Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 410
Filter
1.
Cell Metab ; 33(7): 1449-1465.e6, 2021 07 06.
Article in English | MEDLINE | ID: mdl-34038712

ABSTRACT

The lymphatic vasculature plays important roles in the physiology of the organs in which it resides, though a clear mechanistic understanding of how this crosstalk is mediated is lacking. Here, we performed single-cell transcriptional profiling of human and mouse adipose tissue and found that lymphatic endothelial cells highly express neurotensin (NTS/Nts). Nts expression is reduced by cold and norepinephrine in an α-adrenergic-dependent manner, suggesting a role in adipose thermogenesis. Indeed, NTS treatment of brown adipose tissue explants reduced expression of thermogenic genes. Furthermore, adenoviral-mediated overexpression and knockdown or knockout of NTS in vivo reduced and enhanced cold tolerance, respectively, an effect that is mediated by NTSR2 and ERK signaling. Inhibition of NTSR2 promoted energy expenditure and improved metabolic function in obese mice. These data establish a link between adipose tissue lymphatics and adipocytes with potential therapeutic implications.


Subject(s)
Endothelial Cells/metabolism , Lymphatic Vessels/cytology , Neurotensin/physiology , Thermogenesis , Animals , Energy Metabolism/drug effects , Energy Metabolism/genetics , Lymphatic Vessels/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Obese , Neurotensin/genetics , Neurotensin/metabolism , Neurotensin/pharmacology , Signal Transduction/genetics , Thermogenesis/drug effects , Thermogenesis/genetics
2.
FASEB J ; 35(5): e21371, 2021 05.
Article in English | MEDLINE | ID: mdl-33811694

ABSTRACT

Studies demonstrate a role for neurotensin (NT) in obesity and related comorbidities. Bile acid (BA) homeostasis alterations are associated with obesity. We determined the effect of NT on BA metabolism in obese and non-obese conditions. Plasma and fecal BA profiles were analyzed by LC-MS/MS in male and female NT+/+ and NT-/- mice fed low-fat (LFD) or high-fat diet (HFD) for 6 weeks (early stage of obesity) or greater than 20 weeks (late stage of obesity). The nuclear farnesoid X receptor (FXR) and BA transporter mRNA expression were assessed in ileum, mouse enteroids, and human cell lines. HFD decreased plasma primary and secondary BAs in NT+/+ mice; HFD-induced decrease of plasma BAs was improved in NT-deficient mice. In NT+/+ mice, HFD inhibited ileal FXR and BA transporter expression; HFD-decreased expression of FXR and BA transporters was prevented in NT-/- mice. Compared with LFD-fed NT+/+ mice, LFD-fed NT-/- mice had relatively lower levels of ileal FXR and BA transporter expression. Moreover, NT stimulates the expression of FXR and BA transporters in Caco-2 cells; however, stimulated expression of BA transporters was attenuated in NT-/- enteroids. Therefore, we demonstrate that HFD disrupts the BA metabolism and ileal FXR and BA transporter axis which are improved in the absence of NT, suggesting that NT contributes to HFD-induced disruption of BA metabolism and plays an inhibitory role in the regulation of ileal FXR and BA transporter signaling under obese conditions. Conversely, NT positively regulates the expression of ileal FXR and BA transporters under non-obese conditions. Therefore, NT plays a dual role in obese and non-obese conditions, suggesting possible therapeutic strategies for obesity control.


Subject(s)
Bile Acids and Salts/metabolism , Intestines/physiology , Neurotensin/physiology , Nutrients/metabolism , Obesity/physiopathology , Receptors, Cytoplasmic and Nuclear/metabolism , Animals , Caco-2 Cells , Diet, High-Fat , Female , Humans , Male , Mice
3.
Neuroendocrinology ; 110(1-2): 35-49, 2020.
Article in English | MEDLINE | ID: mdl-31030196

ABSTRACT

BACKGROUND: Hypothalamic neurotensin (Nts)-secreting neurons regulate fundamental physiological processes including metabolism and feeding. However, the role of Nts in modulation of locomotor activity, sleep, and arousal is unclear. We previously identified and characterized Nts neurons in the zebrafish hypothalamus. MATERIALS AND METHODS: In order to study the role of Nts, nts mutant (nts-/-), and overexpressing zebrafish were generated. RESULTS: The expression of both nts mRNA and Nts protein was reduced during the night in wild-type zebrafish. Behavioral assays revealed that locomotor activity was decreased during both day and night, while sleep was increased exclusively during the nighttime in nts-/- larvae. Likewise, inducible overexpression of Nts increased arousal in hsp70:Gal4/uas:Nts larvae. Furthermore, the behavioral response to light-to-dark transitions was reduced in nts-/- larvae. In order to elucidate potential contenders that may mediate Nts action on these behaviors, we profiled the transcriptome of 6 dpf nts-/- larvae. Among other genes, the expression levels of melanin-concentrating hormone receptor 1b were increased in nts-/- larvae. Furthermore, a portion of promelanin-concentrating hormone 1 (pmch1) and pmch2 neurons expressed the nts receptor. In addition, expression of the the two zebrafish melanin-concentrating hormone (Mch) orthologs, Mch1 and Mch2, was increased in nts-/- larvae. CONCLUSION: These results show that the Nts and Mch systems interact and modulate locomotor activity and arousal.


Subject(s)
Arousal/physiology , Hypothalamic Hormones/metabolism , Locomotion/physiology , Melanins/metabolism , Neurotensin/physiology , Pituitary Hormones/metabolism , Zebrafish Proteins/physiology , Zebrafish/metabolism , Animals , Neurotensin/metabolism , Zebrafish Proteins/metabolism
4.
J Neurosci ; 40(3): 632-647, 2020 01 15.
Article in English | MEDLINE | ID: mdl-31744862

ABSTRACT

The central nucleus of the amygdala plays a significant role in alcohol use and other affective disorders; however, the genetically-defined neuronal subtypes and projections that govern these behaviors are not well known. Here we show that neurotensin neurons in the central nucleus of the amygdala of male mice are activated by in vivo ethanol consumption and that genetic ablation of these neurons decreases ethanol consumption and preference in non-ethanol-dependent animals. This ablation did not impact preference for sucrose, saccharin, or quinine. We found that the most robust projection of the central amygdala neurotensin neurons was to the parabrachial nucleus, a brain region known to be important in feeding behaviors, conditioned taste aversion, and alarm. Optogenetic stimulation of projections from these neurons to the parabrachial nucleus is reinforcing, and increases ethanol drinking as well as consumption of sucrose and saccharin solutions. These data suggest that this central amygdala to parabrachial nucleus projection influences the expression of reward-related phenotypes and is a novel circuit promoting consumption of ethanol and palatable fluids.SIGNIFICANCE STATEMENT Alcohol use disorder (AUD) is a major health burden worldwide. Although ethanol consumption is required for the development of AUD, much remains unknown regarding the underlying neural circuits that govern initial ethanol intake. Here we show that ablation of a population of neurotensin-expressing neurons in the central amygdala decreases intake of and preference for ethanol in non-dependent animals, whereas the projection of these neurons to the parabrachial nucleus promotes consumption of ethanol as well as other palatable fluids.


Subject(s)
Alcohol Drinking/psychology , Central Amygdaloid Nucleus/physiology , Food Preferences/physiology , Neurons/physiology , Neurotensin/physiology , Animals , Anxiety/psychology , Central Amygdaloid Nucleus/cytology , Male , Mice , Mice, Inbred C57BL , Motor Activity/physiology , Neural Pathways/cytology , Neural Pathways/physiology , Optogenetics , Parabrachial Nucleus/cytology , Parabrachial Nucleus/physiology , Patch-Clamp Techniques , Reward , Sweetening Agents , Taste/physiology
5.
Oncogene ; 38(24): 4875-4884, 2019 06.
Article in English | MEDLINE | ID: mdl-30770901

ABSTRACT

Castration-resistant prostate cancer (CRPC) with neuroendocrine differentiation (NED) is a lethal disease for which effective therapies are urgently needed. The mechanism underlying development of CRPC with NED, however, remains largely uncharacterized. In this study, we explored and characterized the functional role of neurotensin (NTS) in cell line and animal models of CRPC with NED. NTS was acutely induced by androgen deprivation in animal models of prostate cancer (PCa) and activated downstream signaling leading to NED through activation of neurotensin receptor 1 (NTSR1) and neurotensin receptor 3 (NTSR3), but not neurotensin receptor 2 (NTSR2). Our findings also revealed the existence of a CK8+/CK14+ subpopulation in the LNCaP cell line that expresses high levels of both NTSR1 and NTSR3, and displays an enhanced susceptibility to develop neuroendocrine-like phenotypes upon treatment with NTS. More importantly, NTSR1 pathway inhibition prevented the development of NED and castration resistance in vivo. We propose a novel role of NTS in the development of CRPC with NED, and a possible strategy to prevent the onset of NED by targeting the NTS signaling pathway.


Subject(s)
Cell Transdifferentiation/genetics , Neuroendocrine Cells/physiology , Neurotensin/physiology , Prostate/pathology , Prostatic Neoplasms, Castration-Resistant/pathology , Receptors, Neurotensin/physiology , Adaptor Proteins, Vesicular Transport/physiology , Animals , Cell Line, Tumor , Humans , Male , Mice , Mice, Inbred BALB C , Prostate/metabolism , Prostatic Neoplasms, Castration-Resistant/genetics , Prostatic Neoplasms, Castration-Resistant/physiopathology
6.
J Reprod Dev ; 65(2): 147-153, 2019 Apr 12.
Article in English | MEDLINE | ID: mdl-30662011

ABSTRACT

Previously, we reported that neurotensin (NT), which is expressed in the uterus and oviduct, enhanced bovine sperm capacitation and acrosome reactions. As NT mRNA expression in bovine oviducts increases dramatically in the follicular phase, we hypothesized that NT modulates fertilization and subsequent conception in cattle. The objective of this study was to evaluate the effect of NT on embryo development and blastocyst quality. The rate of embryo cleavage was significantly increased by the addition of NT to the fertilization medium. Furthermore, the total number of cells and numbers of cells in the inner cell mass of blastocysts were significantly increased by NT during in vitro fertilization (IVF). These results suggested that NT enhanced the efficiency of early bovine embryo development and blastocyst quality. The expression of NT receptors (NTRs) in sperm, testes, oocytes, and cumulus cells was evaluated to determine whether NT acted via NTRs in sperm alone or in both male and female reproductive cells during IVF. Immunocytochemistry and reverse transcription polymerase chain reaction revealed that NTR1 and NTR2 were expressed in sperm and testes, but not in oocytes and cumulus cells. We propose that NT selectively acts upon sperm via NTR1 and NTR2 during IVF to improve the cleavage rate and quality of blastocysts, which are important determinants of sperm quality for successful conception. This research supports our hypothesis that NT acts as a key modulator of fertilization and conception in cattle. Further studies are necessary to apply our findings to the industrial framework of bovine reproduction.


Subject(s)
Blastocyst/cytology , Blastocyst/drug effects , Fertilization in Vitro , Neurotensin/pharmacology , Receptors, Neurotensin/physiology , Spermatozoa/drug effects , Acrosome Reaction/drug effects , Acrosome Reaction/genetics , Animals , Blastocyst/physiology , Cattle/embryology , Cells, Cultured , Embryo Culture Techniques/methods , Embryo Culture Techniques/veterinary , Embryo, Mammalian , Embryonic Development/drug effects , Embryonic Development/genetics , Female , Fertilization/drug effects , Fertilization/genetics , Fertilization in Vitro/veterinary , Male , Neurotensin/metabolism , Neurotensin/physiology , Receptors, Neurotensin/genetics , Receptors, Neurotensin/metabolism , Signal Transduction/drug effects , Signal Transduction/genetics , Sperm Capacitation/drug effects , Sperm Capacitation/genetics , Spermatozoa/physiology
7.
Cell Signal ; 52: 147-154, 2018 12.
Article in English | MEDLINE | ID: mdl-30213686

ABSTRACT

Extracellular signal-regulated kinase (ERK), also known as classical mitogen-activated protein kinase, plays critical roles in cell regulation. ERK is activated through phosphorylation by a cascade of protein kinases including MEK. Various ligands activate the MEK/ERK pathway through receptor-dependent cell signaling. In cultured cells, many ligands such as growth factors, hormones, cytokines and vasoactive peptides elicit transient activation of MEK/ERK, often peaking at ~10 min after the cell treatment. Here, we describe a novel biological event, in which ligand-mediated cell signaling results in the dephosphorylation of MEK/ERK. Neuromedin N and neurotensin, peptides derived from the same precursor polypeptide, elicit cell signaling through the neurotensin receptors. In cultured human pulmonary artery smooth muscle cells (PASMCs), but not in human pulmonary artery endothelial cells (PAECs), we found that both neuromedin N and neurotensin promoted the dephosphorylation of ERK and MEK. Human PASMCs were found to express neurotensin receptor (NTR)-1, -2 and -3, while human PAECs only express NTR3. Neuromedin N-mediated dephosphorylation was suppressed by small chemical inhibitors of protein phosphatase 1/2A and peptidyl-prolyl isomerase. Transmission electron microscopy showed the formation of endocytic vesicles in response to neuromedin N treatment, and dephosphorylation did not occur when sorting nexin 9, a critical regulator of the endocytic vesicle formation, was knocked down. We conclude that neuromedin N and neurotensin elicit a unique dephosphorylation signaling in the MEK/ERK pathway that is regulated by endocytosis. Considering the pathophysiological importance of the MEK/ERK pathway, this discovery of the dephosphorylation mechanism should advance the field of cell signaling.


Subject(s)
Endothelial Cells/metabolism , Myocytes, Smooth Muscle/metabolism , Neurotensin/physiology , Peptide Fragments/physiology , Pulmonary Artery/metabolism , Endothelial Cells/cytology , Humans , Intracellular Signaling Peptides and Proteins/pharmacology , MAP Kinase Signaling System , Myocytes, Smooth Muscle/cytology , Nuclear Proteins , Peptidylprolyl Isomerase/antagonists & inhibitors , Protease Nexins/metabolism , Pulmonary Artery/cytology , RNA-Binding Proteins , Receptors, Neurotensin/metabolism
8.
Biochim Biophys Acta Mol Basis Dis ; 1864(3): 900-916, 2018 Mar.
Article in English | MEDLINE | ID: mdl-29288794

ABSTRACT

The peptide neurotensin (Nts) was discovered within the brain over 40years ago and is implicated in regulating analgesia, body temperature, blood pressure, locomotor activity and feeding. Recent evidence suggests, however, that these disparate processes may be controlled via specific populations of Nts neurons and receptors. The neuronal mediators of Nts anorectic action are now beginning to be understood, and, as such, modulating specific Nts pathways might be useful in treating feeding and body weight disorders. This review considers mechanisms through which Nts normally regulates feeding and how disruptions in Nts signaling might contribute to the disordered feeding and body weight of schizophrenia, Parkinson's disease, anorexia nervosa, and obesity. Defining how Nts specifically mediates feeding vs. other aspects of physiology will inform the design of therapeutics that modify body weight without disrupting other important Nts-mediated physiology.


Subject(s)
Appetite Regulation , Brain/metabolism , Feeding and Eating Disorders/genetics , Neurotensin/physiology , Obesity/genetics , Overweight/genetics , Animals , Appetite Regulation/genetics , Body Weight/genetics , Feeding Behavior/physiology , Feeding and Eating Disorders/metabolism , Humans , Neurotensin/genetics , Neurotensin/metabolism , Obesity/metabolism , Overweight/metabolism
9.
Nat Neurosci ; 20(3): 449-458, 2017 Mar.
Article in English | MEDLINE | ID: mdl-28135243

ABSTRACT

Neural networks that control reproduction must integrate social and hormonal signals, tune motivation, and coordinate social interactions. However, the neural circuit mechanisms for these processes remain unresolved. The medial preoptic area (mPOA), an essential node for social behaviors, comprises molecularly diverse neurons with widespread projections. Here we identify a steroid-responsive subset of neurotensin (Nts)-expressing mPOA neurons that interface with the ventral tegmental area (VTA) to form a socially engaged reward circuit. Using in vivo two-photon imaging in female mice, we show that mPOANts neurons preferentially encode attractive male cues compared to nonsocial appetitive stimuli. Ovarian hormone signals regulate both the physiological and cue-encoding properties of these cells. Furthermore, optogenetic stimulation of mPOANts-VTA circuitry promotes rewarding phenotypes, social approach and striatal dopamine release. Collectively, these data demonstrate that steroid-sensitive mPOA neurons encode ethologically relevant stimuli and co-opt midbrain reward circuits to promote prosocial behaviors critical for species survival.


Subject(s)
Neural Pathways/physiology , Neurotensin/physiology , Preoptic Area/physiology , Reward , Social Behavior , Animals , Corpus Striatum/metabolism , Dopamine/metabolism , Estradiol/pharmacology , Estrous Cycle/physiology , Female , Male , Mice , Mice, Transgenic , Neurons/physiology , Neurotensin/metabolism , Odorants , Preoptic Area/drug effects , Preoptic Area/metabolism , Ventral Tegmental Area/physiology
10.
Oncotarget ; 7(43): 70303-70322, 2016 Oct 25.
Article in English | MEDLINE | ID: mdl-27611941

ABSTRACT

Neurotensin (NTS) is a neuropeptide distributed in central nervous and digestive systems. In this study, the significant association between ectopic NTS expression and tumor invasion was confirmed in hepatocellular carcinoma (HCC). In primary HCC tissues, the NTS and neurotensin receptor 1 (NTR1) co-expression (NTS+NTR1+) is a poor prognostic factor correlated with aggressive biological behaviors and poor clinical prognosis. Enhanced epithelial-to-mesenchymal transition (EMT) features, including decreased E-cadherin, increased ß-catenin translocation and N-cadherin expression, were identified in NTS+NTR1+ HCC tissues. Varied NTS-responsible HCC cell lines were established using NTR1 genetically modified Hep3B and HepG2 cells which were used to elucidate the molecular mechanisms regulating NTS-induced EMT and tumor invasion in vitro. Results revealed that inducing exogenous NTS stimulation and enhancing NTR1 expression promoted tumor invasion rather than proliferation by accelerating EMT in HCC cells. The NTS-induced EMT was correlated with the remarkable increase in Wnt1, Wnt3, Wnt5, Axin, and p-GSK3ß expression and was significantly reversed by blocking the NTS signaling via the NTR1 antagonist SR48692 or by inhibiting the activation of the Wnt/ß-catenin pathway via specific inhibitors, such as TSW119 and DKK-1. SR48692 also inhibited the metastases of NTR1-overexpressing HCC xenografts in the lungs in vivo. This finding implied that NTS may be an important stimulus to promote HCC invasion and metastasis both in vitro and in vivo, and NTS signaling enhanced the tumor EMT and invasion potentials by activating the canonical Wnt/ß-catenin signaling pathway. Therefore, NTS may be a valuable therapeutic target to prevent tumor progression in HCC.


Subject(s)
Carcinoma, Hepatocellular/pathology , Epithelial-Mesenchymal Transition , Liver Neoplasms/pathology , Neurotensin/physiology , Receptors, Neurotensin/physiology , Wnt Signaling Pathway/physiology , Adult , Aged , Cell Line, Tumor , Female , Humans , Male , Middle Aged , Neoplasm Metastasis , Neurotensin/analysis , Receptors, Neurotensin/analysis
11.
Zhongguo Zhong Yao Za Zhi ; 40(13): 2524-36, 2015 Jul.
Article in English | MEDLINE | ID: mdl-26697673

ABSTRACT

As a neuropeptide, neurotensin (NTS) is widely expressed in central and peripheral nervous system, which is mainly mediated byneurotensin receptor1 (NTSR1) to activate the related downstream signaling pathways. After summarized the function and mechanism of NTS/NTSR1 in various malignant tumors, we found that NTS/NTSR1 played essential roles during tumor initiation and development. NTS/NTSR1 regulates tumor initiation, proliferation, apoptosis, metastasis and differentiation mainly through three pathways, including IP3/Ca2+ /PKC/MAPKs pathway, MMPs/EGFR/MAPKs (PI3K/Akt) pathway, or Rho-GTPsaes and non-receptor tyrosine kinase pathway. Besides, NTS/NTSR1 is also regulated by some upstream pathways and some traditional Chinese medicine preparations and traditional Chinese medicine therapies. In this article, we summarized the function of NTS/NTSR1 and its mechanisms, and discussed the prospective in its application to clinical diagnosis and drugs targeting.


Subject(s)
Neoplasms/etiology , Neurotensin/physiology , Receptors, Neurotensin/physiology , Animals , ErbB Receptors/physiology , Humans , Medicine, Chinese Traditional , Neurotensin/chemistry , Receptors, Neurotensin/chemistry , Signal Transduction/physiology , rhoA GTP-Binding Protein/physiology
12.
Curr Pharm Des ; 21(26): 3807-12, 2015.
Article in English | MEDLINE | ID: mdl-26044977

ABSTRACT

There is a wealth of evidence that various neuropeptides and their receptor ligands modulate schizophrenia- related behaviors in preclinical animal models, suggesting that neuropeptide systems may represent potential novel therapeutic targets for the treatment of schizophrenia. In particular, neurotensin and tachykinins have been the subject of significant research efforts, generating compelling preclinical data in the schizophrenia field. However, clinical studies with notably selective tachykinin NK3 receptor antagonists in schizophrenia have been disappointing, and they were unable to confirm the promising therapeutic potential from animal studies, thereby questioning the therapeutic utility of these compounds for this condition. This article reviews preclinical and clinical findings on ligands for neurotensin and tachykinin receptors in schizophrenia, and provides possible explanations for the failure so far to develop small-molecule neuropeptide ligands for the treatment of schizophrenia.


Subject(s)
Antipsychotic Agents/therapeutic use , Receptors, Neurotensin/antagonists & inhibitors , Receptors, Tachykinin/antagonists & inhibitors , Schizophrenia/drug therapy , Animals , Antipsychotic Agents/pharmacology , Humans , Ligands , Neuropeptides , Neurotensin/antagonists & inhibitors , Neurotensin/physiology , Receptors, Neurotensin/physiology , Receptors, Tachykinin/physiology , Schizophrenia/physiopathology , Tachykinins/antagonists & inhibitors , Tachykinins/physiology
13.
Biochem Pharmacol ; 93(4): 461-9, 2015 Feb 15.
Article in English | MEDLINE | ID: mdl-25576685

ABSTRACT

Mesenteric adipose tissue hypertrophy is unique to Crohn's disease while the molecular basis of the crosstalk between MAT and the intestinal inflammation is largely unknown. Telmisartan is an angiotensin II type 1 receptor blocker and a peroxisome proliferator-activated receptor-receptor-γ agonist which has beneficial effects on fat distribution and pro-inflammatory adipokine expression. We evaluated the effect of telmisartan upon mesenteric adipose tissue alterations and inflammatory features in IL-10(-)/(-) mice. We found that treatment with telmisartan significantly ameliorated the severity of colitis in IL-10(-)/(-) mice. Additionally, administration of telmisartan was associated with restoration of mesenteric adipose tissue adipocyte morphology and the expression of adipokines. Furthermore, telmisartan treatment suppressed the neurotensin/microRNA-155 pathway in mesenteric adipose tissue from spontaneous colitis which was confirmed by an in vitro study using cultured mesenteric adipose tissue from Crohn's disease patients. Administration of telmisartan showed promising results in spontaneous colitis which was associated with the attenuated mesenteric adipose tissue alteration which at least in part, was associated with its activity in the regulation of the neurotensin/microRNA-155 pathway. These results support the hypothesis that regulating the abnormal immune response in adipose tissue is an important target for the treatment of Crohn's disease.


Subject(s)
Angiotensin II Type 1 Receptor Blockers/therapeutic use , Benzimidazoles/therapeutic use , Benzoates/therapeutic use , Colitis/drug therapy , Intra-Abdominal Fat/drug effects , MicroRNAs/antagonists & inhibitors , Neurotensin/antagonists & inhibitors , Angiotensin II Type 1 Receptor Blockers/pharmacology , Animals , Benzimidazoles/pharmacology , Benzoates/pharmacology , Colitis/metabolism , Colitis/pathology , Inflammation/drug therapy , Inflammation/metabolism , Inflammation/pathology , Intra-Abdominal Fat/metabolism , Intra-Abdominal Fat/pathology , Male , Mice , Mice, Inbred C3H , Mice, Knockout , MicroRNAs/physiology , Neurotensin/physiology , Telmisartan
14.
Int J Cancer ; 136(6): 1475-81, 2015 Mar 15.
Article in English | MEDLINE | ID: mdl-25098665

ABSTRACT

Wnt/ß-catenin signaling plays a pivotal role in regulating cell growth and differentiation by activation of the ß-catenin/T-cell factor (TCF) complex and subsequent regulation of a set of target genes that have one or more TCF-binding elements (TBEs). Hyperactivation of this pathway has been implicated in numerous malignancies including human neuroendocrine tumors (NETs). Neurotensin (NT), an intestinal hormone, induces proliferation of several gastrointestinal (GI) cancers including cancers of the pancreas and colon. Here, we analyzed the human NT promoter in silico and found at least four consensus TBEs within the proximal promoter region. Using a combination of ChIP and luciferase reporter assays, we identified one TBE (located ∼900 bp proximal from the transcription start site) that was immunoprecipitated efficiently by TCF4-targeting antibody; mutation of this site attenuated the responsiveness to ß-catenin. We also confirmed that the promoter activity and the mRNA and protein expression levels of NT were increased by various Wnt pathway activators and decreased by Wnt inhibitors in NET cell lines BON and QGP-1, which express and secrete NT. Similarly, the intracellular content and secretion of NT were induced by Wnt3a in these cells. Finally, inhibition of NT signaling suppressed cell proliferation and anchorage-independent growth and decreased expression levels of growth-related proteins in NET cells. Our results indicate that NT is a direct target of the Wnt/ß-catenin pathway and may be a mediator for NET cell growth.


Subject(s)
Neuroendocrine Tumors/pathology , Neurotensin/physiology , Wnt Signaling Pathway/physiology , beta Catenin/physiology , Cell Line, Tumor , Cell Proliferation/drug effects , Gene Expression Regulation, Neoplastic , Humans , Neurotensin/antagonists & inhibitors , Neurotensin/genetics , Promoter Regions, Genetic , Receptors, Neurotensin/physiology
15.
Clin Endocrinol (Oxf) ; 79(6): 807-11, 2013 Dec.
Article in English | MEDLINE | ID: mdl-23199168

ABSTRACT

OBJECTIVE: Shift work is associated with circadian rhythm disorder, impaired sleep and behavioural changes, including eating habits, predisposing to obesity and metabolic dysfunctions. It involves a neuro-hormonal dysregulation of appetite towards positive energy balance, including increased ghrelin and decreased leptin, but little is known about other hormones, such as xenin, derived from the upper gut (like ghrelin), and lower gut hormones. Our objective was to compare night workers with day workers in relation to appetite-regulating hormones and other metabolic parameters. DESIGN: Cross-sectional, observational study. PARTICIPANTS: Twenty-four overweight women, divided into night shift workers (n = 12) and day shift workers (n = 12). MEASUREMENTS: BMI, waist circumference, fat mass percentage; diet composition; Pittsburgh Sleep Quality Index; lipids; adipokines; meal tolerance test curves of glucose, insulin, ghrelin, PYY3-36, oxyntomodulin, xenin, GLP-1; insulin sensitivity (Stumvoll index). RESULTS: Night workers, as compared with day workers, had greater body fat mass percentage and tendency to greater waist circumference despite similar BMI; greater energy intake; impaired sleep; lower insulin sensitivity; increased triglycerides and tendency to increased C-reactive protein; similar levels of leptin and other adipokines. Night workers had a blunted post-meal suppression of ghrelin (AUCi(0-60 min) 19·4 ± 139·9 vs -141·9 ± 9·0 ng/ml·60 min, P < 0·01); blunted rise of xenin (AUC(0-180 min) 8690·9 ± 2988·2 vs 28 504·4 ± 20 308·3 pg/ml·180 min, P < 0·01) and similar curves of PYY3-36, oxyntomodulin and GPL-1. CONCLUSION: Compared with day workers within the same BMI range, night workers presented a disrupted control of ghrelin and xenin, associated with behavioural changes in diet and sleep and increased adiposity and related metabolic alterations.


Subject(s)
Appetite Regulation/physiology , Gastrointestinal Hormones/physiology , Ghrelin/physiology , Neurotensin/physiology , Work Schedule Tolerance/physiology , Adiposity/physiology , Adult , Cross-Sectional Studies , Digestive System/physiopathology , Energy Intake/physiology , Female , Gastrointestinal Hormones/blood , Ghrelin/blood , Glucagon-Like Peptide 1/blood , Humans , Insulin Resistance/physiology , Neurotensin/blood , Overweight/blood , Overweight/pathology , Overweight/physiopathology , Oxyntomodulin/blood , Peptide Fragments/blood , Peptide YY/blood , Sleep Disorders, Circadian Rhythm/blood , Sleep Disorders, Circadian Rhythm/pathology , Sleep Disorders, Circadian Rhythm/physiopathology
16.
Nat Rev Drug Discov ; 11(6): 462-78, 2012 May 18.
Article in English | MEDLINE | ID: mdl-22596253

ABSTRACT

The search for novel drugs for treating psychiatric disorders is driven by the growing medical need to improve on the effectiveness and side-effect profile of currently available therapies. Given the wealth of preclinical data supporting the role of neuropeptides in modulating behaviour, pharmaceutical companies have been attempting to target neuropeptide receptors for over two decades. However, clinical studies with synthetic neuropeptide ligands have been unable to confirm the promise predicted by studies in animal models. Here, we analyse preclinical and clinical results for neuropeptide receptor ligands that have been studied in clinical trials for psychiatric diseases, including agents that target the receptors for tachykinins, corticotropin-releasing factor, vasopressin and neurotensin, and suggest new ways to exploit the full potential of these candidate drugs.


Subject(s)
Mental Disorders/drug therapy , Psychotropic Drugs/therapeutic use , Receptors, Neuropeptide/physiology , Animals , Corticotropin-Releasing Hormone/physiology , Drug Delivery Systems , Humans , Ligands , Neurotensin/physiology , Receptors, Corticotropin-Releasing Hormone/physiology , Receptors, Neurotensin/drug effects , Receptors, Neurotensin/physiology , Receptors, Tachykinin/physiology , Receptors, Vasopressin/drug effects , Receptors, Vasopressin/physiology , Tachykinins/physiology , Vasopressins/physiology
17.
Behav Brain Res ; 226(2): 597-600, 2012 Jan 15.
Article in English | MEDLINE | ID: mdl-21946307

ABSTRACT

Tridecapeptide neurotensin (NT) acts as a neurotransmitter and/or neuromodulator and plays a role in learning and reinforcement. The central nucleus of amygdala (CeA), which is relatively rich in NT and neurotensin-1 receptors (NTS1), participates in the regulation of memory and learning mechanisms. The aim of this study was to examine the possible effect of NT and NTS1 antagonist (ANT) on passive avoidance learning after their microinjection into the CeA of male wistar rats. NT significantly increased the latency time. Effect of NT was blocked by ANT pretreatment. ANT in itself had no effect. Our results show that in the rat CeA NT facilitates passive avoidance learning via NTS1.


Subject(s)
Amygdala/physiology , Avoidance Learning/physiology , Neurotensin/physiology , Receptors, Neurotensin/physiology , Amygdala/drug effects , Animals , Avoidance Learning/drug effects , Dose-Response Relationship, Drug , Male , Microinjections , Neurotensin/administration & dosage , Pyrazoles/administration & dosage , Pyrazoles/pharmacology , Quinolines/administration & dosage , Quinolines/pharmacology , Rats , Rats, Wistar , Receptors, Neurotensin/antagonists & inhibitors
18.
Hepatology ; 55(3): 807-20, 2012 Mar.
Article in English | MEDLINE | ID: mdl-21994122

ABSTRACT

UNLABELLED: A novel theory in the field of tumor biology postulates that cancer growth is driven by a population of stem-like cells, called tumor-initiating cells (TICs). We previously identified a TIC population derived from hepatocellular carcinoma (HCC) that is characterized by membrane expression of CD133. Here, we describe a novel mechanism by which these cells mediate tumor growth and angiogenesis by systematic comparison of the gene expression profiles between sorted CD133 liver subpopulations through genome-wide microarray analysis. A significantly dysregulated interleukin-8 (IL-8) signaling network was identified in CD133(+) liver TICs obtained from HCC clinical samples and cell lines. IL-8 was found to be overexpressed at both the genomic and proteomic levels in CD133(+) cells isolated from HCC cell lines or clinical samples. Functional studies found enhanced IL-8 secretion in CD133(+) liver TICs to exhibit a greater ability to self-renew, induce tumor angiogenesis, and initiate tumors. In further support of these observations, IL-8 repression in CD133(+) liver TICs by knockdown or neutralizing antibody abolished these effects. Subsequent studies of the IL-8 functional network identified neurotensin (NTS) and CXCL1 to be preferentially expressed in CD133(+) liver TICs. Addition of exogenous NTS resulted in concomitant up-regulation of IL-8 and CXCL1 with simultaneous activation of p-ERK1/2 and RAF-1, both key components of the mitogen-activated protein kinase (MAPK) pathway. Enhanced IL-8 secretion by CD133(+) liver TICs can in turn activate an IL-8-dependent feedback loop that signals through the MAPK pathway. Further, in its role as a liver TIC marker CD133 also plays a functional part in regulating tumorigenesis of liver TICs by way of regulating NTS, IL-8, CXCL1, and MAPK signaling. CONCLUSION: CD133(+) liver TICs promote angiogenesis, tumorigenesis, and self-renewal through NTS-induced activation of the IL-8 signaling cascade.


Subject(s)
Antigens, CD/metabolism , Cell Proliferation , Chemokine CXCL1/physiology , Glycoproteins/metabolism , Interleukin-8/physiology , Liver Neoplasms/pathology , Neoplastic Stem Cells/physiology , Neovascularization, Pathologic/physiopathology , Neurotensin/physiology , Peptides/metabolism , Signal Transduction/physiology , AC133 Antigen , Animals , Antigens, CD/genetics , Carcinoma, Hepatocellular/blood supply , Carcinoma, Hepatocellular/pathology , Carcinoma, Hepatocellular/surgery , Cell Line, Tumor , Cells, Cultured , Feedback, Physiological , Glycoproteins/deficiency , Glycoproteins/genetics , Hepatectomy , Humans , Interleukin-8/deficiency , Interleukin-8/genetics , Liver/blood supply , Liver/pathology , Liver/surgery , Liver Neoplasms/blood supply , Liver Neoplasms/surgery , Mice , Mice, Nude , Mitogen-Activated Protein Kinase Kinases , Neoplastic Stem Cells/immunology , Neurotensin/pharmacology , Peptides/deficiency , Peptides/genetics , Xenograft Model Antitumor Assays
19.
Biochim Biophys Acta ; 1813(10): 1863-71, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21767580

ABSTRACT

In the last decades some reports reveal the neuropeptide neurotensin (NT) as an immune mediator in the Central Nervous System and in the gastrointestinal tract, however its effects on skin immunity were not identified. The present study investigates the effect of NT on signal transduction and on pro/anti-inflammatory function of skin dendritic cells. Furthermore, we investigated how neurotensin can modulate the inflammatory responses triggered by LPS in skin dendritic cells. We observed that fetal-skin dendritic cells (FSDCs) constitutively express NTR1 and NTR3 (neurotensin receptors) and that LPS treatment induces neurotensin expression. In addition, NT downregulated the activation of the inflammatory signaling pathways NF-κB and JNK, as well as, the expression of the cytokines IL-6, TNF-α, IL-10 and the vascular endothelial growth factor (VEGF), while the survival pathway ERK and epidermal growth factor (EGF) were upregulated. Simultaneous dendritic cells exposure to LPS and NT induced a similar cytokine profile to that one induced by NT alone. However, cells pre-treated with NT and then incubated with LPS, completely changed their cytokine profile, upregulating the cytokines tested, without changes on growth factor expression. Overall, our results could open new perspectives in the design of new therapies for skin diseases, like diabetic wound healing, where neuropeptide exposure seems to be beneficial.


Subject(s)
Dendritic Cells/drug effects , Epidermal Growth Factor/genetics , Inflammation Mediators/metabolism , Neurotensin/pharmacology , Skin/drug effects , Animals , Cell Shape/drug effects , Cell Survival/drug effects , Cells, Cultured , Dendritic Cells/metabolism , Down-Regulation/drug effects , Epidermal Growth Factor/metabolism , Fetus/drug effects , Fetus/immunology , Fetus/metabolism , Gene Expression/drug effects , Lipopolysaccharides/pharmacology , Mice , Neurotensin/genetics , Neurotensin/metabolism , Neurotensin/physiology , Receptors, Neurotensin/genetics , Receptors, Neurotensin/metabolism , Skin/immunology , Skin/metabolism , Up-Regulation/drug effects
20.
J Neurosci Res ; 89(10): 1618-26, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21656844

ABSTRACT

The aim of the present in vivo microdialysis study was to investigate whether the tridecapeptide neurotensin (NT) influences the N-methyl-D-aspartate (NMDA) receptor-mediated increase of cortical glutamate transmission in freely moving rats. Intracortical perfusion with NT influenced local extracellular glutamate levels in a bell-shaped, concentration-dependent manner. One hundred and three hundred nanomolar NT concentrations increased glutamate levels (151% ± 7% and 124% ± 3% of basal values, respectively). Higher (1,000 nM) and lower (10 nM) NT concentrations did not alter extracellular glutamate levels. The NT receptor antagonist SR48692 (100 nM) prevented the NT (100 nM)-induced increase in glutamate levels. NMDA (100 and 500 µM) perfusion induced a concentration-dependent increase in extracellular glutamate levels, the lower 10 µM NMDA concentration being ineffective. When NT (10 nM, a concentration by itself ineffective) was added in combination with NMDA (100 µM) to the perfusion medium, a significant greater increase in extracellular glutamate levels (169% ± 7%) was observed with respect to the increase induced by NMDA (100 µM) alone (139% ± 4%). SR48692 (100 nM) counteracted the increase in glutamate levels induced by the treatment with NT (10 nM) plus NMDA (100 µM). The enhancement of cortical glutamate levels induced by NMDA (100 and 500 µM) was partially antagonized by the presence of SR48692, at a concentration (100 nM) that by itself was ineffective in modulating glutamate release. These findings indicate that NT plays a relevant role in the regulation of cortical glutamatergic transmission, especially by modulating the functional activity of cortical NMDA receptors. A possible role in glutamate-mediated neurotoxicity is suggested.


Subject(s)
Cerebral Cortex/metabolism , Glutamic Acid/metabolism , Neurotensin/physiology , Receptors, N-Methyl-D-Aspartate/physiology , Synaptic Transmission/physiology , Animals , Extracellular Fluid/metabolism , Glutamic Acid/physiology , Glutamic Acid/toxicity , Male , Microdialysis/methods , Rats , Rats, Sprague-Dawley , Receptors, N-Methyl-D-Aspartate/antagonists & inhibitors , Receptors, Neurotensin/antagonists & inhibitors , Receptors, Neurotensin/biosynthesis , Up-Regulation/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...