1
|
Laule C, Rahmouni K. Leptin and Associated Neural Pathways Underlying Obesity-Induced Hypertension. Compr Physiol 2025; 15:e8. [PMID: 40293220 PMCID: PMC12038170 DOI: 10.1002/cph4.8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2024] [Revised: 10/24/2024] [Accepted: 11/05/2024] [Indexed: 04/30/2025]
Abstract
Obesity rates have surged to pandemic levels, placing tremendous burden on our society. This chronic and complex disease is related to the development of many life-threatening illnesses including cardiovascular diseases. Hypertension caused by obesity increases the risk for cardiovascular mortality and morbidity by promoting stroke, myocardial infarction, congestive heart failure, and end-stage renal disease. Overwhelming evidence supports neural origins for obesity-induced hypertension and pinpoints the adipose-derived hormone, leptin, and the sympathetic nervous system as major causal factors. Hyperleptinemia in obesity is associated with selective leptin resistance where leptin's renal sympathoexcitatory and pressor effects are preserved while the metabolic actions are impaired. Understanding the mechanisms driving this phenomenon is critical for developing effective therapeutics. This review describes the neural mechanisms of obesity-induced hypertension with a focus on the molecular and neuronal substrates of leptin action.
Collapse
Affiliation(s)
- Connor Laule
- Department of Neuroscience and Pharmacology, University of Iowa Carver College of Medicine, Iowa City, Iowa
| | - Kamal Rahmouni
- Department of Neuroscience and Pharmacology, University of Iowa Carver College of Medicine, Iowa City, Iowa
- Fraternal Order of Eagles Diabetes Research Center, University of Iowa Carver College of Medicine, Iowa City, Iowa
- Obesity Research and Education Initiative, University of Iowa Carver College of Medicine, Iowa City, Iowa
- Iowa Neuroscience Institute, University of Iowa Carver College of Medicine, Iowa City, Iowa
- Veterans Affairs Health Care System, Iowa City, Iowa
| |
Collapse
|
2
|
Engin AB, Engin A. Tryptophan Metabolism in Obesity: The Indoleamine 2,3-Dioxygenase-1 Activity and Therapeutic Options. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2024; 1460:629-655. [PMID: 39287867 DOI: 10.1007/978-3-031-63657-8_21] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/19/2024]
Abstract
Obesity activates both innate and adaptive immune responses in adipose tissue. Adipose tissue macrophages are functional antigen-presenting cells that promote the proliferation of interferon-gamma (IFN-γ)-producing cluster of differentiation (CD)4+ T cells in adipose tissue of obese subjects. The increased formation of neopterin and degradation of tryptophan may result in decreased T-cell responsiveness and lead to immunodeficiency. The activity of inducible indoleamine 2,3-dioxygenase-1 (IDO1) plays a major role in pro-inflammatory, IFN-γ-dominated settings. The expression of several kynurenine pathway enzyme genes is significantly increased in obesity. IDO1 in obesity shifts tryptophan metabolism from serotonin and melatonin synthesis to the formation of kynurenines and increases the ratio of kynurenine to tryptophan as well as with neopterin production. Reduction in serotonin (5-hydroxytryptamine; 5-HT) production provokes satiety dysregulation that leads to increased caloric uptake and obesity. According to the monoamine-deficiency hypothesis, a deficiency of cerebral serotonin is involved in neuropsychiatric symptomatology of depression, mania, and psychosis. Indeed, bipolar disorder (BD) and related cognitive deficits are accompanied by a higher prevalence of overweight and obesity. Furthermore, the accumulation of amyloid-β in Alzheimer's disease brains has several toxic effects as well as IDO induction. Hence, abdominal obesity is associated with vascular endothelial dysfunction. kynurenines and their ratios are prognostic parameters in coronary artery disease. Increased kynurenine/tryptophan ratio correlates with increased intima-media thickness and represents advanced atherosclerosis. However, after bariatric surgery, weight reduction does not lead to the normalization of IDO1 activity and atherosclerosis. IDO1 is involved in the mechanisms of immune tolerance and in the concept of tumor immuno-editing process in cancer development. Serum IDO1 activity is still used as a parameter in cancer development and growth. IDO-producing tumors show a high total IDO immunostaining score, and thus, using IDO inhibitors, such as Epacadostat, Navoximod, and L isomer of 1-methyl-tryptophan, seems an important modality for cancer treatment. There is an inverse correlation between serum folate concentration and body mass index, thus folate deficiency leads to hyperhomocysteinemia-induced oxidative stress. Immune checkpoint blockade targeting cytotoxic T-lymphocyte-associated protein-4 synergizes with imatinib, which is an inhibitor of mitochondrial folate-mediated one-carbon (1C) metabolism. Antitumor effects of imatinib are enhanced by increasing T-cell effector function in the presence of IDO inhibition. Combining IDO targeting with chemotherapy, radiotherapy and/or immunotherapy, may be an effective tool against a wide range of malignancies. However, there are some controversial results regarding the efficacy of IDO1 inhibitors in cancer treatment.
Collapse
Affiliation(s)
- Ayse Basak Engin
- Faculty of Pharmacy, Department of Toxicology, Gazi University, Hipodrom, Ankara, Turkey.
| | - Atilla Engin
- Faculty of Medicine, Department of General Surgery, Gazi University, Besevler, Ankara, Turkey
- Mustafa Kemal Mah. 2137. Sok. 8/14, 06520, Cankaya, Ankara, Turkey
| |
Collapse
|
3
|
Iosif CI, Bashir ZI, Apps R, Pickford J. Cerebellar Prediction and Feeding Behaviour. CEREBELLUM (LONDON, ENGLAND) 2023; 22:1002-1019. [PMID: 36121552 PMCID: PMC10485105 DOI: 10.1007/s12311-022-01476-3] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 09/01/2022] [Indexed: 06/15/2023]
Abstract
Given the importance of the cerebellum in controlling movements, it might be expected that its main role in eating would be the control of motor elements such as chewing and swallowing. Whilst such functions are clearly important, there is more to eating than these actions, and more to the cerebellum than motor control. This review will present evidence that the cerebellum contributes to homeostatic, motor, rewarding and affective aspects of food consumption.Prediction and feedback underlie many elements of eating, as food consumption is influenced by expectation. For example, circadian clocks cause hunger in anticipation of a meal, and food consumption causes feedback signals which induce satiety. Similarly, the sight and smell of food generate an expectation of what that food will taste like, and its actual taste will generate an internal reward value which will be compared to that expectation. Cerebellar learning is widely thought to involve feed-forward predictions to compare expected outcomes to sensory feedback. We therefore propose that the overarching role of the cerebellum in eating is to respond to prediction errors arising across the homeostatic, motor, cognitive, and affective domains.
Collapse
Affiliation(s)
- Cristiana I Iosif
- School of Physiology, Pharmacology and Neuroscience, University of Bristol, Biomedical Sciences Building, University Walk, Bristol, BS8 1TD, UK.
| | - Zafar I Bashir
- School of Physiology, Pharmacology and Neuroscience, University of Bristol, Biomedical Sciences Building, University Walk, Bristol, BS8 1TD, UK
| | - Richard Apps
- School of Physiology, Pharmacology and Neuroscience, University of Bristol, Biomedical Sciences Building, University Walk, Bristol, BS8 1TD, UK
| | - Jasmine Pickford
- School of Physiology, Pharmacology and Neuroscience, University of Bristol, Biomedical Sciences Building, University Walk, Bristol, BS8 1TD, UK
| |
Collapse
|
4
|
Harvey J. Leptin regulation of synaptic function at hippocampal TA-CA1 and SC-CA1 synapses. VITAMINS AND HORMONES 2022; 118:315-336. [PMID: 35180931 DOI: 10.1016/bs.vh.2021.12.002] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
Increasing evidence indicates that the metabolic hormone, leptin markedly influences the functioning of the hippocampus. In particular, exposure to leptin results in persistent changes in synaptic efficacy at both temporoammonic (TA) and Schaffer Collateral (SC) inputs to hippocampal CA1 neurons. The ability of leptin to regulate TA-CA1 and SC-CA1 synapses has important functional implications, as both synaptic connections play important roles in hippocampal-dependent learning and memory. Here we review the modulatory actions of the hormone leptin at these hippocampal CA1 synapses and explore the impact on learning and memory processes.
Collapse
Affiliation(s)
- Jenni Harvey
- Division of Systems Medicine, Ninewells Hospital and Medical School, University of Dundee, Dundee, United Kingdom.
| |
Collapse
|
5
|
Intracellular interplay between cholecystokinin and leptin signalling for satiety control in rats. Sci Rep 2020; 10:12000. [PMID: 32686770 PMCID: PMC7371863 DOI: 10.1038/s41598-020-69035-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2019] [Accepted: 06/29/2020] [Indexed: 11/09/2022] Open
Abstract
Cholecystokinin (CCK) and leptin are satiety-controlling peptides, yet their interactive roles remain unclear. Here, we addressed this issue using in vitro and in vivo models. In rat C6 glioma cells, leptin pre-treatment enhanced Ca2+ mobilization by a CCK agonist (CCK-8s). This leptin action was reduced by Janus kinase inhibitor (AG490) or PI3-kinase inhibitor (LY294002). Meanwhile, leptin stimulation alone failed to mobilize Ca2+ even in cells overexpressing leptin receptors (C6-ObRb). Leptin increased nuclear immunoreactivity against phosphorylated STAT3 (pSTAT3) whereas CCK-8s reduced leptin-induced nuclear pSTAT3 accumulation in these cells. In the rat ventromedial hypothalamus (VMH), leptin-induced action potential firing was enhanced, whereas nuclear pSTAT3 was reduced by co-stimulation with CCK-8s. To further analyse in vivo signalling interplay, a CCK-1 antagonist (lorglumide) was intraperitoneally injected in rats following 1-h restricted feeding. Food access was increased 3-h after lorglumide injection. At this timepoint, nuclear pSTAT3 was increased whereas c-Fos was decreased in the VMH. Taken together, these results suggest that leptin and CCK receptors may both contribute to short-term satiety, and leptin could positively modulate CCK signalling. Notably, nuclear pSTAT3 levels in this experimental paradigm were negatively correlated with satiety levels, contrary to the generally described transcriptional regulation for long-term satiety via leptin receptors.
Collapse
|
6
|
Moravveji A, Sayyah M, Shamsnia E, Vakili Z. Comparing the prolonged effect of interval versus continuous aerobic exercise on blood inflammatory marker of Visfatin level and body mass index of sedentary overweigh/fat female college students. AIMS Public Health 2019; 6:568-576. [PMID: 31909076 PMCID: PMC6940584 DOI: 10.3934/publichealth.2019.4.568] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2019] [Accepted: 12/03/2019] [Indexed: 01/02/2023] Open
Abstract
History and objectives Over weightiness and obesity are usually defined as inflammatory conditions. High ratio of body mass index and Visfatin level recently discovered as markers involved in inflammatory process of obesity. Aerobic exercise is one of the safe interventions to decrease such condition. The purpose of this research was to compare the effect of interval versus continuous aerobic exercise on Visfatin and BMI of sedentary overweight female college students. Materials and Methods Thirty-six healthy sedentary overweight female college students with BMI over 25 or more were randomly assigned into three groups including continuous, interval aerobic exercise and control conditions for eight weeks, three sessions per week. Serum visfatin level was assessed before and after the exercise protocol. The exercise protocol included running a distance of 1200 meters continuously or with rest intervals at 60 to 75 percent of reserved heart rate in the first week that gradually increased by 400 meters on every subsequent week. Results Our study indicated that both aerobic exercise conditions significantly decrease the serum level of visfatin (P = 0.000, P = 0.025, respectively). Both exercise groups also showed a decrease in BMI compared to the control group (P = 0.006, P = 0.004). Conclusion Aerobic exercise has a beneficiary effect on both serum visfatin level and BMI variables involved in inflammation process of obesity regardless of being performed with rest interval or continuously.
