Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 24
Filter
Add more filters










Publication year range
1.
Neuroscience ; 199: 103-15, 2011 Dec 29.
Article in English | MEDLINE | ID: mdl-22001490

ABSTRACT

Identification of novel molecules that can induce neuronal differentiation of embryonic stem (ES) cells is essential for deciphering the molecular mechanisms of early development and for exploring cell therapy approaches. Pituitary adenylate cyclase-activating polypeptide (PACAP) and vasoactive intestinal polypeptide (VIP) are known to be implicated early during ontogenesis in cell proliferation and neuronal differentiation. The aim of the present study was to determine the effects of VIP and PACAP on functional differentiation of ES cells. Quantitative-reverse transcription-polymerase chain reaction analysis showed an inversion of the expression pattern of PAC1 and VPAC1 receptors with time. ES cells expressed genes encoding extracellular signal-regulated kinase 1 and 2 and c-jun amino terminal kinase1. ES cells also expressed T-type α1I and α1G, L-type α1C and α1D, and N-type α1B calcium channel subunit mRNAs. Both peptides modified the shape of undifferentiated ES cells into bipolar cells expressing the neuronal marker neuron-specific enolase (NSE). Immunostaining indicated that PACAP intensified T-type α1I subunit immunoreactivity, whereas VIP increased L-types α1C and α1D, as well as N-type α1B subunit. Electrophysiological recording showed that VIP and PACAP enhanced transient calcium current. Moreover, VIP generated sustained calcium current. These findings demonstrate that PACAP and VIP induce morphological and functional differentiation of ES cells into a neuronal phenotype. Both peptides promote functional maturation of calcium channel subunits, suggesting that they can facilitate the genesis of cellular excitability.


Subject(s)
Cell Differentiation/physiology , Embryonic Stem Cells/metabolism , Gene Expression Regulation, Developmental/physiology , Neural Stem Cells/metabolism , Neurogenesis/physiology , Pituitary Adenylate Cyclase-Activating Polypeptide/metabolism , Vasoactive Intestinal Peptide/metabolism , Animals , Calcium Channels/genetics , Calcium Channels/metabolism , Embryonic Stem Cells/cytology , Gene Expression Profiling , Immunohistochemistry , Mice , Neural Stem Cells/cytology , Pituitary Adenylate Cyclase-Activating Polypeptide/genetics , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Vasoactive Intestinal Peptide/genetics
2.
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
3.
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
4.
Ecology ; 89(2): 555-66, 2008 Feb.
Article in English | MEDLINE | ID: mdl-18409444

ABSTRACT

The Mahalanobis distances have been introduced in habitat selection studies for the estimation of environmental suitability maps (ESMs). The pixels of raster maps of a given area correspond to points in the multidimensional space defined by the mapped environmental variables (ecological space). The Mahalanobis distances measure the distances in this space between these points and the mean of the ecological niche (i.e., the hypothesized optimum for the species) regarding the structure of the niche. The map of these distances over the area of interest is an estimated ESM. Several authors recently noted that the use of a single optimum for the niche of a species may lead to biased predictions of animal occurrence. They proposed to use instead a minimum set of basic habitat requirements, found by partitioning the Mahalanobis distances into a restricted set of biologically meaningful axes. However, the statistical approach they proposed does not take into account the environmental conditions on the area where the niche was sampled (i.e., the environmental availability), and we show that including this availability is necessary. We used their approach as a basis to develop a new exploratory tool, the Mahalanobis distance factor analysis (MADIFA), which performs an additive partitioning of the Mahalanobis distances taking into account this availability. The basic habitat requirements of a species can be derived from the axes of the MADIFA. This method can also be used to compute ESMs using only this small number of basic requirements, therefore including only the biologically relevant information. We also prove that the MADIFA is complementary to the commonly used ecological-niche factor analysis (ENFA). We used the MADIFA method to analyze the niche of the chamois Rupicapra rupicapra in a mountainous area. This method adds to the existing set of tools for the description of the niche.


Subject(s)
Ecosystem , Environment , Models, Biological , Rupicapra/physiology , Algorithms , Altitude , Animals , Factor Analysis, Statistical , Rupicapra/growth & development
5.
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
6.
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
7.
Eur J Neurosci ; 15(9): 1451-60, 2002 May.
Article in English | MEDLINE | ID: mdl-12028355

ABSTRACT

Oxidative stress, resulting from accumulation of reactive oxygen species, plays a critical role in neuronal cell death associated with neurodegenerative diseases and stroke. In the present study, we have investigated the potential neuroprotective effect of pituitary adenylate cyclase-activating polypeptide (PACAP) on oxidative stress-induced apoptosis. Incubation of cerebellar granule cells with PACAP inhibited hydrogen peroxide-evoked cell death in a concentration-dependent manner. The effect of PACAP on granule cell survival was not mimicked by vasoactive intestinal polypeptide and was blocked by the antagonist PACAP6-38. The protective action of PACAP upon hydrogen peroxide-induced neuronal cell death was abolished by the MAP-kinase kinase (MEK) inhibitor U0126 and mimicked by the caspase-3 inhibitor Z-DEVD-FMK. PACAP markedly inhibited hydrogen peroxide-evoked caspase-3 activation and DNA fragmentation. Taken together, these data indicate that PACAP, acting through PACAP receptor type 1, exerts a potent protective effect against neuronal degeneration induced by hydrogen peroxide. The anti-apoptotic effect of PACAP is mediated through the MAP-kinase pathway and can be accounted for by inhibition of caspase-3 activation resulting from oxidative stress.


Subject(s)
Apoptosis/drug effects , Hypoxia-Ischemia, Brain/drug therapy , Neurodegenerative Diseases/drug therapy , Neurons/drug effects , Neuropeptides/pharmacology , Neuroprotective Agents/pharmacology , Oxidative Stress/drug effects , Animals , Apoptosis/physiology , Bucladesine/pharmacology , Caspase 3 , Caspases/drug effects , Caspases/metabolism , Cells, Cultured , Cerebellar Cortex/cytology , Cerebellar Cortex/drug effects , Cerebellar Cortex/metabolism , DNA Fragmentation/drug effects , DNA Fragmentation/physiology , Drug Interactions/physiology , Enzyme Inhibitors/pharmacology , Hydrogen Peroxide/pharmacology , Hypoxia-Ischemia, Brain/metabolism , Hypoxia-Ischemia, Brain/physiopathology , MAP Kinase Signaling System/drug effects , MAP Kinase Signaling System/physiology , Membrane Potentials/drug effects , Membrane Potentials/physiology , Mitochondria/drug effects , Mitochondria/metabolism , Neurodegenerative Diseases/metabolism , Neurodegenerative Diseases/physiopathology , Neurons/metabolism , Neuropeptides/therapeutic use , Neuroprotective Agents/therapeutic use , Oxidative Stress/physiology , Pituitary Adenylate Cyclase-Activating Polypeptide , Rats , Rats, Wistar , Signal Transduction/drug effects , Signal Transduction/physiology
8.
Proc Natl Acad Sci U S A ; 97(24): 13390-5, 2000 Nov 21.
Article in English | MEDLINE | ID: mdl-11087878

ABSTRACT

Caspase-3 knockout mice exhibit thickening of the internal granule cell layer of the cerebellum. Concurrently, it has been shown that intracerebral injection of pituitary adenylate cyclase-activating polypeptide (PACAP) induces a transient increase of the thickness of the cerebellar cortex. In the present study, we have investigated the possible effect of PACAP on caspase activity in cultured cerebellar granule cells from 8-day-old rat. Incubation of granule neurons with PACAP for 24 h promoted cell survival and prevented DNA fragmentation. Exposure of cerebellar granule cells to the specific caspase-3 inhibitor N-benzyloxycarbonyl-Asp-Glu-Val-Asp fluoromethylketone (Z-DEVD-FMK) for 24 h markedly enhanced cell survival and inhibited apoptotic cell death. Time-course studies revealed that PACAP causes a prolonged inhibition of caspase-3 activity without affecting caspase-1. Administration of graded concentrations of PACAP for 3 h induced a dose-dependent inhibition of caspase-3 activity. Incubation of granule cells with both dibutyryl-cAMP (dbcAMP) and phorbol 12-myristate 13-acetate (PMA) mimicked the inhibitory effect of PACAP on caspase-3. Cotreatment of cultured neurons with the protein kinase A inhibitor H89 and the protein kinase C inhibitor chelerythrine abrogated the effect of PACAP on caspase-3 activity. In contrast, the ERK kinase inhibitor U0126 did not affect the action of PACAP on caspase-3 activity. These data demonstrate that PACAP prevents cerebellar granule neurons from apoptotic cell death through a protein kinase A- and protein kinase C-dependent inhibition of caspase-3 activity.


Subject(s)
Apoptosis/physiology , Caspases/metabolism , Cerebellum/physiology , Cysteine Proteinase Inhibitors/pharmacology , Neurons/physiology , Neuropeptides/pharmacology , Neuroprotective Agents/pharmacology , Animals , Apoptosis/drug effects , Caspase 3 , Caspase Inhibitors , Cell Survival/drug effects , Cells, Cultured , Cerebellum/cytology , Neurons/cytology , Neurons/drug effects , Oligopeptides/pharmacology , Pituitary Adenylate Cyclase-Activating Polypeptide , Rats , Rats, Wistar , Vasoactive Intestinal Peptide/pharmacology
9.
J Comp Neurol ; 425(4): 495-509, 2000 Oct 02.
Article in English | MEDLINE | ID: mdl-10975876

ABSTRACT

The distribution and density of pituitary adenylate cyclase-activating polypeptide (PACAP) binding sites as well as PACAP-specific receptor 1 (PAC1-R), vasoactive intestinal polypeptide/PACAP receptor 1 (VPAC1-R), and VPAC2-R mRNAs have been investigated in the rat brain from embryonic day 14 (E14) to postnatal day 8 (P8). Significant numbers of binding sites for the radioiodinated, 27-amino-acid form of PACAP were detected as early as E14 in the neuroepithelia of the metencephalon and the myelencephalon. From E14 to E21, the density of binding sites in the germinative areas increased by 3- to 5-fold. From birth to P12, the density of binding sites gradually declined in all neuroepithelia except in the external granule cell layer of the cerebellum, where the level of binding sites remained high during the first postnatal weeks. Only low to moderate densities of PACAP binding sites were found in regions other than the germinative areas, with the exception of the internal granule cell layer of the cerebellum, which contained a high density of sites. The localization of PACAP receptor mRNAs was investigated by in situ hybridization using [(35)S] uridine triphosphate-specific riboprobes. The evolution of the distribution of PAC1-R and VPAC1-R mRNAs was very similar to that of PACAP binding sites, the concentration of VPAC1-R mRNA being much lower than that of PAC1-R mRNA. In contrast, intense expression of VPAC2-R mRNA was observed in brain regions other than germinative areas, such as the suprachiasmatic, ventral thalamic, and dorsolateral geniculate nuclei. The discrete localization of PACAP binding sites as well as PAC1-R and VPAC1-R mRNAs in neuroepithelia during embryonic life and postnatal development strongly suggests that PACAP, acting through PAC1-R and/or VPAC1-R, may play a crucial role in the regulation of neurogenesis in the rat brain.


Subject(s)
Aging/metabolism , Brain/metabolism , Neuropeptides/metabolism , RNA, Messenger/metabolism , Rats/metabolism , Receptors, Pituitary Hormone/genetics , Animals , Autoradiography , Binding Sites , Brain/growth & development , In Situ Hybridization , Pituitary Adenylate Cyclase-Activating Polypeptide , Rats, Wistar , Receptors, Pituitary Adenylate Cyclase-Activating Polypeptide , Receptors, Pituitary Adenylate Cyclase-Activating Polypeptide , Receptors, Vasoactive Intestinal Peptide/genetics , Receptors, Vasoactive Intestinal Peptide, Type II , Receptors, Vasoactive Intestinal Polypeptide, Type I , Reverse Transcriptase Polymerase Chain Reaction , Tissue Distribution
10.
Pharmacol Rev ; 52(2): 269-324, 2000 Jun.
Article in English | MEDLINE | ID: mdl-10835102

ABSTRACT

Pituitary adenylate cyclase-activating polypeptide (PACAP) is a 38-amino acid peptide that was first isolated from ovine hypothalamic extracts on the basis of its ability to stimulate cAMP formation in anterior pituitary cells. PACAP belongs to the vasoactive intestinal polypeptide (VIP)-glucagon-growth hormone releasing factor-secretin superfamily. The sequence of PACAP has been remarkably well conserved during the evolution from protochordate to mammals, suggesting that PACAP is involved in the regulation of important biological functions. PACAP is widely distributed in the brain and peripheral organs, notably in the endocrine pancreas, gonads, and respiratory and urogenital tracts. Characterization of the PACAP precursor has revealed the existence of a PACAP-related peptide whose activity remains unknown. Two types of PACAP binding sites have been characterized. Type I binding sites exhibit a high affinity for PACAP and a much lower affinity for VIP whereas type II binding sites have similar affinity for PACAP and VIP. Molecular cloning of PACAP receptors has shown the existence of three distinct receptor subtypes, the PACAP-specific PAC1 receptor, which is coupled to several transduction systems, and the two PACAP/VIP-indifferent VPAC1 and VPAC2 receptors, which are primarily coupled to adenylyl cyclase. PAC1 receptors are particularly abundant in the brain and pituitary and adrenal glands whereas VPAC receptors are expressed mainly in the lung, liver, and testis. The wide distribution of PACAP and PACAP receptors has led to an explosion of studies aimed at determining the pharmacological effects and biological functions of the peptide. This report reviews the current knowledge concerning the multiple actions of PACAP in the central nervous system and in various peripheral organs including the endocrine glands, the airways, and the cardiovascular and immune systems, as well as the different effects of PACAP on a number of tumor cell types.


Subject(s)
Neuropeptides/pharmacology , Receptors, Pituitary Hormone/drug effects , Amino Acid Sequence , Animals , Humans , Molecular Sequence Data , Neuropeptides/chemistry , Neuropeptides/genetics , Neuropeptides/physiology , Pituitary Adenylate Cyclase-Activating Polypeptide , Receptors, Pituitary Adenylate Cyclase-Activating Polypeptide , Receptors, Pituitary Adenylate Cyclase-Activating Polypeptide , Receptors, Pituitary Hormone/genetics , Receptors, Vasoactive Intestinal Peptide, Type II , Receptors, Vasoactive Intestinal Polypeptide, Type I
11.
Ann N Y Acad Sci ; 921: 293-9, 2000.
Article in English | MEDLINE | ID: mdl-11193838

ABSTRACT

During development of the rat cerebellum, PAC1 receptors are transiently expressed by neuroblasts of the external granule cell layer (EGL). We have previously shown that PACAP is a potent stimulator of granule cell survival in vitro. In the study reported in this paper, we have investigated the effect of PACAP on the development of the rat cerebellar cortex in vivo. PACAP induces a transient increase in the volume of the cerebellar cortex, with a maximum effect at postnatal day 12, which can be accounted for by an increase in the number of granule cells in the EGL, the molecular layer, and the internal granule cell layer (IGL). The effect of PACAP on the number of granule cells is blocked by the antagonist PACAP(6-38), which, by itself, produces a slight inhibition of the number of granule cells in the IGL. These data indicate that PACAP activates proliferation and/or inhibits programmed cell death of granule cells in the developing rat cerebellum. PACAP also stimulates neuronal migration from the EGL to the IGL. Thus, it appears that PACAP can act in vivo as a neurotrophic factor controlling histogenesis of the cerebellar cortex.


Subject(s)
Cerebellar Cortex/drug effects , Cerebellar Cortex/growth & development , Nerve Growth Factors/pharmacology , Neuropeptides/pharmacology , Animals , Apoptosis/drug effects , Cell Division/drug effects , Cell Movement/drug effects , Cerebellar Cortex/cytology , Nerve Growth Factors/physiology , Neuropeptides/antagonists & inhibitors , Neuropeptides/physiology , Peptide Fragments/pharmacology , Pituitary Adenylate Cyclase-Activating Polypeptide , Rats
13.
Eur J Neurosci ; 11(11): 4051-8, 1999 Nov.
Article in English | MEDLINE | ID: mdl-10583493

ABSTRACT

In rats, rapid eye movement (REM) sleep can be elicited by microinjection of vasoactive intestinal polypeptide (VIP) into the oral pontine reticular nucleus (PnO). In the present study, we investigated whether this area could also be a REM-promoting target for a peptide closely related to VIP: the pituitary adenylyl cyclase-activating polypeptide (PACAP). When administered into the posterior part of the PnO, but not in nearby areas, of freely moving chronically implanted rats, PACAP-27 and PACAP-38 (0.3 and 3 pmol) induced a marked enhancement (60-85% over baseline) of REM sleep for 8 h that could be prevented by prior infusion of the antagonist PACAP-(6-27) (3 pmol) into the same site. Moreover, injections of PACAP into the centre of the posterior PnO resulted in REM sleep enhancement which could last for up to 11 consecutive days. Quantitative autoradiography using [125I]PACAP-27 revealed the presence in the PnO of specific binding sites with high affinity for PACAP-27 and PACAP-38 (IC50 = 2.4 and 3.2 nM, respectively), but very low affinity for VIP (IC50 > 1 microM). These data suggest that PACAP within the PnO may play a key role in REM sleep regulation, and provide evidence for long-term (several days) mechanisms involved in such a control. PAC1 receptors which have a much higher affinity for PACAP than for VIP might mediate this long-term action of PACAP on REM sleep.


Subject(s)
Brain Stem/physiology , Neuropeptides/pharmacology , Neurotransmitter Agents/pharmacology , Pons/physiology , Reticular Formation/physiology , Sleep, REM/drug effects , Animals , Brain Mapping , Brain Stem/drug effects , Male , Microinjections , Neuropeptides/administration & dosage , Neurotransmitter Agents/administration & dosage , Peptide Fragments/pharmacology , Pituitary Adenylate Cyclase-Activating Polypeptide , Pons/drug effects , Rats , Rats, Sprague-Dawley , Reticular Formation/drug effects , Sleep, REM/physiology , Time Factors , Vasoactive Intestinal Peptide/administration & dosage , Vasoactive Intestinal Peptide/pharmacology , Wakefulness/drug effects
14.
Proc Natl Acad Sci U S A ; 96(16): 9415-20, 1999 Aug 03.
Article in English | MEDLINE | ID: mdl-10430957

ABSTRACT

High concentrations of pituitary adenylate cyclase-activating polypeptide (PACAP) receptors are present in the external granule cell layer of the rat cerebellum during postnatal development. In vitro studies have shown that PACAP promotes cell survival and neurite outgrowth on immature cerebellar granule cells in primary culture. In the present study, we have investigated the effect of PACAP on the development of the cerebellar cortex of 8-day-old rats. Incubation of cultured granule cells for 12 or 18 h with PACAP provoked a significant increase in the rate of incorporation of [(3)H]thymidine in cultured granule cells, suggesting that PACAP could stimulate the proliferation of granule cells. After 96 h of treatment, in vivo administration of PACAP provoked a transient increase in the number of granule cells in the molecular layer and in the internal granule cell layer. In contrast, PACAP did not affect the number of Purkinje cells. The augmentation of the number of granule cells evoked by PACAP was significantly inhibited by the PACAP receptor antagonist PACAP(6-38). Administration of PACAP also caused a significant increase in the volume of the cerebellar cortex. The present study provides evidence that PACAP can act in vivo as a trophic factor during rat brain development. Our data indicate that PACAP increases proliferation and/or inhibits programmed cell death of granule cells, as well as stimulating neuronal migration from the external granule cell layer toward the internal granule cell layer.


Subject(s)
Cerebellum/growth & development , Neurons/physiology , Neuropeptides/pharmacology , Neuroprotective Agents/pharmacology , Aging , Animals , Cell Division/drug effects , Cell Survival/drug effects , Cells, Cultured , Cerebellar Cortex/cytology , Cerebellar Cortex/drug effects , Cerebellar Cortex/growth & development , Cerebellum/cytology , Cerebellum/drug effects , DNA/biosynthesis , Dose-Response Relationship, Drug , Kinetics , Neurites/drug effects , Neurites/physiology , Neurons/cytology , Neurons/drug effects , Pituitary Adenylate Cyclase-Activating Polypeptide , Rats , Rats, Wistar , Thymidine/metabolism
15.
Neuroscience ; 85(3): 887-96, 1998 Aug.
Article in English | MEDLINE | ID: mdl-9639281

ABSTRACT

Melatonin secretion from the mammalian pineal gland is strongly stimulated by noradrenaline and also by vasoactive intestinal polypeptide (VIP) and pituitary adenylate cyclase-activating polypeptide (PACAP). Three types of receptors for VIP and PACAP have been characterized so far: VIP1/PACAP receptors and VIP2/PACAP receptors, which possess similar high affinities for VIP and PACAP, and PACAP1 receptors which exhibit a 100-1000-fold higher affinity for PACAP. The aim of the present study was to characterize the receptor subtype(s) mediating the stimulatory effects of VIP and PACAP on melatonin synthesis in the rat pineal gland. Autoradiographic studies showed that PACAP and VIP were equally potent in displacing binding of radioiodinated PACAP27 from pineal sections. Amplification of pineal complementary DNAs by polymerase chain reaction using specific primers for the different receptor subtypes revealed that all three receptor messenger RNAs are expressed and that VIP1/PACAP receptor messenger RNA was predominant over VIP2/PACAP receptor messenger RNA. In vitro, VIP and PACAP stimulated melatonin synthesis with similar high potency and the effect of the two peptides were not additive. The selective VIP1/PACAP receptor agonists [R16]chicken secretin (1-25) and [K15, R16, L27]VIP(1-7)/growth hormone releasing factor(8-27) were significantly more potent than the selective VIP2/PACAP receptor agonist RO 25-1553 in stimulating melatonin secretion. The stimulatory effects of VIP and PACAP were similarly inhibited by the VIP1/PACAP antagonist [acetyl-His1, D-Phe2, K15, R16, L27]VIP(3-7)/growth hormone releasing factor(8-27). These data strongly suggest that VIP and PACAP exert a stimulatory effect on melatonin synthesis mainly through activation of a pineal VIP1/PACAP receptor subtype.


Subject(s)
Adenylyl Cyclases/metabolism , Pineal Gland/chemistry , Pineal Gland/enzymology , Receptors, Pituitary Hormone/analysis , Receptors, Vasoactive Intestinal Peptide/analysis , Adrenergic beta-Agonists/pharmacology , Animals , Autoradiography , Bronchodilator Agents/pharmacology , Enzyme Activation/drug effects , Gene Expression/physiology , Iodine Radioisotopes , Isoproterenol/pharmacology , Male , Melatonin/metabolism , Neuropeptides/pharmacology , Neurotransmitter Agents/pharmacology , Peptide Fragments/pharmacology , Peptides, Cyclic/pharmacology , Pituitary Adenylate Cyclase-Activating Polypeptide , RNA, Messenger/analysis , Rats , Rats, Wistar , Receptors, Pituitary Adenylate Cyclase-Activating Polypeptide , Receptors, Pituitary Hormone/genetics , Receptors, Pituitary Hormone/metabolism , Receptors, Vasoactive Intestinal Peptide/genetics , Receptors, Vasoactive Intestinal Peptide/metabolism , Signal Transduction/drug effects , Signal Transduction/physiology , Vasoactive Intestinal Peptide/analogs & derivatives , Vasoactive Intestinal Peptide/pharmacology
16.
Neuroscience ; 84(3): 801-12, 1998 Jun.
Article in English | MEDLINE | ID: mdl-9579785

ABSTRACT

A high density of pituitary adenylate cyclase-activating polypeptide (PACAP) receptors coupled to both adenylyl cyclase and phospholipase C is found in the external granule cell layer of the rat cerebellum during postnatal development. It has recently been reported that synthetic PACAP promotes cell survival and neurite outgrowth in immature granule cells. In the present study, we have investigated the transduction pathways that mediate the neurotrophic activity of PACAP in cultured granule cells from eight-day-old rat cerebellum. The effect of PACAP on cell survival was mimicked by dibutyryladenosine 3',5'-cyclic-monophosphate but not phorbol 12-myristate 13-acetate suggesting that only the adenylyl cyclase pathway is involved in the neurotrophic activity of PACAP. PACAP also induced a transient increase in c-fos messenger RNA level. The ability of PACAP to stimulate c-fos gene expression was mimicked by dibutyryladenosine 3',5'-cyclic-monophosphate but not phorbol 12-myristate 13-acetate. Similar effects of PACAP on granule cell survival were observed whether the cells were continuously incubated with PACAP for 48 h or only exposed to PACAP during 1 h. The protein kinase A inhibitor H89 significantly reduced the effect of PACAP on c-fos messenger RNA level whereas the specific protein kinase C inhibitor chelerythrine did not modify c-fos gene expression. These data indicate that the action of PACAP on cerebellar granule cell survival and c-fos gene expression are both mediated through the adenylyl cyclase/protein kinase A pathway. The observation that a short-term stimulation by PACAP can be converted into a long-lasting response indicates that the effect of the peptide on cell survival must involve immediate-early gene activation. The fact that a brief exposure to PACAP causes both c-fos gene expression and promotes cell survival strongly suggests that c-fos is involved in the trophic effect of PACAP on immature cerebellar granule cells.


Subject(s)
Cerebellum/cytology , Cyclic AMP-Dependent Protein Kinases/metabolism , Neurons/drug effects , Neuropeptides/pharmacology , Neuroprotective Agents/pharmacology , Proto-Oncogene Proteins c-fos/biosynthesis , Adenylyl Cyclases/metabolism , Animals , Cell Survival/drug effects , Cells, Cultured , Cerebellum/drug effects , Cerebellum/enzymology , Cyclic AMP-Dependent Protein Kinases/antagonists & inhibitors , Enzyme Activation/drug effects , Enzyme Inhibitors/pharmacology , Neurons/enzymology , Pituitary Adenylate Cyclase-Activating Polypeptide , Protein Kinase C/antagonists & inhibitors , Protein Kinase C/metabolism , RNA, Messenger/biosynthesis , Rats , Rats, Wistar , Stimulation, Chemical
17.
Ann Endocrinol (Paris) ; 59(5): 364-405, 1998 Dec.
Article in French | MEDLINE | ID: mdl-9949891

ABSTRACT

Pituitary adenylate cyclase-activating polypeptide (PACAP) has been originally isolated from the sheep hypothalamus on the basis of its ability to stimulate cAMP formation in anterior pituitary cells. Post-translational processing of the PACAP precursor generates two biologically active molecular forms, PACAP38 and PACAP27, and a novel peptide called PACAP-related peptide whose activity remains unknown. The primary structure of PACAP has been remarkably conserved during evolution, from protochordates to mammals, suggesting that the peptide exerts important activities throughout the vertebrate phylum. The sequence of PACAP27 exhibits substantial similarities with those of vasoactive intestinal polypeptide (VIP), glucagon and secretin. The gene encoding the PACAP precursor is widely expressed in the brain and in various peripheral organs, notably in endocrine glands, the gastro-intestinal and uro-genital tracts and the respiratory system. In vivo and in vitro studies have shown that PACAP exerts multiple activities as a hormone, neurohormone, neurotransmitter or trophic factor. For instance, PACAP triggers the release of insulin and glucagon, activates steroidogenesis in the adrenal gland and gonads, and stimulates the secretion of most hypophysial cells. PACAP exerts a potent relaxant activity on smooth muscle fibers in blood vessels, lung and gut. In the brain, PACAP stimulates the electrical activity of various populations of neurons and increases tyrosine hydroxylase gene expression. Recent studies have shown that PACAP exerts a trophic activity during ontogenesis, notably in the adrenal medulla and in the central nervous system. The biological effects of PACAP are mediated through three distinct receptor subtypes which exhibit differential affinities for PACAP and VIP. The PAC1 receptor, which shows high selectivity for PACAP, is coupled to several transduction systems. In contrast, VPAC1 and VPAC2, which bind with the same affinity PACAP and VIP, are mainly coupled to the adenylyl cyclase pathway. The bronchodilatator and vasorelaxant effects of PACAP, as well as the antiproliferative and neuroprotective actions of the peptide, make it a valuable target for new drug development.


Subject(s)
Adenylyl Cyclases/physiology , Neuropeptides/physiology , Pituitary Gland/enzymology , Amino Acid Sequence , Animals , Humans , Molecular Sequence Data , Neuropeptides/chemistry , Peptide Fragments/chemistry , Peptide Fragments/physiology , Pituitary Adenylate Cyclase-Activating Polypeptide , Protein Precursors/chemistry , Protein Precursors/physiology , Receptors, Pituitary Adenylate Cyclase-Activating Polypeptide , Receptors, Pituitary Adenylate Cyclase-Activating Polypeptide , Receptors, Pituitary Hormone/chemistry , Receptors, Pituitary Hormone/physiology , Receptors, Vasoactive Intestinal Peptide, Type II , Receptors, Vasoactive Intestinal Polypeptide, Type I
18.
Ann N Y Acad Sci ; 865: 92-9, 1998 Dec 11.
Article in English | MEDLINE | ID: mdl-9928001

ABSTRACT

In vitro studies have shown that PACAP promotes cell survival and neurite outgrowth in immature cerebellar granule cells. In the present study, we have examined the transduction pathways involved in the neurotrophic activity of PACAP. Incubation of cultured granule cells with graded concentrations of PACAP produced a dose-dependent increase in c-fos mRNA level. The effects of PACAP on c-fos gene expression and granule cell survival were both mimicked by dbcAMP but not by PMA. The maximum effect of PACAP on c-fos gene expression was observed after 1 h of treatment. Similar effects of the peptide on granule cell survival were observed whether the cells were continuously incubated with PACAP for 48 h or only exposed to PACAP during 1 h. The PKA inhibitor H89 significantly reduced the effect of PACAP on c-fos mRNA level, whereas the specific PKC inhibitor chelerytrine had no effect. These data indicate that the action of PACAP on cerebellar granule cell survival and c-fos gene expression are both mediated through the adenylyl cyclase/PKA pathway.


Subject(s)
Cerebellum/physiology , Cyclic AMP-Dependent Protein Kinases/metabolism , Gene Expression Regulation/physiology , Genes, fos , Neurons/physiology , Neuropeptides/pharmacology , Neuroprotective Agents/pharmacology , Animals , Cerebellum/cytology , Enzyme Activation , Gene Expression Regulation/drug effects , Neurites/drug effects , Neurites/physiology , Neurons/cytology , Neurons/drug effects , Neuropeptides/physiology , Pituitary Adenylate Cyclase-Activating Polypeptide , Proto-Oncogene Proteins c-fos/biosynthesis , Proto-Oncogene Proteins c-fos/genetics , Rats
19.
Neuroscience ; 78(2): 419-30, 1997 May.
Article in English | MEDLINE | ID: mdl-9145799

ABSTRACT

High concentrations of pituitary adenylate cyclase-activating polypeptide (PACAP) and its receptors have been detected in the rat cerebellum during ontogenesis. In particular, PACAP receptors are actively expressed in immature granule cells, suggesting that PACAP may act as a neurotrophic factor in the developing rat cerebellum. In the present study, we have investigated the effect of PACAP on cell survival and neurite outgrowth in cultured immature cerebellar granule cells. In control conditions, cultured granule cells undergo programmed cell death. Exposure of cultured cells to PACAP for 24 and 48 h provoked a significant increase in the number of living cells. The effect of PACAP on cell survival was inhibited by the PACAP antagonist PACAP(6-38). Vasoactive intestinal polypeptide was approximately 1000 times less potent than PACAP in promoting cell survival. PACAP also induced a significant increase in the number of processes and in the cumulated length of neurites borne by cultured neuroblasts. The present results demonstrate that PACAP promotes cell survival and neurite outgrowth in cultured immature granule cells. Since PACAP and its receptors are expressed in situ in the rat cerebellar cortex, these data strongly suggest that PACAP plays a physiological role in the survival and differentiation of cerebellar granule cells.


Subject(s)
Cerebellum/cytology , Neurites/drug effects , Neurons/drug effects , Neuropeptides/pharmacology , Animals , Cell Count , Cell Survival/drug effects , Cells, Cultured , Cerebellum/drug effects , Cerebellum/growth & development , Neurofilament Proteins/metabolism , Pituitary Adenylate Cyclase-Activating Polypeptide , Potassium Chloride/pharmacology , Rats , Rats, Wistar , Vasoactive Intestinal Peptide/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...