Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Resultados 1 - 20 de 17.410
Filtrar
Más filtros

Publication year range
1.
Cell ; 186(11): 2475-2491.e22, 2023 05 25.
Artículo en Inglés | MEDLINE | ID: mdl-37178688

RESUMEN

Holistic understanding of physio-pathological processes requires noninvasive 3D imaging in deep tissue across multiple spatial and temporal scales to link diverse transient subcellular behaviors with long-term physiogenesis. Despite broad applications of two-photon microscopy (TPM), there remains an inevitable tradeoff among spatiotemporal resolution, imaging volumes, and durations due to the point-scanning scheme, accumulated phototoxicity, and optical aberrations. Here, we harnessed the concept of synthetic aperture radar in TPM to achieve aberration-corrected 3D imaging of subcellular dynamics at a millisecond scale for over 100,000 large volumes in deep tissue, with three orders of magnitude reduction in photobleaching. With its advantages, we identified direct intercellular communications through migrasome generation following traumatic brain injury, visualized the formation process of germinal center in the mouse lymph node, and characterized heterogeneous cellular states in the mouse visual cortex, opening up a horizon for intravital imaging to understand the organizations and functions of biological systems at a holistic level.


Asunto(s)
Imagenología Tridimensional , Animales , Ratones , Imagenología Tridimensional/métodos , Microscopía Confocal/métodos
2.
Cell ; 184(10): 2715-2732.e23, 2021 05 13.
Artículo en Inglés | MEDLINE | ID: mdl-33852912

RESUMEN

Traumatic brain injury (TBI) is the largest non-genetic, non-aging related risk factor for Alzheimer's disease (AD). We report here that TBI induces tau acetylation (ac-tau) at sites acetylated also in human AD brain. This is mediated by S-nitrosylated-GAPDH, which simultaneously inactivates Sirtuin1 deacetylase and activates p300/CBP acetyltransferase, increasing neuronal ac-tau. Subsequent tau mislocalization causes neurodegeneration and neurobehavioral impairment, and ac-tau accumulates in the blood. Blocking GAPDH S-nitrosylation, inhibiting p300/CBP, or stimulating Sirtuin1 all protect mice from neurodegeneration, neurobehavioral impairment, and blood and brain accumulation of ac-tau after TBI. Ac-tau is thus a therapeutic target and potential blood biomarker of TBI that may represent pathologic convergence between TBI and AD. Increased ac-tau in human AD brain is further augmented in AD patients with history of TBI, and patients receiving the p300/CBP inhibitors salsalate or diflunisal exhibit decreased incidence of AD and clinically diagnosed TBI.


Asunto(s)
Enfermedad de Alzheimer/etiología , Enfermedad de Alzheimer/prevención & control , Lesiones Traumáticas del Encéfalo/complicaciones , Neuroprotección , Proteínas tau/metabolismo , Acetilación , Enfermedad de Alzheimer/metabolismo , Animales , Antiinflamatorios no Esteroideos/uso terapéutico , Biomarcadores/sangre , Biomarcadores/metabolismo , Lesiones Traumáticas del Encéfalo/metabolismo , Línea Celular , Diflunisal/uso terapéutico , Femenino , Gliceraldehído-3-Fosfato Deshidrogenasa (Fosforilante) , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Neuronas/metabolismo , Salicilatos/uso terapéutico , Sirtuina 1/metabolismo , Factores de Transcripción p300-CBP/antagonistas & inhibidores , Factores de Transcripción p300-CBP/metabolismo , Proteínas tau/sangre
3.
Cell ; 181(4): 784-799.e19, 2020 05 14.
Artículo en Inglés | MEDLINE | ID: mdl-32413299

RESUMEN

Swelling of the brain or spinal cord (CNS edema) affects millions of people every year. All potential pharmacological interventions have failed in clinical trials, meaning that symptom management is the only treatment option. The water channel protein aquaporin-4 (AQP4) is expressed in astrocytes and mediates water flux across the blood-brain and blood-spinal cord barriers. Here we show that AQP4 cell-surface abundance increases in response to hypoxia-induced cell swelling in a calmodulin-dependent manner. Calmodulin directly binds the AQP4 carboxyl terminus, causing a specific conformational change and driving AQP4 cell-surface localization. Inhibition of calmodulin in a rat spinal cord injury model with the licensed drug trifluoperazine inhibited AQP4 localization to the blood-spinal cord barrier, ablated CNS edema, and led to accelerated functional recovery compared with untreated animals. We propose that targeting the mechanism of calmodulin-mediated cell-surface localization of AQP4 is a viable strategy for development of CNS edema therapies.


Asunto(s)
Acuaporina 4/metabolismo , Edema/metabolismo , Edema/terapia , Animales , Acuaporina 4/fisiología , Astrocitos/metabolismo , Encéfalo/metabolismo , Edema Encefálico/metabolismo , Calmodulina/metabolismo , Sistema Nervioso Central/metabolismo , Edema/fisiopatología , Masculino , Ratas , Ratas Sprague-Dawley , Médula Espinal/metabolismo , Traumatismos de la Médula Espinal/metabolismo , Trifluoperazina/farmacología
4.
Proc Natl Acad Sci U S A ; 121(19): e2313568121, 2024 May 07.
Artículo en Inglés | MEDLINE | ID: mdl-38648470

RESUMEN

United States (US) Special Operations Forces (SOF) are frequently exposed to explosive blasts in training and combat, but the effects of repeated blast exposure (RBE) on SOF brain health are incompletely understood. Furthermore, there is no diagnostic test to detect brain injury from RBE. As a result, SOF personnel may experience cognitive, physical, and psychological symptoms for which the cause is never identified, and they may return to training or combat during a period of brain vulnerability. In 30 active-duty US SOF, we assessed the relationship between cumulative blast exposure and cognitive performance, psychological health, physical symptoms, blood proteomics, and neuroimaging measures (Connectome structural and diffusion MRI, 7 Tesla functional MRI, [11C]PBR28 translocator protein [TSPO] positron emission tomography [PET]-MRI, and [18F]MK6240 tau PET-MRI), adjusting for age, combat exposure, and blunt head trauma. Higher blast exposure was associated with increased cortical thickness in the left rostral anterior cingulate cortex (rACC), a finding that remained significant after multiple comparison correction. In uncorrected analyses, higher blast exposure was associated with worse health-related quality of life, decreased functional connectivity in the executive control network, decreased TSPO signal in the right rACC, and increased cortical thickness in the right rACC, right insula, and right medial orbitofrontal cortex-nodes of the executive control, salience, and default mode networks. These observations suggest that the rACC may be susceptible to blast overpressure and that a multimodal, network-based diagnostic approach has the potential to detect brain injury associated with RBE in active-duty SOF.


Asunto(s)
Traumatismos por Explosión , Personal Militar , Humanos , Traumatismos por Explosión/diagnóstico por imagen , Adulto , Masculino , Estados Unidos , Imagen por Resonancia Magnética , Femenino , Tomografía de Emisión de Positrones , Cognición/fisiología , Encéfalo/diagnóstico por imagen , Encéfalo/metabolismo , Adulto Joven
5.
Proc Natl Acad Sci U S A ; 120(26): e2301360120, 2023 06 27.
Artículo en Inglés | MEDLINE | ID: mdl-37339206

RESUMEN

Traumatic brain injury (TBI) is a pervasive problem worldwide for which no effective treatment is currently available. Although most studies have focused on the pathology of the injured brain, we have noted that the liver plays an important role in TBI. Using two mouse models of TBI, we found that the enzymatic activity of hepatic soluble epoxide hydrolase (sEH) was rapidly decreased and then returned to normal levels following TBI, whereas such changes were not observed in the kidney, heart, spleen, or lung. Interestingly, genetic downregulation of hepatic Ephx2 (which encodes sEH) ameliorates TBI-induced neurological deficits and promotes neurological function recovery, whereas overexpression of hepatic sEH exacerbates TBI-associated neurological impairments. Furthermore, hepatic sEH ablation was found to promote the generation of A2 phenotype astrocytes and facilitate the production of various neuroprotective factors associated with astrocytes following TBI. We also observed an inverted V-shaped alteration in the plasma levels of four EET (epoxyeicosatrienoic acid) isoforms (5,6-, 8,9-,11,12-, and 14,15-EET) following TBI which were negatively correlated with hepatic sEH activity. However, hepatic sEH manipulation bidirectionally regulates the plasma levels of 14,15-EET, which rapidly crosses the blood-brain barrier. Additionally, we found that the application of 14,15-EET mimicked the neuroprotective effect of hepatic sEH ablation, while 14,15-epoxyeicosa-5(Z)-enoic acid blocked this effect, indicating that the increased plasma levels of 14,15-EET mediated the neuroprotective effect observed after hepatic sEH ablation. These results highlight the neuroprotective role of the liver in TBI and suggest that targeting hepatic EET signaling could represent a promising therapeutic strategy for treating TBI.


Asunto(s)
Lesiones Traumáticas del Encéfalo , Fármacos Neuroprotectores , Animales , Ratones , Fármacos Neuroprotectores/farmacología , Eicosanoides , Astrocitos , Hígado , Epóxido Hidrolasas/genética
6.
Immunol Rev ; 306(1): 58-75, 2022 03.
Artículo en Inglés | MEDLINE | ID: mdl-35067941

RESUMEN

The central nervous system (CNS) has historically been viewed as an immunologically privileged site, but recent studies have uncovered a vast landscape of immune cells that reside primarily along its borders. While microglia are largely responsible for surveying the parenchyma, CNS barrier sites are inhabited by a plethora of different innate and adaptive immune cells that participate in everything from the defense against microbes to the maintenance of neural function. Static and dynamic imaging studies have revolutionized the field of neuroimmunology by providing detailed maps of CNS immune cells as well as information about how these cells move, organize, and interact during steady-state and inflammatory conditions. These studies have also redefined our understanding of neural-immune interactions at a cellular level and reshaped our conceptual view of immune privilege in this specialized compartment. This review will focus on insights gained using imaging techniques in the field of neuroimmunology, with an emphasis on anatomy and CNS immune dynamics during homeostasis, infectious diseases, injuries, and aging.


Asunto(s)
Sistema Nervioso Central , Neuroinmunomodulación , Homeostasis , Humanos
7.
J Neurosci ; 44(32)2024 Aug 07.
Artículo en Inglés | MEDLINE | ID: mdl-38844342

RESUMEN

Sleep slow waves are the hallmark of deeper non-rapid eye movement sleep. It is generally assumed that gray matter properties predict slow-wave density, morphology, and spectral power in healthy adults. Here, we tested the association between gray matter volume (GMV) and slow-wave characteristics in 27 patients with moderate-to-severe traumatic brain injury (TBI, 32.0 ± 12.2 years old, eight women) and compared that with 32 healthy controls (29.2 ± 11.5 years old, nine women). Participants underwent overnight polysomnography and cerebral MRI with a 3 Tesla scanner. A whole-brain voxel-wise analysis was performed to compare GMV between groups. Slow-wave density, morphology, and spectral power (0.4-6 Hz) were computed, and GMV was extracted from the thalamus, cingulate, insula, precuneus, and orbitofrontal cortex to test the relationship between slow waves and gray matter in regions implicated in the generation and/or propagation of slow waves. Compared with controls, TBI patients had significantly lower frontal and temporal GMV and exhibited a subtle decrease in slow-wave frequency. Moreover, higher GMV in the orbitofrontal cortex, insula, cingulate cortex, and precuneus was associated with higher slow-wave frequency and slope, but only in healthy controls. Higher orbitofrontal GMV was also associated with higher slow-wave density in healthy participants. While we observed the expected associations between GMV and slow-wave characteristics in healthy controls, no such associations were observed in the TBI group despite lower GMV. This finding challenges the presumed role of GMV in slow-wave generation and morphology.


Asunto(s)
Lesiones Traumáticas del Encéfalo , Sustancia Gris , Imagen por Resonancia Magnética , Sueño de Onda Lenta , Humanos , Femenino , Masculino , Sustancia Gris/diagnóstico por imagen , Sustancia Gris/patología , Sustancia Gris/fisiopatología , Adulto , Sueño de Onda Lenta/fisiología , Lesiones Traumáticas del Encéfalo/fisiopatología , Lesiones Traumáticas del Encéfalo/diagnóstico por imagen , Lesiones Traumáticas del Encéfalo/patología , Adulto Joven , Polisomnografía , Corteza Cerebral/fisiopatología , Corteza Cerebral/diagnóstico por imagen , Persona de Mediana Edad , Lesiones Encefálicas/fisiopatología , Lesiones Encefálicas/diagnóstico por imagen , Lesiones Encefálicas/patología
8.
J Neurosci ; 44(11)2024 Mar 13.
Artículo en Inglés | MEDLINE | ID: mdl-38326036

RESUMEN

Intercellular adhesion molecule-1 (ICAM-1) is identified as an initiator of neuroinflammatory responses that lead to neurodegeneration and cognitive and sensory-motor deficits in several pathophysiological conditions including traumatic brain injury (TBI). However, the underlying mechanisms of ICAM-1-mediated leukocyte adhesion and transmigration and its link with neuroinflammation and functional deficits following TBI remain elusive. Here, we hypothesize that blocking of ICAM-1 attenuates the transmigration of leukocytes to the brain and promotes functional recovery after TBI. The experimental TBI was induced in vivo by fluid percussion injury (25 psi) in male and female wild-type and ICAM-1-/- mice and in vitro by stretch injury (3 psi) in human brain microvascular endothelial cells (hBMVECs). We treated hBMVECs and animals with ICAM-1 CRISPR/Cas9 and conducted several biochemical analyses and demonstrated that CRISPR/Cas9-mediated ICAM-1 deletion mitigates blood-brain barrier (BBB) damage and leukocyte transmigration to the brain by attenuating the paxillin/focal adhesion kinase (FAK)-dependent Rho GTPase pathway. For analyzing functional outcomes, we used a cohort of behavioral tests that included sensorimotor functions, psychological stress analyses, and spatial memory and learning following TBI. In conclusion, this study could establish the significance of deletion or blocking of ICAM-1 in transforming into a novel preventive approach against the pathophysiology of TBI.


Asunto(s)
Lesiones Traumáticas del Encéfalo , Molécula 1 de Adhesión Intercelular , Animales , Femenino , Humanos , Masculino , Ratones , Encéfalo/metabolismo , Lesiones Traumáticas del Encéfalo/metabolismo , Sistemas CRISPR-Cas , Células Endoteliales/metabolismo , Proteína-Tirosina Quinasas de Adhesión Focal/metabolismo , Leucocitos , Paxillin , Proteínas de Unión al GTP rho/metabolismo
9.
Semin Cell Dev Biol ; 140: 22-34, 2023 05 15.
Artículo en Inglés | MEDLINE | ID: mdl-35786351

RESUMEN

Much of the focus of neuronal cell biology has been devoted to growth cone guidance, synaptogenesis, synaptic activity, plasticity, etc. The axonal shaft too has received much attention, mainly for its astounding ability to transmit action potentials and the transport of material over long distances. For these functions, the axonal cytoskeleton and membrane have been often assumed to play static structural roles. Recent experiments have changed this view by revealing an ultrastructure much richer in features than previously perceived and one that seems to be maintained at a dynamic steady state. The role of mechanics in this is only beginning to be broadly appreciated and appears to involve passive and active modes of coupling different biopolymer filaments, filament turnover dynamics and membrane biophysics. Axons, being unique cellular processes in terms of high aspect ratios and often extreme lengths, also exhibit unique passive mechanical properties that might have evolved to stabilize them under mechanical stress. In this review, we summarize the experiments that have exposed some of these features. It is our view that axonal mechanics deserves much more attention not only due to its significance in the development and maintenance of the nervous system but also due to the susceptibility of axons to injury and neurodegeneration.


Asunto(s)
Axones , Citoesqueleto , Axones/metabolismo , Citoesqueleto/metabolismo , Microtúbulos/metabolismo , Neuronas , Estrés Mecánico
10.
Brain ; 147(6): 2214-2229, 2024 Jun 03.
Artículo en Inglés | MEDLINE | ID: mdl-38802114

RESUMEN

Mild traumatic brain injury (mTBI) has emerged as a potential risk factor for the development of neurodegenerative conditions such as Alzheimer's disease and chronic traumatic encephalopathy. Blast mTBI, caused by exposure to a pressure wave from an explosion, is predominantly experienced by military personnel and has increased in prevalence and severity in recent decades. Yet the underlying pathology of blast mTBI is largely unknown. We examined the expression and localization of AQP4 in human post-mortem frontal cortex and observed distinct laminar differences in AQP4 expression following blast exposure. We also observed similar laminar changes in AQP4 expression and localization and delayed impairment of glymphatic function that emerged 28 days following blast injury in a mouse model of repetitive blast mTBI. In a cohort of veterans with blast mTBI, we observed that blast exposure was associated with an increased burden of frontal cortical MRI-visible perivascular spaces, a putative neuroimaging marker of glymphatic perivascular dysfunction. These findings suggest that changes in AQP4 and delayed glymphatic impairment following blast injury may render the post-traumatic brain vulnerable to post-concussive symptoms and chronic neurodegeneration.


Asunto(s)
Acuaporina 4 , Traumatismos por Explosión , Sistema Glinfático , Adulto , Anciano , Animales , Femenino , Humanos , Masculino , Ratones , Persona de Mediana Edad , Acuaporina 4/metabolismo , Traumatismos por Explosión/complicaciones , Traumatismos por Explosión/patología , Traumatismos por Explosión/metabolismo , Conmoción Encefálica/metabolismo , Conmoción Encefálica/complicaciones , Conmoción Encefálica/patología , Conmoción Encefálica/fisiopatología , Lesiones Traumáticas del Encéfalo/metabolismo , Lesiones Traumáticas del Encéfalo/complicaciones , Lesiones Traumáticas del Encéfalo/patología , Lóbulo Frontal/metabolismo , Lóbulo Frontal/patología , Lóbulo Frontal/diagnóstico por imagen , Sistema Glinfático/metabolismo , Sistema Glinfático/patología , Imagen por Resonancia Magnética , Ratones Endogámicos C57BL , Veteranos
11.
Brain ; 147(5): 1914-1925, 2024 May 03.
Artículo en Inglés | MEDLINE | ID: mdl-38181433

RESUMEN

Autologous bone marrow mononuclear cells (BMMNCs) infused after severe traumatic brain injury have shown promise for treating the injury. We evaluated their impact in children, particularly their hypothesized ability to preserve the blood-brain barrier and diminish neuroinflammation, leading to structural CNS preservation with improved outcomes. We performed a randomized, double-blind, placebo-sham-controlled Bayesian dose-escalation clinical trial at two children's hospitals in Houston, TX and Phoenix, AZ, USA (NCT01851083). Patients 5-17 years of age with severe traumatic brain injury (Glasgow Coma Scale score ≤ 8) were randomized to BMMNC or placebo (3:2). Bone marrow harvest, cell isolation and infusion were completed by 48 h post-injury. A Bayesian continuous reassessment method was used with cohorts of size 3 in the BMMNC group to choose the safest between two doses. Primary end points were quantitative brain volumes using MRI and microstructural integrity of the corpus callosum (diffusivity and oedema measurements) at 6 months and 12 months. Long-term functional outcomes and ventilator days, intracranial pressure monitoring days, intensive care unit days and therapeutic intensity measures were compared between groups. Forty-seven patients were randomized, with 37 completing 1-year follow-up (23 BMMNC, 14 placebo). BMMNC treatment was associated with an almost 3-day (23%) reduction in ventilator days, 1-day (16%) reduction in intracranial pressure monitoring days and 3-day (14%) reduction in intensive care unit (ICU) days. White matter volume at 1 year in the BMMNC group was significantly preserved compared to placebo [decrease of 19 891 versus 40 491, respectively; mean difference of -20 600, 95% confidence interval (CI): -35 868 to -5332; P = 0.01], and the number of corpus callosum streamlines was reduced more in placebo than BMMNC, supporting evidence of preserved corpus callosum connectivity in the treated groups (-431 streamlines placebo versus -37 streamlines BMMNC; mean difference of -394, 95% CI: -803 to 15; P = 0.055), but this did not reach statistical significance due to high variability. We conclude that autologous BMMNC infusion in children within 48 h after severe traumatic brain injury is safe and feasible. Our data show that BMMNC infusion led to: (i) shorter intensive care duration and decreased ICU intensity; (ii) white matter structural preservation; and (iii) enhanced corpus callosum connectivity and improved microstructural metrics.


Asunto(s)
Trasplante de Médula Ósea , Lesiones Traumáticas del Encéfalo , Trasplante Autólogo , Humanos , Niño , Lesiones Traumáticas del Encéfalo/terapia , Masculino , Femenino , Adolescente , Método Doble Ciego , Preescolar , Trasplante de Médula Ósea/métodos , Trasplante Autólogo/métodos , Imagen por Resonancia Magnética , Resultado del Tratamiento , Leucocitos Mononucleares/trasplante , Teorema de Bayes
12.
Brain ; 147(4): 1321-1330, 2024 Apr 04.
Artículo en Inglés | MEDLINE | ID: mdl-38412555

RESUMEN

The pathophysiological underpinnings of critically disrupted brain connectomes resulting in coma are poorly understood. Inflammation is potentially an important but still undervalued factor. Here, we present a first-in-human prospective study using the 18-kDa translocator protein (TSPO) radioligand 18F-DPA714 for PET imaging to allow in vivo neuroimmune activation quantification in patients with coma (n = 17) following either anoxia or traumatic brain injuries in comparison with age- and sex-matched controls. Our findings yielded novel evidence of an early inflammatory component predominantly located within key cortical and subcortical brain structures that are putatively implicated in consciousness emergence and maintenance after severe brain injury (i.e. mesocircuit and frontoparietal networks). We observed that traumatic and anoxic patients with coma have distinct neuroimmune activation profiles, both in terms of intensity and spatial distribution. Finally, we demonstrated that both the total amount and specific distribution of PET-measurable neuroinflammation within the brain mesocircuit were associated with the patient's recovery potential. We suggest that our results can be developed for use both as a new neuroprognostication tool and as a promising biometric to guide future clinical trials targeting glial activity very early after severe brain injury.


Asunto(s)
Lesiones Encefálicas , Coma Postraumatismo Craneoencefálico , Humanos , Coma/complicaciones , Coma Postraumatismo Craneoencefálico/complicaciones , Estudios Prospectivos , Imagen por Resonancia Magnética/métodos , Encéfalo/metabolismo , Lesiones Encefálicas/complicaciones , Hipoxia/complicaciones , Receptores de GABA/metabolismo
13.
Cereb Cortex ; 34(4)2024 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-38629798

RESUMEN

The prevalence of posttraumatic olfactory dysfunction in children after mild traumatic brain injury ranges from 3 to 58%, with potential factors influencing this variation, including traumatic brain injury severity and assessment methods. This prospective longitudinal study examines the association between mild traumatic brain injury and olfactory dysfunction in children. Seventy-five pediatric patients with mild traumatic brain injury and an age-matched healthy control group were enrolled. Olfactory function was assessed using the Sniffin' Sticks battery, which focuses on olfactory threshold and odor identification. The study found that children with mild traumatic brain injury had impaired olfactory function compared with healthy controls, particularly in olfactory threshold scores. The prevalence of olfactory dysfunction in the patient group was 33% and persisted for 1 yr. No significant association was found between traumatic brain injury symptoms (e.g. amnesia, loss of consciousness) and olfactory dysfunction. The study highlights the importance of assessing olfactory function in children after mild traumatic brain injury, given its potential impact on daily life. Although most olfactory dysfunction appears transient, long-term follow-up is essential to fully understand the recovery process. The findings add valuable insights to the limited literature on this topic and urge the inclusion of olfactory assessments in the management of pediatric mild traumatic brain injury.


Asunto(s)
Conmoción Encefálica , Lesiones Traumáticas del Encéfalo , Trastornos del Olfato , Humanos , Niño , Conmoción Encefálica/complicaciones , Estudios de Casos y Controles , Trastornos del Olfato/etiología , Estudios Prospectivos , Estudios Longitudinales , Olfato , Odorantes , Lesiones Traumáticas del Encéfalo/complicaciones
14.
Cereb Cortex ; 34(1)2024 01 14.
Artículo en Inglés | MEDLINE | ID: mdl-37997466

RESUMEN

Blood proteins are emerging as potential biomarkers for mild traumatic brain injury (mTBI). Molecular pathology of mTBI underscores the critical roles of neuronal injury, neuroinflammation, and vascular health in disease progression. However, the temporal profile of blood biomarkers associated with the aforementioned molecular pathology after CT-negative mTBI, their diagnostic and prognostic potential, and their utility in monitoring white matter integrity and progressive brain atrophy remain unclear. Thus, we investigated serum biomarkers and neuroimaging in a longitudinal cohort, including 103 CT-negative mTBI patients and 66 matched healthy controls (HCs). Angiogenic biomarker vascular endothelial growth factor (VEGF) exhibited the highest area under the curve of 0.88 in identifying patients from HCs. Inflammatory biomarker interleukin-1ß and neuronal cell body injury biomarker ubiquitin carboxyl-terminal hydrolase L1 were elevated in acute-stage patients and associated with deterioration of cognitive function from acute-stage to 6-12 mo post-injury period. Notably, axonal injury biomarker neurofilament light (NfL) was elevated in acute-stage patients, with higher levels associated with impaired white matter integrity in acute-stage and progressive gray and white matter atrophy from 3- to 6-12 mo post-injury period. Collectively, our findings emphasized the potential clinical value of serum biomarkers, particularly NfL and VEGF, in diagnosing mTBI and monitoring disease progression.


Asunto(s)
Conmoción Encefálica , Humanos , Conmoción Encefálica/diagnóstico por imagen , Factor A de Crecimiento Endotelial Vascular , Proteínas de Neurofilamentos , Progresión de la Enfermedad , Biomarcadores , Atrofia/patología , Tomografía Computarizada por Rayos X , Encéfalo/diagnóstico por imagen , Encéfalo/patología
15.
Cereb Cortex ; 34(7)2024 Jul 03.
Artículo en Inglés | MEDLINE | ID: mdl-39077916

RESUMEN

The lifetime effects of repetitive head impacts have captured considerable public and scientific interest over the past decade, yet a knowledge gap persists in our understanding of midlife neurological well-being, particularly in amateur level athletes. This study aimed to identify the effects of lifetime exposure to sports-related head impacts on brain morphology in retired, amateur athletes. This cross-sectional study comprised of 37 former amateur contact sports athletes and 21 age- and sex-matched noncontact athletes. High-resolution anatomical, T1 scans were analyzed for the cortical morphology, including cortical thickness, sulcal depth, and sulcal curvature, and cognitive function was assessed using the Dementia Rating Scale-2. Despite no group differences in cognitive functions, the contact group exhibited significant cortical thinning particularly in the bilateral frontotemporal regions and medial brain regions, such as the cingulate cortex and precuneus, compared to the noncontact group. Deepened sulcal depth and increased sulcal curvature across all four lobes of the brain were also notable in the contact group. These data suggest that brain morphology of middle-aged former amateur contact athletes differs from that of noncontact athletes and that lifetime exposure to repetitive head impacts may be associated with neuroanatomical changes.


Asunto(s)
Atletas , Corteza Cerebral , Imagen por Resonancia Magnética , Humanos , Masculino , Femenino , Corteza Cerebral/diagnóstico por imagen , Corteza Cerebral/patología , Corteza Cerebral/anatomía & histología , Estudios Transversales , Persona de Mediana Edad , Traumatismos en Atletas/patología , Traumatismos en Atletas/diagnóstico por imagen , Anciano , Conmoción Encefálica/patología , Conmoción Encefálica/diagnóstico por imagen , Cognición/fisiología
16.
J Neurosci ; 43(2): 319-332, 2023 01 11.
Artículo en Inglés | MEDLINE | ID: mdl-36446585

RESUMEN

Mechanical impact-induced primary injury after traumatic brain injury (TBI) leads to acute microglial pro-inflammatory activation and consequently mediates neurodegeneration, which is a major secondary brain injury mechanism. However, the detailed pathologic cascades have not been fully elucidated, partially because of the pathologic complexity in animal TBI models. Although there are several in vitro TBI models, none of them closely mimic post-TBI microglial activation. In the present study, we aimed to establish an in vitro TBI model, specifically reconstituting the pro-inflammatory activation and associated neurodegeneration following TBI. We proposed three sets of experiments. First, we established a needle scratch injured neuron-induced microglial activation and neurodegeneration in vitro model of TBI. Second, we compared microglial pro-inflammatory cytokines profiles between the in vitro TBI model and TBI in male mice. Additionally, we validated the role of injured neurons-derived damage-associated molecular patterns in amplifying microglial pro-inflammatory pathways using the in vitro TBI model. Third, we applied the in vitro model for the first time to characterize the cellular metabolic profile of needle scratch injured-neuron-activated microglia, and define the role of metabolic reprogramming in mediating pro-inflammatory microglial activation and mediated neurodegeneration. Our results showed that we successfully established a novel in vitro TBI model, which closely mimics primary neuronal injury-triggered microglial pro-inflammatory activation and mediated neurodegeneration after TBI. This in vitro model provides an advanced and highly translational platform for dissecting interactions in the pathologic processes of neuronal injury-microglial activation-neuronal degeneration cascade, and elucidating the detailed underlying cellular and molecular insights after TBI.SIGNIFICANCE STATEMENT Microglial activation is a key component of acute neuroinflammation that leads to neurodegeneration and long-term neurologic outcome deficits after TBI. However, it is not feasible to truly dissect primary neuronal injury-induced microglia activation, and consequently mediated neurodegeneration in vivo Furthermore, there is currently lacking of in vitro TBI models closely mimicking the TBI primary injury-mediated microglial activation. In this study, we successfully established and validated a novel in vitro TBI model of microglial activation, and for the first time, characterized the cellular metabolic profile of microglia in this model. This novel microglial activation in vitro TBI model will help in elucidating microglial inflammatory activation and consequently associated neurodegeneration after TBI.


Asunto(s)
Lesiones Traumáticas del Encéfalo , Microglía , Ratones , Masculino , Animales , Microglía/metabolismo , Lesiones Traumáticas del Encéfalo/patología , Macrófagos/metabolismo , Neuronas/metabolismo , Ratones Endogámicos C57BL
17.
J Proteome Res ; 23(1): 249-263, 2024 01 05.
Artículo en Inglés | MEDLINE | ID: mdl-38064581

RESUMEN

In many cases of traumatic brain injury (TBI), conspicuous abnormalities, such as scalp wounds and intracranial hemorrhages, abate over time. However, many unnoticeable symptoms, including cognitive, emotional, and behavioral dysfunction, often last from several weeks to years after trauma, even for mild injuries. Moreover, the cause of such persistence of symptoms has not been examined extensively. Recent studies have implicated the dysregulation of the molecular system in the injured brain, necessitating an in-depth analysis of the proteome and signaling pathways that mediate the consequences of TBI. Thus, in this study, the brain proteomes of two TBI models were examined by quantitative proteomics during the recovery period to determine the molecular mechanisms of TBI. Our results show that the proteomes in both TBI models undergo distinct changes. A bioinformatics analysis demonstrated robust activation and inhibition of signaling pathways and core proteins that mediate biological processes after brain injury. These findings can help determine the molecular mechanisms that underlie the persistent effects of TBI and identify novel targets for drug interventions.


Asunto(s)
Lesiones Traumáticas del Encéfalo , Lesiones Encefálicas , Animales , Roedores/metabolismo , Proteómica/métodos , Proteoma/genética , Proteoma/metabolismo , Lesiones Traumáticas del Encéfalo/metabolismo , Lesiones Encefálicas/metabolismo
18.
J Proteome Res ; 23(1): 397-408, 2024 01 05.
Artículo en Inglés | MEDLINE | ID: mdl-38096401

RESUMEN

Repeated blast-traumatic brain injury (blast-TBI) has been hypothesized to cause persistent and unusual neurological and psychiatric symptoms in service members returning from war zones. Blast-wave primary effects have been supposed to induce damage and molecular alterations in the brain. However, the mechanisms through which the primary effect of an explosive-driven blast wave generate brain lesions and induce brain consequences are incompletely known. Prior findings from rat brains exposed to two consecutive explosive-driven blasts showed molecular changes (hyperphosphorylated-Tau, AQP4, S100ß, PDGF, and DNA-polymerase-ß) that varied in magnitude and direction across different brain regions. We aimed to compare, in an unbiased manner, the proteomic profile in the hippocampus of double blast vs sham rats using mass spectrometry (MS). Data showed differences in up- and down-regulation for protein abundances in the hippocampus of double blast vs sham rats. Tandem mass tag (TMT)-MS results showed 136 up-regulated and 94 down-regulated proteins between the two groups (10.25345/C52B8VP0X). These TMT-MS findings revealed changes never described before in blast studies, such as increases in MAGI3, a scaffolding protein at cell-cell junctions, which were confirmed by Western blotting analyses. Due to the absence of behavioral and obvious histopathological changes as described in our previous publications, these proteomic data further support the existence of an asymptomatic blast-induced molecular altered status (ABIMAS) associated with specific protein changes in the hippocampus of rats repeatedly expsosed to blast waves generated by explosive-driven detonations.


Asunto(s)
Traumatismos por Explosión , Lesiones Traumáticas del Encéfalo , Sustancias Explosivas , Ratas , Animales , Traumatismos por Explosión/complicaciones , Traumatismos por Explosión/patología , Proteómica , Lesiones Traumáticas del Encéfalo/patología , Hipocampo/patología , Modelos Animales de Enfermedad
19.
Physiol Genomics ; 56(4): 301-316, 2024 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-38145288

RESUMEN

The gut-brain axis interconnects the central nervous system (CNS) and the commensal bacteria of the gastrointestinal tract. The composition of the diet consumed by the host influences the richness of the microbial populations. Traumatic brain injury (TBI) produces profound neurocognitive damage, but it is unknown how diet influences the microbiome following TBI. The present work investigates the impact of a chow diet versus a 60% fat diet (HFD) on fecal microbiome populations in juvenile rats following TBI. Twenty-day-old male rats were placed on one of two diets for 9 days before sustaining either a Sham or TBI via the Closed Head Injury Model of Engineered Rotational Acceleration (CHIMERA). Fecal samples were collected at both 1- and 9-days postinjury. Animals were cognitively assessed in the novel object recognition tests at 8 days postinjury. Fecal microbiota DNA was isolated and sequenced. Twenty days of HFD feeding did not alter body weight, but fat mass was elevated in HFD compared with Chow rats. TBI animals had a greater percentage of entries to the novel object quadrant than Sham counterparts, P < 0.05. The Firmicutes/Bacteroidetes ratio was significantly higher in TBI than in the Sham, P < 0.05. Microbiota of the Firmicutes lineage exhibited perturbations by both injury and diet that were sustained at both time points. Linear regression analyses were performed to associate bacteria with metabolic and neurocognitive endpoints. For example, counts of Lachnospiraceae were negatively associated with percent entries into the novel object quadrant. Taken together, these data suggest that both diet and injury produce robust shifts in microbiota, which may have long-term implications for chronic health.NEW & NOTEWORTHY Traumatic brain injury (TBI) produces memory and learning difficulties. Diet profoundly influences the populations of gut microbiota. Following traumatic brain injury in a pediatric model consuming either a healthy or high-fat diet (HFD), significant shifts in bacterial populations occur, of which, some are associated with diet, whereas others are associated with neurocognitive performance. More work is needed to determine whether these microbes can therapeutically improve learning following trauma to the brain.


Asunto(s)
Lesiones Traumáticas del Encéfalo , Disfunción Cognitiva , Microbioma Gastrointestinal , Humanos , Niño , Ratas , Masculino , Animales , Dieta Alta en Grasa/efectos adversos , Microbioma Gastrointestinal/genética , Bacterias , Lesiones Traumáticas del Encéfalo/microbiología
20.
Glia ; 72(4): 728-747, 2024 04.
Artículo en Inglés | MEDLINE | ID: mdl-38180164

RESUMEN

Senescence is a negative prognostic factor for outcome and recovery following traumatic brain injury (TBI). TBI-induced white matter injury may be partially due to oligodendrocyte demise. We hypothesized that the regenerative capacity of oligodendrocyte precursor cells (OPCs) declines with age. To test this hypothesis, the regenerative capability of OPCs in young [(10 weeks ±2 (SD)] and aged [(62 weeks ±10 (SD)] mice was studied in mice subjected to central fluid percussion injury (cFPI), a TBI model causing widespread white matter injury. Proliferating OPCs were assessed by immunohistochemistry for the proliferating cell nuclear antigen (PCNA) marker and labeled by 5-ethynyl-2'-deoxyuridine (EdU) administered daily through intraperitoneal injections (50 mg/kg) from day 2 to day 6 after cFPI. Proliferating OPCs were quantified in the corpus callosum and external capsule on day 2 and 7 post-injury (dpi). The number of PCNA/Olig2-positive and EdU/Olig2-positive cells were increased at 2dpi (p < .01) and 7dpi (p < .01), respectively, in young mice subjected to cFPI, changes not observed in aged mice. Proliferating Olig2+/Nestin+ cells were less common (p < .05) in the white matter of brain-injured aged mice, without difference in proliferating Olig2+/PDGFRα+ cells, indicating a diminished proliferation of progenitors with different spatial origin. Following TBI, co-staining for EdU/CC1/Olig2 revealed a reduced number of newly generated mature oligodendrocytes in the white matter of aged mice when compared to the young, brain-injured mice (p < .05). We observed an age-related decline of oligodendrogenesis following experimental TBI that may contribute to the worse outcome of elderly patients following TBI.


Asunto(s)
Lesiones Traumáticas del Encéfalo , Lesiones Encefálicas , Sustancia Blanca , Humanos , Anciano , Ratones , Animales , Antígeno Nuclear de Célula en Proliferación , Encéfalo , Oligodendroglía , Ratones Endogámicos C57BL
SELECCIÓN DE REFERENCIAS
Detalles de la búsqueda