1
|
Li X, Li C, Xue W, Wei Z, Shen H, Wu K, Zhu H, Xu H, Wu X, Yi H, Guan J, Yin S. T266M variants of ANGPTL4 improve lipid metabolism by modifying their binding affinity to acetyl-CoA carboxylase in obstructive sleep apnea. Ann Med 2024; 56:2337740. [PMID: 38574398 PMCID: PMC10997356 DOI: 10.1080/07853890.2024.2337740] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/25/2023] [Accepted: 02/27/2024] [Indexed: 04/06/2024] Open
Abstract
BACKGROUND Angiopoietin-like protein 4 (ANGPTL4) is recognized as a crucial regulator in lipid metabolism. Acetyl-CoA carboxylases (ACACAs) play a role in the β-oxidation of fatty acids. Yet, the functions of ANGPTL4 and ACACA in dyslipidemia of obstructive sleep apnea (OSA) remain unclear. METHODS This study included 125 male OSA subjects from the Shanghai Sleep Health Study (SSHS) who were matched for age, body mass index (BMI), and lipid profile. Serum ANGPTL4 levels were measured via ELISA. The ANGPTL4 T266M variants of 4455 subjects along with their anthropometric, fasting biochemical, and standard polysomnographic parameters were collected. Linear regression was used to analyze the associations between quantitative traits and ANGPTL4 T266M. Molecular docking and molecular dynamic simulation were employed to compare the effects of the wild-type ANGPTL4 and its T266M mutation on ACACA. RESULTS Serum ANGPTL4 levels significantly decreased with increasing OSA severity (non-OSA: 59.6 ± 17.4 ng/mL, mild OSA: 50.0 ± 17.5 ng/mL, moderate OSA: 46.3 ± 15.5 ng/mL, severe OSA: 19.9 ± 14.3 ng/mL, respectively, p = 6.02 × 10-16). No associations were found between T266M and clinical characteristics. Molecular docking indicated that mutant ANGTPL4 T266M had stronger binding affinity for the ACACA protein, compared with wild-type ANGPTL4. In terms of protein secondary structure, mutant ANGTPL4 T266M demonstrated greater stability than wild-type ANGPTL4. CONCLUSIONS Serum ANGTPL4 levels were significantly decreased in OSA patients, particularly among individuals with severe OSA. Although functional ANGTPL4 T266M variants were not associated with lipid levels in OSA, ANGTPL4 T266M could enhance binding affinity for the ACACA protein, potentially regulating lipid metabolism.
Collapse
Affiliation(s)
- Xinyi Li
- Department of Otorhinolaryngology Head and Neck Surgery, Shanghai Sixth People’s Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai Key Laboratory of Sleep Disordered Breathing, Otorhinolaryngology Institute of Shanghai JiaoTong University, Shanghai, China
| | - Chenyang Li
- Department of Otorhinolaryngology Head and Neck Surgery, Shanghai Sixth People’s Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai Key Laboratory of Sleep Disordered Breathing, Otorhinolaryngology Institute of Shanghai JiaoTong University, Shanghai, China
| | - Wenjun Xue
- Department of Otorhinolaryngology Head and Neck surgery, Shanghai Eighth People’s Hospital Affiliated to Jiangsu University, Shanghai, China
| | - Zhicheng Wei
- Department of Otorhinolaryngology Head and Neck Surgery, Shanghai Sixth People’s Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai Key Laboratory of Sleep Disordered Breathing, Otorhinolaryngology Institute of Shanghai JiaoTong University, Shanghai, China
| | - Hangdong Shen
- Department of Otorhinolaryngology Head and Neck Surgery, Shanghai Sixth People’s Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai Key Laboratory of Sleep Disordered Breathing, Otorhinolaryngology Institute of Shanghai JiaoTong University, Shanghai, China
| | - Kejia Wu
- Department of Otorhinolaryngology Head and Neck Surgery, Shanghai Sixth People’s Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai Key Laboratory of Sleep Disordered Breathing, Otorhinolaryngology Institute of Shanghai JiaoTong University, Shanghai, China
| | - Huaming Zhu
- Department of Otorhinolaryngology Head and Neck Surgery, Shanghai Sixth People’s Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai Key Laboratory of Sleep Disordered Breathing, Otorhinolaryngology Institute of Shanghai JiaoTong University, Shanghai, China
| | - Huajun Xu
- Department of Otorhinolaryngology Head and Neck Surgery, Shanghai Sixth People’s Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai Key Laboratory of Sleep Disordered Breathing, Otorhinolaryngology Institute of Shanghai JiaoTong University, Shanghai, China
| | - Xiaolin Wu
- Central Laboratory of Shanghai Eighth People’s Hospital, Xuhui Branch of Shanghai Sixth People’s Hospital, P. R. China
| | - Hongliang Yi
- Department of Otorhinolaryngology Head and Neck Surgery, Shanghai Sixth People’s Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai Key Laboratory of Sleep Disordered Breathing, Otorhinolaryngology Institute of Shanghai JiaoTong University, Shanghai, China
| | - Jian Guan
- Department of Otorhinolaryngology Head and Neck Surgery, Shanghai Sixth People’s Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai Key Laboratory of Sleep Disordered Breathing, Otorhinolaryngology Institute of Shanghai JiaoTong University, Shanghai, China
| | - Shankai Yin
- Department of Otorhinolaryngology Head and Neck Surgery, Shanghai Sixth People’s Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai Key Laboratory of Sleep Disordered Breathing, Otorhinolaryngology Institute of Shanghai JiaoTong University, Shanghai, China
| |
Collapse
|
2
|
Juras JA, Pitra S, Smith BN. Systemic Glucose Regulation by a Hindbrain Inhibitory Circuit in a Mouse Model of Type 1 Diabetes. Neuroendocrinology 2024; 114:302-312. [PMID: 38194945 DOI: 10.1159/000536142] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/14/2023] [Accepted: 01/04/2024] [Indexed: 01/11/2024]
Abstract
INTRODUCTION Previous work showed that increasing the electrical activity of inhibitory neurons in the dorsal vagal complex (DVC) is sufficient to increase whole-body glucose concentration in normoglycemic mice. Here we tested the hypothesis that deactivating GABAergic neurons in the dorsal hindbrain of hyperglycemic mice decreases synaptic inhibition of parasympathetic motor neurons in the dorsal motor nucleus of the vagus (DMV) and reduces systemic glucose levels. METHODS Chemogenetic activation or inactivation of GABAergic neurons in the nucleus tractus solitarius (NTS) was used to assess effects of modulating parasympathetic output on blood glucose concentration in normoglycemic and hyperglycemic mice. Patch-clamp electrophysiology in vitro was used to assess cellular effects of chemogenetic manipulation of NTS GABA neurons. RESULTS Chemogenetic activation of GABAergic NTS neurons in normoglycemic mice increased their action potential firing, resulting in increased inhibitory synaptic input to DMV motor neurons and elevated blood glucose concentration. Deactivation of GABAergic DVC neurons in normoglycemic mice altered their electrical activity but did not alter systemic glucose levels. Conversely, stimulation of GABAergic DVC neurons in mice that were hyperglycemic subsequent to treatment with streptozotocin changed their electrical activity but did not alter whole-body glucose concentration, while deactivation of this inhibitory circuit significantly decreased circulating glucose concentration. Peripheral administration of a brain impermeant muscarinic acetylcholine receptor antagonist abolished these effects. CONCLUSION Disinhibiting vagal motor neurons decreases hyperglycemia in a mouse model of type 1 diabetes. This inhibitory brainstem circuit emerges as a key parasympathetic regulator of whole-body glucose homeostasis that undergoes functional plasticity in hyperglycemic conditions.
Collapse
Affiliation(s)
- J Anna Juras
- Department of Neuroscience, University of Kentucky, Lexington, Kentucky, USA
| | - Soledad Pitra
- Department of Neuroscience, University of Kentucky, Lexington, Kentucky, USA
- Department of Biomedical Sciences, Colorado State University, Fort Collins, Colorado, USA
| | - Bret N Smith
- Department of Neuroscience, University of Kentucky, Lexington, Kentucky, USA
- Department of Biomedical Sciences, Colorado State University, Fort Collins, Colorado, USA
| |
Collapse
|
3
|
Engin A. The Unrestrained Overeating Behavior and Clinical Perspective. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2024; 1460:167-198. [PMID: 39287852 DOI: 10.1007/978-3-031-63657-8_6] [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-related co-morbidities decrease life quality, reduce working ability, and lead to early death. In the adult population, eating addiction manifests with excessive food consumption and the unrestrained overeating behavior, which is associated with increased risk of morbidity and mortality and defined as the binge eating disorder (BED). This hedonic intake is correlated with fat preference and the total amount of dietary fat consumption is the most potent risk factor for weight gain. Long-term BED leads to greater sensitivity to the rewarding effects of palatable foods and results in obesity fatefully. Increased plasma concentrations of non-esterified free fatty acids and lipid-overloaded hypertrophic adipocytes may cause insulin resistance. In addition to dietary intake of high-fat diet, sedentary lifestyle leads to increased storage of triglycerides not only in adipose tissue but also ectopically in other tissues. Lipid-induced apoptosis, ceramide accumulation, reactive oxygen species overproduction, endoplasmic reticulum stress, and mitochondrial dysfunction play role in the pathogenesis of lipotoxicity. Food addiction and BED originate from complex action of dopaminergic, opioid, and cannabinoid systems. BED may also be associated with both obesity and major depressive disorder. For preventing morbidity and mortality, as well as decreasing the impact of obesity-related comorbidities in appropriately selected patients, opiate receptor antagonists and antidepressant combination are recommended. Pharmacotherapy alongside behavioral management improves quality of life and reduces the obesity risk; however, the number of licensed drugs is very few. Thus, stereotactic treatment is recommended to break down the refractory obesity and binge eating in obese patient. As recent applications in the field of non-invasive neuromodulation, transcranial magnetic stimulation and transcranial direct current stimulation are thought to be important in image-guided deep brain stimulation in humans. Chronic overnutrition most likely provides repetitive and persistent signals that up-regulate inhibitor of nuclear factor kappa B (NF-κB) kinase beta subunit/NF-κB (IKKβ/NF-κB) in the hypothalamus before the onset of obesity. However, how the mechanisms of high-fat diet-induced peripheral signals affect the hypothalamic arcuate nucleus remain largely unknown.
Collapse
Affiliation(s)
- 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
|
4
|
Hammerschmidt P, Steculorum SM, Bandet CL, Del Río-Martín A, Steuernagel L, Kohlhaas V, Feldmann M, Varela L, Majcher A, Quatorze Correia M, Klar RFU, Bauder CA, Kaya E, Porniece M, Biglari N, Sieben A, Horvath TL, Hornemann T, Brodesser S, Brüning JC. CerS6-dependent ceramide synthesis in hypothalamic neurons promotes ER/mitochondrial stress and impairs glucose homeostasis in obese mice. Nat Commun 2023; 14:7824. [PMID: 38016943 PMCID: PMC10684560 DOI: 10.1038/s41467-023-42595-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2021] [Accepted: 10/17/2023] [Indexed: 11/30/2023] Open
Abstract
Dysregulation of hypothalamic ceramides has been associated with disrupted neuronal pathways in control of energy and glucose homeostasis. However, the specific ceramide species promoting neuronal lipotoxicity in obesity have remained obscure. Here, we find increased expression of the C16:0 ceramide-producing ceramide synthase (CerS)6 in cultured hypothalamic neurons exposed to palmitate in vitro and in the hypothalamus of obese mice. Conditional deletion of CerS6 in hypothalamic neurons attenuates high-fat diet (HFD)-dependent weight gain and improves glucose metabolism. Specifically, CerS6 deficiency in neurons expressing pro-opiomelanocortin (POMC) or steroidogenic factor 1 (SF-1) alters feeding behavior and alleviates the adverse metabolic effects of HFD feeding on insulin sensitivity and glucose tolerance. POMC-expressing cell-selective deletion of CerS6 prevents the diet-induced alterations of mitochondrial morphology and improves cellular leptin sensitivity. Our experiments reveal functions of CerS6-derived ceramides in hypothalamic lipotoxicity, altered mitochondrial dynamics, and ER/mitochondrial stress in the deregulation of food intake and glucose metabolism in obesity.
Collapse
Affiliation(s)
- Philipp Hammerschmidt
- Department of Neuronal Control of Metabolism, Max Planck Institute for Metabolism Research, Gleueler Strasse 50, 50931, Cologne, Germany
- Policlinic for Endocrinology, Diabetes and Preventive Medicine (PEDP), University Hospital Cologne, Kerpener Strasse 26, 50924, Cologne, Germany
- Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD) and Center for Molecular Medicine Cologne (CMMC), University of Cologne, Cologne, Germany
| | - Sophie M Steculorum
- Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD) and Center for Molecular Medicine Cologne (CMMC), University of Cologne, Cologne, Germany
- Max Planck Institute for Metabolism Research, Research Group Neurocircuit Wiring and Function, Cologne, Germany
- National Center for Diabetes Research (DZD), Ingolstädter Landstrasse 1, 85764, Neuherberg, Germany
| | - Cécile L Bandet
- Department of Neuronal Control of Metabolism, Max Planck Institute for Metabolism Research, Gleueler Strasse 50, 50931, Cologne, Germany
- Policlinic for Endocrinology, Diabetes and Preventive Medicine (PEDP), University Hospital Cologne, Kerpener Strasse 26, 50924, Cologne, Germany
- Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD) and Center for Molecular Medicine Cologne (CMMC), University of Cologne, Cologne, Germany
| | - Almudena Del Río-Martín
- Department of Neuronal Control of Metabolism, Max Planck Institute for Metabolism Research, Gleueler Strasse 50, 50931, Cologne, Germany
- Policlinic for Endocrinology, Diabetes and Preventive Medicine (PEDP), University Hospital Cologne, Kerpener Strasse 26, 50924, Cologne, Germany
- Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD) and Center for Molecular Medicine Cologne (CMMC), University of Cologne, Cologne, Germany
| | - Lukas Steuernagel
- Department of Neuronal Control of Metabolism, Max Planck Institute for Metabolism Research, Gleueler Strasse 50, 50931, Cologne, Germany
- Policlinic for Endocrinology, Diabetes and Preventive Medicine (PEDP), University Hospital Cologne, Kerpener Strasse 26, 50924, Cologne, Germany
- Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD) and Center for Molecular Medicine Cologne (CMMC), University of Cologne, Cologne, Germany
| | - Vivien Kohlhaas
- Department of Neuronal Control of Metabolism, Max Planck Institute for Metabolism Research, Gleueler Strasse 50, 50931, Cologne, Germany
- Policlinic for Endocrinology, Diabetes and Preventive Medicine (PEDP), University Hospital Cologne, Kerpener Strasse 26, 50924, Cologne, Germany
- Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD) and Center for Molecular Medicine Cologne (CMMC), University of Cologne, Cologne, Germany
| | - Marvin Feldmann
- Department of Neuronal Control of Metabolism, Max Planck Institute for Metabolism Research, Gleueler Strasse 50, 50931, Cologne, Germany
- Policlinic for Endocrinology, Diabetes and Preventive Medicine (PEDP), University Hospital Cologne, Kerpener Strasse 26, 50924, Cologne, Germany
- Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD) and Center for Molecular Medicine Cologne (CMMC), University of Cologne, Cologne, Germany
- Faculty of Mathematics and Natural Sciences, University of Cologne, Cologne, Germany
| | - Luis Varela
- Yale Center for Molecular and Systems Metabolism, Department of Comparative Medicine, Yale University School of Medicine, 310 Cedar St., BML 330, New Haven, CT, 06520, USA
- Laboratory of Glia-Neuron Interactions in the Control of Hunger. Achucarro Basque Center for Neuroscience, Leioa, 48940, Spain
- Ikerbasque-Basque Foundation for Science, Bilbao, 48013, Spain
| | - Adam Majcher
- Center for Integrative Human Physiology, University of Zürich, Zürich, Switzerland
- Institute of Clinical Chemistry, University Hospital, Zürich, Switzerland
| | - Marta Quatorze Correia
- Department of Neuronal Control of Metabolism, Max Planck Institute for Metabolism Research, Gleueler Strasse 50, 50931, Cologne, Germany
| | - Rhena F U Klar
- Department of Neuronal Control of Metabolism, Max Planck Institute for Metabolism Research, Gleueler Strasse 50, 50931, Cologne, Germany
| | - Corinna A Bauder
- Department of Neuronal Control of Metabolism, Max Planck Institute for Metabolism Research, Gleueler Strasse 50, 50931, Cologne, Germany
- Policlinic for Endocrinology, Diabetes and Preventive Medicine (PEDP), University Hospital Cologne, Kerpener Strasse 26, 50924, Cologne, Germany
- Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD) and Center for Molecular Medicine Cologne (CMMC), University of Cologne, Cologne, Germany
| | - Ecem Kaya
- Department of Neuronal Control of Metabolism, Max Planck Institute for Metabolism Research, Gleueler Strasse 50, 50931, Cologne, Germany
- Policlinic for Endocrinology, Diabetes and Preventive Medicine (PEDP), University Hospital Cologne, Kerpener Strasse 26, 50924, Cologne, Germany
- Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD) and Center for Molecular Medicine Cologne (CMMC), University of Cologne, Cologne, Germany
| | - Marta Porniece
- Department of Neuronal Control of Metabolism, Max Planck Institute for Metabolism Research, Gleueler Strasse 50, 50931, Cologne, Germany
- Policlinic for Endocrinology, Diabetes and Preventive Medicine (PEDP), University Hospital Cologne, Kerpener Strasse 26, 50924, Cologne, Germany
- Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD) and Center for Molecular Medicine Cologne (CMMC), University of Cologne, Cologne, Germany
| | - Nasim Biglari
- Department of Neuronal Control of Metabolism, Max Planck Institute for Metabolism Research, Gleueler Strasse 50, 50931, Cologne, Germany
- Policlinic for Endocrinology, Diabetes and Preventive Medicine (PEDP), University Hospital Cologne, Kerpener Strasse 26, 50924, Cologne, Germany
- Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD) and Center for Molecular Medicine Cologne (CMMC), University of Cologne, Cologne, Germany
| | - Anna Sieben
- Department of Neuronal Control of Metabolism, Max Planck Institute for Metabolism Research, Gleueler Strasse 50, 50931, Cologne, Germany
- Policlinic for Endocrinology, Diabetes and Preventive Medicine (PEDP), University Hospital Cologne, Kerpener Strasse 26, 50924, Cologne, Germany
- Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD) and Center for Molecular Medicine Cologne (CMMC), University of Cologne, Cologne, Germany
| | - Tamas L Horvath
- Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD) and Center for Molecular Medicine Cologne (CMMC), University of Cologne, Cologne, Germany
- Yale Center for Molecular and Systems Metabolism, Department of Comparative Medicine, Yale University School of Medicine, 310 Cedar St., BML 330, New Haven, CT, 06520, USA
- Laboratory of Glia-Neuron Interactions in the Control of Hunger. Achucarro Basque Center for Neuroscience, Leioa, 48940, Spain
- Ikerbasque-Basque Foundation for Science, Bilbao, 48013, Spain
| | - Thorsten Hornemann
- Center for Integrative Human Physiology, University of Zürich, Zürich, Switzerland
- Institute of Clinical Chemistry, University Hospital, Zürich, Switzerland
| | - Susanne Brodesser
- Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD) and Center for Molecular Medicine Cologne (CMMC), University of Cologne, Cologne, Germany
| | - Jens C Brüning
- Department of Neuronal Control of Metabolism, Max Planck Institute for Metabolism Research, Gleueler Strasse 50, 50931, Cologne, Germany.
- Policlinic for Endocrinology, Diabetes and Preventive Medicine (PEDP), University Hospital Cologne, Kerpener Strasse 26, 50924, Cologne, Germany.
- Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD) and Center for Molecular Medicine Cologne (CMMC), University of Cologne, Cologne, Germany.
- National Center for Diabetes Research (DZD), Ingolstädter Landstrasse 1, 85764, Neuherberg, Germany.
| |
Collapse
|
5
|
Tkáč I, Xie T, Shah N, Larson S, Dubinsky JM, Gomez-Pastor R, McLoughlin HS, Orr HT, Eberly LE, Öz G. Regional sex differences in neurochemical profiles of healthy mice measured by magnetic resonance spectroscopy at 9.4 tesla. Front Neurosci 2023; 17:1278828. [PMID: 37954878 PMCID: PMC10634209 DOI: 10.3389/fnins.2023.1278828] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2023] [Accepted: 10/03/2023] [Indexed: 11/14/2023] Open
Abstract
Objective To determine sex differences in the neurochemical concentrations measured by in vivo proton magnetic resonance spectroscopy (1H MRS) of healthy mice on a genetic background commonly used for neurodegenerative disease models. Methods 1H MRS data collected from wild type mice with C57BL/6 or related genetic backgrounds in seven prior studies were used in this retrospective analysis. To be included, data had to be collected at 9.4 tesla magnetic field using advanced 1H MRS protocols, with isoflurane anesthesia and similar animal handling protocols, and a similar number of datasets from male and female mice had to be available for the brain regions analyzed. Overall, 155 spectra from female mice and 166 spectra from male mice (321 in total), collected from six brain regions (brainstem, cerebellum, cortex, hippocampus, hypothalamus, and striatum) at various ages were included. Results Concentrations of taurine, total creatine (creatine + phosphocreatine), ascorbate, glucose and glutamate were consistently higher in male vs. female mice in most brain regions. Striatum was an exception with similar total creatine in male and female mice. The sex difference pattern in the hypothalamus was notably different from other regions. Interaction between sex and age was significant for total creatine and taurine in the cerebellum and hippocampus. Conclusion Sex differences in regional neurochemical levels are small but significant and age-dependent, with consistent male-female differences across most brain regions. The neuroendocrine region hypothalamus displays a different pattern of sex differences in neurochemical levels. Differences in energy metabolism and cellular density may underlie the differences, with higher metabolic rates in females and higher osmoregulatory and antioxidant capacity in males.
Collapse
Affiliation(s)
- Ivan Tkáč
- Center for Magnetic Resonance Research, Department of Radiology, University of Minnesota, Minneapolis, MN, United States
| | - Tiankai Xie
- Division of Biostatistics, School of Public Health, University of Minnesota, Minneapolis, MN, United States
| | - Nitya Shah
- Division of Biostatistics, School of Public Health, University of Minnesota, Minneapolis, MN, United States
| | - Sarah Larson
- Center for Magnetic Resonance Research, Department of Radiology, University of Minnesota, Minneapolis, MN, United States
| | - Janet M. Dubinsky
- Department of Neuroscience, University of Minnesota, Minneapolis, MN, United States
| | - Rocio Gomez-Pastor
- Department of Neuroscience, University of Minnesota, Minneapolis, MN, United States
| | | | - Harry T. Orr
- Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, MN, United States
| | - Lynn E. Eberly
- Center for Magnetic Resonance Research, Department of Radiology, University of Minnesota, Minneapolis, MN, United States
- Division of Biostatistics, School of Public Health, University of Minnesota, Minneapolis, MN, United States
| | - Gülin Öz
- Center for Magnetic Resonance Research, Department of Radiology, University of Minnesota, Minneapolis, MN, United States
| |
Collapse
|
6
|
Chiang JH, Hua XY, Yu AHM, Peh EWY, See E, Jeyakumar Henry C. A Review on Buckwheat and Its Hypoglycemic Bioactive Components in Food Systems. FOOD REVIEWS INTERNATIONAL 2022. [DOI: 10.1080/87559129.2022.2103706] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Affiliation(s)
- Jie Hong Chiang
- Clinical Nutrition Research Centre, Singapore Institute of Food and Biotechnology Innovation, Agency for Science, Technology and Research (A*STAR), Singapore
| | - Xin Yi Hua
- Clinical Nutrition Research Centre, Singapore Institute of Food and Biotechnology Innovation, Agency for Science, Technology and Research (A*STAR), Singapore
| | - Ashley Hui Min Yu
- Clinical Nutrition Research Centre, Singapore Institute of Food and Biotechnology Innovation, Agency for Science, Technology and Research (A*STAR), Singapore
| | - Elaine Wan Yi Peh
- Clinical Nutrition Research Centre, Singapore Institute of Food and Biotechnology Innovation, Agency for Science, Technology and Research (A*STAR), Singapore
| | - E’Ein See
- Clinical Nutrition Research Centre, Singapore Institute of Food and Biotechnology Innovation, Agency for Science, Technology and Research (A*STAR), Singapore
| | - Christiani Jeyakumar Henry
- Clinical Nutrition Research Centre, Singapore Institute of Food and Biotechnology Innovation, Agency for Science, Technology and Research (A*STAR), Singapore
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| |
Collapse
|
7
|
El Mehdi M, Takhlidjt S, Devère M, Arabo A, Le Solliec MA, Maucotel J, Bénani A, Nedelec E, Duparc C, Lefranc B, Leprince J, Anouar Y, Prévost G, Chartrel N, Picot M. The 26RFa (QRFP)/GPR103 neuropeptidergic system in mice relays insulin signalling into the brain to regulate glucose homeostasis. Diabetologia 2022; 65:1198-1211. [PMID: 35476025 DOI: 10.1007/s00125-022-05706-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/07/2022] [Accepted: 03/21/2022] [Indexed: 11/03/2022]
Abstract
AIMS/HYPOTHESIS 26RFa (pyroglutamilated RFamide peptide [QRFP]) is a biologically active peptide that regulates glucose homeostasis by acting as an incretin and by increasing insulin sensitivity at the periphery. 26RFa is also produced by a neuronal population localised in the hypothalamus. In this study we investigated whether 26RFa neurons are involved in the hypothalamic regulation of glucose homeostasis. METHODS 26Rfa+/+, 26Rfa-/- and insulin-deficient male C57Bl/6J mice were used in this study. Mice received an acute intracerebroventricular (i.c.v.) injection of 26RFa, insulin or the 26RFa receptor (GPR103) antagonist 25e and were subjected to IPGTTs, insulin tolerance tests, acute glucose-stimulated insulin secretion tests and pyruvate tolerance tests (PTTs). Secretion of 26RFa by hypothalamic explants after incubation with glucose, leptin or insulin was assessed. Expression and quantification of the genes encoding 26RFa, agouti-related protein, the insulin receptor and GPR103 were evaluated by quantitative reverse transcription PCR and RNAscope in situ hybridisation. RESULTS Our data indicate that i.c.v.-injected 26RFa induces a robust antihyperglycaemic effect associated with an increase in insulin production by the pancreatic islets. In addition, we found that insulin strongly stimulates 26Rfa expression and secretion by the hypothalamus. RNAscope experiments revealed that neurons expressing 26Rfa are mainly localised in the lateral hypothalamic area, that they co-express the gene encoding the insulin receptor and that insulin induces the expression of 26Rfa in these neurons. Concurrently, the central antihyperglycaemic effect of insulin is abolished in the presence of a GPR103 antagonist and in 26RFa-deficient mice. Finally, our data indicate that the hypothalamic 26RFa neurons are not involved in the central inhibitory effect of insulin on hepatic glucose production, but mediate the central effects of the hormone on its own peripheral production. CONCLUSION/INTERPRETATION We have identified a novel mechanism in the hypothalamic regulation of glucose homeostasis, the 26RFa/GPR103 system, and we provide evidence that this neuronal peptidergic system is a key relay for the central regulation of glucose metabolism by insulin.
Collapse
Affiliation(s)
- Mouna El Mehdi
- Inserm, U1239, Laboratory of Neuronal and Neuroendocrine Differentiation and Communication (DC2N), Institute for Research and Innovation in Biomedicine (IRIB), Normandie Université, UNIROUEN, Rouen, France
| | - Saloua Takhlidjt
- Inserm, U1239, Laboratory of Neuronal and Neuroendocrine Differentiation and Communication (DC2N), Institute for Research and Innovation in Biomedicine (IRIB), Normandie Université, UNIROUEN, Rouen, France
| | - Mélodie Devère
- Inserm, U1239, Laboratory of Neuronal and Neuroendocrine Differentiation and Communication (DC2N), Institute for Research and Innovation in Biomedicine (IRIB), Normandie Université, UNIROUEN, Rouen, France
| | - Arnaud Arabo
- Department of Biological Resources (SRB), Institute for Research and Innovation in Biomedicine (IRIB), Normandie Université, UNIROUEN, Rouen, France
| | - Marie-Anne Le Solliec
- Inserm, U1239, Laboratory of Neuronal and Neuroendocrine Differentiation and Communication (DC2N), Institute for Research and Innovation in Biomedicine (IRIB), Normandie Université, UNIROUEN, Rouen, France
| | - Julie Maucotel
- Department of Biological Resources (SRB), Institute for Research and Innovation in Biomedicine (IRIB), Normandie Université, UNIROUEN, Rouen, France
| | - Alexandre Bénani
- Centre for Taste and Feeding Behaviour, CNRS (UMR6265), INRA (UMR1324), AgroSup Dijon, Université de Bourgogne Franche-Comté, Dijon, France
| | - Emmanuelle Nedelec
- Centre for Taste and Feeding Behaviour, CNRS (UMR6265), INRA (UMR1324), AgroSup Dijon, Université de Bourgogne Franche-Comté, Dijon, France
| | - Céline Duparc
- Inserm, U1239, Laboratory of Neuronal and Neuroendocrine Differentiation and Communication (DC2N), Institute for Research and Innovation in Biomedicine (IRIB), Normandie Université, UNIROUEN, Rouen, France
| | - Benjamin Lefranc
- Inserm, U1239, Laboratory of Neuronal and Neuroendocrine Differentiation and Communication (DC2N), Institute for Research and Innovation in Biomedicine (IRIB), Normandie Université, UNIROUEN, Rouen, France
- Cell Imaging Platform of Normandy, Normandie Université, Rouen, France
| | - Jérôme Leprince
- Inserm, U1239, Laboratory of Neuronal and Neuroendocrine Differentiation and Communication (DC2N), Institute for Research and Innovation in Biomedicine (IRIB), Normandie Université, UNIROUEN, Rouen, France
- Cell Imaging Platform of Normandy, Normandie Université, Rouen, France
| | - Youssef Anouar
- Inserm, U1239, Laboratory of Neuronal and Neuroendocrine Differentiation and Communication (DC2N), Institute for Research and Innovation in Biomedicine (IRIB), Normandie Université, UNIROUEN, Rouen, France
| | - Gaëtan Prévost
- Inserm, U1239, Laboratory of Neuronal and Neuroendocrine Differentiation and Communication (DC2N), Institute for Research and Innovation in Biomedicine (IRIB), Normandie Université, UNIROUEN, Rouen, France
- Department of Endocrinology, Diabetes and Metabolic Diseases, Normandie Université, UNIROUEN, Rouen University Hospital, Rouen, France
| | - Nicolas Chartrel
- Inserm, U1239, Laboratory of Neuronal and Neuroendocrine Differentiation and Communication (DC2N), Institute for Research and Innovation in Biomedicine (IRIB), Normandie Université, UNIROUEN, Rouen, France.
| | - Marie Picot
- Inserm, U1239, Laboratory of Neuronal and Neuroendocrine Differentiation and Communication (DC2N), Institute for Research and Innovation in Biomedicine (IRIB), Normandie Université, UNIROUEN, Rouen, France
| |
Collapse
|
8
|
Torres Irizarry VC, Jiang Y, He Y, Xu P. Hypothalamic Estrogen Signaling and Adipose Tissue Metabolism in Energy Homeostasis. Front Endocrinol (Lausanne) 2022; 13:898139. [PMID: 35757435 PMCID: PMC9218066 DOI: 10.3389/fendo.2022.898139] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/17/2022] [Accepted: 04/29/2022] [Indexed: 11/30/2022] Open
Abstract
Obesity has become a global epidemic, and it is a major risk factor for other metabolic disorders such as type 2 diabetes and cardiometabolic disease. Accumulating evidence indicates that there is sex-specific metabolic protection and disease susceptibility. For instance, in both clinical and experimental studies, males are more likely to develop obesity, insulin resistance, and diabetes. In line with this, males tend to have more visceral white adipose tissue (WAT) and less brown adipose tissue (BAT) thermogenic activity, both leading to an increased incidence of metabolic disorders. This female-specific fat distribution is partially mediated by sex hormone estrogens. Specifically, hypothalamic estrogen signaling plays a vital role in regulating WAT distribution, WAT beiging, and BAT thermogenesis. These regulatory effects on adipose tissue metabolism are primarily mediated by the activation of estrogen receptor alpha (ERα) in neurons, which interacts with hormones and adipokines such as leptin, ghrelin, and insulin. This review discusses the contribution of adipose tissue dysfunction to obesity and the role of hypothalamic estrogen signaling in preventing metabolic diseases with a particular focus on the VMH, the central regulator of energy expenditure and glucose homeostasis.
Collapse
Affiliation(s)
- Valeria C. Torres Irizarry
- Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, The University of Illinois at Chicago, Chicago, IL, United States
- Department of Physiology and Biophysics, The University of Illinois at Chicago, Chicago, IL, United States
| | - Yuwei Jiang
- Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, The University of Illinois at Chicago, Chicago, IL, United States
- Department of Physiology and Biophysics, The University of Illinois at Chicago, Chicago, IL, United States
| | - Yanlin He
- Pennington Biomedical Research Center, Louisiana State University System, Baton Rouge, LA, United States
| | - Pingwen Xu
- Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, The University of Illinois at Chicago, Chicago, IL, United States
- Department of Physiology and Biophysics, The University of Illinois at Chicago, Chicago, IL, United States
| |
Collapse
|
9
|
Piper NBC, Whitfield EA, Stewart GD, Xu X, Furness SGB. Targeting appetite and satiety in diabetes and obesity, via G protein-coupled receptors. Biochem Pharmacol 2022; 202:115115. [PMID: 35671790 DOI: 10.1016/j.bcp.2022.115115] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Revised: 05/26/2022] [Accepted: 05/27/2022] [Indexed: 11/17/2022]
Abstract
Type 2 diabetes and obesity have reached pandemic proportions throughout the world, so much so that the World Health Organisation coined the term "Globesity" to help encapsulate the magnitude of the problem. G protein-coupled receptors (GPCRs) are highly tractable drug targets due to their wide involvement in all aspects of physiology and pathophysiology, indeed, GPCRs are the targets of approximately 30% of the currently approved drugs. GPCRs are also broadly involved in key physiologies that underlie type 2 diabetes and obesity including feeding reward, appetite and satiety, regulation of blood glucose levels, energy homeostasis and adipose function. Despite this, only two GPCRs are the target of approved pharmaceuticals for treatment of type 2 diabetes and obesity. In this review we discuss the role of these, and select other candidate GPCRs, involved in various facets of type 2 diabetic or obese pathophysiology, how they might be targeted and the potential reasons why pharmaceuticals against these targets have not progressed to clinical use. Finally, we provide a perspective on the current development pipeline of anti-obesity drugs that target GPCRs.
Collapse
Affiliation(s)
- Noah B C Piper
- Receptor Transducer Coupling Laboratory, School of Biomedical Sciences, Faculty of Medicine, University of Queensland, St. Lucia, QLD 4072, Australia
| | - Emily A Whitfield
- Receptor Transducer Coupling Laboratory, School of Biomedical Sciences, Faculty of Medicine, University of Queensland, St. Lucia, QLD 4072, Australia
| | - Gregory D Stewart
- Drug Discovery Biology Laboratory, Monash Institute of Pharmaceutical Sciences & Department of Pharmacology Monash University, Parkville, VIC 3052, Australia
| | - Xiaomeng Xu
- Drug Discovery Biology Laboratory, Monash Institute of Pharmaceutical Sciences & Department of Pharmacology Monash University, Parkville, VIC 3052, Australia
| | - Sebastian G B Furness
- Receptor Transducer Coupling Laboratory, School of Biomedical Sciences, Faculty of Medicine, University of Queensland, St. Lucia, QLD 4072, Australia; Drug Discovery Biology Laboratory, Monash Institute of Pharmaceutical Sciences & Department of Pharmacology Monash University, Parkville, VIC 3052, Australia.
| |
Collapse
|
10
|
Matty MA, Lau HE, Haley JA, Singh A, Chakraborty A, Kono K, Reddy KC, Hansen M, Chalasani SH. Intestine-to-neuronal signaling alters risk-taking behaviors in food-deprived Caenorhabditis elegans. PLoS Genet 2022; 18:e1010178. [PMID: 35511794 PMCID: PMC9070953 DOI: 10.1371/journal.pgen.1010178] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Accepted: 03/30/2022] [Indexed: 11/19/2022] Open
Abstract
Animals integrate changes in external and internal environments to generate behavior. While neural circuits detecting external cues have been mapped, less is known about how internal states like hunger are integrated into behavioral outputs. Here, we use the nematode C. elegans to examine how changes in internal nutritional status affect chemosensory behaviors. We show that acute food deprivation leads to a reversible decline in repellent, but not attractant, sensitivity. This behavioral change requires two conserved transcription factors MML-1 (MondoA) and HLH-30 (TFEB), both of which translocate from the intestinal nuclei to the cytoplasm during food deprivation. Next, we identify the insulin-like peptide INS-31 as a candidate ligand relaying food-status signals from the intestine to other tissues. Further, we show that neurons likely use the DAF-2 insulin receptor and AGE-1/PI-3 Kinase, but not DAF-16/FOXO to integrate these intestine-released peptides. Altogether, our study shows how internal food status signals are integrated by transcription factors and intestine-neuron signaling to generate flexible behaviors via the gut-brain axis.
Collapse
Affiliation(s)
- Molly A. Matty
- Molecular Neurobiology Laboratory, The Salk Institute for Biological Studies, La Jolla, California, United States of America
| | - Hiu E. Lau
- Molecular Neurobiology Laboratory, The Salk Institute for Biological Studies, La Jolla, California, United States of America
- Division of Biological Sciences, University of California, San Diego, La Jolla, California, United States of America
| | - Jessica A. Haley
- Molecular Neurobiology Laboratory, The Salk Institute for Biological Studies, La Jolla, California, United States of America
- Neurosciences Graduate Program, University of California, San Diego, La Jolla, California, United States of America
| | - Anupama Singh
- Molecular Neurobiology Laboratory, The Salk Institute for Biological Studies, La Jolla, California, United States of America
- Development, Aging and Regeneration Program, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, California, United States of America
| | - Ahana Chakraborty
- Molecular Neurobiology Laboratory, The Salk Institute for Biological Studies, La Jolla, California, United States of America
| | - Karina Kono
- Molecular Neurobiology Laboratory, The Salk Institute for Biological Studies, La Jolla, California, United States of America
| | - Kirthi C. Reddy
- Molecular Neurobiology Laboratory, The Salk Institute for Biological Studies, La Jolla, California, United States of America
| | - Malene Hansen
- Development, Aging and Regeneration Program, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, California, United States of America
| | - Sreekanth H. Chalasani
- Molecular Neurobiology Laboratory, The Salk Institute for Biological Studies, La Jolla, California, United States of America
| |
Collapse
|
11
|
Tripathi R, Banerjee SK, Nirala JP, Mathur R. Simultaneous exposure to electromagnetic field from mobile phone and unimpeded fructose drinking during pre-, peri-, and post-pubertal stages perturbs the hypothalamic and hepatic regulation of energy homeostasis by early adulthood: experimental evidence. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2022; 29:7438-7451. [PMID: 34476698 DOI: 10.1007/s11356-021-15841-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/25/2021] [Accepted: 08/02/2021] [Indexed: 06/13/2023]
Abstract
The present-day children-adolescents ubiquitously use the mobile phones and unrestrictedly consume fructose-laden diet. Unfortunately, a rise in the incidence of insulin resistance and fatty liver syndrome in young adults has also been recorded. To delineate a possible correlate, the effect of exposure to electromagnetic field (EMF) from the mobile phone and unrestricted fructose intake during pre-, peri-, and post-pubertal stages of development on orexigenic and anorexigenic signals arising from the hypothalamus and liver of rats is investigated here. The study design included four arms, i.e., "Normal", "Exposure Only (ExpO)", "Fructose Only (FruO)", and "Exposure with Fructose (EF)", wherein weaned rats received either "normal chow and drinking water" or "normal chow and fructose (15%) drinking solution" in presence and absence of EMF exposure (2 h/day) for 8 weeks. The results indicate that the total calories consumed by the EF were higher by early adulthood than normal, possibly under the influence of the raised levels of the orexigenic hormone, i.e., ghrelin, and it reflected as raised rate of weight gain. At early adulthood, the EF recorded mitigated response and sensitivity of insulin. Despite EF being a "fed-state", both centrally and peripherally, the glycolysis was restrained, but the gluconeogenesis was raised. Additionally, the altered lipid profile and the glycogen levels indicate that the EF developed fatty liver. The energy homeostasis of the EF was compromised as evidenced by (a) reduced expression of the glucosensors-GLUT2 and glucokinase in the hypothalamus and liver and (b) reduced expression of the cellular energy regulator-AMPK, orexigenic peptide-NPY, and anorexigenic peptide-POMC in the hypothalamus. Taken together, the present study evidences that the exposure to EMFfrom the mobile phone and unrestricted fructose intake during childhood-adolescence impairs the central and peripheral pathways that mediate the glucosensing, glucoregulation, feeding, and satiety behavior by early adulthood.
Collapse
Affiliation(s)
- Ruchi Tripathi
- Department of Pharmacology, Delhi Institute of Pharmaceutical Sciences & Research, New Delhi, India
| | - Sanjay Kumar Banerjee
- Drug Discovery Research Centre, Translational Health Science and Technology Institute, Faridabad, India
- Current Address: Department of Biotechnology, National Institute of Pharmaceutical Education and Research, Guwahati, India
| | - Jay Prakash Nirala
- School of Environmental Sciences, Jawaharlal Nehru University, New Delhi, India
| | - Rajani Mathur
- Department of Pharmacology, Delhi Institute of Pharmaceutical Sciences & Research, New Delhi, India.
| |
Collapse
|
12
|
Zhang L, Xu H, Ding N, Li X, Chen X, Chen Z. Beneficial Effects on Brain Micro-Environment by Caloric Restriction in Alleviating Neurodegenerative Diseases and Brain Aging. Front Physiol 2021; 12:715443. [PMID: 34899367 PMCID: PMC8660583 DOI: 10.3389/fphys.2021.715443] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2021] [Accepted: 10/15/2021] [Indexed: 11/18/2022] Open
Abstract
Aging and neurodegenerative diseases are frequently associated with the disruption of the extracellular microenvironment, which includes mesenchyme and body fluid components. Caloric restriction (CR) has been recognized as a lifestyle intervention that can improve long-term health. In addition to preventing metabolic disorders, CR has been shown to improve brain health owing to its enhancing effect on cognitive functions or retarding effect on the progression of neurodegenerative diseases. This article summarizes current findings regarding the neuroprotective effects of CR, which include the modulation of metabolism, autophagy, oxidative stress, and neuroinflammation. This review may offer future perspectives for brain aging interventions.
Collapse
Affiliation(s)
- Li Zhang
- Key Laboratory of Central CNS Regeneration (Ministry of Education), Guangdong-Hong Kong-Macau Institute of CNS Regeneration, Jinan University, Guangzhou, China
- Center for Brain Science and Brain-Inspired Intelligence, Guangdong-Hong Kong-Macao Greater Bay Area, Guangzhou, China
- Bioland Laboratory, Guangzhou Regenerative Medicine and Health Guangdong Laboratory, Guangzhou, China
- Neuroscience and Neurorehabilitation Institute, University of Health and Rehabilitation Sciences, Qingdao, China
| | - Huachong Xu
- School of Traditional Chinese Medicine, Jinan University, Guangzhou, China
| | - Ning Ding
- Key Laboratory of Central CNS Regeneration (Ministry of Education), Guangdong-Hong Kong-Macau Institute of CNS Regeneration, Jinan University, Guangzhou, China
- Medical College, Kunming University of Science and Technology, Kunming, China
| | - Xue Li
- School of Sports Medicine and Health, Chengdu Sport University, Chengdu, China
| | - Xiaoyin Chen
- School of Traditional Chinese Medicine, Jinan University, Guangzhou, China
| | - Zhuangfei Chen
- Medical College, Kunming University of Science and Technology, Kunming, China
| |
Collapse
|
13
|
Jang JH, Kim HK, Seo DW, Ki SY, Park S, Choi SH, Kim DH, Moon SJ, Jeong YT. Whole-Brain Mapping of the Expression Pattern of T1R2, a Subunit Specific to the Sweet Taste Receptor. Front Neuroanat 2021; 15:751839. [PMID: 34776881 PMCID: PMC8581048 DOI: 10.3389/fnana.2021.751839] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2021] [Accepted: 10/04/2021] [Indexed: 12/29/2022] Open
Abstract
Chemosensory receptors are expressed primarily in sensory organs, but their expression elsewhere can permit ligand detection in other contexts that contribute to survival. The ability of sweet taste receptors to detect natural sugars, sugar alcohols, and artificial sweeteners suggests sweet taste receptors are involved in metabolic regulation in both peripheral organs and in the central nervous system. Our limited knowledge of sweet taste receptor expression in the brain, however, has made it difficult to assess their contribution to metabolic regulation. We, therefore, decided to profile the expression pattern of T1R2, a subunit specific to the sweet taste receptor complex, at the whole-brain level. Using T1r2-Cre knock-in mice, we visualized the overall distribution of Cre-labeled cells in the brain. T1r2-Cre is expressed not only in various populations of neurons, but also in glial populations in the circumventricular organs and in vascular structures in the cortex, thalamus, and striatum. Using immunohistochemistry, we found that T1r2 is expressed in hypothalamic neurons expressing neuropeptide Y and proopiomelanocortin in arcuate nucleus. It is also co-expressed with a canonical taste signaling molecule in perivascular cells of the median eminence. Our findings indicate that sweet taste receptors have unidentified functions in the brain and suggest that they may be a novel therapeutic target in the central nervous system.
Collapse
Affiliation(s)
- Jea Hwa Jang
- BK21 Graduate Program, Department of Biomedical Sciences, Korea University College of Medicine, Seoul, South Korea.,Department of Pharmacology, Korea University College of Medicine, Seoul, South Korea
| | - Ha Kyeong Kim
- BK21 Graduate Program, Department of Biomedical Sciences, Korea University College of Medicine, Seoul, South Korea.,Department of Pharmacology, Korea University College of Medicine, Seoul, South Korea
| | - Dong Woo Seo
- BK21 Graduate Program, Department of Biomedical Sciences, Korea University College of Medicine, Seoul, South Korea.,Department of Pharmacology, Korea University College of Medicine, Seoul, South Korea
| | - Su Young Ki
- Department of Pharmacology, Korea University College of Medicine, Seoul, South Korea
| | - Soonhong Park
- Department of Oral Biology, BK21 PLUS Project, Yonsei University College of Dentistry, Seoul, South Korea
| | - Sang-Hyun Choi
- Department of Pharmacology, Korea University College of Medicine, Seoul, South Korea
| | - Dong-Hoon Kim
- BK21 Graduate Program, Department of Biomedical Sciences, Korea University College of Medicine, Seoul, South Korea.,Department of Pharmacology, Korea University College of Medicine, Seoul, South Korea
| | - Seok Jun Moon
- Department of Oral Biology, BK21 PLUS Project, Yonsei University College of Dentistry, Seoul, South Korea
| | - Yong Taek Jeong
- BK21 Graduate Program, Department of Biomedical Sciences, Korea University College of Medicine, Seoul, South Korea.,Department of Pharmacology, Korea University College of Medicine, Seoul, South Korea
| |
Collapse
|
14
|
Folick A, Koliwad SK, Valdearcos M. Microglial Lipid Biology in the Hypothalamic Regulation of Metabolic Homeostasis. Front Endocrinol (Lausanne) 2021; 12:668396. [PMID: 34122343 PMCID: PMC8191416 DOI: 10.3389/fendo.2021.668396] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/16/2021] [Accepted: 05/05/2021] [Indexed: 12/18/2022] Open
Abstract
In mammals, myeloid cells help maintain the homeostasis of peripheral metabolic tissues, and their immunologic dysregulation contributes to the progression of obesity and associated metabolic disease. There is accumulating evidence that innate immune cells also serve as functional regulators within the mediobasal hypothalamus (MBH), a critical brain region controlling both energy and glucose homeostasis. Specifically, microglia, the resident parenchymal myeloid cells of the CNS, play important roles in brain physiology and pathology. Recent studies have revealed an expanding array of microglial functions beyond their established roles as immune sentinels, including roles in brain development, circuit refinement, and synaptic organization. We showed that microglia modulate MBH function by transmitting information resulting from excess nutrient consumption. For instance, microglia can sense the excessive consumption of saturated fats and instruct neurons within the MBH accordingly, leading to responsive alterations in energy balance. Interestingly, the recent emergence of high-resolution single-cell techniques has enabled specific microglial populations and phenotypes to be profiled in unprecedented detail. Such techniques have highlighted specific subsets of microglia notable for their capacity to regulate the expression of lipid metabolic genes, including lipoprotein lipase (LPL), apolipoprotein E (APOE) and Triggering Receptor Expressed on Myeloid Cells 2 (TREM2). The discovery of this transcriptional signature highlights microglial lipid metabolism as a determinant of brain health and disease pathogenesis, with intriguing implications for the treatment of brain disorders and potentially metabolic disease. Here we review our current understanding of how changes in microglial lipid metabolism could influence the hypothalamic control of systemic metabolism.
Collapse
Affiliation(s)
- Andrew Folick
- Diabetes Center, University of California, San Francisco, San Francisco, CA, United States
- Department of Medicine, University of California, San Francisco, San Francisco, CA, United States
| | - Suneil K. Koliwad
- Diabetes Center, University of California, San Francisco, San Francisco, CA, United States
- Department of Medicine, University of California, San Francisco, San Francisco, CA, United States
| | - Martin Valdearcos
- Diabetes Center, University of California, San Francisco, San Francisco, CA, United States
| |
Collapse
|
15
|
Abstract
Energy balance is centrally regulated by the brain through several interacting neuronal systems involving external, peripheral, and central factors within the brain. The hypothalamus integrates these factors and is the key brain area in the regulation of energy balance. In this review, we will explain the structure of the hypothalamus and its role in the regulation of energy balance. An important part of energy balance regulation is the sensing of nutrient status and availability. This review will focus on the sensing of the two main sources of energy by the hypothalamus: glucose and fat. As many common health problems and chronic diseases can be traced back to a disrupted hypothalamic function, we will also discuss hypothalamic sensing of glucose and fats in these pathologies. Finally, we will summarize the current knowledge and discuss how this may be applied clinically and for future research perspectives.
Collapse
|
16
|
Rasal KD, Iquebal MA, Dixit S, Vasam M, Raza M, Sahoo L, Jaiswal S, Nandi S, Mahapatra KD, Rasal A, Udit UK, Meher PK, Murmu K, Angadi UB, Rai A, Kumar D, Sundaray JK. Revealing Alteration in the Hepatic Glucose Metabolism of Genetically Improved Carp, Jayanti Rohu Labeo rohita Fed a High Carbohydrate Diet Using Transcriptome Sequencing. Int J Mol Sci 2020; 21:E8180. [PMID: 33142948 PMCID: PMC7662834 DOI: 10.3390/ijms21218180] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2020] [Revised: 10/20/2020] [Accepted: 10/21/2020] [Indexed: 01/25/2023] Open
Abstract
Although feed cost is the greatest concern in aquaculture, the inclusion of carbohydrates in the fish diet, and their assimilation, are still not well understood in aquaculture species. We identified molecular events that occur due to the inclusion of high carbohydrate levels in the diets of genetically improved 'Jayanti rohu' Labeo rohita. To reveal transcriptional changes in the liver of rohu, a feeding experiment was conducted with three doses of gelatinized starch (20% (control), 40%, and 60%). Transcriptome sequencing revealed totals of 15,232 (4464 up- and 4343 down-regulated) and 15,360 (4478 up- and 4171 down-regulated) differentially expressed genes. Up-regulated transcripts associated with glucose metabolisms, such as hexokinase, PHK, glycogen synthase and PGK, were found in fish fed diets with high starch levels. Interestingly, a de novo lipogenesis mechanism was found to be enriched in the livers of treated fish due to up-regulated transcripts such as FAS, ACCα, and PPARγ. The insulin signaling pathways with enriched PPAR and mTOR were identified by Kyoto Encyclopedia of Genes and Genome (KEGG) as a result of high carbohydrates. This work revealed for the first time the atypical regulation transcripts associated with glucose metabolism and lipogenesis in the livers of Jayanti rohu due to the inclusion of high carbohydrate levels in the diet. This study also encourages the exploration of early nutritional programming for enhancing glucose efficiency in carp species, for sustainable and cost-effective aquaculture production.
Collapse
Affiliation(s)
- Kiran D. Rasal
- Fish Genetics and Biotechnology Division, ICAR-Central Institute of Freshwater Aquaculture, Bhubaneswar 751 002, India; (K.D.R.); (S.D.); (M.V.); (L.S.); (S.N.); (K.D.M.); (A.R.); (U.K.U.); (P.K.M.); (K.M.)
| | - Mir Asif Iquebal
- Centre for Agricultural Bioinformatics (CABin), ICAR-Indian Agricultural Statistics Research Institute, Library Avenue, PUSA, New Delhi 110012, India; (M.A.I.); (M.R.); (S.J.); (U.A.); (A.R.); (D.K.)
| | - Sangita Dixit
- Fish Genetics and Biotechnology Division, ICAR-Central Institute of Freshwater Aquaculture, Bhubaneswar 751 002, India; (K.D.R.); (S.D.); (M.V.); (L.S.); (S.N.); (K.D.M.); (A.R.); (U.K.U.); (P.K.M.); (K.M.)
| | - Manohar Vasam
- Fish Genetics and Biotechnology Division, ICAR-Central Institute of Freshwater Aquaculture, Bhubaneswar 751 002, India; (K.D.R.); (S.D.); (M.V.); (L.S.); (S.N.); (K.D.M.); (A.R.); (U.K.U.); (P.K.M.); (K.M.)
| | - Mustafa Raza
- Centre for Agricultural Bioinformatics (CABin), ICAR-Indian Agricultural Statistics Research Institute, Library Avenue, PUSA, New Delhi 110012, India; (M.A.I.); (M.R.); (S.J.); (U.A.); (A.R.); (D.K.)
| | - Lakshman Sahoo
- Fish Genetics and Biotechnology Division, ICAR-Central Institute of Freshwater Aquaculture, Bhubaneswar 751 002, India; (K.D.R.); (S.D.); (M.V.); (L.S.); (S.N.); (K.D.M.); (A.R.); (U.K.U.); (P.K.M.); (K.M.)
| | - Sarika Jaiswal
- Centre for Agricultural Bioinformatics (CABin), ICAR-Indian Agricultural Statistics Research Institute, Library Avenue, PUSA, New Delhi 110012, India; (M.A.I.); (M.R.); (S.J.); (U.A.); (A.R.); (D.K.)
| | - Samiran Nandi
- Fish Genetics and Biotechnology Division, ICAR-Central Institute of Freshwater Aquaculture, Bhubaneswar 751 002, India; (K.D.R.); (S.D.); (M.V.); (L.S.); (S.N.); (K.D.M.); (A.R.); (U.K.U.); (P.K.M.); (K.M.)
| | - Kanta Das Mahapatra
- Fish Genetics and Biotechnology Division, ICAR-Central Institute of Freshwater Aquaculture, Bhubaneswar 751 002, India; (K.D.R.); (S.D.); (M.V.); (L.S.); (S.N.); (K.D.M.); (A.R.); (U.K.U.); (P.K.M.); (K.M.)
| | - Avinash Rasal
- Fish Genetics and Biotechnology Division, ICAR-Central Institute of Freshwater Aquaculture, Bhubaneswar 751 002, India; (K.D.R.); (S.D.); (M.V.); (L.S.); (S.N.); (K.D.M.); (A.R.); (U.K.U.); (P.K.M.); (K.M.)
| | - Uday Kumar Udit
- Fish Genetics and Biotechnology Division, ICAR-Central Institute of Freshwater Aquaculture, Bhubaneswar 751 002, India; (K.D.R.); (S.D.); (M.V.); (L.S.); (S.N.); (K.D.M.); (A.R.); (U.K.U.); (P.K.M.); (K.M.)
| | - Prem Kumar Meher
- Fish Genetics and Biotechnology Division, ICAR-Central Institute of Freshwater Aquaculture, Bhubaneswar 751 002, India; (K.D.R.); (S.D.); (M.V.); (L.S.); (S.N.); (K.D.M.); (A.R.); (U.K.U.); (P.K.M.); (K.M.)
| | - Khuntia Murmu
- Fish Genetics and Biotechnology Division, ICAR-Central Institute of Freshwater Aquaculture, Bhubaneswar 751 002, India; (K.D.R.); (S.D.); (M.V.); (L.S.); (S.N.); (K.D.M.); (A.R.); (U.K.U.); (P.K.M.); (K.M.)
| | - UB Angadi
- Centre for Agricultural Bioinformatics (CABin), ICAR-Indian Agricultural Statistics Research Institute, Library Avenue, PUSA, New Delhi 110012, India; (M.A.I.); (M.R.); (S.J.); (U.A.); (A.R.); (D.K.)
| | - Anil Rai
- Centre for Agricultural Bioinformatics (CABin), ICAR-Indian Agricultural Statistics Research Institute, Library Avenue, PUSA, New Delhi 110012, India; (M.A.I.); (M.R.); (S.J.); (U.A.); (A.R.); (D.K.)
| | - Dinesh Kumar
- Centre for Agricultural Bioinformatics (CABin), ICAR-Indian Agricultural Statistics Research Institute, Library Avenue, PUSA, New Delhi 110012, India; (M.A.I.); (M.R.); (S.J.); (U.A.); (A.R.); (D.K.)
| | - Jitendra Kumar Sundaray
- Fish Genetics and Biotechnology Division, ICAR-Central Institute of Freshwater Aquaculture, Bhubaneswar 751 002, India; (K.D.R.); (S.D.); (M.V.); (L.S.); (S.N.); (K.D.M.); (A.R.); (U.K.U.); (P.K.M.); (K.M.)
| |
Collapse
|
17
|
Sohn JW, Ho WK. Cellular and systemic mechanisms for glucose sensing and homeostasis. Pflugers Arch 2020; 472:1547-1561. [PMID: 32960363 DOI: 10.1007/s00424-020-02466-2] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2020] [Revised: 08/14/2020] [Accepted: 09/14/2020] [Indexed: 12/25/2022]
Abstract
Glucose is a major source of energy in animals. Maintaining blood glucose levels within a physiological range is important for facilitating glucose uptake by cells, as required for optimal functioning. Glucose homeostasis relies on multiple glucose-sensing cells in the body that constantly monitor blood glucose levels and respond accordingly to adjust its glycemia. These include not only pancreatic β-cells and α-cells that secrete insulin and glucagon, but also central and peripheral neurons regulating pancreatic endocrine function. Different types of cells respond distinctively to changes in blood glucose levels, and the mechanisms involved in glucose sensing are diverse. Notably, recent studies have challenged the currently held views regarding glucose-sensing mechanisms. Furthermore, peripheral and central glucose-sensing cells appear to work in concert to control blood glucose level and maintain glucose and energy homeostasis in organisms. In this review, we summarize the established concepts and recent advances in the understanding of cellular and systemic mechanisms that regulate glucose sensing and its homeostasis.
Collapse
Affiliation(s)
- Jong-Woo Sohn
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology, 291 Daehak-ro, Yuseong-gu, Daejeon, 34141, South Korea.
| | - Won-Kyung Ho
- Department of Physiology, Seoul National University College of Medicine, 103 Daehak-ro, Jongro-gu, Seoul, 03080, South Korea.
- Department of Brain and Cognitive Sciences, Seoul National University College of Natural Sciences, 1 Gwanak-ro, Gwanak-gu, Seoul, 08826, South Korea.
| |
Collapse
|
18
|
Kundap UP, Paudel YN, Shaikh MF. Animal Models of Metabolic Epilepsy and Epilepsy Associated Metabolic Dysfunction: A Systematic Review. Pharmaceuticals (Basel) 2020; 13:ph13060106. [PMID: 32466498 PMCID: PMC7345684 DOI: 10.3390/ph13060106] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2020] [Revised: 05/22/2020] [Accepted: 05/23/2020] [Indexed: 12/13/2022] Open
Abstract
Epilepsy is a serious neurological disorder affecting around 70 million people globally and is characterized by spontaneous recurrent seizures. Recent evidence indicates that dysfunction in metabolic processes can lead to the alteration of neuronal and network excitability, thereby contributing to epileptogenesis. Developing a suitable animal model that can recapitulate all the clinical phenotypes of human metabolic epilepsy (ME) is crucial yet challenging. The specific environment of many symptoms as well as the primary state of the applicable neurobiology, genetics, and lack of valid biomarkers/diagnostic tests are the key factors that hinder the process of developing a suitable animal model. The present systematic review summarizes the current state of available animal models of metabolic dysfunction associated with epileptic disorders. A systematic search was performed by using the Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) model. A range of electronic databases, including google scholar, Springer, PubMed, ScienceDirect, and Scopus, were scanned between January 2000 and April 2020. Based on the selection criteria, 23 eligible articles were chosen and are discussed in the current review. Critical analysis of the selected literature delineated several available approaches that have been modeled into metabolic epilepsy and pointed out several drawbacks associated with the currently available models. The result describes available models of metabolic dysfunction associated with epileptic disorder, such as mitochondrial respiration deficits, Lafora disease (LD) model-altered glycogen metabolism, causing epilepsy, glucose transporter 1 (GLUT1) deficiency, adiponectin responsive seizures, phospholipid dysfunction, glutaric aciduria, mitochondrial disorders, pyruvate dehydrogenase (PDH) α-subunit gene (PDHA1), pyridoxine dependent epilepsy (PDE), BCL2-associated agonist of cell death (BAD), Kcna1 knock out (KO), and long noncoding RNAs (lncRNA) cancer susceptibility candidate 2 (lncRNA CASC2). Finally, the review highlights certain focus areas that may increase the possibilities of developing more suitable animal models and underscores the importance of the rationalization of animal models and evaluation methods for studying ME. The review also suggests the pressing need of developing precise robust animal models and evaluation methods for investigating ME.
Collapse
Affiliation(s)
- Uday Praful Kundap
- Research Center of the University of Montreal Hospital Center (CRCHUM), Department of Neurosciences, Université de Montréal, Montréal, QC H2X 0A9, Canada; (U.P.K.); (Y.N.P.)
- Neuropharmacology Research Strength, Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Selangor 47500, Malaysia
| | - Yam Nath Paudel
- Research Center of the University of Montreal Hospital Center (CRCHUM), Department of Neurosciences, Université de Montréal, Montréal, QC H2X 0A9, Canada; (U.P.K.); (Y.N.P.)
| | - Mohd. Farooq Shaikh
- Neuropharmacology Research Strength, Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Selangor 47500, Malaysia
- Correspondence: ; Tel.: +60-3-551-44-483
| |
Collapse
|
19
|
Tobore TO. Towards a comprehensive theory of obesity and a healthy diet: The causal role of oxidative stress in food addiction and obesity. Behav Brain Res 2020; 384:112560. [DOI: 10.1016/j.bbr.2020.112560] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2019] [Revised: 02/14/2020] [Accepted: 02/14/2020] [Indexed: 02/06/2023]
|
20
|
Garcia-Serrano AM, Duarte JMN. Brain Metabolism Alterations in Type 2 Diabetes: What Did We Learn From Diet-Induced Diabetes Models? Front Neurosci 2020; 14:229. [PMID: 32265637 PMCID: PMC7101159 DOI: 10.3389/fnins.2020.00229] [Citation(s) in RCA: 50] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2019] [Accepted: 03/02/2020] [Indexed: 12/27/2022] Open
Abstract
Type 2 diabetes (T2D) is a metabolic disease with impact on brain function through mechanisms that include glucose toxicity, vascular damage and blood–brain barrier (BBB) impairments, mitochondrial dysfunction, oxidative stress, brain insulin resistance, synaptic failure, neuroinflammation, and gliosis. Rodent models have been developed for investigating T2D, and have contributed to our understanding of mechanisms involved in T2D-induced brain dysfunction. Namely, mice or rats exposed to diabetogenic diets that are rich in fat and/or sugar have been widely used since they develop memory impairment, especially in tasks that depend on hippocampal processing. Here we summarize main findings on brain energy metabolism alterations underlying dysfunction of neuronal and glial cells promoted by diet-induced metabolic syndrome that progresses to a T2D phenotype.
Collapse
Affiliation(s)
- Alba M Garcia-Serrano
- Department of Experimental Medical Science, Faculty of Medicine, Lund University, Lund, Sweden.,Wallenberg Centre for Molecular Medicine, Faculty of Medicine, Lund University, Lund, Sweden
| | - João M N Duarte
- Department of Experimental Medical Science, Faculty of Medicine, Lund University, Lund, Sweden.,Wallenberg Centre for Molecular Medicine, Faculty of Medicine, Lund University, Lund, Sweden
| |
Collapse
|
21
|
Zanos TP. Recording and Decoding of Vagal Neural Signals Related to Changes in Physiological Parameters and Biomarkers of Disease. Cold Spring Harb Perspect Med 2019; 9:a034157. [PMID: 30670469 PMCID: PMC6886457 DOI: 10.1101/cshperspect.a034157] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Our bodies have built-in neural reflexes that continuously monitor organ function and maintain physiological homeostasis. Whereas the field of bioelectronic medicine has mainly focused on the stimulation of neural circuits to treat various conditions, recent studies have started to investigate the possibility of leveraging the sensory arm of these reflexes to diagnose disease states. To accomplish this, neural signals emanating from the body's built-in biosensors and propagating through peripheral nerves must be recorded and decoded to identify the presence or levels of relevant biomarkers of disease. The process of acquiring these signals poses several technical challenges related to the neural interfaces, surgical techniques, and data-processing framework needed to record and analyze them. However, these challenges can be addressed with a rigorous experimental approach and new advances in implantable electrodes, signal processing, and machine learning methods. Outlined in this review are studies decoding vagus nerve activity as it related to inflammatory, metabolic, and cardiopulmonary biomarkers. Successfully decoding peripheral nerve activity related to disease states will not only enable the development of real-time diagnostic devices, but also help advancing truly closed-loop neuromodulation technologies.
Collapse
Affiliation(s)
- Theodoros P Zanos
- Center for Bioelectronic Medicine, The Feinstein Institute for Medical Research, Donald & Barbara Zucker School of Medicine at Hofstra/Northwell, Manhasset, New York 11030
| |
Collapse
|
22
|
Masi EB, Levy T, Tsaava T, Bouton CE, Tracey KJ, Chavan SS, Zanos TP. Identification of hypoglycemia-specific neural signals by decoding murine vagus nerve activity. Bioelectron Med 2019; 5:9. [PMID: 32232099 PMCID: PMC7098244 DOI: 10.1186/s42234-019-0025-z] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2019] [Accepted: 06/06/2019] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Glucose is a crucial energy source. In humans, it is the primary sugar for high energy demanding cells in brain, muscle and peripheral neurons. Deviations of blood glucose levels from normal levels for an extended period of time is dangerous or even fatal, so regulation of blood glucose levels is a biological imperative. The vagus nerve, comprised of sensory and motor fibres, provides a major anatomical substrate for regulating metabolism. While prior studies have implicated the vagus nerve in the neurometabolic interface, its specific role in either the afferent or efferent arc of this reflex remains elusive. METHODS Here we use recently developed methods to isolate and decode specific neural signals acquired from the surface of the vagus nerve in BALB/c wild type mice to identify those that respond robustly to hypoglycemia. We also attempted to decode neural signals related to hyperglycemia. In addition to wild type mice, we analyzed the responses to acute hypo- and hyperglycemia in transient receptor potential cation channel subfamily V member 1 (TRPV1) cell depleted mice. The decoding algorithm uses neural signals as input and reconstructs blood glucose levels. RESULTS Our algorithm was able to reconstruct the blood glucose levels with high accuracy (median error 18.6 mg/dl). Hyperglycemia did not induce robust vagus nerve responses, and deletion of TRPV1 nociceptors attenuated the hypoglycemia-dependent vagus nerve signals. CONCLUSION These results provide insight to the sensory vagal signaling that encodes hypoglycemic states and suggest a method to measure blood glucose levels by decoding nerve signals. TRIAL REGISTRATION Not applicable.
Collapse
Affiliation(s)
| | - Todd Levy
- 2Institute of Bioelectronic Medicine, Feinstein Institute for Medical Research, Manhasset, NY 11030 USA
| | - Tea Tsaava
- 2Institute of Bioelectronic Medicine, Feinstein Institute for Medical Research, Manhasset, NY 11030 USA
| | - Chad E Bouton
- 2Institute of Bioelectronic Medicine, Feinstein Institute for Medical Research, Manhasset, NY 11030 USA
| | - Kevin J Tracey
- Zucker School of Medicine at Hofstra/Northwell, Heampstead, NY USA
- 2Institute of Bioelectronic Medicine, Feinstein Institute for Medical Research, Manhasset, NY 11030 USA
| | - Sangeeta S Chavan
- Zucker School of Medicine at Hofstra/Northwell, Heampstead, NY USA
- 2Institute of Bioelectronic Medicine, Feinstein Institute for Medical Research, Manhasset, NY 11030 USA
| | - Theodoros P Zanos
- Zucker School of Medicine at Hofstra/Northwell, Heampstead, NY USA
- 2Institute of Bioelectronic Medicine, Feinstein Institute for Medical Research, Manhasset, NY 11030 USA
| |
Collapse
|
23
|
Rege SD, Royes L, Tsai B, Zhang G, Yang X, Gomez-Pinilla F. Brain Trauma Disrupts Hepatic Lipid Metabolism: Blame It on Fructose? Mol Nutr Food Res 2019; 63:e1801054. [PMID: 31087499 DOI: 10.1002/mnfr.201801054] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2018] [Revised: 03/21/2019] [Indexed: 02/06/2023]
Abstract
SCOPE The action of brain disorders on peripheral metabolism is poorly understood. The impact of traumatic brain injury (TBI) on peripheral organ function and how TBI effects can be influenced by the metabolic perturbation elicited by fructose ingestion are studied. METHODS AND RESULTS It is found that TBI affects glucose metabolism and signaling proteins for insulin and growth hormone in the liver; these effects are exacerbated by fructose ingestion. Fructose, principally metabolized in the liver, potentiates the action of TBI on hepatic lipid droplet accumulation. Studies in isolated cultured hepatocytes identify GH and fructose as factors for the synthesis of lipids. The liver has a major role in the synthesis of lipids used for brain function and repair. TBI results in differentially expressed genes in the hypothalamus, primarily associated with lipid metabolism, providing cues to understand central control of peripheral alterations. Fructose-fed TBI animals have elevated levels of markers of inflammation, lipid peroxidation, and cell energy metabolism, suggesting the pro-inflammatory impact of TBI and fructose in the liver. CONCLUSION Results reveal the impact of TBI on systemic metabolism and the aggravating action of fructose. The hypothalamic-pituitary-growth axis seems to play a major role in the regulation of the peripheral TBI pathology.
Collapse
Affiliation(s)
- Shraddha D Rege
- Department of Integrative Biology & Physiology, University of California, Los Angeles, Los Angeles, CA, 90095, USA
| | - Luiz Royes
- Department of Integrative Biology & Physiology, University of California, Los Angeles, Los Angeles, CA, 90095, USA.,Centro De Educacao Fisica e Desportos, Universidade Federal de Santa Maria, Santa Maria, Rio Grande do Sul, 97105, Brazil
| | - Brandon Tsai
- Department of Integrative Biology & Physiology, University of California, Los Angeles, Los Angeles, CA, 90095, USA
| | - Guanglin Zhang
- Department of Integrative Biology & Physiology, University of California, Los Angeles, Los Angeles, CA, 90095, USA
| | - Xia Yang
- Department of Integrative Biology & Physiology, University of California, Los Angeles, Los Angeles, CA, 90095, USA
| | - Fernando Gomez-Pinilla
- Department of Integrative Biology & Physiology, University of California, Los Angeles, Los Angeles, CA, 90095, USA.,Department of Neurosurgery, UCLA Brain Injury Research Center, University of California, Los Angeles, Los Angeles, CA, 90095, USA
| |
Collapse
|
24
|
Fuente-Martín E, Mellado-Gil JM, Cobo-Vuilleumier N, Martín-Montalvo A, Romero-Zerbo SY, Diaz Contreras I, Hmadcha A, Soria B, Martin Bermudo F, Reyes JC, Bermúdez-Silva FJ, Lorenzo PI, Gauthier BR. Dissecting the Brain/Islet Axis in Metabesity. Genes (Basel) 2019; 10:genes10050350. [PMID: 31072002 PMCID: PMC6562925 DOI: 10.3390/genes10050350] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2019] [Revised: 05/02/2019] [Accepted: 05/02/2019] [Indexed: 12/17/2022] Open
Abstract
The high prevalence of type 2 diabetes mellitus (T2DM), together with the fact that current treatments are only palliative and do not avoid major secondary complications, reveals the need for novel approaches to treat the cause of this disease. Efforts are currently underway to identify therapeutic targets implicated in either the regeneration or re-differentiation of a functional pancreatic islet β-cell mass to restore insulin levels and normoglycemia. However, T2DM is not only caused by failures in β-cells but also by dysfunctions in the central nervous system (CNS), especially in the hypothalamus and brainstem. Herein, we review the physiological contribution of hypothalamic neuronal and glial populations, particularly astrocytes, in the control of the systemic response that regulates blood glucose levels. The glucosensing capacity of hypothalamic astrocytes, together with their regulation by metabolic hormones, highlights the relevance of these cells in the control of glucose homeostasis. Moreover, the critical role of astrocytes in the response to inflammation, a process associated with obesity and T2DM, further emphasizes the importance of these cells as novel targets to stimulate the CNS in response to metabesity (over-nutrition-derived metabolic dysfunctions). We suggest that novel T2DM therapies should aim at stimulating the CNS astrocytic response, as well as recovering the functional pancreatic β-cell mass. Whether or not a common factor expressed in both cell types can be feasibly targeted is also discussed.
Collapse
Affiliation(s)
- Esther Fuente-Martín
- Andalusian Center of Molecular Biology and Regenerative Medicine-CABIMER, Junta de Andalucia-University of Pablo de Olavide-University of Seville-CSIC, 41092 Seville, Spain.
| | - Jose M Mellado-Gil
- Andalusian Center of Molecular Biology and Regenerative Medicine-CABIMER, Junta de Andalucia-University of Pablo de Olavide-University of Seville-CSIC, 41092 Seville, Spain.
| | - Nadia Cobo-Vuilleumier
- Andalusian Center of Molecular Biology and Regenerative Medicine-CABIMER, Junta de Andalucia-University of Pablo de Olavide-University of Seville-CSIC, 41092 Seville, Spain.
| | - Alejandro Martín-Montalvo
- Andalusian Center of Molecular Biology and Regenerative Medicine-CABIMER, Junta de Andalucia-University of Pablo de Olavide-University of Seville-CSIC, 41092 Seville, Spain.
| | - Silvana Y Romero-Zerbo
- Instituto de Investigación Biomédica de Málaga-IBIMA, UGC Endocrinología y Nutrición. Hospital Regional Universitario de Málaga, 29009 Málaga, Spain.
| | - Irene Diaz Contreras
- Andalusian Center of Molecular Biology and Regenerative Medicine-CABIMER, Junta de Andalucia-University of Pablo de Olavide-University of Seville-CSIC, 41092 Seville, Spain.
- Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), 28029 Madrid, Spain.
| | - Abdelkrim Hmadcha
- Andalusian Center of Molecular Biology and Regenerative Medicine-CABIMER, Junta de Andalucia-University of Pablo de Olavide-University of Seville-CSIC, 41092 Seville, Spain.
- Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), 28029 Madrid, Spain.
| | - Bernat Soria
- Andalusian Center of Molecular Biology and Regenerative Medicine-CABIMER, Junta de Andalucia-University of Pablo de Olavide-University of Seville-CSIC, 41092 Seville, Spain.
- Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), 28029 Madrid, Spain.
| | - Francisco Martin Bermudo
- Andalusian Center of Molecular Biology and Regenerative Medicine-CABIMER, Junta de Andalucia-University of Pablo de Olavide-University of Seville-CSIC, 41092 Seville, Spain.
- Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), 28029 Madrid, Spain.
| | - Jose C Reyes
- Andalusian Center of Molecular Biology and Regenerative Medicine-CABIMER, Junta de Andalucia-University of Pablo de Olavide-University of Seville-CSIC, 41092 Seville, Spain.
| | - Francisco J Bermúdez-Silva
- Instituto de Investigación Biomédica de Málaga-IBIMA, UGC Endocrinología y Nutrición. Hospital Regional Universitario de Málaga, 29009 Málaga, Spain.
- Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), 28029 Madrid, Spain.
| | - Petra I Lorenzo
- Andalusian Center of Molecular Biology and Regenerative Medicine-CABIMER, Junta de Andalucia-University of Pablo de Olavide-University of Seville-CSIC, 41092 Seville, Spain.
| | - Benoit R Gauthier
- Andalusian Center of Molecular Biology and Regenerative Medicine-CABIMER, Junta de Andalucia-University of Pablo de Olavide-University of Seville-CSIC, 41092 Seville, Spain.
- Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), 28029 Madrid, Spain.
| |
Collapse
|
25
|
Matschinsky FM, Wilson DF. The Central Role of Glucokinase in Glucose Homeostasis: A Perspective 50 Years After Demonstrating the Presence of the Enzyme in Islets of Langerhans. Front Physiol 2019; 10:148. [PMID: 30949058 PMCID: PMC6435959 DOI: 10.3389/fphys.2019.00148] [Citation(s) in RCA: 188] [Impact Index Per Article: 31.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2018] [Accepted: 02/07/2019] [Indexed: 01/05/2023] Open
Abstract
It is hypothesized that glucokinase (GCK) is the glucose sensor not only for regulation of insulin release by pancreatic β-cells, but also for the rest of the cells that contribute to glucose homeostasis in mammals. This includes other cells in endocrine pancreas (α- and δ-cells), adrenal gland, glucose sensitive neurons, entero-endocrine cells, and cells in the anterior pituitary. Glucose transport is by facilitated diffusion and is not rate limiting. Once inside, glucose is phosphorylated to glucose-6-phosphate by GCK in a reaction that is dependent on glucose throughout the physiological range of concentrations, is irreversible, and not product inhibited. High glycerol phosphate shuttle, pyruvate dehydrogenase, and pyruvate carboxylase activities, combined with low pentose-P shunt, lactate dehydrogenase, plasma membrane monocarboxylate transport, and glycogen synthase activities constrain glucose-6-phosphate to being metabolized through glycolysis. Under these conditions, glycolysis produces mostly pyruvate and little lactate. Pyruvate either enters the citric acid cycle through pyruvate dehydrogenase or is carboxylated by pyruvate carboxylase. Reducing equivalents from glycolysis enter oxidative phosphorylation through both the glycerol phosphate shuttle and citric acid cycle. Raising glucose concentration increases intramitochondrial [NADH]/[NAD+] and thereby the energy state ([ATP]/[ADP][Pi]), decreasing [Mg2+ADP] and [AMP]. [Mg2+ADP] acts through control of KATP channel conductance, whereas [AMP] acts through regulation of AMP-dependent protein kinase. Specific roles of different cell types are determined by the diverse molecular mechanisms used to couple energy state to cell specific responses. Having a common glucose sensor couples complementary regulatory mechanisms into a tightly regulated and stable glucose homeostatic network.
Collapse
Affiliation(s)
- Franz M Matschinsky
- Department of Biochemistry and Biophysics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - David F Wilson
- Department of Biochemistry and Biophysics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| |
Collapse
|
26
|
Christopoulou E, Tsimihodimos V, Filippatos T, Elisaf M. Apolipoprotein CIII and diabetes. Is there a link? Diabetes Metab Res Rev 2019; 35:e3118. [PMID: 30557902 DOI: 10.1002/dmrr.3118] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/31/2018] [Revised: 12/09/2018] [Accepted: 12/11/2018] [Indexed: 12/24/2022]
Abstract
Apolipoprotein CIII (ApoCIII), a small protein that resides on the surface of lipoprotein particles, is a key regulator of triglyceride metabolism. The inhibition of lipoprotein lipase (LPL), the increased assembly and secretion of very low-density lipoproteins (VLDL) and the decreased reuptake of triglyceride-rich lipoproteins (TRLs) by the liver are mechanisms associating elevated serum ApoCIII levels and hypertriglyceridemia. ApoCIII concentration is high in individuals with diabetes mellitus, indicating a possible positive correlation with impairment of glucose metabolism. The aim of this review (based on a Pubmed search until August 2018) is to present the possible mechanisms linking ApoCIII and deterioration of carbohydrate homeostasis. ApoCIII enhances pancreatic β-cells apoptosis via an increase of the cytoplasmic Ca2+ levels in the insulin-producing cells. In addition, overexpression of ApoCIII enhances non-alcoholic fatty liver disease and exacerbates inflammatory pathways in skeletal muscles, affecting insulin signalling and thereby inducing insulin resistance. Moreover, recent studies reveal a possible mechanism of body weight increase and glucose production through a potential ApoCIII-induced LPL inhibition in the hypothalamus. Also, the presence of ApoCIII on the surface of high-density lipoprotein particles is associated with impairment of their antiglycemic and atheroprotective properties. Modulating ApoCIII may be a potent therapeutic approach to manage hypertriglyceridemia and improve carbohydrate metabolism.
Collapse
Affiliation(s)
- Eliza Christopoulou
- Department of Internal Medicine, School of Medicine, University of Ioannina, Ioannina, Greece
| | - Vasilios Tsimihodimos
- Department of Internal Medicine, School of Medicine, University of Ioannina, Ioannina, Greece
| | - Theodosios Filippatos
- Department of Internal Medicine, School of Medicine, University of Crete, Heraklion, Greece
| | - Moses Elisaf
- Department of Internal Medicine, School of Medicine, University of Ioannina, Ioannina, Greece
| |
Collapse
|
27
|
Joseph B, Shimojo G, Li Z, Thompson-Bonilla MDR, Shah R, Kanashiro A, Salgado HC, Ulloa L. Glucose Activates Vagal Control of Hyperglycemia and Inflammation in Fasted Mice. Sci Rep 2019; 9:1012. [PMID: 30700738 PMCID: PMC6354016 DOI: 10.1038/s41598-018-36298-z] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2018] [Accepted: 11/14/2018] [Indexed: 11/18/2022] Open
Abstract
Sepsis is a leading cause of death in hospitalized patients. Many experimental treatments may have failed in clinical trials for sepsis, in part, because they focused on immune responses of healthy animals that did not mimic the metabolic settings of septic patients. Epidemiological studies show an association between metabolic and immune alterations and over 1/3 of septic patients are diabetic, but the mechanism linking these systems is unknown. Here, we report that metabolic fasting increased systemic inflammation and worsened survival in experimental sepsis. Feeding and administration of glucose in fasted mice activated the vagal tone without affecting blood pressure. Vagal stimulation attenuated hyperglycemia and serum TNF levels in sham but only hyperglycemia in splenectomized mice. Vagal stimulation induced the production of dopamine from the adrenal glands. Experimental diabetes increased hyperglycemia and systemic inflammation in experimental sepsis. Fenoldopam, a specific dopaminergic type-1 agonist, attenuated hyperglycemia and systemic inflammation in diabetic endotoxemic mice. These results indicate that glucose activates vagal control of hyperglycemia and inflammation in fasted septic mice via dopamine.
Collapse
Affiliation(s)
- Biju Joseph
- Department of Surgery, Rutgers-New Jersey Medical School, Newark, NJ, 07103, USA
| | - Guilherme Shimojo
- Department of Surgery, Rutgers-New Jersey Medical School, Newark, NJ, 07103, USA
| | - Zhifeng Li
- Department of Surgery, Rutgers-New Jersey Medical School, Newark, NJ, 07103, USA
| | - Maria Del Rocio Thompson-Bonilla
- Department of Surgery, Rutgers-New Jersey Medical School, Newark, NJ, 07103, USA
- Hospital "October 1st", ISSSTE", 1669 National Polytechnic Institute Ave, Mexico City, Mexico
| | - Roshan Shah
- Department of Surgery, Rutgers-New Jersey Medical School, Newark, NJ, 07103, USA
| | - Alexandre Kanashiro
- Department of Surgery, Rutgers-New Jersey Medical School, Newark, NJ, 07103, USA
- Department of Physiology, Medical School - University of São Paulo, Ribeirão Preto, SP, 14049-900, Brazil
| | - Helio C Salgado
- Department of Physiology, Medical School - University of São Paulo, Ribeirão Preto, SP, 14049-900, Brazil
| | - Luis Ulloa
- Department of Surgery, Rutgers-New Jersey Medical School, Newark, NJ, 07103, USA.
- Center for Immunity and Inflammation, Rutgers-New Jersey Medical School, Newark, NJ, 07103, USA.
| |
Collapse
|
28
|
Comparison of Low Glycaemic Index and High Glycaemic Index Potatoes in Relation to Satiety: A Single-Blinded, Randomised Crossover Study in Humans. Nutrients 2018; 10:nu10111726. [PMID: 30423848 PMCID: PMC6266898 DOI: 10.3390/nu10111726] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2018] [Revised: 10/31/2018] [Accepted: 11/08/2018] [Indexed: 01/19/2023] Open
Abstract
High glycaemic index (GI) foods have been proposed to reduce satiety and thus promote overweight and obesity. Generally, potatoes have a high GI, but they also provide many beneficial nutrients and they are a highly important food source globally. In this study, we investigated how a low GI potato affected subjective satiety as compared to a high GI potato. Twenty healthy men (aged 18–40 years; body mass index (BMI) 18–27 kg/m2) participated in this single-blinded, controlled, randomised crossover trial. On each of the two trial days, the subjects were given a 500-gram portion of either a low or high GI potato variety (Carisma® low GI and Arizona high GI). Subjective appetite sensations were measured at baseline and at +15 min, +45 min, +75 min, +105 min, and +135 min after consumption of the test meal until an ad libitum meal was served at +150 min. No significant differences in the primary endpoint, satiety, were found between the two potato varieties (all p > 0.05). Furthermore, no significant differences were found in the secondary endpoints; hunger, fullness, and prospective food consumption, or ad libitum energy intake (all p > 0.05). In conclusion, the results of this study do not indicate that the GI of potatoes is important for satiety in normal-weight men.
Collapse
|
29
|
Tiwari BK, Abidi AB, Rizvi SI, Pandey KB. Effect of oral supplementation of composite leaf extract of medicinal plants on biomarkers of oxidative stress in induced diabetic Wistar rats. Arch Physiol Biochem 2018; 124:361-366. [PMID: 29207888 DOI: 10.1080/13813455.2017.1411369] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Abstract
Present study was conducted to evaluate the effect of oral supplementation of composite extract of leaves (CLE) of four medicinal plants; Aegle marmelos, Ocimum sanctum, Murraya koenigii and Azadirachta indica on markers of oxidative stress in brain tissues of alloxan-induced diabetic rats in vivo. Enhanced lipid peroxidation, protein oxidation and reduced antioxidative defence systems were measured in brain tissues of diabetic rats. Supplementation of CLE, once in a day for 35 days significantly (p < .05) protected the peroxidation of lipid, oxidation of protein and ameliorated the antioxidant defence in brain tissue of diabetic rats. It was observed that the insulin-like effect of CLE was dose dependent; higher effect at higher doses. The results of the study suggest that supplementation CLE may provide an overall homeostasis and significant neuro-protection through rescuing brain cells from oxidative abuse and accelerating brain antioxidative defence during advanced stage of hyperglycaemia.
Collapse
Affiliation(s)
- Brahm Kumar Tiwari
- a Department of Biochemistry and Biochemical Engineering , Sam Higginbottom Institute of Agriculture, Technology and Sciences (SHIATS) , Allahabad , India
| | - Abu Baker Abidi
- a Department of Biochemistry and Biochemical Engineering , Sam Higginbottom Institute of Agriculture, Technology and Sciences (SHIATS) , Allahabad , India
| | - Syed Ibrahim Rizvi
- b Department of Biochemistry , University of Allahabad , Allahabad , India
| | | |
Collapse
|
30
|
Cavalcanti-de-Albuquerque JP, Kincheski GC, Louzada RA, Galina A, Pierucci APTR, Carvalho DP. Intense physical exercise potentiates glucose inhibitory effect over food intake of male Wistar rats. Exp Physiol 2018; 103:1076-1086. [PMID: 29893447 DOI: 10.1113/ep086916] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2018] [Accepted: 06/06/2018] [Indexed: 11/08/2022]
Abstract
NEW FINDINGS What is the central question of this study? How does an acute session of exercise affect food intake of male Wistar rats? What is the main finding and its importance? Food intake in male Wistar rats is decreased in the first hour after physical exercise independent of the intensity. Moreover, high-intensity exercise potentiates the anorexic effect of peripheral glucose administration. This work raises new feeding-related targets that would explain how exercise drives body weight loss. ABSTRACT Obesity has emerged as a critical metabolic disorder in modern society. An adequate lifestyle with a well-oriented programme of diet and physical exercise (PE) can prevent or potentially even cure obesity. Additionally, PE might lead to weight loss by increasing energy expenditure and decreasing hunger perception. In this article, we hypothesize that an acute exercise session would potentiate the glucose inhibitory effects on food intake in male Wistar rats. Our data show that moderate- or high-intensity PE significantly decreased food intake, although no changes in the expression of feeding-related neuropeptide in the arcuate nucleus of the hypothalamus were found. Exercised animals demonstrated a reduced glucose tolerance and increased blood insulin concentration. Intraperitoneal administration of glucose decreased food intake in control animals. In the animals submitted to moderate-intensity PE, the decrease in food intake promoted by glucose was similar to controls; however, an interaction was observed when glucose was injected in the high-intensity PE group, in which food intake was significantly lower than the effect produced by glucose alone. A different pattern of expression was observed for the monocarboxylate transporter isoforms (MCT1, 2 and 4) and 6-phosphofructo-2-kinase/fructose-2,6-biphosphatase 3 (PFKFBP3) in the hypothalamus, which was dependent on the exercise intensity. In conclusion, PE decreases food intake independently of the intensity. However, an interaction between PE and the anorexic effect of glucose is only observed when a high-intensity exercise is performed. These data show an essential role of exercise intensity in the modulation of the glucose inhibitory effect on food intake.
Collapse
Affiliation(s)
- João Paulo Cavalcanti-de-Albuquerque
- Institute of Biophysics Carlos Chagas Filho, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil.,Institute of Nutrition Josue de Castro, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Grasielle Clotildes Kincheski
- Institute of Medical Biochemistry Leopoldo De Meis, Federal University of Rio de Janeiro, Center of Health Science, Rio de Janeiro, Brazil
| | - Ruy Andrade Louzada
- Institute of Biophysics Carlos Chagas Filho, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Antônio Galina
- Institute of Medical Biochemistry Leopoldo De Meis, Federal University of Rio de Janeiro, Center of Health Science, Rio de Janeiro, Brazil
| | | | - Denise P Carvalho
- Institute of Biophysics Carlos Chagas Filho, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| |
Collapse
|
31
|
Prevot V, Dehouck B, Sharif A, Ciofi P, Giacobini P, Clasadonte J. The Versatile Tanycyte: A Hypothalamic Integrator of Reproduction and Energy Metabolism. Endocr Rev 2018; 39:333-368. [PMID: 29351662 DOI: 10.1210/er.2017-00235] [Citation(s) in RCA: 171] [Impact Index Per Article: 24.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/01/2017] [Accepted: 01/12/2018] [Indexed: 12/16/2022]
Abstract
The fertility and survival of an individual rely on the ability of the periphery to promptly, effectively, and reproducibly communicate with brain neural networks that control reproduction, food intake, and energy homeostasis. Tanycytes, a specialized glial cell type lining the wall of the third ventricle in the median eminence of the hypothalamus, appear to act as the linchpin of these processes by dynamically controlling the secretion of neuropeptides into the portal vasculature by hypothalamic neurons and regulating blood-brain and blood-cerebrospinal fluid exchanges, both processes that depend on the ability of these cells to adapt their morphology to the physiological state of the individual. In addition to their barrier properties, tanycytes possess the ability to sense blood glucose levels, and play a fundamental and active role in shuttling circulating metabolic signals to hypothalamic neurons that control food intake. Moreover, accumulating data suggest that, in keeping with their putative descent from radial glial cells, tanycytes are endowed with neural stem cell properties and may respond to dietary or reproductive cues by modulating hypothalamic neurogenesis. Tanycytes could thus constitute the missing link in the loop connecting behavior, hormonal changes, signal transduction, central neuronal activation and, finally, behavior again. In this article, we will examine these recent advances in the understanding of tanycytic plasticity and function in the hypothalamus and the underlying molecular mechanisms. We will also discuss the putative involvement and therapeutic potential of hypothalamic tanycytes in metabolic and fertility disorders.
Collapse
Affiliation(s)
- Vincent Prevot
- Inserm, Laboratory of Development and Plasticity of the Neuroendocrine Brain, Jean-Pierre Aubert Research Center, Lille, France.,University of Lille, FHU 1000 Days for Health, School of Medicine, Lille, France
| | - Bénédicte Dehouck
- Inserm, Laboratory of Development and Plasticity of the Neuroendocrine Brain, Jean-Pierre Aubert Research Center, Lille, France.,University of Lille, FHU 1000 Days for Health, School of Medicine, Lille, France
| | - Ariane Sharif
- Inserm, Laboratory of Development and Plasticity of the Neuroendocrine Brain, Jean-Pierre Aubert Research Center, Lille, France.,University of Lille, FHU 1000 Days for Health, School of Medicine, Lille, France
| | - Philippe Ciofi
- Inserm, Neurocentre Magendie, Bordeaux, France.,Université de Bordeaux, Bordeaux, France
| | - Paolo Giacobini
- Inserm, Laboratory of Development and Plasticity of the Neuroendocrine Brain, Jean-Pierre Aubert Research Center, Lille, France.,University of Lille, FHU 1000 Days for Health, School of Medicine, Lille, France
| | - Jerome Clasadonte
- Inserm, Laboratory of Development and Plasticity of the Neuroendocrine Brain, Jean-Pierre Aubert Research Center, Lille, France.,University of Lille, FHU 1000 Days for Health, School of Medicine, Lille, France
| |
Collapse
|
32
|
Seo M, Islam SA, Moon SS. Acute anti-obesity effects of intracerebroventricular 11β-HSD1 inhibitor administration in diet-induced obese mice. J Neuroendocrinol 2018; 30:e12580. [PMID: 29418022 DOI: 10.1111/jne.12580] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/20/2017] [Revised: 01/09/2018] [Accepted: 02/01/2018] [Indexed: 11/30/2022]
Abstract
The hypothalamus is the regulatory centre of both appetite and energy balance and endoplasmic reticulum (ER) stress in the hypothalamus is involved in the pathogenesis of obesity. Recently, inhibition of 11 β hydroxysteroid dehydrogenase type1 (11β-HSD1) was reported to have an anti-obesity effect by reducing fat mass. However, the link between the role of 11β-HSD1 in the hypothalamus and obesity has yet to be determined. In the present study, embryonal primary hypothalamic neurones and high-fat diet (HFD) fed mice were used to investigate the anorexigenic effects of 11β-HSD1 inhibitors both in vitro and in vivo. In hypothalamic neurones, carbenoxolone (a non selecitve 11β-HSD inhibitor) alleviated ER stress and ER stress-induced neuropeptide alterations. In HFD mice, i.c.v. administration of carbenoxolone or KR67500 (nonselective and selective 11β-HSD1 inhibitors, respectively) was associated with less weight gain compared to control mice for 24 hours after treatment, presumably by reducing food intake. Furthermore, glucose regulated protein (Grp78), spliced X-box binding protein (Xbp-1s), c/EBP homologous protein (chop) and ER DnaJ homologue protein (Erdj4) expression was decreased in the hypothalami of mice administrated 11β-HSD1 inhibitors compared to controls. Conversely, the phosphorylation of protein kinase B (PKB/Akt), signal transducer and activator of transcription 3 (Stat3), mitogen-activated protein kinase (MAPK/ERK) and S6 kinase1 (S6K1) in the hypothalamus was induced more in mice treated using the same regimes. In conclusion, acute 11β-HSD1 inhibition in the hypothalamus could reduce food intake by decreasing ER stress and increasing insulin, leptin, and mammalian target of rapamycin complex 1 (mTORC1) signalling.
Collapse
Affiliation(s)
- M Seo
- Medical Institute of Dongguk University, Gyeongju, South Korea
- Department of Agricultural Biology, National Institute of Agricultural Sciences, Rural Development Administration, Wanju-gun, South Korea
| | - S A Islam
- Department of Internal Medicine, College of Medicine, Dongguk University, Gyeongju, South Korea
| | - S-S Moon
- Medical Institute of Dongguk University, Gyeongju, South Korea
- Department of Internal Medicine, College of Medicine, Dongguk University, Gyeongju, South Korea
| |
Collapse
|
33
|
Chu SC, Chen PN, Chen JR, Yu CH, Hsieh YS, Kuo DY. Role of hypothalamic leptin-LepRb signaling in NPY-CART-mediated appetite suppression in amphetamine-treated rats. Horm Behav 2018; 98:173-182. [PMID: 29307696 DOI: 10.1016/j.yhbeh.2017.12.019] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/28/2017] [Revised: 12/22/2017] [Accepted: 12/29/2017] [Indexed: 12/15/2022]
Abstract
Leptin is an adipose tissue hormone which plays an important role in regulating energy homeostasis. Amphetamine (AMPH) is a drug of appetite suppressant, which exerts its effect by decreasing the expression of hypothalamic neuropeptide Y (NPY) and increasing that of cocaine- and amphetamine-regulated transcript (CART). This study investigated whether leptin, the leptin receptor (LepRb) and the signal transducer and activator of transcription-3 (STAT3) were involved in NPY/CART-mediated appetite suppression in AMPH-treated rats. Rats were given AMPH daily for four days, and changes in the levels of blood leptin and hypothalamic NPY, CART, LepRb, Janus kinases 2 (JAK2), and STAT3 were assessed and compared. During the AMPH treatment, blood leptin levels and hypothalamic NPY expression decreased, with the largest reduction observed on Day 2. By contrast, the expression of hypothalamic CART, LepRb, JAK2, and STAT3 increased, with the maximum response on Day 2. Furthermore, the binding activity of pSTAT3/DNA increased and was expressed in similar pattern to that of CART, LepRb, and JAK2. An intracerebroventricular infusion of NPY antisense 60min prior to AMPH treatment increased the levels of leptin, as well as the expression in LepRb, JAK2, and CART, whereas an infusion of STAT3 antisense decreased these levels and the expression of these parameters. The results suggest that blood leptin and hypothalamic LepRb-JAK2-STAT3 signaling involved in NPY-CART-regulated appetite suppression in AMPH-treated rats. The findings may aid understanding the role of leptin-LepRb during the treatment of anorectic drugs.
Collapse
Affiliation(s)
- Shu-Chen Chu
- Department of Food Science, Central Taiwan University of Science and Technology, Taichung City 406, Taiwan
| | - Pei-Ni Chen
- Institute of Biochemistry and Biotechnology, Chung Shan Medical University, Chung Shan Medical University Hospital, Taichung City 40201, Taiwan
| | - Jeng-Rung Chen
- Department of Veterinary Medicine, National Chung Hsing University, Taichung City 40201, Taiwan
| | - Ching-Han Yu
- Department of Physiology, Chung Shan Medical University, Chung Shan Medical University Hospital, Taichung City 40201, Taiwan
| | - Yih-Shou Hsieh
- Institute of Biochemistry and Biotechnology, Chung Shan Medical University, Chung Shan Medical University Hospital, Taichung City 40201, Taiwan
| | - Dong-Yih Kuo
- Department of Physiology, Chung Shan Medical University, Chung Shan Medical University Hospital, Taichung City 40201, Taiwan.
| |
Collapse
|
34
|
Szabó I, Hormay E, Csetényi B, Nagy B, Lénárd L, Karádi Z. Multiple functional attributes of glucose-monitoring neurons in the medial orbitofrontal (ventrolateral prefrontal) cortex. Neurosci Biobehav Rev 2018; 85:44-53. [DOI: 10.1016/j.neubiorev.2017.04.024] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2017] [Revised: 04/11/2017] [Accepted: 04/21/2017] [Indexed: 11/28/2022]
|
35
|
Sun L, Liu J, Tian P, Ni Y, Zhao R. The effect of fasting on the appetite-associated factors and energy sensors expression in the hypothalamus of different TI broilers. ANIMAL PRODUCTION SCIENCE 2018. [DOI: 10.1071/an15473] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
Tonic immobility (TI) is a behaviour related to fear and stress response. Birds can exhibit a short (STI) or long (LTI) tonic immobility phenotype on the basis in TI duration. In this study, the differences in the hypothalamic appetite-associated factors and energy sensor gene expression between STI and LTI broilers were evaluated under free feed access or 16-h fasting. The results showed that the concentrations of cholesterol, high density lipoprotein-cholesterol, non-esterified fatty acid, malonaldehyde, lactate dehydrogenase, creatine kinase, alanine aminotransferase and aspartate aminotransferase in plasma were significantly increased (P < 0.05) in fasting broilers compared with broilers fed ad libitum, whereas plasma glucose, triglyceride, and total antioxidant capacity concentrations were decreased (P < 0.05). With respect to TI, however, only low density lipoprotein-cholesterol and glutathione peroxidase concentrations in plasma showed significant differences between LTI and STI broilers, with higher concentrations in LTI compared with STI. Real-time PCR results showed that only NPY mRNA expression demonstrated a tendency to increase in STI broilers compared with LTI (P = 0.095). Fasting downregulated SREBP-1 and its target gene FAS but upregulated CPT1 in the hypothalamus. Additionally, levels of hypothalamic p-GR and p-AMPK protein expression decreased after fasting. These results indicate that a 16-h fasting results in a negative energy status, and is accompanied with changes in expression of hypothalamic energy sensor and appetite-associated factors.
Collapse
|
36
|
Mitochondria-Bound Hexokinase (mt-HK) Activity Differ in Cortical and Hypothalamic Synaptosomes: Differential Role of mt-HK in H 2O 2 Depuration. Mol Neurobiol 2017; 55:5889-5900. [PMID: 29119535 DOI: 10.1007/s12035-017-0807-9] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2017] [Accepted: 10/18/2017] [Indexed: 01/06/2023]
Abstract
Glucose and oxygen are vital for the brain, as these molecules provide energy and metabolic intermediates that are necessary for cell function. The glycolysis pathway and mitochondria play a pivotal role in cell energy metabolism, which is closely related to reactive oxygen species (ROS) production. Hexokinase (HK) is a key enzyme involved in glucose metabolism that modulates the level of brain mitochondrial ROS by recycling ADP for oxidative phosphorylation (OxPhos). Here, we hypothesize that the control of mitochondrial metabolism by hexokinase differs in distinct areas of the brain, such as the cortex and hypothalamus, in which ROS might function as signaling molecules. Thus, we investigated mitochondrial metabolism of synaptosomes derived from both brain regions. Cortical synaptosomes (CSy) show a predominance of glutamatergic synapses, while in the hypothalamic synaptosomes (HSy), the GABAergic synapses predominate. Significant differences of oxygen consumption and ROS production were related to higher mitochondrial complex II activity (succinate dehydrogenase-SDH) in CSy rather than to mitochondrial number. Mitochondrial HK (mt-HK) activity was higher in CSy than in HSy regardless the substrate added. Mitochondrial O2 consumption related to mt-HK activation by 2-deoxyglucose was also higher in CSy. In the presence of substrate for complex II, the activation of synaptosomal mt-HK promoted depuration of ROS in both HSy and CSy, while ROS depuration did not occur in HSy when substrate for complex I was used. The impact of the mt-HK inhibition by glucose-6-phosphate (G6P) was the same in synaptosomes from both areas. Together, the differences found between CSy and HSy indicate specific roles of mt-HK and SDH on the metabolism of each brain region, what probably depends on the main metabolic route that is used by the neurons.
Collapse
|
37
|
Devarakonda K, Stanley S. Investigating metabolic regulation using targeted neuromodulation. Ann N Y Acad Sci 2017; 1411:83-95. [PMID: 29106710 DOI: 10.1111/nyas.13468] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2017] [Revised: 08/11/2017] [Accepted: 08/18/2017] [Indexed: 01/13/2023]
Abstract
The central nervous system (CNS) plays a vital role in regulating energy balance and metabolism. Over the last 50 years, studies in animal models have allowed us to identify critical CNS regions involved in these processes and even crucial cell populations. Now, techniques for genetically and anatomically targeted manipulation of specific neural populations using light (optogenetic), ligands (chemogenetic), or magnetic fields (radiogenetic/magnetogenetic) allow detailed investigation of circuits involved in metabolic regulation. In this review, we provide a brief overview of recent studies using light- and magnetic field-regulated neural activity to investigate the neural circuits contributing to metabolic control.
Collapse
Affiliation(s)
- Kavya Devarakonda
- Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Sarah Stanley
- Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, New York.,Diabetes, Obesity and Metabolism Institute, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, New York
| |
Collapse
|
38
|
Abstract
Understanding of the neural and physiological substrates of hunger and satiety has increased rapidly over the last three decades, and pharmacological targets have already been identified for the treatment of obesity that has moved from pre-clinical screening to therapies approved by regulatory authorities. Initially, this review describes the way in which physiological signals of energy availability interact with hedonic and rewarding properties of food to modulate the neural circuitry that supports eating behaviour. This is followed by a brief account of current and promising targets for drug development and a review of the wide range of preclinical paradigms that model important influences on human eating behaviour, and can be used to guide early stages of the drug development process.
Collapse
|
39
|
Carey M, Gospin R, Goyal A, Tomuta N, Sandu O, Mbanya A, Lontchi-Yimagou E, Hulkower R, Shamoon H, Gabriely I, Hawkins M. Opioid Receptor Activation Impairs Hypoglycemic Counterregulation in Humans. Diabetes 2017; 66:2764-2773. [PMID: 28860128 PMCID: PMC5652610 DOI: 10.2337/db16-1478] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/30/2016] [Accepted: 08/24/2017] [Indexed: 01/08/2023]
Abstract
Although intensive glycemic control improves outcomes in type 1 diabetes mellitus (T1DM), iatrogenic hypoglycemia limits its attainment. Recurrent and/or antecedent hypoglycemia causes blunting of protective counterregulatory responses, known as hypoglycemia-associated autonomic failure (HAAF). To determine whether and how opioid receptor activation induces HAAF in humans, 12 healthy subjects without diabetes (7 men, age 32.3 ± 2.2 years, BMI 25.1 ± 1.0 kg/m2) participated in two study protocols in random order over two consecutive days. On day 1, subjects received two 120-min infusions of either saline or morphine (0.1 μg/kg/min), separated by a 120-min break (all euglycemic). On day 2, subjects underwent stepped hypoglycemic clamps (nadir 60 mg/dL) with evaluation of counterregulatory hormonal responses, endogenous glucose production (EGP, using 6,6-D2-glucose), and hypoglycemic symptoms. Morphine induced an ∼30% reduction in plasma epinephrine response together with reduced EGP and hypoglycemia-associated symptoms on day 2. Therefore, we report the first studies in humans demonstrating that pharmacologic opioid receptor activation induces some of the clinical and biochemical features of HAAF, thus elucidating the individual roles of various receptors involved in HAAF's development and suggesting novel pharmacologic approaches for safer intensive glycemic control in T1DM.
Collapse
Affiliation(s)
- Michelle Carey
- Diabetes Research and Training Center, Albert Einstein College of Medicine, Bronx, NY
- Center for Drug Evaluation and Research, U.S. Food and Drug Administration, Silver Spring, MD
| | - Rebekah Gospin
- Diabetes Research and Training Center, Albert Einstein College of Medicine, Bronx, NY
| | - Akankasha Goyal
- Diabetes Research and Training Center, Albert Einstein College of Medicine, Bronx, NY
| | - Nora Tomuta
- Diabetes Research and Training Center, Albert Einstein College of Medicine, Bronx, NY
| | - Oana Sandu
- Diabetes Research and Training Center, Albert Einstein College of Medicine, Bronx, NY
| | - Armand Mbanya
- Diabetes Research and Training Center, Albert Einstein College of Medicine, Bronx, NY
| | - Eric Lontchi-Yimagou
- Diabetes Research and Training Center, Albert Einstein College of Medicine, Bronx, NY
| | - Raphael Hulkower
- Diabetes Research and Training Center, Albert Einstein College of Medicine, Bronx, NY
| | - Harry Shamoon
- Diabetes Research and Training Center, Albert Einstein College of Medicine, Bronx, NY
| | - Ilan Gabriely
- Diabetes Research and Training Center, Albert Einstein College of Medicine, Bronx, NY
| | - Meredith Hawkins
- Diabetes Research and Training Center, Albert Einstein College of Medicine, Bronx, NY
| |
Collapse
|
40
|
Abstract
A hypercaloric diet combined with a sedentary lifestyle is a major risk factor for the development of insulin resistance, type 2 diabetes mellitus (T2DM) and associated comorbidities. Standard treatment for T2DM begins with lifestyle modification, and includes oral medications and insulin therapy to compensate for progressive β-cell failure. However, current pharmaceutical options for T2DM are limited in that they do not maintain stable, durable glucose control without the need for treatment intensification. Furthermore, each medication is associated with adverse effects, which range from hypoglycaemia to weight gain or bone loss. Unexpectedly, fibroblast growth factor 1 (FGF1) and its low mitogenic variants have emerged as potentially safe candidates for restoring euglycaemia, without causing overt adverse effects. In particular, a single peripheral injection of FGF1 can lower glucose to normal levels within hours, without the risk of hypoglycaemia. Similarly, a single intracerebroventricular injection of FGF1 can induce long-lasting remission of the diabetic phenotype. This Review discusses potential mechanisms by which centrally administered FGF1 improves central glucose-sensing and peripheral glucose uptake in a sustained manner. Specifically, we explore the potential crosstalk between FGF1 and glucose-sensing neuronal circuits, hypothalamic neural stem cells and synaptic plasticity. Finally, we highlight therapeutic considerations of FGF1 and compare its metabolic actions with FGF15 (rodents), FGF19 (humans) and FGF21.
Collapse
Affiliation(s)
- Emanuel Gasser
- Gene Expression Laboratory, Salk Institute for Biological Studies
| | - Christopher P Moutos
- Gene Expression Laboratory, Salk Institute for Biological Studies
- Howard Hughes Medical Institute, Salk Institute for Biological Studies, 10010 North Torrey Pines Road, La Jolla, California 92037, USA
- College of Medicine, University of Arkansas for Medical Sciences, 4301 West Markham Street, Little Rock, Arkansas 72205, USA
| | - Michael Downes
- Gene Expression Laboratory, Salk Institute for Biological Studies
| | - Ronald M Evans
- Gene Expression Laboratory, Salk Institute for Biological Studies
- Howard Hughes Medical Institute, Salk Institute for Biological Studies, 10010 North Torrey Pines Road, La Jolla, California 92037, USA
| |
Collapse
|
41
|
van den Top M, Zhao FY, Viriyapong R, Michael NJ, Munder AC, Pryor JT, Renaud LP, Spanswick D. The impact of ageing, fasting and high-fat diet on central and peripheral glucose tolerance and glucose-sensing neural networks in the arcuate nucleus. J Neuroendocrinol 2017; 29. [PMID: 28834571 DOI: 10.1111/jne.12528] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/11/2017] [Revised: 08/01/2017] [Accepted: 08/17/2017] [Indexed: 12/14/2022]
Abstract
Obesity and ageing are risk factors for diabetes. In the present study, we investigated the effects of ageing, obesity and fasting on central and peripheral glucose tolerance and on glucose-sensing neuronal function in the arcuate nucleus of rats, with a view to providing insight into the central mechanisms regulating glucose homeostasis and how they change or are subject to dysfunction with ageing and obesity. We show that, following a glucose load, central glucose tolerance at the level of the cerebrospinal fluid (CSF) and plasma is significantly reduced in rats maintained on a high-fat diet (HFD). With ageing, up to 2 years, central glucose tolerance was impaired in an age-dependent manner, whereas peripheral glucose tolerance remained unaffected. Ageing-induced peripheral glucose intolerance was improved by a 24-hour fast, whereas central glucose tolerance was not corrected. Pre-wean, immature animals have elevated basal plasma glucose levels and a delayed increase in central glucose levels following peripheral glucose injection compared to mature animals. Electrophysiological recording techniques revealed an energy-status-dependent role for glucose-excited, inhibited and adapting neurones, along with glucose-induced changes in synaptic transmission. We conclude that ageing affects central glucose tolerance, whereas HFD profoundly affects central and peripheral glucose tolerance and, in addition, glucose-sensing neurones adapt function in an energy-status-dependent manner.
Collapse
Affiliation(s)
| | - F-Y Zhao
- NeuroSolutions Ltd, Coventry, UK
| | - R Viriyapong
- Warwick Medical School, University of Warwick, Coventry, UK
- MOAC DTC, University of Warwick, Coventry, UK
| | - N J Michael
- Metabolic Disease and Obesity Program, Biomedicine Discovery Institute, Monash University, Melbourne, VIC, Australia
- Department of Physiology, Monash University, Clayton, VIC, Australia
| | - A C Munder
- Metabolic Disease and Obesity Program, Biomedicine Discovery Institute, Monash University, Melbourne, VIC, Australia
- Department of Physiology, Monash University, Clayton, VIC, Australia
| | - J T Pryor
- Warwick Medical School, University of Warwick, Coventry, UK
- Metabolic Disease and Obesity Program, Biomedicine Discovery Institute, Monash University, Melbourne, VIC, Australia
- Department of Physiology, Monash University, Clayton, VIC, Australia
| | - L P Renaud
- Ottawa Hospital Research Institute, Ottawa Civic Hospital, Ottawa, ON, Canada
| | - D Spanswick
- NeuroSolutions Ltd, Coventry, UK
- Warwick Medical School, University of Warwick, Coventry, UK
- Metabolic Disease and Obesity Program, Biomedicine Discovery Institute, Monash University, Melbourne, VIC, Australia
- Department of Physiology, Monash University, Clayton, VIC, Australia
- Neuroscience Program, Biomedicine Discovery Institute, Monash University, Melbourne, VIC, Australia
| |
Collapse
|
42
|
A comprehensive analysis of mitochondrial genes variants and their association with antipsychotic-induced weight gain. Schizophr Res 2017; 187:67-73. [PMID: 28693754 PMCID: PMC5660917 DOI: 10.1016/j.schres.2017.06.046] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/05/2017] [Revised: 06/21/2017] [Accepted: 06/23/2017] [Indexed: 01/09/2023]
Abstract
Antipsychotic Induced Weight Gain (AIWG) is a common and severe side effect of many antipsychotic medications. Mitochondria play a vital role for whole-body energy homeostasis and there is increasing evidence that antipsychotics modulate mitochondrial function. This study aimed to examine the role of variants in nuclear-encoded mitochondrial genes and the mitochondrial DNA (mtDNA) in conferring risk for AIWG. We selected 168 European-Caucasian individuals from the CATIE sample based upon meeting criteria of multiple weight measures while taking selected antipsychotics (risperidone, quetiapine or olanzapine). We tested the association of 670 nuclear-encoded mitochondrial genes with weight change (%) using MAGMA software. Thirty of these genes showed nominally significant P-values (<0.05). We were able to replicate the association of three genes, CLPB, PARL, and ACAD10, with weight change (%) in an independent prospectively assessed AIWG sample. We analyzed mtDNA variants in a subset of 74 of these individuals using next-generation sequencing. No common or rare mtDNA variants were found to be significantly associated with weight change (%) in our sample. Additionally, analysis of mitochondrial haplogroups showed no association with weight change (%). In conclusion, our findings suggest nuclear-encoded mitochondrial genes play a role in AIWG. Replication in larger sample is required to validate our initial report of mtDNA variants in AIWG.
Collapse
|
43
|
Paeger L, Pippow A, Hess S, Paehler M, Klein AC, Husch A, Pouzat C, Brüning JC, Kloppenburg P. Energy imbalance alters Ca 2+ handling and excitability of POMC neurons. eLife 2017; 6. [PMID: 28762947 PMCID: PMC5538824 DOI: 10.7554/elife.25641] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2017] [Accepted: 06/29/2017] [Indexed: 01/16/2023] Open
Abstract
Satiety-signaling, pro-opiomelanocortin (POMC)-expressing neurons in the arcuate nucleus of the hypothalamus play a pivotal role in the regulation of energy homeostasis. Recent studies reported altered mitochondrial dynamics and decreased mitochondria- endoplasmic reticulum contacts in POMC neurons during diet-induced obesity. Since mitochondria play a crucial role in Ca2+ signaling, we investigated whether obesity alters Ca2+ handling of these neurons in mice. In diet-induced obesity, cellular Ca2+ handling properties including mitochondrial Ca2+ uptake capacity are impaired, and an increased resting level of free intracellular Ca2+ is accompanied by a marked decrease in neuronal excitability. Experimentally increasing or decreasing intracellular Ca2+ concentrations reproduced electrophysiological properties observed in diet-induced obesity. Taken together, we provide the first direct evidence for a diet-dependent deterioration of Ca2+ homeostasis in POMC neurons during obesity development resulting in impaired function of these critical energy homeostasis-regulating neurons. DOI:http://dx.doi.org/10.7554/eLife.25641.001
Collapse
Affiliation(s)
- Lars Paeger
- Biocenter, Institute for Zoology, University of Cologne, Cologne, Germany.,Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases, University of Cologne, Cologne, Germany
| | - Andreas Pippow
- Biocenter, Institute for Zoology, University of Cologne, Cologne, Germany.,Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases, University of Cologne, Cologne, Germany
| | - Simon Hess
- Biocenter, Institute for Zoology, University of Cologne, Cologne, Germany.,Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases, University of Cologne, Cologne, Germany
| | - Moritz Paehler
- Biocenter, Institute for Zoology, University of Cologne, Cologne, Germany.,Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases, University of Cologne, Cologne, Germany
| | - Andreas C Klein
- Biocenter, Institute for Zoology, University of Cologne, Cologne, Germany.,Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases, University of Cologne, Cologne, Germany
| | - Andreas Husch
- Biocenter, Institute for Zoology, University of Cologne, Cologne, Germany.,Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases, University of Cologne, Cologne, Germany
| | - Christophe Pouzat
- MAP5 - Mathématiques Appliquées à Paris 5, CNRS UMR 8145, Paris, France
| | - Jens C Brüning
- Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases, University of Cologne, Cologne, Germany.,Department of Mouse Genetics and Metabolism, Institute for Genetics, Center of Molecular Medicine Cologne, Center for Endocrinology, Diabetes and Preventive Medicine, University Hospital of Cologne, Cologne, Germany.,Max Planck Institute for Metabolism Research, Cologne, Germany
| | - Peter Kloppenburg
- Biocenter, Institute for Zoology, University of Cologne, Cologne, Germany.,Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases, University of Cologne, Cologne, Germany
| |
Collapse
|
44
|
Nilaweera KN, Cabrera-Rubio R, Speakman JR, O'Connor PM, McAuliffe A, Guinane CM, Lawton EM, Crispie F, Aguilera M, Stanley M, Boscaini S, Joyce S, Melgar S, Cryan JF, Cotter PD. Whey protein effects on energy balance link the intestinal mechanisms of energy absorption with adiposity and hypothalamic neuropeptide gene expression. Am J Physiol Endocrinol Metab 2017; 313:E1-E11. [PMID: 28325732 DOI: 10.1152/ajpendo.00356.2016] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/29/2016] [Revised: 03/17/2017] [Accepted: 03/17/2017] [Indexed: 02/08/2023]
Abstract
We tested the hypothesis that dietary whey protein isolate (WPI) affects the intestinal mechanisms related to energy absorption and that the resulting energy deficit is compensated by changes in energy balance to support growth. C57BL/6 mice were provided a diet enriched with WPI with varied sucrose content, and the impact on energy balance-related parameters was investigated. As part of a high-sucrose diet, WPI reduced the hypothalamic expression of pro-opiomelanocortin gene expression and increased energy intake. The energy expenditure was unaffected, but epididymal weight was reduced, indicating an energy loss. Notably, there was a reduction in the ileum gene expression for amino acid transporter SLC6a19, glucose transporter 2, and fatty acid transporter 4. The composition of the gut microbiota also changed, where Firmicutes were reduced. The above changes indicated reduced energy absorption through the intestine. We propose that this mobilized energy in the adipose tissue and caused hypothalamic changes that increased energy intake, acting to counteract the energy deficit arising in the intestine. Lowering the sucrose content in the WPI diet increased energy expenditure. This further reduced epididymal weight and plasma leptin, whereupon hypothalamic ghrelin gene expression and the intestinal weight were both increased. These data suggest that when the intestine-adipose-hypothalamic pathway is subjected to an additional energy loss (now in the adipose tissue), compensatory changes attempt to assimilate more energy. Notably, WPI and sucrose content interact to enable the component mechanisms of this pathway.
Collapse
Affiliation(s)
- Kanishka N Nilaweera
- Food Biosciences Department, Teagasc Food Research Centre, Moorepark, Fermoy, County Cork, Ireland;
| | - Raul Cabrera-Rubio
- Food Biosciences Department, Teagasc Food Research Centre, Moorepark, Fermoy, County Cork, Ireland
| | - John R Speakman
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, China
- Institute of Biological and Environmental Sciences, University of Aberdeen, Aberdeen, Scotland
| | - Paula M O'Connor
- Food Biosciences Department, Teagasc Food Research Centre, Moorepark, Fermoy, County Cork, Ireland
| | - AnneMarie McAuliffe
- Food Biosciences Department, Teagasc Food Research Centre, Moorepark, Fermoy, County Cork, Ireland
| | - Caitriona M Guinane
- Food Biosciences Department, Teagasc Food Research Centre, Moorepark, Fermoy, County Cork, Ireland
| | - Elaine M Lawton
- Food Biosciences Department, Teagasc Food Research Centre, Moorepark, Fermoy, County Cork, Ireland
| | - Fiona Crispie
- Food Biosciences Department, Teagasc Food Research Centre, Moorepark, Fermoy, County Cork, Ireland
| | - Mònica Aguilera
- APC Microbiome Institute, University College Cork, Cork, Ireland
| | - Maurice Stanley
- APC Microbiome Institute, University College Cork, Cork, Ireland
- School of Biochemistry and Cell Biology, University College Cork, Cork, Ireland
| | - Serena Boscaini
- Food Biosciences Department, Teagasc Food Research Centre, Moorepark, Fermoy, County Cork, Ireland
- Department of Anatomy and Neurosciences, University College Cork, Cork, Ireland; and
| | - Susan Joyce
- APC Microbiome Institute, University College Cork, Cork, Ireland
- School of Biochemistry and Cell Biology, University College Cork, Cork, Ireland
| | - Silvia Melgar
- APC Microbiome Institute, University College Cork, Cork, Ireland
| | - John F Cryan
- APC Microbiome Institute, University College Cork, Cork, Ireland
- Department of Anatomy and Neurosciences, University College Cork, Cork, Ireland; and
| | - Paul D Cotter
- Food Biosciences Department, Teagasc Food Research Centre, Moorepark, Fermoy, County Cork, Ireland
- APC Microbiome Institute, University College Cork, Cork, Ireland
| |
Collapse
|
45
|
Kohno D. Sweet taste receptor in the hypothalamus: a potential new player in glucose sensing in the hypothalamus. J Physiol Sci 2017; 67:459-465. [PMID: 28378265 PMCID: PMC10717116 DOI: 10.1007/s12576-017-0535-y] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2017] [Accepted: 03/24/2017] [Indexed: 01/06/2023]
Abstract
The hypothalamic feeding center plays an important role in energy homeostasis. The feeding center senses the systemic energy status by detecting hormone and nutrient levels for homeostatic regulation, resulting in the control of food intake, heat production, and glucose production and uptake. The concentration of glucose is sensed by two types of glucose-sensing neurons in the feeding center: glucose-excited neurons and glucose-inhibited neurons. Previous studies have mainly focused on glucose metabolism as the mechanism underlying glucose sensing. Recent studies have indicated that receptor-mediated pathways also play a role in glucose sensing. This review describes sweet taste receptors in the hypothalamus and explores the role of sweet taste receptors in energy homeostasis.
Collapse
Affiliation(s)
- Daisuke Kohno
- Advanced Scientific Research Leaders Development Unit, Gunma University, 3-39-15 Showa-machi, Maebashi, Gunma, 371-8512, Japan.
- Metabolic Signal Research Center, Institute for Molecular and Cellular Regulation, Gunma University, Maebashi, 371-8512, Japan.
| |
Collapse
|
46
|
Miller GD. Appetite Regulation: Hormones, Peptides, and Neurotransmitters and Their Role in Obesity. Am J Lifestyle Med 2017; 13:586-601. [PMID: 31662725 DOI: 10.1177/1559827617716376] [Citation(s) in RCA: 68] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2017] [Revised: 05/18/2017] [Accepted: 05/31/2017] [Indexed: 12/29/2022] Open
Abstract
Understanding body weight regulation will aid in the development of new strategies to combat obesity. This review examines energy homeostasis and food intake behaviors, specifically with regards to hormones, peptides, and neurotransmitters in the periphery and central nervous system, and their potential role in obesity. Dysfunction in feeding signals by the brain is a factor in obesity. The hypothalamic (arcuate nucleus) and brainstem (nucleus tractus solitaris) areas integrate behavioral, endocrine, and autonomic responses via afferent and efferent pathways from and to the brainstem and peripheral organs. Neurons present in the arcuate nucleus express pro-opiomelanocortin, Neuropeptide Y, and Agouti Related Peptide, with the former involved in lowering food intake, and the latter two acutely increasing feeding behaviors. Action of peripheral hormones from the gut, pancreas, adipose, and liver are also involved in energy homeostasis. Vagal afferent neurons are also important in regulating energy homeostasis. Peripheral signals respond to the level of stored and currently available fuel. By studying their actions, new agents maybe developed that disable orexigenic responses and enhance anorexigenic signals. Although there are relatively few medications currently available for obesity treatment, a number of agents are in development that work through these pathways.
Collapse
Affiliation(s)
- Gary D Miller
- Department of Health and Exercise Science, Wake Forest University, Winston-Salem, North Carolina
| |
Collapse
|
47
|
Paeger L, Karakasilioti I, Altmüller J, Frommolt P, Brüning J, Kloppenburg P. Antagonistic modulation of NPY/AgRP and POMC neurons in the arcuate nucleus by noradrenalin. eLife 2017. [PMID: 28632132 PMCID: PMC5478265 DOI: 10.7554/elife.25770] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
In the arcuate nucleus of the hypothalamus (ARH) satiety signaling (anorexigenic) pro-opiomelanocortin (POMC)-expressing and hunger signaling (orexigenic) agouti-related peptide (AgRP)-expressing neurons are key components of the neuronal circuits that control food intake and energy homeostasis. Here, we assessed whether the catecholamine noradrenalin directly modulates the activity of these neurons in mice. Perforated patch clamp recordings showed that noradrenalin changes the activity of these functionally antagonistic neurons in opposite ways, increasing the activity of the orexigenic NPY/AgRP neurons and decreasing the activity of the anorexigenic POMC neurons. Cell type-specific transcriptomics and pharmacological experiments revealed that the opposing effect on these neurons is mediated by the activation of excitatory α1A - and β- adrenergic receptors in NPY/AgRP neurons, while POMC neurons are inhibited via α2A – adrenergic receptors. Thus, the coordinated differential modulation of the key hypothalamic neurons in control of energy homeostasis assigns noradrenalin an important role to promote feeding. DOI:http://dx.doi.org/10.7554/eLife.25770.001
Collapse
Affiliation(s)
- Lars Paeger
- Biocenter, Institute for Zoology, University of Cologne, Cologne, Germany.,Excellence Cluster on Cellular Stress Responses in Aging Associated Diseases, Cologne, Germany
| | - Ismene Karakasilioti
- Excellence Cluster on Cellular Stress Responses in Aging Associated Diseases, Cologne, Germany.,Max Planck Institute for Metabolism Research, Cologne, Germany.,Center of Molecular Medicine Cologne, University of Cologne, Cologne, Germany.,Center for Endocrinology, Diabetes and Preventive Medicine, University Hospital Cologne, Cologne, Germany
| | - Janine Altmüller
- Center of Molecular Medicine Cologne, University of Cologne, Cologne, Germany.,Cologne Center for Genomics, University of Cologne, Cologne, Germany
| | - Peter Frommolt
- Bioinformatics Facility, Excellence Cluster on Cellular Stress Responses in Aging Associated Diseases, Cologne, Germany
| | - Jens Brüning
- Excellence Cluster on Cellular Stress Responses in Aging Associated Diseases, Cologne, Germany.,Max Planck Institute for Metabolism Research, Cologne, Germany.,Center of Molecular Medicine Cologne, University of Cologne, Cologne, Germany.,Center for Endocrinology, Diabetes and Preventive Medicine, University Hospital Cologne, Cologne, Germany
| | - Peter Kloppenburg
- Biocenter, Institute for Zoology, University of Cologne, Cologne, Germany.,Excellence Cluster on Cellular Stress Responses in Aging Associated Diseases, Cologne, Germany
| |
Collapse
|
48
|
Involvement of CD36 in Modulating the Decrease of NPY and AgRP Induced by Acute Palmitic Acid Stimulation in N1E-115 Cells. Nutrients 2017. [PMID: 28629148 PMCID: PMC5490605 DOI: 10.3390/nu9060626] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Central nervous system (CNS) fatty acid sensing plays an important role in the regulation of food intake, and palmitic acid (PA) is the most important long chain fatty acid (LCFA) in the mammalian diet. To explore the effect of PA on central neuropeptide expression and the role of the cluster of the differentiation of 36 (CD36) in the process, N1E-115 cells were cultured with PA in the presence or absence of sulfosuccinimidyl-oleate (SSO), a CD36 inhibitor. Results showed that 10 μmol/L PA significantly reduced NPY and AgRP mRNA expression after 20 min of exposure, while the expression of CD36 was upregulated. The presence of SSO significantly attenuated the decrease of NPY and AgRP expression that was induced by PA alone, although no notable effect on PA- induced CD36 gene expression was observed. In conclusion, our study suggests the involvement of CD36 in the PA-induced decrease of NPY and AgRP in N1E-115 cells.
Collapse
|
49
|
Müller TD, Finan B, Clemmensen C, DiMarchi RD, Tschöp MH. The New Biology and Pharmacology of Glucagon. Physiol Rev 2017; 97:721-766. [PMID: 28275047 DOI: 10.1152/physrev.00025.2016] [Citation(s) in RCA: 261] [Impact Index Per Article: 32.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
In the last two decades we have witnessed sizable progress in defining the role of gastrointestinal signals in the control of glucose and energy homeostasis. Specifically, the molecular basis of the huge metabolic benefits in bariatric surgery is emerging while novel incretin-based medicines based on endogenous hormones such as glucagon-like peptide 1 and pancreas-derived amylin are improving diabetes management. These and related developments have fostered the discovery of novel insights into endocrine control of systemic metabolism, and in particular a deeper understanding of the importance of communication across vital organs, and specifically the gut-brain-pancreas-liver network. Paradoxically, the pancreatic peptide glucagon has reemerged in this period among a plethora of newly identified metabolic macromolecules, and new data complement and challenge its historical position as a gut hormone involved in metabolic control. The synthesis of glucagon analogs that are biophysically stable and soluble in aqueous solutions has promoted biological study that has enriched our understanding of glucagon biology and ironically recruited glucagon agonism as a central element to lower body weight in the treatment of metabolic disease. This review summarizes the extensive historical record and the more recent provocative direction that integrates the prominent role of glucagon in glucose elevation with its under-acknowledged effects on lipids, body weight, and vascular health that have implications for the pathophysiology of metabolic diseases, and the emergence of precision medicines to treat metabolic diseases.
Collapse
Affiliation(s)
- T D Müller
- Institute for Diabetes and Obesity, Helmholtz Diabetes Center, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany; German Center for Diabetes Research, Neuherberg, Germany; Department of Chemistry, Indiana University, Bloomington, Indiana; Division of Metabolic Diseases, Technische Universität München, Munich, Germany
| | - B Finan
- Institute for Diabetes and Obesity, Helmholtz Diabetes Center, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany; German Center for Diabetes Research, Neuherberg, Germany; Department of Chemistry, Indiana University, Bloomington, Indiana; Division of Metabolic Diseases, Technische Universität München, Munich, Germany
| | - C Clemmensen
- Institute for Diabetes and Obesity, Helmholtz Diabetes Center, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany; German Center for Diabetes Research, Neuherberg, Germany; Department of Chemistry, Indiana University, Bloomington, Indiana; Division of Metabolic Diseases, Technische Universität München, Munich, Germany
| | - R D DiMarchi
- Institute for Diabetes and Obesity, Helmholtz Diabetes Center, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany; German Center for Diabetes Research, Neuherberg, Germany; Department of Chemistry, Indiana University, Bloomington, Indiana; Division of Metabolic Diseases, Technische Universität München, Munich, Germany
| | - M H Tschöp
- Institute for Diabetes and Obesity, Helmholtz Diabetes Center, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany; German Center for Diabetes Research, Neuherberg, Germany; Department of Chemistry, Indiana University, Bloomington, Indiana; Division of Metabolic Diseases, Technische Universität München, Munich, Germany
| |
Collapse
|
50
|
Chamarthi B, Cincotta AH. Effect of bromocriptine-QR therapy on glycemic control in subjects with type 2 diabetes mellitus whose dysglycemia is inadequately controlled on insulin. Postgrad Med 2017; 129:446-455. [DOI: 10.1080/00325481.2017.1315290] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Affiliation(s)
- Bindu Chamarthi
- VeroScience LLC, Tiverton, RI, USA
- Division of Endocrinology, Diabetes and Hypertension, Brigham and Women’s Hospital, Boston, MA, USA
- Department of Medicine, Harvard Medical School, Boston, MA, USA
| | | |
Collapse
|