Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
Más filtros













Base de datos
Intervalo de año de publicación
1.
Stroke ; 53(4): 1386-1395, 2022 04.
Artículo en Inglés | MEDLINE | ID: mdl-35240860

RESUMEN

BACKGROUND: Cortical spreading depolarization (CSD) is a massive neuro-glial depolarization wave, which propagates across the cerebral cortex. In stroke, CSD is a necessary and ubiquitous mechanism for the development of neuronal lesions that initiates in the ischemic core and propagates through the penumbra extending the tissue injury. Although CSD propagation induces dramatic changes in cerebral blood flow, the vascular responses in different ischemic regions and their consequences on reperfusion and recovery remain to be defined. METHODS: Ischemia was performed using the thrombin model of stroke and reperfusion was induced by r-tPA (recombinant tissue-type plasminogen activator) administration in mice. We used in vivo electrophysiology and laser speckle contrast imaging simultaneously to assess both electrophysiological and hemodynamic characteristics of CSD after ischemia onset. Neurological deficits were assessed on day 1, 3, and 7. Furthermore, infarct sizes were quantified using 2,3,5-triphenyltetrazolium chloride on day 7. RESULTS: After ischemia, CSDs were evidenced by the characteristic propagating DC shift extending far beyond the ischemic area. On the vascular level, we observed 2 types of responses: some mice showed spreading hyperemia confined to the penumbra area (penumbral spreading hyperemia) while other showed spreading hyperemia propagating in the full hemisphere (full hemisphere spreading hyperemia). Penumbral spreading hyperemia was associated with severe stroke-induced damage, while full hemisphere spreading hyperemia indicated beneficial infarct outcome and potential viability of the infarct core. In all animals, thrombolysis with r-tPA modified the shape of the vascular response to CSD and reduced lesion volume. CONCLUSIONS: Our results show that different types of spreading hyperemia occur spontaneously after the onset of ischemia. Depending on their shape and distribution, they predict severity of injury and outcome. Furthermore, our data show that modulating the hemodynamic response to CSD may be a promising therapeutic strategy to attenuate stroke outcome.


Asunto(s)
Depresión de Propagación Cortical , Hiperemia , Accidente Cerebrovascular , Animales , Circulación Cerebrovascular , Depresión de Propagación Cortical/fisiología , Humanos , Infarto , Ratones , Accidente Cerebrovascular/diagnóstico por imagen , Accidente Cerebrovascular/tratamiento farmacológico
2.
J Neurosci ; 41(33): 7148-7159, 2021 08 18.
Artículo en Inglés | MEDLINE | ID: mdl-34210784

RESUMEN

Following stroke, the survival of neurons and their ability to reestablish connections is critical to functional recovery. This is strongly influenced by the balance between neuronal excitation and inhibition. In the acute phase of experimental stroke, lethal hyperexcitability can be attenuated by positive allosteric modulation of GABAA receptors (GABAARs). Conversely, in the late phase, negative allosteric modulation of GABAAR can correct the suboptimal excitability and improves both sensory and motor recovery. Here, we hypothesized that octadecaneuropeptide (ODN), an endogenous allosteric modulator of the GABAAR synthesized by astrocytes, influences the outcome of ischemic brain tissue and subsequent functional recovery. We show that ODN boosts the excitability of cortical neurons, which makes it deleterious in the acute phase of stroke. However, if delivered after day 3, ODN is safe and improves motor recovery over the following month in two different paradigms of experimental stroke in mice. Furthermore, we bring evidence that, during the subacute period after stroke, the repairing cortex can be treated with ODN by means of a single hydrogel deposit into the stroke cavity.SIGNIFICANCE STATEMENT Stroke remains a devastating clinical challenge because there is no efficient therapy to either minimize neuronal death with neuroprotective drugs or to enhance spontaneous recovery with neurorepair drugs. Around the brain damage, the peri-infarct cortex can be viewed as a reservoir of plasticity. However, the potential of wiring new circuits in these areas is restrained by a chronic excess of GABAergic inhibition. Here we show that an astrocyte-derived peptide, can be used as a delayed treatment, to safely correct cortical excitability and facilitate sensorimotor recovery after stroke.


Asunto(s)
Inhibidor de la Unión a Diazepam/uso terapéutico , Agonistas de Receptores de GABA-A/uso terapéutico , Neuronas/efectos de los fármacos , Neuropéptidos/uso terapéutico , Fragmentos de Péptidos/uso terapéutico , Receptores de GABA-A/efectos de los fármacos , Accidente Cerebrovascular/tratamiento farmacológico , Adulto , Animales , Astrocitos/metabolismo , Depresión de Propagación Cortical/fisiología , Inhibidor de la Unión a Diazepam/deficiencia , Inhibidor de la Unión a Diazepam/fisiología , Implantes de Medicamentos , Potenciales Evocados Somatosensoriales , Femenino , Agonistas de Receptores de GABA-A/farmacología , Humanos , Hidrogeles , Infarto de la Arteria Cerebral Media/tratamiento farmacológico , Trombosis Intracraneal/tratamiento farmacológico , Trombosis Intracraneal/etiología , Luz , Ratones , Ratones Endogámicos C57BL , N-Metilaspartato/toxicidad , Neuronas/fisiología , Neuropéptidos/deficiencia , Neuropéptidos/fisiología , Técnicas de Placa-Clamp , Fragmentos de Péptidos/deficiencia , Fragmentos de Péptidos/fisiología , Ratas , Rosa Bengala/efectos de la radiación , Rosa Bengala/toxicidad , Método Simple Ciego , Accidente Cerebrovascular/etiología
3.
Molecules ; 26(14)2021 Jul 16.
Artículo en Inglés | MEDLINE | ID: mdl-34299587

RESUMEN

26RFa is a neuropeptide that activates the rhodopsin-like G protein-coupled receptor QRFPR/GPR103. This peptidergic system is involved in the regulation of a wide array of physiological processes including feeding behavior and glucose homeostasis. Herein, the pharmacological profile of a homogenous library of QRFPR-targeting peptide derivatives was investigated in vitro on human QRFPR-transfected cells with the aim to provide possible insights into the structural determinants of the Phe residues to govern receptor activation. Our work advocates to include in next generations of 26RFa(20-26)-based QRFPR agonists effective substitutions for each Phe unit, i.e., replacement of the Phe22 residue by a constrained 1,2,3,4-tetrahydroisoquinoline-3-carboxylic acid moiety, and substitution of both Phe24 and Phe26 by their para-chloro counterpart. Taken as a whole, this study emphasizes that optimized modifications in the C-terminal part of 26RFa are mandatory to design selective and potent peptide agonists for human QRFPR.


Asunto(s)
Sustitución de Aminoácidos , Neuropéptidos , Receptores Acoplados a Proteínas G/agonistas , Animales , Células CHO , Cricetulus , Humanos , Neuropéptidos/química , Neuropéptidos/genética , Neuropéptidos/farmacología , Fenilalanina/química , Fenilalanina/genética , Receptores Acoplados a Proteínas G/metabolismo , Relación Estructura-Actividad
4.
Exp Neurol ; 341: 113696, 2021 07.
Artículo en Inglés | MEDLINE | ID: mdl-33727098

RESUMEN

Days and weeks after an ischemic stroke, the peri-infarct area adjacent to the necrotic tissue exhibits very intense synaptic reorganization aimed at regaining lost functions. In order to enhance functional recovery, it is important to understand the mechanisms supporting neural repair and neuroplasticity in the cortex surrounding the lesion. Brain oscillations of the local field potential (LFP) are rhythmic fluctuations of neuronal excitability that synchronize neuronal activity to organize information processing and plasticity. Although the oscillatory activity of the brain has been probed after stroke in both animals and humans using electroencephalography (EEG), the latter is ineffective to precisely map the oscillatory changes in the peri-infarct zone where synaptic plasticity potential is high. Here, we worked on the hypothesis that the brain oscillatory system is altered in the surviving peri-infarct cortex, which may slow down the functional repair and reduce the recovery. In order to document the relevance of this hypothesis, oscillatory power was measured at various distances from the necrotic core at 7 and 21 days after a permanent cortical ischemia induced in mice. Delta and theta oscillations remained at a normal power in the peri-infarct cortex, in contrast to low gamma oscillations that displayed a gradual decrease, when approaching the border of the lesion. A broadband increase of power was also observed in the homotopic contralateral sites. Thus, the proximal peri-infarct cortex could become a target of therapeutic interventions applied to correct the oscillatory regimen in order to boost post-stroke functional recovery.


Asunto(s)
Isquemia Encefálica/fisiopatología , Infarto Cerebral/fisiopatología , Ritmo Gamma/fisiología , Neocórtex/fisiopatología , Accidente Cerebrovascular/fisiopatología , Animales , Isquemia Encefálica/patología , Infarto Cerebral/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Neocórtex/patología , Accidente Cerebrovascular/patología
5.
Front Endocrinol (Lausanne) ; 11: 566026, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33250858

RESUMEN

Octadecaneuropeptide (ODN) and its precursor diazepam-binding inhibitor (DBI) are peptides belonging to the family of endozepines. Endozepines are exclusively produced by astroglial cells in the central nervous system of mammals, and their release is regulated by stress signals and neuroactive compounds. There is now compelling evidence that the gliopeptide ODN protects cultured neurons and astrocytes from apoptotic cell death induced by various neurotoxic agents. In vivo, ODN causes a very strong neuroprotective action against neuronal degeneration in a mouse model of Parkinson's disease. The neuroprotective activity of ODN is based on its capacity to reduce inflammation, apoptosis, and oxidative stress. The protective effects of ODN are mediated through its metabotropic receptor. This receptor activates a transduction cascade of second messengers to stimulate protein kinase A (PKA), protein kinase C (PKC), and mitogen-activated protein kinase (MAPK)-extracellular signal-regulated kinase (ERK) signaling pathways, which in turn inhibits the expression of proapoptotic factor Bax and the mitochondrial apoptotic pathway. In N2a cells, ODN also promotes survival and stimulates neurite outgrowth. During the ODN-induced neuronal differentiation process, numerous mitochondria and peroxisomes are identified in the neurites and an increase in the amount of cholesterol and fatty acids is observed. The antiapoptotic and neurotrophic properties of ODN, including its antioxidant, antiapoptotic, and pro-differentiating effects, suggest that this gliopeptide and some of its selective and stable derivatives may have therapeutic value for the treatment of some neurodegenerative diseases.


Asunto(s)
Citoprotección/efectos de los fármacos , Inhibidor de la Unión a Diazepam/administración & dosificación , Modelos Animales de Enfermedad , Enfermedades Neurodegenerativas/prevención & control , Neuropéptidos/administración & dosificación , Neuroprotección/efectos de los fármacos , Fármacos Neuroprotectores/administración & dosificación , Fragmentos de Péptidos/administración & dosificación , Animales , Citoprotección/fisiología , Humanos , Ratones , Factores de Crecimiento Nervioso/administración & dosificación , Enfermedades Neurodegenerativas/metabolismo , Enfermedades Neurodegenerativas/patología , Neuroprotección/fisiología , Estrés Oxidativo/efectos de los fármacos , Estrés Oxidativo/fisiología
6.
Pharmacol Ther ; 208: 107386, 2020 04.
Artículo en Inglés | MEDLINE | ID: mdl-31283949

RESUMEN

The existence of specific binding sites for benzodiazepines (BZs) in the brain has prompted the search for endogenous BZ receptor ligands designated by the generic term « endozepines ¼. This has led to the identification of an 86-amino acid polypeptide capable of displacing [3H]diazepam binding to brain membranes, thus called diazepam-binding inhibitor (DBI). It was subsequently found that the sequence of DBI is identical to that of a lipid carrier protein termed acyl-CoA-binding protein (ACBP). The primary structure of DBI/ACBP has been well preserved, suggesting that endozepines exert vital functions. The DBI/ACBP gene is expressed by astroglial cells in the central nervous system, and by various cell types in peripheral organs. Endoproteolytic cleavage of DBI/ACBP generates several bioactive peptides including a triakontatetraneuropeptide that acts as a selective ligand of peripheral BZ receptors/translocator protein, and an octadecaneuropeptide that activates a G protein-coupled receptor and behaves as an allosteric modulator of the GABAAR. Although DBI/ACBP is devoid of a signal peptide, endozepines are released by astrocytes in a regulated manner. Consistent with the diversity and wide distribution of BZ-binding sites, endozepines appear to exert a large array of biological functions and pharmacological effects. Thus, intracerebroventricular administration of DBI or derived peptides induces proconflict and anxiety-like behaviors, and reduces food intake. Reciprocally, the expression of DBI/ACBP mRNA is regulated by stress and metabolic signals. In vitro, endozepines stimulate astrocyte proliferation and protect neurons and astrocytes from apoptotic cell death. Endozepines also regulate neurosteroid biosynthesis and neuropeptide expression, and promote neurogenesis. In peripheral organs, endozepines activate steroid hormone production, stimulate acyl chain ceramide synthesis and trigger pro-inflammatory cytokine secretion. The expression of the DBI/ACBP gene is enhanced in addiction/withdrawal animal models, in patients with neurodegenerative disorders and in various types of tumors. We review herein the current knowledge concerning the various actions of endozepines and discuss the physiopathological implications of these regulatory gliopeptides.


Asunto(s)
Benzodiazepinas/metabolismo , Receptores de GABA-A/metabolismo , Animales , Inhibidor de la Unión a Diazepam/metabolismo , Humanos
7.
J Med Chem ; 61(22): 10185-10197, 2018 11 21.
Artículo en Inglés | MEDLINE | ID: mdl-30358997

RESUMEN

26RFa, the endogenous QRFPR ligand, is implicated in several physiological and pathological conditions such as the regulation of glucose homeostasis and bone mineralization; hence, QRFPR ligands display therapeutic potential. At the molecular level, functional interaction occurs between residues Arg25 of 26RFa and Gln125 of QRFPR. We have designed 26RFa(20-26) analogues incorporating arginine derivatives modified by alkylated substituents. We found that the Arg25 side chain length was necessary to retain the activity of 26RFa(20-26) and that N-monoalkylation of arginine was accommodated by the QRFPR active site. In particular, [(Me)ωArg25]26RFa(20-26) (5b, LV-2186) appeared to be 25-fold more potent than 26RFa(20-26) and displayed a position in a QRFPR homology model slightly different to that of the unmodified heptapeptide. Other peptides were less potent than 26RFa(20-26), exhibited partial agonistic activity, or were totally inactive in accordance to different ligand-bound structures. In vivo, [(Me)ωArg25]26RFa(20-26) exerted a delayed 26RFa-like hypoglycemic effect. Finally, N-methyl substituted arginine-containing peptides represent lead compounds for further development of QRFPR agonists.


Asunto(s)
Arginina/química , Diseño de Fármacos , Guanidina/química , Simulación de Dinámica Molecular , Oligopéptidos/química , Oligopéptidos/síntesis química , Alquilación , Amidas/química , Animales , Células CHO , Técnicas de Química Sintética , Cricetulus , Conformación Proteica
8.
Cell Mol Life Sci ; 75(11): 2075-2091, 2018 06.
Artículo en Inglés | MEDLINE | ID: mdl-29264673

RESUMEN

Parkinson's disease (PD) is a neurodegenerative disorder characterized by a progressive loss of dopamine (DA) neurons through apoptotic, inflammatory and oxidative stress mechanisms. The octadecaneuropeptide (ODN) is a diazepam-binding inhibitor (DBI)-derived peptide, expressed by astrocytes, which protects neurons against oxidative cell damages and apoptosis in an in vitro model of PD. The present study reveals that a single intracerebroventricular injection of 10 ng ODN 1 h after the last administration of 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) prevented the degeneration of DA neurons induced by the toxin in the substantia nigra pars compacta of mice, 7 days after treatment. ODN-mediated neuroprotection was associated with a reduction of the number of glial fibrillary acidic protein-positive reactive astrocytes and a strong inhibition of the expression of pro-inflammatory genes such as interleukins 1ß and 6, and tumor necrosis factor-α. Moreover, ODN blocked the inhibition of the anti-apoptotic gene Bcl-2, and the stimulation of the pro-apoptotic genes Bax and caspase-3, induced by MPTP in the substantia nigra pars compacta. ODN also decreased or even in some cases abolished MPTP-induced oxidative damages, overproduction of reactive oxygen species and accumulation of lipid oxidation products in DA neurons. Furthermore, DBI knockout mice appeared to be more vulnerable than wild-type animals to MPTP neurotoxicity. Taken together, these results show that the gliopeptide ODN exerts a potent neuroprotective effect against MPTP-induced degeneration of nigrostriatal DA neurons in mice, through mechanisms involving downregulation of neuroinflammatory, oxidative and apoptotic processes. ODN may, thus, reduce neuronal damages in PD and other cerebral injuries involving oxidative neurodegeneration.


Asunto(s)
Inhibidor de la Unión a Diazepam/uso terapéutico , Neuropéptidos/uso terapéutico , Fármacos Neuroprotectores/uso terapéutico , Enfermedad de Parkinson/tratamiento farmacológico , Fragmentos de Péptidos/uso terapéutico , Animales , Modelos Animales de Enfermedad , Neuronas Dopaminérgicas/efectos de los fármacos , Neuronas Dopaminérgicas/metabolismo , Neuronas Dopaminérgicas/patología , Regulación de la Expresión Génica/efectos de los fármacos , Humanos , Masculino , Ratones Endogámicos C57BL , Estrés Oxidativo/efectos de los fármacos , Enfermedad de Parkinson/genética , Enfermedad de Parkinson/metabolismo , Enfermedad de Parkinson/patología , Especies Reactivas de Oxígeno/metabolismo
9.
Nanomedicine (Lond) ; 12(8): 879-892, 2017 04.
Artículo en Inglés | MEDLINE | ID: mdl-28339310

RESUMEN

AIM: Develop modified dextran nanoparticles showing potential to assist with drug permeation across the blood-brain barrier for the delivery of neuropeptides. METHODS: Nanoparticles loaded by emulsification with model macromolecular actives were characterized in terms of stability, cytotoxicity and drug-release behavior. Peptide-loaded nanoformulations were tested in an in vivo trout model and in food-deprived mice. RESULTS: Nanoformulations loaded with model peptides showed good stability and appeared nontoxic in low concentration against human brain endothelial cells. They were found to preserve the bioactivity of loaded peptides (angiotensin II) as demonstrated in vivo using a trout model, and to induce a transient reduction of food consumption in mice when loaded with an anorexigenic octaneuropeptide. CONCLUSION: Octylglyceryl dextran-graft-poly(lactic acid) nanoparticles formulated by emulsification demonstrate potential for peptide delivery.


Asunto(s)
Barrera Hematoencefálica/efectos de los fármacos , Encéfalo/efectos de los fármacos , Sistemas de Liberación de Medicamentos , Nanopartículas/administración & dosificación , Péptidos/administración & dosificación , Animales , Encéfalo/diagnóstico por imagen , Línea Celular , Dextranos/química , Humanos , Ratones , Nanopartículas/química , Tamaño de la Partícula , Péptidos/química , Poliésteres/química
10.
J Med Chem ; 55(17): 7516-24, 2012 Sep 13.
Artículo en Inglés | MEDLINE | ID: mdl-22800498

RESUMEN

26RFa, a novel RFamide neuropeptide, is the endogenous ligand of the former orphan receptor GPR103. Intracerebroventricular injection of 26RFa and its C-terminal heptapeptide, 26RFa((20-26)), stimulates food intake in rodents. To develop potent, stable ligands of GPR103 with low molecular weight, we have designed a series of aza-ß(3)-containing 26RFa((20-26)) analogues for their propensity to establish intramolecular hydrogen bonds, and we have evaluated their ability to increase [Ca(2+)](i) in GPR103-transfected cells. We have identified a compound, [Cmpi(21),aza-ß(3)-Hht(23)]26RFa((21-26)), which was 8-fold more potent than 26RFa((20-26)) in mobilizing [Ca(2+)](i). This pseudopeptide was more stable in serum than 26RFa((20-26)) and exerted a longer lasting orexigenic effect in mice. This study constitutes an important step toward the development of 26RFa analogues that could prove useful for the treatment of feeding disorders.


Asunto(s)
Compuestos Aza/farmacología , Diseño de Fármacos , Conducta Alimentaria/efectos de los fármacos , Péptidos/farmacología , Receptores Acoplados a Proteínas G/agonistas , Animales , Dicroismo Circular , Estabilidad de Medicamentos , Enlace de Hidrógeno , Inyecciones Intraventriculares , Masculino , Ratones , Peso Molecular , Péptidos/administración & dosificación
11.
Nat Neurosci ; 14(10): 1276-84, 2011 Sep 11.
Artículo en Inglés | MEDLINE | ID: mdl-21909085

RESUMEN

Astrocytes communicate with synapses by means of intracellular calcium ([Ca(2+)](i)) elevations, but local calcium dynamics in astrocytic processes have never been thoroughly investigated. By taking advantage of high-resolution two-photon microscopy, we identify the characteristics of local astrocyte calcium activity in the adult mouse hippocampus. Astrocytic processes showed intense activity, triggered by physiological transmission at neighboring synapses. They encoded synchronous synaptic events generated by sparse action potentials into robust regional (∼12 µm) [Ca(2+)](i) elevations. Unexpectedly, they also sensed spontaneous synaptic events, producing highly confined (∼4 µm), fast (millisecond-scale) miniature Ca(2+) responses. This Ca(2+) activity in astrocytic processes is generated through GTP- and inositol-1,4,5-trisphosphate-dependent signaling and is relevant for basal synaptic function. Thus, buffering astrocyte [Ca(2+)](i) or blocking a receptor mediating local astrocyte Ca(2+) signals decreased synaptic transmission reliability in minimal stimulation experiments. These data provide direct evidence that astrocytes are integrated in local synaptic functioning in adult brain.


Asunto(s)
Astrocitos/fisiología , Señalización del Calcio/fisiología , Calcio/metabolismo , Sinapsis/metabolismo , Potenciales de Acción/efectos de los fármacos , Potenciales de Acción/fisiología , Adenosina Difosfato/análogos & derivados , Adenosina Difosfato/farmacología , Animales , Animales Recién Nacidos , Astrocitos/efectos de los fármacos , Astrocitos/ultraestructura , Biofisica , Señalización del Calcio/efectos de los fármacos , Comunicación Celular/efectos de los fármacos , Comunicación Celular/fisiología , Quelantes/farmacología , Ácido Egtácico/análogos & derivados , Ácido Egtácico/farmacología , Estimulación Eléctrica/métodos , Inhibidores Enzimáticos/farmacología , Regulación de la Expresión Génica/efectos de los fármacos , Regulación de la Expresión Génica/genética , Heparina/farmacología , Hipocampo/citología , Técnicas In Vitro , Receptores de Inositol 1,4,5-Trifosfato/deficiencia , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Modelos Biológicos , Técnicas de Placa-Clamp , Antagonistas del Receptor Purinérgico P2Y/farmacología , Bloqueadores de los Canales de Sodio/farmacología , Sacarosa/farmacología , Sinapsis/efectos de los fármacos , Transmisión Sináptica/efectos de los fármacos , Transmisión Sináptica/fisiología , Tetrodotoxina/farmacología , Tapsigargina/farmacología
12.
J Neurosci ; 30(45): 15298-303, 2010 Nov 10.
Artículo en Inglés | MEDLINE | ID: mdl-21068334

RESUMEN

Glucose is the primary energetic substrate of the brain, and measurements of its metabolism are the basis of major functional cerebral imaging methods. Contrary to the general view that neurons are fueled solely by glucose in proportion to their energetic needs, recent in vitro and ex vivo analyses suggest that glucose preferentially feeds astrocytes. However, the cellular fate of glucose in the intact brain has not yet been directly observed. We have used a real-time method for measuring glucose uptake in astrocytes and neurons in vivo in male rats by imaging the trafficking of the nonmetabolizable glucose analog 6-deoxy-N-(7-nitrobenz-2-oxa-1,3-diazol-4-yl)-aminoglucose (6-NBDG) using two-photon microscopy. During resting conditions we found that astrocytes and neurons both take up 6-NBDG at the same rate in the barrel cortex of the rat. However, during intense neuronal activity triggered by whisker stimulation, astrocytes rapidly accelerated their uptake, whereas neuronal uptake remained almost unchanged. After the stimulation period, astrocytes returned to their preactivation rates of uptake paralleling the neuronal rate of uptake. These observations suggest that glucose is taken up primarily by astrocytes, supporting the view that functional imaging experiments based on glucose analogs extraction may predominantly reflect the metabolic activity of the astrocytic network.


Asunto(s)
Astrocitos/metabolismo , Glucosa/metabolismo , Neuronas/metabolismo , Corteza Somatosensorial/metabolismo , Análisis de Varianza , Animales , Transporte Biológico , Masculino , Microscopía de Fluorescencia por Excitación Multifotónica , Ratas , Ratas Sprague-Dawley , Vibrisas/fisiología
13.
Exp Neurol ; 210(2): 577-84, 2008 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-18191840

RESUMEN

Urotensin-II (U-II) is a cyclic peptide identified recently in many mammalian species including man. U-II and its receptor are expressed in the central nervous system, in the cardiovascular system and in other peripheral tissues. Although this peptide has been reported initially to be a potent vasoconstrictor, increasing evidence shows that its vascular actions strongly depend on species and vascular beds. Here we analyzed the effects of U-II administration on cerebral blood flow (CBF) under physiological conditions and following cerebral ischemia in rats. Although intravenous injection of U-II had minimal effects on CBF as measured by the technique of laser Doppler flowmetry, its administration (10 nmol) into the lateral cerebral ventricle induced gradual and long lasting increase in CBF (+61% at 1 h post-injection, p<0.05). These U-II-mediated CBF increases were not related to the transient systemic pressor actions of the peptide and were reduced by nitric oxide synthase inhibition (61 vs 17%, p<0.05). Intracerebroventricular administration of U-II following the induction of cerebral ischemia, failed to alter residual CBF in the affected cerebral hemisphere. Nonetheless, following reperfusion (90 min after ischemia), U-II-treated animals displayed a remarkable hyperperfusion compared to vehicle-treated rats (+168%, p<0.05). The volume of infarction was significantly increased in U-II-treated rats (+40%, p<0.05). These results provide the first evidence that U-II increases cerebral blood flow when administered into the cerebral ventricle and exacerbates brain damage following an ischemic insult.


Asunto(s)
Anestesia , Circulación Cerebrovascular/efectos de los fármacos , Isquemia/tratamiento farmacológico , Urotensinas/uso terapéutico , Análisis de Varianza , Animales , Presión Sanguínea/efectos de los fármacos , Infarto Encefálico/etiología , Infarto Encefálico/prevención & control , Modelos Animales de Enfermedad , Isquemia/fisiopatología , Flujometría por Láser-Doppler/métodos , Masculino , Ratas , Ratas Sprague-Dawley , Factores de Tiempo
14.
Stroke ; 38(11): 3007-15, 2007 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-17901379

RESUMEN

BACKGROUND AND PURPOSE: Whereas the effects of chronic arterial hypertension on the cerebral vasculature have been widely studied, its effects on brain tissue have been studied less so. Here we examined if spontaneously hypertensive rats (SHRs) or the normotensive control Wistar Kyoto rats (WKYs) made hypertensive by renal artery stenosis (R-WKYs) are vulnerable to an excitotoxic brain lesion provoked by an overactivation of glutamate receptors. METHODS: Lesion volumes were quantified by histology in WKYs and SHRs subjected to striatal administration of N-methyl-d-aspartate (NMDA) or alpha-amino-3-hydroxy-5-methyl-4-isoxazole propionic acid (AMPA). The expression of AMPA receptors subunits and calcium/calmodulin kinase-II alpha was analyzed by real-time polymerase chain reaction and Western blot. RESULTS: NMDA (50 and 75 nmol) induced similar lesions in both SHRs (10+/-2 mm(3) and 16+/-4 mm(3), respectively) and WKYs (11+/-2 mm(3) and 19+/-7 mm(3), respectively). However, AMPA-induced (2.5 and 5 nmol) lesions were significantly greater in 14-week-old SHRs (14+/-3 mm(3) and 20+/-5 mm(3), respectively) than WKYs (4+/-2 mm(3), P<0.05 and 7+/-4 mm(3), P<0.001, respectively). Furthermore, normotensive 7-week-old SHRs also displayed an aggravated AMPA-induced lesion compared with age-matched WKYs (10+/-3 mm(3) vs 6+/-3 mm(3); P<0.05). Neither NMDA nor AMPA produced increased lesion volumes in R-WKYs (12+/-3 mm(3) and 5+/-4 mm(3), respectively) compared with WKYs. Striatal levels of AMPA receptors subunits, GluR1 and GluR2, were not different between SHRs and WKYs. However, SHRs displayed an increase in phosphorylated form of GluR1 at Ser-831 (P<0.05), as well as in calcium/calmodulin kinase-II alpha (P<0.002). Selective inhibition of this kinase by KN-93 reduced AMPA-induced damage in SHRs (P<0.01 vs vehicle). CONCLUSIONS: These findings show that an increase in phosphorylated GluR1, which increases AMPA receptor conductance, may be involved in the vulnerability of SHRs to AMPA.


Asunto(s)
Daño Encefálico Crónico/etiología , Daño Encefálico Crónico/fisiopatología , Isquemia Encefálica/fisiopatología , Ácido Glutámico/metabolismo , Hipertensión/complicaciones , Receptores AMPA/metabolismo , Animales , Presión Sanguínea/efectos de los fármacos , Presión Sanguínea/fisiología , Daño Encefálico Crónico/genética , Isquemia Encefálica/genética , Isquemia Encefálica/metabolismo , Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/metabolismo , Muerte Celular/efectos de los fármacos , Muerte Celular/genética , Predisposición Genética a la Enfermedad/genética , Hipertensión/genética , Hipertensión/fisiopatología , Activación del Canal Iónico/efectos de los fármacos , Activación del Canal Iónico/fisiología , Masculino , Potenciales de la Membrana/efectos de los fármacos , Potenciales de la Membrana/fisiología , Degeneración Nerviosa/inducido químicamente , Degeneración Nerviosa/metabolismo , Degeneración Nerviosa/fisiopatología , Neurotoxinas/toxicidad , Fosforilación/efectos de los fármacos , Ratas , Ratas Endogámicas SHR , Ratas Endogámicas WKY , Receptores AMPA/agonistas , Flujo Sanguíneo Regional/efectos de los fármacos , Flujo Sanguíneo Regional/fisiología , Resistencia Vascular/efectos de los fármacos , Resistencia Vascular/fisiología , Ácido alfa-Amino-3-hidroxi-5-metil-4-isoxazol Propiónico/toxicidad
15.
J Neurosci ; 27(15): 4036-44, 2007 Apr 11.
Artículo en Inglés | MEDLINE | ID: mdl-17428981

RESUMEN

Spreading depression (SD) is a propagating wave of neuronal depolarization and ionic shifts, seen in stroke and migraine. In vitro, SD is associated with astrocytic [Ca2+] waves, but it is unclear what role they play and whether they influence cerebral blood flow, which is altered in SD. Here we show that SD in vivo is associated with [Ca2+] waves in astrocytes and neurons and with constriction of intracortical arterioles severe enough to result in arrest of capillary perfusion. The vasoconstriction is correlated with fast astrocytic [Ca2+] waves and is inhibited when they are reduced. [Ca2+] waves appear in neurons before astrocytes, and inhibition of astrocytic [Ca2+] waves does not depress SD propagation. This suggests that astrocytes do not drive SD propagation but are responsible for the hemodynamic failure seen deep in the cortex. Similar waves occur in anoxic depolarizations (AD), supporting the notion that SD and AD are related processes.


Asunto(s)
Corteza Cerebral/irrigación sanguínea , Corteza Cerebral/fisiología , Depresión de Propagación Cortical/fisiología , Neuronas/fisiología , Animales , Astrocitos/citología , Astrocitos/fisiología , Velocidad del Flujo Sanguíneo/fisiología , Corteza Cerebral/citología , Microscopía Confocal/métodos , Neuronas/citología , Ratas , Ratas Sprague-Dawley
16.
Circulation ; 111(17): 2241-9, 2005 May 03.
Artículo en Inglés | MEDLINE | ID: mdl-15851587

RESUMEN

BACKGROUND: Accumulating evidence demonstrates a critical involvement of tissue-type plasminogen activator (tPA) in pathological and physiological brain conditions. Determining whether and how vascular tPA can cross the blood-brain barrier (BBB) to enter the brain is thus important, not only during stroke but also in physiological conditions. METHODS AND RESULTS: In the present work, we provide evidence in vivo that intravenous injection of tPA increases NMDA-induced striatal lesion in the absence of BBB leakage. Accordingly, we show that tPA crosses the BBB both after excitotoxic lesion and in control conditions. Indeed, vascular injected tPA can be detected within the brain parenchyma and in the cerebrospinal fluid. By using an in vitro model of BBB, we have confirmed that tPA can cross the intact BBB. Its passage was blocked at 4 degrees C, was saturable, and was independent of its proteolytic activity. We have shown that tPA crosses the BBB by transcytosis, mediated by a member of the LDL receptor-related protein family. CONCLUSIONS: We demonstrate that blood-derived tPA can reach the brain parenchyma without alteration of the BBB. The molecular mechanism of the passage of tPA from blood to brain described here could represent an interesting target to improve thrombolysis in stroke.


Asunto(s)
Barrera Hematoencefálica/metabolismo , Proteína 1 Relacionada con Receptor de Lipoproteína de Baja Densidad/fisiología , Activador de Tejido Plasminógeno/efectos adversos , Activador de Tejido Plasminógeno/farmacocinética , Animales , Encéfalo/efectos de los fármacos , Encéfalo/metabolismo , Encéfalo/patología , Frío , Proteína 1 Relacionada con Receptor de Lipoproteína de Baja Densidad/metabolismo , Masculino , N-Metilaspartato/administración & dosificación , Síndromes de Neurotoxicidad/etiología , Inhibidor 1 de Activador Plasminogénico/administración & dosificación , Transporte de Proteínas , Ratas , Ratas Sprague-Dawley
17.
Stroke ; 33(12): 3019-25, 2002 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-12468806

RESUMEN

BACKGROUND AND PURPOSE: Endothelins act through 2 receptors, namely, ET(A) and ET(B). In the cerebral circulation, ET(A) mediates marked and prolonged vasoconstriction, and its blockade increases cerebral blood flow (CBF) and reduces ischemic brain damage. However, the role of ET(B) receptors remains unclear. In this study we examined, in rats, the kinetics of expression of ET(B) and the effects of ET(B) blockade on changes in CBF and brain damage after focal cerebral ischemia and N-methyl-D-aspartate (NMDA)-induced excitotoxic injury. METHODS: Rats were subjected to transient (60 minutes) focal cerebral ischemia or cortical injection of NMDA. The selective ET(B) antagonist BQ-788 was injected intracerebroventricularly 30 minutes before and 30 minutes after the onset of ischemia. Cortical perfusion was monitored by laser-Doppler flowmetry. The volume of infarction or NMDA-induced cortical lesion was assessed at 24 hours after the insult. The reverse transcription-polymerase chain reaction technique was used to assess ET(B) expression. RESULTS: Cerebral ischemia failed to alter the expression of ET(B) mRNA in both acute and chronic stages. Administration of BQ-788 resulted in an increase in infarction volume (178%; P<0.05) accompanied by a decrease in residual CBF (-26.7% versus control; P<0.01). In these animals we found a positive correlation between the volume of infarction and the severity of the decrease in CBF. NMDA-induced cortical lesions were not affected by the administration of BQ-788. CONCLUSIONS: Our results suggest that the ET(B) antagonist BQ-788 induces deleterious effects that are mediated by the reduction of residual blood flow after ischemia and argue that the optimal therapeutic strategy in stroke would be to target the use of selective ET(A) antagonists and not mixed ET(A)/ET(B) antagonists.


Asunto(s)
Antihipertensivos/efectos adversos , Isquemia Encefálica/patología , Antagonistas de los Receptores de Endotelina , Oligopéptidos/efectos adversos , Piperidinas/efectos adversos , Animales , Antihipertensivos/administración & dosificación , Velocidad del Flujo Sanguíneo/efectos de los fármacos , Isquemia Encefálica/inducido químicamente , Circulación Cerebrovascular/efectos de los fármacos , Modelos Animales de Enfermedad , Agonistas de Aminoácidos Excitadores/administración & dosificación , Agonistas de Aminoácidos Excitadores/toxicidad , Expresión Génica/efectos de los fármacos , Infarto de la Arteria Cerebral Media/patología , Inyecciones Intraventriculares , Masculino , N-Metilaspartato/administración & dosificación , N-Metilaspartato/toxicidad , Oligopéptidos/administración & dosificación , Piperidinas/administración & dosificación , ARN Mensajero/metabolismo , Ratas , Ratas Sprague-Dawley , Receptor de Endotelina B , Receptores de Endotelina/genética
18.
J Cereb Blood Flow Metab ; 22(10): 1165-9, 2002 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-12368653

RESUMEN

To correlate brain metabolic status with the molecular events during cerebral ischemia, a cDNA array was performed after positron emission tomography scanning in a model of focal cerebral ischemia in baboons. Cluster analysis for the expression of 74 genes allowed the identification of 4 groups of genes. In each of the distinct groups, the authors observed a marked inflection in the pattern of gene expression when the CMRo was reduced by 48% to 66%. These patterns of coordinated modifications in gene expression could define molecular checkpoints for the development of an ischemic infarct and a molecular definition of the penumbra.


Asunto(s)
Isquemia Encefálica/genética , Encéfalo/metabolismo , Regulación de la Expresión Génica , Animales , Encéfalo/irrigación sanguínea , Isquemia Encefálica/metabolismo , Arterias Cerebrales/metabolismo , Arterias Cerebrales/patología , Clonación Molecular , Análisis por Conglomerados , ADN Complementario , Modelos Animales de Enfermedad , Masculino , Análisis de Secuencia por Matrices de Oligonucleótidos , Consumo de Oxígeno , Papio
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA