51
|
Stengel A, Mori M, Taché Y. The role of nesfatin-1 in the regulation of food intake and body weight: recent developments and future endeavors. Obes Rev 2013; 14:859-70. [PMID: 23980879 PMCID: PMC3810163 DOI: 10.1111/obr.12063] [Citation(s) in RCA: 59] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/19/2013] [Revised: 06/29/2013] [Accepted: 07/09/2013] [Indexed: 01/07/2023]
Abstract
Nesfatin-1 was discovered in 2006 and introduced as a potential novel anorexigenic modulator of food intake and body weight. The past years have witnessed increasing evidence establishing nesfatin-1 as a potent physiological inhibitor of food intake and body weight and unravelled nesfatin-1's interaction with other brain transmitters to exert its food consumption inhibitory effect. As observed for other anorexigenic brain neuropeptides, nesfatin-1 is also likely to exert additional, if not pleiotropic, actions in the brain and periphery. Recent studies established the prominent expression of the nesfatin-1 precursor, nucleobindin2 (NUCB2), in the stomach and pancreas, where nesfatin-1 influences endocrine secretion. This review will highlight the current experimental state-of-knowledge on the effects of NUCB2/nesfatin-1 on food intake, body weight and glucose homeostasis. Potential implications in human obesity will be discussed in relation to the evidence of changes in circulating levels of NUCB2/nesfatin-1 in disease states, the occurrence of genetic NUCB2 polymorphisms and--in contrast to several other hormones--the independence of leptin signalling known to be blunted under conditions of chronically increased body weight.
Collapse
Affiliation(s)
- A Stengel
- Charité Center for Internal Medicine and Dermatology, Division for General Internal and Psychosomatic Medicine, Charité- Universitätsmedizin Berlin, Berlin, Germany
| | | | | |
Collapse
|
52
|
Abstract
The recently discovered nesfatin-1 is regulated by hunger and satiety. The precursor protein NUCB2 is proteolytically cleaved into three resulting fragments: nesfatin-1, nesfatin-2, and nesfatin-3. The middle segment of nesfatin-1 (M30) is responsible for limiting food intake, while the exact physiological role of nesfatin-2 and nesfatin-3 are not currently known yet. This hormone plays role/roles on diabetic hyperphagia, epilepsy, mood, stress, sleeping, anxiety, hyperpolarization, depolarization, and reproductive functions. This review will address nesfatin, focusing on its discovery and designation, biochemical structure, scientific evidence of its anorexigenic character, the results of the human and animal studies until the present day, its main biochemical and physiological effects, and its possible clinical applications.
Collapse
Affiliation(s)
- Suleyman Aydin
- Department of Medical Biochemistry and Clinical Biochemistry (Firat Hormones Research Group), Medical School, Firat University, 23119, Elazig, Turkey,
| |
Collapse
|
53
|
Jego S, Glasgow SD, Herrera CG, Ekstrand M, Reed SJ, Boyce R, Friedman J, Burdakov D, Adamantidis AR. Optogenetic identification of a rapid eye movement sleep modulatory circuit in the hypothalamus. Nat Neurosci 2013; 16:1637-43. [PMID: 24056699 PMCID: PMC4974078 DOI: 10.1038/nn.3522] [Citation(s) in RCA: 308] [Impact Index Per Article: 25.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2013] [Accepted: 08/23/2013] [Indexed: 12/22/2022]
Abstract
Rapid-Eye Movement (REM) sleep correlates with neuronal activity in the brainstem, basal forebrain and lateral hypothalamus (LH). LH melanin-concentrating hormone (MCH)-expressing neurons are active during sleep, however, their action on REM sleep remains unclear. Using optogenetic tools in newly-generated Tg(Pmch-Cre) mice, we found that acute activation of MCH neurons (ChETA, SSFO) at the onset of REM sleep extended the duration of REM, but not non-REM sleep episode. In contrast, their acute silencing (eNpHR3.0, ArchT) reduced the frequency and amplitude of hippocampal theta rhythm, without affecting REM sleep duration. In vitro activation of MCH neuron terminals induced GABAA-mediated inhibitory post-synaptic currents (IPSCs) in wake-promoting histaminergic neurons of the tuberomammillary nucleus (TMN), while in vivo activation of MCH neuron terminals in TMN or medial septum also prolonged REM sleep episodes. Collectively, these results suggest that activation of MCH neurons maintains REM sleep, possibly through inhibition of arousal circuits in the mammalian brain.
Collapse
Affiliation(s)
- Sonia Jego
- Douglas Institute, Department of Psychiatry, McGill University, Montreal, Canada
| | | | | | | | | | | | | | | | | |
Collapse
|
54
|
Abstract
AIM: To investigate the effect of Nesfatin-1 on gastric acid secretion in rats.
METHODS: Thirty-six male Sprague-Dawley rats were randomly and equally divided into six groups to receive intracerebroventricular injection of 0.05, 0.5 μg of Nesfatin-1 or sterile water and intravenous injection of 10, 50 μg/kg of Nesfatin-1 or sterile water. Gastric secretion was measured using the pylorus-ligation method. Three hours after treatment, rats were killed to remove the stomach and collect the gastric contents. The volume of gastric secretion was measured and the amount of gastric acid was determined by titration with NaOH. H+-K+-ATPase mRNA expression was detected by RT-PCR.
RESULTS: Intracerebroventricular infusion of 0.05 or 0.5 μg of Nesfatin-1 significantly reduced the volume of gastric juice (3.3 mL/3 h ± 0.3 mL/3 h vs 2.4 mL/3 h ± 0.3 mL/3 h; 3.3 mL/3 h ± 0.3 mL/3 h vs 2.5 mL/3 h ± 0.3 mL/3 h, both P < 0.05), inhibited gastric acid output (582.7 μmol/3 h ± 59.3 μmol/3 h vs 373.6 μmol/3 h ± 61.5 μmol/3h, 582.7 μmol/3 h ± 59.3 μmol/3h vs 380.0 μmol/3 h ± 55.8 μmol/3h, both P < 0.05), and decreased gastric H+-K+-ATPase mRNA expression (both P < 0.05). Intravenous injection of 10 or 50 μg/kg of Nesfatin-1 had no significant effect on the volume of gastric juice (3.7 mL/3 h ± 0.7 mL/3 h vs 3.3 mL/3 h ± 0.4 mL/3 h, 3.7 mL/3 h ± 0.7 mL/3 h vs 3.8 mL/3 h ± 0.5 mL/3 h, both P > 0.05), gastric acid output (647.6 μmol/3 h ± 102.8 μmol/3 h vs 573.8 μmol/3 h ± 97.4 μmol/3 h, 647.6 μmol/3 h ± 102.8 μmol/3 h vs 594.4 μmol/3 h ± 121.0 μmol/3 h, both P > 0.05) and gastric H+-K+-ATPase mRNA expression (both P >0.05 ).
CONCLUSION: Nesfatin-1 acts in the central nervous system to inhibit gastric acid secretion in rats.
Collapse
|
55
|
Ablation of neurons expressing melanin-concentrating hormone (MCH) in adult mice improves glucose tolerance independent of MCH signaling. J Neurosci 2013; 33:2009-16. [PMID: 23365238 DOI: 10.1523/jneurosci.3921-12.2013] [Citation(s) in RCA: 55] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
Melanin-concentrating hormone (MCH)-expressing neurons have been ascribed many roles based on studies of MCH-deficient mice. However, MCH neurons express other neurotransmitters, including GABA, nesfatin, and cocaine-amphetamine-regulated transcript. The importance of these other signaling molecules made by MCH neurons remains incompletely characterized. To determine the roles of MCH neurons in vivo, we targeted expression of the human diphtheria toxin receptor (DTR) to the gene for MCH (Pmch). Within 2 weeks of diphtheria toxin injection, heterozygous Pmch(DTR/+) mice lost 98% of their MCH neurons. These mice became lean but ate normally and were hyperactive, especially during a fast. They also responded abnormally to psychostimulants. For these phenotypes, ablation of MCH neurons recapitulated knock-out of MCH, so MCH appears to be the critical neuromodulator released by these neurons. In contrast, MCH-neuron-ablated mice showed improved glucose tolerance when compared with MCH-deficient mutant mice and wild-type mice. We conclude that MCH neurons regulate glucose tolerance through signaling molecules other than MCH.
Collapse
|
56
|
The vertebrate diencephalic MCH system: a versatile neuronal population in an evolving brain. Front Neuroendocrinol 2013; 34:65-87. [PMID: 23088995 DOI: 10.1016/j.yfrne.2012.10.001] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/26/2012] [Revised: 10/05/2012] [Accepted: 10/10/2012] [Indexed: 11/22/2022]
Abstract
Neurons synthesizing melanin-concentrating hormone (MCH) are described in the posterior hypothalamus of all vertebrates investigated so far. However, their anatomy is very different according to species: they are small and periventricular in lampreys, cartilaginous fishes or anurans, large and neuroendocrine in bony fishes, or distributed over large regions of the lateral hypothalamus in many mammals. An analysis of their comparative anatomy alongside recent data about the development of the forebrain, suggests that although very different, MCH neurons of the caudal hypothalamus are homologous. We further hypothesize that their divergent anatomy is linked to divergence in the forebrain - in particular telencephalic evolution.
Collapse
|
57
|
Vas S, Ádori C, Könczöl K, Kátai Z, Pap D, Papp RS, Bagdy G, Palkovits M, Tóth ZE. Nesfatin-1/NUCB2 as a potential new element of sleep regulation in rats. PLoS One 2013; 8:e59809. [PMID: 23560056 PMCID: PMC3613383 DOI: 10.1371/journal.pone.0059809] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2012] [Accepted: 02/18/2013] [Indexed: 11/17/2022] Open
Abstract
Study Objectives Millions suffer from sleep disorders that often accompany severe illnesses such as major depression; a leading psychiatric disorder characterized by appetite and rapid eye movement sleep (REMS) abnormalities. Melanin-concentrating hormone (MCH) and nesfatin-1/NUCB2 (nesfatin) are strongly co - expressed in the hypothalamus and are involved both in food intake regulation and depression. Since MCH was recognized earlier as a hypnogenic factor, we analyzed the potential role of nesfatin on vigilance. Design We subjected rats to a 72 h-long REMS deprivation using the classic flower pot method, followed by a 3 h-long ‘rebound sleep’. Nesfatin mRNA and protein expressions as well as neuronal activity (Fos) were measured by quantitative in situ hybridization technique, ELISA and immunohistochemistry, respectively, in ‘deprived’ and ‘rebound’ groups, relative to controls sacrificed at the same time. We also analyzed electroencephalogram of rats treated by intracerebroventricularly administered nesfatin-1, or saline. Results REMS deprivation downregulated the expression of nesfatin (mRNA and protein), however, enhanced REMS during ‘rebound’ reversed this to control levels. Additionally, increased transcriptional activity (Fos) was demonstrated in nesfatin neurons during ‘rebound’. Centrally administered nesfatin-1 at light on reduced REMS and intermediate stage of sleep, while increased passive wake for several hours and also caused a short-term increase in light slow wave sleep. Conclusions The data designate nesfatin as a potential new factor in sleep regulation, which fact can also be relevant in the better understanding of the role of nesfatin in the pathomechanism of depression.
Collapse
Affiliation(s)
- Szilvia Vas
- Department of Pharmacodynamics, Semmelweis University, Budapest, Hungary
| | | | | | | | | | | | | | | | | |
Collapse
|
58
|
Shimizu H, Mori M. Nesfatin-1: its role in the diagnosis and treatment of obesity and some psychiatric disorders. Methods Mol Biol 2013; 963:327-338. [PMID: 23296620 DOI: 10.1007/978-1-62703-230-8_20] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/01/2023]
Abstract
We discovered a new anorexigenic protein, nesfatin/nucleobindin-2 (NUCB2), which includes an EF-hand, calcium-binding motif. Nesfatin/NUCB2 is converted to nesfatin-1, which may be a physiologically active form in the body. Centrally and systemically administered nesfatin-1 inhibits appetite and body weight gain in rodents. The mid-segment of nesfatin-1 appears to be important in the inhibition of food intake. Intranasal administration of the mid-segment inhibits appetite. Nesfatin-1 may also be involved in the regulation of gastrointestinal function and insulin secretion. We have summarized the recent progress in the research of nesfatin-1.
Collapse
Affiliation(s)
- Hiroyuki Shimizu
- Department of Health and Nutrition, Faculty of Health and Welfare, Takasaki University of Health and Welfare, Takasaki, Gunma, Japan.
| | | |
Collapse
|
59
|
Jego S, Salvert D, Renouard L, Mori M, Goutagny R, Luppi PH, Fort P. Tuberal hypothalamic neurons secreting the satiety molecule Nesfatin-1 are critically involved in paradoxical (REM) sleep homeostasis. PLoS One 2012; 7:e52525. [PMID: 23300698 PMCID: PMC3531409 DOI: 10.1371/journal.pone.0052525] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2012] [Accepted: 11/15/2012] [Indexed: 01/17/2023] Open
Abstract
The recently discovered Nesfatin-1 plays a role in appetite regulation as a satiety factor through hypothalamic leptin-independent mechanisms. Nesfatin-1 is co-expressed with Melanin-Concentrating Hormone (MCH) in neurons from the tuberal hypothalamic area (THA) which are recruited during sleep states, especially paradoxical sleep (PS). To help decipher the contribution of this contingent of THA neurons to sleep regulatory mechanisms, we thus investigated in rats whether the co-factor Nesfatin-1 is also endowed with sleep-modulating properties. Here, we found that the disruption of the brain Nesfatin-1 signaling achieved by icv administration of Nesfatin-1 antiserum or antisense against the nucleobindin2 (NUCB2) prohormone suppressed PS with little, if any alteration of slow wave sleep (SWS). Further, the infusion of Nesfatin-1 antiserum after a selective PS deprivation, designed for elevating PS needs, severely prevented the ensuing expected PS recovery. Strengthening these pharmacological data, we finally demonstrated by using c-Fos as an index of neuronal activation that the recruitment of Nesfatin-1-immunoreactive neurons within THA is positively correlated to PS but not to SWS amounts experienced by rats prior to sacrifice. In conclusion, this work supports a functional contribution of the Nesfatin-1 signaling, operated by THA neurons, to PS regulatory mechanisms. We propose that these neurons, likely releasing MCH as a synergistic factor, constitute an appropriate lever by which the hypothalamus may integrate endogenous signals to adapt the ultradian rhythm and maintenance of PS in a manner dictated by homeostatic needs. This could be done through the inhibition of downstream targets comprised primarily of the local hypothalamic wake-active orexin- and histamine-containing neurons.
Collapse
Affiliation(s)
- Sonia Jego
- Sleep-Waking Neuronal Networks, CNRS - UMR5292; INSERM - U1028, Lyon Neuroscience Research Center (CRNL), Lyon, France
- University Claude Bernard Lyon 1, Lyon, France
- University of Lyon, Lyon, France
| | - Denise Salvert
- Sleep-Waking Neuronal Networks, CNRS - UMR5292; INSERM - U1028, Lyon Neuroscience Research Center (CRNL), Lyon, France
- University Claude Bernard Lyon 1, Lyon, France
- University of Lyon, Lyon, France
| | - Leslie Renouard
- Sleep-Waking Neuronal Networks, CNRS - UMR5292; INSERM - U1028, Lyon Neuroscience Research Center (CRNL), Lyon, France
- University Claude Bernard Lyon 1, Lyon, France
- University of Lyon, Lyon, France
| | - Masatomo Mori
- Department of Medicine and Molecular Science, Gunma University Graduate School of Medicine, Maebashi, Gunma, Japan
| | - Romain Goutagny
- Sleep-Waking Neuronal Networks, CNRS - UMR5292; INSERM - U1028, Lyon Neuroscience Research Center (CRNL), Lyon, France
- University Claude Bernard Lyon 1, Lyon, France
- University of Lyon, Lyon, France
| | - Pierre-Hervé Luppi
- Sleep-Waking Neuronal Networks, CNRS - UMR5292; INSERM - U1028, Lyon Neuroscience Research Center (CRNL), Lyon, France
- University Claude Bernard Lyon 1, Lyon, France
- University of Lyon, Lyon, France
| | - Patrice Fort
- Sleep-Waking Neuronal Networks, CNRS - UMR5292; INSERM - U1028, Lyon Neuroscience Research Center (CRNL), Lyon, France
- University Claude Bernard Lyon 1, Lyon, France
- University of Lyon, Lyon, France
| |
Collapse
|
60
|
Chen YY, Chan RME, Tan KML, Poh LKS, Loke KY, Wang JP, Li H, Hu YH, Wang L, Lee KO, Li GW, Lee YS. The association of a nucleobindin 2 gene (NUCB2) variant with childhood adiposity. Gene 2012; 516:48-52. [PMID: 23266808 DOI: 10.1016/j.gene.2012.12.017] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2012] [Revised: 10/13/2012] [Accepted: 12/02/2012] [Indexed: 11/26/2022]
Abstract
Nucleobindin 2 (NUCB2) is a precursor of nesfatin-1, a hypothalamic anorectic neuropeptide. The association between variants of the NUCB2 gene and adiposity was examined. 142 severely obese Chinese children in Singapore, and 384 normal weight Chinese children from a longitudinal cohort from Da Qing, China, were studied. NUCB2 was screened using PCR and direct sequencing in 29 severely obese children and 24 non-obese children, then screened for a variant c.1012C>G (Q338E, or rs757081) in the rest of the cohort using TaqMan probe. Five variants, including c.1012C>G (Q338E) were found. Genotyping for c.1012C>G found that the GG genotype was significantly less frequent in the obese group; odds ratio for obese subjects carrying the CC and CG genotypes was 2.29 (95% CI 1.17-4.49) in the dominant model, CC genotype 2.86 (95% CI 1.41-5.81) in the additive model, and C allele 1.57 (95% CI 1.17-2.1). The findings were replicated in an independent cohort of 372 obese and 390 normal weight Chinese children, where the odds ratio of obese subjects with CC and CG genotypes was 1.69 (95% CI 1.12-2.55). Within the Da Qing cohort, subjects with the GG genotype had significantly lower BMI and percentage ideal weight for height (WFH) at 5 and 8years of age. Subjects with lower birth weights also had more pronounced difference in WFH and BMI at 5 and 10years of age between GG subjects versus CC/CG subjects. We postulate that GG genotype is protective against excessive weight gain, and factors which predispose to excessive weight gain such as higher birth weights may ameliorate the effect.
Collapse
Affiliation(s)
- Yan Yan Chen
- Endocrinology and Cardiovascular disease Center, Fuwai Hospital and Cardiovascular Institute, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
61
|
Gotoh K, Masaki T, Chiba S, Ando H, Shimasaki T, Mitsutomi K, Fujiwara K, Katsuragi I, Kakuma T, Sakata T, Yoshimatsu H. Nesfatin-1, corticotropin-releasing hormone, thyrotropin-releasing hormone, and neuronal histamine interact in the hypothalamus to regulate feeding behavior. J Neurochem 2012; 124:90-9. [PMID: 23106615 DOI: 10.1111/jnc.12066] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2012] [Revised: 10/16/2012] [Accepted: 10/17/2012] [Indexed: 11/30/2022]
Abstract
Nesfatin-1, corticotropin-releasing hormone (CRH), thyrotropin-releasing hormone (TRH), and hypothalamic neuronal histamine act as anorexigenics in the hypothalamus. We examined interactions among nesfatin-1, CRH, TRH, and histamine in the regulation of feeding behavior in rodents. We investigated whether the anorectic effect of nesfatin-1, α-fluoromethyl histidine (FMH; a specific suicide inhibitor of histidine decarboxylase that depletes hypothalamic neuronal histamine), a CRH antagonist, or anti-TRH antibody affects the anorectic effect of nesfatin-1, whether nesfatin-1 increases CRH and TRH contents and histamine turnover in the hypothalamus, and whether histamine increases nesfatin-1 content in the hypothalamus. We also investigated whether nesfatin-1 decreases food intake in mice with targeted disruption of the histamine H1 receptor (H1KO mice) and if the H1 receptor (H1-R) co-localizes in nesfatin-1 neurons. Nesfatin-1-suppressed feeding was partially attenuated in rats administered with FMH, a CRH antagonist, or anti-TRH antibody, and in H1KO mice. Nesfatin-1 increased CRH and TRH levels and histamine turnover, whereas histamine increased nesfatin-1 in the hypothalamus. Immunohistochemical analysis revealed H1-R expression on nesfatin-1 neurons in the paraventricular nucleus of the hypothalamus. These results indicate that CRH, TRH, and hypothalamic neuronal histamine mediate the suppressive effects of nesfatin-1 on feeding behavior.
Collapse
Affiliation(s)
- Koro Gotoh
- Department of Internal Medicine 1, Faculty of Medicine, Oita University, Yufu, Japan.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
62
|
Xia ZF, Fritze DM, Li JY, Chai B, Zhang C, Zhang W, Mulholland MW. Nesfatin-1 inhibits gastric acid secretion via a central vagal mechanism in rats. Am J Physiol Gastrointest Liver Physiol 2012; 303:G570-7. [PMID: 22723266 PMCID: PMC3468549 DOI: 10.1152/ajpgi.00178.2012] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Nesfatin-1, a novel hypothalamic peptide, inhibits nocturnal feeding behavior and gastrointestinal motility in rodents. The effects of nesfatin-1 on gastrointestinal secretory function, including gastric acid production, have not been evaluated. Nesfatin-1 was injected into the fourth intracerebral ventricle (4V) of chronically cannulated rats to identify a nesfatin dose sufficient to inhibit food intake. Nesfatin-1 (2 μg) inhibited dark-phase food intake, in a dose-dependent fashion, for >3 h. Gastric acid production was evaluated in urethane-anesthetized rats. Nesfatin-1 (2 μg) was introduced via the 4V following endocrine stimulation of gastric acid secretion by pentagastrin (2 μg·kg(-1)·h(-1) iv), vagal stimulation with 2-deoxy-D-glucose (200 mg/kg sc), or no stimulus. Gastric secretions were collected via gastric cannula and neutralized by titration to determine acid content. Nesfatin-1 did not affect basal and pentagastrin-stimulated gastric acid secretion, whereas 2-deoxy-D-glucose-stimulated gastric acid production was inhibited by nesfatin-1 in a dose-dependent manner. c-Fos immunofluorescence in brain sections was used to evaluate in vivo neuronal activation by nesfatin-1 administered via the 4V. Nesfatin-1 caused activation of efferent vagal neurons, as evidenced by a 16-fold increase in the mean number of c-Fos-positive neurons in the dorsal motor nucleus of the vagus (DMNV) in nesfatin-1-treated animals vs. controls (P < 0.01). Finally, nesfatin-induced Ca(2+) signaling was evaluated in primary cultured DMNV neurons from neonatal rats. Nesfatin-1 caused dose-dependent Ca(2+) increments in 95% of cultured DMNV neurons. These studies demonstrate that central administration of nesfatin-1, at doses sufficient to inhibit food intake, results in inhibition of vagally stimulated secretion of gastric acid. Nesfatin-1 activates DMNV efferent vagal neurons in vivo and triggers Ca(2+) signaling in cultured DMNV neurons.
Collapse
Affiliation(s)
- Ze-Feng Xia
- 1Department of General Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; and ,2Department of Surgery, University of Michigan, Ann Arbor, Michigan
| | | | - Ji-Yao Li
- 2Department of Surgery, University of Michigan, Ann Arbor, Michigan
| | - Biaoxin Chai
- 2Department of Surgery, University of Michigan, Ann Arbor, Michigan
| | - Chao Zhang
- 2Department of Surgery, University of Michigan, Ann Arbor, Michigan
| | - Weizhen Zhang
- 2Department of Surgery, University of Michigan, Ann Arbor, Michigan
| | | |
Collapse
|
63
|
Pałasz A, Krzystanek M, Worthington J, Czajkowska B, Kostro K, Wiaderkiewicz R, Bajor G. Nesfatin-1, a unique regulatory neuropeptide of the brain. Neuropeptides 2012; 46:105-12. [PMID: 22225987 DOI: 10.1016/j.npep.2011.12.002] [Citation(s) in RCA: 79] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/19/2011] [Revised: 12/19/2011] [Accepted: 12/19/2011] [Indexed: 12/21/2022]
Abstract
Nesfatin-1, a newly discovered NUCB2-derived satiety neuropeptide is expressed in several neurons of forebrain, hindbrain, brainstem and spinal cord. This novel anorexigenic substance seems to play an important role in hypothalamic pathways regulating food intake and energy homeostasis. Nesfatin-1 immunoreactive cells are detectable in arcuate (ARC), paraventricular (PVN) and supraoptic nuclei (SON), where the peptide is colocalized with POMC/CART, NPY, oxytocin and vasopressin. The nesfatin-1 molecule interacts with a G-protein coupled receptor and its cytophysiological effect depends on inhibitory hyperpolarization of NPY/AgRP neurons in ARC and melanocortin signaling in PVN. Administration of nesfatin-1 significantly inhibits consumatory behavior and decreases weight gain in experimental animals. These recent findings suggest the evidence for nesfatin-1 involvement in other important brain functions such as reproduction, sleep, cognition and anxiety- or stress-related responses. The neuroprotective and antiapoptotic properties of nesfatin-1 were also reported. From the clinical viewpoint it should be noteworthy, that the serum concentration of nesfatin-1 may be a sensitive marker of epileptic seizures. However, the details of nesfatin-1 physiology ought to be clarified, and it may be considered suitable in the future, as a potential drug in the pharmacotherapy of obesity, especially in patients treated with antipsychotics and antidepressants. On the other hand, some putative nesfatin-1 antagonists may improve eating disorders.
Collapse
Affiliation(s)
- Artur Pałasz
- Department of Histology, Medical University of Silesia, Medyków Street 18, 40-752 Katowice, Poland.
| | | | | | | | | | | | | |
Collapse
|
64
|
Li JH, Li XL, Wu J, Jia FY, Lin L. Nesfatin-1 inhibits gastric acid secretion by cultured rat gastric mucosa cells. Shijie Huaren Xiaohua Zazhi 2012; 20:1123-1130. [DOI: 10.11569/wcjd.v20.i13.1123] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
AIM: To clarify the effect of nesfatin-1 on gastric acid secretion and the expression of the H+/K+-ATPase mRNA and protein in rat gastric mucosa cells in vitro.
METHODS: Gastric mucosa cells were isolated from SD rats by enzymolysis and identified by immunofluorescence staining. Cultured rat gastric mucosa cells were divided into control group and nesfatin-1 group, and the nesfatin-1 group was pretreated with different concentrations (0, 10-4, 10-3, 10-2, 10-1 μmol/L) of nesfatin-1 for different durations (0, 1, 2, 3, 4 h). The effect of nesfatin-1 on gastric acid secretion was investigated by monitoring 14C-aminopyrine (14C-AP) accumulation, and the expression of H+/K+-ATPase α and β subunit mRNA and protein was examined by real-time PCR and Western blot.
RESULTS: Pretreatment with nesfatin-1 at a dose of 10-1 or 10-2 μmol/L for 2 or 3 h inhibited gastric acid secretion, but nesfatin-1 at a dose of 10-3 or 10-4 μmol/L had no such effect. Nesfatin-1 at a dose of 10-1 μmol/L inhibited the expression of H+/K+-ATPase α subunit mRNA after pretreatment for 1, 2, or 3 h and inhibited the expression of H+/K+-ATPase β subunit mRNAs after pretreatment for 1 or 2 h. In the dose range between 10-4 to 10-1 μmol/L, nesfatin-1 dose-dependently inhibited the expression of H+/K+-ATPase α subunit and β subunit mRNA after pretreatment for 2 h. Nesfatin-1 at a dose of 10-1 μmmol/L inhibited H+/K+-ATPase α subunit protein expression after pretreatment for 1, 2 or 3 h and inhibited H+/K+-ATPase β subunit protein expression after pretreatment for 2 or 3 h. In the dose range between 10-3 to 10-1 μmol/L, nesfatin-1 dose-dependently inhibited H+/K+-ATPase α and β subunit protein expression after pretreatment for 2 h.
CONCLUSION: Our data suggest that nesfatin-1 inhibits gastric acid secretion by rat gastric mucosa cells in vitro possibly by down-regulating the expression of H+/K+-ATPase mRNA and protein..
Collapse
|
65
|
Chen X, Dong J, Jiang ZY. Nesfatin-1 influences the excitability of glucosensing neurons in the hypothalamic nuclei and inhibits the food intake. ACTA ACUST UNITED AC 2012; 177:21-6. [PMID: 22561448 DOI: 10.1016/j.regpep.2012.04.003] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2012] [Revised: 03/27/2012] [Accepted: 04/23/2012] [Indexed: 10/28/2022]
Abstract
Nesfatin-1 is a recently discovered neuropeptide that has been shown to decrease food intake after lateral, third, or fourth brain ventricle, cisterna magna administration, or PVN injection in ad libitum fed rats. With regards to the understanding of nesfatin-1 brain sites of action, additional microinjection studies will be necessary to define specific nuclei, in addition to the PVN, responsive to nesfatin-1 to get insight into the differential effects on food intake. In the present study, we evaluated nesfatin-1 action to modulate food intake response upon injection into the specific hypothalamic nuclei (PVN, LHA and VMN) in freely fed rats during the dark phase. We extend previous observations by showing that the nesfatin-1 (50 pmol) injected before the onset of the dark period significantly reduced the 1 to 5 h cumulative food intake in rats cannulated into the PVN, LHA, but not in rats cannulated into the VMN. Glucosensing neurons located in the hypothalamus are involved in glucoprivic feeding and homeostatic control of blood glucose. In order to shed light on the mechanisms by which nesfatin-1 exerts its satiety-promoting actions, we examined the effect of nesfatin-1 on the excitability of hypothalamic glucosensing neurons. Nesfatin-1 excited most of the glucose-inhibited (GI) neurons and inhibited most of the glucose-excited (GE) neurons in the PVN. Of 34 GI neurons in the LHA tested, inhibitory effects were seen in 70.6% (24/34) of GI neurons. The main effects were excitatory after intra-VMN administration of nesfatin-1 in GE neurons (27/35, 77.1%). Thus, our data clearly demonstrate that nesfatin-1 may exert at least a part of its physiological actions on the control of food intake as a direct result of its role in modulating the excitability of glucosensing neurons in the PVN, LHA and VMN.
Collapse
Affiliation(s)
- Xi Chen
- Department of Physiology, Qingdao University School of Medicine, Qingdao 266071, China
| | | | | |
Collapse
|
66
|
García-Galiano D, Pineda R, Ilhan T, Castellano JM, Ruiz-Pino F, Sánchez-Garrido MA, Vazquez MJ, Sangiao-Alvarellos S, Romero-Ruiz A, Pinilla L, Diéguez C, Gaytán F, Tena-Sempere M. Cellular distribution, regulated expression, and functional role of the anorexigenic peptide, NUCB2/nesfatin-1, in the testis. Endocrinology 2012; 153:1959-71. [PMID: 22334726 DOI: 10.1210/en.2011-2032] [Citation(s) in RCA: 81] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
Nesfatin-1, product of the precursor NEFA/nucleobindin2 (NUCB2), was initially identified as anorectic hypothalamic neuropeptide, acting in a leptin-independent manner. In addition to its central role in the control of energy homeostasis, evidence has mounted recently that nesfatin-1 is also produced in peripheral metabolic tissues, such as pancreas, adipose, and gut. Moreover, nesfatin-1 has been shown to participate in the control of body functions gated by whole-body energy homeostasis, including puberty onset. Yet, whether, as is the case for other metabolic neuropeptides, NUCB2/nesfatin-1 participates in the direct control of gonadal function remains unexplored. We document here for the first time the expression of NUCB2 mRNA in rat, mouse, and human testes, where NUCB2/nesfatin-1 protein was identified in interstitial mature Leydig cells. Yet in rats, NUCB2/nesfatin-1 became expressed in Sertoli cells upon Leydig cell elimination and was also detected in Leydig cell progenitors. Although NUCB2 mRNA levels did not overtly change in rat testis during pubertal maturation and after short-term fasting, NUCB2/nesfatin-1 content significantly increased along the puberty-to-adult transition and was markedly suppressed after fasting. In addition, testicular NUCB2/nesfatin-1 expression was up-regulated by pituitary LH, because hypophysectomy decreased, whereas human choriogonadotropin (super-agonist of LH receptors) replacement enhanced, NUCB2/nesfatin-1 mRNA and peptide levels. Finally, nesfatin-1 increased human choriogonadotropin-stimulated testosterone secretion by rat testicular explants ex vivo. Our data are the first to disclose the presence and functional role of NUCB2/nesfatin-1 in the testis, where its expression is regulated by developmental, metabolic, and hormonal cues as well as by Leydig cell-derived factors. Our observations expand the reproductive dimension of nesfatin-1, which may operate directly at the testicular level to link energy homeostasis, puberty onset, and gonadal function.
Collapse
Affiliation(s)
- D García-Galiano
- Physiology Section, Department of Cell Biology, Physiology, and Immunology, Faculty of Medicine, University of Córdoba, Córdoba, Spain
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
67
|
Könczöl K, Pintér O, Ferenczi S, Varga J, Kovács K, Palkovits M, Zelena D, Tóth ZE. Nesfatin-1 exerts long-term effect on food intake and body temperature. Int J Obes (Lond) 2012; 36:1514-21. [PMID: 22290539 DOI: 10.1038/ijo.2012.2] [Citation(s) in RCA: 69] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
OBJECTIVE To determine whether the anorexigenic peptide, nesfatin-1 affects energy expenditure, and to follow the time course of its effects. DESIGN Food intake duration, core body temperature, locomotor activity and heart rate of rats were measured by telemetry for 48 h after a single intracerebroventricular injection of 25 or 100 pmol nesfatin-1 applied in the dark or the light phase of the day. Body weight, food and water intake changes were measured daily. Furthermore, cold-responsive nesfatin-1/NUCB2 neurons were mapped in the brain. RESULTS Nesfatin-1 reduced duration of nocturnal food intake for 2 days independently of circadian time injected, and raised body temperature immediately, or with little delay depending on the dose and circadian time applied. The body temperature remained higher during the next light phases of the 48 h observation period, and the circadian curve of temperature flattened. After light phase application, the heart rate was elevated transiently. Locomotion did not change. Daily food and water intake, as well as body weight measurements point to a potential decrease in all parameters on the first day and some degree of compensation on the second day. Cold-activated (Fos positive) nesfatin-1/NUCB2 neurones have been revealed in several brain nuclei involved in cold adaptation. Nesfatin-1 co-localised with prepro-thyrotropin-releasing hormone in cold responsive neurones of the hypothalamic paraventricular nucleus, and in neurones of the nucleus raphe pallidus and obscurus that are premotor neurones regulating brown adipose tissue thermogenesis and skin blood flow. CONCLUSION Nesfatin-1 has a remarkably prolonged effect on food intake and body temperature. Time course of nesfatin-1's effects may be varied depending on the time applied. Many of the nesfatin-1/NUCB2 neurones are cold sensitive, and are positioned in key centres of thermoregulation. Nesfatin-1 regulates energy expenditure a far more potent way than it was recognised before making it a preferable candidate anti-obesity drug.
Collapse
Affiliation(s)
- K Könczöl
- Neuromorphological and Neuroendocrine Research Laboratory, Department of Anatomy, Histology and Embryology, Semmelweis University and the Hungarian Academy of Sciences, Budapest, Hungary.
| | | | | | | | | | | | | | | |
Collapse
|
68
|
Dickens MJ, Cornil CA, Balthazart J. Acute stress differentially affects aromatase activity in specific brain nuclei of adult male and female quail. Endocrinology 2011; 152:4242-51. [PMID: 21878510 PMCID: PMC3199009 DOI: 10.1210/en.2011-1341] [Citation(s) in RCA: 58] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
The rapid and temporary suppression of reproductive behavior is often assumed to be an important feature of the adaptive acute stress response. However, how this suppression operates at the mechanistic level is poorly understood. The enzyme aromatase converts testosterone to estradiol in the brain to activate reproductive behavior in male Japanese quail (Coturnix japonica). The discovery of rapid and reversible modification of aromatase activity (AA) provides a potential mechanism for fast, stress-induced changes in behavior. We investigated the effects of acute stress on AA in both sexes by measuring enzyme activity in all aromatase-expressing brain nuclei before, during, and after 30 min of acute restraint stress. We show here that acute stress rapidly alters AA in the male and female brain and that these changes are specific to the brain nuclei and sex of the individual. Specifically, acute stress rapidly (5 min) increased AA in the male medial preoptic nucleus, a region controlling male reproductive behavior; in females, a similar increase was also observed, but it appeared delayed (15 min) and had smaller amplitude. In the ventromedial and tuberal hypothalamus, regions associated with female reproductive behavior, stress induced a quick and sustained decrease in AA in females, but in males, only a slight increase (ventromedial) or no change (tuberal) in AA was observed. Effects of acute stress on brain estrogen production, therefore, represent one potential way through which stress affects reproduction.
Collapse
Affiliation(s)
- Molly J Dickens
- University of Liège, GIGA Neurosciences, Research Group in Behavioral Neuroendocrinology, 1 Avenue de l'Hopital (B36), 4000 Liège, Belgium.
| | | | | |
Collapse
|
69
|
Stengel A, Taché Y. Minireview: nesfatin-1--an emerging new player in the brain-gut, endocrine, and metabolic axis. Endocrinology 2011; 152:4033-8. [PMID: 21862618 PMCID: PMC3199002 DOI: 10.1210/en.2011-1500] [Citation(s) in RCA: 58] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Nesfatin-1 is a recently identified 82-amino-acid peptide derived from the precursor protein, nucleobindin2 (NUCB2). The brain distribution of NUCB2/nesfatin-1 at the mRNA and protein level along with functional studies in rodents support a role for NUCB2/nesfatin-1 as a novel satiety molecule acting through leptin-independent mechanisms. In addition, nesfatin-1 induces a wide spectrum of central actions to stimulate the pituitary-adrenal axis and sympathetic nervous system and influences visceral functions and emotion. These central actions combined with the activation of NUCB2/nesfatin-1 neurons in the brain by various stressors are indicative of a role in the adaptive response under stressful conditions. In the periphery, evidence is mounting that nesfatin-1 exerts a direct glucose-dependent insulinotropic action on β-cells of the pancreatic islets. However, the cellular mechanisms of nesfatin-1's action remain poorly understood, partly because the receptor through which nesfatin-1 exerts its pleiotropic actions is yet to be identified.
Collapse
Affiliation(s)
- Andreas Stengel
- Department of Medicine, CURE Digestive Diseases Research Center, University of California, Los Angeles, Building 115, Room 117, Veterans Affairs Greater Los Angeles Healthcare System, 11301 Wilshire Boulevard, Los Angeles, California 90073, USA
| | | |
Collapse
|
70
|
Saldanha JF, Carrero JJ, Lobo JC, Stockler-Pinto MB, Leal VO, Calixto A, Geloneze B, Mafra D. The newly identified anorexigenic adipokine nesfatin-1 in hemodialysis patients: Are there associations with food intake, body composition and inflammation? ACTA ACUST UNITED AC 2011; 173:82-5. [PMID: 22036920 DOI: 10.1016/j.regpep.2011.09.010] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2011] [Revised: 09/06/2011] [Accepted: 09/29/2011] [Indexed: 10/15/2022]
Abstract
Nesfatin-1 is a recently identified anorexigenic peptide that has been implicated in appetite regulation, weight loss and/or malnutrition. Anorexia and malnutrition are common features of chronic kidney disease (CKD) that predispose patients to worse outcomes. However, the reasons for the occurrence of anorexia in CKD patients are not fully elucidated. The aim of this study was to investigate the association between nesfatin-1 and protein intake and body composition in patients undergoing hemodialysis (HD). Twenty five HD patients from a private Clinic in Rio de Janeiro, Brazil were studied and compared with 15 healthy subjects that were matched for body mass index (BMI), % body fat mass (by anthropometrics) and age. Appetite was measured using a specific questionnaire, and food intake was evaluated based on 3-day food records. Nesfatin-1 levels were measured by ELISA and leptin, TNF-α and IL-6 levels were determined by a multiplex assay kit. Serum nesfatin-1 levels did not differ between HD patients (0.16±0.07ng/mL) and healthy subjects (0.17±0.10ng/mL). Nesfatin-1 levels showed significant negative correlations with protein intake (r=-0.42; p=0.03), but did not associate with inflammatory markers or appetite scores. Combining patients and controls, we observed positive correlations with BMI (r=0.33; p=0.03), % body fat (r=0.35; p=0.03), leptin (r=0.45; p=0.006) and the triceps skinfold thickness (r=0.36; p=0.02). In multivariate analysis % body fat was the main determinant of nesfatin-1 variance. In conclusion, nesfatin-1 levels did not differ between HD patients and healthy subjects and negatively correlated with protein intake. This pathway is likely not dysregulated in uremia.
Collapse
Affiliation(s)
- J F Saldanha
- Clinical Nutrition Department, Nutrition Faculty, Federal University Fluminense (UFF), Niterói, Brazil.
| | | | | | | | | | | | | | | |
Collapse
|
71
|
Saldanha J, Carrero J, Mafra D. The possible role of nesfatin-1 on appetite regulation in hemodialysis patients. Med Hypotheses 2011; 77:654-7. [DOI: 10.1016/j.mehy.2011.07.006] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2010] [Revised: 06/01/2011] [Accepted: 07/01/2011] [Indexed: 11/26/2022]
|
72
|
Stengel A, Goebel-Stengel M, Jawien J, Kobelt P, Taché Y, Lambrecht NW. Lipopolysaccharide increases gastric and circulating NUCB2/nesfatin-1 concentrations in rats. Peptides 2011; 32:1942-1947. [PMID: 21782869 PMCID: PMC4057044 DOI: 10.1016/j.peptides.2011.07.006] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/27/2011] [Revised: 07/07/2011] [Accepted: 07/07/2011] [Indexed: 02/07/2023]
Abstract
Bacterial lipopolysaccharide (LPS) is an established animal model to study the innate immune response to Gram-negative bacteria mimicking symptoms of infection including reduction of food intake. LPS decreases acyl ghrelin associated with decreased concentrations of circulating ghrelin-O-acyltransferase (GOAT) likely contributing to the anorexigenic effect. We also recently described the prominent expression of the novel anorexigenic hormone, nucleobindin2 (NUCB2)/nesfatin-1 in gastric X/A-like cells co-localized with ghrelin in different pools of vesicles. To investigate whether LPS would affect gastric and circulating NUCB2/nesfatin-1 concentration, ad libitum fed rats were equipped with an intravenous (iv) catheter. LPS was injected intraperitoneally (ip, 100μg/kg) and blood was withdrawn before and at 2, 5, 7 and 24h post injection and processed for NUCB2/nesfatin-1 radioimmunoassay. Gastric corpus was collected to measure NUCB2 mRNA expression by RT-qPCR and NUCB2/nesfatin-1 protein concentration by Western blot. Injection of LPS increased plasma NUCB2/nesfatin-1 concentrations by 43%, 78% and 62% compared to vehicle at 2h, 5h and 7h post injection respectively (p<0.05) and returned to baseline at 24h. The plasma NUCB2/nesfatin-1 increase at 2h was associated with increased corpus NUCB2 mRNA expression (p<0.01), whereas NUCB2 mRNA was not detectable in white blood cells. Likewise, gastric NUCB2 protein concentration was increased by 62% after LPS compared to vehicle (p<0.01). These data show that gastric NUCB2 production and release are increased in response to LPS. These changes are opposite to those of ghrelin in response to LPS supporting a differential gastric regulation of NUCB2/nesfatin-1 and ghrelin expression derived from the same cell by immune challenge.
Collapse
Affiliation(s)
- Andreas Stengel
- CURE/Digestive Diseases Research Center, Center for Neurobiology of Stress, Department of Medicine, Digestive Diseases Division at the University of California Los Angeles, and VA Greater Los Angeles Health Care System, CA 90073, USA
- Department of Medicine, Division Psychosomatic Medicine and Psychotherapy, Charité, Campus Mitte, Universitätsmedizin Berlin, Germany
| | - Miriam Goebel-Stengel
- CURE/Digestive Diseases Research Center, Center for Neurobiology of Stress, Department of Medicine, Digestive Diseases Division at the University of California Los Angeles, and VA Greater Los Angeles Health Care System, CA 90073, USA
- Department of Medicine and Institute of Neurogastroenterology, Martin-Luther-Krankenhaus, Berlin, Germany
| | - Janusz Jawien
- Gastrointestinal Endocrinology, Veterans Affairs Long Beach Healthcare System, Long Beach, CA 90822, USA
| | - Peter Kobelt
- Department of Medicine, Division Psychosomatic Medicine and Psychotherapy, Charité, Campus Mitte, Universitätsmedizin Berlin, Germany
| | - Yvette Taché
- CURE/Digestive Diseases Research Center, Center for Neurobiology of Stress, Department of Medicine, Digestive Diseases Division at the University of California Los Angeles, and VA Greater Los Angeles Health Care System, CA 90073, USA
| | - Nils W.G. Lambrecht
- Gastrointestinal Endocrinology, Veterans Affairs Long Beach Healthcare System, Long Beach, CA 90822, USA
| |
Collapse
|
73
|
Burt J, Alberto CO, Parsons MP, Hirasawa M. Local network regulation of orexin neurons in the lateral hypothalamus. Am J Physiol Regul Integr Comp Physiol 2011; 301:R572-80. [PMID: 21697524 DOI: 10.1152/ajpregu.00674.2010] [Citation(s) in RCA: 75] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
Obesity and inadequate sleep are among the most common causes of health problems in modern society. Thus, the discovery that orexin (hypocretin) neurons play a pivotal role in sleep/wake regulation, energy balance, and consummatory behaviors has sparked immense interest in understanding the regulatory mechanisms of these neurons. The local network consisting of neurons and astrocytes within the lateral hypothalamus and perifornical area (LH/PFA), where orexin neurons reside, shapes the output of orexin neurons and the LH/PFA. Orexin neurons not only send projections to remote brain areas but also contribute to the local network where they release multiple neurotransmitters to modulate its activity. These neurotransmitters have opposing actions, whose balance is determined by the amount released and postsynaptic receptor desensitization. Modulation and negative feedback regulation of excitatory glutamatergic inputs as well as release of astrocyte-derived factors, such as lactate and ATP, can also affect the excitability of orexin neurons. Furthermore, distinct populations of LH/PFA neurons express neurotransmitters with known electrophysiological actions on orexin neurons, such as melanin-concentrating hormone, corticotropin-releasing factor, thyrotropin-releasing hormone, neurotensin, and GABA. These LH/PFA-specific mechanisms may be important for fine tuning the firing activity of orexin neurons to maintain optimal levels of prolonged output to sustain wakefulness and stimulate consummatory behaviors. Building on these exciting findings should shed further light onto the cellular mechanisms of energy balance and sleep-wake regulation.
Collapse
Affiliation(s)
- Julia Burt
- Division of Biomedical Sciences, Faculty of Medicine, Memorial University, St. John's, Newfoundland, Canada
| | | | | | | |
Collapse
|
74
|
Goebel-Stengel M, Wang L, Stengel A, Taché Y. Localization of nesfatin-1 neurons in the mouse brain and functional implication. Brain Res 2011; 1396:20-34. [PMID: 21555116 PMCID: PMC3104076 DOI: 10.1016/j.brainres.2011.04.031] [Citation(s) in RCA: 112] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2010] [Revised: 03/30/2011] [Accepted: 04/17/2011] [Indexed: 12/17/2022]
Abstract
Nesfatin-1 reduces food intake when injected centrally in rodents. We recently described wide distribution of nucleobindin2 (NUCB2)/nesfatin-1 immunoreactivity in rat brain autonomic nuclei activated by various stressors. We used C57BL/6 mice to localize brain NUCB2/nesfatin-1 immunoreactivity and assessed activation of NUCB2/nesfatin 1 neurons after water avoidance stress (WAS). Gastric emptying of a non-nutrient liquid was also determined. NUCB2/nesfatin-1 immunoreactivity was detected in cortical areas including piriform, insular, cingulate and somatomotor cortices, the limbic system including amygdaloid nuclei, hippocampus and septum, the basal ganglia, bed nucleus of the stria terminalis, the thalamus including paraventricular and parafascicular nuclei, the hypothalamus including supraoptic, periventricular, paraventricular (PVN), arcuate nuclei and ventromedial and lateral hypothalamic areas. Intensely labeled NUCB2/nesfatin-1 neurons were detected in a previously undefined region which we named intermediate dorsomedial hypothalamus. In the brainstem, NUCB2/nesfatin-1 immunoreactivity was detected in the raphe nuclei, Edinger-Westphal nucleus, locus coeruleus (LC), lateral parabrachial nucleus, ventrolateral medulla (VLM) and dorsal vagal complex. WAS induced Fos expression in 35% of NUCB2/nesfatin-1-immunoreactive neurons in the PVN, 50% in the LC, 54% in the rostral raphe pallidus, 58% in the VLM, 39% in the middle part of the nucleus of the solitary tract (NTS) and 33% in the caudal NTS. Nesfatin-1 injected intracerebroventricularly significantly decreased gastric emptying. These data showed that NUCB2/nesfatin-1 immunoreactivity is distributed in mouse brain areas involved in the regulation of stress response and visceral functions activated by an acute psychological stressor suggesting that nesfatin-1 might play a role in the efferent component of the stress response.
Collapse
Affiliation(s)
- Miriam Goebel-Stengel
- CURE/Digestive Diseases Research Center, Center for Neurobiology of Stress, Digestive Diseases Division, Department of Medicine, David Geffen School of Medicine, UCLA, VA Greater Los Angeles Healthcare System, Los Angeles, California, USA
| | - Lixin Wang
- CURE/Digestive Diseases Research Center, Center for Neurobiology of Stress, Digestive Diseases Division, Department of Medicine, David Geffen School of Medicine, UCLA, VA Greater Los Angeles Healthcare System, Los Angeles, California, USA
| | - Andreas Stengel
- CURE/Digestive Diseases Research Center, Center for Neurobiology of Stress, Digestive Diseases Division, Department of Medicine, David Geffen School of Medicine, UCLA, VA Greater Los Angeles Healthcare System, Los Angeles, California, USA
| | - Yvette Taché
- CURE/Digestive Diseases Research Center, Center for Neurobiology of Stress, Digestive Diseases Division, Department of Medicine, David Geffen School of Medicine, UCLA, VA Greater Los Angeles Healthcare System, Los Angeles, California, USA
| |
Collapse
|
75
|
Abstract
The protein nucleobindin 2 (NUCB2) or NEFA (DNA binding/EF-hand/acidic amino acid rich region) was identified over a decade ago and implicated in intracellular processes. New developments came with the report that post-translational processing of hypothalamic NUCB2 may result in nesfatin-1, nesfatin-2 and nesfatin-3 and convergent studies showing that nesfatin-1 and full length NUCB2 injected in the brain potently inhibit the dark phase food intake in rodents including leptin receptor deficient Zucker rats. Nesfatin-1 also reduces body weight gain, suggesting a role as a new anorexigenic factor and modulator of energy balance. In light of the obesity epidemic and its associated diseases, underlying new mechanisms regulating food intake may be promising targets in the drug treatment of obese patients particularly as the vast majority of them display reduced leptin sensitivity or leptin resistance while nesfatin-1's mechanism of action is leptin independent. Although much progress on the localization of NUCB2/nesfatin-1 in the brain and periphery as well as on the understanding of nesfatin-1's anorexic effect have been achieved during the past three years, several important mechanisms have yet to be unraveled such as the identification of the nesfatin-1 receptor and the regulation of NUCB2 processing and nesfatin-1 release.
Collapse
Affiliation(s)
- A Stengel
- Department of Medicine, CURE Digestive Diseases Research Center and Center for Neurobiology of Stress, Digestive Diseases Division UCLA, and VA Greater Los Angeles Healthcare System, Los Angeles, CA 90073, USA
| | | | | |
Collapse
|
76
|
Torterolo P, Lagos P, Monti JM. Melanin-concentrating hormone: a new sleep factor? Front Neurol 2011; 2:14. [PMID: 21516258 PMCID: PMC3080035 DOI: 10.3389/fneur.2011.00014] [Citation(s) in RCA: 55] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2010] [Accepted: 03/02/2011] [Indexed: 12/18/2022] Open
Abstract
Neurons containing the neuropeptide melanin-concentrating hormone (MCH) are mainly located in the lateral hypothalamus and the incerto-hypothalamic area, and have widespread projections throughout the brain. While the biological functions of this neuropeptide are exerted in humans through two metabotropic receptors, the MCHR1 and MCHR2, only the MCHR1 is present in rodents. Recently, it has been shown that the MCHergic system is involved in the control of sleep. We can summarize the experimental findings as follows: (1) The areas related to the control of sleep and wakefulness have a high density of MCHergic fibers and receptors. (2) MCHergic neurons are active during sleep, especially during rapid eye movement (REM) sleep. (3) MCH knockout mice have less REM sleep, notably under conditions of negative energy balance. Animals with genetically inactivated MCHR1 also exhibit altered vigilance state architecture and sleep homeostasis. (4) Systemically administered MCHR1 antagonists reduce sleep. (5) Intraventricular microinjection of MCH increases both slow wave sleep (SWS) and REM sleep; however, the increment in REM sleep is more pronounced. (6) Microinjection of MCH into the dorsal raphe nucleus increases REM sleep time. REM seep is inhibited by immunoneutralization of MCH within this nucleus. (7) Microinjection of MCH in the nucleus pontis oralis of the cat enhances REM sleep time and reduces REM sleep latency. All these data strongly suggest that MCH has a potent role in the promotion of sleep. Although both SWS and REM sleep are facilitated by MCH, REM sleep seems to be more sensitive to MCH modulation.
Collapse
Affiliation(s)
- Pablo Torterolo
- Department of Physiology, School of Medicine, University of the Republic Montevideo, Uruguay
| | | | | |
Collapse
|
77
|
Centrally administered nesfatin-1 inhibits feeding behaviour and gastroduodenal motility in mice. Neuroreport 2011; 21:1008-11. [PMID: 20827224 DOI: 10.1097/wnr.0b013e32833f7b96] [Citation(s) in RCA: 97] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Nesfatin-1 was recently identified as a peptide with anorexigenic effects that is localized in the hypothalamus and adipocytes. Not much is known about the effect of nesfatin-1 on gut motility. Food intake was measured after intracerebroventricular administration of nesfatin-1 in food-deprived mice. Antral and duodenal motility was assessed by using a manometric method in conscious fed mice. We found that centrally administered nesfatin-1 decreased food intake and inhibited gastroduodenal motility in mice. These results suggest that nesfatin-1 influences gut motility and feeding behaviour.
Collapse
|
78
|
Goebel M, Stengel A, Wang L, Taché Y. Central nesfatin-1 reduces the nocturnal food intake in mice by reducing meal size and increasing inter-meal intervals. Peptides 2011; 32:36-43. [PMID: 20933030 PMCID: PMC3010516 DOI: 10.1016/j.peptides.2010.09.027] [Citation(s) in RCA: 84] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/25/2010] [Revised: 09/29/2010] [Accepted: 09/29/2010] [Indexed: 10/19/2022]
Abstract
Nesfatin-1 is well established to reduce food intake upon brain injection in rats, while in mice its anorexigenic action and brain expression are largely unexplored. We characterized the influence of intracerebroventricular (icv) and peripheral (intraperitoneal, ip, subcutaneous, sc) injection of nesfatin-1 on dark phase ingestive behavior using an automated feeding monitoring system and co-localized NUCB2/nesfatin-1 immunoreactivity in the associated brain areas. Nesfatin-1 (0.3, 1 or 3 μg/mouse, icv) caused a dose-related reduction of 4-h dark phase food intake by 13%, 27%, and 46% respectively. Nesfatin-1 (3 μg/mouse, icv) action had a 2-h delayed onset, 82% peak inhibition occurring at 3-4h post-injection and was long lasting (30% reduction for 12h period post-injection). Nesfatin-1 (3 μg/mouse, icv)-treated mice had a 46% lower meal frequency associated with 2-times longer inter-meal intervals and a 35% reduction in meal size compared to vehicle during the 1-4h post-injection (p<0.05). NUCB2/nesfatin-1-immunopositive neurons were found in hypothalamic (supraoptic, paraventricular, arcuate, dorsomedial, lateral) and brainstem (dorsal vagal complex) feeding regulatory nuclei. When injected peripherally, neither food intake nor feeding microstructure parameters were altered. These results demonstrate that NUCB2/nesfatin-1 is prominently expressed in mouse hypothalamus and medulla and acts in the brain to curtail the dark phase feeding by inducing satiation and satiety indicated by reduced meal size and prolonged inter-meal intervals respectively. The lack of nesfatin-1 effect when injected peripherally at a 23-times higher dose indicates a primarily central site of the anorexigenic action for nesfatin-1 in mice.
Collapse
Affiliation(s)
- Miriam Goebel
- CURE/Digestive Diseases Research Center, Center for Neurobiology of Stress, Department of Medicine, Digestive Diseases Division at the University of California Los Angeles, and Veterans Affairs Greater Los Angeles Health Care System, CA 90073, USA
| | | | | | | |
Collapse
|
79
|
Abstract
The initial discovery of leptin, an appetite-suppressing hormone originating from fat tissue, substantially supported the idea that fat-borne factors act on the brain to regulate food intake and energy expenditure. Since then, a growing number of cytokines have been found to be released from adipose tissue, thus acting in an endocrine manner. These adipocytokines include not only, e.g., adiponectin, apelin, resistin, and visfatin, but also inflammatory cytokines and steroid hormones such as estrogens and glucocorticoids. They are secreted from their adipose depots and differ in terms of release stimuli, downstream signaling, and their action on the brain. Clearly, adipocytokines play a prominent role in the central control of body weight, and the deregulation of this circuit may lead to the development of obesity and related disorders. In this chapter, we will focus on crosstalk mechanisms and the deregulation of adipocytokines at the expression level and/or sites of central action that eventually will lead to the development and perpetuation of obesity and diabetes.
Collapse
Affiliation(s)
- Carla Schulz
- Department of Internal Medicine I, Luebeck University, Ratzeburger Allee 160, 23538 Luebeck, Germany
| | | | | |
Collapse
|
80
|
Ogiso K, Asakawa A, Amitani H, Nakahara T, Ushikai M, Haruta I, Koyama KI, Amitani M, Harada T, Yasuhara D, Inui A. Plasma nesfatin-1 concentrations in restricting-type anorexia nervosa. Peptides 2011; 32:150-3. [PMID: 20937336 DOI: 10.1016/j.peptides.2010.10.004] [Citation(s) in RCA: 54] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/09/2010] [Revised: 10/04/2010] [Accepted: 10/04/2010] [Indexed: 01/19/2023]
Abstract
Restricting-type anorexia nervosa (AN-R) is characterized by chronic food restriction and severe emaciation due to various cognitive biases such as a distorted self-image. In spite of several treatments, AN-R continues to be a refractory disease because of its unknown pathogenesis. Although previous studies have shown that changes in feeding regulatory peptides such as ghrelin are involved in anorexia, few reports have described the relationship between AN-R and nesfatin-1, a recently identified satiety peptide. Therefore, we examined the plasma nesfatin-1 levels in AN-R patients to determine its role in AN-R. A total of 15 women participated in the study; 7 patients with AN-R and 8 age-matched healthy controls (average BMI, 13.02 ± 0.30 vs. 21.57 ± 0.48, respectively). Our results showed that plasma nesfatin-1 levels were significantly lower in AN-R group than in control group (6.23 ± 0.70 ng/ml vs. 8.91 ± 0.85 ng/ml, respectively, P<0.05). Plasma acyl ghrelin and des-acyl ghrelin levels were significantly higher in AN-R group than in control group (acyl ghrelin: 62.4 ± 10.15 fmol/ml vs. 27.20 ± 5.60 fmol/ml, P<0.01 and des-acyl ghrelin: 300.17 ± 55.95 fmol/ml vs. 107.34 ± 40.63 fmol/ml, P<0.05). Although AN-R is associated with emaciation for a prolonged period, our result suggested that nesfatin-1 levels may be regulated by nutrition status and response to starvation.
Collapse
Affiliation(s)
- Kazuma Ogiso
- Division of Psychosomatic Internal Medicine, Department of Social and Behavioral Medicine, Kagoshima University Graduate School of Medical and Dental Sciences, 8-35-1 Sakuragaoka, Kagoshima 890-8520, Japan
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
81
|
Gonzalez R, Kerbel B, Chun A, Unniappan S. Molecular, cellular and physiological evidences for the anorexigenic actions of nesfatin-1 in goldfish. PLoS One 2010; 5:e15201. [PMID: 21151928 PMCID: PMC2997068 DOI: 10.1371/journal.pone.0015201] [Citation(s) in RCA: 89] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2010] [Accepted: 10/31/2010] [Indexed: 01/08/2023] Open
Abstract
Background Nesfatin-1 is a recently discovered anorexigen encoded in the precursor peptide, nucleobindin-2 (NUCB2) in mammals. To date, nesfatin-1 has not been described in any non-mammalian species, although some information is available in the sequenced genomes of several species. Our objective was to characterize nesfatin-1 in fish. Methodology/Principal Findings In the present study, we employed molecular, immunohistochemical, and physiological studies to characterize the structure, distribution, and appetite regulatory effects of nesfatin-1 in a non-mammalian vertebrate. A very high conservation in NUCB2 sequences, especially in the nesfatin-1 region was found in lower vertebrates. Abundant expression of NUCB2 mRNA was detected in several tissues including the brain and liver of goldfish. Nesfatin-1-like immunoreactive cells are present in the feeding regulatory nucleus of the hypothalamus and in the gastrointestinal tract of goldfish. Approximately 6-fold increase in NUCB2 mRNA levels was found in the liver after 7-day food-deprivation, and a similar increase was also found after short-term fasting. This points toward a possible liver specific role for NUCB2 in the control of metabolism during food-deprivation. Meanwhile, ∼2-fold increase at 1 and 3 h post-feeding and an ∼3-fold reduction after a 7-day food-deprivation was observed in NUCB2 mRNA in the goldfish hypothalamus. In vivo, a single intraperitoneal injection of the full-length native (goldfish; gf) nesfatin-1 at a dose of 50 ng/g body weight induced a 23% reduction of food intake one hour post-injection in goldfish. Furthermore, intracerebroventricular injection of gfnesfatin-1 at a dose of 5 ng/g body weight resulted in ∼50% reduction in food intake. Conclusions/Significance Our results provide molecular, anatomical and functional evidences to support potential anorectic and metabolic roles for endogenous nesfatin-1 in goldfish. Collectively, we provide novel information on NUCB2 in non-mammals and an anorexigenic role for nesfatin-1 in goldfish.
Collapse
Affiliation(s)
- Ronald Gonzalez
- Laboratory of Integrative Neuroendocrinology, Department of Biology, York University, Toronto, Ontario, Canada
| | - Brent Kerbel
- Laboratory of Integrative Neuroendocrinology, Department of Biology, York University, Toronto, Ontario, Canada
| | - Alexander Chun
- Laboratory of Integrative Neuroendocrinology, Department of Biology, York University, Toronto, Ontario, Canada
| | - Suraj Unniappan
- Laboratory of Integrative Neuroendocrinology, Department of Biology, York University, Toronto, Ontario, Canada
- * E-mail:
| |
Collapse
|
82
|
The anorexigenic neuropeptide, nesfatin-1, is indispensable for normal puberty onset in the female rat. J Neurosci 2010; 30:7783-92. [PMID: 20534827 DOI: 10.1523/jneurosci.5828-09.2010] [Citation(s) in RCA: 106] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
The hypothalamic peptide, nesfatin-1, derived from the precursor NEFA/nucleobindin 2 (NUCB2), was recently identified as anorexigenic signal, acting in a leptin-independent manner. Yet its participation in the regulation of other biological functions gated by body energy status remains unexplored. We show herein that NUCB2/nesfatin-1 is involved in the control of female puberty. NUCB2/nesfatin mRNA and protein were detected at the hypothalamus of pubertal female rats, with prominent signals at lateral hypothalamus (LHA), paraventricular (PVN), and supraoptic (SON) nuclei. Hypothalamic NUCB2 expression raised along pubertal transition, with detectable elevations of its mRNA levels at LHA, PVN, and SON, and threefold increase of its total protein content between late-infantile and peripubertal periods. Conditions of negative energy balance, such as 48 h fasting or sustained subnutrition, decreased hypothalamic NUCB2 mRNA and/or protein levels in pubertal females. At this age, central administration of nesfatin-1 induced modest but significant elevations of circulating gonadotropins, whose magnitude was notably augmented in conditions of food deprivation. Continuous intracerebroventricular infusion of antisense morpholino oligonucleotides (as-MONs) against NUCB2 along pubertal maturation, which markedly reduced hypothalamic NUCB2 protein content, delayed vaginal opening and decreased ovarian weights and serum luteinizing hormone (LH) levels. In contrast, in adult female rats, intracerebroventricular injection of nesfatin did not stimulate LH or follicle-stimulating hormone secretion; neither did central as-MON infusion alter preovulatory gonadotropin surges, despite suppression of hypothalamic NUCB2. In sum, our data are the first to disclose the indispensable role of NUCB2/nesfatin-1 in the central networks driving puberty onset, a function that may contribute to its functional coupling to energy homeostasis.
Collapse
|
83
|
Stengel A, Taché Y. Nesfatin-1--role as possible new potent regulator of food intake. REGULATORY PEPTIDES 2010; 163:18-23. [PMID: 20580651 PMCID: PMC2902658 DOI: 10.1016/j.regpep.2010.05.002] [Citation(s) in RCA: 59] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 04/08/2010] [Accepted: 05/11/2010] [Indexed: 01/13/2023]
Abstract
Nesfatin-1 is an 82 amino acid peptide recently discovered in the brain which is derived from nucleobindin2 (NUCB2), a protein that is highly conserved across mammalian species. Nesfatin-1 has received much attention over the past two years due to its reproducible food intake-reducing effect that is linked with recruitment of other hypothalamic peptides regulating feeding behavior. A growing amount of evidence also supports that various stressors activate fore- and hindbrain NUCB2/nesfatin-1 circuitries. In this review, we outline the central nervous system distribution of NUCB2/nesfatin-1, and recent developments on the peripheral expression of NUCB2/nesfatin-1, in particular its co-localization with ghrelin in gastric X/A-like cells and insulin in ss-cells of the endocrine pancreas. Functional studies related to the characteristics of nesfatin-1's inhibitory effects on dark phase food intake are detailed as well as the central activation of NUCB2/nesfatin-1 immunopositive neurons in the response to psychological, immune and visceral stressors. Lastly, potential clinical implications of targeting NUCB2/nesfatin-1 signaling and existing gaps in knowledge to ascertain the role and mechanisms of action of nesfatin-1 are presented.
Collapse
Affiliation(s)
- Andreas Stengel
- Department of Medicine, CURE Digestive Diseases Research Center, Digestive Diseases Division UCLA, Los Angeles, CA 90073, USA
| | | |
Collapse
|
84
|
Zhang AQ, Li XL, Jiang CY, Lin L, Shi RH, Chen JD, Oomura Y. Expression of nesfatin-1/NUCB2 in rodent digestive system. World J Gastroenterol 2010; 16:1735-1741. [PMID: 20380005 PMCID: PMC2852821 DOI: 10.3748/wjg.v16.i14.1735] [Citation(s) in RCA: 89] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/20/2009] [Revised: 01/20/2010] [Accepted: 01/27/2010] [Indexed: 02/06/2023] Open
Abstract
AIM To observe the regional distributions and morphological features of nesfatin-1/nucleobindin-2 (NUCB2) immunoreactive (IR) cells in the rodent digestive system. METHODS Paraffin-embedded sections of seven organs (pancreas, stomach, duodenum, esophagus, liver, small intestine and colon) dissected from sprague-dawley (SD) rats and imprinting-control-region (ICR) mice were prepared. The regional distributions of nesfatin-1/NUCB2 IR cells were observed by immunohistochemical staining. The morphological features of the nesfatin-1/NUCB2 IR cells were evaluated by hematoxylin and eosin (HE) staining. Fresh tissues of the seven organs were prepared for Western blotting to analyze the relative protein levels of NUCB2 in each organ. RESULTS Immunohistochemical staining showed that the nesfatin-1/NUCB2 IR cells were localized in the central part of the pancreatic islets, the lower third and middle portion of the gastric mucosal gland, and the submucous layer of the duodenum in SD rats and ICR mice. HE staining revealed that the morphological features of nesfatin-1/NUCB2 IR cells were mainly islet cells in the pancreas, endocrine cells in the stomach, and Brunner's glands in the duodenum. Western blotting revealed that NUCB2 protein expression was higher in the pancreas, stomach and duodenum than in the esophagus, liver, small intestine and colon (P = 0.000). CONCLUSION Nesfatin-1/NUCB2 IR cells are expressed in the pancreas, stomach and duodenum in rodents. These cells may play an important role in the physiological regulation of carbohydrate metabolism, gastrointestinal function and nutrient absorption.
Collapse
|
85
|
Inhoff T, Stengel A, Peter L, Goebel M, Taché Y, Bannert N, Wiedenmann B, Klapp BF, Mönnikes H, Kobelt P. Novel insight in distribution of nesfatin-1 and phospho-mTOR in the arcuate nucleus of the hypothalamus of rats. Peptides 2010; 31:257-62. [PMID: 19961888 PMCID: PMC4043136 DOI: 10.1016/j.peptides.2009.11.024] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/23/2009] [Revised: 11/24/2009] [Accepted: 11/24/2009] [Indexed: 11/24/2022]
Abstract
Recently, two proteins have been localized in the arcuate nucleus (ARC) and implicated in the regulation of food intake: the serine-threonine-kinase mammalian target of rapamycin (mTOR) as part of the TOR signaling complex 1 (TORC1), and nesfatin-1 derived from the precursor protein nucleobindin2. However, the exact cell types are not well described. Therefore, we performed double-labeling studies for NPY, CART, nesfatin-1 and pmTOR in the ARC. In this study, we showed that nesfatin-1 is not only intracellularly co-localized with cocaine- and amphetamine-regulated transcript (CART) peptide as reported before, but also with phospho-mTOR (pmTOR) and neuropeptide Y (NPY) in ARC neurons. Quantification revealed that 59+/-5% of the pmTOR-immunoreactive (ir) neurons were immunoreactive for nesfatin-1. Moreover, double labeling for nesfatin-1 and NPY exhibited that 19+/-5% of the NPY positive cells were also immunoreactive for nesfatin-1. Furthermore, we could also confirm results from previous studies, showing that the majority of nesfatin-1 neurons are also positive for CART peptide, whereas most of the pmTOR is co-localized with NPY and only to a lesser extent with CART.
Collapse
Affiliation(s)
- Tobias Inhoff
- Department of Medicine, Division Hepatology, Gastroenterology, and Endocrinology, Charité, Campus Virchow, Universitätsmedizin Berlin, Germany
| | - Andreas Stengel
- Department of Medicine, Division of Digestive Diseases, CURE Digestive Diseases Research Center and Center for Neurobiology of Stress, UCLA and Veterans Affairs Greater Los Angeles Healthcare System, Los Angeles, CA, USA
| | - Lisa Peter
- Department of Medicine, Division Hepatology, Gastroenterology, and Endocrinology, Charité, Campus Virchow, Universitätsmedizin Berlin, Germany
| | - Miriam Goebel
- Department of Medicine, Division of Digestive Diseases, CURE Digestive Diseases Research Center and Center for Neurobiology of Stress, UCLA and Veterans Affairs Greater Los Angeles Healthcare System, Los Angeles, CA, USA
| | - Yvette Taché
- Department of Medicine, Division of Digestive Diseases, CURE Digestive Diseases Research Center and Center for Neurobiology of Stress, UCLA and Veterans Affairs Greater Los Angeles Healthcare System, Los Angeles, CA, USA
| | | | - Bertram Wiedenmann
- Department of Medicine, Division Hepatology, Gastroenterology, and Endocrinology, Charité, Campus Virchow, Universitätsmedizin Berlin, Germany
| | - Burghard F. Klapp
- Department of Medicine, Division Psychosomatic Medicine and Psychotherapy, Charité, Campus Mitte, Universitätsmedizin Berlin, Luisenstraße 13 A, 10117 Berlin, Germany
| | - Hubert Mönnikes
- Department of Medicine and Institute of Neurogastroenterology, Martin-Luther-Hospital, Berlin, Germany
| | - Peter Kobelt
- Department of Medicine, Division Hepatology, Gastroenterology, and Endocrinology, Charité, Campus Virchow, Universitätsmedizin Berlin, Germany
- Department of Medicine, Division Psychosomatic Medicine and Psychotherapy, Charité, Campus Mitte, Universitätsmedizin Berlin, Luisenstraße 13 A, 10117 Berlin, Germany
- Corresponding author. Tel.: +49 30 450 559739
| |
Collapse
|
86
|
Griffond B, Risold PY. MCH and feeding behavior-interaction with peptidic network. Peptides 2009; 30:2045-51. [PMID: 19619600 DOI: 10.1016/j.peptides.2009.07.008] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/26/2009] [Revised: 04/17/2009] [Accepted: 07/09/2009] [Indexed: 12/20/2022]
Abstract
Numerous works associate the MCH peptide, and the hypothalamic neurons that produce it, to the feeding behavior and energy homeostasis. It is commonly admitted that MCH is an orexigenic peptide, and MCH neurons could be under the control of arcuate NPY and POMC neurons. However, the literature data is not always concordant. In particular questions about the intrahypothalamic circuit involving other neuropeptides and about the mechanisms through which MCH could act are not yet clearly answered. For example, which receptors mediate a MCH response to NPY or alpha-MSH, does MCH act alone, is there any local anatomical organization within the tuberal LHA? A review of the current literature is then needed to help focus attention on these unresolved and often neglected issues.
Collapse
Affiliation(s)
- B Griffond
- Université de Franche-Comté, Besançon, France
| | | |
Collapse
|
87
|
Peyron C, Sapin E, Leger L, Luppi PH, Fort P. Role of the melanin-concentrating hormone neuropeptide in sleep regulation. Peptides 2009; 30:2052-9. [PMID: 19660508 DOI: 10.1016/j.peptides.2009.07.022] [Citation(s) in RCA: 54] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/03/2009] [Revised: 07/24/2009] [Accepted: 07/24/2009] [Indexed: 10/20/2022]
Abstract
Melanin-concentrating hormone (MCH), a neuropeptide secreted by a limited number of neurons within the tuberal hypothalamus, has been drawn in the field of sleep only fairly recently in 2003. Since then, growing experimental evidence indicates that MCH may play a crucial role in the homeostatic regulation of paradoxical sleep (PS). MCH-expressing neurons fire specifically during PS. When injected icv MCH induces a 200% increase in PS quantities in rats and the lack of MCH induces a decrease in sleep quantities in transgenic mice. Here, we review recent studies suggesting a role for MCH in the regulation of the sleep-wake cycle, in particular PS, including insights on (1) the specific activity of MCH neurons during PS; (2) how they might be controlled across the sleep-wake cycle; (3) how they might modulate PS; (4) and finally whether MCH might take part in the expression of some symptoms observed in primary sleep disorders.
Collapse
Affiliation(s)
- Christelle Peyron
- UMR CNRS, Université Claude Bernard Lyon1, Université de Lyon, Institut Fédératif des Neurosciences de Lyon, France.
| | | | | | | | | |
Collapse
|
88
|
CCK-8S activates c-Fos in a dose-dependent manner in nesfatin-1 immunoreactive neurons in the paraventricular nucleus of the hypothalamus and in the nucleus of the solitary tract of the brainstem. ACTA ACUST UNITED AC 2009; 157:84-91. [DOI: 10.1016/j.regpep.2009.06.009] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2009] [Revised: 06/02/2009] [Accepted: 06/12/2009] [Indexed: 01/16/2023]
|
89
|
Bonnet MS, Pecchi E, Trouslard J, Jean A, Dallaporta M, Troadec JD. Central nesfatin-1-expressing neurons are sensitive to peripheral inflammatory stimulus. J Neuroinflammation 2009; 6:27. [PMID: 19778412 PMCID: PMC2762958 DOI: 10.1186/1742-2094-6-27] [Citation(s) in RCA: 70] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2009] [Accepted: 09/24/2009] [Indexed: 12/05/2022] Open
Abstract
Recently, a novel factor with anorexigenic properties was identified and called nesfatin-1. This protein (82 aac) is not only expressed in peripheral organs but it is also found in neurons located in specific structures including the hypothalamus and the brainstem, two sites strongly involved in food intake regulation. Here, we studied whether some of the neurons that become activated following an injection of an anorectic dose of lipopolysaccharides (LPS) exhibit a nesfatin-1 phenotype. To this end, we used double immunohistochemistry to target the expression of the immediate-early gene c-fos and of nesfatin-1 on coronal frozen sections of the rat brain. The number of c-Fos+/nesfatin-1+ neurons was evaluated in the immunosensitive structures reported to contain nesfatin-1 neurons; i.e. paraventricular hypothalamic nucleus (PVN), supraoptic nucleus (SON), arcuate nucleus (ARC) and nucleus of the solitary tract (NTS). LPS strongly increased the number of c-Fos+/nesfatin-1+ neurons in the PVN, SON and NTS, and to a lesser extent in the ARC. Triple labeling showed that a portion of the nesfatin-1 neurons activated in response to LPS within the NTS are catecholaminergic since they co-express tyrosine hydroxylase (TH). Our data therefore indicate that a portion of nesfatin-1 neurons of both the hypothalamus and brainstem are sensitive to peripheral inflammatory signals, and provide the first clues suggesting that centrally released nesfatin-1 may contribute to the neural mechanisms leading to endotoxaemic anorexia.
Collapse
Affiliation(s)
- Marion S Bonnet
- Centre de Recherche en Neurobiologie-Neurophysiologie de Marseille (CRN2M), UMR 6231 CNRS, Marseille, France
- Département de Physiologie Neurovégétative, USC INRA 2027, Université Paul Cézanne, Université de la Méditerranée, Marseille, France
| | - Emilie Pecchi
- Centre de Recherche en Neurobiologie-Neurophysiologie de Marseille (CRN2M), UMR 6231 CNRS, Marseille, France
- Département de Physiologie Neurovégétative, USC INRA 2027, Université Paul Cézanne, Université de la Méditerranée, Marseille, France
| | - Jérôme Trouslard
- Centre de Recherche en Neurobiologie-Neurophysiologie de Marseille (CRN2M), UMR 6231 CNRS, Marseille, France
- Département de Physiologie Neurovégétative, USC INRA 2027, Université Paul Cézanne, Université de la Méditerranée, Marseille, France
| | - André Jean
- Centre de Recherche en Neurobiologie-Neurophysiologie de Marseille (CRN2M), UMR 6231 CNRS, Marseille, France
- Département de Physiologie Neurovégétative, USC INRA 2027, Université Paul Cézanne, Université de la Méditerranée, Marseille, France
| | - Michel Dallaporta
- Centre de Recherche en Neurobiologie-Neurophysiologie de Marseille (CRN2M), UMR 6231 CNRS, Marseille, France
- Département de Physiologie Neurovégétative, USC INRA 2027, Université Paul Cézanne, Université de la Méditerranée, Marseille, France
| | - Jean-Denis Troadec
- Centre de Recherche en Neurobiologie-Neurophysiologie de Marseille (CRN2M), UMR 6231 CNRS, Marseille, France
- Département de Physiologie Neurovégétative, USC INRA 2027, Université Paul Cézanne, Université de la Méditerranée, Marseille, France
| |
Collapse
|
90
|
Melanin-concentrating hormone directly inhibits GnRH neurons and blocks kisspeptin activation, linking energy balance to reproduction. Proc Natl Acad Sci U S A 2009; 106:17217-22. [PMID: 19805188 DOI: 10.1073/pnas.0908200106] [Citation(s) in RCA: 85] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
A link between energy balance and reproduction is critical for the survival of all species. Energy-consuming reproductive processes need to be aborted in the face of a negative energy balance, yet knowledge of the pathways mediating this link remains limited. Fasting and food restriction that inhibit fertility also upregulate the hypothalamic melanin-concentrating hormone (MCH) system that promotes feeding and decreases energy expenditure; MCH knockout mice are lean and have a higher metabolism but remain fertile. MCH also modulates sleep, drug abuse behavior, and mood, and MCH receptor antagonists are currently being developed as antiobesity and antidepressant drugs. Despite the clinical implications of MCH, the direct postsynaptic effects of MCH have never been reported in CNS neurons. Using patch-clamp recordings in brain slices from multiple lines of transgenic GFP mice, we demonstrate a strong inhibitory effect of MCH on an exclusive population of septal vGluT2-GnRH neurons that is activated by the puberty-triggering and preovulatory luteinizing hormone surge-mediating peptide, kisspeptin. MCH has no effect on kisspeptin-insensitive GnRH, vGluT2, cholinergic, or GABAergic neurons located within the same nucleus. The inhibitory effects of MCH are reproducible and nondesensitizing and are mediated via a direct postsynaptic Ba(2+)-sensitive K(+) channel mechanism involving the MCHR1 receptor. MCH immunoreactive fibers are in close proximity to vGluT2-GFP and GnRH-GFP neurons. Importantly, MCH blocks the excitatory effect of kisspeptin on vGluT2-GnRH neurons. Considering the role of MCH in regulating energy balance and of GnRH and kisspeptin in triggering puberty and maintaining fertility, MCH may provide a critical link between energy balance and reproduction directly at the level of the kisspeptin-activated vGluT2-GnRH neuron.
Collapse
|
91
|
Abstract
An anorexigenic peptide, nesfatin-1 was found in rat hypothalamus, and its expression in the paraventricular nucleus of the hypothalamus was reduced by starvation. Intracerebroventricular administration dose-dependently inhibited food intake for 6 h in male Wistar and leptin resistant, Zucker fatty rats. There may be a crosstalk between nesfatin-1 pathway and melanocortin pathway in the brain. Nesfatin-1 neurons co-express with oxytocin, vasopressin and melanin concentrating hormone in the hypothalamus. Intraperitoneal administration of nesfatin-1 and its mid-segment dose-dependently inhibited food intake for 3 h. Mid-segment of nesfatin-1 decreased food intake under leptin-resistant animal models of obesity. Intraperitoneal administration of the mid-segment of nesfatin-1 increased proopiomelanocortin and cocain- and amphetamine-related peptide mRNA expression in the nucleus of the solitary tract, but not in arcuate nucleus of the hypothalamus. In this review, we summarized recent progress in the research about the possible mechanism of nesfatin-1-induced anorexia.
Collapse
Affiliation(s)
- Hiroyuki Shimizu
- aDepartment of Medicine and Molecular Science, Gunma University Graduate School of Medicine, Maebashi, Japan.
| | | | | | | | | |
Collapse
|
92
|
Goebel M, Stengel A, Lambrecht NW, Wang L, Taché Y. Nesfatin-1 immunoreactivity in rat brain and spinal cord autonomic nuclei. Neurosci Lett 2009; 452:241-6. [PMID: 19348732 PMCID: PMC2674947 DOI: 10.1016/j.neulet.2009.01.064] [Citation(s) in RCA: 147] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2008] [Revised: 01/07/2009] [Accepted: 01/25/2009] [Indexed: 01/16/2023]
Abstract
Nesfatin-1 is one of the peptide products of posttranslational processing of the nucleobindin-2 (NUCB2) gene, suggested to have physiological relevance to suppress food intake and body weight gain in rats. Nesfatin-1-immunoreactive cells have been found in distinct nuclei in the rat brain related to circuitries regulating food intake. Here, we report novel yet undescribed localization of NUCB2/nesfatin-1 at the mRNA and protein level in the rat central nervous system. Immunohistochemical staining revealed the localization of NUCB2/nesfatin-1 in the piriform and insular cortex, endopiriform nucleus, nucleus accumbens, lateral septum, bed nucleus of stria terminalis, central amygdaloid nucleus, medial preoptic area, dorsal raphe nucleus, ambiguus nucleus, ventrolateral medulla and gigantocellular reticular nucleus, as well as Purkinje-cells of the cerebellum. In the spinal cord, nesfatin-1 immunoreactivity (IR) was found in both sympathetic and parasympathetic preganglionic neuronal groups and in the dorsal area X from lower thoracic to sacral segments. The immunohistochemical results were confirmed by RT-PCR in the central amygdaloid nucleus, nucleus accumbens, cerebellum and lumbar spinal cord microdissected by punch technique. The features and distributions of nesfatin-1 IR and mRNA expression in the brain and spinal cord suggest that NUCB2/nesfatin-1 could play a wider role in autonomic regulation of visceral-endocrine functions besides food intake.
Collapse
Affiliation(s)
- Miriam Goebel
- CURE/Digestive Diseases Research Center, Center for Neurobiology of Stress, Digestive Diseases Division, Department of Medicine, David Geffen School of Medicine, UCLA, VA Greater Los Angeles Health System, Los Angeles, California
| | - Andreas Stengel
- CURE/Digestive Diseases Research Center, Center for Neurobiology of Stress, Digestive Diseases Division, Department of Medicine, David Geffen School of Medicine, UCLA, VA Greater Los Angeles Health System, Los Angeles, California
| | - Nils W.G. Lambrecht
- Membrane Biology Laboratory, Digestive Diseases Division, Department of Medicine, David Geffen School of Medicine, UCLA, VA Greater Los Angeles Health System, Los Angeles, California
| | - Lixin Wang
- CURE/Digestive Diseases Research Center, Center for Neurobiology of Stress, Digestive Diseases Division, Department of Medicine, David Geffen School of Medicine, UCLA, VA Greater Los Angeles Health System, Los Angeles, California
| | - Yvette Taché
- CURE/Digestive Diseases Research Center, Center for Neurobiology of Stress, Digestive Diseases Division, Department of Medicine, David Geffen School of Medicine, UCLA, VA Greater Los Angeles Health System, Los Angeles, California
| |
Collapse
|
93
|
Gonzalez R, Tiwari A, Unniappan S. Pancreatic beta cells colocalize insulin and pronesfatin immunoreactivity in rodents. Biochem Biophys Res Commun 2009; 381:643-8. [PMID: 19248766 DOI: 10.1016/j.bbrc.2009.02.104] [Citation(s) in RCA: 115] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2009] [Accepted: 02/20/2009] [Indexed: 12/16/2022]
Abstract
Nesfatin-1 is a recently discovered feeding inhibitory peptide encoded in the precursor protein, nucleobindin 2 (pronesfatin). Previous studies have shown pronesfatin expression in the brain, stomach and pancreas. However, the identity of cells that express nesfatin in the pancreas remain unknown. The objective of this study was to determine which cells in the pancreas of mice and rats express pronesfatin immunoreactivity. We found pronesfatin immunopositive cells exclusively in the pancreatic islets of both CD1 mice and Fischer 344 rats. Our novel results indicate that the insulin producing beta cells colocalize pronesfatin in the islets of both mice and rats. No colocalization of glucagon and pronesfatin was found in mice, while some glucagon positive cells were positive for pronesfatin in rat islets. The abundant presence of pronesfatin immunoreactivity and its colocalization with insulin suggests a potential role for pronesfatin-derived peptides in islet biology and glucose homeostasis in rodents.
Collapse
Affiliation(s)
- Ronald Gonzalez
- Department of Biology, York University, Toronto, Ont., Canada
| | | | | |
Collapse
|
94
|
Abstract
Nesfatin-1 is one of the peptide products of posttranslational processing of the nucleobindin-2 (NUCB2) gene, suggested to have physiological relevance to suppress food intake and body weight gain in rats. Nesfatin-1-immunoreactive cells have been found in distinct nuclei in the rat brain related to circuitries regulating food intake. Here, we report novel yet undescribed localization of NUCB2/nesfatin-1 at the mRNA and protein level in the rat central nervous system. Immunohistochemical staining revealed the localization of NUCB2/nesfatin-1 in the piriform and insular cortex, endopiriform nucleus, nucleus accumbens, lateral septum, bed nucleus of stria terminalis, central amygdaloid nucleus, medial preoptic area, dorsal raphe nucleus, ambiguus nucleus, ventrolateral medulla and gigantocellular reticular nucleus, as well as Purkinje-cells of the cerebellum. In the spinal cord, nesfatin-1 immunoreactivity (IR) was found in both sympathetic and parasympathetic preganglionic neuronal groups and in the dorsal area X from lower thoracic to sacral segments. The immunohistochemical results were confirmed by RT-PCR in the central amygdaloid nucleus, nucleus accumbens, cerebellum and lumbar spinal cord microdissected by punch technique. The features and distributions of nesfatin-1 IR and mRNA expression in the brain and spinal cord suggest that NUCB2/nesfatin-1 could play a wider role in autonomic regulation of visceral-endocrine functions besides food intake.
Collapse
|
95
|
Abstract
Nesfatin/nucleobindin 2 (NUCB2) is expressed in the appetite-control hypothalamic nuclei and brainstem nuclei. Nesfatin/NUCB2 expression in the paraventricular nucleus of the hypothalamus was modulated by starvation and refeeding. Intracerebroventricular administration of nesfatin-1 dose-dependently inhibited food intake for 6 hours in male Wistar and leptin resistant, Zucker fatty rats. Intraperitoneal administration of nesfatin-1 and its mid-segment (M30) dosedependentlyinhibited food intake for 3 hours in male ICR mice. Intraperitoneal administration of M30 also decreased foodintake in leptin-resistant, genetically obese (ob/ob), diabetic (db/db) mice and mice fed a 45% high fat diet for 28 days. Intraperitoneal administration of M30 increased proopiomelanocortin and cocaine- and amphetamine- related peptide mRNA expression in the nucleus of the solitary tract of mice. In addition, intranasal administration of nesfatin-1 significantly inhibited food intake for 6 hours in male Wistar rats. We summarize recent observations about nesfatin-1, and attempt to present future direction of nesfatin-1 research for developing a new anti-obesity treatment.
Collapse
Affiliation(s)
- Hiroyuki Shimizu
- Department of Medicine and Molecular Science, Gunma University Graduate School of Medicine, Maebashi, Japan.
| | | | | | | |
Collapse
|