Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 62
Filter
1.
Neuroscience ; 297: 95-104, 2015 Jun 25.
Article in English | MEDLINE | ID: mdl-25841321

ABSTRACT

Attention-deficit/hyperactivity disorder (ADHD) is a complex neurobehavioral disorder that is characterized by attention difficulties, impulsivity, and hyperactivity. A non-stimulant drug, atomoxetine (ATX), which is a selective noradrenaline reuptake inhibitor, is widely used for ADHD because it exhibits fewer adverse effects compared to conventional psychostimulants. However, little is known about the therapeutic mechanisms of ATX. ATX treatment significantly alleviated hyperactivity of pituitary adenylate cyclase-activating polypeptide (PACAP)-deficient (PACAP(-/-)) mice with C57BL/6J and 129S6/SvEvTac hybrid background. ATX also improved impaired novel object recognition memory and prepulse inhibition in PACAP(-/-) mice with CD1 background. The ATX-induced increases in extracellular noradrenaline and dopamine levels were significantly higher in the prefrontal cortex of PACAP(-/-) mice compared to wild-type mice with C57BL/6J and 129S6/SvEvTac hybrid background. These results suggest that ATX treatment-induced increases in central monoamine metabolism may be involved in the rescue of ADHD-related abnormalities in PACAP(-/-) mice. Our current study suggests that PACAP(-/-) mice are an ideal rodent model with predictive validity for the study of ADHD etiology and drug development. Additionally, the potential effects of differences in genetic background of PACAP(-/-) mice on behaviors are discussed.


Subject(s)
Adrenergic Uptake Inhibitors/therapeutic use , Atomoxetine Hydrochloride/therapeutic use , Cognition Disorders/drug therapy , Hyperkinesis/drug therapy , Memory Disorders/drug therapy , Pituitary Adenylate Cyclase-Activating Polypeptide/deficiency , Prepulse Inhibition/drug effects , Acoustic Stimulation , Analysis of Variance , Animals , Biogenic Monoamines/metabolism , Cognition Disorders/genetics , Dose-Response Relationship, Drug , Exploratory Behavior/drug effects , Hyperkinesis/etiology , Memory Disorders/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Microdialysis , Pituitary Adenylate Cyclase-Activating Polypeptide/genetics , Recognition, Psychology/drug effects
2.
Br J Pharmacol ; 169(3): 512-23, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23517078

ABSTRACT

Inflammatory processes play both regenerative and destructive roles in multiple sclerosis, stroke, CNS trauma, amyotrophic lateral sclerosis and aging-related neurodegenerative diseases such as Alzheimer's, Parkinson's and Huntington's. Endogenous defence mechanisms against these pathologies include those that are directly neuroprotective, and those that modulate the expression of inflammatory mediators in microglia, astrocytes, and invading inflammatory cells. While a number of mechanisms and molecules have been identified that can directly promote neuronal survival, less is known about how the brain protects itself from harmful inflammation, and further, how it co-opts the healing function of the immune system to promote CNS repair. The two closely related neuroprotective peptides, vasoactive intestinal peptide (VIP) and pituitary adenylyl cyclase-activating peptide (PACAP), which are up-regulated in neurons and immune cells after injury and/or inflammation, are known to protect neurons, but also exert powerful in vivo immunomodulatory actions, which are primarily anti-inflammatory. These peptide actions are mediated by high-affinity receptors expressed not only on neurons, but also astrocytes, microglia and peripheral inflammatory cells. Well-established immunomodulatory actions of these peptides are to inhibit macrophage and microglia production and release of inflammatory mediators such as TNF-α and IFN-γ, and polarization of T-cell responses away from Th1 and Th17, and towards a Th2 phenotype. More recent studies have revealed that these peptides can also promote the production of both natural and inducible subsets of regulatory T-cells. The neuroprotective and immunomodulatory actions of VIP and PACAP suggest that receptors for these peptides may be therapeutic targets for neurodegenerative and neuroinflammatory diseases and other forms of CNS injury.


Subject(s)
Brain Diseases/metabolism , Central Nervous System/injuries , Central Nervous System/metabolism , Models, Biological , Pituitary Adenylate Cyclase-Activating Polypeptide/metabolism , Spinal Cord Diseases/metabolism , Vasoactive Intestinal Peptide/metabolism , Animals , Brain Diseases/immunology , Central Nervous System/immunology , Humans , Ligands , Receptors, Pituitary Adenylate Cyclase-Activating Polypeptide/metabolism , Receptors, Vasoactive Intestinal Peptide/metabolism , Signal Transduction , Spinal Cord Diseases/immunology
3.
Neuroscience ; 240: 277-86, 2013 Jun 14.
Article in English | MEDLINE | ID: mdl-23500093

ABSTRACT

Exposure to the pesticide paraquat (PQ) increases the risk of Parkinson's disease (PD), and its effect may be modulated by genetic or other environmental factors. The neuropeptide PACAP (pituitary adenylyl cyclase-activating polypeptide, Adcyap1) has been shown to enhance tyrosine hydroxylase (TH) and VMAT2 expression, protect dopaminergic (DA) neurons against the neurotoxin 6-hydroxydopamine, regulate neuronal mitochondria, and inhibit inflammation. Decreased expression of PACAP may thus interact with environmental factors such as PQ to increase the risk of PD. To mimic a low level environmental exposure to PQ, wild type (WT) and PACAP knockout (KO) mice were given a single [10 mg/kg] dose of PQ, a regimen that did not induce the loss of TH expression or DA neurons in WT mice. This treatment selectively reduced the number of TH-positive cell bodies in the substantia nigra pars compacta (SNpc) selectively in PACAP KO mice. Because inflammation is also a risk factor for PD, we performed a quantitative analysis of SNpc Iba⁺ microglia. As expected, PQ increased the number of larger microglial profiles, indicative of activation, in WT mice. Strikingly, microglial activation was already evident in PACAP KO mice in the basal state. PQ caused no further activation in these mice, although tumor necrosis factor-α gene expression was enhanced. In the periphery, PQ had no effects on the abundance of proinflammatory Th1 or Th17 cells in WT mice, but increased the numbers of anti-inflammatory regulatory T cells (Tregs). PACAP KO mice, in contrast, had elevated numbers of Th17 cells after PQ, and the induction of Tregs was impaired. The results indicate that endogenous PACAP acts to maintain the integrity of DA neurons during exposure to PQ, an action that may be linked to its ability to regulate microglia and/or other immune cells.


Subject(s)
Dopaminergic Neurons/pathology , Neurotoxicity Syndromes/immunology , Neurotoxicity Syndromes/pathology , Pituitary Adenylate Cyclase-Activating Polypeptide/deficiency , Substantia Nigra/pathology , T-Lymphocytes/pathology , Analysis of Variance , Animals , Calcium-Binding Proteins/metabolism , DNA-Binding Proteins , Disease Models, Animal , Dopaminergic Neurons/metabolism , Herbicides/toxicity , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Microfilament Proteins/metabolism , Microglia/pathology , Neurotoxicity Syndromes/etiology , Paraquat/toxicity , Substantia Nigra/drug effects , T-Lymphocytes/drug effects , T-Lymphocytes/metabolism , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/metabolism , Tyrosine 3-Monooxygenase/metabolism
4.
Neuroscience ; 151(1): 63-73, 2008 Jan 02.
Article in English | MEDLINE | ID: mdl-18055122

ABSTRACT

Peripheral nerve injury models are used to investigate processes that can potentially be exploited in CNS injury. A consistent change that occurs in injured peripheral neurons is an induction in expression of pituitary adenylyl cyclase activating peptide (PACAP), a neuropeptide with putative neuroprotective and neuritogenic actions. PACAP-deficient mice were used here to investigate actions of endogenous PACAP after facial nerve injury. Although motor neuron survival after axotomy was not significantly different in PACAP deficient vs. wild type mice, recovery of axon regeneration after crush injury was significantly delayed. The impaired regeneration was associated with 8- to 12-fold increases in gene expression of proinflammatory cytokines tumor necrosis factor-alpha, interferon-gamma, interleukin (IL) -6, and a 90% decrease in the anti-inflammatory cytokine IL-4 at the injury site. Similar cytokine changes and an increased microglial response were observed in the brainstem facial motor nucleus. Because immunocompromised animals such as SCID mice are known to exhibit peripheral nerve regeneration defects, the observations raise the novel hypothesis that PACAP is critically involved in a carefully controlled immune response that is necessary for proper nerve regeneration after injury.


Subject(s)
Inflammation/genetics , Nerve Regeneration/genetics , Pituitary Adenylate Cyclase-Activating Polypeptide/genetics , Pituitary Adenylate Cyclase-Activating Polypeptide/physiology , Animals , Axotomy , Brain Stem/metabolism , Cell Survival , Cytokines/biosynthesis , Facial Nerve/cytology , Facial Nerve/physiology , GAP-43 Protein/metabolism , Galanin/metabolism , Gliosis/pathology , Inflammation/physiopathology , Mice , Mice, Inbred C57BL , Mice, Knockout , Microglia/physiology , Motor Neurons/physiology , Nerve Crush , Nerve Regeneration/physiology , Stilbamidines
5.
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
6.
J Neurophysiol ; 97(3): 2553-8, 2007 Mar.
Article in English | MEDLINE | ID: mdl-17151217

ABSTRACT

Vasoactive intestinal polypeptide (VIP), acting via the VPAC(2) receptor, is a key signaling pathway in the suprachiasmatic nuclei (SCN), the master clock controlling daily rhythms in mammals. Most mice lacking functional VPAC(2) receptors are unable to sustain behavioral rhythms and lack detectable SCN electrical rhythms in vitro. Adult mice that do not produce VIP (VIP/PHI(-/-)) exhibit less severe alterations in wheel-running rhythms, but the effects of this deficiency on the amplitude, phasing, or periodicity of their SCN cellular rhythms are unknown. To investigate this, we used suction electrodes to extracellularly record multiple- and single-unit electrical activity in SCN brain slices from mice with varying degrees of VIP deficiency, ranging from wild-type (VIP/PHI(+/+)) to heterozygous (VIP/PHI(+/-)) and VIP/PHI(-/-) animals. We found decreasing proportions of rhythmic cells in SCN slices from VIP/PHI(+/+) ( approximately 91%, n = 23) through VIP/PHI(-/+) ( approximately 71%, n = 28) to VIP/PHI(-/-) mice (62%; n = 37) and a parallel trend toward decreasing amplitude in the remaining rhythmic cells. SCN neurons from VIP/PHI(-/-) mice exhibited a broad range in the period and phasing of electrical rhythms, concordant with the known alterations in their behavioral rhythms. Further, treatment of VIP/PHI(-/-) slices with a VPAC(2) receptor antagonist significantly reduced the proportion of oscillating neurons, suggesting that VPAC(2) receptors still become activated in the SCN of these mice. The results establish that VIP is important for appropriate periodicity and phasing of SCN neuronal rhythms and suggest that residual VPAC(2) receptor signaling promotes rhythmicity in adult VIP/PHI(-/-) mice.


Subject(s)
Action Potentials/physiology , Circadian Rhythm/genetics , Neurons/physiology , Suprachiasmatic Nucleus/cytology , Vasoactive Intestinal Peptide/deficiency , Action Potentials/drug effects , Analysis of Variance , Animals , Chi-Square Distribution , Circadian Rhythm/drug effects , Female , In Vitro Techniques , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Neurons/drug effects , Receptors, Vasoactive Intestinal Peptide, Type II/antagonists & inhibitors
7.
Ann N Y Acad Sci ; 1070: 5-9, 2006 Jul.
Article in English | MEDLINE | ID: mdl-16888146

ABSTRACT

We have taken advantage of the availability of vasoactive intestinal polypeptide (VIP) knockout (KO) mice to examine the possible influence of deletion of the VIP gene on: (a) airway reactivity and airway inflammation, as indicators of bronchial asthma; (b) mortality from endotoxemia, a model of septic shock; and (c) the pulmonary circulation. VIP KO mice showed: (a) airway hyperresponsiveness to the cholinergic agonist methacholine, as well as peribronchial and perivascular inflammation; (b) a greater susceptibility to death from endotoxemia; and (c) evidence suggestive of pulmonary hypertension.


Subject(s)
Vasoactive Intestinal Peptide/deficiency , Vasoactive Intestinal Peptide/metabolism , Animals , Bronchitis/chemically induced , Bronchitis/genetics , Bronchitis/metabolism , Disease Susceptibility , Endotoxemia/genetics , Endotoxemia/metabolism , Endotoxemia/pathology , Female , Lipopolysaccharides/pharmacology , Male , Methacholine Chloride/pharmacology , Mice , Mice, Inbred C57BL , Mice, Knockout , Survival Rate , Vasoactive Intestinal Peptide/genetics
8.
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
9.
Neuroscience ; 129(1): 93-9, 2004.
Article in English | MEDLINE | ID: mdl-15489032

ABSTRACT

Peripheral nerve inflammation is a common clinical problem that accompanies nerve injury and several diseases including Guillain-Barre syndrome and acute and chronic inflammatory demyelinating polyneuropathy. To determine if neuropeptides are induced in motor neurons after inflammation and to study the mechanisms involved, a nerve cuff soaked in complete Freund's adjuvant (CFA) was applied locally to the facial nerve of Balb/C mice. This procedure resulted in an influx of lymphocytes and macrophages to the affected area and a blockade of retrograde axonal transport distal, but not proximal, to the site of application. The same treatment resulted in a strong ipsilateral induction of pituitary adenylyl cyclase activating peptide (PACAP) gene expression in motor neurons in the facial motor nucleus. Because the changes could have occurred due to the loss of target-derived factors or to the production of new factors by immune cells, we studied the effect of the inflammatory stimulus on PACAP mRNA in mice with severe combined immunodeficiency (SCID). As expected, SCID mice showed a severely reduced influx of T-lymphocytes but not macrophages to the peripheral nerve. Moreover, although retrograde transport distal to the inflammation site was blocked similarly in control and SCID mice, the number of motor neurons expressing PACAP mRNA after CFA application was significantly reduced in SCID mice. The data indicate that the induction of PACAP mRNA during nerve inflammation requires the involvement of lymphocytes. However, because the induction of PACAP gene expression was only partially blocked in SCID mice, macrophages, loss of target-derived factors, or other mechanisms may also contribute to the upregulation of PACAP gene expression in motor neurons after nerve inflammation.


Subject(s)
Facial Nerve/metabolism , Gene Expression/immunology , Inflammation/immunology , Motor Neurons/metabolism , Neuropeptides/genetics , Severe Combined Immunodeficiency/immunology , Animals , Facial Nerve/immunology , Flow Cytometry , Freund's Adjuvant/immunology , Freund's Adjuvant/pharmacology , In Situ Hybridization , Inflammation/chemically induced , Macrophages/drug effects , Macrophages/immunology , Mice , Mice, Inbred BALB C , Motor Neurons/immunology , Neuropeptides/drug effects , Pituitary Adenylate Cyclase-Activating Polypeptide , RNA, Messenger/analysis , T-Lymphocytes/drug effects , T-Lymphocytes/immunology
10.
Cell Mol Life Sci ; 61(18): 2332-42, 2004 Sep.
Article in English | MEDLINE | ID: mdl-15378204

ABSTRACT

The concept that atrial natriuretic peptide (ANP) and the closely related peptides BNP and CNP might be involved in the ontogeny of several organ systems emerged in the late 1980s. While many of the reported in vitro actions have not been examined in the context of organ development in vivo, recent studies demonstrate that mice which lack or overexpress natriuretic peptides or receptors exhibit pronounced skeletal growth defects. This article discusses how natriuretic peptides and other factors appear to regulate bone growth as an example of how natriuretic peptides might participate in the ontogeny of other organ systems. Evidence indicating that natriuretic peptides regulate neural development is then reviewed. Natriuretic peptides and receptors exhibit complex expression patterns in the developing nervous system, where they have been shown to act on neural cells as early as at the embryonic neural tube stage. Interestingly, both bone and brain growth appear to utilize primarily CNP and the CNP-specific type B receptor, and perhaps the type C receptor. In vitro data indicate that CNP may act on developing neurons, astrocytes and Schwann cells like a classical growth factor, regulating proliferation, patterning, phenotypic specification, survival and axonal pathfinding. Natriuretic peptides might also have roles in the vascularization of the embryonic brain, establishment of the blood-brain and blood-nerve barriers, and perhaps in nerve regeneration.


Subject(s)
Bone Development , Central Nervous System/embryology , Central Nervous System/metabolism , Natriuretic Peptides/metabolism , Animals , Cell Culture Techniques , Central Nervous System/blood supply , Central Nervous System/cytology , Humans , Ligands , Natriuretic Peptides/genetics
11.
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
12.
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
13.
Neuropeptides ; 36(1): 50-7, 2002 Feb.
Article in English | MEDLINE | ID: mdl-12147214

ABSTRACT

Neuropeptides act within the pituitary as autocrine or paracrine factors, modulating the synthesis and release of primary pituitary hormones, and possibly regulating cell proliferation and/or plasticity. Manipulation of the endocrine status of rats produces dramatic long-term changes in the pituitary expression of several peptides, including the neuropeptides galanin and vasoactive intestinal peptide (VIP). Whether or not these changes are caused indirectly by hypothalamic factors, or by hormone actions directly in the pituitary, has been only partially addressed. To determine if estrogen or thyroid hormone can act directly within the pituitary to regulate VIP and galanin gene expression, cultured female rat pituitary cells were treated with 10 nM 1,17 beta-estradiol (E2) or triiodothyronine (T(3)). E2 treatment for three days resulted in an approximate 5-fold and 7-fold increase in VIP and galanin mRNA, respectively. In contrast, T(3) treatment reduced the mRNA levels of these neuropeptides to approximately 40% and 30% of control values. A time course study indicated that the actions of estrogen on VIP and galanin mRNA, and of thyroid hormone on VIP mRNA were readily apparent after 24h. The rat pituitary tumor cell line RC-4B/C was found to express easily detectable levels of galanin but not VIP mRNA. Galanin gene expression in these cells was moderately increased by E2 and decreased by T(3). Transfection of a series of luciferase plasmids containing 5 kb to 131 bp of the bovine galanin promoter fused to luciferase revealed cell-type specific enhancer sequences located between -452 and -131 bp of the galanin gene transcription start site. However, transfected plasmids were minimally responsive to E2 and T(3) treatment. Overall the results suggest that E2 and T(3) exert significant local actions in the pituitary on VIP and galanin gene expression. The bovine galanin gene fragment used in these studies contains a potential pituitary cell-type specific enhancer, but appears to lack strong E2-and T(3)-responsive sequences.


Subject(s)
Estradiol/pharmacology , Galanin/biosynthesis , Gene Expression Regulation/drug effects , Pituitary Gland, Anterior/drug effects , Triiodothyronine/pharmacology , Vasoactive Intestinal Peptide/metabolism , Animals , Cattle , Cells, Cultured/drug effects , Enhancer Elements, Genetic , Female , Galanin/genetics , Genes, Reporter , Genes, Synthetic , Luciferases/biosynthesis , Luciferases/genetics , Mice , Organ Specificity , Pituitary Neoplasms/pathology , Promoter Regions, Genetic , RNA, Messenger/biosynthesis , Rats , Rats, Inbred F344 , Rats, Sprague-Dawley , Recombinant Fusion Proteins/biosynthesis , Transfection , Tumor Cells, Cultured
14.
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
15.
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
16.
Mol Cell Endocrinol ; 181(1-2): 165-78, 2001 Jul 05.
Article in English | MEDLINE | ID: mdl-11476950

ABSTRACT

The effects of glucocorticoids on expression of the beta1-adrenergic receptor (beta1AR) gene have been varied. To study the mechanism underling hormonal regulation of the beta1AR, transient transfection of progressively deleted ovine beta1AR promoter fragments was used to identify a 43-bp region (-1274 to -1232 from the translation start site) that contains a novel glucocorticoid regulatory unit (GRU) and confers glucocorticoid responsiveness. Using DNase I footprinting and electrophoretic mobility shift assays (EMSA), we demonstrated the GRU was composed of a palindrome, 5'-TAATTA-3', which is a core binding motif for the homeodomain proteins, an E-box (5'-CACGTG-3'), binding site for the Myc/Max family proteins, and an overlapping glucocorticoid response element (GRE) half-site (5'-TGTTCT-3'). EMSA demonstrated that the GRE half-site is critical for GRU-protein interactions, which also require binding of proteins to the E-box and the homeodomain region. Co-transfection of a plasmid expressing a c-myc antisense construct significantly reduced glucocorticoid responsiveness of the ovine beta1AR promoter. Furthermore, expression of proteins binding to the GRU was shown to be developmentally regulated, being high in embryonic, reduced in newborn and not detectable in adult heart. We conclude that the ovine beta1AR promoter contains a novel, functional GRU and that glucocorticoid receptor (GR) and the Myc/Max family proteins are involved in the cell-specific nuclear factor binding and transactivation via this element. The results suggest an alternative pathway through which glucocorticoids may exert their effects on genes lacking a full consensus GRE.


Subject(s)
Gene Expression Regulation/drug effects , Glucocorticoids/pharmacology , Receptors, Adrenergic, beta-1/genetics , Response Elements/genetics , Sheep/genetics , Transcription Factors , Animals , Base Sequence , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors , Basic-Leucine Zipper Transcription Factors , Cell Line , DNA/genetics , DNA/metabolism , DNA Footprinting , DNA Probes/genetics , DNA Probes/metabolism , DNA-Binding Proteins/metabolism , Gene Expression Regulation, Developmental , Humans , Molecular Sequence Data , Promoter Regions, Genetic/genetics , Proto-Oncogene Proteins c-myc/metabolism , Rats
17.
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
18.
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
19.
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
20.
Invest Ophthalmol Vis Sci ; 41(13): 4085-92, 2000 Dec.
Article in English | MEDLINE | ID: mdl-11095600

ABSTRACT

PURPOSE: Human corneal endothelium, a neural crest-derived tissue, has a very limited regenerative capacity and may depend on trophic factors for its survival throughout life, as well as after injury and during storage before transplantation. The purpose of this study was to determine whether vasoactive intestinal peptide (VIP), a 28-amino acid neurotrophic factor present in human aqueous humor, promotes the survival of corneal endothelium in corneal organ cultures, and whether VIP is produced by the corneal endothelium. METHODS: Thirteen viable human donor corneas that had been received from the Central Florida Lions Eye Bank and stored in preservation medium (Optisol-GS; Chiron Vision, Irvine, CA) at 4 degrees C for 8 to 17 days were bisected. Each half was treated with either 0 or 10 nM VIP (15 minutes) and subjected to H(2)O(2) (1.4 mM, 30 minutes) treatment at 37 degrees C. The numbers of live and dead corneal endothelial (CE) cells isolated from the corneas were then determined under fluorescence microscopy using a live-dead viability-cytotoxicity assay conducted by an observer uninformed of the treatment. The effect of VIP (10(-16) to 10(-6) M) on CE cell survival was also studied in fresh bovine corneas in situ, by using the same assay. The presence of VIP in the corneal endothelium in fresh human donor and bovine eyes was examined by immunocytochemistry, in situ hybridization, and Western blot analysis, whereas VIP in the bovine aqueous humor was assessed by radioimmunoassay. RESULTS: VIP (10 nM) significantly increased CE survival in 10 of 13 human corneas. The mean survival of CE cells (+/-SEM) was 42% +/- 3% in control corneas versus 59% +/- 3% in VIP-treated corneas (P < 0.001). In bovine corneas, VIP at concentrations as low as 10(-10) M demonstrated a significant protective effect. The mean number of dead CE cells on bovine corneas was maximally decreased by 10(-6) M VIP from 46 +/- 5 to 18 +/- 3 per field. In CE cells from fresh human and bovine corneas, VIP immunoreactivity and mRNA were detected. VIP was also present in bovine aqueous humor at 40 +/- 8 pM. CONCLUSION: VIP may be an autocrine trophic factor that protects CE cells from H(2)O(2) in normal aqueous humor and possibly from other oxidative insults.


Subject(s)
Endothelium, Corneal/cytology , Oxidative Stress , Vasoactive Intestinal Peptide/pharmacology , Adolescent , Adult , Aged , Aged, 80 and over , Animals , Aqueous Humor/metabolism , Blotting, Western , Cattle , Cell Survival/drug effects , Cytoprotection/drug effects , Electrophoresis, Polyacrylamide Gel , Endothelium, Corneal/drug effects , Endothelium, Corneal/metabolism , Female , Humans , Hydrogen Peroxide/pharmacology , In Situ Hybridization , Male , Microscopy, Fluorescence , Middle Aged , Organ Culture Techniques , Radioimmunoassay , Receptors, Vasoactive Intestinal Peptide/metabolism , Vasoactive Intestinal Peptide/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...