Collapse
Affiliation(s)
- Alireza Moravveji
- Department of Community and Preventive Medicine, Social Determinant of Health (SDH) Research Center, Kashan University of Medical Sciences, Kashan, Iran
| | - Mansour Sayyah
- Departement of Exercise Sciences, Kashan University of Medical Sciences, Kashan, Iran
| | | | - Zarichehr Vakili
- Department of Pathology, Kashan University of Medical Sciences, Kashan, Iran
| |
Collapse
|
7
|
Suarez AN, Noble EE, Kanoski SE. Regulation of Memory Function by Feeding-Relevant Biological Systems: Following the Breadcrumbs to the Hippocampus. Front Mol Neurosci 2019; 12:101. [PMID: 31057368 PMCID: PMC6482164 DOI: 10.3389/fnmol.2019.00101] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2019] [Accepted: 04/03/2019] [Indexed: 12/15/2022] Open
Abstract
The hippocampus (HPC) controls fundamental learning and memory processes, including memory for visuospatial navigation (spatial memory) and flexible memory for facts and autobiographical events (declarative memory). Emerging evidence reveals that hippocampal-dependent memory function is regulated by various peripheral biological systems that are traditionally known for their roles in appetite and body weight regulation. Here, we argue that these effects are consistent with a framework that it is evolutionarily advantageous to encode and recall critical features surrounding feeding behavior, including the spatial location of a food source, social factors, post-absorptive processing, and other episodic elements of a meal. We review evidence that gut-to-brain communication from the vagus nerve and from feeding-relevant endocrine systems, including ghrelin, insulin, leptin, and glucagon-like peptide-1 (GLP-1), promote hippocampal-dependent spatial and declarative memory via neurotrophic and neurogenic mechanisms. The collective literature reviewed herein supports a model in which various stages of feeding behavior and hippocampal-dependent memory function are closely linked.
Collapse
Affiliation(s)
| | | | - Scott E. Kanoski
- Human and Evolutionary Biology Section, Department of Biological Sciences, University of Southern California, Los Angeles, CA, United States
| |
Collapse
|
8
|
Abstract
PURPOSE OF REVIEW The major health issue of being overweight or obese relates to the development of hypertension, insulin resistance and diabetic complications. One of the major underlying factors influencing the elevated blood pressure in obesity is increased activity of the sympathetic nerves to particular organs such as the kidney. RECENT FINDINGS There is now convincing evidence from animal studies that major signals such as leptin and insulin have a sympathoexcitatory action in the hypothalamus to cause hypertension. Recent studies suggest that this may involve 'neural plasticity' within hypothalamic signalling driven by central actions of leptin mediated via activation of melanocortin receptor signalling and activation of brain neurotrophic factors. This review describes the evidence to support the contribution of the SNS to obesity related hypertension and the major metabolic and adipokine signals.
Collapse
|
9
|
Feng EC, Jiang L. [Effect of leptin on long-term spatial memory of rats with white matter damage in developing brain]. ZHONGGUO DANG DAI ER KE ZA ZHI = CHINESE JOURNAL OF CONTEMPORARY PEDIATRICS 2017; 19:1267-1271. [PMID: 29237528 PMCID: PMC7389803 DOI: 10.7499/j.issn.1008-8830.2017.12.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 06/07/2017] [Accepted: 11/02/2017] [Indexed: 06/07/2023]
Abstract
OBJECTIVE To investigate the neuroprotective effect of leptin by observing its effect on spatial memory of rats with white matter damage in developing brain. METHODS A total of 80 neonatal rats were randomly divided into 3 groups: sham-operation (n=27), model (n=27) and leptin intervention (n=27). The rats in the model and leptin intervention groups were used to prepare a model of white matter damage in developing brain, and the rats in the leptin intervention group were given leptin (100 μg/kg) diluted with normal saline immediately after modelling for 4 consecutive days. The survival rate of the rats was observed and the change in body weight was monitored. When the rats reached the age of 21 days, the Morris water maze test was used to evaluate spatial memory. RESULTS There was no significant difference in the survival rate of rats between the three groups (P>0.05). Within 10 days after birth, the leptin intervention group had similar body weight as the sham-operation group and significantly lower body weight than the model group (P<0.05); more than 10 days after birth, the leptin intervention group had rapid growth with higher body weight than the model and sham-operation groups (P>0.05). The results of place navigation showed that from the second day of experiment, there was a significant difference in the latency period between the three groups (P<0.05); from the fourth day of experiment, the leptin intervention group had a similar latency period as the sham-operation and a significantly shorter latency period than the model group (P<0.05). The results of space search experiment showed that compared with the sham-operation group, the model group had a significant reduction in the number of platform crossings and a significantly longer latency period (P<0.05); compared with the model group, the leptin intervention group had a significantly increased number of platform crossings and a significantly shortened latency period (P<0.05), while there was no significant difference between the leptin intervention and sham-operation groups. CONCLUSIONS Leptin can alleviate spatial memory impairment of rats with white matter damage in developing brain. It thus exerts a neuroprotective effect, and is worthy of further research.
Collapse
Affiliation(s)
- Er-Cui Feng
- School of Biological Science & Medical Engineering, Southeast University, Nanjing 210018, China.
| | | |
Collapse
|
10
|
Presynaptic Regulation of Leptin in a Defined Lateral Hypothalamus-Ventral Tegmental Area Neurocircuitry Depends on Energy State. J Neurosci 2017; 37:11854-11866. [PMID: 29089444 DOI: 10.1523/jneurosci.1942-17.2017] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2017] [Revised: 09/30/2017] [Accepted: 10/16/2017] [Indexed: 12/16/2022] Open
Abstract
Synaptic transmission controls brain activity and behaviors, including food intake. Leptin, an adipocyte-derived hormone, acts on neurons located in the lateral hypothalamic area (LHA) to maintain energy homeostasis and regulate food intake behavior. The specific synaptic mechanisms, cell types, and neural projections mediating this effect remain unclear. In male mice, using pathway-specific retrograde tracing, whole-cell patch-clamp recordings and post hoc cell type identification, we found that leptin reduces excitatory synaptic strength onto both melanin-concentrating hormone- and orexin-expressing neurons projecting from the LHA to the ventral tegmental area (VTA), which may affect dopamine signaling and motivation for feeding. A presynaptic mechanism mediated by distinct intracellular signaling mechanisms may account for this regulation by leptin. The regulatory effects of leptin depend on intact leptin receptor signaling. Interestingly, the synaptic regulatory function of leptin in the LHA-to-VTA neuronal pathway is highly sensitive to energy states: both energy deficiency (acute fasting) and excessive energy storage (high-fat diet-induced obesity) blunt the effect of leptin. These data revealed that leptin may regulate synaptic transmission in the LHA-to-VTA neurocircuitry in an inverted "U-shape" fashion dependent on plasma glucose levels and related to metabolic states.SIGNIFICANCE STATEMENT The lateral hypothalamic area (LHA) to ventral tegmental area (VTA) projection is an important neural pathway involved in balancing whole-body energy states and reward. We found that the excitatory synaptic inputs to both orexin- and melanin-concentrating hormone expressing LHA neurons projecting to the VTA were suppressed by leptin, a peptide hormone derived from adipocytes that signals peripheral energy status to the brain. Interestingly, energy states seem to affect how leptin regulates synaptic transmission since both the depletion of energy induced by acute food deprivation and excessive storage of energy by high-fat diet feeding dampen the suppressive effect of leptin on synaptic transmission. Together, these data show that leptin regulates synaptic transmission and might be important for maintaining energy homeostasis.
Collapse
|
11
|
McGregor G, Harvey J. Food for thought: Leptin regulation of hippocampal function and its role in Alzheimer's disease. Neuropharmacology 2017; 136:298-306. [PMID: 28987937 DOI: 10.1016/j.neuropharm.2017.09.038] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2017] [Revised: 09/27/2017] [Accepted: 09/30/2017] [Indexed: 01/08/2023]
Abstract
Accumulating evidence indicates that diet and body weight are important factors associated with Alzheimer's disease (AD), with a significant increase in AD risk linked to mid-life obesity, and weight loss frequently occurring in the early stages of AD. This has fuelled interest in the hormone leptin, as it is an important hypothalamic regulator of food intake and body weight, but leptin also markedly influences the functioning of the hippocampus; a key brain region that degenerates in AD. Increasing evidence indicates that leptin has cognitive enhancing properties as it facilitates the cellular events that underlie hippocampal-dependent learning and memory. However, significant reductions in leptin's capacity to regulate hippocampal synaptic function occurs with age and dysfunctions in the leptin system are associated with an increased risk of AD. Moreover, leptin is a potential novel target in AD as leptin treatment has beneficial effects in various models of AD. Here we summarise recent advances in leptin neurobiology with particular focus on regulation of hippocampal synaptic function by leptin and the implications of this for neurodegenerative disorders like AD. This article is part of the Special Issue entitled 'Metabolic Impairment as Risk Factors for Neurodegenerative Disorders.'
Collapse
Affiliation(s)
- Gemma McGregor
- Division of Neuroscience, Ninewells Hospital and Medical School, University of Dundee, Dundee DD1 9SY, United Kingdom
| | - Jenni Harvey
- Division of Neuroscience, Ninewells Hospital and Medical School, University of Dundee, Dundee DD1 9SY, United Kingdom.
| |
Collapse
|
12
|
A Hypothalamic Leptin-Glutamate Interaction in the Regulation of Sympathetic Nerve Activity. Neural Plast 2017; 2017:2361675. [PMID: 28845307 PMCID: PMC5560058 DOI: 10.1155/2017/2361675] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2017] [Revised: 05/24/2017] [Accepted: 05/30/2017] [Indexed: 11/17/2022] Open
Abstract
Accumulated evidence indicates that obesity-induced type 2 diabetes (T2D) is associated with enhanced sympathetic activation. The present study was conducted to investigate the role for leptin-glutamate signaling within the hypothalamus in regulating sympathetic nerve activity. In anesthetized rats, microinjections of leptin (5 ng ~ 100 ng) into the arcuate nucleus (ARCN) and paraventricular nucleus (PVN) induced increases in renal sympathetic nerve activity (RSNA), blood pressure (BP), and heart rate (HR). Prior microinjections of NMDA receptor antagonist AP5 (16 pmol) into the ARCN or PVN reduced leptin-induced increases in RSNA, BP, and HR in both ARCN and PVN. Knockdown of a leptin receptor with siRNA inhibited NMDA-induced increases in RSNA, BP, and HR in the ARCN but not in the PVN. Confocal calcium imaging in the neuronal NG108 and astrocytic C6 cells demonstrated that preincubation with leptin induced an increase in intracellular calcium green fluorescence when the cells were challenged with glutamate. In high-fat diet and low-dose streptozotocin-induced T2D rats, we found that leptin receptor and NMDA NR1 receptor expressions in the ARCN and PVN were significantly increased. In conclusion, these studies provide evidence that within the hypothalamic nuclei, leptin-glutamate signaling regulates the sympathetic activation. This may contribute to the sympathoexcitation commonly observed in obesity-related T2D.
Collapse
|
13
|
Wu Y, Fortin DA, Cochrane VA, Chen PC, Shyng SL. NMDA receptors mediate leptin signaling and regulate potassium channel trafficking in pancreatic β-cells. J Biol Chem 2017; 292:15512-15524. [PMID: 28768770 DOI: 10.1074/jbc.m117.802249] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2017] [Revised: 08/01/2017] [Indexed: 12/25/2022] Open
Abstract
NMDA receptors (NMDARs) are Ca2+-permeant, ligand-gated ion channels activated by the excitatory neurotransmitter glutamate and have well-characterized roles in the nervous system. The expression and function of NMDARs in pancreatic β-cells, by contrast, are poorly understood. Here, we report a novel function of NMDARs in β-cells. Using a combination of biochemistry, electrophysiology, and imaging techniques, we now show that NMDARs have a key role in mediating the effect of leptin to modulate β-cell electrical activity by promoting AMP-activated protein kinase (AMPK)-dependent trafficking of KATP and Kv2.1 channels to the plasma membrane. Blocking NMDAR activity inhibited the ability of leptin to activate AMPK, induce KATP and Kv2.1 channel trafficking, and promote membrane hyperpolarization. Conversely, activation of NMDARs mimicked the effect of leptin, causing Ca2+ influx, AMPK activation, and increased trafficking of KATP and Kv2.1 channels to the plasma membrane, and triggered membrane hyperpolarization. Moreover, leptin potentiated NMDAR currents and triggered NMDAR-dependent Ca2+ influx. Importantly, NMDAR-mediated signaling was observed in rat insulinoma 832/13 cells and in human β-cells, indicating that this pathway is conserved across species. The ability of NMDARs to regulate potassium channel surface expression and thus, β-cell excitability provides mechanistic insight into the recently reported insulinotropic effects of NMDAR antagonists and therefore highlights the therapeutic potential of these drugs in managing type 2 diabetes.
Collapse
Affiliation(s)
- Yi Wu
- From the Department of Biochemistry and Molecular Biology, Oregon Health and Science University, Portland, Oregon 97239 and
| | - Dale A Fortin
- From the Department of Biochemistry and Molecular Biology, Oregon Health and Science University, Portland, Oregon 97239 and
| | - Veronica A Cochrane
- From the Department of Biochemistry and Molecular Biology, Oregon Health and Science University, Portland, Oregon 97239 and
| | - Pei-Chun Chen
- the Department of Physiology, College of Medicine, National Cheng Kung University, Tainan 701, Taiwan
| | - Show-Ling Shyng
- From the Department of Biochemistry and Molecular Biology, Oregon Health and Science University, Portland, Oregon 97239 and
| |
Collapse
|
14
|
Shaban H, O’Connor R, Ovsepian SV, Dinan TG, Cryan JF, Schellekens H. Electrophysiological approaches to unravel the neurobiological basis of appetite and satiety: use of the multielectrode array as a screening strategy. Drug Discov Today 2017; 22:31-42. [DOI: 10.1016/j.drudis.2016.09.003] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2016] [Revised: 08/03/2016] [Accepted: 09/06/2016] [Indexed: 01/10/2023]
|
15
|
Engin AB, Engin A. The Interactions Between Kynurenine, Folate, Methionine and Pteridine Pathways in Obesity. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2017; 960:511-527. [PMID: 28585214 DOI: 10.1007/978-3-319-48382-5_22] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Obesity activates both innate and adaptive immune responses in adipose tissue. Elevated levels of eosinophils with depression of monocyte and neutrophil indicate the deficiencies in the immune system of morbidly obese individuals. Actually, adipose tissue macrophages are functional antigen-presenting cells that promote the proliferation of interferon-gamma (IFN-gamma)-producing CD4+ T cells in adipose tissue of obese subjects. Eventually, diet-induced obesity is associated with the loss of tissue homeostasis and development of type 1 inflammatory responses in visceral adipose tissue. Activity of inducible indoleamine 2,3-dioxygenase-1 (IDO-1) plays a major role under pro-inflammatory, IFN-gamma dominated settings. One of the two rate-limiting enzymes which can metabolize tryptophan to kynurenine is IDO-1. Tumor necrosis factor-alpha (TNF-alpha) correlates with IDO-1 in adipose compartments. Actually, IDO-1-mediated tryptophan catabolism due to chronic immune activation is the cause of reduced tryptophan plasma levels and be considered as the driving force for food intake in morbidly obese patients. Thus, decrease in plasma tryptophan levels and subsequent reduction in serotonin (5-HT) production provokes satiety dysregulation that leads to increased caloric uptake and obesity. However, after bariatric surgery, weight reduction does not lead to normalization of IDO-1 activity. Furthermore, there is a connection between arginine and tryptophan metabolic pathways in the generation of reactive nitrogen intermediates. Hence, abdominal obesity is associated with vascular endothelial dysfunction and reduced nitric oxide (NO) availability. IFN-gamma-induced activation of the inducible nitric oxide synthase (iNOS) and dissociation of endothelial adenosine monophosphate activated protein kinase (AMPK)- phosphoinositide 3-kinase (PI3K)-protein kinase B (Akt)- endothelial NO synthase (eNOS) pathway enhances oxidative stress production secondary to high-fat diet. Thus, reduced endothelial NO availability correlates with the increase in plasma non-esterified fatty acids and triglycerides levels. Additionally, in obese patients, folate-deficiency leads to hyperhomocysteinemia. Folic acid confers protection against hyperhomocysteinemia-induced oxidative stress.
Collapse
Affiliation(s)
- Ayse Basak Engin
- Faculty of Pharmacy, Department of Toxicology, Gazi University, Hipodrom, Ankara, Turkey.
| | - Atilla Engin
- Faculty of Medicine, Department of General Surgery, Gazi University, Besevler, Ankara, Turkey.
- , Mustafa Kemal Mah. 2137. Sok. 8/14, 06520, Cankaya, Ankara, Turkey.
| |
Collapse
|
16
|
Desensitization of leptin receptors is coincident with the upregulation of dopamine-related genes in the prefrontal cortex of adolescent mice. Neuroreport 2016; 27:516-21. [DOI: 10.1097/wnr.0000000000000574] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
|
17
|
Gavello D, Carbone E, Carabelli V. Leptin-mediated ion channel regulation: PI3K pathways, physiological role, and therapeutic potential. Channels (Austin) 2016; 10:282-96. [PMID: 27018500 DOI: 10.1080/19336950.2016.1164373] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
Leptin is produced by adipose tissue and identified as a "satiety signal," informing the brain when the body has consumed enough food. Specific areas of the hypothalamus express leptin receptors (LEPRs) and are the primary site of leptin action for body weight regulation. In response to leptin, appetite is suppressed and energy expenditure allowed. Beside this hypothalamic action, leptin targets other brain areas in addition to neuroendocrine cells. LEPRs are expressed also in the hippocampus, neocortex, cerebellum, substantia nigra, pancreatic β-cells, and chromaffin cells of the adrenal gland. It is intriguing how leptin is able to activate different ionic conductances, thus affecting excitability, synaptic plasticity and neurotransmitter release, depending on the target cell. Most of the intracellular pathways activated by leptin and directed to ion channels involve PI3K, which in turn phosphorylates different downstream substrates, although parallel pathways involve AMPK and MAPK. In this review we will describe the effects of leptin on BK, KATP, KV, CaV, TRPC, NMDAR and AMPAR channels and clarify the landscape of pathways involved. Given the ability of leptin to influence neuronal excitability and synaptic plasticity by modulating ion channels activity, we also provide a short overview of the growing potentiality of leptin as therapeutic agent for treating neurological disorders.
Collapse
Affiliation(s)
- Daniela Gavello
- a Department of Drug Science , Lab of Cellular Physiology and Molecular Neuroscience, NIS Center of Excellence, University of Torino , Torino , Italy
| | - Emilio Carbone
- a Department of Drug Science , Lab of Cellular Physiology and Molecular Neuroscience, NIS Center of Excellence, University of Torino , Torino , Italy
| | - Valentina Carabelli
- a Department of Drug Science , Lab of Cellular Physiology and Molecular Neuroscience, NIS Center of Excellence, University of Torino , Torino , Italy
| |
Collapse
|
18
|
McGregor G, Malekizadeh Y, Harvey J. Minireview: Food for thought: regulation of synaptic function by metabolic hormones. Mol Endocrinol 2016; 29:3-13. [PMID: 25470238 DOI: 10.1210/me.2014-1328] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
The peripheral actions of the metabolic hormones, leptin and insulin, are well documented. However, the functions of these hormones are not restricted to the periphery because evidence is growing that both leptin and insulin can readily cross the blood-brain barrier and have widespread central actions. The hippocampus in particular expresses high levels of both insulin and leptin receptors as well as key components of their associated signaling cascades. Moreover, recent studies indicate that both hormones are potential cognitive enhancers. Indeed, it has been demonstrated that both leptin and insulin markedly influence key cellular events that underlie hippocampal learning and memory including activity-dependent synaptic plasticity and the trafficking of glutamate receptors to and away from hippocampal synapses. The hippocampal formation is also a prime site for the neurodegenerative processes that occur during Alzheimer's disease, and impairments in either leptin or insulin function have been linked to central nervous system-driven diseases like Alzheimer's disease. Thus, the capacity of the metabolic hormones, leptin and insulin, to regulate hippocampal synaptic function has significant implications for normal brain function and also central nervous system-driven disease.
Collapse
Affiliation(s)
- Gemma McGregor
- Division of Neuroscience, Medical Research Institute, Ninewells Hospital and Medical School, University of Dundee, Dundee DD1 9SY, United Kingdom
| | | | | |
Collapse
|
19
|
Üner A, Gonçalves GH, Li W, Porceban M, Caron N, Schönke M, Delpire E, Sakimura K, Bjørbæk C. The role of GluN2A and GluN2B NMDA receptor subunits in AgRP and POMC neurons on body weight and glucose homeostasis. Mol Metab 2015; 4:678-91. [PMID: 26500840 PMCID: PMC4588453 DOI: 10.1016/j.molmet.2015.06.010] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/18/2015] [Revised: 06/15/2015] [Accepted: 06/19/2015] [Indexed: 01/03/2023] Open
Abstract
OBJECTIVE Hypothalamic agouti-related peptide (AgRP) and pro-opiomelanocortin (POMC) expressing neurons play critical roles in control of energy balance. Glutamatergic input via n-methyl-d-aspartate receptors (NMDARs) is pivotal for regulation of neuronal activity and is required in AgRP neurons for normal body weight homeostasis. NMDARs typically consist of the obligatory GluN1 subunit and different GluN2 subunits, the latter exerting crucial differential effects on channel activity and neuronal function. Currently, the role of specific GluN2 subunits in AgRP and POMC neurons on whole body energy and glucose balance is unknown. METHODS We used the cre-lox system to genetically delete GluN2A or GluN2B only from AgRP or POMC neurons in mice. Mice were then subjected to metabolic analyses and assessment of AgRP and POMC neuronal function through morphological studies. RESULTS We show that loss of GluN2B from AgRP neurons reduces body weight, fat mass, and food intake, whereas GluN2B in POMC neurons is not required for normal energy balance control. GluN2A subunits in either AgRP or POMC neurons are not required for regulation of body weight. Deletion of GluN2B reduces the number of AgRP neurons and decreases their dendritic length. In addition, loss of GluN2B in AgRP neurons of the morbidly obese and severely diabetic leptin-deficient Lep (ob/ob) mice does not affect body weight and food intake but, remarkably, leads to full correction of hyperglycemia. Lep (ob/ob) mice lacking GluN2B in AgRP neurons are also more sensitive to leptin's anti-obesity actions. CONCLUSIONS GluN2B-containing NMDA receptors in AgRP neurons play a critical role in central control of body weight homeostasis and blood glucose balance via mechanisms that likely involve regulation of AgRP neuronal survival and structure, and modulation of hypothalamic leptin action.
Collapse
Key Words
- AAC, area above the curve
- AMPARs, α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid receptors
- ANCOVA, analysis of covariance
- ANOVA, analysis of variance
- AUC, area under the curve
- AgRP
- AgRP, agouti-related peptide
- CNS, central nervous system
- DAB, 3,3′-diaminobenzidine
- DIO, diet-induced obesity
- DREADD, Designer Receptor Exclusively Activated by Dedigner Drugs
- EPSCs, excitatory post-synaptic synaptic currents
- GABA, gamma-aminobutyric acid
- GTT, glucose tolerance test
- GluN2B
- Glycemia
- HFD, high-fat diet
- HSD, honestly significant difference
- ITT, insulin tolerance test
- KO, knockout
- LTD, long-term depression
- LTP, long-term potentiation
- Lepob/ob mice, obese leptin-deficient mice
- Leptin
- Metabolism
- NMDAR
- NMDARs, N-methyl-d-aspartate receptors
- PBS, phosphate-buffered saline
- POMC, pro-opiomelanocortin
- PVN, paraventricular nucleus
- RT, room temperature
- hrGFP, humanized renilla GFP
Collapse
Affiliation(s)
- Aykut Üner
- Department of Medicine, Division of Endocrinology and Metabolism, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA 02215, USA
| | - Gabriel H.M. Gonçalves
- Department of Medicine, Division of Endocrinology and Metabolism, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA 02215, USA
| | - Wenjing Li
- Department of Medicine, Division of Endocrinology and Metabolism, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA 02215, USA
| | - Matheus Porceban
- Department of Medicine, Division of Endocrinology and Metabolism, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA 02215, USA
| | - Nicole Caron
- Department of Medicine, Division of Endocrinology and Metabolism, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA 02215, USA
| | - Milena Schönke
- Department of Medicine, Division of Endocrinology and Metabolism, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA 02215, USA
| | - Eric Delpire
- Department of Anesthesiology, Vanderbilt University Medical School, Nashville, TN 37232, USA
| | - Kenji Sakimura
- Department of Cellular Neurobiology, Brain Research Institute, Niigata University, Niigata 951-8585, Japan
| | - Christian Bjørbæk
- Department of Medicine, Division of Endocrinology and Metabolism, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA 02215, USA
| |
Collapse
|
20
|
Chowen JA, Argente J. Leptin and the brain. Horm Mol Biol Clin Investig 2015; 7:351-60. [PMID: 25961273 DOI: 10.1515/hmbci.2011.113] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2011] [Accepted: 07/13/2011] [Indexed: 02/03/2023]
Abstract
Leptin, which comes from the Greek root leptos meaning thin, has been the focus of intense investigation since its discovery in 1994. This hormone belongs to the cytokine family and is produced by adipocytes and circulates in proportion to fat mass, thus serving as a satiety signal and informing central metabolic control centers as to the status of peripheral energy stores. However, it participates in numerous other functions both peripherally and centrally, as indicated by the wide distribution of its various receptor isoforms. Leptin is involved in brain development, most notably in development of hypothalamic centers that control metabolism, but also in other brain areas. It acts as a nutritional cue to indicate adequacy of energy stores for pubertal development and reproductive capacity. The effects of this hormone on behavior and cognition are less well studied, but it clearly is involved in specific aspects of these physiological phenomena. As obesity is a major health problem in many areas of the world, the search for pharmacological treatments to decrease appetite and increase energy expenditure is intense. Understanding the mechanisms of actions of all physiological effects of this hormone is of great interest in the pursuit of such treatment.
Collapse
|
21
|
Güven AS, Icagasioglu FD, Duksal F, Sancakdar E, Alaygut D, Uysal EB, Akkar I, Oflaz MB, Kaya A. Serum adiponectin, leptin, and interleukin 6 levels as adipocytokines in children with febrile seizures: The role of adipose tissue in febrile seizures. Hum Exp Toxicol 2014; 34:878-83. [PMID: 25504688 DOI: 10.1177/0960327114561662] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Proinflammatory and anti-inflammatory cytokines have an important role in the pathogenesis of febrile seizures (FS). Adipocytokines like interleukin 6 (IL-6), leptin, and adiponectin released from adipose tissue play a role in inflammation. This study aimed to assess the probable role of adipose tissue in children with FS. We measured serum IL-6, leptin, and adiponectin levels and evaluated clinical and laboratory findings in children with FS (n = 32) and compared the results with the values of children of the same age with febrile illness without seizures (febrile control, FC; n = 26) and healthy control group (HC; n = 29). The serum levels of white blood cells, C-reactive protein, IL-6, leptin, and adiponectin were found to be significantly higher, while serum hemoglobin (Hb) levels were found to be significantly lower in FS and FC groups than in the HC group (p < 0.001). When we compared the FS with the FC group, the serum Hb levels were significantly lower in the FS group than those in the FC group (p = 0.001). There was no significant difference between the FS and FC group with regard to the serum levels of these adipocytokines (p > 0.05). Our data showed that elevated levels of these adipocytokines as acute phase reactants in FS and FC groups did not contribute to the development of FS.
Collapse
Affiliation(s)
- A S Güven
- Division of Pediatric Neurology, Department of Pediatrics, Faculty of Medicine, Cumhuriyet University, Sivas, Turkey
| | - F D Icagasioglu
- Division of Pediatric Neurology, Department of Pediatrics, Faculty of Medicine, Cumhuriyet University, Sivas, Turkey
| | - F Duksal
- Department of Pediatrics, Faculty of Medicine, Cumhuriyet University, Sivas, Turkey
| | - E Sancakdar
- Department of Biochemistry, Faculty of Medicine, Cumhuriyet University, Sivas, Turkey
| | - D Alaygut
- Department of Pediatrics, Faculty of Medicine, Cumhuriyet University, Sivas, Turkey
| | - E B Uysal
- Department of Microbiology, Faculty of Medicine, Cumhuriyet University, Sivas, Turkey
| | - I Akkar
- Department of Pediatrics, Sivas Government Hospital, Sivas, Turkey
| | - M B Oflaz
- Department of Pediatrics, Faculty of Medicine, Cumhuriyet University, Sivas, Turkey
| | - A Kaya
- Department of Pediatrics, Faculty of Medicine, Cumhuriyet University, Sivas, Turkey
| |
Collapse
|
22
|
Guimond D, Diabira D, Porcher C, Bader F, Ferrand N, Zhu M, Appleyard SM, Wayman GA, Gaiarsa JL. Leptin potentiates GABAergic synaptic transmission in the developing rodent hippocampus. Front Cell Neurosci 2014; 8:235. [PMID: 25177272 PMCID: PMC4133691 DOI: 10.3389/fncel.2014.00235] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2014] [Accepted: 07/26/2014] [Indexed: 12/17/2022] Open
Abstract
It is becoming increasingly clear that leptin is not only a hormone regulating energy homeostasis but also a neurotrophic factor impacting a number of brain regions, including the hippocampus. Although leptin promotes the development of GABAergic transmission in the hypothalamus, little is known about its action on the GABAergic system in the hippocampus. Here we show that leptin modulates GABAergic transmission onto developing CA3 pyramidal cells of newborn rats. Specifically, leptin induces a long-lasting potentiation (LLP-GABAA) of miniature GABAA receptor-mediated postsynaptic current (GABAA-PSC) frequency. Leptin also increases the amplitude of evoked GABAA-PSCs in a subset of neurons along with a decrease in the coefficient of variation and no change in the paired-pulse ratio, pointing to an increased recruitment of functional synapses. Adding pharmacological blockers to the recording pipette showed that the leptin-induced LLP-GABAA requires postsynaptic calcium released from internal stores, as well as postsynaptic MAPK/ERK kinases 1 and/or 2 (MEK1/2), phosphoinositide 3 kinase (PI3K) and calcium-calmodulin kinase kinase (CaMKK). Finally, study of CA3 pyramidal cells in leptin-deficient ob/ob mice revealed a reduction in the basal frequency of miniature GABAA-PSCs compared to wild type littermates. In addition, presynaptic GAD65 immunostaining was reduced in the CA3 stratum pyramidale of mutant animals, both results converging to suggest a decreased number of functional GABAergic synapses in ob/ob mice. Overall, these results show that leptin potentiates and promotes the development of GABAergic synaptic transmission in the developing hippocampus likely via an increase in the number of functional synapses, and provide insights into the intracellular pathways mediating this effect. This study further extends the scope of leptin's neurotrophic action to a key regulator of hippocampal development and function, namely GABAergic transmission.
Collapse
Affiliation(s)
- Damien Guimond
- Parc Scientifique de Luminy, Aix-Marseille Université Marseille, France ; Unité 901, Institut National de la Santé et de la Recherche Médicale Marseille, France ; Institut de Neurobiologie de la Méditerranée Marseille, France ; Program in Neuroscience, Department of Integrative Physiology and Neuroscience, Washington State University Pullman, WA, USA
| | - Diabe Diabira
- Parc Scientifique de Luminy, Aix-Marseille Université Marseille, France ; Unité 901, Institut National de la Santé et de la Recherche Médicale Marseille, France ; Institut de Neurobiologie de la Méditerranée Marseille, France
| | - Christophe Porcher
- Parc Scientifique de Luminy, Aix-Marseille Université Marseille, France ; Unité 901, Institut National de la Santé et de la Recherche Médicale Marseille, France ; Institut de Neurobiologie de la Méditerranée Marseille, France
| | - Francesca Bader
- Parc Scientifique de Luminy, Aix-Marseille Université Marseille, France ; Unité 901, Institut National de la Santé et de la Recherche Médicale Marseille, France ; Institut de Neurobiologie de la Méditerranée Marseille, France
| | - Nadine Ferrand
- Parc Scientifique de Luminy, Aix-Marseille Université Marseille, France ; Unité 901, Institut National de la Santé et de la Recherche Médicale Marseille, France ; Institut de Neurobiologie de la Méditerranée Marseille, France
| | - Mingyan Zhu
- Program in Neuroscience, Department of Integrative Physiology and Neuroscience, Washington State University Pullman, WA, USA
| | - Suzanne M Appleyard
- Program in Neuroscience, Department of Integrative Physiology and Neuroscience, Washington State University Pullman, WA, USA
| | - Gary A Wayman
- Program in Neuroscience, Department of Integrative Physiology and Neuroscience, Washington State University Pullman, WA, USA
| | - Jean-Luc Gaiarsa
- Parc Scientifique de Luminy, Aix-Marseille Université Marseille, France ; Unité 901, Institut National de la Santé et de la Recherche Médicale Marseille, France ; Institut de Neurobiologie de la Méditerranée Marseille, France
| |
Collapse
|
23
|
Mancini M, Soldovieri MV, Gessner G, Wissuwa B, Barrese V, Boscia F, Secondo A, Miceli F, Franco C, Ambrosino P, Canzoniero LMT, Bauer M, Hoshi T, Heinemann SH, Taglialatela M. Critical role of large-conductance calcium- and voltage-activated potassium channels in leptin-induced neuroprotection of N-methyl-d-aspartate-exposed cortical neurons. Pharmacol Res 2014; 87:80-6. [PMID: 24973659 DOI: 10.1016/j.phrs.2014.06.010] [Citation(s) in RCA: 14] [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: 05/21/2014] [Revised: 06/17/2014] [Accepted: 06/18/2014] [Indexed: 01/01/2023]
Abstract
In the present study, the neuroprotective effects of the adipokine leptin, and the molecular mechanism involved, have been studied in rat and mice cortical neurons exposed to N-methyl-d-aspartate (NMDA) in vitro. In rat cortical neurons, leptin elicited neuroprotective effects against NMDA-induced cell death, which were concentration-dependent (10-100 ng/ml) and largest when the adipokine was preincubated for 2h before the neurotoxic stimulus. In both rat and mouse cortical neurons, leptin-induced neuroprotection was fully antagonized by paxilline (Pax, 0.01-1 μM) and iberiotoxin (Ibtx, 1-100 nM), with EC50s of 38 ± 10 nM and 5 ± 2 nM for Pax and Ibtx, respectively, close to those reported for Pax- and Ibtx-induced Ca(2+)- and voltage-activated K(+) channels (Slo1 BK channels) blockade; the BK channel opener NS1619 (1-30 μM) induced a concentration-dependent protection against NMDA-induced excitotoxicity. Moreover, cortical neurons from mice lacking one or both alleles coding for Slo1 BK channel pore-forming subunits were insensitive to leptin-induced neuroprotection. Finally, leptin exposure dose-dependently (10-100 ng/ml) increased intracellular Ca(2+) levels in rat cortical neurons. In conclusion, our results suggest that Slo1 BK channel activation following increases in intracellular Ca(2+) levels is a critical step for leptin-induced neuroprotection in NMDA-exposed cortical neurons in vitro, thus highlighting leptin-based intervention via BK channel activation as a potential strategy to counteract neurodegenerative diseases.
Collapse
Affiliation(s)
- Maria Mancini
- Department of Medicine and Health Science, University of Molise, Campobasso, Italy; Department of Science and Technology, University of Sannio, Benevento, Italy; Center for Molecular Biomedicine, Department of Biophysics, Friedrich Schiller University and Jena University Hospital, Jena, Germany
| | | | - Guido Gessner
- Center for Molecular Biomedicine, Department of Biophysics, Friedrich Schiller University and Jena University Hospital, Jena, Germany
| | - Bianka Wissuwa
- Center for Sepsis Control and Care, Jena University Hospital, Jena, Germany
| | - Vincenzo Barrese
- Department of Neuroscience, University of Naples "Federico II", Naples, Italy
| | - Francesca Boscia
- Department of Neuroscience, University of Naples "Federico II", Naples, Italy
| | - Agnese Secondo
- Department of Neuroscience, University of Naples "Federico II", Naples, Italy
| | - Francesco Miceli
- Department of Neuroscience, University of Naples "Federico II", Naples, Italy
| | - Cristina Franco
- Department of Neuroscience, University of Naples "Federico II", Naples, Italy
| | - Paolo Ambrosino
- Department of Medicine and Health Science, University of Molise, Campobasso, Italy
| | | | - Michael Bauer
- Center for Sepsis Control and Care, Jena University Hospital, Jena, Germany
| | - Toshinori Hoshi
- Department of Physiology, University of Pennsylvania, Philadelphia, USA
| | - Stefan H Heinemann
- Center for Molecular Biomedicine, Department of Biophysics, Friedrich Schiller University and Jena University Hospital, Jena, Germany
| | - Maurizio Taglialatela
- Department of Medicine and Health Science, University of Molise, Campobasso, Italy; Department of Neuroscience, University of Naples "Federico II", Naples, Italy.
| |
Collapse
|
24
|
Hasegawa Y, Mukai H, Asashima M, Hojo Y, Ikeda M, Komatsuzaki Y, Ooishi Y, Kawato S. Acute modulation of synaptic plasticity of pyramidal neurons by activin in adult hippocampus. Front Neural Circuits 2014; 8:56. [PMID: 24917791 PMCID: PMC4040441 DOI: 10.3389/fncir.2014.00056] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2013] [Accepted: 05/11/2014] [Indexed: 11/25/2022] Open
Abstract
Activin A is known as a neuroprotective factor produced upon acute excitotoxic injury of the hippocampus (in pathological states). We attempt to reveal the role of activin as a neuromodulator in the adult male hippocampus under physiological conditions (in healthy states), which remains largely unknown. We showed endogenous/basal expression of activin in the hippocampal neurons. Localization of activin receptors in dendritic spines (=postsynapses) was demonstrated by immunoelectron microscopy. The incubation of hippocampal acute slices with activin A (10 ng/mL, 0.4 nM) for 2 h altered the density and morphology of spines in CA1 pyramidal neurons. The total spine density increased by 1.2-fold upon activin treatments. Activin selectively increased the density of large-head spines, without affecting middle-head and small-head spines. Blocking Erk/MAPK, PKA, or PKC prevented the activin-induced spinogenesis by reducing the density of large-head spines, independent of Smad-induced gene transcription which usually takes more than several hours. Incubation of acute slices with activin for 2 h induced the moderate early long-term potentiation (moderate LTP) upon weak theta burst stimuli. This moderate LTP induction was blocked by follistatin, MAPK inhibitor (PD98059) and inhibitor of NR2B subunit of NMDA receptors (Ro25-6981). It should be noted that the weak theta burst stimuli alone cannot induce moderate LTP. These results suggest that MAPK-induced phosphorylation of NMDA receptors (including NR2B) may play an important role for activin-induced moderate LTP. Taken together, the current results reveal interesting physiological roles of endogenous activin as a rapid synaptic modulator in the adult hippocampus.
Collapse
Affiliation(s)
- Yoshitaka Hasegawa
- Department of Biophysics and Life Sciences, Graduate School of Arts and Sciences, The University of Tokyo Meguro, Japan
| | - Hideo Mukai
- Department of Biophysics and Life Sciences, Graduate School of Arts and Sciences, The University of Tokyo Meguro, Japan ; Bioinformatics Project (BIRD), Japan Science and Technology Agency, The University of Tokyo Meguro, Japan ; Core Research for Evolutional Science and Technology Project of Japan Science and Technology Agency, The University of Tokyo Meguro, Japan ; Department of Computer Science, School of Science and Technology, Meiji University Kawasaki, Japan
| | - Makoto Asashima
- Department of Biophysics and Life Sciences, Graduate School of Arts and Sciences, The University of Tokyo Meguro, Japan
| | - Yasushi Hojo
- Department of Biophysics and Life Sciences, Graduate School of Arts and Sciences, The University of Tokyo Meguro, Japan ; Bioinformatics Project (BIRD), Japan Science and Technology Agency, The University of Tokyo Meguro, Japan ; Core Research for Evolutional Science and Technology Project of Japan Science and Technology Agency, The University of Tokyo Meguro, Japan
| | - Muneki Ikeda
- Department of Biophysics and Life Sciences, Graduate School of Arts and Sciences, The University of Tokyo Meguro, Japan
| | - Yoshimasa Komatsuzaki
- Department of Biophysics and Life Sciences, Graduate School of Arts and Sciences, The University of Tokyo Meguro, Japan
| | - Yuuki Ooishi
- Department of Biophysics and Life Sciences, Graduate School of Arts and Sciences, The University of Tokyo Meguro, Japan
| | - Suguru Kawato
- Department of Biophysics and Life Sciences, Graduate School of Arts and Sciences, The University of Tokyo Meguro, Japan ; Bioinformatics Project (BIRD), Japan Science and Technology Agency, The University of Tokyo Meguro, Japan ; Core Research for Evolutional Science and Technology Project of Japan Science and Technology Agency, The University of Tokyo Meguro, Japan ; National MEXT Project in Special Coordinate Funds for Promoting Science and Technology, The University of Tokyo Meguro, Japan
| |
Collapse
|
25
|
Leptin reverses corticosterone-induced inhibition of neural stem cell proliferation through activating the NR2B subunits of NMDA receptors. Biochem Biophys Res Commun 2014; 447:95-100. [DOI: 10.1016/j.bbrc.2014.03.112] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2014] [Accepted: 03/22/2014] [Indexed: 12/31/2022]
|
26
|
Gao Y, Wang C, Pan T, Luo L. Impact of metformin treatment and swimming exercise on visfatin levels in high-fat-induced obesity rats. ACTA ACUST UNITED AC 2014; 58:42-7. [PMID: 24728163 DOI: 10.1590/0004-2730000002840] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2013] [Accepted: 10/24/2013] [Indexed: 01/05/2023]
Abstract
Objective : Visfatin is a recently discovered adipocytokine that contributes to glucose and obesity-related conditions. Until now, its responses to the insulin-sensitizing agent metformin and to exercise are largely unknown. We aim to investigate the impact of metformin treatment and/or swimming exercise on serum visfatin and visfatin levels in subcutaneous adipose tissue (SAT), peri-renal adipose tissue (PAT) and skeletal muscle (SM) of high-fat-induced obesity rats. Materials and methods : Sprague-Dawley rats were fed a normal diet or a high-fat diet for 16 weeks to develop obesity model. The high-fat-induced obesity model rats were then randomized to metformin (MET), swimming exercise (SWI), or adjunctive therapy of metformin and swimming exercise (MAS), besides high-fat obesity control group and a normal control group, all with 10 rats per group. Zoometric and glycemic parameters, lipid profile, and serum visfatin levels were assessed at baseline and after 6 weeks of therapy. Visfatin levels in SAT, PAT and SM were determined by Western Blot. Results : Metformin and swimming exercise improved lipid profile, and increased insulin sensitivity and body weight reduction were observed. Both metformin and swimming exercise down-regulated visfatin levels in SAT and PAT, while the adjunctive therapy conferred greater benefits, but no changes of visfatin levels were observed in SM. Conclusion : Our results indicate that visfatin down-regulation in SAT and PAT may be one of the mechanisms by which metformin and swimming exercise inhibit obesity.
Collapse
Affiliation(s)
- Ya Gao
- The Second Hospital of An Hui Medical University, China
| | - Changjiang Wang
- The First Affiliated Hospital of An Hui Medical University, China
| | - Tianrong Pan
- The Second Hospital of An Hui Medical University, China
| | - Li Luo
- The First Affiliated Hospital of An Hui Medical University, China
| |
Collapse
|
27
|
Irving AJ, Harvey J. Leptin regulation of hippocampal synaptic function in health and disease. Philos Trans R Soc Lond B Biol Sci 2013; 369:20130155. [PMID: 24298156 DOI: 10.1098/rstb.2013.0155] [Citation(s) in RCA: 96] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
The endocrine hormone leptin plays a key role in regulating food intake and body weight via its actions in the hypothalamus. However, leptin receptors are highly expressed in many extra-hypothalamic brain regions and evidence is growing that leptin influences many central processes including cognition. Indeed, recent studies indicate that leptin is a potential cognitive enhancer as it markedly facilitates the cellular events underlying hippocampal-dependent learning and memory, including effects on glutamate receptor trafficking, neuronal morphology and activity-dependent synaptic plasticity. However, the ability of leptin to regulate hippocampal synaptic function markedly declines with age and aberrant leptin function has been linked to neurodegenerative disorders such as Alzheimer's disease (AD). Here, we review the evidence supporting a cognitive enhancing role for the hormone leptin and discuss the therapeutic potential of using leptin-based agents to treat AD.
Collapse
Affiliation(s)
- Andrew J Irving
- Division of Neuroscience, Medical Research Institute, Ninewells Hospital and Medical School, University of Dundee, , Dundee DD1 9SY, UK
| | | |
Collapse
|
28
|
Streijger F, Plunet WT, Lee JHT, Liu J, Lam CK, Park S, Hilton BJ, Fransen BL, Matheson KAJ, Assinck P, Kwon BK, Tetzlaff W. Ketogenic diet improves forelimb motor function after spinal cord injury in rodents. PLoS One 2013; 8:e78765. [PMID: 24223849 PMCID: PMC3817084 DOI: 10.1371/journal.pone.0078765] [Citation(s) in RCA: 86] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2013] [Accepted: 09/16/2013] [Indexed: 11/29/2022] Open
Abstract
High fat, low carbohydrate ketogenic diets (KD) are validated non-pharmacological treatments for some forms of drug-resistant epilepsy. Ketones reduce neuronal excitation and promote neuroprotection. Here, we investigated the efficacy of KD as a treatment for acute cervical spinal cord injury (SCI) in rats. Starting 4 hours following C5 hemi-contusion injury animals were fed either a standard carbohydrate based diet or a KD formulation with lipid to carbohydrate plus protein ratio of 3:1. The forelimb functional recovery was evaluated for 14 weeks, followed by quantitative histopathology. Post-injury 3:1 KD treatment resulted in increased usage and range of motion of the affected forepaw. Furthermore, KD improved pellet retrieval with recovery of wrist and digit movements. Importantly, after returning to a standard diet after 12 weeks of KD treatment, the improved forelimb function remained stable. Histologically, the spinal cords of KD treated animals displayed smaller lesion areas and more grey matter sparing. In addition, KD treatment increased the number of glucose transporter-1 positive blood vessels in the lesion penumbra and monocarboxylate transporter-1 (MCT1) expression. Pharmacological inhibition of MCTs with 4-CIN (α-cyano-4-hydroxycinnamate) prevented the KD-induced neuroprotection after SCI, In conclusion, post-injury KD effectively promotes functional recovery and is neuroprotective after cervical SCI. These beneficial effects require the function of monocarboxylate transporters responsible for ketone uptake and link the observed neuroprotection directly to the function of ketones, which are known to exert neuroprotection by multiple mechanisms. Our data suggest that current clinical nutritional guidelines, which include relatively high carbohydrate contents, should be revisited.
Collapse
Affiliation(s)
- Femke Streijger
- International Collaboration on Repair Discoveries (ICORD), Blusson Spinal Cord Center, Vancouver, British Columbia, Canada
| | - Ward T. Plunet
- International Collaboration on Repair Discoveries (ICORD), Blusson Spinal Cord Center, Vancouver, British Columbia, Canada
| | - Jae H. T. Lee
- International Collaboration on Repair Discoveries (ICORD), Blusson Spinal Cord Center, Vancouver, British Columbia, Canada
| | - Jie Liu
- International Collaboration on Repair Discoveries (ICORD), Blusson Spinal Cord Center, Vancouver, British Columbia, Canada
| | - Clarrie K. Lam
- International Collaboration on Repair Discoveries (ICORD), Blusson Spinal Cord Center, Vancouver, British Columbia, Canada
| | - Soeyun Park
- International Collaboration on Repair Discoveries (ICORD), Blusson Spinal Cord Center, Vancouver, British Columbia, Canada
| | - Brett J. Hilton
- International Collaboration on Repair Discoveries (ICORD), Blusson Spinal Cord Center, Vancouver, British Columbia, Canada
| | - Bas L. Fransen
- International Collaboration on Repair Discoveries (ICORD), Blusson Spinal Cord Center, Vancouver, British Columbia, Canada
| | - Keely A. J. Matheson
- International Collaboration on Repair Discoveries (ICORD), Blusson Spinal Cord Center, Vancouver, British Columbia, Canada
| | - Peggy Assinck
- International Collaboration on Repair Discoveries (ICORD), Blusson Spinal Cord Center, Vancouver, British Columbia, Canada
| | - Brian K. Kwon
- International Collaboration on Repair Discoveries (ICORD), Blusson Spinal Cord Center, Vancouver, British Columbia, Canada
- Department of Zoology, University of British Columbia, Vancouver, British Columbia, Canada
- Combined Neurosurgical and Orthopaedic Spine Program, Department of Orthopaedics, University of British Columbia, Vancouver, British Columbia, Canada
| | - Wolfram Tetzlaff
- International Collaboration on Repair Discoveries (ICORD), Blusson Spinal Cord Center, Vancouver, British Columbia, Canada
- Department of Zoology, University of British Columbia, Vancouver, British Columbia, Canada
- * E-mail:
| |
Collapse
|
29
|
Spatial memory impairment and changes in hippocampal morphology are triggered by high-fat diets in adolescent mice. Is there a role of leptin? Neurobiol Learn Mem 2013; 106:18-25. [DOI: 10.1016/j.nlm.2013.06.012] [Citation(s) in RCA: 101] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2013] [Revised: 05/31/2013] [Accepted: 06/21/2013] [Indexed: 02/06/2023]
|
30
|
Ha S, Baver S, Huo L, Gata A, Hairston J, Huntoon N, Li W, Zhang T, Benecchi EJ, Ericsson M, Hentges ST, Bjørbæk C. Somato-dendritic localization and signaling by leptin receptors in hypothalamic POMC and AgRP neurons. PLoS One 2013; 8:e77622. [PMID: 24204898 PMCID: PMC3812230 DOI: 10.1371/journal.pone.0077622] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2013] [Accepted: 09/13/2013] [Indexed: 11/18/2022] Open
Abstract
Leptin acts via neuronal leptin receptors to control energy balance. Hypothalamic pro-opiomelanocortin (POMC) and agouti-related peptide (AgRP)/Neuropeptide Y (NPY)/GABA neurons produce anorexigenic and orexigenic neuropeptides and neurotransmitters, and express the long signaling form of the leptin receptor (LepRb). Despite progress in the understanding of LepRb signaling and function, the sub-cellular localization of LepRb in target neurons has not been determined, primarily due to lack of sensitive anti-LepRb antibodies. Here we applied light microscopy (LM), confocal-laser scanning microscopy (CLSM), and electron microscopy (EM) to investigate LepRb localization and signaling in mice expressing a HA-tagged LepRb selectively in POMC or AgRP/NPY/GABA neurons. We report that LepRb receptors exhibit a somato-dendritic expression pattern. We further show that LepRb activates STAT3 phosphorylation in neuronal fibers within several hypothalamic and hindbrain nuclei of wild-type mice and rats, and specifically in dendrites of arcuate POMC and AgRP/NPY/GABA neurons of Leprb+/+ mice and in Leprbdb/db mice expressing HA-LepRb in a neuron specific manner. We did not find evidence of LepRb localization or STAT3-signaling in axon-fibers or nerve-terminals of POMC and AgRP/NPY/GABA neurons. Three-dimensional serial EM-reconstruction of dendritic segments from POMC and AgRP/NPY/GABA neurons indicates a high density of shaft synapses. In addition, we found that the leptin activates STAT3 signaling in proximity to synapses on POMC and AgRP/NPY/GABA dendritic shafts. Taken together, these data suggest that the signaling-form of the leptin receptor exhibits a somato-dendritic expression pattern in POMC and AgRP/NPY/GABA neurons. Dendritic LepRb signaling may therefore play an important role in leptin’s central effects on energy balance, possibly through modulation of synaptic activity via post-synaptic mechanisms.
Collapse
Affiliation(s)
- Sangdeuk Ha
- Department of Medicine, Division of Endocrinology and Metabolism, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, Massachusetts, United States of America
| | - Scott Baver
- Department of Medicine, Division of Endocrinology and Metabolism, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, Massachusetts, United States of America
| | - Lihong Huo
- Department of Medicine, Division of Endocrinology and Metabolism, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, Massachusetts, United States of America
| | - Adriana Gata
- Department of Medicine, Division of Endocrinology and Metabolism, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, Massachusetts, United States of America
| | - Joyce Hairston
- Department of Medicine, Division of Endocrinology and Metabolism, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, Massachusetts, United States of America
| | - Nicholas Huntoon
- Department of Medicine, Division of Endocrinology and Metabolism, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, Massachusetts, United States of America
| | - Wenjing Li
- Department of Medicine, Division of Endocrinology and Metabolism, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, Massachusetts, United States of America
| | - Thompson Zhang
- Department of Medicine, Division of Endocrinology and Metabolism, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, Massachusetts, United States of America
| | - Elizabeth J. Benecchi
- Electron Microscopy Facility, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Maria Ericsson
- Electron Microscopy Facility, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Shane T. Hentges
- Department of Biomedical Sciences, Colorado State University, Fort Collins, Colorado, United States of America
| | - Christian Bjørbæk
- Department of Medicine, Division of Endocrinology and Metabolism, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, Massachusetts, United States of America
- * E-mail :
| |
Collapse
|
31
|
Harvey J. Leptin regulation of neuronal morphology and hippocampal synaptic function. Front Synaptic Neurosci 2013; 5:3. [PMID: 23964236 PMCID: PMC3734345 DOI: 10.3389/fnsyn.2013.00003] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2013] [Accepted: 07/12/2013] [Indexed: 12/14/2022] Open
Abstract
The central actions of the hormone leptin in regulating energy homeostasis via the hypothalamus are well documented. However, evidence is growing that this hormone can also modify the structure and function of synapses throughout the CNS. The hippocampus is a region of the forebrain that plays a crucial role in associative learning and memory and is an area also highly vulnerable to neurodegenerative processes. Recent studies indicate that leptin is a potential cognitive enhancer as it modulates the cellular processes underlying hippocampal-dependent learning and memory including dendritic morphology, glutamate receptor trafficking and activity-dependent synaptic plasticity. Here, we review the recent evidence implicating the hormone leptin as a key regulator of hippocampal synaptic function and discuss the role of leptin receptor-driven lipid signaling pathways involved in this process.
Collapse
Affiliation(s)
- Jenni Harvey
- Division of Neuroscience, Medical Research Institute, Ninewells Hospital and Medical School, University of Dundee Dundee, UK
| |
Collapse
|
32
|
Thompson JL, Borgland SL. Presynaptic leptin action suppresses excitatory synaptic transmission onto ventral tegmental area dopamine neurons. Biol Psychiatry 2013; 73:860-8. [PMID: 23305991 DOI: 10.1016/j.biopsych.2012.10.026] [Citation(s) in RCA: 50] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/14/2012] [Revised: 10/31/2012] [Accepted: 10/31/2012] [Indexed: 12/28/2022]
Abstract
BACKGROUND Leptin is an adipocyte-derived cytokine that can act in the brain to suppress feeding and maintain energy homeostasis. Additionally, leptin activates its receptors in the ventral tegmental area (VTA), a critical site for neuroadaptations to rewarding stimuli, to modulate reward-seeking behaviors. Although leptin can decrease intrinsic excitability of dopamine neurons in the VTA, it is unknown whether leptin can modulate excitatory synaptic transmission in this brain region. Because plasticity of glutamatergic synapses onto VTA neurons can encode predictive information about reward, we hypothesized that leptin can decrease excitatory synaptic transmission onto dopamine neurons. METHODS Using whole-cell patch clamp electrophysiology in mouse midbrain slices, we tested the effects of leptin on evoked α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid receptor (AMPAR) or N-methyl-D-aspartate receptor (NMDAR)-mediated excitatory postsynaptic currents (EPSCs) onto VTA dopamine neurons. RESULTS Leptin depressed both AMPAR and NMDAR EPSCs in VTA dopamine neurons and reduced frequency but not amplitude of mini EPSCs. Bath application of the MEK1/2 inhibitor U0126 did not alter leptin-induced suppression of AMPAR EPSCs. However, external, but not internal, application of the phosphoinositol 3-kinase (PI3K) or Janus kinase 2 (Jak2) tyrosine kinase inhibitors abolished leptin-induced synaptic depression. CONCLUSIONS This study demonstrates that leptin causes a presynaptic inhibition of the probability of glutamate release onto VTA dopamine neurons. This synaptic inhibition requires Jak2 and PI3K activation. Leptin-induced weakening of synaptic strength onto dopamine cells may underlie its inhibitory effects on appetitive behavior for rewarding stimuli.
Collapse
Affiliation(s)
- Jennifer L Thompson
- Department of Anesthesiology, Pharmacology and Therapeutics, University of British Columbia, Vancouver, Canada
| | | |
Collapse
|
33
|
Regulation of synaptic functions in central nervous system by endocrine hormones and the maintenance of energy homoeostasis. Biosci Rep 2013; 32:423-32. [PMID: 22582733 PMCID: PMC3804927 DOI: 10.1042/bsr20120026] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023] Open
Abstract
Energy homoeostasis, a co-ordinated balance of food intake and energy expenditure, is regulated by the CNS (central nervous system). The past decade has witnessed significant advances in our understanding of metabolic processes and brain circuitry which responds to a broad range of neural, nutrient and hormonal signals. Accumulating evidence demonstrates altered synaptic plasticity in the CNS in response to hormone signals. Moreover, emerging observations suggest that synaptic plasticity underlies all brain functions, including the physiological regulation of energy homoeostasis, and that impaired synaptic constellation and plasticity may lead to pathological development and conditions. Here, we summarize the current knowledge on the regulation of postsynaptic receptors such as AMPA (α-amino-3-hydroxy-5-methylisoxazole-4-propionic acid), NMDA (N-methyl-D-aspartate) and GABA (γ-aminobutyric acid) receptors, and the presynaptic components by hormone signals. A detailed understanding of the neurobiological mechanisms by which hormones regulate energy homoeostasis may lead to novel strategies in treating metabolic disorders.
Collapse
|
34
|
Khoshdel A, Parvin N, Abbasi M. Selenium and leptin levels in febrile seizure: a case-control study in children. KOREAN JOURNAL OF PEDIATRICS 2013; 56:80-5. [PMID: 23482826 PMCID: PMC3589595 DOI: 10.3345/kjp.2013.56.2.80] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/30/2012] [Revised: 09/11/2012] [Accepted: 10/25/2012] [Indexed: 11/27/2022]
Abstract
PURPOSE Febrile seizures (FS) are seizures that occur between the age of 6 and 60 months, but its pathophysiology still is not fully understood. There is limited information about the correlation between levels of selenium and leptin with FS. This study aimed to determine the relationship between serum levels of selenium and leptin in children with FS. METHODS This case-control study was conducted in a University Hospital in Shahrekord, Iran, in 2011. The serum levels of selenium and leptin of 25 children with simple febrile seizure (case group) were compared with 25 febrile children without seizure (control group) in acute phase and after three months. The levels of serum selenium and leptin were measured by flame atomic absorption spectrophotometer and enzyme-linked immunosorbent assay method, respectively. RESULTS In acute phase, the mean serum level of selenium in case and control groups were 95.88±42.55 and 113.25±54.43 µg/dL, respectively, and difference was not significant (P=0.415), but after three months, this level had a significant increase in both groups (P<0.001). In acute phase, the mean serum leptin level in case and control groups were 0.94±0.5 and 0.98±0.84 ng/mL, respectively, but difference was not significant (P=0.405). After three months, serum leptin level had no significant change in both groups (P=0.882). CONCLUSION These observations suggest that serum levels of selenium and leptin have not specific relation with FS but overllay is lower, however, further study is recommended. Also selenium level in stress and acute phase was significantly lower than recovery phase.
Collapse
Affiliation(s)
- Abolfazl Khoshdel
- Department of Pediatrics, Shahrekord University of Medical Sciences, Shahrekord, Iran
| | | | | |
Collapse
|
35
|
Berman SM, Paz-Filho G, Wong ML, Kohno M, Licinio J, London ED. Effects of leptin deficiency and replacement on cerebellar response to food-related cues. CEREBELLUM (LONDON, ENGLAND) 2013; 12:59-67. [PMID: 22576622 PMCID: PMC3569483 DOI: 10.1007/s12311-012-0360-z] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
Leptin affects eating behavior partly by altering the response of the brain to food-related stimuli. The effects of leptin on brain structure have been observed in the cerebellum, where leptin receptors are most densely expressed, but the function of leptin in the cerebellum remains unclear. We performed a nonrandomized, prospective interventional study of three adults with genetically mediated leptin deficiency. FMRI was recorded three times each year during years 5 and 6 of leptin replacement treatment. Session 1 of each year occurred after 10 months of continuous daily replacement, session 2 after 33-37 days without leptin, and session 3 at 14-23 days after daily replacement was restored. Statistical parametric mapping software (SPM5) was employed to contrast the fMRI blood oxygenation level-dependent response to images of high-calorie foods versus images of brick walls. Covariate analyses quantified the effects of the duration of leptin replacement and concomitant changes in body mass on the cerebral responses. Longer duration of replacement was associated with more activation by food images in a ventral portion of the posterior lobe of the cerebellum, while simultaneous decreases in body mass were associated with decreased activation in a more dorsal portion of the same lobe. These findings indicate that leptin replacement reversibly alters neural function within the posterior cerebellum and modulates plasticity-dependent brain physiology in response to food cues. The results suggest an underexplored role for the posterior cerebellum in the regulation of leptin-mediated processes related to food intake.
Collapse
Affiliation(s)
- Steven M. Berman
- Department of Psychiatry and Biobehavioral Sciences and the Semel Institute, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California 90024
- Brain Research Institute, University of California Los Angeles, Los Angeles, California 90024
| | - Gilberto Paz-Filho
- Department of Translational Medicine, John Curtin School of Medical Research, The Australian National University, Canberra, ACT, Australia 0200
| | - Ma-Li Wong
- Department of Translational Medicine, John Curtin School of Medical Research, The Australian National University, Canberra, ACT, Australia 0200
| | - Milky Kohno
- Department of Psychiatry and Biobehavioral Sciences and the Semel Institute, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California 90024
| | - Julio Licinio
- Department of Translational Medicine, John Curtin School of Medical Research, The Australian National University, Canberra, ACT, Australia 0200
| | - Edythe D. London
- Department of Psychiatry and Biobehavioral Sciences and the Semel Institute, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California 90024
- Department of Molecular and Medical Pharmacology, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California 90024
- Brain Research Institute, University of California Los Angeles, Los Angeles, California 90024
| |
Collapse
|
36
|
Rizig MA, McQuillin A, Ng A, Robinson M, Harrison A, Zvelebil M, Hunt SP, Gurling HM. A gene expression and systems pathway analysis of the effects of clozapine compared to haloperidol in the mouse brain implicates susceptibility genes for schizophrenia. J Psychopharmacol 2012; 26:1218-30. [PMID: 22767372 DOI: 10.1177/0269881112450780] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Clozapine has markedly superior clinical properties compared to other antipsychotic drugs but the side effects of agranulocytosis, weight gain and diabetes limit its use. The reason why clozapine is more effective is not well understood. We studied messenger RNA (mRNA) gene expression in the mouse brain to identify pathways changed by clozapine compared to those changed by haloperidol so that we could identify which changes were specific to clozapine. Data interpretation was performed using an over-representation analysis (ORA) of gene ontology (GO), pathways and gene-by-gene differences. Clozapine significantly changed gene expression in pathways related to neuronal growth and differentiation to a greater extent than haloperidol; including the microtubule-associated protein kinase (MAPK) signalling and GO terms related to axonogenesis and neuroblast proliferation. Several genes implicated genetically or functionally in schizophrenia such as frizzled homolog 3 (FZD3), U2AF homology motif kinase 1 (UHMK1), pericentriolar material 1 (PCM1) and brain-derived neurotrophic factor (BDNF) were changed by clozapine but not by haloperidol. Furthermore, when compared to untreated controls clozapine specifically regulated transcripts related to the glutamate system, microtubule function, presynaptic proteins and pathways associated with synaptic transmission such as clathrin cage assembly. Compared to untreated controls haloperidol modulated expression of neurotoxic and apoptotic responses such as NF-kappa B and caspase pathways, whilst clozapine did not. Pathways involving lipid and carbohydrate metabolism and appetite regulation were also more affected by clozapine than by haloperidol.
Collapse
Affiliation(s)
- Mie A Rizig
- Molecular Psychiatry Laboratory, University College London, London, UK
| | | | | | | | | | | | | | | |
Collapse
|
37
|
Wu X, Hsuchou H, Kastin AJ, Mishra PK, Pan W. Upregulation of astrocytic leptin receptor in mice with experimental autoimmune encephalomyelitis. J Mol Neurosci 2012; 49:446-56. [PMID: 22684620 DOI: 10.1007/s12031-012-9825-2] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2012] [Accepted: 05/28/2012] [Indexed: 01/02/2023]
Abstract
The detrimental role of leptin in experimental autoimmune encephalomyelitis (EAE) is opposite to its neuroprotective role in other neuropathologies. We hypothesize that a shifted cellular distribution of leptin receptors underlies the differential effects of leptin. A robust increase of ObR immunoreactivity was seen along glial fibrillary acidic protein (GFAP)(+) intermediate filaments in reactive astrocytes in the hippocampus and hypothalamus of mice with EAE. Although astrocyte-specific GFAP mRNA and protein were both increased, ObRa mRNA was elevated only after resolution of EAE symptoms, and ObRb mRNA was even decreased at the peak time of symptoms of EAE. A cell type-specific action of leptin may underlie its differential effects.
Collapse
Affiliation(s)
- Xiaojun Wu
- Blood-Brain Barrier Group, Pennington Biomedical Research Center, Baton Rouge, LA 70808, USA
| | | | | | | | | |
Collapse
|
38
|
Pan W, Hsuchou H, Jayaram B, Khan RS, Huang EYK, Wu X, Chen C, Kastin AJ. Leptin action on nonneuronal cells in the CNS: potential clinical applications. Ann N Y Acad Sci 2012; 1264:64-71. [PMID: 22530983 PMCID: PMC3407332 DOI: 10.1111/j.1749-6632.2012.06472.x] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Leptin, an adipocyte-derived cytokine, crosses the blood–brain barrier to act on many regions of the central nervous system (CNS). It participates in the regulation of energy balance, inflammatory processes, immune regulation, synaptic formation, memory condensation, and neurotrophic activities. This review focuses on the newly identified actions of leptin on astrocytes. We first summarize the distribution of leptin receptors in the brain, with a focus on the hypothalamus, where the leptin receptor is known to mediate essential feeding suppression activities, and on the hippocampus, where leptin facilitates memory, reduces neurodegeneration, and plays a dual role in seizures. We will then discuss regulation of the nonneuronal leptin system in obesity. Its relationship with neuronal leptin signaling is illustrated by in vitro assays in primary astrocyte culture and by in vivo studies on mice after pretreatment with a glial metabolic inhibitor or after cell-specific deletion of intracellular signaling leptin receptors. Overall, the glial leptin system shows robust regulation and plays an essential role in obesity. Strategies to manipulate this nonneuronal leptin signaling may have major clinical impact.
Collapse
Affiliation(s)
- Weihong Pan
- Blood-Brain Barrier Group, Pennington Biomedical Research Center, Baton Rouge, Lousiana 70808, USA.
| | | | | | | | | | | | | | | |
Collapse
|
39
|
Valladolid-Acebes I, Merino B, Principato A, Fole A, Barbas C, Lorenzo MP, García A, Del Olmo N, Ruiz-Gayo M, Cano V. High-fat diets induce changes in hippocampal glutamate metabolism and neurotransmission. Am J Physiol Endocrinol Metab 2012; 302:E396-402. [PMID: 22114023 DOI: 10.1152/ajpendo.00343.2011] [Citation(s) in RCA: 95] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Obesity and high-fat (HF) diets have a deleterious impact on hippocampal function and lead to impaired synaptic plasticity and learning deficits. Because all of these processes need an adequate glutamatergic transmission, we have hypothesized that nutritional imbalance triggered by these diets might eventually concern glutamate (Glu) neural pathways within the hippocampus. Glu is withdrawn from excitatory synapses by specific uptake mechanisms involving neuronal (EAAT-3) and glial (GLT-1, GLAST) transporters, which regulate the time that synaptically released Glu remains in the extracellular space and, consequently, the duration and location of postsynaptic receptor activation. The goal of the present study was to evaluate in mouse hippocampus the effect of a short-term high-fat dietary treatment on 1) Glu uptake kinetics, 2) the density of Glu carriers and Glu-degrading enzymes, 3) the density of Glu receptor subunits, and 4) synaptic transmission and plasticity. Here, we show that HF diet triggers a 50% decrease of the Michaelis-Menten constant together with a 300% increase of the maximal velocity of the uptake process. Glial Glu carriers GLT-1 and GLAST were upregulated in HF mice (32 and 27%, respectively), whereas Glu-degrading enzymes glutamine synthase and GABA-decarboxilase appeared to be downregulated in these animals. In addition, HF diet hippocampus displayed diminished basal synaptic transmission and hindered NMDA-induced long-term depression (NMDA-LTD). This was coincident with a reduced density of the NR2B subunit of NMDA receptors. All of these results are compatible with the development of leptin resistance within the hippocampus. Our data show that HF diets upregulate mechanisms involved in Glu clearance and simultaneously impair Glu metabolism. Neurochemical changes occur concomitantly with impaired basal synaptic transmission and reduced NMDA-LTD. Taken together, our results suggest that HF diets trigger neurochemical changes, leading to a desensitization of NMDA receptors within the hippocampus, which might account for cognitive deficits.
Collapse
Affiliation(s)
- Ismael Valladolid-Acebes
- Departamento de Ciencias Farmacéuticas y de la Alimentación, Facultad de Farmacia, Universidad CEU-San Pablo, Madrid, Spain
| | | | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Moult PR, Harvey J. NMDA receptor subunit composition determines the polarity of leptin-induced synaptic plasticity. Neuropharmacology 2011; 61:924-36. [PMID: 21752339 PMCID: PMC3300059 DOI: 10.1016/j.neuropharm.2011.06.021] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2011] [Revised: 06/21/2011] [Accepted: 06/22/2011] [Indexed: 11/16/2022]
Abstract
Leptin is a hormone that crosses the blood-brain barrier and regulates numerous CNS functions. The hippocampus in particular is an important site for leptin action. Indeed, leptin markedly influences excitatory synaptic transmission and synaptic plasticity in this brain region. Recent studies indicate that leptin modulation of hippocampal excitatory synaptic transmission is age-dependent however the cellular basis for this is unclear. Here we show that early in development leptin evokes a transient (P11-18) or persistent (P5-8) depression of synaptic transmission, whereas leptin evokes a long lasting increase (LTP) in synaptic strength in adulthood. The synaptic depressions induced by leptin required activation of NMDA receptor GluN2B subunits and the ERK signalling cascade. Conversely, leptin-induced LTP in adult was mediated by GluN2A subunits and involved PI 3-kinase dependent signalling. In addition, low-frequency stimulus (LFS)-evoked LTD occluded the persistent effects of leptin at P5-8 and vice versa. Similarly, synaptically-induced LTP occluded the persistent increase in synaptic transmission induced by leptin, indicating that similar expression mechanisms underlie leptin-induced LTD and LFS-induced LTD at P5-8, and leptin-induced LTP and HFS-induced LTP in adult. These findings have important implications for the role of leptin in hippocampal synaptic function during early neuronal development and in aging.
Collapse
Affiliation(s)
- Peter R Moult
- Centre for Neuroscience, Division of Medical Sciences, Ninewells Hospital and Medical School, University of Dundee, Dundee, UK
| | | |
Collapse
|
41
|
Pekary AE, Sattin A, Blood J. Rapid modulation of TRH and TRH-like peptide release in rat brain and peripheral tissues by leptin. Brain Res 2010; 1345:9-18. [PMID: 20546704 DOI: 10.1016/j.brainres.2010.05.039] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2010] [Accepted: 05/13/2010] [Indexed: 11/15/2022]
Abstract
Leptin is not only a feedback modulator of feeding and energy expenditure but also regulates reproductive functions, CNS development and mood. Obesity and major depression are growing public health concerns which may derive, in part, from disregulation of leptin feedback at the level of the hypothalamic feeding centers and mood regulators within the limbic system. Identifying downstream mediators of leptin action may provide therapeutic opportunities. We and others have previously reported that thyrotropin-releasing hormone (TRH, pGlu-His-Pro-NH(2)) and TRH-like peptides (pGlu-X-Pro-NH(2), where "X" can be any amino acid residue) have neuroprotective, antidepressant, anti-epileptic, analeptic, anti-ataxic, and anorectic properties. For this reason, young, adult male Sprague-Dawley rats were injected ip with 1mg/kg rat leptin and peptide and protein levels were measured in brain and peripheral tissues at 0, 0.5, 1 and 2h later. Eleven brain regions: pyriform cortex (PYR), entorhinal cortex (ENT), cerebellum (CBL), nucleus accumbens (NA), frontal cortex (FCX), amygdala (AY), posterior cingulate (PCNG), striatum (STR), hippocampus (HC), medulla oblongata (MED) and anterior cingulate (ACNG) and five peripheral tissues (adrenals, testes, epididymis, pancreas and prostate) were analyzed. TRH and six TRH-like peptide levels in STR fell by 0.5h consistent with leptin-induced release of these peptides: STR (7 downward arrow). Significant changes in TRH and TRH-like peptide levels for other brain regions were: CBL (5 downward arrow), ENT (5 downward arrow), HC (4 downward arrow), AY (4 downward arrow), FCX (3 downward arrow), and ACNG (1 downward arrow). The rapid modulation of TRH and TRH-like peptide release combined with their similarity in behavioral, neuroendocrine, immunomodulatory, metabolic and steroidogenic effects to that of leptin is consistent with these peptides participating in downstream signaling.
Collapse
Affiliation(s)
- A E Pekary
- Research Service, VA Greater Los Angeles Healthcare System, Los Angeles, CA 90073, USA.
| | | | | |
Collapse
|
42
|
Lynch JJ, Shek EW, Castagné V, Mittelstadt SW. The proconvulsant effects of leptin on glutamate receptor-mediated seizures in mice. Brain Res Bull 2010; 82:99-103. [PMID: 20152888 DOI: 10.1016/j.brainresbull.2010.02.003] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2009] [Revised: 02/01/2010] [Accepted: 02/03/2010] [Indexed: 10/19/2022]
Abstract
The metabolic-related hormone, leptin has been suggested for clinical use as an anticonvulsant based upon data generated from in vitro and in vivo non-human studies. However, a number of other non-human experiments have demonstrated proconvulsant activity for leptin. The current study investigated potential pro- and anticonvulsant effects of leptin during exposure to either glutamate (the major endogenous excitatory neurotransmitter) or three subtype-selective glutamate receptor agonists (N-methyl-d-aspartic acid [NMDA], alpha-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid [AMPA], and kainate). Male C57BL/6JRj mice were pretreated with leptin (0.1-10mg/kg, i.p.) and then administered doses of the glutamate receptor agonists (i.p.) that had been previously shown to result in clonic convulsions in approximately half of the animals tested. Leptin had no clear convulsant-related effects with either glutamate or AMPA, but it exhibited dose-related, proconvulsant activity (decreased latency to first occurrence of various convulsion-related signs, and increased percentage of animals exhibiting such signs) with both NMDA and kainate. The proconvulsant effects of leptin observed during the current study suggest that a cautious approach should be taken when administering leptin to individuals who may be prone to seizures.
Collapse
Affiliation(s)
- James J Lynch
- Department of Integrative Pharmacology, Global Pharmaceutical Research and Development, Abbott Laboratories, 100 Abbott Park Road, Abbott Park, IL 60064-6119, USA.
| | | | | | | |
Collapse
|
43
|
|
44
|
Zhao W, Wu X, Xie H, Ke Y, Yung WH. Permissive Role of Insulin in the Expression of Long-Term Potentiation in the Hippocampus of Immature Rats. Neurosignals 2010; 18:236-45. [DOI: 10.1159/000324040] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2010] [Accepted: 12/30/2010] [Indexed: 01/21/2023] Open
|
45
|
Wozniak SE, Gee LL, Wachtel MS, Frezza EE. Adipose tissue: the new endocrine organ? A review article. Dig Dis Sci 2009; 54:1847-56. [PMID: 19052866 DOI: 10.1007/s10620-008-0585-3] [Citation(s) in RCA: 354] [Impact Index Per Article: 22.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/20/2008] [Accepted: 10/13/2008] [Indexed: 02/07/2023]
Abstract
Fat is either white or brown, the latter being found principally in neonates. White fat, which comprises adipocytes, pre-adipocytes, macrophages, endothelial cells, fibroblasts, and leukocytes, actively participates in hormonal and inflammatory systems. Adipokines include hormones such as leptin, adiponectin, visfatin, apelin, vaspin, hepcidine, chemerin, omentin, and inflammatory cytokines, including tumor necrosis factor alpha (TNF), monocyte chemoattractant protein-1 (MCP-1), and plasminogen activator protein (PAI). Multiple roles in metabolic and inflammatory responses have been assigned to adipokines; this review describes the molecular actions and clinical significance of the more important adipokines. The array of adipokines evidences diverse roles for adipose tissue, which looms large in the mediators of inflammation and metabolism. For this reason, treating obesity is more than a reduction of excess fat; it is also the treatment of obesity's comorbidities, many of which will some day be treated by drugs that counteract derangements induced by adipokine excesses.
Collapse
Affiliation(s)
- Susan E Wozniak
- Department of Surgery, Texas Tech University Health Sciences Center, Lubbock, TX, USA
| | | | | | | |
Collapse
|
46
|
Narita K, Kosaka H, Okazawa H, Murata T, Wada Y. Relationship between plasma leptin level and brain structure in elderly: a voxel-based morphometric study. Biol Psychiatry 2009; 65:992-4. [PMID: 19026409 DOI: 10.1016/j.biopsych.2008.10.006] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/01/2008] [Revised: 10/07/2008] [Accepted: 10/07/2008] [Indexed: 01/08/2023]
Abstract
BACKGROUND Recent accumulating lines of evidence reveal that leptin is associated with synaptic plasticity and neuroprotective activity in the brain. METHODS In this preliminary study with a cross-sectional design, we examined the relationship between plasma leptin level and total or regional gray matter (GM) volume in 34 elderly subjects (mean age 64.5 years) with normal fasting glucose level and without dementia and metabolic syndrome by voxel-based morphometry of magnetic resonance imaging scans. RESULTS Plasma leptin level showed no significant correlation with total GM volume but showed a significantly positive correlation with GM volumes in the right hippocampus, left parahippocampus, and right cerebellum with adjustments for age, gender, body mass index (BMI), and waist-to-hip ratio (W/H). Also, after adjustments for age, gender, BMI, W/H, and intracranial volume, plasma leptin level significantly positively correlated with GM volumes in the right hippocampus and bilateral cerebella but not with that in the left parahippocampus. CONCLUSIONS The results of this pilot study would be beneficial for our understanding of the neuroprotective effects of leptin on human brain aging.
Collapse
Affiliation(s)
- Kosuke Narita
- Department of Neuropsychiatry, University of Fukui, Fukui 910-1193, Japan
| | | | | | | | | |
Collapse
|
47
|
Morrison CD. Leptin signaling in brain: A link between nutrition and cognition? BIOCHIMICA ET BIOPHYSICA ACTA 2009; 1792:401-8. [PMID: 19130879 PMCID: PMC2670357 DOI: 10.1016/j.bbadis.2008.12.004] [Citation(s) in RCA: 161] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 07/28/2008] [Revised: 12/05/2008] [Accepted: 12/07/2008] [Indexed: 12/22/2022]
Abstract
Leptin is a protein hormone that acts within the hypothalamus to suppress food intake and decrease body adiposity, but it is increasingly clear that the hypothalamus is not the only site of leptin action, nor food intake the only biological effect of leptin. Instead, leptin is a pleiotropic hormone that impinges on many brain areas, and in doing so alters food intake, motivation, learning, memory, cognitive function, neuroprotection, reproduction, growth, metabolism, energy expenditure, and more. This diversity of function also means that a dysregulation of leptin secretion and signaling can have far reaching effects. To date research on leptin signaling has focused primarily on the hypothalamus, and the result is a relative lack of information regarding the impact of leptin signaling and leptin resistance in non-hypothalamic areas, despite a growing literature implicating leptin in the regulation of neuronal structure and function in the hippocampus, cortex and other brain areas associated with cognition.
Collapse
|
48
|
Hsuchou H, He Y, Kastin AJ, Tu H, Markadakis EN, Rogers RC, Fossier PB, Pan W. Obesity induces functional astrocytic leptin receptors in hypothalamus. Brain 2009; 132:889-902. [PMID: 19293246 PMCID: PMC2668946 DOI: 10.1093/brain/awp029] [Citation(s) in RCA: 140] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2008] [Revised: 12/31/2008] [Accepted: 01/28/2009] [Indexed: 11/12/2022] Open
Abstract
The possible role of astrocytes in the regulation of feeding has been overlooked. It is well-established that the endothelial cells constituting the blood-brain barrier transport leptin from blood to brain and that hypothalamic neurons respond to leptin to induce anorexic signaling. However, few studies have addressed the role of astrocytes in either leptin transport or cellular activation. We recently showed that the obese agouti viable yellow mouse has prominent astrocytic expression of the leptin receptor. In this study, we test the hypothesis that diet-induced obesity increases astrocytic leptin receptor expression and function in the hypothalamus. Double-labelling immunohistochemistry and confocal microscopic analysis showed that all astrocytes in the hypothalamus express leptin receptors. In adult obese mice, 2 months after being placed on a high-fat diet, there was a striking increase of leptin receptor (+) astrocytes, most prominent in the dorsomedial hypothalamus and arcuate nucleus. Agouti viable yellow mice with their adult-onset obesity showed similar changes, but the increase of leptin receptor (+) astrocytes was barely seen in ob/ob or db/db mice with their early-onset obesity and defective leptin systems. The marked leptin receptor protein expression in the astrocytes, shown with several antibodies against different receptor epitopes, was supported by RT-PCR detection of leptin receptor-a and -b mRNAs in primary hypothalamic astrocytes. Unexpectedly, the protein expression of GFAP, a marker of astrocytes, was also increased in adult-onset obesity. Real-time confocal imaging showed that leptin caused a robust increase of calcium signalling in primary astrocytes from the hypothalamus, confirming their functionality. The results indicate that metabolic changes in obese mice can rapidly alter leptin receptor expression and astrocytic activity, and that leptin receptor is responsible for leptin-induced calcium signalling in astrocytes. This novel and clinically relevant finding opens new avenues in astrocyte biology.
Collapse
Affiliation(s)
- Hung Hsuchou
- Pennington Biomedical Research Center, 6400 Perkins Road, Baton Rouge, LA 70808, USA
| | | | | | | | | | | | | | | |
Collapse
|
49
|
Abstract
Synthesized and released by the adipose tissue, leptin is the widely studied 167-amino acid hormonal protein product of the obesity gene. Originally leptin was defined in association with satiety and energy balance and claimed to be an anti-obesity factor that functioned via a feedback effect from adipocytes to hypothalamus. There is a growing body of evidence that emphasizes the importance of leptin in the regulation of food intake and body weight in animals and humans, alike. Other research findings point out that it plays a role in the regulation of the metabolism, sexual development, reproduction, hematopoiesis, immunity, gastrointestinal functions, sympathetic activation, and angiogenesis. The aim of this review is to evaluate the relation between leptin and the central nervous system (CNS).
Collapse
Affiliation(s)
- Yusuf Ziya Ziylan
- Department of Physiology, Istanbul Medical School, Istanbul University, Capa, Istanbul, Turkey
| | | | | |
Collapse
|
50
|
Solovyova N, Moult PR, Milojkovic B, Lambert JJ, Harvey J. Bi-directional modulation of fast inhibitory synaptic transmission by leptin. J Neurochem 2009; 108:190-201. [PMID: 19094063 PMCID: PMC2605943 DOI: 10.1111/j.1471-4159.2008.05751.x] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
The hormone leptin has widespread actions in the CNS. Indeed, leptin markedly influences hippocampal excitatory synaptic transmission and synaptic plasticity. However, the effects of leptin on fast inhibitory synaptic transmission in the hippocampus have not been evaluated. Here, we show that leptin modulates GABA(A) receptor-mediated synaptic transmission onto hippocampal CA1 pyramidal cells. Leptin promotes a rapid and reversible increase in the amplitude of evoked GABA(A) receptor-mediated inhibitory synaptic currents (IPSCs); an effect that was paralleled by increases in the frequency and amplitude of miniature IPSCs, but with no change in paired pulse ratio or coefficient of variation, suggesting a post-synaptic expression mechanism. Following washout of leptin, a persistent depression (inhibitory long-lasting depression) of evoked IPSCs was observed. Whole-cell dialysis or bath application of inhibitors of phosphoinositide 3 (PI 3)-kinase or Akt prevented leptin-induced enhancement of IPSCs indicating involvement of a post-synaptic PI 3-kinase/Akt-dependent pathway. In contrast, blockade of PI 3-kinase or Akt activity failed to alter the ability of leptin to induce inhibitory long-lasting depression, suggesting that this process is independent of PI 3-kinase/Akt. In conclusion these data indicate that the hormone leptin bi-directionally modulates GABA(A) receptor-mediated synaptic transmission in the hippocampus. These findings have important implications for the role of this hormone in regulating hippocampal pyramidal neuron excitability.
Collapse
Affiliation(s)
- Natasha Solovyova
- Neurosciences Institute, Division of Pathology & Neuroscience, Ninewells Hospital & Medical School, University of Dundee, Dundee, UK
| | | | | | | | | |
Collapse
|