Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 26
Filtrar
Más filtros













Base de datos
Intervalo de año de publicación
1.
Org Lett ; 25(36): 6688-6692, 2023 09 15.
Artículo en Inglés | MEDLINE | ID: mdl-37671859

RESUMEN

Enzymatic reduction of diphenylmethanimine derivatives has rarely been reported owing to their steric hindrance. Herein, imine reductase (IRED) from Nocardia cyriacigeorgica rationally engineered with an efficient strategy of focused rational iterative site-specific mutagenesis (FRISM) was selected for the reduction of a series of N-cyclopropylmethyl-1-aryl-1-phenylmethylimines. Two highly enantioselective IRED variants were identified, providing various bulky amine products with moderate to high yields and high ee values (up to >99%). This work provided an effective method to construct these important pharmaceutical intermediates.


Asunto(s)
Aminas , Bencilaminas , Iminas , Mutagénesis Sitio-Dirigida , Catálisis
2.
Mol Neurodegener ; 14(1): 44, 2019 11 27.
Artículo en Inglés | MEDLINE | ID: mdl-31775817

RESUMEN

BACKGROUND: Traumatic brain injury (TBI) is a major cause of CNS neurodegeneration and has no disease-altering therapies. It is commonly associated with a specific type of biomechanical disruption of the axon called traumatic axonal injury (TAI), which often leads to axonal and sometimes perikaryal degeneration of CNS neurons. We have previously used genome-scale, arrayed RNA interference-based screens in primary mouse retinal ganglion cells (RGCs) to identify a pair of related kinases, dual leucine zipper kinase (DLK) and leucine zipper kinase (LZK) that are key mediators of cell death in response to simple axotomy. Moreover, we showed that DLK and LZK are the major upstream triggers for JUN N-terminal kinase (JNK) signaling following total axonal transection. However, the degree to which DLK/LZK are involved in TAI/TBI is unknown. METHODS: Here we used the impact acceleration (IA) model of diffuse TBI, which produces TAI in the visual system, and complementary genetic and pharmacologic approaches to disrupt DLK and LZK, and explored whether DLK and LZK play a role in RGC perikaryal and axonal degeneration in response to TAI. RESULTS: Our findings show that the IA model activates DLK/JNK/JUN signaling but, in contrast to axotomy, many RGCs are able to recover from the injury and terminate the activation of the pathway. Moreover, while DLK disruption is sufficient to suppress JUN phosphorylation, combined DLK and LZK inhibition is required to prevent RGC cell death. Finally, we show that the FDA-approved protein kinase inhibitor, sunitinib, which has activity against DLK and LZK, is able to produce similar increases in RGC survival. CONCLUSION: The mitogen-activated kinase kinase kinases (MAP3Ks), DLK and LZK, participate in cell death signaling of CNS neurons in response to TBI. Moreover, sustained pharmacologic inhibition of DLK is neuroprotective, an effect creating an opportunity to potentially translate these findings to patients with TBI.


Asunto(s)
Lesiones Traumáticas del Encéfalo/metabolismo , Supervivencia Celular/fisiología , Quinasas Quinasa Quinasa PAM/metabolismo , Neuronas/metabolismo , Animales , Lesiones Traumáticas del Encéfalo/patología , Modelos Animales de Enfermedad , Leucina Zippers/efectos de los fármacos , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Masculino , Ratones Endogámicos C57BL , Neuronas/efectos de los fármacos , Inhibidores de Proteínas Quinasas/farmacología , Células Ganglionares de la Retina/metabolismo
3.
PLoS One ; 14(11): e0224846, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31710637

RESUMEN

Optogenetically engineered human neural progenitors (hNPs) are viewed as promising tools in regenerative neuroscience because they allow the testing of the ability of hNPs to integrate within nervous system of an appropriate host not only structurally, but also functionally based on the responses of their differentiated progenies to light. Here, we transduced H9 embryonic stem cell-derived hNPs with a lentivirus harboring human channelrhodopsin (hChR2) and differentiated them into a forebrain lineage. We extensively characterized the fate and optogenetic functionality of hChR2-hNPs in vitro with electrophysiology and immunocytochemistry. We also explored whether the in vivo phenotype of ChR2-hNPs conforms to in vitro observations by grafting them into the frontal neocortex of rodents and analyzing their survival and neuronal differentiation. Human ChR2-hNPs acquired neuronal phenotypes (TUJ1, MAP2, SMI-312, and synapsin 1 immunoreactivity) in vitro after an average of 70 days of coculturing with CD1 astrocytes and progressively displayed both inhibitory and excitatory neurotransmitter signatures by immunocytochemistry and whole-cell patch clamp recording. Three months after transplantation into motor cortex of naïve or injured mice, 60-70% of hChR2-hNPs at the transplantation site expressed TUJ1 and had neuronal cytologies, whereas 60% of cells also expressed ChR2. Transplant-derived neurons extended axons through major commissural and descending tracts and issued synaptophysin+ terminals in the claustrum, endopiriform area, and corresponding insular and piriform cortices. There was no apparent difference in engraftment, differentiation, or connectivity patterns between injured and sham subjects. Same trends were observed in a second rodent host, i.e. rat, where we employed longer survival times and found that the majority of grafted hChR2-hNPs differentiated into GABAergic neurons that established dense terminal fields and innervated mostly dendritic profiles in host cortical neurons. In physiological experiments, human ChR2+ neurons in culture generated spontaneous action potentials (APs) 100-170 days into differentiation and their firing activity was consistently driven by optical stimulation. Stimulation generated glutamatergic and GABAergic postsynaptic activity in neighboring ChR2- cells, evidence that hChR2-hNP-derived neurons had established functional synaptic connections with other neurons in culture. Light stimulation of hChR2-hNP transplants in vivo generated complicated results, in part because of the variable response of the transplants themselves. Our findings show that we can successfully derive hNPs with optogenetic properties that are fully transferrable to their differentiated neuronal progenies. We also show that these progenies have substantial neurotransmitter plasticity in vitro, whereas in vivo they mostly differentiate into inhibitory GABAergic neurons. Furthermore, neurons derived from hNPs have the capacity of establishing functional synapses with postsynaptic neurons in vitro, but this outcome is technically challenging to explore in vivo. We propose that optogenetically endowed hNPs hold great promise as tools to explore de novo circuit formation in the brain and, in the future, perhaps launch a new generation of neuromodulatory therapies.


Asunto(s)
Células Madre Embrionarias Humanas/citología , Células-Madre Neurales/citología , Neuronas/citología , Optogenética , Animales , Astrocitos/citología , Astrocitos/efectos de la radiación , Axones/metabolismo , Axones/efectos de la radiación , Diferenciación Celular/efectos de la radiación , Linaje de la Célula/efectos de la radiación , Supervivencia Celular/efectos de la radiación , Channelrhodopsins/metabolismo , Células Madre Embrionarias Humanas/efectos de la radiación , Humanos , Lentivirus/metabolismo , Luz , Ratones Desnudos , Corteza Motora/metabolismo , Células-Madre Neurales/efectos de la radiación , Plasticidad Neuronal/efectos de la radiación , Neuronas/efectos de la radiación , Neurotransmisores/metabolismo , Fenotipo , Estimulación Luminosa , Ratas Desnudas , Transmisión Sináptica/efectos de la radiación
4.
Hum Mol Genet ; 25(16): 3515-3523, 2016 08 15.
Artículo en Inglés | MEDLINE | ID: mdl-27378696

RESUMEN

Multiple missense mutations in Leucine-rich repeat kinase 2 (LRRK2) have been linked to Parkinson's disease (PD), the most common degenerative movement disorder. LRRK2 is expressed by both neurons and microglia, the residential immune cells in the brain. Increasing evidence supports a role of LRRK2 in modulating microglial activity, of which Lrrk2-null rodent microglia display less inflammatory response to endotoxin lipopolysaccharide (LPS). The underlying molecular mechanism, however, remains elusive. Chemokine (C-X3-C) receptor 1 (CX3CR1), predominantly expressed by microglia, suppresses microglial inflammation while promotes migration. Using whole-genome microarray screening, we found that Cx3cr1 mRNA levels were substantially higher in microglia derived from Lrrk2 knockout (Lrrk2-/-) mice. The total and cell surface levels of CX3CR1 proteins were also remarkably increased. In correlation with the enhanced CX3CR1 expression, Lrrk2-null microglia migrated faster and travelled longer distance toward the source of fractalkine (CX3CL1), an endogenous ligand of CX3CR1. To investigate the impact of CX3CR1 elevation in vivo, we compared LPS-induced inflammation in the striatum of Lrrk2-/- knockout mice with Cx3cr1 heterozygous and homozygous knockout background. We found that a complete loss of Cx3cr1 restored the responsiveness of Lrrk2-/- microglia to LPS stimulation. In conclusion, our findings reveal a previously unknown regulatory role for LRRK2 in CX3CR1 signalling and suggest that an increase of CX3CR1 activity contributes to the attenuated inflammatory responses in Lrrk2-null microglia.


Asunto(s)
Inflamación/genética , Proteína 2 Quinasa Serina-Treonina Rica en Repeticiones de Leucina/genética , Enfermedad de Parkinson/genética , Receptores de Quimiocina/genética , Animales , Receptor 1 de Quimiocinas CX3C , Cuerpo Estriado/metabolismo , Cuerpo Estriado/patología , Humanos , Inflamación/inducido químicamente , Inflamación/patología , Lipopolisacáridos/administración & dosificación , Activación de Macrófagos/efectos de los fármacos , Ratones Noqueados , Microglía/metabolismo , Microglía/patología , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Neuronas/patología , Enfermedad de Parkinson/patología , Receptores de Quimiocina/biosíntesis , Transducción de Señal/genética
5.
Exp Neurol ; 277: 268-274, 2016 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-26784004

RESUMEN

Mechanisms of primary blast injury caused by overpressure are not fully understood. In particular, the presence and time course of neuroinflammation are unknown and so are the signatures of reactive inflammatory cells, especially the neuroprotective versus injurious roles of microglia. In general, chronic microglial activation in the injured brain suggests a pro-degenerative role for these reactive cells. In this study, we investigated the temporal dynamics of microglial activation in the brain of mice exposed to mild-moderate blast in a shock tube. Because, in our previous work, we had found that torso shielding with rigid Plexiglas attenuates traumatic axonal injury in the brain, we also evaluated neuroinflammatory microglial responses in animals with torso protection at 7 days post blast injury. Because of the prominent involvement of the visual system in blast TBI in rodents, activated microglial cells were counted in the optic tract at various time points post-injury with stereological methods. Cell counts (activated microglial cell densities) from subjects exposed to blast TBI were compared with counts from corresponding sham animals. We found that mild-moderate blast injury causes focal activation of microglia in certain white matter tracts, including the visual pathway. In the optic tract, the density of activated microglial profiles gradually intensified from 3 to 15 days post-injury and then became attenuated at 30 days. Torso protection significantly reduced microglial activation at 7 days. These findings shed light into mechanisms of primary blast neurotrauma and may suggest novel diagnostic and monitoring methods for patients. They leave open the question of whether microglial activation post blast is protective or detrimental, although response is time limited. Finally, our findings confirm the protective role of torso shielding and stress the importance of improved or optimized body gear for warfighters or other individuals at risk for blast exposure.


Asunto(s)
Traumatismos por Explosión/complicaciones , Encefalitis/etiología , Encefalitis/prevención & control , Equipos de Seguridad , Torso/fisiología , Análisis de Varianza , Animales , Antígenos CD/metabolismo , Antígenos de Diferenciación Mielomonocítica/metabolismo , Proteínas de Unión al Calcio/metabolismo , Modelos Animales de Enfermedad , Canal de Potasio Kv1.3/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Proteínas de Microfilamentos/metabolismo , Microglía/patología , Tracto Óptico/patología , Factores de Tiempo
6.
Exp Neurol ; 275 Pt 3: 436-449, 2016 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-25450468

RESUMEN

Repetitive mild traumatic brain injury (mTBI) is implicated in chronic neurological illness. The development of animal models of repetitive mTBI in mice is essential for exploring mechanisms of these chronic diseases, including genetic vulnerability by using transgenic backgrounds. In this study, the rat model of impact acceleration (IA) was redesigned for the mouse cranium and used in two clinically relevant repetitive mTBI paradigms. We first determined, by using increments of weight dropped from 1m that the 40g weight was most representative of mTBI and was not associated with fractures, brain contusions, anoxic-ischemic injury, mortality, or significant neurological impairments. Quantitative evaluation of traumatic axonal injury (TAI) in the optic nerve/tract, cerebellum and corpus callosum confirmed that weight increase produced a graded injury. We next evaluated two novel repetitive mTBI paradigms (1 time per day or 3 times per day at days 0, 1, 3, and 7) and compared the resulting TAI, neuronal cell death, and neuroinflammation to single hit mTBI at sub-acute (7days) and chronic time points (10weeks) post-injury. Both single and repetitive mTBI caused TAI in the optic nerve/tract, cerebellum, corticospinal tract, lateral lemniscus and corpus callosum. Reactive microglia with phagocytic phenotypes were present at injury sites. Severity of axonal injury corresponded to impact load and frequency in the optic nerve/tract and cerebellum. Both single and repeat injury protocols were associated with retinal ganglion cell loss and optic nerve degeneration; these outcomes correlated with impact load and number/frequency. No phosphorylated tau immunoreactivity was detected in the brains of animals subjected to repetitive mTBI. Our findings establish a new model of repetitive mTBI model featured by TAI in discrete CNS tracts, especially the visual system and cerebellum. Injury in retina and optic nerve provides a sensitive measure of severity of mTBI, thus enabling further studies on mechanisms and experimental therapeutics. Our model can also be useful in exploring mechanisms of chronic neurological disease caused by repetitive mTBI in wild-type and transgenic mice.


Asunto(s)
Aceleración/efectos adversos , Axones/patología , Lesiones Encefálicas/patología , Modelos Animales de Enfermedad , Degeneración Nerviosa/patología , Células Ganglionares de la Retina/patología , Animales , Lesiones Encefálicas/complicaciones , Inflamación/etiología , Inflamación/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Degeneración Nerviosa/etiología , Nervio Óptico/patología
8.
Exp Neurol ; 273: 168-76, 2015 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-26311071

RESUMEN

Chronic traumatic encephalopathy (CTE) is associated with repetitive mild traumatic brain injury (mTBI) in the context of contact and collision sports, but not all exposed individuals develop this condition. In addition, experiments in animal models in several laboratories have shown that non-transgenic mice do not develop tauopathy after exposure to repetitive mTBI schedules. It is thus reasonable to assume that genetic factors may play an etiological role in the development of CTE. More than 40 mutations in the tau gene are known to confer proneness to aggregation and are thought to cause neurodegenerative diseases including frontotemporal degeneration (FTD). Transgenic mice harboring these mutations can be used to ask the question whether repetitive mTBI can accelerate onset and course of tauopathy or worsen the outcomes of transgenic disease. In this study, we exposed mice harboring the tau P301S transgene associated with FTD to repetitive mTBI schedules by impact acceleration (IA) that we have previously characterized. We explored the progression of tauopathy in the retina and neocortex based on density of neuronal profiles loaded with tau pS422, a marker of advanced tau hyperphosphorylation. We found that the density of tau pS422 (+) retinal ganglion cells (RGCs) increased twenty fold with one mTBI hit, a little over fifty fold with four mTBI hits and sixty fold with 12 mTBI hits. The severity of mTBI burden (number of hits) was a significant factor in tauopathy outcome. On the other hand, we found no association between repetitive mTBI and density of pS422 (+) neuronal profiles in neocortex, a region that is not featured by significant TAI in our repetitive mTBI model. We observed similar, but less prominent, trends in tauopathy-prone transgenic mice harboring all 6 isoforms of wild-type human tau without mouse tau. Our findings indicate that repetitive mTBI accelerates tauopathy under diverse genetic conditions predisposing to tau aggregation and suggest a vulnerability-stress model in understanding some cases of acquired neurodegenerative disease after repetitive mTBI.


Asunto(s)
Lesiones Encefálicas/complicaciones , Mutación/genética , Retina/patología , Tauopatías/patología , Proteínas tau/genética , Análisis de Varianza , Animales , Recuento de Células , Corteza Cerebral/patología , Modelos Animales de Enfermedad , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Prolina/genética , Tractos Piramidales/patología , Retina/metabolismo , Células Ganglionares de la Retina/patología , Serina/genética , Tauopatías/complicaciones , Tauopatías/genética , Vías Visuales/metabolismo , Vías Visuales/patología , gamma-Sinucleína/metabolismo
9.
Stem Cell Res Ther ; 6: 93, 2015 May 14.
Artículo en Inglés | MEDLINE | ID: mdl-25971252

RESUMEN

INTRODUCTION: Diffuse axonal injury is an extremely common type of traumatic brain injury encountered in motor vehicle crashes, sports injuries, and in combat. Although many cases of diffuse axonal injury result in chronic disability, there are no current treatments for this condition. Its basic lesion, traumatic axonal injury, has been aggressively modeled in primate and rodent animal models. The inexorable axonal and perikaryal degeneration and dysmyelination often encountered in traumatic axonal injury calls for regenerative therapies, including therapies based on stem cells and precursors. Here we explore the proof of concept that treatments based on transplants of human oligodendrocyte progenitor cells can replace or remodel myelin and, eventually, contribute to axonal regeneration in traumatic axonal injury. METHODS: We derived human oligodendrocyte progenitor cells from the human embryonic stem cell line H9, purified and characterized them. We then transplanted these human oligodendrocyte progenitor cells into the deep sensorimotor cortex next to the corpus callosum of nude rats subjected to traumatic axonal injury based on the impact acceleration model of Marmarou. We explored the time course and spatial distribution of differentiation and structural integration of these cells in rat forebrain. RESULTS: At the time of transplantation, over 90 % of human oligodendrocyte progenitor cells expressed A2B5, PDGFR, NG2, O4, Olig2 and Sox10, a profile consistent with their progenitor or early oligodendrocyte status. After transplantation, these cells survived well and migrated massively via the corpus callosum in both injured and uninjured brains. Human oligodendrocyte progenitor cells displayed a striking preference for white matter tracts and were contained almost exclusively in the corpus callosum and external capsule, the striatopallidal striae, and cortical layer 6. Over 3 months, human oligodendrocyte progenitor cells progressively matured into myelin basic protein(+) and adenomatous polyposis coli protein(+) oligodendrocytes. The injured environment in the corpus callosum of impact acceleration subjects tended to favor maturation of human oligodendrocyte progenitor cells. Electron microscopy revealed that mature transplant-derived oligodendrocytes ensheathed host axons with spiral wraps intimately associated with myelin sheaths. CONCLUSIONS: Our findings suggest that, instead of differentiating locally, human oligodendrocyte progenitor cells migrate massively along white matter tracts and differentiate extensively into ensheathing oligodendrocytes. These features make them appealing candidates for cellular therapies of diffuse axonal injury aiming at myelin remodeling and axonal protection or regeneration.


Asunto(s)
Lesiones Encefálicas/terapia , Oligodendroglía/citología , Trasplante de Células Madre , Células Madre/citología , Animales , Encéfalo/metabolismo , Encéfalo/patología , Diferenciación Celular , Línea Celular , Movimiento Celular , Supervivencia Celular , Modelos Animales de Enfermedad , Células Madre Embrionarias Humanas/citología , Humanos , Inmunohistoquímica , Masculino , Microscopía Electrónica , Proteína Básica de Mielina/metabolismo , Oligodendroglía/ultraestructura , Ratas , Ratas Desnudas , Receptor alfa de Factor de Crecimiento Derivado de Plaquetas/metabolismo , Células Madre/metabolismo
11.
Acta Neuropathol Commun ; 2: 153, 2014 Nov 25.
Artículo en Inglés | MEDLINE | ID: mdl-25422066

RESUMEN

INTRODUCTION: Blast injury to brain, a hundred-year old problem with poorly characterized neuropathology, has resurfaced as health concern in recent deployments in Iraq and Afghanistan. To characterize the neuropathology of blast injury, we examined the brains of veterans for the presence of amyloid precursor protein (APP)-positive axonal swellings typical of diffuse axonal injury (DAI) and compared them to healthy controls as well as controls with opiate overdose, anoxic-ischemic encephalopathy, and non-blast TBI (falls and motor vehicle crashes). RESULTS: In cases with blast history, we found APP (+) axonal abnormalities in several brain sites, especially the medial dorsal frontal white matter. In white matter, these abnormalities were featured primarily by clusters of axonal spheroids or varicosities in a honeycomb pattern with perivascular distribution. Axonal abnormalities colocalized with IBA1 (+) reactive microglia and had an appearance that was distinct from classical DAI encountered in TBI due to motor vehicle crashes. Opiate overdose cases also showed APP (+) axonal abnormalities, but the intensity of these lesions was lower compared to cases with blast histories and there was no clear association of such lesions with microglial activation. CONCLUSIONS: Our findings demonstrate that many cases with history of blast exposure are featured by APP (+) axonopathy that may be related to blast exposure, but an important role for opiate overdose, antemortem anoxia, and concurrent blunt TBI events in war theater or elsewhere cannot be discounted.


Asunto(s)
Traumatismos por Explosión/complicaciones , Encéfalo/metabolismo , Encéfalo/patología , Lesión Axonal Difusa/metabolismo , Lesión Axonal Difusa/patología , Accidentes por Caídas , Adolescente , Adulto , Precursor de Proteína beta-Amiloide/metabolismo , Axones/metabolismo , Axones/patología , Proteínas de Unión al Calcio , Proteínas de Unión al ADN/metabolismo , Lesión Axonal Difusa/etiología , Sobredosis de Droga/metabolismo , Sobredosis de Droga/patología , Femenino , Humanos , Masculino , Proteínas de Microfilamentos , Microglía/metabolismo , Microglía/patología , Persona de Mediana Edad , Vehículos a Motor , Trastornos Relacionados con Opioides/metabolismo , Trastornos Relacionados con Opioides/patología , Veteranos , Adulto Joven
12.
Regen Med ; 7(6): 785-97, 2012 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-23164079

RESUMEN

AIM: To explore the hypothesis that grafts of exogenous stem cells in the spinal cord of athymic rats or rats with transgenic motor neuron disease can induce endogenous stem cells and initiate intrinsic repair mechanisms that can be exploited in amyotrophic lateral sclerosis therapeutics. MATERIALS & METHODS: Human neural stem cells (NSCs) were transplanted into the lower lumbar spinal cord of healthy rats or rats with transgenic motor neuron disease to explore whether signals related to stem cells can initiate intrinsic repair mechanisms in normal and amyotrophic lateral sclerosis subjects. Patterns of migration and differentiation of NSCs in the gray and white matter, with emphasis on the central canal region and ependymal cell-driven neurogenesis, were analyzed. RESULTS: Findings suggest that there is extensive cross-signaling between transplanted NSCs and a putative neurogenic niche in the ependyma of the lower lumbar cord. The formation of a neuronal cord from NSC-derived cells next to ependyma suggests that this structure may serve a mediating or auxiliary role for ependymal induction. CONCLUSION: These findings raise the possibility that NSCs may stimulate endogenous neurogenesis and initiate intrinsic repair mechanisms in the lower spinal cord.


Asunto(s)
Diferenciación Celular , Movimiento Celular , Enfermedad de la Neurona Motora , Células-Madre Neurales , Médula Espinal/metabolismo , Nicho de Células Madre , Esclerosis Amiotrófica Lateral/patología , Esclerosis Amiotrófica Lateral/terapia , Animales , Epéndimo/metabolismo , Epéndimo/patología , Humanos , Enfermedad de la Neurona Motora/metabolismo , Enfermedad de la Neurona Motora/patología , Enfermedad de la Neurona Motora/terapia , Células-Madre Neurales/metabolismo , Células-Madre Neurales/trasplante , Ratas , Ratas Transgénicas , Médula Espinal/patología , Trasplante Heterólogo
13.
J Neuropathol Exp Neurol ; 70(5): 399-416, 2011 May.
Artículo en Inglés | MEDLINE | ID: mdl-21487304

RESUMEN

The increased use of explosives in recent wars has increased the number of veterans with blast injuries. Of particular interest is blast injury to the brain, and a key question is whether the primary overpressure wave of the blast is injurious or whether brain injury from blast is mostly due to secondary and tertiary effects. Using a shock tube generating shock waves comparable to open-field blast waves, we explored the effects of blast on parenchymatous organs of mice with emphasis on the brain. The main injuries in nonbrain organs were hemorrhages in the lung interstitium and alveolar spaces and hemorrhagic infarcts in liver, spleen, and kidney. Neuropathological and behavioral outcomes of blast were studied at mild blast intensity, that is, 68 ± 8 kPag (9.9 ± 1.2 psig) static pressure, 103 kPag (14.9 psig) total pressure and 183 ± 14 kPag (26.5 ± 2.1 psig) membrane rupture pressure. Under these conditions, we observed multifocal axonal injury, primarily in the cerebellum/brainstem, the corticospinal system, and the optic tract. We also found prolonged behavioral and motor abnormalities, including deficits in social recognition and spatial memory and in motor coordination. Shielding of the torso ameliorated axonal injury and behavioral deficits. These findings indicate that long CNS axon tracts are particularly vulnerable to the effects of blast, even at mild intensities that match the exposure of most veterans in recent wars. Prevention of some of these neurological effects by torso shielding may generate new ideas as to how to protect military and civilian populations in blast scenarios.


Asunto(s)
Traumatismos por Explosión/patología , Lesiones Encefálicas/patología , Encéfalo/patología , Degeneración Nerviosa/patología , Neuronas/patología , Animales , Axones/patología , Traumatismos por Explosión/fisiopatología , Encéfalo/fisiopatología , Lesiones Encefálicas/fisiopatología , Modelos Animales de Enfermedad , Inmunohistoquímica , Masculino , Memoria/fisiología , Ratones , Actividad Motora/fisiología , Degeneración Nerviosa/fisiopatología , Tinción con Nitrato de Plata
14.
Neurosci Lett ; 494(3): 222-6, 2011 May 02.
Artículo en Inglés | MEDLINE | ID: mdl-21402124

RESUMEN

Stem cells provide novel sources of cell therapies for motor neuron disease that have recently entered clinical trials. In the present study, we transplanted human neural stem cells (NSCs) into the ventral horn of both the lumbar (L4-L5) and cervical (C4-C5) protuberance of SOD1 G93A rats, in an effort to test the feasibility and general efficacy of a dual grafting paradigm addressing several muscle groups in the front limbs, hind limbs and the respiratory apparatus. Transplantation was done prior to the onset of motor neuron disease. Compared with animals that had received dead NSC grafts (serving as controls), rats with live NSCs grafted at the two spinal levels lived 17 days longer. Disease onset in dually grafted animals was delayed by 10 days compared to control animals. Disease duration in NSC-grafted animals was longer by 7 days compared to controls. Our results support the potential of NSC grafts at multiple levels of spinal cord as future cellular therapy for motor neuron disease.


Asunto(s)
Enfermedad de la Neurona Motora/cirugía , Células-Madre Neurales/trasplante , Médula Espinal/cirugía , Trasplante de Células Madre/métodos , Esclerosis Amiotrófica Lateral , Animales , Vértebras Cervicales , Modelos Animales de Enfermedad , Humanos , Región Lumbosacra , Ratas , Ratas Transgénicas , Médula Espinal/patología , Superóxido Dismutasa/genética , Superóxido Dismutasa-1
15.
Neurobiol Dis ; 41(2): 538-51, 2011 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-21074615

RESUMEN

Current experimental models of blast injuries used to study blast-induced neurotrauma (BINT) vary widely, which makes the comparison of the experimental results extremely challenging. Most of the blast injury models replicate the ideal Friedländer type of blast wave, without the capability to generate blast signatures with multiple shock fronts and refraction waves as seen in real-life conditions; this significantly reduces their clinical and military relevance. Here, we describe the pathophysiological consequences of graded blast injuries and BINT generated by a newly developed, highly controlled, and reproducible model using a modular, multi-chamber shock tube capable of tailoring pressure wave signatures and reproducing complex shock wave signatures seen in theater. While functional deficits due to blast exposure represent the principal health problem for today's warfighters, the majority of available blast models induces tissue destruction rather than mimic functional deficits. Thus, the main goal of our model is to reliably reproduce long-term neurological impairments caused by blast. Physiological parameters, functional (motor, cognitive, and behavioral) outcomes, and underlying molecular mechanisms involved in inflammation measured in the brain over the 30 day post-blast period showed this model is capable of reproducing major neurological changes of clinical BINT.


Asunto(s)
Traumatismos por Explosión/diagnóstico , Traumatismos por Explosión/patología , Lesiones Encefálicas/diagnóstico , Lesiones Encefálicas/patología , Presión/efectos adversos , Animales , Cámaras de Exposición Atmosférica/efectos adversos , Cámaras de Exposición Atmosférica/normas , Presión Atmosférica , Traumatismos por Explosión/fisiopatología , Lesiones Encefálicas/fisiopatología , Modelos Animales de Enfermedad , Ambiente Controlado , Masculino , Ratones , Ratones Endogámicos C57BL
16.
Tissue Eng Part A ; 17(5-6): 855-63, 2011 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-20973749

RESUMEN

The potential of human embryonic stem (ES) cells as experimental therapies for neuronal replacement has recently received considerable attention. In view of the organization of the mature nervous system into distinct neural circuits, key challenges of such therapies are the directed differentiation of human ES cell-derived neural precursors (NPs) into specific neuronal types and the directional growth of axons along specified trajectories. In the present study, we cultured human NPs derived from the NIH-approved ES line BGO1 on polycaprolactone fiber matrices of different diameter (i.e., nanofibers and microfibers) and orientation (i.e., aligned and random); fibers were coated with poly-L-ornithine/laminin to mimic the extracellular matrix and support the adhesion, viability, and differentiation of NPs. On aligned fibrous meshes, human NPs adopt polarized cell morphology with processes extending along the axis of the fiber, whereas NPs on plain tissue culture surfaces or random fiber substrates form nonpolarized neurite networks. Under differentiation conditions, human NPs cultured on aligned fibrous substrates show a higher rate of neuronal differentiation than other matrices; 62% and 86% of NPs become TUJ1 (+) early neurons on aligned micro- and nanofibers, respectively, whereas only 32% and 27% of NPs acquire the same fate on random micro- and nanofibers. Metabolic cell activity/viability studies reveal that fiber alignment and diameter also have an effect on NP viability, but only in the presence of mitogens. Our findings demonstrate that fibrous substrates serve as an artificial extracellular matrix and provide a microenviroment that influences key aspects of the neuronal differentiation of ES-derived NPs.


Asunto(s)
Diferenciación Celular/efectos de los fármacos , Células Madre Embrionarias/citología , Nanofibras/química , Células-Madre Neurales/citología , Células-Madre Neurales/efectos de los fármacos , Neuronas/citología , Poliésteres/farmacología , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Células Cultivadas , Células Madre Embrionarias/efectos de los fármacos , Células Madre Embrionarias/metabolismo , Técnica del Anticuerpo Fluorescente , Humanos , Proteínas de Filamentos Intermediarios/metabolismo , Mitógenos/farmacología , Nanofibras/ultraestructura , Proteínas del Tejido Nervioso/metabolismo , Nestina , Células-Madre Neurales/metabolismo , Células-Madre Neurales/ultraestructura , Péptidos/farmacología
18.
Stem Cells ; 27(10): 2414-26, 2009 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-19609935

RESUMEN

Stem cell grafts have been advocated as experimental treatments for neurological diseases by virtue of their ability to offer trophic support for injured neurons and, theoretically, to replace dead neurons. Human embryonic stem cells (HESCs) are a rich source of neural precursors (NPs) for grafting, but have been questioned for their tendency to form tumors. Here we studied the ability of HESC-derived NP grafts optimized for cell number and differentiation stage prior to transplantation, to survive and stably differentiate and integrate in the basal forebrain (neostriatum) of young adult nude rats over long periods of time (6 months). NPs were derived from adherent monolayer cultures of HESCs exposed to noggin. After transplantation, NPs showed a drastic reduction in mitotic activity and an avid differentiation into neurons that projected via major white matter tracts to a variety of forebrain targets. A third of NP-derived neurons expressed the basal forebrain-neostriatal marker dopamine-regulated and cyclic AMP-regulated phosphoprotein. Graft-derived neurons formed mature synapses with host postsynaptic structures, including dendrite shafts and spines. NPs inoculated in white matter tracts showed a tendency toward glial (primarily astrocytic) differentiation, whereas NPs inoculated in the ventricular epithelium persisted as nestin(+) precursors. Our findings demonstrate the long-term ability of noggin-derived human NPs to structurally integrate tumor-free into the mature mammalian forebrain, while maintaining some cell fate plasticity that is strongly influenced by particular central nervous system (CNS) niches.


Asunto(s)
Células Madre Embrionarias/fisiología , Células Madre Embrionarias/trasplante , Neostriado/fisiología , Trasplante de Células Madre/métodos , Células Madre/fisiología , Trasplante Heterólogo/fisiología , Animales , Proteínas Portadoras/metabolismo , Proteínas Portadoras/farmacología , Diferenciación Celular/fisiología , Línea Celular Tumoral , Supervivencia Celular/fisiología , Células Madre Embrionarias/citología , Supervivencia de Injerto/fisiología , Conos de Crecimiento/fisiología , Conos de Crecimiento/ultraestructura , Humanos , Masculino , Ratones , Ratones Endogámicos ICR , Neostriado/citología , Neostriado/cirugía , Vías Nerviosas/citología , Vías Nerviosas/fisiología , Neuroglía/citología , Neuroglía/metabolismo , Neuronas/citología , Neuronas/metabolismo , Fosfoproteínas/metabolismo , Ratas , Ratas Desnudas , Células Madre/citología , Sinapsis/ultraestructura
19.
J Neurosci Methods ; 179(2): 292-9, 2009 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-19428540

RESUMEN

Nitric oxide (NO) is a gas messenger with diverse physiological roles in the nervous system, from modulation of synaptic plasticity and neurogenesis to the mediation of neuronal death. NO production in the brain is catalyzed by three isoforms of NO synthase (NOS) including neuronal NOS (nNOS), inducible NOS and endothelial NOS. In this report, we demonstrate a method for in vitro and in vivo silencing of nNOS using RNAi strategies. Because of their efficiency in infecting postmitotic cells like neurons, lentiviral vectors were used as nNOS shRNA carriers. Of the siRNA sequences screened, one corresponding to exon 10 of the rat nNOS specifically and efficiently inhibited nNOS expression at the mRNA and protein level. In vitro experiments using rat cortical neurons showed the general efficacy of shRNA vectors in silencing constitutively expressed nNOS. To demonstrate the anatomical specificity of nNOS silencing in vivo, vectors were used to selectively knock-down the endogenous nNOS expression in cortical GABAergic interneurons of rat piriform cortex. Our findings show that the method reported here can achieve stable and highly effective nNOS suppression in an anatomically defined brain region. The ability of our nNOS silencing vectors to effectively and precisely silence nNOS expression shows their value as research tools for further studies of the role of nNOS in specific brain circuits. Furthermore, our findings raise the possibility for future considerations of lentiviral strategies as therapies for diseases of the nervous system involving NO neurotoxic cascades.


Asunto(s)
Regulación hacia Abajo/genética , Técnicas de Silenciamiento del Gen , Óxido Nítrico Sintasa de Tipo I/genética , Vías Olfatorias/enzimología , Interferencia de ARN/fisiología , Animales , Células Cultivadas , Regulación Enzimológica de la Expresión Génica/genética , Vectores Genéticos/genética , Interneuronas/enzimología , Lentivirus/genética , Óxido Nítrico/biosíntesis , Óxido Nítrico Sintasa de Tipo I/metabolismo , Vías Olfatorias/citología , Ratas , Ratas Sprague-Dawley , Ácido gamma-Aminobutírico/metabolismo
20.
J Comp Neurol ; 514(4): 297-309, 2009 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-19326469

RESUMEN

Cell replacement strategies for degenerative and traumatic diseases of the nervous system depend on the functional integration of grafted cells into host neural circuitry, a condition necessary for the propagation of physiological signals and, perhaps, targeting of trophic support to injured neurons. We have recently shown that human neural stem cell (NSC) grafts ameliorate motor neuron disease in SOD1 transgenic rodents. Here we study structural aspects of integration of neuronally differentiated human NSCs in the motor circuitry of SOD1 G93A rats. Human NSCs were grafted into the lumbar protuberance of 8-week-old SOD1 G93A rats; the results were compared to those on control Sprague-Dawley rats. Using pre-embedding immuno-electron microscopy, we found human synaptophysin (+) terminals contacting the perikarya and proximal dendrites of host alpha motor neurons. Synaptophysin (+) terminals had well-formed synaptic vesicles and were associated with membrane specializations primarily in the form of symmetrical synapses. To analyze the anatomy of motor circuits engaging differentiated NSCs, we injected the retrograde transneuronal tracer Bartha-pseudorabies virus (PRV) or the retrograde marker cholera toxin B (CTB) into the gastrocnemius muscle/sciatic nerve of SOD1 rats before disease onset and also into control rats. With this tracing, NSC-derived neurons were labeled with PRV but not CTB, a pattern suggesting that PRV entered NSC-derived neurons via transneuronal transfer from host motor neurons but not via direct transport from the host musculature. Our results indicate an advanced degree of structural integration, via functional synapses, of differentiated human NSCs into the segmental motor circuitry of SOD1-G93A rats.


Asunto(s)
Enfermedad de la Neurona Motora/fisiopatología , Neuronas/citología , Neuronas/fisiología , Médula Espinal/fisiología , Trasplante de Células Madre , Superóxido Dismutasa/genética , Animales , Diferenciación Celular , Toxina del Cólera , Modelos Animales de Enfermedad , Femenino , Humanos , Masculino , Enfermedad de la Neurona Motora/genética , Neuronas Motoras/fisiología , Músculo Esquelético/inervación , Músculo Esquelético/fisiología , Neuronas/ultraestructura , Ratas , Ratas Sprague-Dawley , Ratas Transgénicas , Nervio Ciático/fisiología , Médula Espinal/citología , Médula Espinal/ultraestructura , Superóxido Dismutasa-1 , Sinapsis/fisiología , Sinapsis/ultraestructura , Vesículas Sinápticas/fisiología , Sinaptofisina/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA