo Photometry Reveals Insulin and 2-Deoxyglucose Maintain Prolonged Inhibition of VMH Vglut2 Neurons in Male Mice. Endocrinology 2022; 163:6631280. [PMID: 35788848 DOI: 10.1210/endocr/bqac095] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/09/2022] [Indexed: 11/19/2022]
Abstract
The ventromedial hypothalamic (VMH) nucleus is a well-established hub for energy and glucose homeostasis. In particular, VMH neurons are thought to be important for initiating the counterregulatory response to hypoglycemia, and ex vivo electrophysiology and immunohistochemistry data indicate a clear role for VMH neurons in sensing glucose concentration. However, the temporal response of VMH neurons to physiologically relevant changes in glucose availability in vivo has been hampered by a lack of available tools for measuring neuronal activity over time. Since the majority of neurons within the VMH are glutamatergic and can be targeted using the vesicular glutamate transporter Vglut2, we expressed cre-dependent GCaMP7s in Vglut2 cre mice and examined the response profile of VMH to intraperitoneal injections of glucose, insulin, and 2-deoxyglucose (2DG). We show that reduced available glucose via insulin-induced hypoglycemia and 2DG-induced glucoprivation, but not hyperglycemia induced by glucose injection, inhibits VMH Vglut2 neuronal population activity in vivo. Surprisingly, this inhibition was maintained for at least 45 minutes despite prolonged hypoglycemia and initiation of a counterregulatory response. Thus, although VMH stimulation, via pharmacological, electrical, or optogenetic approaches, is sufficient to drive a counterregulatory response, our data suggest VMH Vglut2 neurons are not the main drivers required to do so, since VMH Vglut2 neuronal population activity remains suppressed during hypoglycemia and glucoprivation.
Collapse
Affiliation(s)
- Sasha Rawlinson
- Monash Biomedicine Discovery Institute and Department of Physiology, Monash University, Clayton, Victoria, 3800, Australia
| | - Alex Reichenbach
- Monash Biomedicine Discovery Institute and Department of Physiology, Monash University, Clayton, Victoria, 3800, Australia
| | - Rachel E Clarke
- Department of Neurosciences, Medical University of South Carolina, Charleston, South Carolina 29425, USA
| | - Juan Nuñez-Iglesias
- Monash Biomedicine Discovery Institute and Department of Anatomy and Developmental Biology, Monash University, Clayton, Victoria, 3800, Australia
| | - Harry Dempsey
- Monash Biomedicine Discovery Institute and Department of Physiology, Monash University, Clayton, Victoria, 3800, Australia
| | - Sarah H Lockie
- Monash Biomedicine Discovery Institute and Department of Physiology, Monash University, Clayton, Victoria, 3800, Australia
| | - Zane B Andrews
- Monash Biomedicine Discovery Institute and Department of Physiology, Monash University, Clayton, Victoria, 3800, Australia
| |
Collapse
|
9
|
Sprengell M, Kubera B, Peters A. Brain Mass (Energy) Resistant to Hyperglycaemic Oversupply: A Systematic Review. Front Neurosci 2021; 15:740502. [PMID: 34803585 PMCID: PMC8600366 DOI: 10.3389/fnins.2021.740502] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2021] [Accepted: 10/13/2021] [Indexed: 12/09/2022] Open
Abstract
Cerebral energy supply is determined by the energy content of the blood. Accordingly, the brain is undersupplied during hypoglycaemia. Whether or not there is an additional cerebral energy demand that depends upon the energy content of the brain is considered differently in two opposing theoretical approaches. The Selfish-Brain theory postulates that the brain actively demands energy from the body when needed, while long-held theories, the gluco-lipostatic theory and its variants, deny such active brain involvement and view the brain as purely passively supplied. Here we put the competing theories to the test. We conducted a systematic review of a condition in which the rival theories make opposite predictions, i.e., experimental T1DM. The Selfish-Brain theory predicts that induction of experimental type 1 diabetes causes minor mass (energy) changes in the brain as opposed to major glucose changes in the blood. This prediction becomes our hypothesis to be tested here. A total of 608 works were screened by title and abstract, and 64 were analysed in full text. According to strict selection criteria defined in our PROSPERO preannouncement and complying with PRISMA guidelines, 18 studies met all inclusion criteria. Thirteen studies provided sufficient data to test our hypothesis. The 13 evaluable studies (15 experiments) showed that the diabetic groups had blood glucose concentrations that differed from controls by +294 ± 96% (mean ± standard deviation) and brain mass (energy) that differed from controls by −4 ± 13%, such that blood changes were an order of magnitude greater than brain changes (T = 11.5, df = 14, p < 0.001). This finding confirms not only our hypothesis but also the prediction of the Selfish-Brain theory, while the predictions of the gluco-lipostatic theory and its variants were violated. The current paper completes a three-part series of systematic reviews, the two previous papers deal with a distal and a proximal bottleneck in the cerebral brain supply, i.e., caloric restriction and cerebral artery occlusion. All three papers demonstrate that accurate predictions are only possible if one regards the brain as an organ that regulates its energy concentrations independently and occupies a primary position in a hierarchically organised energy metabolism. Systematic Review Registration:https://www.crd.york.ac.uk/prospero/display_record.php?RecordID=156816, PROSPERO, identifier: CRD42020156816.
Collapse
Affiliation(s)
- Marie Sprengell
- Center of Brain, Behavior and Metabolism (CBBM), Medical Clinic 1, University of Lübeck, Lübeck, Germany
| | - Britta Kubera
- Center of Brain, Behavior and Metabolism (CBBM), Medical Clinic 1, University of Lübeck, Lübeck, Germany
| | - Achim Peters
- Center of Brain, Behavior and Metabolism (CBBM), Medical Clinic 1, University of Lübeck, Lübeck, Germany
| |
Collapse
|
10
|
Sprengell M, Kubera B, Peters A. Proximal Disruption of Brain Energy Supply Raises Systemic Blood Glucose: A Systematic Review. Front Neurosci 2021; 15:685031. [PMID: 34248487 PMCID: PMC8264130 DOI: 10.3389/fnins.2021.685031] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2021] [Accepted: 05/28/2021] [Indexed: 12/17/2022] Open
Abstract
This work joins a series that methodically tests the predictions of the Selfish-Brain theory. The theory postulates a vital ability of the mammalian brain, namely to give priority to its own energy metabolism. The brain behaves “selfishly” in this respect. For the cerebral artery occlusion studied here, the theory predicts an increase in blood glucose concentration, what becomes the hypothesis to be tested. We conducted a systematic review of cerebral-artery-occlusion papers to test whether or not the included studies could confirm this hypothesis. We identified 239 records, screened 231 works by title or abstract, and analyzed 89 by full text. According to strict selection criteria (set out in our PROSPERO preregistration, complying with PRISMA guidelines), 7 papers provided enough information to decide on the hypothesis. Our hypothesis could be fully confirmed for the 3 to 24 h after the onset of a transient 2 h or permanent occlusion. As for the mechanism, the theory predicts that the energy-deprived brain suppresses insulin secretion via the sympathoadrenal system, thereby preventing insulin-mediated glucose uptake into muscle and fat and, as a result, enhancing insulin-independent glucose uptake via the blood-brain barrier. Evidence from our included studies actually demonstrated cerebral insulin suppression. In all, the current work confirms the second major prediction of the Selfish-Brain theory that relates to a proximal bottleneck of the cerebral supply chain, cerebral artery occlusion. Its first major prediction relates to a distal supply bottleneck, caloric restriction, and is fulfilled as shown by our previous work, whereas the prediction of the long held gluco-lipostatic theory, which sees the brain as only passively supplied, is violated (Sprengell et al., 2021). The crucial point was that caloric restriction elicits smaller changes in mass (energy) in the brain than in the body. Taken together, the evidence from the current and previous work clearly shows that the most accurate predictions are possible with a theory that views the brain as an independently self-regulating energy compartment occupying a primary position in energy metabolism.
Collapse
Affiliation(s)
- Marie Sprengell
- Medical Clinic 1, Center of Brain, Behavior and Metabolism (CBBM), University of Lübeck, Lübeck, Germany
| | - Britta Kubera
- Medical Clinic 1, Center of Brain, Behavior and Metabolism (CBBM), University of Lübeck, Lübeck, Germany
| | - Achim Peters
- Medical Clinic 1, Center of Brain, Behavior and Metabolism (CBBM), University of Lübeck, Lübeck, Germany
| |
Collapse
|
11
|
Vickneson K, Blackburn J, Gallagher JR, Evans ML, de Galan BE, Pedersen-Bjergaard U, Thorens B, McNeilly AD, McCrimmon RJ. Cold-induced dishabituation in rodents exposed to recurrent hypoglycaemia. Diabetologia 2021; 64:1436-1441. [PMID: 33730186 PMCID: PMC8099849 DOI: 10.1007/s00125-021-05425-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/21/2020] [Accepted: 01/19/2021] [Indexed: 11/04/2022]
Abstract
AIMS/HYPOTHESIS Recurrent hypoglycaemia in people with diabetes leads to progressive suppression of counterregulatory hormonal responses to subsequent hypoglycaemia. Recently it has been proposed that the mechanism underpinning this is a form of adaptive memory referred to as habituation. To test this hypothesis, we use two different durations of cold exposure to examine whether rodents exposed to recurrent hypoglycaemia exhibit two characteristic features of habituation, namely stimulus generalisation and dishabituation. METHODS In the first study (stimulus generalisation study), hyperinsulinaemic-hypoglycaemic (2.8 mmol/l) glucose clamps were performed in non-diabetic rodents exposed to prior moderate-duration cold (4°C for 3 h) or control conditions. In the second study (dishabituation study), rodents exposed to prior recurrent hypoglycaemia or saline (154 mmol/l NaCl) injections over 4 weeks underwent a longer-duration cold (4°C for 4.5 h) exposure followed 24 h later by a hyperinsulinaemic-hypoglycaemic (2.8 mmol/l) glucose clamp. Output measures were counterregulatory hormone responses during experimental hypoglycaemia. RESULTS Moderate-duration cold exposure blunted the adrenaline (epinephrine) response (15,266 ± 1920 vs 7981 ± 1258 pmol/l, Control vs Cold; p < 0.05) to next day hypoglycaemia in healthy non-diabetic rodents. In contrast, the suppressed adrenaline response (Control 5912 ± 1417 vs recurrent hypoglycaemia 1836 ± 736 pmol/l; p < 0.05) that is associated with recurrent hypoglycaemia was restored following longer-duration cold exposure (recurrent hypoglycaemia + Cold 4756 ± 826 pmol/l; not significant vs Control). CONCLUSIONS/INTERPRETATION Non-diabetic rodents exhibit two cardinal features of habituation, namely stimulus generalisation and dishabituation. These findings provide further support for the hypothesis that suppressed counterregulatory responses following exposure to recurrent hypoglycaemia in diabetes result from habituation.
Collapse
Affiliation(s)
| | - Jessica Blackburn
- Division of Systems Medicine, School of Medicine, University of Dundee, Dundee, UK
| | - Jennifer R Gallagher
- Division of Systems Medicine, School of Medicine, University of Dundee, Dundee, UK
| | - Mark L Evans
- Wellcome Trust/MRC Institute of Metabolic Science, Cambridge, Cambridge, UK
| | - Bastiaan E de Galan
- Radboud University Medical Center, Radboud University Nijmegen, Nijmegen, the Netherlands
- Maastricht University Medical Center+, Maastricht University, Maastricht, the Netherlands
- CARIM School for Cardiovascular Medicine, Maastricht University, Maastricht, the Netherlands
| | | | - Bernard Thorens
- Faculty of Biology and Medicine, University of Lausanne, Lausanne, Switzerland
| | - Alison D McNeilly
- Division of Systems Medicine, School of Medicine, University of Dundee, Dundee, UK
| | - Rory J McCrimmon
- Division of Systems Medicine, School of Medicine, University of Dundee, Dundee, UK.
| |
Collapse
|
12
|
Garcia SM, Hirschberg PR, Sarkar P, Siegel DM, Teegala SB, Vail GM, Routh VH. Insulin actions on hypothalamic glucose-sensing neurones. J Neuroendocrinol 2021; 33:e12937. [PMID: 33507001 PMCID: PMC10561189 DOI: 10.1111/jne.12937] [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] [Received: 10/27/2020] [Revised: 12/22/2020] [Accepted: 12/29/2020] [Indexed: 12/13/2022]
Abstract
Subsequent to the discovery of insulin 100 years ago, great strides have been made in understanding its function, especially in the brain. It is now clear that insulin is a critical regulator of the neuronal circuitry controlling energy balance and glucose homeostasis. This review focuses on the effects of insulin and diabetes on the activity and glucose sensitivity of hypothalamic glucose-sensing neurones. We highlight the role of electrophysiological data in understanding how insulin regulates glucose-sensing neurones. A brief introduction describing the benefits and limitations of the major electrophysiological techniques used to investigate glucose-sensing neurones is provided. The mechanisms by which hypothalamic neurones sense glucose are discussed with an emphasis on those glucose-sensing neurones already shown to be modulated by insulin. Next, the literature pertaining to how insulin alters the activity and glucose sensitivity of these hypothalamic glucose-sensing neurones is described. In addition, the effects of impaired insulin signalling during diabetes and the ramifications of insulin-induced hypoglycaemia on hypothalamic glucose-sensing neurones are covered. To the extent that it is known, we present hypotheses concerning the mechanisms underlying the effects of these insulin-related pathologies. To conclude, electrophysiological data from the hippocampus are evaluated aiming to provide clues regarding how insulin might influence neuronal plasticity in glucose-sensing neurones. Although much has been accomplished subsequent to the discovery of insulin, the work described in our review suggests that the regulation of central glucose sensing by this hormone is both important and understudied.
Collapse
Affiliation(s)
- Stephanie M Garcia
- Department of Pharmacology, Physiology and Neuroscience, Rutgers, New Jersey Medical School, The State University of New Jersey, Newark, NJ, USA
| | - Pamela R Hirschberg
- Department of Pharmacology, Physiology and Neuroscience, Rutgers, New Jersey Medical School, The State University of New Jersey, Newark, NJ, USA
| | - Pallabi Sarkar
- Department of Pharmacology, Physiology and Neuroscience, Rutgers, New Jersey Medical School, The State University of New Jersey, Newark, NJ, USA
| | - Dashiel M Siegel
- Department of Pharmacology, Physiology and Neuroscience, Rutgers, New Jersey Medical School, The State University of New Jersey, Newark, NJ, USA
| | - Suraj B Teegala
- Department of Pharmacology, Physiology and Neuroscience, Rutgers, New Jersey Medical School, The State University of New Jersey, Newark, NJ, USA
| | - Gwyndolin M Vail
- Department of Pharmacology, Physiology and Neuroscience, Rutgers, New Jersey Medical School, The State University of New Jersey, Newark, NJ, USA
| | - Vanessa H Routh
- Department of Pharmacology, Physiology and Neuroscience, Rutgers, New Jersey Medical School, The State University of New Jersey, Newark, NJ, USA
| |
Collapse
|
13
|
Sprengell M, Kubera B, Peters A. Brain More Resistant to Energy Restriction Than Body: A Systematic Review. Front Neurosci 2021; 15:639617. [PMID: 33633541 PMCID: PMC7900631 DOI: 10.3389/fnins.2021.639617] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2020] [Accepted: 01/07/2021] [Indexed: 01/10/2023] Open
Abstract
The gluco-lipostatic theory and its modern variants assume that blood glucose and energy stores are controlled in closed-loop feedback processes. The Selfish Brain theory is based on the same assumptions, but additionally postulates that the brain, as an independent energy compartment, self-regulates its energy concentration with the highest priority. In some clinical situations these two theories make opposite predictions. To investigate one of these situations, namely caloric restriction, we formulated a hypothesis which, if confirmed, would match the predictions of the Selfish Brain theory—but not those of the gluco-lipostatic theory. Hypothesis: Calorie restriction causes minor mass (energy) changes in the brain as opposed to major changes in the body. We conducted a systematic review of caloric-restriction studies to test whether or not the evaluated studies confirmed this hypothesis. We identified 3,157 records, screened 2,804 works by title or abstract, and analyzed 232 by full text. According to strict selection criteria (set out in our PROSPERO preregistration, complying with PRISMA guidelines, and the pre-defined hypothesis-decision algorithm), 8 papers provided enough information to decide on the hypothesis: In animals, high-energy phosphates were measured by 31P-nuclear magnetic resonance, and organ and total body weights were measured by scales, while in humans organ sizes were determined by magnetic resonance imaging. All 8 decidable papers confirmed the hypothesis, none spoke against it. The evidence presented here clearly shows that the most accurate predictions are possible with a theory that regards the brain as independently self-regulating and as occupying a primary position in a hierarchically organized energy metabolism.
Collapse
Affiliation(s)
- Marie Sprengell
- Center of Brain, Behavior and Metabolism (CBBM), University of Lübeck, Lübeck, Germany
| | - Britta Kubera
- Center of Brain, Behavior and Metabolism (CBBM), University of Lübeck, Lübeck, Germany
| | - Achim Peters
- Center of Brain, Behavior and Metabolism (CBBM), University of Lübeck, Lübeck, Germany
| |
Collapse
|
14
|
Wen S, Nguyen T, Gong M, Yuan X, Wang C, Jin J, Zhou L. An Overview of Similarities and Differences in Metabolic Actions and Effects of Central Nervous System Between Glucagon-Like Peptide-1 Receptor Agonists (GLP-1RAs) and Sodium Glucose Co-Transporter-2 Inhibitors (SGLT-2is). Diabetes Metab Syndr Obes 2021; 14:2955-2972. [PMID: 34234493 PMCID: PMC8254548 DOI: 10.2147/dmso.s312527] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/24/2021] [Accepted: 06/15/2021] [Indexed: 12/17/2022] Open
Abstract
GLP-1 receptor agonists (GLP-1RAs) and SGLT-2 inhibitors (SGLT-2is) are novel antidiabetic medications associated with considerable cardiovascular benefits therapying treatment of diabetic patients. GLP-1 exhibits atherosclerosis resistance, whereas SGLT-2i acts to ameliorate the neuroendocrine state in the patients with chronic heart failure. Despite their distinct modes of action, both factors share pathways by regulating the central nervous system (CNS). While numerous preclinical and clinical studies have demonstrated that GLP-1 can access various nuclei associated with energy homeostasis and hedonic eating in the CNS via blood-brain barrier (BBB), research on the activity of SGLT-2is remains limited. In our previous studies, we demonstrated that both GLP-1 receptor agonists (GLP-1RAs) liraglutide and exenatide, as well as an SGLT-2i, dapagliflozin, could activate various nuclei and pathways in the CNS of Sprague Dawley (SD) rats and C57BL/6 mice, respectively. Moreover, our results revealed similarities and differences in neural pathways, which possibly regulated different metabolic effects of GLP-1RA and SGLT-2i via sympathetic and parasympathetic systems in the CNS, such as feeding, blood glucose regulation and cardiovascular activities (arterial blood pressure and heart rate control). In the present article, we extensively discuss recent preclinical studies on the effects of GLP-1RAs and SGLT-2is on the CNS actions, with the aim of providing a theoretical explanation on their mechanism of action in improvement of the macro-cardiovascular risk and reducing incidence of diabetic complications. Overall, these findings are expected to guide future drug design approaches.
Collapse
Affiliation(s)
- Song Wen
- Department of Endocrinology, Shanghai Pudong Hospital, Fudan University, Shanghai, 201399, People’s Republic of China
| | - Thiquynhnga Nguyen
- Department of Endocrinology, Shanghai Pudong Hospital, Fudan University, Shanghai, 201399, People’s Republic of China
| | - Min Gong
- Department of Endocrinology, Shanghai Pudong Hospital, Fudan University, Shanghai, 201399, People’s Republic of China
| | - Xinlu Yuan
- Department of Endocrinology, Shanghai Pudong Hospital, Fudan University, Shanghai, 201399, People’s Republic of China
| | - Chaoxun Wang
- Department of Endocrinology, Shanghai Pudong Hospital, Fudan University, Shanghai, 201399, People’s Republic of China
| | - Jianlan Jin
- Department of Endocrinology, Shanghai Pudong Hospital, Fudan University, Shanghai, 201399, People’s Republic of China
| | - Ligang Zhou
- Department of Endocrinology, Shanghai Pudong Hospital, Fudan University, Shanghai, 201399, People’s Republic of China
- Shanghai Key Laboratory of Vascular Lesions Regulation and Remodeling, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai, People’s Republic of China
- Correspondence: Ligang Zhou Department of Endocrinology, Shanghai Pudong Hospital, Fudan University, Shanghai, 201399, People’s Republic of ChinaTel +8613611927616 Email
| |
Collapse
|
15
|
Stoelzel CR, Zhang Y, Cincotta AH. Circadian-timed dopamine agonist treatment reverses high-fat diet-induced diabetogenic shift in ventromedial hypothalamic glucose sensing. Endocrinol Diabetes Metab 2020; 3:e00139. [PMID: 32704560 PMCID: PMC7375120 DOI: 10.1002/edm2.139] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2020] [Accepted: 03/28/2020] [Indexed: 12/19/2022] Open
Abstract
INTRODUCTION Within the ventromedial hypothalamus (VMH), glucose inhibitory (GI) neurons sense hypoglycaemia while glucose excitatory (GE) neurons sense hyperglycaemia to initiate counter control mechanisms under normal conditions. However, potential electrophysiological alterations of these two neuronal types in vivo in insulin-resistant states have never been simultaneously fully documented. Further, the anti-diabetic effect of dopamine agonism on this VMH system under insulin resistance has not been studied. METHODS This study examined the impact of a high-fat diet (HFD) on in vivo electrophysiological recordings from VMH GE and GI neurons and the ability of circadian-timed dopamine agonist therapy to reverse any adverse effect of the HFD on such VMH activities and peripheral glucose metabolism. RESULTS HFD significantly inhibited VMH GE neuronal electrophysiological response to local hyperglycaemia (36.3%) and augmented GI neuronal excitation response to local hypoglycaemia (47.0%). Bromocriptine (dopamine agonist) administration at onset of daily activity (but not during the daily sleep phase) completely reversed both VMH GE and GI neuronal aberrations induced by HFD. Such timed treatment also normalized glucose intolerance and insulin resistance. These VMH and peripheral glucose metabolism effects of circadian-timed bromocriptine may involve its known effect to reduce elevated VMH noradrenergic activity in insulin-resistant states as local VMH administration of norepinephrine was observed to significantly inhibit VMH GE neuronal sensing of local hyperglycaemia in insulin-sensitive animals on regular chow diet (52.4%). CONCLUSIONS HFD alters VMH glucose sensing in a manner that potentiates hyperglycaemia and this effect on the VMH can be reversed by appropriately circadian-timed dopamine agonist administration.
Collapse
|
16
|
He Y, Xu P, Wang C, Xia Y, Yu M, Yang Y, Yu K, Cai X, Qu N, Saito K, Wang J, Hyseni I, Robertson M, Piyarathna B, Gao M, Khan SA, Liu F, Chen R, Coarfa C, Zhao Z, Tong Q, Sun Z, Xu Y. Estrogen receptor-α expressing neurons in the ventrolateral VMH regulate glucose balance. Nat Commun 2020; 11:2165. [PMID: 32358493 PMCID: PMC7195451 DOI: 10.1038/s41467-020-15982-7] [Citation(s) in RCA: 44] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2020] [Accepted: 04/06/2020] [Indexed: 12/20/2022] Open
Abstract
Brain glucose-sensing neurons detect glucose fluctuations and prevent severe hypoglycemia, but mechanisms mediating functions of these glucose-sensing neurons are unclear. Here we report that estrogen receptor-α (ERα)-expressing neurons in the ventrolateral subdivision of the ventromedial hypothalamic nucleus (vlVMH) can sense glucose fluctuations, being glucose-inhibited neurons (GI-ERαvlVMH) or glucose-excited neurons (GE-ERαvlVMH). Hypoglycemia activates GI-ERαvlVMH neurons via the anoctamin 4 channel, and inhibits GE-ERαvlVMH neurons through opening the ATP-sensitive potassium channel. Further, we show that GI-ERαvlVMH neurons preferentially project to the medioposterior arcuate nucleus of the hypothalamus (mpARH) and GE-ERαvlVMH neurons preferentially project to the dorsal Raphe nuclei (DRN). Activation of ERαvlVMH to mpARH circuit and inhibition of ERαvlVMH to DRN circuit both increase blood glucose. Thus, our results indicate that ERαvlVMH neurons detect glucose fluctuations and prevent severe hypoglycemia in mice.
Collapse
Affiliation(s)
- Yanlin He
- Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Pingwen Xu
- Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Chunmei Wang
- Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Yan Xia
- Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Meng Yu
- Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Yongjie Yang
- Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Kaifan Yu
- Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Xing Cai
- Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Na Qu
- Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Kenji Saito
- Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Julia Wang
- Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Ilirjana Hyseni
- Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Matthew Robertson
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Badrajee Piyarathna
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Min Gao
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Sohaib A Khan
- Department of Cell and Cancer Biology, Vontz Center for Molecular Studies, University of Cincinnati, College of Medicine, Cincinnati, OH, 45267, USA
| | - Feng Liu
- Departments of Pharmacology, University of Texas Health at San Antonio, San Antonio, TX, 78229, USA
| | - Rui Chen
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Cristian Coarfa
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Zhongming Zhao
- Center for Precision Health, School of Biomedical Informatics, The University of Texas Health Science Center at Houston, Houston, TX, 77030, USA
| | - Qingchun Tong
- Brown Foundation Institute of Molecular Medicine, University of Texas Health Science Center at Houston, Houston, TX, 77030, USA
| | - Zheng Sun
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, 77030, USA
- Department of Medicine, Division of Diabetes, Endocrinology and Metabolism, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Yong Xu
- Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, Houston, TX, 77030, USA.
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, 77030, USA.
| |
Collapse
|
17
|
Nguyen T, Wen S, Gong M, Yuan X, Xu D, Wang C, Jin J, Zhou L. Dapagliflozin Activates Neurons in the Central Nervous System and Regulates Cardiovascular Activity by Inhibiting SGLT-2 in Mice. Diabetes Metab Syndr Obes 2020; 13:2781-2799. [PMID: 32848437 PMCID: PMC7425107 DOI: 10.2147/dmso.s258593] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/28/2020] [Accepted: 07/10/2020] [Indexed: 02/06/2023] Open
Abstract
PURPOSE This study investigates the possible effect and central mechanism of novel antidiabetic medication sodium glucose transporter-2 (SGLT-2i) on the cardiovascular activity. MATERIAL AND METHODS Thirty-four normal male C57BL/6 mice were randomly assigned to 2 groups to receive single Dapagliflozin (1.52mg/kg) dose via intragastric gavage or a comparable dose of saline. Glycemic level (BG), blood pressure (BP) and heart rate (HR) were measured 2 hours after administration of the respective treatments. Immunohistochemical tests were performed to determine the effect of SGLT-2i on neural localization of SGLT-2 and c-Fos, a neural activator. The distributional relationships of SGLT-2 and c-Fos were examined by immunofluorescence. RESULTS Administration of SGLT-2i significantly decreased BP but did not affect the HR. There was no difference in BG between the two groups. Results showed that SGLT-2 was localized to specific regions involved in autonomic control. Expression of c-Fos was significantly higher in major critical nuclei in the aforementioned regions in groups treated with Dapagliflozin. CONCLUSION This study demonstrates that SGLT-2 is expressed in CNS tissues involved in autonomic control and possibly influence cardiovascular function. Dapagliflozin influences central autonomic activity via unidentified pathways by inhibiting central or peripheral SGLT-2. These results provide a new concept that sympathetic inhibition by SGLT-2i can be mediated by central autonomic system, a mechanism that explains how SGLT-2i improves the cardiovascular function.
Collapse
Affiliation(s)
- Thiquynhnga Nguyen
- Department of Endocrinology, Shanghai Pudong Hospital, Fudan University, Shanghai201399, People’s Republic of China
| | - Song Wen
- Department of Endocrinology, Shanghai Pudong Hospital, Fudan University, Shanghai201399, People’s Republic of China
| | - Min Gong
- Department of Endocrinology, Shanghai Pudong Hospital, Fudan University, Shanghai201399, People’s Republic of China
| | - Xinlu Yuan
- Department of Endocrinology, Shanghai Pudong Hospital, Fudan University, Shanghai201399, People’s Republic of China
| | - Dongxiang Xu
- Department of Endocrinology, Shanghai Pudong Hospital, Fudan University, Shanghai201399, People’s Republic of China
| | - Chaoxun Wang
- Department of Endocrinology, Shanghai Pudong Hospital, Fudan University, Shanghai201399, People’s Republic of China
| | - Jianlan Jin
- Department of Endocrinology, Shanghai Pudong Hospital, Fudan University, Shanghai201399, People’s Republic of China
| | - Ligang Zhou
- Department of Endocrinology, Shanghai Pudong Hospital, Fudan University, Shanghai201399, People’s Republic of China
- Correspondence: Ligang Zhou Department of Endocrinology, Shanghai Pudong Hospital, Fudan University, Shanghai201399, People’s Republic of ChinaTel +86 13611927616 Email
| |
Collapse
|
18
|
Gong M, Wen S, Nguyen T, Wang C, Jin J, Zhou L. Converging Relationships of Obesity and Hyperuricemia with Special Reference to Metabolic Disorders and Plausible Therapeutic Implications. Diabetes Metab Syndr Obes 2020; 13:943-962. [PMID: 32280253 PMCID: PMC7125338 DOI: 10.2147/dmso.s232377] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/25/2019] [Accepted: 03/09/2020] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND Obesity and hyperuricemia mutually influence metabolic syndrome. This study discusses the metabolic relationships between obesity and hyperuricemia in terms of pathophysiology, complications, and treatments. METHODS We searched for preclinical or clinical studies on the pathophysiology, complications, and therapy of obesity and hyperuricemia on the PubMed database. RESULTS In this systemic review, we summarized our searching results on topics of pathophysiology, complications and therapeutic strategy. In pathophysiology, we firstly introduce genetic variations for obesity, hyperuricemia and their relationships by genetic studies. Secondly, we talk about the epigenetic influences on obesity and hyperuricemia. Thirdly, we describe the central metabolic regulation and the role of hyperuricemia. Then, we refer to the character of adipose tissue inflammation and oxidative stress in the obesity and hyperuricemia. In the last part of this topic, we reviewed the critical links of gut microbiota in the obesity and hyperuricemia. In the following part, we review the pathophysiology of major complications in obesity and hyperuricemia including insulin resistance and type 2 diabetes mellitus, chronic kidney disease, cardiovascular diseases, and cancers. Finally, we recapitulate the therapeutic strategies especially the novel pharmaceutic interventions for obesity and hyperuricemia, which concurrently show the mutual metabolic influences between two diseases. CONCLUSION The data reviewed here delineate the metabolic relationships between obesity and hyperuricemia, and provide a comprehensive overview of the therapeutic targets for the management of metabolic syndromes.
Collapse
Affiliation(s)
- Min Gong
- Department of Endocrinology, Shanghai Pudong Hospital, Fudan University, Shanghai201399, People’s Republic of China
| | - Song Wen
- Department of Endocrinology, Shanghai Pudong Hospital, Fudan University, Shanghai201399, People’s Republic of China
| | - Thiquynhnga Nguyen
- Department of Endocrinology, Shanghai Pudong Hospital, Fudan University, Shanghai201399, People’s Republic of China
| | - Chaoxun Wang
- Department of Endocrinology, Shanghai Pudong Hospital, Fudan University, Shanghai201399, People’s Republic of China
| | - Jianlan Jin
- Department of Endocrinology, Shanghai Pudong Hospital, Fudan University, Shanghai201399, People’s Republic of China
| | - Ligang Zhou
- Department of Endocrinology, Shanghai Pudong Hospital, Fudan University, Shanghai201399, People’s Republic of China
- Correspondence: Ligang Zhou Department of Endocrinology, Shanghai Pudong Hospital, Fudan University, Shanghai201399, ChinaTel +8613611927616 Email
| |
Collapse
|
19
|
Hirschberg PR, Sarkar P, Teegala SB, Routh VH. Ventromedial hypothalamus glucose-inhibited neurones: A role in glucose and energy homeostasis? J Neuroendocrinol 2020; 32:e12773. [PMID: 31329314 PMCID: PMC7074896 DOI: 10.1111/jne.12773] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/01/2019] [Revised: 06/18/2019] [Accepted: 07/14/2019] [Indexed: 12/20/2022]
Abstract
The ventromedial hypothalamus (VMH) plays a complex role in glucose and energy homeostasis. The VMH is necessary for the counter-regulatory response to hypoglycaemia (CRR) that increases hepatic gluconeogenesis to restore euglycaemia. On the other hand, the VMH also restrains hepatic glucose production during euglycaemia and stimulates peripheral glucose uptake. The VMH is also important for the ability of oestrogen to increase energy expenditure. This latter function is mediated by VMH modulation of the lateral/perifornical hypothalamic area (lateral/perifornical hypothalamus) orexin neurones. Activation of VMH AMP-activated protein kinase (AMPK) is necessary for the CRR. By contrast, VMH AMPK inhibition favours decreased basal glucose levels and is required for oestrogen to increase energy expenditure. Specialised VMH glucose-sensing neurones confer the ability to sense and respond to changes in blood glucose levels. Glucose-excited (GE) neurones increase and glucose-inhibited (GI) neurones decrease their activity as glucose levels rise. VMH GI neurones, in particular, appear to be important in the CRR, although a role for GE neurones cannot be discounted. AMPK mediates glucose sensing in VMH GI neurones suggesting that, although activation of these neurones is important for the CRR, it is necessary to silence them to lower basal glucose levels and enable oestrogen to increase energy expenditure. In support of this, we found that oestrogen reduces activation of VMH GI neurones in low glucose by inhibiting AMPK. In this review, we present the evidence underlying the role of the VMH in glucose and energy homeostasis. We then discuss the role of VMH glucose-sensing neurones in mediating these effects, with a strong emphasis on oestrogenic regulation of glucose sensing and how this may affect glucose and energy homeostasis.
Collapse
Affiliation(s)
- Pamela R Hirschberg
- Department of Pharmacology, Physiology and Neurosciences, Rutgers New Jersey Medical School, The State University of New Jersey, Newark, NJ, USA
| | - Pallabi Sarkar
- Department of Pharmacology, Physiology and Neurosciences, Rutgers New Jersey Medical School, The State University of New Jersey, Newark, NJ, USA
| | - Suraj B Teegala
- Department of Pharmacology, Physiology and Neurosciences, Rutgers New Jersey Medical School, The State University of New Jersey, Newark, NJ, USA
| | - Vanessa H Routh
- Department of Pharmacology, Physiology and Neurosciences, Rutgers New Jersey Medical School, The State University of New Jersey, Newark, NJ, USA
| |
Collapse
|
20
|
Ichikawa R, Takano K, Fujimoto K, Motomiya T, Kobayashi M, Kitamura T, Shichiri M. Basal glucagon hypersecretion and response to oral glucose load in prediabetes and mild type 2 diabetes. Endocr J 2019; 66:663-675. [PMID: 31142688 DOI: 10.1507/endocrj.ej18-0372] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
Dysregulation of glucagon secretion plays an important role in the pathogenesis of type 2 diabetes (T2DM). However it hasn't been elucidated involvement of glucagon dysregulation in pathophysiology of T2DM. Recently a new glucagon sandwich enzyme-linked immunosorbent assay (ELISA) became available that can measure plasma glucagon level with higher accuracy and simpler procedure than the conventional RIA method. We performed OGTT for adult subjects aged 20-69 years to define normal glucose tolerance (NGT, n = 25), borderline glucose intolerance (defined as pre-diabetes mellitus: preDM, n = 15), or diabetes mellitus (DM, n = 13), and we measured glucagon levels with this new ELISA method at fasting and during OGTT. Plasma glucose, insulin, glucagon and active GLP-1 were also measured. This study took place in diabetes outpatient clinic in Kitasato University Hospital and an affiliated outpatient clinic. PreDM and DM exhibited higher fasting plasma glucagon levels than NGT (34.4 ± 4.6 and 44.1 ± 5.0 vs. 20.6 ± 3.6 pg/mL), and statistical significance was observed between NGT and DM (p < 0.05). There was significant correlation between fasting glucagon level and indexes of insulin sensitivity. During OGTT, glucagon levels were less suppressed in DM and preDM than in NGT, whereas no apparent relationship was observed between glucagon and GLP-1 secretion. Significant positive correlation was observed between glucagon levels during OGTT and fasting TG. In conclusion, subjects with mild T2DM exhibited fasting hyperglucagonemia and insufficient suppression to oral glucose load compared to NGT subjects.
Collapse
Affiliation(s)
- Raishi Ichikawa
- Department of Diabetes, Endocrinology & Metabolism, Kitasato University, School of Medicine, Kanagawa 252-0374, Japan
| | - Koji Takano
- Department of Diabetes, Endocrinology & Metabolism, Kitasato University, School of Medicine, Kanagawa 252-0374, Japan
| | - Kazumi Fujimoto
- Department of Diabetes, Endocrinology & Metabolism, Kitasato University, School of Medicine, Kanagawa 252-0374, Japan
| | | | - Masaki Kobayashi
- Metabolic Signal Research Center, Institute for Molecular and Cellular Regulation, Gunma University, Gunma 371-8512, Japan
| | - Tadahiro Kitamura
- Metabolic Signal Research Center, Institute for Molecular and Cellular Regulation, Gunma University, Gunma 371-8512, Japan
| | - Masayoshi Shichiri
- Department of Diabetes, Endocrinology & Metabolism, Kitasato University, School of Medicine, Kanagawa 252-0374, Japan
| |
Collapse
|
21
|
Stanley S, Moheet A, Seaquist ER. Central Mechanisms of Glucose Sensing and Counterregulation in Defense of Hypoglycemia. Endocr Rev 2019; 40:768-788. [PMID: 30689785 PMCID: PMC6505456 DOI: 10.1210/er.2018-00226] [Citation(s) in RCA: 59] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/07/2018] [Accepted: 01/17/2019] [Indexed: 12/12/2022]
Abstract
Glucose homeostasis requires an organism to rapidly respond to changes in plasma glucose concentrations. Iatrogenic hypoglycemia as a result of treatment with insulin or sulfonylureas is the most common cause of hypoglycemia in humans and is generally only seen in patients with diabetes who take these medications. The first response to a fall in glucose is the detection of impending hypoglycemia by hypoglycemia-detecting sensors, including glucose-sensing neurons in the hypothalamus and other regions. This detection is then linked to a series of neural and hormonal responses that serve to prevent the fall in blood glucose and restore euglycemia. In this review, we discuss the current state of knowledge about central glucose sensing and how detection of a fall in glucose leads to the stimulation of counterregulatory hormone and behavior responses. We also review how diabetes and recurrent hypoglycemia impact glucose sensing and counterregulation, leading to development of impaired awareness of hypoglycemia in diabetes.
Collapse
Affiliation(s)
- Sarah Stanley
- Diabetes, Obesity and Metabolism Institute, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Amir Moheet
- Division of Diabetes, Endocrinology, and Metabolism, Department of Medicine, University of Minnesota, Minneapolis, Minnesota
| | - Elizabeth R Seaquist
- Division of Diabetes, Endocrinology, and Metabolism, Department of Medicine, University of Minnesota, Minneapolis, Minnesota
| |
Collapse
|
22
|
Wen S, Wang C, Gong M, Zhou L. An overview of energy and metabolic regulation. SCIENCE CHINA-LIFE SCIENCES 2018; 62:771-790. [PMID: 30367342 DOI: 10.1007/s11427-018-9371-4] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/05/2018] [Accepted: 08/23/2018] [Indexed: 12/21/2022]
Abstract
The physiology and behaviors related to energy balance are monitored by the nervous and humoral systems. Because of the difficulty in treating diabetes and obesity, elucidating the energy balance mechanism and identifying critical targets for treatment are important research goals. Therefore, the purpose of this article is to describe energy regulation by the central nervous system (CNS) and peripheral humoral pathway. Homeostasis and rewarding are the basis of CNS regulation. Anorexigenic or orexigenic effects reflect the activities of the POMC/CART or NPY/AgRP neurons within the hypothalamus. Neurotransmitters have roles in food intake, and responsive brain nuclei have different functions related to food intake, glucose monitoring, reward processing. Peripheral gut- or adipose-derived hormones are the major source of peripheral humoral regulation systems. Nutrients or metabolites and gut microbiota affect metabolism via a discrete pathway. We also review the role of peripheral organs, the liver, adipose tissue, and skeletal muscle in peripheral regulation. We discuss these topics and how the body regulates metabolism.
Collapse
Affiliation(s)
- Song Wen
- Department of Endocrinology, Shanghai Pudong Hospital, Fudan University, Shanghai, 201399, China
| | - Chaoxun Wang
- Department of Endocrinology, Shanghai Pudong Hospital, Fudan University, Shanghai, 201399, China
| | - Min Gong
- Department of Endocrinology, Shanghai Pudong Hospital, Fudan University, Shanghai, 201399, China
| | - Ligang Zhou
- Department of Endocrinology, Shanghai Pudong Hospital, Fudan University, Shanghai, 201399, China.
| |
Collapse
|
23
|
Alvarsson A, Stanley SA. Remote control of glucose-sensing neurons to analyze glucose metabolism. Am J Physiol Endocrinol Metab 2018; 315:E327-E339. [PMID: 29812985 PMCID: PMC6171010 DOI: 10.1152/ajpendo.00469.2017] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/22/2017] [Revised: 04/25/2018] [Accepted: 05/23/2018] [Indexed: 12/13/2022]
Abstract
The central nervous system relies on a continual supply of glucose, and must be able to detect glucose levels and regulate peripheral organ functions to ensure that its energy requirements are met. Specialized glucose-sensing neurons, first described half a century ago, use glucose as a signal and modulate their firing rates as glucose levels change. Glucose-excited neurons are activated by increasing glucose concentrations, while glucose-inhibited neurons increase their firing rate as glucose concentrations fall and decrease their firing rate as glucose concentrations rise. Glucose-sensing neurons are present in multiple brain regions and are highly expressed in hypothalamic regions, where they are involved in functions related to glucose homeostasis. However, the roles of glucose-sensing neurons in healthy and disease states remain poorly understood. Technologies that can rapidly and reversibly activate or inhibit defined neural populations provide invaluable tools to investigate how specific neural populations regulate metabolism and other physiological roles. Optogenetics has high temporal and spatial resolutions, requires implants for neural stimulation, and is suitable for modulating local neural populations. Chemogenetics, which requires injection of a synthetic ligand, can target both local and widespread populations. Radio- and magnetogenetics offer rapid neural activation in localized or widespread neural populations without the need for implants or injections. These tools will allow us to better understand glucose-sensing neurons and their metabolism-regulating circuits.
Collapse
Affiliation(s)
- Alexandra Alvarsson
- Diabetes, Obesity, and Metabolism Institute, Icahn School of Medicine at Mount Sinai , New York, New York
| | - Sarah A Stanley
- Diabetes, Obesity, and Metabolism Institute, Icahn School of Medicine at Mount Sinai , New York, New York
- Department of Neuroscience, Icahn School of Medicine at Mount Sinai , New York, New York
| |
Collapse
|
24
|
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: 2.5] [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
|
25
|
Koekkoek LL, Mul JD, la Fleur SE. Glucose-Sensing in the Reward System. Front Neurosci 2017; 11:716. [PMID: 29311793 PMCID: PMC5742113 DOI: 10.3389/fnins.2017.00716] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2017] [Accepted: 12/07/2017] [Indexed: 01/14/2023] Open
Abstract
Glucose-sensing neurons are neurons that alter their activity in response to changes in extracellular glucose. These neurons, which are an important mechanism the brain uses to monitor changes in glycaemia, are present in the hypothalamus, where they have been thoroughly investigated. Recently, glucose-sensing neurons have also been identified in brain nuclei which are part of the reward system. However, little is known about the molecular mechanisms by which they function, and their role in the reward system. We therefore aim to provide an overview of molecular mechanisms that have been studied in the hypothalamic glucose-sensing neurons, and investigate which of these transporters, enzymes and channels are present in the reward system. Furthermore, we speculate about the role of glucose-sensing neurons in the reward system.
Collapse
Affiliation(s)
- Laura L Koekkoek
- Department of Endocrinology and Metabolism, Academic Medical Center, University of Amsterdam, Amsterdam, Netherlands.,Laboratory of Endocrinology, Department of Clinical Chemistry, Academic Medical Center, University of Amsterdam, Amsterdam, Netherlands.,Metabolism and Reward Group, Netherlands Institute for Neuroscience, Institute of the Royal Netherlands Academy of Arts and Sciences (KNAW), Amsterdam, Netherlands
| | - Joram D Mul
- Department of Endocrinology and Metabolism, Academic Medical Center, University of Amsterdam, Amsterdam, Netherlands.,Laboratory of Endocrinology, Department of Clinical Chemistry, Academic Medical Center, University of Amsterdam, Amsterdam, Netherlands.,Metabolism and Reward Group, Netherlands Institute for Neuroscience, Institute of the Royal Netherlands Academy of Arts and Sciences (KNAW), Amsterdam, Netherlands
| | - Susanne E la Fleur
- Department of Endocrinology and Metabolism, Academic Medical Center, University of Amsterdam, Amsterdam, Netherlands.,Laboratory of Endocrinology, Department of Clinical Chemistry, Academic Medical Center, University of Amsterdam, Amsterdam, Netherlands.,Metabolism and Reward Group, Netherlands Institute for Neuroscience, Institute of the Royal Netherlands Academy of Arts and Sciences (KNAW), Amsterdam, Netherlands
| |
Collapse
|
26
|
McNeilly AD, Gallagher JR, Huang JTJ, Ashford MLJ, McCrimmon RJ. High-Intensity Exercise as a Dishabituating Stimulus Restores Counterregulatory Responses in Recurrently Hypoglycemic Rodents. Diabetes 2017; 66:1696-1702. [PMID: 28270522 DOI: 10.2337/db16-1533] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/12/2016] [Accepted: 02/21/2017] [Indexed: 11/13/2022]
Abstract
Hypoglycemia is a major adverse effect of insulin therapy for people with type 1 diabetes (T1D). Profound defects in the normal counterregulatory response to hypoglycemia explain the frequency of hypoglycemia occurrence in T1D. Defective counterregulation results to a large extent from prior exposure to hypoglycemia per se, leading to a condition called impaired awareness of hypoglycemia (IAH), the cause of which is unknown. In the current study, we investigate the hypothesis that IAH develops through a special type of adaptive memory referred to as habituation. To test this hypothesis, we used a novel intense stimulus (high-intensity exercise) to demonstrate two classic features of a habituated response, namely dishabituation and response recovery. We demonstrate that after recurrent hypoglycemia the introduction of a novel dishabituating stimulus (a single burst of high-intensity exercise) in male Sprague-Dawley rats restores the defective hypoglycemia counterregulatory response. In addition, the rats showed an enhanced response to the novel stimulus (response recovery). We make the further observation using proteomic analysis of hypothalamic extracts that high-intensity exercise in recurrently hypoglycemic rats increases levels of a number of proteins linked with brain-derived neurotrophic factor signaling. These findings may lead to novel therapeutic approaches for individuals with T1D and IAH.
Collapse
Affiliation(s)
- Alison D McNeilly
- Division of Molecular and Clinical Medicine, University of Dundee, Dundee, U.K
| | | | - Jeffrey T-J Huang
- Division of Molecular and Clinical Medicine, University of Dundee, Dundee, U.K
| | - Michael L J Ashford
- Division of Molecular and Clinical Medicine, University of Dundee, Dundee, U.K
| | - Rory J McCrimmon
- Division of Molecular and Clinical Medicine, University of Dundee, Dundee, U.K.
| |
Collapse
|
27
|
Grommen SVH, Scott MK, Darras VM, De Groef B. Spatial and temporal expression profiles of urocortin 3 mRNA in the brain of the chicken (Gallus gallus). J Comp Neurol 2017; 525:2583-2591. [PMID: 28395119 DOI: 10.1002/cne.24223] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2017] [Revised: 03/20/2017] [Accepted: 04/03/2017] [Indexed: 01/15/2023]
Abstract
Urocortin 3 (UCN3) is a neuropeptide believed to regulate stress-coping responses by binding to type 2 corticotropin-releasing hormone receptors. Here, we report the cloning and brain distribution of UCN3 mRNA in a sauropsid-the chicken, Gallus gallus. Mature chicken UCN3 is predicted to be a 40-amino acid peptide showing high sequence similarity to human (93%), mouse (93%), and Xenopus (88%) UCN3. During the last third of embryonic development, UCN3 mRNA levels changed differentially in the various brain parts. In all brain parts, UCN3 mRNA levels tended to increase toward hatching, except for caudal brainstem, where a gradual decrease was observed during the last week of embryonic development. In cerebellum, a rapid increase in gene expression occurred between embryonic days 17 and 19. Using in situ hybridization, UCN3 mRNA was found to be expressed predominantly in the hypothalamus, pons, and medulla of posthatch chick brains, but not in some areas that are among the main expression sites in rodents, such as the brain areas where in mammals the median preoptic nucleus and the medial amygdala are located. This suggests that the roles of UCN3 in chicken, and perhaps sauropsids in general, are not all identical to those in rodents.
Collapse
Affiliation(s)
- Sylvia V H Grommen
- Department of Physiology, Anatomy and Microbiology, La Trobe University, Bundoora, Victoria, 3086, Australia
| | - Melissa K Scott
- Department of Physiology, Anatomy and Microbiology, La Trobe University, Bundoora, Victoria, 3086, Australia
| | - Veerle M Darras
- Laboratory of Comparative Endocrinology, Department of Biology, KU Leuven, B-3000, Leuven, Belgium
| | - Bert De Groef
- Department of Physiology, Anatomy and Microbiology, La Trobe University, Bundoora, Victoria, 3086, Australia
| |
Collapse
|
28
|
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: 4.0] [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
|
29
|
Otlivanchik O, Sanders NM, Dunn-Meynell A, Levin BE. Orexin signaling is necessary for hypoglycemia-induced prevention of conditioned place preference. Am J Physiol Regul Integr Comp Physiol 2015; 310:R66-73. [PMID: 26511522 DOI: 10.1152/ajpregu.00066.2015] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2015] [Accepted: 10/19/2015] [Indexed: 01/24/2023]
Abstract
While the neural control of glucoregulatory responses to insulin-induced hypoglycemia is beginning to be elucidated, brain sites responsible for behavioral responses to hypoglycemia are relatively poorly understood. To help elucidate central control mechanisms associated with hypoglycemia unawareness, we first evaluated the effect of recurrent hypoglycemia on a simple behavioral measure, the robust feeding response to hypoglycemia, in rats. First, food intake was significantly, and similarly, increased above baseline saline-induced intake (1.1 ± 0.2 g; n = 8) in rats experiencing a first (4.4 ± 0.3; n = 8) or third daily episode of recurrent insulin-induced hypoglycemia (IIH, 3.7 ± 0.3 g; n = 9; P < 0.05). Because food intake was not impaired as a result of prior IIH, we next developed an alternative animal model of hypoglycemia-induced behavioral arousal using a conditioned place preference (CPP) model. We found that hypoglycemia severely blunted previously acquired CPP in rats and that recurrent hypoglycemia prevented this blunting. Pretreatment with a brain penetrant, selective orexin receptor-1 antagonist, SB-334867A, blocked hypoglycemia-induced blunting of CPP. Recurrently hypoglycemic rats also showed decreased preproorexin expression in the perifornical hypothalamus (50%) but not in the adjacent lateral hypothalamus. Pretreatment with sertraline, previously shown to prevent hypoglycemia-associated glucoregulatory failure, did not prevent blunting of hypoglycemia-induced CPP prevention by recurrent hypoglycemia. This work describes the first behavioral model of hypoglycemia unawareness and suggests a role for orexin neurons in mediating behavioral responses to hypoglycemia.
Collapse
Affiliation(s)
- Oleg Otlivanchik
- Graduate School of Biomedical Sciences, Rutgers University, Newark, New Jersey; Department of Neurology, New Jersey Medical School, Rutgers University, Newark, New Jersey
| | | | - Ambrose Dunn-Meynell
- Graduate School of Biomedical Sciences, Rutgers University, Newark, New Jersey; Department of Neurology, New Jersey Medical School, Rutgers University, Newark, New Jersey; Neurology Service, Veterans Affairs Medical Center, East Orange, New Jersey; and
| | - Barry E Levin
- Department of Neurology, New Jersey Medical School, Rutgers University, Newark, New Jersey; Neurology Service, Veterans Affairs Medical Center, East Orange, New Jersey; and
| |
Collapse
|
30
|
Fan X, Chan O, Ding Y, Zhu W, Mastaitis J, Sherwin R. Reduction in SGLT1 mRNA Expression in the Ventromedial Hypothalamus Improves the Counterregulatory Responses to Hypoglycemia in Recurrently Hypoglycemic and Diabetic Rats. Diabetes 2015; 64:3564-72. [PMID: 26130763 PMCID: PMC4587643 DOI: 10.2337/db15-0022] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/06/2015] [Accepted: 06/23/2015] [Indexed: 12/11/2022]
Abstract
The objective of this study was to determine whether the sodium-glucose transporter SGLT1 in the ventromedial hypothalamus (VMH) plays a role in glucose sensing and in regulating the counterregulatory response to hypoglycemia, and if so, whether knockdown of in the VMH can improve counterregulatory responses to hypoglycemia in diabetic rats or rats exposed to recurrent bouts of hypoglycemia (RH). Normal Sprague-Dawley rats as well as RH or streptozotocin (STZ)-diabetic rats received bilateral VMH microinjections of an adenoassociated viral vector containing either the SGLT1 short hairpin RNA (shRNA) or a scrambled RNA sequence. Subsequently, these rats underwent a hypoglycemic clamp to assess hormone responses. In a subgroup of rats, glucose kinetics was determined using tritiated glucose. The shRNA reduced VMH SGLT1 expression by 53% in nondiabetic rats, and this augmented glucagon and epinephrine responses and hepatic glucose production during hypoglycemia. Similarly, SGLT1 knockdown improved the glucagon and epinephrine responses in RH rats and restored the impaired epinephrine response to hypoglycemia in STZ-diabetic animals. These findings suggest that SGLT1 in the VMH plays a significant role in the detection and activation of counterregulatory responses to hypoglycemia. Inhibition of SGLT1 may offer a potential therapeutic target to diminish the risk of hypoglycemia in diabetes.
Collapse
Affiliation(s)
- Xiaoning Fan
- Department of Internal Medicine, Section of Endocrinology, Yale School of Medicine, New Haven, CT
| | - Owen Chan
- Department of Internal Medicine, Section of Endocrinology, Yale School of Medicine, New Haven, CT
| | - Yuyan Ding
- Department of Internal Medicine, Section of Endocrinology, Yale School of Medicine, New Haven, CT
| | - Wanling Zhu
- Department of Internal Medicine, Section of Endocrinology, Yale School of Medicine, New Haven, CT
| | - Jason Mastaitis
- Department of Internal Medicine, Section of Endocrinology, Yale School of Medicine, New Haven, CT
| | - Robert Sherwin
- Department of Internal Medicine, Section of Endocrinology, Yale School of Medicine, New Haven, CT
| |
Collapse
|
31
|
Resveratrol ameliorates mitochondrial dysfunction but increases the risk of hypoglycemia following hemorrhagic shock. J Trauma Acute Care Surg 2015; 77:926-33. [PMID: 25248062 DOI: 10.1097/ta.0000000000000358] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
BACKGROUND Hemorrhagic shock (HS) may contribute to organ failure, by profoundly altering mitochondrial function. Resveratrol (RSV), a naturally occurring polyphenol, has been shown to promote mitochondrial function and regulate glucose homeostasis in diabetes. We hypothesized that RSV during resuscitation would ameliorate HS-induced mitochondrial dysfunction and improve hyperglycemia following acute blood loss. METHODS With the use a decompensated HS model, male Long-Evans rats (n = 6 per group) were resuscitated with lactated Ringer's solution with or without RSV (30 mg/kg) and were killed before hemorrhage (sham), at severe shock, following resuscitation, and 18 hours after resuscitation. At each time point, the liver and kidney mitochondria were isolated to assess individual respiratory complexes (CI, CII, and CIV) and the production of reactive oxygen species (ROS). Blood samples were assayed for glucose, insulin, corticosterone, total glucagon-like peptide (GLP-1), glucagon, and serum cytokine levels. The Homeostatic Model Assessment-Insulin Resistance index was used to quantify insulin resistance. RESULTS RSV supplementation following HS significantly improved mitochondrial function and decreased mitochondrial ROS production in both liver and kidney. RSV-treated animals had significantly lower blood glucose levels following resuscitation when compared with sham animals (116.0 ± 20.2 mg/dL vs. 227.7 ± 8.3 mg/dL, p < 0.05) or those resuscitated with lactated Ringer's solution (116.0 ± 20.2 mg/dL vs. 359.0 ± 79.5 mg/dL, p < 0.05). RSV supplementation was associated with significantly decreased plasma insulin levels (1.0 ± 0.4 ng/mL vs. 6.5 ± 3.7 ng/mL, p < 0.05), increased total GLP-1 levels (385.8 ± 56.6 ng/mL vs. 187.3 ± 11.1 ng/mL, p < 0.05), and a lower natural Log Homeostatic Model Assessment-Insulin Resistance index (1.30 ± 0.42 vs. 4.18 ± 0.68, p < 0.05) but had minimal effect on plasma corticosterone, glucagon, or cytokine levels. CONCLUSION Resuscitation with RSV restores mitochondrial function and decreases insulin resistance but may be associated with increased hypoglycemia. The observed antiglycemic effects of RSV may be mediated by decreased mitochondrial ROS and increased GLP-1 secretion.
Collapse
|
32
|
Bouret S, Levin BE, Ozanne SE. Gene-environment interactions controlling energy and glucose homeostasis and the developmental origins of obesity. Physiol Rev 2015; 95:47-82. [PMID: 25540138 PMCID: PMC4281588 DOI: 10.1152/physrev.00007.2014] [Citation(s) in RCA: 97] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Obesity and type 2 diabetes mellitus (T2DM) often occur together and affect a growing number of individuals in both the developed and developing worlds. Both are associated with a number of other serious illnesses that lead to increased rates of mortality. There is likely a polygenic mode of inheritance underlying both disorders, but it has become increasingly clear that the pre- and postnatal environments play critical roles in pushing predisposed individuals over the edge into a disease state. This review focuses on the many genetic and environmental variables that interact to cause predisposed individuals to become obese and diabetic. The brain and its interactions with the external and internal environment are a major focus given the prominent role these interactions play in the regulation of energy and glucose homeostasis in health and disease.
Collapse
Affiliation(s)
- Sebastien Bouret
- The Saban Research Institute, Neuroscience Program, Childrens Hospital Los Angeles, University of Southern California, Los Angeles, California; Inserm U837, Jean-Pierre Aubert Research Center, University Lille 2, Lille, France; Neurology Service, Veterans Administration Medical Center, East Orange, New Jersey; Department of Neurology and Neurosciences, Rutgers, New Jersey Medical School, Newark, New Jersey; and University of Cambridge Institute of Metabolic Science and MRC Metabolic Diseases Unit, Cambridge, United Kingdom
| | - Barry E Levin
- The Saban Research Institute, Neuroscience Program, Childrens Hospital Los Angeles, University of Southern California, Los Angeles, California; Inserm U837, Jean-Pierre Aubert Research Center, University Lille 2, Lille, France; Neurology Service, Veterans Administration Medical Center, East Orange, New Jersey; Department of Neurology and Neurosciences, Rutgers, New Jersey Medical School, Newark, New Jersey; and University of Cambridge Institute of Metabolic Science and MRC Metabolic Diseases Unit, Cambridge, United Kingdom
| | - Susan E Ozanne
- The Saban Research Institute, Neuroscience Program, Childrens Hospital Los Angeles, University of Southern California, Los Angeles, California; Inserm U837, Jean-Pierre Aubert Research Center, University Lille 2, Lille, France; Neurology Service, Veterans Administration Medical Center, East Orange, New Jersey; Department of Neurology and Neurosciences, Rutgers, New Jersey Medical School, Newark, New Jersey; and University of Cambridge Institute of Metabolic Science and MRC Metabolic Diseases Unit, Cambridge, United Kingdom
| |
Collapse
|
33
|
Routh VH, Hao L, Santiago AM, Sheng Z, Zhou C. Hypothalamic glucose sensing: making ends meet. Front Syst Neurosci 2014; 8:236. [PMID: 25540613 PMCID: PMC4261699 DOI: 10.3389/fnsys.2014.00236] [Citation(s) in RCA: 122] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2014] [Accepted: 11/26/2014] [Indexed: 01/12/2023] Open
Abstract
The neuroendocrine system governs essential survival and homeostatic functions. For example, growth is needed for development, thermoregulation maintains optimal core temperature in a changing environment, and reproduction ensures species survival. Stress and immune responses enable an organism to overcome external and internal threats while the circadian system regulates arousal and sleep such that vegetative and active functions do not overlap. All of these functions require a significant portion of the body's energy. As the integrator of the neuroendocrine system, the hypothalamus carefully assesses the energy status of the body in order to appropriately partition resources to provide for each system without compromising the others. While doing so the hypothalamus must ensure that adequate glucose levels are preserved for brain function since glucose is the primary fuel of the brain. To this end, the hypothalamus contains specialized glucose sensing neurons which are scattered throughout the nuclei controlling distinct neuroendocrine functions. We hypothesize that these neurons play a key role in enabling the hypothalamus to partition energy to meet these peripheral survival needs without endangering the brain's glucose supply. This review will first describe the varied mechanisms underlying glucose sensing in neurons within discrete hypothalamic nuclei. We will then evaluate the way in which peripheral energy status regulates glucose sensitivity. For example, during energy deficit such as fasting specific hypothalamic glucose sensing neurons become sensitized to decreased glucose. This increases the gain of the information relay when glucose availability is a greater concern for the brain. Finally, changes in glucose sensitivity under pathological conditions (e.g., recurrent insulin-hypoglycemia, diabetes) will be addressed. The overall goal of this review is to place glucose sensing neurons within the context of hypothalamic control of neuroendocrine function.
Collapse
Affiliation(s)
- Vanessa H Routh
- Department of Pharmacology and Physiology, New Jersey Medical School, Rutgers University Newark, NJ, USA
| | - Lihong Hao
- Department of Pharmacology and Physiology, New Jersey Medical School, Rutgers University Newark, NJ, USA ; Department of Pharmacology and Physiology and Graduate School of the Biomedical Sciences, New Jersey Medical School, Rutgers University Newark, NJ, USA
| | - Ammy M Santiago
- Department of Pharmacology and Physiology, New Jersey Medical School, Rutgers University Newark, NJ, USA ; Department of Pharmacology and Physiology and Graduate School of the Biomedical Sciences, New Jersey Medical School, Rutgers University Newark, NJ, USA
| | - Zhenyu Sheng
- Department of Pharmacology and Physiology, New Jersey Medical School, Rutgers University Newark, NJ, USA
| | - Chunxue Zhou
- Department of Pharmacology and Physiology, New Jersey Medical School, Rutgers University Newark, NJ, USA ; Department of Pharmacology and Physiology and Graduate School of the Biomedical Sciences, New Jersey Medical School, Rutgers University Newark, NJ, USA
| |
Collapse
|
34
|
Ogunnowo-Bada EO, Heeley N, Brochard L, Evans ML. Brain glucose sensing, glucokinase and neural control of metabolism and islet function. Diabetes Obes Metab 2014; 16 Suppl 1:26-32. [PMID: 25200293 PMCID: PMC4405079 DOI: 10.1111/dom.12334] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/29/2014] [Accepted: 06/05/2014] [Indexed: 11/30/2022]
Abstract
It is increasingly apparent that the brain plays a central role in metabolic homeostasis, including the maintenance of blood glucose. This is achieved by various efferent pathways from the brain to periphery, which help control hepatic glucose flux and perhaps insulin-stimulated insulin secretion. Also, critically important for the brain given its dependence on a constant supply of glucose as a fuel--emergency counter-regulatory responses are triggered by the brain if blood glucose starts to fall. To exert these control functions, the brain needs to detect rapidly and accurately changes in blood glucose. In this review, we summarize some of the mechanisms postulated to play a role in this and examine the potential role of the low-affinity hexokinase, glucokinase, in the brain as a key part of some of this sensing. We also discuss how these processes may become altered in diabetes and related metabolic diseases.
Collapse
Affiliation(s)
- E O Ogunnowo-Bada
- Wellcome Trust-MRC Institute of Metabolic Science, IMS Metabolic Research Laboratories, University of CambridgeCambridge, UK
| | - N Heeley
- Wellcome Trust-MRC Institute of Metabolic Science, IMS Metabolic Research Laboratories, University of CambridgeCambridge, UK
| | - L Brochard
- Wellcome Trust-MRC Institute of Metabolic Science, IMS Metabolic Research Laboratories, University of CambridgeCambridge, UK
| | - M L Evans
- Wellcome Trust-MRC Institute of Metabolic Science, IMS Metabolic Research Laboratories, University of CambridgeCambridge, UK
- Correspondence to: Mark Evans, Wellcome Trust-MRC Institute of Metabolic Science, University of Cambridge, IMS Metabolic Research Laboratories, Box 289 Addenbrookes Hospital, Hills Road, Cambridge CB2 0QQ, UK. E-mail:
| |
Collapse
|
35
|
The role of central nervous system on hypoglycemia and the feasibility of the brain theory in traditional Chinese medicine on treatment of diabetes mellitus. JOURNAL OF INTEGRATIVE MEDICINE-JIM 2014; 12:1-6. [DOI: 10.1016/s2095-4964(14)60008-x] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
|
36
|
Resch JM, Maunze B, Gerhardt AK, Magnuson SK, Phillips KA, Choi S. Intrahypothalamic pituitary adenylate cyclase-activating polypeptide regulates energy balance via site-specific actions on feeding and metabolism. Am J Physiol Endocrinol Metab 2013; 305:E1452-63. [PMID: 24148346 PMCID: PMC3882380 DOI: 10.1152/ajpendo.00293.2013] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
Numerous studies have demonstrated that both the hypothalamic paraventricular nuclei (PVN) and ventromedial nuclei (VMN) regulate energy homeostasis through behavioral and metabolic mechanisms. Receptors for pituitary adenylate cyclase-activating polypeptide (PACAP) are abundantly expressed in these nuclei, suggesting PACAP may be critical for the regulation of feeding behavior and body weight. To characterize the unique behavioral and physiological responses attributed to select hypothalamic cell groups, PACAP was site-specifically injected into the PVN or VMN. Overall food intake was significantly reduced by PACAP at both sites; however, meal pattern analysis revealed that only injections into the PVN produced significant reductions in meal size, duration, and total time spent eating. PACAP-mediated hypophagia in both the PVN and VMN was abolished by PAC1R antagonism, whereas pretreatment with a VPACR antagonist had no effect. PACAP injections into the VMN produced unique changes in metabolic parameters, including significant increases in core body temperature and spontaneous locomotor activity that was PAC1R dependent whereas, PVN injections of PACAP had no effect. Finally, PACAP-containing afferents were identified using the neuronal tracer cholera toxin subunit B (CTB) injected unilaterally into the PVN or VMN. CTB signal from PVN injections was colocalized with PACAP mRNA in the medial anterior bed nucleus of the stria terminalis, VMN, and lateral parabrachial nucleus (LPB), whereas CTB signal from VMN injections was highly colocalized with PACAP mRNA in the medial amygdala and LPB. These brain regions are known to influence energy homeostasis perhaps, in part, through PACAP projections to the PVN and VMN.
Collapse
Affiliation(s)
- Jon M Resch
- Department of Biomedical Sciences, Marquette University, Milwaukee, Wisconsin
| | | | | | | | | | | |
Collapse
|
37
|
Chan O, Sherwin R. Influence of VMH fuel sensing on hypoglycemic responses. Trends Endocrinol Metab 2013; 24:616-24. [PMID: 24063974 PMCID: PMC3909530 DOI: 10.1016/j.tem.2013.08.005] [Citation(s) in RCA: 61] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/28/2013] [Revised: 08/20/2013] [Accepted: 08/27/2013] [Indexed: 12/12/2022]
Abstract
Hypoglycemia produces complex neural and hormonal responses that restore glucose levels to normal. Glucose, metabolic substrates and their transporters, neuropeptides and neurotransmitters alter the firing rate of glucose-sensing neurons in the ventromedial hypothalamus (VMH); these monitor energy status and regulate the release of neurotransmitters that instigate a suitable counter-regulatory response. Under normal physiological conditions, these mechanisms maintain blood glucose concentrations within narrow margins. However, antecedent hypoglycemia and diabetes can lead to adaptations within the brain that impair counter-regulatory responses. Clearly, the mechanisms employed to detect and regulate the response to hypoglycemia, and the pathophysiology of defective counter-regulation in diabetes, are complex and need to be elucidated to permit the development of therapies that prevent or reduce the risk of hypoglycemia.
Collapse
Affiliation(s)
- Owen Chan
- Yale University School of Medicine, Department of Internal Medicine - Section of Endocrinology, New Haven, CT, 06520 U.S.A
| | - Robert Sherwin
- Yale University School of Medicine, Department of Internal Medicine - Section of Endocrinology, New Haven, CT, 06520 U.S.A
| |
Collapse
|
38
|
The effect of high fat diet and saturated fatty acids on insulin signaling in the amygdala and hypothalamus of rats. Brain Res 2013; 1537:191-200. [DOI: 10.1016/j.brainres.2013.09.025] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2013] [Revised: 09/13/2013] [Accepted: 09/19/2013] [Indexed: 12/22/2022]
|
39
|
Stanley S, Domingos AI, Kelly L, Garfield A, Damanpour S, Heisler L, Friedman J. Profiling of Glucose-Sensing Neurons Reveals that GHRH Neurons Are Activated by Hypoglycemia. Cell Metab 2013; 18:596-607. [PMID: 24093682 DOI: 10.1016/j.cmet.2013.09.002] [Citation(s) in RCA: 79] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/13/2013] [Revised: 07/29/2013] [Accepted: 08/22/2013] [Indexed: 12/17/2022]
Abstract
Comprehensive transcriptional profiling of glucose-sensing neurons is challenging because of low expression levels of glucokinase (Gck) and other key proteins that transduce a glucose signal. To overcome this, we generated and validated transgenic mice with a neuronal/endocrine-specific Gck promoter driving cre expression and mated them to mice with cre-dependent expression of an EGFP-tagged ribosomal protein construct (EEF1A1-LSL.EGFPL10) that can be used to map and profile cells. We found significant Gck expression in hypothalamic and limbic regions in cells that are activated following administration of glucose or 2-deoxyglucose. Transcriptional profiling from Gck-cre/EEF1A1-LSL.EGFPL10 mice enriched known and previously unknown glucose-sensing populations including neurons expressing growth hormone releasing hormone (GHRH). Electrophysiological recordings show that hypoglycemia activates GHRH neurons, suggesting a mechanistic link between hypoglycemia and growth hormone release. These studies provide a means for mapping glucose-sensitive neurons and for generating transcriptional profiles from other cell types expressing cre in a cell-specific manner.
Collapse
Affiliation(s)
- Sarah Stanley
- Laboratory of Molecular Genetics, Rockefeller University, New York, NY 10065, USA
| | | | | | | | | | | | | |
Collapse
|
40
|
Diepenbroek C, Serlie MJ, Fliers E, Kalsbeek A, la Fleur SE. Brain areas and pathways in the regulation of glucose metabolism. Biofactors 2013; 39:505-13. [PMID: 23913677 DOI: 10.1002/biof.1123] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/05/2013] [Accepted: 05/28/2013] [Indexed: 11/11/2022]
Abstract
Glucose is the most important source of fuel for the brain and its concentration must be kept within strict boundaries to ensure the organism's optimal fitness. To maintain glucose homeostasis, an optimal balance between glucose uptake and glucose output is required. Besides managing acute changes in plasma glucose concentrations, the brain controls a daily rhythm in glucose concentrations. The various nuclei within the hypothalamus that are involved in the control of both these processes are well known. However, novel studies indicate an additional role for brain areas that are originally appreciated in other processes than glucose metabolism. Therefore, besides the classic hypothalamic pathways, we will review cortico-limbic brain areas and their role in glucose metabolism.
Collapse
Affiliation(s)
- Charlene Diepenbroek
- Department of Endocrinology and Metabolism, Academic Medical Center, University of Amsterdam, Amsterdam, the Netherlands
| | | | | | | | | |
Collapse
|
41
|
Peters A, Kubera B, Hubold C, Langemann D. The corpulent phenotype-how the brain maximizes survival in stressful environments. Front Neurosci 2013; 7:47. [PMID: 23565074 PMCID: PMC3613700 DOI: 10.3389/fnins.2013.00047] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2013] [Accepted: 03/13/2013] [Indexed: 11/14/2022] Open
Abstract
The reactivity of the stress system may change during the life course. In many—but not all—humans the stress reactivity decreases, once the individual is chronically exposed to a stressful and unsafe environment (e.g., poverty, work with high demands, unhappy martial relationship). Such an adaptation is referred to as habituation. Stress habituation allows alleviating the burden of chronic stress, particularly cardiovascular morbidity and mortality. Interestingly, two recent experiments demonstrated low stress reactivity during a mental or psychosocial challenge in subjects with a high body mass. In this focused review we attempt to integrate these experimental findings in a larger context. Are these data compatible with data sets showing a prolonged life expectancy in corpulent people? From the perspective of neuroenergetics, we here raise the question whether “obesity” is unhealthy at all. Is the corpulent phenotype possibly the result of “adaptive phenotypic plasticity” allowing optimized survival in stressful environments?
Collapse
Affiliation(s)
- Achim Peters
- Clinical Research Group: Brain Metabolism, Neuroenergetics, Obesity and Diabetes, University of Luebeck Luebeck, Germany
| | | | | | | |
Collapse
|
42
|
Park-York M, Boghossian S, Oh H, York DA. PKCθ expression in the amygdala regulates insulin signaling, food intake and body weight. Obesity (Silver Spring) 2013; 21:755-64. [PMID: 23712979 DOI: 10.1002/oby.20278] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/02/2012] [Accepted: 05/18/2012] [Indexed: 11/10/2022]
Abstract
OBJECTIVE To investigate the signaling mechanisms that might underlie the loss of anorectic response to insulin injections into the central nucleus of the amygdala (CeA) within 3 days of feeding a high fat diet. DESIGN AND METHODS Protein samples from amygdala and hypothalamus of rats fed high or low fat diets were subjected to a phosphorylation screening assay. The effects of dietary fat intake on the expression and activation of protein kinase C theta (PKCθ) in brain regions was studied. Finally, lentiviral vectors were used to overexpress rat PKCθ unilaterally or bilaterally into the CeA of rats and the effects on food intake, body weight and insulin stimulation of Akt phosphorylation were studied. RESULTS The level of pMARCKS (Myristoylated alanine-rich C-kinase substrate), a major substrate of PKCθ, was increased 116% in amygdala of high fat diet fed rats but reduced in the hypothalamus. High fat diets increased the level of PKCθ in a region specific manner in the brain and this PKCθ was activated by membrane association. Overexpressing rat PKCθ either unilaterally or bilaterally into the CeA inhibited insulin stimulation of Akt signaling and blocked the anorectic response to insulin injected into the amygdala. Bilaterally injected PKCθ rats gained more weight and body fat and had increased food intake when fed a high fat diet compared to the control rats that received a lentiviral-Green Fluorescent Protein construct. CONCLUSION The data suggest that insulin may have a physiological role within the amygdala to regulate energy balance.
Collapse
|
43
|
PKCθ over expression in the central nucleus of the amygdala or hypothalamus has differential effects on energy balance and peripheral glucose homeostasis. Brain Res 2013; 1498:85-94. [DOI: 10.1016/j.brainres.2012.12.042] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2012] [Revised: 12/19/2012] [Accepted: 12/29/2012] [Indexed: 01/08/2023]
|
44
|
Abstract
Islet hormones, especially insulin and glucagon, are important for glucose homeostasis. Insulin is a necessity for life, and disturbed insulin release results in disordered blood glucose regulation. Although isolated islets are fully capable of detecting changes in their local environment (particularly glucose fluctuations) and altering hormone release appropriately, experimentally manipulating pancreatic innervation alters islet hormone release in the whole animal. This article describes how brain may play a role in influencing and directing secretion of insulin and glucagon as a key part of the integrated physiology of blood glucose homeostasis.
Collapse
Affiliation(s)
- Mayowa A Osundiji
- Department of Medicine, Institute of Metabolic Science, University of Cambridge, Addenbrooke's Hospital, Cambridge, UK
| | | |
Collapse
|
45
|
Routh VH, Donovan CM, Ritter S. 2. Hypoglycemia Detection. TRANSLATIONAL ENDOCRINOLOGY & METABOLISM 2012; 3:47-87. [PMID: 24910721 PMCID: PMC4045627 DOI: 10.1210/team.9781936704200.ch2] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
|
46
|
Hurst P, Garfield AS, Marrow C, Heisler LK, Evans ML. Recurrent hypoglycemia is associated with loss of activation in rat brain cingulate cortex. Endocrinology 2012; 153:1908-14. [PMID: 22396449 PMCID: PMC3328129 DOI: 10.1210/en.2011-1827] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/29/2011] [Accepted: 01/26/2012] [Indexed: 12/30/2022]
Abstract
A subset of people with diabetes fail to mount defensive counterregulatory responses (CRR) to hypoglycemia. Although the mechanisms by which this occurs remain unclear, recurrent exposure to hypoglycemia may be an important etiological factor. We hypothesized that loss of CRR to recurrent exposure to hypoglycemia represents a type of stress desensitization, in which limbic brain circuitry involved in modulating stress responses might be implicated. Here, we compared activation of limbic brain regions associated with stress desensitization during acute hypoglycemia (AH) and recurrent hypoglycemia (RH). Healthy Sprague Dawley rats were exposed to either acute or recurrent 3-d hypoglycemia. We also examined whether changes in neuronal activation were caused directly by the CRR itself by infusing epinephrine, glucagon, and corticosterone without hypoglycemia. AH increased neuronal activity as quantified by c-fos immunoreactivity (FOS-IR) in the cingulate cortex and associated ectorhinal and perirhinal cortices but not in an adjacent control area (primary somatosensory cortex). FOS-IR was not observed after hormone infusion, suggesting that AH-associated activation was caused by hypoglycemia rather than by CRR. Importantly, AH FOS-IR activation was significantly blunted in rats exposed to RH. In conclusion, analogous with other models of stress habituation, activation in the cingulate cortex and associated brain areas is lost with exposure to RH. Our data support the hypothesis that limbic brain areas may be associated with the loss of CRR to RH in diabetes.
Collapse
Affiliation(s)
- Paul Hurst
- University of Cambridge Metabolic Research Laboratories/Department of Medicine/National Institute for Health Research Cambridge Biomedical Research Centre, Cambridge CB2 0QQ, United Kingdom
| | | | | | | | | |
Collapse
|
47
|
Kubera B, Hubold C, Zug S, Wischnath H, Wilhelm I, Hallschmid M, Entringer S, Langemann D, Peters A. The brain's supply and demand in obesity. FRONTIERS IN NEUROENERGETICS 2012; 4:4. [PMID: 22408618 PMCID: PMC3297086 DOI: 10.3389/fnene.2012.00004] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/05/2011] [Accepted: 02/24/2012] [Indexed: 11/13/2022]
Abstract
During psychosocial stress, the brain demands extra energy from the body to satisfy its increased needs. For that purpose it uses a mechanism referred to as "cerebral insulin suppression" (CIS). Specifically, activation of the stress system suppresses insulin secretion from pancreatic beta-cells, and in this way energy-particularly glucose-is allocated to the brain rather than the periphery. It is unknown, however, how the brain of obese humans organizes its supply and demand during psychosocial stress. To answer this question, we examined 20 obese and 20 normal weight men in two sessions (Trier Social Stress Test and non-stress control condition followed by either a rich buffet or a meager salad). Blood samples were continuously taken and subjects rated their vigilance and mood by standard questionnaires. First, we found a low reactive stress system in obesity. While obese subjects showed a marked hormonal response to the psychosocial challenge, the cortisol response to the subsequent meal was absent. Whereas the brains of normal weight subjects demanded for extra energy from the body by using CIS, CIS was not detectable in obese subjects. Our findings suggest that the absence of CIS in obese subjects is due to the absence of their meal-related cortisol peak. Second, normal weight men were high reactive during psychosocial stress in changing their vigilance, thereby increasing their cerebral energy need, whereas obese men were low reactive in this respect. Third, normal weight subjects preferred carbohydrates after stress to supply their brain, while obese men preferred fat and protein instead. We conclude that the brain of obese people organizes its need, supply, and demand in a low reactive manner.
Collapse
Affiliation(s)
- Britta Kubera
- Medical Clinic 1, University of LübeckLübeck, Germany
| | | | - Sophia Zug
- Medical Clinic 1, University of LübeckLübeck, Germany
| | | | - Ines Wilhelm
- Department of Neuroendocrinology, University of LübeckLübeck, Germany
| | | | - Sonja Entringer
- Department of Psychiatry and Human Behavior, University of California, IrvineCA, USA
| | - Dirk Langemann
- Institute of Technical Computational Mathematics, University of BraunschweigBraunschweig, Germany
| | - Achim Peters
- Medical Clinic 1, University of LübeckLübeck, Germany
| |
Collapse
|
48
|
Beall C, Ashford ML, McCrimmon RJ. The physiology and pathophysiology of the neural control of the counterregulatory response. Am J Physiol Regul Integr Comp Physiol 2011; 302:R215-23. [PMID: 22071156 DOI: 10.1152/ajpregu.00531.2011] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
Despite significant technological and pharmacological advancements, insulin replacement therapy fails to adequately replicate β-cell function, and so glucose control in type 1 diabetes mellitus (T1D) is frequently erratic, leading to periods of hypoglycemia. Moreover, the counterregulatory response (CRR) to falling blood glucose is impaired in diabetes, leading to an increased risk of severe hypoglycemia. It is now clear that the brain plays a significant role in the development of defective glucose counterregulation and impaired hypoglycemia awareness in diabetes. In this review, the basic intracellular glucose-sensing mechanisms are discussed, as well as the neural networks that respond to and coordinate the body's response to a hypoglycemic challenge. Subsequently, we discuss how the body responds to repeated hypoglycemia and how these adaptations may explain, at least in part, the development of impaired glucose counterregulation in diabetes.
Collapse
Affiliation(s)
- Craig Beall
- Medical Research Institute, Division of Cardiovascular and Diabetes Medicine, Ninewells Hospital and Medical School, Univ. of Dundee, Dundee, DD1 9SY, United Kingdom
| | | | | |
Collapse
|
49
|
Ritter S, Li AJ, Wang Q, Dinh TT. Minireview: The value of looking backward: the essential role of the hindbrain in counterregulatory responses to glucose deficit. Endocrinology 2011; 152:4019-32. [PMID: 21878511 PMCID: PMC3444967 DOI: 10.1210/en.2010-1458] [Citation(s) in RCA: 62] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
This review focuses on evidence indicating a key role for the hindbrain in mobilizing behavioral, autonomic and endocrine counterregulatory responses to acute and profound glucose deficit, and identifies hindbrain norepinephrine (NE) and epinephrine (E) neurons as essential mediators of some of these responses. It has become clear that hindbrain NE/E neurons are functionally diverse. However, considerable progress has been made in identifying the particular NE/E neurons important for particular glucoregulatory responses. Although it is not yet known whether NE/E neurons are directly activated by glucose deficit, compelling evidence indicates that if they are not, the primary glucoreceptor cells must be located in the immediate vicinity these neurons. Hindbrain studies identifying cellular markers associated with glucose-sensing functions in other brain regions are discussed, as are studies examining the relationship of these markers to counterregulatory responses of NE/E neurons. Further investigations to identify glucose-sensing cells (neurons, ependymocytes, or glia) controlling counterregulatory responses are crucial, as are studies to determine the specific functions of glucose-sensing cells throughout the brain. Likewise, examination of the roles (if any) of hindbrain counterregulatory systems in managing glucose homeostasis under basal, nonglucoprivic conditions will also be important for a full understanding of energy homeostasis. Nevertheless, the accumulated evidence demonstrates that hindbrain glucose sensors and NE/E neurons are essential players in triggering counterregulatory responses to emergencies of glucose deficit.
Collapse
Affiliation(s)
- Sue Ritter
- Department of Veterinary and Comparative Anatomy, Pharmacology, College of Veterinary Medicine, Washington State University, Pullman, Washington 99164-6520, USA.
| | | | | | | |
Collapse
|
50
|
Abstract
We have reported that corticotropin-releasing factor (CRF) receptor subtypes, CRF1 and CRF2, are involved in stress-induced anorexia. To clarify in which brain regions the CRF receptor is involved in mediating stress-induced anorexia, we examined the effect of microinjecting CRF1-selective or CRF2-selective antagonist into the lateral septum or the bed nucleus of the stria terminalis (BNST), which are implicated in regulating stress response. The results demonstrated that injecting antisauvagine-30 into the lateral septum or the BNST significantly attenuated restraint-induced anorexia, whereas injecting antalarmin into these regions did not affect anorexia. These results suggest that the CRF2 receptor in the lateral septum and the BNST is involved in the stress-induced inhibitory mechanism of feeding behavior.
Collapse
|