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

Bases de datos
Tipo del documento
Intervalo de año de publicación
1.
Epilepsia ; 60(5): 1005-1016, 2019 05.
Artículo en Inglés | MEDLINE | ID: mdl-31032909

RESUMEN

OBJECTIVE: Dogs with spontaneous or acquired epilepsy exhibit resemblance in etiology and disease course to humans, potentially offering a translational model of the human disease. Blood-brain barrier dysfunction (BBBD) has been shown to partake in epileptogenesis in experimental models of epilepsy. To test the hypothesis that BBBD can be detected in dogs with naturally occurring seizures, we developed a linear dynamic contrast-enhanced magnetic resonance imaging (DCE-MRI) analysis algorithm that was validated in clinical cases of seizing dogs and experimental epileptic rats. METHODS: Forty-six dogs with naturally occurring seizures of different etiologies and 12 induced epilepsy rats were imaged using DCE-MRI. Six healthy dogs and 12 naive rats served as control. DCE-MRI was analyzed by linear-dynamic method. BBBD scores were calculated in whole brain and in specific brain regions. Immunofluorescence analysis for transforming growth factor beta (TGF-ß) pathway proteins was performed on the piriform cortex of epileptic dogs. RESULTS: We found BBBD in 37% of dogs with seizures. A significantly higher cerebrospinal fluid to serum albumin ratio was found in dogs with BBBD relative to dogs with intact blood-brain barrier (BBB). A significant difference was found between epileptic and control rats when BBBD scores were calculated for the piriform cortex at 48 hours and 1 month after status epilepticus. Mean BBBD score of the piriform lobe in idiopathic epilepsy (IE) dogs was significantly higher compared to control. Immunohistochemistry results suggested active TGF-ß signaling and neuroinflammation in the piriform cortex of dogs with IE, showing increased levels of serum albumin colocalized with glial acidic fibrillary protein and pSMAD2 in an area where BBBD had been detected by linear DCE-MRI. SIGNIFICANCE: Detection of BBBD in dogs with naturally occurring epilepsy provides the ground for future studies for evaluation of novel treatment targeting the disrupted BBB. The involvement of the piriform lobe seen using our linear DCE-MRI protocol and algorithm emphasizes the possibility of using dogs as a translational model for the human disease.


Asunto(s)
Barrera Hematoencefálica , Enfermedades de los Perros/fisiopatología , Epilepsia/veterinaria , Imagen por Resonancia Magnética/métodos , Neuroimagen/métodos , Albúminas/líquido cefalorraquídeo , Algoritmos , Animales , Neoplasias Encefálicas/complicaciones , Neoplasias Encefálicas/fisiopatología , Neoplasias Encefálicas/veterinaria , Medios de Contraste , Convulsivantes/toxicidad , Enfermedades de los Perros/sangre , Enfermedades de los Perros/líquido cefalorraquídeo , Enfermedades de los Perros/diagnóstico por imagen , Perros , Epilepsia/diagnóstico por imagen , Epilepsia/metabolismo , Epilepsia/fisiopatología , Gliosis/etiología , Paraoxon/toxicidad , Corteza Piriforme/irrigación sanguínea , Corteza Piriforme/diagnóstico por imagen , Corteza Piriforme/metabolismo , Corteza Piriforme/patología , Estudios Prospectivos , Ratas , Albúmina Sérica/análisis , Transducción de Señal , Estado Epiléptico/inducido químicamente , Estado Epiléptico/fisiopatología , Factor de Crecimiento Transformador beta/fisiología
2.
Brain ; 141(2): 422-458, 2018 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-29360998

RESUMEN

The mechanisms underpinning concussion, traumatic brain injury, and chronic traumatic encephalopathy, and the relationships between these disorders, are poorly understood. We examined post-mortem brains from teenage athletes in the acute-subacute period after mild closed-head impact injury and found astrocytosis, myelinated axonopathy, microvascular injury, perivascular neuroinflammation, and phosphorylated tau protein pathology. To investigate causal mechanisms, we developed a mouse model of lateral closed-head impact injury that uses momentum transfer to induce traumatic head acceleration. Unanaesthetized mice subjected to unilateral impact exhibited abrupt onset, transient course, and rapid resolution of a concussion-like syndrome characterized by altered arousal, contralateral hemiparesis, truncal ataxia, locomotor and balance impairments, and neurobehavioural deficits. Experimental impact injury was associated with axonopathy, blood-brain barrier disruption, astrocytosis, microgliosis (with activation of triggering receptor expressed on myeloid cells, TREM2), monocyte infiltration, and phosphorylated tauopathy in cerebral cortex ipsilateral and subjacent to impact. Phosphorylated tauopathy was detected in ipsilateral axons by 24 h, bilateral axons and soma by 2 weeks, and distant cortex bilaterally at 5.5 months post-injury. Impact pathologies co-localized with serum albumin extravasation in the brain that was diagnostically detectable in living mice by dynamic contrast-enhanced MRI. These pathologies were also accompanied by early, persistent, and bilateral impairment in axonal conduction velocity in the hippocampus and defective long-term potentiation of synaptic neurotransmission in the medial prefrontal cortex, brain regions distant from acute brain injury. Surprisingly, acute neurobehavioural deficits at the time of injury did not correlate with blood-brain barrier disruption, microgliosis, neuroinflammation, phosphorylated tauopathy, or electrophysiological dysfunction. Furthermore, concussion-like deficits were observed after impact injury, but not after blast exposure under experimental conditions matched for head kinematics. Computational modelling showed that impact injury generated focal point loading on the head and seven-fold greater peak shear stress in the brain compared to blast exposure. Moreover, intracerebral shear stress peaked before onset of gross head motion. By comparison, blast induced distributed force loading on the head and diffuse, lower magnitude shear stress in the brain. We conclude that force loading mechanics at the time of injury shape acute neurobehavioural responses, structural brain damage, and neuropathological sequelae triggered by neurotrauma. These results indicate that closed-head impact injuries, independent of concussive signs, can induce traumatic brain injury as well as early pathologies and functional sequelae associated with chronic traumatic encephalopathy. These results also shed light on the origins of concussion and relationship to traumatic brain injury and its aftermath.awx350media15713427811001.


Asunto(s)
Traumatismos en Atletas/complicaciones , Conmoción Encefálica/etiología , Traumatismos Craneocerebrales/complicaciones , Traumatismos Craneocerebrales/etiología , Tauopatías/etiología , Lesiones del Sistema Vascular/etiología , Potenciales de Acción/fisiología , Adolescente , Animales , Atletas , Encéfalo/patología , Proteínas de Unión al Calcio , Estudios de Cohortes , Simulación por Computador , Traumatismos Craneocerebrales/diagnóstico por imagen , Proteínas de Unión al ADN/metabolismo , Modelos Animales de Enfermedad , Femenino , Regulación de la Expresión Génica/fisiología , Hipocampo/fisiopatología , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Proteínas de Microfilamentos , Modelos Neurológicos , Corteza Prefrontal/fisiopatología , Receptores CCR2/genética , Receptores CCR2/metabolismo , Receptores de Interleucina-8A/genética , Receptores de Interleucina-8A/metabolismo , Adulto Joven
3.
Brain ; 140(6): 1692-1705, 2017 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-28444141

RESUMEN

A biomarker that will enable the identification of patients at high-risk for developing post-injury epilepsy is critically required. Microvascular pathology and related blood-brain barrier dysfunction and neuroinflammation were shown to be associated with epileptogenesis after injury. Here we used prospective, longitudinal magnetic resonance imaging to quantitatively follow blood-brain barrier pathology in rats following status epilepticus, late electrocorticography to identify epileptic animals and post-mortem immunohistochemistry to confirm blood-brain barrier dysfunction and neuroinflammation. Finally, to test the pharmacodynamic relevance of the proposed biomarker, two anti-epileptogenic interventions were used; isoflurane anaesthesia and losartan. Our results show that early blood-brain barrier pathology in the piriform network is a sensitive and specific predictor (area under the curve of 0.96, P < 0.0001) for epilepsy, while diffused pathology is associated with a lower risk. Early treatments with either isoflurane anaesthesia or losartan prevented early microvascular damage and late epilepsy. We suggest quantitative assessment of blood-brain barrier pathology as a clinically relevant predictive, diagnostic and pharmaco!dynamics biomarker for acquired epilepsy.


Asunto(s)
Anestésicos por Inhalación/farmacología , Bloqueadores del Receptor Tipo 1 de Angiotensina II/farmacología , Barrera Hematoencefálica/diagnóstico por imagen , Barrera Hematoencefálica/fisiopatología , Isoflurano/farmacología , Losartán/farmacología , Imagen por Resonancia Magnética/métodos , Estado Epiléptico/diagnóstico por imagen , Estado Epiléptico/fisiopatología , Anestesia por Inhalación , Anestésicos por Inhalación/administración & dosificación , Bloqueadores del Receptor Tipo 1 de Angiotensina II/administración & dosificación , Animales , Biomarcadores , Barrera Hematoencefálica/efectos de los fármacos , Modelos Animales de Enfermedad , Electrocorticografía , Isoflurano/administración & dosificación , Losartán/administración & dosificación , Masculino , Estudios Prospectivos , Ratas , Ratas Sprague-Dawley , Estado Epiléptico/tratamiento farmacológico
4.
Brain ; 138(Pt 1): 69-79, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-25367022

RESUMEN

The insula, a structure involved in higher order representation of interoceptive states, has recently been implicated in drug craving and social stress. Here, we performed brain magnetic resonance imaging to measure volumes of the insula and amygdala, a structure with reciprocal insular connections, in 26 alcohol-dependent patients and 24 healthy volunteers (aged 22-56 years, nine females in each group). We used an established morphometry method to quantify total and regional insular volumes. Volumetric measurements of the amygdala were obtained using a model-based segmentation/registration tool. In alcohol-dependent patients, anterior insula volumes were bilaterally reduced compared to healthy volunteers (left by 10%, right by 11%, normalized to total brain volumes). Furthermore, alcohol-dependent patients, compared with healthy volunteers, had bilaterally increased amygdala volumes. The left amygdala was increased by 28% and the right by 29%, normalized to total brain volumes. Post-mortem studies of the anterior insula showed that the reduced anterior insular volume may be associated with a population of von Economo neurons, which were 60% diminished in subjects with a history of alcoholism (n = 6) as compared to subjects without a history of alcoholism (n = 6) (aged 32-56 years, all males). The pattern of neuroanatomical change observed in our alcohol-dependent patients might result in a loss of top-down control of amygdala function, potentially contributing to impaired social cognition as well as an inability to control negatively reinforced alcohol seeking and use.


Asunto(s)
Alcoholismo/patología , Amígdala del Cerebelo/patología , Corteza Cerebral/patología , Neuronas/patología , Adulto , Alcoholismo/epidemiología , Análisis de Varianza , Femenino , Humanos , Procesamiento de Imagen Asistido por Computador , Imagen por Resonancia Magnética , Masculino , Persona de Mediana Edad , Trastornos del Humor/etiología , Adulto Joven
5.
J Neurosci ; 31(39): 13921-35, 2011 Sep 28.
Artículo en Inglés | MEDLINE | ID: mdl-21957254

RESUMEN

The SRY-box (Sox) transcription factors regulate oligodendrocyte differentiation, but their signaling targets are largely unknown. We have identified a major signal transduction pathway regulated by Sox containing gene 17 (Sox17) in the oligodendrocyte lineage. Microarray analysis in oligodendrocyte progenitor cells (OPCs) after Sox17 attenuation revealed upregulated genes associated with cell cycle control and activation of the Wingless and integration site (Wnt)/ß-catenin pathway. Sox17 knockdown also increases the levels of cyclin D1, Axin2, and activated ß-catenin. In OPCs, the expression pattern of Sox17, cyclin D1, and secreted Frizzled-related protein-1 in the presence of platelet-derived growth factor (PDGF) was coordinately accelerated by addition of thyroid hormone, indicating differentiation-induced regulation of Sox17 targets. In developing white matter, decreased total ß-catenin, activated ß-catenin, and cyclin D1 levels coincided with the peak of Sox17 expression, and immunoprecipitates showed a developmentally regulated interaction among Sox17, T-cell transcription factor 4, and ß-catenin proteins. In OPCs, PDGF stimulated phosphorylation of glycogen synthase 3ß and the Wnt coreceptor LRP6, and enhanced ß-catenin-dependent gene expression. Sox17 overexpression inhibited PDGF-induced TOPFLASH and cyclin D1 promoter activity, and decreased endogenous cyclin D1, activated ß-catenin, as well as total ß-catenin levels. Recombinant Sox17 prevented Wnt3a from repressing myelin protein expression, and inhibition of Sox17-mediated proteasomal degradation of ß-catenin blocked myelin protein induction. These results indicate that Sox17 suppresses cyclin D1 expression and cell proliferation by directly antagonizing ß-catenin, whose activity in OPCs is stimulated not only by Wnt3a, but also by PDGF. Our identification of downstream targets of Sox17 thus defines signaling pathways and molecular mechanisms in OPCs that are regulated by Sox17 during cell cycle exit and the onset of differentiation in oligodendrocyte development.


Asunto(s)
Proteínas HMGB/fisiología , Oligodendroglía/fisiología , Factores de Transcripción SOXF/fisiología , Transducción de Señal/fisiología , Células Madre/fisiología , Proteínas Wnt/fisiología , beta Catenina/fisiología , Animales , Células Cultivadas , Técnicas de Sustitución del Gen , Proteínas HMGB/antagonistas & inhibidores , Proteínas HMGB/genética , Humanos , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Proteínas de la Mielina/antagonistas & inhibidores , Proteínas de la Mielina/biosíntesis , Células 3T3 NIH , Ratas , Ratas Sprague-Dawley , Proteínas Recombinantes/antagonistas & inhibidores , Proteínas Recombinantes/genética , Proteínas Recombinantes/farmacología , Factores de Transcripción SOXF/antagonistas & inhibidores , Factores de Transcripción SOXF/genética , beta Catenina/antagonistas & inhibidores
6.
Proc Natl Acad Sci U S A ; 106(4): 1267-72, 2009 Jan 27.
Artículo en Inglés | MEDLINE | ID: mdl-19147841

RESUMEN

Pro- and mature neurotrophins often elicit opposing biological effects. For example, mature brain-derived neurotrophic factor (mBDNF) is critical for long-term potentiation induced by high-frequency stimulation, whereas proBDNF facilitate long-term depression induced by low-frequency stimulation. Because mBDNF is derived from proBDNF by endoproteolytic cleavage, mechanisms regulating the cleavage of proBDNF may control the direction of BDNF regulation. Using methods that selectively detect proBDNF or mBDNF, we show that low-frequency stimulation induced predominant proBDNF secretion in cultured hippocampal neurons. In contrast, high-frequency stimulation preferentially increased extracellular mBDNF. Inhibition of extracellular, but not intracellular cleavage of proBDNF greatly reduced high-frequency stimulation-induced extracellular mBDNF. Moreover, high-frequency, but not low-frequency stimulation selectively induced the secretion of tissue plasminogen activator, a key protease involved in extracellular proBDNF to mBDNF conversion. Thus, high-frequency neuronal activity controls the ratio of extracellular proBDNF/mBDNF by regulating the secretion of extracellular proteases. Our study demonstrates activity-dependent control of extracellular proteolytic cleavage of a secretory protein, and reveals an important mechanism that controls diametrically opposed functions of BDNF isoforms.


Asunto(s)
Factor Neurotrófico Derivado del Encéfalo/metabolismo , Espacio Extracelular/metabolismo , Neuronas/fisiología , Animales , Western Blotting , Estimulación Eléctrica , Espacio Intracelular/metabolismo , Neuronas/citología , Neuronas/metabolismo , Isoformas de Proteínas/metabolismo , Precursores de Proteínas/metabolismo , Ratas , Activador de Tejido Plasminógeno/deficiencia , Activador de Tejido Plasminógeno/metabolismo
7.
Sci Transl Med ; 11(521)2019 12 04.
Artículo en Inglés | MEDLINE | ID: mdl-31801888

RESUMEN

A growing body of evidence shows that epileptic activity is frequent but often undiagnosed in patients with Alzheimer's disease (AD) and has major therapeutic implications. Here, we analyzed electroencephalogram (EEG) data from patients with AD and found an EEG signature of transient slowing of the cortical network that we termed paroxysmal slow wave events (PSWEs). The occurrence per minute of the PSWEs was correlated with level of cognitive impairment. Interictal (between seizures) PSWEs were also found in patients with epilepsy, localized to cortical regions displaying blood-brain barrier (BBB) dysfunction, and in three rodent models with BBB pathology: aged mice, young 5x familial AD model, and status epilepticus-induced epilepsy in young rats. To investigate the potential causative role of BBB dysfunction in network modifications underlying PSWEs, we infused the serum protein albumin directly into the cerebral ventricles of naïve young rats. Infusion of albumin, but not artificial cerebrospinal fluid control, resulted in high incidence of PSWEs. Our results identify PSWEs as an EEG manifestation of nonconvulsive seizures in patients with AD and suggest BBB pathology as an underlying mechanism and as a promising therapeutic target.


Asunto(s)
Enfermedad de Alzheimer/fisiopatología , Barrera Hematoencefálica/fisiopatología , Corteza Cerebral/fisiopatología , Electroencefalografía , Epilepsia/fisiopatología , Anciano , Envejecimiento/patología , Animales , Demencia/fisiopatología , Humanos , Masculino , Ratones , Red Nerviosa/fisiopatología , Perfusión , Ratas , Albúmina Sérica/metabolismo
8.
Sci Transl Med ; 11(521)2019 12 04.
Artículo en Inglés | MEDLINE | ID: mdl-31801886

RESUMEN

Aging involves a decline in neural function that contributes to cognitive impairment and disease. However, the mechanisms underlying the transition from a young-and-healthy to aged-and-dysfunctional brain are not well understood. Here, we report breakdown of the vascular blood-brain barrier (BBB) in aging humans and rodents, which begins as early as middle age and progresses to the end of the life span. Gain-of-function and loss-of-function manipulations show that this BBB dysfunction triggers hyperactivation of transforming growth factor-ß (TGFß) signaling in astrocytes, which is necessary and sufficient to cause neural dysfunction and age-related pathology in rodents. Specifically, infusion of the serum protein albumin into the young rodent brain (mimicking BBB leakiness) induced astrocytic TGFß signaling and an aged brain phenotype including aberrant electrocorticographic activity, vulnerability to seizures, and cognitive impairment. Furthermore, conditional genetic knockdown of astrocytic TGFß receptors or pharmacological inhibition of TGFß signaling reversed these symptomatic outcomes in aged mice. Last, we found that this same signaling pathway is activated in aging human subjects with BBB dysfunction. Our study identifies dysfunction in the neurovascular unit as one of the earliest triggers of neurological aging and demonstrates that the aging brain may retain considerable latent capacity, which can be revitalized by therapeutic inhibition of TGFß signaling.


Asunto(s)
Envejecimiento/patología , Barrera Hematoencefálica/patología , Barrera Hematoencefálica/fisiopatología , Transducción de Señal , Factor de Crecimiento Transformador beta/metabolismo , Adulto , Anciano , Anciano de 80 o más Años , Albúminas/metabolismo , Animales , Astrocitos/efectos de los fármacos , Astrocitos/metabolismo , Barrera Hematoencefálica/efectos de los fármacos , Enfermedad Crónica , Disfunción Cognitiva/patología , Disfunción Cognitiva/fisiopatología , Técnicas de Silenciamiento del Gen , Hipocampo/efectos de los fármacos , Hipocampo/patología , Hipocampo/fisiopatología , Humanos , Ratones Transgénicos , Persona de Mediana Edad , Inhibidores de Proteínas Quinasas/farmacología , Receptor Tipo I de Factor de Crecimiento Transformador beta/antagonistas & inhibidores , Receptor Tipo I de Factor de Crecimiento Transformador beta/metabolismo , Adulto Joven
9.
Sci Rep ; 7(1): 7711, 2017 08 09.
Artículo en Inglés | MEDLINE | ID: mdl-28794441

RESUMEN

Brain damage due to stroke or traumatic brain injury (TBI), both leading causes of serious long-term disability, often leads to the development of epilepsy. Patients who develop post-injury epilepsy tend to have poor functional outcomes. Emerging evidence highlights a potential role for blood-brain barrier (BBB) dysfunction in the development of post-injury epilepsy. However, common mechanisms underlying the pathological hyperexcitability are largely unknown. Here, we show that comparative transcriptome analyses predict remodeling of extracellular matrix (ECM) as a common response to different types of injuries. ECM-related transcriptional changes were induced by the serum protein albumin via TGFß signaling in primary astrocytes. In accordance with transcriptional responses, we found persistent degradation of protective ECM structures called perineuronal nets (PNNs) around fast-spiking inhibitory interneurons, in a rat model of TBI as well as in brains of human epileptic patients. Exposure of a naïve brain to albumin was sufficient to induce the transcriptional and translational upregulation of molecules related to ECM remodeling and the persistent breakdown of PNNs around fast-spiking inhibitory interneurons, which was contingent on TGFß signaling activation. Our findings provide insights on how albumin extravasation that occurs upon BBB dysfunction in various brain injuries can predispose neural circuitry to the development of chronic inhibition deficits.


Asunto(s)
Matriz Extracelular/metabolismo , Expresión Génica , Neuronas/metabolismo , Transducción de Señal , Factor de Crecimiento Transformador beta/metabolismo , Astrocitos/metabolismo , Barrera Hematoencefálica/metabolismo , Lesiones Traumáticas del Encéfalo/etiología , Lesiones Traumáticas del Encéfalo/metabolismo , Lesiones Traumáticas del Encéfalo/patología , Corteza Cerebral/metabolismo , Corteza Cerebral/patología , Biología Computacional/métodos , Matriz Extracelular/genética , Perfilación de la Expresión Génica , Hipocampo/metabolismo , Hipocampo/patología , Humanos , Inflamación/genética , Inflamación/metabolismo , Inflamación/patología , Interneuronas/metabolismo , Losartán/farmacología , Receptor Tipo I de Factor de Crecimiento Transformador beta/metabolismo , Activación Transcripcional , Transcriptoma
10.
FEBS Lett ; 542(1-3): 74-8, 2003 May 08.
Artículo en Inglés | MEDLINE | ID: mdl-12729901

RESUMEN

We studied the neuroprotective effects of valproic acid (VPA), a primary mood stabilizer and anticonvulsant, in cultured rat cerebral cortical neurons (CCNs). CCNs underwent spontaneous cell death when their age increased in culture. As shown by mitochondrial activity and calcein-AM assays, treatment of CCNs with VPA starting from day 9 in vitro markedly increased viability and prolonged the life span of the cultures. The neuroprotective action of VPA was time-dependent and occurred at therapeutic levels with a maximal effect at about 0.5 mM. LiCl (1 mM) also protected CCNs from aging-induced, spontaneous cell death but less effectively. VPA-induced neuroprotection in aging CCN cultures was associated with a robust increase in histone H3 acetylation levels and the protective effect was mimicked by treatment with a histone deacetylase inhibitor, trichostatin A, but not by VPA analogs which are inactive in blocking histone deacetylase. Our results suggest a role of histone deacetylase inhibition in mediating the neuroprotective action of VPA.


Asunto(s)
Anticonvulsivantes/farmacología , Antimaníacos/farmacología , Corteza Cerebral/citología , Inhibidores de Histona Desacetilasas , Neuronas/efectos de los fármacos , Fármacos Neuroprotectores/farmacología , Ácido Valproico/farmacología , Envejecimiento , Animales , Muerte Celular/efectos de los fármacos , Células Cultivadas , Relación Dosis-Respuesta a Droga , Inhibidores Enzimáticos/farmacología , Neuronas/citología , Neuronas/enzimología , Ratas
11.
Invest Ophthalmol Vis Sci ; 45(11): 3955-63, 2004 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-15505042

RESUMEN

PURPOSE: To characterize properties of Pdlim2, a novel PDZ and LIM domain-containing protein. METHODS: cDNA encoding Pdlim2 was identified in a cDNA library of transcripts expressed in the tissues of the rat eye irido-corneal angle. The expression pattern of the Pdlim2 gene was studied by Northern blot analysis and in situ hybridization. Proteins interacting with Pdlim2 were identified by pull-down assay and mass spectrometry. Intracellular localization of Pdlim2 was investigated by confocal microscopy. RESULTS: Rat Pdlim2 protein belongs to the ALP subfamily of proteins containing the PDZ domain in the N-terminal portion and the LIM domain in the C-terminal portion of the protein. The Pdlim2 gene was specifically expressed in the corneal epithelial cells, but not in the corneal stroma and endothelium nor in other ocular tissues. Pdlim2 was also expressed in the lung. In rat corneal and lung extracts, alpha-actinin-1, alpha-actinin-4, filamin A, and myosin heavy polypeptide 9 were co-immunoprecipitated with Pdlim2. Myosin VI was co-immunoprecipitated with Pdlim2 from corneal but not lung extracts. alpha-Actinins were the most abundant among immunoprecipitated proteins. Direct interaction of Pdlim2 with alpha-actinins and filamin was confirmed using pull-down assays and gel overlay assay with purified proteins. Pdlim2 and alpha-actinins were co-localized mainly to stress fibers after transfection into COS-7 cells. In transfected COS-7 cells, complexes of Pdlim2 and alpha-actinin-1 were preferentially located along the basal aspect. CONCLUSIONS: These results suggest that Pdlim2, like other ALP subfamily members, may act as an adapter that directs other proteins to the cytoskeleton.


Asunto(s)
Actinina/metabolismo , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Proteínas Contráctiles/metabolismo , Epitelio Corneal/metabolismo , Proteínas de Microfilamentos/metabolismo , Proteínas Adaptadoras Transductoras de Señales/genética , Secuencia de Aminoácidos , Animales , Northern Blotting , Células COS/metabolismo , Chlorocebus aethiops , Filaminas , Expresión Génica , Biblioteca de Genes , Hibridación in Situ , Iris/metabolismo , Pulmón/metabolismo , Espectrometría de Masas , Microscopía Confocal , Datos de Secuencia Molecular , Ratas , Ratas Wistar , Transfección
12.
Invest Ophthalmol Vis Sci ; 45(9): 3081-90, 2004 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-15326124

RESUMEN

PURPOSE: To characterize gene expression pattern in the combined tissues of the rat iridocorneal angle by expressed sequence tag (EST) analysis, as part of the NEIBank project. METHODS: RNA was extracted from dissected tissues of the rat iridocorneal angle (iris, ciliary body, trabecular meshwork, and Schlemm's canal) and used to construct unamplified, non-normalized cDNA libraries in the pSPORT1 vector. Approximately 5000 clones were sequenced from the 5'-end. Clones were clustered and identified using the GRIST software, a procedure based on BLAST comparisons. Complete sequences of several novel cDNAs showing eye-preferred expression patterns were obtained. The expression patterns of several genes have been investigated by Northern blot and in situ hybridization, as well as by RT-PCR. RESULTS: After analysis and removal of non-mRNA sequences, 2195 independent clusters, potentially representing individual eye angle-expressed clones were obtained. The expression profile of the combined rat eye angle tissues was more similar to that of the human iris than to human trabecular meshwork. Several cDNAs encoding transcription factors essential for normal eye development and function including Pax-6, Six3, c-Maf, Maf1, Sox-4, Foxc1, Rx, and Ldb2 were present among sequenced clones. A number of tested cDNAs showed eye-preferred expression patterns. Myocilin, which is abundant in human eye angle tissues, was not observed in the rat collection; however, transcripts for three other olfactomedin-domain proteins were seen. Latrotoxin receptor (CL1AA) and optimedin were shown to be expressed in the iris and ciliary body, as well as in the ganglion and inner nuclear cell layers of the retina, whereas the rat orthologue of the human HNOEL-iso gene was expressed in the iris and sclera and less actively in the trabecular meshwork, retina, and optic nerve. CONCLUSIONS: The iridocorneal libraries are a good source of novel uncharacterized genes and molecular markers for the tissues of the eye angle. Although myocilin is not abundantly expressed in rat eye angle, other olfactomedin-containing genes are expressed there and may play important roles in normal eye function and disease.


Asunto(s)
Córnea/metabolismo , Etiquetas de Secuencia Expresada , Proteínas del Ojo/metabolismo , Perfilación de la Expresión Génica , Iris/metabolismo , Secuencia de Aminoácidos , Animales , Clonación Molecular , Proteínas del Ojo/genética , Expresión Génica , Biblioteca de Genes , Datos de Secuencia Molecular , Ratas , Ratas Wistar
13.
J Neurosci Methods ; 113(1): 59-62, 2002 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-11741722

RESUMEN

Despite enormous progress in the development of new morphological techniques, there is still not a simple technique for visualization of the fiber architecture in the mammalian brain. To develop such a technique, thick (400-600 microm) sections of the rat, mice, calf or postmortal human brain were fixed in paraformaldehyde, dehydrated in a series of ethanol and finally immersed in methyl salicylate. The major principle of this newly developed method was to make the neural tissue transparent, and then utilize the ability of neuronal fibers to deflect and deviate light directed from the side to render them visible. Dark-field illumination was used to create illuminating rays of light arriving at an angle exceeding the collecting angle of the objective lens, thus causing only the axonal pathways to be visible as a bright silver silhouette against a dark background. As a result, a three-dimensional structure of the whole white matter of the brain slice became clearly viewable. This technique worked equally well for mammalian brain frontal, sagittal and horizontal sections, as well as for the spinal cord sections. The method was appropriate for verification of axonal fiber courses in brain slice preparations used in electrophysiological experiments, including special applications, such as visualization of axonal bundles within neural transplants. Due to its simplicity, the technique can be successfully used even in an amateur laboratory having basic microscopy equipment and reagents.


Asunto(s)
Axones/ultraestructura , Vías Nerviosas/ultraestructura , Salicilatos , Animales , Encéfalo/citología , Encéfalo/ultraestructura , Fijadores , Masculino , Ratones , Ratones Endogámicos C3H , Fibras Nerviosas/ultraestructura , Vías Nerviosas/citología , Ratas , Ratas Wistar
14.
Seishin Shinkeigaku Zasshi ; 105(1): 81-6, 2003.
Artículo en Japonés | MEDLINE | ID: mdl-12701214

RESUMEN

Lithium has long been one of the primary drugs used to treat bipolar mood disorder. However, neither the etiology of this disease nor the therapeutic mechanism(s) of this drug is well understood. Several lines of clinical evidence suggest that lithium has neurotrophic actions. For example chronic lithium treatment increases the volume of gray matter and the content of N-acetyl-aspartate, a cell survival marker, in bipolar mood disorder patients (Moore et al., 2000). Moreover, treatment with this mood-stabilizer suppresses the decrease in the volume of the subgenual pre-frontal cortex found in bipolar patients (Drevets, 2001). To elucidate molecular mechanisms underlying the neuroprotective and neurotrophic actions of lithium, we employed a preparation of cultured cortical neurons prepared form embryonic rats. We found that treatment with therapeutic doses (0.2-1.2 mM) of lithium robustly protects cortical neurons from multiple insults, notably glutamate-induced excitotoxicity. The neuroprotection against glutamate excitotoxicity is time-dependent, requiring treatment for 5-6 days for maximal effect, and is associated with a reduction in NMDA receptor-mediated Ca2+ influx. The latter is correlated with a decrease in Tyrosine 1472 phosphorylation levels in the NR2B subunit of NMDA receptors and a loss of Src kinase activity which is involved in NR2B tyrosine phosphorylation. Neither the activity of total tyrosine protein kinase nor that of tyrosine protein phosphatase is affected by this drug, indicating the selectivity of the modulation. Lithium neuroprotection against excitotoxicity is inhibited by a BDNF-neutralizing antibody and K252a, a Trk antagonist. Lithium treatment time-dependently increases the intracellular level of BDNF in cortical neurons and activates its receptor, TrkB. The neuroprotection can be completely blocked by either heterozygous or homozygous knockout of the BDNF gene. These results suggest a central role of BDNF and TrkB in mediating the neuroprotective effects of this mood-stabilizer. Finally, long-term lithium treatment of cortical neurons stimulates the proliferation of their progenitor cells detected by co-labeling with BrdU and nestin. Lithium pretreatment also blocks the decrease in progenitor proliferation induced by glutamate, glucocorticoids and haloperidol, suggesting a role in CNS neuroplasticity. We used animal models to investigate further therapeutic potentials for lithium. In the MCAO/reperfusion model of stroke, we found that post-insult treatment with lithium robustly reduced infarct volume and neurological deficits. These beneficial effects were evident when therapeutic concentrations of lithium were injected at least up to 3 h after ischemic onset. The neuroprotection was associated with activation of heat-shock factor-1 and induction of heat-shock protein-70, a cytoprotective protein. In a rat excitotoxic model of Huntington's disease, the excitotoxin-induced loss of striatal medium-sized neurons was markedly reduced by lithium. This lithium protection was correlated with up-regulation of cytoprotective Bcl-2 and down-regulation of apoptotic proteins p53 and Bax, and neurons showing DNA damage and caspase-3 activation. Taken together, our results provide a new insight into the molecular mechanisms involved in lithium neuroprotection against glutamate excitotoxicity. Moreover, these novel molecular and cellular actions might contribute to the neurotrophic and neuroprotective actions of this mood-stabilizer in patients, and could be related to its clinical efficacy for treating mood disorder patients. Clearly, mood-stabilizers may have expanded use for treating excitotoxin-related neurodegenerative diseases.


Asunto(s)
Litio/farmacología , Neuronas/efectos de los fármacos , Animales , Técnicas In Vitro , Litio/uso terapéutico , Ratas
15.
J Neurosci Methods ; 236: 125-47, 2014 Oct 30.
Artículo en Inglés | MEDLINE | ID: mdl-25169049

RESUMEN

The study of CNS glial cell function requires experimental methods to detect, purify, and manipulate each cell population with fidelity and specificity. With the identification and cloning of cell- and stage-specific markers, glial cell analysis techniques have grown beyond physical methods of tissue dissociation and cell culture, and become highly specific with immunoselection of cell cultures in vitro and genetic targeting in vivo. The unique plasticity of glial cells offers the potential for cell replacement therapies in neurological disease that utilize neural cells derived from transplanted neural stem and progenitor cells. In this mini-review, we outline general physical and genetic approaches for macroglial cell generation. We summarize cell culture methods to obtain astrocytes and oligodendrocytes and their precursors, from developing and adult tissue, as well as approaches to obtain human neural progenitor cells through the establishment of stem cells. We discuss popular targeting rodent strains designed for cell-specific detection, selection and manipulation of neuroglial cell progenitors and their committed progeny. Based on shared markers between astrocytes and stem cells, we discuss genetically modified mouse strains with overlapping expression, and highlight SOX-expressing strains available for targeting of stem and progenitor cell populations. We also include recently established mouse strains for detection, and tag-assisted RNA and miRNA analysis. This discussion aims to provide a brief overview of the rapidly expanding collection of experimental approaches and genetic resources for the isolation and targeting of macroglial cells, their sources, progeny and gene products to facilitate our understanding of their properties and potential application in pathology.


Asunto(s)
Astrocitos/fisiología , Separación Celular/métodos , Marcación de Gen/métodos , Oligodendroglía/fisiología , Animales , Técnicas de Cultivo de Célula , Humanos , Modelos Genéticos , Células Madre/fisiología
16.
J Med Case Rep ; 7: 83, 2013 Mar 19.
Artículo en Inglés | MEDLINE | ID: mdl-23510050

RESUMEN

INTRODUCTION: Isolated aphonia induced by acute stroke is a rare phenomenon with only a few cases reported in the literature. CASE PRESENTATION: We report an unusual case of a 44-year-old African-American man with a history of hypertension, smoking and cocaine use who developed acute aphonia secondary to simultaneous ischemic infarctions of the bilateral putamen nuclei. CONCLUSION: We describe the clinical presentation of acute aphonia induced by bilateral putamen nuclei ischemic infarctions, correlating clinical symptoms with injury localization. We further highlight the anatomic and functional organization of the neural pathways involved.

17.
Invest Ophthalmol Vis Sci ; 52(5): 2584-92, 2011 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-21228389

RESUMEN

PURPOSE: Olfactomedin 2 (OLFM2) belongs to the family of olfactomedin domain-containing proteins. Genetic data suggest its association with glaucoma in Japanese patients. However, its functions are still elusive. In this study, the properties of mammalian OLFM2 were investigated. METHODS: Expression of the rat and mouse Olfm2 gene was studied by using real-time PCR and in situ hybridization. Substitutions were introduced into OLFM2 by mutagenesis in vitro. Intracellular localization of OLFM2 was studied by confocal microscopy after transient transfection in HEK293 cells. Interaction of OLFM2 with olfactomedin 1 (Olfm1), olfactomedin 3 (Olfm3), myocilin, and gliomedin was studied by using co-immunoprecipitation. RESULTS: Two major human OLFM2 mRNAs encode secreted proteins with a length of 454 and 478 amino acids. OLFM2 is more closely related to OLFM1 and -3 than to any other family members. Olfm2 showed the most dynamic expression pattern compared with Olfm1 and -3 during mouse eye development and was expressed preferentially in the developing retinal ganglion cell layer. Among three OLFM2 substitutions tested (T86M, R144Q, and L420S), only L420S completely blocked secretion of the protein. OLFM2 interacted with Olfm1 and -3, but not with myocilin and gliomedin. Co-transfection of the L420S mutant with wild-type Olfm1 and -3 significantly inhibited secretion of Olfm1 and -3. CONCLUSIONS: Highly conserved OLFM2 protein may play an important role in the course of retinal and eye development. Severe mutations in one of the closely related olfactomedin domain-containing proteins (Olfm1-3) may block the secretion and probably the activity of all three family members, leading to more pronounced diseases of the retina than the knockout of individual genes.


Asunto(s)
Proteínas de la Matriz Extracelular/genética , Ojo/embriología , Regulación del Desarrollo de la Expresión Génica/fisiología , Glicoproteínas/genética , Dominios y Motivos de Interacción de Proteínas/genética , Células Ganglionares de la Retina/metabolismo , Secuencia de Aminoácidos , Animales , Moléculas de Adhesión Celular Neuronal/metabolismo , Proteínas del Citoesqueleto/metabolismo , Proteínas de la Matriz Extracelular/metabolismo , Ojo/crecimiento & desarrollo , Proteínas del Ojo/metabolismo , Técnica del Anticuerpo Fluorescente Indirecta , Glicoproteínas/metabolismo , Células HEK293/metabolismo , Humanos , Inmunoprecipitación , Hibridación in Situ , Ratones , Microscopía Confocal , Datos de Secuencia Molecular , Mutagénesis Sitio-Dirigida , Unión Proteica , Mapeo de Interacción de Proteínas , ARN Mensajero/metabolismo , Ratas , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Homología de Secuencia de Aminoácido , Transfección
18.
Annu Rev Pharmacol Toxicol ; 45: 269-90, 2005.
Artículo en Inglés | MEDLINE | ID: mdl-15822178

RESUMEN

Increasing evidence supports the notion that glyceraldehyde-3-phosphate dehydrogenase (GAPDH) is a protein with multiple functions, including its surprising role in apoptosis. GAPDH is overexpressed and accumulates in the nucleus during apoptosis induced by a variety of insults in diverse cell types. Knockdown of GAPDH using an antisense strategy demonstrates its involvement in the apoptotic cascade in which GAPDH nuclear translocation appears essential. Knowledge concerning the mechanisms underlying GAPDH nuclear translocation and subsequent cell death is growing. Additional evidence suggests that GAPDH may be an intracellular sensor of oxidative stress during early apoptosis. Abnormal expression, nuclear accumulation, changes in physical properties, and loss of glycolytic activity of GAPDH have been found in cellular and transgenic models as well as postmortem tissues of several neurodegenerative diseases. The interaction of GAPDH with disease-related proteins as well as drugs used to treat these diseases suggests that it is a potential molecular target for drug development.


Asunto(s)
Apoptosis/fisiología , Gliceraldehído-3-Fosfato Deshidrogenasa (Fosforilante)/metabolismo , Enfermedades Neurodegenerativas/enzimología , Animales , Muerte Celular/fisiología , Humanos , Estrés Oxidativo/fisiología
19.
Mol Cell Neurosci ; 22(3): 285-97, 2003 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-12691731

RESUMEN

Huntington's disease is due to an expansion of CAG repeats in the huntingtin gene. Huntingtin interacts with several proteins including glyceraldehyde-3-phosphate dehydrogenase (GAPDH). We performed immunohistochemical analysis of GAPDH expression in the brains of transgenic mice carrying the huntingtin gene with 89 CAG repeats. In all wild-type animals examined, GAPDH was evenly distributed among the different cell types throughout the brain. In contrast, the majority of transgenic mice showed GAPDH overexpression, with the most prominent GAPDH changes observed in the caudate putamen, globus pallidus, neocortex, and hippocampal formation. Double staining for NeuN and GFAP revealed that GAPDH overexpression occurred exclusively in neurons. Nissl staining analysis of the neocortex and caudate putamen indicated 24 and 27% of cell loss in transgenic mice, respectively. Subcellular fluorescence analysis revealed a predominant increase in GAPDH immunostaining in the nucleus. Thus, we conclude that mutation of huntingtin is associated with GAPDH overexpression and nuclear translocation in discrete populations of brain neurons.


Asunto(s)
Gliceraldehído-3-Fosfato Deshidrogenasa (Fosforilante)/genética , Enfermedad de Huntington/genética , Enfermedad de Huntington/metabolismo , Animales , Núcleo Caudado/enzimología , Núcleo Caudado/patología , Modelos Animales de Enfermedad , Regulación Enzimológica de la Expresión Génica , Gliceraldehído-3-Fosfato Deshidrogenasa (Fosforilante)/metabolismo , Proteína Huntingtina , Enfermedad de Huntington/patología , Ratones , Ratones Transgénicos , Neocórtex/enzimología , Neocórtex/patología , Proteínas del Tejido Nervioso/genética , Proteínas Nucleares/genética , Expansión de Repetición de Trinucleótido
20.
Proc Natl Acad Sci U S A ; 100(10): 6210-5, 2003 May 13.
Artículo en Inglés | MEDLINE | ID: mdl-12732732

RESUMEN

Lithium has long been a primary drug used to treat bipolar mood disorder, even though the drug's therapeutic mechanisms remain obscure. Recent studies demonstrate that lithium has neuroprotective effects against glutamate-induced excitotoxicity in cultured neurons and in vivo. The present study was undertaken to examine whether postinsult treatment with lithium reduces brain damage induced by cerebral ischemia. We found that s.c. injection of lithium dose dependently (0.5-3 mEq/kg) reduced infarct volume in the rat model of middle cerebral artery occlusionreperfusion. Infarct volume was reduced at a therapeutic dose of 1 mEq/kg even when administered up to 3 h after the onset of ischemia. Neurological deficits induced by ischemia were also reduced by daily administration of lithium over 1 week. Moreover, lithium treatment decreased the number of neurons showing DNA damage in the ischemic brain. These neuroprotective effects were associated with an up-regulation of cytoprotective heat shock protein 70 (HSP70) in the ischemic brain hemisphere as determined by immunohistochemistry and Western blotting analysis. Lithium-induced HSP70 up-regulation in the ischemic hemisphere was preceded by an increase in the DNA binding activity of heat shock factor 1, which regulates the transcription of HSP70. Physical variables and cerebral blood flow were unchanged by lithium treatment. Our results suggest that postinsult lithium treatment reduces both ischemia-induced brain damage and associated neurological deficits. Moreover, the heat shock response is likely to be involved in lithium's neuroprotective actions. Additionally, our studies indicate that lithium may have clinical utility for the treatment of patients with acute stroke.


Asunto(s)
Daño Encefálico Crónico/prevención & control , Infarto Cerebral/prevención & control , Ataque Isquémico Transitorio/fisiopatología , Litio/uso terapéutico , Animales , Apoptosis/efectos de los fármacos , Presión Sanguínea/efectos de los fármacos , Temperatura Corporal/efectos de los fármacos , Infarto Cerebral/patología , Circulación Cerebrovascular/efectos de los fármacos , Ataque Isquémico Transitorio/tratamiento farmacológico , Masculino , Ratas , Ratas Sprague-Dawley , Reflejo de Sobresalto , Reperfusión
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA