1
|
Stefanakis K, Upadhyay J, Ramirez-Cisneros A, Patel N, Sahai A, Mantzoros CS. Leptin physiology and pathophysiology in energy homeostasis, immune function, neuroendocrine regulation and bone health. Metabolism 2024; 161:156056. [PMID: 39481533 DOI: 10.1016/j.metabol.2024.156056] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/04/2024] [Revised: 10/28/2024] [Accepted: 10/28/2024] [Indexed: 11/02/2024]
Abstract
Since its discovery and over the past thirty years, extensive research has significantly expanded our understanding of leptin and its diverse roles in human physiology, pathophysiology and therapeutics. A prototypical adipokine initially identified for its critical function in appetite regulation and energy homeostasis, leptin has been revealed to also exert profound effects on the hypothalamic-pituitary-gonadal, thyroid, adrenal and growth hormone axis, differentially between animals and humans, as well as in regulating immune function. Beyond these roles, leptin plays a pivotal role in significantly affecting bone health by promoting bone formation and regulating bone metabolism both directly and indirectly through its neuroendocrine actions. The diverse actions of leptin are particularly notable in leptin-deficient animal models and in conditions characterized by low circulating leptin levels, such as lipodystrophies and relative energy deficiency. Conversely, the effectiveness of leptin is attenuated in leptin-sufficient states, such as obesity and other high-adiposity conditions associated with hyperleptinemia and leptin tolerance. This review attempts to consolidate 30 years of leptin research with an emphasis on its physiology and pathophysiology in humans, including its promising therapeutic potential. We discuss preclinical and human studies describing the pathophysiology of energy deficiency across organ systems and the significant role of leptin in regulating neuroendocrine, immune, reproductive and bone health. We finally present past proof of concept clinical trials of leptin administration in leptin-deficient subjects that have demonstrated positive neuroendocrine, reproductive, and bone health outcomes, setting the stage for future phase IIb and III randomized clinical trials in these conditions.
Collapse
Affiliation(s)
- Konstantinos Stefanakis
- Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Jagriti Upadhyay
- Department of Medicine, Lahey Hospital and Medical Center, Burlington, MA, USA
| | - Arantxa Ramirez-Cisneros
- Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Nihar Patel
- Department of Medicine, Lahey Hospital and Medical Center, Burlington, MA, USA
| | - Akshat Sahai
- Vassar Brothers Medical Center, Poughkeepsie, NY, USA
| | - Christos S Mantzoros
- Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA; Department of Medicine, Boston VA Healthcare System, Boston, MA, USA.
| |
Collapse
|
2
|
Targa G, Mottarlini F, Rizzi B, Taddini S, Parolaro S, Fumagalli F, Caffino L. Anorexia-Induced Hypoleptinemia Drives Adaptations in the JAK2/STAT3 Pathway in the Ventral and Dorsal Hippocampus of Female Rats. Nutrients 2024; 16:1171. [PMID: 38674862 PMCID: PMC11054075 DOI: 10.3390/nu16081171] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2024] [Revised: 04/04/2024] [Accepted: 04/12/2024] [Indexed: 04/28/2024] Open
Abstract
Leptin is an appetite-regulating adipokine that is reduced in patients with anorexia nervosa (AN), a psychiatric disorder characterized by self-imposed starvation, and has been linked to hyperactivity, a hallmark of AN. However, it remains unknown how leptin receptor (LepR) and its JAK2-STAT3 downstream pathway in extrahypothalamic brain areas, such as the dorsal (dHip) and ventral (vHip) hippocampus, crucial for spatial memory and emotion regulation, may contribute to the maintenance of AN behaviors. Taking advantage of the activity-based anorexia (ABA) model (i.e., the combination of food restriction and physical activity), we observed reduced leptin plasma levels in adolescent female ABA rats at the acute phase of the disorder [post-natal day (PND) 42], while the levels increased over control levels following a 7-day recovery period (PND49). The analysis of the intracellular leptin pathway revealed that ABA rats showed an overall decrease of the LepR/JAK2/STAT3 signaling in dHip at both time points, while in vHip we observed a transition from hypo- (PND42) to hyperactivation (PND49) of the pathway. These changes might add knowledge on starvation-induced fluctuations in leptin levels and in hippocampal leptin signaling as initial drivers of the transition from adaptative mechanisms to starvation toward the maintenance of aberrant behaviors typical of AN patients, such as perpetuating restraint over eating.
Collapse
Affiliation(s)
- Giorgia Targa
- Department of Pharmacological and Biomolecular Sciences ‘Rodolfo Paoletti’, Università degli Studi di Milano, Via Balzaretti 9, 20133 Milano, Italy; (G.T.); (F.M.); (B.R.); (S.T.); (S.P.); (F.F.)
| | - Francesca Mottarlini
- Department of Pharmacological and Biomolecular Sciences ‘Rodolfo Paoletti’, Università degli Studi di Milano, Via Balzaretti 9, 20133 Milano, Italy; (G.T.); (F.M.); (B.R.); (S.T.); (S.P.); (F.F.)
| | - Beatrice Rizzi
- Department of Pharmacological and Biomolecular Sciences ‘Rodolfo Paoletti’, Università degli Studi di Milano, Via Balzaretti 9, 20133 Milano, Italy; (G.T.); (F.M.); (B.R.); (S.T.); (S.P.); (F.F.)
- Center for Neuroscience, University of Camerino, 62032 Camerino, Italy
| | - Sofia Taddini
- Department of Pharmacological and Biomolecular Sciences ‘Rodolfo Paoletti’, Università degli Studi di Milano, Via Balzaretti 9, 20133 Milano, Italy; (G.T.); (F.M.); (B.R.); (S.T.); (S.P.); (F.F.)
| | - Susanna Parolaro
- Department of Pharmacological and Biomolecular Sciences ‘Rodolfo Paoletti’, Università degli Studi di Milano, Via Balzaretti 9, 20133 Milano, Italy; (G.T.); (F.M.); (B.R.); (S.T.); (S.P.); (F.F.)
| | - Fabio Fumagalli
- Department of Pharmacological and Biomolecular Sciences ‘Rodolfo Paoletti’, Università degli Studi di Milano, Via Balzaretti 9, 20133 Milano, Italy; (G.T.); (F.M.); (B.R.); (S.T.); (S.P.); (F.F.)
| | - Lucia Caffino
- Department of Pharmacological and Biomolecular Sciences ‘Rodolfo Paoletti’, Università degli Studi di Milano, Via Balzaretti 9, 20133 Milano, Italy; (G.T.); (F.M.); (B.R.); (S.T.); (S.P.); (F.F.)
| |
Collapse
|
3
|
Liu Z, Xiao T, Liu H. Leptin signaling and its central role in energy homeostasis. Front Neurosci 2023; 17:1238528. [PMID: 38027481 PMCID: PMC10644276 DOI: 10.3389/fnins.2023.1238528] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2023] [Accepted: 10/17/2023] [Indexed: 12/01/2023] Open
Abstract
Leptin plays a critical role in regulating appetite, energy expenditure and body weight, making it a key factor in maintaining a healthy balance. Despite numerous efforts to develop therapeutic interventions targeting leptin signaling, their effectiveness has been limited, underscoring the importance of gaining a better understanding of the mechanisms through which leptin exerts its functions. While the hypothalamus is widely recognized as the primary site responsible for the appetite-suppressing and weight-reducing effects of leptin, other brain regions have also been increasingly investigated for their involvement in mediating leptin's action. In this review, we summarize leptin signaling pathways and the neural networks that mediate the effects of leptin, with a specific emphasis on energy homeostasis.
Collapse
Affiliation(s)
- Zhaoxun Liu
- Nursing Department, The Third Xiangya Hospital, Central South University, Changsha, Hunan, China
- Department of Emergency, The Third Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Tao Xiao
- Nursing Department, The Third Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Hailan Liu
- USDA/ARS Children’s Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, Houston, TX, United States
| |
Collapse
|
4
|
Nozari Y, Park C, Brietzke E, Iacobucci M, Gill H, McIntyre RS. Correlation between improved leptin signaling and cognitive function post bariatric surgery. J Affect Disord 2023; 326:225-231. [PMID: 36736790 DOI: 10.1016/j.jad.2023.01.100] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/27/2021] [Revised: 01/23/2023] [Accepted: 01/27/2023] [Indexed: 02/04/2023]
Abstract
Determining whether changes in leptin signaling plays a role in the improvement of cognitive function post-bariatric surgery may aid in the understanding and development of novel therapeutic approaches targeting cognitive dysfunction through the greater understanding of processes connecting obesity and brain health. Several studies have explored the effects of cognition post bariatric surgery, and others have studied leptin and its changes post surgery. However the amalgamation of the effects of leptin signaling in relation to cognition post bariatric surgery have yet to be considered as key tools in the understanding of cognitive dysfunction in obese subjects with leptin resistance or insensitivity. This review serves to highlight the potential correlations, to further elucidate the effect of improved leptin signaling on cognition post bariatric surgery, and to propose a direct cause for the improvement of cognitive function via the amelioration of the leptin Janus kinase/Signal transducer and activator of transcription (JAK/STAT) signaling pathway as a result of the reversal of inflammatory processes involved in diseased individuals.
Collapse
Affiliation(s)
- Y Nozari
- Mood Disorders Psychopharmacology Unit, Toronto Western Hospital, Toronto, ON, Canada; University of Toronto HBSc, Toronto, ON, Canada.
| | - C Park
- Mood Disorders Psychopharmacology Unit, Toronto Western Hospital, Toronto, ON, Canada; University of Toronto MSc, Toronto, ON, Canada
| | - E Brietzke
- Department of Psychiatry, Queen's University School of Medicine, Kingston, ON, Canada; Centre for Neuroscience Studies (CNS), Queen's University, Kingston, ON, Canada
| | - M Iacobucci
- Mood Disorders Psychopharmacology Unit, Toronto Western Hospital, Toronto, ON, Canada; University of Toronto HBSc, Toronto, ON, Canada
| | - H Gill
- Mood Disorders Psychopharmacology Unit, Toronto Western Hospital, Toronto, ON, Canada; Institute of Medical Science, University of Toronto, Toronto, ON, Canada
| | - R S McIntyre
- Mood Disorders Psychopharmacology Unit, Toronto Western Hospital, Toronto, ON, Canada; Department of Pharmacology, University of Toronto, Toronto, ON, Canada; Department of Psychiatry, University of Toronto, Toronto, ON, Canada; Brain and Cognition Discovery Foundation, Toronto, ON, Canada
| |
Collapse
|
5
|
Leptin in the Commissural Nucleus of the Tractus Solitarius (cNTS) and Anoxic Stimulus in the Carotid Body Chemoreceptors Increases cNTS Leptin Signaling Receptor and Brain Glucose Retention in Rats. MEDICINA (KAUNAS, LITHUANIA) 2022; 58:medicina58040550. [PMID: 35454388 PMCID: PMC9025962 DOI: 10.3390/medicina58040550] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/23/2022] [Revised: 04/10/2022] [Accepted: 04/12/2022] [Indexed: 11/29/2022]
Abstract
Background and Objectives: The commissural nucleus of the tractus solitarius (cNTS) not only responds to glucose levels directly, but also receives afferent signals from the liver, and from the carotid chemoreceptors (CChR). In addition, leptin, through its receptors in the cNTS, regulates food intake, body weight, blood glucose levels, and brain glucose retention (BGR). These leptin effects on cNTS are thought to be mediated through the sympathetic–adrenal system. How these different sources of information converging in the NTS regulate blood glucose levels and brain glucose retention remains largely unknown. The goal of the present study was to determine whether the local administration of leptin in cNTS alone, or after local anoxic stimulation using sodium cyanide (NaCN) in the carotid sinus, modifies the expression of leptin Ob-Rb and of c-Fos mRNA. We also investigated how leptin, alone, or in combination with carotid sinus stimulation, affected brain glucose retention. Materials and Methods: The experiments were carried out in anesthetized male Wistar rats artificially ventilated to maintain homeostatic values for pO2, pCO2, and pH. We had four groups: (a) experimental 1, leptin infusion in cNTS and NaCN in the isolated carotid sinus (ICS; n = 10); (b) experimental 2, leptin infusion in cNTS and saline in the ICS (n = 10); (c) control 1, artificial cerebrospinal fluid (aCSF) in cNTS and NaCN in the ICS (n = 10); (d) control 2, aCSF in cNTS and saline in the ICS (n = 10). Results: Leptin in cNTS, preceded by NaCN in the ICS increased BGR and leptin Ob-Rb mRNA receptor expression, with no significant increases in c-Fos mRNA in the NTSc. Conclusions: Leptin in the cNTS enhances brain glucose retention induced by an anoxic stimulus in the carotid chemoreceptors, through an increase in Ob-Rb receptors, without persistent changes in neuronal activation.
Collapse
|
6
|
Kamstra K, Rizwan MZ, Grattan DR, Horsfield JA, Tups A. Leptin regulates glucose homeostasis via the canonical Wnt pathway in the zebrafish. FASEB J 2022; 36:e22207. [PMID: 35188286 DOI: 10.1096/fj.202101764r] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2021] [Revised: 01/12/2022] [Accepted: 02/01/2022] [Indexed: 11/11/2022]
Abstract
Leptin is best known for its role in adipostasis, but it also regulates blood glucose levels. The molecular mechanism by which leptin controls glucose homeostasis remains largely unknown. Here, we use a zebrafish model to show that Wnt signaling mediates the glucoregulatory effects of leptin. Under normal feeding conditions, leptin regulates glucose homeostasis but not adipostasis in zebrafish. In times of nutrient excess, however, we found that leptin also regulates body weight and size. Using a Wnt signaling reporter fish, we show that leptin activates the canonical Wnt pathway in vivo. Utilizing two paradigms for hyperglycemia, it is revealed that leptin regulates glucose homeostasis via the Wnt pathway, as pharmacological inhibition of this pathway impairs the glucoregulatory actions of leptin. Our results may shed new light on the evolution of the physiological function of leptin.
