Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 37
Filter
1.
Curr Res Neurobiol ; 5: 100113, 2023.
Article in English | MEDLINE | ID: mdl-38020806

ABSTRACT

Apnea of prematurity (AOP) affects more than 50% of preterm infants and leads to perinatal intermittent hypoxia (IH) which is a major cause of morbimortality worldwide. At birth, the human cerebellar cortex is still immature, making it vulnerable to perinatal events. Additionally, studies have shown a correlation between cerebellar functions and the deficits observed in children who have experienced AOP. Yet, the cerebellar alterations underpinning this link remain poorly understood. To gain insight into the involvement of the cerebellum in perinatal hypoxia-related consequences, we developed a mouse model of AOP. Our previous research has revealed that IH induces oxidative stress in the developing cerebellum, as evidenced by the over-expression of genes involved in reactive oxygen species production and the under-expression of genes encoding antioxidant enzymes. These changes suggest a failure of the defense system against oxidative stress and could be responsible for neuronal death in the cerebellum. Building upon these findings, we conducted a transcriptomic study of the genes involved in the processes that occur during cerebellar development. Using real-time PCR, we analyzed the expression of these genes at different developmental stages and in various cell types. This enabled us to pinpoint a timeframe of vulnerability at P8, which represents the age with the highest number of downregulated genes in the cerebellum. Furthermore, we discovered that our IH protocol affects several molecular pathways, including proliferation, migration, and differentiation. This indicates that IH can impact the development of different cell types, potentially contributing to the histological and behavioral deficits observed in this model. Overall, our data strongly suggest that the cerebellum is highly sensitive to IH, and provide valuable insights into the cellular and molecular mechanisms underlying AOP. In the long term, these findings may contribute to the identification of novel therapeutic targets for improving the clinical management of this prevalent pathology.

2.
Cell Biosci ; 12(1): 148, 2022 Sep 06.
Article in English | MEDLINE | ID: mdl-36068642

ABSTRACT

BACKGROUND: Apnea of prematurity (AOP) is caused by respiratory control immaturity and affects nearly 50% of premature newborns. This pathology induces perinatal intermittent hypoxia (IH), which leads to neurodevelopmental disorders. The impact on the brain has been well investigated. However, despite its functional importance and immaturity at birth, the involvement of the cerebellum remains poorly understood. Therefore, this study aims to identify the effects of IH on cerebellar development using a mouse model of AOP consisting of repeated 2-min cycles of hypoxia and reoxygenation over 6 h and for 10 days starting on postnatal day 2 (P2). RESULTS: At P12, IH-mice cerebella present higher oxidative stress associated with delayed maturation of the cerebellar cortex and decreased dendritic arborization of Purkinje cells. Moreover, mice present with growth retardation and motor disorders. In response to hypoxia, the developing cerebellum triggers compensatory mechanisms resulting in the unaltered organization of the cortical layers from P21 onwards. Nevertheless, some abnormalities remain in adult Purkinje cells, such as the dendritic densification, the increase in afferent innervation, and axon hypomyelination. Moreover, this compensation seems insufficient to allow locomotor recovery because adult mice still show motor impairment and significant disorders in spatial learning. CONCLUSIONS: All these findings indicate that the cerebellum is a target of intermittent hypoxia through alterations of developmental mechanisms leading to long-term functional deficits. Thus, the cerebellum could contribute, like others brain structures, to explaining the pathophysiology of AOP.

3.
Mar Environ Res ; 169: 105393, 2021 Jul.
Article in English | MEDLINE | ID: mdl-34217095

ABSTRACT

The Kerguelen Islands (49°26'S, 69°50'E) represent a unique environment due to their geographical isolation, which protects them from anthropogenic pollution. The ability of the endemic mussel, part of the Mytilus complex, to cope with moderate heat stress was explored using omic tools. Transcripts involved in six major metabolic functions were selected and the qRT-PCR data indicated mainly changes in aerobic and anaerobic energy metabolism and stress response. Proteomic comparisons revealed a typical stress response pattern with cytoskeleton modifications and elements suggesting increased energy metabolism. Results also suggest conservation of protein homeostasis by the long-lasting presence of HSP while a general decrease in transcription is observed. The overall findings are consistent with an adaptive response to moderate stresses in mussels in good physiological condition, i.e. living in a low-impact site, and with the literature concerning this model species. Therefore, local blue mussels could be advantageously integrated into biomonitoring strategies, especially in the context of Global Change.


Subject(s)
Mytilus edulis , Mytilus , Animals , Antarctic Regions , Heat-Shock Response , Mytilus edulis/genetics , Proteomics
4.
Biochim Biophys Acta Gen Subj ; 1863(11): 129410, 2019 11.
Article in English | MEDLINE | ID: mdl-31401178

ABSTRACT

BACKGROUND: Neurodegenerative disorders, such as Parkinson's disease (PD), are characterized by neuronal death involving, among other events, mitochondrial dysfunction and excitotoxicity. Along these lines, several attempts have been made to slow this pathology but none have been yet discovered. Based on its capacity to cross the blood-brain barrier and provide neuronal protection in vitro and in vivo, the pituitary adenylate cyclase-activating polypeptide (PACAP) represents a promising lead molecule. Pharmacological studies showed that PACAP interacts with three different G protein-coupled receptors, i.e. PAC1, VPAC1 and VPAC2. However, only PAC1 is associated with neuronal anti-apoptotic actions, whilst VPAC activation might cause adverse effects. In the context of the development of PAC1-selective agonists, PACAP(1-23) (PACAP23) appears as the shortest known PACAP bioactive fragment. METHODS: Hence, the capacity of this peptide to bind PACAP receptors and protect neuroblastoma cells was evaluated under conditions of mitochondrial dysfunction and glutamate excitotoxicity. In addition, its ability to activate downstream signaling events involving G proteins (Gαs and Gαq), EPAC, and calcium was also assessed. RESULTS: Compared to the endogenous peptide, PACAP23 showed a reduced affinity towards PAC1, although this fragment exerted potent neuroprotection. However, surprisingly, some disparities were observed for PACAP23 signaling compared to full length PACAP, suggesting that downstream signaling related to neuroprotection is distinctly regulated following subtle differences in their PAC1 interactions. CONCLUSIONS: Altogether, this study demonstrates the potent neuroprotective action of amidated PACAP23. GENERAL SIGNIFICANCE: PACAP23 represents an attractive template for development of shorter PACAP-derived neuroprotective molecules.


Subject(s)
Calcium Signaling/drug effects , Neuroprotective Agents , Peptides , Pituitary Adenylate Cyclase-Activating Polypeptide , Animals , CHO Cells , Cricetulus , Humans , Neuroprotective Agents/chemistry , Neuroprotective Agents/pharmacology , Peptides/chemistry , Peptides/pharmacology , Pituitary Adenylate Cyclase-Activating Polypeptide/chemistry , Pituitary Adenylate Cyclase-Activating Polypeptide/pharmacology , Receptors, Vasoactive Intestinal Peptide, Type II/metabolism , Receptors, Vasoactive Intestinal Polypeptide, Type I/metabolism
5.
Mol Neurobiol ; 55(11): 8263-8277, 2018 Nov.
Article in English | MEDLINE | ID: mdl-29526016

ABSTRACT

Deficits in hippocampal synaptic plasticity result in cognitive impairment in Huntington's disease (HD). Pituitary adenylate cyclase-activating polypeptide (PACAP) is a neuropeptide that exerts neuroprotective actions, mainly through the PAC1 receptor. However, the role of PACAP in cognition is poorly understood, and no data exists in the context of Huntington's disease (HD). Here, we investigated the ability of PACAP receptor stimulation to enhance memory development in HD. First, we observed a hippocampal decline of all three PACAP receptor expressions, i.e., PAC1, VPAC1, and VPAC2, in two different HD mouse models, R6/1 and HdhQ7/Q111, from the onset of cognitive dysfunction. In hippocampal post-mortem human samples, we found a specific decrease of PAC1, without changes in VPAC1 and VPAC2 receptors. To determine whether activation of PACAP receptors could contribute to improve memory performance, we conducted daily intranasal administration of PACAP38 to R6/1 mice at the onset of cognitive impairment for seven days. We found that PACAP treatment rescued PAC1 level in R6/1 mice, promoted expression of the hippocampal brain-derived neurotrophic factor, and reduced the formation of mutant huntingtin aggregates. Furthermore, PACAP administration counteracted R6/1 mice memory deficits as analyzed by the novel object recognition test and the T-maze spontaneous alternation task. Importantly, the effect of PACAP on cognitive performance was associated with an increase of VGlut-1 and PSD95 immunolabeling in hippocampus of R6/1 mice. Taken together, these results suggest that PACAP, acting through stimulation of PAC1 receptor, may have a therapeutic potential to counteract cognitive deficits induced in HD.


Subject(s)
Hippocampus/physiopathology , Huntington Disease/physiopathology , Memory/physiology , Neuronal Plasticity/drug effects , Pituitary Adenylate Cyclase-Activating Polypeptide/pharmacology , Administration, Intranasal , Animals , Brain-Derived Neurotrophic Factor/metabolism , Cognition Disorders/physiopathology , Disease Models, Animal , Disks Large Homolog 4 Protein/metabolism , Gene Expression Regulation/drug effects , Hippocampus/pathology , Humans , Huntingtin Protein/metabolism , Male , Mice, Inbred C57BL , Mice, Transgenic , Middle Aged , Pituitary Adenylate Cyclase-Activating Polypeptide/administration & dosage , Protein Aggregates , Receptors, Pituitary Adenylate Cyclase-Activating Polypeptide/metabolism , Vesicular Glutamate Transport Protein 1/metabolism
6.
Neurogastroenterol Motil ; 30(7): e13305, 2018 07.
Article in English | MEDLINE | ID: mdl-29411462

ABSTRACT

BACKGROUND: Anorexia nervosa, a restrictive eating disorder, is often associated with gastrointestinal disorders, particularly a delayed gastric emptying. However, the mechanisms remained poorly documented. Thus, we aimed to evaluate gastric emptying and antrum protein metabolism in the Activity-Based Anorexia model (ABA). METHODS: Females C57Bl/6 mice were randomized into 3 groups: Control, ABA, and Limited Food Access (LFA). Food access has been progressively limited from 6 h/day at day 6 to 3 h/day at day 9 and until day 17. ABA mice had free access to an activity wheel. Gastric emptying was assessed. On gastric extracts, a proteomic analysis was performed, as well as an evaluation of protein synthesis and protein oxidation. KEY RESULTS: Both LFA and ABA mice exhibited a delayed gastric emptying compared with Controls (P < .05). Proteomic approach revealed 15 proteins that were differentially expressed. Among these proteins, we identified 2 clusters of interest contributing to (i) the organization of muscle fiber with ACTA2, VCL, KRT19, KRT8, and DES proteins and (ii) "heat shock proteins" with STIP1, HSPD1, and HSPA8 proteins. ABA mice specifically exhibited an increased rate of gastric oxidized proteins. CONCLUSIONS AND INFERENCES: Delayed gastric emptying observed in anorectic conditions appears to be secondary to malnutrition. However, an oxidative stress is specifically present in the stomach of ABA mice. Its role remains to be further studied.


Subject(s)
Anorexia/metabolism , Gastric Emptying/physiology , Gastroparesis/metabolism , Protein Carbonylation/physiology , Pyloric Antrum/metabolism , Animals , Anorexia/complications , Anorexia/physiopathology , Female , Gastroparesis/etiology , Gastroparesis/physiopathology , Mice , Mice, Inbred C57BL , Random Allocation , Spectrometry, Mass, Electrospray Ionization/methods
7.
Mol Neurobiol ; 53(8): 5203-16, 2016 10.
Article in English | MEDLINE | ID: mdl-26399645

ABSTRACT

The cerebellum is a structure of the central nervous system involved in balance, motor coordination, and voluntary movements. The elementary circuit implicated in the control of locomotion involves Purkinje cells, which receive excitatory inputs from parallel and climbing fibers, and are regulated by cerebellar interneurons. In mice as in human, the cerebellar cortex completes its development mainly after birth with the migration, differentiation, and synaptogenesis of granule cells. These cellular events are under the control of numerous extracellular matrix molecules including pleiotrophin (PTN). This cytokine has been shown to regulate the morphogenesis of Purkinje cells ex vivo and in vivo via its receptor PTPζ. Since Purkinje cells are the unique output of the cerebellar cortex, we explored the consequences of their PTN-induced atrophy on the function of the cerebellar neuronal circuit in mice. Behavioral experiments revealed that, despite a normal overall development, PTN-treated mice present a delay in the maturation of their flexion reflex. Moreover, patch clamp recording of Purkinje cells revealed a significant increase in the frequency of spontaneous excitatory postsynaptic currents in PTN-treated mice, associated with a decrease of climbing fiber innervations and an abnormal perisomatic localization of the parallel fiber contacts. At adulthood, PTN-treated mice exhibit coordination impairment on the rotarod test associated with an alteration of the synchronization gait. Altogether these histological, electrophysiological, and behavior data reveal that an early ECM disruption of PTN composition induces short- and long-term defaults in the establishment of proper functional cerebellar circuit.


Subject(s)
Carrier Proteins/pharmacology , Cerebellum/growth & development , Cytokines/pharmacology , Extracellular Space/chemistry , Nerve Net/growth & development , Neurons/metabolism , Aging/metabolism , Animals , Carrier Proteins/administration & dosage , Cell Shape/drug effects , Cytokines/administration & dosage , Excitatory Postsynaptic Potentials/drug effects , Female , Gait/drug effects , Humans , Locomotion/drug effects , Male , Mice , Neurons/drug effects
8.
Neuroscience ; 290: 472-84, 2015 Apr 02.
Article in English | MEDLINE | ID: mdl-25639232

ABSTRACT

Astrocytes synthesize and release endozepines, a family of regulatory neuropeptides, including diazepam-binding inhibitor (DBI) and its processing fragments such as the octadecaneuropeptide (ODN). At the molecular level, ODN interacts with two types of receptors, i.e. it acts as an inverse agonist of the central-type benzodiazepine receptor (CBR), and as an agonist of a G protein-coupled receptor (GPCR). ODN exerts a wide range of biological effects mediated through these two receptors and, in particular, it regulates astrocyte activity through an autocrine/paracrine mechanism involving the metabotropic receptor. More recently, it has been shown that Müller glial cells secrete phosphorylated DBI and that bisphosphorylated ODN ([bisphospho-Thr(3,9)]ODN, bpODN) has a stronger affinity for CBR than ODN. The aim of the present study was thus to investigate whether bpODN is released by mouse cortical astrocytes and to compare its potency to ODN. Using a radioimmunoassay and mass spectrometry analysis we have shown that bpODN as well as ODN were released in cultured astrocyte supernatants. Both bpODN and ODN increased astrocyte calcium event frequency but in a very different range of concentration. Indeed, ODN stimulatory effect decreased at concentrations over 10(-10)M whereas bpODN increased the calcium event frequency at similar doses. In vivo effects of bpODN and ODN were analyzed in two behavioral paradigms involving either the metabotropic receptor (anorexia) or the CBR (anxiety). As previously described, ODN (100ng, icv) induced a significant reduction of food intake. Similar effect was achieved with bpODN but at a 10 times higher dose (1000 ng, icv). Similarly, and contrasting with our hypothesis, bpODN was also 10 times less potent than ODN to induce anxiety-related behavior in the elevated zero maze test. Thus, the present data do not support that phosphorylation of ODN is involved in receptor selectivity but indicate that it rather weakens ODN activity.


Subject(s)
Astrocytes/metabolism , Diazepam Binding Inhibitor/metabolism , Diazepam Binding Inhibitor/pharmacology , Neuropeptides/metabolism , Neuropeptides/pharmacology , Peptide Fragments/metabolism , Peptide Fragments/pharmacology , Animals , Anti-Obesity Agents/pharmacology , Anxiety/chemically induced , Calcium/metabolism , Cells, Cultured , Diazepam Binding Inhibitor/analysis , Eating/drug effects , Exploratory Behavior/drug effects , Male , Maze Learning , Mice , Mice, Inbred C57BL , Neuropeptides/analysis , Peptide Fragments/analysis , Psychotropic Drugs/pharmacology , Rats
9.
Transl Psychiatry ; 4: e458, 2014 Oct 07.
Article in English | MEDLINE | ID: mdl-25290265

ABSTRACT

The molecular mechanisms at the origin of eating disorders (EDs), including anorexia nervosa (AN), bulimia and binge-eating disorder (BED), are currently unknown. Previous data indicated that immunoglobulins (Igs) or autoantibodies (auto-Abs) reactive with α-melanocyte-stimulating hormone (α-MSH) are involved in regulation of feeding and emotion; however, the origin of such auto-Abs is unknown. Here, using proteomics, we identified ClpB heat-shock disaggregation chaperone protein of commensal gut bacteria Escherichia coli as a conformational antigen mimetic of α-MSH. We show that ClpB-immunized mice produce anti-ClpB IgG crossreactive with α-MSH, influencing food intake, body weight, anxiety and melanocortin receptor 4 signaling. Furthermore, chronic intragastric delivery of E. coli in mice decreased food intake and stimulated formation of ClpB- and α-MSH-reactive antibodies, while ClpB-deficient E. coli did not affect food intake or antibody levels. Finally, we show that plasma levels of anti-ClpB IgG crossreactive with α-MSH are increased in patients with AN, bulimia and BED, and that the ED Inventory-2 scores in ED patients correlate with anti-ClpB IgG and IgM, which is similar to our previous findings for α-MSH auto-Abs. In conclusion, this work shows that the bacterial ClpB protein, which is present in several commensal and pathogenic microorganisms, can be responsible for the production of auto-Abs crossreactive with α-MSH, associated with altered feeding and emotion in humans with ED. Our data suggest that ClpB-expressing gut microorganisms might be involved in the etiology of EDs.


Subject(s)
Autoantibodies/immunology , Escherichia coli Proteins/immunology , Escherichia coli/immunology , Feeding and Eating Disorders/blood , Feeding and Eating Disorders/immunology , Heat-Shock Proteins/immunology , alpha-MSH/immunology , Adolescent , Adult , Animals , Disease Models, Animal , Electrophoresis, Polyacrylamide Gel , Endopeptidase Clp , Female , Humans , Immunoblotting , Male , Mice , Mice, Inbred C57BL , Young Adult
10.
Br J Pharmacol ; 171(19): 4425-39, 2014 Oct.
Article in English | MEDLINE | ID: mdl-24913445

ABSTRACT

BACKGROUND AND PURPOSE: The neuropeptide 26RFa and its cognate receptor GPR103 are involved in the control of food intake and bone mineralization. Here, we have tested, experimentally, the predicted ligand-receptor interactions by site-directed mutagenesis of GPR103 and designed point-substituted 26RFa analogues. EXPERIMENTAL APPROACH: Using the X-ray structure of the ß2 -adrenoceptor, a 3-D molecular model of GPR103 has been built. The bioactive C-terminal octapeptide 26RFa(19-26) , KGGFSFRF-NH2 , was docked in this GPR103 model and the ligand-receptor complex was submitted to energy minimization. KEY RESULTS: In the most stable complex, the Phe-Arg-Phe-NH2 part was oriented inside the receptor cavity, whereas the N-terminal Lys residue remained outside. A strong intermolecular interaction was predicted between the Arg(25) residue of 26RFa and the Gln(125) residue located in the third transmembrane helix of GPR103. To confirm this interaction experimentally, we tested the ability of 26RFa and Arg-modified 26RFa analogues to activate the wild-type and the Q125A mutant receptors transiently expressed in CHO cells. 26RFa (10(-6) M) enhanced [Ca(2+) ]i in wild-type GPR103-transfected cells, but failed to increase [Ca(2+) ]i in Q125A mutant receptor-expressing cells. Moreover, asymmetric dimethylation of the side chain of arginine led to a 26RFa analogue, [ADMA(25) ]26RFa(20-26) , that was unable to activate the wild-type GPR103, but antagonized 26RFa-evoked [Ca(2+) ]i increase. CONCLUSION AND IMPLICATIONS: Altogether, these data provide strong evidence for a functional interaction between the Arg(25) residue of 26RFa and the Gln(125) residue of GPR103 upon ligand-receptor activation, which can be exploited for the rational design of potent GPR103 agonists and antagonists.


Subject(s)
Models, Molecular , Neuropeptides/metabolism , Receptors, G-Protein-Coupled , Amino Acid Sequence , Animals , CHO Cells , Cricetinae , Cricetulus , Humans , Molecular Sequence Data , Mutagenesis, Site-Directed , Oligopeptides/metabolism , Receptors, Adrenergic, beta-2/chemistry , Receptors, G-Protein-Coupled/agonists , Receptors, G-Protein-Coupled/chemistry , Receptors, G-Protein-Coupled/genetics , Receptors, G-Protein-Coupled/metabolism , Sequence Alignment , Structure-Activity Relationship
11.
Curr Pharm Des ; 17(10): 1002-24, 2011.
Article in English | MEDLINE | ID: mdl-21524253

ABSTRACT

In neurological insults, such as cerebral ischemia and traumatic brain injury, complex molecular mechanisms involving inflammation and apoptosis are known to cause severe neuronal cell loss, emphasizing the necessity of developing therapeutic strategies targeting simultaneously these two processes. Over the last decade, numerous in vitro and in vivo studies have demonstrated the unique therapeutical potential of pituitary adenylate cyclase-activating polypeptide (PACAP) for the treatment of neuronal disorders involving apoptotic cell death and neuroinflammation. The neuroprotective activity of PACAP is based on its capacity to reduce the production of deleterious cytokines from activated microglia, to stimulate the release of neuroprotective agents from astrocytes and to inhibit pro-apoptotic intracellular pathways. However, the use of PACAP as a clinically applicable drug is hindered by its peptidic nature. As most natural peptides, native PACAP shows poor metabolic stability, low bioavailability, inadequate distribution and rapid blood clearance. Moreover, injection of PACAP to human can induce peripheral adverse side effects. Therefore, targeted chemical modifications and/or conjugation of PACAP to different macromolecules are required to improve the pharmacokinetic and pharmacological properties of PACAP. This review presents the chemical, biochemical and pharmacological strategies that are currently under development to convert PACAP from a hypophysiotropic neurohormone into a clinically relevant neuroprotective drug.


Subject(s)
Drug Discovery/methods , Neuroprotective Agents/therapeutic use , Pituitary Adenylate Cyclase-Activating Polypeptide/physiology , Pituitary Adenylate Cyclase-Activating Polypeptide/therapeutic use , Animals , Apoptosis/drug effects , Astrocytes/drug effects , Astrocytes/metabolism , Astrocytes/pathology , Brain Injuries/drug therapy , Brain Injuries/immunology , Brain Injuries/pathology , Brain Ischemia/drug therapy , Brain Ischemia/immunology , Brain Ischemia/pathology , Cell Survival/drug effects , Cytokines/immunology , Humans , Nerve Growth Factors/metabolism , Neuroprotective Agents/adverse effects , Pituitary Adenylate Cyclase-Activating Polypeptide/adverse effects , Pituitary Adenylate Cyclase-Activating Polypeptide/metabolism , Receptors, G-Protein-Coupled/metabolism , Receptors, Pituitary Adenylate Cyclase-Activating Polypeptide/metabolism , Receptors, Pituitary Adenylate Cyclase-Activating Polypeptide/physiology
12.
Mol Cell Neurosci ; 43(1): 60-71, 2010 Jan.
Article in English | MEDLINE | ID: mdl-19286457

ABSTRACT

A body of evidence points to the matricial CCN proteins as key regulators of organogenesis. NOV/CCN3, a founder CCN member, is expressed in the developing central nervous system but its functions during neural development have not been studied yet. Here we describe the pattern of NOV expression during rat cerebellar postnatal development and show that NOV expression increases during the second postnatal week, a critical period for the maturation of granule neuron precursors (GNP). NOV transcripts are specifically produced by Purkinje neurons and NOV protein localises extracellularly in the molecular layer and the inner part of the external granule layer, at a key position to control GNP proliferation and migration. In vitro, NOV reduces Sonic Hedgehog-induced GNP proliferation through beta3 integrins and stimulation of GSK3-beta activity whereas NOV stimulates GNP migration through distinct RGD-dependent integrins. These findings identify a new paracrine role of NOV in the development of cerebellar granule neurons.


Subject(s)
Cerebellum/cytology , Nephroblastoma Overexpressed Protein/metabolism , Neurons/physiology , Stem Cells/physiology , Animals , Cell Movement/physiology , Cell Proliferation , Cells, Cultured , Cerebellum/growth & development , Cerebellum/metabolism , Glycogen Synthase Kinase 3/metabolism , Glycogen Synthase Kinase 3 beta , Hedgehog Proteins/genetics , Hedgehog Proteins/metabolism , Humans , Integrin beta3/metabolism , Nephroblastoma Overexpressed Protein/genetics , Neurons/cytology , Oligopeptides/metabolism , Paracrine Communication/physiology , Purkinje Cells/cytology , Purkinje Cells/physiology , Rats , Rats, Wistar , Stem Cells/cytology
13.
Curr Med Chem ; 16(33): 4462-80, 2009.
Article in English | MEDLINE | ID: mdl-19835562

ABSTRACT

Pituitary adenylate cyclase-activating polypeptide (PACAP) is a 38-amino acid peptide that was initially isolated from hypothalamus extracts on the basis of its ability to stimulate the production of cAMP in cultured pituitary cells. Recent studies have shown that PACAP exerts potent neuroprotective effects not only in vitro but also in in vivo models of Parkinson's disease, Huntington's disease, traumatic brain injury and stroke. The protective effects of PACAP are based on its capacity to prevent neuronal apoptosis by acting directly on neurons or indirectly through the release of neuroprotective factors by astrocytes. These biological activities are mainly mediated through activation of the PAC1 receptor which is currently considered as a potential target for the treatment of neurodegenerative diseases. However, the use of native PACAP, the endogenous ligand of PAC1, as an efficient neuroprotective drug is actually limited by its rapid degradation. Moreover, injection of PACAP to human induces peripheral side effects which are mainly mediated through VPAC1 and VPAC2 receptors. Strategies to overcome these compromising conditions include the development of metabolically stable analogs of PACAP acting as selective agonists of the PAC1 receptor. This review presents an overview of the structure-activity relationships of PACAP and summarizes the molecular and conformational requirements for activation of PAC1 receptor. The applicability of PACAP analogs as therapeutic agents for treatment of neurodegenerative diseases is also discussed.


Subject(s)
Neuroprotective Agents/chemistry , Pituitary Adenylate Cyclase-Activating Polypeptide/chemistry , Amino Acid Sequence , Animals , Humans , Molecular Sequence Data , Neuroprotective Agents/pharmacology , Pituitary Adenylate Cyclase-Activating Polypeptide/metabolism , Rats , Receptors, Pituitary Adenylate Cyclase-Activating Polypeptide/metabolism , Structure-Activity Relationship
14.
Neuroscience ; 160(2): 434-51, 2009 May 05.
Article in English | MEDLINE | ID: mdl-19236905

ABSTRACT

The distribution and density of pituitary adenylate cyclase-activating polypeptide (PACAP) binding sites have been investigated in the brain of the primates Jacchus callithrix (marmoset) and Macaca fascicularis (macaque) using [(125)I]-PACAP27 as a radioligand. PACAP binding sites were widely expressed in the brain of these two species with particularly high densities in the septum, hypothalamus and habenula. A moderate density of recognition sites was seen in all subdivisions of the cerebral cortex with a heterogenous distribution, the highest concentrations occurring in layers I and VI while the underlying white matter was almost devoid of binding sites. Reverse transcriptase-polymerase chain reaction (RT-PCR) analysis revealed intense expression of the mRNAs encoding the short and hop-1 variants of pituitary adenylate cyclase-activating polypeptide-specific receptor (PAC1-R) in the cortex of both marmoset and macaque, whereas vasoactive intestinal polypeptide/pituitary adenylate cyclase-activating polypeptide mutual receptor, subtype 1 (VPAC1-R) and vasoactive intestinal polypeptide/pituitary adenylate cyclase-activating polypeptide mutual receptor, subtype 2 (VPAC2-R) mRNAs were expressed at a much lower level. In situ hybridization histochemistry showed intense expression of PAC1-R and weak expression of VPAC1-R mRNAs in layer IV of the cerebral cortex. Incubation of cortical tissue slices with PACAP induced a dose-dependent stimulation of cyclic AMP formation, indicating that PACAP binding sites correspond to functional receptors. Moreover, treatment of primate cortical slices with 100 nM PACAP significantly reduced the activity of caspase-3, a key enzyme of the apoptotic cascade. The present results indicate that PACAP should exert the same neuroprotective effect in the brain of primates as in rodents and suggest that PAC1-R agonists may have a therapeutic value to prevent neuronal cell death after stroke or in specific neurodegenerative diseases.


Subject(s)
Brain Mapping , Brain/metabolism , Pituitary Adenylate Cyclase-Activating Polypeptide/metabolism , Receptors, Pituitary Adenylate Cyclase-Activating Polypeptide/metabolism , Animals , Callithrix , Female , Habenula/metabolism , Hypothalamus/metabolism , Macaca fascicularis , Male , Pituitary Adenylate Cyclase-Activating Polypeptide/genetics , RNA, Messenger/analysis , Receptors, Pituitary Adenylate Cyclase-Activating Polypeptide/classification , Receptors, Pituitary Adenylate Cyclase-Activating Polypeptide/genetics , Septum of Brain/metabolism , Species Specificity , Tissue Distribution
15.
J Mol Neurosci ; 36(1-3): 8-15, 2008 Nov.
Article in English | MEDLINE | ID: mdl-18574733

ABSTRACT

Pituitary adenylate cyclase-activating polypeptide (PACAP) is a neuropeptide that belongs to the secretin/glucagon/vasoactive intestinal polypeptide superfamily. The PACAPergic system is actively expressed in the developing cerebellum of mammals. In particular, PACAP receptors are expressed by granule cell precursors suggesting a role of the peptide in neurogenesis of this cell type. Consistent with this hypothesis, several studies reported antiapoptotic effects of PACAP in the developing cerebellum. On the other hand, the sphingomyelin metabolites ceramides are recognized as important signaling molecules that play pivotal roles during neuronal development. Ceramides, which production can be induced by death factors such as FasL or TNFalpha, are involved in the control of cell survival during brain development through activation of caspase-dependent mechanisms. The present review focuses on the interactions between PACAP and ceramides in the control of granule cell survival and on the transduction mechanisms associated with the anti- and proapoptotic effects of PACAP and ceramides, respectively.


Subject(s)
Apoptosis/physiology , Ceramides/metabolism , Cerebellum , Neurons/physiology , Pituitary Adenylate Cyclase-Activating Polypeptide/metabolism , Animals , Ceramides/chemistry , Cerebellum/cytology , Cerebellum/growth & development , Molecular Structure , Neurons/cytology , Second Messenger Systems/physiology
16.
Anal Bioanal Chem ; 390(7): 1861-71, 2008 Apr.
Article in English | MEDLINE | ID: mdl-18278588

ABSTRACT

Bacterial surface-associated proteins play crucial roles in host-pathogen interactions and pathogenesis. The identification of these proteins represents an important goal of bacterial proteomics for vaccine development, but also for environmental concerns such as microbial biosensing. Here, we developed such an approach for Legionella pneumophila, a bacterium that causes severe pneumonia. We propose a complementary strategy consisting of (1) a fluorescent labelling of surface-exposed proteins in parallel with (2) a fractionation of the outer-membrane protein extract. These two distinct protein populations were subsequently separated using two-dimensional gel electrophoresis and characterised by mass spectrometry. Within these populations, we found proteins which were expected for the compartments studied, but also a great number of proteins never experimentally described, and also a non-negligible fraction of proteins never described in these fractions. These data provided new routes of inspection for transport and host recognition for Legionella pneumophila. In addition, these results on the membranome and surfaceome show that Legionella in the stationary phase of growth possesses the major determinants to infect host cells.


Subject(s)
Bacterial Outer Membrane Proteins/chemistry , Fluorescent Dyes/chemistry , Legionella pneumophila/chemistry , Peptide Mapping/methods , Proteomics/methods , Cell Fractionation , Electrophoresis, Gel, Two-Dimensional/methods , Legionella pneumophila/growth & development , Reproducibility of Results , Sensitivity and Specificity , Staining and Labeling , Surface Properties
17.
J Neuroendocrinol ; 19(5): 321-7, 2007 May.
Article in English | MEDLINE | ID: mdl-17425606

ABSTRACT

Pituitary adenylate cyclase-activating polypeptide (PACAP) was originally isolated from ovine hypothalamus on the basis of its hypophysiotrophic activity. It has subsequently been shown that PACAP and its receptors are widely distributed in the central nervous system of adult mammals, indicating that PACAP may act as a neurotransmitter and/or neuromodulator. It has also been found that PACAP and its receptors are expressed in germinative neuroepithelia, suggesting that PACAP could be involved in neurogenesis. There is now compelling evidence that PACAP exerts neurotrophic activities in the developing cerebellum and in embryonic stem (ES) cells. In particular, the presence of PACAP receptors has been demonstrated in the granule layer of the immature cerebellar cortex, and PACAP has been shown to promote survival, inhibit migration and activate neurite outgrowth of granule cell precursors. In cerebellar neuroblasts, PACAP is a potent inhibitor of the mitochondrial apoptotic pathway through activation of the MAPkinase extracellular regulated kinase. ES cells and embryoid bodies (EB) also express PACAP receptors and PACAP facilitates neuronal orientation and induces the appearance of an electrophysiological activity. Taken together, the anti-apoptotic and pro-differentiating effects of PACAP characterised in cerebellar neuroblasts as well as ES and EB cells indicate that PACAP acts not only as a neurohormone and a neurotransmitter, but also as a growth factor.


Subject(s)
Apoptosis/physiology , Cell Differentiation/physiology , Cerebellum/cytology , Embryonic Stem Cells/cytology , Neurons/cytology , Pituitary Adenylate Cyclase-Activating Polypeptide/physiology , Animals , Cerebellum/growth & development , Cerebellum/physiology , Embryonic Stem Cells/physiology , Gene Expression Regulation, Developmental , Humans , Nerve Growth Factor/physiology , Neurons/physiology
18.
Neuroscience ; 146(2): 697-712, 2007 May 11.
Article in English | MEDLINE | ID: mdl-17383102

ABSTRACT

Migration of immature neurons is essential for forming the cortical layers and nuclei. Impairment of migration results in aberrant neuronal cytoarchitecture, which leads to various neurological disorders. Neurons alter the mode, tempo and rate of migration when they translocate through different cortical layers, but little is known about the mechanisms underlying this process. Here we show that endogenous pituitary adenylate cyclase-activating polypeptide (PACAP) has short-term and cortical-layer-specific effects on granule cell migration in the early postnatal mouse cerebellum. Application of exogenous PACAP significantly slowed the migration of isolated granule cells and shortened the leading process in the microexplant cultures of the postnatal day (P)0-3 cerebella. Interestingly, in the cerebellar slices of P10 mice, application of exogenous PACAP significantly inhibited granule cell migration in the external granular layer (EGL) and molecular layer (ML), but failed to alter the movement in the Purkinje cell layer (PCL) and internal granular layer (IGL). In contrast, application of PACAP antagonist accelerated granule cell migration in the PCL, but did not change the movement in the EGL, ML and IGL. Inhibition of the cAMP signaling and the activity of phospholipase C significantly reduced the effects of exogenous PACAP on granule cell migration. The PACAP action on granule cell migration was transient, and lasted for approximately 2 h. The duration of PACAP action on granule cell migration was determined by the desensitization of its receptors and prolonged by inhibiting the protein kinase C. Endogenous PACAP was present sporadically in the bottom of the ML, intensively in the PCL, and throughout the IGL. Collectively, these results indicated that PACAP acts on granule cell migration as "a brake (stop signal) for cell movement." Furthermore, these results suggest that endogenous PACAP slows granule cell migration when the cells enter the PACAP-rich PCL, and 2 h later the desensitization of PACAP receptors allows the cells to accelerate the rate of migration and to actively move within the PACAP-rich IGL. Therefore, endogenous PACAP may provide a cue that regulates granule cell migration in a cerebellar cortical-layer-specific manner.


Subject(s)
Cell Movement/drug effects , Cerebellar Cortex/cytology , Neurons/physiology , Pituitary Adenylate Cyclase-Activating Polypeptide/pharmacology , Analysis of Variance , Animals , Animals, Newborn , Calbindins , Calcium/metabolism , Cells, Cultured , Drug Interactions , Enzyme Inhibitors/pharmacology , In Vitro Techniques , Mice , Neurons/drug effects , Peptide Fragments/pharmacology , Pituitary Adenylate Cyclase-Activating Polypeptide/antagonists & inhibitors , Pituitary Adenylate Cyclase-Activating Polypeptide/metabolism , S100 Calcium Binding Protein G/metabolism , Time Factors
19.
Eur J Neurosci ; 19(6): 1446-58, 2004 Mar.
Article in English | MEDLINE | ID: mdl-15066141

ABSTRACT

Abstract Activation of potassium (K(+)) currents plays a critical role in the control of programmed cell death. Because pituitary adenylate cyclase-activating polypeptide (PACAP) has been shown to inhibit the apoptotic cascade in the cerebellar cortex during development, we have investigated the effect of PACAP on K(+) currents in cultured cerebellar granule cells using the patch-clamp technique in the whole-cell configuration. Two types of outward K(+) currents, a transient K(+) current (I(A)) and a delayed rectifier K(+) current (I(K)) were characterized using two different voltage protocols and specific inhibitors of K(+) channels. Application of PACAP induced a reversible reduction of the I(K) amplitude, but did not affect I(A), while the PACAP-related peptide vasoactive intestinal polypeptide had no effect on either types of K(+) currents. Repeated applications of PACAP induced gradual attenuation of the electrophysiological response. In the presence of guanosine 5'-[gammathio]triphosphate (GTPgammaS), PACAP provoked a marked and irreversible I(K) depression, whereas cell dialysis with guanosine 5'-[betathio]diphosphate GDPbetaS totally abolished the effect of PACAP. Pre-treatment of the cells with pertussis toxin did not modify the effect of PACAP on I(K). In contrast, cholera toxin suppressed the PACAP-induced inhibition of I(K). Exposure of granule cells to dibutyryl cyclic adenosine monophosphate (dbcAMP) mimicked the inhibitory effect of PACAP on I(K). Addition of the specific protein kinase A inhibitor H89 in the patch pipette solution prevented the reduction of I(K) induced by both PACAP and dbcAMP. PACAP provoked a sustained increase of the resting membrane potential in cerebellar granule cells cultured either in high or low KCl-containing medium, and this long-term depolarizing effect of PACAP was mimicked by the I(K) specific blocker tetraethylammonium chloride (TEA). In addition, pre-incubation of granule cells with TEA suppressed the effect of PACAP on resting membrane potential. TEA mimicked the neuroprotective effect of PACAP against ethanol-induced apoptotic cell death, and the increase of caspase-3 activity observed after exposure of granule cells to ethanol was also significantly inhibited by TEA. Taken together, the present results demonstrate that, in rat cerebellar granule cells, PACAP reduces the delayed outward rectifier K(+) current by activating a type 1 PACAP (PAC1) receptor coupled to the adenylyl cyclase/protein kinase A pathway through a cholera toxin-sensitive Gs protein. Our data also show that PACAP and TEA induce long-term depolarization of the resting membrane potential, promote cell survival and inhibit caspase-3 activity, suggesting that PACAP-evoked inhibition of I(K) contributes to the anti-apoptotic effect of the peptide on cerebellar granule cells.


Subject(s)
Apoptosis/drug effects , Cyclic AMP-Dependent Protein Kinases/metabolism , Cyclic AMP/metabolism , Guanosine Diphosphate/analogs & derivatives , Neuropeptides/pharmacology , Potassium Channels, Voltage-Gated , Potassium Channels/drug effects , Signal Transduction/drug effects , Sulfonamides , 4-Aminopyridine/pharmacology , Animals , Animals, Newborn , Caspase 3 , Caspases/metabolism , Cell Survival/drug effects , Cells, Cultured , Central Nervous System Depressants/pharmacology , Cerebellum/cytology , Cholera Toxin/pharmacology , Cyclic AMP/physiology , Cyclic AMP-Dependent Protein Kinases/physiology , Delayed Rectifier Potassium Channels , Drug Interactions , Electric Conductivity , Enzyme Inhibitors/pharmacology , Ethanol/pharmacology , Guanosine 5'-O-(3-Thiotriphosphate)/pharmacology , Guanosine Diphosphate/pharmacology , Isoquinolines/pharmacology , Membrane Potentials/drug effects , Models, Neurological , Neural Inhibition/drug effects , Neurons/drug effects , Patch-Clamp Techniques/methods , Peptide Fragments/pharmacology , Pertussis Toxin/pharmacology , Pituitary Adenylate Cyclase-Activating Polypeptide , Potassium Channel Blockers/pharmacology , Rats , Rats, Wistar , Tetraethylammonium/pharmacology , Tetrodotoxin/pharmacology , Thionucleotides/pharmacology , Time Factors , Vasoactive Intestinal Peptide/pharmacology
20.
Science ; 296(5573): 1648-9, 2002 May 31.
Article in English | MEDLINE | ID: mdl-12040181

ABSTRACT

A key issue in signal transduction is how signaling pathways common to many systems-so-called canonical signaling cassettes-integrate signals from molecules having a wide spectrum of activities, such as hormones and neurotrophins, to deliver distinct biological outcomes. The neuroendocrine cell line PC12, derived from rat pheochromocytoma, provides an example of how one canonical signaling cassette-the Raf --> mitogen-activated protein kinase kinase (MEK) --> extracellular signal-regulated kinase (ERK) pathway-can promote distinct outcomes, which in this case include neuritogenesis, gene induction, and proliferation. Two growth hormones, epidermal growth factor (EGF) and nerve growth factor (NGF), use the same pathway to cause PC12 proliferation and differentiation, respectively. In addition, pituitary adenylate cyclase-activating polypeptide (PACAP), a neurotransmitter that also causes differentiation, uses the same canonical cassette as NGF but in a different way. The Connections Map for PC12 Cell Differentiation brings into focus the complex array of specific cellular responses that rely on canonical signal transduction systems.


Subject(s)
Cell Differentiation , MAP Kinase Signaling System , PC12 Cells/physiology , Animals , Cell Division , Cyclic AMP/metabolism , Epidermal Growth Factor/metabolism , Epidermal Growth Factor/pharmacology , Mitogen-Activated Protein Kinases/metabolism , Models, Biological , Nerve Growth Factor/metabolism , Nerve Growth Factor/pharmacology , Neurites/physiology , Neuropeptides/metabolism , Neuropeptides/pharmacology , Pituitary Adenylate Cyclase-Activating Polypeptide , Rats , Receptor, trkA/metabolism , Receptors, Cell Surface/metabolism , Response Elements , Transcription, Genetic
SELECTION OF CITATIONS
SEARCH DETAIL
...