Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
1.
Neuromodulation ; 21(2): 160-167, 2018 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-28960670

RESUMEN

INTRODUCTION AND OBJECTIVES: Cognitive impairment is a significant comorbidity of temporal lobe epilepsy that is associated with extensive hippocampal cell loss. Deep brain stimulation (DBS) of the anterior thalamic nucleus (ANT) has been used for the treatment of refractory partial seizures. In the pilocarpine model of epilepsy, ANT DBS applied during status epilepticus (SE) reduces hippocampal inflammation and apoptosis. When given to chronic epileptic animals it reduces hippocampal excitability and seizure frequency. Here, we tested whether ANT DBS delivered during SE and the silent phase of the pilocarpine model would reduce cognitive impairment when animals became chronically epileptic. MATERIALS AND METHODS: SE was induced by a systemic pilocarpine injection (320 mg/kg). Immediately after SE onset, rats were assigned to receive DBS during the first six hours of SE (n = 8; DBSa group) or during SE + the silent period (i.e., 6 h/day until the animals developed the first spontaneous recurrent seizure; n = 10; DBSs group). Four months following SE, animals underwent water maze testing and histological evaluation. Nonstimulated chronic epileptic animals (n = 13; PCTL group) and age-matched naïve rats (n = 11, CTL group) were used as controls. Results were analyzed by repeated-measures analyses of variance (RM_ANOVA) and one-way ANOVAs, followed by Newman-Keuls post hoc tests. RESULTS: Although all groups learned the spatial task, epileptic animals with or without DBS spent significantly less time in the platform quadrant, denoting a spatial memory deficit (p < 0.02). Despite these negative behavioral results, we found that animals given DBS had a significantly higher number of cells in the CA1 region and dentate gyrus. Mossy fiber sprouting was similar among all epileptic groups. CONCLUSIONS: Despite lesser hippocampal neuronal loss, ANT DBS delivered either during SE or during SE and the silent phase of the pilocarpine model did not mitigate memory deficits in chronic epileptic rats.


Asunto(s)
Núcleos Talámicos Anteriores/fisiología , Estimulación Encefálica Profunda/métodos , Epilepsia del Lóbulo Temporal/terapia , Aprendizaje Espacial/fisiología , Animales , Modelos Animales de Enfermedad , Epilepsia del Lóbulo Temporal/inducido químicamente , Hipocampo/metabolismo , Hipocampo/patología , Estudios Longitudinales , Masculino , Aprendizaje por Laberinto/efectos de los fármacos , Agonistas Muscarínicos/toxicidad , Pilocarpina/toxicidad , Distribución Aleatoria , Ratas , Ratas Wistar , Aprendizaje Espacial/efectos de los fármacos
2.
Acta Neurochir Suppl ; 122: 329-33, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27165931

RESUMEN

Intracranial pressure (ICP) is a major neurological parameter in animals and humans. ICP is a function of the relationship between the contents of the cranium (brain parenchyma, cerebrospinal fluid, and blood) and the volume of the skull. Increased ICP can cause serious physiological effects or even death in patients who do not quickly receive proper care, which includes ICP monitoring. Epilepsies are a set of central nervous system disorders resulting from abnormal and excessive neuronal discharges, usually associated with hypersynchronism and/or hyperexcitability. Temporal lobe epilepsy (TLE) is one of the most common forms of epilepsy and is also refractory to medication. ICP characteristics of subjects with epilepsy have not been elucidated because there are few studies associating these two important neurological factors. In this work, an invasive (ICPi) and the new minimally invasive (ICPmi) methods were used to evaluate ICP features in rats with chronic epilepsy, induced by the experimental model of pilocarpine, capable of generating the main features of human TLE in these animals.


Asunto(s)
Epilepsia del Lóbulo Temporal/fisiopatología , Hipocampo/diagnóstico por imagen , Presión Intracraneal/fisiología , Animales , Anticonvulsivantes/uso terapéutico , Encéfalo/diagnóstico por imagen , Encéfalo/patología , Enfermedad Crónica , Modelos Animales de Enfermedad , Epilepsia/inducido químicamente , Epilepsia/tratamiento farmacológico , Epilepsia/patología , Epilepsia/fisiopatología , Epilepsia del Lóbulo Temporal/inducido químicamente , Epilepsia del Lóbulo Temporal/tratamiento farmacológico , Epilepsia del Lóbulo Temporal/patología , Hipocampo/patología , Imagen por Resonancia Magnética , Masculino , Agonistas Muscarínicos/toxicidad , Tamaño de los Órganos , Pilocarpina/toxicidad , Ratas , Ratas Wistar , Tiopental/uso terapéutico
3.
Brain Res Bull ; 147: 133-139, 2019 04.
Artículo en Inglés | MEDLINE | ID: mdl-30658130

RESUMEN

BACKGROUND: Deep brain stimulation (DBS) refers to the delivery of electric current to specific deep brain structures through implanted electrodes. Recently approved for use in United States, DBS to the anterior nucleus of thalamus (ANT) is a safe and effective alternative treatment for medically refractory seizures. Despite the anti-seizure effects of ANT DBS, preclinical and clinical studies have failed to demonstrate it actions at a whole brain level. OBJECTIVE: Here, we used a magnetic resonance imaging (MRI)-based approach in healthy adult rats to investigate the effects of ANT DBS through the circuit of Papez, which has central role in the generation and propagation of limbic seizures, in temporal lobe epilepsy (TLE). METHODS: After ANT electrode implantation and recovery, ANT DBS and SHAM (sham animals had electrodes implanted but were not stimulated) rats received one single injection of the contrast enhancer, manganese chloride (60 mg/kg, ip). Twelve hours after, rats underwent the baseline scan using the MEMRI (Manganese-Enhanced Magnetic Resonance Imaging) technique. We used the same MEMRI and parvalbumin sequence to follow the DBS delivered during 1 h (130 Hz and 200 µA). Perfusion was followed by subsequent c-Fos and parvalbumin immunostaining of brain sections. RESULTS: Acute unilateral ANT DBS significantly reduced the overall manganese uptake and consequently, the MEMRI contrast in the circuit of Papez. Additionally, c-Fos expression was bilaterally increased in the cingulate cortex and posterior hypothalamus, areas directly connected to ANT, as well as in amygdala and subiculum, within the limbic circuitry. CONCLUSION: Our data indicate that MEMRI can be used to detect whole-brain responses to DBS, as the high frequency stimulation parameters used here caused a significant reduction of cell activity in the circuit of Papez that might help to explain the antiepileptic effects of ANT DBS.


Asunto(s)
Núcleos Talámicos Anteriores/metabolismo , Convulsiones/terapia , Amígdala del Cerebelo/metabolismo , Animales , Núcleo Celular/metabolismo , Estimulación Encefálica Profunda/métodos , Electrodos Implantados , Epilepsia/metabolismo , Epilepsia/terapia , Epilepsia del Lóbulo Temporal/terapia , Hipocampo/metabolismo , Sistema Límbico , Imagen por Resonancia Magnética/métodos , Masculino , Proteínas Proto-Oncogénicas c-fos/metabolismo , Ratas , Ratas Wistar , Convulsiones/metabolismo , Tálamo/metabolismo
4.
Sci Rep ; 9(1): 9973, 2019 07 10.
Artículo en Inglés | MEDLINE | ID: mdl-31292491

RESUMEN

EPNs comprise a heterogeneous group of neuroepithelial tumors, accounting for about 10% of all intracranial tumors in children and up to 30% of brain tumors in those younger than 3 years. Actually, the pattern therapy for low-grade EPNs includes complete surgical resection followed by radiation therapy. Total surgical excision is often not possible due to tumor location. The aim of this study was to evaluate, for the first time, the anti-tumor activity of Amblyomin-X in 4 primary cultures derived from pediatric anaplastic posterior fossa EPN, Group A (anaplastic, WHO grade III) and one primary culture of a high grade neuroepithelial tumor with MN1 alteration, which was initially misdiagnosed as EPN: i) by in vitro assays: comparisons of temozolomide and cisplatin; ii) by intracranial xenograft model. Amblyomin-X was able to induce cell death in EPN cells in a more significant percentage compared to cisplatin. The cytotoxic effects of Amblyomin-X were not detected on hFSCs used as control, as opposed to cisplatin-treatment, which promoted a substantial effect in the hAFSCs viability. TEM analysis showed ultrastructural alterations related to the process of cell death: mitochondrial degeneration, autophagosomes and aggregate-like structures. MRI and histopathological analyzes demonstrated significant tumor mass regression. Our results suggest that Amblyomin-X has a selective effect on tumor cells by inducing apoptotic cell death and may be a therapeutic option for Group AEPNs.


Asunto(s)
Antineoplásicos/farmacología , Ependimoma/tratamiento farmacológico , Proteínas y Péptidos Salivales/farmacología , Adulto , Animales , Apoptosis/efectos de los fármacos , Proteínas de Artrópodos , Niño , Preescolar , Femenino , Células Madre Fetales/citología , Células Madre Fetales/metabolismo , Humanos , Masculino , Ratas Wistar , Ensayos Antitumor por Modelo de Xenoinjerto/métodos
5.
Cell Transplant ; 28(9-10): 1306-1320, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31161782

RESUMEN

Amniotic fluid has been investigated as new cell source for stem cells in the development of future cell-based transplantation. This study reports isolation of viable human amniotic fluid-derived stem cells, labeled with multimodal iron oxide nanoparticles, and its effect on focal cerebral ischemia-reperfusion injury in Wistar rats. Middle cerebral artery occlusion of 60 min followed by reperfusion for 1 h, 6 h, and 24 h was employed in the present study to produce ischemia and reperfusion-induced cerebral injury in rats. Tests were employed to assess the functional outcome of the sensorimotor center activity in the brain, through a set of modified neurological severity scores used to assess motor and exploratory capacity 24 h, 14, and 28 days after receiving cellular therapy via tail vein. In our animal model of stroke, transplanted cells migrated to the ischemic focus, infarct volume decreased, and motor deficits improved. Therefore, we concluded that these cells appear to have beneficial effects on the ischemic brain, possibly based on their ability to enhance endogenous repair mechanisms.


Asunto(s)
Líquido Amniótico/metabolismo , Conducta Animal , Isquemia Encefálica , Trasplante de Células Madre , Células Madre/metabolismo , Accidente Cerebrovascular , Adulto , Animales , Isquemia Encefálica/metabolismo , Isquemia Encefálica/patología , Isquemia Encefálica/fisiopatología , Isquemia Encefálica/terapia , Modelos Animales de Enfermedad , Femenino , Xenoinjertos , Humanos , Embarazo , Ratas , Ratas Wistar , Células Madre/patología , Accidente Cerebrovascular/metabolismo , Accidente Cerebrovascular/patología , Accidente Cerebrovascular/fisiopatología , Accidente Cerebrovascular/terapia
6.
Stem Cell Res Ther ; 9(1): 310, 2018 11 09.
Artículo en Inglés | MEDLINE | ID: mdl-30413179

RESUMEN

BACKGROUND: Previous studies have demonstrated remarkable tropism of mesenchymal stem cells (MSCs) toward malignant gliomas, making these cells a potential vehicle for delivery of therapeutic agents to disseminated glioblastoma (GBM) cells. However, the potential contribution of MSCs to tumor progression is a matter of concern. It has been suggested that CD133+ GBM stem cells secrete a variety of chemokines, including monocytes chemoattractant protein-1 (MCP-1/CCL2) and stromal cell-derived factor-1(SDF-1/CXCL12), which could act in this tropism. However, the role in the modulation of this tropism of the subpopulation of CD133+ cells, which initiate GBM and the mechanisms underlying the tropism of MSCs to CD133+ GBM cells and their effects on tumor development, remains poorly defined. METHODS/RESULTS: We found that isolated and cultured MSCs (human umbilical cord blood MSCs) express CCR2 and CXCR4, the respective receptors for MCP-1/CCL2 and SDF-1/CXCL12, and demonstrated, in vitro, that MCP-1/CCL2 and SDF-1/CXC12, secreted by CD133+ GBM cells from primary cell cultures, induce the migration of MSCs. In addition, we confirmed that after in vivo GBM tumor establishment, by stereotaxic implantation of the CD133+ GBM cells labeled with Qdots (705 nm), MSCs labeled with multimodal iron oxide nanoparticles (MION) conjugated to rhodamine-B (Rh-B) (MION-Rh), infused by caudal vein, were able to cross the blood-brain barrier of the animal and migrate to the tumor region. Evaluation GBM tumors histology showed that groups that received MSC demonstrated tumor development, glial invasiveness, and detection of a high number of cycling cells. CONCLUSIONS: Therefore, in this study, we validated the chemotactic effect of MCP-1/CCL2 and SDF-1/CXCL12 in mediating the migration of MSCs toward CD133+ GBM cells. However, we observed that, after infiltrating the tumor, MSCs promote tumor growth in vivo probably by release of exosomes. Thus, the use of these cells as a therapeutic carrier strategy to target GBM cells must be approached with caution.


Asunto(s)
Antígeno AC133/metabolismo , Neoplasias Encefálicas/patología , Glioblastoma/patología , Células Madre Mesenquimatosas/metabolismo , Células Madre Neoplásicas/patología , Tropismo , Animales , Neoplasias Encefálicas/ultraestructura , Carcinogénesis/metabolismo , Carcinogénesis/patología , Ensayos de Migración Celular , Proliferación Celular , Separación Celular , Quimiocinas/metabolismo , Glioblastoma/ultraestructura , Humanos , Inmunofenotipificación , Masculino , Células Madre Mesenquimatosas/ultraestructura , Modelos Biológicos , Células Madre Neoplásicas/ultraestructura , Puntos Cuánticos/metabolismo , Ratas Wistar , Receptores de Quimiocina/metabolismo , Esferoides Celulares/patología , Células Tumorales Cultivadas
7.
Oncotarget ; 9(31): 21731-21743, 2018 Apr 24.
Artículo en Inglés | MEDLINE | ID: mdl-29774098

RESUMEN

BACKGROUND: Ependymoma (EPN), the third most common pediatric brain tumor, is a central nervous system (CNS) malignancy originating from the walls of the ventricular system. Surgical resection followed by radiation therapy has been the primary treatment for most pediatric intracranial EPNs. Despite numerous studies into the prognostic value of histological classification, the extent of surgical resection and adjuvant radiotherapy, there have been relatively few studies into the molecular and cellular biology of EPNs. RESULTS: We elucidated the ultrastructure of the cultured EPN cells and characterized their profile of immunophenotypic pluripotency markers (CD133, CD90, SSEA-3, CXCR4). We established an experimental EPN model by the intracerebroventricular infusion of EPN cells labeled with multimodal iron oxide nanoparticles (MION), thereby generating a tumor and providing a clinically relevant animal model. MRI analysis was shown to be a valuable tool when combined with effective MION labeling techniques to accompany EPN growth. CONCLUSIONS: We demonstrated that GFAP/CD133+CD90+/CD44+ EPN cells maintained key histopathological and growth characteristics of the original patient tumor. The characterization of EPN cells and the experimental model could facilitate biological studies and preclinical drug screening for pediatric EPNs. METHODS: In this work, we established notoriously challenging primary cell culture of anaplastic EPNs (WHO grade III) localized in the posterior fossa (PF), using EPNs obtained from 1 to 10-year-old patients (n = 07), and then characterized their immunophenotype and ultrastructure to finally develop a xenograft model.

8.
Behav Brain Res ; 296: 331-338, 2016 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-26416672

RESUMEN

Neonates that suffer oxygen deprivation during birth can have long lasting cognitive deficits, such as memory and learning impairments. Hippocampus, one of the main structures that participate in memory and learning processes, is a plastic and dynamic structure that conserves during life span the property of generating new cells which can become neurons, the so-called neurogenesis. The present study investigated whether a model of rat neonatal anoxia, that causes only respiratory distress, is able to alter the hippocampal volume, the neurogenesis rate and has functional implications in adult life. MRI analysis revealed significant hippocampal volume decrease in adult rats who had experienced neonatal anoxia compared to control animals for rostral, caudal and total hippocampus. In addition, these animals also had 55.7% decrease of double-labelled cells to BrdU and NeuN, reflecting a decrease in neurogenesis rate. Finally, behavioral analysis indicated that neonatal anoxia resulted in disruption of spatial working memory, similar to human condition, accompanied by an anxiogenic effect. The observed behavioral alterations caused by oxygen deprivation at birth might represent an outcome of the decreased hippocampal neurogenesis and volume, evidenced by immunohistochemistry and MRI analysis. Therefore, based on current findings we propose this model as suitable to explore new therapeutic approaches.


Asunto(s)
Ansiedad/etiología , Conducta Animal/fisiología , Hipocampo/patología , Hipoxia/complicaciones , Trastornos de la Memoria/etiología , Memoria a Corto Plazo/fisiología , Neurogénesis/fisiología , Memoria Espacial/fisiología , Factores de Edad , Animales , Animales Recién Nacidos , Ansiedad/fisiopatología , Modelos Animales de Enfermedad , Imagen por Resonancia Magnética , Masculino , Trastornos de la Memoria/fisiopatología , Ratas , Ratas Wistar
9.
Front Neurol ; 6: 161, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26217304

RESUMEN

Magnetic resonance imaging (MRI) is an excellent non-invasive tool to investigate biological systems. The administration of the paramagnetic divalent ion manganese (Mn(2+)) enhances MRI contrast in vivo. Due to similarities between Mn(2+) and calcium (Ca(2+)), the premise of manganese-enhanced MRI (MEMRI) is that the former may enter neurons and other excitable cells through voltage-gated Ca(2+) channels. As such, MEMRI has been used to trace neuronal pathways, define morphological boundaries, and study connectivity in morphological and functional imaging studies. In this article, we provide a brief overview of MEMRI and discuss recently published data to illustrate the usefulness of this method, particularly in animal models.

10.
Epilepsy Res ; 108(4): 644-52, 2014 May.
Artículo en Inglés | MEDLINE | ID: mdl-24630048

RESUMEN

Manganese-enhanced MRI (MEMRI) has been considered a surrogate marker of Ca(+2) influx into activated cells and tracer of neuronal active circuits. However, the induction of status epilepticus (SE) by kainic acid does not result in hippocampal MEMRI hypersignal, in spite of its high cell activity. Similarly, short durations of status (5 or 15min) induced by pilocarpine did not alter the hippocampal MEMRI, while 30 min of SE even reduced MEMRI signal Thus, this study was designed to investigate possible explanations for the absence or decrease of MEMRI signal after short periods of SE. We analyzed hippocampal caspase-3 activation (to evaluate apoptosis), T2 relaxometry (tissue water content) and aquaporin 4 expression (water-channel protein) of rats subjected to short periods of pilocarpine-induced SE. For the time periods studied here, apoptotic cell death did not contribute to the decrease of the hippocampal MEMRI signal. However, T2 relaxation was higher in the group of animals subjected to 30min of SE than in the other SE or control groups. This result is consistent with higher AQP-4 expression during the same time period. Based on apoptosis and tissue water content analysis, the low hippocampal MEMRI signal 30min after SE can potentially be attributed to local edema rather than to cell death.


Asunto(s)
Caspasa 3/metabolismo , Hipocampo/patología , Estado Epiléptico/patología , Animales , Acuaporinas/metabolismo , Hipocampo/metabolismo , Imagen por Resonancia Magnética/métodos , Masculino , Pilocarpina , Proteínas Proto-Oncogénicas c-fos/metabolismo , Ratas , Estado Epiléptico/inducido químicamente , Estado Epiléptico/metabolismo
11.
Einstein (Sao Paulo) ; 10(2): 247-52, 2012.
Artículo en Inglés, Portugués | MEDLINE | ID: mdl-23052465

RESUMEN

Magnetic resonance images are useful in the study of experimental models of temporal lobe epilepsy. The manganese-enhanced MRI (MEMRI) technique is of interest since it combines the effects caused by manganese on the increased contrast in activated cell populations, when competing with calcium in synaptic transmission. Thus, the purpose of this study was to investigate the temporal evolution of the contrast related to manganese in the acute phase of temporal lobe epilepsy induced by systemic pilocarpine and compare it to the expression of the c-Fos protein. During this phase, the intensity of the MEMRI signal was analyzed at three different time points (5, 15 or 30 minutes) after the onset of status epilepticus (SE). The group that was maintained in status epilepticus for 30 minutes showed a decrease in intensity of the signal in CA1 and the dentate gyrus (DG). There were no differences between the control group and the other groups treated with pilocarpine. The expression of the protein, c-Fos, in the same animals showed that even in the short-duration status epilepticus (5 minutes), there was already maximal cellular activation in subregions of the hippocampus (DG, CA1 and CA3). Under the experimental conditions tested, our data suggest that the MEMRI signal was not sensitive for the identification of detectable variations of cell activation in the acute phase of the pilocarpine model. Our findings are not consistent with the idea that manganese contrast reflects primarily alterations in cellular activity during SE when other signal-modifying elements can act.


Asunto(s)
Medios de Contraste , Epilepsia del Lóbulo Temporal/patología , Manganeso , Proteínas Proto-Oncogénicas c-fos/análisis , Enfermedad Aguda , Animales , Giro Dentado/patología , Modelos Animales de Enfermedad , Epilepsia del Lóbulo Temporal/inducido químicamente , Aumento de la Imagen/métodos , Interpretación de Imagen Asistida por Computador , Imagen por Resonancia Magnética , Masculino , Pilocarpina , Ratas , Ratas Wistar
12.
Einstein (Sao Paulo) ; 10(2): 164-70, 2012.
Artículo en Inglés, Portugués | MEDLINE | ID: mdl-23052451

RESUMEN

OBJECTIVE: The aim of the current study was to monitor the migration of superparamagnetic iron oxide nanoparticle (SPION)-labeled C6 cells, which were used to induce glioblastoma tumor growth in an animal model, over time using magnetic resonance imaging (MRI), with the goal of aiding in tumor prognosis and therapy. METHODS: Two groups of male Wistar rats were used for the tumor induction model. In the first group (n=3), the tumors were induced via the injection of SPION-labeled C6 cells. In the second group (n=3), the tumors were induced via the injection of unlabeled C6 cells. Prussian Blue staining was performed to analyze the SPION distribution within the C6 cells in vitro. Tumor-inducing C6 cells were injected into the right frontal cortex, and subsequent tumor monitoring and SPION detection were performed using T2- and T2*-weighted MRI at a 2T field strength. In addition, cancerous tissue was histologically analyzed after performing the MRI studies. RESULTS: The in vitro qualitative evaluation demonstrated adequate distribution and satisfactory cell labeling of the SPIONs. At 14 or 21 days after C6 injection, a SPION-induced T2- and T2*-weighted MRI signal reduction was observed within the lesion located in the left frontal lobe on parasagittal topography. Moreover, histological staining of the tumor tissue with Prussian Blue revealed a broad distribution of SPIONs within the C6 cells. CONCLUSION: MRI analyses exhibit potential for monitoring the tumor growth of C6 cells efficiently labeled with SPIONs.


Asunto(s)
Neoplasias Encefálicas/patología , Rastreo Celular/métodos , Compuestos Férricos , Glioma/patología , Imagen por Resonancia Magnética/métodos , Nanopartículas de Magnetita , Animales , Línea Celular Tumoral , Glioblastoma/patología , Masculino , Ratas , Ratas Wistar , Coloración y Etiquetado , Factores de Tiempo
13.
Einstein (Sao Paulo) ; 10(1): 11-5, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-23045819

RESUMEN

OBJECTIVE: The objective was to establish a pattern of tumor growth of the C6 model of glioblastoma multiform in Wistar rats via magnetic resonance imaging (MRI) for the subsequent verification of tumor volume reduction due to magnetic hyperthermia therapy. METHODS: Young male Wistar rats weighing between 250 and 300 g were used for the C6 model. After the rats were anesthetized (55 mg/ kg ketamine and 11 mg/kg xylazine), C6 lineage tumorigenic cells suspended in culture medium (10(5) cells in 10 microl) were stereotaxically injected into the right frontal cortex (bregma coordinates: 2.0 mm anteroposterior, 3.0 mm laterolateral, and 2.5 mm depth) of the rats using a Hamilton syringe. For the control group, the rats were injected with culture medium without cells. MRI scans were performed at 14, 21, and 28 d after the injection using a 2.0 T MRI scanner (Bruker BioSpec, Germany). The animals were anesthetized with 55 mg/kg ketamine and 11 mg/kg xylazine before being examined. Coronal multilayers were acquired using a standard spin echo sequence with the following parameters: repetition/echo time = 4.000 ms/67.1 ms, field of view = 3.50, matrix = 192, slice thickness = 0.4 mm, and slice separation = 0 mm. RESULTS: The MRI analysis enabled a clear visualization of the tumor mass, and it was possible to establish the tumor volume parameters on the various days that were examined. The volume at 14 d after induction was 13.7 +/- 2.5 mm3. On days 21 and 28, the tumor volumes were 31.7 +/- 6.5 mm3 and 122.1 +/- 11.8 mm3, respectively. CONCLUSION: These results demonstrated that it is possible to evaluate the C6 model tumor volume in rats, which will allow for the future implementation and verification of magnetic hyperthermia therapy.


Asunto(s)
Neoplasias Encefálicas/terapia , Glioblastoma/terapia , Hipertermia Inducida/métodos , Magnetoterapia/métodos , Imagen por Resonancia Magnética , Animales , Neoplasias Encefálicas/patología , Línea Celular Tumoral/trasplante , Lóbulo Frontal/patología , Glioblastoma/patología , Masculino , Ratas , Ratas Wistar , Carga Tumoral
15.
Einstein (Säo Paulo) ; 10(2)apr.-jun. 2012. ilus, graf
Artículo en Inglés, Portugués | LILACS | ID: lil-644893

RESUMEN

Magnetic resonance images are useful in the study of experimental models of temporal lobe epilepsy. The manganese-enhanced MRI (MEMRI) technique is of interest since it combines the effects caused by manganese on the increased contrast in activated cell populations, when competing with calcium in synaptic transmission. Thus, the purpose of this study was to investigate the temporal evolution of the contrast related to manganese in the acute phase of temporal lobe epilepsy induced by systemic pilocarpine and compare it to the expression of the c-Fos protein. During this phase, the intensity of the MEMRI signal was analyzed at three different time points (5, 15 or 30 minutes) after the onset of status epilepticus (SE). The group that was maintained in status epilepticus for 30 minutes showed a decrease in intensity of the signal in CA1 and the dentate gyrus (DG). There were no differences between the control group and the other groups treated with pilocarpine. The expression of the protein, c-Fos, in the same animals showed that even in the short-duration status epilepticus (5 minutes), there was already maximal cellular activation in subregions of the hippocampus (DG, CA1 and CA3). Under the experimental conditions tested, our data suggest that the MEMRI signal was not sensitive for the identification of detectable variations of cell activation in the acute phase of the pilocarpine model. Our findings are not consistent with the idea that manganese contrast reflects primarily alterations in cellular activity during SE when other signal-modifying elements can act.


As imagens de ressonância magnética são úteis no estudo de modelos experimentais de epilepsia do lobo temporal. A técnica manganese-enhanced MRI (MEMRI) é de interesse por combinar os efeitos provocados pelo manganês no aumento do contraste de populações celulares ativadas, ao competir com o cálcio na transmissão sináptica. Assim, o propósito deste estudo foi investigar a evolução temporal do contraste provocado pelo manganês na fase aguda da epilepsia do lobo temporal induzida por pilocarpina sistêmica e compará-las à expressão da proteína c-Fos. Nessa fase, a intensidade do sinal MEMRI foi analisada em três diferentes pontos temporais (5, 15 ou 30 minutos) após o início do status epilepticus (SE). O grupo que foi mantido em status epilepticus por 30 minutos mostrou diminuição na intensidade de sinal no CA1 e giro denteado (GD). Não houve diferenças entre o Grupo Controle e os outros grupos tratados com pilocarpina. A expressão da proteína c-Fos, nos mesmos animais, mostrou que, mesmo no status epilepticus de curta duração (5 minutos) já há ativação celular máxima nas sub-regiões do hipocampo (GD, CA1 e CA3). Nas condições experimentais testadas, nossos dados sugerem que o sinal MEMRI não foi sensível para identificar variações detectáveis da ativação celular na fase aguda do modelo de pilocarpina. Nossos achados não são consistentes com a ideia que o contraste por manganês reflete primariamente alterações na atividade celular durante o SE quando outros elementos modificadores do sinal podem atuar.


Asunto(s)
Epilepsia , Hipocampo , Imagen por Resonancia Magnética , Manganeso
16.
Einstein (Säo Paulo) ; 10(2)apr.-jun. 2012. ilus
Artículo en Inglés, Portugués | LILACS | ID: lil-644879

RESUMEN

Objective: The aim of the current study was to monitor the migration of superparamagnetic iron oxide nanoparticle (SPION)-labeled C6 cells, which were used to induce glioblastoma tumor growth in an animal model, over time using magnetic resonance imaging (MRI), with the goal of aiding in tumor prognosis and therapy. Methods: Two groups of male Wistar rats were used for the tumor induction model. In the first group (n=3), the tumors were induced via the injection of SPION-labeled C6 cells. In the second group (n=3), the tumors were induced via the injection of unlabeled C6 cells. Prussian Blue staining was performed to analyze the SPION distribution within the C6 cells in vitro. Tumor-inducing C6 cells were injected into the right frontal cortex, and subsequent tumor monitoring and SPION detection were performed using T2- and T2*-weighted MRI at a 2T strength. In addition, cancerous tissue was histologically analyzed after performing the MRI studies. Results: The in vitro qualitative evaluation demonstrated adequate distribution and satisfactory cell labeling of the SPIONs. At 14 or 21 days after C6 injection, a SPIONinduced T2- and T2*-weighted MRI signal reduction was observed within the lesion located in the left frontal lobe on parasagittal topography. Moreover, histological staining of the tumor tissue with Prussian Blue revealed a broad distribution of SPIONs within the C6 cells. Conclusion: MRI analyses exhibit potential for monitoring the tumor growth of C6 cells efficiently labeled with SPIONs.


Objetivo: Realizar monitoramento temporal por imagem por ressonância magnética da migração de células C6 marcadas com nanopartículas superparamagnéticas de óxido de ferro utilizadas na indução de tumor de glioblastoma no modelo animal, com o intuito de auxiliar no prognóstico e na terapêutica de tumores. Métodos: Para o modelo animal de indução de tumor, foram utilizados ratos Wistar machos, divididos em dois grupos. No primeiro grupo (n=3), o tumor foi induzido por células C6 marcadas com nanopartículas superparamagnéticas de óxido de ferro e, no segundo grupo, (n=3) o tumor foi induzido por C6 não marcadas. Foi realizada análise in vitro da distribuição intracelular das nanopartículas superparamagnéticas de óxido de ferro nas células C6 mediante coloração de azul da prússia. As células C6 para a indução de tumor foram implantadas no córtex frontal direito. Posteriormente, foram realizados o monitoramento tumoral e a detecção das nanopartículas superparamagnéticas de óxido de ferro por sequências de imagem por ressonância magnética ponderadas em T2 e T2*, em campo de 2T. Após os estudos de imagem por ressonância magnética, o tecido tumoral foi submetido à análise histológica. Resultados: A avaliação qualitativa do estudo in vitro mostrou boa distribuição e satisfatória marcação celular com nanopartículas superparamagnéticas de óxido de ferro. No monitoramento realizado por imagem por ressonância magnética, foi observada, no 14o e 21o dia, redução do sinal em T2 e T2*, induzida pelas nanopartículas superparamagnéticas de óxido de ferro, na lesão localizada no lobo frontal esquerdo em topografia parassagital. Por meio da marcação com azul da prússia, a análise histológica do tecido tumoral revelou que, nas células C6, ainda encontramos uma vasta distribuição das nanopartículas superparamagnéticas de óxido de ferro. Conclusão: A imagem por ressonância magnética apresenta-se com alto potencial para o monitoramento das células C6 marcadas eficientemente com nanopartículas superparamagnéticas de óxido de ferro na avaliação do crescimento tumoral.


Asunto(s)
Ratas , Glioma , Imagen por Resonancia Magnética , Nanopartículas
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA