中文网站正在持续更新中,请密切关注我们康肽生物的最新动态,或点击访问右上角的英文官方网站 www.phoenixpeptide.com
PHOENIX PHARMACEUTICALS, INC. TOP HOME PAGE
Top » Catalog English Version | My Account | 联系我们 | China



 多肽



 标记多肽 



 多肽激素文库



 抗体 



 免疫试剂盒 



 生物标志物阵列 



 多肽样品检测



 自定义肽链合成及GMP



 产品目录索取



 样品准备



 提问和解答


Neuromedin S (NmS)
A Novel anorexigenic hormone and Endogenous Ligand for Orphan GPCR FM-4/TGR-1

Related Products

Identification and functional analysis of a novel ligand for G protein-coupledreceptor, Neuromedin S.

We identified a novel 36-amino acid neuropeptide in rat brain as an endogenous ligand for the G protein-coupled receptors FM-3/GPR66 and FM-4/TGR-1, which were identified to date as the neuromedin U (NMU) receptors, and designated this peptide neuromedin S (NMS) because it was specifically expressed in the suprachiasmatic nucleus (SCN) of the hypothalamus. NMS shared a C-terminal core structure with NMU. NMS mRNA was highly expressed in the central nervous system, spleen and testis. In rat brain, NMS expression was restricted to the ventrolateral portion of the SCN and has a diurnal peak under light/dark cycling, but remains stable under constant darkness. Intracerebroventricular (ICV) administration of NMS in rats induced nonphotic type phase shifts in the circadian rhythm of locomotor activity. ICV injection of NMS also decreased 12-h food intake during the dark period in rats. This anorexigenic effect was more potent than that observed with the same dose of NMU. ICV administration of NMS increased proopiomelanocortin (POMC) mRNA expression in the arcuate nucleus (Arc) and corticotropin-releasing hormone mRNA in the paraventricular nucleus, and induced c-Fos expression in the POMC neurons in the Arc. These findings suggest that NMS is implicated in the regulation of circadian rhythm and feeding behavior.

Miyazato M,  et al. Regul Pept. 2007 Aug 24; [Epub ahead of print].


Novel Role of the Anorexigenic Peptide Neuromedin U in the Control of LH Secretion and its Regulation by Gonadal Hormones and Photoperiod.

Neuromedin U (NMU) is a widely spread neuropeptide, with predominant expression at the gastrointestinal tract and brain, putatively involved in the regulation of a diversity of biological functions, including food intake, energy balance and circadian rhythms; all closely related to reproduction. Yet, the implication of NMU in the control of the gonadotropic axis remains scarcely studied. We report herein analyses on the hypothalamic expression and function of NMU in different physiological and experimental states of rat reproductive system. Expression of NMU mRNA at the hypothalamus was persistently detected along female postnatal development, with maximum levels in adulthood that fluctuated across the cycle and were modulated by ovarian steroids. Acute central administration of NMU evoked increases of serum LH levels in pubertal female rats, while repeated injection of NMU tended to advance vaginal opening. Likewise, central injection of NMU increased serum LH concentrations in cyclic female rats, with peak responses in estrus. In contrast, NMU significantly inhibited pre-elevated LH secretion in gonadectomized and kisspeptin-treated rats. Finally, in acyclic females due to photoperiodic manipulation (constant light), hypothalamic NMU mRNA levels were markedly depressed but relative LH responses to exogenous NMU were significantly augmented. Altogether, our present data support a predominant stimulatory role of NMU in the control of the female gonadotropic axis, which appears under the influence of developmental, hormonal and photoperiodic cues, and might contribute to the joint regulation of energy balance, biological rhythms and reproduction. Key words: Neuromedin U (NMU), Luteinizing hormone (LH), Neuromedin S (NMS), Estrus cycle, Female rat.

Vigo E,  et al. Am J Physiol Endocrinol Metab. 2007 Aug 28; [Epub ahead of print]


Neuromedin S exerts an antidiuretic action in rats.

We recently identified neuromedin S (NMS) as an endogenous ligand for the FM-4/TGR-1 receptor. Here, we examined the possible involvement of central NMS in regulation of urinary output and vasopressin (AVP) release in rats. Intracerebroventricular (icv) injection of NMS induced a dose-dependent increase in the plasma level of AVP, followed by a decrease of nocturnal urinary output. Expression of cFos after icv injection of NMS was observed in the supachiasmatic nucleus (SCN), arcuate nucleus, paraventricular nucleus (PVN), and supraoptic nucleus (SON). The cFos expressing cells in PVN and SON, but not SCN, were then double-stained using antibodies against the vasopressin. On the other hand, icv injection of neuromedin U, which also binds to the FM-4/TGR-1 receptor, required a concentration ten times higher than that of NMS in order to exert the same antidiuretic potency. These results suggest that central NMS may exert a physiological antidiuretic action via vasopressin release.

Sakamoto T,  et al. Biochem Biophys Res Commun. 2007 Sep 21;361(2):457-61. Epub 2007 Jul 18.


Expression of neuromedins S and U and their receptors in the hypothalamus andendocrine glands of the rat.

Neuromedin S (NMS) and neuromedin U (NMU) are regulatory peptides that share the C-terminal amino-acid sequence and act via common G protein-coupled receptors called NMUR1 and NMUR2. Semiquantitative real time-PCR showed that in the rat hypothalamus and testis NMS gene expression was markedly higher than that of the NMU gene, while the reverse occurred in the anterior pituitary and thyroid gland. Low expression of both genes was detected in the thymus, adrenal gland and ovary, whereas in the pancreatic islets only the expression of NMU mRNA was detected. In the rat hypothalamus the expression of the NMUR2 gene was strikingly higher than that of the NMUR1 gene; in contrast, in the testis and ovary the very low expression of NMUR2 contrasted with the relatively high expression of the NMUR1 gene. In the other glands examined only expression of the NMUR1 gene was found. The marked differences in the level of expression of NMU, NMS and their receptors in the hypothalamus and endocrine glands of the rat suggest that in this species such neuromedins may play different roles in the functional regulation of neuroendocrine axes.

Rucinski M,  et al. Int J Mol Med. 2007 Aug;20(2):255-9.


Neuromedin s as novel putative regulator of luteinizing hormone secretion.

Neuromedin S (NMS), a 36 amino acid peptide structurally related to neuromedin U, was recently identified in rat brain as ligand for the G protein-coupled receptor FM4/TGR-1, also termed neuromedin U receptor type-2 (NMU2R). Central expression of NMS appears restricted to the suprachiasmatic nucleus, and NMS has been involved in the regulation of dark-light rhythms and suppression of food intake. Reproduction is known to be tightly regulated by metabolic and photoperiodic cues. Yet the potential contribution of NMS to the control of reproductive axis remains unexplored. We report herein analyses of hypothalamic expression of NMS and NMU2R genes, as well as LH responses to NMS, in different developmental and functional states of the female rat. Expression of NMS and NMU2R genes was detected at the hypothalamus along postnatal development, with significant adulthood). In adult females, hypothalamic expression of NMS (which was confined to suprachiasmatic nucleus) and NMU2R significantly varied during the estrous cycle (maximum at proestrus) and was lowered after ovariectomy and enhanced after progesterone supplementation. Central administration of NMS evoked modest LH secretory responses in pubertal and cyclic females at diestrus, whereas exaggerated LH secretory bursts were elicited by NMS at estrus and after short-term fasting. Conversely, NMS significantly decreased elevated LH concentrations of ovariectomized rats. In summary, we provide herein novel evidence for the ability of NMS to modulate LH secretion in the female rat. Moreover, hypothalamic expression of NMS and NMU2R genes appeared dependent on the functional state of the female reproductive axis. Our data are the first to disclose the potential implication of NMS in the regulation of gonadotropic axis, a function that may contribute to the integration of circadian rhythms, energy balance, and reproduction.

Vigo E,  et al. Endocrinology. 2007 Feb;148(2):813-23. Epub 2006 Nov 16.

Neuromedin S Neuromedin S Nueromedin S

Antibodies are available for Neuromendin S (NMS) amd NMS Prepro (70-103) Immunohistochemistry

Western_Blot NMS NMS
NMS NMS

 

Identification of neuromedin S and its possible role in the mammalian circadian oscillator system

The discovery of neuropeptides has resulted in an increased understanding of novel regulatory mechanisms of certain physiological phenomena. Here we identify a novel neuropeptide of 36 amino-acid residues in rat brain as an endogenous ligand for the orphan G protein-coupled receptor FM-4/TGR-1, which was identified to date as the neuromedin U (NMU) receptor, and designate this peptide 'neuromedin S (NMS)' because it is specifically expressed in the suprachiasmatic nuclei (SCN) of the hypothalamus. NMS shares a C-terminal core structure with NMU. The NMS precursor contains another novel peptide. NMS mRNA is highly expressed in the central nervous system, spleen and testis. In rat brain, NMS expression is restricted to the core of the SCN and has a diurnal peak under light/dark cycling, but remains stable under constant darkness. Intracerebroventricular administration of NMS in rats activates SCN neurons and induces nonphotic type phase shifts in the circadian rhythm of locomotor activity. These findings suggest that NMS in the SCN is implicated in the regulation of circadian rhythms through autocrine and/or paracrine actions.

Mori K, et al. EMBO J. 2005 Jan 06; [Epub ahead of print]

Neuromedin S is a novel anorexigenic hormone

A novel 36-amino acid neuropeptide, neuromedin S (NMS), has recently been identified in rat brain and has been shown to be an endogenous ligand for two orphan G protein-coupled receptors, FM-3/GPR66 and FM-4/TGR-1. These receptors have been identified as neuromedin U (NMU) receptor type-1 and type-2, respectively. In this study, the physiological role of the novel peptide, NMS, on feeding regulation was investigated. Intracerebroventricular (icv) injection of NMS decreased 12-h food intake during the dark period in rats. This anorexigenic effect was more potent and persistent than that observed with the same dose of NMU. Neuropeptide Y, ghrelin and agouti-related protein-induced food intake was counteracted by co-administration of NMS. Icv administration of NMS increased proopiomelanocortin (POMC) mRNA expression in the arcuate nucleus (Arc) and CRH (CRH) mRNA in the paraventricular nucleus (PVN). Pretreatment with SHU9119 (antagonist for -melanocyte-stimulating hormone) and -helical CRF- (9-41) (antagonist for CRH) attenuated NMS-induced suppression of 24 h food intake. After icv injection of NMS, Fos-immunoreactive cells were detected in both the PVN and Arc. When neuronal multiple unit activity was recorded in the PVN before and after icv injection of NMS, a significant increase in firing rate was observed 5 min after administration, and this increase continued for 100 min. These results suggest that the novel peptide, NMS, may be a potent anorexigenic hormone in the hypothalamus, and that expression of POMC mRNA in the Arc and CRH mRNA in the PVN may be involved in NMS action on feeding.

Ida T., et al. Endocrinology 2005, June 23, 10.1210/en.2005-0107

5 4 6
 
NMU receptor type-1 (NMU1R) NMU receptor type-2 (NMU2R)
FM-3/GPR66 FM-4/TGR-1
Peripheral tissues CNS: PVN & SCN
NMU = NMS NMS > NMU
 
1 3 2

5 4 6
 
Neuromedin U Neuromedin S
Brain-gut neuropeptide Neuro & Immunopeptide
Suppression of feeding Circadian rhythm regulation in the SCN
Regulation of energy homeostasis Immune response in the spleen
Elevation of arterial blood pressures and control of local blood flow Spermatogenesis in the testis
 
1 3 2

Related Products

Sequence Neuromedin S

Structure of NMS. (A) Amino-acid sequences of human, rat and mouse prepro-NMS. Identical residues are shaded. The dotted line denotes the predicted signal peptide. The arrowheads indicate proteolytic processing sites. The asterisk shows a glycine residue, which serves as an amide donor for C-terminal amidation. NMS sequences are boxed. Sequences conserved between NMS and NMU are indicated by a solid underline. The sequence data for the human, rat and mouse NMS cDNAs have been submitted to the DDBJ/EMBL/GenBank databases under accession nos. AB164464, AB164465 and AB164466, respectively. (B) Sequence comparison of NMS and NMU. Human, rat and mouse NMS and NMU sequences are aligned. Residues identical between peptides are shaded. Conserved core sequences are indicated by a solid underline. (C) Schematic representation of the preproproteins of rat NMS and NMU. The preproproteins of NMS and NMU are represented by boxes divided into protein domain, proportional to their length. The open and filled arrowheads indicate cleavage sites by signal peptidase and proprotein convertase, respectively. The sequences of the proteolytic processing sites are shown. The basic amino-acid residues recognized by proprotein convertase are underlined. SP, signal peptide.

Mori K, et al. EMBO J. 2005 Jan 06; [Epub ahead of print]

Effects of NMS on rat systemic blood pressure compared with the effects of NMU. (C, D) Rat systemic blood pressure. The indicated amounts of NMS (C) and NMU (D) were administered to rats at the time indicated by the arrow. Both assays were performed as described in "Supplementary materials and methods."

Mori K, et al. EMBO J. 2005 Jan 06; [Epub ahead of print]


Related Products

Pharmacological characterization of synthetic NMS using human FM-3/GPR66 and FM-4/TGR-1 stably expressed in CHO cells.

(A, B) Dose-response relationships of [Ca2+]i change for human NMS (filled circle), human NMU (open circle), rat NMS (filled triangle) and rat NMU (open triangle) in CHO/FM-3 (A) and CHO/FM-4 (B) cells. Data points are meanss.e.m. of triplicates for each experiment. Insets show the time course of [Ca2+]i changes induced by human NMS (solid line) and human NMU (dotted line). Each peptide (10-8 M) was added at the time indicated by the arrow. (C, D) Competitive radioligand binding analysis. [125I-Tyr0]-human NMS binding to FM-3/GPR66 (C) and FM-4/TGR-1 (D) was displaced by increasing concentrations of human NMS (filled circle) and human NMU (open circle). Data were determined in triplicate.

Mori K, et al. EMBO J. 2005 Jan 06; [Epub ahead of print]

neuromedin S

Expression studies of rat NMS.

(A, B) Quantitative RT-PCR analysis of the NMS mRNA in a rat multiple-tissue cDNA panel. Each column represents the means.e.m. of triplicate experiments. LHA, lateral hypothalamic area; VMH, ventromedial hypothalamus; SCN, suprachiasmatic nucleus; ME, median eminence; PVN, paraventricular nucleus; ARC, arcuate nucleus; SON, supraoptic nucleus; DR, dorsal raphe; SN, substantia nigra; LC, locus coeruleus; NTS, nucleus of the solitary tract. (C) Autoradiogram of the NMS mRNA expression in a coronal section of the rat brain. Scale bar, 2 mm. (D-F) Distribution of VIP (D), NMS (E) and AVP (F) mRNA in the rat SCN. Serial sections were used. The SCN is indicated with a dotted line. Scale bar, 500 m.

Mori K, et al. EMBO J. 2005 Jan 06; [Epub ahead of print]

neuromedin S
Related Products

Expression pattern of rat NMS within the SCN.

(A, B) Temporal expression profiles of the NMS mRNA. Animals were maintained under 12-h light/dark cycles (A) or constant darkness for 2 days (B). The amount of NMS mRNA was quantified by in situ hybridization analysis. The experiments were performed side by side. Data represent the meanss.e.m. of 3-4 animals. Open and filled horizontal bars indicate light and dark periods, respectively. (C, D) Responses of NMS expression to light exposure under conditions of constant darkness and to a dark pulse during the light period of a 12-h light/dark cycle. Animals maintained in constant darkness for 2 days were exposed to a light pulse for 30 min at CT6 (C), or animals maintained under 12-h light/dark cycles were exposed to a dark pulse for 30 min at ZT6 (D). Brain samples were collected 1 h after exposure to the light or dark pulse. Data are presented as the meanss.e.m. of 3-4 animals, and are shown as relative changes in NMS mRNA levels.

Mori K, et al. EMBO J. 2005 Jan 06; [Epub ahead of print]


Related Products

In vivo experiments with rat NMS.

(A-D) Phase shifts of the circadian rhythm of locomotor activity induced by ICV administration of NMS. Representative doubled-plotted actograms of locomotor activity are shown. Rats were ICV administered with rat NMS (1 nmol) at CT6 (A), CT12 (B), CT18 (C) and CT23 (D). Each arrowhead indicates the time of NMS administration. Each line shows the regression lines drawn based on the daily onset of locomotor activity before and after administration. (E) Phase-response plot for ICV administration of 1 nmol NMS. The plus and minus values indicate phase advance and delay, respectively. n=3-5. The data points at CT12, 13, 15, 18 and 19 overlap. (F) Dependence of circadian rhythm phase shift on NMS administration. ICV administration of NMS at CT6 and CT23 induced significant phase advance and delay, respectively, in a dose-dependent manner. n=4-5 per group. (G, H) Induction of c-Fos protein expression within the SCN following ICV administration of NMS. Rats were administered at CT6 with 1 nmol NMS (G) or saline (H). 3v, third ventricle. Scale bar, 150 m.

Mori K, et al. EMBO J. 2005 Jan 06; [Epub ahead of print]

NMS

Comparison of food intake after intracerebroventricular administration of NMS and NMU to rats (n = 8-10 in each group).

Each bar and vertical line represents the mean ± SEM. (A) Dark-phase feeding. Food intake of free-feeding rats was examined during a period of 12 h from 1900 h to 0700 h. Each reagent was injected at 1845 h (a, P < 0.05; b, P < 0.01 vs. saline; #, P < 0.01 vs. NMU at the same dose as NMS). (B) The inhibitory effects on food intake after injection. Food intake was examined over a period of 24 h from 1900 h to 1900 h for 3 days after injection. Each reagent (1 nmol) was injected at 1845 h. (a, P < 0.05; b, P < 0.01 vs. saline; £, P < 0.05; §, P < 0.01 vs. NMU at the same dose as NMS). (C) Two-hour food intake in rats that had fasted for 8 h and then received each reagent at 0900 h (b, P < 0.01 vs. saline in fasting rats; #, P < 0.01 vs. NMU at same dose as NMS in fasting rats). (D) Effect of co-administration of NPY (0.5 nmol), ghrelin (0.5 nmol) or AGRP (1 nmol) with NMU or NMS on 2-h food intake in free-feeding rats. Each reagent was injected at 0845 h (a, P < 0.05 vs. NPY + saline; #, P < 0.05 vs. NPY + 1.0 nmol NMU; b, P < 0.01 vs. ghrelin + saline; c, P < 0.05; d, P < 0.01 vs. AGRP + saline).

Ida T., et al. Endocrinology 2005, June 23, 10.1210/en.2005-0107

Involvement of a-MSH and CRH in NMS-elicited feeding behavior.

Quantitative RT-PCR on NMS administered rats (n = 8 per group). Level of POMC mRNA (A) and CRH mRNA (B). (C):Effect of pre-treatments with a-MSH antagonist (SHU9119) or CRH antagonist (a-helical CRF-(9-41)) on food intake reduction by NMS in 8-h fasted rats. Each antagonist was injected at 0745 h, and then NMS was injected at 0845 h. Food intake was examined during a 2-h period from 0900 h to 1100 h. Asterisks indicate the significant difference (P < 0.05). (D): Effect of pre-treatments with a-MSH antagonist (SHU9119) or CRH antagonist (a-helical CRF-(9-41)) on food intake reduction by NMS in intact rats. Each antagonist was injected at 0745 h, and then NMS was injected at 0845 h, then, 24 h food intake was examined. Asterisks indicate the significant difference (P < 0.05).

Ida T., et al. Endocrinology 2005, June 23, 10.1210/en.2005-0107

Related Products

BF Fg

Representative photomicrographs

c-Fos-immunoreactive cell nuclei in selected brain regions after administration of 1 nmol NMS (A, B, C) or NMU (D, E, F). The brain regions shown include the PVN (A, D), Arc (B, E) and SON (C, F). Scale bar = 200 µm (for all panels).

Ida T., et al. Endocrinology 2005, June 23, 10.1210/en.2005-0107

Effect of icv injection of NMS or NMU on multiple-unit activity (MUA) in the PVN.

(A, B): Frequency-time histograms of firing rate of the PVN after administration of NMS or NMU in representative rats. NMS (A) or NMU (B) was injected 15 min (arrows) after stable MUA volley. Abscissa: time (min), ordinate: spike/sec. MUA profiles in one representative rat are shown. (C): Summary of the effects of icv injection of NMS or NMU on MUA volley frequency at 30 min intervals for 2-h. Each column and vertical bar represents the mean ± SEM (n = 3-4) (a, P < 0.05 vs. saline; b, P < 0.01 vs. saline). (D):Location of the MUA electrode tip. Recording sites of MUA volleys were located adjacent to the PVN.

Ida T., et al. Endocrinology 2005, June 23, 10.1210/en.2005-0107

Neuromedin S;%045-84%;%045-85%;%045-86%;%045-87%;%045-88%;%045-89%;%045-90%;%045-91%;%045-92%;%045-93%;%045-94%


分类搜索
关键字搜索
按字母搜索
A B C D E F G H I J K L M N
O P Q R S T U V W X Y Z

Copyright © 2024 PHOENIX BIOTECH