Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 94
Filter
Add more filters










Publication year range
1.
Biochem Pharmacol ; 183: 114355, 2021 01.
Article in English | MEDLINE | ID: mdl-33279496

ABSTRACT

The therapeutic mechanism of action of methionine aminopeptidase 2 (MetAP2) inhibitors for obesity-diabetes has not yet been fully defined. Xenin, a K-cell derived peptide hormone, possesses an N-terminal Met amino acid residue. Thus, elevated xenin levels could represent a potential pharmacological mechanism of MetAP2 inhibitors, since long-acting xenin analogues have been shown to improve obesity-diabetes. The present study has assessed the ability of the MetAP2 inhibitor, TNP-470, to augment the antidiabetic utility of the incretin-enhancer drug, sitagliptin, in high fat fed (HFF) mice. TNP-470 (1 mg/kg) and sitagliptin (25 mg/kg) were administered once-daily alone, or in combination, to diabetic HFF mice (n = 10) for 18 days. Individual therapy with TNP-470 or sitagliptin resulted in numerous metabolic benefits including reduced blood glucose, increased circulating and pancreatic insulin and improved glucose tolerance, insulin sensitivity, pyruvate tolerance and overall pancreatic islet architecture. Further assessment of metabolic rate revealed that all treatments reduced respiratory exchange ratio and increased locomotor activity. All sitagliptin treated mice also exhibited increased energy expenditure. In addition, treatment with TNP-470 alone, or in combination with sitagliptin, reduced food intake and body weight, as well as elevating plasma and intestinal xenin. Importantly, combined sitagliptin and TNP-470 therapy was associated with further significant benefits beyond that observed by either treatment alone. This included more rapid restoration of normoglycaemia, superior glucose tolerance, increased circulating GIP concentrations and an enhanced pancreatic beta:alpha cell ratio. In conclusion, these data demonstrate that TNP-470 increases plasma and intestinal xenin levels, and augments the antidiabetic advantages of sitagliptin.


Subject(s)
Aminopeptidases/antagonists & inhibitors , Diet, High-Fat/adverse effects , Hypoglycemic Agents/administration & dosage , Metalloendopeptidases/antagonists & inhibitors , Neurotensin/biosynthesis , O-(Chloroacetylcarbamoyl)fumagillol/administration & dosage , Sitagliptin Phosphate/administration & dosage , Aminopeptidases/metabolism , Animals , Blood Glucose/drug effects , Blood Glucose/metabolism , Diabetes Mellitus, Experimental/drug therapy , Diabetes Mellitus, Experimental/metabolism , Male , Metalloendopeptidases/metabolism , Mice , Mice, Inbred C57BL , Up-Regulation/drug effects , Up-Regulation/physiology
2.
Neuropharmacology ; 154: 13-21, 2019 08.
Article in English | MEDLINE | ID: mdl-30266601

ABSTRACT

Animals must ingest water via drinking to maintain fluid homeostasis, yet the neurons that specifically promote drinking behavior are incompletely characterized. The lateral hypothalamic area (LHA) as a whole is essential for drinking behavior but most LHA neurons indiscriminately promote drinking and feeding. By contrast, activating neurotensin (Nts)-expressing LHA neurons (termed LHA Nts neurons) causes mice to immediately drink water with a delayed suppression of feeding. We therefore hypothesized that LHA Nts neurons are sufficient to induce drinking behavior and that these neurons specifically bias for fluid intake over food intake. To test this hypothesis we used designer receptors exclusively activated by designer drugs (DREADDs) to selectively activate LHA Nts neurons and studied the impact on fluid intake, fluid preference and feeding. Activation of LHA Nts neurons stimulated drinking in water-replete and dehydrated mice, indicating that these neurons are sufficient to promote water intake regardless of homeostatic need. Interestingly, mice with activated LHA Nts neurons drank any fluid that was provided regardless of its palatability, but if given a choice they preferred water or palatable solutions over unpalatable (quinine) or dehydrating (hypertonic saline) solutions. Notably, acute activation of LHA Nts neurons robustly promoted fluid but not food intake. Overall, our study confirms that activation of LHA Nts neurons is sufficient to induce drinking behavior and biases for fluid intake. Hence, LHA Nts neurons may be important targets for orchestrating the appropriate ingestive behavior necessary to maintain fluid homeostasis. This article is part of the Special Issue entitled 'Hypothalamic Control of Homeostasis'.


Subject(s)
Drinking/physiology , Hypothalamic Area, Lateral/metabolism , Neurons/metabolism , Neurotensin/biosynthesis , Animals , Choice Behavior/physiology , Gene Expression , Male , Mice , Mice, Transgenic , Neurotensin/genetics
3.
Biol Chem ; 399(1): 79-92, 2017 12 20.
Article in English | MEDLINE | ID: mdl-28822218

ABSTRACT

Modulation of neuropeptide receptors is important for pancreatic ß-cell function. Here, islet distribution and effects of the neurotensin (NT) receptor modulators, xenin and NT, was examined. Xenin, but not NT, significantly improved glucose disposal and insulin secretion, in mice. However, both peptides stimulated insulin secretion from rodent ß-cells at 5.6 mm glucose, with xenin having similar insulinotropic actions at 16.7 mm glucose. In contrast, NT inhibited glucose-induced insulin secretion. Similar observations were made in human 1.1B4 ß-cells and isolated mouse islets. Interestingly, similar xenin levels were recorded in pancreatic and small intestinal tissue. Arginine and glucose stimulated xenin release from islets. Streptozotocin treatment decreased and hydrocortisone treatment increased ß-cell mass in mice. Xenin co-localisation with glucagon was increased by streptozotocin, but unaltered in hydrocortisone mice. This corresponded to elevated plasma xenin levels in streptozotocin mice. In addition, co-localisation of xenin with insulin was increased by hydrocortisone, and decreased by streptozotocin. Further in vitro investigations revealed that xenin and NT protected ß-cells against streptozotocin-induced cytotoxicity. Xenin augmented rodent and human ß-cell proliferation, whereas NT displayed proliferative actions only in human ß-cells. These data highlight the involvement of NT signalling pathways for the possible modulation of ß-cell function.


Subject(s)
Insulin-Secreting Cells/cytology , Neurotensin/biosynthesis , Animals , Apoptosis/drug effects , Calcium/metabolism , Cell Line , Cell Proliferation/drug effects , Cell Survival/drug effects , DNA Damage , Dose-Response Relationship, Drug , Gene Expression Regulation/drug effects , Glucose/pharmacology , Humans , Insulin/metabolism , Insulin Secretion , Insulin-Secreting Cells/drug effects , Insulin-Secreting Cells/metabolism , Intracellular Space/drug effects , Intracellular Space/metabolism , Membrane Potentials/drug effects , Mice , Neurotensin/metabolism
4.
J Control Release ; 220(Pt B): 766-82, 2015 Dec 28.
Article in English | MEDLINE | ID: mdl-26471393

ABSTRACT

Pancreatic cancer is highly aggressive, malignant, and notoriously difficult to cure using conventional cancer therapies. These conventional therapies have significant limitations due to excessive extracellular matrix (ECM) of pancreatic cancer and poor cancer specificity. The excess ECM prevents infiltration of drugs into the inner layer of the solid tumor. Therefore, novel treatment modalities that can specifically target the tumor and degrade the ECM are required for effective therapy. In the present study, we used ECM-degrading and Wnt signal-disrupting oncolytic adenovirus (oAd/DCN/LRP) to achieve a desirable therapeutic outcome against pancreatic cancer. In addition, to overcome the limitations in systemic delivery of oncolytic Ad (oAd) and to specifically target pancreatic cancer, neurotensin peptide (NT)-conjugated polyethylene glycol (PEG) was chemically crosslinked to the surface of Ad, generating a systemically injectable hybrid system, oAd/DCN/LRP-PEG-NT. We tested the targeting and therapeutic efficacy of oAd/DCN/LRP-PEG-NT toward neurotensin receptor 1 (NTR)-overexpressing pancreatic cancer cells, both in vitro and in vivo. The oAd/DCN/LRP-PEG-NT elicited increased NTR-selective cancer cell killing and transduction efficiency when compared with a cognate control lacking NT (oAd/DCN/LRP-PEG). Furthermore, systemic administration of oAd/DCN/LRP-PEG-NT significantly decreased induction of innate and adaptive immune responses against Ad, and blood retention time was markedly prolonged by PEGylation. Moreover, NTR-targeting oAd elicited greater in vivo tumor growth suppression when compared with naked oAd and 9.5 × 10(6)-fold increased tumor-to-liver ratio. This significantly enhanced antitumor effect of oAd/DCN/LRP-PEG-NT was mediated by active viral replication and viral spreading, which was facilitated by ECM degradation and inhibition of Wnt signaling-related factors (Wnt, ß-catenin, and/or vimentin) in the tumor tissues. Taken together, these results demonstrate that oAd/DCN/LRP-PEG-NT has strong therapeutic potential for systemic treatment of NTR-overexpressing pancreatic cancer due to its NTR-targeting ability, enhanced therapeutic efficacy, and safety.


Subject(s)
Adenoviridae/genetics , Decorin/genetics , Genetic Therapy/methods , Neurotensin/genetics , Oncolytic Virotherapy/methods , Oncolytic Viruses/genetics , Pancreatic Neoplasms/therapy , Receptors, Neurotensin/metabolism , Tumor Burden/drug effects , Wnt Signaling Pathway/genetics , Adaptive Immunity , Adenoviridae/growth & development , Adenoviridae/metabolism , Animals , Cell Line, Tumor , Cell Survival , Decorin/biosynthesis , Gene Expression Regulation, Viral , Genetic Therapy/adverse effects , Humans , Immunity, Innate , Male , Mice, Nude , Neurotensin/biosynthesis , Neurotensin/immunology , Oncolytic Virotherapy/adverse effects , Oncolytic Viruses/growth & development , Oncolytic Viruses/immunology , Oncolytic Viruses/metabolism , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/metabolism , Pancreatic Neoplasms/pathology , Pancreatic Neoplasms/virology , Polyethylene Glycols/chemistry , Time Factors , Transduction, Genetic , Viral Load , Virus Replication , Xenograft Model Antitumor Assays
5.
Sci Rep ; 5: 11192, 2015 Jun 08.
Article in English | MEDLINE | ID: mdl-26053252

ABSTRACT

This study assessed the expression, distribution and function of neurotensin (NTs) and two main neurotensin receptors (NTSR), NTSR1 and NTSR2 in normal rat urinary bladders. NTs is primarily located in the suburothelium and the interstitium of smooth muscle bundles. The NTSR1 and NTSR2 receptor subtypes are found to co-localize with smooth muscle cells (SMCs). NTs not only can directly act on bladder SMCs to induce intracellular calcium mobilization by activating the phospholipase C/inositol triphosphate (PLC/IP3) pathway, promoting extracellular calcium influx through a non-selective cation channels, but may be also involved in the modulation of the cholinergic system. Nowadays, the selective antimuscarinic drugs (solifenacin) and the selective beta 3-adrenergic agonist (mirabegron) are used as the first-line pharmacotherapy for overactive bladder (OAB), but without satisfactory treatment benefits in some patients. This study provided evidence suggesting that bladder NTs may play an important role in the regulation of micturition. Further research is needed to investigate the effects of NTs on bladder contractility and the underlying mechanism, which might reveal that the administration of NTSR antagonists can potentially relieve the symptoms of OAB by coordination with antimuscarinic pharmacotherapy.


Subject(s)
Neurotensin/metabolism , Receptors, Neurotensin/metabolism , Urinary Bladder, Overactive/physiopathology , Urinary Bladder/physiology , Animals , Calcium/metabolism , Cholinergic Neurons/metabolism , Female , Muscarinic Antagonists/pharmacology , Muscle Contraction/drug effects , Muscle, Smooth/drug effects , Neurotensin/biosynthesis , Rats , Rats, Sprague-Dawley , Receptors, Neurotensin/antagonists & inhibitors , Receptors, Neurotensin/biosynthesis
6.
Exp Biol Med (Maywood) ; 239(1): 6-12, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24198343

ABSTRACT

Diabetic foot ulcers are an important complication of diabetes mellitus characterized by chronic, non-healing ulcers resulting from poor proliferation and migration of fibroblasts and keratinocytes, thus impairing a correct re-epithelialization of wounded tissues. This healing process can be modulated by neuropeptides released from peripheral nerves; however, little is known regarding the role of neurotensin (NT) as a modulator of human keratinocyte function under hyperglycemic conditions. Therefore, this work is focused on the effect of NT in human keratinocytes, under normal and hyperglycemic conditions at different functional levels, namely NT receptors, cytokine, and growth factor expression, as well as proliferation and migration. Human keratinocyte cells were maintained at either 10/30 mM glucose and treated with or without NT (10 nM). The results show that NT did not affect keratinocyte viability. In addition, NT and all NT receptor expression levels were significantly reduced by hyperglycemia; however, NT treatment stimulated expression of NT and neurotensin receptor 2 (NTR2) while neurotensin receptor 1 (NTR1) and neurotensin receptor 3 (NTR3) expression levels were unchanged. Keratinocyte proliferation was not affected by NT and hyperglycemia, while cell migration was reduced by NT treatment. These results demonstrated that hyperglycemic conditions strongly impaired endogenous NT and NTR2 expression in keratinocytes. Despite the addition of exogenous NT to stimulate the endogenous NT and NTR2 expression, these changes do not translate into functional modifications on keratinocytes, particularly in terms of migration, proliferation, and production of cytokines or growth factors. These results suggest that NT production by keratinocytes may exert a paracrine effect on other skin cells, namely fibroblasts, macrophages, and dendritic cells for correct wound healing.


Subject(s)
Diabetic Foot/metabolism , Keratinocytes/metabolism , Neurotensin/biosynthesis , Paracrine Communication , Receptors, Neurotensin/biosynthesis , Wound Healing , Cell Line , Cell Movement , Cell Proliferation , Cell Survival , Cytokines/metabolism , Diabetic Foot/pathology , Fibroblasts/metabolism , Fibroblasts/pathology , Gene Expression Regulation , Humans , Hyperglycemia/metabolism , Hyperglycemia/pathology , Keratinocytes/pathology , Macrophages/metabolism , Macrophages/pathology
7.
Neurogastroenterol Motil ; 25(1): e70-9, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23095091

ABSTRACT

BACKGROUND: Roux-en-Y gastric bypass (RYGB) surgery is very effective in reducing excess body weight and improving glucose homeostasis in obese subjects. Changes in the pattern of gut hormone secretion are thought to play a major role, but the mechanisms leading to both changed hormone secretion and beneficial effects remain unclear. Specifically, it is not clear whether changes in the number of hormone-secreting enteroendocrine cells, or changes in the releasing stimuli, or both, are important. METHODS: We estimated numbers of enteroendocrine cells after immunohistochemical staining in fixed tissue samples from rats at 10-11 months after RYGB. KEY RESULTS: Numbers of glucagon-like peptide-1 (GLP-1) (L-cells, co-expressing peptide YY (PYY)), cholecystokinin (CCK), neurotensin, and 5-HT-immunoreactive cells were significantly increased in the Roux and common limbs, but not the biliopancreatic limb in RYGB rats compared with sham-operated, obese rats fed high-fat diet, and chow-fed controls. This increase was mostly accounted for by general hyperplasia of all intestinal wall layers of the nutrient-perfused Roux and common limbs, and less to increased density of expression. The number of ghrelin cells in the bypassed stomach was not different among the three groups. CONCLUSIONS & INFERENCES: The findings suggest that the number of enteroendocrine cells increases passively as the gut adapts, and that the increased total number of L- and I-cells is likely to contribute to the higher circulating levels of GLP-1, PYY, and CCK, potentially leading to suppression of food intake and stimulation of insulin secretion. Whether changes in releasing stimuli also contribute to altered circulating levels will have to be determined in future studies.


Subject(s)
Cholecystokinin/biosynthesis , Enteroendocrine Cells/cytology , Gastric Bypass , Glucagon-Like Peptide 1/biosynthesis , Neurotensin/biosynthesis , Serotonin/biosynthesis , Animals , Enteroendocrine Cells/metabolism , Immunohistochemistry , Male , Rats , Rats, Sprague-Dawley
8.
Endocrinology ; 153(12): 5782-95, 2012 Dec.
Article in English | MEDLINE | ID: mdl-23064014

ABSTRACT

Enteroendocrine cells such as duodenal cholecystokinin (CCK cells) are generally thought to be confined to certain segments of the gastrointestinal (GI) tract and to store and release peptides derived from only a single peptide precursor. In the current study, however, transgenic mice expressing enhanced green fluorescent protein (eGFP) under the control of the CCK promoter demonstrated a distribution pattern of CCK-eGFP positive cells that extended throughout the intestine. Quantitative PCR and liquid chromatography-mass spectrometry proteomic analyses of isolated, FACS-purified CCK-eGFP-positive cells demonstrated expression of not only CCK but also glucagon-like peptide 1 (GLP-1), gastric inhibitory peptide (GIP), peptide YY (PYY), neurotensin, and secretin, but not somatostatin. Immunohistochemistry confirmed this expression pattern. The broad coexpression phenomenon was observed both in crypts and villi as demonstrated by immunohistochemistry and FACS analysis of separated cell populations. Single-cell quantitative PCR indicated that approximately half of the duodenal CCK-eGFP cells express one peptide precursor in addition to CCK, whereas an additional smaller fraction expresses two peptide precursors in addition to CCK. The coexpression pattern was further confirmed through a cell ablation study based on expression of the human diphtheria toxin receptor under the control of the proglucagon promoter, in which activation of the receptor resulted in a marked reduction not only in GLP-1 cells, but also PYY, neurotensin, GIP, CCK, and secretin cells, whereas somatostatin cells were spared. Key elements of the coexpression pattern were confirmed by immunohistochemical double staining in human small intestine. It is concluded that a lineage of mature enteroendocrine cells have the ability to coexpress members of a group of functionally related peptides: CCK, secretin, GIP, GLP-1, PYY, and neurotensin, suggesting a potential therapeutic target for the treatment and prevention of diabetes and obesity.


Subject(s)
Cholecystokinin/biosynthesis , Enteroendocrine Cells/cytology , Gastric Inhibitory Polypeptide/biosynthesis , Gene Expression Regulation , Glucagon-Like Peptide 1/biosynthesis , Neurotensin/biosynthesis , Peptide YY/metabolism , Animals , Cell Lineage , Cell Separation , Diabetes Mellitus/prevention & control , Enteroendocrine Cells/metabolism , Flow Cytometry , Ghrelin/metabolism , Green Fluorescent Proteins/metabolism , Humans , Immunohistochemistry/methods , Intestinal Mucosa/metabolism , Mice , Mice, Transgenic , Obesity/prevention & control , Promoter Regions, Genetic
9.
Cancer Biol Ther ; 12(5): 427-35, 2011 Sep 01.
Article in English | MEDLINE | ID: mdl-21725197

ABSTRACT

Neurotensin receptor-1 (NTSR-1) is a G-protein coupled receptor (GPCR) that has been recently identified as a mediator of cancer progression. NTSR-1 and its endogenous ligand, neurotensin (NTS), are co-expressed in several breast cancer cell lines and breast cancer tumor samples. Based on our previously published study demonstrating that intact structured membrane microdomains (SMDs) are required for NTSR-1 mitogenic signaling, we hypothesized that regulated receptor palmitoylation is responsible for NTSR-1 localization and signaling within SMDs upon NTS stimulation. Site-directed mutagenesis and pharmacological strategies were utilized to assess NTRS-1 post-translational modifications in an over-expression cell model (HEK293T) as well as a native breast cancer cell model (MDA-MB-231). NTSR-1 palmitoylation was confirmed by multiple chemical and fluororadiographic methodologies. NTSR-1 glycosylation was confirmed by pharmacological (tunicamycin) and chemical (PGNaseF and O-type glycosidase) approaches. Physiological correlates including cell viability (MTS assay), apoptosis (caspase 3/7 assay) and ERK phosphorylation were utilized to assess the consequences of NTRS-1 palmitoylation. The interaction between palmitoylated NTRS-1 and Gαq/11 within SMDS was confirmed with immunopreciptation analysis of detergent-free isolated fractions of caveolin-rich microdomains. We identified dual-palmitoylation at Cys381 and Cys383 of endogenously-expressed NTSR-1 in MDA-MB-231 breast adeno-carcinomas as well as exogenously-expressed NTSR-1 in HEK293T cells (which do not normally express NTSR-1). Pharmacological inhibition of NTSR-1 palmitoylation in MDA-MB-231 cells as well as NTSR-1-expressing HEK293T cells diminished NTS-mediated ERK 1/2 phosphorylation. Additionally, NTSR-1 mutated at Cys381 and Cys383 showed diminished ERK1/2 stimulation and reduced ability to protect HEK293T cells against apoptosis induced by serum starvation. Mechanistically, mutated C381,383S-NTSR-1 showed reduced ability to interact with Gαq/11 and diminished localization to structured membrane microdomains (SMDs), where Gαq/11 preferentially resides. We also demonstrated that only glycosylated isoforms of NTRS-1 localize within SMDs by palmitotylation. Collectively, our data establish palmitoylation as a novel pharmacological target to inhibit NTSR-1 mitogenic signaling in breast cancer cells.


Subject(s)
Cell Membrane/drug effects , Lipoylation/drug effects , MAP Kinase Signaling System , Neurotensin/pharmacology , Receptors, Neurotensin/metabolism , Apoptosis/drug effects , Breast Neoplasms , Cell Line , Cell Membrane/metabolism , Extracellular Signal-Regulated MAP Kinases/metabolism , Female , GTP-Binding Protein alpha Subunits, Gq-G11/metabolism , Glycosylation/drug effects , Humans , Mutagenesis, Site-Directed , Neurotensin/biosynthesis , Palmitates/pharmacology , Phosphorylation , Receptors, Mitogen/metabolism , Receptors, Neurotensin/genetics , Tunicamycin/pharmacology
10.
Am J Physiol Cell Physiol ; 301(1): C213-26, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21508335

ABSTRACT

The mammalian target of rapamycin (mTOR) signaling exists in two complexes: mTORC1 and mTORC2. Neurotensin (NT), an intestinal hormone secreted by enteroendocrine (N) cells in the small bowel, has important physiological effects in the gastrointestinal tract. The human endocrine cell line BON abundantly expresses the NT gene and synthesizes and secretes NT in a manner analogous to that of N cells. Here, we demonstrate that the inhibition of mTORC1 by rapamycin (mTORC1 inhibitor), torin1 (both mTORC1 and mTORC2 inhibitor) or short hairpin RNA-mediated knockdown of mTOR, regulatory associated protein of mTOR (RAPTOR), and p70 S6 kinase (p70S6K) increased basal NT release via upregulating NT gene expression in BON cells. c-Jun activity was increased by rapamycin or torin1 or p70S6K knockdown. c-Jun overexpression dramatically increased NT promoter activity, which was blocked by PD98059, an mitogen-activated protein kinase kinase (MEK) inhibitor. Furthermore, overexpression of MEK1 or extracellular signal-regulated kinase 1 (ERK1) increased c-Jun expression and NT promoter activity. More importantly, PD98059 blocked rapamycin- or torin1-enhanced NT secretion. Consistently, rapamycin and torin1 also increased NT gene expression in Hep3B cells, a human hepatoma cell line that, similar to BON, expresses high levels of NT. Phosphorylation of c-Jun and ERK1/2 was also increased by rapamycin and torin1 in Hep3B cells. Finally, we showed activation of mTOR in BON cells treated with amino acids, high glucose, or serum and, concurrently, the attenuation of ERK1/2 and c-Jun phosphorylation and NT secretion. Together, mTORC1, as a nutrient sensor, negatively regulates NT secretion via the MEK/ERK/c-Jun signaling pathway. Our results identify a physiological link between mTORC1 and MEK/ERK signaling in controlling intestinal hormone gene expression and secretion.


Subject(s)
Endocrine Cells/metabolism , Mitogen-Activated Protein Kinases/metabolism , Neurotensin/genetics , Neurotensin/metabolism , Proteins/antagonists & inhibitors , Proto-Oncogene Proteins c-jun/metabolism , Signal Transduction , Adaptor Proteins, Signal Transducing/genetics , Cell Line , Extracellular Signal-Regulated MAP Kinases/metabolism , Flavonoids/pharmacology , Gastrointestinal Tract , Gene Expression Regulation , Humans , MAP Kinase Signaling System/drug effects , Mechanistic Target of Rapamycin Complex 1 , Multiprotein Complexes , Naphthyridines/pharmacology , Neurotensin/biosynthesis , Protein Kinase Inhibitors/pharmacology , Proteins/genetics , Proteins/metabolism , RNA Interference , RNA, Small Interfering , Regulatory-Associated Protein of mTOR , Ribosomal Protein S6 Kinases, 70-kDa/genetics , Sirolimus/pharmacology , TOR Serine-Threonine Kinases
11.
J Alzheimers Dis ; 22(3): 819-28, 2010.
Article in English | MEDLINE | ID: mdl-20858966

ABSTRACT

Ghrelin and neurotensin (NTS) are neuroendocrine peptides that exert opposite effects on food intake and energy homeostasis, but share comparable actions in improving memory and learning. Ghrelin and NTS mediate their effects via receptors with high evolutionary identity: two ghrelin G-protein coupled receptors (GPCRs; GHS-R1a/1b) and three NTS-receptors, two GPCRs (NTSR1/2) and one non-GPCR (NTSR3). Because ghrelin and NTS systems are tightly linked to energy balance regulation and cognitive processes, they have been proposed to be altered in Alzheimer's disease (AD), a dementia syndrome markedly influenced by the metabolic status. Although it has been demonstrated that ghrelin and NTS can attenuate AD-related cognitive impairment, a comprehensive analysis of these systems in AD has not been conducted. Here, we used quantitative real time-RT-PCR to analyze expression of the ghrelin/NTS axis in one of the cortical regions most affected in AD, the temporal gyrus. Results unveiled a striking reduction of mRNA levels for ghrelin, and its newly discovered In2-ghrelin variant, as well as for the enzyme responsible for ghrelin acylation, ghrelin-O-acyltransferase and GHS-R1a, while expression of GHS-R1b was markedly increased. In addition, expression levels of NTSR1 and NTSR2 were profoundly decreased in AD, whereas mRNA levels of NTS only declined slightly, and those of NTSR3 (which is involved in neuronal apoptosis) did not vary. Taken together, our results provide the first quantitative evidence showing that ghrelin/NTS systems are markedly altered in the brain of AD patients, thereby suggesting that these systems may contribute to the severe cognitive deficit observed in this pathology.


Subject(s)
Alzheimer Disease/metabolism , Ghrelin/biosynthesis , Neurotensin/biosynthesis , Receptors, Ghrelin/biosynthesis , Receptors, Neurotensin/biosynthesis , Temporal Lobe/metabolism , Aged , Aged, 80 and over , Alzheimer Disease/genetics , Alzheimer Disease/pathology , Cognition Disorders/genetics , Cognition Disorders/metabolism , Cognition Disorders/pathology , Female , Gene Expression Regulation , Ghrelin/genetics , Humans , Male , Neurotensin/genetics , Receptors, Ghrelin/genetics , Receptors, Neurotensin/genetics , Temporal Lobe/pathology
12.
Biol Reprod ; 83(4): 641-7, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20592307

ABSTRACT

Leiomyomas or fibroids are the most frequently diagnosed tumors of the female genital tract, and their growth seems to be steroid-hormone dependent by a yet undetermined cellular and molecular mechanism. Sexual hormones induce the secretion of growth factor peptides and the expression of their receptors, stimulating cell proliferation. One of these factors is neurotensin, and increasing evidence suggests that it can promote growth of different cancer cells. Since there are no data on neurotensin expression in normal and tumoral uterine tissue, we have analyzed the expression of NTS and NTSR1 receptor using immunohistochemistry for protein detection, in situ hybridization to detect cells expressing NTS mRNA, and RT-PCR to detect NTSR1 transcript as well as any of the alternative splice variants recently described for this receptor. We found that NTS and NTSR1 are expressed in connective cells of normal myometrium. In leiomyomas, immunoreactivity for NTS and NTSR1 receptor is colocalized in the smooth muscle cells that are also transcribing NTS. Women receiving high doses of steroids for in vitro fertilization showed tumor growth and increased immunoreactivity for neurotensin and NTSR1 receptor. Interestingly, alternative splice variants of NTSR1 receptor were detected only in tumoral tissue. These findings suggest a role of steroid hormones inducing neurotensin expression in leiomyoma smooth muscle cells. In these cells, NTS could act autocrinally through NTSR1 receptor, promoting their proliferation.


Subject(s)
Leiomyoma/metabolism , Myometrium/metabolism , Neurotensin/biosynthesis , Receptors, Neurotensin/biosynthesis , Uterine Neoplasms/metabolism , Adult , Female , Gene Expression Regulation, Neoplastic , Humans , Immunohistochemistry , In Situ Hybridization , Leiomyoma/genetics , Middle Aged , Neurotensin/genetics , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , RNA, Neoplasm/biosynthesis , RNA, Neoplasm/genetics , Receptors, Neurotensin/genetics , Reverse Transcriptase Polymerase Chain Reaction , Uterine Neoplasms/genetics
13.
Int J Cancer ; 123(8): 1816-23, 2008 Oct 15.
Article in English | MEDLINE | ID: mdl-18661521

ABSTRACT

Distant metastasis is a major factor associated with poor prognosis in head and neck squamous cell carcinomas (HNSCC), but little is known of its molecular mechanisms. New markers that predict clinical outcome, in particular the ability of primary tumors to develop metastatic tumors, are urgently needed. Based on a genome-wide gene expression analysis using clinical specimens of HNSCC, we narrowed our focus to the analysis of the neurotensin (NTS) and neurotensin receptor 1 (NTSR1) oncogenic signal pathways. Kaplan-Meier curves and log rank tests revealed that high mRNA expression levels of NTS and NTSR1 had a significant adverse effect on metastasis-free survival rate, suggesting a contribution of this pathway in HNSCC cancer progression. In HNSCC cells, which expressed NTSR1, a NTS agonist promoted cellular invasion, migration and induction of several mRNAs, such as interleukin 8 and matrix metalloproteinase 1 transcripts. In addition, knock down of NTSR1 expression with small interfering RNAs resulted in reduction of cellular invasion and migration in HNSCC cell lines. Our findings suggest a critical role for the NTS and NTSR1 oncogenic pathways in invasion and migration of HNSCC cells during the metastatic process. Our study raises the possibility that NTS and NTSR1 could be a useful predictive marker of poor prognosis in patients with HNSCC and a molecular therapeutic target in antimetastatic strategies for HNSCCs.


Subject(s)
Carcinoma, Squamous Cell/metabolism , Head and Neck Neoplasms/metabolism , Neurotensin/metabolism , Receptors, Neurotensin/metabolism , Carcinoma, Squamous Cell/genetics , Carcinoma, Squamous Cell/pathology , Cell Line, Tumor , Cell Movement/physiology , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Head and Neck Neoplasms/genetics , Head and Neck Neoplasms/pathology , Humans , Matrix Metalloproteinase 1/biosynthesis , Matrix Metalloproteinase 1/genetics , Matrix Metalloproteinase 12/biosynthesis , Matrix Metalloproteinase 12/genetics , Neoplasm Invasiveness , Neurotensin/biosynthesis , Neurotensin/genetics , Osteopontin/biosynthesis , Osteopontin/genetics , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Receptors, Neurotensin/biosynthesis , Receptors, Neurotensin/genetics , Signal Transduction
15.
Neuroimmunomodulation ; 13(3): 170-8, 2006.
Article in English | MEDLINE | ID: mdl-17183192

ABSTRACT

OBJECTIVE: Intraperitoneal injection of the endotoxin lipopolysaccharide (LPS) produces inflammation accompanied by activation of the immune system and the secretion of cytokines. Cytokines stimulate the hypothalamo-pituitary-adrenal (HPA) axis to release the anti-inflammatory corticosterone which controls its own production by acting on the HPA axis. Upstream in the HPA axis are neuroendocrine corticotrophin-releasing hormone (CRH) neurons located in the paraventricular nucleus (PVN), whose multipeptidergic phenotype changes during inflammation: while CRH mRNA is up-regulated in these conditions, neurotensin (NT) mRNA expression is induced de novo. The negative feedback control of glucocorticoids on CRH production is well documented; however, their action on NT production in the PVN of the hypothalamus is poorly documented. The aim of this study was to determine if glucocorticoids modulate the de novo production of NT during inflammation. METHODS: Using quantitative in situ hybridization histochemistry, we examined whether the absence (adrenalectomy) or excess (corticosterone implants) of glucocorticoids modulate de novo production of NT mRNA in the PVN during inflammation induced by LPS treatment. RESULTS: A relatively low dose of LPS (50 microg/kg) that is not efficient to induce NT mRNA production in the PVN becomes efficient after adrenalectomy. Moreover, corticosterone excess reduces LPS-induced production of NT mRNA in the PVN. CONCLUSION: Glucocorticoids exert a negative control on NT mRNA production in the PVN of the hypothalamus, and this effect requires that NT mRNA production be triggered, such as during inflammation.


Subject(s)
Glucocorticoids/metabolism , Lipopolysaccharides/metabolism , Neurons/metabolism , Neurotensin/biosynthesis , Paraventricular Hypothalamic Nucleus/metabolism , Adrenalectomy , Adrenocorticotropic Hormone/metabolism , Animals , Down-Regulation , Hypothalamo-Hypophyseal System/metabolism , In Situ Hybridization , Inflammation/metabolism , Male , Pituitary-Adrenal System/metabolism , RNA, Messenger/analysis , Rats , Rats, Wistar
16.
Eur J Neurosci ; 24(1): 116-34, 2006 Jul.
Article in English | MEDLINE | ID: mdl-16882012

ABSTRACT

The ventral tegmental area (VTA) is involved in reward-related behaviours and the actions of psychostimulant drugs. It is influenced by afferents expressing a variety of neurotransmitters and neuromodulators; the innervation containing neurotensin is among the densest of these. Intra-VTA neurotensin activates dopaminergic neurons and plays an important role in the development of behavioural sensitization to psychostimulant drugs and possibly in schizophrenia. Using gold-coupled wheatgerm agglutinin as retrograde tracer in combination with nonisotopic in situ hybridization for neurotensin mRNA or neurotensin antibodies after colchicine treatment, the present study was undertaken to demonstrate the neurotensinergic neurons projecting to the VTA and determine whether (and in which subpopulations) neurotensin expression is regulated in VTA-projecting neurons after administrations of the psychostimulant drug methamphetamine or the antipsychotic haloperidol. This study reveals the lateral preoptico-rostral lateral hypothalamic continuum and the medial preoptic area as main sources for the neurotensin afferents of the VTA. Fewer neurotensinergic, VTA-projecting neurons are situated in the dorsal raphe, pedunculopontine and laterodorsal tegmental nuclei, lateral hypothalamic area, ventral endopiriform area, lateral septum, accumbens shell, parabrachial nucleus and different parts of the extended amygdala. The number of neurotensinergic VTA-projecting neurons increased significantly only after methamphetamine administration and exclusively in the accumbens shell. It is concluded that the widespread neurotensinergic VTA-projecting neurons, situated in areas involved in different reward-related behaviours, are well suited to convey distinct reward information to the VTA. The up-regulation of neurotensin expression selectively in VTA-projecting neurons in the accumbens shell following methamphetamine administration may be an important factor in the development of behavioural sensitization.


Subject(s)
Afferent Pathways/metabolism , Antipsychotic Agents/pharmacology , Central Nervous System Stimulants/pharmacology , Neurotensin/metabolism , Ventral Tegmental Area/physiology , Afferent Pathways/drug effects , Animals , Colchicine/pharmacology , Haloperidol/pharmacology , Immunohistochemistry , In Situ Hybridization , Male , Methamphetamine/pharmacology , Neurotensin/biosynthesis , Neurotensin/genetics , RNA, Messenger/biosynthesis , Rats , Rats, Sprague-Dawley , Ventral Tegmental Area/anatomy & histology
17.
J Psychiatry Neurosci ; 31(4): 229-45, 2006 Jul.
Article in English | MEDLINE | ID: mdl-16862241

ABSTRACT

The peptide neurotensin has been studied for more than 30 years. Although it is widely distributed in the central and peripheral nervous systems, neurotensin has been more intensely studied with regard to its interactions with the central dopamine system. A number of claims have been made regarding its possible implication in many diseases of the central nervous system, including schizophrenia. In this review, we describe briefly the basic biology of this neuropeptide, and then we consider the strengths and the weaknesses of the data that suggest a role for neurotensin in schizophrenia, drug abuse, Parkinson's disease, pain, central control of blood pressure, eating disorders, cancer, neurodegenerative disorders and inflammation.


Subject(s)
Central Nervous System Diseases/physiopathology , Central Nervous System/physiopathology , Neurotensin/physiology , Animals , Biogenic Monoamines/metabolism , Biogenic Monoamines/physiology , Humans , Neurotensin/biosynthesis , Pain/physiopathology , Parkinson Disease/physiopathology , Schizophrenia/physiopathology , Substance-Related Disorders/physiopathology , Synaptic Transmission
18.
Cancer Res ; 66(12): 6243-9, 2006 Jun 15.
Article in English | MEDLINE | ID: mdl-16778199

ABSTRACT

Emerging evidence supports neurotensin as a trophic and antiapoptotic factor, mediating its control via the high-affinity neurotensin receptor (NT1 receptor) in several human solid tumors. In a series of 51 patients with invasive ductal breast cancers, 34% of all tumors were positive for neurotensin and 91% positive for NT1 receptor. We found a coexpression of neurotensin and NT1 receptor in a large proportion (30%) of ductal breast tumors, suggesting a contribution of the neurotensinergic signaling cascade within breast cancer progression. Functionally expressed NT1 receptor, in the highly malignant MDA-MB-231 human breast cancer cell line, coordinated a series of transforming functions, including cellular migration, invasion, induction of the matrix metalloproteinase (MMP)-9 transcripts, and MMP-9 gelatinase activity. Disruption of NT1 receptor signaling by silencing RNA or use of a specific NT1 receptor antagonist, SR48692, caused the reversion of these transforming functions and tumor growth of MDA-MB-231 cells xenografted in nude mice. Our findings support the contribution of neurotensin in human breast cancer progression and point out the utility to develop therapeutic molecules targeting neurotensin or NT1 receptor signaling cascade. These strategies would increase the range of therapeutic approaches and be beneficial for specific patients.


Subject(s)
Adenocarcinoma/metabolism , Adenocarcinoma/pathology , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Neurotensin/biosynthesis , Receptors, Neurotensin/biosynthesis , Animals , Carcinoma in Situ/metabolism , Carcinoma in Situ/pathology , Carcinoma, Ductal, Breast/metabolism , Carcinoma, Ductal, Breast/pathology , Cell Growth Processes/physiology , Cell Line, Tumor , Cell Movement/physiology , Disease Progression , Enzyme Activation , Female , Humans , Immunohistochemistry , Matrix Metalloproteinase 9/metabolism , Mice , Middle Aged , Neoplasm Invasiveness , Transplantation, Heterologous
19.
J Neurosci ; 25(41): 9497-506, 2005 Oct 12.
Article in English | MEDLINE | ID: mdl-16221860

ABSTRACT

Neurotensin (NT) is implicated in the regulation of energy homeostasis, in addition to its many described physiological functions. NT is postulated to mediate, in part, the effects of leptin in the hypothalamus. We generated clonal, immortalized hypothalamic cell lines, N-39 and N-36/1, which are the first representative NT-expressing cell models available for the investigation of NT gene regulation and control mechanisms. The cell lines express the Ob-Rb leptin receptor neuropeptide Y (NPY)-Y1, Y2, Y4, Y5 receptors, melanocortin 4 receptor, insulin receptor, and the NT receptor. NT mRNA levels are induced by approximately 1.5-fold to twofold with leptin, insulin, and alpha-melanocyte stimulating hormone treatments but not by NPY. Leptin-mediated induction of NT gene expression was biphasic at 10(-11) and 10(-7) M. The leptin responsive region was localized to within -381 to -250 bp of the 5' regulatory region of the NT gene. Furthermore, we demonstrated direct leptin-mediated signal transducers and activators of transcription (STAT) binding to this region at 10(-11) m, but not 10(-7) m leptin, in chromatin precipitation assays. Leptin-induced NT regulation was attenuated by dominant-negative STAT3 protein expression. These data support the hypothesis that NT may have a direct role in the neuroendocrine control of feeding and energy homeostasis.


Subject(s)
Gene Expression Regulation/physiology , Insulin/physiology , Leptin/physiology , Models, Neurological , Neurotensin/biosynthesis , Neurotensin/genetics , alpha-MSH/physiology , Animals , Anorexia/genetics , Anorexia/metabolism , Cell Line, Transformed , Cells, Cultured , Clone Cells , Hypothalamus/drug effects , Hypothalamus/metabolism , Mice , Neurotensin/physiology
20.
Hear Res ; 205(1-2): 1-6, 2005 Jul.
Article in English | MEDLINE | ID: mdl-15953510

ABSTRACT

The distribution of neurotensin-containing cell bodies and fibers has been observed in the central and peripheral nervous system, including sensory ganglia, but no description has been found in the peripheral auditory system. Here, we investigated the presence of neurotensin immunoreactivity in the cochlea of the adult Wistar rat. Strong neurotensin immunoreactivity was detected in the cytoplasm of the inner hair cells (IHC) and Deiters' cells of the organ of Corti. Outer hair cells (OHC) show weak immunoreaction. Neurotensin immunoreactivity was also found in the neurons and fibers of the spiral ganglia. Quantitative microdensitometric image analysis of the neurotensin immunoreactivity showed a strong immunoreaction in the hair cells of organ of Corti and a moderate to strong labeling in the spiral ganglion neurons. A series of double immunolabeling experiments demonstrated a strong neurotensin immunoreactivity in the parvalbumin immunoreactive IHC and also in the calbindin immunoreactive Deiters' cells. Weak neurotensin immunoreactivity was seen in the calbindin positive OHC. Neurofilament and parvalbumin immunoreactive neurons and fibers in the spiral ganglia showed neurotensin immunoreactivity. Calbindin immunoreactivity was not detected in the spiral ganglion neurons, which are labeled by neurotensin immunoreactivity. The presence of neurotensin in the cochlea may be related to its modulation of neurotransmission in the peripheral auditory pathway.


Subject(s)
Neurons/chemistry , Neurotensin/analysis , Organ of Corti/chemistry , Animals , Hair Cells, Auditory/chemistry , Hair Cells, Auditory/immunology , Image Processing, Computer-Assisted , Immunohistochemistry , Male , Microscopy, Confocal , Microscopy, Fluorescence , Neurons/immunology , Neurotensin/biosynthesis , Neurotensin/immunology , Organ of Corti/immunology , Rats , Rats, Wistar , Specific Pathogen-Free Organisms , Spiral Ganglion/chemistry , Spiral Ganglion/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...