Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 60
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Eur J Immunol ; 53(9): e2350370, 2023 09.
Artículo en Inglés | MEDLINE | ID: mdl-37366289

RESUMEN

Fingolimod has generally shown neuroprotective effects in stroke models. Here, we tested the hypothesis that fingolimod modulates T-cell cytokine production towards a regulatory phenotype. Second, we investigated how fingolimod altered the Treg suppressive function and the sensitivity of effector T cells to regulation. Mice that had underwent the permanent electrocoagulation of the left middle cerebral artery received saline or fingolimod (0.5 mg/kg) daily for 10-days post-ischaemia. Fingolimod improved neurobehavioural recovery compared to saline control and increased Treg frequency in the periphery and brain. Tregs from fingolimod-treated animals had a higher expression of CCR8. Fingolimod increased the frequencies of CD4+ IL-10+ , CD4+ IFN-γ+ and CD4+ IL-10+ IFN-γ+ cells in spleen and blood, and CD4+ IL-17+ cells in the spleen, with only minor effects on CD8+ T-cell cytokine production. Treg from post-ischaemic mice had reduced suppressive function compared to Treg from non-ischaemic mice. Fingolimod treatment rescued this function against saline-treated but not fingolimod-treated CD4+ effector T cells. In conclusion, fingolimod seems to improve the suppressive function of Treg post-stroke while also increasing the resistance of CD4+ effector cells to this suppression. Fingolimod's capacity to increase both effector and regulatory functions may explain the lack of consistent improvement in functional recovery in experimental brain ischaemia.


Asunto(s)
Isquemia Encefálica , Clorhidrato de Fingolimod , Ratones , Animales , Clorhidrato de Fingolimod/farmacología , Clorhidrato de Fingolimod/uso terapéutico , Linfocitos T Reguladores/metabolismo , Interleucina-10/metabolismo , Expresión Génica , Isquemia Encefálica/tratamiento farmacológico , Isquemia Encefálica/metabolismo
2.
Mol Pharm ; 21(7): 3310-3320, 2024 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-38856116

RESUMEN

Siponimod is a promising agent for the inhibition of ocular neovascularization in diabetic retinopathy and age-related macular degeneration. Siponimod's development for ophthalmological application is hindered by the limited information available on the drug's solubility, stability, ocular pharmacokinetics (PK), and toxicity in vivo. In this study, we investigated the aqueous stability of siponimod under stress conditions (up to 60 °C) and its degradation behavior in solution. Additionally, siponimod's ocular PK and toxicity were investigated using intravitreal injection of two different doses (either 1300 or 6500 ng) in an albino rabbit model. Siponimod concentration was quantified in the extracted vitreous, and the PK parameters were calculated. The drug half-life after administration of the low and high doses was 2.8 and 3.9 h, respectively. The data obtained in vivo was used to test the ability of published in silico models to predict siponimod's PK accurately. Two models that correlated siponimod's molecular descriptors with its elimination from the vitreous closely predicted the half-life. Furthermore, 24 h and 7 days after intravitreal injections, the retinas showed no signs of toxicity. This study provides important information necessary for the formulation and development of siponimod for ophthalmologic applications. The short half-life of siponimod necessitates the development of a sustained drug delivery system to maintain therapeutic concentrations over an extended period, while the lack of short-term ocular toxicity observed in the retinas of siponimod-treated rabbits supports possible clinical use.


Asunto(s)
Azetidinas , Inyecciones Intravítreas , Animales , Conejos , Azetidinas/farmacocinética , Azetidinas/administración & dosificación , Semivida , Cuerpo Vítreo/efectos de los fármacos , Cuerpo Vítreo/metabolismo , Masculino , Retina/efectos de los fármacos , Retina/metabolismo , Ojo/efectos de los fármacos , Ojo/metabolismo , Retinopatía Diabética/tratamiento farmacológico , Inhibidores de la Angiogénesis/farmacocinética , Inhibidores de la Angiogénesis/administración & dosificación , Inhibidores de la Angiogénesis/toxicidad , Solubilidad , Degeneración Macular/tratamiento farmacológico , Compuestos de Bencilo
3.
J Pharmacol Exp Ther ; 386(2): 224-241, 2023 08.
Artículo en Inglés | MEDLINE | ID: mdl-37188532

RESUMEN

Sphingosine-1-phosphate (S1P) receptors control endothelial cell proliferation, migration, and survival. Evidence of the ability of S1P receptor modulators to influence multiple endothelial cell functions suggests their potential use for antiangiogenic effect. The main purpose of our study was to investigate the potential of siponimod for the inhibition of ocular angiogenesis in vitro and in vivo. We investigated the effects of siponimod on the metabolic activity (thiazolyl blue tetrazolium bromide assay), cell toxicity (lactate dehydrogenase release), basal proliferation and growth factor-induced proliferation (bromodeoxyuridine assay), and migration (transwell migration assay) of human umbilical vein endothelial cells (HUVEC) and retinal microvascular endothelial cells (HRMEC). The effects of siponimod on HRMEC monolayer integrity, barrier function under basal conditions, and tumor necrosis factor alpha (TNF-α)-induced disruption were assessed using the transendothelial electrical resistance and fluorescein isothiocyanate-dextran permeability assays. Siponimod's effect on TNF-α-induced distribution of barrier proteins in HRMEC was investigated using immunofluorescence. Finally, the effect of siponimod on ocular neovascularization in vivo was assessed using suture-induced corneal neovascularization in albino rabbits. Our results show that siponimod did not affect endothelial cell proliferation or metabolic activity but significantly inhibited endothelial cell migration, increased HRMEC barrier integrity, and reduced TNF-α-induced barrier disruption. Siponimod also protected against TNF-α-induced disruption of claudin-5, zonula occludens-1, and vascular endothelial-cadherin in HRMEC. These actions are mainly mediated by sphingosine-1-phosphate receptor 1 modulation. Finally, siponimod prevented the progression of suture-induced corneal neovascularization in albino rabbits. In conclusion, the effects of siponimod on various processes known to be involved in angiogenesis support its therapeutic potential in disorders associated with ocular neovascularization. SIGNIFICANCE STATEMENT: Siponimod is an extensively characterized sphingosine-1-phosphate receptor modulator already approved for the treatment of multiple sclerosis. It inhibited retinal endothelial cell migration, potentiated endothelial barrier function, protected against tumor necrosis factor alpha-induced barrier disruption, and also inhibited suture-induced corneal neovascularization in rabbits. These results support its use for a novel therapeutic indication in the management of ocular neovascular diseases.


Asunto(s)
Neovascularización de la Córnea , Factor de Necrosis Tumoral alfa , Animales , Humanos , Conejos , Retina , Neovascularización Patológica , Células Endoteliales de la Vena Umbilical Humana , Células Cultivadas
4.
J Headache Pain ; 24(1): 62, 2023 May 26.
Artículo en Inglés | MEDLINE | ID: mdl-37237336

RESUMEN

BACKGROUND AND AIMS: Cortical spreading depression (CSD), a transient neuronal and glial depolarization that propagates slowly across the cerebral cortex, is the putative electrophysiological event underlying migraine aura and a headache trigger. Migraine is three times more prevalent in women than men, linked to circulating female hormones. High estrogen levels or estrogen withdrawal may be a migraine trigger for many women. We, therefore, aimed to examine whether sex, gonadectomy, and female hormone supplementation and withdrawal affect the susceptibility to CSD. METHODS: To determine CSD susceptibility, we recorded the frequency of CSDs triggered during 2-h topical KCl application in intact or gonadectomized female and male rats, without or with estradiol or progesterone supplementation via daily intraperitoneal injections. Estrogen or progesterone treatment followed by withdrawal was studied in a separate cohort. To take the first step towards identifying potential mechanisms, we studied glutamate and GABAA receptor binding using autoradiography. RESULTS: The CSD frequency in intact female rats was higher than intact male and ovariectomized rats. We did not detect a change in CSD frequency during different stages of the estrous cycle in intact females. Daily estrogen injections for three weeks did not change CSD frequency. However, one-week estrogen withdrawal after two weeks of treatment significantly increased CSD frequency compared with the vehicle group in gonadectomized females. The same protocol of estrogen treatment and withdrawal was ineffective in gonadectomized males. In contrast to estrogen, daily progesterone injections for three weeks elevated CSD susceptibility, and one-week withdrawal after two weeks of treatment partially normalized this effect. Autoradiography did not reveal significant changes in glutamate or GABAA receptor binding density after estrogen treatment and withdrawal. CONCLUSIONS: These data suggest that females are more susceptible to CSD, and sexual dimorphism is abrogated by gonadectomy. Moreover, estrogen withdrawal after prolonged daily treatment enhances CSD susceptibility. These findings may have implications for estrogen-withdrawal migraine, although the latter tends to be without aura.


Asunto(s)
Depresión de Propagación Cortical , Trastornos Migrañosos , Ratas , Femenino , Masculino , Animales , Depresión de Propagación Cortical/fisiología , Progesterona/farmacología , Receptores de GABA-A , Estrógenos/farmacología , Glutamatos/farmacología
5.
Stroke ; 53(12): 3741-3750, 2022 12.
Artículo en Inglés | MEDLINE | ID: mdl-36252110

RESUMEN

BACKGROUND: It has been reported that the S1P (sphingosine 1-phosphate) receptor modulator fingolimod reduces infarction in rodent models of stroke. Recent studies have suggested that circadian rhythms affect stroke and neuroprotection. Therefore, this study revisited the use of fingolimod in mouse focal cerebral ischemia to test the hypothesis that efficacy might depend on whether experiments were performed during the inactive sleep or active wake phases of the circadian cycle. METHODS: Two different stroke models were implemented in male C57Bl/6 mice-transient middle cerebral artery occlusion and permanent distal middle cerebral artery occlusion. Occlusion occurred either during inactive or active circadian phases. Mice were treated with 1 mg/kg fingolimod at 30- or 60-minute postocclusion and 1 day later for permanent and transient middle cerebral artery occlusion, respectively. Infarct volume, brain swelling, hemorrhagic transformation, and behavioral outcome were assessed at 2 or 3 days poststroke. Three independent experiments were performed in 2 different laboratories. RESULTS: Fingolimod decreased peripheral lymphocyte number in naive mice, as expected. However, it did not significantly affect infarct volume, brain swelling, hemorrhagic transformation, or behavioral outcome at 2 or 3 days after transient or permanent focal cerebral ischemia during inactive or active circadian phases of stroke onset. CONCLUSIONS: Outcomes were not improved by fingolimod in either transient or permanent focal cerebral ischemia during both active and inactive circadian phases. These negative findings suggest that further testing of fingolimod in clinical trials may not be warranted unless translational studies can identify factors associated with fingolimod's efficacy or lack thereof.


Asunto(s)
Edema Encefálico , Isquemia Encefálica , Accidente Cerebrovascular , Animales , Ratones , Masculino , Clorhidrato de Fingolimod/farmacología , Clorhidrato de Fingolimod/uso terapéutico , Infarto de la Arteria Cerebral Media/tratamiento farmacológico , Edema Encefálico/tratamiento farmacológico , Esfingosina , Accidente Cerebrovascular/tratamiento farmacológico , Ratones Endogámicos C57BL , Hemorragia/tratamiento farmacológico , Modelos Animales de Enfermedad
6.
J Neuroinflammation ; 18(1): 37, 2021 Jan 30.
Artículo en Inglés | MEDLINE | ID: mdl-33516262

RESUMEN

BACKGROUND: The role of the immune system in stroke is well-recognised. Fingolimod, an immunomodulatory agent licensed for the management of relapsing-remitting multiple sclerosis, has been shown to provide benefit in rodent models of stroke. Its mechanism of action, however, remains unclear. We hypothesised fingolimod increases the number and/or function of regulatory T cells (Treg), a lymphocyte population which promotes stroke recovery. The primary aim of this study was to rigorously investigate the effect of fingolimod on Tregs in a mouse model of brain ischaemia. The effect of fingolimod in mice with common stroke-related comorbidities (ageing and hypercholesteremia) was also investigated. METHODS: Young (15-17 weeks), aged C57BL/6 mice (72-73 weeks), and ApoE-/- mice fed a high-fat diet (20-21 weeks) underwent permanent electrocoagulation of the left middle cerebral artery. Mice received either saline or fingolimod (0.5 mg/kg or 1 mg/kg) at 2, 24, and 48 h post-ischaemia via intraperitoneal injection. Another cohort of young mice (8-9, 17-19 weeks) received short-term (5 days) or long-term (10 days) fingolimod (0.5 mg/kg) treatment. Flow cytometry was used to quantify Tregs in blood, spleen, and lymph nodes. Immunohistochemistry was used to quantify FoxP3+ cell infiltration into the ischaemic brain. RESULTS: Fingolimod significantly increased the frequency of Tregs within the CD4+ T cell population in blood and spleen post-ischaemia in all three mouse cohorts compared to untreated ischemic mice. The highest splenic Treg frequency in fingolimod-treated mice was observed in ApoE-/- mice (9.32 ± 1.73% vs. 7.8 ± 3.01% in young, 6.09 ± 1.64% in aged mice). The highest circulating Treg frequency was also noted in ApoE-/- mice (8.39 ± 3.26% vs. 5.43 ± 2.74% in young, 4.56 ± 1.60% in aged mice). Fingolimod significantly increased the number of FoxP3+ cells in the infarct core of all mice. The most pronounced effects were seen when mice were treated for 10 days post-ischaemia. CONCLUSIONS: Fingolimod increases Treg frequency in spleen and blood post-ischaemia and enhances the number of FoxP3+ cells in the ischaemic brain. The effect of fingolimod on this regulatory cell population may underlie its neuroprotective activity and could be exploited as part of future stroke therapy.


Asunto(s)
Envejecimiento/efectos de los fármacos , Isquemia Encefálica/tratamiento farmacológico , Modelos Animales de Enfermedad , Clorhidrato de Fingolimod/farmacología , Inmunosupresores/farmacología , Linfocitos T Reguladores/efectos de los fármacos , Envejecimiento/inmunología , Animales , Isquemia Encefálica/inmunología , Dieta Alta en Grasa/efectos adversos , Femenino , Clorhidrato de Fingolimod/uso terapéutico , Inmunosupresores/uso terapéutico , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Linfocitos T Reguladores/inmunología
7.
Immunol Cell Biol ; 97(1): 5-16, 2019 01.
Artículo en Inglés | MEDLINE | ID: mdl-30022515

RESUMEN

Stroke is a major cause of morbidity and mortality worldwide. Despite the intensive search for new therapies, hundreds of agents targeting various pathophysiological mechanisms have failed clinical trials, and the thrombolytic agent tissue plasminogen activator is currently the only FDA-approved medication for the treatment of acute ischemic stroke. The immune system is involved in all stages of stroke, from the pathogenesis of risk factors to neurotoxicity, to tissue remodeling and repair. There is a bidirectional interaction between the brain and the immune system, with stroke-induced immunosuppression and subsequent infection a principal source of patient mortality. Newer work also points to a role for the gut microbiota in the immune response to stroke, while clinical sequelae such as dementia might now also be explained in immune terms. However, the exact roles of innate and adaptive components have not been fully elucidated, with studies reporting both detrimental and beneficial functions. Time is a key determinant in defining whether immunity and inflammation are neuroprotective or neurotoxic. The local inflammatory milieu also has a clear influence on many proposed treatments. This review examines the individual components of the immune response to stroke, highlighting the most promising future stroke immunotherapies.


Asunto(s)
Inmunoterapia , Accidente Cerebrovascular/inmunología , Inmunidad Adaptativa , Microbioma Gastrointestinal/inmunología , Humanos , Inmunidad Innata , Accidente Cerebrovascular/patología , Accidente Cerebrovascular/terapia , Activador de Tejido Plasminógeno/uso terapéutico
8.
Pharmacol Res ; 125(Pt B): 232-245, 2017 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-28855094

RESUMEN

The lipid mediator sphingosine 1-phosphate (S1P) affects cellular functions in most systems. Interest in its therapeutic potential has increased following the discovery of its G protein-coupled receptors and the recent availability of agents that can be safely administered in humans. Although the role of S1P in bone biology has been the focus of much less research than its role in the nervous, cardiovascular and immune systems, it is becoming clear that this lipid influences many of the functions, pathways and cell types that play a key role in bone maintenance and repair. Indeed, S1P is implicated in many osteogenesis-related processes including stem cell recruitment and subsequent differentiation, differentiation and survival of osteoblasts, and coupling of the latter cell type with osteoclasts. In addition, S1P's role in promoting angiogenesis is well-established. The pleiotropic effects of S1P on bone and blood vessels have significant potential therapeutic implications, as current therapeutic approaches for critical bone defects show significant limitations. Because of the complex effects of S1P on bone, the pharmacology of S1P-like agents and their physico-chemical properties, it is likely that therapeutic delivery of S1P agents will offer significant advantages compared to larger molecular weight factors. Hence, it is important to explore novel methods of utilizing S1P agents therapeutically, and improve our understanding of how S1P and its receptors modulate bone physiology and repair.


Asunto(s)
Huesos/metabolismo , Lisofosfolípidos/metabolismo , Osteogénesis/fisiología , Esfingosina/análogos & derivados , Animales , Humanos , Neovascularización Fisiológica/fisiología , Osteoblastos/metabolismo , Osteoclastos/metabolismo , Transducción de Señal , Esfingosina/metabolismo , Células Madre/metabolismo
9.
Stroke ; 47(5): 1312-1318, 2016 05.
Artículo en Inglés | MEDLINE | ID: mdl-27032444

RESUMEN

BACKGROUND AND PURPOSE: The debate over the fact that experimental drugs proposed for the treatment of stroke fail in the translation to the clinical situation has attracted considerable attention in the literature. In this context, we present a retrospective pooled analysis of a large data set from preclinical studies, to examine the effects of early versus late administration of intravenous recombinant tissue-type plasminogen activator. METHODS: We collected data from 26 individual studies from 9 international centers (13 researchers; 716 animals) that compared recombinant tissue-type plasminogen activator with controls, in a unique mouse model of thromboembolic stroke induced by an in situ injection of thrombin into the middle cerebral artery. Studies were classified into early (<3 hours) versus late (≥3 hours) drug administration. Final infarct volumes, assessed by histology or magnetic resonance imaging, were compared in each study, and the absolute differences were pooled in a random-effect meta-analysis. The influence of time of administration was tested. RESULTS: When compared with saline controls, early recombinant tissue-type plasminogen activator administration was associated with a significant benefit (absolute difference, -6.63 mm(3); 95% confidence interval, -9.08 to -4.17; I(2)=76%), whereas late recombinant tissue-type plasminogen activator treatment showed a deleterious effect (+5.06 mm(3); 95% confidence interval, +2.78 to +7.34; I(2)=42%; Pint<0.00001). Results remained unchanged after subgroup analyses. CONCLUSIONS: Our results provide the basis needed for the design of future preclinical studies on recanalization therapies using this model of thromboembolic stroke in mice. The power analysis reveals that a multicenter trial would require 123 animals per group instead of 40 for a single-center trial.


Asunto(s)
Isquemia Encefálica/tratamiento farmacológico , Fibrinolíticos/farmacología , Accidente Cerebrovascular/tratamiento farmacológico , Activador de Tejido Plasminógeno/farmacología , Animales , Isquemia Encefálica/patología , Modelos Animales de Enfermedad , Fibrinolíticos/administración & dosificación , Infarto de la Arteria Cerebral Media/tratamiento farmacológico , Infarto de la Arteria Cerebral Media/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Accidente Cerebrovascular/patología , Activador de Tejido Plasminógeno/administración & dosificación
10.
J Biol Chem ; 289(30): 20845-57, 2014 Jul 25.
Artículo en Inglés | MEDLINE | ID: mdl-24928515

RESUMEN

Sphingosine kinase 2 (SPK2) and autophagy are both involved in brain preconditioning, but whether preconditioning-induced SPK2 up-regulation and autophagy activation are linked mechanistically remains to be elucidated. In this study, we used in vitro and in vivo models to explore the role of SPK2-mediated autophagy in isoflurane and hypoxic preconditioning. In primary mouse cortical neurons, both isoflurane and hypoxic preconditioning induced autophagy. Isoflurane and hypoxic preconditioning protected against subsequent oxygen glucose deprivation or glutamate injury, whereas pretreatment with autophagy inhibitors (3-methyladenine or KU55933) abolished preconditioning-induced tolerance. Pretreatment with SPK2 inhibitors (ABC294640 and SKI-II) or SPK2 knockdown prevented preconditioning-induced autophagy. Isoflurane also induced autophagy in mouse in vivo as shown by Western blots for LC3 and p62, LC3 immunostaining, and electron microscopy. Isoflurane-induced autophagy in mice lacking the SPK1 isoform (SPK1(-/-)), but not in SPK2(-/-)mice. Sphingosine 1-phosphate and the sphingosine 1-phosphate receptor agonist FTY720 did not protect against oxygen glucose deprivation in cultured neurons and did not alter the expression of LC3 and p62, suggesting that SPK2-mediated autophagy and protections are not S1P-dependent. Beclin 1 knockdown abolished preconditioning-induced autophagy, and SPK2 inhibitors abolished isoflurane-induced disruption of the Beclin 1/Bcl-2 association. These results strongly indicate that autophagy is involved in isoflurane preconditioning both in vivo and in vitro and that SPK2 contributes to preconditioning-induced autophagy, possibly by disrupting the Beclin 1/Bcl-2 interaction.


Asunto(s)
Autofagia , Corteza Cerebral/metabolismo , Precondicionamiento Isquémico , Neuronas/metabolismo , Fosfotransferasas (Aceptor de Grupo Alcohol)/metabolismo , Adamantano/análogos & derivados , Adamantano/farmacología , Anestésicos por Inhalación/farmacología , Animales , Proteínas Reguladoras de la Apoptosis/genética , Proteínas Reguladoras de la Apoptosis/metabolismo , Beclina-1 , Células Cultivadas , Corteza Cerebral/citología , Inhibidores Enzimáticos/farmacología , Isoflurano/farmacología , Lactosilceramidos/genética , Lactosilceramidos/metabolismo , Ratones , Ratones Noqueados , Morfolinas/farmacología , Neuronas/citología , Fosfotransferasas (Aceptor de Grupo Alcohol)/antagonistas & inhibidores , Fosfotransferasas (Aceptor de Grupo Alcohol)/genética , Proteínas Proto-Oncogénicas c-bcl-2/genética , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Piridinas/farmacología , Pironas/farmacología , Factor de Transcripción TFIIH , Factores de Transcripción/genética , Factores de Transcripción/metabolismo
11.
Stroke ; 45(5): 1468-73, 2014 May.
Artículo en Inglés | MEDLINE | ID: mdl-24723319

RESUMEN

BACKGROUND AND PURPOSE: Small subcortical white matter infarcts are a common stroke subtype often associated with cognitive deficits. The lack of relevant models confined to white matter has limited the investigation of its pathophysiology. Here, we examine tissue and functional outcome after an ischemic lesion within corpus callosum in wild-type (WT) mice and in mice null for a gene, NOTCH3, linked to white matter ischemic injury in patients. METHODS: WT and NOTCH3 knockout mice were subjected to stereotactic microinjections of the potent vasoconstrictor endothelin-1 at the level of periventricular white matter to induce a focal ischemic lesion. Infarct location was confirmed by MRI, and brains were examined for lesion size and histology; behavioral deficits were assessed ≤1 month in WT mice. RESULTS: Ischemic damage featured an early cerebral blood flow deficit, blood-brain barrier opening, and a lesion largely confined to white matter. At later stages, myelin and axonal degeneration and microglial/macrophage infiltration were found. WT mice displayed prolonged cognitive deficit when tested using a novel object recognition task. NOTCH3 mutants showed larger infarcts and greater cognitive deficit at 7 days post stroke. CONCLUSIONS: Taken together, these data show the usefulness of microinjections of endothelin-1 into periventricular white matter to study focal infarcts and cognitive deficit in WT mice. In short-term studies, stroke outcome was worse in NOTCH3 null mice, consistent with the notion that the lack of the NOTCH3 receptor affects white matter stroke susceptibility.


Asunto(s)
Infarto Cerebral/fisiopatología , Cuerpo Calloso/fisiopatología , Leucoencefalopatías/fisiopatología , Trastornos de la Memoria/fisiopatología , Receptores Notch/deficiencia , Reconocimiento en Psicología/fisiología , Animales , Conducta Animal/fisiología , Infarto Cerebral/genética , Infarto Cerebral/patología , Cuerpo Calloso/efectos de los fármacos , Cuerpo Calloso/patología , Modelos Animales de Enfermedad , Endotelina-1/administración & dosificación , Endotelina-1/farmacología , Predisposición Genética a la Enfermedad/genética , Leucoencefalopatías/genética , Leucoencefalopatías/patología , Masculino , Trastornos de la Memoria/genética , Trastornos de la Memoria/patología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Distribución Aleatoria , Receptor Notch3 , Receptores Notch/genética
12.
Circ J ; 78(4): 795-802, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24632793

RESUMEN

This review focuses on the role of sphingosine-1-phosphate (S1P) signaling in the heart, with particular emphasis on how it could be modulated therapeutically in the context of myocardial infarction (MI). After a brief general description of sphingolipid metabolism and signaling, this review will examine the relationship between S1P and the beneficial effects of high-density lipoprotein (HDL), and finally focus on the known actions of S1P on different mechanisms relevant to MI pathophysiology (cardiomyocyte protection, fibrosis, remodeling, arrhythmia, control of vascular tone and potential repair mechanisms). The potential of particular enzyme isoforms or receptor subtypes for the development of therapeutic agents for MI will also be explored.


Asunto(s)
Lisofosfolípidos/metabolismo , Infarto del Miocardio , Miocitos Cardíacos , Transducción de Señal , Esfingosina/análogos & derivados , Animales , Humanos , Lipoproteínas HDL/metabolismo , Infarto del Miocardio/metabolismo , Infarto del Miocardio/patología , Infarto del Miocardio/fisiopatología , Infarto del Miocardio/terapia , Miocitos Cardíacos/metabolismo , Miocitos Cardíacos/patología , Esfingosina/metabolismo
13.
Circulation ; 125(2): 335-45, 2012 Jan 17.
Artículo en Inglés | MEDLINE | ID: mdl-22144569

RESUMEN

BACKGROUND: Migraine is an independent risk factor for stroke. Mechanisms underlying this association are unclear. Familial hemiplegic migraine (FHM), a migraine subtype that also carries an increased stroke risk, is a useful model for common migraine phenotypes because of shared aura and headache features, trigger factors, and underlying glutamatergic mechanisms. METHODS AND RESULTS: Here, we show that FHM type 1 (FHM1) mutations in Ca(V)2.1 voltage-gated Ca(2+) channels render the brain more vulnerable to ischemic stroke. Compared with wild-type mice, 2 FHM1 mutant mouse strains developed earlier onset of anoxic depolarization and more frequent peri-infarct depolarizations associated with rapid expansion of infarct core on diffusion-weighted magnetic resonance imaging and larger perfusion deficits on laser speckle flowmetry. Cerebral blood flow required for tissue survival was higher in the mutants, leading to infarction with milder ischemia. As a result, mutants developed larger infarcts and worse neurological outcomes after stroke, which were selectively attenuated by a glutamate receptor antagonist. CONCLUSIONS: We propose that enhanced susceptibility to ischemic depolarizations akin to spreading depression predisposes migraineurs to infarction during mild ischemic events, thereby increasing the stroke risk.


Asunto(s)
Isquemia Encefálica/fisiopatología , Trastornos Migrañosos/genética , Mutación , Accidente Cerebrovascular/etiología , Animales , Canales de Calcio Tipo N/genética , Circulación Cerebrovascular , Depresión de Propagación Cortical , Susceptibilidad a Enfermedades , Imagen por Resonancia Magnética , Ratones , Trastornos Migrañosos/complicaciones , Migraña con Aura/genética , Accidente Cerebrovascular/genética
14.
Stroke ; 44(2): 505-11, 2013 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-23287783

RESUMEN

BACKGROUND AND PURPOSE: The sphingosine 1-phosphate receptor agonist fingolimod reduces infarct size in rodent models of stroke and enhances blood-brain barrier integrity. Based on these observations, we hypothesized that combination of fingolimod with tissue plasminogen activator (tPA) would reduce the risk of hemorrhagic transformation associated with delayed administration of tPA. METHODS: We evaluated the effects of fingolimod in a mouse model of thromboembolic stroke, in which both the beneficial effect of reperfusion associated with early tPA treatment and hemorrhagic transformation associated with delayed administration mimic clinical observations in humans. RESULTS: Our results demonstrate that fingolimod treatment attenuates the neurological deficit and reduces infarct volume after in situ thromboembolic occlusion of the middle cerebral artery. Combination of fingolimod and tPA improves the neurological outcome of the thrombolytic therapy and reduces the risk of hemorrhagic transformation associated with delayed administration of tPA. CONCLUSIONS: This study confirms the protective efficacy of fingolimod as a treatment against ischemic stroke in another rodent model of stroke (thromboembolic occlusion), and suggests that fingolimod could potentially be used in combination with tPA to reduce the risk of brain hemorrhage.


Asunto(s)
Hemorragia Cerebral/prevención & control , Modelos Animales de Enfermedad , Glicoles de Propileno/administración & dosificación , Esfingosina/análogos & derivados , Tromboembolia/tratamiento farmacológico , Activador de Tejido Plasminógeno/administración & dosificación , Animales , Hemorragia Cerebral/patología , Clorhidrato de Fingolimod , Masculino , Ratones , Ratones Endogámicos C57BL , Fármacos Neuroprotectores/administración & dosificación , Esfingosina/administración & dosificación , Tromboembolia/patología , Factores de Tiempo , Resultado del Tratamiento
15.
Biochem Biophys Res Commun ; 442(1-2): 92-8, 2013 Dec 06.
Artículo en Inglés | MEDLINE | ID: mdl-24246675

RESUMEN

OBJECTIVE: Nicotinamide rescues ß-cell damage and diabetes in rodents, but a large-scale clinical trial failed to show the benefit of nicotinamide in the prevention of type 1 diabetes. Recent studies have shown that Sirt1 deacetylase, a putative protector of ß-cells, is inhibited by nicotinamide. We investigated the effects of isonicotinamide, which is a derivative of nicotinamide and does not inhibit Sirt1, on streptozotocin (STZ)-induced diabetes in mice. RESEARCH DESIGN AND METHODS: Male C57BL/6 mice were administered with three different doses of STZ (65, 75, and 100 mg/kg BW) alone or in combination with subsequent high-fat feeding. The mice were treated with isonicotinamide (250 mg/kg BW/day) or phosphate-buffered saline for 10 days. The effects of isonicotinamide on STZ-induced diabetes were assessed by blood glucose levels, glucose tolerance test, and immunohistochemistry. RESULTS: Isonicotinamide effectively prevented hyperglycemia induced by higher doses of STZ (75 and 100mg/kg BW) alone and low-dose STZ (65 mg/kg BW) followed by 6-week high-fat diet in mice. The protective effects of isonicotinamide were associated with decreased apoptosis of ß-cells and reductions in both insulin content and insulin-positive area in the pancreas of STZ-administered mice. In addition, isonicotinamide inhibited STZ-induced apoptosis in cultured isolated islets. CONCLUSIONS: These data clearly demonstrate that isonicotinamide exerts anti-diabetogenic effects by preventing ß-cell damage after STZ administration. These findings warrant further investigations on the protective effects of isonicotinamide and related compounds against ß-cell damage in diabetes.


Asunto(s)
Citoprotección , Diabetes Mellitus Experimental/prevención & control , Hipoglucemiantes/administración & dosificación , Células Secretoras de Insulina/efectos de los fármacos , Niacinamida/administración & dosificación , Animales , Apoptosis/efectos de los fármacos , Diabetes Mellitus Experimental/inducido químicamente , Dieta Alta en Grasa/efectos adversos , Células Secretoras de Insulina/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Sirtuina 1/antagonistas & inhibidores , Estreptozocina/administración & dosificación
16.
Nat Rev Neurol ; 19(8): 489-505, 2023 08.
Artículo en Inglés | MEDLINE | ID: mdl-37438431

RESUMEN

Migraine is a leading cause of disability in more than one billion people worldwide, yet it remains universally underappreciated, even by individuals with the condition. Among other shortcomings, current treatments (often repurposed agents) have limited efficacy and potential adverse effects, leading to low treatment adherence. After the introduction of agents that target the calcitonin gene-related peptide pathway, another new drug class, the ditans - a group of selective serotonin 5-HT1F receptor agonists - has just reached the international market. Here, we review preclinical studies from the late 1990s and more recent clinical research that contributed to the development of the ditans and led to their approval for acute migraine treatment by the US Food and Drug Administration and the European Medicines Agency.


Asunto(s)
Trastornos Migrañosos , Receptores de Serotonina , Humanos , Receptores de Serotonina/uso terapéutico , Trastornos Migrañosos/tratamiento farmacológico , Trastornos Migrañosos/inducido químicamente , Antagonistas del Receptor Peptídico Relacionado con el Gen de la Calcitonina/farmacología , Antagonistas del Receptor Peptídico Relacionado con el Gen de la Calcitonina/uso terapéutico , Péptido Relacionado con Gen de Calcitonina , Receptor de Serotonina 5-HT1F
17.
J Neurosci ; 31(15): 5755-63, 2011 Apr 13.
Artículo en Inglés | MEDLINE | ID: mdl-21490217

RESUMEN

Familial hemiplegic migraine type 1, a monogenic migraine variant with aura, is linked to gain-of-function mutations in the CACNA1A gene encoding Ca(V)2.1 channels. The S218L mutation causes severe channel dysfunction, and paroxysmal migraine attacks can be accompanied by seizures, coma, and hemiplegia; patients expressing the R192Q mutation exhibit hemiplegia only. Familial hemiplegic migraine knock-in mice expressing the S218L or R192Q mutation are highly susceptible to cortical spreading depression, the electrophysiological surrogate for migraine aura, and develop severe and prolonged motor deficits after spreading depression. The S218L mutants also develop coma and seizures and sometimes die. To investigate underlying mechanisms for these symptoms, we used multielectrode electrophysiological recordings, diffusion-weighted magnetic resonance imaging, and c-fos immunohistochemistry to trace spreading depression propagation into subcortical structures. We showed that unlike the wild type, cortical spreading depression readily propagated into subcortical structures in both familial hemiplegic migraine type 1 mutants. Whereas the facilitated subcortical spread appeared limited to the striatum in R192Q, hippocampal and thalamic spread was detected in the S218L mutants with an allele-dosage effect. Both strains exhibited increased susceptibility to subcortical spreading depression and reverberating spreading depression waves. Altogether, these data show that spreading depression propagates between cortex, basal ganglia, diencephalon, and hippocampus in genetically susceptible brains, which could explain the prolonged hemiplegia, coma, and seizure phenotype in this variant of migraine with aura.


Asunto(s)
Depresión de Propagación Cortical/fisiología , Animales , Animales Modificados Genéticamente , Fenómenos Fisiológicos Cardiovasculares , Ataxia Cerebelosa/genética , Ataxia Cerebelosa/fisiopatología , Corteza Cerebral/fisiopatología , Coma/fisiopatología , Depresión de Propagación Cortical/genética , Fenómenos Electrofisiológicos , Femenino , Genotipo , Hipocampo/fisiopatología , Imagen por Resonancia Magnética , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Mutantes Neurológicos , Trastornos Migrañosos/genética , Trastornos Migrañosos/fisiopatología , Cloruro de Potasio/farmacología , Proteínas Proto-Oncogénicas c-fos/genética , Caracteres Sexuales , Tálamo/fisiopatología
18.
Stroke ; 43(1): 199-204, 2012 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-21980199

RESUMEN

BACKGROUND AND PURPOSE: Cerebral preconditioning provides insights into endogenous mechanisms that protect the brain from ischemic injury. Hypoxia and the anesthetic isoflurane are powerful preconditioning agents. Recent data show that sphingosine 1-phosphate receptor stimulation improves outcome in rodent models of stroke. Endogenous sphingosine 1-phosphate levels are controlled by the expression and activity of sphingosine kinases (SPK). We hypothesize that SPK upregulation mediates preconditioning induced by isoflurane and hypoxia and reduces ischemic injury. METHODS: Male wild-type C57BL/J, SPK1(-/-) and SPK2(-/-) mice were exposed to isoflurane or hypoxia preconditioning before transient middle cerebral artery occlusion. Infarct volume and neurological outcome were measured 24 hours later. SPK inhibitors (SKI-II and ABC294640) were used to test the involvement of SPK2. Expressions of SPK1, SPK2, and hypoxia-inducible factor 1α were determined. Primary cultures of mouse cortical neurons were exposed to isoflurane before glutamate- or hydrogen peroxide-induced cell death. RESULTS: Isoflurane preconditioning and hypoxia preconditioning significantly reduced infarct volume and improved neurological outcome in wild-type and SPK1(-/-) mice but not in SPK2(-/-) mice. Pretreatment with SKI-II or ABC294640 abolished the isoflurane preconditioning-induced tolerance. Western blot showed a rapid and sustained increase in SPK2 level, whereas SPK1 level was similar between preconditioned mice and controls. Hypoxia-inducible factor 1α was upregulated in wild-type isoflurane-preconditioned mice but not in SPK2(-/-). Isoflurane preconditioning protected primary neurons against cell death, which was abolished in ABC294640-treated cells. CONCLUSIONS: Applying genetic and pharmacological approaches, we demonstrate that neuronal SPK2 isoform plays an important role in cerebral preconditioning.


Asunto(s)
Isquemia Encefálica/metabolismo , Encéfalo/metabolismo , Precondicionamiento Isquémico/métodos , Neuronas/metabolismo , Fosfotransferasas (Aceptor de Grupo Alcohol)/metabolismo , Animales , Encéfalo/patología , Isquemia Encefálica/patología , Subunidad alfa del Factor 1 Inducible por Hipoxia/genética , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Masculino , Ratones , Ratones Noqueados , Neuronas/patología , Fosfotransferasas (Aceptor de Grupo Alcohol)/genética
19.
Lab Invest ; 92(10): 1492-502, 2012 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-22906986

RESUMEN

Conventional histopathology with hematoxylin & eosin (H&E) has been the gold standard for histopathological diagnosis of a wide range of diseases. However, it is not performed in vivo and requires thin tissue sections obtained after tissue biopsy, which carries risk, particularly in the central nervous system. Here we describe the development of an alternative, multicolored way to visualize tissue in real-time through the use of coherent Raman imaging (CRI), without the use of dyes. CRI relies on intrinsic chemical contrast based on vibrational properties of molecules and intrinsic optical sectioning by nonlinear excitation. We demonstrate that multicolor images originating from CH(2) and CH(3) vibrations of lipids and protein, as well as two-photon absorption of hemoglobin, can be obtained with subcellular resolution from fresh tissue. These stain-free histopathological images show resolutions similar to those obtained by conventional techniques, but do not require tissue fixation, sectioning or staining of the tissue analyzed.


Asunto(s)
Rastreo Celular/métodos , Técnicas de Preparación Histocitológica , Espectrometría Raman/métodos , Tomografía de Coherencia Óptica/métodos , Animales , Neoplasias Encefálicas/patología , Línea Celular Tumoral , Enfermedades Desmielinizantes/patología , Modelos Animales de Enfermedad , Hemoglobinas/química , Humanos , Lípidos/química , Ratones , Ratones Endogámicos C57BL , Ratones Desnudos , Proteínas/química , Coloración y Etiquetado , Accidente Cerebrovascular/patología , Tomografía de Coherencia Óptica/instrumentación
20.
J Neurochem ; 123 Suppl 2: 58-64, 2012 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-23050643

RESUMEN

The cerebral endothelium can be a vital source of signaling factors such as brain-derived neurotrophic factor that defends the neuronal parenchyma against stress and injury. But the underlying mechanisms remain to be fully defined. Here, we use cell models to ask how vascular neuroprotection is sustained. Human brain endothelial cells were grown in culture, and conditioned media were transferred to primary rat cortical neurons. Brain endothelial cell-conditioned media activated neuronal Akt signaling and protected neurons against hypoxia and oxygen-glucose deprivation. Blockade of Akt phosphorylation with the PI3-kinase inhibitor LY294002 negated this vascular neuroprotective effect. Upstream of Akt signaling, the brain-derived neurotrophic factor receptor TrkB (neurotrophic tyrosine kinase receptor, type 2) was involved because depletion with TrkB/Fc eliminated the ability of endothelial-conditioned media to protect neurons against hypoxia. Downstream of Akt signaling, activation of GSK-3ß (glycogen synthase kinase 3 beta), caspase 9, caspase 3 and Bad pathways were detected. Taken together, these findings suggest that the molecular basis for vascular neuroprotection involves TrkB-Akt signaling that ameliorates neuronal apoptosis. Further investigation of these mechanisms may reveal new approaches for augmenting endogenous vascular neuroprotection in stroke, brain injury, and neurodegeneration.


Asunto(s)
Hipoxia de la Célula/efectos de los fármacos , Medios de Cultivo Condicionados/farmacología , Fármacos Neuroprotectores/farmacología , Proteínas Proto-Oncogénicas c-akt/metabolismo , Receptor trkB/metabolismo , Transducción de Señal/efectos de los fármacos , Análisis de Varianza , Animales , Hipoxia de la Célula/fisiología , Supervivencia Celular/efectos de los fármacos , Células Cultivadas , Corteza Cerebral/citología , Cromonas/farmacología , Embrión de Mamíferos , Células Endoteliales/química , Inhibidores Enzimáticos/farmacología , Glucosa/deficiencia , Glucógeno Sintasa Quinasa 3/metabolismo , Glucógeno Sintasa Quinasa 3 beta , Humanos , Hipoxia/tratamiento farmacológico , Hipoxia/metabolismo , Microvasos/citología , Morfolinas/farmacología , Neuronas , Fosforilación/efectos de los fármacos , Ratas
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA