Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Front Immunol ; 11: 293, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32194553

RESUMEN

Inflammation is considered a mechanistic driver of Alzheimer's disease, thought to increase tau phosphorylation, the first step to the formation of neurofibrillary tangles (NFTs). To further understand how inflammation impacts the development of tau pathology, we used (hTau) mice, which express all six, non-mutated, human tau isoforms, but with an altered ratio of tau isoforms favoring 3R tau due to the concomitant loss of murine tau (mTau) that is predominantly 4R. Such an imbalance pattern has been related to susceptibility to NFTs formation, but whether or not this also affects susceptibility to systemic inflammation and related changes in tau phosphorylation is not known. To reduce the predominance of 3R tau by increasing 4R tau availability, we bred hTau mice on a heterozygous mTau background and compared the impact of systemic inflammation induced by lipopolysaccharide (LPS) in hTau mice hetero- or homozygous mTau knockout. Three-month-old male wild-type (Wt), mTau+/-, mTau-/-, hTau/mTau+/-, and hTau/mTau-/- mice were administered 100, 250, or 330 µg/kg of LPS or its vehicle phosphate buffer saline (PBS) [intravenously (i.v.), n = 8-9/group]. Sickness behavior, reflected by behavioral suppression in the spontaneous alternation task, hippocampal tau phosphorylation, measured by western immunoblotting, and circulating cytokine levels were quantified 4 h after LPS administration. The persistence of the LPS effects (250 µg/kg) on these measures, and food burrowing behavior, was assessed at 24 h post-inoculation in Wt, mTau+/-, and hTau/mTau+/- mice (n = 9-10/group). In the absence of immune stimulation, increasing 4R tau levels in hTau/mTau+/- exacerbated pS202 and pS396/404 tau phosphorylation, without altering total tau levels or worsening early behavioral perturbations characteristic of hTau/mTau-/- mice. We also show for the first time that modulating 4R tau levels in hTau mice affects the response to systemic inflammation. Behavior was suppressed in all genotypes 4 h following LPS administration, but hTau/mTau+/- exhibited more severe sickness behavior at the 100 µg/kg dose and a milder behavioral and cytokine response than hTau/mTau-/- mice at the 330 µg/kg dose. All LPS doses decreased tau phosphorylation at both epitopes in hTau/mTau+/- mice, but pS202 levels were selectively reduced at the 100 µg/kg dose in hTau/mTau-/- mice. Behavioral suppression and decreased tau phosphorylation persisted at 24 h following LPS administration in hTau/mTau+/- mice.


Asunto(s)
Hipocampo/metabolismo , Inflamación/complicaciones , Tauopatías/etiología , Proteínas tau/metabolismo , Animales , Citocinas/biosíntesis , Lipopolisacáridos/toxicidad , Masculino , Ratones , Ratones Endogámicos C57BL , Fosforilación , Isoformas de Proteínas/análisis , Proteínas tau/análisis
2.
Acta Neuropathol Commun ; 8(1): 13, 2020 02 04.
Artículo en Inglés | MEDLINE | ID: mdl-32019610

RESUMEN

Tau deposition in the brain is a pathological hallmark of many neurodegenerative disorders, including Alzheimer's disease (AD). During the course of these tauopathies, tau spreads throughout the brain via synaptically-connected pathways. Such propagation of pathology is thought to be mediated by tau species ("seeds") containing the microtubule binding region (MTBR) composed of either three repeat (3R) or four repeat (4R) isoforms. The tau MTBR also forms the core of the neuropathological filaments identified in AD brain and other tauopathies. Multiple approaches are being taken to limit tau pathology, including immunotherapy with anti-tau antibodies. Given its key structural role within fibrils, specifically targetting the MTBR with a therapeutic antibody to inhibit tau seeding and aggregation may be a promising strategy to provide disease-modifying treatment for AD and other tauopathies. Therefore, a monoclonal antibody generating campaign was initiated with focus on the MTBR. Herein we describe the pre-clinical generation and characterisation of E2814, a humanised, high affinity, IgG1 antibody recognising the tau MTBR. E2814 and its murine precursor, 7G6, as revealed by epitope mapping, are antibodies bi-epitopic for 4R and mono-epitopic for 3R tau isoforms because they bind to sequence motif HVPGG. Functionally, both antibodies inhibited tau aggregation in vitro. They also immunodepleted a variety of MTBR-containing tau protein species. In an in vivo model of tau seeding and transmission, attenuation of deposition of sarkosyl-insoluble tau in brain could also be observed in response to antibody treatment. In AD brain, E2814 bound different types of tau filaments as shown by immunogold labelling and recognised pathological tau structures by immunohistochemical staining. Tau fragments containing HVPGG epitopes were also found to be elevated in AD brain compared to PSP or control. Taken together, the data reported here have led to E2814 being proposed for clinical development.


Asunto(s)
Enfermedad de Alzheimer/inmunología , Enfermedad de Alzheimer/terapia , Anticuerpos Monoclonales/inmunología , Inmunización Pasiva/métodos , Proteínas tau/genética , Proteínas tau/inmunología , Enfermedad de Alzheimer/patología , Animales , Anticuerpos Monoclonales/farmacología , Lóbulo Frontal/metabolismo , Lóbulo Frontal/patología , Inmunoglobulina G/inmunología , Inmunoglobulina G/farmacología , Masculino , Ratones Transgénicos , Agregación Patológica de Proteínas/inmunología , Isoformas de Proteínas/inmunología , Isoformas de Proteínas/farmacología
3.
Exp Gerontol ; 94: 103-107, 2017 08.
Artículo en Inglés | MEDLINE | ID: mdl-27979768

RESUMEN

Alzheimer's disease (AD), the predominant form of dementia, is highly correlated with the abnormal hyperphosphorylation and aggregation of tau. Immune responses are key drivers of AD and how they contribute to tau pathology in human disease remains largely unknown. This review summarises current knowledge on the association between inflammatory processes and tau pathology. While, preclinical evidence suggests that inflammation can indeed induce tau hyperphosphorylation at both pre- and post-tangles epitopes, a better understanding of whether this develops into advanced pathological features such as neurofibrillary tangles is needed. Microglial cells, the immune phagocytes in the central nervous system, appear to play a key role in regulating tau pathology, but the underlying mechanisms are not fully understood. Their activation can be detrimental via the secretion of pro-inflammatory mediators, particularly interleukin-1ß, but also potentially beneficial through phagocytosis of extracellular toxic tau oligomers. Nevertheless, anti-inflammatory treatments in animal models were found protective, but whether or not they affect microglial phagocytosis of tau species is unknown. However, one major challenge to our understanding of the role of inflammation in the progression of tau pathology is the preclinical models used to address this question. They mostly rely on the use of septic doses of lipopolysaccharide that do not reflect the inflammatory conditions experienced AD patients, questioning whether the impact of inflammation on tau pathology in these models is dose-dependent and relevant to the human disease. The use of more translational models of inflammation corroborated with verification in clinical investigations are necessary to progress our understanding of the interplay between inflammation and tau pathology.


Asunto(s)
Enfermedad de Alzheimer/metabolismo , Encéfalo/metabolismo , Mediadores de Inflamación/metabolismo , Inflamación/metabolismo , Agregación Patológica de Proteínas , Proteínas tau/metabolismo , Enfermedad de Alzheimer/tratamiento farmacológico , Enfermedad de Alzheimer/patología , Enfermedad de Alzheimer/fisiopatología , Animales , Antiinflamatorios/uso terapéutico , Encéfalo/efectos de los fármacos , Encéfalo/patología , Encéfalo/fisiopatología , Humanos , Inflamación/tratamiento farmacológico , Inflamación/patología , Inflamación/fisiopatología , Fármacos Neuroprotectores/uso terapéutico , Fosforilación , Transducción de Señal
4.
Br J Pharmacol ; 174(5): 370-385, 2017 03.
Artículo en Inglés | MEDLINE | ID: mdl-28009436

RESUMEN

BACKGROUND AND PURPOSE: AMPA receptor positive allosteric modulators represent a potential therapeutic strategy to improve cognition in people with schizophrenia. These studies collectively constitute the preclinical pharmacology data package used to build confidence in the pharmacology of this molecule and enable a clinical trial application. EXPERIMENTAL APPROACH: [N-[(2S)-5-(6-fluoro-3-pyridinyl)-2,3-dihydro 1H-inden-2-yl]-2-propanesulfonamide] (UoS12258) was profiled in a number of in vitro and in vivo studies to highlight its suitability as a novel therapeutic agent. KEY RESULTS: We demonstrated that UoS12258 is a selective, positive allosteric modulator of the AMPA receptor. At rat native hetero-oligomeric AMPA receptors, UoS12258 displayed a minimum effective concentration of approximately 10 nM in vitro and enhanced AMPA receptor-mediated synaptic transmission at an estimated free brain concentration of approximately 15 nM in vivo. UoS12258 reversed a delay-induced deficit in novel object recognition in rats after both acute and sub-chronic dosing. Sub-chronic dosing reduced the minimum effective dose from 0.3 to 0.03 mg·kg-1 . UoS12258 was also effective at improving performance in two other cognition models, passive avoidance in scopolamine-impaired rats and water maze learning and retention in aged rats. In side-effect profiling studies, UoS12258 did not produce significant changes in the maximal electroshock threshold test at doses below 10 mg·kg-1 . CONCLUSION AND IMPLICATIONS: We conclude that UoS12258 is a potent and selective AMPA receptor modulator exhibiting cognition enhancing properties in several rat behavioural models superior to other molecules that have previously entered clinical evaluation.


Asunto(s)
Conducta Animal/efectos de los fármacos , Indenos/farmacología , Nootrópicos/farmacología , Receptores AMPA/efectos de los fármacos , Sulfonamidas/farmacología , Regulación Alostérica/efectos de los fármacos , Animales , Reacción de Prevención/efectos de los fármacos , Cognición/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Electrochoque , Humanos , Indenos/administración & dosificación , Indenos/toxicidad , Masculino , Aprendizaje por Laberinto/efectos de los fármacos , Nootrópicos/administración & dosificación , Nootrópicos/toxicidad , Ratas , Ratas Sprague-Dawley , Ratas Wistar , Receptores AMPA/metabolismo , Reconocimiento en Psicología/efectos de los fármacos , Escopolamina/toxicidad , Sulfonamidas/administración & dosificación , Sulfonamidas/toxicidad
5.
Bioorg Med Chem Lett ; 25(24): 5792-6, 2015 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-26531152

RESUMEN

Herein we describe a series of tetrahydrobenzotriazoles as novel, potent metabotropic glutamate receptor subtype 5 (mGlu5) positive allosteric modulators (PAMs). Exploration of the SAR surrounding the tetrahydrobenzotriazole core ultimately led to the identification of 29 as a potent mGlu5 PAM with a low maximal glutamate potency fold shift, acceptable in vitro DMPK parameters and in vivo PK profile and efficacy in the rat novel object recognition (NOR) assay. As a result 29 was identified as a suitable compound for progression to in vivo safety evaluation.


Asunto(s)
Antipsicóticos/química , Receptor del Glutamato Metabotropico 5/antagonistas & inhibidores , Triazoles/química , Regulación Alostérica/efectos de los fármacos , Animales , Antipsicóticos/metabolismo , Antipsicóticos/farmacología , Astrocitos/citología , Astrocitos/efectos de los fármacos , Astrocitos/metabolismo , Cognición/efectos de los fármacos , Modelos Animales de Enfermedad , Evaluación Preclínica de Medicamentos , Semivida , Humanos , Microsomas/metabolismo , Ratas , Receptor del Glutamato Metabotropico 5/metabolismo , Relación Estructura-Actividad , Triazoles/metabolismo , Triazoles/farmacología
6.
Bioorg Med Chem Lett ; 20(18): 5434-8, 2010 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-20709550

RESUMEN

A series of N-substituted 3-(4-piperidinyl)-1,3-benzoxazolinones and oxindoles are reported which were found to be potent and selective muscarinic M1 agonists. By control of the physicochemical characteristics of the series, particularly the lipophilicity, compounds with good metabolic stability and excellent brain penetration were identified. An exemplar of the series was shown to be pro-cognitive in the novel object recognition rat model of temporal induced memory deficit.


Asunto(s)
Benzoxazoles/farmacocinética , Indoles/farmacocinética , Trastornos de la Memoria/tratamiento farmacológico , Agonistas Muscarínicos/farmacocinética , Nootrópicos/farmacocinética , Receptor Muscarínico M1/metabolismo , Animales , Benzoxazoles/química , Benzoxazoles/uso terapéutico , Encéfalo/metabolismo , Indoles/química , Indoles/uso terapéutico , Agonistas Muscarínicos/química , Agonistas Muscarínicos/uso terapéutico , Nootrópicos/química , Nootrópicos/uso terapéutico , Oxindoles , Ratas
7.
Eur J Pharmacol ; 603(1-3): 147-9, 2009 Jan 28.
Artículo en Inglés | MEDLINE | ID: mdl-19111716

RESUMEN

The muscarinic acetylcholine receptor (mAChR) agonist, xanomeline, attenuates amphetamine-induced activity in WT mice. This effect is abolished in mice lacking the M(4) muscarinic acetylcholine receptor (M(4) mAChR KO) and partially attenuated in mice lacking M(1) muscarinic acetylcholine receptor (M(1) mAChR KO). Collectively, these data suggest that the efficacy exhibited by xanomeline in the mouse amphetamine-induced hyperactivity model, is mediated predominantly by M(4) muscarinic acetylcholine receptors, and that M(1) muscarinic acetylcholine receptors may play a more minor role. This supports the hypothesis that activation of M(4), and to a lesser extent M(1) muscarinic acetylcholine receptors, may represent a potential target for the treatment of psychosis seen in schizophrenia.


Asunto(s)
Anfetamina/toxicidad , Agonistas Muscarínicos/farmacología , Piridinas/farmacología , Receptor Muscarínico M1/deficiencia , Receptor Muscarínico M1/genética , Receptor Muscarínico M4/deficiencia , Receptor Muscarínico M4/genética , Tiadiazoles/farmacología , Animales , Hipercinesia/inducido químicamente , Hipercinesia/tratamiento farmacológico , Ratones , Ratones Noqueados , Agonistas Muscarínicos/uso terapéutico , Piridinas/uso terapéutico , Especificidad por Sustrato , Tiadiazoles/uso terapéutico
8.
Psychopharmacology (Berl) ; 201(4): 483-94, 2009 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-18762914

RESUMEN

OBJECTIVES: To test the novel nonimidazole histamine H3 receptor antagonist 5-[(3-cyclobutyl-2,3,4,5-tetrahydro-1H-3-benzazapin-7-yl)oxy]-N-methyl-2-pyrazinecarboxamide (GSK207040) in a series of behavioral and neurochemical paradigms designed to evaluate its antipsychotic potential. MATERIALS AND METHODS: Acute orally administered GSK207040 was investigated for its capacity to reverse a 24-h-induced deficit in novel object recognition memory, deficits in prepulse inhibition (PPI) induced by isolation rearing, and hyperlocomotor activity induced by amphetamine. The acute neurochemical effects of GSK207040 were explored by analyzing rat anterior cingulate cortex microdialysates for levels of dopamine, noradrenaline, and acetylcholine and by c-fos immunohistochemistry. The potential for interaction with the antipsychotic dopamine D2 receptor antagonist haloperidol was explored behaviorally (spontaneous locomotor activity and catalepsy), biochemically (plasma prolactin), and via ex vivo receptor occupancy determinations. RESULTS: GSK207040 significantly enhanced object recognition memory (3 mg/kg) and attenuated isolation rearing-induced deficits in PPI (1.0 and 3.2 mg/kg) but did not reverse amphetamine-induced increases in locomotor activity. There was no evidence of an interaction of GSK207040 with haloperidol. GSK207040 (3.2 mg/kg) raised extracellular concentrations of dopamine, noradrenaline, and acetylcholine in the anterior cingulate cortex and c-fos expression in the core of the nucleus accumbens was increased at doses of 3.2 and 10.0 mg/kg. CONCLUSIONS: The behavioral and neurochemical profile of GSK207040 supports the potential of histamine H3 receptor antagonism to treat the cognitive and sensory gating deficits of schizophrenia. However, the failure of GSK207040 to reverse amphetamine-induced locomotor hyperactivity suggests that the therapeutic utility of histamine H(3) receptor antagonism versus positive symptoms is less likely, at least following acute administration.


Asunto(s)
Antipsicóticos/farmacología , Benzazepinas/farmacología , Antagonistas de los Receptores Histamínicos/farmacología , Pirazinas/farmacología , Esquizofrenia/tratamiento farmacológico , Administración Oral , Anfetamina/farmacología , Animales , Antipsicóticos/administración & dosificación , Conducta Animal/efectos de los fármacos , Benzazepinas/administración & dosificación , Relación Dosis-Respuesta a Droga , Antagonistas de los Receptores Histamínicos/administración & dosificación , Hipercinesia/inducido químicamente , Hipercinesia/prevención & control , Masculino , Memoria/efectos de los fármacos , Pirazinas/administración & dosificación , Ratas , Ratas Sprague-Dawley , Receptores Histamínicos H3/efectos de los fármacos , Reconocimiento en Psicología/efectos de los fármacos , Esquizofrenia/fisiopatología , Aislamiento Social/psicología
9.
Neuropsychopharmacology ; 33(7): 1642-52, 2008 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-17728699

RESUMEN

Neurokinin-3 (NK3) receptors are concentrated in forebrain and basal ganglia structures within the mammalian CNS. This distribution, together with the modulatory influence of NK3 receptors on monoaminergic neurotransmission, has led to the hypothesis that NK3 receptor antagonists may have therapeutic efficacy in the treatment of psychiatric disorders. Here we describe the in vitro and in vivo characterization of the highly selective NK3 receptor antagonist talnetant (SB-223412). Talnetant has high affinity for recombinant human NK3 receptors (pKi 8.7) and demonstrates selectivity over other neurokinin receptors (pKi NK2 = 6.6 and NK1<4). In native tissue-binding studies, talnetant displayed high affinity for the guinea pig NK3 receptor (pKi 8.5). Functionally, talnetant competitively antagonized neurokinin B (NKB)-induced responses at the human recombinant receptor in both calcium and phosphoinositol second messenger assay systems (pA2 of 8.1 and 7.7, respectively). In guinea pig brain slices, talnetant antagonized NKB-induced increases in neuronal firing in the medial habenula (pKB = 7.9) and senktide-induced increases in neuronal firing in the substantia nigra pars compacta (pKB = 7.7) with no diminution of maximal agonist efficacy, suggesting competitive antagonism at native NK3 receptors. Talnetant (3-30 mg/kg i.p.) significantly attenuated senktide-induced 'wet dog shake' behaviors in the guinea pig in a dose-dependent manner. Microdialysis studies demonstrated that acute administration of talnetant (30 mg/kg i.p.) produced significant increases in extracellular dopamine and norepinephrine in the medial prefrontal cortex and attenuated haloperidol-induced increases in nucleus accumbens dopamine levels in the freely moving guinea pigs. Taken together, these data demonstrate that talnetant is a selective, competitive, brain-penetrant NK3 receptor antagonist with the ability to modulate mesolimbic and mesocortical dopaminergic neurotransmission and hence support its potential therapeutic utility in the treatment of schizophrenia.


Asunto(s)
Encéfalo/efectos de los fármacos , Quinolinas/farmacología , Receptores de Neuroquinina-3/antagonistas & inhibidores , Animales , Encéfalo/citología , Calcio/metabolismo , Línea Celular Transformada , Línea Celular Tumoral , Dopamina/metabolismo , Relación Dosis-Respuesta a Droga , Cobayas , Humanos , Técnicas In Vitro , Masculino , Neuronas/efectos de los fármacos , Neurotransmisores/metabolismo , Unión Proteica/efectos de los fármacos , Receptores de Neuroquinina-3/efectos de los fármacos , Receptores de Neuroquinina-3/metabolismo
11.
J Pharmacol Exp Ther ; 321(3): 1032-45, 2007 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-17327487

RESUMEN

6-[(3-Cyclobutyl-2,3,4,5-tetrahydro-1H-3-benzazepin-7-yl)oxy]-N-methyl-3-pyridinecarboxamide hydrochloride (GSK189254) is a novel histamine H(3) receptor antagonist with high affinity for human (pK(i) = 9.59 -9.90) and rat (pK(i) = 8.51-9.17) H(3) receptors. GSK189254 is >10,000-fold selective for human H(3) receptors versus other targets tested, and it exhibited potent functional antagonism (pA(2) = 9.06 versus agonist-induced changes in cAMP) and inverse agonism [pIC(50) = 8.20 versus basal guanosine 5'-O-(3-[(35)S]thio)triphosphate binding] at the human recombinant H(3) receptor. In vitro autoradiography demonstrated specific [(3)H]GSK189254 binding in rat and human brain areas, including cortex and hippocampus. In addition, dense H(3) binding was detected in medial temporal cortex samples from severe cases of Alzheimer's disease, suggesting for the first time that H(3) receptors are preserved in late-stage disease. After oral administration, GSK189254 inhibited cortical ex vivo R-(-)-alpha-methyl[imidazole-2,5(n)-(3)H]histamine dihydrochloride ([(3)H]R-alpha-methylhistamine) binding (ED(50) = 0.17 mg/kg) and increased c-Fos immunoreactivity in prefrontal and somatosensory cortex (3 mg/kg). Microdialysis studies demonstrated that GSK189254 (0.3-3 mg/kg p.o.) increased the release of acetylcholine, noradrenaline, and dopamine in the anterior cingulate cortex and acetylcholine in the dorsal hippocampus. Functional antagonism of central H(3) receptors was demonstrated by blockade of R-alpha-methylhistamine-induced dipsogenia in rats (ID(50) = 0.03 mg/kg p.o.). GSK189254 significantly improved performance of rats in diverse cognition paradigms, including passive avoidance (1 and 3 mg/kg p.o.), water maze (1 and 3 mg/kg p.o.), object recognition (0.3 and 1 mg/kg p.o.), and attentional set shift (1 mg/kg p.o.). These data suggest that GSK189254 may have therapeutic potential for the symptomatic treatment of dementia in Alzheimer's disease and other cognitive disorders.


Asunto(s)
Benzazepinas/farmacología , Encéfalo/efectos de los fármacos , Antagonistas de los Receptores Histamínicos/farmacología , Niacinamida/análogos & derivados , Nootrópicos/farmacología , Receptores Histamínicos H3/metabolismo , Adolescente , Adulto , Anciano , Anciano de 80 o más Años , Enfermedad de Alzheimer/metabolismo , Enfermedad de Alzheimer/patología , Enfermedad de Alzheimer/fisiopatología , Animales , Benzazepinas/metabolismo , Benzazepinas/farmacocinética , Unión Competitiva , Encéfalo/metabolismo , Encéfalo/patología , Línea Celular , Perros , Agonistas de los Receptores Histamínicos/metabolismo , Agonistas de los Receptores Histamínicos/farmacocinética , Agonistas de los Receptores Histamínicos/farmacología , Antagonistas de los Receptores Histamínicos/metabolismo , Antagonistas de los Receptores Histamínicos/farmacocinética , Humanos , Masculino , Aprendizaje por Laberinto/efectos de los fármacos , Ratones , Persona de Mediana Edad , Neurotransmisores/metabolismo , Niacinamida/metabolismo , Niacinamida/farmacocinética , Niacinamida/farmacología , Nootrópicos/metabolismo , Nootrópicos/farmacocinética , Ratas , Ratas Sprague-Dawley , Ratas Wistar , Receptores Histamínicos H3/análisis , Sus scrofa
12.
Brain Res ; 1149: 50-7, 2007 May 29.
Artículo en Inglés | MEDLINE | ID: mdl-17382304

RESUMEN

SB-277011-A is a dopamine D(3) receptor antagonist that exhibits over 100-fold selectivity over dopamine D(2) receptors and a broad spectrum of other receptor, ion channels, and enzymes. We employed c-Fos immunohistochemistry to characterise the functional neuroanatomical effects of acute administration of SB-277011-A and observed a time-dependent increase in the density of c-Fos-like positive nuclei in rat forebrain with maximal effects observed 2 h post-dose. The relative influence of the different brain regions on the overall effect of SB-277011-A was ranked by partial least squares discriminant analysis loadings plot which indicated that sites within the nucleus accumbens exerted the greatest influence on the separation of the vehicle and SB-277011-A treatment groups. At the 2 h time-point, c-Fos-like expression was shown to be significantly elevated (p<0.05) in the core and shell of the nucleus accumbens, at both rostral and caudal levels, and in the lateral septum. No significant changes were detected in the caudate nucleus (lateral or medial) or in the cingulate, infralimbic prefrontal, or somatosensory cortices. The capacity of SB-277011-A to trigger immediate early gene expression in these limbic regions of rat brain adds to a growing consensus of the potential utility of dopamine D(3) receptor antagonism in psychiatric disorders including schizophrenia and drug dependency.


Asunto(s)
Nitrilos/farmacología , Prosencéfalo/efectos de los fármacos , Proteínas Proto-Oncogénicas c-fos/efectos de los fármacos , Receptores de Dopamina D3/antagonistas & inhibidores , Tetrahidroisoquinolinas/farmacología , Animales , Inmunohistoquímica , Masculino , Prosencéfalo/metabolismo , Proteínas Proto-Oncogénicas c-fos/biosíntesis , Ratas , Ratas Sprague-Dawley , Factores de Tiempo
13.
Eur J Pharmacol ; 493(1-3): 95-8, 2004 Jun 16.
Artículo en Inglés | MEDLINE | ID: mdl-15189768

RESUMEN

The vasoactive peptide urotensin-II and its receptor, GPR14 (now known as UT receptor), are localised in the mammalian central nervous system. Accordingly, various centrally mediated effects of urotensin-II on behaviour, neuroendocrine hormones and neurochemistry have been described. To investigate neuroanatomical substrates for the central actions of urotensin-II, expression of the immediate early gene c-fos was examined following intracerebroventricular administration to rats. Urotensin-II increased Fos expression in the cingulate cortex and periaqueductal grey, suggesting important central roles for urotensin-II and its receptor.


Asunto(s)
Ligandos , Neuropéptidos/farmacología , Proteínas Proto-Oncogénicas c-fos/genética , Proteínas Proto-Oncogénicas c-fos/metabolismo , Receptores Acoplados a Proteínas G/efectos de los fármacos , Urotensinas/farmacología , Animales , Relación Dosis-Respuesta a Droga , Expresión Génica/efectos de los fármacos , Expresión Génica/genética , Giro del Cíngulo/efectos de los fármacos , Giro del Cíngulo/fisiología , Inyecciones Intraventriculares , Masculino , Núcleos Talámicos de la Línea Media/efectos de los fármacos , Núcleos Talámicos de la Línea Media/fisiología , Neuropéptidos/química , Sustancia Gris Periacueductal/efectos de los fármacos , Sustancia Gris Periacueductal/fisiología , Proteínas Proto-Oncogénicas c-fos/inmunología , Ratas , Ratas Sprague-Dawley , Receptores Acoplados a Proteínas G/fisiología , Reino Unido , Urotensinas/química
14.
Psychopharmacology (Berl) ; 177(1-2): 1-14, 2004 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-15205870

RESUMEN

RATIONALE: Neuromedin-U (NmU) is an agonist at NMU1R and NMU2R. The brain distribution of NmU and its receptors, in particular NMU2R, suggests widespread central roles for NmU. In agreement, centrally administered NmU affects feeding behaviour, energy expenditure and pituitary output. Further central nervous system (CNS) roles for NmU warrant investigation. OBJECTIVES: To investigate the CNS role of NmU by mapping NMU1R and NMU2R mRNA and measuring the behavioural, endocrine, neurochemical and c-fos response to intracerebroventricular (i.c.v.) NmU. METHODS: Binding affinity and functional potency of rat NmU was determined at human NMU1R and NMU2R. Expression of NMU1R and NMU2R mRNA in rat and human tissue was determined using semi-quantitative reverse-transcription polymerase chain reaction. In in-vivo studies, NmU was administered i.c.v. to male Sprague-Dawley rats, and changes in grooming, motor activity and pre-pulse inhibition (PPI) were assessed. In further studies, plasma endocrine hormones, [DOPAC + HVA]/[dopamine] and [5-HIAA]/[5-HT] ratios and levels of Fos-like immunoreactivity (FLI) were measured 20 min post-NmU (i.c.v.). RESULTS: NmU bound to NMU1R ( K(I), 0.11+/-0.02 nM) and NMU2R ( K(I), 0.21+/-0.05 nM) with equal affinity and was equally active at NMU1R (EC(50), 1.25+/-0.05 nM) and NMU2R (EC(50), 1.10+/-0.20 nM) in a functional assay. NMU2R mRNA expression was found at the highest levels in the CNS regions of both rat and human tissues. NMU1R mRNA expression was restricted to the periphery of both species with the exception of the rat amygdala. NmU caused a marked increase in grooming and motor activity but did not affect PPI. Further, NmU decreased plasma prolactin but did not affect levels of corticosterone, luteinising hormone or thyroid stimulating hormone. NmU elevated levels of 5-HT in the frontal cortex and hypothalamus, with decreased levels of its metabolites in the hippocampus and hypothalamus, but did not affect dopamine function. NmU markedly increased FLI in the nucleus accumbens, frontal cortex and central amygdala. CONCLUSIONS: These data provide further evidence for widespread roles for NmU and its receptors in the brain.


Asunto(s)
Sistema Nervioso Central/efectos de los fármacos , Sistema Nervioso Central/metabolismo , Proteínas de la Membrana/agonistas , Proteínas de la Membrana/metabolismo , Neuropéptidos/administración & dosificación , Receptores de Neurotransmisores/agonistas , Receptores de Neurotransmisores/metabolismo , Animales , Línea Celular , Relación Dosis-Respuesta a Droga , Humanos , Inyecciones Intraventriculares , Ratas , Ratas Sprague-Dawley , Porcinos
15.
Eur J Neurosci ; 18(6): 1706-10, 2003 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-14511349

RESUMEN

Brief N-methyl-D-aspartate (NMDA) receptor blockade in neonatal rats has been reported to increase neuronal apoptosis. We replicated this finding using MK-801 (0.5 mg/kg) administered twice on postnatal day 7, and then studied the long-term consequences. In adulthood, treated rats showed reduced volume and neuronal number within the hippocampus, and altered hippocampal NMDA receptor (NR1 subunit) expression. Synaptophysin mRNA was decreased in the thalamus (laterodorsal nucleus). Adult MK-801-treated females had prepulse inhibition deficits and increased locomotor activity. The data show that a transient and limited glutamatergic intervention during development can have chronic behavioural, structural and molecular effects. The effects are reminiscent of alterations reported in schizophrenia and, as such, are consistent with hypotheses advocating a role for NMDA receptor hypofunction, and aberrant apoptosis, in the neurodevelopmental pathogenesis of the disorder.


Asunto(s)
Conducta Animal/efectos de los fármacos , Maleato de Dizocilpina/farmacología , Antagonistas de Aminoácidos Excitadores/farmacología , Receptores de N-Metil-D-Aspartato/antagonistas & inhibidores , Tiempo , Animales , Animales Recién Nacidos , Apoptosis/efectos de los fármacos , Recuento de Células , Femenino , Regulación del Desarrollo de la Expresión Génica/efectos de los fármacos , Hipocampo/anatomía & histología , Hipocampo/crecimiento & desarrollo , Hipocampo/metabolismo , Inmunohistoquímica , Hibridación in Situ , Etiquetado Corte-Fin in Situ , Inhibición Psicológica , Masculino , Actividad Motora/efectos de los fármacos , Neuronas/efectos de los fármacos , Ratas , Ratas Sprague-Dawley , Receptores de N-Metil-D-Aspartato/genética , Factores Sexuales , Sinaptofisina/genética , Tálamo/crecimiento & desarrollo , Tálamo/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...