51
|
Yuan J, Gilbert ER, Cline MA. The central anorexigenic mechanism of amylin in Japanese quail ( Coturnix japonica ) involves pro-opiomelanocortin, calcitonin receptor, and the arcuate nucleus of the hypothalamus. Comp Biochem Physiol A Mol Integr Physiol 2017; 210:28-34. [DOI: 10.1016/j.cbpa.2017.05.011] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2017] [Revised: 05/19/2017] [Accepted: 05/22/2017] [Indexed: 01/10/2023]
|
52
|
Chhabra KH, Morgan DA, Tooke BP, Adams JM, Rahmouni K, Low MJ. Reduced renal sympathetic nerve activity contributes to elevated glycosuria and improved glucose tolerance in hypothalamus-specific Pomc knockout mice. Mol Metab 2017; 6:1274-1285. [PMID: 29031726 PMCID: PMC5641634 DOI: 10.1016/j.molmet.2017.07.005] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/16/2017] [Revised: 07/02/2017] [Accepted: 07/10/2017] [Indexed: 11/11/2022] Open
Abstract
Objective Hypothalamic arcuate nucleus-specific pro-opiomelanocortin deficient (ArcPomc−/−) mice exhibit improved glucose tolerance despite massive obesity and insulin resistance. We demonstrated previously that their improved glucose tolerance is due to elevated glycosuria. However, the underlying mechanisms that link glucose reabsorption in the kidney with ArcPomc remain unclear. Given the function of the hypothalamic melanocortin system in controlling sympathetic outflow, we hypothesized that reduced renal sympathetic nerve activity (RSNA) in ArcPomc−/− mice could explain their elevated glycosuria and consequent enhanced glucose tolerance. Methods We measured RSNA by multifiber recording directly from the nerves innervating the kidneys in ArcPomc−/− mice. To further validate the function of RSNA in glucose reabsorption, we denervated the kidneys of WT and diabetic db/db mice before measuring their glucose tolerance and urine glucose levels. Moreover, we performed western blot and immunohistochemistry to determine kidney GLUT2 and SGLT2 levels in either ArcPomc−/− mice or the renal-denervated mice. Results Consistent with our hypothesis, we found that basal RSNA was decreased in ArcPomc−/− mice relative to their wild type (WT) littermates. Remarkably, both WT and db/db mice exhibited elevated glycosuria and improved glucose tolerance after renal denervation. The elevated glycosuria in obese ArcPomc−/−, WT and db/db mice was due to reduced renal GLUT2 levels in the proximal tubules. Overall, we show that renal-denervated WT and diabetic mice recapitulate the phenotype of improved glucose tolerance and elevated glycosuria associated with reduced renal GLUT2 levels observed in obese ArcPomc−/− mice. Conclusion Hence, we conclude that ArcPomc is essential in maintaining basal RSNA and that elevated glycosuria is a possible mechanism to explain improved glucose tolerance after renal denervation in drug resistant hypertensive patients. Hypothalamic POMC is essential in maintaining basal renal sympathetic nerve activity. Renal denervation improves glucose tolerance in wild-type and db/db mice by elevating their glycosuria. Decreased renal GLUT2 is responsible for elevated glycosuria in mice with suppressed renal sympathetic nerve activity.
Collapse
Affiliation(s)
- Kavaljit H Chhabra
- Department of Molecular and Integrative Physiology, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Donald A Morgan
- Department of Pharmacology, University of Iowa, Iowa City, IA, USA
| | - Benjamin P Tooke
- Department of Molecular and Integrative Physiology, University of Michigan Medical School, Ann Arbor, MI, USA; Case Western Reserve University, Cleveland, OH, USA
| | - Jessica M Adams
- Department of Molecular and Integrative Physiology, University of Michigan Medical School, Ann Arbor, MI, USA; Department of Pediatrics and Communicable Diseases, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Kamal Rahmouni
- Department of Pharmacology, University of Iowa, Iowa City, IA, USA
| | - Malcolm J Low
- Department of Molecular and Integrative Physiology, University of Michigan Medical School, Ann Arbor, MI, USA; Department of Internal Medicine, Division of Metabolism, Endocrinology and Diabetes, University of Michigan Medical School, Ann Arbor, MI, USA.
| |
Collapse
|
53
|
Riera CE, Tsaousidou E, Halloran J, Follett P, Hahn O, Pereira MMA, Ruud LE, Alber J, Tharp K, Anderson CM, Brönneke H, Hampel B, Filho CDDM, Stahl A, Brüning JC, Dillin A. The Sense of Smell Impacts Metabolic Health and Obesity. Cell Metab 2017; 26:198-211.e5. [PMID: 28683287 DOI: 10.1016/j.cmet.2017.06.015] [Citation(s) in RCA: 126] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/07/2016] [Revised: 04/09/2017] [Accepted: 06/16/2017] [Indexed: 01/09/2023]
Abstract
Olfactory inputs help coordinate food appreciation and selection, but their role in systemic physiology and energy balance is poorly understood. Here we demonstrate that mice upon conditional ablation of mature olfactory sensory neurons (OSNs) are resistant to diet-induced obesity accompanied by increased thermogenesis in brown and inguinal fat depots. Acute loss of smell perception after obesity onset not only abrogated further weight gain but also improved fat mass and insulin resistance. Reduced olfactory input stimulates sympathetic nerve activity, resulting in activation of β-adrenergic receptors on white and brown adipocytes to promote lipolysis. Conversely, conditional ablation of the IGF1 receptor in OSNs enhances olfactory performance in mice and leads to increased adiposity and insulin resistance. These findings unravel a new bidirectional function for the olfactory system in controlling energy homeostasis in response to sensory and hormonal signals.
Collapse
Affiliation(s)
- Celine E Riera
- Howard Hughes Medical Institute and Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA, USA; The Paul F. Glenn Center for Aging Research, University of California, Berkeley, Berkeley, CA, USA; Diabetes and Obesity Research Institute, Cedars-Sinai Medical Center, 8700 Beverly Boulevard, Los Angeles, CA, USA
| | - Eva Tsaousidou
- Department of Neuronal Control of Metabolism, Max Planck Institute for Metabolism Research, Gleueler Strasse 50, Cologne, Germany; Center for Endocrinology, Diabetes and Preventive Medicine (CEDP), University Hospital Cologne, Kerpener Strasse 26, Cologne, Germany; Max Planck Institute for Biology of Ageing, Cologne, Germany and Excellence Cluster on Cellular Stress Responses in Aging Associated Diseases (CECAD) Cologne, Germany; Department of Genetics and Complex Diseases and Sabri Ülker Center, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Jonathan Halloran
- Howard Hughes Medical Institute and Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA, USA; The Paul F. Glenn Center for Aging Research, University of California, Berkeley, Berkeley, CA, USA
| | - Patricia Follett
- The Salk Institute for Biological Studies, 10010 North Torrey Pines Road, La Jolla, CA, USA
| | - Oliver Hahn
- Max Planck Institute for Biology of Ageing, Cologne, Germany and Excellence Cluster on Cellular Stress Responses in Aging Associated Diseases (CECAD) Cologne, Germany
| | - Mafalda M A Pereira
- Department of Neuronal Control of Metabolism, Max Planck Institute for Metabolism Research, Gleueler Strasse 50, Cologne, Germany; Center for Endocrinology, Diabetes and Preventive Medicine (CEDP), University Hospital Cologne, Kerpener Strasse 26, Cologne, Germany
| | - Linda Engström Ruud
- Department of Neuronal Control of Metabolism, Max Planck Institute for Metabolism Research, Gleueler Strasse 50, Cologne, Germany; Center for Endocrinology, Diabetes and Preventive Medicine (CEDP), University Hospital Cologne, Kerpener Strasse 26, Cologne, Germany
| | - Jens Alber
- Department of Neuronal Control of Metabolism, Max Planck Institute for Metabolism Research, Gleueler Strasse 50, Cologne, Germany; Center for Endocrinology, Diabetes and Preventive Medicine (CEDP), University Hospital Cologne, Kerpener Strasse 26, Cologne, Germany
| | - Kevin Tharp
- Nutritional Sciences and Toxicology, University of California, Berkeley, Berkeley, CA, USA
| | - Courtney M Anderson
- Nutritional Sciences and Toxicology, University of California, Berkeley, Berkeley, CA, USA
| | - Hella Brönneke
- Department of Neuronal Control of Metabolism, Max Planck Institute for Metabolism Research, Gleueler Strasse 50, Cologne, Germany; Center for Endocrinology, Diabetes and Preventive Medicine (CEDP), University Hospital Cologne, Kerpener Strasse 26, Cologne, Germany
| | - Brigitte Hampel
- Department of Neuronal Control of Metabolism, Max Planck Institute for Metabolism Research, Gleueler Strasse 50, Cologne, Germany; Center for Endocrinology, Diabetes and Preventive Medicine (CEDP), University Hospital Cologne, Kerpener Strasse 26, Cologne, Germany
| | | | - Andreas Stahl
- Nutritional Sciences and Toxicology, University of California, Berkeley, Berkeley, CA, USA
| | - Jens C Brüning
- Department of Neuronal Control of Metabolism, Max Planck Institute for Metabolism Research, Gleueler Strasse 50, Cologne, Germany; Center for Endocrinology, Diabetes and Preventive Medicine (CEDP), University Hospital Cologne, Kerpener Strasse 26, Cologne, Germany; Max Planck Institute for Biology of Ageing, Cologne, Germany and Excellence Cluster on Cellular Stress Responses in Aging Associated Diseases (CECAD) Cologne, Germany.
| | - Andrew Dillin
- Howard Hughes Medical Institute and Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA, USA; The Paul F. Glenn Center for Aging Research, University of California, Berkeley, Berkeley, CA, USA.
| |
Collapse
|
54
|
Rouault AAJ, Srinivasan DK, Yin TC, Lee AA, Sebag JA. Melanocortin Receptor Accessory Proteins (MRAPs): Functions in the melanocortin system and beyond. Biochim Biophys Acta Mol Basis Dis 2017; 1863:2462-2467. [PMID: 28499989 DOI: 10.1016/j.bbadis.2017.05.008] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2017] [Revised: 05/02/2017] [Accepted: 05/05/2017] [Indexed: 12/27/2022]
Abstract
G-protein coupled receptors (GPCRs) are regulated by numerous proteins including kinases, G-proteins, β-arrestins and accessory proteins. Several families of GPCR accessory proteins like Receptor Activity Modifying Proteins, Receptor Transporting Proteins and Melanocortin Receptor Accessory Proteins (MRAPs) have been identified as regulator of receptor trafficking, signaling and ligand specificity. The MRAP family contains two members, MRAP1 and MRAP2, responsible for the formation of a functional ACTH receptor and for the regulation of energy homeostasis respectively. Like all known GPCR accessory proteins, MRAPs are single transmembrane proteins, however, they form a unique structure since they assemble as an anti-parallel homodimer. Moreover, the accepted idea that MRAPs are specific regulators of melanocortin receptors was recently challenged by the discovery that MRAP2 inhibits the activity of prokineticin receptors. Recent studies are starting to explain the role of the unusual structure of MRAPs and to illustrate the importance of MRAP2 for the maintenance of both energy and glucose homeostasis. This article is part of a Special Issue entitled: Melanocortin Receptors - edited by Ya-Xiong Tao.
Collapse
Affiliation(s)
- Alix A J Rouault
- University of Iowa, Carver College of Medicine, Department of Molecular Physiology and Biophysics, Fraternal Order of Eagle Diabetes Research Center, Iowa City, IA 52242, United States
| | - Dinesh K Srinivasan
- University of Iowa, Carver College of Medicine, Department of Molecular Physiology and Biophysics, Fraternal Order of Eagle Diabetes Research Center, Iowa City, IA 52242, United States
| | - Terry C Yin
- University of Iowa, Carver College of Medicine, Department of Molecular Physiology and Biophysics, Fraternal Order of Eagle Diabetes Research Center, Iowa City, IA 52242, United States
| | - Abigail A Lee
- University of Iowa, Carver College of Medicine, Department of Molecular Physiology and Biophysics, Fraternal Order of Eagle Diabetes Research Center, Iowa City, IA 52242, United States
| | - Julien A Sebag
- University of Iowa, Carver College of Medicine, Department of Molecular Physiology and Biophysics, Fraternal Order of Eagle Diabetes Research Center, Iowa City, IA 52242, United States.
| |
Collapse
|
55
|
Xiao Y, Deng Y, Yuan F, Xia T, Liu H, Li Z, Liu Z, Ying H, Liu Y, Zhai Q, Chen S, Guo F. ATF4/ATG5 Signaling in Hypothalamic Proopiomelanocortin Neurons Regulates Fat Mass via Affecting Energy Expenditure. Diabetes 2017; 66:1146-1158. [PMID: 28213613 DOI: 10.2337/db16-1546] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/15/2016] [Accepted: 02/13/2017] [Indexed: 11/13/2022]
Abstract
Although many biological functions of activating transcription factor 4 (ATF4) have been identified, a role of hypothalamic ATF4 in the regulation of energy homeostasis is poorly understood. In this study, we showed that hypothalamic proopiomelanocortin (POMC) neuron-specific ATF4 knockout (PAKO) mice are lean and have higher energy expenditure. Furthermore, PAKO mice were resistant to high-fat diet-induced obesity, glucose intolerance, and leptin resistance. Moreover, the expression of autophagy protein 5 (ATG5) was increased or decreased by ATF4 knockdown or overexpression, respectively, and ATF4 inhibited the transcription of ATG5 by binding to the basic zipper-containing protein sites on its promoter. Importantly, mice with double knockout of ATF4 and ATG5 in POMC neurons gained more fat mass and reduced energy expenditure compared with PAKO mice under a high-fat diet. Finally, the effect of ATF4 deletion in POMC neurons was possibly mediated via enhanced ATG5-dependent autophagy and α-melanocyte-stimulating hormone production in the hypothalamus. Taken together, these results identify the beneficial role of hypothalamic ATF4/ATG5 axis in the regulation of energy expenditure, obesity, and obesity-related metabolic disorders, which suggests that ATF4/ATG5 axis in the hypothalamus may be a new potential therapeutic target for treating obesity and obesity-related metabolic diseases.
Collapse
Affiliation(s)
- Yuzhong Xiao
- Key Laboratory of Nutrition and Metabolism, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, China
| | - Yalan Deng
- Key Laboratory of Nutrition and Metabolism, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, China
| | - Feixiang Yuan
- Key Laboratory of Nutrition and Metabolism, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, China
| | - Tingting Xia
- Key Laboratory of Nutrition and Metabolism, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, China
| | - Hao Liu
- Key Laboratory of Nutrition and Metabolism, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, China
| | - Zhigang Li
- Key Laboratory of Nutrition and Metabolism, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, China
| | - Zhixue Liu
- Key Laboratory of Nutrition and Metabolism, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, China
| | - Hao Ying
- Key Laboratory of Nutrition and Metabolism, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, China
| | - Yi Liu
- Key Laboratory of Nutrition and Metabolism, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, China
| | - Qiwei Zhai
- Key Laboratory of Nutrition and Metabolism, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, China
| | - Shanghai Chen
- Key Laboratory of Nutrition and Metabolism, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, China
| | - Feifan Guo
- Key Laboratory of Nutrition and Metabolism, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, China
| |
Collapse
|
56
|
Shipp SL, Wang G, Cline MA, Gilbert ER. Chick subcutaneous and abdominal adipose tissue depots respond differently in lipolytic and adipogenic activity to α-melanocyte stimulating hormone (α-MSH). Comp Biochem Physiol A Mol Integr Physiol 2017; 209:56-64. [PMID: 28438719 DOI: 10.1016/j.cbpa.2017.04.013] [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: 01/21/2017] [Revised: 04/13/2017] [Accepted: 04/13/2017] [Indexed: 12/12/2022]
Abstract
In birds, α-MSH is anorexigenic, but effects on adipose tissue are unknown. Four day-old chicks were intraperitoneally injected with 0 (vehicle), 5, 10, or 50μg of α-MSH and subcutaneous and abdominal adipose tissue collected at 60min for RNA isolation (n=10). Plasma was collected post-euthanasia at 60 and 180min for measuring non-esterified fatty acids (NEFA) and α-MSH (n=10). Relative to the vehicle, food intake was reduced in the 50μg-treated group. Plasma NEFAs were greater in 10μg than vehicle-treated chicks at 3h. Plasma α-MSH was 3.06±0.57ng/ml. In subcutaneous tissue, melanocortin receptor 5 (MC5R) mRNA was increased in 10μg, MC2R and CCAAT-enhancer-binding protein β (C/EBPβ) mRNAs increased in 50μg, peroxisome proliferator-activated receptor γ and C/EBPα decreased in 5, 10 and 50μg, and Ki67 mRNA decreased in 50μg α-MSH-injected chicks, compared to vehicle-injected chicks. In abdominal tissue, adipose triglyceride lipase mRNA was greater in 10μg α-MSH- than vehicle-treated chicks. Cells isolated from abdominal fat that were treated with 10 and 100nM α-MSH for 4h expressed more MC5R and perilipin-1 than control cells (n=6). Cells that received 100nM α-MSH expressed more fatty acid binding protein 4 and comparative gene identification-58 mRNA than control cells. Glycerol-3-phosphate dehydrogenase (G3PDH) activity was greater in cells at 9days post-differentiation that were treated with 1 and 100nM α-MSH for 4h than in control cells (n=3). Results suggest that α-MSH increases lipolysis and reduces adipogenesis in adipose tissue.
Collapse
Affiliation(s)
- Steven L Shipp
- Department of Animal and Poultry Sciences, Virginia Tech, Blacksburg, VA 24061, United States
| | - Guoqing Wang
- Department of Animal and Poultry Sciences, Virginia Tech, Blacksburg, VA 24061, United States
| | - Mark A Cline
- Department of Animal and Poultry Sciences, Virginia Tech, Blacksburg, VA 24061, United States
| | - Elizabeth R Gilbert
- Department of Animal and Poultry Sciences, Virginia Tech, Blacksburg, VA 24061, United States.
| |
Collapse
|
57
|
Dorfman MD, Krull JE, Scarlett JM, Guyenet SJ, Sajan MP, Damian V, Nguyen HT, Leitges M, Morton GJ, Farese RV, Schwartz MW, Thaler JP. Deletion of Protein Kinase C λ in POMC Neurons Predisposes to Diet-Induced Obesity. Diabetes 2017; 66:920-934. [PMID: 28073831 PMCID: PMC5360303 DOI: 10.2337/db16-0482] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/18/2016] [Accepted: 01/02/2017] [Indexed: 12/16/2022]
Abstract
Effectors of the phosphoinositide 3-kinase (PI3K) signal transduction pathway contribute to the hypothalamic regulation of energy and glucose homeostasis in divergent ways. Here we show that central nervous system (CNS) action of the PI3K signaling intermediate atypical protein kinase C (aPKC) constrains food intake, weight gain, and glucose intolerance in both rats and mice. Pharmacological inhibition of CNS aPKC activity acutely increases food intake and worsens glucose tolerance in chow-fed rodents and causes excess weight gain during high-fat diet (HFD) feeding. Similarly, selective deletion of the aPKC isoform Pkc-λ in proopiomelanocortin (POMC) neurons disrupts leptin action, reduces melanocortin content in the paraventricular nucleus, and markedly increases susceptibility to obesity, glucose intolerance, and insulin resistance specifically in HFD-fed male mice. These data implicate aPKC as a novel regulator of energy and glucose homeostasis downstream of the leptin-PI3K pathway in POMC neurons.
Collapse
Affiliation(s)
- Mauricio D Dorfman
- UW Diabetes Institute and Department of Medicine, University of Washington, Seattle, WA
| | - Jordan E Krull
- UW Diabetes Institute and Department of Medicine, University of Washington, Seattle, WA
| | - Jarrad M Scarlett
- UW Diabetes Institute and Department of Medicine, University of Washington, Seattle, WA
| | - Stephan J Guyenet
- UW Diabetes Institute and Department of Medicine, University of Washington, Seattle, WA
| | - Mini P Sajan
- Department of Internal Medicine, University of South Florida College of Medicine, Tampa, FL
- Research & Internal Medicine Services, James A. Haley VA Medical Center, Tampa, FL
| | - Vincent Damian
- UW Diabetes Institute and Department of Medicine, University of Washington, Seattle, WA
| | - Hong T Nguyen
- UW Diabetes Institute and Department of Medicine, University of Washington, Seattle, WA
| | - Michael Leitges
- The Biotechnology Centre of Oslo, University of Oslo, Oslo, Norway
| | - Gregory J Morton
- UW Diabetes Institute and Department of Medicine, University of Washington, Seattle, WA
| | - Robert V Farese
- Department of Internal Medicine, University of South Florida College of Medicine, Tampa, FL
- Research & Internal Medicine Services, James A. Haley VA Medical Center, Tampa, FL
| | - Michael W Schwartz
- UW Diabetes Institute and Department of Medicine, University of Washington, Seattle, WA
| | - Joshua P Thaler
- UW Diabetes Institute and Department of Medicine, University of Washington, Seattle, WA
| |
Collapse
|
58
|
Klein MO, MacKay H, Edwards A, Park S, Kiss ACI, Felicio LF, Abizaid A. POMC and NPY mRNA expression during development is increased in rat offspring brain from mothers fed with a high fat diet. Int J Dev Neurosci 2017; 64:14-20. [DOI: 10.1016/j.ijdevneu.2017.03.004] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2016] [Revised: 02/21/2017] [Accepted: 03/10/2017] [Indexed: 01/06/2023] Open
Affiliation(s)
- Marianne Orlandini Klein
- Department of NeuroscienceCarleton UniversityOttawaONCanada
- Department of PharmacologyInstitute of Biomedical Science, University of São PauloSão PauloSPBrazil
| | - Harry MacKay
- Department of NeuroscienceCarleton UniversityOttawaONCanada
| | | | - Su‐Bin Park
- Department of NeuroscienceCarleton UniversityOttawaONCanada
| | | | - Luciano Freitas Felicio
- Department of PharmacologyInstitute of Biomedical Science, University of São PauloSão PauloSPBrazil
- Department of PathologySchool of Veterinary Medicine, University of São PauloSão PauloSPBrazil
| | | |
Collapse
|
59
|
Wang L, Sui L, Panigrahi SK, Meece K, Xin Y, Kim J, Gromada J, Doege CA, Wardlaw SL, Egli D, Leibel RL. PC1/3 Deficiency Impacts Pro-opiomelanocortin Processing in Human Embryonic Stem Cell-Derived Hypothalamic Neurons. Stem Cell Reports 2017; 8:264-277. [PMID: 28132887 PMCID: PMC5312251 DOI: 10.1016/j.stemcr.2016.12.021] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2016] [Revised: 12/20/2016] [Accepted: 12/21/2016] [Indexed: 12/19/2022] Open
Abstract
We recently developed a technique for generating hypothalamic neurons from human pluripotent stem cells. Here, as proof of principle, we examine the use of these cells in modeling of a monogenic form of severe obesity: PCSK1 deficiency. The cognate enzyme, PC1/3, processes many prohormones in neuroendocrine and other tissues. We generated PCSK1 (PC1/3)-deficient human embryonic stem cell (hESC) lines using both short hairpin RNA and CRISPR-Cas9, and investigated pro-opiomelanocortin (POMC) processing using hESC-differentiated hypothalamic neurons. The increased levels of unprocessed POMC and the decreased ratios (relative to POMC) of processed POMC-derived peptides in both PCSK1 knockdown and knockout hESC-derived neurons phenocopied POMC processing reported in PC1/3-null mice and PC1/3-deficient patients. PC1/3 deficiency was associated with increased expression of melanocortin receptors and PRCP (prolylcarboxypeptidase, a catabolic enzyme for α-melanocyte stimulating hormone (αMSH)), and reduced adrenocorticotropic hormone secretion. We conclude that the obesity accompanying PCSK1 deficiency may not be primarily due to αMSH deficiency. Stem cell-derived hypothalamic neurons are used to study human obesity shRNA and CRISPR-Cas9 were used to generate models of PCSK1 deficiency PC1/3 deficiency impaired POMC processing in arcuate-like neurons Adaptive changes occurred in “downstream” POMC processing enzymes
Collapse
Affiliation(s)
- Liheng Wang
- Division of Molecular Genetics, Department of Pediatrics and Naomi Berrie Diabetes Center, College of Physicians and Surgeons, Columbia University, 1150 St. Nicholas Avenue, Room 620A, New York, NY 10032, USA; Department of Medicine and Naomi Berrie Diabetes Center, College of Physicians and Surgeons, Columbia University, New York, NY 10032, USA
| | - Lina Sui
- Division of Molecular Genetics, Department of Pediatrics and Naomi Berrie Diabetes Center, College of Physicians and Surgeons, Columbia University, 1150 St. Nicholas Avenue, Room 620A, New York, NY 10032, USA
| | - Sunil K Panigrahi
- Department of Medicine and Naomi Berrie Diabetes Center, College of Physicians and Surgeons, Columbia University, New York, NY 10032, USA
| | - Kana Meece
- Department of Medicine and Naomi Berrie Diabetes Center, College of Physicians and Surgeons, Columbia University, New York, NY 10032, USA
| | - Yurong Xin
- Regeneron Pharmaceuticals, Tarrytown, NY 10591, USA
| | - Jinrang Kim
- Regeneron Pharmaceuticals, Tarrytown, NY 10591, USA
| | | | - Claudia A Doege
- Department of Pathology and Cell Biology and Naomi Berrie Diabetes Center, College of Physicians and Surgeons, Columbia University, New York, NY 10032, USA
| | - Sharon L Wardlaw
- Department of Medicine and Naomi Berrie Diabetes Center, College of Physicians and Surgeons, Columbia University, New York, NY 10032, USA
| | - Dieter Egli
- Division of Molecular Genetics, Department of Pediatrics and Naomi Berrie Diabetes Center, College of Physicians and Surgeons, Columbia University, 1150 St. Nicholas Avenue, Room 620A, New York, NY 10032, USA; New York Stem Cell Foundation Research Institute, 3960 Broadway, New York, NY 10032, USA
| | - Rudolph L Leibel
- Division of Molecular Genetics, Department of Pediatrics and Naomi Berrie Diabetes Center, College of Physicians and Surgeons, Columbia University, 1150 St. Nicholas Avenue, Room 620A, New York, NY 10032, USA; Department of Medicine and Naomi Berrie Diabetes Center, College of Physicians and Surgeons, Columbia University, New York, NY 10032, USA.
| |
Collapse
|
60
|
Burnett LC, LeDuc CA, Sulsona CR, Paull D, Rausch R, Eddiry S, Carli JFM, Morabito MV, Skowronski AA, Hubner G, Zimmer M, Wang L, Day R, Levy B, Fennoy I, Dubern B, Poitou C, Clement K, Butler MG, Rosenbaum M, Salles JP, Tauber M, Driscoll DJ, Egli D, Leibel RL. Deficiency in prohormone convertase PC1 impairs prohormone processing in Prader-Willi syndrome. J Clin Invest 2017; 127:293-305. [PMID: 27941249 PMCID: PMC5199710 DOI: 10.1172/jci88648] [Citation(s) in RCA: 104] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2016] [Accepted: 10/20/2016] [Indexed: 12/17/2022] Open
Abstract
Prader-Willi syndrome (PWS) is caused by a loss of paternally expressed genes in an imprinted region of chromosome 15q. Among the canonical PWS phenotypes are hyperphagic obesity, central hypogonadism, and low growth hormone (GH). Rare microdeletions in PWS patients define a 91-kb minimum critical deletion region encompassing 3 genes, including the noncoding RNA gene SNORD116. Here, we found that protein and transcript levels of nescient helix loop helix 2 (NHLH2) and the prohormone convertase PC1 (encoded by PCSK1) were reduced in PWS patient induced pluripotent stem cell-derived (iPSC-derived) neurons. Moreover, Nhlh2 and Pcsk1 expression were reduced in hypothalami of fasted Snord116 paternal knockout (Snord116p-/m+) mice. Hypothalamic Agrp and Npy remained elevated following refeeding in association with relative hyperphagia in Snord116p-/m+ mice. Nhlh2-deficient mice display growth deficiencies as adolescents and hypogonadism, hyperphagia, and obesity as adults. Nhlh2 has also been shown to promote Pcsk1 expression. Humans and mice deficient in PC1 display hyperphagic obesity, hypogonadism, decreased GH, and hypoinsulinemic diabetes due to impaired prohormone processing. Here, we found that Snord116p-/m+ mice displayed in vivo functional defects in prohormone processing of proinsulin, pro-GH-releasing hormone, and proghrelin in association with reductions in islet, hypothalamic, and stomach PC1 content. Our findings suggest that the major neuroendocrine features of PWS are due to PC1 deficiency.
Collapse
Affiliation(s)
- Lisa C. Burnett
- Institute of Human Nutrition
- Department of Pediatrics, Division of Molecular Genetics, and
- Naomi Berrie Diabetes Center, Columbia University, New York, New York, USA
| | - Charles A. LeDuc
- Department of Pediatrics, Division of Molecular Genetics, and
- Naomi Berrie Diabetes Center, Columbia University, New York, New York, USA
- New York Obesity Research Center, New York, New York, USA
| | - Carlos R. Sulsona
- Department of Pediatrics, Division of Genetics and Metabolism, University of Florida College of Medicine Gainesville, Florida, USA
| | - Daniel Paull
- The New York Stem Cell Foundation Research Institute, New York, New York, USA
| | - Richard Rausch
- Department of Pediatrics, Division of Molecular Genetics, and
- Naomi Berrie Diabetes Center, Columbia University, New York, New York, USA
| | - Sanaa Eddiry
- Centre de Physiopathologie de Toulouse-Purpan, Université de Toulouse, CNRS UMR 5282, INSERM UMR 1043, Université Paul Sabatier, Toulouse, France
| | - Jayne F. Martin Carli
- Department of Pediatrics, Division of Molecular Genetics, and
- Naomi Berrie Diabetes Center, Columbia University, New York, New York, USA
- Department of Biochemistry and Molecular Biophysics, Columbia University, New York, New York, USA
| | - Michael V. Morabito
- Department of Pediatrics, Division of Molecular Genetics, and
- Naomi Berrie Diabetes Center, Columbia University, New York, New York, USA
| | - Alicja A. Skowronski
- Institute of Human Nutrition
- Department of Pediatrics, Division of Molecular Genetics, and
- Naomi Berrie Diabetes Center, Columbia University, New York, New York, USA
| | | | - Matthew Zimmer
- The New York Stem Cell Foundation Research Institute, New York, New York, USA
| | - Liheng Wang
- Department of Pediatrics, Division of Molecular Genetics, and
- Naomi Berrie Diabetes Center, Columbia University, New York, New York, USA
| | - Robert Day
- Institut de pharmacologie de Sherbrooke, Department of Surgery, Division of Urology, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, Quebec, Canada
| | - Brynn Levy
- Department of Pathology and Cell Biology, Columbia University, New York, New York, USA
| | - Ilene Fennoy
- Department of Pediatrics, Division of Pediatric Diabetes, Endocrinology and Metabolism, Columbia University, New York, New York, USA
| | - Beatrice Dubern
- Institute of Cardiometabolism and Nutrition, Assistance Publique Hôpitaux de Paris, Sorbonne University, University Pierre et Marie-Curie, INSERM UMRS 1166, Paris, France
| | - Christine Poitou
- Institute of Cardiometabolism and Nutrition, Assistance Publique Hôpitaux de Paris, Sorbonne University, University Pierre et Marie-Curie, INSERM UMRS 1166, Paris, France
| | - Karine Clement
- Institute of Cardiometabolism and Nutrition, Assistance Publique Hôpitaux de Paris, Sorbonne University, University Pierre et Marie-Curie, INSERM UMRS 1166, Paris, France
| | - Merlin G. Butler
- Department of Psychiatry and Behavioral Sciences, Division of Research and Genetics, Kansas University Medical Center, Kansas City, Kansas, USA
| | - Michael Rosenbaum
- Department of Pediatrics, Division of Molecular Genetics, and
- Naomi Berrie Diabetes Center, Columbia University, New York, New York, USA
| | - Jean Pierre Salles
- Centre de Physiopathologie de Toulouse-Purpan, Université de Toulouse, CNRS UMR 5282, INSERM UMR 1043, Université Paul Sabatier, Toulouse, France
- Unité d’Endocrinologie, Hôpital des Enfants, and
| | - Maithe Tauber
- Centre de Physiopathologie de Toulouse-Purpan, Université de Toulouse, CNRS UMR 5282, INSERM UMR 1043, Université Paul Sabatier, Toulouse, France
- Unité d’Endocrinologie, Hôpital des Enfants, and
- Centre de Référence du Syndrome de Prader-Willi, CHU Toulouse, Toulouse, France
| | - Daniel J. Driscoll
- Department of Pediatrics, Division of Genetics and Metabolism, University of Florida College of Medicine Gainesville, Florida, USA
- Center for Epigenetics, University of Florida College of Medicine, Gainesville, Florida, USA
| | - Dieter Egli
- Department of Pediatrics, Division of Molecular Genetics, and
- Naomi Berrie Diabetes Center, Columbia University, New York, New York, USA
- The New York Stem Cell Foundation Research Institute, New York, New York, USA
| | - Rudolph L. Leibel
- Department of Pediatrics, Division of Molecular Genetics, and
- Naomi Berrie Diabetes Center, Columbia University, New York, New York, USA
- New York Obesity Research Center, New York, New York, USA
| |
Collapse
|
61
|
Chernysheva MP, Nozdrachev AD. Neuroendocrine hypothalamus as a homeostat of endogenous time. J EVOL BIOCHEM PHYS+ 2017. [DOI: 10.1134/s002209301701001x] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
62
|
Engin A. Diet-Induced Obesity and the Mechanism of Leptin Resistance. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2017; 960:381-397. [PMID: 28585208 DOI: 10.1007/978-3-319-48382-5_16] [Citation(s) in RCA: 49] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Leptin signaling blockade by chronic overstimulation of the leptin receptor or hypothalamic pro-inflammatory responses due to elevated levels of saturated fatty acid can induce leptin resistance by activating negative feedback pathways. Although, long form leptin receptor (Ob-Rb) initiates leptin signaling through more than seven different signal transduction pathways, excessive suppressor of cytokine signaling-3 (SOCS-3) activity is a potential mechanism for the leptin resistance that characterizes human obesity. Because the leptin-responsive metabolic pathways broadly integrate with other neurons to control energy balance, the methods used to counteract the leptin resistance has extremely limited effect. In this chapter, besides the impairment of central and peripheral leptin signaling pathways, limited access of leptin to central nervous system (CNS) through blood-brain barrier, mismatch between high leptin and the amount of leptin receptor expression, contradictory effects of cellular and circulating molecules on leptin signaling, the connection between leptin signaling and endoplasmic reticulum (ER) stress and self-regulation of leptin signaling has been discussed in terms of leptin resistance.
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
|
63
|
Navarro M. The Role of the Melanocortin System in Drug and Alcohol Abuse. INTERNATIONAL REVIEW OF NEUROBIOLOGY 2017; 136:121-150. [DOI: 10.1016/bs.irn.2017.06.009] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
|
64
|
Page-Wilson G, Nguyen KT, Atalayer D, Meece K, Bainbridge HA, Korner J, Gordon RJ, Panigrahi SK, White A, Smiley R, Wardlaw SL. Evaluation of CSF and plasma biomarkers of brain melanocortin activity in response to caloric restriction in humans. Am J Physiol Endocrinol Metab 2017; 312:E19-E26. [PMID: 27894065 PMCID: PMC5283881 DOI: 10.1152/ajpendo.00330.2016] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/29/2016] [Accepted: 11/11/2016] [Indexed: 11/22/2022]
Abstract
The melanocortin neuronal system, which consists of hypothalamic proopiomelanocortin (POMC) and agouti-related protein (AgRP) neurons, is a leptin target that regulates energy balance and metabolism, but studies in humans are limited by a lack of reliable biomarkers to assess brain melanocortin activity. The objective of this study was to measure the POMC prohormone and its processed peptide, β-endorphin (β-EP), in cerebrospinal fluid (CSF) and AgRP in CSF and plasma after calorie restriction to validate their utility as biomarkers of brain melanocortin activity. CSF and plasma were obtained from 10 lean and obese subjects after fasting (40 h) and refeeding (24 h), and from 8 obese subjects before and after 6 wk of dieting (800 kcal/day) to assess changes in neuropeptide and hormone levels. After fasting, plasma leptin decreased to 35%, and AgRP increased to 153% of baseline. During refeeding, AgRP declined as leptin increased; CSF β-EP increased, but POMC did not change. Relative changes in plasma and CSF leptin were blunted in obese subjects. After dieting, plasma and CSF leptin decreased to 46% and 70% of baseline, CSF POMC and β-EP decreased, and plasma AgRP increased. At baseline, AgRP correlated negatively with insulin and homeostasis model assessment (HOMA-IR), and positively with the Matsuda index. Thus, following chronic calorie restriction, POMC and β-EP declined in CSF, whereas acutely, only β-EP changed. Plasma AgRP, however, increased after both acute and chronic calorie restriction. These results support the use of CSF POMC and plasma AgRP as biomarkers of hypothalamic melanocortin activity and provide evidence linking AgRP to insulin sensitivity.
Collapse
Affiliation(s)
- Gabrielle Page-Wilson
- Department of Medicine, Columbia University College of Physicians & Surgeons, New York, New York
| | - Kim T Nguyen
- Department of Medicine, Columbia University College of Physicians & Surgeons, New York, New York
| | - Deniz Atalayer
- Department of Medicine, Columbia University College of Physicians & Surgeons, New York, New York
| | - Kana Meece
- Department of Medicine, Columbia University College of Physicians & Surgeons, New York, New York
| | - Heather A Bainbridge
- Department of Medicine, Columbia University College of Physicians & Surgeons, New York, New York
| | - Judith Korner
- Department of Medicine, Columbia University College of Physicians & Surgeons, New York, New York
| | - Rebecca J Gordon
- Department of Pediatrics, Columbia University College of Physicians & Surgeons, New York, New York
| | - Sunil K Panigrahi
- Department of Medicine, Columbia University College of Physicians & Surgeons, New York, New York
| | - Anne White
- Faculties of Life Sciences and Medical and Human Sciences, University of Manchester, Manchester, United Kingdom; and
| | - Richard Smiley
- Department of Anesthesiology, Columbia University College of Physicians & Surgeons, New York, New York
| | - Sharon L Wardlaw
- Department of Medicine, Columbia University College of Physicians & Surgeons, New York, New York;
| |
Collapse
|
65
|
Nillni EA. The metabolic sensor Sirt1 and the hypothalamus: Interplay between peptide hormones and pro-hormone convertases. Mol Cell Endocrinol 2016; 438:77-88. [PMID: 27614022 DOI: 10.1016/j.mce.2016.09.002] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/31/2016] [Revised: 09/05/2016] [Accepted: 09/05/2016] [Indexed: 01/11/2023]
Abstract
The last decade had witnessed a tremendous progress in our understanding of the causes of metabolic diseases including obesity. Among the contributing factors regulating energy balance are nutrient sensors such as sirtuins. Sirtuin1 (Sirt1), a NAD + - dependent deacetylase is affected by diet, environmental stress, and also plays a critical role in metabolic health by deacetylating proteins in many tissues, including liver, muscle, adipose tissue, heart, endothelium, and in the complexity of the hypothalamus. Because of its dependence on NAD+, Sirt1 also functions as a nutrient/redox sensor, and new novel data show a function of this enzyme in the maturation of hypothalamic peptide hormones controlling energy balance either through regulation of specific nuclear transcription factors or by regulating specific pro-hormone convertases (PCs) involved in the post-translational processing of pro-hormones. The post-translational processing mechanism of pro-hormones is critical in the pathogenesis of obesity as recently shown that metabolic and physiological triggers affect the biosynthesis and processing of many peptides hormones. Specific regulation of pro-hormone processing is likely another key step where final amounts of bioactive peptides can be tightly regulated. Different factors stimulate or inhibit pro-hormones biosynthesis in concert with an increase in the PCs involved in the maturation of bioactive hormones. Adding more complexity to the system, the new studies describe here suggest that Sirt1 could also regulate the fate of peptide hormone biosynthesis. The present review summarizes the recent progress in hypothalamic SIRT1 research with a particular emphasis on the tissue-specific control of neuropeptide hormone maturation. The series of studies done in mouse and rat models strongly advocate for the first time that a deacetylating enzyme could be a regulator in the maturation of peptide hormones and their processing enzymes. These discoveries are the culmination of the first in-depth understanding of the metabolic role of Sirt1 in the brain. It suggests that Sirt1 behaves differently in the brain than in organs such as the liver and pancreas, where the enzyme has been more commonly studied.
Collapse
Affiliation(s)
- Eduardo A Nillni
- The Warren Alpert Medical School, Department of Molecular Biology, Cell Biology and Biochemistry, Brown University, Providence, RI 02912, USA.
| |
Collapse
|
66
|
Fleury N, Geldenhuys S, Gorman S. Sun Exposure and Its Effects on Human Health: Mechanisms through Which Sun Exposure Could Reduce the Risk of Developing Obesity and Cardiometabolic Dysfunction. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2016; 13:E999. [PMID: 27727191 PMCID: PMC5086738 DOI: 10.3390/ijerph13100999] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/31/2016] [Revised: 09/30/2016] [Accepted: 10/01/2016] [Indexed: 01/10/2023]
Abstract
Obesity is a significant burden on global healthcare due to its high prevalence and associations with chronic health conditions. In our animal studies, ongoing exposure to low dose ultraviolet radiation (UVR, found in sunlight) reduced weight gain and the development of signs of cardiometabolic dysfunction in mice fed a high fat diet. These observations suggest that regular exposure to safe levels of sunlight could be an effective means of reducing the burden of obesity. However, there is limited knowledge around the nature of associations between sun exposure and the development of obesity and cardiometabolic dysfunction, and we do not know if sun exposure (independent of outdoor activity) affects the metabolic processes that determine obesity in humans. In addition, excessive sun exposure has strong associations with a number of negative health consequences such as skin cancer. This means it is very important to "get the balance right" to ensure that we receive benefits without increasing harm. In this review, we detail the evidence around the cardiometabolic protective effects of UVR and suggest mechanistic pathways through which UVR could be beneficial.
Collapse
Affiliation(s)
- Naomi Fleury
- Telethon Kids Institute, University of Western Australia, P.O. Box 855, Perth 6872, Australia.
| | - Sian Geldenhuys
- Telethon Kids Institute, University of Western Australia, P.O. Box 855, Perth 6872, Australia.
| | - Shelley Gorman
- Telethon Kids Institute, University of Western Australia, P.O. Box 855, Perth 6872, Australia.
| |
Collapse
|
67
|
Shipp SL, Cline MA, Gilbert ER. Recent advances in the understanding of how neuropeptide Y and α-melanocyte stimulating hormone function in adipose physiology. Adipocyte 2016; 5:333-350. [PMID: 27994947 DOI: 10.1080/21623945.2016.1208867] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/01/2016] [Revised: 06/28/2016] [Accepted: 06/28/2016] [Indexed: 12/20/2022] Open
Abstract
Communication between the brain and the adipose tissue has been the focus of many studies in recent years, with the "brain-fat axis" identified as a system that orchestrates the assimilation and usage of energy to maintain body mass and adequate fat stores. It is now well-known that appetite-regulating peptides that were studied as neurotransmitters in the central nervous system can act both on the hypothalamus to regulate feeding behavior and also on the adipose tissue to modulate the storage of energy. Energy balance is thus partly controlled by factors that can alter both energy intake and storage/expenditure. Two such factors involved in these processes are neuropeptide Y (NPY) and α-melanocyte stimulating hormone (α-MSH). NPY, an orexigenic factor, is associated with promoting adipogenesis in both mammals and chickens, while α-MSH, an anorexigenic factor, stimulates lipolysis in rodents. There is also evidence of interaction between the 2 peptides. This review aims to summarize recent advances in the study of NPY and α-MSH regarding their role in adipose tissue physiology, with an emphasis on the cellular and molecular mechanisms. A greater understanding of the brain-fat axis and regulation of adiposity by bioactive peptides may provide insights on strategies to prevent or treat obesity and also enhance nutrient utilization efficiency in agriculturally-important species.
Collapse
|
68
|
Jeong JH, Woo YJ, Chua S, Jo YH. Single-Cell Gene Expression Analysis of Cholinergic Neurons in the Arcuate Nucleus of the Hypothalamus. PLoS One 2016; 11:e0162839. [PMID: 27611685 PMCID: PMC5017726 DOI: 10.1371/journal.pone.0162839] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2016] [Accepted: 08/29/2016] [Indexed: 01/18/2023] Open
Abstract
The cholinoceptive system in the hypothalamus, in particular in the arcuate nucleus (ARC), plays a role in regulating food intake. Neurons in the ARC contain multiple neuropeptides, amines, and neurotransmitters. To study molecular and neurochemical heterogeneity of ARC neurons, we combine single-cell qRT-PCR and single-cell whole transcriptome amplification methods to analyze expression patterns of our hand-picked 60 genes in individual neurons in the ARC. Immunohistochemical and single-cell qRT-PCR analyses show choline acetyltransferase (ChAT)-expressing neurons in the ARC. Gene expression patterns are remarkably distinct in each individual cholinergic neuron. Two-thirds of cholinergic neurons express tyrosine hydroxylase (Th) mRNA. A large subset of these Th-positive cholinergic neurons is GABAergic as they express the GABA synthesizing enzyme glutamate decarboxylase and vesicular GABA transporter transcripts. Some cholinergic neurons also express the vesicular glutamate transporter transcript gene. POMC and POMC-processing enzyme transcripts are found in a subpopulation of cholinergic neurons. Despite this heterogeneity, gene expression patterns in individual cholinergic cells appear to be highly regulated in a cell-specific manner. In fact, membrane receptor transcripts are clustered with their respective intracellular signaling and downstream targets. This novel population of cholinergic neurons may be part of the neural circuitries that detect homeostatic need for food and control the drive to eat.
Collapse
Affiliation(s)
- Jae Hoon Jeong
- Division of Endocrinology, Department of Medicine, Albert Einstein College of Medicine, Bronx, United States of America
| | - Young Jae Woo
- Department of Genetics, Albert Einstein College of Medicine, Bronx, United States of America
| | - Streamson Chua
- Division of Endocrinology, Department of Medicine, Albert Einstein College of Medicine, Bronx, United States of America
| | - Young-Hwan Jo
- Division of Endocrinology, Department of Medicine, Albert Einstein College of Medicine, Bronx, United States of America
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, United States of America
- * E-mail:
| |
Collapse
|
69
|
Cawley NX, Li Z, Loh YP. 60 YEARS OF POMC: Biosynthesis, trafficking, and secretion of pro-opiomelanocortin-derived peptides. J Mol Endocrinol 2016; 56:T77-97. [PMID: 26880796 PMCID: PMC4899099 DOI: 10.1530/jme-15-0323] [Citation(s) in RCA: 115] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/08/2016] [Accepted: 02/15/2016] [Indexed: 12/15/2022]
Abstract
Pro-opiomelanocortin (POMC) is a prohormone that encodes multiple smaller peptide hormones within its structure. These peptide hormones can be generated by cleavage of POMC at basic residue cleavage sites by prohormone-converting enzymes in the regulated secretory pathway (RSP) of POMC-synthesizing endocrine cells and neurons. The peptides are stored inside the cells in dense-core secretory granules until released in a stimulus-dependent manner. The complexity of the regulation of the biosynthesis, trafficking, and secretion of POMC and its peptides reflects an impressive level of control over many factors involved in the ultimate role of POMC-expressing cells, that is, to produce a range of different biologically active peptide hormones ready for action when signaled by the body. From the discovery of POMC as the precursor to adrenocorticotropic hormone (ACTH) and β-lipotropin in the late 1970s to our current knowledge, the understanding of POMC physiology remains a monumental body of work that has provided insight into many aspects of molecular endocrinology. In this article, we describe the intracellular trafficking of POMC in endocrine cells, its sorting into dense-core secretory granules and transport of these granules to the RSP. Additionally, we review the enzymes involved in the maturation of POMC to its various peptides and the mechanisms involved in the differential processing of POMC in different cell types. Finally, we highlight studies pertaining to the regulation of ACTH secretion in the anterior and intermediate pituitary and POMC neurons of the hypothalamus.
Collapse
Affiliation(s)
- Niamh X Cawley
- Section on Cellular NeurobiologyEunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, Maryland, USA
| | - Zhaojin Li
- Section on Cellular NeurobiologyEunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, Maryland, USA
| | - Y Peng Loh
- Section on Cellular NeurobiologyEunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, Maryland, USA
| |
Collapse
|
70
|
α-Melanocyte-stimulating hormone ameliorates ocular surface dysfunctions and lesions in a scopolamine-induced dry eye model via PKA-CREB and MEK-Erk pathways. Sci Rep 2015; 5:18619. [PMID: 26685899 PMCID: PMC4685655 DOI: 10.1038/srep18619] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2015] [Accepted: 11/23/2015] [Indexed: 11/08/2022] Open
Abstract
Dry eye is a highly prevalent, chronic, and multifactorial disease that compromises quality of life and generates socioeconomic burdens. The pathogenic factors of dry eye disease (DED) include tear secretion abnormalities, tear film instability, and ocular surface inflammation. An effective intervention targeting the pathogenic factors is needed to control this disease. Here we applied α-Melanocyte-stimulating hormone (α-MSH) twice a day to the ocular surface of a scopolamine-induced dry eye rat model. The results showed that α-MSH at different doses ameliorated tear secretion, tear film stability, and corneal integrity, and corrected overexpression of proinflammatory factors, TNF-α, IL-1β, and IFN-γ, in ocular surface of the dry eye rats. Moreover, α-MSH, at 10(-4) μg/μl, maintained corneal morphology, inhibited apoptosis, and restored the number and size of conjunctival goblet cells in the dry eye rats. Mechanistically, α-MSH activated both PKA-CREB and MEK-Erk pathways in the dry eye corneas and conjunctivas; pharmacological blockade of either pathway abolished α-MSH's protective effects, suggesting that both pathways are necessary for α-MSH's protection under dry eye condition. The peliotropic protective functions and explicit signaling mechanism of α-MSH warrant translation of the α-MSH-containing eye drop into a novel and effective intervention to DED.
Collapse
|
71
|
α-Melanocyte-stimulating hormone prevents glutamate excitotoxicity in developing chicken retina via MC4R-mediated down-regulation of microRNA-194. Sci Rep 2015; 5:15812. [PMID: 26507936 PMCID: PMC4623527 DOI: 10.1038/srep15812] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2015] [Accepted: 09/30/2015] [Indexed: 11/29/2022] Open
Abstract
Glutamate excitotoxicity is a common pathology to blinding ischemic retinopathies, such as diabetic retinopathy, glaucoma, and central retinal vein or artery occlusion. The development of an effective interventional modality to glutamate excitotoxicity is hence important to preventing blindness. Herein we showed that α-melanocyte-stimulating hormone (α-MSH) time-dependently protected against glutamate-induced cell death and tissue damage in an improved embryonic chicken retinal explant culture system. α-MSH down-regulated microRNA-194 (miR-194) expression during the glutamate excitotoxicity in the retinal explants. Furthermore, pharmacological antagonists to melanocortin 4 receptor (MC4R) and lentivirus-mediated overexpression of pre-miR-194 abrogated the suppressing effects of α-MSH on glutamate-induced activities of caspase 3 or 7, the ultimate enzymes for glutamate-induced cell death. These results suggest that the protective effects of α-MSH may be due to the MC4R mediated-down-regulation of miR-194 during the glutamate-induced excitotoxicity. Finally, α-MSH attenuated cell death and recovered visual functions in glutamate-stimulated post-hatch chick retinas. These results demonstrate the previously undescribed protective effects of α-MSH against glutamate-induced excitotoxic cell death in the cone-dominated retina both in vitro and in vivo, and indicate a novel molecular mechanism linking MC4R-mediated signaling to miR-194.
Collapse
|
72
|
Page-Wilson G, Meece K, White A, Rosenbaum M, Leibel RL, Smiley R, Wardlaw SL. Proopiomelanocortin, agouti-related protein, and leptin in human cerebrospinal fluid: correlations with body weight and adiposity. Am J Physiol Endocrinol Metab 2015; 309:E458-65. [PMID: 26152765 PMCID: PMC4556883 DOI: 10.1152/ajpendo.00206.2015] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/28/2015] [Accepted: 07/01/2015] [Indexed: 11/22/2022]
Abstract
Leptin and its neuronal targets, which produce proopiomelanocortin (POMC) and agouti-related protein (AgRP), regulate energy balance. This study characterized leptin, POMC, and AgRP in the cerebrospinal fluid (CSF) of 47 healthy human subjects, 23 lean and 24 overweight/obese (OW/OB), as related to BMI, adiposity, plasma leptin, soluble leptin receptor (s-OB-R), and insulin. POMC was measured since the POMC prohormone is the predominant POMC peptide in CSF and correlates with hypothalamic POMC in rodents. Plasma AgRP was similarly characterized. CSF leptin was 83-fold lower than in plasma and correlated strongly with BMI, body fat, and insulin. The relative amount of leptin transported into CSF declined with increasing BMI, ranging from 4.5 to 0.52%, consistent with a saturable transport mechanism. CSF sOB-R was 78-fold lower than in plasma and correlated negatively with plasma and CSF leptin. CSF POMC was higher in lean vs. OW/OB subjects (P < 0.001) and correlated negatively with CSF leptin (r = -0.60, P < 0.001) and with plasma leptin, insulin, BMI, and adiposity. CSF AgRP was not different in lean vs. OW/OB; however, plasma AgRP was higher in lean subjects (P = 0.001) and correlated negatively with BMI, adiposity, leptin, insulin, and HOMA (P < 0.005). Thus, CSF measurements may provide useful biomarkers for brain leptin and POMC activity. The striking negative correlation between CSF leptin and POMC could be secondary to leptin resistance and/or neuronal changes associated with obesity but may also indicate that POMC plays a primary role in regulating body weight and adiposity. The role of plasma AgRP as a neuroendocrine biomarker deserves further study.
Collapse
Affiliation(s)
- Gabrielle Page-Wilson
- Department of Medicine, Columbia University College of Physicians and Surgeons, New York, New York
| | - Kana Meece
- Department of Medicine, Columbia University College of Physicians and Surgeons, New York, New York
| | - Anne White
- Faculties of Life Sciences and Medical and Human Sciences, University of Manchester, Manchester, United Kingdom
| | - Michael Rosenbaum
- Department of Pediatrics, Columbia University College of Physicians and Surgeons, New York, New York; and
| | - Rudolph L Leibel
- Department of Pediatrics, Columbia University College of Physicians and Surgeons, New York, New York; and
| | - Richard Smiley
- Department of Anesthesiology, Columbia University College of Physicians and Surgeons, New York, New York
| | - Sharon L Wardlaw
- Department of Medicine, Columbia University College of Physicians and Surgeons, New York, New York;
| |
Collapse
|
73
|
Alsters SIM, Goldstone AP, Buxton JL, Zekavati A, Sosinsky A, Yiorkas AM, Holder S, Klaber RE, Bridges N, van Haelst MM, le Roux CW, Walley AJ, Walters RG, Mueller M, Blakemore AIF. Truncating Homozygous Mutation of Carboxypeptidase E (CPE) in a Morbidly Obese Female with Type 2 Diabetes Mellitus, Intellectual Disability and Hypogonadotrophic Hypogonadism. PLoS One 2015; 10:e0131417. [PMID: 26120850 PMCID: PMC4485893 DOI: 10.1371/journal.pone.0131417] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2015] [Accepted: 06/02/2015] [Indexed: 01/25/2023] Open
Abstract
Carboxypeptidase E is a peptide processing enzyme, involved in cleaving numerous peptide precursors, including neuropeptides and hormones involved in appetite control and glucose metabolism. Exome sequencing of a morbidly obese female from a consanguineous family revealed homozygosity for a truncating mutation of the CPE gene (c.76_98del; p.E26RfsX68). Analysis detected no CPE expression in whole blood-derived RNA from the proband, consistent with nonsense-mediated decay. The morbid obesity, intellectual disability, abnormal glucose homeostasis and hypogonadotrophic hypogonadism seen in this individual recapitulates phenotypes in the previously described fat/fat and Cpe knockout mouse models, evidencing the importance of this peptide/hormone-processing enzyme in regulating body weight, metabolism, and brain and reproductive function in humans.
Collapse
Affiliation(s)
- Suzanne I. M. Alsters
- Section of Investigative Medicine, Division of Diabetes, Endocrinology, and Metabolism, Department of Medicine, Imperial College London, London, United Kingdom
| | - Anthony P. Goldstone
- Imperial Centre for Endocrinology, Imperial College Healthcare NHS Trust, Hammersmith Hospital, London, United Kingdom
- Centre for Neuropsychopharmacology and Computational, Cognitive and Clinical Neuroimaging Laboratory, Division of Brain Sciences, Department of Medicine, Imperial College London, Hammersmith Hospital, London, United Kingdom
- Metabolic and Molecular Imaging Group, MRC Clinical Sciences Centre, Imperial College London, Hammersmith Hospital, London, United Kingdom
- * E-mail: (AG); (AB)
| | - Jessica L. Buxton
- Section of Investigative Medicine, Division of Diabetes, Endocrinology, and Metabolism, Department of Medicine, Imperial College London, London, United Kingdom
- Centre for Cardiovascular Genetics, UCL Institute of Cardiovascular Science, London, United Kingdom
| | - Anna Zekavati
- NIHR Imperial BRC Genomics Facility, Faculty of Medicine, Imperial College London, London, United Kingdom
| | - Alona Sosinsky
- NIHR Imperial BRC Genomics Facility, Faculty of Medicine, Imperial College London, London, United Kingdom
| | - Andrianos M. Yiorkas
- Section of Investigative Medicine, Division of Diabetes, Endocrinology, and Metabolism, Department of Medicine, Imperial College London, London, United Kingdom
| | - Susan Holder
- NW Thames Regional Genetics Service, Kennedy Galton Centre, North West London Hospitals NHS Trust, Northwick Park Hospital, Harrow, United Kingdom
| | - Robert E. Klaber
- Department of Paediatrics, Imperial College Healthcare NHS Trust, St Mary's Hospital, London, United Kingdom
| | - Nicola Bridges
- Department of Paediatric Endocrinology, Chelsea and Westminster Hospital, London, United Kingdom
| | - Mieke M. van Haelst
- Department of Medical Genetics, Wilhelmina Children’s Hospital, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Carel W. le Roux
- Section of Investigative Medicine, Division of Diabetes, Endocrinology, and Metabolism, Department of Medicine, Imperial College London, London, United Kingdom
- Diabetes Complications Research Centre, Conway Institute, School of Medicine and Medical Science, University College Dublin, Dublin, Ireland
| | - Andrew J. Walley
- Department of Epidemiology and Biostatistics, School of Public Health, Imperial College London, London, United Kingdom
| | - Robin G. Walters
- Clinical Trial Service Unit and Epidemiological Studies Unit (CTSU), Nuffield Department of Population Health, University of Oxford, Oxford, United Kingdom
| | - Michael Mueller
- NIHR Imperial BRC Genomics Facility, Faculty of Medicine, Imperial College London, London, United Kingdom
| | - Alexandra I. F. Blakemore
- Section of Investigative Medicine, Division of Diabetes, Endocrinology, and Metabolism, Department of Medicine, Imperial College London, London, United Kingdom
- * E-mail: (AG); (AB)
| |
Collapse
|
74
|
Ramírez S, Claret M. Hypothalamic ER stress: A bridge between leptin resistance and obesity. FEBS Lett 2015; 589:1678-87. [PMID: 25913783 DOI: 10.1016/j.febslet.2015.04.025] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2015] [Revised: 04/14/2015] [Accepted: 04/15/2015] [Indexed: 12/21/2022]
Abstract
The prevalence of obesity has increased worldwide at an alarming rate. However, non-invasive pharmacological treatments remain elusive. Leptin resistance is a general feature of obesity, thus strategies aimed at enhancing the sensitivity to this hormone may constitute an excellent therapeutical approach to counteract current obesity epidemics. Nevertheless, the etiology and neuronal basis of leptin resistance remains an enigma. A recent hypothesis gaining substantial experimental support is that hypothalamic endoplasmic reticulum (ER) stress plays a causal role in the development of leptin resistance and obesity. The objective of this review article is to provide an updated view on current evidence connecting hypothalamic ER stress with leptin resistance. We discuss the experimental findings supporting this hypothesis, as well as the potential causes and underlying mechanisms leading to this metabolic disorder. Understanding these mechanisms may provide key insights into the development of novel intervention approaches.
Collapse
Affiliation(s)
- Sara Ramírez
- Diabetes and Obesity Research Laboratory, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036 Barcelona, Spain
| | - Marc Claret
- Diabetes and Obesity Research Laboratory, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036 Barcelona, Spain; CIBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), 08036 Barcelona, Spain
| |
Collapse
|
75
|
Faulkner LD, Dowling AR, Stuart RC, Nillni EA, Hill JW. Reduced melanocortin production causes sexual dysfunction in male mice with POMC neuronal insulin and leptin insensitivity. Endocrinology 2015; 156:1372-85. [PMID: 25590244 PMCID: PMC4399313 DOI: 10.1210/en.2014-1788] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Proopiomelanocortin (POMC)-derived peptides like α-melanocyte-stimulating hormone (MSH) substantially improve hepatic insulin sensitivity and regulate energy expenditure. Melanocortinergic agents are also powerful inducers of sexual arousal that are being investigated for a possible therapeutic role in erectile dysfunction. It is currently unclear whether reduced melanocortin (MC) activity may contribute to the sexual dysfunction accompanying obesity and type 2 diabetes. Male rodents with leptin and insulin resistance targeted to POMC neurons (leptin receptor [LepR]/insulin receptor [IR]POMC mice) exhibit obesity, hyperinsulinemia, hyperglycemia, and systemic insulin resistance. In this study, we demonstrate that LepR/IRPOMC males are also subfertile due to dramatic alterations in sexual behavior. Remarkably, these reproductive changes are accompanied by decreased α-MSH production not present when a single receptor type is deleted. Unexpectedly, behavioral sensitivity to α-MSH and MC receptor expression are also reduced in LepR/IRPOMC males, a potential adaptation of the MC system to altered α-MSH production. Together, these results suggest that concurrent insulin and leptin resistance in POMC neurons in individuals with obesity or type 2 diabetes can reduce endogenous α-MSH levels and impair sexual function.
Collapse
Affiliation(s)
- Latrice D Faulkner
- Department of Physiology and Pharmacology (L.D.F., A.R.D., J.W.H.), College of Medicine, The University of Toledo, Toledo, Ohio 43614-2598; and Division of Endocrinology (R.C.S., E.A.N.), Department of Medicine, The Warren Alpert Medical School of Brown University, Providence, Rhode Island 02912-9107
| | | | | | | | | |
Collapse
|
76
|
Cyr NE, Steger JS, Toorie AM, Yang JZ, Stuart R, Nillni EA. Central Sirt1 regulates body weight and energy expenditure along with the POMC-derived peptide α-MSH and the processing enzyme CPE production in diet-induced obese male rats. Endocrinology 2015; 156:961-74. [PMID: 25549049 PMCID: PMC4330311 DOI: 10.1210/en.2014-1970] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
In the periphery, the nutrient-sensing enzyme Sirtuin 1 (silent mating type information regulation 2 homolog 1 [Sirt1]) reduces body weight in diet-induced obese (DIO) rodents. However, the role of hypothalamic Sirt1 in body weight and energy balance regulation is debated. The first studies to reveal that central Sirt1 regulates body weight came from experiments in our laboratory using Sprague-Dawley rats. Central inhibition of Sirt1 decreased body weight and food intake as a result of a forkhead box protein O1 (FoxO1)-mediated increase in the anorexigenic proopiomelanocortin (POMC) and decrease in the orexigenic Agouti-related peptide in the hypothalamic arcuate nucleus. Here, we demonstrate that central inhibition of Sirt1 in DIO decreased body weight and increased energy expenditure at higher levels as compared with the lean counterpart. Brain Sirt1 inhibition in DIO increased acetylated FoxO1, which in turn increased phosphorylated FoxO1 via improved insulin/phosphorylated AKT signaling. Elevated acetylated FoxO1 and phosphorylated FoxO1 increased POMC along with the α-melanocyte-stimulating hormone (α-MSH) maturation enzyme carboxypeptidase E, which resulted in more of the bioactive POMC product α-MSH released into the paraventricular nucleus. Increased in α-MSH led to augmented TRH levels and circulating T3 levels (triiodothyronine, thyroid hormone). These results indicate that inhibiting hypothalamic Sirt1 in DIO enhances the activity of the hypothalamic-pituitary-thyroid axis, which stimulates energy expenditure. Because we show that blocking central Sirt1 causes physiological changes that promote a negative energy balance in an obese individual, our results support brain Sirt1 as a significant target for weight loss therapeutics.
Collapse
Affiliation(s)
- Nicole E Cyr
- Division of Endocrinology (N.E.C., J.S.S., A.M.T., J.Z.Y., R.S., E.A.N.), Department of Medicine, The Warren Alpert Medical School of Brown University/Rhode Island Hospital, Providence, Rhode Island 02903; and Department of Molecular Biology, Cell Biology, and Biochemistry (E.A.N.), Brown University, Providence, Rhode Island 02912
| | | | | | | | | | | |
Collapse
|
77
|
Wang L, Meece K, Williams DJ, Lo KA, Zimmer M, Heinrich G, Martin Carli J, Leduc CA, Sun L, Zeltser LM, Freeby M, Goland R, Tsang SH, Wardlaw SL, Egli D, Leibel RL. Differentiation of hypothalamic-like neurons from human pluripotent stem cells. J Clin Invest 2015; 125:796-808. [PMID: 25555215 DOI: 10.1172/jci79220] [Citation(s) in RCA: 83] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2014] [Accepted: 11/20/2014] [Indexed: 12/29/2022] Open
Abstract
The hypothalamus is the central regulator of systemic energy homeostasis, and its dysfunction can result in extreme body weight alterations. Insights into the complex cellular physiology of this region are critical to the understanding of obesity pathogenesis; however, human hypothalamic cells are largely inaccessible for direct study. Here, we developed a protocol for efficient generation of hypothalamic neurons from human embryonic stem cells (ESCs) and induced pluripotent stem cells (iPSCs) obtained from patients with monogenetic forms of obesity. Combined early activation of sonic hedgehog signaling followed by timed NOTCH inhibition in human ESCs/iPSCs resulted in efficient conversion into hypothalamic NKX2.1+ precursors. Application of a NOTCH inhibitor and brain-derived neurotrophic factor (BDNF) further directed the cells into arcuate nucleus hypothalamic-like neurons that express hypothalamic neuron markers proopiomelanocortin (POMC), neuropeptide Y (NPY), agouti-related peptide (AGRP), somatostatin, and dopamine. These hypothalamic-like neurons accounted for over 90% of differentiated cells and exhibited transcriptional profiles defined by a hypothalamic-specific gene expression signature that lacked pituitary markers. Importantly, these cells displayed hypothalamic neuron characteristics, including production and secretion of neuropeptides and increased p-AKT and p-STAT3 in response to insulin and leptin. Our results suggest that these hypothalamic-like neurons have potential for further investigation of the neurophysiology of body weight regulation and evaluation of therapeutic targets for obesity.
Collapse
|
78
|
Kang DY, Kim HC. Functional relevance of three proopiomelanocortin (POMC) genes in darkening camouflage, blind-side hypermelanosis, and appetite of Paralichthys olivaceus. Comp Biochem Physiol B Biochem Mol Biol 2015; 179:44-56. [DOI: 10.1016/j.cbpb.2014.09.002] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2014] [Revised: 08/21/2014] [Accepted: 09/10/2014] [Indexed: 12/12/2022]
|
79
|
Zhang C, Murugan S, Boyadjieva N, Jabbar S, Shrivastava P, Sarkar DK. Beta-endorphin cell therapy for cancer prevention. Cancer Prev Res (Phila) 2014; 8:56-67. [PMID: 25403848 DOI: 10.1158/1940-6207.capr-14-0254] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
β-Endorphin (BEP)-producing neuron in the hypothalamus plays a key role in bringing the stress axis to a state of homeostasis and maintaining body immune defense system. Long-term delivery of BEP to obtain beneficial effect on chemoprevention is challenging, as the peptides rapidly develop tolerance. Using rats as animal models, we show here that transplantation of BEP neurons into the hypothalamus suppressed carcinogens- and hormone-induced cancers in various tissues and prevented growth and metastasis of established tumors via activation of innate immune functions. In addition, we show that intracerebroventricular administration of nanosphere-attached dibutyryl cyclic adenosine monophosphate (dbcAMP) increased the number of BEP neurons in the hypothalamus, reduced the stress response, enhanced the innate immune function, and prevented tumor cell growth, progression, and metastasis. BEP neuronal supplementation did not produce any deleterious effects on general health but was beneficial in suppressing age-induced alterations in physical activity, metabolic, and immune functions. We conclude that the neuroimmune system has significant control over cancer growth and progression, and that activation of the neuroimmune system via BEP neuronal supplementation/induction may have therapeutic value for cancer prevention and improvement of general health.
Collapse
Affiliation(s)
- Changqing Zhang
- Rutgers Endocrine Program, The State University of New Jersey, New Brunswick, New Jersey. Endocrinology and Animal Biosciences Graduate Program, Rutgers, The State University of New Jersey, New Brunswick, New Jersey
| | - Sengottuvelan Murugan
- Rutgers Endocrine Program, The State University of New Jersey, New Brunswick, New Jersey. Department of Animal Sciences, The State University of New Jersey, New Brunswick, New Jersey
| | - Nadka Boyadjieva
- Rutgers Endocrine Program, The State University of New Jersey, New Brunswick, New Jersey. Department of Animal Sciences, The State University of New Jersey, New Brunswick, New Jersey
| | - Shaima Jabbar
- Rutgers Endocrine Program, The State University of New Jersey, New Brunswick, New Jersey. Endocrinology and Animal Biosciences Graduate Program, Rutgers, The State University of New Jersey, New Brunswick, New Jersey
| | - Pallavi Shrivastava
- Rutgers Endocrine Program, The State University of New Jersey, New Brunswick, New Jersey. Department of Animal Sciences, The State University of New Jersey, New Brunswick, New Jersey
| | - Dipak K Sarkar
- Rutgers Endocrine Program, The State University of New Jersey, New Brunswick, New Jersey. Endocrinology and Animal Biosciences Graduate Program, Rutgers, The State University of New Jersey, New Brunswick, New Jersey. Department of Animal Sciences, The State University of New Jersey, New Brunswick, New Jersey.
| |
Collapse
|
80
|
Jeong JK, Kim JG, Lee BJ. Participation of the central melanocortin system in metabolic regulation and energy homeostasis. Cell Mol Life Sci 2014; 71:3799-809. [PMID: 24894870 PMCID: PMC11113577 DOI: 10.1007/s00018-014-1650-z] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2013] [Revised: 04/23/2014] [Accepted: 05/12/2014] [Indexed: 10/25/2022]
Abstract
Obesity and metabolic disorders, such as type 2 diabetes and hypertension, have attracted considerable attention as life-threatening diseases not only in developed countries but also worldwide. Additionally, the rate of obesity in young people all over the world is rapidly increasing. Accumulated evidence suggests that the central nervous system may participate in the development of and/or protection from obesity. For example, in the brain, the hypothalamic melanocortin system senses and integrates central and peripheral metabolic signals and controls the degree of energy expenditure and feeding behavior, in concert with metabolic status, to regulate whole-body energy homeostasis. Currently, researchers are studying the mechanisms by which peripheral metabolic molecules control feeding behavior and energy balance through the central melanocortin system. Accordingly, recent studies have revealed that some inflammatory molecules and transcription factors participate in feeding behavior and energy balance by controlling the central melanocortin pathway, and have thus become new candidates as therapeutic targets to fight metabolic diseases such as obesity and diabetes.
Collapse
Affiliation(s)
- Jin Kwon Jeong
- Department of Biology and Biochemistry, Center for Nuclear Receptors and Cell Signaling, University of Houston, Houston, TX 77004 USA
| | - Jae Geun Kim
- Program in Integrative Cell Signaling and Neurobiology of Metabolism, Section of Comparative Medicine, Yale University School of Medicine, New Haven, CT 06520 USA
| | - Byung Ju Lee
- Department of Biological Sciences, University of Ulsan, Ulsan, 680-749 South Korea
| |
Collapse
|
81
|
Bekdash R, Zhang C, Sarkar D. Fetal alcohol programming of hypothalamic proopiomelanocortin system by epigenetic mechanisms and later life vulnerability to stress. Alcohol Clin Exp Res 2014; 38:2323-30. [PMID: 25069392 DOI: 10.1111/acer.12497] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2014] [Accepted: 05/24/2014] [Indexed: 01/01/2023]
Abstract
Hypothalamic proopiomelanocortin (POMC) neurons, one of the major regulators of the hypothalamic-pituitary-adrenal (HPA) axis, immune functions, and energy homeostasis, are vulnerable to the adverse effects of fetal alcohol exposure (FAE). These effects are manifested in POMC neurons by a decrease in Pomc gene expression, a decrement in the levels of its derived peptide β-endorphin and a dysregulation of the stress response in the adult offspring. The HPA axis is a major neuroendocrine system with pivotal physiological functions and mode of regulation. This system has been shown to be perturbed by prenatal alcohol exposure. It has been demonstrated that the perturbation of the HPA axis by FAE is long-lasting and is linked to molecular, neurophysiological, and behavioral changes in exposed individuals. Recently, we showed that the dysregulation of the POMC system function by FAE is induced by epigenetic mechanisms such as hypermethylation of Pomc gene promoter and an alteration in histone marks in POMC neurons. This developmental programming of the POMC system by FAE altered the transcriptome in POMC neurons and induced a hyperresponse to stress in adulthood. These long-lasting epigenetic changes influenced subsequent generations via the male germline. We also demonstrated that the epigenetic programming of the POMC system by FAE was reversed in adulthood with the application of the inhibitors of DNA methylation or histone modifications. Thus, prenatal environmental influences, such as alcohol exposure, could epigenetically modulate POMC neuronal circuits and function to shape adult behavioral patterns. Identifying specific epigenetic factors in hypothalamic POMC neurons that are modulated by fetal alcohol and target Pomc gene could be potentially useful for the development of new therapeutic approaches to treat stress-related diseases in patients with fetal alcohol spectrum disorders.
Collapse
Affiliation(s)
- Rola Bekdash
- Endocrinology Program, Department of Animal Sciences, Rutgers, The State University of New Jersey, New Brunswick, New Jersey; Neuroscience Graduate Program, The State University of New Jersey, New Brunswick, New Jersey
| | | | | |
Collapse
|
82
|
Cyr NE, Steger JS, Toorie AM, Yang JZ, Stuart R, Nillni EA. Central Sirt1 regulates body weight and energy expenditure along with the POMC-derived peptide α-MSH and the processing enzyme CPE production in diet-induced obese male rats. Endocrinology 2014; 155:2423-35. [PMID: 24773342 PMCID: PMC4060185 DOI: 10.1210/en.2013-1998] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
In the periphery, the nutrient-sensing enzyme Sirtuin 1 (silent mating type information regulation 2 homolog 1 [Sirt1]) reduces body weight in diet-induced obese (DIO) rodents. However, the role of Sirt1 in the brain, particularly the hypothalamus, in body weight and energy balance regulation is debated. Among the first studies to reveal that central Sirt1 regulates body weight came from experiments in our laboratory using Sprague Dawley rats. In that study, central inhibition of Sirt1 decreased body weight and food intake as a result of a Forkhead box protein O1 (FoxO1)-mediated increase in the anorexigenic proopiomelanocortin (POMC) and decrease in the orexigenic Agouti-related peptide in the hypothalamic arcuate nucleus. Here, we demonstrate that central inhibition of Sirt1 in DIO decreased body weight and increased energy expenditure at higher levels as compared with the lean counterpart. Brain Sirt1 inhibition in DIO increased acetylated FoxO1, which, in turn, increased phosphorylated FoxO1 via improved insulin/pAKT signaling. Elevated acetylated FoxO1 and phosphorylated FoxO1 increased POMC along with the α-MSH maturation enzyme carboxypeptidase E, which resulted in more of the bioactive POMC product α-MSH released into the paraventricular nucleus. Increased in α-MSH led to augmented TRH levels and circulating T3 levels (thyroid hormone). These results indicate that inhibiting hypothalamic Sirt1 in DIO enhances the activity of the hypothalamic-pituitary-thyroid axis, which stimulates energy expenditure. Because we show that blocking central Sirt1 causes physiological changes that promote a negative energy balance in an obese individual, our results support brain Sirt1 as a significant target for weight loss therapeutics.
Collapse
Affiliation(s)
- Nicole E Cyr
- Division of Endocrinology (N.E.C, J.S.S., A.M.T., J.Z.Y, R.S., E.A.N.), Department of Medicine, The Warren Alpert Medical School of Brown University/Rhode Island Hospital, Providence, Rhode Island 02903; and Department of Molecular Biology, Cell Biology and Biochemistry (E.A.N.), Brown University, Providence, Rhode Island 02912
| | | | | | | | | | | |
Collapse
|
83
|
Wardlaw SL, Burant CF, Klein S, Meece K, White A, Kasten T, Lucey BP, Bateman RJ. Continuous 24-hour leptin, proopiomelanocortin, and amino acid measurements in human cerebrospinal fluid: correlations with plasma leptin, soluble leptin receptor, and amino acid levels. J Clin Endocrinol Metab 2014; 99:2540-8. [PMID: 24670082 PMCID: PMC4079306 DOI: 10.1210/jc.2013-4087] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
CONTEXT In order to characterize diurnal changes in central leptin and its target neuropeptide, proopiomelanocortin (POMC), we measured leptin and POMC in cerebrospinal fluid (CSF) as related to changes in plasma leptin and soluble leptin receptor (sOB-R) levels. CSF and plasma levels of 20 amino acids (AA) were also measured because AA can affect brain POMC. DESIGN AND PARTICIPANTS Stored CSF and plasma samples obtained from eight healthy subjects who served as controls for a previous study were evaluated. CSF was collected hourly over 33 h via indwelling subarachnoid catheter. Leptin, sOB-R, and POMC were measured by sensitive ELISA and AA by gas chromatography-mass spectrometry. RESULTS There was a diurnal rhythm for plasma leptin with a peak at 2200 h (144% of baseline) and there was a similar diurnal rhythm for CSF leptin with a peak (117%) 3-5 h after the plasma peak. Plasma sOB-R was lowest at 0300 h and correlated negatively with plasma and CSF leptin. A diurnal rhythm for POMC in CSF was also detected with a peak (125%) at 0100 h. A positive correlation existed between CSF POMC and leptin in individual subjects over time. CSF levels of many AA increased at night. There was a significant correlation between CSF POMC and 10 AA, including leucine, isoleucine, tryptophan, and tyrosine. CONCLUSIONS Diurnal changes occur in leptin and POMC in human CSF that likely reflect changes in central leptin and melanocortin activity. Our results suggest that nocturnal elevations in leptin, AA, and POMC may help to suppress appetite and feeding at night.
Collapse
Affiliation(s)
- Sharon L Wardlaw
- Department of Medicine (S.L.W., K.M.), Columbia University College of Physicians & Surgeons, New York, New York 10032; Department of Internal Medicine (C.F.B.), University of Michigan Medical School, Ann Arbor, Michigan 48019; Center for Human Nutrition and Atkins Center for Excellence in Obesity Medicine (S.K.), Washington University School of Medicine, St Louis, Missouri 63110; Faculties of Life Sciences and Medical and Human Sciences (A.W.), University of Manchester, Manchester M13 9PL, United Kingdom; and Department of Neurology (T.K., B.P.L., R.J.B), Washington University School of Medicine, St Louis, Missouri 63110
| | | | | | | | | | | | | | | |
Collapse
|
84
|
Sanathara NM, Moreas J, Mahavongtrakul M, Sinchak K. Estradiol upregulates progesterone receptor and orphanin FQ colocalization in arcuate nucleus neurons and opioid receptor-like receptor-1 expression in proopiomelanocortin neurons that project to the medial preoptic nucleus in the female rat. Neuroendocrinology 2014; 100:103-18. [PMID: 24821192 PMCID: PMC4225187 DOI: 10.1159/000363324] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/24/2013] [Accepted: 04/29/2014] [Indexed: 12/13/2022]
Abstract
BACKGROUND Ovarian steroids regulate sexual receptivity in the female rat by acting on neurons that converge on proopiomelanocortin (POMC) neurons in the arcuate nucleus of the hypothalamus (ARH) that project to the medial preoptic nucleus (MPN). Estradiol rapidly activates these neurons to release β-endorphin that activates MPN μ-opioid receptors (MOP) to inhibit lordosis. Lordosis is facilitated by the subsequent action of progesterone that deactivates the estradiol-induced MPN MOP activation. Orphanin FQ (OFQ/N; also known as nociceptin) infusions into the ARH, like progesterone, deactivate MPN MOP and facilitate lordosis in estradiol-primed rats. OFQ/N reduces the activity of ARH β-endorphin neurons through post- and presynaptic mechanisms via its cognate receptor, ORL-1. METHODS We tested the hypotheses that progesterone receptors (PR) are expressed in ARH OFQ/N neurons by immunohistochemistry and ORL-1 is expressed in POMC neurons that project to the MPN by combining Fluoro-Gold injection into the MPN and double-label fluorescent in situ hybridization (FISH). We also hypothesized that estradiol increases coexpression of PR-OFQ/N and ORL-1-POMC in ARH neurons of ovariectomized rats. RESULTS The number of PR- and OFQ/N-immunopositive ARH neurons was increased as was their colocalization by estradiol treatment. FISH for ORL-1 and POMC mRNA revealed a subpopulation of ARH neurons that was triple labeled, indicating these neurons project to the MPN and coexpress ORL-1 and POMC mRNA. Estradiol was shown to upregulate ORL-1 and POMC expression in MPN-projecting ARH neurons. CONCLUSION Estradiol upregulates the ARH OFQ/N-ORL-1 system projecting to the MPN that regulates lordosis.
Collapse
Affiliation(s)
- Nayna M Sanathara
- Department of Biological Sciences, California State University, Long Beach, Long Beach, Calif., USA
| | | | | | | |
Collapse
|
85
|
Mercer AJ, Stuart RC, Attard CA, Otero-Corchon V, Nillni EA, Low MJ. Temporal changes in nutritional state affect hypothalamic POMC peptide levels independently of leptin in adult male mice. Am J Physiol Endocrinol Metab 2014; 306:E904-15. [PMID: 24518677 PMCID: PMC3989737 DOI: 10.1152/ajpendo.00540.2013] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Hypothalamic proopiomelanocortin (POMC) neurons constitute a critical anorexigenic node in the central nervous system (CNS) for maintaining energy balance. These neurons directly affect energy expenditure and feeding behavior by releasing bioactive neuropeptides but are also subject to signals directly related to nutritional state such as the adipokine leptin. To further investigate the interaction of diet and leptin on hypothalamic POMC peptide levels, we exposed 8- to 10-wk-old male POMC-Discosoma red fluorescent protein (DsRed) transgenic reporter mice to either 24-48 h (acute) or 2 wk (chronic) food restriction, high-fat diet (HFD), or leptin treatment. Using semiquantitative immunofluorescence and radioimmunoassays, we discovered that acute fasting and chronic food restriction decreased the levels of adrenocorticotropic hormone (ACTH), α-melanocyte-stimulating hormone (α-MSH), and β-endorphin in the hypothalamus, together with decreased DsRed fluorescence, compared with control ad libitum-fed mice. Furthermore, acute but not chronic HFD or leptin administration selectively increased α-MSH levels in POMC fibers and increased DsRed fluorescence in POMC cell bodies. HFD and leptin treatments comparably increased circulating leptin levels at both time points, suggesting that transcription of Pomc and synthesis of POMC peptide products are not modified in direct relation to the concentration of plasma leptin. Our findings indicate that negative energy balance persistently downregulated POMC peptide levels, and this phenomenon may be partially explained by decreased leptin levels, since these changes were blocked in fasted mice treated with leptin. In contrast, sustained elevation of plasma leptin by HFD or hormone supplementation did not significantly alter POMC peptide levels, indicating that enhanced leptin signaling does not chronically increase Pomc transcription and peptide synthesis.
Collapse
Affiliation(s)
- Aaron J Mercer
- Department of Molecular & Integrative Physiology, University of Michigan, Ann Arbor, Michigan
| | | | | | | | | | | |
Collapse
|
86
|
Zhang L, Dong L, Liu X, Jiang Y, Zhang L, Zhang X, Li X, Zhang Y. α-Melanocyte-stimulating hormone protects retinal vascular endothelial cells from oxidative stress and apoptosis in a rat model of diabetes. PLoS One 2014; 9:e93433. [PMID: 24695675 PMCID: PMC3973693 DOI: 10.1371/journal.pone.0093433] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2013] [Accepted: 03/04/2014] [Indexed: 12/14/2022] Open
Abstract
AIMS Oxidative stress and apoptosis are among the earliest lesions of diabetic retinopathy. This study sought to examine the anti-oxidative and anti-apoptotic effects of α-melanocyte-stimulating hormone (α-MSH) in early diabetic retinas and to explore the underlying mechanisms in retinal vascular endothelial cells. METHODS Sprague-Dawley rats were injected intravenously with streptozocin to induce diabetes. The diabetic rats were injected intravitreally with α-MSH or saline. At week 5 after diabetes, the retinas were analyzed for reactive oxygen species (ROS) and gene expression. One week later, the retinas were processed for terminal deoxynucleotidyl transferase dUTP nick-end labeling staining and transmission electron microscopy. Retinal vascular endothelial cells were stimulated by high glucose (HG) with or without α-MSH. The expression of Forkhead box O genes (Foxos) was examined through real-time PCR. The Foxo4 gene was overexpressed in endothelial cells by transient transfection prior to α-MSH or HG treatment, and oxidative stress and apoptosis were analyzed through CM-H2DCFDA and annexin-V assays, respectively. RESULTS In diabetic retinas, the levels of H2O2 and ROS and the total anti-oxidant capacity were normalized, the apoptotic cell number was reduced, and the ultrastructural injuries were ameliorated by α-MSH. Treatment with α-MSH also corrected the aberrant changes in eNOS, iNOS, ICAM-1, and TNF-α expression levels in diabetic retinas. Furthermore, α-MSH inhibited Foxo4 up-regulation in diabetic retinas and in endothelial cells exposed to HG, whereas Foxo4 overexpression abrogated the anti-oxidative and anti-apoptotic effects of α-MSH in HG-stimulated retinal vascular endothelial cells. CONCLUSIONS α-MSH normalized oxidative stress, reduced apoptosis and ultrastructural injuries, and corrected gene expression levels in early diabetic retinas. The protective effects of α-MSH in retinal vascular endothelial cells may be mediated through the inhibition of Foxo4 up-regulation induced by HG. This study suggests an α-MSH-mediated potential intervention approach to early diabetic retinopathy and a novel regulatory mechanism involving Foxo4.
Collapse
Affiliation(s)
- Lijuan Zhang
- Tianjin Medical University Eye Hospital/Eye Institute, Tianjin, China
| | - Lijie Dong
- Tianjin Medical University Eye Hospital/Eye Institute, Tianjin, China
| | - Xun Liu
- Tianjin Medical University Eye Hospital/Eye Institute, Tianjin, China
| | - Yuanfeng Jiang
- Tianjin Medical University Eye Hospital/Eye Institute, Tianjin, China
| | - Lingjun Zhang
- Tianjin Medical University Eye Hospital/Eye Institute, Tianjin, China
| | - Xiaomin Zhang
- Tianjin Medical University Eye Hospital/Eye Institute, Tianjin, China
| | - Xiaorong Li
- Tianjin Medical University Eye Hospital/Eye Institute, Tianjin, China
| | - Yan Zhang
- Tianjin Medical University Eye Hospital/Eye Institute, Tianjin, China
| |
Collapse
|
87
|
Acs P, Bauer PO, Mayer B, Bera T, Macallister R, Mezey E, Pastan I. A novel form of ciliopathy underlies hyperphagia and obesity in Ankrd26 knockout mice. Brain Struct Funct 2014; 220:1511-28. [PMID: 24633808 DOI: 10.1007/s00429-014-0741-9] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2014] [Accepted: 02/19/2014] [Indexed: 02/06/2023]
Abstract
Human ciliopathies are genetic disorders caused by mutations in genes responsible for the formation and function of primary cilia. Some are associated with hyperphagia and obesity (e.g., Bardet-Biedl Syndrome, Alström Syndrome), but the mechanisms underlying these problems are not fully understood. The human gene ANKRD26 is located on 10p12, a locus that is associated with some forms of hereditary obesity. Previously, we reported that disruption of this gene causes hyperphagia, obesity and gigantism in mice. In the present study, we looked for the mechanisms that induce hyperphagia in the Ankrd26-/- mice and found defects in primary cilia in regions of the central nervous system that control appetite and energy homeostasis.
Collapse
Affiliation(s)
- Peter Acs
- Laboratory of Molecular Biology, National Cancer Institute, NIH, Bethesda, USA
| | | | | | | | | | | | | |
Collapse
|
88
|
Caruso V, Lagerström MC, Olszewski PK, Fredriksson R, Schiöth HB. Synaptic changes induced by melanocortin signalling. Nat Rev Neurosci 2014; 15:98-110. [PMID: 24588018 DOI: 10.1038/nrn3657] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
The melanocortin system has a well-established role in the regulation of energy homeostasis, but there is growing evidence of its involvement in memory, nociception, mood disorders and addiction. In this Review, we focus on the role of the melanocortin 4 receptor and provide an integrative view of the molecular mechanisms that lead to melanocortin-induced changes in synaptic plasticity within these diverse physiological systems. We also highlight the importance of melanocortin peptides and receptors in chronic pain syndromes, memory impairments, depression and drug abuse, and the possibility of targeting them for therapeutic purposes.
Collapse
|
89
|
Eerola K, Nordlund W, Virtanen S, Dickens AM, Mattila M, Ruohonen ST, Chua SC, Wardlaw SL, Savontaus M, Savontaus E. Lentivirus-mediated α-melanocyte-stimulating hormone overexpression in the hypothalamus decreases diet induced obesity in mice. J Neuroendocrinol 2013; 25:1298-1307. [PMID: 24118213 DOI: 10.1111/jne.12109] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/28/2013] [Revised: 09/20/2013] [Accepted: 09/21/2013] [Indexed: 11/29/2022]
Abstract
Melanocyte stimulating hormone (MSH) derived from the pro-hormone pro-opiomelanocortin (POMC) has potent effects on metabolism and feeding that lead to reduced body weight in the long-term. To determine the individual roles of POMC derived peptides and their sites of action, we created a method for the delivery of single MSH peptides using lentiviral vectors and studied the long-term anti-obesity effects of hypothalamic α-MSH overexpression in mice. An α-MSH lentivirus (LVi-α-MSH-EGFP) vector carrying the N'-terminal part of POMC and the α-MSH sequence was generated and shown to produce bioactive peptide in an in vitro melanin synthesis assay. Stereotaxis was used to deliver the LVi-α-MSH-EGFP or control LVi-EGFP vector to the arcuate nucleus (ARC) of the hypothalamus of male C57Bl/6N mice fed on a high-fat diet. The effects of 6-week-treatment on body weight, food intake, glucose tolerance and organ weights were determined. Additionally, a 14-day pairfeeding study was conducted to assess whether the weight decreasing effect of the LVi-α-MSH-EGFP treatment is dependent on decreased food intake. The 6-week LVi-α-MSH-EGFP treatment reduced weight gain (8.4 ± 0.4 g versus 12.3 ± 0.6 g; P < 0.05), which was statistically significant starting from 1 week after the injections. The weight of mesenteric fat was decreased and glucose tolerance was improved compared to LVi-EGFP treated mice. Food intake was decreased during the first week in the LVi-α-MSH-EGFP treated mice but subsequently increased to the level of LVi-EGFP treated mice. The LVi-EGFP injected control mice gained more weight even when pairfed to the level of food intake by LVi-α-MSH-EGFP treated mice. We demonstrate that gene transfer of α-MSH, a single peptide product of POMC, into the ARC of the hypothalamus, reduces obesity and improves glucose tolerance, and that factors other than decreased food intake also influence the weight decreasing effects of α-MSH overexpression in the ARC. Furthermore, viral MSH vectors delivered stereotaxically provide a novel tool for further exploration of chronic site-specific effects of POMC peptides.
Collapse
Affiliation(s)
- K Eerola
- Department of Pharmacology, Drug Development and Therapeutics, and Turku Center for Disease Modeling, University of Turku, Turku, Finland
- Turku Centre for Biotechnology, University of Turku, Turku, Finland
- FinPharma Doctoral Program, Drug Discovery Section, Turku, Finland
| | - W Nordlund
- Department of Pharmacology, Drug Development and Therapeutics, and Turku Center for Disease Modeling, University of Turku, Turku, Finland
| | - S Virtanen
- Department of Pharmacology, Drug Development and Therapeutics, and Turku Center for Disease Modeling, University of Turku, Turku, Finland
- Turku Centre for Biotechnology, University of Turku, Turku, Finland
| | - A M Dickens
- Department of Pharmacology, Drug Development and Therapeutics, and Turku Center for Disease Modeling, University of Turku, Turku, Finland
- Turku PET Centre, Medicity/PET Preclinical Imaging, University of Turku, Turku, Finland
| | - M Mattila
- Turku Centre for Biotechnology, University of Turku, Turku, Finland
- Medical Biochemistry and Genetics, University of Turku, Turku, Finland
| | - S T Ruohonen
- Department of Pharmacology, Drug Development and Therapeutics, and Turku Center for Disease Modeling, University of Turku, Turku, Finland
| | - S C Chua
- Albert Einstein College of Medicine, New York, NY, USA
| | - S L Wardlaw
- Department of Medicine, Columbia University College of Physicians & Surgeons, New York, NY, USA
| | - M Savontaus
- Turku Centre for Biotechnology, University of Turku, Turku, Finland
- Turku Heart Center, Turku University Hospital and University of Turku, Turku, Finland
| | - E Savontaus
- Department of Pharmacology, Drug Development and Therapeutics, and Turku Center for Disease Modeling, University of Turku, Turku, Finland
- Unit of Clinical Pharmacology, Turku University Hospital, Turku, Finland
| |
Collapse
|
90
|
Schneeberger M, Dietrich MO, Sebastián D, Imbernón M, Castaño C, Garcia A, Esteban Y, Gonzalez-Franquesa A, Rodríguez IC, Bortolozzi A, Garcia-Roves PM, Gomis R, Nogueiras R, Horvath TL, Zorzano A, Claret M. Mitofusin 2 in POMC neurons connects ER stress with leptin resistance and energy imbalance. Cell 2013; 155:172-87. [PMID: 24074867 DOI: 10.1016/j.cell.2013.09.003] [Citation(s) in RCA: 406] [Impact Index Per Article: 33.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2013] [Revised: 07/09/2013] [Accepted: 08/22/2013] [Indexed: 11/26/2022]
Abstract
Mitofusin 2 (MFN2) plays critical roles in both mitochondrial fusion and the establishment of mitochondria-endoplasmic reticulum (ER) interactions. Hypothalamic ER stress has emerged as a causative factor for the development of leptin resistance, but the underlying mechanisms are largely unknown. Here, we show that mitochondria-ER contacts in anorexigenic pro-opiomelanocortin (POMC) neurons in the hypothalamus are decreased in diet-induced obesity. POMC-specific ablation of Mfn2 resulted in loss of mitochondria-ER contacts, defective POMC processing, ER stress-induced leptin resistance, hyperphagia, reduced energy expenditure, and obesity. Pharmacological relieve of hypothalamic ER stress reversed these metabolic alterations. Our data establish MFN2 in POMC neurons as an essential regulator of systemic energy balance by fine-tuning the mitochondrial-ER axis homeostasis and function. This previously unrecognized role for MFN2 argues for a crucial involvement in mediating ER stress-induced leptin resistance.
Collapse
Affiliation(s)
- Marc Schneeberger
- Diabetes and Obesity Research Laboratory, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036 Barcelona, Spain; Department of Endocrinology and Nutrition, Hospital Clínic. School of Medicine, University of Barcelona, 08036 Barcelona, Spain; Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), 08036 Barcelona, Spain
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
91
|
Emaresi G, Ducrest AL, Bize P, Richter H, Simon C, Roulin A. Pleiotropy in the melanocortin system: expression levels of this system are associated with melanogenesis and pigmentation in the tawny owl (Strix aluco). Mol Ecol 2013; 22:4915-30. [PMID: 24033481 DOI: 10.1111/mec.12438] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2013] [Revised: 06/26/2013] [Accepted: 06/27/2013] [Indexed: 01/22/2023]
Abstract
The adaptive function of melanin-based coloration is a long-standing debate. A recent genetic model suggested that pleiotropy could account for covariations between pigmentation, behaviour, morphology, physiology and life history traits. We explored whether the expression levels of genes belonging to the melanocortin system (MC1R, POMC, PC1/3, PC2 and the antagonist ASIP), which have many pleiotropic effects, are associated with melanogenesis (through variation in the expression of the genes MITF, SLC7A11, TYR, TYRP1) and in turn melanin-based coloration. We considered the tawny owl (Strix aluco) because individuals vary continuously from light to dark reddish, and thus, colour variation is likely to stem from differences in the levels of gene expression. We measured gene expression in feather bases collected in nestlings at the time of melanin production. As expected, the melanocortin system was associated with the expression of melanogenic genes and pigmentation. Offspring of darker reddish fathers expressed PC1/3 to lower levels but tended to express PC2 to higher levels. The convertase enzyme PC1/3 cleaves the POMC prohormone to obtain ACTH, while the convertase enzyme PC2 cleaves ACTH to produce α-melanin-stimulating hormone (α-MSH). ACTH regulates glucocorticoids, hormones that modulate stress responses, while α-MSH induces eumelanogenesis. We therefore conclude that the melanocortin system, through the convertase enzymes PC1/3 and PC2, may account for part of the interindividual variation in melanin-based coloration in nestling tawny owls. Pleiotropy may thus account for the covariation between phenotypic traits involved in social interactions (here pigmentation) and life history, morphology, behaviour and physiology.
Collapse
Affiliation(s)
- Guillaume Emaresi
- Department of Ecology and Evolution, Biophore Building, University of Lausanne, 1015 Lausanne, Switzerland
| | | | | | | | | | | |
Collapse
|
92
|
Mul JD, Spruijt BM, Brakkee JH, Adan RAH. Melanocortin MC(4) receptor-mediated feeding and grooming in rodents. Eur J Pharmacol 2013; 719:192-201. [PMID: 23872405 DOI: 10.1016/j.ejphar.2013.04.060] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2013] [Revised: 03/27/2013] [Accepted: 04/03/2013] [Indexed: 02/04/2023]
Abstract
Decades ago it was recognized that the pharmacological profile of melanocortin ligands that stimulated grooming behavior in rats was strikingly similar to that of Xenopus laevis melanophore pigment dispersion. After cloning of the melanocortin MC1 receptor, expressed in melanocytes, and the melanocortin MC4 receptor, expressed mainly in brain, the pharmacological profiles of these receptors appeared to be very similar and it was demonstrated that these receptors mediate melanocortin-induced pigmentation and grooming respectively. Grooming is a low priority behavior that is concerned with care of body surface. Activation of central melanocortin MC4 receptors is also associated with meal termination, and continued postprandial stimulation of melanocortin MC4 receptors may stimulate natural postprandial grooming behavior as part of the behavioral satiety sequence. Indeed, melanocortins fail to suppress food intake or induce grooming behavior in melanocortin MC4 receptor-deficient rats. This review will focus on how melanocortins affect grooming behavior through the melanocortin MC4 receptor, and how melanocortin MC4 receptors mediate feeding behavior. This review also illustrates how melanocortins were the most likely candidates to mediate grooming and feeding based on the natural behaviors they induced.
Collapse
Affiliation(s)
- Joram D Mul
- Metabolic Diseases Institute, University of Cincinnati, 2170 East Galbraith Road, 45237 Cincinnati, Ohio, USA.
| | - Berry M Spruijt
- Department of Biology, Faculty of Beta Sciences, University of Utrecht, Padualaan 8, 3584 CH Utrecht, The Netherlands
| | - Jan H Brakkee
- Rudolf Magnus Institute of Neuroscience, Department of Neuroscience and Pharmacology, University Medical Center Utrecht, Universiteitsweg 100, 3584 CG Utrecht, The Netherlands
| | - Roger A H Adan
- Rudolf Magnus Institute of Neuroscience, Department of Neuroscience and Pharmacology, University Medical Center Utrecht, Universiteitsweg 100, 3584 CG Utrecht, The Netherlands
| |
Collapse
|
93
|
Younes-Rapozo V, Moura EG, Manhães AC, Pinheiro CR, Santos-Silva AP, de Oliveira E, Lisboa PC. Maternal nicotine exposure during lactation alters hypothalamic neuropeptides expression in the adult rat progeny. Food Chem Toxicol 2013; 58:158-68. [PMID: 23623838 DOI: 10.1016/j.fct.2013.04.036] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2013] [Revised: 04/08/2013] [Accepted: 04/16/2013] [Indexed: 11/28/2022]
Abstract
Maternal exposure to nicotine during lactation causes hyperleptinemia in the pups and, at adulthood, these animals are overweight and hyperleptinemic, while, in their hypothalamus, the leptin signaling pathway is reduced, evidencing a central leptin resistance. Then, we evaluated the expression of pro-opiomelanocortin (POMC), alpha-melanocyte stimulating hormone (α-MSH), cocaine and amphetamine-regulated transcript (CART), neuropeptide Y (NPY), agouti-related peptide (AgRP) and others in different hypothalamic nuclei in order to better understand the mechanisms underlying the obese phenotype observed in these animals at adulthood. On the 2nd postnatal day (P2), dams were subcutaneously implanted with osmotic minipumps releasing nicotine (NIC-6 mg/kg/day) or saline for 14 days. Offspring were killed in P180 and immunohistochemistry and Western blot analysis were carried out. Significance data had p<0.05. Adult NIC offspring showed more intense NPY staining in the paraventricular nucleus (PVN) (+21%) and increased number of POMC-positive cells in the: arcuate nucleus (+33%), as an increase in fiber density of α-MSH in PVN (+85%). However, the number of CART-positive cells was reduced in the PVN (-25%). CRH staining was more intense in NIC offspring (+136%). Orexins and AgRP were not altered. Thus, maternal nicotine exposure changes hypothalamic neuropeptides in the adult progeny that is partially compatible with leptin resistance.
Collapse
Affiliation(s)
- Viviane Younes-Rapozo
- Laboratório de Fisiologia Endócrina, Instituto de Biologia Roberto Alcantara Gomes, Universidade do Estado do Rio de Janeiro, Rio de Janeiro, Brazil
| | | | | | | | | | | | | |
Collapse
|
94
|
Suzuki H, Asakawa A, Amitani H, Nakamura N, Inui A. Ghrelin and cachexia in chronic kidney disease. Pediatr Nephrol 2013; 28:521-6. [PMID: 22760416 DOI: 10.1007/s00467-012-2241-6] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/03/2012] [Revised: 05/27/2012] [Accepted: 05/30/2012] [Indexed: 12/14/2022]
Abstract
Ghrelin is a growth hormone (GH) secretagogue and a potent orexigenic factor that stimulates feeding by interacting with hypothalamic feeding-regulatory nuclei. Its multifaceted effects are potentially beneficial as a treatment in human disease states. In both adult and pediatric chronic kidney disease (CKD) patients, decreased appetite plays a major role in wasting, which in turn is linked to morbidity and mortality; wasting has also been linked to high levels of leptin and proinflammatory cytokines. The beneficial effects of ghrelin treatment in CKD are potentially mediated by multiple concurrent actions, including the stimulation of appetite-regulating centers, anti-inflammatory effects, and direct kidney effects. Further evaluation of this appetite-regulating hormone in CKD is needed to confirm previous findings and to determine the underlying mechanisms.
Collapse
Affiliation(s)
- Hajime Suzuki
- Department of Psychosomatic Internal Medicine, Kagoshima University Graduate School of Medical and Dental Sciences, 8-35-1 Sakuragaoka, Kagoshima, 890-8520, Japan
| | | | | | | | | |
Collapse
|
95
|
Cashion A, Stanfill A, Thomas F, Xu L, Sutter T, Eason J, Ensell M, Homayouni R. Expression levels of obesity-related genes are associated with weight change in kidney transplant recipients. PLoS One 2013; 8:e59962. [PMID: 23544116 PMCID: PMC3609773 DOI: 10.1371/journal.pone.0059962] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2012] [Accepted: 02/24/2013] [Indexed: 11/23/2022] Open
Abstract
Background The aim of this study was to investigate the association of gene expression profiles in subcutaneous adipose tissue with weight change in kidney transplant recipients and to gain insights into the underlying mechanisms of weight gain. Methodology/Principal Findings A secondary data analysis was done on a subgroup (n = 26) of existing clinical and gene expression data from a larger prospective longitudinal study examining factors contributing to weight gain in transplant recipients. Measurements taken included adipose tissue gene expression profiles at time of transplant, baseline and six-month weight, and demographic data. Using multivariate linear regression analysis controlled for race and gender, expression levels of 1553 genes were significantly (p<0.05) associated with weight change. Functional analysis using Gene Ontology and Kyoto Encyclopedia of Genes and Genomes classifications identified metabolic pathways that were enriched in this dataset. Furthermore, GeneIndexer literature mining analysis identified a subset of genes that are highly associated with obesity in the literature and Ingenuity pathway analysis revealed several significant gene networks associated with metabolism and endocrine function. Polymorphisms in several of these genes have previously been linked to obesity. Conclusions/Significance We have successfully identified a set of molecular pathways that taken together may provide insights into the mechanisms of weight gain in kidney transplant recipients. Future work will be done to determine how these pathways may contribute to weight gain.
Collapse
Affiliation(s)
- Ann Cashion
- Department of Acute and Chronic Care, College of Nursing, University of Tennessee Health Science Center, Memphis, Tennessee, United States of America.
| | | | | | | | | | | | | | | |
Collapse
|
96
|
Cyr NE, Toorie AM, Steger JS, Sochat MM, Hyner S, Perello M, Stuart R, Nillni EA. Mechanisms by which the orexigen NPY regulates anorexigenic α-MSH and TRH. Am J Physiol Endocrinol Metab 2013; 304:E640-50. [PMID: 23321476 PMCID: PMC3602689 DOI: 10.1152/ajpendo.00448.2012] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/06/2012] [Accepted: 01/12/2013] [Indexed: 01/24/2023]
Abstract
Protein posttranslational processing is a cellular mechanism fundamental to the generation of bioactive peptides, including the anorectic α-melanocyte-stimulating hormone (α-MSH) and thyrotropin-releasing hormone (TRH) peptides produced in the hypothalamic arcuate (ARC) and paraventricular (PVN) nuclei, respectively. Neuropeptide Y (NPY) promotes positive energy balance in part by suppressing α-MSH and TRH. The mechanism by which NPY regulates α-MSH output, however, is not well understood. Our results reveal that NPY inhibited the posttranslational processing of α-MSH's inactive precursor proopiomelanocortin (POMC) by decreasing the prohormone convertase-2 (PC2). We also found that early growth response protein-1 (Egr-1) and NPY-Y1 receptors mediated the NPY-induced decrease in PC2. NPY given intra-PVN also decreased PC2 in PVN samples, suggesting a reduction in PC2-mediated pro-TRH processing. In addition, NPY attenuated the α-MSH-induced increase in TRH production by two mechanisms. First, NPY decreased α-MSH-induced CREB phosphorylation, which normally enhances TRH transcription. Second, NPY decreased the amount of α-MSH in the PVN. Collectively, these results underscore the significance of the interaction between NPY and α-MSH in the central regulation of energy balance and indicate that posttranslational processing is a mechanism that plays a specific role in this interaction.
Collapse
Affiliation(s)
- Nicole E Cyr
- Division of Endocrinology, Department of Medicine, The Warren Alpert Medical School of Brown University/Rhode Island Hospital, Providence, RI 02903, USA
| | | | | | | | | | | | | | | |
Collapse
|
97
|
Douris N, Maratos-Flier E. Two paths diverge in the brain: melanin-concentrating hormone controls hepatic and adipose metabolism. Gastroenterology 2013; 144:501-4. [PMID: 23347675 DOI: 10.1053/j.gastro.2013.01.029] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/02/2022]
|
98
|
Aller MA, Arias JI, Prieto I, Gilsanz C, Arias A, Yang H, Arias J. Surgical inflammatory stress: the embryo takes hold of the reins again. Theor Biol Med Model 2013; 10:6. [PMID: 23374964 PMCID: PMC3577641 DOI: 10.1186/1742-4682-10-6] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2012] [Accepted: 01/18/2013] [Indexed: 01/07/2023] Open
Abstract
The surgical inflammatory response can be a type of high-grade acute stress response associated with an increasingly complex trophic functional system for using oxygen. This systemic neuro-immune-endocrine response seems to induce the re-expression of 2 extraembryonic-like functional axes, i.e. coelomic-amniotic and trophoblastic-yolk-sac-related, within injured tissues and organs, thus favoring their re-development. Accordingly, through the up-regulation of two systemic inflammatory phenotypes, i.e. neurogenic and immune-related, a gestational-like response using embryonic functions would be induced in the patient's injured tissues and organs, which would therefore result in their repair. Here we establish a comparison between the pathophysiological mechanisms that are produced during the inflammatory response and the physiological mechanisms that are expressed during early embryonic development. In this way, surgical inflammation could be a high-grade stress response whose pathophysiological mechanisms would be based on the recapitulation of ontogenic and phylogenetic-related functions. Thus, the ultimate objective of surgical inflammation, as a gestational process, is creating new tissues/organs for repairing the injured ones. Since surgical inflammation and early embryonic development share common production mechanisms, the factors that hamper the wound healing reaction in surgical patients could be similar to those that impair the gestational process.
Collapse
Affiliation(s)
- Maria-Angeles Aller
- Department of Surgery, School of Medicine, Complutense University of Madrid, Madrid, Spain
| | - Jose-Ignacio Arias
- General and Digestive Surgery Unit, Monte Naranco Hospital, Oviedo, Asturias, Spain
| | - Isabel Prieto
- Department of General and Digestive Surgery, La Paz Hospital, Autonomous University, Madrid, Spain
| | - Carlos Gilsanz
- General and Digestive Surgery Unit, Sudeste University Hospital, Arganda del Rey, Madrid, Spain
| | - Ana Arias
- Department of Medicine, Puerta de Hierro Hospital, Autonomous University, Madrid, Spain
| | - Heping Yang
- Division of Gastroenterology and Liver Disease, USC Research Centre for Liver Diseases, Los Angeles, CA, USA
| | - Jaime Arias
- Department of Surgery, School of Medicine, Complutense University of Madrid, Madrid, Spain
| |
Collapse
|
99
|
Jeong JK, Diano S. Prolyl carboxypeptidase and its inhibitors in metabolism. Trends Endocrinol Metab 2013; 24:61-7. [PMID: 23245768 PMCID: PMC3893043 DOI: 10.1016/j.tem.2012.11.001] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/11/2012] [Revised: 10/30/2012] [Accepted: 11/04/2012] [Indexed: 01/09/2023]
Abstract
Proopiomelanocortin (POMC)-expressing neurons in the hypothalamus integrate a variety of central and peripheral metabolic inputs, and regulate energy homeostasis by controlling energy expenditure and food intake. To accomplish this, a precise balance of production and degradation of α-melanocyte-stimulating hormone (α-MSH), an anorexigenic neuropeptide and product of the POMC gene, in the hypothalamus, is crucial. Prolyl carboxypeptidase (PRCP) is a key enzyme that degrades α-MSH to an inactive form unable to inhibit food intake. Because it represents a new therapeutic target for the treatment of metabolic disorders, such as obesity and diabetes, efforts have been made to generate potent, brain-penetrant PRCP inhibitors. Here, we discuss the role of PRCP on energy metabolism and the development of PRCP inhibitors.
Collapse
Affiliation(s)
- Jin Kwon Jeong
- Program in Integrative Cell Signaling and Neurobiology of Metabolism, Yale University School of Medicine, New Haven, CT 06520, USA
| | | |
Collapse
|
100
|
Aller MA, Blanco-Rivero J, Arias JI, Balfagon G, Arias J. The wound-healing response and upregulated embryonic mechanisms: brothers-in-arms forever. Exp Dermatol 2012; 21:497-503. [PMID: 22716244 DOI: 10.1111/j.1600-0625.2012.01525.x] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The cutaneous wound-healing reaction occurs in overlapping but inter-related phases, which ultimately result in fibrosis. The pathophysiological mechanisms involved in fibrotic diseases, including organ-related and even systemic diseases, such as systemic sclerosis, could represent the successive systemic upregulation of extraembryonic-like phenotypes, that is, amniotic and vitelline phenotypes. These two extraembryonic-like phenotypes act on the injured tissue to induce a process similar to gastrulation, which occurs during the early phases of embryo development. The amniotic-like phenotype plays a leading role in the development of neurogenic responses with significant hydroelectrolytic alterations that essentially represent the development of open microcirculation within the injured tissue. In turn, through the overlapping expression of a vitelline-like phenotype, a bone marrow-related response is produced. Interstitial infiltration by molecular and cellular mediators contributed by amniotic- and vitelline-like functions provides the functional and metabolic autonomy needed for inducing new tissue formation through mechanisms similar to those that act in gastrulation during the early phases of embryonic development. Thus, while a new tissue is formed, it quickly evolves into fibrotic tissue because of premature senescence. Mechanisms related to extraembryonic-like functions have been suggested in the following physiological and pathological processes: embryonic development; wound-healing reactions occurring during adult life; and senescence. The existence of this sort of basic self-organizing fractal-like functional pattern is an essential characteristic of our way of life.
Collapse
Affiliation(s)
- María-Angeles Aller
- Department of Surgery I, School of Medicine, Complutense University of Madrid, Madrid, Spain
| | | | | | | | | |
Collapse
|