Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 41
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Exp Neurol ; 374: 114714, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-38325653

RESUMEN

Traumatic brain injury (TBI) is a leading cause of disability and increases the risk of developing neurodegenerative diseases. The mechanisms linking TBI to neurodegeneration remain to be defined. It has been proposed that the induction of cellular senescence after injury could amplify neuroinflammation and induce long-term tissue changes. The induction of a senescence response post-injury in the immature brain has yet to be characterised. We carried out two types of brain injury in juvenile CD1 mice: invasive TBI using controlled cortical impact (CCI) and repetitive mild TBI (rmTBI) using weight drop injury. The analysis of senescence-related signals showed an increase in γH2AX-53BP1 nuclear foci, p53, p19ARF, and p16INK4a expression in the CCI group, 5 days post-injury (dpi). At 35 days, the difference was no longer statistically significant. Gene expression showed the activation of different senescence pathways in the ipsilateral and contralateral hemispheres in the injured mice. CCI-injured mice showed a neuroinflammatory early phase after injury (increased Iba1 and GFAP expression), which persisted for GFAP. After CCI, there was an increase at 5 days in p16INK4, whereas in rmTBI, a significant increase was seen at 35 dpi. Both injuries caused a decrease in p21 at 35 dpi. In rmTBI, other markers showed no significant change. The PCR array data predicted the activation of pathways connected to senescence after rmTBI. These results indicate the induction of a complex cellular senescence and glial reaction in the immature mouse brain, with clear differences between an invasive brain injury and a repetitive mild injury.


Asunto(s)
Conmoción Encefálica , Lesiones Traumáticas del Encéfalo , Lesiones Encefálicas , Ratones , Animales , Conmoción Encefálica/complicaciones , Enfermedades Neuroinflamatorias , Lesiones Traumáticas del Encéfalo/complicaciones , Senescencia Celular , Ratones Endogámicos C57BL , Modelos Animales de Enfermedad
2.
J Trauma Acute Care Surg ; 95(4): 481-489, 2023 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-37249511

RESUMEN

BACKGROUND: Cardiac dysfunction (CD) has emerged as a key contributor to delayed organ failure and late mortality in patients surviving the initial traumatic hemorrhagic response. Inflammatory processes are implicated in the initial stages of this CD; however, downstream pathways leading to a characteristic rapid fall in stroke volume and cardiac output are not yet fully defined. Currently, no cardioprotective treatments are available. We investigated the role of myocardial oxidative stress in the pathogenesis of CD associated to traumatic hemorrhagic injury, and its related metabolomic profile. METHODS: Ex vivo tissue from a 3-hour murine model of pressure-controlled trauma hemorrhagic shock (THS) was analyzed. Animals were randomized to echocardiography-guided crystalloid fluid resuscitation or a control group (sham: cannulation and anesthesia only, or naïve: no intervention). Trauma hemorrhagic shock and naïve samples were assessed by immunohistochemistry for nuclear 8-hydroxy-2'-deoxyguanosine expression as a marker of oxidative stress. Metabolomic analysis of THS and sham group tissue was performed by LC-MS. RESULTS: 8-Hydroxy-2'-deoxyguanosine expression across the myocardium was significantly higher following THS injury compared to naïve group (33.01 ± 14.40% vs. 15.08 ± 3.96%, p < 0.05). Trauma hemorrhagic shock injury significantly increased lysine ( p = 0.022), and decreased aconitate ( p = 0.016) and glutamate ( p = 0.047) in the myocardium, indicating activation of a catabolic metabolism and oxidative stress response. CONCLUSION: We confirm the acute development of oxidative stress lesions and altered cardiac energy metabolism following traumatic hemorrhage injury, providing insight into the relationship between inflammatory damage and impaired cardiac contractility. These findings may provide targets for development of novel cardioprotective therapeutics aiming to decrease late mortality from trauma.


Asunto(s)
Lesiones por Aplastamiento , Choque Hemorrágico , Animales , Humanos , Ratones , 8-Hidroxi-2'-Desoxicoguanosina , Corazón , Hemorragia/etiología , Hemorragia/terapia , Miocardio , Choque Hemorrágico/terapia
3.
Lab Anim ; 57(1): 87-88, 2023 02.
Artículo en Inglés | MEDLINE | ID: mdl-36825390
6.
Lab Anim ; 57(2): 170-181, 2023 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-36204973

RESUMEN

Nurturing a culture of care remains a key strategy and needs to be well integrated in the education programmes for laboratory animal professionals. Addressing attitudes is a complex task that must ensure reflective learning approaches. Teaching strategies must facilitate a safe space to talk openly about emotions and caring responsibilities. We reflect on two training initiatives focusing on culture of care. Firstly, the 'Care-full Stories' tool, which uses fictionalised prompts (storytelling) to encourage participants to share their own stories from working in animal research. Feedback on its impact on establishing a safe space for sharing experiences and the importance of appreciating diverse perspectives between staff is discussed. Secondly, we provide feedback on the development of training approaches on animal research integrity and culture of care with low- middle-income international communities. Strategic targets addressing the multicultural diversity of the communities, recognising their specific needs and their access to resources, must be well defined. It is important to acknowledge the interconnection between people, animals and their shared natural environment when defining the culture of care concept and addressing the teaching approaches. We discuss both the positive outcomes and challenges of these two learning experiences to support innovation when planning tools for teaching culture of care. Accounting for 'how' and 'where' the training will be delivered remains key to its successful uptake and local sustainability. Supporting improved educational tools to ascertain why caring has an impact on our professional lives will have a direct impact on the wellbeing of laboratory animal professionals worldwide.

7.
Front Mol Neurosci ; 15: 991112, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36267701

RESUMEN

C-type natriuretic peptide (CNP) is highly expressed in the central nervous system (CNS) and key to neuronal development; however, a broader role for CNP in the CNS remains unclear. To address this deficit, we investigated behavioral, sensory and motor abnormalities and blood-brain barrier (BBB) integrity in a unique mouse model with inducible, global deletion of CNP (gbCNP-/-). gbCNP-/- mice and wild-type littermates at 12 (young adult) and 65 (aged) weeks of age were investigated for changes in gait and motor coordination (CatWalk™ and rotarod tests), anxiety-like behavior (open field and elevated zero maze tests), and motor and sensory function (modified neurological severity score [mNSS] and primary SHIRPA screen). Vascular permeability was assessed in vivo (Miles assay) with complementary in vitro studies conducted in primary murine brain endothelial cells. Young adult gbCNP-/- mice had normal gait but reduced motor coordination, increased locomotor activity in the open field and elevated zero maze, and had a higher mNSS score. Aged gbCNP-/- animals developed recurrent spontaneous seizures and had impaired gait and wide-ranging motor and sensory dysfunction. Young adult and aged gbCNP-/- mice exhibited increased BBB permeability, which was partially restored in vitro by CNP administration. Cultured brain endothelial cells from gbCNP-/- mice had an abnormal ZO-1 protein distribution. These data suggest that lack of CNP in the CNS impairs tight junction protein arrangement and increases BBB permeability, which is associated with changes in locomotor activity, motor coordination and late-onset seizures.

9.
PLoS One ; 17(6): e0268962, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35704595

RESUMEN

The early detection of traumatic brain injuries can directly impact the prognosis and survival of patients. Preceding attempts to automate the detection and the assessment of the severity of traumatic brain injury continue to be based on clinical diagnostic methods, with limited tools for disease outcomes in large populations. Despite advances in machine and deep learning tools, current approaches still use simple trends of statistical analysis which lack generality. The effectiveness of deep learning to extract information from large subsets of data can be further emphasised through the use of more elaborate architectures. We therefore explore the use of a multiple input, convolutional neural network and long short-term memory (LSTM) integrated architecture in the context of traumatic injury detection through predicting the presence of brain injury in a murine preclinical model dataset. We investigated the effectiveness and validity of traumatic brain injury detection in the proposed model against various other machine learning algorithms such as the support vector machine, the random forest classifier and the feedforward neural network. Our dataset was acquired using a home cage automated (HCA) system to assess the individual behaviour of mice with traumatic brain injury or non-central nervous system (non-CNS) injured controls, whilst housed in their cages. Their distance travelled, body temperature, separation from other mice and movement were recorded every 15 minutes, for 72 hours weekly, for 5 weeks following intervention. The HCA behavioural data was used to train a deep learning model, which then predicts if the animals were subjected to a brain injury or just a sham intervention without brain damage. We also explored and evaluated different ways to handle the class imbalance present in the uninjured class of our training data. We then evaluated our models with leave-one-out cross validation. Our proposed deep learning model achieved the best performance and showed promise in its capability to detect the presence of brain trauma in mice.


Asunto(s)
Lesiones Traumáticas del Encéfalo , Aprendizaje Profundo , Algoritmos , Animales , Lesiones Traumáticas del Encéfalo/diagnóstico , Humanos , Aprendizaje Automático , Ratones , Redes Neurales de la Computación
12.
Lab Anim ; 55(2): 189, 2021 04.
Artículo en Inglés | MEDLINE | ID: mdl-33847179
13.
14.
Theranostics ; 11(1): 346-360, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33391479

RESUMEN

Rationale: Traumatic brain injury (TBI) leads to neurological impairment, with no satisfactory treatments available. Classical ketogenic diets (KD), which reduce reliance on carbohydrates and provide ketones as fuel, have neuroprotective potential, but their high fat content reduces compliance, and experimental evidence suggests they protect juvenile brain against TBI, but not adult brain, which would strongly limit their applicability in TBI. Methods: We designed a new-KD with a fat to carbohydrate plus protein ratio of 2:1, containing medium chain triglycerides (MCT), docosahexaenoic acid (DHA), low glycaemic index carbohydrates, fibres and the ketogenic amino acid leucine, and evaluated its neuroprotective potential in adult TBI. Adult male C57BL6 mice were injured by controlled cortical impact (CCI) and assessed for 70 days, during which they received a control diet or the new-KD. Results: The new-KD, that markedly increased plasma Beta-hydroxybutyrate (ß-HB), significantly attenuated sensorimotor deficits and corrected spatial memory deficit. The lesion size, perilesional inflammation and oxidation were markedly reduced. Oligodendrocyte loss appeared to be significantly reduced. TBI activated the mTOR pathway and the new-KD enhanced this increase and increased histone acetylation and methylation. Conclusion: The behavioural improvement and tissue protection provide proof of principle that this new formulation has therapeutic potential in adult TBI.


Asunto(s)
Lesiones Traumáticas del Encéfalo/dietoterapia , Encéfalo/patología , Dieta Cetogénica/métodos , Memoria Espacial , Ácido 3-Hidroxibutírico/sangre , Acetilación , Animales , Ataxia/fisiopatología , Encéfalo/metabolismo , Lesiones Traumáticas del Encéfalo/metabolismo , Lesiones Traumáticas del Encéfalo/patología , Lesiones Traumáticas del Encéfalo/fisiopatología , Carbohidratos de la Dieta , Grasas de la Dieta , Fibras de la Dieta , Proteínas en la Dieta , Modelos Animales de Enfermedad , Ácidos Docosahexaenoicos , Epigénesis Genética , Índice Glucémico , Código de Histonas , Inflamación/metabolismo , Inflamación/patología , Cojera Animal/fisiopatología , Leucina , Masculino , Metilación , Ratones , Prueba del Laberinto Acuático de Morris , Oligodendroglía/patología , Paresia/fisiopatología , Equilibrio Postural , Prueba de Desempeño de Rotación con Aceleración Constante , Trastornos de la Sensación/fisiopatología , Transducción de Señal , Serina-Treonina Quinasas TOR , Triglicéridos
16.
Clin Sci (Lond) ; 134(13): 1715-1734, 2020 07 17.
Artículo en Inglés | MEDLINE | ID: mdl-32648582

RESUMEN

Sepsis is a major worldwide healthcare issue with unmet clinical need. Despite extensive animal research in this area, successful clinical translation has been largely unsuccessful. We propose one reason for this is that, sometimes, the experimental question is misdirected or unrealistic expectations are being made of the animal model. As sepsis models can lead to a rapid and substantial suffering - it is essential that we continually review experimental approaches and undertake a full harm:benefit impact assessment for each study. In some instances, this may require refinement of existing sepsis models. In other cases, it may be replacement to a different experimental system altogether, answering a mechanistic question whilst aligning with the principles of reduction, refinement and replacement (3Rs). We discuss making better use of patient data to identify potentially useful therapeutic targets which can subsequently be validated in preclinical systems. This may be achieved through greater use of construct validity models, from which mechanistic conclusions are drawn. We argue that such models could provide equally useful scientific data as face validity models, but with an improved 3Rs impact. Indeed, construct validity models may not require sepsis to be modelled, per se. We propose that approaches that could support and refine clinical translation of research findings, whilst reducing the overall welfare burden on research animals.


Asunto(s)
Modelos Animales de Enfermedad , Sepsis/patología , Investigación Biomédica Traslacional , Animales , Ensayos Clínicos como Asunto , Humanos , Sepsis/fisiopatología
17.
Front Immunol ; 11: 10, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32117219

RESUMEN

The targeted delivery of therapies to diseased tissues offers a safe opportunity to achieve optimal efficacy while limiting systemic exposure. These considerations apply to many disease indications but are especially relevant for rheumatoid arthritis (RA), as RA is a systemic autoimmune disease which affects multiple joints. We have identified an antibody that is specific to damaged arthritic cartilage (anti-ROS-CII) that can be used to deliver treatments specifically to arthritic joints, yielding augmented efficacy in experimental arthritis. In the current study, we demonstrate that scaffolds enriched with bioactive payloads can be delivered precisely to an inflamed joint and achieve superior efficacy outcomes consistent with the pharmacological properties of these payloads. As a scaffold, we have used extracellular vesicles (EVs) prepared from human neutrophils (PMNs), which possess intrinsic anti-inflammatory properties and the ability to penetrate inflamed arthritic cartilage. EV fortified with anti-ROS-CII (EV/anti-ROS-CII) retained anti-ROS-CII specificity and bound exclusively to the damaged cartilage. Following systemic administration, EV/anti-ROS-CII (a) exhibited the ability to localize specifically in the arthritic joint in vivo and (b) was able to specifically target single (viral IL-10 or anti-TNF) or combined (viral IL-10 and anti-TNF) anti-inflammatory treatments to the arthritic joint, which accelerated attenuation of clinical and synovial inflammation. Overall, this study demonstrates the attainability of targeting a pro-resolving biological scaffold to the arthritic joint. The potential of targeting scaffolds such as EV, nanoparticles, or a combination thereof alongside combined therapeutics is paramount for designing systemically administered broad-spectrum of anti-inflammatory treatments.


Asunto(s)
Antiinflamatorios/administración & dosificación , Anticuerpos Monoclonales/administración & dosificación , Anticuerpos Monoclonales/inmunología , Artritis Experimental/tratamiento farmacológico , Artritis Reumatoide/tratamiento farmacológico , Cartílago/inmunología , Cartílago/patología , Sistemas de Liberación de Medicamentos/métodos , Vesículas Extracelulares , Animales , Femenino , Voluntarios Sanos , Humanos , Interleucina-10/administración & dosificación , Articulación de la Rodilla/efectos de los fármacos , Leucocitos/citología , Ratones , Ratones Endogámicos C57BL , Resultado del Tratamiento , Factor de Necrosis Tumoral alfa/inmunología , Proteínas Virales/administración & dosificación
18.
J Neurotrauma ; 37(1): 66-79, 2020 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-31256709

RESUMEN

Traumatic brain injury (TBI) can lead to life-changing neurological deficits, which reflect the fast-evolving secondary injury post-trauma. There is a need for acute protective interventions, and the aim of this study was to explore in an experimental TBI model the neuroprotective potential of a single bolus of a neuroactive omega-3 fatty acid, docosahexaenoic acid (DHA), administered in a time window feasible for emergency services. Adult mice received a controlled cortical impact injury (CCI) and neurological impairment was assessed with the modified Neurological Severity Score (mNSS) up to 28 days post-injury. DHA (500 nmol/kg) or saline were injected intravenously at 30 min post-injury. The lipid mediator profile was assessed in the injured hemisphere at 3 h post-CCI. After completion of behavioral tests and lesion assessment using magnetic resonance imaging, over 7 days or 28 days post-TBI, the tissue was analyzed by immunohistochemistry. The single DHA bolus significantly reduced the injury-induced neurological deficit and increased pro-resolving mediators in the injured brain. DHA significantly reduced lesion size, the microglia and astrocytic reaction, and oxidation, and decreased the accumulation of beta-amyloid precursor protein (APP), indicating a reduced axonal injury at 7 days post-TBI. DHA reduced the neurofilament light levels in plasma at 28 days. Therefore, an acute single bolus of DHA post-TBI, in a time window relevant for acute emergency intervention, can induce a long-lasting and significant improvement in neurological outcome, and this is accompanied by a marked upregulation of neuroprotective mediators, including the DHA-derived resolvins and protectins.


Asunto(s)
Lesiones Traumáticas del Encéfalo/patología , Encéfalo/efectos de los fármacos , Ácidos Docosahexaenoicos/farmacología , Fármacos Neuroprotectores/farmacología , Recuperación de la Función/efectos de los fármacos , Animales , Encéfalo/patología , Metabolismo de los Lípidos/efectos de los fármacos , Ratones
19.
Front Immunol ; 10: 2774, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31866998

RESUMEN

Cardiac dysfunction (CD) importantly contributes to mortality in trauma patients, who survive their initial injuries following successful hemostatic resuscitation. This poor outcome has been correlated with elevated biomarkers of myocardial injury, but the pathophysiology triggering this CD remains unknown. We investigated the pathophysiology of acute CD after trauma using a mouse model of trauma hemorrhage shock (THS)-induced CD with echocardiographic guidance of fluid resuscitation, to assess the THS impact on myocardial integrity and function. Mice were subjected to trauma (soft tissue and bone fracture) and different degrees of hemorrhage severity (pressure controlled ~MABP < 35 mmHg or <65 mmHg) for 1 h, to characterize the acute impact on cardiac function. In a second study, mice were subjected to trauma and hemorrhage (MABP < 35 mmHg) for 1 h, then underwent two echocardiographic-guided resuscitations to baseline stroke volume at 60 and 120 min, and were monitored up to 180 min to study the longer impact of THS following resuscitation. Naïve and sham animals were used as controls. At 60 min post-THS injury, animals showed a lower cardiac output (CO) and stroke volume (SV) and an early rise of heart fatty acid-binding protein (H-FABP = 167 ± 38 ng/ml; 90% increase from shams, 3.54 ± 3.06 ng/ml), when subjected to severe hemorrhage and injury. Despite resuscitation, these animals maintained lower CO (6 ml/min vs. 23 ml/min), lower SV (10 µl vs. 46 µl; both ~75% decreased), and higher H-FABP (levels (340 ± 115 ng/ml vs. 10.3 ± 0.2 ng/ml; all THS vs. shams, P < 0.001) at 180 min post-THS injury. Histopathological and flow-cytometry analysis of the heart confirmed an influx of circulatory leukocytes, compared to non-injured hearts. Myocardial injury was supported by an increase of troponin I and h-FABP and the widespread ultrastructural disorganization of the morphology of sarcomeres and mitochondria. DNA fragmentation and chromatin condensation driven by leakage of apoptosis-inducing factor (AIF) may suggest a mitochondria-driven progressive cell death. THS modeling in the mouse results in cardiomyocyte damage and reduced myocardial function, which mimics the cardiac dysfunction seen in trauma patients. This CD model may, therefore, provide further understanding to the mechanisms underlying CD and act as a tool for developing cardioprotective therapeutics to improve survival after injury.


Asunto(s)
Hemorragia/complicaciones , Hemorragia/etiología , Modelos Cardiovasculares , Disfunción Ventricular/etiología , Disfunción Ventricular/fisiopatología , Heridas y Lesiones/complicaciones , Animales , Biomarcadores , Modelos Animales de Enfermedad , Corazón/fisiopatología , Hemodinámica , Humanos , Masculino , Ratones , Miocardio/metabolismo , Choque Hemorrágico/etiología
20.
J Neurosci Methods ; 321: 49-63, 2019 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-30991030

RESUMEN

BACKGROUND: Neurotrauma patients face major neurological sequelae. The failure in the preclinical-to-clinical translation of candidate therapies could be due to poor evaluation of rodent behaviours after neurotrauma. NEW METHOD: A home cage automated system was used to study the long term behaviour of individual rats with traumatic brain injury (TBI), spinal cord injury (SCI) and non-CNS injured controls, whilst group-housed in their home cages. Naïve rats were used as baseline controls. Automated locomotor activity and body temperature recordings were carried out 24 h /day for 3 days/week during 12 weeks post-injury. Behavioural patterns, including aggression, rearing, grooming, feeding and drinking were analysed from automated video recordings during week 1, 6 and 12. RESULTS: SCI animals showed a lower locomotor activity compared to TBI or control animals during light and dark phases. TBI animals showed a higher aggression during the dark phase in the first week post-injury compared to SCI or control animals. Individual grooming and rearing were reduced in SCI animals compared to TBI and control animals in the first week post-injury during the dark phase. No differences in drinking or feeding were detected between groups. Locomotor activity did not differ between naïve male and female rats, but body temperature differ between light and dark phases for both. STANDARD METHODS: Injury severity was compared to standard SCI and TBI behaviour scores (BBB and mNSS, respectively) and histological analysis. CONCLUSIONS: This study demonstrates the practical benefits of using a non-intrusive automated home cage recording system to observe long term individual behaviour of group-housed SCI and TBI rats.


Asunto(s)
Conducta Animal , Lesiones Traumáticas del Encéfalo/diagnóstico , Lesiones Traumáticas del Encéfalo/psicología , Traumatismos de la Médula Espinal/diagnóstico , Traumatismos de la Médula Espinal/psicología , Grabación en Video/métodos , Animales , Temperatura Corporal , Modelos Animales de Enfermedad , Femenino , Aseo Animal , Locomoción , Masculino , Reconocimiento de Normas Patrones Automatizadas , Ratas Sprague-Dawley
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...