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










Publication year range
1.
Cells ; 12(23)2023 12 04.
Article in English | MEDLINE | ID: mdl-38067196

ABSTRACT

Research in the field of pharmacology aims to generate new treatments for pathologies. Nowadays, there are an increased number of chronic disorders that severely and durably handicap many patients. Among the most widespread pathologies, obesity, which is often associated with diabetes, is constantly increasing in incidence, and in parallel, neurodegenerative and mood disorders are increasingly affecting many people. For years, these pathologies have been so frequently observed in the population in a concomitant way that they are considered as comorbidities. In fact, common mechanisms are certainly at work in the etiology of these pathologies. The main purpose of this review is to show the value of anticipating the effect of baseline treatment of a condition on its comorbidity in order to obtain concomitant positive actions. One of the implications would be that by understanding and targeting shared molecular mechanisms underlying these conditions, it may be possible to tailor drugs that address both simultaneously. To this end, we firstly remind readers of the close link existing between depression and diabetes and secondly address the potential benefit of the pleiotropic actions of two major active molecules used to treat central and peripheral disorders, first a serotonin reuptake inhibitor (Prozac ®) and then GLP-1R agonists. In the second part, by discussing the therapeutic potential of new experimental antidepressant molecules, we will support the concept that a better understanding of the intracellular signaling pathways targeted by pharmacological agents could lead to future synergistic treatments targeting solely positive effects for comorbidities.


Subject(s)
Depression , Diabetes Mellitus , Humans , Depression/drug therapy , Diabetes Mellitus/drug therapy , Comorbidity , Antidepressive Agents/pharmacology , Antidepressive Agents/therapeutic use , Mood Disorders/drug therapy
2.
Eur J Med Chem ; 231: 114157, 2022 Mar 05.
Article in English | MEDLINE | ID: mdl-35131536

ABSTRACT

Alpha-linolenic acid (ALA), an essential omega-3 polyunsaturated fatty acid found in plants, exerts neuroprotection and anti-inflammatory effects in chronic and acute CNS disease models. However, the underlying mechanisms are not yet understood. Since ALA is not incorporated into the brain, the observed health benefits may result from some of its metabolites. The putative formation of dihydroxylated ALA derivatives (called linotrins) was recently shown in vitro in the presence of lipoxygenases. However, the in vitro biosynthesis of linotrins was neither stereoselective nor quantitatively efficient for studying their physiological roles as enantiomeric pure forms. Herein, we report the first stereo-controlled synthesis that features regio- and stereoselective hydrometalations of alkynes for assembling the sensitive E,Z,E-conjugated trienes, as well as LC-MS investigations that provide evidence of linotrins occurrence in plants. Moreover, strong anti-inflammatory effects on microglia highlight the potential physiological importance of linotrins and open new perspectives in search of CNS therapeutics.


Subject(s)
Microglia , Oxylipins , Humans , Inflammation/drug therapy , Lipopolysaccharides/pharmacology , Microglia/metabolism , Oxylipins/metabolism , Oxylipins/pharmacology , alpha-Linolenic Acid/metabolism , alpha-Linolenic Acid/pharmacology
3.
Pharmacol Res ; 167: 105539, 2021 05.
Article in English | MEDLINE | ID: mdl-33737242

ABSTRACT

Deterioration of insulin secretion and pancreatic beta-cell mass by inflammatory attacks is one of the main pathophysiological features of type 2 diabetes (T2D). Therefore, preserving beta-cell mass and stimulating insulin secretion only in response to glucose for avoiding the hypoglycemia risks, are the most state-of-the-art option for the treatment of T2D. In this study we tested two correlated hypothesis that 1/ the endogenous peptide released from sortilin, known as PE, that stimulates insulin secretion only in response to glucose, protects beta-cells against death induced by cytokines, and 2/ Spadin and Mini-Spadin, two synthetic peptides derived from PE, that mimic the effects of PE in insulin secretion, also provide beneficial effect on beta-cells survival. We show that PE and its derivatives by inducing a rise of intracellular calcium concentration by depolarizing the membrane protect beta-cells against death induced by Interleukin-1ß. Using biochemical, confocal imaging and cell biology techniques, we reveal that the protective effects of PE and its derivatives rely on the activation of the CaM-Kinase pathway, and on the phosphorylation and activation of the transcription factor CREB. In addition, Mini-Spadin promotes beta-cell proliferation, suggesting its possible regenerative effect. This study highlights new possible roles of PE in pancreatic beta-cell survival and its derivatives as pharmacological tools against diabetes.


Subject(s)
Adaptor Proteins, Vesicular Transport/pharmacology , Cell Survival/drug effects , Cyclic AMP Response Element-Binding Protein/metabolism , Insulin-Secreting Cells/drug effects , Peptides/pharmacology , Adaptor Proteins, Vesicular Transport/chemistry , Animals , Cell Line , Diabetes Mellitus, Type 2/drug therapy , Diabetes Mellitus, Type 2/metabolism , Insulin Secretion/drug effects , Insulin-Secreting Cells/cytology , Insulin-Secreting Cells/metabolism , Peptides/chemistry , Rats , Signal Transduction/drug effects
4.
J Ethnopharmacol ; 263: 113147, 2020 Dec 05.
Article in English | MEDLINE | ID: mdl-32736058

ABSTRACT

ETHNOPHARMACOLOGICAL RELEVANCE: Macaranga Thou. (Euphorbiaceae) is a large genus that comprises over 300 species distributed between Western Africa and the islands of the South Pacific. Plants of this genus have a long-standing history of use in traditional medicine for different purposes, including the treatment of inflammation. Fresh and dried leaves of certain Macaranga species (e.g. M. tanarius (L.) Müll.Arg.), have been used to treat cuts, bruises, boils, swellings, sores and covering of wounds in general. Several reports described Macaranga spp. being a rich source of polyphenols, such as prenylated stilbenoids and flavonoids, mostly responsible for its biological activity. Similarly, an abundant content of prenylated stilbenes was also described in M. siamensis S.J.Davies, species recently identified (2001) in Thailand. While the respective biological activity of the prenylated stilbenes from M. siamensis was poorly investigated to date, our recent study pointed out the interest as the natural source of several novel anti-inflammatory stilbenoids isolated from this species. AIM OF THE STUDY: This work investigated the potential anti-inflammatory effects of the stilbenoid macasiamenene F (MF) isolated from M. siamensis S.J.Davies (Euphorbiaceae) on the lipopolysaccharide (LPS)-induced inflammation-like response of monocytes and microglia, major cells involved in the peripheral and central inflammatory response, respectively. MATERIALS AND METHODS: LPS-induced stimulation of TLR4 signaling led to the activation of inflammatory pathways in in vitro models of THP-1 and THP-1-XBlue™-MD2-CD14 human monocytes, BV-2 mouse microglia, and an ex vivo model of brain-sorted mouse microglia. The ability of the stilbenoid MF to intervene in the IкB/NF-кB and MAPKs/AP-1 inflammatory cascade was investigated. The gene and protein expressions of the pro-inflammatory cytokines IL-1ß and TNF-α were evaluated at the transcription and translation levels. The protective effect of MF against LPS-triggered microglial loss was assessed by cell counting and the LDH assay. RESULTS: MF demonstrated beneficial effects, reducing both monocyte and microglial inflammation as assessed in vitro. It efficiently inhibited the degradation of IкBα, thereby reducing the NF-кB activity and TNF-α expression in human monocytes. Furthermore, the LPS-induced expression of IL-1ß and TNF-α in microglia was dampened by pre-, co-, or post-treatment with MF. In addition to its anti-inflammatory effect, MF demonstrated a cytoprotective effect against the LPS-induced death of BV-2 microglia. CONCLUSION: Our research into anti-inflammatory and protective effects of MF has shown that it is a promising candidate for further in vitro and in vivo investigations of MF interventions with respect to acute and chronic inflammation, including potentially beneficial effects on the inflammatory component of brain diseases such as stroke and Alzheimer's disease.


Subject(s)
Anti-Inflammatory Agents/therapeutic use , Cytoprotection/drug effects , Euphorbiaceae , Microglia/drug effects , Monocytes/drug effects , Prenylation/drug effects , Stilbenes/therapeutic use , Animals , Anti-Inflammatory Agents/isolation & purification , Anti-Inflammatory Agents/pharmacology , Cell Line, Tumor , Cells, Cultured , Cytoprotection/physiology , Dose-Response Relationship, Drug , Humans , Inflammation/drug therapy , Inflammation/metabolism , Inflammation Mediators/antagonists & inhibitors , Inflammation Mediators/metabolism , Lipopolysaccharides/toxicity , Male , Mice , Mice, Inbred C57BL , Microglia/metabolism , Monocytes/metabolism , Plant Extracts/isolation & purification , Plant Extracts/pharmacology , Plant Extracts/therapeutic use , Prenylation/physiology , Stilbenes/isolation & purification , Stilbenes/pharmacology
5.
Neuromolecular Med ; 21(4): 432-444, 2019 12.
Article in English | MEDLINE | ID: mdl-31489567

ABSTRACT

Diabetes affects more than 425 million people worldwide, a scale approaching pandemic proportion. Diabetes represents a major risk factor for stroke, and therefore is actively addressed for stroke prevention. However, how diabetes affects stroke severity has not yet been extensively considered, which is surprising given the evident but understudied common mechanistic features of both pathologies. The increase in number of diabetic people, incidence of stroke in the presence of this specific risk factor, and the exacerbation of ischemic brain damage in diabetic conditions (at least in animal models) warrants the need to integrate this comorbidity in preclinical studies of brain ischemia to develop novel therapeutic approaches. Therefore, a better understanding of the commonalties involved in the course of both diseases would offer the promise of discovering novel neuroprotective pathways that would be more appropriated to clinical scenarios. In this article, we will review the relevant mechanisms that have been identified as common traits of both pathologies and that could be, to our knowledge, potential targets in both pathologies.


Subject(s)
Diabetes Mellitus/physiopathology , Ischemic Stroke/physiopathology , Signal Transduction/physiology , Animals , Brain Damage, Chronic/etiology , Cardiovascular Diseases/epidemiology , Clinical Trials as Topic , Comorbidity , Diabetes Complications/physiopathology , Diabetes Mellitus/drug therapy , Diabetes Mellitus/epidemiology , Diet/adverse effects , Disease Models, Animal , Humans , Hyperlipidemias/epidemiology , Hyperlipidemias/physiopathology , Hypoglycemic Agents/pharmacology , Hypoglycemic Agents/therapeutic use , Ischemic Stroke/drug therapy , Ischemic Stroke/epidemiology , Ischemic Stroke/prevention & control , Metabolic Syndrome/epidemiology , Metabolic Syndrome/physiopathology , Neuroprotective Agents/therapeutic use , Obesity/epidemiology , Prevalence , Risk Factors , Sedentary Behavior , Signal Transduction/drug effects
6.
Front Pharmacol ; 9: 1561, 2018.
Article in English | MEDLINE | ID: mdl-30697159

ABSTRACT

The pharmacological properties and physiological roles of the type I receptor sortilin, also called neurotensin receptor-3, are various and complex. Sortilin is involved in important biological functions from neurotensin and pro-Nerve Growth Factor signaling in the central nervous system to regulation of glucose and lipid homeostasis in the periphery. The peripheral functions of sortilin being less extensively addressed, the focus of the current review is to discuss recent works describing sortilin-induced molecular mechanisms regulating blood glucose homeostasis and insulin signaling. Thus, an overview of several roles ascribed to sortilin in diabetes and other metabolic diseases are presented. Investigations on crucial cellular pathways involved in the protective effect of sortilin receptor on beta cells, including recent discoveries about regulation of cell fate, are also detailed. In addition, we provide a special focus on insulin secretion regulation involving complexes between sortilin and neurotensin receptors. The last section comments on the future research areas which should be developed to address the function of new effectors of the sortilin system in the endocrine apparatus.

7.
Int J Mol Sci ; 17(11)2016 Nov 08.
Article in English | MEDLINE | ID: mdl-27834811

ABSTRACT

The aim of the present review is to unravel the mechanisms of action of the soluble form of the neurotensin (NT) receptor-3 (NTSR3), also called Sortilin, in numerous physiopathological processes including cancer development, cardiovascular diseases and depression. Sortilin/NTSR3 is a transmembrane protein thought to exert multiple functions both intracellularly and at the level of the plasma membrane. The Sortilin/NTSR3 extracellular domain is released by shedding from all the cells expressing the protein. Although the existence of the soluble form of Sortilin/NTSR3 (sSortilin/NTSR3) has been evidenced for more than 10 years, the studies focusing on the role of this soluble protein at the mechanistic level remain rare. Numerous cancer cells, including colonic cancer cells, express the receptor family of neurotensin (NT), and particularly Sortilin/NTSR3. This review aims to summarize the functional role of sSortilin/NTSR3 characterized in the colonic cancer cell line HT29. This includes mechanisms involving signaling cascades through focal adhesion kinase (FAK), a key pathway leading to the weakening of cell-cell and cell-extracellular matrix adhesions, a series of events which could be responsible for cancer metastasis. Finally, some future approaches targeting the release of sNTSR3 through the inhibition of matrix metalloproteases (MMPs) are suggested.


Subject(s)
Adaptor Proteins, Vesicular Transport/genetics , Cardiovascular Diseases/genetics , Colorectal Neoplasms/genetics , Depression/genetics , Focal Adhesion Kinase 1/genetics , Gene Expression Regulation, Neoplastic , Adaptor Proteins, Vesicular Transport/blood , Adaptor Proteins, Vesicular Transport/metabolism , Cardiovascular Diseases/blood , Cardiovascular Diseases/pathology , Cell Adhesion , Cell Communication , Cell Membrane/metabolism , Cell Membrane/pathology , Colorectal Neoplasms/blood , Colorectal Neoplasms/pathology , Depression/blood , Depression/pathology , Focal Adhesion Kinase 1/metabolism , HT29 Cells , Humans , Matrix Metalloproteinases/genetics , Matrix Metalloproteinases/metabolism , Protein Domains , Signal Transduction
8.
J Diabetes Res ; 2016: 3142175, 2016.
Article in English | MEDLINE | ID: mdl-28105440

ABSTRACT

Inhibition of the potassium channels TREK-1 by spadin (SPA) is currently thought to be a promising therapeutic target for the treatment of depression. Since these channels are expressed in pancreatic ß-cells, we investigated their role in the control of insulin secretion and glucose homeostasis. In this study, we confirmed the expression of TREK-1 channels in the insulin secreting MIN6-B1 ß-cell line and in mouse islets. We found that their blockade by SPA potentiated insulin secretion induced by potassium chloride dependent membrane depolarization. Inhibition of TREK-1 by SPA induced a decrease of the resting membrane potential (ΔVm ~ 12 mV) and increased the cytosolic calcium concentration. In mice, administration of SPA enhanced the plasma insulin level stimulated by glucose, confirming its secretagogue effect observed in vitro. Taken together, this work identifies SPA as a novel potential pharmacological agent able to control insulin secretion and glucose homeostasis.


Subject(s)
Calcium/metabolism , Insulin-Secreting Cells/drug effects , Insulin/metabolism , Islets of Langerhans/drug effects , Peptides/pharmacology , Potassium Channels, Tandem Pore Domain/metabolism , Animals , Cell Line , Cytosol/metabolism , Glucose/pharmacology , Insulin Secretion , Insulin-Secreting Cells/metabolism , Islets of Langerhans/metabolism , Male , Membrane Potentials/drug effects , Mice
9.
Genes Cancer ; 5(7-8): 240-249, 2014 Jul.
Article in English | MEDLINE | ID: mdl-25221642

ABSTRACT

The neurotensin (NT) receptor-3 (NTSR3), also called sortilin is a multifunctional protein localized at the intracellular and plasma membrane level. The extracellular domain of NTSR3 (sNTSR3) is released by shedding from several cell lines including colonic cancer cells. This soluble protein acts as an active ligand through its ability to bind, to be internalized in the human adenocarcinoma epithelial HT29 cells and to stimulate the PI3 kinase pathway. The aim of this study was to investigate cellular responses induced by sNTSR3 in HT29 cells. The cellular functions of sNTSR3 were monitored by immunofluocytochemistry, electron microscopy and quantitative PCR in order to characterize the cell shape and the expression of adhesion proteins. We evidenced that sNTSR3 significantly regulates the cellular morphology as well as the cell-cell and the cell-matrix adherens properties by decreasing the expession of several integrins and by modifying the structure of desmosomes. Altogether, these properties lead to an increase of cell detachment upon sNTSR3 treatment on HT29, HCT116 and SW620 cancer cells. Our results indicate that sNTSR3 may induce the first phase of a process which weaken HT29 epithelial properties including desmosome architecture, cell spreading, and initiation of cell separation, all events which could be responsible for cancer metastasis.

10.
Int J Biochem Cell Biol ; 45(5): 952-9, 2013 May.
Article in English | MEDLINE | ID: mdl-23395631

ABSTRACT

The neurotensin (NT) receptor-3 (NTSR3), also called sortilin, is thought to display several functions including a role as a receptor or a co-receptor, in the sorting to plasma membrane and to lysosomes, and in the regulated secretion. The aim of this study was to investigate the function of the soluble form of NTSR3 (sNTSR3) released from several cell lines including colonic cancer cells. The human adenocarcinoma epithelial cell line HT29 has been used to monitor the release, the binding and internalization of sNTSR3 by radioreceptor assays and confocal microscopy. The modulation of the intracellular signaling pathways by the protein has been investigated by using Fura-2 fluorescence calcium imaging microscopy and Western blots analysis. We demonstrated that sNTSR3 specifically binds and internalizes into HT29 cells. This binding, independent from the transactivation of the epidermal growth factor receptor, leads to the increase of intracellular calcium concentration and to the activation of a FAK/Src-dependent activation of the PI3 kinase pathway. In conclusion, sNTSR3 released from the membrane bound NTSR3 is a functional protein able to activate intracellular pathways involved in cell survival but probably not in cell growth.


Subject(s)
Adaptor Proteins, Vesicular Transport/metabolism , Colonic Neoplasms/metabolism , Focal Adhesion Kinase 1/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Calcium/metabolism , Cell Growth Processes/physiology , Colonic Neoplasms/enzymology , ErbB Receptors/metabolism , HT29 Cells , Humans , Phosphatidylinositol 3-Kinases/genetics , Protein Kinase C-alpha/metabolism , Signal Transduction
11.
Cells ; 2(1): 124-35, 2013 Mar 04.
Article in English | MEDLINE | ID: mdl-24709648

ABSTRACT

The neuropeptide, neurotensin, exerts numerous biological functions, including an efficient anti-apoptotic role, both in the central nervous system and in the periphery. This review summarizes studies that clearly evidenced the protective effect of neurotensin through its three known receptors. The pivotal involvement of the neurotensin receptor-3, also called sortilin, in the molecular mechanisms of the anti-apoptotic action of neurotensin has been analyzed in neuronal cell death, in cancer cell growth and in pancreatic beta cell protection. The relationships between the anti-apoptotic role of neurotensin and important physiological and pathological contexts are discussed in this review.

12.
Article in English | MEDLINE | ID: mdl-23230428

ABSTRACT

The pharmacological roles of the neuropeptide neurotensin through its three known receptors are various and complex. Neurotensin is involved in several important biological functions including analgesia and hypothermia in the central nervous system and also food intake and glucose homeostasis in the periphery. This review focuses on recent works dealing with molecular mechanisms regulating blood glucose level and insulin secretion upon neurotensin action. Investigations on crucial cellular components involved in the protective effect of the peptide on beta cells are also detailed. The role of xenin, a neurotensin-related peptide, on the regulation of insulin release by glucose-dependent insulinotropic polypeptide is summarized. The last section comments on the future research areas which should be developed to address the function of new effectors of the neurotensinergic system in the endocrine pancreas.

13.
Biochem Biophys Res Commun ; 414(1): 118-22, 2011 Oct 14.
Article in English | MEDLINE | ID: mdl-21945442

ABSTRACT

Neurotensin (NT) promotes the proliferation of human colonic cancer cells by undefined mechanisms. We already demonstrated that, in the human colon adenocarcinoma cell line HT29, the effects of NT were mediated by a complex formed between the NT receptor-1 (NTSR1) and-3 (NTSR3). Here we examined cellular mechanisms that led to NT-induced MAP kinase phosphorylation and growth factors receptors transactivation in colonic cancer cells and proliferation in HT29 cells. With the aim to identify upstream signaling involved in NT-elicited MAP kinase activation, we found that the stimulatory effects of the peptide were totally independent from the activation of the epidermal growth factor receptor (EGFR) both in the HT29 and the HCT116 cells. NT was unable to promote phosphorylation of EGFR and to compete with EGF for its binding to the receptor. Pharmacological approaches allowed us to differentiate EGF and NT signaling in HT29 cells since only NT activation of Erk1/2 was shown to be sensitive to PKC inhibitors and since only NT increased the intracellular level of calcium. We also observed that NT was not able to transactivate Insulin-like growth factor receptor. Our findings indicate that, in the HT29 and HCT116 cell lines, NT stimulates MAP kinase phosphorylation and cell growth by a pathway which does not involve EGF system but rather NT receptors which transduce their own intracellular effectors. These results indicate that depending on the cell line used, blocking EGFR is not the general rule to inhibit NT-induced cancer cell proliferation.


Subject(s)
Adenocarcinoma/pathology , Colonic Neoplasms/pathology , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Neurotensin/pharmacology , Adaptor Proteins, Vesicular Transport/agonists , Adaptor Proteins, Vesicular Transport/metabolism , Adenocarcinoma/metabolism , Cell Proliferation/drug effects , Colonic Neoplasms/metabolism , Epidermal Growth Factor/pharmacology , ErbB Receptors/agonists , ErbB Receptors/metabolism , HCT116 Cells , HT29 Cells , Humans , Phosphorylation , Receptors, Neurotensin/agonists , Receptors, Neurotensin/metabolism
14.
Int J Biochem Cell Biol ; 42(10): 1681-8, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20601081

ABSTRACT

Neurotensin (NT) is secreted from neurons and gastrointestinal endocrine cells. We previously reported that the three NT receptors (NTSRs) are expressed in pancreatic islets and beta cell lines on which we observed a protective effect of NT against cytotoxic agents. In this study, we explored the role of NT on insulin secretion in the endocrine pancreatic beta cells. We observed that NT stimulates insulin secretion at low glucose level and has a small inhibiting effect on stimulated insulin secretion from isolated islets or INS-1E cells. We studied the mechanisms by which NT elicited calcium concentration changes using fura-2 loaded islets or INS-1E cells. NT increases calcium influx through the opening of cationic channels. Similar calcium influxes were observed after treatment with NTSR selective ligands. NT-evoked calcium regulation involves PKC and the translocation of PKCalpha and PKCepsilon to the plasma membrane. Part of NT effects appears to be also mediated by PKA but not via the Erk pathway. Taken together, these data provide evidence for an important endocrine role of NT in the regulation of the secretory function of beta cells.


Subject(s)
Insulin-Secreting Cells/metabolism , Insulin/biosynthesis , Neurotensin/pharmacology , Protein Kinase C-alpha/metabolism , Protein Kinase C-epsilon/metabolism , Animals , Calcium Signaling/drug effects , Cell Line , Cyclic AMP-Dependent Protein Kinases/genetics , Cyclic AMP-Dependent Protein Kinases/metabolism , Cytoprotection , Enzyme Activation/drug effects , Insulin/genetics , Insulin/metabolism , Insulin Secretion , Insulin-Secreting Cells/cytology , Insulin-Secreting Cells/drug effects , Piperidines/pharmacology , Protein Kinase C-alpha/genetics , Protein Kinase C-epsilon/genetics , Pyrazoles/pharmacology , Quinolines/pharmacology , Rats , Receptors, Neurotensin/agonists
15.
PLoS Biol ; 8(4): e1000355, 2010 Apr 13.
Article in English | MEDLINE | ID: mdl-20405001

ABSTRACT

Current antidepressant treatments are inadequate for many individuals, and when they are effective, they require several weeks of administration before a therapeutic effect can be observed. Improving the treatment of depression is challenging. Recently, the two-pore domain potassium channel TREK-1 has been identified as a new target in depression, and its antagonists might become effective antidepressants. In mice, deletion of the TREK-1 gene results in a depression-resistant phenotype that mimics antidepressant treatments. Here, we validate in mice the antidepressant effects of spadin, a secreted peptide derived from the propeptide generated by the maturation of the neurotensin receptor 3 (NTSR3/Sortilin) and acting through TREK-1 inhibition. NTSR3/Sortilin interacted with the TREK-1 channel, as shown by immunoprecipitation of TREK-1 and NTSR3/Sortilin from COS-7 cells and cortical neurons co-expressing both proteins. TREK-1 and NTSR3/Sortilin were colocalized in mouse cortical neurons. Spadin bound specifically to TREK-1 with an affinity of 10 nM. Electrophysiological studies showed that spadin efficiently blocked the TREK-1 activity in COS-7 cells, cultured hippocampal pyramidal neurons, and CA3 hippocampal neurons in brain slices. Spadin also induced in vivo an increase of the 5-HT neuron firing rate in the Dorsal Raphe Nucleus. In five behavioral tests predicting an antidepressant response, spadin-treated mice showed a resistance to depression as found in TREK-1 deficient mice. More importantly, an intravenous 4-d treatment with spadin not only induced a strong antidepressant effect but also enhanced hippocampal phosphorylation of CREB protein and neurogenesis, considered to be key markers of antidepressant action after chronic treatment with selective serotonin reuptake inhibitors. This work also shows the development of a reliable method for dosing the propeptide in serum of mice by using AlphaScreen technology. These findings point out spadin as a putative antidepressant of new generation with a rapid onset of action. Spadin can be regarded as the first natural antidepressant peptide identified. It corresponds to a new concept to address the treatment of depression.


Subject(s)
Adaptor Proteins, Vesicular Transport/metabolism , Antidepressive Agents/chemistry , Peptides/metabolism , Potassium Channels, Tandem Pore Domain/metabolism , Adaptor Proteins, Vesicular Transport/chemistry , Adaptor Proteins, Vesicular Transport/genetics , Adaptor Proteins, Vesicular Transport/pharmacology , Animals , Antidepressive Agents/metabolism , Antidepressive Agents/therapeutic use , COS Cells , Chlorocebus aethiops , Cyclic AMP Response Element-Binding Protein/metabolism , Depressive Disorder/drug therapy , Drug Design , Humans , Male , Mice , Mice, Inbred C57BL , Motor Activity/drug effects , Patch-Clamp Techniques , Peptides/chemistry , Peptides/genetics , Peptides/pharmacology , Peptides/therapeutic use , Potassium Channel Blockers/metabolism , Potassium Channels, Tandem Pore Domain/antagonists & inhibitors , Potassium Channels, Tandem Pore Domain/genetics , Raphe Nuclei/drug effects , Serotonin/metabolism , Synaptic Transmission/drug effects
16.
Int J Biochem Cell Biol ; 41(12): 2398-402, 2009 Dec.
Article in English | MEDLINE | ID: mdl-19891061

ABSTRACT

The neuropeptide neurotensin (NT) has been recently shown to protect pancreatic beta cells from toxic agents-induced apoptosis through interaction with the NT receptor-2 (NTSR2) and activation of the phosphatidylinositol-3 kinase pathway. However, expression of the NT receptor-3/sortilin (NTSR3) in the mouse pancreatic beta cell line -TC3 led us to investigate its possible functional role in these cells. By using siRNA, immunoprecipitation, co-localization and caspase-3 assays,we provide evidence for a functional endogenous interaction between NTSR2 and NTSR3. Expression of both receptors is necessary for the protective action of NT on staurosporine-induced caspase-3 activity in -TC3 cells. Moreover, NTSR2 and NTSR3 co-immunoprecipitate and are co-localized at the plasma membrane. Thus, the NT response in beta cells is controlled by the formation of a functional complex between NTSR2 and NTSR3.


Subject(s)
Apoptosis/immunology , Insulin-Secreting Cells/metabolism , Neurotensin/metabolism , Receptors, Neurotensin/metabolism , Animals , Caspase 3/metabolism , Cell Line , Cytoprotection , Insulin-Secreting Cells/immunology , Insulin-Secreting Cells/pathology , Mice , Neurotensin/immunology , Phosphatidylinositol 3-Kinases/metabolism , Protein Binding , Protein Transport , RNA, Small Interfering/genetics , Receptors, Neurotensin/genetics , Receptors, Neurotensin/immunology , Signal Transduction , Staurosporine/metabolism
17.
Int J Biochem Cell Biol ; 40(10): 2296-302, 2008.
Article in English | MEDLINE | ID: mdl-18456542

ABSTRACT

The survival of pancreatic beta cells depends on the balance between external cytotoxic and protective molecular systems. The neuropeptide neurotensin (NT) has been shown to regulate certain functions of the endocrine pancreas including insulin and glucagon release. However, the mechanism of action of NT as well as the identification of receptors involved in the pancreatic functions of the peptide remained to be studied. We demonstrate here that NT is an efficient protective agent of pancreatic beta cells against cytotoxic agents. Both beta-TC3 and INS-1E cell lines and the mouse pancreatic islet cells express the three known NT receptors. The incubation of beta cells with NT protects cells from apoptosis induced either by staurosporine or by IL-1beta. In beta-TC3 cells, NT activates both MAP and PI-3 kinases pathways and strongly reduces the staurosporine or the Il-1beta-induced caspase-3 activity by a mechanism involving Akt activation. The NTSR2 agonist levocabastine displays the same protective effect than NT whereas the NTSR1 antagonist is unable to block the effect of NT suggesting the predominant involvement of the NTSR2 in the action of NT on beta cells. These results clearly indicate for the first time that NT is able to protect endocrine beta cells from external cytotoxic agents, a role well correlated with its release in the circulation after a meal.


Subject(s)
Apoptosis/drug effects , Cytoprotection/drug effects , Insulin-Secreting Cells/cytology , Insulin-Secreting Cells/drug effects , Neurotensin/pharmacology , Animals , Caspase 3/metabolism , Cell Line , Gene Expression Regulation/drug effects , Insulin-Secreting Cells/enzymology , Intracellular Space/drug effects , Intracellular Space/metabolism , Mice , Rats , Receptors, Neurotensin/genetics , Receptors, Neurotensin/metabolism , Signal Transduction/drug effects
18.
FEBS J ; 274(21): 5518-32, 2007 Nov.
Article in English | MEDLINE | ID: mdl-17916086

ABSTRACT

The small G protein Rap1 regulates diverse cellular processes such as integrin activation, cell adhesion, cell-cell junction formation and cell polarity. It is crucial to identify Rap1 effectors to better understand the signalling pathways controlling these processes. Krev interaction trapped 1 (Krit1), a protein with FERM (band four-point-one/ezrin/radixin/moesin) domain, was identified as a Rap1 partner in a yeast two-hybrid screen, but this interaction was not confirmed in subsequent studies. As the evidence suggests a role for Krit1 in Rap1-dependent pathways, we readdressed this question. In the present study, we demonstrate by biochemical assays that Krit1 interacts with Rap1A, preferentially its GTP-bound form. We show that, like other FERM proteins, Krit1 adopts two conformations: a closed conformation in which its N-terminal NPAY motif interacts with its C-terminus and an opened conformation bound to integrin cytoplasmic domain associated protein (ICAP)-1, a negative regulator of focal adhesion assembly. We show that a ternary complex can form in vitro between Krit1, Rap1 and ICAP-1 and that Rap1 binds the Krit1 FERM domain in both closed and opened conformations. Unlike ICAP-1, Rap1 does not open Krit1. Using sedimentation assays, we show that Krit1 binds in vitro to microtubules through its N- and C-termini and that Rap1 and ICAP-1 inhibit Krit1 binding to microtubules. Consistently, YFP-Krit1 localizes on cyan fluorescent protein-labelled microtubules in baby hamster kidney cells and is delocalized from microtubules upon coexpression with activated Rap1V12. Finally, we show that Krit1 binds to phosphatidylinositol 4,5-P(2)-containing liposomes and that Rap1 enhances this binding. Based on these results, we propose a model in which Krit1 would be delivered by microtubules to the plasma membrane where it would be captured by Rap1 and ICAP-1.


Subject(s)
Intracellular Signaling Peptides and Proteins/metabolism , Membrane Proteins/metabolism , Microtubule-Associated Proteins/metabolism , Microtubules/metabolism , rap1 GTP-Binding Proteins/metabolism , Adaptor Proteins, Signal Transducing , Amino Acid Motifs , Animals , Binding Sites , Cricetinae , Membranes/metabolism , Microtubule-Associated Proteins/chemistry , Models, Biological , Models, Molecular , Protein Conformation , Transfection , Two-Hybrid System Techniques
19.
J Biol Chem ; 279(17): 17004-12, 2004 Apr 23.
Article in English | MEDLINE | ID: mdl-14739276

ABSTRACT

Arf1 is a small G protein involved in vesicular trafficking, and although it is only distantly related to Ras, it adopts a similar three-dimensional structure. In the present work, we study Arf1 bound to GDP and GTP and its interactions with one of its guanosine nucleotide exchange factors, ARNO-Sec7. The (31)P NMR spectra of Arf1.GDP.Mg(2+) and Arf1.GTP.Mg(2+) share the general features typical for all small G proteins studied so far. Especially, the beta-phosphate resonances of the bound nucleotide are shifted strongly downfield compared with the resonance positions of the free magnesium complexes of GDP and GTP. However, no evidence for an equilibrium between two conformational states of Arf1.GDP.Mg(2+) or Arf1.GTP.Mg(2+) could be observed as it was described earlier for Ras and Ran. Glu(156) of ARNO-Sec7 has been suggested to play as "glutamic acid finger" an important role in the nucleotide exchange mechanism. In the millimolar concentration range used in the NMR experiments, wild type ARNO-Sec7 and ARNO-Sec7(E156D) do weakly interact with Arf1.GDP.Mg(2+) but do not form a strong complex with magnesium-free Arf1.GDP. Only wild type ARNO-Sec7 competes weakly with GDP on Arf1.GDP.Mg(2+) and leads to a release of GDP when added to the solution. The catalytically inactive mutants ARNO-Sec7(E156A) and ARNO-Sec7(E156K) induce a release of magnesium from Arf1.GDP.Mg(2+) but do not promote GDP release. In addition, ARNO-Sec7 does not interact or only very weakly interacts with the GTP-bound form of Arf1, opposite to the observation made earlier for Ran, where the nucleotide exchange factor RCC1 forms a complex with Ran.GTP.Mg(2+) and is able to displace the bound GTP.


Subject(s)
ADP-Ribosylation Factor 1/chemistry , GTPase-Activating Proteins/chemistry , Guanine Nucleotide Exchange Factors/chemistry , Catalysis , Catalytic Domain , Escherichia coli/metabolism , GTPase-Activating Proteins/metabolism , Glutamic Acid/chemistry , Guanine Nucleotide Exchange Factors/metabolism , Guanosine Diphosphate/chemistry , Guanosine Triphosphate/chemistry , Ions , Magnesium/chemistry , Magnetic Resonance Spectroscopy , Nucleotides/chemistry , Phosphorus/chemistry , Protein Binding , Protein Conformation
SELECTION OF CITATIONS
SEARCH DETAIL
...