Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
Add more filters










Publication year range
1.
iScience ; 26(11): 108110, 2023 Nov 17.
Article in English | MEDLINE | ID: mdl-37860691

ABSTRACT

In neuropathic pain, recent evidence has highlighted a sex-dependent role of the P2X4 receptor in spinal microglia in the development of tactile allodynia following nerve injury. Here, using internalization-defective P2X4mCherryIN knockin mice (P2X4KI), we demonstrate that increased cell surface expression of P2X4 induces hypersensitivity to mechanical stimulations and hyperexcitability in spinal cord neurons of both male and female naive mice. During neuropathy, both wild-type (WT) and P2X4KI mice of both sexes develop tactile allodynia accompanied by spinal neuron hyperexcitability. These responses are selectively associated with P2X4, as they are absent in global P2X4KO or myeloid-specific P2X4KO mice. We show that P2X4 is de novo expressed in reactive microglia in neuropathic WT and P2X4KI mice of both sexes and that tactile allodynia is relieved by pharmacological blockade of P2X4 or TrkB. These results show that the upregulation of P2X4 in microglia is crucial for neuropathic pain, regardless of sex.

2.
Cell Mol Life Sci ; 80(5): 138, 2023 May 05.
Article in English | MEDLINE | ID: mdl-37145189

ABSTRACT

Numerous evidences support that microglia contributes to the progression of Alzheimer's disease. P2X4 receptors are ATP-gated channels with high calcium permeability, which are de novo expressed in a subset of reactive microglia associated with various pathological contexts, contributing to microglial functions. P2X4 receptors are mainly localized in lysosomes and trafficking to the plasma membrane is tightly regulated. Here, we investigated the role of P2X4 in the context of Alzheimer's disease (AD). Using proteomics, we identified Apolipoprotein E (ApoE) as a specific P2X4 interacting protein. We found that P2X4 regulates lysosomal cathepsin B (CatB) activity promoting ApoE degradation; P2rX4 deletion results in higher amounts of intracellular and secreted ApoE in both bone-marrow-derived macrophage (BMDM) and microglia from APPswe/PSEN1dE9 brain. In both human AD brain and APP/PS1 mice, P2X4 and ApoE are almost exclusively expressed in plaque-associated microglia. In 12-month-old APP/PS1 mice, genetic deletion of P2rX4 reverses topographical and spatial memory impairment and reduces amount of soluble small aggregates of Aß1-42 peptide, while no obvious alteration of plaque-associated microglia characteristics is observed. Our results support that microglial P2X4 promotes lysosomal ApoE degradation, indirectly altering Aß peptide clearance, which in turn might promotes synaptic dysfunctions and cognitive deficits. Our findings uncover a specific interplay between purinergic signaling, microglial ApoE, soluble Aß (sAß) species and cognitive deficits associated with AD.


Subject(s)
Alzheimer Disease , Animals , Humans , Mice , Alzheimer Disease/metabolism , Amyloid beta-Protein Precursor/metabolism , Apolipoproteins E/metabolism , Disease Models, Animal , Memory Disorders , Mice, Transgenic , Microglia/metabolism , Plaque, Amyloid/metabolism , Receptors, Purinergic P2X4/metabolism
3.
J Neuroinflammation ; 19(1): 234, 2022 Sep 24.
Article in English | MEDLINE | ID: mdl-36153535

ABSTRACT

BACKGROUND: Research in recent years firmly established that microglial cells play an important role in the pathogenesis of Alzheimer's disease (AD). In parallel, a series of studies showed that, under both homeostatic and pathological conditions, microglia are a heterogeneous cell population. In AD, amyloid-ß (Aß) plaque-associated microglia (PAM) display a clearly distinct phenotype compared to plaque-distant microglia (PCM), suggesting that these two microglia subtypes likely differently contribute to disease progression. So far, molecular characterization of PAM was performed indirectly using single cell RNA sequencing (scRNA-seq) approaches or based on markers that are supposedly up-regulated in this microglia subpopulation. METHODS: In this study based on a well-characterized AD mouse model, we combined cell-specific laser capture microdissection and RNA-seq analysis to i) identify, without preconceived notions of the molecular and/or functional changes that would affect these cells, the genes and gene networks that are dysregulated in PAM or PCM at three critical stages of the disease, and ii) to investigate the potential contribution of both plaque-associated and plaque-distant microglia. RESULTS: First, we established that our approach allows selective isolation of microglia, while preserving spatial information and preventing transcriptome changes induced by classical purification approaches. Then, we identified, in PAM and PCM subpopulations, networks of co-deregulated genes and analyzed their potential functional roles in AD. Finally, we investigated the dynamics of microglia transcriptomic remodeling at early, intermediate and late stages of the disease and validated select findings in postmortem human AD brain. CONCLUSIONS: Our comprehensive study provides useful transcriptomic information regarding the respective contribution of PAM and PCM across the Aß pathology progression. It highlights specific pathways that would require further study to decipher their roles across disease progression. It demonstrates that the proximity of microglia to Aß-plaques dramatically alters the microglial transcriptome and reveals that these changes can have both positive and negative impacts on the surrounding cells. These opposing effects may be driven by local microglia heterogeneity also demonstrated by this study. Our approach leads to molecularly define the less well studied plaque-distant microglia. We show that plaque-distant microglia are not bystanders of the disease, although the transcriptomic changes are far less striking compared to what is observed in plaque-associated microglia. In particular, our results suggest they may be involved in Aß oligomer detection and in Aß-plaque initiation, with increased contribution as the disease progresses.


