Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
Más filtros











Base de datos
Intervalo de año de publicación
1.
PLoS One ; 16(12): e0261599, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34905584

RESUMEN

[This corrects the article DOI: 10.1371/journal.pone.0233263.].

2.
Sci Rep ; 10(1): 11991, 2020 07 20.
Artículo en Inglés | MEDLINE | ID: mdl-32686718

RESUMEN

Traumatic brain injury (TBI) results in a cascade of cellular responses, which produce neuroinflammation, partly due to microglial activation. Transforming from surveying to primed phenotypes, microglia undergo considerable molecular changes. However, specific microglial profiles in rat remain elusive due to tedious methodology and limited availability of reagents. Here, we present a flow cytometry-based analysis of rat microglia 24 h after TBI using the controlled cortical impact model, validated with a bioinformatics approach. Isolated microglia are analyzed for morphological changes and their expression of activation markers using flow cytometry, traditional gating-based analysis methods and support the data by employing bioinformatics statistical tools. We use CD45, CD11b/c, and p2y12 receptor to identify microglia and evaluate their activation state using CD32, CD86, RT1B, CD200R, and CD163. The results from logic-gated flow cytometry analysis was validated with bioinformatics-based analysis and machine learning algorithms to detect quantitative changes in morphology and marker expression in microglia due to activation following TBI.


Asunto(s)
Biomarcadores/metabolismo , Lesiones Traumáticas del Encéfalo/metabolismo , Biología Computacional , Citometría de Flujo , Microglía/metabolismo , Animales , Lesiones Traumáticas del Encéfalo/patología , Polaridad Celular , Tamaño de la Célula , Microglía/patología , Ratas Sprague-Dawley
3.
PLoS One ; 15(5): e0233263, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32453741

RESUMEN

In the U.S., approximately 1.7 million people suffer traumatic brain injury each year, with many enduring long-term consequences and significant medical and rehabilitation costs. The primary injury causes physical damage to neurons, glia, fiber tracts and microvasculature, which is then followed by secondary injury, consisting of pathophysiological mechanisms including an immune response, inflammation, edema, excitotoxicity, oxidative damage, and cell death. Most attempts at intervention focus on protection, repair or regeneration, with regenerative medicine becoming an intensively studied area over the past decade. The use of stem cells has been studied in many disease and injury models, using stem cells from a variety of sources and applications. In this study, human adipose-derived mesenchymal stromal cells (MSCs) were administered at early (3 days) and delayed (14 days) time points after controlled cortical impact (CCI) injury in rats. Animals were routinely assessed for neurological and vestibulomotor deficits, and at 32 days post-injury, brain tissue was processed by flow cytometry and immunohistochemistry to analyze neuroinflammation. Treatment with HB-adMSC at either 3d or 14d after injury resulted in significant improvements in neurocognitive outcome and a change in neuroinflammation one month after injury.


Asunto(s)
Lesiones Traumáticas del Encéfalo/terapia , Trasplante de Células Madre Mesenquimatosas , Tejido Adiposo/citología , Animales , Encéfalo/patología , Lesiones Traumáticas del Encéfalo/patología , Lesiones Traumáticas del Encéfalo/psicología , Modelos Animales de Enfermedad , Humanos , Inflamación/patología , Masculino , Aprendizaje por Laberinto , Células Madre Mesenquimatosas/citología , Destreza Motora , Neurogénesis , Ratas Sprague-Dawley , Factores de Tiempo
4.
Heliyon ; 5(10): e02532, 2019 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-31667385

RESUMEN

BACKGROUND: Human multipotent adult progenitor cells (MAPC®) are an emerging therapy for traumatic brain injury (TBI); however, clinically translating a therapy involves overcoming many factors in vivo which are not present in pre-clinical testing. In this study we examined clinical parameters in vitro that may impact cell therapy efficacy. METHODS: MAPC were infused through varying gauged needles and catheters with and without chlorhexidine, and their viability tested with trypan blue exclusion. MAPC were co-cultured with phenytoin and celecoxib at relevant clinical concentrations for 1 h and 24 h. Anti-inflammatory potency was tested using a stimulated rat splenocyte co-culture and ELISA for TNF-α production. MAPC were cultured under different osmolar concentrations and stained with propidium iodide for viability. Anti-inflammatory potency was tested by co-culture of MAPC with naïve lymphocytes activated by CD3/CD28 beads, and Click-iT® Plus EdU was used to quantify proliferation by flow cytometry. RESULTS: The mean viability of the MAPC infused via needles was 95 ± 1%; no difference was seen with varying flow rate, but viability was notably reduced by chlorhexidine. MAPC function was not impaired by co-culture with phenytoin, celecoxib, or combination with both. Co-culture with phenytoin showed a decrease in TNF-α production as compared to the MAPC control. MAPC cultured at varying osmolar concentrations all had viabilities greater than 90% with no statistical difference between them. Co-culture of MAPC with CD3/CD28 activated PBMCs showed a significant reduction in proliferation as measured by EdU uptake. DISCUSSION: Needle diameter, phenytoin, celecoxib, and a relevant range of osmolarities do not impair MAPC viability or anti-inflammatory potency in vitro.

5.
Sci Rep ; 8(1): 480, 2018 01 11.
Artículo en Inglés | MEDLINE | ID: mdl-29323194

RESUMEN

No current clinical intervention can alter the course of acute spinal cord injury (SCI), or appreciably improve neurological outcome. Mesenchymal stromal cells (MSCs) have been shown to modulate the injury sequelae of SCI largely via paracrine effects, although the mechanisms remain incompletely understood. One potential modality is through secretion of extracellular vesicles (EVs). In this study, we investigate whether systemic administration of EVs isolated from human MSCs (MSCEv) has the potential to be efficacious as an alternative to cell-based therapy for SCI. Additionally, we investigate whether EVs isolated from human MSCs stimulated with pro-inflammatory cytokines have enhanced anti-inflammatory effects when administered after SCI. Immunohistochemistry supported the quantitative analysis, demonstrating a diminished inflammatory response with apparent astrocyte and microglia disorganization in cord tissue up to 10 mm caudal to the injury site. Locomotor recovery scores showed significant improvement among animals treated with MSCEv. Significant increases in mechanical sensitivity threshold were observed in animals treated with EVs from either naïve MSC (MSCEvwt) or stimulated MSC (MSCEv+), with a statistically significant increase in threshold for MSCEv+-treated animals when compared to those that received MSCEvwt. In conclusion, these data show that treatment of acute SCI with extracellular vesicles derived from human MSCs attenuates neuroinflammation and improves functional recovery.


Asunto(s)
Vesículas Extracelulares/trasplante , Microglía/metabolismo , Traumatismos de la Médula Espinal/terapia , Animales , Astrocitos/citología , Astrocitos/metabolismo , Modelos Animales de Enfermedad , Vesículas Extracelulares/metabolismo , Humanos , Inflamación , Locomoción/fisiología , Masculino , Células Madre Mesenquimatosas/citología , Células Madre Mesenquimatosas/metabolismo , Microglía/citología , Neutrófilos/citología , Neutrófilos/metabolismo , Ratas , Ratas Sprague-Dawley , Receptores de IgG/metabolismo , Recuperación de la Función , Bazo/citología , Bazo/inmunología , Resultado del Tratamiento
6.
Stem Cells ; 36(1): 79-90, 2018 01.
Artículo en Inglés | MEDLINE | ID: mdl-29076623

RESUMEN

Extracellular vesicles (EVs) secreted by mesenchymal stromal cells (MSCs) have been proposed to be a key mechanistic link in the therapeutic efficacy of cells in response to cellular injuries through paracrine effects. We hypothesize that inflammatory stimulation of MSCs results in the release of EVs that have greater anti-inflammatory effects. The present study evaluates the immunomodulatory abilities of EVs derived from inflammation-stimulated and naive MSCs (MSCEv+ and MSCEv, respectively) isolated using a current Good Manufacturing Practice-compliant tangential flow filtration system. Detailed characterization of both EVs revealed differences in protein composition, cytokine profiles, and RNA content, despite similarities in size and expression of common surface markers. MSCEv+ further attenuated release of pro-inflammatory cytokines in vitro when compared to MSCEv, with a distinctly different pattern of EV-uptake by activated primary leukocyte subpopulations. The efficacy of EVs was partially attributed to COX2/PGE2 expression. The present study demonstrates that inflammatory stimulation of MSCs renders release of EVs that have enhanced anti-inflammatory properties partially due to COX2/PGE2 pathway alteration. Stem Cells 2018;36:79-90.


Asunto(s)
Vesículas Extracelulares/metabolismo , Inflamación/metabolismo , Células Madre Mesenquimatosas/metabolismo , Microscopía Electrónica de Transmisión/métodos , Humanos
7.
Photoacoustics ; 2(2): 75-80, 2014 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-25302157

RESUMEN

Surgical drainage of intracranial hematomas is often required within the first four hours after traumatic brain injury (TBI) to avoid death or severe disability. Although CT and MRI permit hematoma diagnosis, they can be used only at a major health-care facility. This delays hematoma diagnosis and therapy. We proposed to use an optoacoustic technique for rapid, noninvasive diagnosis of hematomas. In this study we developed a near-infrared OPO-based optoacoustic system for hematoma diagnosis and cerebral venous blood oxygenation monitoring in rats. A specially-designed blast device was used to inflict TBI in anesthetized rats. Optoacoustic signals were recorded from the superior sagittal sinus and hematomas that allowed for measurements of their oxygenations. These results indicate that the optoacoustic technique may be used for early diagnosis of hematomas and may provide important information for improving outcomes in patients with TBI.

8.
Mol Pain ; 3: 13, 2007 Jun 05.
Artículo en Inglés | MEDLINE | ID: mdl-17550594

RESUMEN

Corticotropin-releasing factor receptor CRF1 has been implicated in the neurobiological mechanisms of anxiety and depression. The amygdala plays an important role in affective states and disorders such as anxiety and depression. The amygdala is also emerging as a neural substrate of pain affect. However, the involvement of the amygdala in the interaction of pain and anxiety remains to be determined. This study tested the hypothesis that CRF1 receptors in the amygdala are critically involved in pain-related anxiety. Anxiety-like behavior was determined in adult male rats using the elevated plus maze (EPM) test. The open-arm preference (ratio of open arm entries to the total number of entries) was measured. Nocifensive behavior was assessed by measuring hindlimb withdrawal thresholds for noxious mechanical stimulation of the knee. Measurements were made in normal rats and in rats with arthritis induced in one knee by intraarticular injections of kaolin/carrageenan. A selective CRF1 receptor antagonist (NBI27914) or vehicle was administered systemically (i.p.) or into the central nucleus of the amygdala (CeA, by microdialysis). The arthritis group showed a decreased preference for the open arms in the EPM and decreased hindlimb withdrawal thresholds. Systemic or intraamygdalar (into the CeA) administration of NBI27914, but not vehicle, inhibited anxiety-like behavior and nocifensive pain responses, nearly reversing the arthritis pain-related changes. This study shows for the first time that CRF1 receptors in the amygdala contribute critically to pain-related anxiety-like behavior and nocifensive responses in a model of arthritic pain. The results are a direct demonstration that the clinically well-documented relationship between pain and anxiety involves the amygdala.


Asunto(s)
Amígdala del Cerebelo/metabolismo , Ansiedad/fisiopatología , Dolor/fisiopatología , Receptores de Hormona Liberadora de Corticotropina/fisiología , Amígdala del Cerebelo/fisiopatología , Compuestos de Anilina/administración & dosificación , Compuestos de Anilina/farmacología , Animales , Ansiedad/etiología , Ansiedad/metabolismo , Artritis Experimental/inducido químicamente , Artritis Experimental/complicaciones , Artritis Experimental/fisiopatología , Carragenina , Miembro Posterior/fisiopatología , Caolín , Masculino , Aprendizaje por Laberinto , Dolor/complicaciones , Pirimidinas/administración & dosificación , Pirimidinas/farmacología , Ratas , Ratas Sprague-Dawley , Receptores de Hormona Liberadora de Corticotropina/antagonistas & inhibidores
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA