Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
País de afiliación
Intervalo de año de publicación
1.
Childs Nerv Syst ; 35(5): 815-822, 2019 05.
Artículo en Inglés | MEDLINE | ID: mdl-30826957

RESUMEN

PURPOSE: German pediatric guidelines for severe traumatic brain injury (TBI) management expired in 2011. Thus, divergent evidence-based institutional protocols are predominantly being followed. We performed a survey of current Pediatric Intensive Care Unit (PICU) management of isolated severe TBI in Germany to reveal potential varying practices. METHODS: Seventy German PICUs were invited to join an anonymous online survey from February to May 2017. Twenty-nine participants (41.4%) successfully completed the survey (17 university hospitals and 12 district hospitals). The majority of items were polar (yes/no) or scaled (e.g., never - always). Main topics were imaging, neurosurgery, neuromonitoring, adjuvant therapy, and medication. Severity of TBI was defined via Glasgow Coma Scale. RESULTS: The majority of respondents (93.1%) had internal TBI standards, and patients were mainly administered to interdisciplinary trauma units. The use of advanced neuromonitoring techniques, intracranial hypertension management, and drug treatment differed between PICUs. Routine administration of hypertonic saline in TBI-associated cerebral edema was performed by 3.4%, while it was never an option for 31.0% of the participants. Prophylactic anticonvulsive therapy was restrictively performed. If indicated, the main anticonvulsive drugs used were phenobarbital and levetiracetam. Neuroendocrine follow-up was recommended/performed by 58.6% of the PICUs. CONCLUSIONS: This survey provides an overview of the current PICU practices of isolated severe TBI management in Germany and demonstrates a wide instrumental and therapeutical range, revealing an unmet need for the revised national guideline and further (international) clinical trials for the treatment of severe TBI in pediatrics.


Asunto(s)
Lesiones Traumáticas del Encéfalo/epidemiología , Lesiones Traumáticas del Encéfalo/terapia , Unidades de Cuidado Intensivo Pediátrico/normas , Guías de Práctica Clínica como Asunto/normas , Índice de Severidad de la Enfermedad , Encuestas y Cuestionarios/normas , Adolescente , Niño , Preescolar , Femenino , Alemania/epidemiología , Humanos , Lactante , Recién Nacido , Masculino , Resultado del Tratamiento , Adulto Joven
2.
Acta Neuropathol Commun ; 12(1): 124, 2024 Aug 06.
Artículo en Inglés | MEDLINE | ID: mdl-39107839

RESUMEN

Epithelioid glioblastoma (eGB), a very aggressive and rare brain tumour, is associated with a dismal median overall survival. Effective therapies for patients with eGB, particularly with leptomeningeal dissemination, are still lacking. Here, we describe a case of a 25-year-old male diagnosed with an intramedullary cervical tumour with subsequent leptomeningeal disease. Histopathology identified a highly necrotising, epithelioid-type tumour with high cell density, most compatible with the diagnosis of an eGB. DNA analysis revealed an unprecedented B-Raf protooncogene, serine/threonine kinase (BRAF) gene variant in exon 15 (ENST00000288602.6, c.1799_1810delinsATG, p.(V600_W604delinsDG)), triggering activation of the mitogen-activated protein kinase (MAPK) pathway. Consequently, we initiated MAPK inhibitor (MAPKi) therapy, utilizing a combination of BRAF and mitogen-activated protein kinase kinase (MEK) inhibitors. Liquid chromatography-tandem mass spectrometry analysis confirmed the drugs' presence in the patient's cerebrospinal fluid, indicating their capacity to cross the blood-brain barrier. Remarkably, the patient responded very well to therapy and transitioned from a near-comatose state to significantly improved health, sustained for over three months. This study highlights that MAPKi, particularly targeted towards novel BRAFV600 mutations, might offer promising advancements in eGB treatment strategies.


Asunto(s)
Neoplasias Encefálicas , Glioblastoma , Mutación , Inhibidores de Proteínas Quinasas , Proteínas Proto-Oncogénicas B-raf , Humanos , Proteínas Proto-Oncogénicas B-raf/genética , Masculino , Adulto , Glioblastoma/genética , Glioblastoma/tratamiento farmacológico , Glioblastoma/patología , Inhibidores de Proteínas Quinasas/uso terapéutico , Inhibidores de Proteínas Quinasas/farmacología , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/tratamiento farmacológico , Neoplasias Encefálicas/patología , Quinasas de Proteína Quinasa Activadas por Mitógenos/antagonistas & inhibidores , Quinasas de Proteína Quinasa Activadas por Mitógenos/metabolismo , Quinasas de Proteína Quinasa Activadas por Mitógenos/genética
3.
Nat Cell Biol ; 3(12): 1035-42, 2001 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-11781564

RESUMEN

Excitotoxic brain lesions initially result in the primary destruction of brain parenchyma, after which microglial cells migrate towards the sites of injury. At these sites, the cells produce large quantities of oxygen radicals and cause secondary damage that accounts for most of the loss of brain function. Here we show that this microglial migration is strongly controlled in living brain tissue by expression of the integrin CD11a, regulated by the nuclear enzyme poly(ADP-ribose) polymerase-1 (PARP-1) through the formation of a nuclear PARP-NF-kappaB-protein complex. Downregulation of PARP or CD11a by transfection with antisense DNA abrogated microglial migration almost completely and prevented neurons from secondary damage.


Asunto(s)
Antígeno-1 Asociado a Función de Linfocito/genética , Microglía/fisiología , Poli(ADP-Ribosa) Polimerasas/metabolismo , Animales , Encefalopatías/fisiopatología , Movimiento Celular/fisiología , Células Cultivadas , ADN sin Sentido , Agonistas de Aminoácidos Excitadores/farmacología , Expresión Génica/fisiología , Ratones , Microglía/citología , N-Metilaspartato/farmacología , FN-kappa B/metabolismo , Degeneración Nerviosa/fisiopatología , Neurotoxinas/farmacología , Fagocitosis/fisiología , ARN Mensajero/análisis , Transfección
4.
Klin Monbl Augenheilkd ; 228(11): 963-6, 2011 Nov.
Artículo en Alemán | MEDLINE | ID: mdl-21901662

RESUMEN

This paper analyses the case reports for three children in which a papilledema occurred before the age of one year. Furthermore, an analysis is also given of three further case reports for children aged less than one year in which, in spite of open fontanelle, no papilledema was found, however, a dilatation of the sub-arachnoidal space was demonstrated echographically. Even in children less than one year of age in which an open fontanelle still exists and in whom a neuro-paediatric clarification of internal hydrocepalus is made, in spite of opththalmoscopically inconspicuous findings for the papilla an echography is indispensable for the evaluation of the sub-arachnoidal space. Here, the early recognition of a dilatation of the retro-bulbar sub-arachnoidal space can possibly prevent the occurrence of a consecutive optic atrophy. At the present time, the data available do not allow the recommendation of an upper age limit for an echographic examination.


Asunto(s)
Fontanelas Craneales/diagnóstico por imagen , Ecocardiografía/métodos , Papiledema/diagnóstico por imagen , Espacio Subaracnoideo/diagnóstico por imagen , Fontanelas Craneales/patología , Dilatación Patológica/diagnóstico por imagen , Humanos , Lactante
5.
Acta Neurochir Suppl ; 106: 247-50, 2010.
Artículo en Inglés | MEDLINE | ID: mdl-19812958

RESUMEN

High S100B serum levels are considered to reflect brain injury severity. However, the dynamics of S100B passage from the cerebral compartment into the blood remain unclear. We examined the temporal profile of S100B release into the cerebrospinal fluid (CSF) and blood in acute brain injury.In patients treated with ventricular drainage (subarachnoid hemorrhage, SAH, n = 23; traumatic brain injury, TBI, n = 19), we measured S100B levels in the serum and CSF. The Glasgow Coma Score (GCS) was assessed daily. Statistical analysis was performed by the Mann-Whitney rank sum test for group differences and by the Pearson correlation.In normal controls (n = 6), S100B levels in the serum (0.05 +/- 0.01 microg/L) comprised around 10% of the CSF concentration (0.66 +/- 0.08 microg/L). Following brain injury, S100B levels were significantly increased in the serum (p < 0.05 in SAH day 2-5, TBI day 1-8) and excessively increased in the CSF (p < 0.05 in SAH and TBI day 1-10). For the individual patient, there was no consistent correlation between S100B levels in serum or CSF and GCS. We therefore calculated the ratio of S100B serum/CSF. Following brain injury, the S100B passage from the CSF to the blood was significantly impaired. Further, higher ratios were correlated with better neurological function (p = 0.002).Because stimulated active S100B release may serve as a repair mechanism, a higher S100B serum/CSF ratio may contribute to neurological recovery.


Asunto(s)
Lesiones Encefálicas/sangre , Lesiones Encefálicas/líquido cefalorraquídeo , Factores de Crecimiento Nervioso/sangre , Factores de Crecimiento Nervioso/líquido cefalorraquídeo , Proteínas S100/sangre , Proteínas S100/líquido cefalorraquídeo , Adulto , Anciano , Estudios de Casos y Controles , Femenino , Escala de Coma de Glasgow , Humanos , Masculino , Persona de Mediana Edad , Subunidad beta de la Proteína de Unión al Calcio S100 , Estadística como Asunto , Estadísticas no Paramétricas , Hemorragia Subaracnoidea/sangre , Hemorragia Subaracnoidea/líquido cefalorraquídeo , Factores de Tiempo , Adulto Joven
7.
Oncogene ; 36(40): 5593-5608, 2017 10 05.
Artículo en Inglés | MEDLINE | ID: mdl-28553953

RESUMEN

Activating transcription factor 4 (ATF4) is a critical mediator of metabolic and oxidative homeostasis and cell survival. ATF4 is elevated in response to diverse microenvironmental stresses, including starvation, ER stress damages and exposure to toxic factors. Here we show that ATF4 expression fosters the malignancy of primary brain tumors (WHO grade III and IV gliomas) and increases proliferation and tumor angiogenesis. Hence, ATF4 expression promotes cell migration and anchorage-independent cell growth, whereas siRNA-mediated knockdown of ATF4 attenuates these features of malignancy in human gliomas. Further experiments revealed that ATF4-dependent tumor promoting effects are mediated by transcriptional targeting the glutamate antiporter xCT/SCL7A11 (also known as system Xc-). Thus, xCT is elevated as a consequence of ATF4 activation. We further found evidence that ATF4-induced proliferation can be attenuated by pharmacological or genetic xCT inhibition and ferroptosis inducers such as sorafenib, erastin and GPx4 inhibitor RSL3. Further, fostered xCT expression promotes cell survival and growth in ATF4 knockdown cells. Moreover, increased xCT levels ameliorate sorafenib and erastin-induced ferroptosis. Conversely, ATF4 knockdown renders cells susceptible for erastin, sorafenib and RSL3-induced ferroptosis. We further identified that ATF4 promotes tumor-mediated neuronal cell death which can be alleviated by xCT inhibition. Moreover, elevated ATF4 expression in gliomas promotes tumor angiogenesis. Noteworthy, ATF4-induced angiogenesis could be diminished by ferroptosis inducers erastin and by GPx4 inhibitor RSL3. Our data provide proof-of-principle evidence that ATF4 fosters proliferation and induces a toxic microenvironmental niche. Furthermore, ATF4 increases tumor angiogenesis and shapes the vascular architecture in a xCT-dependent manner. Thus, inhibition of ATF4 is a valid target for diminishing tumor growth and vasculature via sensitizing tumor cells for ferroptosis.


Asunto(s)
Factor de Transcripción Activador 4/metabolismo , Sistema de Transporte de Aminoácidos y+/metabolismo , Neoplasias Encefálicas/irrigación sanguínea , Muerte Celular , Glioma/irrigación sanguínea , Neovascularización Patológica , Factor de Transcripción Activador 4/genética , Sistema de Transporte de Aminoácidos y+/genética , Neoplasias Encefálicas/metabolismo , Línea Celular Tumoral , Proliferación Celular , Regulación Neoplásica de la Expresión Génica , Glioma/metabolismo , Ácido Glutámico/metabolismo , Humanos , Hierro/metabolismo , Neuronas/patología
8.
Oncogene ; 35(48): 6246-6261, 2016 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-27157615

RESUMEN

Microglial cells in the brain tumor microenvironment are associated with enhanced glioma malignancy. They persist in an immunosuppressive M2 state at the peritumoral site and promote the growth of gliomas. Here, we investigated the underlying factors contributing to the abolished immune surveillance. We show that brain tumors escape pro-inflammatory M1 conversion of microglia via CD74 activation through the secretion of the cytokine macrophage migration inhibitory factor (MIF), which results in a M2 shift of microglial cells. Interruption of this glioma-microglial interaction through an antibody-neutralizing approach or small interfering RNA (siRNA)-mediated inhibition prolongs survival time in glioma-implanted mice by reinstating the microglial pro-inflammatory M1 function. We show that MIF-CD74 signaling inhibits interferon (IFN)-γ secretion in microglia through phosphorylation of microglial ERK1/2 (extracellular signal-regulated protein kinases 1 and 2). The inhibition of MIF signaling or its receptor CD74 promotes IFN-γ release and amplifies tumor death either through pharmacological inhibition or through siRNA-mediated knockdown. The reinstated IFN-γ secretion leads both to direct inhibition of glioma growth as well as inducing a M2 to M1 shift in glioma-associated microglia. Our data reveal that interference with the MIF signaling pathway represents a viable therapeutic option for the restoration of IFN-γ-driven immune surveillance.


Asunto(s)
Antígenos de Diferenciación de Linfocitos B/metabolismo , Transformación Celular Neoplásica/metabolismo , Glioma/metabolismo , Antígenos de Histocompatibilidad Clase II/metabolismo , Factores Inhibidores de la Migración de Macrófagos/metabolismo , Microglía/metabolismo , Transducción de Señal , Animales , Comunicación Autocrina , Línea Celular Tumoral , Transformación Celular Neoplásica/genética , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Técnicas de Silenciamiento del Gen , Glioma/diagnóstico por imagen , Glioma/genética , Glioma/patología , Xenoinjertos , Humanos , Interferón gamma/metabolismo , Ratones , Microglía/inmunología , Modelos Biológicos , Fagocitosis , Ratas
9.
Brain Res ; 864(1): 44-51, 2000 May 02.
Artículo en Inglés | MEDLINE | ID: mdl-10793185

RESUMEN

We used the fluorescent dye Fluoro-Jade, capable of selectively staining degenerating neurons and their processes, in order to analyze degenerative effects of transecting the hippocampus from its main input, the entorhinal cortex in vivo and in organotypical hippocampal slice culture. Degenerating fibers stained with Fluoro-Jade were present as early as 1 day postlesion in the outer molecular layer of the dentate gyrus and could be detected up to 30 days postlesion. However, the intensity of the Fluoro-Jade staining in the outer molecular layer faded from postlesional day 20 onward. Punctate staining, various cells and neural processes became visible in this area suggesting that degenerating processes were phagocytosed by microglial cells or astrocytes. We conclude that Fluoro-Jade is an early and sensitive marker for studying degenerating neurites in the hippocampal system.


Asunto(s)
Desnervación/efectos adversos , Corteza Entorrinal/patología , Hipocampo/patología , Degeneración Nerviosa/patología , Vías Nerviosas/patología , Animales , Corteza Entorrinal/fisiopatología , Colorantes Fluorescentes , Hipocampo/fisiopatología , Masculino , Degeneración Nerviosa/fisiopatología , Vías Nerviosas/fisiopatología , Ratas , Ratas Wistar , Factores de Tiempo
10.
Oncogene ; 30(1): 43-53, 2011 Jan 06.
Artículo en Inglés | MEDLINE | ID: mdl-20802520

RESUMEN

Malignant glioma represents one of the most aggressive and lethal human neoplasias. A hallmark of gliomas is their rapid proliferation and destruction of vital brain tissue, a process in which excessive glutamate release by glioma cells takes center stage. Pharmacologic antagonism with glutamate signaling through ionotropic glutamate receptors attenuates glioma progression in vivo, indicating that glutamate release by glioma cells is a prerequisite for rapid glioma growth. Glutamate has been suggested to promote glioma cell proliferation in an autocrine or paracrine manner, in particular by activation of the (RS)-α-amino-3-hydroxy-5-methylisoxazole-4-propionic acid hydrate (AMPA) subtype of glutamate receptors. Here, we dissect the effects of glutamate secretion on glioma progression. Glioma cells release glutamate through the amino-acid antiporter system X(c)(-), a process that is mechanistically linked with cystine incorporation. We show that disrupting glutamate secretion by interfering with the system X(c)(-) activity attenuates glioma cell proliferation solely cystine dependently, whereas glutamate itself does not augment glioma cell growth in vitro. Neither AMPA receptor agonism nor antagonism affects glioma growth in vitro. On a molecular level, AMPA insensitivity is concordant with a pronounced transcriptional downregulation of AMPA receptor subunits or overexpression of the fully edited GluR2 subunit, both of which block receptor activity. Strikingly, AMPA receptor inhibition in tumor-implanted brain slices resulted in markedly reduced tumor progression associated with alleviated neuronal cell death, suggesting that the ability of glutamate to promote glioma progression strictly requires the tumor microenvironment. Concerning a potential pharmacotherapy, targeting system X(c)(-) activity disrupts two major pathophysiological properties of glioma cells, that is, the induction of excitotoxic neuronal cell death and incorporation of cystine required for rapid proliferation.


Asunto(s)
Neoplasias Encefálicas/metabolismo , Neoplasias Encefálicas/patología , Cistina/metabolismo , Glioma/metabolismo , Glioma/patología , Ácido Glutámico/metabolismo , Animales , Neoplasias Encefálicas/genética , Procesos de Crecimiento Celular/fisiología , Línea Celular Tumoral , Cistina/genética , Cistina/farmacología , Regulación hacia Abajo , Glioma/genética , Ácido Glutámico/genética , Ácido Glutámico/farmacología , Humanos , Ratones , Ratas , Receptores AMPA/antagonistas & inhibidores , Receptores AMPA/genética , Receptores AMPA/metabolismo , Transducción de Señal , Transfección
11.
Rofo ; 182(4): 322-6, 2010 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-19862651

RESUMEN

PURPOSE: To evaluate the potential of MSCT and a novel AmicroCT system to assess the volume of malignant brain tumors in rats compared to histology. MATERIALS AND METHODS: Fourteen rats underwent stereotactic implantation of GFP-marked F 98-glioma cells. On day 10 after implantation, animals received double-dose contrast-enhanced AmicroCT and MSCT imaging using Iomeprol. MSCT- and AmicroCT-derived tumor volumes were calculated and compared to histology (fluorescence staining) as the gold standard. RESULTS: There was good correlation between the AmicroCT-derived tumor volume (69 A+ or - 23 mm(3)) and histology (81 A + or - 14 mm(3); p > 0.14). MSCT, however, showed significantly smaller tumor volumes (55 A + or - 25 mm(3)) compared to histology (p < 0.01) but was able to detect the tumors in all animals. CONCLUSION: AmicroCT allows in vivo imaging of the contrast-enhancing parts of experimental gliomas with high correlation to histology. Although MSCT is less suitable for assessing exact tumor volume, this method reliably detects tumors in rats. Due to the high spatial resolution, AmicroCT-systems could play an important role for fusion imaging, e. g. to assess experimental brain gliomas with multimodal AmicroCT/PET- or AmicroCT/MRI-fusion images.


Asunto(s)
Enfermedades de los Ganglios Basales/diagnóstico por imagen , Neoplasias Encefálicas/diagnóstico por imagen , Glioma/diagnóstico por imagen , Procesamiento de Imagen Asistido por Computador/métodos , Tomografía Computarizada de Haz Cónico Espiral/métodos , Microtomografía por Rayos X/métodos , Animales , Ganglios Basales/diagnóstico por imagen , Ganglios Basales/patología , Enfermedades de los Ganglios Basales/patología , Neoplasias Encefálicas/patología , Línea Celular Tumoral , Medios de Contraste , Glioma/patología , Yopamidol/análogos & derivados , Microscopía Fluorescente , Trasplante de Neoplasias , Ratas , Ratas Endogámicas F344 , Sensibilidad y Especificidad , Estadística como Asunto
13.
Glia ; 31(3): 262-6, 2000 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-10941152

RESUMEN

Most CNS pathologies are accompanied by the occurrence of activated, phagocytic microglial cells. We intended to investigate whether (1) isolated microglial cells removed from the CNS cytokine network sustain their capacity to acquire an activated phenotype when challenged with cellular or noncellular debris; and (2) different substrates lead to different patterns of microglial activation. It was observed that although removed from their usual surroundings microglial cells preserve their ability to transform to an amoeboid morphology, form multinucleated giant cells, and enhance their expression of MHC class II when exposed to membranes of neuronal or glial origin. Furthermore, cellular substrates derived from primary hippocampal neuronal cultures, neuroblastic cells (B50), or glial cells were all able to induce similar morphological changes and enhanced expression of MHC class II. In contrast, phagocytosis of Latex beads induced an amoeboid morphology but no increase in the expression of immunologically relevant molecules. Interferon-beta (IFN-beta), a substance clinically used in the treatment of the relapsing-remitting form of multiple sclerosis, was shown to inhibit the phagocytosis-induced upregulation of MHC-class II. In summary, phagocytic microglial cells are independent from the CNS cytokine network in their transition from a resting to an activated phenotype; and different cellular substrates, regardless whether they are of neuronal, glial, or even malignant origin, result in similar morphological and functional changes.


Asunto(s)
Regulación de la Expresión Génica/fisiología , Genes MHC Clase II/genética , Células Gigantes/patología , Microglía/patología , Neuroglía/patología , Neuronas/patología , Regulación hacia Arriba/fisiología , Animales , Animales Recién Nacidos , Células Cultivadas , Regulación de la Expresión Génica/efectos de los fármacos , Genes MHC Clase II/efectos de los fármacos , Células Gigantes/metabolismo , Microglía/efectos de los fármacos , Microglía/metabolismo , Ratas , Ratas Wistar , Regulación hacia Arriba/efectos de los fármacos
14.
Hum Mol Genet ; 12(19): 2481-9, 2003 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-12915451

RESUMEN

Proximal spinal muscular atrophy (SMA) is a common neuromuscular disorder causing infant death in half of all patients. Homozygous absence of the survival motor neuron gene (SMN1) is the primary cause of SMA, while SMA severity is mainly determined by the number of SMN2 copies. One SMN2 copy produces only about 10% of full-length protein identical to SMN1, whereas the majority of SMN2 transcripts is aberrantly spliced due to a silent mutation within an exonic splicing enhancer in exon 7. However, correct splicing can be restored by over-expression of the SR-like splicing factor Htra2-beta 1. We show that in fibroblast cultures derived from SMA patients treated with therapeutic doses (0.5-500 microM) of valproic acid (VPA), the level of full-length SMN2 mRNA/protein increased 2- to 4-fold. Importantly, this up-regulation of SMN could be most likely attributed to increased levels of Htra2-beta 1 which facilitates the correct splicing of SMN2 RNA as well as to an SMN gene transcription activation. Especially at low VPA concentrations, the restored SMN level depended on the number of SMN2 copies. Moreover, VPA was able to increase SMN protein levels through transcription activation in organotypic hippocampal brain slices from rats. Finally, VPA also increased the expression of further SR proteins, which may have important implications for other disorders affected by alternative splicing. Since VPA is a drug highly successfully used in long-term epilepsy therapy, our findings open the exciting perspective for a first causal therapy of an inherited disease by elevating the SMN2 transcription level and restoring its correct splicing.


Asunto(s)
Fibroblastos/metabolismo , GABAérgicos/uso terapéutico , Atrofia Muscular Espinal/genética , Proteínas del Tejido Nervioso/genética , Ácido Valproico/uso terapéutico , Animales , Anticuerpos Monoclonales/metabolismo , Western Blotting , Células Cultivadas , Relación Dosis-Respuesta a Droga , Exones , Fibroblastos/efectos de los fármacos , Eliminación de Gen , Regulación de la Expresión Génica/efectos de los fármacos , Terapia Genética , Hipocampo/efectos de los fármacos , Hipocampo/metabolismo , Humanos , Atrofia Muscular Espinal/metabolismo , Atrofia Muscular Espinal/terapia , Proteínas del Tejido Nervioso/clasificación , Técnicas de Cultivo de Órganos , Empalme del ARN , ARN Mensajero/análisis , Ratas , Ratas Wistar , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Factores de Tiempo , Transcripción Genética/efectos de los fármacos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA