Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
1.
Neurobiol Dis ; 48(3): 339-47, 2012 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-22782080

RESUMEN

Matrix metalloproteinases (MMPs) are involved in tissue repair, cell death and morphogenesis. We investigated the role of the gelatinases MMP-2 and MMP-9 in the pathogenesis of neuronal death induced by prolonged seizures in the developing brain. Seven-day-old rats, MMP-9 knockout mice and transgenic rats overexpressing MMP-9 received intraperitoneal injections of pilocarpine, 250 mg/kg, to induce seizures. After 6-72 h pups were sacrificed, tissue from different brain regions was isolated and expression of MMP-9 mRNA and protein was analyzed by real-time PCR or Western blot. Additionally, brains were fixed and processed for TUNEL-staining, immunohistochemistry and in situ zymography. We found increased numbers of TUNEL-positive cells 24 h after pilocarpine-induced seizures, most pronounced in cortical areas and the dentate gyrus, and less pronounced in thalamus. At 6-24 h, MMP-9 mRNA levels showed significant elevation compared to sham-treated controls; this effect resolved by 48 h, whereas MMP-2 mRNA levels remained stable. Cortical gelatinolytic activity, monitored by in situ zymography, was enhanced following pilocarpine-induced seizures. The MMP inhibitor GM 6001 ameliorated cell death following pilocarpine-induced seizures in infant rats. MMP-9 knockout mice were less susceptible to seizure-induced brain injury. Transgenic rats overexpressing MMP-9 were equally susceptible to seizure-induced brain injury as wild type rats. Our results suggest a significant contribution of MMP-9 to cell death after pilocarpine-induced seizures in the developing brain. As indicated by Western blot analysis, MMP-9 activation may be linked to activation of the Erk/CREB-pathway. The findings implicate involvement of MMP-9 in the pathophysiology of brain injury following seizures in the developing brain.


Asunto(s)
Apoptosis/fisiología , Encéfalo/enzimología , Metaloproteinasa 9 de la Matriz/metabolismo , Convulsiones/enzimología , Animales , Western Blotting , Encéfalo/patología , Convulsivantes/toxicidad , Humanos , Inmunohistoquímica , Etiquetado Corte-Fin in Situ , Metaloproteinasa 9 de la Matriz/deficiencia , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Degeneración Nerviosa , Pilocarpina/toxicidad , ARN Mensajero/análisis , Ratas , Ratas Transgénicas , Ratas Wistar , Reacción en Cadena en Tiempo Real de la Polimerasa , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Convulsiones/inducido químicamente , Convulsiones/patología
2.
Nat Neurosci ; 11(4): 476-87, 2008 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-18344994

RESUMEN

Intrinsic antioxidant defenses are important for neuronal longevity. We found that in rat neurons, synaptic activity, acting via NMDA receptor (NMDAR) signaling, boosted antioxidant defenses by making changes to the thioredoxin-peroxiredoxin (Prx) system. Synaptic activity enhanced thioredoxin activity, facilitated the reduction of overoxidized Prxs and promoted resistance to oxidative stress. Resistance was mediated by coordinated transcriptional changes; synaptic NMDAR activity inactivated a previously unknown Forkhead box O target gene, the thioredoxin inhibitor Txnip. Conversely, NMDAR blockade upregulated Txnip in vivo and in vitro, where it bound thioredoxin and promoted vulnerability to oxidative damage. Synaptic activity also upregulated the Prx reactivating genes Sesn2 (sestrin 2) and Srxn1 (sulfiredoxin), via C/EBPbeta and AP-1, respectively. Mimicking these expression changes was sufficient to strengthen antioxidant defenses. Trans-synaptic stimulation of synaptic NMDARs was crucial for boosting antioxidant defenses; chronic bath activation of all (synaptic and extrasynaptic) NMDARs induced no antioxidative effects. Thus, synaptic NMDAR activity may influence the progression of pathological processes associated with oxidative damage.


Asunto(s)
Antioxidantes/metabolismo , Estrés Oxidativo/fisiología , Peroxirredoxinas/metabolismo , Receptores de N-Metil-D-Aspartato/metabolismo , Tiorredoxinas/metabolismo , Animales , Proteínas Portadoras/metabolismo , Células Cultivadas , Corteza Cerebral/citología , Corteza Cerebral/metabolismo , Regulación de la Expresión Génica/fisiología , Ratones , Neuronas/metabolismo , Proteínas Nucleares , Oxidorreductasas actuantes sobre Donantes de Grupos Sulfuro/metabolismo , Peroxidasas , Proteínas/metabolismo , Ratas , Transducción de Señal/fisiología , Sinapsis/metabolismo , Transmisión Sináptica/fisiología , Transcripción Genética/fisiología
3.
Ann Neurol ; 64(1): 42-52, 2008 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-18067175

RESUMEN

OBJECTIVE: Marijuana and alcohol are most widely abused drugs among women of reproductive age. Neurocognitive deficits have been reported in children whose mothers used marijuana during pregnancy. Maternal consumption of ethanol is known to cause serious developmental deficits METHODS: Infant rats and mice received systemic injections of Delta(9)-tetrahydrocannabinol (THC; 1-10mg/kg) or the synthetic cannabinoid WIN 55,212-2 (1-10mg/kg), alone or in combination with subtoxic and toxic ethanol doses, and apoptotic neurodegeneration was studied in the brains RESULTS: Acute administration of THC (1-10mg/kg), the principal psychoactive cannabinoid of marijuana, markedly enhanced proapoptotic properties of ethanol in the neonatal rat brain. THC did not induce neurodegeneration when administered alone. Neuronal degeneration became disseminated and severe when THC was combined with a mildly intoxicating ethanol dose (3gm/kg), with the effect of this drug combination resembling the massive apoptotic death observed when administering ethanol alone at much higher doses. The detrimental effect of THC was mimicked by the synthetic cannabinoid WIN 55,212-2 (1-10mg/kg) and counteracted by the CB(1) receptor antagonist SR141716A (0.4mg/kg). THC enhanced the proapoptotic effect of the GABA(A) agonist phenobarbital and the N-methyl-D-aspartate receptor antagonist dizocilpine. Interestingly, infant CB(1) receptor knock-out mice were less susceptible to the neurotoxic effect of ethanol. Furthermore, the CB(1) receptor antagonist SR141716A ameliorated neurotoxicity of ethanol INTERPRETATION: These observations indicate that CB(1) receptor activation modulates GABAergic and glutamatergic neurotransmission and primes the developing brain to suffer apoptotic neuronal death.


Asunto(s)
Envejecimiento/fisiología , Trastornos del Sistema Nervioso Inducidos por Alcohol/inducido químicamente , Encéfalo/efectos de los fármacos , Encéfalo/crecimiento & desarrollo , Cannabinoides/agonistas , Etanol/agonistas , Neurotoxinas/agonistas , Trastornos del Sistema Nervioso Inducidos por Alcohol/metabolismo , Trastornos del Sistema Nervioso Inducidos por Alcohol/fisiopatología , Animales , Animales Recién Nacidos , Benzoxazinas/agonistas , Benzoxazinas/toxicidad , Encéfalo/fisiopatología , Cannabinoides/toxicidad , Muerte Celular/efectos de los fármacos , Muerte Celular/fisiología , Depresores del Sistema Nervioso Central/agonistas , Depresores del Sistema Nervioso Central/toxicidad , Relación Dosis-Respuesta a Droga , Dronabinol/agonistas , Dronabinol/toxicidad , Resistencia a Medicamentos/efectos de los fármacos , Resistencia a Medicamentos/fisiología , Sinergismo Farmacológico , Etanol/toxicidad , Antagonistas de Aminoácidos Excitadores/toxicidad , Agonistas del GABA/toxicidad , Ratones , Ratones Noqueados , Morfolinas/agonistas , Morfolinas/toxicidad , Naftalenos/agonistas , Naftalenos/toxicidad , Degeneración Nerviosa/inducido químicamente , Degeneración Nerviosa/metabolismo , Degeneración Nerviosa/fisiopatología , Neurotoxinas/toxicidad , Ratas , Ratas Wistar , Receptor Cannabinoide CB1/agonistas , Receptor Cannabinoide CB1/antagonistas & inhibidores
4.
Ann Neurol ; 64(4): 434-45, 2008 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-18991352

RESUMEN

OBJECTIVE: Sedative and anticonvulsant drugs, which inhibit N-methyl-D-aspartate receptor-mediated excitation or enhance GABA-mediated action, may cause apoptotic neurodegeneration in the developing mammalian brain. Here we explored whether such agents influence early postnatal neurogenesis. METHODS: The N-methyl-D-aspartate antagonist MK801 and the GABA subtype A agonists phenobarbital and diazepam were administered to infant rats, and cell proliferation and neurogenesis were studied in the brain using 5-bromo-2'-deoxyuridine and doublecortin immunohistochemistry and stereology. Using confocal microscopy, we quantified neurogenesis in the dentate gyrus on postnatal day 15 (P15) after treatment with MK801 or phenobarbital on P6 to P10. Learning and memory were assessed at the age of 6 months after early postnatal treatment with phenobarbital. RESULTS: MK801, phenobarbital, and diazepam reduced numbers of newly born cells in the brain. We found no evidence that these agents caused apoptosis of 5-bromo-2'-deoxyuridine-positive cells. In the dentate gyrus, many of the newly formed cells differentiated toward a neuronal phenotype. Phenobarbital and MK801 reduced numbers of newly formed neurons in the dentate gyrus. At the age of 6 months, phenobarbital-treated rats had fewer neurons in the dentate gyrus and performed worse than saline-treated littermates in water maze learning and memory task. INTERPRETATION: These findings show that blockade of N-methyl-D-aspartate receptor-mediated excitation and enhancement of GABA subtype A receptor activation impair cell proliferation and inhibit neurogenesis in the immature rat brain. Because many sedative and antiepileptic drugs used in pediatric medicine act via these mechanisms, our findings raise concerns about their potential impact on human brain development.


Asunto(s)
Anticonvulsivantes/farmacología , Hipnóticos y Sedantes/farmacología , Neurogénesis/efectos de los fármacos , Factores de Edad , Análisis de Varianza , Animales , Animales Recién Nacidos , Apoptosis/efectos de los fármacos , Conducta Animal/efectos de los fármacos , Bromodesoxiuridina/metabolismo , Recuento de Células/métodos , Proliferación Celular/efectos de los fármacos , Proteína Doblecortina , Femenino , Hipocampo/anatomía & histología , Masculino , Aprendizaje por Laberinto/efectos de los fármacos , Ratas , Ratas Wistar , Factores de Tiempo
5.
Ann N Y Acad Sci ; 1053: 269-86, 2005 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-16179532

RESUMEN

To reveal whether increased Ca2+ permeability of glutamate AMPA channels triggered by the transgene for GluR-B(N) induces decline in motor functions and neurodegeneration in the spinal cord, we evaluated growth, motor coordination, and spinal reflexes in transgenic GluR-B(N) and wild-type (wt) mice. To reveal whether the transgenic GluR-B(N) expression aggravates the course of motoneuron disease in SOD1 mice, we mated heterozygous GluR-B(N) and SOD1 [C57BL6Ico-TgN(hSOD1-G93A)1Gur] mice to generate double-transgenic progeny. The phenotypic sequelae in mice carrying mutations were evaluated by monitoring growth, motor coordination, and survival. Neuronal degeneration was assessed by morphological and stereological analysis of spinal cord and brain. We found that transgenic expression in mice of GluR-B(N)-containing glutamate AMPA receptors with increased Ca2+ permeability leads to a late-onset degeneration of neurons in the spinal cord and decline of motor functions. Neuronal death progressed over the entire life span, but manifested clinically in late adulthood, resembling the course of a slow neurodegenerative disorder. Additional transgenic expression of mutated human SOD1 accelerated disease progression, aggravated severity of motor decline, and decreased survival. These observations reveal that moderate, but persistently elevated Ca2+ influx via glutamate AMPA channels causes degeneration of spinal motoneurons and motor decline over the span of life. These features resemble the course of sporadic amyotrophic lateral sclerosis (ALS) in humans and suggest that modified function of glutamate AMPA channels may be causally linked to pathogenesis of ALS.


Asunto(s)
Enfermedad de la Neurona Motora/fisiopatología , Receptores AMPA/genética , Receptores AMPA/fisiología , Animales , Animales Modificados Genéticamente , Ansiedad/genética , Ansiedad/psicología , Peso Corporal/fisiología , Cobalto/metabolismo , Electromiografía , Conducta Exploratoria/fisiología , Marcha/fisiología , Hibridación in Situ , Ratones , Actividad Motora/fisiología , Mutación/fisiología , Fenotipo , Equilibrio Postural/fisiología , Reflejo/fisiología , Médula Espinal/metabolismo , Superóxido Dismutasa/genética , Superóxido Dismutasa-1 , Sobrevida , Temblor/genética , Temblor/fisiopatología
6.
Neurotox Res ; 19(4): 638-48, 2011 May.
Artículo en Inglés | MEDLINE | ID: mdl-20661683

RESUMEN

Matrix metalloproteinases (MMPs) play an essential role in tissue repair, cell death, and morphogenesis. The aim of the present study was to investigate potential involvement of selected MMPs in the pathogenesis of neuronal apoptosis induced by the NMDA antagonist MK-801 (dizocilpine) or the GABA(A) agonist phenobarbital in infant rats, transgenic rats overexpressing MMP-9 and MMP-9 knockout mice. Seven-day-old rats or knockout mice received intraperitoneal injections of MK-801, 1 mg/kg, or phenobarbital, 50 mg/kg. At different survival intervals following administration of the compounds (1-72 h), pups were sacrificed, tissue from different brain regions was isolated, and the expression and activity of MMP-2 and MMP-9 were analyzed by real-time PCR, western blot, and zymography. In addition, brains were fixed and processed for TUNEL staining. In all the brain regions analyzed, we found an increased number of TUNEL-positive cells 24 h after administration of MK-801. After treatment, we detected no significant increase in MMP-2 or MMP-9 mRNA expression in cortical areas. No changes in the MMP-9 protein expression or gelatinolytic activity of MMP-2 were observed in conjunction with MK-801 or phenobarbital-induced neuroapoptosis in any brain region analyzed. The extent of neurodegeneration induced by MK-801 or phenobarbital was not altered in MMP-9 transgenic rats and was increased in MMP-9 knockout mice compared to wild-type rats and mice. Treatment with the panmetalloproteinase inhibitor GM6001 did not confer protection against MK-801-induced apoptotic cell death in the developing rat brain. Our results suggest that activation of MMP-9 and MMP-2 does not contribute to pathogenesis of neuronal apoptosis caused by NMDA antagonists or GABA(A) agonists in the developing rat and mouse brain.


Asunto(s)
Apoptosis/fisiología , Encéfalo/enzimología , Maleato de Dizocilpina/toxicidad , Metaloproteinasa 2 de la Matriz/metabolismo , Metaloproteinasa 9 de la Matriz/metabolismo , Neuronas/citología , Neuronas/efectos de los fármacos , Fenobarbital/toxicidad , Animales , Animales Recién Nacidos , Apoptosis/efectos de los fármacos , Encéfalo/efectos de los fármacos , Encéfalo/crecimiento & desarrollo , Metaloproteinasa 2 de la Matriz/fisiología , Metaloproteinasa 9 de la Matriz/fisiología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Neuronas/enzimología , Ratas , Ratas Wistar
7.
Neurobiol Dis ; 25(3): 526-35, 2007 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-17188498

RESUMEN

Matrix metalloproteinases (MMPs) play an essential role in tissue repair, cell death and morphogenesis and may constitute therapeutic targets for acute brain injuries. In this study, we investigated the expression of 72 kDa and 92 kDa collagenases MMP-2 and MMP-9 at transcriptional, functional and protein expression level following traumatic brain injury in infant rats. Seven-day-old Wistar rats were subjected to head trauma using a weight drop device. Pups were sacrificed at defined time points (2-72 h) after trauma and brains were processed for molecular studies (semiquantitative and real-time PCR, Western blot, gelatin zymography) and histology. Trauma triggered widespread cell death in the cortex, basal ganglia and white matter. mRNA levels for MMP-2 and -9 were increased in the brain at 12-72 h after trauma. Protein expression of the analyzed MMPs and activity of MMP-2 were increased at 12 h and peaked at 24 h after trauma. Intraperitoneal injection of GM6001 (Ilomastat), an MMP inhibitor, 2 h after trauma, substantially attenuated traumatic brain injury in a dose-dependent manner. These findings causally link the MMPs to trauma-induced neuronal cell death in the immature rodent brain. MMPs might serve as useful targets for therapeutic approaches aimed at preserving neuronal function in the immature brain in the context of mechanical injury.


Asunto(s)
Lesiones Encefálicas/metabolismo , Lesiones Encefálicas/fisiopatología , Encéfalo/enzimología , Metaloproteinasa 2 de la Matriz/metabolismo , Metaloproteinasa 9 de la Matriz/metabolismo , Factores de Edad , Animales , Apoptosis/efectos de los fármacos , Encéfalo/crecimiento & desarrollo , Encéfalo/patología , Lesiones Encefálicas/tratamiento farmacológico , Dipéptidos/farmacología , Regulación Enzimológica de la Expresión Génica , Etiquetado Corte-Fin in Situ , Metaloproteinasa 2 de la Matriz/genética , Metaloproteinasa 9 de la Matriz/genética , Inhibidores de la Metaloproteinasa de la Matriz , Degeneración Nerviosa/tratamiento farmacológico , Degeneración Nerviosa/metabolismo , Degeneración Nerviosa/fisiopatología , Inhibidores de Proteasas/farmacología , ARN Mensajero/metabolismo , Ratas , Ratas Wistar , Inhibidor Tisular de Metaloproteinasa-1/genética , Inhibidor Tisular de Metaloproteinasa-1/metabolismo , Inhibidor Tisular de Metaloproteinasa-2/genética , Inhibidor Tisular de Metaloproteinasa-2/metabolismo
8.
Proteomics Clin Appl ; 1(7): 640-9, 2007 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-21136719

RESUMEN

Traumatic brain injury (TBI) is a major cause of morbidity and mortality among children and adolescents. To gain insight into developmental events influenced by TBI, we analyzed subacute mouse brain proteome changes in a percussion head trauma model at P7 ipsi- and contralateral to the site of injury. The comparison of brain proteomes of trauma mice and controls revealed reproducible changes in the intensity of 28 proteins (30 protein spots) in response to trauma. The changes detected suggest that TBI leads to apoptosis, inflammation, and oxidative stress. These changes were consistent with our results of histological and biochemical evaluation of the brains which revealed widespread apoptotic neurodegeneration, microglia activation, and increased levels of protein carbonyls. Furthermore, we detected changes in proteins involved in neuronal migration as well as axonal and dendritic growth and guidance, suggesting interference of trauma with these developmental events.

9.
Neurobiol Dis ; 25(3): 614-22, 2007 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-17188500

RESUMEN

Focal mechanical cortical trauma triggers diffuse apoptotic neurodegeneration in the developing rat brain which is associated with invasion of brain tissue with inflammatory mediators. We hypothesized that caspase-1 and the two caspase-1-processed cytokines, interleukin (IL)-1beta and IL-18, are involved in trauma-induced neuronal cell death in the developing brain. 7-day-old Wistar rats or C57/BL6 mice were subjected to head trauma using a weight drop device. Animals were sacrificed at defined time points following trauma and brains were processed for histology and molecular analyses. Neuronal cell death in the immature brain peaked at 12-24 h and was accompanied by a marked increase of mRNA and protein levels for caspase-1, IL-1beta and IL-18 within 2 to 12 h following the injury. Caspase-1 levels were elevated for 72 h, whereas IL-1beta decreased earlier at 48 h. IL-18 remained high over a period of 3 days and decreased to normal levels by day 7 after the injury. Intraperitoneal injection of recombinant human IL-18-binding protein (IL-18BP), a specific inhibitor of IL-18, attenuated traumatic brain injury. Mice deficient in IL-18 (IL-18-/-) were protected against trauma-induced brain damage. These findings indicate that IL-18 is involved in trauma-induced neuronal cell death in the immature rodent brain and might serve as a potential therapeutic target.


Asunto(s)
Lesiones Encefálicas/inmunología , Lesiones Encefálicas/metabolismo , Caspasa 1/metabolismo , Interleucina-18/metabolismo , Interleucina-1beta/metabolismo , Animales , Apoptosis/fisiología , Encéfalo/enzimología , Encéfalo/inmunología , Encéfalo/patología , Lesiones Encefálicas/patología , Caspasa 1/genética , Interleucina-18/genética , Interleucina-1beta/genética , Ratones , Ratones Endogámicos C57BL , Ratones Mutantes , Degeneración Nerviosa/inmunología , Degeneración Nerviosa/metabolismo , Degeneración Nerviosa/patología , ARN Mensajero/metabolismo , Ratas , Ratas Wistar
10.
Exp Neurol ; 193(2): 497-503, 2005 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-15869952

RESUMEN

Antiepileptic drugs (AEDs) used to treat seizures in pregnant women, infants, and young children can cause cognitive impairment. One mechanism implicated in the development of neurocognitive deficits is a pathologic enhancement of physiologically occurring apoptotic neuronal death in the developing brain. We investigated whether the newer antiepileptic drug levetiracetam (LEV) and the older antiepileptic drug sulthiame (SUL) have neurotoxic properties in the developing rat brain. SUL significantly enhanced neuronal death in the brains of rat pups ages 0 to 7 days at doses of 100 mg/kg and above, whereas LEV did not show this neurotoxic effect. Dosages of both drugs used in the context of this study comply with an effective anticonvulsant dose range applied in rodent seizure models. Thus, LEV is an AED which lacks neurotoxicity in the developing rat brain and should be considered in the treatment of epilepsy in pregnant women, infants, and toddlers once general safety issues have been properly addressed.


Asunto(s)
Encéfalo/efectos de los fármacos , Neurotoxinas/toxicidad , Piracetam/análogos & derivados , Piracetam/toxicidad , Tiazinas/toxicidad , Factores de Edad , Animales , Animales Recién Nacidos , Encéfalo/crecimiento & desarrollo , Encéfalo/patología , Muerte Celular/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Femenino , Levetiracetam , Masculino , Degeneración Nerviosa/inducido químicamente , Degeneración Nerviosa/patología , Ratas , Ratas Wistar
11.
Proc Natl Acad Sci U S A ; 102(16): 5826-31, 2005 Apr 19.
Artículo en Inglés | MEDLINE | ID: mdl-15827116

RESUMEN

Amyotrophic lateral sclerosis (ALS) is a devastating disorder of the central nervous system in middle and old age that leads to progressive loss of spinal motoneurons. Transgenic mice overexpressing mutated human Cu(2+)/Zn(2+) superoxide dismutase 1 (SOD1) reproduce clinical features of the familial form of ALS. However, changes in SOD1 activity do not correlate with severity of motor decline in sporadic cases, indicating that targets unrelated to superoxide metabolism contribute to the pathogenesis of the disease. We show here that transgenic expression in mice of GluR-B(N)-containing L-alpha-amino-3-hydroxy-5-methylisoxazole-4-proprionate (AMPA) receptors with increased Ca(2+) permeability leads to late-onset degeneration of neurons in the spinal cord and decline of motor functions. Neuronal death progresses over the entire lifespan but manifests clinically in late adulthood, resembling the course of a slow neurodegenerative disorder. Additional transgenic expression of mutated human SOD1 accelerates disease progression, aggravates the severity of motor decline, and decreases survival. These observations link persistently elevated Ca(2+) influx through AMPA channels with progressive motor decline and late-onset degeneration of spinal motoneurons, indicating that functionally altered AMPA channels may be causally related to pathogenesis of sporadic ALS in humans.


Asunto(s)
Esclerosis Amiotrófica Lateral/metabolismo , Subunidades de Proteína/metabolismo , Receptores AMPA/metabolismo , Esclerosis Amiotrófica Lateral/etiología , Esclerosis Amiotrófica Lateral/patología , Animales , Encéfalo/citología , Encéfalo/patología , Calcio/metabolismo , Cobalto/metabolismo , Electromiografía , Humanos , Hibridación in Situ , Ratones , Ratones Endogámicos BALB C , Ratones Transgénicos , Actividad Motora/fisiología , Neuronas/metabolismo , Neuronas/patología , Neuronas/ultraestructura , Subunidades de Proteína/genética , Receptores AMPA/genética , Reflejo/fisiología , Médula Espinal/citología , Médula Espinal/patología , Superóxido Dismutasa/genética , Superóxido Dismutasa/metabolismo , Superóxido Dismutasa-1
12.
Ann Neurol ; 56(3): 351-60, 2004 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-15349862

RESUMEN

Neurotoxicity of anticancer agents complicates treatment of children with cancer. We investigated neurotoxic effects of common cytotoxic drugs in neuronal cultures and in the developing rat brain. When neurons were exposed to cisplatin (5-100 microM), cyclophosphamide (5-100 microM), methotrexate (5-100 microM), vinblastin (0.1-1 microM), or thiotepa (5-100 microM), a concentration-dependent neurotoxic effect was observed. Neurotoxicity was potentiated by nontoxic glutamate concentrations. The N-methyl-D-aspartate receptor antagonist MK 801 (10 microM), the AMPA receptor antagonists GYKI 52466 (10 microM) and NBQX (10 microM), and the pancaspase inhibitor Ac-DEVD-CHO (1 nM) ameliorated neurotoxicity of cytotoxic drugs. To investigate neurotoxicity in vivo, we administered to 7-day-old rats the following: cisplatin (5-15 mg/kg i.p.), cyclophosphamide (200-600 mg/kg i.p.), thiotepa (15-45 mg/kg), or ifosfamide (100-500 mg/kg) and their brains were analyzed at 4 to 24 hours. Cytotoxic drugs produced widespread lesions within cortex, thalamus, hippocampal dentate gyrus, and caudate nucleus in a dose-dependent fashion. Early histological analysis demonstrated dendritic swelling and relative preservation of axonal terminals, which are morphological features indicating excitotoxicity. After longer survival periods, degenerating neurons displayed morphological features consistent with active cell death. These results demonstrate that anticancer drugs are potent neurotoxins in vitro and in vivo; they activate excitotoxic mechanisms but also trigger active neuronal death.


Asunto(s)
Antineoplásicos/toxicidad , Neuronas/efectos de los fármacos , Neuronas/patología , Animales , Muerte Celular/efectos de los fármacos , Muerte Celular/fisiología , Células Cultivadas , Corteza Cerebral/efectos de los fármacos , Corteza Cerebral/patología , Corteza Cerebral/fisiología , Relación Dosis-Respuesta a Droga , Ratas , Ratas Wistar
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA