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











Base de datos
Intervalo de año de publicación
1.
J Neurosci Res ; 93(10): 1519-25, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26068706

RESUMEN

A major component of the damaging effect after traumatic brain injury (TBI) is activation of the inflammatory system. In particular, chemokines and chemokine-regulated factors become activated in resident brain cells and signal to different invading immune cells. For evaluation of the effect on invading cells 3 days after injury, mice were treated with a single initial dose of the anti-inflammatory agent Rabeximod in an experimental TBI model. For comparison, mice subjected to TBI were similarly injected with cyclophosphamide. TBI resulted in reduced body weight, an effect further enhanced by administration of Rabeximod, without obvious influence on motor performance. As revealed by quantitative RT-PCR, microglial upregulation of chemokine Ccl3 in response to TBI was unaffected by Rabeximod. Also, injury-induced expression of Cxcl10 in plasmacytoid dendritic cells (DCs) and endothelial expression of platelet selectin (Selp) were uninfluenced by Rabeximod. In contrast, Rabeximod robustly reduced the H2-Aa transcript characteristic for classical DCs defined by CD11c/Itgax in the injured brain. In addition, the expression of lysozyme 2 in large phagocytic cells was impaired by Rabeximod. These results show that Rabeximod exerts a selective and potent inhibition of cells serving cortical antigen presentation after brain trauma.


Asunto(s)
Antiinflamatorios/uso terapéutico , Presentación de Antígeno/efectos de los fármacos , Lesiones Encefálicas/tratamiento farmacológico , Corteza Cerebral/patología , Indoles/uso terapéutico , Quinoxalinas/uso terapéutico , Animales , Peso Corporal/efectos de los fármacos , Peso Corporal/fisiología , Ciclofosfamida/uso terapéutico , Citocinas/genética , Citocinas/metabolismo , Células Dendríticas/efectos de los fármacos , Células Dendríticas/metabolismo , Modelos Animales de Enfermedad , Regulación de la Expresión Génica/efectos de los fármacos , Masculino , Ratones , Ratones Endogámicos C57BL , ARN Mensajero/metabolismo , Factores de Tiempo
2.
PLoS One ; 9(8): e104754, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25153123

RESUMEN

Brain trauma is known to activate inflammatory cells via various chemokine signals although their interactions remain to be characterized. Mice deficient in Ccl3, Ccr2 or Cxcl10 were compared with wildtype mice after controlled cortical impact injury. Expression of Ccl3 in wildtypes was rapidly upregulated in resident, regularly spaced reactive microglia. Ccl3-deficiency enhanced endothelial expression of platelet selectin and invasion of peripheral inflammatory cells. Appearance of Ccr2 transcripts, encoding the Ccl2 receptor, reflected invasion of lysozyme 2-expressing phagocytes and classical antigen-presenting dendritic cells expressing major histocompatibility complex class II. Ccr2 also directed clustered plasmacytoid dendritic cells positive for the T-cell attracting chemokine Cxcl10. A reduction in Ccr2 and dendritic cells was found in injured wildtype cortex after cyclophosphamide treatment resembling effects of Ccr2-deficiency. The findings demonstrate the feasibility to control inflammation in the injured brain by regulating chemokine-dependent pathways.


Asunto(s)
Lesiones Encefálicas/patología , Quimiocinas/fisiología , Células Dendríticas/fisiología , Animales , Lesiones Encefálicas/inmunología , Lesiones Encefálicas/metabolismo , Quimiocina CCL3/genética , Quimiocina CCL3/metabolismo , Quimiocina CXCL10/genética , Quimiocina CXCL10/metabolismo , Quimiocinas/inmunología , Quimiocinas/metabolismo , Ciclofosfamida/farmacología , Células Dendríticas/efectos de los fármacos , Células Dendríticas/inmunología , Eliminación de Gen , Inflamación/genética , Inflamación/metabolismo , Inflamación/fisiopatología , Ratones , Fagocitos/metabolismo , Fagocitos/fisiología , Receptores CCR2/genética , Receptores CCR2/metabolismo , Transducción de Señal
3.
PLoS One ; 7(3): e33090, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22461890

RESUMEN

Clearing of dead cells is a fundamental process to limit tissue damage following brain injury. Engulfment has classically been believed to be performed by professional phagocytes, but recent data show that non-professional phagocytes are highly involved in the removal of cell corpses in various situations. The role of astrocytes in cell clearance following trauma has however not been studied in detail. We have found that astrocytes actively collect and engulf whole dead cells in an in vitro model of brain injury and thereby protect healthy neurons from bystander cell death. Time-lapse experiments showed that migrating neurons that come in contact with free-floating cell corpses induced apoptosis, while neurons that migrate through groups of dead cells, garnered by astrocytes, remain unaffected. Furthermore, apoptotic cells are present within astrocytes in the mouse brain following traumatic brain injury (TBI), indicating a possible role for astrocytes in engulfment of apoptotic cells in vivo. qRT-PCR analysis showed that members of both ced pathways and Megf8 are expressed in the cell culture, indicating their possible involvement in astrocytic engulfment. Moreover, addition of dead cells had a positive effect on the protein expression of MEGF10, an ortholog to CED1, known to initiate phagocytosis by binding to phosphatidylserine. Although cultured astrocytes have an immense capacity for engulfment, seemingly without adverse effects, the ingested material is stored rather than degraded. This finding might explain the multinuclear astrocytes that are found at the lesion site in patients with various brain disorders.


Asunto(s)
Apoptosis/fisiología , Astrocitos/fisiología , Neuronas/fisiología , Fagocitosis/fisiología , Animales , Astrocitos/metabolismo , Astrocitos/ultraestructura , Western Blotting , Lesiones Encefálicas/fisiopatología , Comunicación Celular/fisiología , Movimiento Celular/fisiología , Células Cultivadas , Femenino , Proteína Ácida Fibrilar de la Glía/genética , Proteína Ácida Fibrilar de la Glía/metabolismo , Masculino , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Ratones , Ratones Endogámicos C57BL , Microscopía Confocal , Microscopía Electrónica de Transmisión , Oligodendroglía/fisiología , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Estrés Mecánico
4.
Eur J Neurosci ; 34(1): 110-23, 2011 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-21623956

RESUMEN

Increasing evidence suggests that interleukin-1ß (IL-1ß) is a key mediator of the inflammatory response following traumatic brain injury (TBI). Recently, we showed that intracerebroventricular administration of an IL-1ß-neutralizing antibody was neuroprotective following TBI in mice. In the present study, an anti-IL-1ß antibody or control antibody was administered intraperitoneally following controlled cortical injury (CCI) TBI or sham injury in 105 mice and we extended our histological, immunological and behavioral analysis. First, we demonstrated that the treatment antibody reached target brain regions of brain-injured animals in high concentrations (> 11 nm) remaining up to 8 days post-TBI. At 48 h post-injury, the anti-IL-1ß treatment attenuated the TBI-induced hemispheric edema (P < 0.05) but not the memory deficits evaluated using the Morris water maze (MWM). Neutralization of IL-1ß did not influence the TBI-induced increases (P < 0.05) in the gene expression of the Ccl3 and Ccr2 chemokines, IL-6 or Gfap. Up to 20 days post-injury, neutralization of IL-1ß was associated with improved visuospatial learning in the MWM, reduced loss of hemispheric tissue and attenuation of the microglial activation caused by TBI (P < 0.05). Motor function using the rotarod and cylinder tests was not affected by the anti-IL-1ß treatment. Our results suggest an important negative role for IL-1ß in TBI. The improved histological and behavioral outcome following anti-IL-1ß treatment also implies that further exploration of IL-1ß-neutralizing compounds as a treatment option for TBI patients is warranted.


Asunto(s)
Edema Encefálico/etiología , Edema Encefálico/patología , Lesiones Encefálicas/complicaciones , Lesiones Encefálicas/patología , Trastornos del Conocimiento/etiología , Interleucina-1beta/metabolismo , Animales , Anticuerpos/uso terapéutico , Conducta Animal/fisiología , Edema Encefálico/fisiopatología , Lesiones Encefálicas/fisiopatología , Quimiocinas/genética , Quimiocinas/metabolismo , Trastornos del Conocimiento/fisiopatología , Modelos Animales de Enfermedad , Proteína Ácida Fibrilar de la Glía/genética , Proteína Ácida Fibrilar de la Glía/metabolismo , Humanos , Interleucina-6/genética , Interleucina-6/metabolismo , Aprendizaje/fisiología , Masculino , Memoria/fisiología , Ratones , Ratones Endogámicos C57BL , Microglía/citología , Microglía/metabolismo , Pruebas Neuropsicológicas , Resultado del Tratamiento
5.
Restor Neurol Neurosci ; 28(3): 311-21, 2010.
Artículo en Inglés | MEDLINE | ID: mdl-20479526

RESUMEN

PURPOSE: Astroglial responses after traumatic brain injury are difficult to detect with routine morphological methods. The aims for this study were to compare the temporal and spatial expression pattern of vimentin- and glial fibrillary acidic protein (GFAP) in a weight drop model of mild cerebral contusion injury in the rat. We also wanted to study the vimentin response with immunohistochemistry and vimentin mRNA RT-PCR analysis in severe cortical contusion injury produced by the controlled cortical impact in the mouse. METHODS: Vimentin and GFAP immunohistochemistry (1 day, 3 days and 7 days) combined with vimentin mRNA RT-PCR analysis (1 h, 4 h, 22 h, 3 days and 7 days) were used after experimental traumatic brain injury in the rat and mouse. RESULTS: Increases in post-traumatic vimentin mRNA levels in the cortex and in the hippocampus appeared together with vimentin immunoreactivity in astrocytes in the perimeter of the cortical lesion, in the subcortical white matter and in the hippocampus starting at one day after severe trauma. GFAP immunostaining revealed hypertrophic astrocytes peaking at day 3 in the perifocal cortical region. There was no significant increase in GFAP immunoreactivity in the white matter in the rat. However, in the mouse there was a slight increase in the number of GFAP positive cells in this region, 3 days after trauma. Overall the pattern of vimentin immunoreactivity was very similar in the rat and mouse. CONCLUSIONS: Vimentin immunoreactivity was more sensitive than the GFAP staining method to demonstrate the distribution and time course of astrocyte reactions after a contusion injury, especially in the white matter distant from the cortical lesion.


Asunto(s)
Astrocitos/metabolismo , Lesiones Encefálicas/metabolismo , Proteína Ácida Fibrilar de la Glía/biosíntesis , Gliosis/metabolismo , Proteínas del Tejido Nervioso/biosíntesis , Vimentina/biosíntesis , Animales , Astrocitos/patología , Lesiones Encefálicas/patología , Lesiones Encefálicas/fisiopatología , Corteza Cerebral/metabolismo , Corteza Cerebral/patología , Corteza Cerebral/fisiopatología , Modelos Animales de Enfermedad , Proteína Ácida Fibrilar de la Glía/genética , Gliosis/patología , Gliosis/fisiopatología , Hipocampo/metabolismo , Hipocampo/patología , Hipocampo/fisiopatología , Inmunohistoquímica , Masculino , Ratones , Ratones Endogámicos C57BL , Proteínas del Tejido Nervioso/genética , ARN Mensajero/biosíntesis , Distribución Aleatoria , Ratas , Ratas Sprague-Dawley , Regulación hacia Arriba/genética , Vimentina/genética
6.
Eur J Neurosci ; 31(5): 852-63, 2010 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-20374285

RESUMEN

Traumatic brain injury (TBI) in the mouse results in the rapid appearance of scattered clusters of cells expressing the chemokine Cxcl10 in cortical and subcortical areas. To extend the observation of this unique pattern, we used neuropathological mouse models using quantitative reverse transcriptase-polymerase chain reaction, gene array analysis, in-situ hybridization and flow cytometry. As for TBI, cell clusters of 150-200 mum expressing Cxcl10 characterize the cerebral cortex of mice carrying a transgene encoding the Swedish mutation of amyloid precursor protein, a model of amyloid Alzheimer pathology. The same pattern was found in experimental autoimmune encephalomyelitis in mice modelling multiple sclerosis. In contrast, mice carrying a SOD1(G93A) mutant mimicking amyotrophic lateral sclerosis pathology lacked such cell clusters in the cerebral cortex, whereas clusters appeared in the brainstem and spinal cord. Mice homozygous for a null mutation of the Cxcl10 gene did not show detectable levels of Cxcl10 transcript after TBI, confirming the quantitative reverse transcriptase-polymerase chain reaction and in-situ hybridization signals. Moreover, unbiased microarray expression analysis showed that Cxcl10 was among 112 transcripts in the neocortex upregulated at least threefold in both TBI and ageing TgSwe mice, many of them involved in inflammation. The identity of the Cxcl10(+) cells remains unclear but flow cytometry showed increased numbers of activated microglia/macrophages as well as myeloid dendritic cells in the TBI and experimental autoimmune encephalomyelitis models. It is concluded that the Cxcl10(+) cells appear in the inflamed central nervous system and may represent a novel population of cells that it may be possible to target pharmacologically in a broad range of neurodegenerative conditions.


Asunto(s)
Lesiones Encefálicas/metabolismo , Lesiones Encefálicas/patología , Quimiocina CXCL10/biosíntesis , Enfermedades Neurodegenerativas/metabolismo , Enfermedades Neurodegenerativas/patología , Animales , Separación Celular , Modelos Animales de Enfermedad , Citometría de Flujo , Expresión Génica , Perfilación de la Expresión Génica , Hibridación in Situ , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Análisis de Secuencia por Matrices de Oligonucleótidos , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
7.
J Neurotrauma ; 26(8): 1307-14, 2009 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-19317611

RESUMEN

Cerebral gene expression changes in response to traumatic brain injury will provide useful information in the search for future trauma treatment. In order to characterize the outcome of mild brain injury, we studied C57BL/6J mice in a weight-drop, closed head injury model. At various times post-injury, mRNA was isolated from neocortex and hippocampus and transcriptional alterations were studied using quantitative reverse transcriptase PCR and gene array analysis. At three days post-injury, the results showed unilateral injury responses, both in neocortex and hippocampus, with the main effect seen on the side of the skull hit by the dropping weight. Upregulated transcripts encoded products characterizing reactive astrocytes, phagocytes, microglia, and immune-reactive cells. Markers for oligodendrocytes and T-cells were not altered. Notably, strong differences in the responses among individual mice were seen (e.g., for the Gfap transcript expressed by reactive astrocytes and the chemokine Ccl3 transcript expressed by activated microglial cells). In conclusion, mild TBI chiefly activates transcripts leading to tissue signaling, inflammatory processes, and chemokine signaling, as in focal brain injury, suggesting putative targets for drug development.


Asunto(s)
Lesiones Encefálicas/metabolismo , Traumatismos Cerrados de la Cabeza/metabolismo , Hipocampo/metabolismo , Inflamación/metabolismo , Neocórtex/metabolismo , Análisis de Varianza , Animales , Astrocitos/metabolismo , Lesiones Encefálicas/genética , Expresión Génica , Traumatismos Cerrados de la Cabeza/genética , Inflamación/genética , Masculino , Ratones , Microglía/metabolismo , Análisis de Secuencia por Matrices de Oligonucleótidos , Fagocitos/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Estadísticas no Paramétricas
8.
J Neurosci ; 28(33): 8189-98, 2008 Aug 13.
Artículo en Inglés | MEDLINE | ID: mdl-18701681

RESUMEN

Ubiquitin-positive intraneuronal inclusions are a consistent feature of the major human neurodegenerative diseases, suggesting that dysfunction of the ubiquitin proteasome system is central to disease etiology. Research using inhibitors of the 20S proteasome to model Parkinson's disease is controversial. We report for the first time that specifically 26S proteasomal dysfunction is sufficient to trigger neurodegenerative disease. Here, we describe novel conditional genetic mouse models using the Cre/loxP system to spatially restrict inactivation of Psmc1 (Rpt2/S4) to neurons of either the substantia nigra or forebrain (e.g., cortex, hippocampus, and striatum). PSMC1 is an essential subunit of the 26S proteasome and Psmc1 conditional knock-out mice display 26S proteasome depletion in targeted neurons, in which the 20S proteasome is not affected. Impairment of specifically ubiquitin-mediated protein degradation caused intraneuronal Lewy-like inclusions and extensive neurodegeneration in the nigrostriatal pathway and forebrain regions. Ubiquitin and alpha-synuclein neuropathology was evident, similar to human Lewy bodies, but interestingly, inclusion bodies contained mitochondria. We support this observation by demonstrating mitochondria in an early form of Lewy body (pale body) from Parkinson's disease patients. The results directly confirm that 26S dysfunction in neurons is involved in the pathology of neurodegenerative disease. The model demonstrates that 26S proteasomes are necessary for normal neuronal homeostasis and that 20S proteasome activity is insufficient for neuronal survival. Finally, we are providing the first reproducible genetic platform for identifying new therapeutic targets to slow or prevent neurodegeneration.


Asunto(s)
Encéfalo/enzimología , Cuerpos de Inclusión/enzimología , Cuerpos de Lewy/enzimología , Degeneración Nerviosa/enzimología , Degeneración Nerviosa/genética , Neuronas/enzimología , Complejo de la Endopetidasa Proteasomal/deficiencia , Animales , Encéfalo/patología , Femenino , Humanos , Cuerpos de Inclusión/genética , Cuerpos de Inclusión/patología , Cuerpos de Lewy/genética , Cuerpos de Lewy/patología , Masculino , Ratones , Ratones Noqueados , Ratones Transgénicos , Degeneración Nerviosa/patología , Neuronas/patología , Complejo de la Endopetidasa Proteasomal/genética , Complejo de la Endopetidasa Proteasomal/fisiología
9.
Genesis ; 46(7): 368-72, 2008 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-18615710

RESUMEN

Growth differentiation factor-1 (GDF1), a TGF-beta superfamily member, participates in early embryo patterning. Later functions are implied by the Gdf1 expression in the peripheral and central nervous system. Such roles of the gene have been difficult to study, because Gdf1 null mice die during late embryogenesis. Here, we report the production of a mouse carrying a conditional Gdf1 allele, with exon 2 flanked by loxP sites. Crossing these mice with CaMKIIalpha-Cre mice resulted in Gdf1 ablation in the forebrain postnatally. Such mice displayed no behavioral changes or altered expression levels in a set of hippocampal genes examined. However, excision of the floxed Gdf1 exon caused increased expression of the remaining part of the bicistronic Uog1-Gdf1 transcript in the hippocampus. This indicates that the transcript level is regulated by a negative feedback-loop, sensing presence of either the protein or the mRNA region encoded by Gdf1 exon 2.


Asunto(s)
Alelos , Tipificación del Cuerpo/genética , Regulación de la Expresión Génica/fisiología , Péptidos y Proteínas de Señalización Intercelular/genética , Prosencéfalo/embriología , Animales , Cruzamientos Genéticos , Cartilla de ADN/genética , Componentes del Gen , Vectores Genéticos/genética , Factor 1 de Diferenciación de Crecimiento , Hibridación in Situ , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Ratones , Ratones Transgénicos , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
10.
J Neurotrauma ; 25(8): 959-74, 2008 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-18665806

RESUMEN

Cerebral gene expressions change in response to traumatic brain injury (TBI), and future trauma treatment may improve with increased knowledge about these regulations. We subjected C57BL/6J mice to injury by controlled cortical impact (CCI). At various time points post-injury, mRNA from neocortex and hippocampus was isolated, and transcriptional alterations studied using quantitative real-time polymerase chain reaction (PCR) and gene array analysis. Spatial distribution of enhanced expression was characterized by in situ hybridization. Products of the upregulated transcripts serve functions in a range of cellular mechanisms, including stress, inflammation and immune responses, and tissue remodeling. We also identified increased transcript levels characterizing reactive astrocytes, oligodendrocytes, and microglia, and furthermore, we demonstrated a novel pattern of scattered cell clusters expressing the chemokine Cxcl10. Notably, a sustained increase in integrin alpha X (Itgax), characterizing antigen-presenting dendritic cells, was found with the transcript located to similar cell clusters. In contrast, T-cell receptor alpha transcript showed only a modest increase. The induced P-selectin (Selp) expression level in endothelial cells, and chemokines from microglia, may guide perivascular accumulation of extravasating inflammatory monocytes differentiating into dendritic cells. In conclusion, our study shows that following TBI, secondary injury chiefly involves inflammatory processes and chemokine signaling, which comprise putative targets for pharmaceutical neuroprotection.


Asunto(s)
Lesiones Encefálicas/metabolismo , Lesiones Encefálicas/patología , Quimiocinas/metabolismo , Animales , Antígeno CD11c/genética , Antígeno CD11c/metabolismo , Quimiocinas/genética , Análisis por Conglomerados , Células Dendríticas/fisiología , Ratones , Ratones Endogámicos C57BL , Microglía/fisiología , ARN Mensajero/metabolismo
11.
BMC Neurosci ; 9: 43, 2008 Apr 29.
Artículo en Inglés | MEDLINE | ID: mdl-18445277

RESUMEN

BACKGROUND: The Adhesion G protein-coupled receptors (GPCRs) are membrane-bound receptors with long N termini. This family has 33 members in humans. Several Adhesion GPCRs are known to have important physiological functions in CNS development and immune system response mediated by large cell surface ligands. However, the majority of Adhesion GPCRs are still poorly studied orphans with unknown functions. RESULTS: In this study we performed the extensive tissue localization analysis of the entire Adhesion GPCR family in rat and mouse. By applying the quantitative real-time PCR technique we have produced comparable expression profile for each of the members in the Adhesion family. The results are compared with literature data and data from the Allen Brain Atlas project. Our results suggest that the majority of the Adhesion GPCRs are either expressed in the CNS or ubiquitously. In addition the Adhesion GPCRs from the same phylogenetic group have either predominant CNS or peripheral expression, although each of their expression profile is unique. CONCLUSION: Our findings indicate that many of Adhesion GPCRs are expressed, and most probably, have function in CNS. The related Adhesion GPCRs are well conserved in their structure and interestingly have considerable overlap in their expression profiles, suggesting similarities among the physiological roles for members within many of the phylogenetically related clusters.


Asunto(s)
Sistema Nervioso Central/metabolismo , Perfilación de la Expresión Génica/métodos , Proteínas de la Membrana/genética , ARN Mensajero/genética , Receptores Acoplados a Proteínas G/genética , Animales , Evolución Molecular , Glicoproteínas de Membrana , Proteínas de la Membrana/química , Proteínas de la Membrana/clasificación , Ratones , Biología Molecular/métodos , Familia de Multigenes/genética , Filogenia , Complejo GPIb-IX de Glicoproteína Plaquetaria , ARN Mensajero/análisis , Ratas , Receptores Acoplados a Proteínas G/química , Receptores Acoplados a Proteínas G/clasificación , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa/métodos , Especificidad de la Especie
12.
J Mol Neurosci ; 35(2): 179-93, 2008 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-18418736

RESUMEN

Members of the solute carrier families (SLC) 32, 36, and 38, together also designated the beta-group of SLCs, are known to transport neutral amino acids. In this paper, we show that these three families were present before the split of the animal lineage and that they are likely to share a common decent. We also show that the APF transporters found in plants are most likely homologous to the mammalian beta-group, suggesting that this type of transporters arouse early in the evolution of eukaryotes. We performed detailed tissue expression analysis of all the members of the beta-group in rat and found several examples of highly specific expression patterns, with SLC38A7 being exclusively found in liver, SLC38A5 in blood, and SLC38A4 in muscle and liver. Moreover, we found that SLC38A10 is expressed in several endocrine organs. We also found that SLC38A1 is highly up regulated in the cortex from rats treated with diazepam and that SLC38A2 is significantly down regulated in the same tissue. In addition, we performed a detailed expression analysis of SLC38A1 and SLC38A6 in mouse brain using in situ hybridization, which showed that both these transporters are widely expressed in the brain.


Asunto(s)
Sistema de Transporte de Aminoácidos A/genética , Sistemas de Transporte de Aminoácidos Neutros/genética , Sistemas de Transporte de Aminoácidos/genética , Evolución Molecular , Proteínas del Tejido Nervioso/genética , Filogenia , Proteínas del Transporte Vesicular de Aminoácidos Inhibidores/genética , Animales , Anticonvulsivantes/farmacología , Corteza Cerebral/efectos de los fármacos , Corteza Cerebral/fisiología , Diazepam/farmacología , Dopaminérgicos/farmacología , Sistema Endocrino/fisiología , Expresión Génica/efectos de los fármacos , Expresión Génica/fisiología , Humanos , Hibridación in Situ , Levodopa/farmacología , Hígado/fisiología , Masculino , Ratones , Ratones Endogámicos , Músculo Esquelético/fisiología , Ratas , Ratas Wistar
13.
J Neurosci Res ; 84(1): 47-57, 2006 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-16583403

RESUMEN

Three genetic mouse models were examined to define effects of bone morphogenetic protein (BMP) signalling on gene expression in normal and injured adult brain. CaMKII-Cre eliminated the BMP receptor Acvr1 (Alk2) and the common TGFbeta superfamily signal mediator Smad4 or activated a constitutively active Acvr1 in postnatal forebrain neurons. All mutants followed mendelian ratios, with no overt phenotypic changes. In situ hybridization demonstrated normal patterns of the dendritic marker MAP2 (Mtap2) throughout cortex despite neuron-specific losses of Acvr1 or Smad4. However, strong up-regulation of Mtap2 transcript in these mice was found by quantitative RT-PCR (qRT-PCR), indicating that Mtap2 is normally suppressed by BMP. Traumatic brain injury (TBI) resulted in increases of histone-associated DNA fragments in both control and Smad4-deficient cortex. Several cell-type-specific transcripts known to be involved in injury-related responses were measured by qRT-PCR. Gfap mRNA was strongly up-regulated in controls as well as in the loss-of-BMP-signalling mutants. Notably, activated Acvr1 signalling gave significantly lower TBI-induced up-regulations of Gfap and Phox2a mRNA levels, indicating reductions in astroglial and neuronal reactions to injury. Strong impairment in injury-induced Timp1 transcript up-regulation was also seen in these mice. In contrast, osteopontin (Spp1) transcript levels in activated microglia were not reduced by Acvr1 signalling. Altogether, the data suggest that BMP signalling is dispensable in adult cortical neurons but that augmented BMP signalling affects molecular changes associated with neuronal lesions.


Asunto(s)
Proteínas Morfogenéticas Óseas/genética , Lesiones Encefálicas/metabolismo , Regulación de la Expresión Génica/fisiología , Expresión Génica/fisiología , Transducción de Señal/fisiología , Receptores de Activinas Tipo I/genética , Análisis de Varianza , Animales , Conducta Animal/fisiología , Peso Corporal/genética , Lesiones Encefálicas/patología , Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina , Proteínas Quinasas Dependientes de Calcio-Calmodulina/genética , Muerte Celular/fisiología , Proteínas Fluorescentes Verdes/biosíntesis , Historia Medieval , Hibridación in Situ/métodos , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , ARN Mensajero/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa/métodos , Proteína Smad4/genética
14.
J Neurotrauma ; 21(9): 1168-82, 2004 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-15453987

RESUMEN

Mitogen-activated protein kinase (MAPK) cascades are membrane-to-nucleus signaling modules that recently have been implicated as mediators of cellular injury. In this study, we investigated the involvement of the MAP kinase p44/p42 (extracellular signal-regulated kinase [ERK1/2]) in traumatic brain injury (TBI) in rats. There was a strong increase in activated, phosphorylated ERK 1/2 (p-ERK 1/2) protein at 10 min up to 24 h after the injury. Expression of p-ERK occurred in cells identified as neurons, astrocytes, and microglia. Most of the cells expressing p-ERK were TUNEL positive at later time points. Treatment with the MEK inhibitor U0126 or the free radical scavenger S-PBN, both with neuroprotective properties in TBI, attenuated the early activation of ERK and resulted in less activation of caspase-3 and subsequent DNA fragmentation. Post-treatment with U0126 resulted in a significant decrease (-60%) in cortical cavity size and cortical atrophy at 2 weeks after trauma. Overall, the results suggest that ERK activation is initiated by increased oxygen radical activity and that overactivation of ERK sets off secondary cell death mechanisms in TBI. Clinical studies are warranted to evaluate the concept of MEK inhibition in head-injured patients.


Asunto(s)
Apoptosis/fisiología , Lesiones Encefálicas/metabolismo , Lesiones Encefálicas/patología , Radicales Libres/metabolismo , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Animales , Lesiones Encefálicas/enzimología , Muerte Celular/fisiología , Activación Enzimática/fisiología , Masculino , Proteínas Quinasas Activadas por Mitógenos/fisiología , Ratas , Ratas Sprague-Dawley
15.
Genesis ; 40(2): 67-73, 2004 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-15452869

RESUMEN

Catecholaminergic neurons are affected in several neurological and psychiatric diseases. Tyrosine hydroxylase (TH) is the first, rate-limiting enzyme in catecholamine synthesis. We report a knockin mouse expressing Cre-recombinase from the 3'-untranslated region of the endogenous Th gene by means of an internal ribosomal entry sequence (IRES). The resulting Cre expression matches the normal pattern of TH expression, while the pattern and level of TH are not altered in the knockin mouse. Crossings with two different LacZ reporter mice demonstrated Cre-mediated genomic recombination in TH expressing tissues. In addition, LacZ was found in some unexpected cell populations (including oocytes), indicating recombination due to transient developmental TH expression. Our novel knockin mouse can be used for generation of tissue-specific or general knockouts (depending on scheme of crossing) in mice carrying genes flanked by loxP sites. This knockin mouse can also be used for tracing cell lineages expressing TH during development.


Asunto(s)
Integrasas/metabolismo , Recombinación Genética , Tirosina 3-Monooxigenasa/genética , Proteínas Virales/metabolismo , Regiones no Traducidas 3' , Glándulas Suprarrenales/metabolismo , Animales , Química Encefálica/inmunología , Electroporación , Femenino , Genes Reporteros , Heterocigoto , Inmunohistoquímica , Operón Lac , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Células Madre , Distribución Tisular/genética , Transgenes , beta-Galactosidasa/metabolismo
16.
Mol Cell Neurosci ; 25(2): 345-54, 2004 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-15019950

RESUMEN

Bone morphogenetic proteins (BMPs) 4 and 6 as well as MEK inhibitors PD98059 and U0126 potentiate neurotrophin 3 (NT3)- and neurturin (NTN)-induced neurite outgrowth and survival of peripheral neurons from the E9 chicken embryo. Preexposure to BMP4 or PD98059 was sufficient to prime the potentiation of subsequently added NT3. Phosphorylation of Erk2, induced by NT3, was reduced by MEK inhibition but unaffected by BMP signaling. Real-time PCR showed that neither BMP stimulation nor MEK inhibition increased Trk receptor expression and that the BMP-induced genes Smad6 and Id1 were not upregulated by PD98059. In contrast, both MEK inhibition and BMP signaling suppressed transcription of the serum-response element (SRE)-driven Egr1 gene. A reporter assay using NGF-stimulated PC12 cells demonstrated that MEK/Erk/Elk-driven transcriptional activity was inhibited by Smad1/5 and by PD98059. Thus, suppression of SRE-controlled transcription represents a likely convergence point for pathways regulating neurotrophic responses.


Asunto(s)
Diferenciación Celular/fisiología , Ganglios/enzimología , Sistema de Señalización de MAP Quinasas/fisiología , Factores de Crecimiento Nervioso/metabolismo , Neuronas/enzimología , Sistema Nervioso Periférico/enzimología , Factores de Transcripción , Animales , Proteínas Morfogenéticas Óseas/metabolismo , Proteínas Morfogenéticas Óseas/farmacología , Diferenciación Celular/efectos de los fármacos , Embrión de Pollo , Proteínas de Unión al ADN/metabolismo , Proteínas de Unión al ADN/farmacología , Sinergismo Farmacológico , Inhibidores Enzimáticos/farmacología , Ganglios/citología , Ganglios/crecimiento & desarrollo , Genes Reguladores/efectos de los fármacos , Genes Reguladores/genética , MAP Quinasa Quinasa 1 , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Quinasas de Proteína Quinasa Activadas por Mitógenos/antagonistas & inhibidores , Quinasas de Proteína Quinasa Activadas por Mitógenos/metabolismo , Factores de Crecimiento Nervioso/farmacología , Neuritas/efectos de los fármacos , Neuritas/enzimología , Neuritas/ultraestructura , Neuronas/citología , Neuronas/efectos de los fármacos , Neurotrofina 3/metabolismo , Neurotrofina 3/farmacología , Células PC12 , Sistema Nervioso Periférico/citología , Sistema Nervioso Periférico/crecimiento & desarrollo , Proteínas Proto-Oncogénicas/efectos de los fármacos , Proteínas Proto-Oncogénicas/genética , Ratas , Elemento de Respuesta al Suero/efectos de los fármacos , Elemento de Respuesta al Suero/genética , Proteínas Smad , Proteína Smad1 , Transactivadores/metabolismo , Transactivadores/farmacología , Proteína Elk-1 con Dominio ets
17.
Exp Neurol ; 180(2): 167-71, 2003 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-12684030

RESUMEN

Olfactory ensheathing cells (OEC) constitute a specialized population of glia that accompany primary olfactory axons and have been reported to facilitate axonal regeneration after spinal cord injury in vivo. In the present report we describe OEC neurotrophic factor expression and neurotrophic properties of OECs in vitro. Investigation of the rat olfactory system during development and adulthood by radioactive in situ hybridization revealed positive labeling in the olfactory nerve layer for the neurotrophic molecules S-100beta, CNTF, BMP-7/OP-1, and artemin, as well as for the neurotrophic factor receptors RET and TrkC. Ribonuclease protection assay of cultured OEC revealed expression of NGF, BDNF, GDNF, and CNTF mRNA, while NT3 and NT4 mRNA were not detectable. In vitro bioassays of neurotrophic activity involved coculturing of adult OEC with embryonic chick ganglia and demonstrated increased neurite outgrowth from sympathetic, ciliary, and Remak's ganglia. However, when culturing the ganglia with OEC-conditioned medium, neurite outgrowth was not stimulated to any detectable extent. Our results suggest that the neurotrophic properties of OEC may involve secretion of neurotrophic molecules but that cellular interactions are crucial.


Asunto(s)
Proteínas de Drosophila , Factores de Crecimiento Nervioso/biosíntesis , Neuroglía/citología , Neuroglía/metabolismo , Nervio Olfatorio/citología , Nervio Olfatorio/metabolismo , Animales , Bioensayo , Comunicación Celular/fisiología , Células Cultivadas , Embrión de Pollo , Técnicas de Cocultivo , Medios de Cultivo Condicionados/farmacología , Ganglios/citología , Ganglios/embriología , Receptores del Factor Neurotrófico Derivado de la Línea Celular Glial , Hibridación in Situ , Factores de Crecimiento Nervioso/genética , Neuritas/efectos de los fármacos , Neuritas/fisiología , Ensayos de Protección de Nucleasas , Bulbo Olfatorio/citología , Bulbo Olfatorio/embriología , Proteínas Proto-Oncogénicas/biosíntesis , Proteínas Proto-Oncogénicas/genética , Proteínas Proto-Oncogénicas c-ret , ARN Mensajero/biosíntesis , Ratas , Ratas Sprague-Dawley , Proteínas Tirosina Quinasas Receptoras/biosíntesis , Proteínas Tirosina Quinasas Receptoras/genética , Receptor trkC/biosíntesis , Receptor trkC/genética
18.
J Neurosci Res ; 72(4): 444-53, 2003 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-12704806

RESUMEN

We investigated the use of the mouse tyrosine hydroxylase (TH) gene to drive knock-in constructs in catecholaminergic neurons. Two targeting constructs representing truncated forms of either of the BMP receptors ALK-2 or BMPR-II preceded by an internal ribosome entry site (IRES) were introduced into the 3' untranslated region of TH. An frt-flanked neomycin-resistance (neo(r)) cassette was placed in the 3' end of the targeting constructs. Mice homozygous for the knock-in alleles showed various degrees of hypokinetic behavior, depending mainly on whether the neo(r) cassette was removed. In situ hybridization and immunohistochemistry showed that TH mRNA and protein were variously down-regulated in these mouse strains. Reduced levels of dopamine and noradrenalin were found in several brain areas. However, number and morphology of neurons in substantia nigra and their projections to striatum appeared normal in the neo(r)-positive TH hypomorphic mice as examined by markers for L-aromatic amino acid decarboxylase and the dopamine transporter. Elimination of the neo(r) cassette from the knock-in alleles partially restored TH and dopamine levels. The present neo(r)-positive TH hypomorphic mice show that nigrostriatal innervation develops independently of TH and should find use as a model for conditions of reduced catecholamine synthesis, as seen in, for example, L-dihydroxyphenylalanine-responsive dystonia/infantile parkinsonism.


Asunto(s)
Dopamina/metabolismo , Proteínas , Sustancia Negra/enzimología , Tirosina 3-Monooxigenasa/genética , Tirosina 3-Monooxigenasa/metabolismo , Receptores de Activinas Tipo I/genética , Animales , Receptores de Proteínas Morfogenéticas Óseas de Tipo II , Dopamina/análisis , Resistencia a Medicamentos , Genotipo , Inmunohistoquímica , Hibridación in Situ , Ratones , Ratones Transgénicos , Modelos Animales , Mutagénesis Insercional , Neomicina , Norepinefrina/análisis , Norepinefrina/metabolismo , Trastornos Parkinsonianos/fisiopatología , Proteínas Serina-Treonina Quinasas/genética , ARN Mensajero/análisis , Serotonina/análisis , Serotonina/metabolismo , Tirosina 3-Monooxigenasa/deficiencia
19.
J Neurosci Res ; 68(2): 248-53, 2002 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-11948670

RESUMEN

We have developed a transgenic mouse expressing the Cre recombinase under control of a tetracycline-responsive promoter. Using a CamKIIalpha-driven tTA transgenic strain and a lacZ reporter mouse, we obtained the expected neuronal pattern of recombination in the olfactory lobe, cortex, striatum, hippocampus and Purkinje cells. Moreover, recombination can be completely abolished by feeding the mice doxycycline in their drinking water. We also show that it is possible to get a different pattern of recombination by changing the timing of the doxycycline-mediated shutdown of Cre expression. By starting the doxycycline treatment at birth, we restrict recombination to striatum only. This approach should be applicable to other inducible transgenic strains, thus increasing the number of available tissue-specific patterns for conditional knockouts. Also, our tetO-Cre transgene can be combined with any of the increasing number of tetracycline transactivator transgenic strains to direct specifically inducible genomic recombination to several areas of the brain.


Asunto(s)
Encéfalo/efectos de los fármacos , Doxiciclina/administración & dosificación , Regulación de la Expresión Génica , Integrasas/genética , Recombinación Genética/efectos de los fármacos , Transgenes , Proteínas Virales/genética , Animales , Encéfalo/citología , Doxiciclina/farmacología , Genoma , Integrasas/fisiología , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Neuronas/efectos de los fármacos , Regiones Promotoras Genéticas/efectos de los fármacos , Factores de Tiempo , Proteínas Virales/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA