Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 30
Filter
Add more filters










Publication year range
1.
Cell Death Differ ; 15(10): 1542-52, 2008 Oct.
Article in English | MEDLINE | ID: mdl-18551134

ABSTRACT

In mammals, programmed cell death (PCD) is a central event during brain development. Trophic factors have been shown to prevent PCD in postmitotic neurons. Similarly, cytokines have neurotrophic effects involving regulation of neuronal survival. Nevertheless, neuronal PCD is only partially understood and host determinants are incompletely defined. The present study provides evidence that the cytokine interleukin-9 (IL-9) and its receptor specifically control PCD of neurons in the murine newborn neocortex. IL-9 antiapoptotic action appeared to be time-restricted to early postnatal stages as both ligand and receptor transcripts were mostly expressed in neocortex between postnatal days 0 and 10. This period corresponds to the physiological peak of apoptosis for postmitotic neurons in mouse neocortex. In vivo studies showed that IL-9/IL-9 receptor pathway inhibits apoptosis in the newborn neocortex. Furthermore, in vitro studies demonstrated that IL-9 and its receptor are mainly expressed in neurons. IL-9 effects were mediated by the activation of the JAK/STAT (janus kinase/signal transducer and activator of transcription) pathway, whereas nuclear factor-kappaB (NF-kappaB) or Erk pathways were not involved in mediating IL-9-induced inhibition of cell death. Finally, IL-9 reduced the expression of the mitochondrial pro-apoptotic factor Bax whereas Bcl-2 level was not significantly affected. Together, these data suggest that IL-9/IL-9 receptor signaling pathway represents a novel endogenous antiapoptotic mechanism for cortical neurons by controlling JAK/STAT and Bax levels.


Subject(s)
Apoptosis/physiology , Cerebral Cortex/cytology , Interleukin-9/metabolism , Neurons/physiology , Receptors, Interleukin-9/metabolism , Signal Transduction/physiology , Animals , Animals, Newborn , Caspase 3/metabolism , Cells, Cultured , Humans , Janus Kinases/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , STAT Transcription Factors/metabolism , bcl-2-Associated X Protein/metabolism
2.
Peptides ; 28(9): 1688-99, 2007 Sep.
Article in English | MEDLINE | ID: mdl-17606312

ABSTRACT

In 1970, Drs. Said and Mutt isolated a novel peptide from porcine intestinal extracts with powerful vasoactive properties, and named it vasoactive intestinal peptide (VIP). Since then, the biological actions of VIP in the gut as well as its signal transduction pathways have been extensively studied. A variety of in vitro and in vivo studies have indicated that VIP, expressed in intrinsic non-adrenergic non-cholinergic (NANC) neurons, is a potent regulator of gastrointestinal (GI) motility, water absorption and ion flux, mucus secretion and immune homeostasis. These VIP actions are believed to be mediated mainly by interactions with highly expressed VPAC(1) receptors and the production of nitric oxide (NO). Furthermore, VIP has been implicated in numerous physiopathological conditions affecting the human gut, including pancreatic endocrine tumors secreting VIP (VIPomas), insulin-dependent diabetes, Hirschsprung's disease, and inflammatory bowel syndromes such as Crohn's disease and ulcerative colitis. To further understand the physiological roles of VIP on the GI tract, we have begun to analyze the anatomical and physiological phenotype of C57BL/6 mice lacking the VIP gene. Herein, we demonstrate that the overall intestinal morphology and light microscopic structure is significantly altered in VIP(-/-) mice. Macroscopically there is an overall increase in weight, and decrease in length of the bowel compared to wild type (WT) controls. Microscopically, the phenotype was characterized by thickening of smooth muscle layers, increased villi length, and higher abundance of goblet cells. Alcian blue staining indicated that the latter cells were deficient in mucus secretion in VIP(-/-) mice. The differences became more pronounced from the duodenum to the distal jejunum or ileum of the small bowel but, became much less apparent or absent in the colon with the exception of mucus secretion defects. Further examination of the small intestine revealed larger axonal trunks and unusual unstained patches in myenteric plexus. Physiologically, the VIP(-/-) mice showed an impairment in intestinal transit. Moreover, unlike WT C57BL/6 mice, a significant percentage of VIP(-/-) mice died in the first postnatal year with overt stenosis of the gut.


Subject(s)
Gastrointestinal Tract/physiopathology , Hirschsprung Disease/physiopathology , Ileus/physiopathology , Mutation , Vasoactive Intestinal Peptide/physiology , Animals , Gastrointestinal Motility/genetics , Gastrointestinal Motility/physiology , Gastrointestinal Tract/metabolism , Gastrointestinal Tract/pathology , Hirschsprung Disease/genetics , Hirschsprung Disease/pathology , Ileus/metabolism , Ileus/pathology , Immunohistochemistry , Mice , Mice, Inbred C57BL , Mice, Knockout , Vasoactive Intestinal Peptide/genetics , Vasoactive Intestinal Peptide/metabolism
3.
Cell Signal ; 18(11): 2013-21, 2006 Nov.
Article in English | MEDLINE | ID: mdl-16723209

ABSTRACT

Atrial natriuretic peptide (ANP) and the closely-related peptides BNP and CNP are highly conserved cardiovascular hormones. They bind to single transmembrane-spanning receptors, triggering receptor-intrinsic guanylyl cyclase activity. The "truncated" type-C natriuretic peptide receptor (NPR-C) has long been called a clearance receptor because it lacks the intracellular guanylyl cyclase domain, though data suggest it might negatively couple to adenylyl cyclase via G(i). Here we report the molecular cloning and characterization of the Xenopus laevis type-C natriuretic peptide receptor (XNPR-C). Analysis confirms the presence of a short intracellular C-terminus, as well as a high similarity to fish and mammalian NPR-C. Injection of XNPR-C mRNA into Xenopus oocytes resulted in expression of high affinity [(125)I]ANP binding sites that were competitively and completely displaced by natriuretic analogs and the unrelated neuropeptide vasoactive intestinal peptide (VIP). Measurement of cAMP levels in mRNA-injected oocytes revealed that XNPR-C is negatively coupled to adenylyl cyclase in a pertussis toxin-sensitive manner. When XNPR-C was co-expressed with PAC(1) receptors for pituitary adenylyl cyclase-activating polypeptide (PACAP), VIP and natriuretic peptides counteracted the cAMP induction by PACAP. These results suggest that VIP and natriuretic peptides can potentially modulate the action of PACAP in cells where these receptors are co-expressed.


Subject(s)
Oocytes/metabolism , Receptors, Atrial Natriuretic Factor/genetics , Receptors, Atrial Natriuretic Factor/metabolism , Receptors, Pituitary Adenylate Cyclase-Activating Polypeptide/metabolism , Signal Transduction , Vasoactive Intestinal Peptide/metabolism , Adenylyl Cyclases/metabolism , Amino Acid Sequence , Animals , Binding, Competitive , Cloning, Molecular , DNA, Complementary/genetics , Humans , Molecular Sequence Data , Oocytes/drug effects , RNA, Messenger/metabolism , RNA, Messenger/pharmacology , Rats , Receptors, Pituitary Adenylate Cyclase-Activating Polypeptide/antagonists & inhibitors , Sequence Alignment , Vasoactive Intestinal Peptide/pharmacology , Xenopus
4.
Dev Biol ; 271(1): 161-75, 2004 Jul 01.
Article in English | MEDLINE | ID: mdl-15196958

ABSTRACT

Atrial natriuretic peptide (ANP) binding sites have been detected in the embryonic brain, but the specific receptor subtypes and biological functions for ANP family ligands therein remain undefined. We now characterize the patterns of gene expression for the natriuretic peptides [ANP, brain natriuretic peptide (BNP), type-C natriuretic peptide (CNP)] and their receptors (NPR-A, NPR-B, NPR-C) at several early stages in the embryonic mouse nervous system by in situ hybridization, and begin to define the potential developmental actions using cell culture models of peripheral (PNS) and central nervous systems (CNS). In the CNS, gene transcripts for CNP were present at the onset of neurogenesis, embryonic day 10.5 (E10.5), primarily in the dorsal part of the ventricular zone (VZ) throughout the hindbrain and spinal cord. On E14.5, new CNP signals were observed in the ventrolateral spinal cord where motor neurons reside, and in bands of cells surrounding the spinal cord and hindbrain, localized to dura and/or cartilage primordia. ANP and BNP gene transcripts were not detected in embryonic brain, but were highly abundant in the heart. The CNP-specific receptor (NPR-B) gene was expressed in cells just outside the VZ, in regions where post-mitotic neurons are differentiating. Gene expression for NPR-C, which recognizes all natriuretic peptides, was present in the roof plate of the hindbrain and spinal cord and in bilateral stripes just dorsolateral to the floor plate at E12.5. In the PNS, NPR-B and NPR-C transcripts were highly expressed in dorsal root sensory (DRG) and cranial ganglia beginning at E10.5, with NPR-C signal also prominent in adjoining nerves, consistent with Schwann cell localization. In contrast, NPR-A gene expression was undetectable in neural tissues. To define ontogenetic functions, we employed embryonic DRG and hindbrain cell cultures. The natriuretic peptides potently stimulated DNA synthesis in neuron-depleted as well as neuron-containing Schwann cell cultures and differentially inhibited neurite outgrowth in DRG sensory neuron cultures. CNP also exhibited modest survival-promoting effects for sensory neurons. In marked contrast to PNS effects, the peptides inhibited proliferation of neural precursor cells of the E10.5 hindbrain. Moreover, CNP, alone and in combination with sonic hedgehog (Shh), induced the expression of the Shh target gene gli-1 in hindbrain cultures, suggesting that natriuretic peptides may also modify patterning events in the embryonic brain. These studies reveal widespread, but discrete patterns of natriuretic peptide and receptor gene expression in the early embryonic nervous system, and suggest that the peptides play region- and stage-specific roles during the development of the peripheral and central nervous systems.


Subject(s)
Gene Expression Regulation, Developmental , Natriuretic Peptides/metabolism , Nervous System/embryology , RNA, Messenger/metabolism , Receptors, Atrial Natriuretic Factor/metabolism , Animals , Cells, Cultured , DNA Primers , DNA Replication/genetics , Ganglia, Spinal/cytology , Ganglia, Spinal/embryology , Hedgehog Proteins , Immunohistochemistry , In Situ Hybridization , Kruppel-Like Transcription Factors , Mice , Natriuretic Peptides/physiology , Nervous System/metabolism , Polymerase Chain Reaction/methods , Receptors, Atrial Natriuretic Factor/physiology , Trans-Activators/metabolism , Transcription Factors/metabolism , Zinc Finger Protein GLI1
5.
Am J Physiol Regul Integr Comp Physiol ; 287(5): R1194-201, 2004 Nov.
Article in English | MEDLINE | ID: mdl-15217792

ABSTRACT

Previous studies indicate that light information reaches the suprachiasmatic nucleus through a subpopulation of retinal ganglion cells that contain both glutamate and pituitary adenylyl cyclase-activating peptide (PACAP). Although the role of glutamate in this pathway has been well studied, the involvement of PACAP and its receptors is only beginning to be understood. To investigate the functions of PACAP in vivo, we developed a mouse model in which the gene coding for PACAP was disrupted by targeted homologous recombination. RIA was used to confirm a lack of detectable PACAP protein in these mice. PACAP-deficient mice exhibited significant impairment in the magnitude of the response to brief light exposures with both light-induced phase delays and advances of the circadian system impacted. This mutation equally impacted phase shifts induced by bright and dim light exposure. Despite these effects on phase shifting, the loss of PACAP had only limited effects on the generation of circadian oscillations, as measured by rhythms in wheel-running activity. Unlike melanopsin-deficient mice, the mice lacking PACAP exhibited no loss of function in the direct light-induced inhibition of locomotor activity, i.e., masking. Finally, the PACAP-deficient mice exhibited normal phase shifts in response to exposure to discrete dark treatments. The results reported here show that the loss of PACAP produced selective deficits in the light response of the circadian system.


Subject(s)
Circadian Rhythm/physiology , Light , Neuropeptides/physiology , Animals , Behavior, Animal/physiology , DNA, Complementary/biosynthesis , DNA, Complementary/genetics , Darkness , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Motor Activity/physiology , Neuropeptides/deficiency , Neuropeptides/genetics , Pituitary Adenylate Cyclase-Activating Polypeptide , Radioimmunoassay , Stem Cell Transplantation
6.
J Comp Neurol ; 441(3): 266-75, 2001 Dec 17.
Article in English | MEDLINE | ID: mdl-11745649

ABSTRACT

The genes encoding pituitary adenylyl cyclase-activating peptide (PACAP) and its selective type I receptor (PAC1) are expressed in the embryonic mouse neural tube, where they may be involved in neurogenesis and neural tube development. We examined here the early expression and potential actions of PACAP and PAC1 in the vertebrate developmental model Xenopus laevis. PACAP and PAC1 mRNAs were first detected by RT-PCR in stage 16-18 embryos (18 hours after fertilization). Two distinct PACAP precursor mRNAs were identified. One encoded both growth hormone-releasing hormone and PACAP, whereas the other encoded only full-length PACAP. Unlike that in the adult, the latter represented the predominant embryonic PACAP mRNA species. In situ hybridization revealed that PACAP and PAC1 mRNAs were restricted to neural cells. PAC1 gene expression was observed mainly in the ventricular zone in the ventral parts of the prosencephalon, mensencephalon, rhombencephalon, and anterior spinal cord. In contrast, PACAP mRNA was localized exclusively in postmitotic cells in the dorsolateral parts of the rhombencephalon and entire spinal cord. Most PACAP mRNA-containing cells were characterized as Rohon-Beard neurons. Exposure of early embryos to UV irradiation, which ventralizes embryos and inhibits neural induction, reduced the expression of PACAP and PAC1 genes. These results suggest that PACAP may be involved in the early development of the embryonic Xenopus neural tube.


Subject(s)
Nervous System/embryology , Neuropeptides/genetics , Receptors, Pituitary Hormone/genetics , Xenopus laevis/embryology , Xenopus laevis/genetics , Animals , Embryo, Nonmammalian/drug effects , Embryo, Nonmammalian/physiology , Embryo, Nonmammalian/radiation effects , Gene Expression Regulation, Developmental/drug effects , Gene Expression Regulation, Developmental/radiation effects , Lithium Chloride/pharmacology , Pituitary Adenylate Cyclase-Activating Polypeptide , RNA, Messenger/metabolism , Receptors, Pituitary Adenylate Cyclase-Activating Polypeptide , Receptors, Pituitary Adenylate Cyclase-Activating Polypeptide , Receptors, Pituitary Hormone/physiology , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction , Tissue Distribution , Ultraviolet Rays , Xenopus Proteins
7.
J Biol Chem ; 276(47): 43668-76, 2001 Nov 23.
Article in English | MEDLINE | ID: mdl-11553633

ABSTRACT

To identify neural tumor cell lines that could be used as models to study growth-related natriuretic peptide actions, we determined the effects of these peptides on the proliferation of human and rodent neuroblastoma cell lines. Subnanomolar concentrations of atrial natriuretic peptide (ANP) and type C natriuretic peptide (CNP) stimulated proliferation in all four cell lines. These actions were associated with cGMP elevation and were blocked by a protein kinase G inhibitor. These data imply the involvement of guanylyl cyclase (GC)-coupled natriuretic receptors. However, higher concentrations of ANP and CNP, and low concentrations of des-[Gln(18),Ser(19),Gly(20),Leu(21),Gly(22)]-ANP(4-23)-NH(2) (desANP(4-23)) (analog for NPR-C receptor) exerted antiproliferative actions in three of the cell lines. These effects were insensitive to a protein kinase G inhibitor and to HS-142-1, suggesting that growth-inhibitory actions involved a non-GC receptor. They did not appear to involve cAMP, protein kinase A, protein kinase C, or calcium mobilization but were abolished when constitutive mitogen-activated protein kinase activity was inhibited. Radioligand binding experiments revealed the presence of a uniform class of binding sites in NG108 cells and multiple binding sites in Neuro2a cells. Northern and reverse transcriptase-polymerase chain reaction analyses revealed differential gene expression for NPR-A/B/C in NG108 and Neuro2a cells. The results indicate that natriuretic peptides stimulate neuroblastoma cell proliferation through type NPR-A/B (GC) receptors. Higher concentrations of ANP and CNP exerted a mitogen-activated protein kinase-dependent antiproliferative action mediated by a non-GC receptor that interacts with desANP(4-23) with relatively high affinity.


Subject(s)
Atrial Natriuretic Factor/pharmacology , Cell Division/drug effects , Guanylate Cyclase/metabolism , Neuroblastoma/pathology , Atrial Natriuretic Factor/chemistry , Base Sequence , Molecular Sequence Data , Receptors, Atrial Natriuretic Factor/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Tumor Cells, Cultured
8.
Neuropeptides ; 35(2): 117-26, 2001 Apr.
Article in English | MEDLINE | ID: mdl-11384207

ABSTRACT

In early reports on 125I-VIP binding experiments in liver membranes, it has been proposed that, the VIP binding sites were partially sensitive to GTP. Here we confirm that the VIP binding sites of chicken liver membranes consisted mainly in bivalent VIP/PACAP receptors and that about 50% of the 125I-VIP binding capacity was not affected by the GTP analogue GppNHp. Part of these bivalent receptors also appeared to represent PHI binding sites. In GppNHp-treated membranes, the GTP-insensitive VIP binding sites displayed a 17-fold higher relative affinity than in control membranes for the VIP analogue PHI. Such data suggested that GTP-insensitive VIP receptors may correspond to a subclass of high-affinity PHI receptors. Cross-linking of 125 I-VIP or 125 I-PHI to their receptors, revealed 2 components of 48 and 60 kDa. The radiolabelling of the 60 kDa component was strongly affected by increasing concentrations of the GTP analogue but was modestly abolished by an excess of PHI. Conversely, the radiolabelling of the 48 kDa molecular form was not affected by the GTP analogue but was efficiently abolished by increasing concentrations of PHI. Taken together, the data suggest that the 48 kDa component expressed in chicken liver membranes display the properties of a GTP-insensitive VIP/PHI receptor that can be pharmacologically discriminated from the GTP-sensitive 60 kDa form, through its much higher affinity for PHI.


Subject(s)
Guanosine Triphosphate/metabolism , Liver/metabolism , Peptide PHI/pharmacology , Receptors, Vasoactive Intestinal Peptide/metabolism , Animals , Cell Membrane/metabolism , Chickens , Cross-Linking Reagents/metabolism , Cross-Linking Reagents/pharmacology , Guanosine Triphosphate/analogs & derivatives , Guanylyl Imidodiphosphate/pharmacology , Iodine Radioisotopes , Neuropeptides/metabolism , Neuropeptides/pharmacology , Peptide PHI/metabolism , Pituitary Adenylate Cyclase-Activating Polypeptide , Radioligand Assay , Receptors, Pituitary Adenylate Cyclase-Activating Polypeptide , Receptors, Pituitary Hormone/metabolism , Vasoactive Intestinal Peptide/metabolism , Vasoactive Intestinal Peptide/pharmacology
9.
J Neurosci ; 21(11): 3849-59, 2001 Jun 01.
Article in English | MEDLINE | ID: mdl-11356873

ABSTRACT

The neuropeptide pituitary adenylyl cyclase-activating peptide (PACAP) and one of its receptors (PAC(1)) are expressed in embryonic neural tube, where they appear to regulate neurogenesis and patterning. We now show that PAC(1) gene expression is also present in neonatal rats in the ventricular and subventricular zones and in the optic chiasm, areas that are rich in oligodendrocyte (OL) progenitors (OLP). Because actions of PACAP on OLP have not been reported, we examined the effects of PACAP on the proliferation of purified OLP in culture and on myelinogenesis in cerebellar slices. Northern analyses on total RNA from purified glial cell subtypes revealed an abundant 7 kb hybridizing transcript in OLP, which was confirmed to correspond to the PAC(1) receptor by reverse transcription-PCR. The presence of this receptor was also corroborated by radioligand binding and cAMP assay. In cultured OL, receptor density decreased during maturation but was partially counterbalanced by the appearance of sites that bound both PACAP and the related peptide vasoactive intestinal peptide. PACAP increased DNA synthesis in OLP cultures almost twofold and increased the bromodeoxyuridine-labeling index in O4-positive OLP. PACAP treatment also resulted in decreased sulfate incorporation into sulfatide in cultures of differentiating OL. The PACAP effect on sulfatide synthesis was fully reproduced in a cerebellar explant model. These findings indicate that PACAP may act at two stages during OL development to (1) stimulate proliferation and (2) delay maturation and/or myelinogenesis.


Subject(s)
Cell Differentiation/drug effects , DNA/metabolism , Neuropeptides/metabolism , Oligodendroglia/cytology , Stem Cells/metabolism , Animals , Animals, Newborn , Binding, Competitive/drug effects , Bromodeoxyuridine , Cell Division/drug effects , Cells, Cultured , Cerebellum/cytology , Cerebellum/drug effects , Cerebellum/metabolism , Cerebral Ventricles/cytology , Cerebral Ventricles/metabolism , Cerebral Ventricles/surgery , Gene Expression , In Situ Hybridization , In Vitro Techniques , Myelin Sheath/metabolism , Neuropeptides/pharmacology , Optic Chiasm/cytology , Optic Chiasm/metabolism , Pituitary Adenylate Cyclase-Activating Polypeptide , RNA, Messenger/metabolism , Radioligand Assay , Rats , Rats, Wistar , Receptors, Pituitary Adenylate Cyclase-Activating Polypeptide , Receptors, Pituitary Hormone/biosynthesis , Receptors, Pituitary Hormone/genetics , Reverse Transcriptase Polymerase Chain Reaction , Stem Cells/drug effects
10.
Biochem Biophys Res Commun ; 284(1): 211-8, 2001 Jun 01.
Article in English | MEDLINE | ID: mdl-11374892

ABSTRACT

SK-N-SH human neuroblastoma subclones differ widely in basal and second messenger induction of the gene encoding the neuropeptide vasoactive intestinal peptide (VIP). These differences were recapitulated by a chimeric gene which consisted of 5.2 kb of the human VIP gene 5' flanking sequence fused to a reporter. Subsequent gene deletion experiments revealed several regulatory regions on the gene, including a 645-bp sequence located approximately 4.0 upstream from the transcription start site. Here we examined this upstream region in detail. Inhibitory sequences were found to be present on each end of the 645-bp fragment. When removed, basal transcription increased more than 50-fold. Subsequent deletion/mutation analysis showed that the 213-bp fragment contained at least two enhancer elements. One of these was localized to an AT-rich 42-bp sequence shown by others to bind Oct proteins in neuroblastoma cells, while the other corresponded to a composite AP-1/ets element. In addition to these enhancers, a 28-bp sequence on the 213-bp fragment with no apparent homology to known silencers inhibited transcription. The studies provide molecular details of a complex regulatory region on the VIP gene that is likely to be used to finely tune the level of gene transcription in vivo.


Subject(s)
Gene Expression Regulation, Neoplastic , Regulatory Sequences, Nucleic Acid , Transcription, Genetic , Vasoactive Intestinal Peptide/genetics , 5' Untranslated Regions/genetics , Base Sequence , Consensus Sequence , DNA-Binding Proteins/metabolism , Enhancer Elements, Genetic , Gene Silencing , Genes, Reporter , Host Cell Factor C1 , Humans , Molecular Sequence Data , Mutagenesis, Site-Directed , Neuroblastoma/metabolism , Octamer Transcription Factor-1 , Octamer Transcription Factor-2 , Sequence Deletion , Transcription Factor AP-1/metabolism , Transcription Factors/metabolism , Transfection , Tumor Cells, Cultured , Vasoactive Intestinal Peptide/biosynthesis
11.
Endocrinology ; 141(9): 3366-76, 2000 Sep.
Article in English | MEDLINE | ID: mdl-10965909

ABSTRACT

Pituitary adenylyl cyclase-activating peptide (PACAP) appears to regulate several neuroendocrine functions in the frog, but its messenger RNA (mRNA) structure and brain distribution are unknown. To understand the potential role of PACAP in the male frog hypothalamic-pituitary-gonadal axis, we cloned the frog Xenopus laevis PACAP mRNA and determined its distribution in the brain. We then analyzed the castration-induced alterations of mRNA expression for PACAP and its selective type I receptor (PAC1) in the hypothalamic anterior preoptic area, a region known to regulate reproductive function. The PACAP mRNA encodes a peptide precursor predicted to give rise to both GH-releasing hormone and PACAP. The deduced peptide sequence of PACAP-38 was nearly identical to that of human PACAP with one amino acid substitution. Abundant PACAP mRNA was detected in the brain, but not several other tissues, including the testis. In situ hybridization revealed strong expression of the PACAP gene in the dorsal pallium, ventral hypothalamus, and nuclei of cerebellum. PACAP mRNA signals were weak to moderate in the hypothalamic anterior preoptic area and were absent in the pituitary. Castration induced an increase in the expression of PACAP and PAC1 receptor mRNAs in the hypothalamic anterior preoptic area after 3 days. Replacement with testosterone prevented the castration-induced changes. These results provide a molecular basis for studying the physiological functions of PACAP in frog brain and suggest that PACAP may be involved in the feedback regulation of hypothalamic-pituitary-gonadal axis.


Subject(s)
Brain Chemistry/physiology , Growth Hormone-Releasing Hormone/biosynthesis , Neuropeptides/biosynthesis , Orchiectomy , Amino Acid Sequence , Animals , Base Sequence , Blotting, Northern , Cloning, Molecular , DNA, Complementary/biosynthesis , DNA, Complementary/genetics , Feedback/physiology , Humans , In Situ Hybridization , Larva , Male , Molecular Sequence Data , Pituitary Adenylate Cyclase-Activating Polypeptide , Preoptic Area/metabolism , RNA, Messenger/biosynthesis , Reverse Transcriptase Polymerase Chain Reaction , Testosterone/pharmacology , Tissue Distribution , Up-Regulation , Xenopus Proteins , Xenopus laevis
12.
Eur J Pharmacol ; 391(1-2): 21-9, 2000 Mar 10.
Article in English | MEDLINE | ID: mdl-10720631

ABSTRACT

HT29 cells display an undifferentiated phenotype in culture. However, numerous treatments are able to induce both epithelial differentiation and cell growth inhibition. We have previously demonstrated that adenosine and its analogues act through specific adenosine receptors to modulate cell proliferation in HT29 and other human colon adenocarcinoma cell lines. Among the treatments tested, the most potent inhibition of HT29 cell growth was induced by deprivation of extracellular adenosine using adenosine deaminase. Here, we investigated the capacity of adenosine deaminase to initiate epithelial differentiation. After 1 month of daily addition of 10 U/ml adenosine deaminase to the culture medium, HT29 cells were cloned by limited dilution. Among the clones obtained, we focused our attention on clone 13. Microscopic visualization and proliferation studies indicated that cells from this clone grew very slowly and in a pseudo-monolayer, in marked contrast with the situation observed in the mother HT29 cell line. In addition, clone 13 cells displayed epithelial features that mimic the enterocytic differentiation of Caco-2 cells. These modifications were accompanied by dramatic changes in the activity of adenosine receptors, as demonstrated by pharmacological studies. In contrast to the original HT29 cells, clone 13 as well as Caco-2 cells displayed (i) a very low number of adenosine A(1) receptors, and (ii) increases in intracellular cAMP levels when challenged with adenosine analogues. It is hypothesized that a loss of adenosine A(1) receptors, with no change or a concomitant increase in adenosine A(2) receptors, results in the emergence of adenosine A(2) receptor-mediated differentiation and inhibition of proliferation, through a cAMP-dependent pathway.


Subject(s)
Adenosine/physiology , Epithelial Cells/physiology , Receptors, Purinergic P1/physiology , Adenosine/analogs & derivatives , Adenosine/metabolism , Adenosine Deaminase/chemistry , Adenosine-5'-(N-ethylcarboxamide)/pharmacology , Caco-2 Cells , Cell Differentiation/physiology , Cell Division/physiology , Clone Cells/physiology , Clone Cells/ultrastructure , Culture Media , Cyclic AMP/metabolism , Epithelial Cells/drug effects , HT29 Cells , Humans , Purinergic P1 Receptor Antagonists , Receptors, Purinergic P1/biosynthesis , Xanthines/pharmacology
13.
Endocrinology ; 141(2): 657-65, 2000 Feb.
Article in English | MEDLINE | ID: mdl-10650947

ABSTRACT

Pituitary adenylate cyclase-activating polypeptide (PACAP) has been found to modulate neuroendocrine functions in the frog brain and pituitary, but the nucleotide sequence and brain distribution of messenger RNA (mRNA) for the selective type I receptor for PACAP (PAC1) in the frog are still unknown. Here, we report the isolation and characterization of a PAC1 receptor complementary DNA (cDNA) clone from a frog (Xenopus laevis) tadpole brain cDNA library. This cDNA encoded a 466-amino acid protein that has 74% homology with human PAC1 receptor and 48% homology to the frog vasoactive intestinal peptide/PACAP receptor. Injection of in vitro synthesized mRNA of the cloned cDNA into Xenopus oocytes resulted in expression of selective high affinity PACAP receptors (Kd = 47 pM). IC50 values for PACAP-38, PACAP-27, and VIP were 27 pM, 110 pM and >1 microM, respectively. These results indicated that the cloned cDNA represents a Xenopus PACAP-preferring PAC1 receptor. Northern hybridization revealed that PAC1 receptor mRNA was present at high levels in the brain. In situ hybridization showed that the PAC1 receptor gene was expressed highly in the pallium, preoptic nucleus, and nucleus of cerebellum, and moderately in the Purkinje cell layer of the cerebellum. Moderate PAC1 receptor mRNA signals were detected in the distal lobe of the pituitary. A knowledge of the molecular structure and expression pattern of the PAC1 receptor will facilitate further investigation of the physiological roles of PAC1 receptor in the frog brain.


Subject(s)
Brain/metabolism , RNA, Messenger/metabolism , Receptors, Pituitary Hormone/genetics , Receptors, Pituitary Hormone/metabolism , Transcription, Genetic , Amino Acid Sequence , Animals , Base Sequence , Cloning, Molecular , DNA, Complementary , Female , Gene Library , Goldfish , Humans , In Vitro Techniques , Kinetics , Molecular Sequence Data , Neuropeptides/metabolism , Oocytes/physiology , Pituitary Adenylate Cyclase-Activating Polypeptide , Polymerase Chain Reaction , RNA, Messenger/genetics , Ranidae , Receptors, Pituitary Adenylate Cyclase-Activating Polypeptide , Receptors, Pituitary Adenylate Cyclase-Activating Polypeptide , Receptors, Pituitary Hormone/chemistry , Recombinant Proteins/metabolism , Sequence Alignment , Sequence Homology, Amino Acid , Vasoactive Intestinal Peptide/metabolism , Xenopus Proteins , Xenopus laevis
14.
Ann N Y Acad Sci ; 921: 129-36, 2000.
Article in English | MEDLINE | ID: mdl-11193816

ABSTRACT

Pituitary adenylate cyclase activating peptide (PACAP) may play a role in neurogenesis, nerve injury, and neural tumor growth. A PACAP ligand receptor system functionally coupled to cAMP production was found to be expressed in the embryonic mouse neural tube at the onset of neurogenesis. PACAP was found to inhibit DNA synthesis and antagonize sonic hedgehog signaling in cells isolated from the neural tube, suggesting that PACAP interacts with patterning factors to regulate neurogenesis and phenotypic specification in the developing CNS. PACAP and PACAP receptor (PAC1) mRNA levels were strongly increased and decreased, respectively, in motor neurons in adult rats after facial nerve axotomy, indicating that PACAP may also act in nerve regeneration. Experiments using a neuroblastoma tumor cell line model indicate that PACAP may execute growth-related functions by activating MAP kinase in addition to cAMP-dependent protein kinase A.


Subject(s)
Nervous System/drug effects , Nervous System/embryology , Neuropeptides/pharmacology , Neuropeptides/physiology , Animals , Axotomy , Cell Division/drug effects , Cell Division/physiology , Cell Survival/drug effects , Cell Survival/physiology , DNA/biosynthesis , In Vitro Techniques , Mice , Motor Neurons/physiology , Nerve Regeneration/drug effects , Nerve Regeneration/physiology , Neuroblastoma/pathology , Neuropeptides/genetics , Pituitary Adenylate Cyclase-Activating Polypeptide , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats , Receptors, Pituitary Adenylate Cyclase-Activating Polypeptide , Receptors, Pituitary Adenylate Cyclase-Activating Polypeptide , Receptors, Pituitary Hormone/genetics , Receptors, Pituitary Hormone/physiology , Signal Transduction
15.
J Neurosci Res ; 57(6): 953-61, 1999 Sep 15.
Article in English | MEDLINE | ID: mdl-10467267

ABSTRACT

It has been demonstrated that pituitary adenylate cyclase activating polypeptide (PACAP) promotes the survival of neurons in culture and can inhibit neuronal cell death after experimental injury. Furthermore, peripheral axotomy results in increased PACAP gene expression in sensory and sympathetic neurons, suggesting that PACAP might be a mediator in the injury response in certain parts of the nervous system. However, changes in PACAP expression have not been reported in injured motor neurons, despite the significant problem of motor neuron degeneration in injury and in several neurological diseases. We examined here changes in gene expression of PACAP and two high-affinity PACAP receptors, PAC(1) and VPAC(2), in adult rat motor neurons after facial nerve axotomy by in situ hybridization. PACAP gene expression was very low in facial motor neurons of normal rats. However, a robust time-dependent increase in PACAP mRNA was observed in the facial motor nucleus in most or all axotomized motor neurons. This induction was detectable 6 hr after axotomy, and peaked at 48 hr, when expression on the injured side averaged more than 20-fold higher than that on the contralateral side. Thereafter, PACAP mRNA levels decreased slightly, but remained more than 10-fold elevated for as long as 30 days after axotomy. In contrast to PACAP, gene expression for both the PAC(1) and VPAC(2) receptor was high in facial motor neurons of normal rats. No significant change was observed for VPAC(2) receptor gene expression in facial motor neurons after axotomy, whereas gene expression for the PAC(1) receptor became significantly decreased. The results indicate that the PACAP ligand receptor system is tightly regulated in the facial motor nucleus after axotomy, providing evidence that PACAP may be involved in motor injury responses.


Subject(s)
Facial Nerve/metabolism , Motor Neurons/metabolism , Neuropeptides/metabolism , Receptors, Pituitary Hormone/genetics , Animals , Axotomy , Facial Nerve/cytology , Gene Expression , In Situ Hybridization , Male , Pituitary Adenylate Cyclase-Activating Polypeptide , Rats , Rats, Wistar , Receptors, Pituitary Adenylate Cyclase-Activating Polypeptide
16.
J Biol Chem ; 273(31): 19685-90, 1998 Jul 31.
Article in English | MEDLINE | ID: mdl-9677397

ABSTRACT

The growth rate of rodent embryonic neuroblasts and human neuroblastoma cell lines is regulated in part by autocrine or paracrine actions of neuropeptides of the family that includes vasoactive intestinal peptide (VIP), peptide histidine isoleucine (PHI), and pituitary adenylate cyclase-activating peptide (PACAP). These peptides act via seven transmembrane G-protein-linked receptors coupled to cAMP elevation, phospholipase C activation, intracellular Ca2+ release, and/or of mitogen-activated protein (MAP) kinase activation. Here we investigated the action of these peptides on the mouse neuroblastoma cell line Neuro2a. PHI and VIP inhibited proliferation at concentrations as low as 10(-13) M and 10(-10) M, respectively. In contrast, PACAP action was biphasic, with stimulation occurring at subnanomolar doses and inhibition at higher doses. Peptide actions were studied further by measuring cAMP and ERK1/2 MAP kinase activity and by assessing 3H-thymidine incorporation in conjunction with a panel of signal transduction pathways inhibitors. The data obtained indicated that the PHI-inhibitory and PACAP-stimulatory activities were mediated by corresponding changes in activity of the MAP kinase pathway and independent of protein kinase A (PKA) or protein kinase C (PKC). In contrast, the inhibitory actions of VIP and PACAP were specifically blocked by antagonists of PKA. Northern blot analysis revealed gene expression for only the PACAP-preferring (PAC1) receptor. However, binding experiments using 125I-labeled PACAP27, PHI, and VIP, demonstrated the presence of PACAP-preferring sites, bivalent VIP/PACAP sites, and PHI-binding sites that did not interact with VIP. The studies demonstrate potent regulatory actions of PACAP, PHI, and VIP on neuroblastoma cell proliferation which appear to be mediated by multiple subsets of receptors which differentially couple to MAP kinase and PKA signaling pathways.


Subject(s)
Cell Division/drug effects , Neuroblastoma/metabolism , Peptide PHI/pharmacology , Animals , Calcium-Calmodulin-Dependent Protein Kinases/metabolism , Cyclic AMP/metabolism , Humans , Mice , Neuropeptides/pharmacology , Nucleic Acid Synthesis Inhibitors/pharmacology , Pituitary Adenylate Cyclase-Activating Polypeptide , RNA, Messenger/metabolism , Receptors, Pituitary Adenylate Cyclase-Activating Polypeptide , Receptors, Pituitary Adenylate Cyclase-Activating Polypeptide , Receptors, Pituitary Hormone/metabolism , Signal Transduction/drug effects , Tumor Cells, Cultured , Vasoactive Intestinal Peptide/pharmacology
17.
Eur J Pharmacol ; 341(2-3): 289-97, 1998 Jan 12.
Article in English | MEDLINE | ID: mdl-9543251

ABSTRACT

Several lines of evidence suggest that extracellular adenosine interacting with specific cell surface receptors may influence cell growth and differentiation of cancer cells in culture. The data presented here demonstrate that various treatments of human colonic adenocarcinoma HT29 cells in the presence of exogenously added adenosine deaminase, which converts extracellular adenosine into inosine, resulted in a significant decrease of the proliferation. Cell growth inhibition was also observed in the presence of adenosine A1 receptor antagonists. These various treatments also induced a significant elevation of basal intracellular cAMP levels. This strongly indicated that extracellular adenosine was maintaining low intracellular cAMP levels in HT29 cells. A partial pharmacological characterization of the binding of the adenosine A1 receptor agonist [3H]CCPA (2-chloro-N6-cyclopentyl[2,3,4,5-(3)H]adenosine), and the adenosine A1 receptor antagonist [3H]DPCPX (cyclopentyl-1,3-dipropyl[2,3-(3)H]xanthine), to HT29 cells is also provided. Together the data support the idea that A1-adenosine receptors are expressed in HT29 cells and might mediate part of the above described effects of adenosine on cell proliferation.


Subject(s)
Adenosine/analogs & derivatives , Cell Division/drug effects , Cyclic AMP/metabolism , HT29 Cells/drug effects , Purinergic P1 Receptor Antagonists , Adenosine/metabolism , Adenosine/pharmacology , Adenosine Deaminase/metabolism , Adenosine Deaminase/pharmacology , Adenosine Deaminase Inhibitors , Binding, Competitive , HT29 Cells/metabolism , Humans , Receptors, Purinergic P1/drug effects , Receptors, Purinergic P1/metabolism
18.
Eur J Pharmacol ; 341(2-3): 299-308, 1998 Jan 12.
Article in English | MEDLINE | ID: mdl-9543252

ABSTRACT

In a previous study, we provided evidence that extracellular adenosine modulates growth of the poorly differentiated colonic adenocarcinoma cells HT29 and proposed that adenosine A1 receptors might mediate proliferative effects. We now extend our investigations to a group of colonic adenocarcinoma cells at different stages of enterocytic differentiation. In HT29, DLD-1, Caco-2 and SW403, proliferation was decreased in the presence of adenosine deaminase (5 or 10 U/ml), 5'-N-ethylcarboxamido-adenosine (NECA; 1 microM), xanthine amine congener and 8-phenyltheophylline (both at 1 nM or 1 microM). NECA stimulated cAMP accumulation in all cell lines except for HT29. In the presence of forskolin (adenyl cyclase activator) cAMP accumulation was inhibited at sub-nanomolar concentrations of NECA and stimulated at micromolar concentrations in all four cell lines. The inhibitory response disappeared in the presence of 50 nM cyclopentyladenosine (CPA). The binding of [3H]cyclopentyl-1,3-dipropylxanthine and [3H]NECA was also investigated in the four cell lines. Results of displacement experiments were consistent with the idea that poorly differentiated cells with high proliferation rates (e.g. HT29) express mainly adenosine A receptors. In contrast, displacement curves with more differentiated cells exhibiting low proliferation rates (e.g. Caco-2, DLD-1, SW403) displayed two components. The high-affinity component was no longer seen in competition experiments performed in the presence of [3H]NECA and 50 nM CPA. Together, our results further support the idea that extracellular adenosine stimulates cell proliferation in colonic adenocarcinoma cells. The effects might involve cAMP-coupled adenosine receptors.


Subject(s)
Adenosine/analogs & derivatives , Cyclic AMP/metabolism , Receptors, Purinergic P1/drug effects , Adenosine/physiology , Adenosine Deaminase/pharmacology , Adenosine-5'-(N-ethylcarboxamide)/pharmacology , Antineoplastic Agents/pharmacology , Binding, Competitive , Caco-2 Cells/drug effects , Caco-2 Cells/metabolism , Cell Differentiation/drug effects , Cell Division/drug effects , Colon/cytology , Colon/drug effects , HT29 Cells/drug effects , HT29 Cells/metabolism , Humans , Receptors, Purinergic P1/metabolism , Tumor Cells, Cultured/drug effects
19.
Cell Signal ; 10(1): 13-26, 1998 Jan.
Article in English | MEDLINE | ID: mdl-9502113

ABSTRACT

Human colonic adenocarcinoma cell lines have conserved several features of the native tissue. Among these is the expression of cell surface receptors for hormones and neurotransmitters that may be involved in the regulation of proliferation and differentiation processes in these cancer cells. Here, we confirm that high-affinity binding sites for the Vasoactive Intestinal Polypeptide (VIP) and for the VIP analogue Pituitary Adenylate-Cyclase Activating Polypeptide (PACAP), were expressed in 4 human colonic adenocarcinoma cell lines, HT29, SW403, DLD-1 and Caco-2, that spontaneously displayed variable phenotypic properties in culture. We demonstrated that after long-term treatments, VIP and PACAP significantly reduced cell proliferation in the 4 cell lines and modulated intracellular cAMP and cGMP levels. Furthermore, conspicuous differences were observed from one cell type to another concerning expression of the receptor subsets or the effects of the neuropeptides on cell growth and on cyclic nucleotides production.


Subject(s)
Receptors, Pituitary Hormone/metabolism , Receptors, Vasoactive Intestinal Peptide/metabolism , Adenocarcinoma , Caco-2 Cells , Cell Division/drug effects , Colonic Neoplasms , Cyclic AMP/metabolism , Cyclic GMP/metabolism , Growth Inhibitors/metabolism , Growth Inhibitors/pharmacology , HT29 Cells , Humans , Neuropeptides/metabolism , Neuropeptides/pharmacology , Pituitary Adenylate Cyclase-Activating Polypeptide , Receptors, Pituitary Adenylate Cyclase-Activating Polypeptide , Receptors, Pituitary Hormone/biosynthesis , Receptors, Vasoactive Intestinal Peptide/biosynthesis , Tumor Cells, Cultured , Vasoactive Intestinal Peptide/metabolism , Vasoactive Intestinal Peptide/pharmacology
20.
Peptides ; 18(6): 835-41, 1997.
Article in English | MEDLINE | ID: mdl-9285932

ABSTRACT

Neuroendocrine tumors, neuroblastoma in particular, commonly express the neuropeptides vasoactive intestinal peptide (VIP) and pituitary adenylate cyclase activating peptide (PACAP) and their receptors. Retinoic acid (RA) has been shown to induce differentiation of neuroblastoma cell lines, possibly by augmenting or interfering with neuropeptide autocrine loops. We sought to determine which receptor gene subtypes are expressed in selected human neuroblastoma cell lines (SH-SY5Y, IMR-32, and LA-N-5), and the effect of RA on the VIP/PACAP ligand/receptor system. Expression of both PACAP1 and VIP1/PACAP2 receptor genes was detected by Northern analysis, which characteristically encode Type I (PACAP-preferring), and Type II (bivalent VIP/PACAP) receptors, respectively. Binding experiments carried out on IMR-32 cells, using 125I VIP and 125I PACAP-27 as tracers, corroborated that both receptor subtypes were expressed. In contrast to RA upregulation of VIP binding (confirmed here in IMR-32 cells), levels of both receptor mRNAs were reduced after RA treatment. VIP mRNA in each cell line was increased by RA, whereas PACAP mRNA, detected in IMR-32 cells only, was reduced. The studies indicate that several components of the VIP/PACAP autocrine system are regulated in neuroblastoma cell lines during RA differentiation.


Subject(s)
Neuroblastoma/metabolism , Receptors, Pituitary Hormone/genetics , Receptors, Vasoactive Intestinal Peptide/genetics , Tretinoin/pharmacology , Autocrine Communication , Gene Expression , Humans , Ligands , Neuropeptides/genetics , Neuropeptides/metabolism , Pituitary Adenylate Cyclase-Activating Polypeptide , RNA, Messenger/metabolism , Receptors, Pituitary Adenylate Cyclase-Activating Polypeptide , Receptors, Pituitary Adenylate Cyclase-Activating Polypeptide , Receptors, Pituitary Hormone/metabolism , Receptors, Vasoactive Intestinal Peptide/metabolism , Receptors, Vasoactive Intestinal Polypeptide, Type I , Tumor Cells, Cultured , Up-Regulation/drug effects , Vasoactive Intestinal Peptide/genetics , Vasoactive Intestinal Peptide/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...