Collapse
Affiliation(s)
- Kaj Kamstra
- Centre for Neuroendocrinology and Brain Health Research Centre, University Otago, Dunedin, New Zealand.,Department of Physiology, Dunedin School of Medicine, University of Otago, Dunedin, New Zealand
| | - Mohammed Z Rizwan
- Centre for Neuroendocrinology and Brain Health Research Centre, University Otago, Dunedin, New Zealand.,Department of Anatomy, Dunedin School of Medicine, University of Otago, Dunedin, New Zealand
| | - David R Grattan
- Centre for Neuroendocrinology and Brain Health Research Centre, University Otago, Dunedin, New Zealand.,Department of Anatomy, Dunedin School of Medicine, University of Otago, Dunedin, New Zealand
| | - Julia A Horsfield
- Department of Pathology, University of Otago, Dunedin, New Zealand.,Maurice Wilkins Centre for Molecular Biodiscovery, University of Auckland, Auckland, New Zealand
| | - Alexander Tups
- Centre for Neuroendocrinology and Brain Health Research Centre, University Otago, Dunedin, New Zealand.,Department of Physiology, Dunedin School of Medicine, University of Otago, Dunedin, New Zealand
| |
Collapse
|
7
|
Yoo S, Kim J, Lyu P, Hoang TV, Ma A, Trinh V, Dai W, Jiang L, Leavey P, Duncan L, Won JK, Park SH, Qian J, Brown SP, Blackshaw S. Control of neurogenic competence in mammalian hypothalamic tanycytes. SCIENCE ADVANCES 2021; 7:eabg3777. [PMID: 34049878 PMCID: PMC8163082 DOI: 10.1126/sciadv.abg3777] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/31/2020] [Accepted: 04/09/2021] [Indexed: 05/07/2023]
Abstract
Hypothalamic tanycytes, radial glial cells that share many features with neuronal progenitors, can generate small numbers of neurons in the postnatal hypothalamus, but the identity of these neurons and the molecular mechanisms that control tanycyte-derived neurogenesis are unknown. In this study, we show that tanycyte-specific disruption of the NFI family of transcription factors (Nfia/b/x) robustly stimulates tanycyte proliferation and tanycyte-derived neurogenesis. Single-cell RNA sequencing (scRNA-seq) and single-cell assay for transposase-accessible chromatin sequencing (scATAC-seq) analysis reveals that NFI (nuclear factor I) factors repress Sonic hedgehog (Shh) and Wnt signaling in tanycytes and modulation of these pathways blocks proliferation and tanycyte-derived neurogenesis in Nfia/b/x-deficient mice. Nfia/b/x-deficient tanycytes give rise to multiple mediobasal hypothalamic neuronal subtypes that can mature, fire action potentials, receive synaptic inputs, and selectively respond to changes in internal states. These findings identify molecular mechanisms that control tanycyte-derived neurogenesis, which can potentially be targeted to selectively remodel the hypothalamic neural circuitry that controls homeostatic physiological processes.
Collapse
Affiliation(s)
- Sooyeon Yoo
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University, Baltimore, MD 21205, USA
- Department of Pathology, Seoul National University Hospital, 71 Daehak-ro, Jongno-gu 03082, Republic of Korea
| | - Juhyun Kim
- Department of Psychiatry and Behavioral Science, Johns Hopkins University, Baltimore, MD 21205, USA
| | - Pin Lyu
- Department of Ophthalmology, Johns Hopkins University, Baltimore, MD 21205, USA
| | - Thanh V Hoang
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University, Baltimore, MD 21205, USA
| | - Alex Ma
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University, Baltimore, MD 21205, USA
| | - Vickie Trinh
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University, Baltimore, MD 21205, USA
| | - Weina Dai
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University, Baltimore, MD 21205, USA
| | - Lizhi Jiang
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University, Baltimore, MD 21205, USA
| | - Patrick Leavey
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University, Baltimore, MD 21205, USA
| | - Leighton Duncan
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University, Baltimore, MD 21205, USA
| | - Jae-Kyung Won
- Department of Pathology, Seoul National University Hospital, 71 Daehak-ro, Jongno-gu 03082, Republic of Korea
| | - Sung-Hye Park
- Department of Pathology, Seoul National University Hospital, 71 Daehak-ro, Jongno-gu 03082, Republic of Korea
| | - Jiang Qian
- Department of Ophthalmology, Johns Hopkins University, Baltimore, MD 21205, USA
| | - Solange P Brown
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University, Baltimore, MD 21205, USA
- Kavli Neuroscience Discovery Institute, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Seth Blackshaw
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University, Baltimore, MD 21205, USA.
- Department of Ophthalmology, Johns Hopkins University, Baltimore, MD 21205, USA
- Kavli Neuroscience Discovery Institute, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
- Department of Neurology, Johns Hopkins University, Baltimore, MD 21205, USA
- Institute for Cell Engineering, Johns Hopkins University, Baltimore, MD 21205, USA
| |
Collapse
|
8
|
Liu H, Du T, Li C, Yang G. STAT3 phosphorylation in central leptin resistance. Nutr Metab (Lond) 2021; 18:39. [PMID: 33849593 PMCID: PMC8045279 DOI: 10.1186/s12986-021-00569-w] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2021] [Accepted: 04/03/2021] [Indexed: 12/20/2022] Open
Abstract
Mechanism exploitation of energy homeostasis is urgently required because of the worldwide prevailing of obesity-related metabolic disorders in human being. Although it is well known that leptin plays a central role in regulating energy balance by suppressing food intake and promoting energy expenditure, the existence of leptin resistance in majority of obese individuals hampers the utilization of leptin therapy against these disorders. However, the mechanism of leptin resistance is largely unknown in spite of the globally enormous endeavors. Current theories to interpret leptin resistance include the impairment of leptin transport, attenuation of leptin signaling, chronic inflammation, ER tress, deficiency of autophagy, as well as leptin itself. Leptin-activated leptin receptor (LepRb) signals in hypothalamus via several pathways, in which JAK2-STAT3 pathway, the most extensively investigated one, is considered to mediate the major action of leptin in energy regulation. Upon leptin stimulation the phosphorylation of STAT3 is one of the key events in JAK2-STAT3 pathway, followed by the dimerization and nuclear translocation of this molecule. Phosphorylated STAT3 (p-STAT3), as a transcription factor, binds to and regulates its target gene such as POMC gene, playing the physiological function of leptin. Regarding POMC gene in hypothalamus however little is known about the detail of its interaction with STAT3. Moreover the status of p-STAT3 and its significance in hypothalamus of DIO mice needs to be well elucidated. This review comprehends literatures on leptin and leptin resistance and especially discusses what STAT3 phosphorylation would contribute to central leptin resistance.
Collapse
Affiliation(s)
- Huimin Liu
- College of Life Science, Henan Agricultural University, 95 Wen Hua Road, Zhengzhou, 450002, China
| | - Tianxin Du
- College of Life Science, Henan Agricultural University, 95 Wen Hua Road, Zhengzhou, 450002, China
| | - Chen Li
- College of Life Science, Henan Agricultural University, 95 Wen Hua Road, Zhengzhou, 450002, China
| | - Guoqing Yang
- College of Life Science, Henan Agricultural University, 95 Wen Hua Road, Zhengzhou, 450002, China.
| |
Collapse
|
9
|
Boucsein A, Kamstra K, Tups A. Central signalling cross-talk between insulin and leptin in glucose and energy homeostasis. J Neuroendocrinol 2021; 33:e12944. [PMID: 33615588 DOI: 10.1111/jne.12944] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/29/2020] [Revised: 01/10/2021] [Accepted: 01/27/2021] [Indexed: 12/28/2022]
Abstract
Energy homeostasis is controlled by an intricate regulatory system centred in the brain. The peripheral adiposity signals insulin and leptin play a crucial role in this system by informing the brain of the energy status of the body and mediating their catabolic effects through signal transduction in hypothalamic areas that control food intake, energy expenditure and glucose metabolism. Disruptions of insulin and leptin signalling can result in diabetes and obesity. The central signalling cross-talk between insulin and leptin is essential for maintenance of normal healthy energy homeostasis. An important role of leptin in glucoregulation has been revealed. Typically regarded as being controlled by insulin, the control of glucose homeostasis critically depends on functional leptin action. Leptin, on the other hand, is able to lower glucose levels in the absence of insulin, although insulin is necessary for long-term stabilisation of euglycaemia. Evidence from rodent models and human patients suggests that leptin improves insulin sensitivity in type 1 diabetes. The signalling cross-talk between insulin and leptin is likely conveyed by the WNT/β-catenin pathway. Leptin activates WNT/β-catenin signalling, leading to inhibition of glycogen synthase kinase-3β, a key inhibitor of insulin action, thereby facilitating improved insulin signal transduction and sensitisation of insulin action. Interestingly, insights into the roles of insulin and leptin in insects and fish indicate that leptin may have initially evolved as a glucoregulatory hormone and that its anorexigenic and body weight regulatory function was acquired throughout evolution. Furthermore, the regulation of both central and peripheral control of energy homeostasis is tightly controlled by the circadian clock, allowing adaptation of homeostatic processes to environmental cues.
Collapse
Affiliation(s)
- Alisa Boucsein
- Centre for Neuroendocrinology, Department of Physiology, School of Biomedical Sciences, University of Otago, Dunedin, New Zealand
| | - Kaj Kamstra
- Centre for Neuroendocrinology, Department of Physiology, School of Biomedical Sciences, University of Otago, Dunedin, New Zealand
| | - Alexander Tups
- Centre for Neuroendocrinology, Department of Physiology, School of Biomedical Sciences, University of Otago, Dunedin, New Zealand
- Maurice Wilkins Centre for Molecular Biodiscovery, University of Auckland, Auckland, New Zealand
| |
Collapse
|
10
|
Yoo S, Cha D, Kim S, Jiang L, Cooke P, Adebesin M, Wolfe A, Riddle R, Aja S, Blackshaw S. Tanycyte ablation in the arcuate nucleus and median eminence increases obesity susceptibility by increasing body fat content in male mice. Glia 2020; 68:1987-2000. [PMID: 32173924 PMCID: PMC7423758 DOI: 10.1002/glia.23817] [Citation(s) in RCA: 53] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2019] [Revised: 02/28/2020] [Accepted: 02/28/2020] [Indexed: 12/11/2022]
Abstract
Tanycytes are radial glial cells located in the mediobasal hypothalamus. Recent studies have proposed that tanycytes play an important role in hypothalamic control of energy homeostasis, although this has not been directly tested. Here, we report the phenotype of mice in which tanycytes of the arcuate nucleus and median eminence were conditionally ablated in adult mice. Although the cerebrospinal fluid-hypothalamic barrier was rendered more permeable following tanycyte ablation, neither the blood-hypothalamic barrier nor leptin-induced pSTAT3 activation in hypothalamic parenchyma were affected. We observed a significant increase in visceral fat distribution accompanying insulin insensitivity in male mice, without significant effect on either body weight or food intake. A high-fat diet tended to accelerate overall body weight gain in tanycyte-ablated mice, but the development of visceral adiposity and insulin insensitivity was comparable to wildtype. Thermoneutral housing exacerbated fat accumulation and produced a shift away from fat oxidation in tanycyte-ablated mice. These results clarify the extent to which tanycytes regulate energy balance, and demonstrate a role for tanycytes in regulating fat metabolism.
Collapse
Affiliation(s)
- Sooyeon Yoo
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - David Cha
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Soohyun Kim
- Department of Orthopedic Surgery, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Lizhi Jiang
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Patrick Cooke
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Mobolanie Adebesin
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Andrew Wolfe
- Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Ryan Riddle
- Department of Orthopedic Surgery, Johns Hopkins University School of Medicine, Baltimore, Maryland
- Baltimore Veterans Administration Medical Center, Baltimore, Maryland
| | - Susan Aja
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Seth Blackshaw
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, Maryland
- Department of Ophthalmology, Johns Hopkins University School of Medicine, Baltimore, Maryland
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, Maryland
- Center for Human Systems Biology, Johns Hopkins University School of Medicine, Baltimore, Maryland
- Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland
| |
Collapse
|
11
|
Hristov M, Landzhov B, Yakimova K. Cafeteria diet-induced obesity reduces leptin-stimulated NADPH-diaphorase reactivity in the hypothalamic arcuate nucleus of rats. Acta Histochem 2020; 122:151616. [PMID: 33066838 DOI: 10.1016/j.acthis.2020.151616] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2020] [Revised: 08/08/2020] [Accepted: 08/17/2020] [Indexed: 02/07/2023]
Abstract
Leptin is an adipokine that plays an important role in the regulation of energy homeostasis. The failure of endogenous and exogenous leptin to mediate its effects (for example, at suppressing appetite and decreasing body weight) has been termed leptin resistance. Hyperleptinemia and leptin resistance can be well demonstrated in animals in which obesity is induced by consumption of a palatable, high-calorie diet (e.g., cafeteria diet-induced obesity). Since leptin receptor signaling is known to be impaired in the hypothalamic arcuate nucleus (ARC) of obese rodents, we investigated the effect of leptin on nicotinamide adenine dinucleotide phosphate-diaphorase (NADPH-d) reactivity in the ARC of male Wistar rats with cafeteria diet-induced obesity. Our results have shown that after intraperitoneal administration of leptin, the number of NADPH-d positive neurons in the ARC was significantly lower in obese rats compared with that observed in normal weight rats. Additionally, we have found that leptin-induced NADPH-d staining in ARC neurons and the adjacent ependyma was decreased in obese rats. The results presented here suggest that the ability of leptin to activate nitric oxide synthase in neurons within the ARC as well as tanycytes and ependymal cells of the third ventricle is reduced in rats made obese by a cafeteria diet. We speculate that impairment in leptin-induced NO production presents a potential mechanism, involved in the pathogenesis of obesity and obesity-related disease states.
Collapse
Affiliation(s)
- Milen Hristov
- Department of Pharmacology and Toxicology, Faculty of Medicine, Medical University of Sofia, 2 "Zdrave" St., 1431 Sofia, Bulgaria.
| | - Boycho Landzhov
- Department of Anatomy, Histology and Embryology, Faculty of Medicine, Medical University of Sofia, 2 "Zdrave" St., 1431 Sofia, Bulgaria
| | - Krassimira Yakimova
- Department of Pharmacology and Toxicology, Faculty of Medicine, Medical University of Sofia, 2 "Zdrave" St., 1431 Sofia, Bulgaria
| |
Collapse
|
12
|
Nogueira G, Solon C, Carraro RS, Engel DF, Ramalho AF, Sidarta-Oliveira D, Gaspar RS, Bombassaro B, Vasques AC, Geloneze B, Vinolo MA, Donato Junior J, Velloso LA. Interleukin-17 acts in the hypothalamus reducing food intake. Brain Behav Immun 2020; 87:272-285. [PMID: 31863824 DOI: 10.1016/j.bbi.2019.12.012] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/06/2019] [Revised: 11/20/2019] [Accepted: 12/16/2019] [Indexed: 12/12/2022] Open
Abstract
Interleukin-17 (IL-17) is expressed in the intestine in response to changes in the gut microbiome landscape and plays an important role in intestinal and systemic inflammatory diseases. There is evidence that dietary factors can also modify the expression of intestinal IL-17. Here, we hypothesized that, similar to several other gut-produced factors, IL-17 may act in the hypothalamus to modulate food intake. We confirm that food intake increases IL-17 expression in the mouse ileum and human blood. There is no expression of IL-17 in the hypothalamus; however, IL-17 receptor A is expressed in both pro-opiomelanocortin (POMC) and agouti-related peptide (AgRP) neurons. Upon systemic injection, IL-17 promoted a rapid increase in hypothalamic POMC expression, which was followed by a late increase in the expression of AgRP. Both systemic and intracerebroventricular injections of IL-17 reduced calorie intake without affecting whole-body energy expenditure. Systemic but not intracerebroventricular injection of IL-17 increase brown adipose tissue temperature. Thus, IL-17 is a gut-produced factor that is controlled by diet and modulates food intake by acting in the hypothalamus. Our findings provide the first evidence of a cytokine that is acutely regulated by food intake and plays a role in the regulation of eating.
Collapse
Affiliation(s)
- Guilherme Nogueira
- Laboratory of Cell Signalling-Obesity and Comorbidities Research Center, University of Campinas, Campinas, Brazil
| | - Carina Solon
- Laboratory of Cell Signalling-Obesity and Comorbidities Research Center, University of Campinas, Campinas, Brazil
| | - Rodrigo S Carraro
- Laboratory of Cell Signalling-Obesity and Comorbidities Research Center, University of Campinas, Campinas, Brazil
| | - Daiane F Engel
- Laboratory of Cell Signalling-Obesity and Comorbidities Research Center, University of Campinas, Campinas, Brazil
| | - Albina F Ramalho
- Laboratory of Cell Signalling-Obesity and Comorbidities Research Center, University of Campinas, Campinas, Brazil
| | - Davi Sidarta-Oliveira
- Laboratory of Cell Signalling-Obesity and Comorbidities Research Center, University of Campinas, Campinas, Brazil
| | - Rodrigo S Gaspar
- Laboratory of Cell Signalling-Obesity and Comorbidities Research Center, University of Campinas, Campinas, Brazil
| | - Bruna Bombassaro
- Laboratory of Cell Signalling-Obesity and Comorbidities Research Center, University of Campinas, Campinas, Brazil
| | - Ana C Vasques
- Laboratory of Investigation in Metabolism and Diabetes, University of Campinas, Campinas, Brazil
| | - Bruno Geloneze
- Laboratory of Investigation in Metabolism and Diabetes, University of Campinas, Campinas, Brazil
| | - Marco A Vinolo
- Laboratory of Immunoinflammation, Department of Genetics, Evolution, Microbiology and Immunology, University of Campinas, Campinas, Brazil
| | - Jose Donato Junior
- Department of Physiology and Biophysics, Institute of Biomedical Sciences, University of Sao Paulo, Sao Paulo, Brazil
| | - Licio A Velloso
- Laboratory of Cell Signalling-Obesity and Comorbidities Research Center, University of Campinas, Campinas, Brazil.
| |
Collapse
|
13
|
Venema W, Severi I, Perugini J, Di Mercurio E, Mainardi M, Maffei M, Cinti S, Giordano A. Ciliary Neurotrophic Factor Acts on Distinctive Hypothalamic Arcuate Neurons and Promotes Leptin Entry Into and Action on the Mouse Hypothalamus. Front Cell Neurosci 2020; 14:140. [PMID: 32528252 PMCID: PMC7253709 DOI: 10.3389/fncel.2020.00140] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2020] [Accepted: 04/24/2020] [Indexed: 12/13/2022] Open
Abstract
In humans and experimental animals, the administration of ciliary neurotrophic factor (CNTF) reduces food intake and body weight. To gain further insights into the mechanism(s) underlying its satiety effect, we: (i) evaluated the CNTF-dependent activation of the Janus kinase 2 (JAK2) and signal transducer and activator of transcription 3 (STAT3) pathway in mouse models where neuropeptide Y (NPY) and pro-opiomelanocortin (POMC) neurons can be identified by green fluorescent protein (GFP); and (ii) assessed whether CNTF promotes leptin signaling in hypothalamic feeding centers. Immunohistochemical experiments enabled us to establish that intraperitoneal injection of mouse recombinant CNTF activated the JAK2-STAT3 pathway in a substantial proportion of arcuate nucleus (ARC) NPY neurons (18.68% ± 0.60 in 24-h fasted mice and 25.50% ± 1.17 in fed mice) but exerted a limited effect on POMC neurons (4.15% ± 0.33 in 24-h fasted mice and 2.84% ± 0.45 in fed mice). CNTF-responsive NPY neurons resided in the ventromedial ARC, facing the median eminence (ME), and were surrounded by albumin immunoreactivity, suggesting that they are located outside the blood-brain barrier (BBB). In both normally fed and high-fat diet (HFD) obese animals, CNTF activated extracellular signal-regulated kinase signaling in ME β1- and β2-tanycytes, an effect that has been linked to the promotion of leptin entry into the brain. Accordingly, compared to the animals treated with leptin, mice treated with leptin/CNTF showed: (i) a significantly greater leptin content in hypothalamic protein extracts; (ii) a significant increase in phospho-STAT3 (P-STAT3)-positive neurons in the ARC and the ventromedial hypothalamic nucleus of normally fed mice; and (iii) a significantly increased number of P-STAT3-positive neurons in the ARC and dorsomedial hypothalamic nucleus of HFD obese mice. Collectively, these data suggest that exogenously administered CNTF reduces food intake by exerting a leptin-like action on distinctive NPY ARC neurons and by promoting leptin signaling in hypothalamic feeding centers.
Collapse
Affiliation(s)
- Wiebe Venema
- Section of Neuroscience and Cell Biology, Department of Experimental and Clinical Medicine, Università Politecnica Delle Marche, Ancona, Italy
| | - Ilenia Severi
- Section of Neuroscience and Cell Biology, Department of Experimental and Clinical Medicine, Università Politecnica Delle Marche, Ancona, Italy
| | - Jessica Perugini
- Section of Neuroscience and Cell Biology, Department of Experimental and Clinical Medicine, Università Politecnica Delle Marche, Ancona, Italy
| | - Eleonora Di Mercurio
- Section of Neuroscience and Cell Biology, Department of Experimental and Clinical Medicine, Università Politecnica Delle Marche, Ancona, Italy
| | - Marco Mainardi
- Institute of Neuroscience, National Research Council, Pisa, Italy
| | | | - Saverio Cinti
- Section of Neuroscience and Cell Biology, Department of Experimental and Clinical Medicine, Università Politecnica Delle Marche, Ancona, Italy
- Center of Obesity, Università Politecnica delle Marche-United Hospitals, Ancona, Italy
| | - Antonio Giordano
- Section of Neuroscience and Cell Biology, Department of Experimental and Clinical Medicine, Università Politecnica Delle Marche, Ancona, Italy
| |
Collapse
|
14
|
Skinner NJ, Rizwan MZ, Grattan DR, Tups A. Chronic Light Cycle Disruption Alters Central Insulin and Leptin Signaling as well as Metabolic Markers in Male Mice. Endocrinology 2019; 160:2257-2270. [PMID: 31276158 DOI: 10.1210/en.2018-00935] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/02/2018] [Accepted: 06/20/2019] [Indexed: 01/25/2023]
Abstract
Recent evidence suggests that the circadian timing system plays a role in energy and glucose homeostasis, and disruptions to this system are a risk factor for the development of metabolic disorders. We exposed animals to a constantly shifting lighting environment comprised of a 6-hour advance, occurring every 6 days, to chronically disrupt their circadian timing system. This treatment caused a gradual increase in body weight of 12 ± 2% after 12 phase shifts, compared with a 6 ± 1% increase in mice under control lighting conditions. Additionally, after the fifth phase shift, light cycle-disrupted (CD) animals showed a reversal in their diurnal pattern of energy homeostasis and locomotor activity, followed by a subsequent loss of this rhythm. To investigate potential molecular mechanisms mediating these metabolic alterations, we assessed central leptin and insulin sensitivity. We discovered that CD mice had a decrease in central leptin signaling, as indicated by a reduction in the number of phosphorylated signal transducer and activator of transcription 3 immunoreactive cells in the arcuate nucleus of the hypothalamus. Furthermore, CD animals exhibited a marked increase in fasting blood glucose (269.4 ± 21.1 mg/dL) compared with controls (108.8 ± 21.3 mg/dL). This dramatic increase in fasting glucose levels was not associated with an increase in insulin levels, suggesting impairments in pancreatic insulin release. Peripheral hyperglycemia was accompanied by central alterations in insulin signaling at the level of phospho Akt and insulin receptor substrate 1, suggesting that light cycle disruption alters central insulin signaling. These results provide mechanistic insights into the association between light cycle disruption and metabolic disease.
Collapse
Affiliation(s)
- Nathan J Skinner
- Centre for Neuroendocrinology and Brain Health Research Centre, Department of Physiology, School of Biomedical Sciences, University of Otago, Dunedin, New Zealand
| | - Mohammed Z Rizwan
- Centre for Neuroendocrinology and Brain Health Research Centre, Department of Physiology, School of Biomedical Sciences, University of Otago, Dunedin, New Zealand
- Centre for Neuroendocrinology and Brain Health Research Centre, Department of Anatomy, School of Biomedical Sciences, University of Otago, Dunedin, New Zealand
| | - David R Grattan
- Centre for Neuroendocrinology and Brain Health Research Centre, Department of Anatomy, School of Biomedical Sciences, University of Otago, Dunedin, New Zealand
| | - Alexander Tups
- Centre for Neuroendocrinology and Brain Health Research Centre, Department of Physiology, School of Biomedical Sciences, University of Otago, Dunedin, New Zealand
| |
Collapse
|
15
|
Augustine RA, Knowles PJ, Khant Aung Z, Grattan DR, Ladyman SR. Impaired hypothalamic leptin sensitivity in pseudopregnant rats treated with chronic prolactin to mimic pregnancy. J Neuroendocrinol 2019; 31:e12702. [PMID: 30803074 DOI: 10.1111/jne.12702] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/31/2018] [Revised: 01/27/2019] [Accepted: 02/20/2019] [Indexed: 02/04/2023]
Abstract
Pregnancy in rodents is associated with hyperphagia, increased fat deposition, elevated leptin concentrations and insensitivity to the satiety action of leptin. To investigate the hormonal mechanisms involved in the development of this state of pregnancy-induced leptin resistance, we have used a pseudopregnancy rat model. We have previously demonstrated that pseudopregnant rats have a normal feeding response to leptin, although, if pseudopregnancy is extended using chronic i.c.v. ovine prolactin infusion along with progesterone implants, then leptin no longer suppresses food intake. The present study aimed to investigate the effect of chronically high lactogen levels, as seen in mid-pregnancy, on leptin-induced activation of hypothalamic Janus kinase/signal transducer and activator of transcription (JAK/STAT) signal transduction and mRNA expression of leptin (LepR-B) and prolactin (Prlr-L) receptors, using pseudopregnant rats chronically infused with ovine prolactin. Groups of virgin (dioestrous) and pseudopregnant rats were treated with chronic i.c.v. infusion of either prolactin (2.5 μg μL-1 h-1 for 5 days) or vehicle (artificial cerebrospinal fluid [aCSF]) via a minipump connected to a cannula surgically implanted into the lateral ventricle. Rats were fasted overnight and then received an i.c.v. injection of leptin (400 ng) or vehicle (aCSF) and were perfused 30 minutes later. In chronic vehicle-infused pseudopregnant rats, i.c.v. leptin increased the number of phosphorylated STAT3 positive cells in the arcuate nucleus and ventromedial nucleus (VMH) of the hypothalamus, similar to all acute-leptin treated virgin groups. This effect of leptin, however, was not observed in the pseudopregnant rats that were chronically infused with prolactin. A quantitative polymerase chain reaction analysis also showed decreased expression of LepR-B in the arcuate and VMH nuclei, as well as decreased Prlr-L in the arcuate nucleus of prolactin-infused "extended pseudopregnancy" rats. These data suggest that the attenuation of the leptin-induced suppression of food intake caused by chronically high lactogen levels in pseudopregnant rats is associated with impaired leptin-induced activation of the JAK/STAT pathway in specific hypothalamic nuclei.
Collapse
Affiliation(s)
- Rachael A Augustine
- Centre for Neuroendocrinology and Department of Physiology, University of Otago, Dunedin, New Zealand
| | - Penelope J Knowles
- Centre for Neuroendocrinology and Department of Anatomy, University of Otago, Dunedin, New Zealand
| | - Zin Khant Aung
- Centre for Neuroendocrinology and Department of Anatomy, University of Otago, Dunedin, New Zealand
| | - David R Grattan
- Centre for Neuroendocrinology and Department of Anatomy, University of Otago, Dunedin, New Zealand
- Maurice Wilkins Centre for Molecular Biodiscovery, Auckland, New Zealand
| | - Sharon R Ladyman
- Centre for Neuroendocrinology and Department of Anatomy, University of Otago, Dunedin, New Zealand
- Maurice Wilkins Centre for Molecular Biodiscovery, Auckland, New Zealand
| |
Collapse
|
16
|
Yoo S, Cha D, Kim DW, Hoang TV, Blackshaw S. Tanycyte-Independent Control of Hypothalamic Leptin Signaling. Front Neurosci 2019; 13:240. [PMID: 30941008 PMCID: PMC6433882 DOI: 10.3389/fnins.2019.00240] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2019] [Accepted: 03/01/2019] [Indexed: 12/29/2022] Open
Abstract
Leptin is secreted by adipocytes to regulate appetite and body weight. Recent studies have reported that tanycytes actively transport circulating leptin across the brain barrier into the hypothalamus, and are required for normal levels of hypothalamic leptin signaling. However, direct evidence for leptin receptor (LepR) expression is lacking, and the effect of tanycyte-specific deletion of LepR has not been investigated. In this study, we analyze the expression and function of the tanycytic LepR in mice. Using single-molecule fluorescent in situ hybridization (smfISH), RT-qPCR, single-cell RNA sequencing (scRNA-Seq), and selective deletion of the LepR in tanycytes, we are unable to detect expression of LepR in the tanycytes. Tanycyte-specific deletion of LepR likewise did not affect leptin-induced pSTAT3 expression in hypothalamic neurons, regardless of whether leptin was delivered by intraperitoneal or intracerebroventricular injection. Finally, we use activity-regulated scRNA-Seq (act-Seq) to comprehensively profile leptin-induced changes in gene expression in all cell types in mediobasal hypothalamus. Clear evidence for leptin signaling is only seen in endothelial cells and subsets of neurons, although virtually all cell types show leptin-induced changes in gene expression. We thus conclude that LepR expression in tanycytes is either absent or undetectably low, that tanycytes do not directly regulate hypothalamic leptin signaling through a LepR-dependent mechanism, and that leptin regulates gene expression in diverse hypothalamic cell types through both direct and indirect mechanisms.
Collapse
Affiliation(s)
- Sooyeon Yoo
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University, Baltimore, MD, United States
| | - David Cha
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University, Baltimore, MD, United States
| | - Dong Won Kim
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University, Baltimore, MD, United States
| | - Thanh V Hoang
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University, Baltimore, MD, United States
| | - Seth Blackshaw
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University, Baltimore, MD, United States.,Department of Ophthalmology, Johns Hopkins University, Baltimore, MD, United States.,Department of Neurology, Johns Hopkins University, Baltimore, MD, United States.,Center for Human Systems Biology, Johns Hopkins University, Baltimore, MD, United States.,School of Medicine, Institute for Cell Engineering, Johns Hopkins University, Baltimore, MD, United States
| |
Collapse
|
17
|
van den Pol AN, Acuna C, Davis JN, Huang H, Zhang X. Defining the caudal hypothalamic arcuate nucleus with a focus on anorexic excitatory neurons. J Physiol 2019; 597:1605-1625. [PMID: 30618146 PMCID: PMC6418765 DOI: 10.1113/jp277152] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2018] [Accepted: 01/03/2019] [Indexed: 01/11/2023] Open
Abstract
KEY POINTS Excitatory glutamate neurons are sparse in the rostral hypothalamic arcuate nucleus (ARC), the subregion that has received the most attention in the past. In striking contrast, excitatory neurons are far more common (by a factor of 10) in the caudal ARC, an area which has received relatively little attention. These glutamate cells may play a negative role in energy balance and food intake. They can show an increase in phosphorylated Stat-3 in the presence of leptin, are electrically excited by the anorectic neuromodulator cholecystokinin, and inhibited by orexigenic neuromodulators neuropeptide Y, met-enkephalin, dynorphin and the catecholamine dopamine. The neurons project local axonal connections that excite other ARC neurons including proopiomelanocortin neurons that can play an important role in obesity. These data are consistent with models suggesting that the ARC glutamatergic neurons may play both a rapid and a slower role in acting as anorectic neurons in CNS control of food intake and energy homeostasis. ABSTRACT Here we interrogate a unique class of excitatory neurons in the hypothalamic arcuate nucleus (ARC) that utilizes glutamate as a fast neurotransmitter using mice expressing GFP under control of the vesicular glutamate transporter 2 (vGluT2) promoter. These neurons show a unique distribution, synaptic characterization, cellular physiology and response to neuropeptides involved in energy homeostasis. Although apparently not previously appreciated, the caudal ARC showed a far greater density of vGluT2 cells than the rostral ARC, as seen in transgenic vGluT2-GFP mice and mRNA analysis. After food deprivation, leptin induced an increase in phosphorylated Stat-3 in vGluT2-positive neurons, indicating a response to hormonal cues of energy state. Based on whole-cell recording electrophysiology in brain slices, vGluT2 neurons were spontaneously active with a spike frequency around 2 Hz. vGluT2 cells were responsive to a number of neuropeptides related to energy homeostasis; they were excited by the anorectic peptide cholecystokinin, but inhibited by orexigenic neuropeptide Y, dynorphin and met-enkephalin, consistent with an anorexic role in energy homeostasis. Dopamine, associated with the hedonic aspect of enhancing food intake, inhibited vGluT2 neurons. Optogenetic excitation of vGluT2 cells evoked EPSCs in neighbouring neurons, indicating local synaptic excitation of other ARC neurons. Microdrop excitation of ARC glutamate cells in brain slices rapidly increased excitatory synaptic activity in anorexigenic proopiomelanocortin neurons. Together these data support the perspective that vGluT2 cells may be more prevalent in the ARC than previously appreciated, and play predominantly an anorectic role in energy metabolism.
Collapse
Affiliation(s)
| | - Claudio Acuna
- Department of NeurosurgeryYale University School of MedicineNew HavenCT06520USA
| | - John N. Davis
- Department of NeurosurgeryYale University School of MedicineNew HavenCT06520USA
| | - Hao Huang
- Department of NeurosurgeryYale University School of MedicineNew HavenCT06520USA
| | - Xiaobing Zhang
- Department of NeurosurgeryYale University School of MedicineNew HavenCT06520USA
| |
Collapse
|
18
|
McGregor G, Harvey J. Leptin Regulation of Synaptic Function at Hippocampal TA-CA1 and SC-CA1 Synapses: Implications for Health and Disease. Neurochem Res 2019; 44:650-660. [PMID: 28819795 PMCID: PMC6420429 DOI: 10.1007/s11064-017-2362-1] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2017] [Revised: 07/05/2017] [Accepted: 07/21/2017] [Indexed: 12/16/2022]
Abstract
Growing evidence indicates that the endocrine hormone leptin regulates hippocampal synaptic function in addition to its established role as a hypothalamic satiety signal. Indeed, numerous studies show that leptin facilitates the cellular events that underlie hippocampal learning and memory including activity-dependent synaptic plasticity and glutamate receptor trafficking, indicating that leptin may be a potential cognitive enhancer. Although there has been extensive investigation into the modulatory role of leptin at hippocampal Schaffer collateral (SC)-CA1 synapses, recent evidence indicates that leptin also potently regulates excitatory synaptic transmission at the anatomically distinct temporoammonic (TA) input to hippocampal CA1 neurons. The cellular mechanisms underlying activity-dependent synaptic plasticity at TA-CA1 synapses differ from those at SC-CA1 synapses and the TA input is implicated in spatial and episodic memory formation. Furthermore, the TA input is an early target for neurodegeneration in Alzheimer's disease (AD) and aberrant leptin function is linked to AD. Here, we review the evidence that leptin regulates hippocampal synaptic function at both SC- and TA-CA1 synapses and discuss the consequences for neurodegenerative disorders like AD.
Collapse
Affiliation(s)
- Gemma McGregor
- Division of Neuroscience, School of Medicine, Ninewells Hospital and Medical School, University of Dundee, Dundee, DD1 9SY, UK
| | - Jenni Harvey
- Division of Neuroscience, School of Medicine, Ninewells Hospital and Medical School, University of Dundee, Dundee, DD1 9SY, UK.
| |
Collapse
|
19
|
Lee YH, Hsu HC, Kao PC, Shiao YJ, Yeh SHH, Shie FS, Hsu SM, Yeh CW, Liu HK, Yang SB, Tsay HJ. Augmented Insulin and Leptin Resistance of High Fat Diet-Fed APPswe/PS1dE9 Transgenic Mice Exacerbate Obesity and Glycemic Dysregulation. Int J Mol Sci 2018; 19:2333. [PMID: 30096853 PMCID: PMC6121904 DOI: 10.3390/ijms19082333] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2018] [Revised: 08/02/2018] [Accepted: 08/03/2018] [Indexed: 11/16/2022] Open
Abstract
Alzheimer's disease (AD), a progressive neurodegenerative disease is highly associated with metabolic syndromes. We previously demonstrated that glycemic dysregulation and obesity are augmented in high fat diet (HFD)-treated APPswe/PS1dE9 (APP/PS1) transgenic mice. In the current study, the underlying mechanism mediating exacerbated metabolic stresses in HFD APP/PS1 transgenic mice was further examined. APP/PS1 mice developed insulin resistance and, consequently, impaired glucose homeostasis after 10 weeks on HFD. [18F]-2-fluoro-2-deoxy-d-glucose ([18F]-FDG) positron emission tomography showed that interscapular brown adipose tissue is vulnerable to HFD and AD-related pathology. Chronic HFD induced hyperphagia, with limited effects on basal metabolic rates in APP/PS1 transgenic mice. Excessive food intake may be caused by impairment of leptin signaling in the hypothalamus because leptin failed to suppress the food intake of HFD APP/PS1 transgenic mice. Leptin-induced pSTAT3 signaling in the arcuate nucleus was attenuated. Dysregulated energy homeostasis including hyperphagia and exacerbated obesity was elicited prior to the presence of the amyloid pathology in the hypothalamus of HFD APP/PS1 transgenic mice; nevertheless, cortical neuroinflammation and the level of serum Aβ and IL-6 were significantly elevated. Our study demonstrates the pivotal role of AD-related pathology in augmenting HFD-induced insulin and leptin resistance and impairing hypothalamic regulation of energy homeostasis.
Collapse
Affiliation(s)
- Yi-Heng Lee
- Institute of Neuroscience, School of Life Science, National Yang-Ming University, Taipei 112, Taiwan.
- Taiwan International Graduate Program in Interdisciplinary Neuroscience, National Yang-Ming University and Academia Sinica, Taipei 112, Taiwan.
| | - Hao-Chieh Hsu
- Institute of Anatomy and Cell Biology, School of Medicine, National Yang-Ming University, Taipei 112, Taiwan.
| | - Pei-Chen Kao
- Institute of Neuroscience, School of Life Science, National Yang-Ming University, Taipei 112, Taiwan.
| | - Young-Ji Shiao
- National Research Institute of Chinese Medicine, Ministry of Health and Welfare, Taipei 112, Taiwan.
- Institute of Biopharmaceutical Science, National Yang-Ming University, Taipei 112, Taiwan.
| | - Skye Hsin-Hsien Yeh
- Brain Research Center, National Yang-Ming University, Taipei 112, Taiwan.
- Aging and Health Research Center, National Yang-Ming University, Taipei 112, Taiwan.
| | - Feng-Shiun Shie
- Center for Neuropsychiatric Research, National Health Research Institutes, No. 35 Keyan Road, Zhunan Town, Miaoli County 350, Taiwan.
| | - Shu-Meng Hsu
- Institute of Neuroscience, School of Life Science, National Yang-Ming University, Taipei 112, Taiwan.
| | - Chih-Wen Yeh
- Institute of Neuroscience, School of Life Science, National Yang-Ming University, Taipei 112, Taiwan.
| | - Hui-Kang Liu
- National Research Institute of Chinese Medicine, Ministry of Health and Welfare, Taipei 112, Taiwan.
- Ph.D. Program for Clinical Drug Discovery from Botanical Herbs, Taipei Medical University, Taipei 110, Taiwan.
| | - Shi-Bing Yang
- Taiwan International Graduate Program in Interdisciplinary Neuroscience, National Yang-Ming University and Academia Sinica, Taipei 112, Taiwan.
- Institute of Biomedical Sciences, Academia Sinica, Taipei 115, Taiwan.
| | - Huey-Jen Tsay
- Institute of Neuroscience, School of Life Science, National Yang-Ming University, Taipei 112, Taiwan.
- Taiwan International Graduate Program in Interdisciplinary Neuroscience, National Yang-Ming University and Academia Sinica, Taipei 112, Taiwan.
| |
Collapse
|
20
|
Hypothalamic inflammation and malfunctioning glia in the pathophysiology of obesity and diabetes: Translational significance. Biochem Pharmacol 2018; 153:123-133. [PMID: 29337002 DOI: 10.1016/j.bcp.2018.01.024] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2017] [Accepted: 01/09/2018] [Indexed: 12/25/2022]
|
21
|
Seo M, Islam SA, Moon SS. Acute anti-obesity effects of intracerebroventricular 11β-HSD1 inhibitor administration in diet-induced obese mice. J Neuroendocrinol 2018; 30:e12580. [PMID: 29418022 DOI: 10.1111/jne.12580] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/20/2017] [Revised: 01/09/2018] [Accepted: 02/01/2018] [Indexed: 11/30/2022]
Abstract
The hypothalamus is the regulatory centre of both appetite and energy balance and endoplasmic reticulum (ER) stress in the hypothalamus is involved in the pathogenesis of obesity. Recently, inhibition of 11 β hydroxysteroid dehydrogenase type1 (11β-HSD1) was reported to have an anti-obesity effect by reducing fat mass. However, the link between the role of 11β-HSD1 in the hypothalamus and obesity has yet to be determined. In the present study, embryonal primary hypothalamic neurones and high-fat diet (HFD) fed mice were used to investigate the anorexigenic effects of 11β-HSD1 inhibitors both in vitro and in vivo. In hypothalamic neurones, carbenoxolone (a non selecitve 11β-HSD inhibitor) alleviated ER stress and ER stress-induced neuropeptide alterations. In HFD mice, i.c.v. administration of carbenoxolone or KR67500 (nonselective and selective 11β-HSD1 inhibitors, respectively) was associated with less weight gain compared to control mice for 24 hours after treatment, presumably by reducing food intake. Furthermore, glucose regulated protein (Grp78), spliced X-box binding protein (Xbp-1s), c/EBP homologous protein (chop) and ER DnaJ homologue protein (Erdj4) expression was decreased in the hypothalami of mice administrated 11β-HSD1 inhibitors compared to controls. Conversely, the phosphorylation of protein kinase B (PKB/Akt), signal transducer and activator of transcription 3 (Stat3), mitogen-activated protein kinase (MAPK/ERK) and S6 kinase1 (S6K1) in the hypothalamus was induced more in mice treated using the same regimes. In conclusion, acute 11β-HSD1 inhibition in the hypothalamus could reduce food intake by decreasing ER stress and increasing insulin, leptin, and mammalian target of rapamycin complex 1 (mTORC1) signalling.
Collapse
Affiliation(s)
- M Seo
- Medical Institute of Dongguk University, Gyeongju, South Korea
- Department of Agricultural Biology, National Institute of Agricultural Sciences, Rural Development Administration, Wanju-gun, South Korea
| | - S A Islam
- Department of Internal Medicine, College of Medicine, Dongguk University, Gyeongju, South Korea
| | - S-S Moon
- Medical Institute of Dongguk University, Gyeongju, South Korea
- Department of Internal Medicine, College of Medicine, Dongguk University, Gyeongju, South Korea
| |
Collapse
|
22
|
Yuan X, Caron A, Wu H, Gautron L. Leptin Receptor Expression in Mouse Intracranial Perivascular Cells. Front Neuroanat 2018; 12:4. [PMID: 29410615 PMCID: PMC5787097 DOI: 10.3389/fnana.2018.00004] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2017] [Accepted: 01/08/2018] [Indexed: 12/28/2022] Open
Abstract
Past studies have suggested that non-neuronal brain cells express the leptin receptor. However, the identity and distribution of these leptin receptor-expressing non-neuronal brain cells remain debated. This study assessed the distribution of the long form of the leptin receptor (LepRb) in non-neuronal brain cells using a reporter mouse model in which LepRb-expressing cells are permanently marked by tdTomato fluorescent protein (LepRb-CretdTomato). Double immunohistochemistry revealed that, in agreement with the literature, the vast majority of tdTomato-tagged cells across the mouse brain were neurons (i.e., based on immunoreactivity for NeuN). Non-neuronal structures also contained tdTomato-positive cells, including the choroid plexus and the perivascular space of the meninges and, to a lesser extent, the brain. Based on morphological criteria and immunohistochemistry, perivascular cells were deduced to be mainly pericytes. Notably, tdTomato-positive cells were immunoreactive for vitronectin and platelet derived growth factor receptor beta (PDGFBR). In situ hybridization studies confirmed that most tdTomato-tagged perivascular cells were enriched in leptin receptor mRNA (all isoforms). Using qPCR studies, we confirmed that the mouse meninges were enriched in Leprb and, to a greater extent, the short isoforms of the leptin receptor. Interestingly, qPCR studies further demonstrated significantly altered expression for Vtn and Pdgfrb in the meninges and hypothalamus of LepRb-deficient mice. Collectively, our data demonstrate that the only intracranial non-neuronal cells that express LepRb in the adult mouse are cells that form the blood-brain barrier, including, most notably, meningeal perivascular cells. Our data suggest that pericytic leptin signaling plays a role in the integrity of the intracranial perivascular space and, consequently, may provide a link between obesity and numerous brain diseases.
Collapse
Affiliation(s)
- Xuefeng Yuan
- Division of Hypothalamic Research and Department of Internal Medicine, The University of Texas Southwestern Medical Center, Dallas, TX, United States.,Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Alexandre Caron
- Division of Hypothalamic Research and Department of Internal Medicine, The University of Texas Southwestern Medical Center, Dallas, TX, United States
| | - Hua Wu
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Laurent Gautron
- Division of Hypothalamic Research and Department of Internal Medicine, The University of Texas Southwestern Medical Center, Dallas, TX, United States
| |
Collapse
|
23
|
Abstract
Our understanding of adipose tissue as an endocrine organ has been transformed over the last 20 years. During this time, a number of adipocyte-derived factors or adipokines have been identified. This article will review evidence for how adipokines acting via the central nervous system (CNS) regulate normal physiology and disease pathology. The reported CNS-mediated effects of adipokines are varied and include the regulation of energy homeostasis, autonomic nervous system activity, the reproductive axis, neurodevelopment, cardiovascular function, and cognition. Due to the wealth of information available and the diversity of their known functions, the archetypal adipokines leptin and adiponectin will be focused on extensively. Other adipokines with established CNS actions will also be discussed. Due to the difficulties associated with studying CNS function on a molecular level in humans, the majority of our knowledge, and as such the studies described in this paper, comes from work in experimental animal models; however, where possible the relevant data from human studies are also highlighted. © 2017 American Physiological Society. Compr Physiol 7:1359-1406, 2017.
Collapse
Affiliation(s)
- Craig Beall
- Biomedical Neuroscience Research Group, Institute of Biomedical and Clinical Sciences, University of Exeter Medical School, Exeter, Devon, United Kingdom
| | - Lydia Hanna
- Biomedical Neuroscience Research Group, Institute of Biomedical and Clinical Sciences, University of Exeter Medical School, Exeter, Devon, United Kingdom
| | - Kate L J Ellacott
- Biomedical Neuroscience Research Group, Institute of Biomedical and Clinical Sciences, University of Exeter Medical School, Exeter, Devon, United Kingdom
| |
Collapse
|
24
|
Ramos-Lobo AM, Donato J. The role of leptin in health and disease. Temperature (Austin) 2017; 4:258-291. [PMID: 28944270 DOI: 10.1080/23328940.2017.1327003] [Citation(s) in RCA: 88] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2016] [Revised: 04/27/2017] [Accepted: 05/02/2017] [Indexed: 02/07/2023] Open
Abstract
Leptin is a master regulator of energy balance and body adiposity. Additionally, leptin exerts important control on glucose homeostasis, thermogenesis, autonomic nervous system and neuroendocrine axes. In metabolic diseases, such as obesity and diabetes mellitus, leptin signaling may be compromised, indicating the important role of this hormone in the etiology and pathophysiological manifestations of these conditions. In the present manuscript, we reviewed important concepts of leptin signaling, as well as about the effects of leptin on several biologic functions. We also discussed the possible therapeutic use of leptin administration and how our current obesogenic environment contributes to the development of leptin resistance. Our objective was to provide a comprehensive and state-of-the-art review about the importance of leptin to maintain the homeostasis and during pathological conditions.
Collapse
Affiliation(s)
- Angela M Ramos-Lobo
- Department of Physiology and Biophysics, Institute of Biomedical Sciences, University of São Paulo, São Paulo, SP, Brazil
| | - Jose Donato
- Department of Physiology and Biophysics, Institute of Biomedical Sciences, University of São Paulo, São Paulo, SP, Brazil
| |
Collapse
|
25
|
Ji S, Tokizane K, Ohkawa Y, Ohmi Y, Banno R, Okajima T, Kiyama H, Furukawa K, Furukawa K. Increased a-series gangliosides positively regulate leptin/Ob receptor-mediated signals in hypothalamus of GD3 synthase-deficient mice. Biochem Biophys Res Commun 2016; 479:453-460. [DOI: 10.1016/j.bbrc.2016.09.077] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2016] [Accepted: 09/15/2016] [Indexed: 12/14/2022]
|
26
|
Senzacqua M, Severi I, Perugini J, Acciarini S, Cinti S, Giordano A. Action of Administered Ciliary Neurotrophic Factor on the Mouse Dorsal Vagal Complex. Front Neurosci 2016; 10:289. [PMID: 27445662 PMCID: PMC4921504 DOI: 10.3389/fnins.2016.00289] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2016] [Accepted: 06/10/2016] [Indexed: 12/13/2022] Open
Abstract
Ciliary neurotrophic factor (CNTF) induces weight loss in obese rodents and humans through activation of the hypothalamic Jak-STAT (Janus kinase-signal transducer and activator of transcription) signaling pathway. Here, we tested the hypothesis that CNTF also affects the brainstem centers involved in feeding and energy balance regulation. To this end, wild-type and leptin-deficient (ob/ob and db/db) obese mice were acutely treated with intraperitoneal recombinant CNTF. Coronal brainstem sections were processed for immunohistochemical detection of STAT3, STAT1, STAT5 phosphorylation and c-Fos. In wild-type mice, CNTF treatment for 45 min induced STAT3, STAT1, and STAT5 phosphorylation in neurons as well as glial cells of the area postrema; here, the majority of CNTF-responsive cells activated multiple STAT isoforms, and a significant proportion of CNTF-responsive glial cells bore the immaturity and plasticity markers nestin and vimentin. After 120 min CNTF treatment, c-Fos expression was intense in glial cells and weak in neurons of the area postrema, it was intense in several neurons of the rostral and caudal solitary tract nucleus (NTS), and weak in some cholinergic neurons of the dorsal motor nucleus of the vagus. In the ob/ob and db/db mice, Jak-STAT activation and c-Fos expression were similar to those induced in wild-type mouse brainstem. Treatment with CNTF (120 min, to induce c-Fos expression) and leptin (25 min, to induce STAT3 phosphorylation) demonstrated the co-localization of the two transcription factors in a small neuron population in the caudal NTS portion. Finally, weak immunohistochemical CNTF staining, detected in funiculus separans, and meningeal glial cells, matched the modest amount of CNTF found by RT-qPCR in micropunched area postrema tissue, which in contrast exhibited a very high amount of CNTF receptor. Collectively, the present findings show that the area postrema and the NTS exhibit high, distinctive responsiveness to circulating exogenous and, probably, endogenous CNTF.
Collapse
Affiliation(s)
- Martina Senzacqua
- Department of Experimental and Clinical Medicine, Università Politecnica delle MarcheAncona, Italy
| | - Ilenia Severi
- Department of Experimental and Clinical Medicine, Università Politecnica delle MarcheAncona, Italy
| | - Jessica Perugini
- Department of Experimental and Clinical Medicine, Università Politecnica delle MarcheAncona, Italy
| | - Samantha Acciarini
- Department of Experimental and Clinical Medicine, Università Politecnica delle MarcheAncona, Italy
| | - Saverio Cinti
- Department of Experimental and Clinical Medicine, Università Politecnica delle MarcheAncona, Italy
- Center of Obesity, Università Politecnica delle Marche-United HospitalsAncona, Italy
| | - Antonio Giordano
- Department of Experimental and Clinical Medicine, Università Politecnica delle MarcheAncona, Italy
| |
Collapse
|
27
|
Kwon O, Kim KW, Kim MS. Leptin signalling pathways in hypothalamic neurons. Cell Mol Life Sci 2016; 73:1457-77. [PMID: 26786898 PMCID: PMC11108307 DOI: 10.1007/s00018-016-2133-1] [Citation(s) in RCA: 188] [Impact Index Per Article: 20.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2015] [Revised: 12/20/2015] [Accepted: 01/07/2016] [Indexed: 12/25/2022]
Abstract
Leptin is the most critical hormone in the homeostatic regulation of energy balance among those so far discovered. Leptin primarily acts on the neurons of the mediobasal part of hypothalamus to regulate food intake, thermogenesis, and the blood glucose level. In the hypothalamic neurons, leptin binding to the long form leptin receptors on the plasma membrane initiates multiple signaling cascades. The signaling pathways known to mediate the actions of leptin include JAK-STAT signaling, PI3K-Akt-FoxO1 signaling, SHP2-ERK signaling, AMPK signaling, and mTOR-S6K signaling. Recent evidence suggests that leptin signaling in hypothalamic neurons is also linked to primary cilia function. On the other hand, signaling molecules/pathways mitigating leptin actions in hypothalamic neurons have been extensively investigated in an effort to treat leptin resistance observed in obesity. These include SOCS3, tyrosine phosphatase PTP1B, and inflammatory signaling pathways such as IKK-NFκB and JNK signaling, and ER stress-mitochondrial signaling. In this review, we discuss leptin signaling pathways in the hypothalamus, with a particular focus on the most recently discovered pathways.
Collapse
Affiliation(s)
- Obin Kwon
- Division of Endocrinology and Metabolism, Asan Medical Center, University of Ulsan College of Medicine, 88 Olympic-ro 43-gil, Songpa-gu, Seoul, 05505, Korea
- Appetite Regulation Laboratory, Asan Institute for Life Science, Seoul, 05505, Korea
| | - Ki Woo Kim
- Department of Pharmacology, Yonsei University Wonju College of Medicine, Wonju, 26426, Korea
- Department of Global Medical Science, Yonsei University Wonju College of Medicine, Wonju, 26426, Korea
| | - Min-Seon Kim
- Division of Endocrinology and Metabolism, Asan Medical Center, University of Ulsan College of Medicine, 88 Olympic-ro 43-gil, Songpa-gu, Seoul, 05505, Korea.
- Appetite Regulation Laboratory, Asan Institute for Life Science, Seoul, 05505, Korea.
| |
Collapse
|
28
|
Woo SM, Choi YK, Kim AJ, Yun YJ, Shin YC, Cho SG, Ko SG. Sip-jeon-dea-bo-tang, a traditional herbal medicine, ameliorates cisplatin-induced anorexia via the activation of JAK1/STAT3-mediated leptin and IL-6 production in the fat tissue of mice. Mol Med Rep 2016; 13:2967-72. [PMID: 26936678 PMCID: PMC4805058 DOI: 10.3892/mmr.2016.4889] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2015] [Accepted: 01/04/2016] [Indexed: 02/02/2023] Open
Abstract
Despite its therapeutic advantages, chemotherapy can also cause adverse effects, including anorexia and loss of appetite. Although numerous patients with cancer have been reported to suffer from anorexia during or following chemotherapy, treatment options for anorexia remain to be determined. In Asian countries, traditional medicines are widely used to treat problems with appetite; sip-jeon-dea-bo-tang (SJDBT) is one of those medicines used for the treatment of anorexia. The present study demonstrated that SJDBT ameliorated cisplatin-induced anorexia. In a mouse model of chemotherapy-induced anorexia, oral administration of SJDBT prevented the cisplatin-induced reduction of food intake, inhibiting weight loss. The results of multiplex assays showed that SJDBT only altered the levels of interleukin (IL)-6 and leptin in the serum and fat tissue. In addition, SJDBT maintained the serum leptin level and increased the serum IL-6 level, whereas cisplatin reduced the levels of both serum leptin and IL-6. Furthermore, SJDBT was revealed to increase the levels of leptin and IL-6 in the fat tissue by activating the JAK1/STAT3 signaling pathway. In conclusion, the present results revealed that SJDBT ameliorated cisplatin-induced anorexia, suggesting its usefulness in the prevention of anorexia during chemotherapy.
Collapse
Affiliation(s)
- Sang-Mi Woo
- Department of Preventive Medicine, College of Republic of Korean Medicine, Kyung Hee University, Seoul 130‑701, Republic of Korea
| | - Youn Kyung Choi
- Department of Preventive Medicine, College of Republic of Korean Medicine, Kyung Hee University, Seoul 130‑701, Republic of Korea
| | - Ah-Jeong Kim
- Department of Preventive Medicine, College of Republic of Korean Medicine, Kyung Hee University, Seoul 130‑701, Republic of Korea
| | - Yee Jin Yun
- Department of Preventive Medicine, College of Republic of Korean Medicine, Kyung Hee University, Seoul 130‑701, Republic of Korea
| | - Yong Cheol Shin
- Department of Preventive Medicine, College of Republic of Korean Medicine, Kyung Hee University, Seoul 130‑701, Republic of Korea
| | - Sung-Gook Cho
- Department of Preventive Medicine, College of Republic of Korean Medicine, Kyung Hee University, Seoul 130‑701, Republic of Korea
| | - Seong Gyu Ko
- Department of Preventive Medicine, College of Republic of Korean Medicine, Kyung Hee University, Seoul 130‑701, Republic of Korea
| |
Collapse
|
29
|
Severi I, Senzacqua M, Mondini E, Fazioli F, Cinti S, Giordano A. Activation of transcription factors STAT1 and STAT5 in the mouse median eminence after systemic ciliary neurotrophic factor administration. Brain Res 2015; 1622:217-229. [PMID: 26133794 DOI: 10.1016/j.brainres.2015.06.028] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2015] [Revised: 05/29/2015] [Accepted: 06/21/2015] [Indexed: 12/26/2022]
Abstract
Exogenously administered ciliary neurotrophic factor (CNTF) causes weight loss in obese rodents and humans through leptin-like activation of the Jak-STAT3 signaling pathway in hypothalamic arcuate neurons. Here we report for the first time that 40min after acute systemic treatment, rat recombinant CNTF (intraperitoneal injection of 0.3mg/kg of body weight) induced nuclear translocation of the tyrosine-phosphorylated forms of STAT1 and STAT5 in the mouse median eminence and other circumventricular organs, including the vascular organ of the lamina terminalis and the subfornical organ. In the tuberal hypothalamus of treated mice, specific nuclear immunostaining for phospo-STAT1 and phospho-STAT5 was detected in ependymal cells bordering the third ventricle floor and lateral recesses, and in median eminence cells. Co-localization studies documented STAT1 and STAT5 activation in median eminence β-tanycytes and underlying radial glia-like cells. A few astrocytes in the arcuate nucleus responded to CNTF by STAT5 activation. The vast majority of median eminence tanycytes and radial glia-like cells showing phospho-STAT1 and phospho-STAT5 immunoreactivity were also positive for phospho-STAT3. In contrast, STAT3 was the sole STAT isoform activated by CNTF in arcuate nucleus and median eminence neurons. Finally, immunohistochemical evaluation of STAT activation 20, 40, 80, and 120min from the injection demonstrated that cell activation was accompanied by c-Fos expression. Collectively, our findings show that CNTF activates STAT3, STAT1, and STAT5 in vivo. The distinctive activation pattern of these STAT isoforms in the median eminence may disclose novel targets and pathways through which CNTF regulates food intake.
Collapse
Affiliation(s)
- Ilenia Severi
- Department of Experimental and Clinical Medicine, Università Politecnica delle Marche, Ancona, Italy
| | - Martina Senzacqua
- Department of Experimental and Clinical Medicine, Università Politecnica delle Marche, Ancona, Italy
| | - Eleonora Mondini
- Department of Experimental and Clinical Medicine, Università Politecnica delle Marche, Ancona, Italy
| | - Francesca Fazioli
- Department of Clinical and Molecular Sciences, Università Politecnica delle Marche, Ancona, Italy
| | - Saverio Cinti
- Department of Experimental and Clinical Medicine, Università Politecnica delle Marche, Ancona, Italy; Center of Obesity, Università Politecnica delle Marche-United Hospitals, Ancona, Italy
| | - Antonio Giordano
- Department of Experimental and Clinical Medicine, Università Politecnica delle Marche, Ancona, Italy.
| |
Collapse
|
30
|
Cheng L, Yu Y, Szabo A, Wu Y, Wang H, Camer D, Huang XF. Palmitic acid induces central leptin resistance and impairs hepatic glucose and lipid metabolism in male mice. J Nutr Biochem 2015; 26:541-8. [PMID: 25724108 DOI: 10.1016/j.jnutbio.2014.12.011] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2014] [Revised: 11/19/2014] [Accepted: 12/08/2014] [Indexed: 12/12/2022]
Abstract
The consumption of diets rich in saturated fat largely contributes to the development of obesity in modern societies. A diet high in saturated fats can induce inflammation and impair leptin signaling in the hypothalamus. However, the role of saturated fatty acids on hypothalamic leptin signaling, and hepatic glucose and lipid metabolism remains largely undiscovered. In this study, we investigated the effects of intracerebroventricular (icv) administration of a saturated fatty acid, palmitic acid (PA, C16:0), on central leptin sensitivity, hypothalamic leptin signaling, inflammatory molecules and hepatic energy metabolism in C57BL/6J male mice. We found that the icv administration of PA led to central leptin resistance, evidenced by the inhibition of central leptin's suppression of food intake. Central leptin resistance was concomitant with impaired hypothalamic leptin signaling (JAK2-STAT3, PKB/Akt-FOXO1) and a pro-inflammatory response (TNF-α, IL1-β, IL-6 and pIκBa) in the mediobasal hypothalamus and paraventricular hypothalamic nuclei. Furthermore, the pre-administration of icv PA blunted the effect of leptin-induced decreases in mRNA expression related to gluconeogenesis (G6Pase and PEPCK), glucose transportation (GLUT2) and lipogenesis (FAS and SCD1) in the liver of mice. Therefore, elevated central PA concentrations can induce pro-inflammatory responses and leptin resistance, which are associated with disorders of energy homeostasis in the liver as a result of diet-induced obesity.
Collapse
Affiliation(s)
- Licai Cheng
- School of Medicine, University of Wollongong and Illawarra Health and Medical Research Institute, NSW 2522, Australia
| | - Yinghua Yu
- School of Medicine, University of Wollongong and Illawarra Health and Medical Research Institute, NSW 2522, Australia; Schizophrenia Research Institute (SRI), 405 Liverpool St, Sydney, NSW 2010, Australia
| | - Alexander Szabo
- School of Medicine, University of Wollongong and Illawarra Health and Medical Research Institute, NSW 2522, Australia; ANSTO Life Sciences, Australian Nuclear Science and Technology Organisation (ANSTO), Lucas Heights, NSW 2234, Australia
| | - Yizhen Wu
- School of Medicine, University of Wollongong and Illawarra Health and Medical Research Institute, NSW 2522, Australia
| | - Hongqin Wang
- School of Medicine, University of Wollongong and Illawarra Health and Medical Research Institute, NSW 2522, Australia
| | - Danielle Camer
- School of Medicine, University of Wollongong and Illawarra Health and Medical Research Institute, NSW 2522, Australia
| | - Xu-Feng Huang
- School of Medicine, University of Wollongong and Illawarra Health and Medical Research Institute, NSW 2522, Australia; Schizophrenia Research Institute (SRI), 405 Liverpool St, Sydney, NSW 2010, Australia.
| |
Collapse
|
31
|
Zhang L, Song H, Ge Y, Ji G, Yao Z. Temporal relationship between diet-induced steatosis and onset of insulin/leptin resistance in male Wistar rats. PLoS One 2015; 10:e0117008. [PMID: 25658428 PMCID: PMC4319780 DOI: 10.1371/journal.pone.0117008] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2014] [Accepted: 12/17/2014] [Indexed: 02/06/2023] Open
Abstract
Rats fed with high-fat-high-sucrose (HFHS) diet are known to manifest metabolic syndrome including hyperinsulinemia, hyperleptinemia, hyperglycemia, diabetic dyslipidemia, and hepatic steatosis. The aim of the current study is to determine the temporal relationships between the development of hepatic steatosis and the onset of insulin and leptin resistance in hypothalamus and liver in male Wistar rats (six weeks of age) fed chow or HFHS diet for up to 8 weeks. Fasting plasma glucose, lipids/lipoproteins, insulin and leptin levels were quantified, histopathologic score of hepatic steatosis and inflammation were assessed, and the responses of common checkpoints of insulin and leptin signalling responsible for lipogenesis and gluconeogenesis were analyzed. In addition, acute insulin or leptin administration was performed at different stages of HFHS dieting to determine the responsiveness of the respective signalling pathways. Hyperinsulinemia, hyperglycemia, dyslipidemia, and increased homeostasis model assessment of basal insulin resistance occurred 1-week after HFHS dieting, coinciding with upregulation of suppressor of cytokine signalling 3 in both hypothalamus and liver. However, hepatosteatosis, accompanied with increased expression of sterol regulatory element binding protein 1c and phosphoenolpyruvate carboxykinase, did not manifest until 4- to 8-week after HFHS dieting. Lowered insulin sensitivity (shown by decreased insulin receptor substrate 1 and protein kinase B phosphorylation) occurred approximately 2 weeks prior to leptin resistance (shown by impaired signal transducer and activator of transcription 3 activation) in both the liver and hypothalamus. Acute insulin/leptin administration also demonstrated the impaired insulin or leptin signalling transduction. These data suggest that lowered insulin sensitivity and leptin resistance occurred at least 2-3 weeks earlier than the manifestation of hepatosteatosis in rats fed HFHS diet.
Collapse
Affiliation(s)
- Li Zhang
- Institute of Digestive Disease, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200032, China
| | - Haiyan Song
- Institute of Digestive Disease, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200032, China
| | - Yingli Ge
- Institute of Digestive Disease, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200032, China
| | - Guang Ji
- Institute of Digestive Disease, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200032, China
- E-institute of Shanghai Municipal Education Commission, Shanghai University of Traditional Chinese Medicine, Shanghai 200032, China
| | - Zemin Yao
- Department of Biochemistry, Microbiology & Immunology, Ottawa Institute of Systems Biology, University of Ottawa, Ottawa, Canada
| |
Collapse
|
32
|
Aguilar-Valles A, Inoue W, Rummel C, Luheshi GN. Obesity, adipokines and neuroinflammation. Neuropharmacology 2015; 96:124-34. [PMID: 25582291 DOI: 10.1016/j.neuropharm.2014.12.023] [Citation(s) in RCA: 120] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2014] [Revised: 12/16/2014] [Accepted: 12/19/2014] [Indexed: 12/14/2022]
Abstract
Global levels of obesity are reaching epidemic proportions, leading to a dramatic increase in incidence of secondary diseases and the significant economic burden associated with their treatment. These comorbidities include diabetes, cardiovascular disease, and some psychopathologies, which have been linked to a low-grade inflammatory state. Obese individuals exhibit an increase in circulating inflammatory mediators implicated as the underlying cause of these comorbidities. A number of these molecules are also manufactured and released by white adipose tissue (WAT), in direct proportion to tissue mass and are collectively known as adipokines. In the current review we focused on the role of two of the better-studied members of this family namely, leptin and adiponectin, with particular emphasis on their role in neuro-immune interactions, neuroinflammation and subsequent brain diseases. This article is part of a Special Issue entitled 'Neuroimmunology and Synaptic Function'.
Collapse
Affiliation(s)
- Argel Aguilar-Valles
- Department of Neuroscience, Université de Montréal and Goodman Cancer Centre, Department of Biochemistry, McGill University, Montréal, Canada
| | - Wataru Inoue
- Robarts Research Institute, Department of Physiology and Pharmacology, Schulich School of Medicine and Dentistry, University of Western Ontario, London, Ontario N6A 5B7, Canada
| | - Christoph Rummel
- Department of Veterinary-Physiology and -Biochemistry, Justus-Liebig-University Giessen, Frankfurter Strasse 100, D-35392 Giessen, Germany
| | - Giamal N Luheshi
- Douglas Mental Health University Institute, Department of Psychiatry, McGill University, Montreal, Quebec H4H 1R3, Canada.
| |
Collapse
|
33
|
Qiu J, Kleineidam A, Gouraud S, Yao ST, Greenwood M, Hoe SZ, Hindmarch C, Murphy D. The use of protein-DNA, chromatin immunoprecipitation, and transcriptome arrays to describe transcriptional circuits in the dehydrated male rat hypothalamus. Endocrinology 2014; 155:4380-90. [PMID: 25144923 PMCID: PMC4256826 DOI: 10.1210/en.2014-1448] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
The supraoptic nucleus (SON) of the hypothalamus is responsible for maintaining osmotic stability in mammals through its elaboration of the antidiuretic hormone arginine vasopressin. Upon dehydration, the SON undergoes a function-related plasticity, which includes remodeling of morphology, electrical properties, and biosynthetic activity. This process occurs alongside alterations in steady state transcript levels, which might be mediated by changes in the activity of transcription factors. In order to identify which transcription factors might be involved in changing patterns of gene expression, an Affymetrix protein-DNA array analysis was carried out. Nuclear extracts of SON from dehydrated and control male rats were analyzed for binding to the 345 consensus DNA transcription factor binding sequences of the array. Statistical analysis revealed significant changes in binding to 26 consensus elements, of which EMSA confirmed increased binding to signal transducer and activator of transcription (Stat) 1/Stat3, cellular Myelocytomatosis virus-like cellular proto-oncogene (c-Myc)-Myc-associated factor X (Max), and pre-B cell leukemia transcription factor 1 sequences after dehydration. Focusing on c-Myc and Max, we used quantitative PCR to confirm previous transcriptomic analysis that had suggested an increase in c-Myc, but not Max, mRNA levels in the SON after dehydration, and we demonstrated c-Myc- and Max-like immunoreactivities in SON arginine vasopressin-expressing cells. Finally, by comparing new data obtained from Roche-NimbleGen chromatin immunoprecipitation arrays with previously published transcriptomic data, we have identified putative c-Myc target genes whose expression changes in the SON after dehydration. These include known c-Myc targets, such as the Slc7a5 gene, which encodes the L-type amino acid transporter 1, ribosomal protein L24, histone deactylase 2, and the Rat sarcoma proto-oncogene (Ras)-related nuclear GTPase.
Collapse
Affiliation(s)
- Jing Qiu
- School of Clinical Sciences (J.Q., A.K., S.G., S.T.Y., M.G., C.H., D.M.), University of Bristol, Bristol BS1 3NY, United Kingdom; and Department of Physiology (S.Z.H., C.H., D.M.), Faculty of Medicine, University of Malaya, Kuala Lumpur 50603, Malaysia
| | | | | | | | | | | | | | | |
Collapse
|
34
|
Damm J, Wiegand F, Harden LM, Wenisch S, Gerstberger R, Rummel C, Roth J. Intraperitoneal and subcutaneous injections of the TLR9 agonist ODN 1668 in rats: brain inflammatory responses are related to peripheral IL-6 rather than interferons. J Neuroimmunol 2014; 277:105-17. [PMID: 25465287 DOI: 10.1016/j.jneuroim.2014.10.007] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2014] [Revised: 10/20/2014] [Accepted: 10/21/2014] [Indexed: 12/30/2022]
Abstract
Subcutaneous or intraperitoneal administration of Toll-like receptor (TLR)-9 agonist, ODN 1668 caused moderate fever and anorexia. In comparison to stimulation of other intracellular TLRs, activation of TLR9 did not result in pronounced peripheral induction of interferons, but rather induced interleukin-6. Expression of cytokines (TNFα, IL-1β) and inducible forms of enzymes for prostaglandin E2 synthesis occurred in the brain, in conjunction with a moderate activation of the transcription factors STAT3 and NF-IL6 in brain endothelial cells. The lack of a septic-like state in ODN 1668-treated rats reinforces the therapeutic value of this drug.
Collapse
Affiliation(s)
- J Damm
- Institute of Veterinary-Physiology and -Biochemistry, Justus-Liebig-University of Giessen, Frankfurter Strasse 100, D-35392 Giessen, Germany
| | - F Wiegand
- Institute of Veterinary-Physiology and -Biochemistry, Justus-Liebig-University of Giessen, Frankfurter Strasse 100, D-35392 Giessen, Germany
| | - L M Harden
- Institute of Veterinary-Physiology and -Biochemistry, Justus-Liebig-University of Giessen, Frankfurter Strasse 100, D-35392 Giessen, Germany; Brain Function Research Group, School of Physiology, Faculty of Health Sciences, University of the Witwatersrand, 7 York Road, Parktown, Johannesburg, South Africa
| | - S Wenisch
- Institute of Veterinary-Anatomy, Justus-Liebig-University of Giessen, Frankfurter Strasse 98, D-35392 Giessen, Germany
| | - R Gerstberger
- Institute of Veterinary-Physiology and -Biochemistry, Justus-Liebig-University of Giessen, Frankfurter Strasse 100, D-35392 Giessen, Germany
| | - C Rummel
- Institute of Veterinary-Physiology and -Biochemistry, Justus-Liebig-University of Giessen, Frankfurter Strasse 100, D-35392 Giessen, Germany
| | - J Roth
- Institute of Veterinary-Physiology and -Biochemistry, Justus-Liebig-University of Giessen, Frankfurter Strasse 100, D-35392 Giessen, Germany.
| |
Collapse
|
35
|
Sun B, Song L, Tamashiro KLK, Moran TH, Yan J. Large litter rearing improves leptin sensitivity and hypothalamic appetite markers in offspring of rat dams fed high-fat diet during pregnancy and lactation. Endocrinology 2014; 155:3421-33. [PMID: 24926823 PMCID: PMC5393320 DOI: 10.1210/en.2014-1051] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/21/2014] [Accepted: 06/06/2014] [Indexed: 01/09/2023]
Abstract
Maternal high-fat (HF) diet has long-term consequences on the offspring's metabolic phenotype. Here, we determined the effects of large litter (LL) rearing in offspring of rat dams fed HF diet during gestation and lactation. Pregnant Sprague-Dawley rats were maintained on standard chow (CHOW) or HF diet throughout gestation and lactation. Pups were raised in normal litters (NLs) (10 pups/dam) or LLs (16 pups/dam) during lactation, resulting in 4 groups: CHOW-NL, CHOW-LL, HF-NL, and HF-LL. The offspring were weaned onto to either CHOW or HF diet on postnatal day 21. Male and female pups with maternal HF diet (HF-NL) had greater body weight and adiposity, higher plasma leptin levels, impaired glucose tolerance, abnormal hypothalamic leptin signaling pathways (lower leptin receptor-b [OB-Rb] and signal transducer and activator of transcription 3, higher suppressor of cytokine signaling 3 mRNA expression) and appetite markers (lower neuropeptide Y and Agouti-related peptide mRNA expression), and reduced phospho-signal transducer and activator of transcription 3 level in response to leptin in the arcuate nucleus at weaning, whereas LL rearing normalized these differences. When weaned onto CHOW diet, adult male offspring from HF diet-fed dams continued to have greater adiposity, higher leptin levels, and lower hypothalamic OB-Rb, and LL rearing improved them. When weaned onto HF diet, both adult male and female offspring with maternal HF diet had greater body weight and adiposity, higher leptin levels, impaired glucose tolerance, lower OB-Rb, and higher suppressor of cytokine signaling 3 in hypothalamus compared with those of CHOW dams, whereas LL rearing improved most of them except male OB-Rb expression. Our data suggest that LL rearing improves hypothalamic leptin signaling pathways and appetite markers in an age- and sex-specific manner in this model.
Collapse
Affiliation(s)
- Bo Sun
- Department of Physiology and Pathophysiology (B.S., L.S., J.Y.), Xi'an Jiaotong University School of Medicine, Xi'an, Shaanxi 710061, People's Republic of China; and Department of Psychiatry and Behavioral Sciences (K.L.K.T., T.H.M.), Johns Hopkins University School of Medicine, Baltimore, Maryland 21205
| | | | | | | | | |
Collapse
|
36
|
The neuroanatomical function of leptin in the hypothalamus. J Chem Neuroanat 2014; 61-62:207-20. [PMID: 25007719 DOI: 10.1016/j.jchemneu.2014.05.004] [Citation(s) in RCA: 59] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2013] [Revised: 05/09/2014] [Accepted: 05/28/2014] [Indexed: 02/07/2023]
Abstract
The anorexigenic hormone leptin plays an important role in the control of food intake and feeding-related behavior, for an important part through its action in the hypothalamus. The adipose-derived hormone modulates a complex network of several intercommunicating orexigenic and anorexigenic neuropeptides in the hypothalamus to reduce food intake and increase energy expenditure. In this review we present an updated overview of the functional role of leptin in respect to feeding and feeding-related behavior per distinct hypothalamic nuclei. In addition to the arcuate nucleus, which is a major leptin sensitive hub, leptin-responsive neurons in other hypothalamic nuclei, including the, dorsomedial-, ventromedial- and paraventricular nucleus and the lateral hypothalamic area, are direct targets of leptin. However, leptin also modulates hypothalamic neurons in an indirect manner, such as via the melanocortin system. The dissection of the complexity of leptin's action on the networks involved in energy balance is subject of recent and future studies. A full understanding of the role of hypothalamic leptin in the regulation of energy balance requires cell-specific manipulation using of conditional deletion and expression of leptin receptors. In addition, optogenetic and pharmacogenetic tools in combination with other pharmacological (such as the recent discovery of a leptin receptor antagonist) and neuronal tracing techniques to map the circuit, will be helpful to understand the role of leptin receptor expressing neurons. Better understanding of these circuits and the involvement of leptin could provide potential sites for therapeutic interventions in obesity and metabolic diseases characterized by dysregulation of energy balance.
Collapse
|
37
|
Ladyman SR, Grattan DR. JAK-STAT and feeding. JAKSTAT 2014; 2:e23675. [PMID: 24058809 PMCID: PMC3710322 DOI: 10.4161/jkst.23675] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2012] [Revised: 01/17/2013] [Accepted: 01/18/2013] [Indexed: 12/26/2022] Open
Abstract
The regulation of energy balance requires a complex system to homeostatically maintain the adult body at a precise set point. The central nervous system, particularly the hypothalamus, plays a key role in integrating a variety of signals that can relay information about the body's energy stores. As part of this system, numerous cytokines and hormones contribute to the regulation of food intake and energy homeostasis. Cytokines, and some hormones, are known to act through JAK-STAT intracellular signaling pathways. The hormone leptin, which plays a vital role in appetite regulation, signals through the JAK-STAT pathway, and it is through this involvement that the JAK-STAT pathway has become an established component in the mechanisms regulating food intake within the body. Emerging research, however, is now showing that this involvement of JAK-STAT is not limited to its activation by leptin. Furthermore, while the JAK-STAT pathway may simply act to transmit the anorectic signal of circulating factors, this intracellular signaling pathway may also become impaired when normal regulation of energy balance is disrupted. Thus, altered JAK-STAT signaling may contribute to the breakdown of the normal homeostatic mechanisms maintaining body weight in obesity.
Collapse
Affiliation(s)
- Sharon R Ladyman
- Centre for Neuroendocrinology and Department of Anatomy; School of Medical Sciences; University of Otago; Dunedin, New Zealand
| | | |
Collapse
|
38
|
Maniscalco JW, Rinaman L. Systemic leptin dose-dependently increases STAT3 phosphorylation within hypothalamic and hindbrain nuclei. Am J Physiol Regul Integr Comp Physiol 2014; 306:R576-85. [PMID: 24523344 DOI: 10.1152/ajpregu.00017.2014] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Leptin released peripherally acts within the central nervous system (CNS) to modulate numerous physiological and behavioral functions. Histochemical identification of leptin-responsive CNS cells can reveal the specific cellular phenotypes and neural circuits through which leptin signaling modulates these functions. Leptin signaling elicits phosphorylation of signal transducer and activator of transcription 3 (pSTAT3), making pSTAT3-immunoreactivity (ir) a useful proxy for identifying leptin-responsive cells. Relatively low systemic doses of leptin (i.e., 10-130 μg/kg body wt) are sufficient to decrease food intake, inhibit gastric emptying, and increase sympathetic activity, but there are no histological reports of central pSTAT3-ir following leptin doses within this range. Considering this, we quantified central pSTAT3-ir in rats after intraperitoneal injections of leptin at doses ranging from 50 to 800 μg/kg body wt. Tissue sections were processed to identify pSTAT3-ir alone or in combination with immunolabeling for cocaine- and amphetamine-regulated transcript (CART), glucagon-like peptide-1 (GLP-1), prolactin-releasing peptide (PrRP), or dopamine-β-hydroxylase (DβH). Leptin doses as low as 50, 100, and 200 μg/kg body wt significantly increased the number of pSTAT3-ir cells in the arcuate nucleus of the hypothalamus (ARC), nucleus of the solitary tract (NTS), and ventromedial nucleus of the hypothalamus, respectively, and also led to robust pSTAT3 labeling in neural processes. The differential dose-dependent increases in pSTAT3-ir across brain regions provide new information regarding central leptin sensitivity. Within the ARC, CART-ir and pSTAT3-ir were often colocalized, consistent with evidence of leptin sensitivity in this neural population. Conversely, within the NTS, pSTAT3 only rarely colocalized with PrRP and/or DβH, and never with GLP-1.
Collapse
Affiliation(s)
- James W Maniscalco
- Department of Neuroscience, University of Pittsburgh, Pittsburgh, Pennsylvania
| | | |
Collapse
|
39
|
Koch CE, Lowe C, Pretz D, Steger J, Williams LM, Tups A. High-fat diet induces leptin resistance in leptin-deficient mice. J Neuroendocrinol 2014; 26:58-67. [PMID: 24382295 DOI: 10.1111/jne.12131] [Citation(s) in RCA: 70] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/18/2013] [Revised: 11/27/2013] [Accepted: 12/23/2013] [Indexed: 12/22/2022]
Abstract
The occurrence of type II diabetes is highly correlated with obesity, although the mechanisms linking the two conditions are incompletely understood. Leptin is a potent insulin sensitiser and, in leptin-deficient, insulin insensitive, Lep(ob/ob) mice, leptin improves glucose tolerance, indicating that leptin resistance may link obesity to insulin insensitivity. Leptin resistance occurs in response to a high-fat diet (HFD) and both hyperleptinaemia and inflammation have been proposed as causative mechanisms. Scrutinising the role of hyperleptinaemia in this process, central hyperleptinaemia in Lep(ob/ob) mice was induced by chronic i.c.v. infusion of leptin (4.2 μg/day) over 10 days. This treatment led to a dramatic decline in body weight and food intake, as well as an improvement in glucose tolerance. Transfer to HFD for 4 days markedly arrested the beneficial effects of leptin on these parameters. Because Lep(ob/ob) mice are exquisitely sensitive to leptin, the possibility that leptin could reverse HFD-induced glucose intolerance in these animals was investigated. HFD led to increased body weight and glucose intolerance compared to a low-fat diet (LFD). Older and heavier Lep(ob/ob) mice were used as body weight-matched controls. Mice in each group received either i.p. leptin (1.25 mg/kg) or vehicle, and glucose tolerance, food intake and the number of phosphorylated signal transducer and activator of transcription (pSTAT)3 immunoreactive cells in the arcuate nucleus (ARC) and ventromedial hypothalamus (VMH) were analysed. Leptin improved glucose tolerance (P = 0. 019) and reduced food intake in Lep(ob/ob) mice on LFD (P ≤ 0.001) but was ineffective in mice on HFD. Furthermore, when leptin was administered centrally, the glucose tolerance of Lep(ob/ob) mice on HFD was significantly impaired (P = 0.007). Although leptin induced the number of pSTAT3 immunoreactive cells in the ARC and VMH of Lep(ob/ob) mice on LFD, HFD was associated with elevated pSTAT3 immunoreactivity in vehicle-treated Lep(ob/ob) mice that was unaffected by leptin treatment, suggesting central leptin resistance. Negating central inflammation by co-administering a c-Jun n-terminal kinase (JNK) inhibitor reinstated the glucose-lowering effects of leptin. These findings demonstrate that Lep(ob/ob) mice develop leptin resistance on a HFD independent of hyperleptinaemia and also indicate that the JNK inflammatory pathway plays a key role in the induction of diet-induced glucose intolerance.
Collapse
Affiliation(s)
- C E Koch
- Department of Animal Physiology, Faculty of Biology, Philipps University Marburg, Marburg, Germany
| | | | | | | | | | | |
Collapse
|
40
|
de Carvalho Borges B, Rorato R, Uchoa ET, Marangon P, da Silva GSF, de Paula FJ, Branco LGS, Antunes-Rodrigues J, Elias LLK. High-fat diet induces site-specific unresponsiveness to LPS-stimulated STAT3 activation in the hypothalamus. Am J Physiol Regul Integr Comp Physiol 2014; 306:R34-44. [DOI: 10.1152/ajpregu.00147.2013] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Hypophagia induced by inflammation is associated with Janus kinase (JAK)-2/signal transducer and activator of transcription (STAT) 3 signaling pathway, and leptin-mediated hypophagia is also mediated by JAK2-STAT3 pathway. We have previously reported that lipopolysaccharide (LPS) did not reduce food intake in leptin-resistant high-fat diet (HFD) rats but maintained body weight loss. We investigated whether changes in p-STAT3 expression in the hypothalamus and brain stem could account for the desensitization of hypophagia in HFD animals after a low LPS dose (100 μg/kg). Wistar rats fed standard diet (3.95 kcal/g) or HFD (6.3 kcal/g) for 8 wk were assigned into control diet-saline, control diet-LPS, HFD-saline, and HFD-LPS groups. LPS reduced feeding in the control diet but not HFD. This group showed no p-STAT3 expression in the paraventricular nucleus (PVN) and ventromedial hypothalamic nucleus (VMH), but sustained, though lower than control, p-STAT3 in the nucleus of the solitary tract (NTS) and raphe pallidus (RPa). LPS decreased body weight in HFD rats and increased Fos expression in the NTS. LPS increased body temperature, oxygen consumption, and energy expenditure in both control diet and HFD rats, and this response was more pronounced in HFD-LPS group. Brown adipose tissue (BAT) thermogenesis and increased energy expenditure seem to contribute to body weight loss in HFD-LPS. This response might be related with increased brain stem activation. In conclusion, LPS activates STAT3-mediated pathway in the hypothalamus and brain stem, leading to hypophagia, however, LPS effects on food intake, but not body weight loss, are abolished by leptin resistance induced by HFD. The preserved STAT3 phosphorylation in the brain stem suggests that unresponsiveness to LPS on STAT3 activation under HFD might be selective to the hypothalamus.
Collapse
Affiliation(s)
| | | | | | | | - Glauber S. F. da Silva
- Department of Physiology and
- Department of Morphology, Physiology and Basic Pathology, Dental School of Ribeirao Preto, University of Sao Paulo, Brazil
| | - Francisco José de Paula
- Department of Internal Medicine, School of Medicine of Ribeirao Preto, University of Sao Paulo, Brazil; and
| | - Luiz G. S. Branco
- Department of Morphology, Physiology and Basic Pathology, Dental School of Ribeirao Preto, University of Sao Paulo, Brazil
| | | | | |
Collapse
|
41
|
Murphy KT, Schwartz GJ, Nguyen NLT, Mendez JM, Ryu V, Bartness TJ. Leptin-sensitive sensory nerves innervate white fat. Am J Physiol Endocrinol Metab 2013; 304:E1338-47. [PMID: 23612999 PMCID: PMC3680695 DOI: 10.1152/ajpendo.00021.2013] [Citation(s) in RCA: 57] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Leptin, the primary white adipose tissue (WAT) adipokine, is thought to convey lipid reserve information to the brain via the circulation. Because WAT responds to environmental/internal signals in a fat pad-specific (FPS) manner, systemic signals such as leptin would fail to communicate such distinctive information. Saturation of brain leptin transport systems also would fail to convey increased lipid levels beyond that point. WAT possesses sensory innervation exemplified by proven sensory-associated peptides in nerves within the tissue and by viral sensory nerve-specific transneuronal tract tracer, H129 strain of herpes simplex virus 1 labeling of dorsal root ganglia (DRG) pseudounipolar neurons, spinal cord and central sensory circuits. Leptin as a paracrine factor activating WAT sensory innervation could supply the brain with FPS information. Therefore, we tested for and found the presence of the long form of the leptin receptor (Ob-Rb) on DRG pseudounipolar neurons immunohistochemically labeled after injections of Fluorogold, a retrograde tract tracer, into inguinal WAT (IWAT). Intra-IWAT leptin injections (300 ng) significantly elevated IWAT nerve spike rate within 5 min and persisted for at least 30 min. Intra-IWAT leptin injections also induced significant c-Fos immunoreactivity (ir), indicating neural activation across DRG pseudounipolar sensory neurons labeled with Fluorogold IWAT injections. Intraperitoneal leptin injection did not increase c-Fos-ir in DRG or the arcuate nucleus, nor did it increase arcuate signal transducer and activator of transcription 3 phosphorylation-ir. Collectively, these results strongly suggest that endogenous leptin secreted from white adipocytes functions as a paracrine factor to activate spinal sensory nerves innervating the tissue.
Collapse
Affiliation(s)
- Keegan T Murphy
- Department of Biology, Obesity Reversal Center, Georgia State University, Atlanta, Georgia; and
| | | | | | | | | | | |
Collapse
|
42
|
Hatzelmann T, Harden LM, Roth J, Gerstberger R. Antipyretic effect of central [Pyr1]apelin13 on LPS-induced fever in the rat. ACTA ACUST UNITED AC 2013; 184:6-13. [DOI: 10.1016/j.regpep.2013.03.018] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2012] [Revised: 01/15/2013] [Accepted: 03/03/2013] [Indexed: 12/20/2022]
|
43
|
Velmurugan S, Russell JA, Leng G. Systemic leptin increases the electrical activity of supraoptic nucleus oxytocin neurones in virgin and late pregnant rats. J Neuroendocrinol 2013; 25:383-90. [PMID: 23298261 DOI: 10.1111/jne.12016] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/11/2012] [Revised: 11/12/2012] [Accepted: 12/20/2012] [Indexed: 12/14/2022]
Abstract
In the rat hypothalamus, fasting attenuates the expression of oxytocin and this can be reversed by exogenous leptin administration. In the present study, we investigated the effects of systemically administered leptin on the electrical activity of magnocellular neurones in the supraoptic nucleus of urethane-anaesthetised rats. In virgin female rats, systemic leptin significantly excited identified oxytocin neurones with no detected effects on the patterning of activity, as reflected by hazard function analyses. The lowest dose that was consistently effective was 100 μg/i.v., and this dose had no significant effect on vasopressin neurones. In virgin rats fasted overnight, the spontaneous firing rate of oxytocin neurones was significantly lower than in unfasted rats, although leptin had a similar excitatory effect as in unfasted rats. In late pregnant rats (days 19-21 of pregnancy), spontaneous firing rates of oxytocin neurones were higher than in virgins, and the initial response to leptin was similar to that in virgin rats, although the increase in activity was more persistent. In fasted pregnant rats, the mean spontaneous firing rate of oxytocin neurones was again lower than in unfasted rats, although leptin had no significant effect even at the higher dose of 1 mg/rat. Thus, fasting reduced the spontaneous firing rates of oxytocin neurones in nonpregnant rats, and this effect could be reversed by the excitatory effects of leptin. Pregnant rats showed some evidence of leptin resistance but only after an overnight fast.
Collapse
Affiliation(s)
- S Velmurugan
- Centre for Integrative Physiology, School of Biomedical Sciences, College of Medicine and Veterinary Medicine, University of Edinburgh, Edinburgh, UK
| | | | | |
Collapse
|
44
|
Jung CH, Kim MS. Molecular mechanisms of central leptin resistance in obesity. Arch Pharm Res 2013; 36:201-7. [DOI: 10.1007/s12272-013-0020-y] [Citation(s) in RCA: 86] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2012] [Accepted: 12/24/2012] [Indexed: 02/06/2023]
|
45
|
Enhanced leptin sensitivity, reduced adiposity, and improved glucose homeostasis in mice lacking exchange protein directly activated by cyclic AMP isoform 1. Mol Cell Biol 2012; 33:918-26. [PMID: 23263987 DOI: 10.1128/mcb.01227-12] [Citation(s) in RCA: 73] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
The prototypic second messenger cyclic AMP (cAMP) is essential for controlling cellular metabolism, including glucose and lipid homeostasis. In mammals, the majority of cAMP functions are mediated by cAMP-dependent protein kinase (PKA) and exchange proteins directly activated by cAMP (Epacs). To explore the physiological functions of Epac1, we generated Epac1 knockout mice. Here we report that Epac1 null mutants have reduced white adipose tissue and reduced plasma leptin levels but display heightened leptin sensitivity. Epac1-deficient mice are more resistant to high-fat diet-induced obesity, hyperleptinemia, and glucose intolerance. Furthermore, pharmacological inhibition of Epac by use of an Epac-specific inhibitor reduces plasma leptin levels in vivo and enhances leptin signaling in organotypic hypothalamic slices. Taken together, our results demonstrate that Epac1 plays an important role in regulating adiposity and energy balance.
Collapse
|
46
|
Folch J, Pedrós I, Patraca I, Sureda F, Junyent F, Beas-Zarate C, Verdaguer E, Pallàs M, Auladell C, Camins A. Neuroprotective and anti-ageing role of leptin. J Mol Endocrinol 2012; 49:R149-56. [PMID: 22967480 DOI: 10.1530/jme-12-0151] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
Leptin (Lep), an adipose-derived hormone, exerts very important functions in the body mainly on energy storage and availability. The physiological effects of Lep controlling the body weight and suppressing appetite are mediated by the long form of Lep receptor in the hypothalamus. Lep receptor activates several downstream molecules involved in key pathways related to cell survival such as STAT3, PI3K, MAPK, AMPK, CDK5 and GSK3β. Collectively, these pathways act in a coordinated manner and form a network that is fully involved in Lep physiological response. Although the major interest in Lep is related to its role in the regulation of energy balance, and since resistance to Lep affects is the primary risk factor for obesity, the interest on their effects on brain cognition and neuroprotection is increasing. Thus, Lep and Lep mimetic compounds now await and deserve systematic exploration as the orchestrator of protective responses in the nervous system. Moreover, Lep might promote the activation of a cognitive process that may retard or even partially reverse selected aspects of Alzheimer's disease or ageing memory loss.
Collapse
Affiliation(s)
- Jaume Folch
- Unitat de Farmacologia i Farmacognòsia, Institut de Biomedicina, Centros de Investigación Biomédica en Red Enfermedades Neurodegenerativas, Universitat de Barcelona, Nucli Universitari de Pedralbes, 08028 Barcelona, Spain
| | | | | | | | | | | | | | | | | | | |
Collapse
|
47
|
Sun B, Purcell RH, Terrillion CE, Yan J, Moran TH, Tamashiro KL. Maternal high-fat diet during gestation or suckling differentially affects offspring leptin sensitivity and obesity. Diabetes 2012; 61:2833-41. [PMID: 22751689 PMCID: PMC3478561 DOI: 10.2337/db11-0957] [Citation(s) in RCA: 193] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Maternal high-fat (HF) diet throughout gestation and suckling has long-term consequences on the offspring's metabolic phenotype. Here we determine the relative contribution of pre- or postnatal maternal HF diet on offspring's metabolic phenotype. Pregnant Sprague-Dawley rats were maintained on normal chow or HF diet throughout gestation and suckling. All litters were cross-fostered to chow or HF dams on postnatal day (PND)1, resulting in four groups. Body weight, body composition, and glucose tolerance were measured at weaning and in adulthood. Leptin sensitivity was assessed by signal transducer and activator of transcription (STAT)3 activation on PND10 and PND21. Pups cross-fostered to HF dams gained more body weight than chow pups by PND7 and persisted until weaning. Postnatal HF pups had greater adiposity, higher plasma leptin concentration, impaired glucose tolerance, and reduced phosphorylated STAT3 in response to leptin in the arcuate nucleus at weaning. After weaning, male offspring cross-fostered to HF dams were hyperphagic and maintained greater body weight than postnatal chow pups. Postnatal HF diet during suckling continued to impair glucose tolerance in male and female offspring in adulthood. Maternal HF diet during suckling has a greater influence in determining offspring's metabolic phenotype than prenatal HF diet exposure and could provide insight regarding optimal perinatal nutrition for mothers and children.
Collapse
Affiliation(s)
- Bo Sun
- Department of Physiology and Pathophysiology, Xi’an Jiaotong University School of Medicine, Xi’an, Shaanxi, People’s Republic of China
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Ryan H. Purcell
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Chantelle E. Terrillion
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Jianqun Yan
- Department of Physiology and Pathophysiology, Xi’an Jiaotong University School of Medicine, Xi’an, Shaanxi, People’s Republic of China
- Corresponding authors: Kellie L.K. Tamashiro, , and Jianqun Yan,
| | - Timothy H. Moran
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Kellie L.K. Tamashiro
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, Maryland
- Corresponding authors: Kellie L.K. Tamashiro, , and Jianqun Yan,
| |
Collapse
|
48
|
Neuroanatomical determinants of the sympathetic nerve responses evoked by leptin. Clin Auton Res 2012; 23:1-7. [PMID: 22714900 DOI: 10.1007/s10286-012-0168-4] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2012] [Accepted: 05/18/2012] [Indexed: 01/04/2023]
Abstract
Leptin is an adipocyte-derived hormone that relays a satiety signal to the brain. The effect of leptin on the sympathetic nervous system is an important aspect in the regulation of energy homeostasis as well as several other physiological functions. The arcuate nucleus of the hypothalamus is considered a major site for the regulation of physiological processes by leptin. However, there is growing recognition that other hypothalamic and extra-hypothalamic brain nuclei are important for leptin regulation of physiological processes including sympathetic nerve traffic. The current review discusses the various hypothalamic and extra-hypothalamic nuclei that have been implicated in leptin-induced increase in regional sympathetic nerve activity. The continuous rise in the prevalence of obesity underscores the importance of understanding the underlying neural mechanisms regulating sympathetic traffic to different tissues to design effective strategies to reverse obesity and associated diseases.
Collapse
|
49
|
Reidelberger R, Haver A, Chelikani PK, Apenteng B, Perriotte-Olson C, Anders K, Steenson S, Blevins JE. Effects of leptin replacement alone and with exendin-4 on food intake and weight regain in weight-reduced diet-induced obese rats. Am J Physiol Endocrinol Metab 2012; 302:E1576-85. [PMID: 22510712 PMCID: PMC3378160 DOI: 10.1152/ajpendo.00058.2012] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Abstract
Weight loss in obese humans produces a relative leptin deficiency, which is postulated to activate potent orexigenic and energy conservation mechanisms to restrict weight loss and promote weight regain. Here we determined whether leptin replacement alone or with GLP-1 receptor agonist exendin-4 attenuates weight regain or promotes greater weight loss in weight-reduced diet-induced obese (DIO) rats. Forty percent restriction in daily intake of a high-fat diet in DIO rats for 4 wk reduced body weight by 12%, body fat by 29%, and plasma leptin by 67% and normalized leptin sensitivity. When food restriction ended, body weight, body fat, and plasma leptin increased rapidly. Daily administration of leptin [3-h intraperitoneal (ip) infusions (4 nmol·kg(-1)·h(-1))] at onset and end of dark period for 3 wk did not attenuate hyperphagia and weight regain, nor did it affect mean daily meal sizes or meal numbers. Exendin-4 (50 pmol·kg(-1)·h(-1)) infusions during the same intervals prevented postrestriction hyperphagia and weight regain by normalizing meal size. Coadministration of leptin and exendin-4 did not reduce body weight more than exendin-4 alone. Instead, leptin began to attenuate the inhibitory effects of exendin-4 on food intake, meal size, and weight regain by the end of the second week of administration. Plasma leptin in rats receiving leptin was sevenfold greater than in rats receiving vehicle and 17-fold greater than in rats receiving exendin-4. Together, these results do not support the hypothesis that leptin replacement alone or with exendin-4 attenuates weight regain or promotes greater weight loss in weight-reduced DIO rats.
Collapse
Affiliation(s)
- Roger Reidelberger
- Veterans Affairs Research Service, Veterans Affairs Nebraska-Western Iowa Health Care System, Omaha, 68105, USA.
| | | | | | | | | | | | | | | |
Collapse
|
50
|
Dendritically targeted Bdnf mRNA is essential for energy balance and response to leptin. Nat Med 2012; 18:564-71. [PMID: 22426422 PMCID: PMC3327556 DOI: 10.1038/nm.2687] [Citation(s) in RCA: 152] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2011] [Accepted: 01/24/2012] [Indexed: 01/09/2023]
Abstract
Mutations in the Bdnf gene, which produces transcripts with either short or long 3' untranslated regions (3' UTRs), cause human obesity; however, the precise role of brain-derived neurotrophic factor (BDNF) in the regulation of energy balance is unknown. Here we show the relationship between Bdnf mRNA with a long 3' UTR (long 3' UTR Bdnf mRNA), leptin, neuronal activation and body weight. We found that long 3' UTR Bdnf mRNA was enriched in the dendrites of hypothalamic neurons and that insulin and leptin could stimulate its translation in dendrites. Furthermore, mice harboring a truncated long Bdnf 3' UTR developed severe hyperphagic obesity, which was completely reversed by viral expression of long 3' UTR Bdnf mRNA in the hypothalamus. In these mice, the ability of leptin to activate hypothalamic neurons and inhibit food intake was compromised despite normal activation of leptin receptors. These results reveal a novel mechanism linking leptin action to BDNF expression during hypothalamic-mediated regulation of body weight, while also implicating dendritic protein synthesis in this process.
Collapse
|