Subject(s)
Alzheimer Disease , Microglia , Alzheimer Disease/pathology , Amyloid beta-Peptides/metabolism , Amyloid beta-Protein Precursor/genetics , Amyloid beta-Protein Precursor/metabolism , Animals , Disease Models, Animal , Disease Progression , Humans , Mice , Mice, Transgenic , Microglia/metabolism , Plaque, Amyloid/pathology , Transcriptome
4.
eNeuro ; 8(1)2021.
Article in English | MEDLINE | ID: mdl-33380526

ABSTRACT

ATP is an extracellular signaling molecule involved in numerous physiological and pathologic processes. However, in situ characterization of the spatiotemporal dynamic of extracellular ATP is still challenging because of the lack of sensor with appropriate specificity, sensitivity, and kinetics. Here, we report the development of biosensors based on the fusion of cation permeable ATP receptors (P2X) to genetically encoded calcium sensors [genetically encoded calcium indicator (GECI)]. By combining the features of P2X receptors with the high signal-to-noise ratio of GECIs, we generated ultrasensitive green and red fluorescent sniffers that detect nanomolar ATP concentrations in situ and also enable the tracking of P2X receptor activity. We provide the proof of concept that these sensors can dynamically track ATP release evoked by depolarization in mouse neurons or by extracellular hypotonicity. Targeting these P2X-based biosensors to diverse cell types should advance our knowledge of extracellular ATP dynamics in vivo.


Subject(s)
Receptors, Purinergic P2 , Adenosine Triphosphate , Animals , Calcium , Mice , Neurons , Receptors, Purinergic P2/genetics , Signal Transduction
5.
Brain Behav Immun ; 91: 404-417, 2021 01.
Article in English | MEDLINE | ID: mdl-33190798

ABSTRACT

Microglia, the resident immune cells of the brain, have recently emerged as key players in Alzheimer Disease (AD) pathogenesis, but their roles in AD remain largely elusive and require further investigation. Microglia functions are readily altered when isolated from their brain environment, and microglia reporter mice thus represent valuable tools to study the contribution of these cells to neurodegenerative diseases such as AD. The CX3CR1+/eGFP mice is one of the most popular microglia reporter mice, and has been used in numerous studies to investigate in vivo microglial functions, including in the context of AD research. However, until now, the impact of CX3CR1 haplodeficiency on the typical features of Alzheimer Disease has not been studied in depth. To fill this gap, we generated APPswe/PSEN1dE9:CX3CR1+/eGFP mice and analyzed these mice for Alzheimer's like pathology and neuroinflammation hallmarks. More specifically, using robust multifactorial statistical and multivariate analyses, we investigated the impact of CX3CR1 deficiency in both males and females, at three typical stages of the pathology progression: at early stage when Amyloid-ß (Aß) deposition just starts, at intermediate stage during Aß accumulation phase and at more advanced stages when Aß plaque number stabilizes. We found that CX3CR1 haplodeficiency had little impact on the progression of the pathology in the APPswe/PSEN1dE9 model and demonstrated that the APPswe/PSEN1dE9:CX3CR1+/eGFP line is a relevant and useful model to study the role of microglia in Alzheimer Disease. In addition, although Aß plaques density is higher in females compared to age-matched males, we show that their glial reaction, inflammation status and memory deficits are not different.


Subject(s)
Alzheimer Disease , CX3C Chemokine Receptor 1 , Alzheimer Disease/genetics , Amyloid beta-Peptides/metabolism , Amyloid beta-Protein Precursor/metabolism , Animals , Brain/metabolism , CX3C Chemokine Receptor 1/genetics , Disease Models, Animal , Disease Progression , Female , Male , Mice , Mice, Transgenic , Microglia/metabolism , Plaque, Amyloid
6.
Article in English | MEDLINE | ID: mdl-32425766

ABSTRACT

Neuronal hippocampal cultures are simple and valuable models for studying neuronal function. While embryonic cultures are widely used for different applications, mouse postnatal cultures are still challenging, lack reproducibility and/or exhibit inappropriate neuronal activity. Yet, postnatal cultures have major advantages such as allowing genotyping of pups before culture and reducing the number of experimental animals. Herein we describe a simple and fast protocol for culturing and genetically manipulating hippocampal neurons from P0 to P3 mice. This protocol provides reproducible cultures exhibiting a consistent neuronal development, normal excitatory over inhibitory neuronal ratio and a physiological neuronal activity. We also describe simple and efficient procedures for genetic manipulation of neurons using transfection reagent or lentiviral particles. Overall, this method provides a detailed and validated protocol allowing to explore cellular mechanisms and neuronal activity in postnatal hippocampal neurons in culture.

7.
Glia ; 68(9): 1692-1728, 2020 09.
Article in English | MEDLINE | ID: mdl-31958188

ABSTRACT

Development, physiological functions, and pathologies of the brain depend on tight interactions between neurons and different types of glial cells, such as astrocytes, microglia, oligodendrocytes, and oligodendrocyte precursor cells. Assessing the relative contribution of different glial cell types is required for the full understanding of brain function and dysfunction. Over the recent years, several technological breakthroughs were achieved, allowing "glio-scientists" to address new challenging biological questions. These technical developments make it possible to study the roles of specific cell types with medium or high-content workflows and perform fine analysis of their mutual interactions in a preserved environment. This review illustrates the potency of several cutting-edge experimental approaches (advanced cell cultures, induced pluripotent stem cell (iPSC)-derived human glial cells, viral vectors, in situ glia imaging, opto- and chemogenetic approaches, and high-content molecular analysis) to unravel the role of glial cells in specific brain functions or diseases. It also illustrates the translation of some techniques to the clinics, to monitor glial cells in patients, through specific brain imaging methods. The advantages, pitfalls, and future developments are discussed for each technique, and selected examples are provided to illustrate how specific "gliobiological" questions can now be tackled.


Subject(s)
Astrocytes , Neuroglia , Humans , Microglia , Neurons , Oligodendroglia
8.
Sci Rep ; 8(1): 964, 2018 01 17.
Article in English | MEDLINE | ID: mdl-29343707

ABSTRACT

Chronic inflammatory and neuropathic pains are major public health concerns. Potential therapeutic targets include the ATP-gated purinergic receptors (P2RX) that contribute to these pathological types of pain in several different cell types. The purinergic receptors P2RX2 and P2RX3 are expressed by a specific subset of dorsal root ganglion neurons and directly shape pain processing by primary afferents. In contrast the P2RX4 and P2RX7 are mostly expressed in myeloid cells, where activation of these receptors triggers the release of various pro-inflammatory molecules. Here, we demonstrate that P2RX4 also controls calcium influx in mouse dorsal root ganglion neurons. P2RX4 is up-regulated in pain-processing neurons during long lasting peripheral inflammation and it co-localizes with Brain-Derived Neurotrophic Factor (BDNF). In the dorsal horn of the spinal cord, BDNF-dependent signaling pathways, phosphorylation of Erk1/2 and of the GluN1 subunit as well as the down regulation of the co-transporter KCC2, which are triggered by peripheral inflammation are impaired in P2RX4-deficient mice. Our results suggest that P2RX4, expressed by sensory neurons, controls neuronal BDNF release that contributes to hyper-excitability during chronic inflammatory pain and establish P2RX4 in sensory neurons as a new potential therapeutic target to treat hyperexcitability during chronic inflammatory pain.


Subject(s)
Brain-Derived Neurotrophic Factor/metabolism , Ganglia, Spinal/metabolism , Inflammation/metabolism , Receptors, Purinergic P2X4/metabolism , Sensory Receptor Cells/metabolism , Signal Transduction/physiology , Animals , Female , Hyperalgesia/metabolism , Male , Mice , Mice, Inbred C57BL , Neuralgia/metabolism , Spinal Cord Dorsal Horn/metabolism , Up-Regulation/physiology
9.
PLoS One ; 11(3): e0150793, 2016.
Article in English | MEDLINE | ID: mdl-26963100

ABSTRACT

Kainic acid (KA) is routinely used to elicit status epilepticus (SE) and epileptogenesis. Among the available KA administration protocols, intranasal instillation (IN) remains understudied. Dosages of KA were instilled IN in mice. Racine Scale and Video-EEG were used to assess and quantify SE onset. Time spent in SE and spike activity was quantified for each animal and confirmed by power spectrum analysis. Immunohistochemistry and qPCR were performed to define brain inflammation occurring after SE, including activated microglial phenotypes. Long term video-EEG recording was also performed. Titration of IN KA showed that a dose of 30 mg/kg was associated with low mortality while eliciting SE. IN KA provoked at least one behavioral and electrographic SE in the majority of the mice (>90%). Behavioral and EEG SE were accompanied by a rapid and persistent microglial-astrocytic cell activation and hippocampal neurodegeneration. Specifically, microglial modifications involved both pro- (M1) and anti-inflammatory (M2) genes. Our initial long-term video-EEG exploration conducted using a small cohort of mice indicated the appearance of spike activity or SE. Our study demonstrated that induction of SE is attainable using IN KA in mice. Typical pro-inflammatory brain changes were observed in this model after SE, supporting disease pathophysiology. Our results are in favor of the further development of IN KA as a means to study seizure disorders. A possibility for tailoring this model to drug testing or to study mechanisms of disease is offered.


Subject(s)
Behavior, Animal/drug effects , Electroencephalography , Kainic Acid/pharmacology , Status Epilepticus/physiopathology , Administration, Intranasal , Animals , Astrocytes/metabolism , Astrocytes/pathology , Disease Models, Animal , Inflammation/metabolism , Inflammation/pathology , Inflammation/physiopathology , Mice , Mice, Transgenic , Microglia/metabolism , Microglia/pathology , Status Epilepticus/drug therapy , Status Epilepticus/metabolism , Status Epilepticus/pathology
10.
Diabetologia ; 57(3): 532-41, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24317793

ABSTRACT

AIMS/HYPOTHESIS: Beta cell failure due to progressive secretory dysfunction and limited expansion of beta cell mass is a key feature of type 2 diabetes. Beta cell function and mass are controlled by glucose and hormones/neurotransmitters that activate G protein-coupled receptors or receptor tyrosine kinases. We have investigated the role of ß-arrestin (ARRB)2, a scaffold protein known to modulate such receptor signalling, in the modulation of beta cell function and mass, with a specific interest in glucagon-like peptide-1 (GLP-1), muscarinic and insulin receptors. METHODS: ß-arrestin2-knockout mice and their wild-type littermates were fed a normal or a high-fat diet (HFD). Glucose tolerance, insulin sensitivity and insulin secretion were assessed in vivo. Beta cell mass was evaluated in pancreatic sections. Free cytosolic [Ca(2+)] and insulin secretion were determined using perifused islets. The insulin signalling pathway was evaluated by western blotting. RESULTS: Arrb2-knockout mice exhibited impaired glucose tolerance and insulin secretion in vivo, but normal insulin sensitivity compared with wild type. Surprisingly, the absence of ARRB2 did not affect glucose-stimulated insulin secretion or GLP-1- and acetylcholine-mediated amplifications from perifused islets, but it decreased the islet insulin content and beta cell mass. Additionally, there was no compensatory beta cell mass expansion through proliferation in response to the HFD. Furthermore, Arrb2 deletion altered the islet insulin signalling pathway. CONCLUSIONS/INTERPRETATION: ARRB2 is unlikely to be involved in the regulation of insulin secretion, but it is required for beta cell mass plasticity. Additionally, we provide new insights into the mechanisms involved in insulin signalling in beta cells.


Subject(s)
Arrestins/metabolism , Diabetes Mellitus, Experimental/metabolism , Glucagon-Like Peptide 1/metabolism , Insulin-Secreting Cells/metabolism , Insulin/metabolism , Pancreas/metabolism , Animals , Blotting, Western , Diet, High-Fat , Insulin Secretion , Mice , Mice, Knockout , Real-Time Polymerase Chain Reaction , Receptor, Insulin , Signal Transduction , beta-Arrestin 2 , beta-Arrestins
11.
J Biol Chem ; 285(3): 1989-2002, 2010 Jan 15.
Article in English | MEDLINE | ID: mdl-19915011

ABSTRACT

Strategies based on activating GLP-1 receptor (GLP-1R) are intensively developed for the treatment of type 2 diabetes. The exhaustive knowledge of the signaling pathways linked to activated GLP-1R within the beta-cells is of major importance. In beta-cells, GLP-1 activates the ERK1/2 cascade by diverse pathways dependent on either Galpha(s)/cAMP/cAMP-dependent protein kinase (PKA) or beta-arrestin 1, a scaffold protein. Using pharmacological inhibitors, beta-arrestin 1 small interfering RNA, and islets isolated from beta-arrestin 1 knock-out mice, we demonstrate that GLP-1 stimulates ERK1/2 by two temporally distinct pathways. The PKA-dependent pathway mediates rapid and transient ERK1/2 phosphorylation that leads to nuclear translocation of the activated kinases. In contrast, the beta-arrestin 1-dependent pathway produces a late ERK1/2 activity that is restricted to the beta-cell cytoplasm. We further observe that GLP-1 phosphorylates the cytoplasmic proapoptotic protein Bad at Ser-112 but not at Ser-155. We find that the beta-arrestin 1-dependent ERK1/2 activation engaged by GLP-1 mediates the Ser-112 phosphorylation of Bad, through p90RSK activation, allowing the association of Bad with the scaffold protein 14-3-3, leading to its inactivation. beta-Arrestin 1 is further found to mediate the antiapoptotic effect of GLP-1 in beta-cells through the ERK1/2-p90RSK-phosphorylation of Bad. This new regulatory mechanism engaged by activated GLP-1R involving a beta-arrestin 1-dependent spatiotemporal regulation of the ERK1/2-p90RSK activity is now suspected to participate in the protection of beta-cells against apoptosis. Such signaling mechanism may serve as a prototype to generate new therapeutic GLP-1R ligands.


Subject(s)
Apoptosis/drug effects , Arrestins/metabolism , Glucagon-Like Peptide 1/pharmacology , Insulin-Secreting Cells/metabolism , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , bcl-Associated Death Protein/metabolism , 14-3-3 Proteins/metabolism , Animals , Cell Line , Cyclic AMP/metabolism , Cyclic AMP-Dependent Protein Kinases/metabolism , Cytosol/drug effects , Cytosol/metabolism , Enzyme Activation/drug effects , Insulin-Secreting Cells/drug effects , Mice , Phosphorylation/drug effects , Ribosomal Protein S6 Kinases, 90-kDa/metabolism , Serine , Signal Transduction/drug effects , Time Factors , bcl-Associated Death Protein/chemistry , beta-Arrestin 1 , beta-Arrestins
12.
Diabetes ; 58(5): 1105-15, 2009 May.
Article in English | MEDLINE | ID: mdl-19223597

ABSTRACT

OBJECTIVE: In type 2 diabetes, chronic hyperglycemia is detrimental to beta-cells, causing apoptosis and impaired insulin secretion. The transcription factor cAMP-responsive element-binding protein (CREB) is crucial for beta-cell survival and function. We investigated whether prolonged exposure of beta-cells to high glucose affects the functional integrity of CREB. RESEARCH DESIGN AND METHODS: INS-1E cells and rat and human islets were used. Gene expression was analyzed by RT-PCR and Western blotting. Apoptosis was detected by cleaved caspase-3 emergence, DNA fragmentation, and electron microscopy. RESULTS: Chronic exposure of INS-1E cells and rat and human islets to high glucose resulted in decreased CREB protein expression, phosphorylation, and transcriptional activity associated with apoptosis and impaired beta-cell function. High-glucose treatment increased CREB polyubiquitination, while treatment of INS-1E cells with the proteasome inhibitor MG-132 prevented the decrease in CREB content. The emergence of apoptosis in INS-1E cells with decreased CREB protein expression knocked down by small interfering RNA suggested that loss of CREB protein content induced by high glucose contributes to beta-cell apoptosis. Loading INS-1E cells or human islets with a cell-permeable peptide mimicking the proteasomal targeting sequence of CREB blocked CREB degradation and protected INS-1E cells and human islets from apoptosis induced by high glucose. The insulin secretion in response to glucose and the insulin content were preserved in human islets exposed to high glucose and loaded with the peptide. CONCLUSIONS: These studies demonstrate that the CREB degradation by the ubiquitin-proteasome pathway contributes to beta-cell dysfunction and death upon glucotoxicity and provide new insight into the cellular mechanisms of glucotoxicity.


Subject(s)
Cyclic AMP Response Element Modulator/metabolism , Glucose/toxicity , Insulin-Secreting Cells/pathology , Islets of Langerhans/pathology , Proteasome Endopeptidase Complex/metabolism , Ubiquitin/metabolism , Animals , Apoptosis/drug effects , Brain Death , CREB-Binding Protein/drug effects , CREB-Binding Protein/metabolism , Cell Line , Cell Survival/drug effects , Cyclic AMP Response Element Modulator/drug effects , DNA Fragmentation , Diabetes Mellitus, Experimental/metabolism , Humans , Insulin/metabolism , Insulin Secretion , Insulin-Secreting Cells/drug effects , Islets of Langerhans/drug effects , Rats , Rats, Wistar , Reverse Transcriptase Polymerase Chain Reaction
13.
J Biol Chem ; 284(7): 4332-42, 2009 Feb 13.
Article in English | MEDLINE | ID: mdl-19074139

ABSTRACT

In pancreatic beta-cells, the pituitary adenylate cyclase-activating polypeptide (PACAP) exerts a potent insulin secretory effect via PAC(1) and VPAC receptors (Rs) through the Galpha(s)/cAMP/protein kinase A pathway. Here, we investigated the mechanisms linking PAC(1)R to ERK1/2 activation in INS-1E beta-cells and pancreatic islets. PACAP caused a transient (5 min) increase in ERK1/2 phosphorylation via PAC(1)Rs and promoted nuclear translocation of a fraction of cytosolic p-ERK1/2. Both protein kinase A- and Src-dependent pathways mediated this transient ERK1/2 activation. Moreover, PACAP potentiated glucose-induced long-lasting ERK1/2 activation. Blocking Ca(2+) influx abolished glucose-induced ERK1/2 activation and PACAP potentiating effect. Glucose stimulation during KCl depolarization showed that, in addition to the triggering signal (rise in cytosolic [Ca(2+)]), the amplifying pathway was also involved in glucose-induced sustained ERK1/2 activation and was required for PACAP potentiation. The finding that at 30 min glucose-induced p-ERK1/2 was detected in both cytosol and nucleus while the potentiating effect of PACAP was only observed in the cytosol, suggested the involvement of the scaffold protein beta-arrestin. Indeed, beta-arrestin 1 (beta-arr1) depletion (in beta-arr1 knockout mouse islets or in INS-1E cells by siRNA) completely abolished PACAP potentiation of long-lasting ERK1/2 activation by glucose. Finally, PACAP potentiated glucose-induced CREB transcriptional activity and IRS-2 mRNA expression mainly via the ERK1/2 signaling pathway, and likewise, beta-arr1 depletion reduced the PACAP potentiating effect on IRS-2 expression. These results establish for the first time that PACAP potentiates glucose-induced long-lasting ERK1/2 activation via a beta-arr1-dependent pathway and thus provide new insights concerning the mechanisms of PACAP and glucose actions in pancreatic beta-cells.


Subject(s)
Arrestins/metabolism , Glucose/pharmacology , Insulin-Secreting Cells/metabolism , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Receptors, Pituitary Adenylate Cyclase-Activating Polypeptide/metabolism , Sweetening Agents/pharmacology , Animals , Arrestins/genetics , Calcium/metabolism , Calcium Signaling/drug effects , Calcium Signaling/physiology , Cell Line , Cell Nucleus/genetics , Cell Nucleus/metabolism , Cyclic AMP Response Element-Binding Protein/genetics , Cyclic AMP Response Element-Binding Protein/metabolism , Cytosol/metabolism , Enzyme Activation/drug effects , Enzyme Activation/physiology , Gene Expression Regulation/drug effects , Gene Expression Regulation/physiology , Glucose/metabolism , Insulin Receptor Substrate Proteins/biosynthesis , Insulin Receptor Substrate Proteins/genetics , Insulin-Secreting Cells/cytology , MAP Kinase Signaling System/drug effects , MAP Kinase Signaling System/physiology , Male , Mice , Mice, Knockout , Mitogen-Activated Protein Kinase 1/genetics , Mitogen-Activated Protein Kinase 3/genetics , Phosphorylation/drug effects , Phosphorylation/physiology , Pituitary Adenylate Cyclase-Activating Polypeptide/genetics , Pituitary Adenylate Cyclase-Activating Polypeptide/metabolism , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Receptors, Pituitary Adenylate Cyclase-Activating Polypeptide/genetics , Sweetening Agents/metabolism , Time Factors , beta-Arrestin 1 , beta-Arrestins
14.
Pharm Res ; 23(11): 2665-71, 2006 Nov.
Article in English | MEDLINE | ID: mdl-17048117

ABSTRACT

PURPOSE: A series of C2-substituted ATP analogues was previously shown to have potent insulin-secreting properties, yet with poor tissue-selectivity for the pancreatic beta-cell. The present study was designed to evaluate the binding profile on beta-cell membranes and the effects on insulin release and pancreatic vascular resistance of a second generation of P2Y(1) receptor agonists, based on C2-substitution of the adenosine 5'-O-(1-boranotriphosphate) scaffold. MATERIALS AND METHODS: Functional experiments were performed in the rat isolated pancreas model; binding studies with ATP-alpha-[(35)S] were performed in membrane homogenates from the rat insulinoma INS-1 cell line. The diastereoisomers of the compounds are designated by A and B. RESULTS: Under 8.3 mmol l(-1) glucose, 2-methylthio-ATP-alpha-B, A isomer, induced a biphasic and concentration dependent insulin response; its maximal efficacy reaches ninefold the baseline secretion and its EC(50) is 28.1 nmol l(-1). No significant effect of this isomer was observed on vascular resistance, whereas the B isomer, which was a less potent insulin secretagogue, consistently induced a transient vasoconstriction. Interestingly, the insulin response induced by 2-methylthio-ATP-alpha-B, A isomer, was clearly glucose-dependent. This drug competes with ATP-alpha-[(35)S] binding in a complex two sites interaction model, with a K(0.5) value of 17.7 nmol l(-1). 2-Chloro-ATP-alpha-B had a similar insulin-secreting profile as 2-methylthio-ATP-alpha-B, with a lower tissue-selectivity. The non-substituted ATP-alpha-B analog, A isomer, was less potent than the C2-substituted derivatives (A isomers) and had a vasorelaxant effect. CONCLUSIONS: We conclude that 2-methylthio-ATP-alpha-B, A isomer, is a potent and tissue-selective P2Y receptor agonist with high efficacy. Its insulin-releasing action is glucose-dependent, which gives interest to this compound as a drug candidate for treating type 2 diabetes.


Subject(s)
Adenosine Triphosphate/analogs & derivatives , Boranes/pharmacology , Insulin/metabolism , Purinergic P2 Receptor Agonists , Receptors, Purinergic P2/physiology , Adenosine Triphosphate/metabolism , Animals , Dose-Response Relationship, Drug , Insulin Secretion , Male , Rats , Rats, Wistar , Receptors, Purinergic P2Y1 , Thionucleotides/metabolism , Vascular Resistance/drug effects
15.
J Agric Food Chem ; 53(1): 151-7, 2005 Jan 12.
Article in English | MEDLINE | ID: mdl-15631522

ABSTRACT

Insulin resistance and oxidative stress act synergistically in the development of cardiovascular complications. The present study compared the efficacy of three polyphenolic extracts in their capacity to prevent hypertension, cardiac hypertrophy, increased production of reactive oxygen species (ROS) by the aorta or the heart, and increased expression of cardiac NAD(P)H oxidase in a model of insulin resistance. Rats were fed a 60%-enriched fructose food and were treated once a day (gavage) for 6 weeks with 10 mL/kg of water only (F group) or the same amount of solution containing a red grape skin polyphenolic extract enriched in anthocyanins (ANT), a grape seed extract enriched in procyanidins and rich in galloylated procyanidins (PRO), or the commercial preparation Vitaflavan (VIT), rich in catechin oligomers. All treatments were administered at the same dose of 21 mg/kg of polyphenols. Our data indicate that (a) the ANT treatment prevented hypertension, cardiac hypertrophy, and production of ROS, (b) the PRO treatment prevented insulin resistance, hypertriglyceridemia, and overproduction of ROS but had only minor effects on hypertension or hypertrophy, while (c) Vitaflavan prevented hypertension, cardiac hypertrophy, and overproduction of ROS. All polyphenolic treatments prevented the increased expression of the p91phox NADPH oxidase subunit. In summary, our study suggest that (a) the pathogeny of cardiac hypertrophy in the fructose-fed rat disease involves both hypertension and hyperproduction of ROS, (b) polyphenolic extracts enriched in different types of polyphenols possess differential effects on insulin resistance, hypertension, and cardiac hypertrophy, and (c) polyphenols modulate the expression of NAD(P)H oxidase.


Subject(s)
Cardiomegaly/prevention & control , Flavonoids/administration & dosage , Hypertension/prevention & control , Insulin Resistance , NADPH Oxidases/metabolism , Phenols/administration & dosage , Animals , Fructose/administration & dosage , Fruit/chemistry , Heart Ventricles/enzymology , NADPH Oxidases/analysis , Plant Extracts/administration & dosage , Plant Extracts/chemistry , Polyphenols , Rats , Rats, Sprague-Dawley , Reactive Oxygen Species/metabolism , Seeds/chemistry , Vitis/chemistry
16.
J Agric Food Chem ; 52(18): 5593-7, 2004 Sep 08.
Article in English | MEDLINE | ID: mdl-15373398

ABSTRACT

The effects of a red wine polyphenolic extract (RWPE), ethanol, or both combined were evaluated in insulin resistant rats. Rats were fed for 6 weeks with fructose (60%)-enriched food and force-fed with (a) water only (F group), (b) aqueous solution of RWPE (100 mg/kg, FP group), (c) 10% (v/v) mixture of ethanol and water (FE group), or (d) solution containing the same amount of the RWPE and ethanol (FPE group). Animals fed a standard chow (C group) were used for comparison purpose. After 6 weeks, blood pressure was higher in F (130.0 x b1 1.7 mm Hg) than in C animals (109.6 x b1 0.9 mm Hg) and similar to the C group in all other fructose-fed treatment groups. Relative heart weight was higher in F (3.10 x b1 0.05) than in C (2.78 x b1 0.07) and significantly lower in FP (2.92 x b1 0.04) and FPE (2.87 x b1 0.08 mg/g) than in F animals. Left ventricle and aorta productions of reactive oxygen species (O2*-) were higher in F than in C groups and lowered by the RWPE but not by the ethanol treatment. Ethanol but not the RWPE treatment reduced the degree of insulin resistance in the fructose-fed rats. In summary, our study showed that polyphenols are able to prevent cardiac hypertrophy and production of reactive oxygen species in the insulin resistant fructose-fed rat.


Subject(s)
Cardiomegaly/prevention & control , Ethanol/administration & dosage , Flavonoids/administration & dosage , Hypertension/prevention & control , Insulin Resistance , Phenols/administration & dosage , Wine/analysis , Animals , Anions , Dietary Carbohydrates/administration & dosage , Fructose/administration & dosage , Male , Polyphenols , Rats , Rats, Sprague-Dawley , Reactive Oxygen Species/metabolism , Superoxides/metabolism
17.
Diabetes ; 53(6): 1467-74, 2004 Jun.
Article in English | MEDLINE | ID: mdl-15161750

ABSTRACT

We previously showed that pancreatic beta-cells express a neuronal isoform of nitric oxide synthase (nNOS) that controls insulin secretion by exerting two enzymatic activities: nitric oxide (NO) production and cytochrome c reductase activity. We now bring evidence that two inhibitors of nNOS, N-omega-nitro-l-arginine methyl ester (l-NAME) and 7-nitroindazole (7-NI), increase glucose-induced insulin secretion but affect beta-cell function differently. In the presence of l-NAME, insulin response is monophasic, whereas 7-NI preserves the normal biphasic secretory pattern. In addition, the alterations of beta-cell functional response induced by the inhibitors also differ by their sensitivity to a substitutive treatment with sodium nitroprusside, a chemical NO donor. These differences are probably related to the nature of the two inhibitors. Indeed, using low-temperature SDS-PAGE and real-time analysis of nNOS dimerization by surface plasmon resonance, we could show that 7-NI, which competes with arginine and tetrahydrobiopterin (BH(4)), an essential cofactor for nNOS dimer formation, inhibits dimerization of the enzyme, whereas the substrate-based inhibitor l-NAME stabilizes the homodimeric state of nNOS. The latter effect could be reproduced by the two endogenous inhibitors of NOS, N-omega-methyl-l-arginine and asymmetric dimethylarginine, and resulted interestingly in a reduced ability of the protein inhibitor of nNOS (PIN) to dissociate nNOS dimers. We conclude that intracellular factors able to induce abnormalities in the nNOS monomer/dimer equilibrium could lead to pancreatic beta-cell dysfunction.


Subject(s)
Arginine/analogs & derivatives , Arginine/pharmacology , Drosophila Proteins , Glucose/pharmacology , Insulin/metabolism , Nitric Oxide Synthase/chemistry , Nitric Oxide Synthase/metabolism , Animals , Carrier Proteins/pharmacology , Cell Line , Dimerization , Dyneins , Enzyme Inhibitors/pharmacology , Indazoles/pharmacology , Insulin Secretion , Kinetics , Male , Miconazole/pharmacology , NG-Nitroarginine Methyl Ester/pharmacology , Nitric Oxide Donors/pharmacology , Nitric Oxide Synthase/drug effects , Nitric Oxide Synthase Type I , Nitroarginine/pharmacology , Nitroprusside/pharmacology , Rats , Rats, Wistar , Surface Plasmon Resonance
SELECTION OF CITATIONS
SEARCH DETAIL
...