Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 26
Filter
1.
Nicotine Tob Res ; 24(5): 736-744, 2022 03 26.
Article in English | MEDLINE | ID: mdl-34897512

ABSTRACT

INTRODUCTION: The Population Assessment of Tobacco and Health (PATH) Study is a nationally representative cohort of tobacco product users and nonusers. The study's main purpose is to obtain longitudinal epidemiologic data on tobacco use and exposure among the US population. AIMS AND METHODS: Nicotine biomarkers-cotinine (COT) and trans-3'-hydroxycotinine (HCT)-were measured in blood samples collected from adult daily tobacco users and nonusers during Wave 1 of the PATH Study (2013-2014; n = 5012; one sample per participant). Participants' tobacco product use and exposure to secondhand smoke were categorized based on questionnaire responses. Nonusers were subdivided into never users and recent former users. Daily tobacco users were classified into seven tobacco product use categories: exclusive users of cigarette, smokeless tobacco, electronic cigarette, cigar, pipe, and hookah, as well as polyusers. We calculated sample-weighted geometric mean (GM) concentrations of cotinine, HCT, and the nicotine metabolite ratio (NMR) and evaluated their associations with tobacco use with adjustment for potential confounders. RESULTS: The GMs (95% confidence intervals) of COT and HCT concentrations for daily tobacco users were 196 (184 to 208) and 72.5 (67.8 to 77.4) ng/mL, and for nonusers they were 0.033 (0.028 to 0.037) and 0.021 (0.018 to 0.023) ng/mL. Exclusive smokeless tobacco users had the highest COT concentrations of all user groups examined. The GM NMR in daily users was 0.339 (95% confidence interval: 0.330 to 0.350). CONCLUSIONS: These nationally representative estimates of serum nicotine biomarkers could be the basis for reference ranges characterizing nicotine exposure for daily tobacco users and nonusers in the US adult population. IMPLICATIONS: This report summarizes the serum nicotine biomarker measurements in Wave 1 of the PATH Study. We are reporting the first estimates of HCT in serum for daily tobacco users and nonusers in the noninstitutionalized, civilian US adult population; the first nationally representative serum COT estimates for daily exclusive users of different tobacco products and daily polyusers; and the first nationally representative estimate of the serum NMR in daily tobacco users by age, race/ethnicity, and sex.


Subject(s)
Electronic Nicotine Delivery Systems , Tobacco Use Disorder , Adult , Biomarkers , Cotinine/analogs & derivatives , Humans , Nicotine , Nicotiana , Tobacco Use Disorder/epidemiology
2.
J Neurotrauma ; 38(17): 2454-2472, 2021 09 01.
Article in English | MEDLINE | ID: mdl-33843262

ABSTRACT

Loss of plasmalemmal integrity may mediate cell death after traumatic brain injury (TBI). Prior studies in controlled cortical impact (CCI) indicated that the membrane resealing agent Kollidon VA64 improved histopathological and functional outcomes. Kollidon VA64 was therefore selected as the seventh therapy tested by the Operation Brain Trauma Therapy consortium, across three pre-clinical TBI rat models: parasagittal fluid percussion injury (FPI), CCI, and penetrating ballistic-like brain injury (PBBI). In each model, rats were randomized to one of four exposures (7-15/group): (1) sham; (2) TBI+vehicle; (3) TBI+Kollidon VA64 low-dose (0.4 g/kg); and (4) TBI+Kollidon VA64 high-dose (0.8 g/kg). A single intravenous VA64 bolus was given 15 min post-injury. Behavioral, histopathological, and serum biomarker outcomes were assessed over 21 days generating a 22-point scoring matrix per model. In FPI, low-dose VA64 produced zero points across behavior and histopathology. High-dose VA64 worsened motor performance compared with TBI-vehicle, producing -2.5 points. In CCI, low-dose VA64 produced intermediate benefit on beam balance and the Morris water maze (MWM), generating +3.5 points, whereas high-dose VA64 showed no effects on behavior or histopathology. In PBBI, neither dose altered behavior or histopathology. Regarding biomarkers, significant increases in glial fibrillary acidic protein (GFAP) levels were seen in TBI versus sham at 4 h and 24 h across models. Benefit of low-dose VA64 on GFAP was seen at 24 h only in FPI. Ubiquitin C-terminal hydrolase-L1 (UCH-L1) was increased in TBI compared with vehicle across models at 4 h but not at 24 h, without treatment effects. Overall, low dose VA64 generated +4.5 points (+3.5 in CCI) whereas high dose generated -2.0 points. The modest/inconsistent benefit observed reduced enthusiasm to pursue further testing.


Subject(s)
Brain Injuries, Traumatic/drug therapy , Pyrrolidines/therapeutic use , Vinyl Compounds/therapeutic use , Animals , Behavior, Animal , Brain Injuries, Traumatic/pathology , Brain Injuries, Traumatic/psychology , Disease Models, Animal , Glial Fibrillary Acidic Protein/metabolism , Male , Rats , Rats, Sprague-Dawley , Recovery of Function
3.
J Neurotrauma ; 38(16): 2323-2334, 2021 08 15.
Article in English | MEDLINE | ID: mdl-33544034

ABSTRACT

Mitochondrial dysfunction is a pivotal target for neuroprotection strategies for traumatic brain injury (TBI). However, comprehensive time-course evaluations of mitochondrial dysfunction are lacking in the pre-clinical penetrating TBI (PTBI) model. The current study was designed to characterize temporal responses of mitochondrial dysfunction from 30 min to 2 weeks post-injury after PTBI. Anesthetized adult male rats were subjected to either PTBI or sham craniectomy (n = 6 animals per group × 7 time points). Animals were euthanized at 30 min, 3 h, 6 h, 24 h, 3 days, 7 days, and 14 days post-PTBI, and mitochondria were isolated from the ipsilateral hemisphere of brain regions near the injury core (i.e., frontal cortex [FC] and striatum [ST]) and a more distant region from the injury core (i.e., hippocampus [HIP]). Mitochondrial bioenergetics parameters were measured in real time using the high-throughput procedures of the Seahorse Flux Analyzer (Agilent Technologies, Santa Clara, CA). The post-injury time course of FC + ST showed a biphasic mitochondrial bioenergetics dysfunction response, indicative of reduced adenosine triphosphate synthesis rate and maximal respiratory capacity after PTBI. An initial phase of energy crisis was detected at 30 min (-42%; p < 0.05 vs. sham), which resolved to baseline levels between 3 and 6 h (non-significant vs. sham). This was followed by a second and more robust phase of bioenergetics dysregulation detected at 24 h that remained unresolved out to 14 days post-injury (-55% to -90%; p < 0.05 vs. sham). In contrast, HIP mitochondria showed a delayed onset of mitochondrial dysfunction at 7 days (-74%; p < 0.05 vs. sham) that remained evident out to 14 days (-51%; p < 0.05 vs. sham) post-PTBI. Collectively, PTBI-induced mitochondrial dysfunction responses were time and region specific, evident differentially at the injury core and distant region of PTBI. The current results provide the basis that mitochondrial dysfunction may be targeted differentially based on region specificity post-PTBI. Even more important, these results suggest that therapeutic interventions targeting mitochondrial dysfunction may require extended dosing regimens to achieve clinical efficacy after TBI.


Subject(s)
Brain Injuries, Traumatic/metabolism , Energy Metabolism/physiology , Head Injuries, Penetrating/metabolism , Mitochondria/physiology , Animals , Disease Models, Animal , Male , Rats , Rats, Sprague-Dawley , Time Factors
4.
Toxicol Appl Pharmacol ; 386: 114826, 2020 01 01.
Article in English | MEDLINE | ID: mdl-31730783

ABSTRACT

The widespread use and high abuse liability of tobacco products has received considerable public health attention, in particular for youth, who are vulnerable to nicotine addiction. In this study, adult and adolescent squirrel monkeys were used to evaluate age-related metabolism and pharmacokinetics of nicotine after intravenous administration. A physiologically-based pharmacokinetic (PBPK) model was created to characterize the pharmacokinetic behaviors of nicotine and its metabolites, cotinine, trans-3'-hydroxycotinine (3'-OH cotinine), and trans-3'-hydroxycotinine glucuronide (3'-OH cotinine glucuronide) for both adult and adolescent squirrel monkeys. The PBPK nicotine model was first calibrated for adult squirrel monkeys utilizing in vitro nicotine metabolic data, plasma concentration-time profiles and cumulative urinary excretion data for nicotine and metabolites. Further model refinement was conducted when the calibrated adult model was scaled to the adolescents, because adolescents appeared to clear nicotine and cotinine more rapidly relative to adults. More specifically, the resultant model parameters representing systemic clearance of nicotine and cotinine for adolescent monkeys were approximately two- to three-fold of the adult values on a per body weight basis. The nonhuman primate PBPK model in general captured experimental observations that were used for both model calibration and evaluation, with acceptable performance metrics for precision and bias. The model also identified differences in nicotine pharmacokinetics between adolescent and adult nonhuman primates which might also be present in humans.


Subject(s)
Nicotine/pharmacokinetics , Age Factors , Animals , Cotinine/metabolism , Cotinine/urine , Injections, Intravenous , Liver/metabolism , Male , Nicotine/administration & dosage , Nicotine/blood , Nicotine/urine , Saimiri
5.
Front Neurol ; 10: 699, 2019.
Article in English | MEDLINE | ID: mdl-31312174

ABSTRACT

Close-head concussive injury, as one of the most common forms of traumatic brain injury (TBI), has been shown to induce cognitive deficits that are long lasting. A concussive impact model was previously established in our lab that produces clinically relevant signs of concussion and induced acute pathological changes in rats. To evaluate the long-term effects of repeated concussions in this model, we utilized a comprehensive Morris water maze (MWM) paradigm for cognitive assessments at 1 and 6 months following repeated concussive impacts in rats. As such, adult Sprague-Dawley rats received either anesthesia (sham) or repeated concussive impacts (4 consecutive impacts at 1 h interval). At 1 month post-injury, results of the spatial learning task showed that the average latencies to locate the hidden "escape" platform were significantly longer in the injured rats over the last 2 days of the MWM testing compared to sham controls (p < 0.05). In the memory retention task, rats subjected to repeated concussive impacts also spent significantly less time in the platform zone searching for the missing platform during the probe trial (p < 0.05). On the working memory task, the injured rats showed a trend toward worse performance, but this failed to reach statistical significance compared to sham controls (p = 0.07). At 6 months post-injury, no differences were detected between the injured group and sham controls in either the spatial learning or probe trials. However, rats with repeated concussive impacts exhibited significantly worsened working memory performance compared to sham controls (p < 0.05). In addition, histopathological assessments for axonal neurodegeneration using silver stain showed that repeated concussive impacts induced significantly more axonal degeneration in the corpus callosum compared to sham controls (p < 0.05) at 1 month post-injury, whereas such difference was not observed at 6 months post-injury. Overall, the results show that repeated concussive impacts in our model produced significant cognitive deficits in both spatial learning abilities and in working memory abilities in a time-dependent fashion that may be indicative of progressive pathology and warrant further investigation.

6.
Front Neurol ; 10: 605, 2019.
Article in English | MEDLINE | ID: mdl-31244764

ABSTRACT

Mitochondria constitute a central role in brain energy metabolism, and play a pivotal role in the development of secondary pathophysiology and subsequent neuronal cell death following traumatic brain injury (TBI). Under normal circumstances, the brain consumes glucose as the preferred energy source for adenosine triphosphate (ATP) production over ketones. To understand the comprehensive picture of substrate-specific mitochondrial bioenergetics responses following TBI, adult male rats were subjected to either 10% unilateral penetrating ballistic-like brain injury (PBBI) or sham craniectomy (n = 5 animals per group). At 24 h post-injury, mitochondria were isolated from pooled brain regions (frontal cortex and striatum) of the ipsilateral hemisphere. Mitochondrial bioenergetics parameters were measured ex vivo in the presence of four sets of metabolic substrates: pyruvate+malate (PM), glutamate+malate (GM), succinate (Succ), and ß-hydroxybutyrate+malate (BHBM). Additionally, mitochondrial matrix dehydrogenase activities [i.e., pyruvate dehydrogenase complex (PDHC), alpha-ketoglutarate dehydrogenase complex (α-KGDHC), and glutamate dehydrogenase (GDH)] and mitochondrial membrane-bound dehydrogenase activities [i.e., electron transport chain (ETC) Complex I, II, and IV] were compared between PBBI and sham groups. Furthermore, mitochondrial coenzyme contents, including NAD(t) and FAD(t), were quantitatively measured in both groups. Collectively, PBBI led to an overall significant decline in the ATP synthesis rates (43-50%; * p < 0.05 vs. sham) when measured using each of the four sets of substrates. The PDHC and GDH activities were significantly reduced in the PBBI group (42-53%; * p < 0.05 vs. sham), whereas no significant differences were noted in α-KGDHC activity between groups. Both Complex I and Complex IV activities were significantly reduced following PBBI (47-81%; * p < 0.05 vs. sham), whereas, Complex II activity was comparable between groups. The NAD(t) and FAD(t) contents were significantly decreased in the PBBI group (27-35%; * p < 0.05 vs. sham). The decreased ATP synthesis rates may be due to the significant reductions in brain mitochondrial dehydrogenase activities and coenzyme contents observed acutely following PBBI. These results provide a basis for the use of "alternative biofuels" for achieving higher ATP production following severe penetrating brain trauma.

7.
J Trauma Acute Care Surg ; 83(1 Suppl 1): S16-S24, 2017 07.
Article in English | MEDLINE | ID: mdl-28628600

ABSTRACT

BACKGROUND: Brain-derived neurotrophic factor (BDNF) and insulin-like growth factor-1 (IGF-1) are essential for neuroplasticity and neuronal survival. Despite the importance of these endogenous factors in mediating posttraumatic recovery, little is known about their response after penetrating type traumatic brain injury. The objective of this study was to quantify the expression levels BDNF and IGF-1, two well-known neuroplasticity mediators, after penetrating ballistic-like brain injury (PBBI). METHODS: Rats were randomly assigned to receive unilateral sham or PBBI injuries. Using enzyme-linked immunosorbent assay and immunohistochemistry, we performed a comprehensive evaluation of BDNF and IGF-1 expression at acute (1 hour, 6 hours, 1 day) and subacute (2, 3, 7, and 14 days) timepoints after injury. RESULTS: BDNF and IGF-1 expression was transiently upregulated in both cortex and hippocampus after PBBI. Although BDNF levels increased at acute timepoints, IGF-1 expression peaked at 3 days in cortical homogenates. Although there was loss of staining in cells bordering the cavity, increased BDNF and IGF-1 immunoreactivity was observed in scattered neurons away from the contusion site. Glial upregulation of both growth factors was observed at early timepoints in the hippocampus. CONCLUSION: Our findings demonstrate that PBBI results in a brief upregulation of BDNF and IGF-1 during early posttraumatic period, providing critical information for interventions aiming to enhance neuronal survival and brain plasticity.


Subject(s)
Brain-Derived Neurotrophic Factor/metabolism , Head Injuries, Penetrating/metabolism , Insulin-Like Growth Factor I/metabolism , Animals , Enzyme-Linked Immunosorbent Assay , Immunohistochemistry , Male , Military Medicine , Random Allocation , Rats , Rats, Sprague-Dawley
8.
J Neurotrauma ; 34(19): 2768-2789, 2017 10 01.
Article in English | MEDLINE | ID: mdl-28326890

ABSTRACT

Closed-head concussive injury is one of the most common causes of traumatic brain injury (TBI). Isolated concussions frequently produce acute neurological impairments, and individuals typically recover spontaneously within a short time frame. In contrast, brain injuries resulting from multiple concussions can result in cumulative damage and elevated risk of developing chronic brain pathologies. Increased attention has focused on identification of diagnostic markers that can prognostically serve as indices of brain health after injury, revealing the temporal profile of vulnerability to a second insult. Such markers may demarcate adequate recovery periods before concussed patients can return to required activities. We developed a noninvasive closed-head impact model that captures the hallmark symptoms of concussion in the absence of gross tissue damage. Animals were subjected to single or repeated concussive impact and examined using a battery of neurological, vestibular, sensorimotor, and molecular metrics. A single concussion induced transient, but marked, acute neurological impairment, gait alterations, neuronal death, and increased glial fibrillary acidic protein (GFAP) expression in brain tissue. As expected, repeated concussions exacerbated sensorimotor dysfunction, prolonged gait abnormalities, induced neuroinflammation, and upregulated GFAP and tau. These animals also exhibited chronic functional neurological impairments with sustained astrogliosis and white matter thinning. Acute changes in molecular signatures correlated with behavioral impairments, whereas increased times to regaining consciousness and balance impairments were associated with higher GFAP and neuroinflammation. Overall, behavioral consequences of either single or repeated concussive impact injuries appeared to resolve more quickly than the underlying molecular, metabolic, and neuropathological abnormalities. This observation, which is supported by similar studies in other mTBI models, underscores the critical need to develop more objective prognostic measures for guiding return-to-play decisions.


Subject(s)
Brain Concussion , Disease Models, Animal , Animals , Brain Concussion/complications , Brain Concussion/pathology , Brain Concussion/physiopathology , Male , Rats , Rats, Sprague-Dawley
9.
J Trauma Acute Care Surg ; 81(5): 860-867, 2016 11.
Article in English | MEDLINE | ID: mdl-27769083

ABSTRACT

BACKGROUND: Energy metabolic dysfunction is a key determinant of cellular damage following traumatic brain injury and may be worsened by additional insults. This study evaluated the acute/subacute effects of combined hypoxemia (HX) and hemorrhagic shock (HS) on cerebral interstitial levels of glucose, lactate, and pyruvate in a rat model of penetrating ballistic-like brain injury (PBBI). METHODS: Rats were randomly assigned into the sham control, PBBI, and combined injury (P + HH) groups. The P + HH group received PBBI followed by 30-minute HX and 30 minute HS. Samples were collected from striatum (perilesional region) using intracerebral microdialysis at 1 to 3 hours after injury and then at 1 to 3, 7, and 14 days after injury. Glucose, lactate, and pyruvate were measured in the dialysate samples. RESULTS: Glucose levels dropped significantly up to 24 hours following injury in both PBBI and P + HH groups (p < 0.05). A reduction in pyruvate was observed in the PBBI group from 24 to 72 hours after injury (vs. sham). In the P + HH group, the pyruvate was significantly reduced from 2 to 24 hours after injury (p < 0.05 vs. PBBI). This prominent reduction persisted for 14 days after injury. In contrast, lactate levels were significantly increased in the PBBI group during the first 24 hours after injury and remained elevated out to 7 days. The P + HH group exhibited a similar trend of lactate increase as did the PBBI group. Critically, P + HH further increased the lactate-to-pyruvate ratio by more than twofold (vs. PBBI) during the first 24 hours. The ratio reached a peak at 2 hours and then gradually decreased, but the level remained significantly higher than that in the sham control from 2 to 14 days after injury (p < 0.05). CONCLUSION: This study identified the temporal profile of energy-related neurochemical dysregulation induced by PBBI and combined injury in the perilesional region. Furthermore, combined HX and HS further reduced the pyruvate level and increased the lactate-to-pyruvate ratio following PBBI, indicating the exacerbation of posttraumatic metabolic perturbation.


Subject(s)
Brain/metabolism , Head Injuries, Penetrating/metabolism , Hypoxia/metabolism , Shock, Hemorrhagic/metabolism , Animals , Brain/pathology , Disease Models, Animal , Glucose/metabolism , Head Injuries, Penetrating/complications , Head Injuries, Penetrating/pathology , Hypoxia/etiology , Lactic Acid/metabolism , Male , Microdialysis , Pyruvic Acid/metabolism , Random Allocation , Rats , Rats, Sprague-Dawley , Shock, Hemorrhagic/etiology
10.
Methods Mol Biol ; 1462: 89-100, 2016.
Article in English | MEDLINE | ID: mdl-27604714

ABSTRACT

The central nervous system (CNS) is protected by blood-brain barrier (BBB) and blood-cerebrospinal-fluid (CSF) barrier that limit toxic agents and most molecules from penetrating the brain and spinal cord. However, these barriers also prevent most pharmaceuticals from entering into the CNS. Drug delivery to the CNS following neurotrauma is complicated. Although studies have shown BBB permeability increases in various TBI models, it remains as the key mitigating factor for delivering drugs into the CNS. The commonly used methods for drug delivery in preclinical neurotrauma studies include intraperitoneal, subcutaneous, intravenous, and intracerebroventricular delivery. It should be noted that for a drug to be successfully translated into the clinic, it needs to be administered preclinically as it would be anticipated to be administered to patients. And this likely leads to better dose selection of the drug, as well as recognition of any possible side effects, prior to transition into a clinical trial. Additionally, novel approach that is noninvasive and yet circumvents BBB, such as drug delivery through nerve pathways innervating the nasal passages, needs to be investigated in animal models, as it may provide a viable drug delivery method for patients who sustain mild CNS injury or require chronic treatments. Therefore, the focus of this chapter is to present rationales and methods for delivering drugs by IV infusion via the jugular vein, and intranasally in preclinical studies.


Subject(s)
Drug Delivery Systems , Pharmaceutical Preparations/administration & dosage , Trauma, Nervous System/drug therapy , Animals , Brain Injuries, Traumatic/drug therapy , Brain Injuries, Traumatic/etiology , Brain Injuries, Traumatic/pathology , Disease Models, Animal , Drug Administration Routes , Drug Delivery Systems/methods , Male , Rats , Trauma, Nervous System/etiology , Trauma, Nervous System/pathology
11.
Methods Mol Biol ; 1462: 445-58, 2016.
Article in English | MEDLINE | ID: mdl-27604733

ABSTRACT

Animal models of traumatic brain injury (TBI) provide important tools for studying the pathobiology of brain trauma and for evaluating therapeutic or diagnostic targets. Incorporation of additional insults such as hemorrhagic shock (HS) and/or hypoxemia (HX) into these models more closely recreates clinical scenarios as TBI often occurs in conjunction with these systemic insults (i.e., polytrauma). We have developed a rat model of polytrauma that combines penetrating TBI, HS and HX. Following brain trauma, HX was induced by reducing the inspired oxygen while HS was induced by withdrawing blood to lower the mean arterial pressure. The physiological, histological, and behavioral aspects of this animal model have been characterized and have demonstrated exacerbating effects of systemic insults on penetrating TBI. As such, this model may facilitate the use of simultaneous assessments of multiple mechanisms and provide a platform for testing novel diagnostic and therapeutic targets.


Subject(s)
Brain Injuries, Traumatic/etiology , Brain Injuries, Traumatic/pathology , Disease Models, Animal , Hypoxia/etiology , Shock, Hemorrhagic/etiology , Shock, Hemorrhagic/pathology , Animals , Brain Injuries, Traumatic/physiopathology , Hypoxia/pathology , Hypoxia/physiopathology , Male , Rats , Shock, Hemorrhagic/physiopathology , Trauma, Nervous System/etiology , Trauma, Nervous System/pathology , Trauma, Nervous System/physiopathology
12.
Methods Mol Biol ; 1462: 539-51, 2016.
Article in English | MEDLINE | ID: mdl-27604737

ABSTRACT

The Morris water maze (MWM) task is one of the most widely used and versatile tools in behavioral neuroscience for evaluating spatial learning and memory. With regard to detecting cognitive deficits following central nervous system (CNS) injuries, MWM has been commonly utilized in various animal models of neurotrauma, such as fluid percussion injury (FPI), cortical controlled impact (CCI) injury, weight-drop impact injury, and penetrating ballistic-like brain injury (PBBI). More importantly, it serves as a therapeutic index for assessing the efficacy of treatment interventions on cognitive performance following neurotrauma. Thus, it is critical to design an MWM testing paradigm that is sensitive yet discriminating for the purpose of evaluating potential therapeutic interventions. In this chapter, we discuss how multiple test manipulations, including the size of platform, numbers of trials per day, the frequency of retesting intervals, and the texture of platform surface, impact MWM's ability to detect cognitive deficits using a rat model of PBBI.


Subject(s)
Brain Injuries, Traumatic/etiology , Brain Injuries, Traumatic/physiopathology , Cognition , Disease Models, Animal , Maze Learning , Animals , Cognition Disorders/etiology , Cognition Disorders/physiopathology , Male , Memory , Rats , Spatial Learning
13.
Methods Mol Biol ; 1462: 597-610, 2016.
Article in English | MEDLINE | ID: mdl-27604740

ABSTRACT

Mitochondrial dysfunction is one of the key posttraumatic neuropathological events observed in various experimental models of traumatic brain injury (TBI). The extent of mitochondrial dysfunction has been associated with the severity and time course of secondary injury following brain trauma. Critically, several mitochondrial targeting preclinical drugs used in experimental TBI models have shown improved mitochondrial bioenergetics, together with cortical tissue sparing and cognitive behavioral outcome. Mitochondria, being a central regulator of cellular metabolic pathways and energy producer of cells, are of a great interest for researchers aiming to adopt cutting-edge methodology for mitochondrial bioenergetics assessment. The traditional way of mitochondrial bioenergetics analysis utilizing a Clark-type oxygen electrode (aka. oxytherm) is time-consuming and labor-intensive. In the present chapter, we describe an advanced and high-throughput method for mitochondrial bioenergetics assessments utilizing the Seahorse Biosciences XF(e)24 Flux Analyzer. This allows for simultaneous measurement of multiple samples with higher efficiency than the oxytherm procedure. This chapter provides helpful guidelines for conducting mitochondrial isolation and studying mitochondrial bioenergetics in brain tissue homogenates following experimental TBI.


Subject(s)
Energy Metabolism , High-Throughput Screening Assays , Mitochondria/metabolism , Trauma, Nervous System/etiology , Trauma, Nervous System/metabolism , Animals , Cell Respiration/drug effects , Drug Discovery , Drug Evaluation, Preclinical , Male , Metabolomics/methods , Mitochondria/drug effects , Oxidative Phosphorylation , Rats , Reactive Oxygen Species/metabolism , Trauma, Nervous System/drug therapy
14.
J Neurotrauma ; 33(6): 523-37, 2016 Mar 15.
Article in English | MEDLINE | ID: mdl-26670792

ABSTRACT

Nicotinamide (vitamin B3) was the first drug selected for cross-model testing by the Operation Brain Trauma Therapy (OBTT) consortium based on a compelling record of positive results in pre-clinical models of traumatic brain injury (TBI). Adult male Sprague-Dawley rats were exposed to either moderate fluid percussion injury (FPI), controlled cortical impact injury (CCI), or penetrating ballistic-like brain injury (PBBI). Nicotinamide (50 or 500 mg/kg) was delivered intravenously at 15 min and 24 h after injury with subsequent behavioral, biomarker, and histopathological outcome assessments. There was an intermediate effect on balance beam performance with the high (500 mg/kg) dose in the CCI model, but no significant therapeutic benefit was detected on any other motor task across the OBTT TBI models. There was an intermediate benefit on working memory with the high dose in the FPI model. A negative effect of the low (50 mg/kg) dose, however, was observed on cognitive outcome in the CCI model, and no cognitive improvement was observed in the PBBI model. Lesion volume analysis showed no treatment effects after either FPI or PBBI, but the high dose of nicotinamide resulted in significant tissue sparing in the CCI model. Biomarker assessments included measurements of glial fibrillary acidic protein (GFAP) and ubiquitin carboxyl-terminal hydrolase-1 (UCH-L1) in blood at 4 or 24 h after injury. Negative effects (both doses) were detected on biomarker levels of GFAP after FPI and on biomarker levels of UCH-L1 after PBBI. The high dose of nicotinamide, however, reduced GFAP levels after both PBBI and CCI. Overall, our results showed a surprising lack of benefit from the low dose nicotinamide. In contrast, and partly in keeping with the literature, some benefit was achieved with the high dose. The marginal benefits achieved with nicotinamide, however, which appeared sporadically across the TBI models, has reduced enthusiasm for further investigation by the OBTT Consortium.


Subject(s)
Brain Injuries, Traumatic , Niacinamide/administration & dosage , Recovery of Function/drug effects , Vitamin B Complex/administration & dosage , Animals , Biomarkers/blood , Disease Models, Animal , Dose-Response Relationship, Drug , Glial Fibrillary Acidic Protein/blood , Male , Rats , Rats, Sprague-Dawley , Ubiquitin Thiolesterase/blood
15.
J Neurotrauma ; 33(6): 553-66, 2016 Mar 15.
Article in English | MEDLINE | ID: mdl-26671075

ABSTRACT

Operation Brain Trauma Therapy (OBTT) is a consortium of investigators using multiple pre-clinical models of traumatic brain injury (TBI) to bring acute therapies to clinical trials. To screen therapies, we used three rat models (parasagittal fluid percussion injury [FPI], controlled cortical impact [CCI], and penetrating ballistic-like brain injury [PBBI]). We report results of the third therapy (cyclosporin-A; cyclosporine; [CsA]) tested by OBTT. At each site, rats were randomized to treatment with an identical regimen (TBI + vehicle, TBI + CsA [10 mg/kg], or TBI + CsA [20 mg/kg] given intravenously at 15 min and 24 h after injury, and sham). We assessed motor and Morris water maze (MWM) tasks over 3 weeks after TBI and lesion volume and hemispheric tissue loss at 21 days. In FPI, CsA (10 mg/kg) produced histological protection, but 20 mg/kg worsened working memory. In CCI, CsA (20 mg/kg) impaired MWM performance; surprisingly, neither dose showed benefit on any outcome. After PBBI, neither dose produced benefit on any outcome, and mortality was increased (20 mg/kg) partly caused by the solvent vehicle. In OBTT, CsA produced complex effects with histological protection at the lowest dose in the least severe model (FPI), but only deleterious effects as model severity increased (CCI and PBBI). Biomarker assessments included measurements of glial fibrillary acidic protein (GFAP) and ubiquitin C-terminal hydrolase-L1 (UCH-L1) in blood at 4 or 24 h after injury. No positive treatment effects were seen on biomarker levels in any of the models, whereas significant increases in 24 h UCH-L1 levels were seen with CsA (20 mg/kg) after CCI and 24 h GFAP levels in both CsA treated groups in the PBBI model. Lack of behavioral protection in any model, indicators of toxicity, and a narrow therapeutic index reduce enthusiasm for clinical translation.


Subject(s)
Brain Injuries, Traumatic , Cyclosporine/pharmacology , Immunosuppressive Agents/pharmacology , Recovery of Function/drug effects , Animals , Biomarkers/blood , Disease Models, Animal , Glial Fibrillary Acidic Protein/blood , Male , Random Allocation , Rats , Rats, Sprague-Dawley , Ubiquitin Thiolesterase/blood
16.
J Neurotrauma ; 33(6): 606-14, 2016 Mar 15.
Article in English | MEDLINE | ID: mdl-26671284

ABSTRACT

Operation Brain Trauma Therapy (OBTT) is a fully operational, rigorous, and productive multicenter, pre-clinical drug and circulating biomarker screening consortium for the field of traumatic brain injury (TBI). In this article, we synthesize the findings from the first five therapies tested by OBTT and discuss both the current work that is ongoing and potential future directions. Based on the results generated from the first five therapies tested within the exacting approach used by OBTT, four (nicotinamide, erythropoietin, cyclosporine A, and simvastatin) performed below or well below what was expected based on the published literature. OBTT has identified, however, the early post-TBI administration of levetiracetam as a promising agent and has advanced it to a gyrencephalic large animal model--fluid percussion injury in micropigs. The sixth and seventh therapies have just completed testing (glibenclamide and Kollidon VA 64), and an eighth drug (AER 271) is in testing. Incorporation of circulating brain injury biomarker assessments into these pre-clinical studies suggests considerable potential for diagnostic and theranostic utility of glial fibrillary acidic protein in pre-clinical studies. Given the failures in clinical translation of therapies in TBI, rigorous multicenter, pre-clinical approaches to therapeutic screening such as OBTT may be important for the ultimate translation of therapies to the human condition.


Subject(s)
Brain Injuries, Traumatic/drug therapy , Drug Evaluation, Preclinical/trends , Neuroprotective Agents/therapeutic use , Animals , Disease Models, Animal , Drug Evaluation, Preclinical/methods , Male , Neurology/methods , Neurology/trends , Rats , Rats, Sprague-Dawley
17.
J Neurotrauma ; 33(1): 147-56, 2016 Jan 01.
Article in English | MEDLINE | ID: mdl-25789543

ABSTRACT

Acute traumatic brain injury (TBI) is associated with neurological dysfunction, changes in brain proteins, and increased serum biomarkers. However, the relationship between these brain proteins and serum biomarkers, and the ability of these serum biomarkers to indicate a neuroprotective/therapeutic response, remains elusive. Penetrating ballistic-like brain injury (PBBI) was used to systematically analyze several key TBI biomarkers, glial fibrillary acidic protein (GFAP) and its break-down products (BDPs)-ubiquitin C-terminal hydrolase-L1 (UCH-L1), α-II spectrin, and α-II spectrin BDPs (SBDPs)-in brain tissues and serum during an extended acute-subacute time-frame. In addition, neurological improvement and serum GFAP theranostic value was evaluated after neuroprotective treatment. In brain tissues, total GFAP increased more than three-fold 2 to 7 d after PBBI. However, this change was primarily due to GFAP-BDPs which increased to 2.7-4.8 arbitrary units (AU). Alpha-II spectrin was nearly ablated 3 d after PBBI, but somewhat recovered after 7 d. In conjunction with α-II spectrin loss, SBDP-145/150 increased approximately three-fold 2 to 7 d after PBBI (vs. sham, p<0.05). UCH-L1 protein levels were slightly decreased 7 d after PBBI but otherwise were unaffected. Serum GFAP was elevated by 3.2- to 8.8-fold at 2 to 4 h (vs. sham; p<0.05) and the 4 h increase was strongly correlated to 3 d GFAP-BDP abundance (r=0.66; p<0.05). Serum GFAP showed such a strong injury effect that it also was evaluated after therapeutic intervention with cyclosporin A (CsA). Administration of 2.5 mg/kg CsA significantly reduced serum GFAP elevation by 22.4-fold 2 h after PBBI (vs. PBBI+vehicle; p<0.05) and improved neurological function 1 d post-injury. Serum biomarkers, particularly GFAP, may be correlative tools of brain protein changes and feasible theranostic markers of TBI progression and recovery.


Subject(s)
Glial Fibrillary Acidic Protein/metabolism , Head Injuries, Penetrating/metabolism , Spectrin/metabolism , Ubiquitin Thiolesterase/metabolism , Animals , Biomarkers/blood , Disease Models, Animal , Glial Fibrillary Acidic Protein/blood , Head Injuries, Penetrating/blood , Male , Rats , Rats, Sprague-Dawley , Ubiquitin Thiolesterase/blood
18.
Ther Hypothermia Temp Manag ; 6(1): 30-9, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26684246

ABSTRACT

Brain hypothermia has been considered as a promising alternative to whole-body hypothermia in treating acute neurological disease, for example, traumatic brain injury. Previously, we demonstrated that 2-hours selective brain cooling (SBC) effectively mitigated acute (≤24 hours postinjury) neurophysiological dysfunction induced by a penetrating ballistic-like brain injury (PBBI) in rats. This study evaluated neuroprotective effects of extended SBC (4 or 8 hours in duration) on sub-acute secondary injuries between 3 and 21 days postinjury (DPI). SBC (34°C) was achieved via extraluminal cooling of rats' bilateral common carotid arteries (CCA). Depending on the experimental design, SBC was introduced either immediately or with a 2- or 4-hour delay after PBBI and maintained for 4 or 8 hours. Neuroprotective effects of SBC were evaluated by measuring brain lesion volume, axonal injury, neuroinflammation, motor and cognitive functions, and post-traumatic seizures. Compared to untreated PBBI animals, 4 or 8 hours SBC treatment initiated immediately following PBBI produced comparable neuroprotective benefits against PBBI-induced early histopathology at 3 DPI as evidenced by significant reductions in brain lesion volume, axonal pathology (beta-amyloid precursor protein staining), neuroinflammation (glial fibrillary acetic protein stained-activated astrocytes and rat major histocompatibility complex class I stained activated microglial cell), and post-traumatic nonconvulsive seizures. In the later phase of the injury (7-21 DPI), significant improvement on motor function (rotarod test) was observed under most SBC protocols, including the 2-hour delay in SBC initiation. However, SBC treatment failed to improve cognitive performance (Morris water maze test) measured 13-17 DPI. The protective effects of SBC on delayed axonal injury (silver staining) were evident out to 14 DPI. In conclusion, the CCA cooling method of SBC produced neuroprotection measured across multiple domains that were evident days/weeks beyond the cooling duration and in the absence of overt adverse effects. These "proof-of-concept" results suggest that SBC may provide an attractive neuroprotective approach for clinical considerations.


Subject(s)
Brain Injuries, Traumatic/therapy , Head Injuries, Penetrating/therapy , Hypothermia, Induced , Animals , Brain/pathology , Brain Injuries, Traumatic/pathology , Electroencephalography , Head Injuries, Penetrating/pathology , Isoflurane , Male , Maze Learning , Models, Animal , Random Allocation , Rats, Sprague-Dawley , Rotarod Performance Test
19.
J Trauma Acute Care Surg ; 79(4 Suppl 2): S130-8, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26406425

ABSTRACT

BACKGROUND: Traumatic brain injury often occurs with concomitant hypoxemia (HX) and hemorrhagic shock (HS), leading to poor outcomes. This study characterized the acute physiology and subacute behavioral consequences of these additional insults in a model of penetrating ballistic-like brain injury (PBBI). METHODS: Rats were randomly assigned into sham control, HX + HS (HH), 5% PBBI alone, 5% PBBI + HH, 10% PBBI alone, and 10% PBBI + HH groups. Mean arterial pressure, heart rate, and breathing rate were monitored continuously. In the combined injury groups, animals were subjected to 30-minute HX (Pao2, 30-40 mm Hg) and then 30-min HS (mean arterial pressure, 40 mm Hg) followed by fluid resuscitation with lactated Ringer's solution after PBBI or sham PBBI. Motor function was assessed using the rotarod task at 7 days and 14 days after injury. Cognitive function was assessed in the Morris water maze task from 13 days to 17 days after injury. RESULTS: Combined HH caused acute bradycardia that was reversed by fluid resuscitation. During HX phase, tachypnea was observed in all HH groups. Persistent bradypnea was detected in 10% PBBI + HH group during the resuscitation phase. PBBI produced significant decrements in motor performance (vs. sham and HH groups). Additional insults significantly worsened motor deficits following 5% PBBI but not 10% PBBI. Both 5% PBBI and 10% PBBI produced significant cognitive deficits in the Morris water maze task with worsened deficits evident following the more severe injury (i.e., 10% PBBI). Alternatively, rats subjected to 5% PBBI + HH exhibited cognitive impairment that was significantly worse compared with 5% PBBI alone, whereas this worsening effect was not detected in the 10% PBBI groups. CONCLUSION: This study characterized the physiological responses and neurobehavioral profiles following combined PBBI and HH. Ten percent PBBI produces motor and cognitive deficits, which may exceed a sensitivity threshold capacity. In contrast, 5% PBBI produces a lower, albeit significant, magnitude of deficits and thus provides a more sensitive screen for evaluating the cumulative effects of additional insults, which were indeed demonstrated to significantly worsen outcome.


Subject(s)
Head Injuries, Penetrating/physiopathology , Hypotension/physiopathology , Hypoxia/physiopathology , Animals , Blood Gas Analysis , Disease Models, Animal , Hemodynamics/physiology , Male , Maze Learning/physiology , Postural Balance/physiology , Psychomotor Performance/physiology , Random Allocation , Rats , Rats, Sprague-Dawley , Resuscitation/methods , Trauma Severity Indices
20.
Restor Neurol Neurosci ; 33(2): 189-203, 2015.
Article in English | MEDLINE | ID: mdl-25588460

ABSTRACT

PURPOSE: The present work compared the behavioral outcomes of ACCS therapy delivered either intravenously (i.v.) or intracerebroventricularly (i.c.v.) after penetrating ballistic-like brain injury (PBBI). Histological markers for neuroinflammation and neurodegeneration were employed to investigate the potential therapeutic mechanism of ACCS. METHODS: Experiment-1, ACCS was administered either i.v. or i.c.v. for 1 week post-PBBI. Outcome metrics included behavioral (rotarod and Morris water maze) and gross morphological assessments. Experiment-2, rats received ACCS i.c.v for either 1 or 2 weeks post-PBBI. The inflammatory response was determined by immunohistochemistry for neutrophils and microglia reactivity. Neurodegeneration was visualized using silver staining. RESULTS: Both i.v. and i.c.v. delivery of ACCS improved motor outcome but failed to improve cognitive outcome or tissue sparing. Importantly, only i.c.v. ACCS treatment produced persistent motor improvements at a later endpoint. The i.c.v. ACCS treatment significantly reduced PBBI-induced increase in myeloperoxidase (MPO) and ionized calcium binding adaptor molecule 1 (Iba1) expression. Concomitant reduction of both Iba1 and silver staining were detected in corpus callosum with i.c.v. ACCS treatment. CONCLUSIONS: ACCS, as a treatment for TBI, showed promise with regard to functional (motor) recovery and demonstrated strong capability to modulate neuroinflammatory responses that may underline functional recovery. However, the majority of beneficial effects appear restricted to the i.c.v. route of ACCS delivery, which warrants future studies examining delivery routes (e.g. intranasal delivery) which are more clinically viable for the treatment of TBI.


Subject(s)
Cytokines/administration & dosage , Head Injuries, Penetrating/drug therapy , Motor Activity/drug effects , Neuroimmunomodulation/drug effects , Neuroprotective Agents/administration & dosage , Amnion , Animals , Brain/drug effects , Brain/pathology , Brain/physiopathology , Calcium-Binding Proteins/metabolism , Disease Models, Animal , Head Injuries, Penetrating/pathology , Head Injuries, Penetrating/physiopathology , Immunohistochemistry , Male , Maze Learning/drug effects , Maze Learning/physiology , Microfilament Proteins/metabolism , Motor Activity/physiology , Neuroimmunomodulation/physiology , Peroxidase/metabolism , Random Allocation , Rats, Sprague-Dawley , Rotarod Performance Test , Solutions
SELECTION OF CITATIONS
SEARCH DETAIL
...