Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 38
Filtrar
Más filtros

Banco de datos
Tipo del documento
País de afiliación
Intervalo de año de publicación
1.
J Biomech Eng ; 145(6)2023 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-36562120

RESUMEN

Trauma to the brain is a biomechanical problem where the initiating event is a dynamic loading (blunt, inertial, blast) to the head. To understand the relationship between the mechanical parameters of the injury and the spatial and temporal deformation patterns in the brain, there is a need to develop a reusable and adaptable experimental traumatic brain injury (TBI) model that can measure brain motion under varying parameters. In this effort, we aim to directly measure brain deformation (strain and strain rates) in different brain regions in a human head model using a drop tower. METHODS: Physical head models consisting of a half, sagittal plane skull, brain, and neck were constructed and subjected to crown and frontal impacts at two impact speeds. All tests were recorded with a high-speed camera at 1000 frames per second. Motion of visual markers within brain surrogates were used to track deformations and calculate spatial strain histories in 6 brain regions of interest. Principal strains, strain rates and strain impulses were calculated and reported. RESULTS: Higher impact velocities corresponded to higher strain values across all impact scenarios. Crown impacts were characterized by high, long duration strains distributed across the parietal, frontal and hippocampal regions whereas frontal impacts were characterized by sharply rising and falling strains primarily found in the parietal, frontal, hippocampal and occipital regions. High strain rates were associated with short durations and impulses indicating fast but short-lived strains. 2.23 m/s (5 mph) crown impacts resulted in 53% of the brain with shear strains higher than 0.15 verses 32% for frontal impacts. CONCLUSIONS: The results reveal large differences in the spatial and temporal strain responses between crown and forehead impacts. Overall, the results suggest that for the same speed, crown impact leads to higher magnitude strain patterns than a frontal impact. The data provided by this model provides unique insight into the spatial and temporal deformation patterns that have not been provided by alternate surrogate models. The model can be used to investigate how anatomical, material and loading features and parameters can affect deformation patterns in specific regions of interest in the brain.


Asunto(s)
Traumatismos Craneocerebrales , Cráneo , Humanos , Fenómenos Biomecánicos , Cabeza/fisiología , Encéfalo
2.
Cells Tissues Organs ; 210(2): 105-117, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34198287

RESUMEN

Biomaterial-based scaffolds used in nerve conduits including channels for confining regenerating axons and 3-dimensional (3D) gels as substrates for growth have made improvements in models of nerve repair. Many biomaterial strategies, however, continue to fall short of autologous nerve grafts, which remain the current gold standard in repairing severe nerve lesions (<20 mm). Intraluminal nerve conduit fibers have also shown considerable promise in directing regenerating axons in vitro and in vivo and have gained increasing interest for nerve repair. It is unknown, however, how growing axons respond to a fiber when encountered in a 3D environment. In this study, we considered a construct consisting of a compliant collagen hydrogel matrix and a fiber component to assess contact-guided axon growth. We investigated preferential axon outgrowth on synthetic and natural polymer fibers by utilizing small-diameter microfibers of poly-L-lactic acid and type I collagen representing 2 different fiber stiffnesses. We found that axons growing freely in a 3D hydrogel culture preferentially attach, turn and follow fibers with outgrowth rates and distances that far exceed outgrowth in a hydrogel alone. Wet-spun type I collagen from rat tail tendon performed the best, associated with highly aligned and accelerated outgrowth. This study also evaluated the response of dorsal root ganglion neurons from adult rats to provide data more relevant to axon regenerative potential in nerve repair. We found that ECM treatments on fibers enhanced the regeneration of adult axons indicating that both the physical and biochemical presentation of the fibers are essential for enhancing axon guidance and growth.


Asunto(s)
Tejido Nervioso , Andamios del Tejido , Animales , Axones , Regeneración Nerviosa , Ratas , Ingeniería de Tejidos
3.
Glia ; 68(10): 2070-2085, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-32170885

RESUMEN

Myelin loss in the brain is a common occurrence in traumatic brain injury (TBI) that results from impact-induced acceleration forces to the head. Fast and abrupt head motions, either resulting from violent blows and/or jolts, cause rapid stretching of the brain tissue, and the long axons within the white matter tracts are especially vulnerable to such mechanical strain. Recent studies have shown that mechanotransduction plays an important role in regulating oligodendrocyte progenitors cell differentiation into oligodendrocytes. However, little is known about the impact of mechanical strain on mature oligodendrocytes and the stability of their associated myelin sheaths. We used an in vitro cellular stretch device to address these questions, as well as characterize a mechanotransduction mechanism that mediates oligodendrocyte responses. Mechanical stretch caused a transient and reversible myelin protein loss in oligodendrocytes. Cell death was not observed. Myelin protein loss was accompanied by an increase in intracellular Ca2+ and Erk1/2 activation. Chelating Ca2+ or inhibiting Erk1/2 activation was sufficient to block the stretch-induced loss of myelin protein. Further biochemical analyses revealed that the stretch-induced myelin protein loss was mediated by the release of Ca2+ from the endoplasmic reticulum (ER) and subsequent Ca2+ -dependent activation of Erk1/2. Altogether, our findings characterize an Erk1/2-dependent mechanotransduction mechanism in mature oligodendrocytes that de-stabilizes the myelination program.


Asunto(s)
Calcio/metabolismo , Sistema de Señalización de MAP Quinasas/fisiología , Mecanotransducción Celular/fisiología , Proteínas de la Mielina/deficiencia , Oligodendroglía/metabolismo , Animales , Animales Recién Nacidos , Quelantes del Calcio/farmacología , Ionóforos de Calcio/farmacología , Células Cultivadas , Corteza Cerebral/citología , Corteza Cerebral/efectos de los fármacos , Corteza Cerebral/metabolismo , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Mecanotransducción Celular/efectos de los fármacos , Vaina de Mielina/efectos de los fármacos , Vaina de Mielina/metabolismo , Oligodendroglía/efectos de los fármacos , Ratas
4.
J Neurosci Res ; 98(10): 2027-2044, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-32741029

RESUMEN

Millions suffer a traumatic brain injury (TBI) each year wherein the outcomes associated with injury can vary greatly between individuals. This study postulates that variations in each biomechanical parameter of a head trauma lead to differences in histological and behavioral outcome measures that should be considered collectively in assessing injury. While trauma severity typically scales with the magnitude of injury, much less is known about the effects of rate and duration of the mechanical insult. In this study, a newly developed voice-coil fluid percussion injury system was used to investigate the effects of injury rate and fluid percussion impulse on a collection of post-injury outcomes in male rats. Collectively the data suggest a potential shift in the specificity and progression of neuronal injury and function rather than a general scaling of injury severity. While a faster, shorter fluid percussion first presents as a mild TBI, neuronal loss and some behavioral tasks were similar among the slower and faster fluid percussion injuries. This study concludes that the sequelae of neuronal degeneration and behavioral outcomes are related to the complete temporal profile of the fluid percussion and do not scale only with peak pressure.


Asunto(s)
Fenómenos Biomecánicos/fisiología , Lesiones Traumáticas del Encéfalo/patología , Lesiones Traumáticas del Encéfalo/fisiopatología , Aprendizaje por Laberinto/fisiología , Animales , Lesiones Traumáticas del Encéfalo/psicología , Supervivencia Celular/fisiología , Masculino , Ratas , Ratas Sprague-Dawley , Factores de Tiempo
5.
Brain Behav Immun ; 59: 190-199, 2017 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-27614125

RESUMEN

Traumatic brain injury (TBI), even at mild levels, can activate matrix metalloproteinases (MMPs) and the induction of neuroinflammation that can result in blood brain barrier breakdown and neurodegeneration. MMP2 has a significant role in neuroinflammation and neurodegeneration by modulating the chemokine CXCL12α (stromal cell derived factor SDF-1α) signaling pathway and the induction of apoptosis. SDF-1α is responsible for cell proliferation and differentiation throughout the nervous system and is also implicated in various neurodegenerative illnesses. We hypothesized that TBI leads to MMP2 activation and cleavage of the N-terminal 4 amino acid residues of CXCL12α with generation of the highly neurotoxic fragment SDF-1(5-67). Using an in vitro stretch-injury model of rat neuronal cultures and the in vivo fluid percussion injury (FPI) model in rats, we found that oxidative stress has a significant role in the activation of MMP2. This is initiated by the induction of free radical generating enzyme NADPH oxidase 1 (NOX1). Induction of NOX1 correlated well with the signatures of oxidative stress marker, 4HNE in the injured neuronal cultures and cerebral cortex of rats. Further, using MMP2 siRNA and pharmacological MMP2 inhibitor, ARP100, we established the neurodegenerative role of MMP2 in cleaving SDF-1α to a neurotoxic fragment SDF-1(5-67). By immunofluorescence, western blotting and TUNEL experiments, we show the cleaved form of SDF leads to apoptotic cell death in neurons. This work identifies a new potential therapeutic target to reduce the complications of brain damage in TBI.


Asunto(s)
Lesiones Traumáticas del Encéfalo/enzimología , Quimiocina CXCL12/metabolismo , Metaloproteinasa 2 de la Matriz/metabolismo , Degeneración Nerviosa/enzimología , Degeneración Nerviosa/genética , Animales , Apoptosis/efectos de los fármacos , Lesiones Traumáticas del Encéfalo/genética , Caspasa 3/biosíntesis , Caspasa 3/genética , Supervivencia Celular/genética , Células Cultivadas , Quimiocina CXCL12/genética , Activación Enzimática , Técnicas de Silenciamiento del Gen , Metaloproteinasa 2 de la Matriz/genética , Inhibidores de la Metaloproteinasa de la Matriz/farmacología , NADPH Oxidasa 1/biosíntesis , NADPH Oxidasa 1/genética , Neuronas/efectos de los fármacos , Estrés Oxidativo , ARN Interferente Pequeño/farmacología , Ratas , Ratas Sprague-Dawley
6.
J Neurosci Res ; 92(10): 1350-1361, 2014 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-24799156

RESUMEN

Traumatic brain injury (TBI) can occur from physical trauma from a wide spectrum of insults ranging from explosions to falls. The biomechanics of the trauma can vary in key features, including the rate and magnitude of the insult. Although the effect of peak injury pressure on neurological outcome has been examined in the fluid percussion injury (FPI) model, it is unknown whether differences in rate of rise of the injury waveform modify cellular and physiological changes after TBI. Using a programmable FPI device, we examined juvenile rats subjected to a constant peak pressure at two rates of injury: a standard FPI rate of rise and a faster rate of rise to the same peak pressure. Immediate postinjury assessment identified fewer seizures and relatively brief loss of consciousness after fast-rise injuries than after standard-rise injuries at similar peak pressures. Compared with rats injured at standard rise, fewer silver-stained injured neuronal profiles and degenerating hilar neurons were observed 4-6 hr after fast-rise FPI. However, 1 week postinjury, both fast- and standard-rise FPI resulted in hilar cell loss and enhanced perforant path-evoked granule cell field excitability compared with sham controls. Notably, the extent of neuronal loss and increase in dentate excitability were not different between rats injured at fast and standard rates of rise to peak pressure. Our data indicate that reduced cellular damage and improved immediate neurological outcome after fast rising primary concussive injuries mask the severity of the subsequent cellular and neurophysiological pathology and may be unreliable as a predictor of prognosis.


Asunto(s)
Lesiones Encefálicas/etiología , Lesiones Encefálicas/patología , Hipocampo/patología , Percusión/efectos adversos , Análisis de Varianza , Animales , Animales Recién Nacidos , Lesiones Encefálicas/complicaciones , Lesiones Encefálicas/mortalidad , Ondas Encefálicas/fisiología , Distribución de Chi-Cuadrado , Modelos Animales de Enfermedad , Fluoresceínas , Hipocampo/metabolismo , Técnicas In Vitro , Neuronas/metabolismo , Neuronas/patología , Fosfopiruvato Hidratasa/metabolismo , Ratas , Convulsiones/etiología , Factores de Tiempo
7.
J Neurotrauma ; 41(11-12): 1450-1468, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38269433

RESUMEN

Blast-induced traumatic brain injury is typically regarded as a signature medical concern for military personnel who are exposed to explosive devices in active combat zones. However, soldiers as well as law enforcement personnel may be repeatedly exposed to low-level blasts during training sessions with heavy weaponries as part of combat readiness. Service personnel who sustain neurotrauma from repeated low-level blast (rLLB) exposure do not display overt pathological symptoms immediately but rather develop mild symptoms including cognitive impairments, attention deficits, mood changes, irritability, and sleep disturbances over time. Recently, we developed a rat model of rLLB by applying controlled low-level blast pressures (≤ 70 kPa) repeated five times successively to mimic the pressures experienced by service members. Using this model, we assessed anxiety-like symptoms, motor coordination, and short-term memory as a function of time. We also investigated the role of the NLRP3 inflammasome, a complex involved in chronic microglial activation and pro-inflammatory cytokine interleukin (IL)-1ß release, in rLLB-induced neuroinflammation. NLRP3 and caspase-1 protein expression, microglial activation, and IL-1ß release were examined as factors likely contributing to these neurobehavioral changes. Animals exposed to rLLB displayed acute and chronic short-term memory impairments and chronic anxiety-like symptoms accompanied by increased microglial activation, NLRP3 expression, and IL-1ß release. Treatment with MCC950, an NLRP3 inflammasome complex inhibitor, suppressed microglial activation, reduced NLRP3 expression and IL-1ß release, and improved short-term memory deficits after rLLB exposure. Collectively, this study demonstrates that rLLB induces chronic neurobehavioral and neuropathological changes by increasing NLRP3 inflammasome protein expression followed by cytokine IL-1ß release.


Asunto(s)
Traumatismos por Explosión , Modelos Animales de Enfermedad , Furanos , Indenos , Trastornos de la Memoria , Microglía , Proteína con Dominio Pirina 3 de la Familia NLR , Enfermedades Neuroinflamatorias , Ratas Sprague-Dawley , Sulfonamidas , Animales , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Proteína con Dominio Pirina 3 de la Familia NLR/antagonistas & inhibidores , Indenos/farmacología , Traumatismos por Explosión/complicaciones , Ratas , Trastornos de la Memoria/etiología , Trastornos de la Memoria/metabolismo , Enfermedades Neuroinflamatorias/metabolismo , Enfermedades Neuroinflamatorias/tratamiento farmacológico , Masculino , Microglía/metabolismo , Microglía/efectos de los fármacos , Sulfonamidas/farmacología , Furanos/farmacología , Lesiones Traumáticas del Encéfalo/metabolismo , Lesiones Traumáticas del Encéfalo/complicaciones , Inflamasomas/metabolismo
8.
Front Cell Neurosci ; 17: 1111403, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37066078

RESUMEN

Introduction: In vitro models of traumatic brain injury (TBI) commonly use neurons isolated from the central nervous system. Limitations with primary cortical cultures, however, can pose challenges to replicating some aspects of neuronal injury associated with closed head TBI. The known mechanisms of axonal degeneration from mechanical injury in TBI are in many ways similar to degenerative disease, ischemia, and spinal cord injury. It is therefore possible that the mechanisms that result in axonal degeneration in isolated cortical axons after in vitro stretch injury are shared with injured axons from different neuronal types. Dorsal root ganglia neurons (DRGN) are another neuronal source that may overcome some current limitations including remaining healthy in culture for long periods of time, ability to be isolated from adult sources, and myelinated in vitro. Methods: The current study sought to characterize the differential responses between cortical and DRGN axons to mechanical stretch injury associated with TBI. Using an in vitro model of traumatic axonal stretch injury, cortical and DRGN neurons were injured at a moderate (40% strain) and severe stretch (60% strain) and acute alterations in axonal morphology and calcium homeostasis were measured. Results: DRGN and cortical axons immediately form undulations in response to severe injury, experience similar elongation and recovery within 20 min after the initial injury, and had a similar pattern of degeneration over the first 24 h after injury. Additionally, both types of axons experienced comparable degrees of calcium influx after both moderate and severe injury that was prevented through pre-treatment with tetrodotoxin in cortical neurons and lidocaine in DRGNs. Similar to cortical axons, stretch injury also causes calcium activated proteolysis of sodium channel in DRGN axons that is prevented by treatment with lidocaine or protease inhibitors. Discussion: These findings suggest that DRGN axons share the early response of cortical neurons to a rapid stretch injury and the associated secondary injury mechanisms. The utility of a DRGN in vitro TBI model may allow future studies to explore TBI injury progression in myelinated and adult neurons.

9.
Crit Rev Biomed Eng ; 39(3): 241-59, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21967304

RESUMEN

Neural tissue engineering offers tremendous promise to combat the effects of disease, aging, or injury in the nervous system. Here we review neural tissue engineering with respect to the design of living tissue to directly replace damaged or diseased neural tissue, or to augment the capacity for nervous system regeneration and restore lost function. This article specifically addresses the development and implementation of tissue engineered three-dimensional (3-D) neural constructs and biohybridized neural-electrical microsystems. Living 3-D neural constructs may be "pre-engineered" in vitro with controlled neuroanatomical and functional characteristics for neuroregeneration, to recapitulate lost neuroanatomy, or to serve as a nervous tissue interface to a device. One application being investigated is developing constructs of axonal tracts that, upon transplantation, may facilitate nervous system repair by directly restoring lost connections or by serving as a targeted scaffold to promote host regeneration by exploiting axon-mediated axonal regeneration. In another application, living nervous tissue engineered constructs are being investigated to biohybridize neural-electrical interface microsystems for functional integration with the nervous system. With this design, in vivo neuritic ingrowth and synaptic integration may occur with the living component, potentially exploiting a more natural integration with the nonorganic interface. Overall, the use of tissue engineered 3-D neural constructs may significantly advance regeneration or device-based deficit mitigation in the nervous system that has not been achieved by non-tissue engineering approaches.


Asunto(s)
Técnicas de Cultivo de Célula , Regeneración Nerviosa/fisiología , Enfermedades del Sistema Nervioso/terapia , Neuronas/citología , Neuronas/fisiología , Ingeniería de Tejidos/métodos , Animales , Materiales Biocompatibles , Supervivencia Celular , Humanos , Microfluídica/instrumentación , Microfluídica/métodos , Ingeniería de Tejidos/instrumentación
10.
Crit Rev Biomed Eng ; 39(2): 81-124, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21488817

RESUMEN

Damage to the peripheral nervous system is surprisingly common and occurs primarily from trauma or a complication of surgery. Although recovery of nerve function occurs in many mild injuries, outcomes are often unsatisfactory following severe trauma. Nerve repair and regeneration presents unique clinical challenges and opportunities, and substantial contributions can be made through the informed application of biomedical engineering strategies. This article reviews the clinical presentations and classification of nerve injuries, in addition to the state of the art for surgical decision-making and repair strategies. This discussion presents specific challenges that must be addressed to realistically improve the treatment of nerve injuries and promote widespread recovery. In particular, nerve defects a few centimeters in length use a sensory nerve autograft as the standard technique; however, this approach is limited by the availability of donor nerve and comorbidity associated with additional surgery. Moreover, we currently have an inadequate ability to noninvasively assess the degree of nerve injury and to track axonal regeneration. As a result, wait-and-see surgical decisions can lead to undesirable and less successful "delayed" repair procedures. In this fight for time, degeneration of the distal nerve support structure and target progresses, ultimately blunting complete functional recovery. Thus, the most pressing challenges in peripheral nerve repair include the development of tissue-engineered nerve grafts that match or exceed the performance of autografts, the ability to noninvasively assess nerve damage and track axonal regeneration, and approaches to maintain the efficacy of the distal pathway and targets during the regenerative process. Biomedical engineering strategies can address these issues to substantially contribute at both the basic and applied levels, improving surgical management and functional recovery following severe peripheral nerve injury.


Asunto(s)
Ingeniería Biomédica/métodos , Regeneración Tisular Dirigida/métodos , Regeneración Nerviosa , Traumatismos de los Nervios Periféricos , Traumatismos del Sistema Nervioso/cirugía , Animales , Humanos , Nervios Periféricos/anatomía & histología , Nervios Periféricos/fisiopatología , Nervios Periféricos/cirugía , Ratas
11.
Front Neurol ; 11: 990, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33013653

RESUMEN

Blast exposure has been identified to be the most common cause for traumatic brain injury (TBI) in soldiers. Over the years, rodent models to mimic blast exposures and the behavioral outcomes observed in veterans have been developed extensively. However, blast tube design and varying experimental parameters lead to inconsistencies in the behavioral outcomes reported across research laboratories. This review aims to curate the behavioral outcomes reported in rodent models of blast TBI using shockwave tubes or open field detonations between the years 2008-2019 and highlight the important experimental parameters that affect behavioral outcome. Further, we discuss the role of various design parameters of the blast tube that can affect the nature of blast exposure experienced by the rodents. Finally, we assess the most common behavioral tests done to measure cognitive, motor, anxiety, auditory, and fear conditioning deficits in blast TBI (bTBI) and discuss the advantages and disadvantages of these tests.

12.
Exp Neurol ; 332: 113378, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-32553593

RESUMEN

Soldiers are often exposed to more than one traumatic brain injury (TBI) over the course of their service. In recent years, more attention has been drawn to the increased risk of neurological deficits caused by the 'blast plus' polytrauma, which typically is a blast trauma combined with other forms of TBI. In this study, we investigated the behavioral and neuronal deficits resulting from a blast plus injury involving a mild-moderate blast followed by a mild blunt trauma using the fluid percussion injury model. We identified that the blast injury predisposed the brain to increased cognitive deficits, chronic ventricular enlargement, increased neurodegeneration at acute time points and chronic neuronal loss. Interestingly, a single blast and single blunt injury differed in their onset and manifestation of cognitive and regional neuronal loss. We also identified the presence of cleaved RIP1 from caspase 8 mediated apoptosis in the blunt injury while the blast injury did not activate immediate apoptosis but led to decreased hilar neuronal survival over time.


Asunto(s)
Traumatismos por Explosión/psicología , Lesiones Traumáticas del Encéfalo/psicología , Enfermedades del Sistema Nervioso/psicología , Heridas no Penetrantes/psicología , Animales , Apoptosis/genética , Lesiones Traumáticas del Encéfalo/complicaciones , Caspasa 8/genética , Supervivencia Celular , Ventrículos Cerebrales/patología , Trastornos del Conocimiento/etiología , Trastornos del Conocimiento/psicología , Masculino , Memoria a Corto Plazo , Enfermedades del Sistema Nervioso/etiología , Neuronas/patología , Proteínas Serina-Treonina Quinasas/genética , Ratas , Ratas Sprague-Dawley , Proteína Serina-Treonina Quinasas de Interacción con Receptores , Memoria Espacial
13.
Front Neurosci ; 14: 600136, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33408609

RESUMEN

At birth, there are 100 billion neurons in the human brain, with functional neural circuits extending through the spine to the epidermis of the feet and toes. Following birth, limbs and vertebrae continue to grow by several orders of magnitude, forcing established axons to grow by up to 200 cm in length without motile growth cones. The leading regulatory paradigm suggests that biomechanical expansion of mitotic tissue exerts tensile force on integrated nervous tissue, which synchronizes ongoing growth of spanning axons. Here, we identify unique transcriptional changes in embryonic rat DRG and cortical neurons while the corresponding axons undergo physiological levels of controlled mechanical stretch in vitro. Using bioreactors containing cultured neurons, we recapitulated the peak biomechanical increase in embryonic rat crown-rump-length. Biologically paired sham and "stretch-grown" DRG neurons spanned 4.6- and 17.2-mm in length following static or stretch-induced growth conditions, respectively, which was associated with 456 significant changes in gene transcription identified by genome-wide cDNA microarrays. Eight significant genes found in DRG were cross-validated in stretch-grown cortical neurons by qRT-PCR, which included upregulation of Gpat3, Crem, Hmox1, Hpse, Mt1a, Nefm, Sprr1b, and downregulation of Nrep. The results herein establish a link between biomechanics and gene transcription in mammalian neurons, which elucidates the mechanism underlying long-term growth of axons, and provides a basis for new research in therapeutic axon regeneration.

14.
Am J Forensic Med Pathol ; 30(4): 339-45, 2009 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-19901817

RESUMEN

The risk of traumatic brain injury (TBI) while riding roller coasters has received substantial attention. Case reports of TBI around the time of riding roller coasters have led many medical professionals to assert that the high gravitational forces (G-forces) induced by roller coasters pose a significant TBI risk. Head injury research, however, has shown that G-forces alone cannot predict TBI. Established head injury criterions and procedures were employed to compare the potential of TBI between daily activities and roller coaster riding. Three-dimensional head motions were measured during 3 different roller coaster rides, a pillow fight, and car crash simulations. Data was analyzed and compared with published data, using similar analyses of head motions. An 8.05 m/s car crash lead to the largest head injury criterion measure of 28.1 and head impact power of 3.41, over 6 times larger than the roller coaster rides of 4.1 and 0.36. Notably, the linear and rotational components of head acceleration during roller coaster rides were milder than those induced by many common activities. As such, there appears to be an extremely low risk of TBI due to the head motions induced by roller coaster rides.


Asunto(s)
Lesiones Encefálicas/fisiopatología , Gravitación , Movimientos de la Cabeza/fisiología , Aceleración , Accidentes de Tránsito , Adolescente , Adulto , Ropa de Cama y Ropa Blanca , Fenómenos Biomecánicos , Niño , Femenino , Humanos , Imagenología Tridimensional , Puntaje de Gravedad del Traumatismo , Masculino , Modelos Biológicos
15.
Rev Sci Instrum ; 90(7): 075116, 2019 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-31370428

RESUMEN

Blast simulators facilitate the creation of shock waves and measurement of pressure morphology in a controlled laboratory setting and are currently a vital model for replicating blast-induced neurotrauma. Due to the maintenance and operation cost of conventional blast simulators, we developed a pneumatic, table-top, gas-driven shock tube to test an alternative method of shock wave generation using a membrane-less driver section. Its unique operational mechanism based on air gun technology does not rely on a plastic membrane rupture for the generation of pressure pulses, allowing the simulator to be quickly reset and thus decreasing the experimental turnaround time. The focus of this study is to demonstrate that this proof-of-concept device can generate shock waves with diverse characteristics based on the selection of driver gas, driver pressurization, and driven section material. Pressure waves were generated using compressed nitrogen or helium at 15 psig and 80 psig and were analyzed based on their velocity and profile shape characteristics. At 15 psig, independent of the type of driver gas, driver pressurization, and driven section material, pressure pulses travelled at sonic velocities. At 80 psig, generation of shock waves was observed in all conditions. The choice of the driver gas affected the velocities of the resulting pressure waves and the shape of pressure waveforms, particularly the peak overpressure and rise time values. Our results demonstrate that depending on the selection of driver gas and magnitude of driver pressurization, the shock wave signatures can be controlled and altered using a piston-based driver section.

16.
Mol Neurobiol ; 56(8): 5332-5345, 2019 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-30603958

RESUMEN

Traumatic brain injury (TBI) contributes a major cause of death, disability, and mental health disorders. Most TBI patients suffer long-term post-traumatic stress disorder, cognitive dysfunction, and disability. The underlying molecular and cellular mechanisms of such neuropathology progression in TBI remain elusive. In part, it is due to non-standardized classification of mild, moderate, and severe injury in various animal models of TBI. Thus, a better diagnosis and treatment requires a better understanding of the injury mechanisms in a well-defined severity of mild, moderate, and severe injury in different models that may potentially reflect the various types of human brain injuries. The purpose of this review article is to highlight the classification of mild, moderate, and severe injury in various animal models of TBI with special focus on mixed injury that represents a translational concussive head injury. We will classify animal models of TBI broadly into focal injury, diffuse injury, and mixed injury. Focal injury, a localized injury, is represented by animal models of controlled cortical impact, penetrating ballistic-like brain injury, and Feeney or Shohami weight drop injury. A global diffuse injury is best represented by shock tube model of primary blast injury, and Marmarou or Maryland weight drop model. A mixed injury consists of focal and diffuse injury which reproduces the concussive clinical syndrome, and it is best studied in animal model of lateral fluid percussion injury.


Asunto(s)
Lesiones Traumáticas del Encéfalo/patología , Índice de Severidad de la Enfermedad , Animales , Conducta Animal , Traumatismos por Explosión/patología , Conmoción Encefálica/patología , Modelos Animales de Enfermedad , Humanos
17.
J Neural Eng ; 5(4): 374-84, 2008 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-18827311

RESUMEN

Neural-electrical interface platforms are being developed to extracellularly monitor neuronal population activity. Polyaniline-based electrically conducting polymer fibers are attractive substrates for sustained functional interfaces with neurons due to their flexibility, tailored geometry and controlled electro-conductive properties. In this study, we addressed the neurobiological considerations of utilizing small diameter (<400 microm) fibers consisting of a blend of electrically conductive polyaniline and polypropylene (PA-PP) as the backbone of encapsulated tissue-engineered neural-electrical relays. We devised new approaches to promote survival, adhesion and neurite outgrowth of primary dorsal root ganglion neurons on PA-PP fibers. We attained a greater than ten-fold increase in the density of viable neurons on fiber surfaces to approximately 700 neurons mm(-2) by manipulating surrounding surface charges to bias settling neuronal suspensions toward fibers coated with cell-adhesive ligands. This stark increase in neuronal density resulted in robust neuritic extension and network formation directly along the fibers. Additionally, we encapsulated these neuronal networks on PA-PP fibers using agarose to form a protective barrier while potentially facilitating network stability. Following encapsulation, the neuronal networks maintained integrity, high viability (>85%) and intimate adhesion to PA-PP fibers. These efforts accomplished key prerequisites for the establishment of functional electrical interfaces with neuronal populations using small diameter PA-PP fibers-specifically, improved neurocompatibility, high-density neuronal adhesion and neuritic network development directly on fiber surfaces.


Asunto(s)
Modelos Neurológicos , Neuronas/fisiología , Polímeros/química , Ingeniería de Tejidos , Compuestos de Anilina/química , Animales , Materiales Biocompatibles , Adhesión Celular/fisiología , Supervivencia Celular , Colágeno/química , Conductividad Eléctrica , Electrónica , Ganglios Espinales/citología , Ganglios Espinales/fisiología , Hidrogeles/química , Microscopía Confocal , Microscopía de Contraste de Fase , Red Nerviosa/fisiología , Neuritas/fisiología , Polipropilenos/química , Ratas , Ratas Sprague-Dawley , Sefarosa/química
18.
J Neurosurg ; 108(2): 343-7, 2008 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-18240932

RESUMEN

OBJECT: Although neuron transplantation to repair the nervous system has shown promise in animal models, there are few practical sources of viable neurons for clinical application and insufficient approaches to bridge extensive nerve damage in patients. Therefore, the authors sought a clinically relevant source of neurons that could be engineered into transplantable nervous tissue constructs. The authors chose to evaluate human dorsal root ganglion (DRG) neurons due to their robustness in culture. METHODS: Cervical DRGs were harvested from 16 live patients following elective ganglionectomies, and thoracic DRGs were harvested from 4 organ donor patients. Following harvest, the DRGs were digested in a dispase-collagenase treatment to dissociate neurons for culture. In addition, dissociated human DRG neurons were placed in a specially designed axon expansion chamber that induces continuous mechanical tension on axon fascicles spanning 2 populations of neurons originally plated approximately 100 microm apart. RESULTS: The adult human DRG neurons, positively identified by neuronal markers, survived at least 3 months in culture while maintaining the ability to generate action potentials. Stretch-growth of axon fascicles in the expansion chamber occurred at the rate of 1 mm/day to a length of 1 cm, creating the first engineered living human nervous tissue constructs. CONCLUSIONS: These data demonstrate the promise of adult human DRG neurons as an alternative transplant material due to their availability, viability, and capacity to be engineered. Also, these data show the feasibility of harvesting DRGs from living patients as a source of neurons for autologous transplant as well as from organ donors to serve as an allograft source of neurons.


Asunto(s)
Ganglios Espinales/citología , Neuronas/citología , Ingeniería de Tejidos/métodos , Andamios del Tejido , Recolección de Tejidos y Órganos , Potenciales de Acción/fisiología , Adulto , Axones/fisiología , Axones/ultraestructura , Técnicas de Cultivo de Célula , Supervivencia Celular , Preescolar , Estudios de Factibilidad , Femenino , Humanos , Masculino , Persona de Mediana Edad , Neuronas/fisiología , Neuronas/trasplante , Estrés Mecánico
19.
Neurol Res ; 30(10): 1063-7, 2008 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-19079981

RESUMEN

OBJECTIVE: We have previously described a technique developed in our laboratory to create transplantable living axon tracts of several centimeters in length. In this paper, we describe how these engineered neural tissue constructs can be used to create a novel neuroelectrical interface with the regenerating peripheral nervous system, to potentially enable afferent and efferent communications with prosthetic devices. METHODS: Using continuous mechanical tension, we have generated axon tracts of up to 10 cm in length, spanning two populations of neurons in vitro. We have now adapted this stretch-growth paradigm to include a mechanically compliant multi-electrode array that is attached to one of the neuron populations. Once the desired axon length has been reached, the neuroelectrode construct is completely embedded in a supportive hydrogel matrix and affixed to the transected sciatic nerve. RESULTS: Building upon our previous work with peripheral nerve repair, we have designed our neural interface to ensure transplant stability and firm attachment to the electrode array substrate. DISCUSSION: Our preliminary findings indicate that the interface not only maintains its orientation, but also is conducive to host nerve ingrowth. Our ongoing analysis seeks to characterize transplanted neuronal survival, synaptic integration, and functional connectivity. This research provides an opportunity to evaluate an entirely new approach in restoring motor and sensory functions of patients with peripheral nerve damage.


Asunto(s)
Regeneración Tisular Dirigida/métodos , Regeneración Nerviosa/fisiología , Enfermedades del Sistema Nervioso Periférico/cirugía , Ingeniería de Tejidos/métodos , Interfaz Usuario-Computador , Animales , Humanos
20.
Prog Brain Res ; 161: 143-69, 2007.
Artículo en Inglés | MEDLINE | ID: mdl-17618975

RESUMEN

Mild to moderate cases of traumatic brain injury (TBI) are very common, but are not always associated with the overt pathophysiogical changes seen following severe trauma. While neuronal death has been considered to be a major factor, the pervasive memory, cognitive and motor function deficits suffered by many mild TBI patients do not always correlate with cell loss. Therefore, we assert that functional impairment may result from alterations in surviving neurons. Current research has begun to explore CNS synaptic circuits after traumatic injury. Here we review significant findings made using in vivo and in vitro models of TBI that provide mechanistic insight into injury-induced alterations in synaptic electrophysiology. In the hippocampus, research now suggests that TBI regionally alters the delicate balance between excitatory and inhibitory neurotransmission in surviving neurons, disrupting the normal functioning of synaptic circuits. In another approach, a simplified model of neuronal stretch injury in vitro, has been used to directly explore how injury impacts the physiology and cell biology of neurons in the absence of alterations in blood flow, blood brain barrier integrity, or oxygenation associated with in vivo models of brain injury. This chapter discusses how these two models alter excitatory and inhibitory synaptic transmission at the receptor, cellular and circuit levels and how these alterations contribute to cognitive impairment and a reduction in seizure threshold associated with human concussive brain injury.


Asunto(s)
Sistema Nervioso Central/lesiones , Sistema Nervioso Central/fisiopatología , Traumatismos del Sistema Nervioso/fisiopatología , Animales , Encéfalo/patología , Lesiones Encefálicas/fisiopatología , Modelos Animales de Enfermedad , Electrofisiología , Humanos , Plasticidad Neuronal/fisiología , Receptores de Neurotransmisores/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA