Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 22
Filtrar
Más filtros











Base de datos
Intervalo de año de publicación
1.
Transl Res ; 268: 40-50, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38246342

RESUMEN

Traumatic brain injury (TBI) has a significant impact on cognitive function, affecting millions of people worldwide. Myelin loss is a prominent pathological feature of TBI, while well-functioning myelin is crucial for memory and cognition. Utilizing drug repurposing to identify effective drug candidates for TBI treatment has gained attention. Notably, recent research has highlighted the potential of clemastine, an FDA-approved allergy medication, as a promising pro-myelinating drug. Therefore, in this study, we aim to investigate whether clemastine can enhance myelination and alleviate cognitive impairment following mild TBI using a clinically relevant rat model of TBI. Mild diffuse TBI was induced using the Closed-Head Impact Model of Engineered Rotational Acceleration (CHIMERA). Animals were treated with either clemastine or an equivalent volume of the vehicle from day 1 to day 14 post-injury. Following treatment, memory-related behavioral tests were conducted, and myelin pathology in the cortex and hippocampus was assessed through immunofluorescence staining and ProteinSimple® capillary-based immunoassay. Our results showed that TBI leads to significant myelin loss, axonal damage, glial activation, and a decrease in mature oligodendrocytes in both the cortex and hippocampus. The TBI animals also exhibited notable deficits in memory-related tests. In contrast, animals treated with clemastine showed an increase in mature oligodendrocytes, enhanced myelination, and improved performance in the behavioral tests. These preliminary findings support the therapeutic value of clemastine in alleviating TBI-induced cognitive impairment, with substantial clinical translational potential. Our findings also underscore the potential of remyelinating therapies for TBI.


Asunto(s)
Axones , Clemastina , Disfunción Cognitiva , Modelos Animales de Enfermedad , Vaina de Mielina , Ratas Sprague-Dawley , Animales , Clemastina/farmacología , Clemastina/uso terapéutico , Vaina de Mielina/efectos de los fármacos , Vaina de Mielina/patología , Vaina de Mielina/metabolismo , Disfunción Cognitiva/tratamiento farmacológico , Disfunción Cognitiva/patología , Axones/efectos de los fármacos , Axones/patología , Masculino , Ratas , Lesiones Traumáticas del Encéfalo/tratamiento farmacológico , Lesiones Traumáticas del Encéfalo/patología , Lesiones Traumáticas del Encéfalo/complicaciones , Traumatismos Difusos del Encéfalo/efectos de los fármacos , Traumatismos Difusos del Encéfalo/patología , Hipocampo/efectos de los fármacos , Hipocampo/patología
2.
J Neurosci ; 42(48): 9082-9096, 2022 11 30.
Artículo en Inglés | MEDLINE | ID: mdl-36257689

RESUMEN

Traumatic brain injury (TBI) is associated with chronic psychiatric complications and increased risk for development of neurodegenerative pathology. Aged individuals account for most TBI-related hospitalizations and deaths. Nonetheless, neurobiological mechanisms that underlie worsened functional outcomes after TBI in the elderly remain unclear. Therefore, this study aimed to identify pathways that govern differential responses to TBI with age. Here, adult (2 months of age) and aged (16-18 months of age) male C57BL/6 mice were subjected to diffuse brain injury (midline fluid percussion), and cognition, gliosis, and neuroinflammation were determined 7 or 30 d postinjury (dpi). Cognitive impairment was evident 7 dpi, independent of age. There was enhanced morphologic restructuring of microglia and astrocytes 7 dpi in the cortex and hippocampus of aged mice compared with adults. Transcriptional analysis revealed robust age-dependent amplification of cytokine/chemokine, complement, innate immune, and interferon-associated inflammatory gene expression in the cortex 7 dpi. Ingenuity pathway analysis of the transcriptional data showed that type I interferon (IFN) signaling was significantly enhanced in the aged brain after TBI compared with adults. Age prolonged inflammatory signaling and microgliosis 30 dpi with an increased presence of rod microglia. Based on these results, a STING (stimulator of interferon genes) agonist, DMXAA, was used to determine whether augmenting IFN signaling worsened cortical inflammation and gliosis after TBI. DMXAA-treated Adult-TBI mice showed comparable expression of myriad genes that were overexpressed in the cortex of Aged-TBI mice, including Irf7, Clec7a, Cxcl10, and Ccl5 Overall, diffuse TBI promoted amplified IFN signaling in aged mice, resulting in extended inflammation and gliosis.SIGNIFICANCE STATEMENT Elderly individuals are at higher risk of complications following traumatic brain injury (TBI). Individuals >70 years old have the highest rates of TBI-related hospitalization, neurodegenerative pathology, and death. Although inflammation has been linked with poor outcomes in aging, the specific biological pathways driving worsened outcomes after TBI in aging remain undefined. In this study, we identify amplified interferon-associated inflammation and gliosis in aged mice following TBI that was associated with persistent inflammatory gene expression and microglial morphologic diversity 30 dpi. STING (stimulator of interferon genes) agonist DMXAA was used to demonstrate a causal link between augmented interferon signaling and worsened neuroinflammation after TBI. Therefore, interferon signaling may represent a therapeutic target to reduce inflammation-associated complications following TBI.


Asunto(s)
Traumatismos Difusos del Encéfalo , Lesiones Traumáticas del Encéfalo , Animales , Ratones , Masculino , Gliosis/etiología , Gliosis/metabolismo , Ratones Endogámicos C57BL , Interferones , Traumatismos Difusos del Encéfalo/metabolismo , Traumatismos Difusos del Encéfalo/patología , Microglía/metabolismo , Lesiones Traumáticas del Encéfalo/patología , Encéfalo/metabolismo , Inflamación/metabolismo
3.
ASN Neuro ; 14: 17590914221099112, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35503242

RESUMEN

Traumatic brain injury (TBI) has consequences that last for years following injury. While TBI can precipitate a variety of diffuse pathologies, the mechanisms involved in injury-induced neuronal membrane disruption remain elusive. The lysosomal cysteine protease, Cathepsin B (Cath B), and specifically its redistribution into the cytosol has been implicated in cell death. Little is known about Cath B or neuronal membrane disruption chronically following diffuse TBI. Therefore, the current study evaluated Cath B and diffuse neuronal membrane disruption over a more chronic post-injury window (6 h-4 w). We evaluated Cath B in adult male Sprague-Dawley rats following central fluid percussion injury (CFPI). Expression of Cath B, as well as Cath B-associated pro (Bak and AIF) and anti-apoptotic (Bcl-xl) proteins, were assessed using western blot analysis. Cath B activity was also assessed. Localization of Cath B was evaluated in the membrane disrupted and non-disrupted population following CFPI using immunohistochemistry paired with quantitative image analysis and ultrastructural verification. There was no difference in expression or activity of Cath B or any of the associated proteins between sham and CFPI at any time post-injury. Immunohistological studies, however, showed a sub-cellular re-localization of Cath B at 2 w and 4 w post-injury in the membrane disrupted neuronal population as compared to the time-point matched non-disrupted neurons. Both membrane disruption and Cath B relocalization appear linked to neuronal atrophy. These observations are indicative of a late secondary pathology that represents an opportunity for therapeutic treatment of these neurons following diffuse TBI. Summary Statement Lysosomal cathepsin B relocalizes to the cytosol in neurons with disrupted plasmalemmal membranes weeks following diffuse brain injury. Both the membrane disrupted and cathepsin B relocalized neuronal subpopulations displayed smaller soma and nucleus size compared to non-pathological neurons, indicating atrophy.


Asunto(s)
Traumatismos Difusos del Encéfalo , Lesiones Traumáticas del Encéfalo , Animales , Atrofia/metabolismo , Atrofia/patología , Traumatismos Difusos del Encéfalo/metabolismo , Traumatismos Difusos del Encéfalo/patología , Lesiones Traumáticas del Encéfalo/patología , Catepsina B/análisis , Catepsina B/metabolismo , Masculino , Neuronas/metabolismo , Ratas , Ratas Sprague-Dawley
4.
J Neurosci ; 42(20): 4215-4228, 2022 05 18.
Artículo en Inglés | MEDLINE | ID: mdl-35440489

RESUMEN

Traumatic brain injury (TBI) is associated with an increased risk of cognitive, psychiatric, and neurodegenerative complications that may develop after injury. Increased microglial reactivity following TBI may underlie chronic neuroinflammation, neuropathology, and exaggerated responses to immune challenges. Therefore, the goal of this study was to force turnover of trauma-associated microglia that develop after diffuse TBI and determine whether this alleviated chronic inflammation, improved functional recovery and attenuated reduced immune reactivity to lipopolysaccharide (LPS) challenge. Male mice received a midline fluid percussion injury (mFPI) and 7 d later were subjected to a forced microglia turnover paradigm using CSF1R antagonism (PLX5622). At 30 d postinjury (dpi), cortical gene expression, dendritic complexity, myelin content, neuronal connectivity, cognition, and immune reactivity were assessed. Myriad neuropathology-related genes were increased 30 dpi in the cortex, and 90% of these gene changes were reversed by microglial turnover. Reduced neuronal connectivity was evident 30 dpi and these deficits were attenuated by microglial turnover. TBI-associated dendritic remodeling and myelin alterations, however, remained 30 dpi independent of microglial turnover. In assessments of functional recovery, increased depressive-like behavior, and cognitive impairment 30 dpi were ameliorated by microglia turnover. To investigate microglial priming and reactivity 30 dpi, mice were injected intraperitoneally with LPS. This immune challenge caused prolonged lethargy, sickness behavior, and microglial reactivity in the TBI mice. These extended complications with LPS in TBI mice were prevented by microglia turnover. Collectively, microglial turnover 7 dpi alleviated behavioral and cognitive impairments associated with microglial priming and immune reactivity 30 dpi.SIGNIFICANCE STATEMENT A striking feature of traumatic brain injury (TBI), even mild injuries, is that over 70% of individuals have long-term neuropsychiatric complications. Chronic inflammatory processes are implicated in the pathology of these complications and these issues can be exaggerated by immune challenge. Therefore, our goal was to force the turnover of microglia 7 d after TBI. This subacute 7 d postinjury (dpi) time point is a critical transitional period in the shift toward chronic inflammatory processes and microglia priming. This forced microglia turnover intervention in mice attenuated the deficits in behavior and cognition 30 dpi. Moreover, microglia priming and immune reactivity after TBI were also reduced with microglia turnover. Therefore, microglia represent therapeutic targets after TBI to reduce persistent neuroinflammation and improve recovery.


Asunto(s)
Traumatismos Difusos del Encéfalo , Lesiones Traumáticas del Encéfalo , Disfunción Cognitiva , Animales , Traumatismos Difusos del Encéfalo/metabolismo , Traumatismos Difusos del Encéfalo/patología , Lesiones Traumáticas del Encéfalo/patología , Disfunción Cognitiva/metabolismo , Inflamación/metabolismo , Lipopolisacáridos/toxicidad , Masculino , Ratones , Ratones Endogámicos C57BL , Microglía/metabolismo
5.
J Neurotrauma ; 39(5-6): 411-422, 2022 03.
Artículo en Inglés | MEDLINE | ID: mdl-35018831

RESUMEN

Cerebellar dysfunction after traumatic brain injury (TBI) is commonly suspected based on clinical symptoms, although cerebellar pathology has rarely been investigated. To address the hypothesis that the cerebellar axon-myelin unit is altered by diffuse TBI, we used the central fluid percussion injury (cFPI) model in adult mice to create widespread axonal injury by delivering the impact to the forebrain. We specifically focused on changes in myelin components (myelin basic protein [MBP], 2',3'-cyclic nucleotide 3'-phosphodiesterase [CNPase], nodal/paranodal domains [neurofascin (Nfasc), ankyrin-G], and phosphorylated neurofilaments [SMI-31, SMI-312]) in the cerebellum, remote from the impact, at two, seven, and 30 days post-injury (dpi). When compared with sham-injured controls, cerebellar MBP and CNPase protein levels were decreased at 2 dpi that remained reduced up to 30 dpi. Diffuse TBI induced different effects on neuronal (Nfasc 186, Nfasc 140) and glial (Nfasc 155) neurofascin isoforms that play a key role in the assembly of the nodes of Ranvier. Expression of Nfasc 140 in the cerebellum increased at 7 dpi, in contrast to Nfasc 155 levels, which were decreased. Although neurofascin binding partner ankyrin-G protein levels decreased acutely after cFPI, its expression levels increased at 7 dpi and remained unchanged up to 30 dpi. The TBI-induced reduction in neurofilament phosphorylation (SMI-31) observed in the cerebellum was closely associated with decreased levels of the myelin proteins MBP and CNPase. This is the first evidence of temporal and spatial structural changes in the axon-myelin unit in the cerebellum, remote from the location of the impact site, in a diffuse TBI model in mice.


Asunto(s)
Traumatismos Difusos del Encéfalo , Lesiones Traumáticas del Encéfalo , 2',3'-Nucleótido Cíclico Fosfodiesterasas/metabolismo , Animales , Ancirinas/metabolismo , Axones/patología , Traumatismos Difusos del Encéfalo/patología , Lesiones Traumáticas del Encéfalo/patología , Cerebelo/metabolismo , Ratones , Vaina de Mielina/metabolismo
6.
J Forensic Leg Med ; 82: 102226, 2021 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-34375839

RESUMEN

Traumatic brain injury (TBI) is recognised as a serious global public health problem that imposes a heavy socioeconomic burden on society. The vast majority of cases result from road traffic accidents and falls, and the injuries are mainly attributed to velocity-related mechanisms. Lethal cases are mostly found to suffer from severe diffuse brain injuries (DBI), comprising diffuse vascular injury, diffuse axonal injury (DAI), generalized cerebral edema and ischemic-hypoxic injury. Coup and contrecoup brain contusions may also occur. This study set out to describe the pathological findings of severe DBI in terms of survival times and Abbreviated Injury Scale (AIS) severity scores. The autopsy data from 2 recent years (2018 and 2019) were reviewed to recruit over 800 cases presenting with severe head injuries. Many demographic characteristics of TBI were identified (for example, causes, victim genders and victim ages). These were revealed to be like those previously reported in the literature, confirming that there are shared risk factors across the globe. The hallmarks of severe TBI-such as a unimodal survival distribution and a period for detecting DAI via conventional staining-were also evident, as per previous reports. However, it was noticed that the histopathological detection rates of DAI surged after 72 h, which might be because these injuries are mediated by secondary axotomy. This study also analysed real brain weights to identify the time period for the development of cerebral edema in humans; this period seems to have never been reported. The increment time of cerebral edema reached a peak in 12 h, after which the condition sustained for at least 72 h. This may be a golden period in clinical practice as well as a prognostic factor in forensic medicine.


Asunto(s)
Escala Resumida de Traumatismos , Edema Encefálico/patología , Traumatismos Difusos del Encéfalo/patología , Lesiones Traumáticas del Encéfalo/epidemiología , Lesiones Traumáticas del Encéfalo/patología , Adolescente , Adulto , Anciano , Anciano de 80 o más Años , Autopsia , Niño , Preescolar , Femenino , Traumatismos Cerrados de la Cabeza/epidemiología , Traumatismos Cerrados de la Cabeza/patología , Humanos , Lactante , Masculino , Persona de Mediana Edad , Estudios Retrospectivos , Análisis de Supervivencia , Tailandia/epidemiología , Adulto Joven
7.
J Neurotrauma ; 37(17): 1918-1932, 2020 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-32178582

RESUMEN

Oculomotor deficits, such as insufficiencies in accommodation, convergence, and saccades, are common following traumatic brain injury (TBI). Previous studies in patients with mild TBI attributed these deficits to insufficient activation of subcortical oculomotor nuclei, although the exact mechanism is unknown. A possible cause for neuronal dysfunction in these regions is biomechanically induced plasma membrane permeability. We used our established porcine model of head rotational TBI to investigate whether cell permeability changes occurred in subcortical oculomotor areas following single or repetitive TBI, with repetitive injuries separated by 15 min, 3 days, or 7 days. Swine were subjected to sham conditions or head rotational acceleration in the sagittal plane using a HYGE pneumatic actuator. Two hours prior to the final injury, the cell-impermeant dye Lucifer Yellow was injected into the ventricles to diffuse throughout the interstitial space to assess plasmalemmal permeability. Animals were sacrificed 15 min after the final injury for immunohistological analysis. Brain regions examined for cell membrane permeability included caudate, substantia nigra pars reticulata, superior colliculus, and cranial nerve oculomotor nuclei. We found that the distribution of permeabilized neurons varied depending on the number and spacing of injuries. Repetitive injuries separated by 15 min or 3 days resulted in the most permeability. Many permeabilized cells lost neuron-specific nuclear protein reactivity, although no neuronal loss occurred acutely after injury. Microglia contacted and appeared to begin phagocytosing permeabilized neurons in repetitively injured animals. These pathologies within oculomotor areas may mediate transient dysfunction and/or degeneration that may contribute to oculomotor deficits following diffuse TBI.


Asunto(s)
Traumatismos Difusos del Encéfalo/patología , Lesiones Traumáticas del Encéfalo/patología , Membrana Celular/patología , Neuronas/patología , Complejo Nuclear Oculomotor/patología , Animales , Encéfalo/metabolismo , Encéfalo/patología , Traumatismos Difusos del Encéfalo/metabolismo , Lesiones Traumáticas del Encéfalo/metabolismo , Membrana Celular/metabolismo , Femenino , Neuronas/metabolismo , Complejo Nuclear Oculomotor/metabolismo , Porcinos
8.
J Vis Exp ; (156)2020 02 10.
Artículo en Inglés | MEDLINE | ID: mdl-32090988

RESUMEN

Traumatic brain injury (TBI) is a leading cause of acquired epilepsy. TBI can result in a focal or diffuse brain injury. Focal injury is a result of direct mechanical forces, sometimes penetrating through the cranium, creating a direct lesion in the brain tissue. These are visible during brain imaging as areas with contusion, laceration, and hemorrhage. Focal lesions induce neuronal death and glial scar formation and are present in 20%-25% of all people who incur a TBI. However, in the majority of TBI cases, injury is caused by acceleration-deceleration forces and subsequent tissue shearing, resulting in nonfocal, diffuse damage. A subpopulation of TBI patients continues to develop post-traumatic epilepsy (PTE) after a latency period of months or years. Currently, it is impossible to predict which patients will develop PTE, and seizures in PTE patients are challenging to control, necessitating further research. Until recently, the field was limited to only two animal/rodent models with validated spontaneous post-traumatic seizures, both presenting with large focal lesions with massive tissue loss in the cortex and sometimes subcortical structures. In contrast to these approaches, it was determined that diffuse TBI induced using a modified weight drop model is sufficient to initiate development of spontaneous convulsive and non-convulsive seizures, even in the absence of focal lesions or tissue loss. Similar to human patients with acquired post-traumatic epilepsy, this model presents with a latency period after injury before seizure onset. In this protocol, the community will be provided with a new model of post-traumatic epilepsy, detailing how to induce diffuse non-lesional TBI followed by continuous long-term video-electroencephalographic animal monitoring over the course of several months. This protocol will detail animal handling, the weight drop procedure, the electrode placement for two acquisition systems, and the frequent challenges encountered during each of the steps of surgery, postoperative monitoring, and data acquisition.


Asunto(s)
Traumatismos Difusos del Encéfalo/patología , Lesiones Traumáticas del Encéfalo/complicaciones , Lesiones Traumáticas del Encéfalo/patología , Modelos Animales de Enfermedad , Epilepsia Postraumática/etiología , Epilepsia Postraumática/patología , Animales , Encéfalo/patología , Encéfalo/fisiopatología , Traumatismos Difusos del Encéfalo/fisiopatología , Lesiones Traumáticas del Encéfalo/fisiopatología , Corteza Cerebral/patología , Epilepsia Postraumática/fisiopatología , Masculino , Ratones , Ratones Endogámicos C57BL , Convulsiones/etiología , Convulsiones/patología
9.
Neuroimage Clin ; 25: 102136, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-31865019

RESUMEN

BACKGROUND: Diffuse traumatic brain injury (TBI) is known to lead to microstructural changes within both white and grey matter detected in vivo with diffusion tensor imaging (DTI). Numerous studies have shown alterations in fractional anisotropy (FA) and mean diffusivity (MD) within prominent white matter tracts, but few have linked these to changes within the grey matter with confirmation via histological assessment. This is especially important as alterations in the grey matter may be predictive of long-term functional deficits. METHODS: A total of 33 male Sprague Dawley rats underwent severe closed-head TBI. Eight animals underwent tensor-based morphometry (TBM) and DTI at baseline (pre-TBI), 24 hours (24 h), 7, 14, and 30 days post-TBI. Immunohistochemical analysis for the detection of ionised calcium-binding adaptor molecule 1 (IBA1) to assess microglia number and percentage of activated cells, ß-amyloid precursor protein (APP) as a marker of axonal injury, and myelin basic protein (MBP) to investigate myelination was performed at each time-point. RESULTS: DTI showed significant alterations in FA and RD in numerous white matter tracts including the corpus callosum, internal and external capsule, and optic tract and in the grey-matter in the cortex, thalamus, and hippocampus, with the most significant effects observed at 14 D post-TBI. TBM confirmed volumetric changes within the hippocampus and thalamus. Changes in DTI were in line with significant axonal injury noted at 24 h post-injury via immunohistochemical analysis of APP, with widespread microglial activation seen within prominent white matter tracts and the grey matter, which persisted to 30 D within the hippocampus and thalamus. Microstructural alterations in MBP+ve fibres were also noted within the hippocampus and thalamus, as well as the cortex. CONCLUSION: This study confirms the widespread effects of diffuse TBI on white matter tracts which could be detected via DTI and extends these findings to key grey matter regions, with a comprehensive investigation of the whole brain. In particular, the hippocampus and thalamus appear to be vulnerable to ongoing pathology post-TBI, with DTI able to detect these alterations supporting the clinical utility in evaluating these regions post-TBI.


Asunto(s)
Traumatismos Difusos del Encéfalo/patología , Lesiones Traumáticas del Encéfalo/patología , Encéfalo/patología , Sustancia Gris/patología , Sustancia Blanca/patología , Animales , Imagen de Difusión Tensora , Masculino , Ratas , Ratas Sprague-Dawley
10.
Curr Opin Neurol ; 32(6): 786-795, 2019 12.
Artículo en Inglés | MEDLINE | ID: mdl-31633494

RESUMEN

PURPOSE OF REVIEW: Diffuse or traumatic axonal injury is one of the principal pathologies encountered in traumatic brain injury (TBI) and the resulting axonal loss, disconnection, and brain atrophy contribute significantly to clinical morbidity and disability. The seminal discovery of the slow Wallerian degeneration mice (Wld) in which transected axons do not degenerate but survive and function independently for weeks has transformed concepts on axonal biology and raised hopes that axonopathies may be amenable to specific therapeutic interventions. Here we review mechanisms of axonal degeneration and also describe how these mechanisms may inform biological therapies of traumatic axonopathy in the context of TBI. RECENT FINDINGS: In the last decade, SARM1 [sterile a and Toll/interleukin-1 receptor (TIR) motif containing 1] and the DLK (dual leucine zipper bearing kinase) and LZK (leucine zipper kinase) MAPK (mitogen-activated protein kinases) cascade have been established as the key drivers of Wallerian degeneration, a complex program of axonal self-destruction which is activated by a wide range of injurious insults, including insults that may otherwise leave axons structurally robust and potentially salvageable. Detailed studies on animal models and postmortem human brains indicate that this type of partial disruption is the main initial pathology in traumatic axonopathy. At the same time, the molecular dissection of Wallerian degeneration has revealed that the decision that commits axons to degeneration is temporally separated from the time of injury, a window that allows potentially effective pharmacological interventions. SUMMARY: Molecular signals initiating and triggering Wallerian degeneration appear to be playing an important role in traumatic axonopathy and recent advances in understanding their nature and significance is opening up new therapeutic opportunities for TBI.


Asunto(s)
Axones , Traumatismos Difusos del Encéfalo , Lesiones Traumáticas del Encéfalo , Degeneración Walleriana , Animales , Axones/metabolismo , Axones/patología , Traumatismos Difusos del Encéfalo/metabolismo , Traumatismos Difusos del Encéfalo/patología , Lesiones Traumáticas del Encéfalo/tratamiento farmacológico , Lesiones Traumáticas del Encéfalo/metabolismo , Lesiones Traumáticas del Encéfalo/patología , Humanos , Degeneración Walleriana/tratamiento farmacológico , Degeneración Walleriana/metabolismo , Degeneración Walleriana/patología
11.
J Neurotrauma ; 36(22): 3172-3182, 2019 11 15.
Artículo en Inglés | MEDLINE | ID: mdl-31280698

RESUMEN

With an emphasis on traumatic axonal injury (TAI), frequency and evolution of traumatic intracranial lesions on 3T clinical magnetic resonance imaging (MRI) were assessed in a combined hospital and community-based study of patients with mild traumatic brain injury (mTBI). The findings were related to post-concussion symptoms (PCS) at 3 and 12 months. Prospectively, 194 patients (16-60 years of age) were recruited from the emergency departments at a level 1 trauma center and a municipal outpatient clinic into the Trondheim mTBI follow-up study. MRI was acquired within 72 h (n = 194) and at 3 (n = 165) and 12 months (n = 152) in patients and community controls (n = 78). The protocol included T2, diffusion weighted imaging, fluid attenuated inversion recovery (FLAIR), and susceptibility weighted imaging (SWI). PCS was assessed with British Columbia Post Concussion Symptom Inventory in patients and controls. Traumatic lesions were present in 12% on very early MRI, and in 5% when computed tomography (CT) was negative. TAI was found in 6% and persisted for 12 months on SWI, whereas TAI lesions on FLAIR disappeared or became less conspicuous on follow-up. PCS occurred in 33% of patients with lesions on MRI and in 19% in patients without lesions at 3 months (p = 0.12) and in 21% with lesions and 14% without lesions at 12 months (p = 0.49). Very early MRI depicted cases of TAI in patients with mTBI with microbleeds persisting for 12 months. Patients with traumatic lesions may have a more protracted recovery, but the study was underpowered to detect significant differences for PCS because of the low frequency of trauma-related MRI lesions.


Asunto(s)
Conmoción Encefálica/diagnóstico por imagen , Traumatismos Difusos del Encéfalo/diagnóstico por imagen , Síndrome Posconmocional/diagnóstico por imagen , Adulto , Conmoción Encefálica/patología , Traumatismos Difusos del Encéfalo/patología , Femenino , Hospitales , Humanos , Estudios Longitudinales , Imagen por Resonancia Magnética , Masculino , Síndrome Posconmocional/patología , Atención Primaria de Salud , Estudios Prospectivos , Adulto Joven
12.
Neuroscience ; 388: 152-170, 2018 09 15.
Artículo en Inglés | MEDLINE | ID: mdl-30036662

RESUMEN

Changes in inhibition following traumatic brain injury (TBI) appear to be one of the major factors that contribute to excitation:inhibition imbalance. Neuron pathology, interneurons in particular evolves from minutes to weeks post injury and follows a complex time course. Previously, we showed that in the long-term in diffuse TBI (dTBI), there was select reduction of specific dendrite-targeting neurons in sensory cortex and hippocampus while in motor cortex there was up-regulation of specific dendrite-targeting neurons. We now investigated the time course of dTBI effects on interneurons in neocortex and hippocampus. Brains were labeled with antibodies against calbindin (CB), parvalbumin (PV), calretinin (CR) neuropeptide Y (NPY), and somatostatin (SOM) at 24 h and 2 weeks post dTBI. We found time-dependent, brain area-specific changes in inhibition at 24 h and 2 weeks. At 24 h post-injury, reduction of dendrite-targeting inhibitory neurons occurred in sensory cortex and hippocampus. At 2 weeks, we found compensatory changes in the somatosensory cortex and CA2/3 of hippocampus affected at 24 h, with affected interneuronal populations returning to sham levels. However, DG of hippocampus now showed reduction of dendrite-targeting inhibitory neurons. Finally, with respect to motor cortex, there was an upregulation of dendrite-targeting interneurons in the supragranular layers at 24 h returning to normal levels by 2 weeks. Overall, our findings reconfirm that dendritic inhibition is particularly susceptible to brain trauma, but also show that there are complex brain-area-specific changes in inhibitory neuronal numbers and in compensatory changes, rather than a simple monotonic progression of changes post-dTBI.


Asunto(s)
Traumatismos Difusos del Encéfalo/fisiopatología , Lesiones Traumáticas del Encéfalo/fisiopatología , Corteza Cerebral/fisiopatología , Hipocampo/fisiopatología , Neuronas/fisiología , Animales , Traumatismos Difusos del Encéfalo/patología , Lesiones Traumáticas del Encéfalo/patología , Corteza Cerebral/patología , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Hipocampo/patología , Masculino , Inhibición Neural/fisiología , Neuronas/patología , Distribución Aleatoria , Ratas Sprague-Dawley , Factores de Tiempo
13.
J Neurotrauma ; 35(23): 2837-2849, 2018 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-29690837

RESUMEN

Traumatic brain injury (TBI) commonly results in injury to the components of the white matter tracts, causing post-injury cognitive deficits. The myelin-producing oligodendrocytes (OLs) are vulnerable to TBI, although may potentially be replaced by proliferating oligodendrocyte progenitor cells (OPCs). The cytokine interleukin-1ß (IL-1ß) is a key mediator of the complex inflammatory response, and when neutralized in experimental TBI, behavioral outcome was improved. To evaluate the role of IL-1ß on oligodendrocyte cell death and OPC proliferation, 116 adult male mice subjected to sham injury or the central fluid percussion injury (cFPI) model of traumatic axonal injury, were analyzed at two, seven, and 14 days post-injury. At 30 min post-injury, mice were randomly administered an IL-1ß neutralizing or a control antibody. OPC proliferation (5-ethynyl 2'- deoxyuridine (EdU)/Olig2 co-labeling) and mature oligodendrocyte cell loss was evaluated in injured white matter tracts. Microglia/macrophages immunohistochemistry and ramification using Sholl analysis were also evaluated. Neutralizing IL-1ß resulted in attenuated cell death, indicated by cleaved caspase-3 expression, and attenuated loss of mature OLs from two to seven days post-injury in brain-injured animals. IL-1ß neutralization also attenuated the early, two day post-injury increase of microglia/macrophage immunoreactivity and altered their ramification. The proliferation of OPCs in brain-injured animals was not altered, however. Our data suggest that IL-1ß is involved in the TBI-induced loss of OLs and early microglia/macrophage activation, although not the OPC proliferation. Attenuated oligodendrocyte cell loss may contribute to the improved behavioral outcome observed by IL-1ß neutralization in this mouse model of diffuse TBI.


Asunto(s)
Traumatismos Difusos del Encéfalo/patología , Lesiones Traumáticas del Encéfalo/patología , Interleucina-1beta/antagonistas & inhibidores , Oligodendroglía/patología , Animales , Traumatismos Difusos del Encéfalo/inmunología , Traumatismos Difusos del Encéfalo/metabolismo , Lesiones Traumáticas del Encéfalo/inmunología , Lesiones Traumáticas del Encéfalo/metabolismo , Interleucina-1beta/inmunología , Masculino , Ratones , Ratones Endogámicos C57BL , Distribución Aleatoria
14.
Sci Rep ; 8(1): 2387, 2018 02 05.
Artículo en Inglés | MEDLINE | ID: mdl-29402984

RESUMEN

Traumatic brain injury (TBI) is one of the most important death and disability cause, involving substantial costs, also in economic terms, when considering the young age of the involved subject. Aim of this paper is to report a series of patients treated at our institutions, to verify neurological results at six months or survival; in fatal cases we searched for ßAPP, GFAP, IL-1ß, NFL, Spectrin II, TUNEL and miR-21, miR-16, and miR-92 expressions in brain samples, to verify DAI diagnosis and grade as strong predictor of survival and inflammatory response. Concentrations of 8OHdG as measurement of oxidative stress was performed. Immunoreaction of ß-APP, IL-1ß, GFAP, NFL, Spectrin II and 8OHdG were significantly increased in the TBI group with respect to control group subjects. Cell apoptosis, measured by TUNEL assay, were significantly higher in the study group than control cases. Results indicated that miR-21, miR-92 and miR-16 have a high predictive power in discriminating trauma brain cases from controls and could represent promising biomarkers as strong predictor of survival, and for the diagnosis of postmortem traumatic brain injury.


Asunto(s)
Biomarcadores/análisis , Traumatismos Difusos del Encéfalo/patología , Encéfalo/patología , Perfilación de la Expresión Génica , Técnicas de Diagnóstico Molecular/métodos , Adolescente , Adulto , Anciano , Femenino , Humanos , Masculino , Persona de Mediana Edad , Pronóstico , Índice de Severidad de la Enfermedad , Análisis de Supervivencia , Adulto Joven
15.
J Neurotrauma ; 35(14): 1694-1704, 2018 07 15.
Artículo en Inglés | MEDLINE | ID: mdl-29390943

RESUMEN

Traumatic brain injury (TBI) is a prevalent disease with significant costs. Although progress has been made in understanding the complex pathobiology of focal lesions associated with TBI, questions remain regarding the diffuse responses to injury. Expression of the transient receptor potential melastatin 4 (Trpm4) channel is linked to cytotoxic edema during hemorrhagic contusion expansion. However, little is known about Trpm4 following diffuse TBI. To explore Trpm4 expression in diffuse TBI, rats were subjected to a diffuse central fluid percussion injury (CFPI) and survived for 1.5 h to 8 weeks. The total number of Trpm4+ cells, as well as individual cellular intensity/expression of Trpm4, were assessed. Hemotoxylin and eosin (H&E) labeling was performed to evaluate cell damage/death potentially associated with Trpm4 expression following diffuse TBI. Finally, ultrastructural assessments were performed to evaluate the integrity of Trpm4+ cells and the potential for swelling associated with Trpm4 expression. Trpm4 was primarily restricted to astrocytes within the hippocampus and peaked at 6 h post-injury. While the number of Trpm4+ astrocytes returned to sham levels by 8 weeks post-CFPI, cellular intensity occurred in region-specific waves following injury. Correlative H&E assessments demonstrated little evidence of hippocampal damage, suggesting that Trpm4 expression by astrocytes does not precipitate cell death following diffuse TBI. Additionally, ultrastructural assessments showed Trpm4+ astrocytes exhibited twice the soma size compared with Trpm4- astrocytes, indicating that astrocyte swelling is associated with Trpm4 expression. This study provides a foundation for future investigations into the role of Trpm4 in astrocyte swelling and edema following diffuse TBI.


Asunto(s)
Astrocitos/metabolismo , Astrocitos/patología , Edema Encefálico/patología , Lesiones Traumáticas del Encéfalo/patología , Canales Catiónicos TRPM/metabolismo , Animales , Edema Encefálico/etiología , Edema Encefálico/metabolismo , Traumatismos Difusos del Encéfalo/metabolismo , Traumatismos Difusos del Encéfalo/patología , Lesiones Traumáticas del Encéfalo/complicaciones , Lesiones Traumáticas del Encéfalo/metabolismo , Masculino , Ratas , Ratas Sprague-Dawley
16.
Ann Pathol ; 38(2): 103-109, 2018 Apr.
Artículo en Francés | MEDLINE | ID: mdl-29429858

RESUMEN

According to the French High Authority for Health, sudden unexpected death in infants (SUDI) is defined as "a sudden death that occurs in an infant, whereas nothing in its known history could have predicted it". This is an exclusion diagnosis. There are great interregional disparities despite the professional recommendations established in February 2007. For the examination of the brain, instructions are not adapted to current and research practice. The role of the pathologist, like anyone involved in SUDI, is to eliminate an abuse head trauma and to determine the cause of death. Major neuropathological lesions by definition do not exist. Lesions of hypoxia/ischemia are the most frequent but not specific. The accessibility of anti-APP immunoblotting has highlighted the role of anoxia in the development of axonal diffuse damages. Many studies are looking for a neurological substratum of the SUDI (neuropathological and/or neurobiochinic). This article aims to define a detailed sampling protocol based on foreign consensus and current data of science in order to assist pathologists and to promote a homogeneous data bank in France.


Asunto(s)
Autopsia/métodos , Encéfalo/patología , Muerte Súbita del Lactante/patología , Traumatismos Difusos del Encéfalo/patología , Lesiones Traumáticas del Encéfalo/diagnóstico , Lesiones Traumáticas del Encéfalo/patología , Causas de Muerte , Bases de Datos Factuales , Diagnóstico Diferencial , Francia , Humanos , Hipoxia-Isquemia Encefálica/diagnóstico , Hipoxia-Isquemia Encefálica/patología , Lactante , Muerte Súbita del Lactante/diagnóstico , Muerte Súbita del Lactante/epidemiología , Muerte Súbita del Lactante/etiología
17.
Sci Rep ; 7(1): 13211, 2017 10 16.
Artículo en Inglés | MEDLINE | ID: mdl-29038483

RESUMEN

Determining regions of altered brain physiology after diffuse brain injury is challenging. Microglia, brain immune cells with ramified and dynamically moving processes, constantly surveil the parenchyma for dysfunction which, when present, results in a changed morphology. Our purpose was to define the spatiotemporal changes in microglia morphology over 28 days following rat midline fluid percussion injury (mFPI) as a first step in exploiting microglia morphology to reflect altered brain physiology. Microglia morphology was quantified from histological sections using Image J skeleton and fractal analysis procedures at three time points and in three regions post-mFPI: impact site, primary somatosensory cortex barrel field (S1BF), and a remote region. Microglia ramification (process length/cell and endpoints/cell) decreased in the impact and S1BF but not the remote region (p < 0.05). Microglia complexity was decreased in the S1BF (p = 0.003) and increased in the remote region (p < 0.02). Rod-shaped microglia were present in the S1BF and had a 1.8:1.0 length:width ratio. An in-depth quantitative morphologic analysis revealed diverse and widespread changes to microglia morphology in the cortex post-mFPI. Due to their close link to neuronal function, changes in microglia morphology, summarized in this study, likely reflect altered physiology with diverse and widespread impact on neuronal and circuit function.


Asunto(s)
Traumatismos Difusos del Encéfalo/patología , Microglía/patología , Corteza Somatosensorial/patología , Animales , Traumatismos Difusos del Encéfalo/fisiopatología , Fractales , Masculino , Percusión , Ratas , Ratas Sprague-Dawley , Corteza Somatosensorial/fisiopatología
18.
J Neurotrauma ; 34(2): 414-422, 2017 01 15.
Artículo en Inglés | MEDLINE | ID: mdl-27142118

RESUMEN

Increasing evidence suggests that traumatic brain injury (TBI) may raise the risk of developing late-onset Parkinson's disease (PD). Recently, the peroxisome proliferation-activated receptor gamma (PPARγ) agonist pioglitazone has been demonstrated to be neuroprotective in animal models of neurodegeneration. The present study investigates the vulnerability of the nigrostriatal system after TBI, and intervention with pioglitazone treatment. Adult male Sprague-Dawley rats were subjected to sham or moderate midline fluid percussion brain injury (mFPI), followed by an intraperitoneal injection of 10 mg/kg pioglitazone or vehicle beginning 30 min after the injury and subsequently every 24 h for 5 days. Following injury, pro-inflammatory cytokines and chemokine were acutely increased in the striatum and substantia nigra within 6 h. Dopaminergic axonal damage and microglial activation were revealed using immunohistochemistry in the medial forebrain bundle at 1 day post-injury. Microglial activation identified by Iba1 and OX-6 immunostaining was persistently increased in the substantia nigra pars compacta 7 to 28 days post-injury. Further, brain injury induced significant dopaminergic neuronal loss, which was quantified by tyrosine hydroxylase immunostaining and retrograde fluorescent tracer fluorogold labeling in the nigra at 28 days. Loss of neurons was accompanied by increased extracellular dopamine (DA) turnover in the striatum, indicating enhanced dopaminergic activity in functional compensation after nigrostriatal damage. Strikingly, pioglitazone treatment greatly attenuated microglial activation and improved dopaminergic neuronal survival in the nigrostriatal system, which may promote locomotor recovery. These results suggest that interventions that attenuate secondary inflammation could be a feasible therapeutic treatment to improve outcome after TBI.


Asunto(s)
Traumatismos Difusos del Encéfalo/metabolismo , Neuronas Dopaminérgicas/metabolismo , Mediadores de Inflamación/metabolismo , Neostriado/metabolismo , Sustancia Negra/metabolismo , Tiazolidinedionas/uso terapéutico , Animales , Traumatismos Difusos del Encéfalo/efectos de los fármacos , Traumatismos Difusos del Encéfalo/patología , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/fisiología , Neuronas Dopaminérgicas/efectos de los fármacos , Neuronas Dopaminérgicas/patología , Inflamación/tratamiento farmacológico , Inflamación/metabolismo , Inflamación/patología , Mediadores de Inflamación/antagonistas & inhibidores , Masculino , Neostriado/efectos de los fármacos , Neostriado/patología , Degeneración Nerviosa/metabolismo , Degeneración Nerviosa/patología , Degeneración Nerviosa/prevención & control , Pioglitazona , Ratas , Ratas Sprague-Dawley , Sustancia Negra/efectos de los fármacos , Sustancia Negra/patología , Tiazolidinedionas/farmacología
19.
J Neurotrauma ; 34(1): 213-219, 2017 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-27306143

RESUMEN

In the pathophysiology of traumatic brain injury (TBI), the amygdala remains understudied, despite involvement in processing emotional and stressful stimuli associated with anxiety disorders, such as post-traumatic stress disorder (PTSD). Because the basolateral amygdala (BLA) integrates inputs from sensory and other limbic structures coordinating emotional learning and memory, injury-induced changes in circuitry may contribute to psychiatric sequelae of TBI. This study quantified temporal changes in dendritic complexity of BLA neurons after experimental diffuse TBI, modeled by midline fluid percussion injury. At post-injury days (PIDs) 1, 7, and 28, brain tissue from sham and brain-injured adult, male rats was processed for Golgi, glial fibrillary acidic protein (GFAP), or silver stain and analyzed to quantify BLA dendritic branch intersections, activated astrocytes, and regional neuropathology, respectively. Compared to sham, brain-injured rats at all PIDs showed enhanced dendritic branch intersections in both pyramidal and stellate BLA neuronal types, as evidenced by Sholl analysis. GFAP staining in the BLA was significantly increased at PID1 and 7 in comparison to sham. However, the BLA was relatively spared from neuropathology, demonstrated by an absence of argyrophilic accumulation over time, in contrast to other brain regions. These data suggest an early and persistent enhancement of dendritic complexity within the BLA after a single diffuse TBI. Increased dendritic complexity would alter information processing into and through the amygdala, contributing to emotional symptoms post-TBI, including PTSD.


Asunto(s)
Complejo Nuclear Basolateral/patología , Traumatismos Difusos del Encéfalo/patología , Lesiones Traumáticas del Encéfalo/patología , Dendritas/patología , Animales , Lesiones Traumáticas del Encéfalo/complicaciones , Hipertrofia , Masculino , Ratas , Ratas Sprague-Dawley , Factores de Tiempo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA