Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters











Publication year range
1.
J Clin Invest ; 120(8): 2745-54, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20628197

ABSTRACT

Gait disorders and postural instability, which are commonly observed in elderly patients with Parkinson disease (PD), respond poorly to dopaminergic agents used to treat other parkinsonian symptoms. The brain structures underlying gait disorders and falls in PD and aging remain to be characterized. Using functional MRI in healthy human subjects, we have shown here that activity of the mesencephalic locomotor region (MLR), which is composed of the pedunculopontine nucleus (PPN) and the adjacent cuneiform nucleus, was modulated by the speed of imagined gait, with faster imagined gait activating a discrete cluster within the MLR. Furthermore, the presence of gait disorders in patients with PD and in aged monkeys rendered parkinsonian by MPTP intoxication correlated with loss of PPN cholinergic neurons. Bilateral lesioning of the cholinergic part of the PPN induced gait and postural deficits in nondopaminergic lesioned monkeys. Our data therefore reveal that the cholinergic neurons of the PPN play a central role in controlling gait and posture and represent a possible target for pharmacological treatment of gait disorders in PD.


Subject(s)
Gait , Parasympathetic Nervous System/physiology , Parkinson Disease/physiopathology , Pedunculopontine Tegmental Nucleus/physiology , Postural Balance , Accidental Falls , Adult , Aged , Aged, 80 and over , Animals , Female , Humans , MPTP Poisoning/physiopathology , Macaca , Male
2.
Biochem Biophys Res Commun ; 387(2): 283-8, 2009 Sep 18.
Article in English | MEDLINE | ID: mdl-19580790

ABSTRACT

Bombesin receptor subtype-3 (BRS-3) is an orphan G protein-coupled receptor in the bombesin receptor family that still awaits identification of its natural ligand. BRS-3 deficient mice develop a mild late-onset obesity with metabolic defects, implicating BRS-3 plays a role in feeding and metabolism. We describe here the pharmacological characterization of a synthetic compound, 16a, which serves as a potent agonist for BRS-3. This compound is selective for BRS-3 as it does not activate neuromedin B or gastrin-releasing peptide receptors, two most closely related bombesin receptors, as well as a series of other GPCRs. We assessed the receptor trafficking of BRS-3 and found that compound 16a promoted beta-arrestin translocation to the cell membrane. Neither central nor peripheral administration of compound 16a affects locomotor activity in mice. Therefore compound 16a is a potential tool to study the function of the BRS-3 system in vitro and possibly in vivo.


Subject(s)
Indoles/pharmacology , Receptors, Bombesin/agonists , Animals , Arrestins/metabolism , Cell Membrane/metabolism , Humans , Male , Mice , Mice, Inbred C57BL , Motor Activity/drug effects , Protein Transport/drug effects , Rats , beta-Arrestins
3.
Insect Biochem Mol Biol ; 37(4): 318-29, 2007 Apr.
Article in English | MEDLINE | ID: mdl-17368195

ABSTRACT

Adenosine receptors (AdoR) are members of the G protein-coupled receptor superfamily and mediate extracellular adenosine signaling, but the mechanism of adenosine signaling is still unclear. Here we report the first characterization of an insect AdoR, encoded by the Drosophila gene CG9753. Adenosine stimulation of Chinese hamster ovary cells carrying transiently expressed CG9753 led to a dose-dependent increase of intracellular cAMP and calcium, but untransfected controls showed no such response, showing that CG9753 encodes a functional AdoR. Endogenous CG9753 transcripts were detected in the brain, imaginal discs, ring gland and salivary glands of third-instar Drosophila larvae, and CG9753 overexpression in vivo caused lethality or severe developmental anomalies. These developmental defects were reduced by adenosine depletion, consistent with the proposed function of the CG9753 product as an AdoR. Overexpression of the G protein subunit Galpha(s) or of the catalytic subunit of protein kinase A (PKA) partially mimicked and enhanced the defects caused by ectopic expression of AdoR. Our results suggest that AdoR is an essential part of the adenosine signaling pathway and Drosophila offers a unique opportunity to use genetic analysis to study conserved aspects of the adenosine signaling pathway.


Subject(s)
Cyclic AMP/physiology , Drosophila/metabolism , Receptors, Purinergic P1/metabolism , Second Messenger Systems/physiology , Adenosine/metabolism , Amino Acid Sequence , Animals , CHO Cells , Cricetinae , Cricetulus , Drosophila/genetics , Drosophila/physiology , Gene Expression Regulation, Developmental , Hemolymph , Injections , Molecular Sequence Data , Phenotype , Receptors, Purinergic P1/genetics , Sequence Alignment
4.
J Mol Endocrinol ; 37(1): 39-50, 2006 Aug.
Article in English | MEDLINE | ID: mdl-16901922

ABSTRACT

Two glycoprotein hormone subunits, (glycoprotein hormone alpha2-subunit GPA2) and (glycoprotein hormone beta5-subunit GPB5) have been recently discovered which, when expressed in vitro, heterodimerize to form a new hormone called thyrostimulin. Thyrostimulin activates the thyroid-stimulating hormone receptor (TSHR) and has thyrotropic activity. Immunological studies have indicated that both subunits co-localize in pituitary cells. To explore the function of thyrostimulin in the rat, we have cloned rat GPA2 and GPB5, reconstituted the heterodimers in vitro, and confirmed that rat thyrostimulin activates TSHR with an affinity similar to that of TSH. In situ hybridization of the pituitary showed that while GPA2 is expressed in the anterior lobe, GPB5 is not detected in any of the lobes. A quantitative analysis showed that the co-localization of GPA2 and GPB5 is restricted in the rat to the eye and the testis. We found that GPB5 can be detected in the pituitary by quantitative-PCR, but at extremely low levels, 2000-fold lower than TSH beta-subunit (GPBtsh). Furthermore, the levels of GPB5 remain constant during the estrus cycle, while those of GPA2 vary. Finally, we found that none of the thyrostimulin subunits was induced by TRH in pituitary cell culture. These data point at the thyrostimulin system as being functionally different to the TSH system.


Subject(s)
Glycoproteins/metabolism , Pituitary Gland/metabolism , Protein Subunits/metabolism , Amino Acid Sequence , Animals , Base Sequence , Cattle , Cells, Cultured , Estrous Cycle/physiology , Glycoproteins/genetics , Humans , In Situ Hybridization , Molecular Sequence Data , Pituitary Gland/cytology , Protein Subunits/genetics , Rats , Rats, Sprague-Dawley , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Sequence Alignment , Thyrotropin/metabolism , Tissue Distribution
5.
J Comp Neurol ; 497(3): 367-83, 2006 Jul 20.
Article in English | MEDLINE | ID: mdl-16736466

ABSTRACT

In the rat, the neuropeptide B/neuropeptide W (NPB/NPW) system is composed of two ligands, neuropeptide B (NPB) and neuropeptide W (NPW), and one receptor, GPR7. Although preliminary analyses show roles in feeding, hormone secretion, and analgesia, the lack of a detailed anatomical map impairs our understanding of the NPB/NPW system. We demonstrate in this report the expression patterns of GPR7, NPB, and NPW precursor messenger ribonucleic acid (mRNA) in the rat brain by using in situ hybridization and in situ binding experiments. The amygdala expresses the highest levels of GPR7 mRNA and binding signals. Other nuclei with high levels of expression and binding are the suprachiasmatic and the ventral tuberomamillary nuclei. Moderate levels are seen in the dorsal endopiriform, dorsal tenia tecta, bed nucleus, and the red nucleus. Low levels are in the olfactory bulb, parastrial nucleus, hypothalamus, laterodorsal tegmentum, superior colliculus, locus coeruleus, and the nucleus of the solitary tract. Although the NPB precursor is mostly expressed at low levels in the brain, moderate expression is seen in anterior olfactory nucleus, piriform cortex, median preoptic nucleus, basolateral amygdala, hippocampus, medial tuberal nucleus, substantia nigra, dorsal raphe nucleus, Edinger-Westphal nucleus, and the locus coeruleus. To our surprise, the expression of NPW precursor was not detected. Our study greatly expands the preliminary in situ data previously reported. With this map of the NPB/NPW system in the rat brain, a better understanding of the functional implications of the system in various behavioral paradigms is now possible.


Subject(s)
Brain Mapping/methods , Brain/metabolism , In Situ Hybridization/methods , Neuropeptides/metabolism , RNA, Messenger/metabolism , Receptors, G-Protein-Coupled/metabolism , Receptors, Neuropeptide/metabolism , Animals , Brain/anatomy & histology , Gene Expression Profiling/methods , Male , Molecular Probes/metabolism , Neurons/cytology , Neurons/metabolism , Rats , Rats, Sprague-Dawley
6.
Eur J Pharmacol ; 539(3): 145-50, 2006 Jun 13.
Article in English | MEDLINE | ID: mdl-16697365

ABSTRACT

The mas-like G protein-coupled receptors form a subfamily of G protein-coupled receptors that includes variable member numbers across different species and that have been shown to bind a wide variety of ligands from peptides to amino acid derivatives. While screening a library of peptides against different orphan G protein-coupled receptors, we found that human salusin beta activates the mouse mas-like G protein-coupled receptor, mMrgA1 with an EC(50) of about 300 nM. Salusin beta is a bioactive peptide recently discovered through bioinformatics analysis which stimulates arginine-vasopressin release from rat pituitary and causes rapid and profound hypotension and bradycardia. However, when we further analyzed the generality of the mMrgA1 activation, we found that human salusin beta does not activate corresponding human mas-like G protein-coupled receptors. Our results show that human salusin beta is a surrogate ligand of the mouse MrgA1 and raises a cautionary flag for experiments that analyze the pharmacological profiles of mas-like G protein-coupled receptors from different species.


Subject(s)
Adenosine Triphosphatases/metabolism , Receptors, G-Protein-Coupled/metabolism , Adenosine Triphosphatases/genetics , Amino Acid Sequence , Animals , Cell Line , Dose-Response Relationship, Drug , Humans , Intercellular Signaling Peptides and Proteins , Ligands , Mice , Molecular Sequence Data , Rats , Receptors, G-Protein-Coupled/genetics
7.
Neuroreport ; 17(8): 813-6, 2006 May 29.
Article in English | MEDLINE | ID: mdl-16708020

ABSTRACT

GPR39, an orphan G protein-coupled receptor, has been recently identified as the receptor for the bioactive peptide obestatin. Obestatin is secreted from the stomach and acts as an anti-appetite hormone. This activity is induced whether obestatin is administered intraperitoneally or intracerebroventricularly. GPR39 is known to be expressed in the central nervous system but its precise localization is unknown. In view of the growing importance of this system, we decided to study the sites of GPR39 mRNA expression by in-situ hybridization. We find the highest levels of GPR39 mRNA in the amygdala, the hippocampus, and the auditory cortex and low levels in several other brain regions. Surprisingly, we find no expression of GPR39 in the hypothalamus, expected to be the site of the anorexigenic action of obestatin.


Subject(s)
Appetite Regulation/physiology , Brain/metabolism , Peptide Hormones/metabolism , Receptors, G-Protein-Coupled/genetics , Amygdala/anatomy & histology , Amygdala/metabolism , Animals , Auditory Cortex/anatomy & histology , Auditory Cortex/metabolism , Brain/anatomy & histology , Gene Expression/physiology , Hippocampus/anatomy & histology , Hippocampus/metabolism , Hypothalamus/anatomy & histology , Hypothalamus/metabolism , Male , Mice , Mice, Inbred C57BL , RNA, Messenger/metabolism
8.
Pharmacol Ther ; 110(3): 525-32, 2006 Jun.
Article in English | MEDLINE | ID: mdl-16289308

ABSTRACT

Due to their diversity, G-protein-coupled receptors (GPCRs) are major regulators of intercellular interactions. They exert their actions by being activated by a vast array of natural ligands, referred to in this article as "transmitters". Yet each GPCR is highly selective in its ligand recognition. Traditionally, the transmitters were found first and served to characterize the receptors pharmacologically. Since the end of the 1980s, however, it is the GPCRs that are first to be found because they are identified molecularly by homology screening approaches. But the GPCRs found this way suffer of one drawback, they lack their natural transmitters, they are "orphan" GPCRs. Searching for transmitters of orphan GPCRs has given birth to the reverse pharmacology approach that uses orphan GPCRs as targets to identify their transmitters. The most salient successes of the reverse pharmacology approach were the discoveries of 9 novel neuropeptide families. These have enriched our understanding of several important behavioral responses. But the application of reverse pharmacology has also led to some surprising results that question some basic pharmacological concepts. This review aims at describing the history of the orphan GPCRs and their impact on our understanding of biology.


Subject(s)
Receptors, G-Protein-Coupled/physiology , Animals , Humans , Ligands , Signal Transduction
9.
FEBS J ; 272(22): 5694-702, 2005 Nov.
Article in English | MEDLINE | ID: mdl-16279935

ABSTRACT

The discovery of novel biologically active peptides has led to an explosion in our understanding of the molecular mechanisms that underlie the regulation of sleep and wakefulness. Urotensin II (UII), a peptide originally isolated from fish and known for its strong cardiovascular effects in mammals, is another surprising candidate in the regulatory network of sleep. The UII receptor was found to be expressed by cholinergic neurons of laterodorsal and pedunculopontine tegmental nuclei, an area known to be of utmost importance for the on- and offset of rapid eye movement (REM) sleep. Recently, physiological data have provided further evidence that UII is indeed a modulator of REM sleep. The peptide directly excites cholinergic mesopontine neurons and increases the rate of REM sleep episodes. These new results and its emerging behavioral effects establish UII as a neurotransmitter/neuromodulator in mammals and should spark further interest into the neurobiological role of the peptide.


Subject(s)
Arousal , Heart/physiology , Neuropeptides/physiology , Neurophysiology , Sleep, REM/physiology , Urotensins/physiology , Acetylcholine/metabolism , Acetylcholine/pharmacology , Acetylcholine/physiology , Animals , Models, Neurological , Neuropeptides/chemistry , Neuropeptides/genetics , Pons/physiology , Protein Isoforms , Receptors, G-Protein-Coupled/physiology , Receptors, Presynaptic/physiology , Sleep, REM/drug effects , Tegmentum Mesencephali/physiology , Urotensins/chemistry , Urotensins/genetics
10.
J Neurophysiol ; 94(5): 3648-52, 2005 Nov.
Article in English | MEDLINE | ID: mdl-16222076

ABSTRACT

Metastin is an antimetastatic peptide encoded by the KiSS-1 gene in cancer cells. Recent studies found that metastin is a ligand for the orphan G-protein-coupled receptor GPR54, which is highly expressed in specific brain regions such as the hypothalamus and parts of the hippocampus. This study shows that activation of GPR54 by submicromolar concentrations of metastin reversibly enhances excitatory synaptic transmission in hippocampal dentate granule cells in a mitogen-activated protein (MAP) kinase-dependent manner. Synaptic enhancement by metastin was suppressed by intracellular application of the G-protein inhibitor GDP-beta-S and the calcium chelator BAPTA. Analysis of miniature excitatory postsynaptic currents (mEPSCs) revealed an increase in the mean amplitude but no change in event frequency. This indicates that GPR54 and the mechanism responsible for the increase in EPSCs are postsynaptic. Metastin-induced synaptic potentiation was abolished by 50 microM PD98059 and 20 microM U0126, two inhibitors of the MAP kinases ERK1 and ERK2. The effect was also blocked by inhibitors of calcium/calmodulin-dependent kinases and tyrosine kinases. RT-PCR experiments showed that both KiSS-1 and GPR54 are expressed in the hippocampal dentate gyrus. Metastin is thus a novel endogenous factor that modulates synaptic excitability in the dentate gyrus through mechanisms involving MAP kinases, which in turn may be controlled upstream by calcium-activated kinases and tyrosine kinases.


Subject(s)
Action Potentials/physiology , Excitatory Postsynaptic Potentials/physiology , Hippocampus/physiology , MAP Kinase Signaling System/physiology , Mitogen-Activated Protein Kinases/metabolism , Proteins/pharmacology , Synaptic Transmission/physiology , Action Potentials/drug effects , Animals , Cells, Cultured , Excitatory Postsynaptic Potentials/drug effects , Hippocampus/drug effects , Kisspeptins , Rats , Rats, Sprague-Dawley , Synaptic Transmission/drug effects , Tumor Suppressor Proteins
11.
Brain Res ; 1059(2): 139-48, 2005 Oct 19.
Article in English | MEDLINE | ID: mdl-16183039

ABSTRACT

Urotensin II (UII) is a vasomodulatory peptide that was not predicted to elicit CNS activity. However, because we have recently shown that the urotensin II receptor (UII-R) is selectively expressed in rat mesopontine cholinergic (MPCh) neurons, we hypothesize that UII may have a central function. The present study demonstrates that the UII system is able to modulate MPCh neuron activity. Brain slice experiments demonstrate that UII excites MPCh neurons of the mouse laterodorsal tegmentum (LDTg) by activating a slow inward current. Furthermore, microinfusion of UII into the ventral tegmental area produces a sustained increase in dopamine efflux in the nucleus accumbens, as measured by in vivo chronoamperometry. In agreement with UII activation of MPCh neurons, intracerebroventricular injections of UII significantly modulate ambulatory movements in both rats and mice but do not significantly affect startle habituation or prepulse inhibition. The present study establishes that UII is a neuromodulator that may be exploited to target disorders involving MPCh dysfunction.


Subject(s)
Cholinergic Fibers/physiology , Neurons/physiology , Pons/physiology , Urotensins/physiology , Animals , Electrophysiology , Habituation, Psychophysiologic/physiology , Male , Membrane Potentials/physiology , Mice , Motor Activity/physiology , Nucleus Accumbens/cytology , Nucleus Accumbens/physiology , Organ Culture Techniques , Pons/cytology , Rats , Rats, Wistar , Reflex, Startle/physiology , Species Specificity , Tegmentum Mesencephali/cytology , Tegmentum Mesencephali/physiology
12.
Eur J Pharmacol ; 519(1-2): 191-3, 2005 Sep 05.
Article in English | MEDLINE | ID: mdl-16111673

ABSTRACT

The G protein-coupled receptor MrgX2 belongs to the large family of the Mas-related genes or sensory neuron-specific G protein-coupled receptors. The MrgX2 receptor has been shown to be activated by the peptides cortistatin and proadrenomedullin N-terminal peptides (PAMP). Here we investigated the structure activity relationship of PAMP and identified key structural features that are shared with cortistatin and might explain why two apparently unrelated peptides are able to activate a single G protein-coupled receptor.


Subject(s)
Neuropeptides/metabolism , Protein Precursors/metabolism , Proteins/metabolism , Receptors, Neuropeptide/metabolism , Adrenomedullin , Amino Acid Motifs/genetics , Amino Acid Sequence , Binding Sites/genetics , Binding, Competitive , Humans , Molecular Sequence Data , Nerve Tissue Proteins , Neuropeptides/chemistry , Neuropeptides/genetics , Protein Precursors/chemistry , Protein Precursors/genetics , Proteins/chemistry , Proteins/genetics , Receptors, G-Protein-Coupled
13.
J Neurosci ; 25(23): 5465-74, 2005 Jun 08.
Article in English | MEDLINE | ID: mdl-15944374

ABSTRACT

Urotensin II (UII) is a cyclic neuropeptide with strong vasoconstrictive activity in the peripheral vasculature. UII receptor mRNA is also expressed in the CNS, in particular in cholinergic neurons located in the mesopontine tegmental area, including the pedunculopontine tegmental (PPT) and lateral dorsal tegmental nuclei. This distribution suggests that the UII system is involved in functions regulated by acetylcholine, such as the sleep-wake cycle. Here, we tested the hypothesis that UII influences cholinergic PPT neuron activity and alters rapid eye movement (REM) sleep patterns in rats. Local administration of UII into the PPT nucleus increases REM sleep without inducing changes in the cortical blood flow. Intracerebroventricular injection of UII enhances both REM sleep and wakefulness and reduces slow-wave sleep 2. Intracerebroventricular, but not local, administration of UII increases cortical blood flow. Moreover, whole-cell recordings from rat-brain slices show that UII selectively excites cholinergic PPT neurons via an inward current and membrane depolarization that were accompanied by membrane conductance decreases. This effect does not depend on action potential generation or fast synaptic transmission because it persisted in the presence of TTX and antagonists of ionotropic glutamate, GABA, and glycine receptors. Collectively, these results suggest that UII plays a role in the regulation of REM sleep independently of its cerebrovascular actions by directly activating cholinergic brainstem neurons.


Subject(s)
Acetylcholinesterase/metabolism , Neurons/physiology , Sleep, REM/physiology , Tegmentum Mesencephali/physiology , Urotensins/physiology , Animals , Cerebrovascular Circulation , Electroencephalography , Electromyography , In Vitro Techniques , Injections, Intraventricular , Male , Neurons/metabolism , Patch-Clamp Techniques , Pedunculopontine Tegmental Nucleus/cytology , Pedunculopontine Tegmental Nucleus/physiology , Rats , Rats, Sprague-Dawley , Receptors, G-Protein-Coupled/metabolism , Tegmentum Mesencephali/blood supply , Tegmentum Mesencephali/cytology , Urotensins/pharmacology , Wakefulness
SELECTION OF CITATIONS
SEARCH DETAIL