Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 103
Filtrar
Mais filtros

Base de dados
País/Região como assunto
Tipo de documento
País de afiliação
Intervalo de ano de publicação
1.
Proc Natl Acad Sci U S A ; 117(29): 17269-17277, 2020 07 21.
Artigo em Inglês | MEDLINE | ID: mdl-32611818

RESUMO

Traumatic brain injury (TBI) is the strongest environmental risk factor for the accelerated development of neurodegenerative diseases. There are currently no therapeutics to address this due to lack of insight into mechanisms of injury progression, which are challenging to study in mammalian models. Here, we have developed and extensively characterized a head-specific approach to TBI in Drosophila, a powerful genetic system that shares many conserved genes and pathways with humans. The Drosophila TBI (dTBI) device inflicts mild, moderate, or severe brain trauma by precise compression of the head using a piezoelectric actuator. Head-injured animals display features characteristic of mammalian TBI, including severity-dependent ataxia, life span reduction, and brain degeneration. Severe dTBI is associated with cognitive decline and transient glial dysfunction, and stimulates antioxidant, proteasome, and chaperone activity. Moreover, genetic or environmental augmentation of the stress response protects from severe dTBI-induced brain degeneration and life span deficits. Together, these findings present a tunable, head-specific approach for TBI in Drosophila that recapitulates mammalian injury phenotypes and underscores the ability of the stress response to mitigate TBI-induced brain degeneration.


Assuntos
Lesões Encefálicas Traumáticas/metabolismo , Encéfalo/metabolismo , Drosophila/fisiologia , Neuroglia/metabolismo , Animais , Comportamento Animal , Lesões Encefálicas Traumáticas/diagnóstico por imagem , Lesões Encefálicas Traumáticas/patologia , Modelos Animais de Doenças , Cabeça , Humanos , Masculino , Doenças Neurodegenerativas/metabolismo , Neuroglia/patologia , Estresse Fisiológico
2.
Nano Lett ; 22(11): 4315-4324, 2022 06 08.
Artigo em Inglês | MEDLINE | ID: mdl-35588529

RESUMO

Extracellular vesicles (EVs) have attracted enormous attention for their diagnostic and therapeutic potential. However, it has proven challenging to achieve the sensitivity to detect individual nanoscale EVs, the specificity to distinguish EV subpopulations, and a sufficient throughput to study EVs among an enormous background. To address this fundamental challenge, we developed a droplet-based optofluidic platform to quantify specific individual EV subpopulations at high throughput. The key innovation of our platform is parallelization of droplet generation, processing, and analysis to achieve a throughput (∼20 million droplets/min) more than 100× greater than typical microfluidics. We demonstrate that the improvement in throughput enables EV quantification at a limit of detection = 9EVs/µL, a >100× improvement over gold standard methods. Additionally, we demonstrate the clinical potential of this system by detecting human EVs in complex media. Building on this work, we expect this technology will allow accurate quantification of rare EV subpopulations for broad biomedical applications.


Assuntos
Vesículas Extracelulares , Ensaio de Imunoadsorção Enzimática , Humanos , Microfluídica
3.
Neuroimage ; 251: 119002, 2022 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-35176490

RESUMO

The brain is a complex network consisting of neuron cell bodies in the gray matter and their axonal projections, forming the white matter tracts. These neurons are supported by an equally complex vascular network as well as glial cells. Traumatic brain injury (TBI) can lead to the disruption of the structural and functional brain networks due to disruption of both neuronal cell bodies in the gray matter as well as their projections and supporting cells. To explore how an impact can alter the function of brain networks, we integrated a finite element (FE) brain mechanics model with linked models of brain dynamics (Kuramoto oscillator) and vascular perfusion (Balloon-Windkessel) in this study. We used empirical resting-state functional magnetic resonance imaging (MRI) data to optimize the fit of our brain dynamics and perfusion models to clinical data. Results from the FE model were used to mimic injury in these optimized brain dynamics models: injury to the nodes (gray matter) led to a decrease in the nodal oscillation frequency, while damage to the edges (axonal connections/white matter) progressively decreased coupling among connected nodes. A total of 53 cases, including 33 non-injurious and 20 concussive head impacts experienced by professional American football players were simulated using this integrated model. We examined the correlation of injury outcomes with global measures of structural connectivity, neural dynamics, and functional connectivity of the brain networks when using different lesion methods. Results show that injurious head impacts cause significant alterations in global network topology regardless of lesion methods. Changes between the disrupted and healthy functional connectivity (measured by Pearson correlation) consistently correlated well with injury outcomes (AUC≥0.75), although the predictive performance is not significantly different (p>0.05) to that of traditional kinematic measures (angular acceleration). Intriguingly, our lesion model for gray matter damage predicted increases in global efficiency and clustering coefficient with increases in injury risk, while disrupting axonal connections led to lower network efficiency and clustering. When both injury mechanisms were combined into a single injury prediction model, the injury prediction performance depended on the thresholds used to determine neurodegeneration and mechanical tolerance for axonal injury. Together, these results point towards complex effects of mechanical trauma to the brain and provide a new framework for understanding brain injury at a causal mechanistic level and developing more effective diagnostic methods and therapeutic interventions.


Assuntos
Lesões Encefálicas Traumáticas , Substância Branca , Fenômenos Biomecânicos , Encéfalo/patologia , Lesões Encefálicas Traumáticas/patologia , Simulação por Computador , Humanos , Imageamento por Ressonância Magnética , Redes Neurais de Computação , Substância Branca/patologia
4.
Hippocampus ; 32(3): 231-250, 2022 03.
Artigo em Inglês | MEDLINE | ID: mdl-34978378

RESUMO

Proper function of the hippocampus is critical for executing cognitive tasks such as learning and memory. Traumatic brain injury (TBI) and other neurological disorders are commonly associated with cognitive deficits and hippocampal dysfunction. Although there are many existing models of individual subregions of the hippocampus, few models attempt to integrate the primary areas into one system. In this work, we developed a computational model of the hippocampus, including the dentate gyrus, CA3, and CA1. The subregions are represented as an interconnected neuronal network, incorporating well-characterized ex vivo slice electrophysiology into the functional neuron models and well-documented anatomical connections into the network structure. In addition, since plasticity is foundational to the role of the hippocampus in learning and memory as well as necessary for studying adaptation to injury, we implemented spike-timing-dependent plasticity among the synaptic connections. Our model mimics key features of hippocampal activity, including signal frequencies in the theta and gamma bands and phase-amplitude coupling in area CA1. We also studied the effects of spike-timing-dependent plasticity impairment, a potential consequence of TBI, in our model and found that impairment decreases broadband power in CA3 and CA1 and reduces phase coherence between these two subregions, yet phase-amplitude coupling in CA1 remains intact. Altogether, our work demonstrates characteristic hippocampal activity with a scaled network model of spiking neurons and reveals the sensitive balance of plasticity mechanisms in the circuit through one manifestation of mild traumatic injury.


Assuntos
Concussão Encefálica , Transtornos Cognitivos , Hipocampo , Humanos , Aprendizagem , Plasticidade Neuronal/fisiologia , Neurônios
5.
Neural Comput ; 33(1): 67-95, 2021 01.
Artigo em Inglês | MEDLINE | ID: mdl-33253030

RESUMO

Mild traumatic brain injury (mTBI) presents a significant health concern with potential persisting deficits that can last decades. Although a growing body of literature improves our understanding of the brain network response and corresponding underlying cellular alterations after injury, the effects of cellular disruptions on local circuitry after mTBI are poorly understood. Our group recently reported how mTBI in neuronal networks affects the functional wiring of neural circuits and how neuronal inactivation influences the synchrony of coupled microcircuits. Here, we utilized a computational neural network model to investigate the circuit-level effects of N-methyl D-aspartate receptor dysfunction. The initial increase in activity in injured neurons spreads to downstream neurons, but this increase was partially reduced by restructuring the network with spike-timing-dependent plasticity. As a model of network-based learning, we also investigated how injury alters pattern acquisition, recall, and maintenance of a conditioned response to stimulus. Although pattern acquisition and maintenance were impaired in injured networks, the greatest deficits arose in recall of previously trained patterns. These results demonstrate how one specific mechanism of cellular-level damage in mTBI affects the overall function of a neural network and point to the importance of reversing cellular-level changes to recover important properties of learning and memory in a microcircuit.


Assuntos
Concussão Encefálica/fisiopatologia , Transtornos da Memória/fisiopatologia , Rememoração Mental/fisiologia , Redes Neurais de Computação , Receptores de N-Metil-D-Aspartato/fisiologia , Encéfalo/fisiopatologia , Humanos , Rede Nervosa/fisiopatologia , Plasticidade Neuronal/fisiologia , Aprendizado de Máquina não Supervisionado
6.
J Biomech Eng ; 142(9)2020 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-32266930

RESUMO

With an increasing focus on long-term consequences of concussive brain injuries, there is a new emphasis on developing tools that can accurately predict the mechanical response of the brain to impact loading. Although finite element models (FEM) estimate the brain response under dynamic loading, these models are not capable of delivering rapid (∼seconds) estimates of the brain's mechanical response. In this study, we develop a multibody spring-mass-damper model that estimates the regional motion of the brain to rotational accelerations delivered either about one anatomic axis or across three orthogonal axes simultaneously. In total, we estimated the deformation across 120 locations within a 50th percentile human brain. We found the multibody model (MBM) correlated, but did not precisely predict, the computed finite element response (average relative error: 18.4 ± 13.1%). We used machine learning (ML) to combine the prediction from the MBM and the loading kinematics (peak rotational acceleration, peak rotational velocity) and significantly reduced the discrepancy between the MBM and FEM (average relative error: 9.8 ± 7.7%). Using an independent sports injury testing set, we found the hybrid ML model also correlated well with predictions from a FEM (average relative error: 16.4 ± 10.2%). Finally, we used this hybrid MBM-ML approach to predict strains appearing in different locations throughout the brain, with average relative error estimates ranging from 8.6% to 25.2% for complex, multi-axial acceleration loading. Together, these results show a rapid and reasonably accurate method for predicting the mechanical response of the brain for single and multiplanar inputs, and provide a new tool for quickly assessing the consequences of impact loading throughout the brain.


Assuntos
Encéfalo , Análise de Elementos Finitos , Fenômenos Biomecânicos , Lesões Encefálicas , Modelos Biológicos , Rotação
7.
Neurobiol Dis ; 119: 13-25, 2018 11.
Artigo em Inglês | MEDLINE | ID: mdl-30031156

RESUMO

Cytosolic PSD-95 interactor (cypin), the primary guanine deaminase in the brain, plays key roles in shaping neuronal circuits and regulating neuronal survival. Despite this pervasive role in neuronal function, the ability for cypin activity to affect recovery from acute brain injury is unknown. A key barrier in identifying the role of cypin in neurological recovery is the absence of pharmacological tools to manipulate cypin activity in vivo. Here, we use a small molecule screen to identify two activators and one inhibitor of cypin's guanine deaminase activity. The primary screen identified compounds that change the initial rate of guanine deamination using a colorimetric assay, and secondary screens included the ability of the compounds to protect neurons from NMDA-induced injury and NMDA-induced decreases in frequency and amplitude of miniature excitatory postsynaptic currents. Hippocampal neurons pretreated with activators preserved electrophysiological function and survival after NMDA-induced injury in vitro, while pretreatment with the inhibitor did not. The effects of the activators were abolished when cypin was knocked down. Administering either cypin activator directly into the brain one hour after traumatic brain injury significantly reduced fear conditioning deficits 5 days after injury, while delivering the cypin inhibitor did not improve outcome after TBI. Together, these data demonstrate that cypin activation is a novel approach for improving outcome after TBI and may provide a new pathway for reducing the deficits associated with TBI in patients.


Assuntos
Lesões Encefálicas Traumáticas/metabolismo , Lesões Encefálicas Traumáticas/prevenção & controle , Guanina Desaminase/metabolismo , Animais , Lesões Encefálicas Traumáticas/fisiopatologia , Células COS , Células Cultivadas , Chlorocebus aethiops , Dimetil Sulfóxido/farmacologia , Medo/efeitos dos fármacos , Medo/fisiologia , Guanina Desaminase/antagonistas & inibidores , Compostos Heterocíclicos com 3 Anéis/farmacologia , Hipocampo/efeitos dos fármacos , Hipocampo/metabolismo , Hipocampo/fisiopatologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , N-Metilaspartato/farmacologia , Técnicas de Cultura de Órgãos , Ratos
8.
Acta Neuropathol ; 135(5): 711-726, 2018 05.
Artigo em Inglês | MEDLINE | ID: mdl-29460006

RESUMO

Although concussion is now recognized as a major health issue, its non-lethal nature has limited characterization of the underlying pathophysiology. In particular, potential neuropathological changes have typically been inferred from non-invasive techniques or post-mortem examinations of severe traumatic brain injury (TBI). Here, we used a swine model of head rotational acceleration based on human concussion to examine blood-brain barrier (BBB) integrity after injury in association with diffuse axonal injury and glial responses. We then determined the potential clinical relevance of the swine concussion findings through comparisons with pathological changes in human severe TBI, where post-mortem examinations are possible. At 6-72 h post-injury in swine, we observed multifocal disruption of the BBB, demonstrated by extravasation of serum proteins, fibrinogen and immunoglobulin-G, in the absence of hemorrhage or other focal pathology. BBB disruption was observed in a stereotyped distribution consistent with biomechanical insult. Specifically, extravasated serum proteins were frequently observed at interfaces between regions of tissue with differing material properties, including the gray-white boundary, periventricular and subpial regions. In addition, there was substantial overlap of BBB disruption with regions of axonal pathology in the white matter. Acute perivascular cellular uptake of blood-borne proteins was observed to be prominent in astrocytes (GFAP-positive) and neurons (MAP-2-positive), but not microglia (IBA1-positive). Parallel examination of human severe TBI revealed similar patterns of serum extravasation and glial uptake of serum proteins, but to a much greater extent than in the swine model, attributed to the higher injury severity. These data suggest that BBB disruption represents a new and important pathological feature of concussion.


Assuntos
Barreira Hematoencefálica/patologia , Barreira Hematoencefálica/fisiopatologia , Concussão Encefálica/patologia , Concussão Encefálica/fisiopatologia , Adolescente , Adulto , Animais , Astrócitos/patologia , Astrócitos/fisiologia , Biomarcadores/sangue , Fenômenos Biomecânicos , Permeabilidade Capilar , Modelos Animais de Doenças , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Suínos , Porco Miniatura , Adulto Jovem
9.
J Neurosci ; 34(12): 4200-13, 2014 Mar 19.
Artigo em Inglês | MEDLINE | ID: mdl-24647941

RESUMO

Alterations in the activity of neural circuits are a common consequence of traumatic brain injury (TBI), but the relationship between single-neuron properties and the aggregate network behavior is not well understood. We recently reported that the GluN2B-containing NMDA receptors (NMDARs) are key in mediating mechanical forces during TBI, and that TBI produces a complex change in the functional connectivity of neuronal networks. Here, we evaluated whether cell-to-cell heterogeneity in the connectivity and aggregate contribution of GluN2B receptors to [Ca(2+)]i before injury influenced the functional rewiring, spontaneous activity, and network plasticity following injury using primary rat cortical dissociated neurons. We found that the functional connectivity of a neuron to its neighbors, combined with the relative influx of calcium through distinct NMDAR subtypes, together contributed to the individual neuronal response to trauma. Specifically, individual neurons whose [Ca(2+)]i oscillations were largely due to GluN2B NMDAR activation lost many of their functional targets 1 h following injury. In comparison, neurons with large GluN2A contribution or neurons with high functional connectivity both independently protected against injury-induced loss in connectivity. Mechanistically, we found that traumatic injury resulted in increased uncorrelated network activity, an effect linked to reduction of the voltage-sensitive Mg(2+) block of GluN2B-containing NMDARs. This uncorrelated activation of GluN2B subtypes after injury significantly limited the potential for network remodeling in response to a plasticity stimulus. Together, our data suggest that two single-cell characteristics, the aggregate contribution of NMDAR subtypes and the number of functional connections, influence network structure following traumatic injury.


Assuntos
Lesões Encefálicas/metabolismo , Rede Nervosa/metabolismo , Plasticidade Neuronal/fisiologia , Neurônios/metabolismo , Receptores de N-Metil-D-Aspartato/metabolismo , Animais , Lesões Encefálicas/fisiopatologia , Córtex Cerebral/metabolismo , Córtex Cerebral/fisiopatologia , Rede Nervosa/fisiopatologia , Fenótipo , Ratos , Ratos Sprague-Dawley , Receptores de N-Metil-D-Aspartato/genética
10.
Brain ; 136(Pt 1): 65-80, 2013 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-23293266

RESUMO

The recent public awareness of the incidence and possible long-term consequences of traumatic brain injury only heightens the need to develop effective approaches for treating this neurological disease. In this report, we identify a new therapeutic target for traumatic brain injury by studying the role of astrocytes, rather than neurons, after neurotrauma. We use in vivo multiphoton imaging and show that mechanical forces during trauma trigger intercellular calcium waves throughout the astrocytes, and these waves are mediated by purinergic signalling. Subsequent in vitro screening shows that astrocyte signalling through the 'mechanical penumbra' affects the activity of neural circuits distant from the injury epicentre, and a reduction in the intercellular calcium waves within astrocytes restores neural activity after injury. In turn, the targeting of different purinergic receptor populations leads to a reduction in hippocampal cell death in mechanically injured organotypic slice cultures. Finally, the most promising therapeutic candidate from our in vitro screen (MRS 2179, a P2Y1 receptor antagonist) also improves histological and cognitive outcomes in a preclinical model of traumatic brain injury. This work shows the potential of studying astrocyte signalling after trauma to yield new and effective therapeutic targets for treating traumatic brain injury.


Assuntos
Difosfato de Adenosina/análogos & derivados , Astrócitos/efeitos dos fármacos , Lesões Encefálicas/tratamento farmacológico , Antagonistas do Receptor Purinérgico P2Y/farmacologia , Recuperação de Função Fisiológica/efeitos dos fármacos , Transdução de Sinais/efeitos dos fármacos , Difosfato de Adenosina/farmacologia , Difosfato de Adenosina/uso terapêutico , Animais , Astrócitos/metabolismo , Lesões Encefálicas/metabolismo , Cálcio/metabolismo , Células Cultivadas , Córtex Cerebral/efeitos dos fármacos , Córtex Cerebral/lesões , Córtex Cerebral/metabolismo , Humanos , Aprendizagem em Labirinto/efeitos dos fármacos , Camundongos , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Antagonistas do Receptor Purinérgico P2Y/uso terapêutico , Ratos , Ratos Sprague-Dawley
11.
Mol Ther ; 21(12): 2258-67, 2013 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-23831593

RESUMO

Neural stem cell (NSC) therapy represents a potentially powerful approach for gene transfer in the diseased central nervous system. However, transplanted primary, embryonic stem cell- and induced pluripotent stem cell-derived NSCs generate largely undifferentiated progeny. Understanding how physiologically immature cells influence host activity is critical to evaluating the therapeutic utility of NSCs. Earlier inquiries were limited to single-cell recordings and did not address the emergent properties of neuronal ensembles. To interrogate cortical networks post-transplant, we used voltage sensitive dye imaging in mouse neocortical brain slices, which permits high temporal resolution analysis of neural activity. Although moderate NSC engraftment largely preserved host physiology, subtle defects in the activation properties of synaptic inputs were induced. High-density engraftment severely dampened cortical excitability, markedly reducing the amplitude, spatial extent, and velocity of propagating synaptic potentials in layers 2-6. These global effects may be mediated by specific disruptions in excitatory network structure in deep layers. We propose that depletion of endogenous cells in engrafted neocortex contributes to circuit alterations. Our data provide the first evidence that nonintegrating cells cause differential host impairment as a function of engrafted load. Moreover, they emphasize the necessity for efficient differentiation methods and proper controls for engraftment effects that interfere with the benefits of NSC therapy.


Assuntos
Sobrevivência de Enxerto , Neocórtex/fisiologia , Células-Tronco Neurais/fisiologia , Células-Tronco Neurais/transplante , Animais , Diferenciação Celular , Movimento Celular , Técnicas de Transferência de Genes , Camundongos , Camundongos Endogâmicos C57BL , Camundongos SCID , Neocórtex/crescimento & desenvolvimento , Neurônios/fisiologia , Imagens com Corantes Sensíveis à Voltagem
12.
J Biomech Eng ; 136(2): 021008, 2014 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-24384610

RESUMO

Traumatic brain injury (TBI) is a significant public health problem, on pace to become the third leading cause of death worldwide by 2020. Moreover, emerging evidence linking repeated mild traumatic brain injury to long-term neurodegenerative disorders points out that TBI can be both an acute disorder and a chronic disease. We are at an important transition point in our understanding of TBI, as past work has generated significant advances in better protecting us against some forms of moderate and severe TBI. However, we still lack a clear understanding of how to study milder forms of injury, such as concussion, or new forms of TBI that can occur from primary blast loading. In this review, we highlight the major advances made in understanding the biomechanical basis of TBI. We point out opportunities to generate significant new advances in our understanding of TBI biomechanics, especially as it appears across the molecular, cellular, and whole organ scale.


Assuntos
Traumatismos por Explosões/fisiopatologia , Lesões Encefálicas/fisiopatologia , Encéfalo/fisiopatologia , Efeitos Psicossociais da Doença , Modelos Biológicos , Humanos , Pressão
13.
J Biomech Eng ; 136(9): 091004, 2014 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-24950710

RESUMO

Although blast-induced traumatic brain injury (bTBI) is well recognized for its significance in the military population, the unique mechanisms of primary bTBI remain undefined. Animate models of primary bTBI are critical for determining these potentially unique mechanisms, but the biomechanical characteristics of many bTBI models are poorly understood. In this study, we examine some common shock tube configurations used to study blast-induced brain injury in the laboratory and define the optimal configuration to minimize the effect of torso overpressure and blast-induced head accelerations. Pressure transducers indicated that a customized animal holder successfully reduced peak torso overpressures to safe levels across all tested configurations. However, high speed video imaging acquired during the blast showed significant head accelerations occurred when animals were oriented perpendicular to the shock tube axis. These findings of complex head motions during blast are similar to previous reports [Goldstein et al., 2012, "Chronic Traumatic Encephalopathy in Blast-Exposed Military Veterans and a Blast Neurotrauma Mouse Model," Sci. Transl. Med., 4(134), 134ra160; Sundaramurthy et al., 2012, "Blast-Induced Biomechanical Loading of the Rat: An Experimental and Anatomically Accurate Computational Blast Injury Model," J. Neurotrauma, 29(13), pp. 2352-2364; Svetlov et al., 2010, "Morphologic and Biochemical Characterization of Brain Injury in a Model of Controlled Blast Overpressure Exposure," J. Trauma, 69(4), pp. 795-804]. Under the same blast input conditions, minimizing head acceleration led to a corresponding elimination of righting time deficits. However, we could still achieve righting time deficits under minimal acceleration conditions by significantly increasing the peak blast overpressure. Together, these data show the importance of characterizing the effect of blast overpressure on head kinematics, with the goal of producing models focused on understanding the effects of blast overpressure on the brain without the complicating factor of superimposed head accelerations.


Assuntos
Aceleração/efeitos adversos , Lesões Encefálicas/etiologia , Lesões Encefálicas/fisiopatologia , Explosões , Neurologia , Animais , Modelos Animais de Doenças , Cabeça/fisiologia , Masculino , Camundongos , Movimento
14.
J Trauma Acute Care Surg ; 96(1): 26-34, 2024 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-37853567

RESUMO

BACKGROUND: Early but not late tranexamic acid (TXA) after TBI preserves blood-brain-barrier integrity, but it is unclear if and how dose timing affects cognitive recovery beyond hours postinjury. We hypothesized that early (1 hour post-TBI) but not late (24 hours post-TBI) TXA administration improves cognitive recovery for 14 days. METHODS: CD1 male mice (n = 25) were randomized to severe TBI (injury [I], by controlled cortical impact) or sham craniotomy (S) followed by intravenous saline at 1 hour (placebo [P1]) or 30 mg/kg TXA at 1 hour (TXA1) or 24 hours (TXA24). Daily body weights, Garcia Neurological Test scores, brain/lung water content, and Morris water maze exercises quantifying swimming traffic in the platform quadrant (zone [Z] 1) and platform area (Z5) were recorded for up to 14 days. RESULTS: Among injured groups, I-TXA1 demonstrated fastest weight gain for 14 days and only I-TXA1 showed rapid (day 1) normalization of Garcia Neurological Test ( p = 0.01 vs. I-P1, I-TXA24). In cumulative spatial trials, compared with I-TXA1, I-TXA24 hindered learning (distance to Z5 and % time in Z1, p < 0.05). Compared with I-TXA1, I-TXA24 showed poorer memory with less Z5 time (0.51 vs. 0.16 seconds, p < 0.01) and Z5 crossing frequency. Unexpectedly, TXA in uninjured animals (S-TXA1) displayed faster weight gain but inferior learning and memory. CONCLUSION: Early TXA appears beneficial for cognitive and behavioral outcomes following TBI, although administration 24 hours postinjury consistently impairs cognitive recovery. Tranexamic acid in sham animals may lead to adverse effects on cognition.


Assuntos
Lesões Encefálicas Traumáticas , Ácido Tranexâmico , Animais , Masculino , Camundongos , Encéfalo , Lesões Encefálicas Traumáticas/complicações , Lesões Encefálicas Traumáticas/tratamento farmacológico , Aprendizagem em Labirinto , Ácido Tranexâmico/farmacologia , Aumento de Peso
15.
Artigo em Inglês | MEDLINE | ID: mdl-38767935

RESUMO

BACKGROUND: Traumatic brain injury (TBI) induces cognitive deficits driven by neuroinflammation and cerebral edema. The commonly used atypical antipsychotic, quetiapine (QTP), has been recently shown to improve post-TBI outcomes. We hypothesized that QTP would thereby improve animal learning and memory 2 weeks after severe TBI. METHODS: CD1 male mice (n = 35) underwent severe TBI (controlled cortical impact, injury, I) or sham craniotomy (S), followed by BID saline (P, placebo) or QTP (10 or 20 mg/kg, IP) for 2 weeks. Animals underwent Morris Water Maze (MWM) exercises to gauge spatial learning and memory. The distance and time required for swimming animals to reach the platform area (Zone 5, Z5) located in quadrant 1 (Zone 1, Z1) was calculated from digital video recordings analyzed using Ethovision software. Animal bodyweights were recorded daily and on day 14, injured cerebral hemispheres were procured for edema determination (wet-to-dry ratio). Intergroup differences were evaluated with ANOVA/Bonferroni correction (p < 0.05). RESULTS: On day 14, animal weight loss recovery was lowest in I + P compared to I + QTP20 and I + QTP10 (p ≤ 0.01 for either). Cerebral edema was greatest in I + P, and only significantly decreased in I + QTP20 (p < 0.05). Both QTP doses similarly improved spatial learning by significantly reducing latency time and travel distance to target zones (p < 0.05). In probe memory trials, only I + QTP20 and not I + QTP10 significantly favored animal reaching or crossing into target zones (p < 0.05). CONCLUSION: Post-TBI QTP reduces brain edema and improves spatial learning and memory with a potential dose dependence impact benefiting memory up to 14 days. These data suggest an unanticipated QTP benefit following brain injury that should be specifically explored.

16.
Artigo em Inglês | MEDLINE | ID: mdl-38415197

RESUMO

Over the past two decades Biomedical Engineering has emerged as a major discipline that bridges societal needs of human health care with the development of novel technologies. Every medical institution is now equipped at varying degrees of sophistication with the ability to monitor human health in both non-invasive and invasive modes. The multiple scales at which human physiology can be interrogated provide a profound perspective on health and disease. We are at the nexus of creating "avatars" (herein defined as an extension of "digital twins") of human patho/physiology to serve as paradigms for interrogation and potential intervention. Motivated by the emergence of these new capabilities, the IEEE Engineering in Medicine and Biology Society, the Departments of Biomedical Engineering at Johns Hopkins University and Bioengineering at University of California at San Diego sponsored an interdisciplinary workshop to define the grand challenges that face biomedical engineering and the mechanisms to address these challenges. The Workshop identified five grand challenges with cross-cutting themes and provided a roadmap for new technologies, identified new training needs, and defined the types of interdisciplinary teams needed for addressing these challenges. The themes presented in this paper include: 1) accumedicine through creation of avatars of cells, tissues, organs and whole human; 2) development of smart and responsive devices for human function augmentation; 3) exocortical technologies to understand brain function and treat neuropathologies; 4) the development of approaches to harness the human immune system for health and wellness; and 5) new strategies to engineer genomes and cells.

17.
J Biol Chem ; 287(6): 4348-59, 2012 Feb 03.
Artigo em Inglês | MEDLINE | ID: mdl-22179603

RESUMO

N-methyl-D-aspartate receptors (NMDARs), critical mediators of both physiologic and pathologic neurological signaling, have previously been shown to be sensitive to mechanical stretch through the loss of its native Mg(2+) block. However, the regulation of this mechanosensitivity has yet to be further explored. Furthermore, as it has become apparent that NMDAR-mediated signaling is dependent on specific NMDAR subtypes, as governed by the identity of the NR2 subunit, a crucial unanswered question is the role of subunit composition in observed NMDAR mechanosensitivity. Here, we used a recombinant system to assess the mechanosensitivity of specific subtypes and demonstrate that the mechanosensitive property is uniquely governed by the NR2B subunit. NR1/NR2B NMDARs displayed significant stretch sensitivity, whereas NR1/NR2A NMDARs did not respond to stretch. Furthermore, NR2B mechanosensitivity was regulated by PKC activity, because PKC inhibition reduced stretch responses in transfected HEK 293 cells and primary cortical neurons. Finally, using NR2B point mutations, we identified a PKC phosphorylation site, Ser-1323 on NR2B, as a unique critical regulator of stretch sensitivity. These data suggest that the selective mechanosensitivity of NR2B can significantly impact neuronal response to traumatic brain injury and illustrate that the mechanical tone of the neuron can be dynamically regulated by PKC activity.


Assuntos
Lesões Encefálicas/metabolismo , Mecanotransdução Celular , Neurônios/metabolismo , Proteína Quinase C/metabolismo , Receptores de N-Metil-D-Aspartato/metabolismo , Animais , Lesões Encefálicas/genética , Lesões Encefálicas/patologia , Células HEK293 , Humanos , Neurônios/patologia , Mutação Puntual , Proteína Quinase C/genética , Ratos , Receptores de N-Metil-D-Aspartato/genética , Transfecção
18.
J Neurochem ; 125(6): 909-20, 2013 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-23305291

RESUMO

The calpain family of calcium-dependent proteases has been implicated in a variety of diseases and neurodegenerative pathologies. Prolonged activation of calpains results in proteolysis of numerous cellular substrates including cytoskeletal components and membrane receptors, contributing to cell demise despite coincident expression of calpastatin, the specific inhibitor of calpains. Pharmacological and gene-knockout strategies have targeted calpains to determine their contribution to neurodegenerative pathology; however, limitations associated with treatment paradigms, drug specificity, and genetic disruptions have produced inconsistent results and complicated interpretation. Specific, targeted calpain inhibition achieved by enhancing endogenous calpastatin levels offers unique advantages in studying pathological calpain activation. We have characterized a novel calpastatin-overexpressing transgenic mouse model, demonstrating a substantial increase in calpastatin expression within nervous system and peripheral tissues and associated reduction in protease activity. Experimental activation of calpains via traumatic brain injury resulted in cleavage of α-spectrin, collapsin response mediator protein-2, and voltage-gated sodium channel, critical proteins for the maintenance of neuronal structure and function. Calpastatin overexpression significantly attenuated calpain-mediated proteolysis of these selected substrates acutely following severe controlled cortical impact injury, but with no effect on acute hippocampal neurodegeneration. Augmenting calpastatin levels may be an effective method for calpain inhibition in traumatic brain injury and neurodegenerative disorders.


Assuntos
Lesões Encefálicas/metabolismo , Proteínas de Ligação ao Cálcio/metabolismo , Animais , Lesões Encefálicas/patologia , Proteínas de Ligação ao Cálcio/genética , Feminino , Efeito Fundador , Hipocampo/patologia , Humanos , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Masculino , Camundongos , Camundongos Transgênicos , Canal de Sódio Disparado por Voltagem NAV1.2/metabolismo , Degeneração Neural/patologia , Proteínas do Tecido Nervoso/metabolismo , Príons/genética , Regiões Promotoras Genéticas , Proteólise , Espectrina/metabolismo
19.
Commun Biol ; 6(1): 1278, 2023 12 18.
Artigo em Inglês | MEDLINE | ID: mdl-38110605

RESUMO

Plasticity and homeostatic mechanisms allow neural networks to maintain proper function while responding to physiological challenges. Despite previous work investigating morphological and synaptic effects of brain-derived neurotrophic factor (BDNF), the most prevalent growth factor in the central nervous system, how exposure to BDNF manifests at the network level remains unknown. Here we report that BDNF treatment affects rodent hippocampal network dynamics during development and recovery from glutamate-induced excitotoxicity in culture. Importantly, these effects are not obvious when traditional activity metrics are used, so we delve more deeply into network organization, functional analyses, and in silico simulations. We demonstrate that BDNF partially restores homeostasis by promoting recovery of weak and medium connections after injury. Imaging and computational analyses suggest these effects are caused by changes to inhibitory neurons and connections. From our in silico simulations, we find that BDNF remodels the network by indirectly strengthening weak excitatory synapses after injury. Ultimately, our findings may explain the difficulties encountered in preclinical and clinical trials with BDNF and also offer information for future trials to consider.


Assuntos
Fator Neurotrófico Derivado do Encéfalo , Sinapses , Fator Neurotrófico Derivado do Encéfalo/farmacologia , Fator Neurotrófico Derivado do Encéfalo/metabolismo , Células Cultivadas , Sinapses/metabolismo , Neurônios/fisiologia , Ácido Glutâmico/metabolismo
20.
J Neurochem ; 121(5): 793-805, 2012 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-22428606

RESUMO

Although enhanced calpain activity is well documented after traumatic brain injury (TBI), the pathways targeting specific substrate proteolysis are less defined. Our past work demonstrated that calpain cleaves voltage gated sodium channel (NaCh) α-subunits in an in vitro TBI model. In this study, we investigated the pathways leading to NaCh cleavage utilizing our previously characterized in vitro TBI model, and determined the location of calpain activation within neuronal regions following stretch injury to micropatterned cultures. Calpain specific breakdown products of α-spectrin appeared within axonal, dendritic, and somatic regions 6 h after injury, concurrent with the appearance of NaCh α-subunit proteolysis in both whole cell or enriched axonal preparations. Direct pharmacological activation of either NMDA receptors (NMDArs) or NaChs resulted in NaCh proteolysis. Likewise, a chronic (6 h) dual inhibition of NMDArs/NaChs but not L-type voltage gated calcium channels significantly reduced NaCh proteolysis 6 h after mechanical injury. Interestingly, an early, transient (30 min) inhibition of NMDArs alone significantly reduced NaCh proteolysis. Although a chronic inhibition of calpain significantly reduced proteolysis, a transient inhibition of calpain immediately after injury failed to significantly attenuate NaCh proteolysis. These data suggest that both NMDArs and NaChs are key contributors to calpain activation after mechanical injury, and that a larger temporal window of sustained calpain activation needs consideration in developing effective treatments for TBI.


Assuntos
Lesões Encefálicas/metabolismo , Calpaína/metabolismo , Receptores de N-Metil-D-Aspartato/metabolismo , Canais de Sódio/metabolismo , Animais , Western Blotting , Ativação Enzimática/fisiologia , Imuno-Histoquímica , Neurônios , Proteólise , Ratos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA