Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 63
Filtrar
1.
J Intern Med ; 2024 Jul 07.
Artículo en Inglés | MEDLINE | ID: mdl-38973251

RESUMEN

BACKGROUND: Spinocerebellar ataxia 4 (SCA4), characterized in 1996, features adult-onset ataxia, polyneuropathy, and linkage to chromosome 16q22.1; its underlying mutation has remained elusive. OBJECTIVE: To explore the radiological and neuropathological abnormalities in the entire neuroaxis in SCA4 and search for its mutation. METHODS: Three Swedish families with undiagnosed ataxia went through clinical, neurophysiological, and neuroimaging tests, including PET studies and genetic investigations. In four cases, neuropathological assessments of the neuroaxis were performed. Genetic testing included short read whole genome sequencing, short tandem repeat analysis with ExpansionHunter de novo, and long read sequencing. RESULTS: Novel features for SCA4 include dysautonomia, motor neuron affection, and abnormal eye movements. We found evidence of anticipation; neuroimaging demonstrated atrophy in the cerebellum, brainstem, and spinal cord. [18F]FDG-PET demonstrated brain hypometabolism and [11C]Flumazenil-PET reduced binding in several brain lobes, insula, thalamus, hypothalamus, and cerebellum. Moderate to severe loss of Purkinje cells in the cerebellum and of motor neurons in the anterior horns of the spinal cord along with pronounced degeneration of posterior tracts was also found. Intranuclear, mainly neuronal, inclusions positive for p62 and ubiquitin were sparse but widespread in the CNS. This finding prompted assessment for nucleotide expansions. A polyglycine stretch encoding GGC expansions in the last exon of the zink finger homeobox 3 gene was identified segregating with disease and not found in 1000 controls. CONCLUSIONS: SCA4 is a neurodegenerative disease caused by a novel GGC expansion in the coding region of ZFHX3, and its spectrum is expanded to include dysautonomia and neuromuscular manifestations.

2.
Artículo en Inglés | MEDLINE | ID: mdl-38780782

RESUMEN

PURPOSE: Swine are one of the major animal species used in translational research, with unique advantages given the similar anatomic and physiologic characteristics as man, but the investigator needs to be familiar with important differences. This article targets clinical anesthesiologists who are proficient in human monitoring. We summarize our experience during the last two decades, with the aim to facilitate for clinical and non-clinical researchers to improve in porcine research. METHODS: This was a retrospective review of 337 swine with a mean (SD) weight 60 (4.2) kg at the Experimental Traumatology laboratory at Södersjukhuset (Stockholm south general hospital) between 2003 and 2023, including laboratory parameters and six CT-angiography examinations. RESULTS: Swine may be ventilated through the snout using a size 2 neonatal mask. Intubate using a 35 cm miller laryngoscope and an intubating introducer. Swine are prone to alveolar atelectasis and often require alveolar recruitment. Insert PA-catheters through a cut-down technique in the internal jugular vein, and catheters in arteries and veins using combined cut-down and Seldinger techniques. Cardiopulmonary resuscitation is possible and lateral chest compressions are most effective. Swine are prone to lethal ventricular arrhythmias, which may be reversed by defibrillation. Most vital parameters are similar to man, with the exception of a higher core temperature, higher buffer bases and increased coagulation. Anesthesia methods are similar to man, but swine require five times the dose of ketamine. CONCLUSION: Swine share anatomical and physiological features with man, which allows for seamless utilization of clinical monitoring equipment, medication, and physiological considerations.

3.
J Brachial Plex Peripher Nerve Inj ; 19(1): e6-e12, 2024 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-38263956

RESUMEN

Brachial plexus birth injuries cause diminished motor function in the upper extremity. The most common sequel is internal rotation contracture. A number of these patients also suffer from cocontractions, preventing the use of an otherwise good passive range of motion in the shoulder. One theory behind the co-contracture problem is that injured nerve fibers grow into distal support tissue not corresponding to the proximal support tissue, resulting in reinnervation of the wrong muscle groups. To further elucidate this hypothesis, we used rat neonates to investigate a possible model for the study of cocontractions in brachial plexus birth injuries. Five-day-old rats were subjected to a crush injury to the C5-C6 roots. After a healing period of 4 weeks, the infraspinatus muscle was injected with Fluoro-Gold. A week later, the animals were perfused and spinal cords harvested and sectioned. Differences in the uptake of Fluoro-Gold and NeuN positive cells of between sides of the spinal cord were recorded. We found a larger amount of Fluoro-Gold positive cells on the uninjured side, while the injured side had positive cells dispersed over a longer area in the craniocaudal direction. Our findings indicate that the method can be used to trace Fluoro-Gold from muscle through a neuroma. Our results also indicate that a neuroma in continuity somewhat prevents the correct connection from being established between the motor neuron pool in the spinal cord and target muscle and that some neurons succumb to a crushing injury. We also present future research ideas.

4.
Artículo en Inglés | MEDLINE | ID: mdl-38059837

RESUMEN

OBJECTIVE: The majority of traumatic brain injuries (TBIs) are classified as mild and occur in young individuals. The course of recovery varies but can result in chronic or troubling outcomes. The impact of age on TBI outcomes in young adults before complete brain maturation is not well studied. METHODS: In this study, we compared the effects of mild TBI on cognitive performance and self-reported TBI symptoms and posttraumatic stress disorder (PTSD) in 903 soldiers in 3 different age groups: 24 years or younger, 25 to 27 years, and 28 to 40 years. The soldiers had returned from war zones in Iraq and were screened for TBI within a few days of return. Cognitive performance was measured with the Automated Neuropsychological Assessment Metrics of Military TBI Version 4 (ANAM4). Symptoms associated with mild TBI were self-reported on the Neurobehavioral Symptom Inventory, and the PTSD Checklist-Civilian Version (PCL-C). RESULTS: Soldiers with TBI in every age group had significantly higher prevalence of most symptoms than those with no TBI. Soldiers with TBI also reported more chronic pain sites, regardless of age. Soldiers aged 28 to 40 years with TBI had the lowest cognitive performance scores (ANAM) across several subtests, both unadjusted and adjusted. The Global Deficit Score was significantly higher for soldiers aged 28 to 40 years and 25 to 27 years with TBI than for soldiers younger than 24 years with no TBI. After adjusting for PTSD symptoms, education, and number of lifetime TBIs, the overall test battery mean for soldiers aged 28 to 40 years with TBI was significantly lower than for soldiers younger than 24 years with no TBI. CONCLUSION: Soldiers with mild TBI in the younger age group show more symptoms associated to frontal lobe function while soldiers in the older group suffer more cognitive impairment. This may warrant further study as it may indicate a propensity to later cognitive decline among soldiers who were older at the time of injury.

5.
Ann Biomed Eng ; 51(12): 2762-2771, 2023 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-37532895

RESUMEN

Behind armor blunt trauma (BABT) is a non-penetrating injury caused by the rapid deformation of body armor, by a projectile, which may in extreme circumstances cause death. The understanding of the mechanisms is still low, in relation to what is needed for safety threshold levels. Few models of graded kinetic energy transfer to the body exist. We established an experimental model for graded BABT. The cold gas cannon was air-driven, consisted of a pressure vessel, a barrel, and a pressure actuator. It required short training to operate and was constructed by standard components. It produced standardized expulsion of plastic projectiles with 65 mm and weight 58 g. Velocity correlated linearly to pressure (R 0.9602, p < 0.0001), equation Y = 6.558*X + 46.50. Maximum tested pressure was 10 bar, velocity 110 m/s and kinetic energy (Ek) 351 J. Crossbred male swine (n = 10) mean weight (SD) 56 ± 3 kg, were subjected to BABT, mean Ek (SD) 318 (61) J, to a fix point on the right lateral thorax. Pulmonary contusion was confirmed by physiological parameters pO2 (p < 0.05), SaO2 (p < 0.01), pCO2 (p < 0.01), etCO2 (p < 0.01), MPAP (p < 0.01), Cstat (p < 0.01), intrapulmonary shunt (Q's/Q't) (p < 0.05), and qualified trans-thoracic ultrasound (p < 0.0001). The consistent injury profile enabled for the addition of future experimental interventions.


Asunto(s)
Contusiones , Traumatismos Torácicos , Heridas no Penetrantes , Masculino , Porcinos , Animales , Balística Forense , Traumatismos Torácicos/diagnóstico por imagen , Ropa de Protección/efectos adversos , Heridas no Penetrantes/diagnóstico por imagen , Pulmón
6.
Neurotrauma Rep ; 4(1): 107-117, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-36895820

RESUMEN

Monitoring protein biomarker levels in the cerebrospinal fluid (CSF) can help assess injury severity and outcome after traumatic brain injury (TBI). Determining injury-induced changes in the proteome of brain extracellular fluid (bECF) can more closely reflect changes in the brain parenchyma, but bECF is not routinely available. The aim of this pilot study was to compare time-dependent changes of S100 calcium-binding protein B (S100B), neuron-specific enolase (NSE), total Tau, and phosphorylated Tau (p-Tau) levels in matching CSF and bECF samples collected at 1, 3, and 5 days post-injury from severe TBI patients (n = 7; GCS 3-8) using microcapillary-based western analysis. We found that time-dependent changes in CSF and bECF levels were most pronounced for S100B and NSE, but there was substantial patient-to-patient variability. Importantly, the temporal pattern of biomarker changes in CSF and bECF samples showed similar trends. We also detected two different immunoreactive forms of S100B in both CSF and bECF samples, but the contribution of the different immunoreactive forms to total immunoreactivity varied from patient to patient and time point to time point. Our study is limited, but it illustrates the value of both quantitative and qualitative analysis of protein biomarkers and the importance of serial sampling for biofluid analysis after severe TBI.

7.
Front Neurol ; 14: 1094723, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-36761348
8.
Front Psychol ; 13: 795759, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35967608

RESUMEN

Conduct after capture (CAC) training is for personnel at risk of being captured. To be effective, it needs to be stressful. But how do we know if it is stressful enough? This study uses biomarkers and cognitive measures to evaluate CAC. Soldiers undergoing CAC were measured by the stress hormone cortisol from saliva samples at baseline and during training. The training consisted of being taken capture and put through a number of realistic and threatening scenarios, targeting survival strategies taught in the preceding week. Between scenarios, the trainees were held in a holding cell where they were monitored by a guard. The saliva samples were taken in conjunction with the scenarios. The whole training took place over a period of ~24 h. Cognitive performance was measured at baseline and after training. Three groups took part Group A (n = 20) was taken after 48 h of intense tasks leaving them in a poor resting state. Group B (n = 23) was well rested at CAC onset. Group C (n = 10) was part of a survival, evasion, resistance, and escape (SERE) instructor course. The CAC training was the same for all groups. Group A exhibited a high increase in cortisol during CAC, compared to baseline levels were multiple times as high as "expected" values. Group B exhibited elevated levels slightly lower than those of group A, they also "dropped" to "normal" levels during the latter part of the exercise. Group C displayed the least increase with only slightly elevated levels. CAC training is stressful and cortisol levels were elevated enough to satisfy the prerequisite for effective stress inoculation. No cognitive performance drop could be identified; however, several participants "froze" during the exercise due to intensive stress.

9.
Int J Mol Sci ; 23(2)2022 Jan 10.
Artículo en Inglés | MEDLINE | ID: mdl-35054909

RESUMEN

Hippocampal dysfunction contributes to multiple traumatic brain injury sequala. Female rodents' outcome is superior to male which has been ascribed the neuroprotective sex hormones 17ß-estradiol and progesterone. Cytochrome P450 1B1 (CYP1B1) is an oxidative enzyme influencing the neuroinflammatory response by creating inflammatory mediators and metabolizing neuroprotective 17ß-estradiol and progesterone. In this study, we aimed to describe hippocampal CYP1B1 mRNA expression, protein presence of CYP1B1 and its key redox partner Cytochrome P450 reductase (CPR) in both sexes, as well as the effect of penetrating traumatic brain injury (pTBI). A total 64 adult Sprague Dawley rats divided by sex received pTBI or sham-surgery and were assigned survival times of 1-, 3-, 5- or 7 days. CYP1B1 mRNA was quantified using in-situ hybridization and immunohistochemistry performed to verify protein colocalization. CYP1B1 mRNA expression was present in all subregions but greatest in CA2 irrespective of sex, survival time or intervention. At 3-, 5- and 7 days post-injury, expression in CA2 was reduced in male rats subjected to pTBI compared to sham-surgery. Females subjected to pTBI instead exhibited increased expression in all CA subregions 3 days post-injury, the only time point expression in CA2 was greater in females than in males. Immunohistochemical analysis confirmed neuronal CYP1B1 protein in all hippocampal subregions, while CPR was limited to CA1 and CA2. CYP1B1 mRNA is constitutively expressed in both sexes. In response to pTBI, females displayed a more urgent but brief regulatory response than males. This indicates there may be sex-dependent differences in CYP1B1 activity, possibly influencing inflammation and neuroprotection in pTBI.


Asunto(s)
Lesiones Traumáticas del Encéfalo/etiología , Lesiones Traumáticas del Encéfalo/metabolismo , Citocromo P-450 CYP1B1/genética , Expresión Génica , Hipocampo/metabolismo , Animales , Biomarcadores , Lesiones Traumáticas del Encéfalo/patología , Citocromo P-450 CYP1B1/metabolismo , Modelos Animales de Enfermedad , Ciclo Estral , Femenino , Hipocampo/patología , Inmunohistoquímica , Hibridación in Situ , Masculino , ARN Mensajero/genética , Ratas , Factores Sexuales
10.
Mil Med ; 2022 Jan 03.
Artículo en Inglés | MEDLINE | ID: mdl-34977944

RESUMEN

INTRODUCTION: Mental health issues from intense or prolonged stress are a common concern in regard to military deployment. Deployments can objectively vary in stress exposure, but it is the individuals' perception of that stress that affects sustainability, mental health, and combat fitness, which calls for the need of a protocol to evaluate and maintain a current estimation of stress impact. So, how can we assess the impact of stressors during different phases of deployment? MATERIALS AND METHODS: We used three psychological self-rating forms, the PSS14-Perceived Stress Scale, SMBM-Shirom Melamed Burnout Measure, and KSQ-Karolinska Sleep Questionnaire, to measure the impact of stress before (T1), during (T2), and at homecoming (T3). We also wanted to see if T1 or T2 results could predict T3 results to be able to better prepare the homecoming program.The forms were handed out to Swedish soldiers deployed in Mali in 2017. The forms were collected as a way to assess the status of the mental health load at three timepoints based on the personnel function as a way to assess the current "psychological fitness level". RESULTS: The results show that stress measured using PSS14 was high at homecoming. The same result was observed for SMBM. No measures from T1 or T2 could however predict the T3 results. CONCLUSIONS: Taken together, we found that screening of all contingent staff is relatively easy and provides personnel with relevant data on mental health and stress at the current time. We also found that test results correlated between T1 and T2 but not with T3. This indicates that there might be different stressors that affect staff at different timepoints.

11.
Ann Biomed Eng ; 50(1): 78-85, 2022 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-34907465

RESUMEN

Underwater blast differs from blast in air. The increased density and viscosity of water relative to air cause injuries to occur almost exclusively as primary blast, and may cause disorientation in a diver, which may lead to inability to protect the airway and cause drowning. However, cognitive impairments from under water blast wave exposure have not been properly investigated, and no experimental model has been described. We established an experimental model (water shock tube) for simulating the effects of underwater blast pressure waves in rodents, and to investigate neurology in relation to organ injury. The model produced standardized pressure waves (duration of the primary peak 3.5 ms, duration of the entire complex waveform including all subsequent reflections 325 ms, mean impulse 141-281 kPa-ms, mean peak pressure 91-194 kPa). 31 rats were randomized to control (n = 6), exposure 90 kPa (n = 8), 152 kPa (n = 8), and 194 kPa (n = 9). There was a linear trend between the drop height of the water shock tube and electroencephalography (EEG) changes (p = 0.014), while no differences in oxygen saturation, heart rate, S100b or macroscopic bleedings were detected. Microscopic bleedings were detected in lung, intestines, and meninges. Underwater pressure waves caused changes in EEG, at pressures when mild hemorrhage occurred in organs, suggesting an impact on brain functions. The consistent injury profile enabled for the addition of future experimental interventions.


Asunto(s)
Traumatismos por Explosión , Animales , Sistema Nervioso Central , Estudios de Factibilidad , Modelos Teóricos , Ratas , Roedores
12.
Front Cell Neurosci ; 15: 658662, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34140880

RESUMEN

Noise exposure is the most important external factor causing acquired hearing loss in humans, and it is strongly associated with the production of reactive oxygen species (ROS) in the cochlea. Several studies reported that the administration of various compounds with antioxidant effects can treat oxidative stress-induced hearing loss. However, traditional systemic drug administration to the human inner ear is problematic and has not been successful in a clinical setting. Thus, there is an urgent need to develop rescue treatment for patients with acute acoustic injuries. Hydrogen gas has antioxidant effects, rapid distribution, and distributes systemically after inhalation.The purpose of this study was to determine the protective efficacy of a single dose of molecular hydrogen (H2) on cochlear structures. Guinea pigs were divided into six groups and sacrificed immediately after or at 1 or 2 weeks. The animals were exposed to broadband noise for 2 h directly followed by 1-h inhalation of 2% H2 or room air. Electrophysiological hearing thresholds using frequency-specific auditory brainstem response (ABR) were measured prior to noise exposure and before sacrifice. ABR thresholds were significantly lower in H2-treated animals at 2 weeks after exposure, with significant preservation of outer hair cells in the entire cochlea. Quantification of synaptophysin immunoreactivity revealed that H2 inhalation protected the cochlear inner hair cell synaptic structures containing synaptophysin. The inflammatory response was greater in the stria vascularis, showing increased Iba1 due to H2 inhalation.Repeated administration of H2 inhalation may further improve the therapeutic effect. This animal model does not reproduce conditions in humans, highlighting the need for additional real-life studies in humans.

13.
Front Neurol ; 11: 540144, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33178100

RESUMEN

Most blast-induced traumatic brain injuries (bTBI) are mild in severity and culpable for the lingering and persistent neuropsychological complaints in affected individuals. There is evidence that the prevalence of symptoms post-exposure may be sex-specific. Our laboratory has focused on changes in the monoamine and the neuropeptide, galanin, systems in male rodents following primary bTBI. In this study, we aimed to replicate these findings in female rodents. Brainstem sections from the locus coeruleus (LC) and dorsal raphe nuclei (DRN) were processed for in situ hybridisation at 1 and 7 days post-bTBI. We investigated changes in the transcripts for tyrosine hydroxylase (TH), tryptophan hydroxylase two (TPH2) and galanin. Like in males, we found a transient increase in TH transcript levels bilaterally in the female LC. Changes in TPH2 mRNA were more pronounced and extensive in the DRN of females compared to males. Galanin mRNA was increased bilaterally in the LC and DRN, although this increase was not apparent until day 7 in the LC. Serum analysis revealed an increase in corticosterone, but only in exposed females. These changes occurred without any visible signs of white matter injury, cell death, or blood-brain barrier breakdown. Taken together, in the apparent absence of visible structural damage to the brain, the monoamine and galanin systems, two key players in emotional regulation, are activated deferentially in males and females following primary blast exposure. These similarities and differences should be considered when developing and evaluating diagnostic and therapeutic interventions for bTBI.

14.
Front Neurol ; 11: 836, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32982907

RESUMEN

Mild traumatic brain injury (mTBI, also known as a concussion) as a consequence of battlefield blast exposure or blunt force trauma has been of increasing concern to militaries during recent conflicts. This concern is due to the frequency of exposure to improvised explosive devices for forces engaged in operations both in Iraq and Afghanistan coupled with the recognition that mTBI may go unreported or undetected. Blasts can lead to mTBI through a variety of mechanisms. Debate continues as to whether exposure to a primary blast wave alone is sufficient to create brain injury in humans, and if so, exactly how this occurs with an intact skull. Resources dedicated to research in this area have also varied substantially among contributing NATO countries. Most of the research has been conducted in the US, focused on addressing uncertainties in management practices. Development of objective diagnostic tests should be a top priority to facilitate both diagnosis and prognosis, thereby improving management. It is expected that blast exposure and blunt force trauma to the head will continue to be a potential source of injury during future conflicts. An improved understanding of the effects of blast exposure will better enable military medical providers to manage mTBI cases and develop optimal protective measures. Without the immediate pressures that come with a high operational tempo, the time is right to look back at lessons learned, make full use of available data, and modify mitigation strategies with both available evidence and new evidence as it comes to light. Toward that end, leveraging our cooperation with the civilian medical community is critical because the military experience over the past 10 years has led to a renewed interest in many similar issues pertaining to mTBI in the civilian world. Such cross-fertilization of knowledge will undoubtedly benefit all. This paper highlights similarities and differences in approach to mTBI patient care in NATO and partner countries and provides a summary of and lessons learned from a NATO lecture series on the topic of mTBI, demonstrating utility of having patients present their experiences to a medical audience, linking practical clinical care to policy approaches.

15.
J Neurotrauma ; 37(12): 1381-1391, 2020 06 15.
Artículo en Inglés | MEDLINE | ID: mdl-32013731

RESUMEN

Brain protein biomarker clearance to blood in traumatic brain injury (TBI) is not fully understood. The aim of this study was to analyze the effect that a disrupted blood-brain barrier (BBB) had on biomarker clearance. Seventeen severe TBI patients admitted to Karolinska University Hospital were prospectively included. Cerebrospinal fluid (CSF) and blood concentrations of S100 calcium binding protein B (S100B) and neuron-specific enolase (NSE) were analyzed every 6-12 h for ∼1 week. Blood and CSF albumin were analyzed every 12-24 h, and BBB integrity was assessed using the CSF:blood albumin quotient (QA). We found that time-dependent changes in the CSF and blood levels of the two biomarkers were similar, but that the correlation between the biomarkers and QA was lower for NSE (ρ = 0.444) than for S100B (ρ = 0.668). Because data were longitudinal, we also conducted cross correlation analyses, which indicated a directional flow and lag-time of biomarkers from CSF to blood. For S100B, this lag-time could be ascribed to BBB integrity, whereas for NSE it could not. Upon inferential modelling, using generalized least square estimation (S100B) or linear mixed models (NSE), QA (p = 0.045), time from trauma (p < 0.001), time from trauma2 (p = 0.023), and CSF biomarker levels (p = 0.008) were independent predictors of S100B in blood. In contrast, for NSE, only time from trauma was significant (p < 0.001). These findings are novel and important, but must be carefully interpreted because of different characteristics between the two proteins. Nonetheless, we present the first data that indicate that S100B and NSE are cleared differently from the central nervous system, and that both the disrupted BBB and additional alternative pathways, such as the recently described glymphatic system, may play a role. This is of importance both for clinicians aiming to utilize these biomarkers and for the pathophysiological understanding of brain protein clearance, but warrants further examination.


Asunto(s)
Barrera Hematoencefálica/metabolismo , Lesiones Traumáticas del Encéfalo/metabolismo , Sistema Glinfático/metabolismo , Fosfopiruvato Hidratasa/metabolismo , Subunidad beta de la Proteína de Unión al Calcio S100/metabolismo , Índice de Severidad de la Enfermedad , Adulto , Biomarcadores/sangre , Biomarcadores/líquido cefalorraquídeo , Biomarcadores/metabolismo , Barrera Hematoencefálica/patología , Lesiones Traumáticas del Encéfalo/diagnóstico , Femenino , Humanos , Estudios Longitudinales , Masculino , Persona de Mediana Edad , Estudios Prospectivos , Adulto Joven
16.
Front Neurol ; 10: 811, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31417487

RESUMEN

Traumatic brain injury (TBI) is followed by a secondary inflammation in the brain. The inflammatory response includes prostanoid synthesis by the inducible enzyme cyclooxygenase-2 (COX-2). Inhibition of COX-2 is associated with improved functional outcome in experimental TBI models, although central nervous system-specific effects are not fully understood. Animal studies report better outcomes in females than males. The exact mechanisms for this gender dichotomy remain unknown. In an initial study we reported increased COX-2 expression in male rats, compared to female, following experimental TBI. It is possible that COX-2 induction is directly associated with increased cell death after TBI. Therefore, we designed a sequential study to investigate the blocking of COX-2 specifically, using the established COX-2 inhibitor diclofenac. Male Sprague-Dawley rats weighing between 250 and 350 g were exposed to focal penetrating TBI and randomly selected for diclofenac treatment (5 µg intralesionally, immediately following TBI) (n = 8), controls (n = 8), sham operation (n = 8), and normal (no manipulation) (n = 4). After 24 h, brains were removed, fresh frozen, cut into 14 µm coronal sections and subjected to COX-2 immunofluorescence, Fluoro Jade, TUNEL, and lesion area analyses. Diclofenac treatment decreased TUNEL staining indicative of apoptosis with a mean change of 54% (p < 0.05) and lesion area with a mean change of 55% (p < 0.005). Neuronal degeneration measured by Fluoro Jade and COX-2 protein expression levels were not affected. In conclusion, COX-2 inhibition by diclofenac was associated with decreased apoptosis and lesion area after focal penetrating TBI and may be of interest for further studies of clinical applications.

17.
J Clin Neurosci ; 64: 227-233, 2019 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-30948313

RESUMEN

Traumatic brain injury (TBI) is followed by a secondary inflammation in the brain. Neuroprotectin D1 (NPD1) is synthesized from docosahexaenoic acid (DHA) and has anti-inflammatory and antiapoptotic effects in experimental models of neurodegenerative disease and brain ischemia-reperfusion. It is not known whether intralesional administration of NPD1 ameliorates inflammation and cell death after severe TBI. We therefore investigated the effects of NPD1 following a severe form of focal penetrating TBI. A total of 30 male Sprague-Dawley rats weighing between 350 and 450 g were exposed to focal penetrating TBI or sham surgery. The rats were randomized to NPD1 treatment (50 ng intralesionally, immediately following TBI) or no treatment. The rats were sacrificed at 24 or 72 h. All subgroups consisted of 5 rats. Brains were removed, fresh frozen, cut in 14-µm coronal sections and subjected to Fluoro-Jade, TUNEL, MnSOD, 3-NT, COX-2, Ox-42 and NF-κB immuno-staining and lesion size analyses. NPD1 decreased the lesion area at 72 h compared to no treatment with a mean change 42% (NPD1 14.1 mm2; no treatment 24.5 mm2) (p < 0.01). No difference was detected in markers for neuronal degeneration, apoptosis, anti-inflammatory or antioxidative enzymes, or immune cells. In conclusion, single-dose intralesional administration of NPD1 had brain tissue sparing effects after focal penetrating TBI, which may be beneficial in preventing brain tissue damage, making NPD1 a potential candidate for further clinical applications. Exact mechanisms of action could not be determined and it is possible that continuous or multiple administration regimens may increase efficacy in sequential preclinical studies.


Asunto(s)
Antiinflamatorios/farmacología , Lesiones Traumáticas del Encéfalo/patología , Ácidos Docosahexaenoicos/farmacología , Animales , Apoptosis/efectos de los fármacos , Traumatismos Penetrantes de la Cabeza/patología , Inflamación , Masculino , Ratas , Ratas Sprague-Dawley
18.
J Neurotrauma ; 36(11): 1724-1737, 2019 06.
Artículo en Inglés | MEDLINE | ID: mdl-30628544

RESUMEN

Traumatic brain injury (TBI) triggers multiple pathobiological responses with differing onsets, magnitudes, and durations. Identifying the therapeutic window of individual pathologies is critical for successful pharmacological treatment. Dozens of experimental pharmacotherapies have been successfully tested in rodent models, yet all of them (to date) have failed in clinical trials. The differing time scales of rodent and human biological and pathological processes may have contributed to these failures. We compared rodent versus human time scales of TBI-induced changes in cerebral glucose metabolism, inflammatory processes, axonal integrity, and water homeostasis based on published data. We found that the trajectories of these pathologies run on different timescales in the two species, and it appears that there is no universal "conversion rate" between rodent and human pathophysiological processes. For example, the inflammatory process appears to have an abbreviated time scale in rodents versus humans relative to cerebral glucose metabolism or axonal pathologies. Limitations toward determining conversion rates for various pathobiological processes include the use of differing outcome measures in experimental and clinical TBI studies and the rarity of longitudinal studies. In order to better translate time and close the translational gap, we suggest 1) using clinically relevant outcome measures, primarily in vivo imaging and blood-based proteomics, in experimental TBI studies and 2) collecting data at multiple post-injury time points with a frequency exceeding the expected information content by two or three times. Combined with a big data approach, we believe these measures will facilitate the translation of promising experimental treatments into clinical use.


Asunto(s)
Lesiones Traumáticas del Encéfalo/metabolismo , Lesiones Traumáticas del Encéfalo/fisiopatología , Modelos Animales de Enfermedad , Animales , Humanos , Factores de Tiempo
19.
Front Neurol ; 9: 479, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29973912

RESUMEN

Repeated mild blast-induced traumatic brain injury (rmbTBI), caused by recurrent exposure to low levels of explosive blast, is a significant concern for military health systems. However, the pathobiology of rmbTBI is currently poorly understood. Animal models are important tools to identify the molecular changes of rmbTBI, but comparisons across different models can present their own challenges. In this study, we compared two well-established rodent models of mbTBI, the "KI model" and the "USU/WRAIR model." These two models create different pulse forms, in terms of peak pressure and duration. Following single and double exposures to mild levels of blast, we used in situ hybridization (ISH) to assess changes in mRNA levels of tyrosine hydroxylase (TH), tryptophan hydroxylase (TPH2), and galanin in the locus coeruleus (LC) and dorsal raphe nucleus (DRN). These systems and their transmitters are known to mediate responses to stress and anxiety. We found increased mRNA levels of TH, TPH2 and galanin in the LC and DRN of single-exposed rats relative to sham rats in the KI but not the USU/WRAIR model. Sham mRNA values measured in the USU/WRAIR model were substantially higher than their KI counterparts. Double exposure caused similarly significant increases in mRNA values in the KI model but not the USU/WRAIR model, except TPH2 and galanin levels in the DRN. We detected no cumulative effect of injury in either model at the used inter-injury interval (30 min), and there were no detectable neuropathological changes in any experimental group at 1 day post-injury. The apparent lack of early response to injury as compared to sham in the USU/WRAIR model is likely caused by stressors (e.g., transportation and noise), associated with the experimental execution, that were absent in the KI model. This study is the first to directly compare two established rodent models of rmbTBI, and to highlight the challenges of comparing findings from different animal models. Additional studies are needed to understand the role of stress, dissect the effects of psychological and physical injuries and to identify the window of increased cerebral vulnerability, i.e., the inter-injury interval that results in a cumulative effect following repeated blast exposure.

20.
eNeuro ; 5(2)2018.
Artículo en Inglés | MEDLINE | ID: mdl-29568799

RESUMEN

Traumatic brain injury (TBI) leads to a deleterious and multifactorial secondary inflammatory response in the brain. Oxidative stress from the inflammation likely contributes to the brain damage although it is unclear to which extent. A largely unexplored approach is to consider phenotypic regulation of oxidative stress levels. Genetic polymorphism influences inflammation in the central nervous system and it is possible that the antioxidative response differs between phenotypes and affects the severity of the secondary injury. We therefore compared the antioxidative response in inbred rat strains dark agouti (DA) to piebald viral glaxo (PVG). DA has high susceptibility to inflammatory challenges and PVG is protected. Primary neuronal cell cultures were exposed to peroxynitrite (ONOO-), nitric oxide (NO), superoxide (O2-), and 4-hydroxynonenal (4-HNE). Our findings demonstrated a phenotypic control of the neuronal antioxidative response, specific to manganese O2- dismutase (MnSOD). DA neurons had increased levels of MnSOD, equal levels of peroxiredoxin 5 (PRDX5), decreased oxidative stress markers 3-nitrotyrosine (3-NT) and 4-HNE and decreased neuronal death detected by lactate dehydrogenase (LDH) release after 24 h, and higher oxidative stress levels by CellROX than PVG after 2 h. It is possible that DA neurons had a phenotypic adaptation to a fiercer inflammatory environment. ONOO- was confirmed as the most powerful oxidative damage mediator, while 4-HNE caused few oxidative effects. Inducible NO synthase (iNOS) was not induced, suggesting that inflammatory, while not oxidative stimulation was required. These findings indicate that phenotypic antioxidative regulation affects the secondary inflammation, which should be considered in future individualized treatments and when evaluating antioxidative pharmacological interventions.


Asunto(s)
Lesiones Traumáticas del Encéfalo , Modelos Animales de Enfermedad , Neuronas Dopaminérgicas , Hipocampo , Inflamación , Estrés Oxidativo , Animales , Lesiones Traumáticas del Encéfalo/genética , Lesiones Traumáticas del Encéfalo/inmunología , Lesiones Traumáticas del Encéfalo/metabolismo , Técnicas de Cultivo de Célula , Susceptibilidad a Enfermedades , Neuronas Dopaminérgicas/inmunología , Neuronas Dopaminérgicas/metabolismo , Embrión de Mamíferos , Hipocampo/inmunología , Hipocampo/metabolismo , Inflamación/genética , Inflamación/inmunología , Inflamación/metabolismo , Estrés Oxidativo/genética , Estrés Oxidativo/inmunología , Ratas , Ratas Endogámicas
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...