Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 92
Filter
1.
Neurotrauma Rep ; 5(1): 254-266, 2024.
Article in English | MEDLINE | ID: mdl-38515547

ABSTRACT

Blast-related traumatic brain injury (bTBI) is a major cause of neurological disorders in the U.S. military that can adversely impact some civilian populations as well and can lead to lifelong deficits and diminished quality of life. Among these types of injuries, the long-term sequelae are poorly understood because of variability in intensity and number of the blast exposure, as well as the range of subsequent symptoms that can overlap with those resulting from other traumatic events (e.g., post-traumatic stress disorder). Despite the valuable insights that rodent models have provided, there is a growing interest in using injury models using species with neuroanatomical features that more closely resemble the human brain. With this purpose, we established a gyrencephalic model of blast injury in ferrets, which underwent blast exposure applying conditions that closely mimic those associated with primary blast injuries to warfighters. In this study, we evaluated brain biochemical, microstructural, and behavioral profiles after blast exposure using in vivo longitudinal magnetic resonance imaging, histology, and behavioral assessments. In ferrets subjected to blast, the following alterations were found: 1) heightened impulsivity in decision making associated with pre-frontal cortex/amygdalar axis dysfunction; 2) transiently increased glutamate levels that are consistent with earlier findings during subacute stages post-TBI and may be involved in concomitant behavioral deficits; 3) abnormally high brain N-acetylaspartate levels that potentially reveal disrupted lipid synthesis and/or energy metabolism; and 4) dysfunction of pre-frontal cortex/auditory cortex signaling cascades that may reflect similar perturbations underlying secondary psychiatric disorders observed in warfighters after blast exposure.

2.
J Neurotrauma ; 41(7-8): 1000-1004, 2024 Apr.
Article in English | MEDLINE | ID: mdl-37905505

ABSTRACT

Mild concussive events without loss of consciousness are typically left untreated and can result in neurological abnormalities at later stages of life. No systematic studies have been carried out to determine the effect of concussion or repeated mild concussive episodes on brain vulnerability towards blast exposure. We have evaluated the effect of repeated mild concussive events on the vulnerability of brain to blast exposure using neurobehavioral functional assessments. Rats were subjected to either repeated mild concussive impacts (two impacts 1 week apart using a modified Marmarou weight drop model), a single blast exposure (19 psi using an advanced blast simulator), or a single blast exposure one day after the second mild concussive impact. Neurobehavioral changes were monitored using rotating pole test, open field exploration test, and novel object recognition test. Rotating pole test results indicated that vestibulomotor function was unaffected by blast or repeated mild concussive impacts, but significant impairment was observed in the blast exposed animals who had prior repeated mild concussive impacts. Novel object recognition test revealed short-term memory loss at 1 month post-blast only in rats subjected to both repeated mild concussive impacts and blast. Horizontal activity count, ambulatory activity count, center time and margin time legacies in the open field exploratory activity test indicated that only those rats exposed to both repeated mild concussive impacts and blast develop anxiety-like behaviors at both acute and sub-acute time-points. The results indicate that a history of repeated mild concussive episodes heightens brain vulnerability to blast exposure.


Subject(s)
Blast Injuries , Brain Concussion , Military Personnel , Rats , Animals , Humans , Brain Concussion/complications , Brain , Amnesia , Afghan Campaign 2001- , Blast Injuries/complications
3.
J Neurotrauma ; 2024 Apr 05.
Article in English | MEDLINE | ID: mdl-38047526

ABSTRACT

Blast exposure can cause auditory deficits that have a lasting, significant impact on patients. Although the effects of blast on auditory functions localized to the ear have been well documented, the impact of blast on central auditory processing is largely undefined. Understanding the structural and functional alterations in the central nervous system (CNS) associated with blast injuries is crucial for unraveling blast-induced pathophysiological pathways and advancing development of therapeutic interventions. In this study, we used electrophysiology in combination with optogenetics assay, proteomic analysis, and morphological evaluation to investigate the impairment of synaptic connectivity in the auditory cortex (AC) of mice following blast exposure. Our results show that the long-range functional connectivity between the medial geniculate nucleus (MGN) and AC was impaired in the acute phase of blast injury. We also identified impaired synaptic transmission and dendritic spine alterations within 7 days of blast exposure, which recovered at 28 days post-blast. Additionally, proteomic analysis identified a few differentially expressed proteins in the cortex that are involved in synaptic signaling and plasticity. These findings collectively suggest that blast-induced alterations in the sound signaling network in the auditory cortex may underlie hearing deficits in the acute and sub-acute phases after exposure to shockwaves. This study may shed light on the perturbations underlying blast-induced auditory dysfunction and provide insights into the potential therapeutic windows for improving auditory outcomes in blast-exposed individuals.

4.
Mil Med ; 188(Suppl 6): 271-279, 2023 11 08.
Article in English | MEDLINE | ID: mdl-37948226

ABSTRACT

INTRODUCTION: Combat injuries are complex and multimodal. Most injuries to the extremities occur because of explosive devices such as improvised explosive devices. Blast exposure dramatically increases the risk of infection in combat wounds, and there is limited available information on the best antibiotic treatments for these injuries. We previously demonstrated that mice exposed to blast displayed a delayed clearance of cefazolin from the plasma and liver; further semi-mechanistic modeling determined that cefazolin concentrations in the skin of these mice were reduced. Our objective was to investigate the effects of blast on the pharmacokinetics of antibiotics of different types used for the treatment of combat wounds in the rat model. MATERIALS AND METHODS: Male Sprague Dawley rats were exposed to blast overpressure followed by injection of a bolus of animal equivalent doses of an antibiotic (cefazolin, cefepime, ertapenem, or clindamycin) into the tail vein at 1-hour post-blast exposure. Blood was collected at predetermined time points via repeated sampling from the tail vein. Animals were also euthanized at predetermined time points, at which time liver, kidney, skin, and blood via cardiac puncture were collected. Antibiotic concentrations were determined by ultra-performance liquid chromatography-tandem mass spectrometry. RESULTS: Blast-exposed rats exhibited a similar rate of clearance compared to non-blasted rats in the blood, liver, kidney, and skin, which is inconsistent with the data regarding cefazolin in blast-exposed mice. CONCLUSIONS: Our results in rats do not recapitulate our previous observation of delayed cefazolin clearance in mice following the blast overpressure exposure. Although using rats permitted us to collect multiple blood samples from the same animals, rats may not be a suitable model for measuring the pharmacokinetics of antibiotics following blast. The interpretation of the results may be challenging because of variation in data among rat subjects in the same sample groups.


Subject(s)
Anti-Bacterial Agents , Blast Injuries , Humans , Rats , Male , Mice , Animals , Rats, Sprague-Dawley , Anti-Bacterial Agents/therapeutic use , Blast Injuries/drug therapy , Cefazolin/therapeutic use , Explosions , Disease Models, Animal
5.
Mil Med ; 188(Suppl 6): 288-294, 2023 11 08.
Article in English | MEDLINE | ID: mdl-37948259

ABSTRACT

INTRODUCTION: Simulation of blast exposure in the laboratory has been inconsistent across laboratories. This is primarily because of adoption of the shock wave-generation techniques that are used in aerodynamic tests as opposed to application of blast exposures that are relevant to combat and training environments of a Warfighter. Because of the differences in blast signatures, characteristically different pathological consequences are observed among the preclinical studies. This is also further confounded by the varied exposure positioning of the animal subject (e.g., inside the blast simulator vs. at the mouth of the simulator). In this study, we compare biomechanical responses to blast exposures created in an advanced blast simulator (ABS) that generates "free-field"-like blast exposure with those produced by a traditionally applied cylindrical blast simulator (CBS) that generates a characteristically different blast signature. In addition, we have tested soft-armor vest protective responses with the ABS and CBS to compare the biomechanical responses to this form of personal protective equipment in each setting in a rodent model. MATERIALS AND METHODS: Anesthetized male Sprague-Dawley rats (n = 6) were surgically probed with an intrathoracic pressure (ITP) transducer and an intracranial pressure (ICP) transducer directed into the lateral cerebral ventricle (Millar, Inc.). An ABS for short-duration blast or a CBS for long-duration blast was used to expose animals to an incident blast overpressure of 14.14 psi (impulse: 30.27 psi*msec) or 16.3 psi (impulse: 71.9 psi*msec) using a custom-made holder (n = 3-4/group). An external pitot probe located near the animal was used to measure the total pressure (tip) and static gauge (side-on) pressure. Data were recorded using a TMX-18 data acquisition system (AstroNova Inc.). MATLAB was used to analyze the recordings to identify the peak amplitudes and rise times of the pressure traces. Peak ICP, peak ITP, and their impulses were normalized by expressing them relative to the associated peak static pressure. RESULTS: Normalized impulse (ABS: 1.02 ± 0.03 [vest] vs. 1.02 ± 0.01 [no-vest]; CBS: 1.21 ± 0.07 [vest] vs. 1.01 ± 0.01 [no-vest]) and peak pressure for ICP (ABS: 1.03 ± 0.03 [vest] vs. 0.99 ± 0.04 [no-vest]; CBS: 1.06 ± 0.08 [vest] vs. 1.13 ± 0.06 [no-vest]) remained unaltered when comparisons are made between vest and no-vest groups, and the normalized peak ITP (ABS: 1.50 ± 0.02 [vest] vs. 1.24 ± 0.16 [no-vest]; CBS: 1.71 ± 0.20 [vest] vs. 1.37 ± 0.06 [no-vest]) showed a trend of an increase in the vest group compared to the no-vest group. However, impulses in short-duration ABS (0.94 ± 0.06 [vest] vs. 0.92 ± 0.13 [no-vest]) blast remained unaltered, whereas a significant increase of ITP impulse (1.21 ± 0.07 [vest] vs. 1.17 ± 0.01 [no-vest]) in CBS was observed. CONCLUSIONS: The differences in the biomechanical response between ABS and CBS could be potentially attributed to the higher dynamic pressures that are imparted from long-duration CBS blasts, which could lead to chest compression and rapid acceleration/deceleration. In addition, ICP and ITP responses occur independently of each other, with no evidence of thoracic surge.


Subject(s)
Blast Injuries , Rats , Animals , Male , Rats, Sprague-Dawley , Explosions , Computer Simulation , Personal Protective Equipment
6.
Front Neurol ; 14: 1237647, 2023.
Article in English | MEDLINE | ID: mdl-37877029

ABSTRACT

Introduction: Mild traumatic brain injury (mTBI) caused by repetitive low-intensity blast overpressure (relBOP) in military personnel exposed to breaching and heavy weapons is often unrecognized and is understudied. Exposure to relBOP poses the risk of developing abnormal behavioral and psychological changes such as altered cognitive function, anxiety, and depression, all of which can severely compromise the quality of the life of the affected individual. Due to the structural and anatomical heterogeneity of the brain, understanding the potentially varied effects of relBOP in different regions of the brain could lend insights into the risks from exposures. Methods: In this study, using a rodent model of relBOP and western blotting for protein expression we showed the differential expression of various neuropathological proteins like TDP-43, tight junction proteins (claudin-5, occludin, and glial fibrillary acidic protein (GFAP)) and a mechanosensitive protein (piezo-2) in different regions of the brain at different intensities and frequency of blast. Results: Our key results include (i) significant increase in claudin-5 after 1x blast of 6.5 psi in all three regions and no definitive pattern with higher number of blasts, (ii) significant increase in piezo-2 at 1x followed by significant decrease after multiple blasts in the cortex, (iii) significant increase in piezo-2 with increasing number of blasts in frontal cortex and mixed pattern of expression in hippocampus and (iv) mixed pattern of TDP-3 and GFAP expression in all the regions of brain. Discussion: These results suggest that there are not definitive patterns of changes in these marker proteins with increase in intensity and/or frequency of blast exposure in any particular region; the changes in expression of these proteins are different among the regions. We also found that the orientation of blast exposure (e.g. front vs. side exposure) affects the altered expression of these proteins.

7.
Int J Mol Sci ; 24(16)2023 Aug 08.
Article in English | MEDLINE | ID: mdl-37628746

ABSTRACT

Due to use of explosive devices and heavy weapons systems in modern conflicts, the effect of BW on the brain and body is of increasing concern. These exposures have been commonly linked with neurodegenerative diseases and psychiatric disorders in veteran populations. A likely neurobiological link between exposure to blasts and the development of neurobehavioral disorders, such as depression and PTSD, could be neuroinflammation triggered by the blast wave. In this study, we exposed rats to single or repeated BW (up to four exposures-one per day) at varied intensities (13, 16, and 19 psi) to mimic the types of blast exposures that service members may experience in training and combat. We then measured a panel of neuroinflammatory markers in the brain tissue with a multiplex cytokine/chemokine assay to understand the pathophysiological process(es) associated with single and repeated blast exposures. We found that single and repeated blast exposures promoted neuroinflammatory changes in the brain that are similar to those characterized in several neurological disorders; these effects were most robust after 13 and 16 psi single and repeated blast exposures, and they exceeded those recorded after 19 psi repeated blast exposures. Tumor necrosis factor-alpha and IL-10 were changed by 13 and 16 psi single and repeated blast exposures. In conclusion, based upon the growing prominence of negative psychological health outcomes in veterans and soldiers with a history of blast exposures, identifying the molecular etiology of these disorders, such as blast-induced neuroinflammation, is necessary for rationally establishing countermeasures and treatment regimens.


Subject(s)
Cytokines , Neuroinflammatory Diseases , Animals , Rats , Brain , Tumor Necrosis Factor-alpha , Biological Assay
8.
J Neurotrauma ; 2023 Jul 21.
Article in English | MEDLINE | ID: mdl-37279302

ABSTRACT

Blast-induced traumatic brain injury (bTBI) has been identified as the signature injury of Operation Iraqi Freedom and Operation Enduring Freedom. Although the incidence of bTBI increased significantly after the introduction of improvised explosive devices, the mechanism of the injury is still uncertain, which is negatively impacting the development of suitable countermeasures. Identification of suitable biomarkers that could aid in the proper diagnosis of and prognosis for both acute and chronic bTBI is essential since bTBI frequently is occult and may not be associated with overtly detectable injuries to the head. Lysophosphatidic acid (LPA) is a bioactive phospholipid generated by activated platelets, astrocytes, choroidal plexus cells and microglia and is reported to play major roles in stimulating inflammatory processes. The levels of LPA in the cerebrospinal fluid (CSF) have been reported to increase acutely after non-blast related brain injuries. In the present study, we have evaluated the utility of LPA levels measured in the CSF and plasma of laboratory rats as an acute and chronic biomarker of brain injury resulting from single and tightly coupled repeated blast overpressure exposures. In the CSF, many LPA species increased at acute time-points, returned to normal levels at 1 month, and increased again at 6 months and 1 year post-blast overpressure exposures. In the plasma, several LPA species increased acutely, returned to normal levels by 24 h, and were significantly decreased at 1 year post-blast overpressure exposures. These decreases in LPA species in the plasma were associated with decreased levels of lysophosphatidyl choline, suggesting a defective upstream biosynthetic pathway of LPAs in the plasma. Notably, the changes in LPA levels in the CSF (but not plasma) negatively correlated with neurobehavioral functions in these rats, suggesting that CSF levels of LPAs may provide a suitable biomarker of bTBI that reflects severity of injury.

9.
Neurosci Lett ; 810: 137364, 2023 07 27.
Article in English | MEDLINE | ID: mdl-37391063

ABSTRACT

Although blast-induced traumatic brain injury (bTBI) has been designated as the signature injury of recent combat operations, its precise pathological mechanism(s) has not been identified thus far. Prior preclinical studies on bTBI demonstrated acute neuroinflammatory cascades which are known to be contributing to neurodegeneration. Danger-associated chemical patterns are released from the injured cells, which activate non-specific pattern recognition receptors, such as toll-like receptors (TLRs) leading to increased expression of inflammatory genes and release of cytokines. Upregulation of specific TLRs in the brain has been described as a mechanism of injury in diverse brain injury models unrelated to blast exposure. However, the expression profile of various TLRs in bTBI has not been investigated thus far. Hence, we have evaluated the expression of transcripts for TLR1-TLR10 in the brain of a gyrencephalic animal model of bTBI. We exposed ferrets to tightly coupled repeated blasts and determined the differential expression of TLRs (TLR1-10) by quantitative RT-PCR in multiple brain regions at 4 hr, 24 hr, 7 days and 28 days post-blast injury. The results obtained indicate that multiple TLRs are upregulated in the brain at 4 hr, 24 hr, 7 days and 28 days post-blast. Specifically, upregulation of TLR2, TLR4 and TLR9 was noted in different brain regions, suggesting that multiple TLRs might play a role in the pathophysiology of bTBI and that drugs that can inhibit multiple TLRs might have enhanced efficacy to attenuate brain damage and thereby improve bTBI outcome. Taken together, these results suggest that several TLRs are upregulated in the brain after bTBI and participate in the inflammatory response and thereby provide new insights into the disease pathogenesis. Therefore, inhibition of multiple TLRs, including TLR2, 4 and 9, simultaneously might be a potential therapeutic strategy for the treatment of bTBI.


Subject(s)
Blast Injuries , Brain Injuries, Traumatic , Brain Injuries , Animals , Ferrets , Up-Regulation , Toll-Like Receptor 2 , Toll-Like Receptor 1 , Brain , Brain Injuries, Traumatic/drug therapy , Brain Injuries/drug therapy , Brain Injuries/pathology , Toll-Like Receptors
10.
Arthrosc Sports Med Rehabil ; 5(1): e201-e206, 2023 Feb.
Article in English | MEDLINE | ID: mdl-36866312

ABSTRACT

Purpose: To evaluate the recurrent dislocation risk and patient-reported outcomes of peroneus longus allograft tissue for medial patellofemoral ligament (MPFL) reconstruction. Methods: Patients who underwent MPFL reconstruction with peroneus longus allograft at an academic center between 2008 and 2016 were identified. Record review and patient contact were used to identify any cases of recurrent patellar dislocation and collect patient-reported outcomes scores (Knee injury and Osteoarthritis Outcome Score [KOOS], Norwich Patellar Instability score, Marx activity scale). Patients with 1-year minimum follow-up were included. Outcomes were quantified and the proportion of patients reaching a previously defined patient acceptable symptom state (PASS) for patellar instability was determined. Results: Sixty-one patients (42 female and 19 male) underwent MPFL reconstruction with peroneus longus allograft during the study period. Forty-six patients (76%) with 1-year minimum follow up were contacted at a mean of 3.5 years postoperative. The mean age at time of surgery was 22 ± 7.2 years. Patient-reported outcomes data were available in 34 patients. Mean KOOS subscale scores were as follows: Symptoms 83.2 ± 19.1, Pain 85.2 ± 17.6, Activities of Daily Living 89.9 ± 14.8, Sports 75 ± 26.2, and Quality of Life 72.6 ± 25.7. The mean Norwich Patellar Instability score was 14.9% ± 17.4%. The mean Marx activity score was 6.0 ± 5.2. No recurrent dislocations were noted during the study period. Sixty-three percent of patients who underwent isolated MPFL reconstruction met PASS thresholds in at least 4 of 5 KOOS subscales. Conclusions: The use of a peroneus longus allograft in MPFL reconstruction in conjunction with other indicated procedures results in a low re-dislocation risk and a high proportion of patients meeting PASS criteria for patient-reported outcome scores 3 to 4 years postoperatively. Level of Evidence: IV, case series.

11.
Ann Biomed Eng ; 51(7): 1616-1626, 2023 Jul.
Article in English | MEDLINE | ID: mdl-36913085

ABSTRACT

The biomechanics and efficacy of personal protective equipment in mitigating injuries from blast overpressure remain unclear. The objectives of this study were to define intrathoracic pressures in response to blast wave (BW) exposure and biomechanically evaluate a soft-armor vest (SA) at diminishing these perturbations. Male Sprague-Dawley rats were instrumented with pressure sensors in the thorax and were exposed laterally to multiple exposures ranging from 33 to 108 kPa BW with SA and without SA. There were significant increases in rise time, peak negative pressure, and negative impulse in the thoracic cavity compared to the BW. Esophageal measurements were increased to a greater extent when compared to the carotid and the BW for all parameters (except positive impulse, which decreased). SA minimally altered the pressure parameters and energy content. This study establishes the relationship of external blast flow conditions and intra-body biomechanical responses in the thoracic cavity of rodents with and without SA.


Subject(s)
Blast Injuries , Rats , Animals , Male , Rodentia , Rats, Sprague-Dawley , Biomechanical Phenomena , Explosions
12.
Brain Sci ; 12(10)2022 Oct 04.
Article in English | MEDLINE | ID: mdl-36291274

ABSTRACT

Blast-induced traumatic brain injury (bTBI) frequently results in sleep-wake disturbances. However, limited studies have investigated the molecular signaling mechanisms underlying these sleep disturbances, and potentially efficacious therapies are lacking. We investigated the levels of melatonin and genes involved in melatonin synthesis pathway in the pineal glands of Sprague Dawley rats exposed to single and tightly coupled repeated blasts during the night and daytime. Rats were exposed to single and tightly coupled repeated blasts using an advanced blast simulator. The plasma, cerebrospinal fluid (CSF), and pineal gland were collected at 6 h, 24 h, or 1 month postblast at two different time points: one during the day (1000 h) and one at night (2200 h). Differential expressions of genes involved in pineal melatonin synthesis were quantified using quantitative real-time polymerase chain reaction (qRT-PCR). Plasma and CSF melatonin levels were assessed using a commercial melatonin ELISA kit. The plasma and CSF melatonin levels showed statistically significant decreases at 6 h and 24 h in the blast-exposed rats euthanized in the night (in dim light), with no significant alterations noted in rats euthanized in the morning (daylight) at all three-time points. Blast-exposed rats showed statistically significant decreases in Tph1, Aanat, Asmt, and Mtnr1b mRNA levels, along with increased Tph2 mRNA, in the pineal gland samples collected at night at 6 h and 24 h. No significant changes in the mRNA levels of these genes were noted at 1 month. These findings imply that the melatonin circadian rhythm is disrupted following blast exposure, which may be a factor in the sleep disturbances that blast victims frequently experience.

13.
Knee ; 38: 178-183, 2022 Oct.
Article in English | MEDLINE | ID: mdl-36063612

ABSTRACT

BACKGROUND: Rehabilitation protocols following medial patellofemoral ligament (MPFL) reconstruction were historically restrictive, with patients often immobilized and/or given weightbearing restrictions. However, more recently published protocols have been more aggressive. We compared patient-reported outcomes and recurrent dislocation risk between patients treated with a restrictive rehabilitation program (early post-operative bracing and weightbearing restrictions) and an accelerated rehabilitation protocol (no post-operative bracing or weightbearing restrictions) following MPFL reconstruction. METHODS: Patients who underwent isolated MPFL reconstruction at an academic center between 2008 and 2016 were identified. Patient demographics, anatomical measurements, surgical details, and outcomes were collected. During this period, the rehabilitation protocol at the center transitioned from a restrictive to an accelerated rehabilitation protocol. Failure risk and patient-reported outcomes were compared based on rehabilitation protocol. RESULTS: Of the163 isolated MPFL reconstructions performed during the study period, 123 (75%) were available for minimum one-year follow up at a mean of 4.0 years post-operative. Overall, 53 knees (43%) underwent the accelerated rehabilitation protocol and the remaining 70 knees (57%) underwent the restrictive protocol. There were 3 recurrent dislocations during the study period (2.4%), all of which occurred in the restrictive rehabilitation group. Multiple linear regression demonstrated that being in the accelerated rehabilitation group was not associated with poorer Knee injury and Osteoarthritis Outcome Score (KOOS) subscales controlling for age, sex, body mass index, Caton-Deschamps Index, tibial tubercle-trochlear groove distance, sulcus angle, MPFL graft choice, and length of follow-up. CONCLUSION: An accelerated rehabilitation protocol without immobilization or weightbearing restrictions does not increase risk of recurrent patellar dislocation or poorer patient-reported outcome following isolated MPFL reconstruction.


Subject(s)
Joint Dislocations , Joint Instability , Patellar Dislocation , Patellofemoral Joint , Humans , Joint Instability/etiology , Joint Instability/surgery , Ligaments, Articular/surgery , Patellar Dislocation/surgery , Patellofemoral Joint/injuries , Patellofemoral Joint/surgery , Recurrence
14.
Science ; 377(6613): eabl3921, 2022 09 23.
Article in English | MEDLINE | ID: mdl-36137018

ABSTRACT

The vertebrate intestine forms by asymmetric gut rotation and elongation, and errors cause lethal obstructions in human infants. Rotation begins with tissue deformation of the dorsal mesentery, which is dependent on left-sided expression of the Paired-like transcription factor Pitx2. The conserved morphogen Nodal induces asymmetric Pitx2 to govern embryonic laterality, but organ-level regulation of Pitx2 during gut asymmetry remains unknown. We found Nodal to be dispensable for Pitx2 expression during mesentery deformation. Intestinal rotation instead required a mechanosensitive latent transforming growth factor-ß (TGFß), tuning a second wave of Pitx2 that induced reciprocal tissue stiffness in the left mesentery as mechanical feedback with the right side. This signaling regulator, an accelerator (right) and brake (left), combines biochemical and biomechanical inputs to break gut morphological symmetry and direct intestinal rotation.


Subject(s)
Gastrulation , Gene Expression Regulation, Developmental , Homeodomain Proteins , Intestines , Mechanotransduction, Cellular , Nodal Protein , Transcription Factors , Transforming Growth Factor beta , Animals , Chick Embryo , Gastrulation/genetics , Gastrulation/physiology , Homeodomain Proteins/genetics , Homeodomain Proteins/pharmacology , Intestines/embryology , Mechanotransduction, Cellular/genetics , Mechanotransduction, Cellular/physiology , Mice , Nodal Protein/genetics , Transcription Factors/genetics , Transcription Factors/pharmacology , Transforming Growth Factor beta/metabolism , Homeobox Protein PITX2
15.
Sci Rep ; 12(1): 14605, 2022 08 26.
Article in English | MEDLINE | ID: mdl-36028539

ABSTRACT

Animal studies provide valuable insights on how the interaction of blast waves with the head may injure the brain. However, there is no acceptable methodology to scale the findings from animals to humans. Here, we propose an experimental/computational approach to project observed blast-induced molecular changes in the rat brain to the human brain. Using a shock tube, we exposed rats to a range of blast overpressures (BOPs) and used a high-fidelity computational model of a rat head to correlate predicted biomechanical responses with measured changes in glial fibrillary acidic protein (GFAP) in rat brain tissues. Our analyses revealed correlates between model-predicted strain rate and measured GFAP changes in three brain regions. Using these correlates and a high-fidelity computational model of a human head, we determined the equivalent BOPs in rats and in humans that induced similar strain rates across the two species. We used the equivalent BOPs to project the measured GFAP changes in the rat brain to the human. Our results suggest that, relative to the rat, the human requires an exposure to a blast wave of a higher magnitude to elicit similar brain-tissue responses. Our proposed methodology could assist in the development of safety guidelines for blast exposure.


Subject(s)
Blast Injuries , Brain Injuries , Animals , Brain , Explosions , Head , Humans , Rats
16.
Hematol Oncol Clin North Am ; 36(4): 829-851, 2022 08.
Article in English | MEDLINE | ID: mdl-35778331

ABSTRACT

The field of gene therapy has experienced tremendous growth in the last decade ranging from improvements in the design of viral vectors for gene addition of therapeutic gene cassettes to the discovery of site-specific nucleases targeting transgenes to desired locations in the genome. Such advancements have not only enabled the development of disease models but also created opportunities for the development of tailored therapeutic approaches. There are 3 main methods of gene modification that can be used for the prevention or treatment of disease. This includes viral vector-mediated gene therapy to supply or bypass a missing/defective gene, gene editing enabled by programmable nucleases to create sequence-specific alterations in the genome, and gene silencing to reduce the expression of a gene or genes. These gene-modification platforms can be delivered either in vivo, for which the therapy is injected directed into a patient's body, or ex vivo, in which cells are harvested from a patient and modified in a laboratory setting, and then returned to the patient.


Subject(s)
Gene Editing , Genetic Therapy , Endonucleases/metabolism , Genetic Vectors/genetics , Humans , Transgenes
17.
Orthop J Sports Med ; 10(6): 23259671221104414, 2022 Jun.
Article in English | MEDLINE | ID: mdl-35783469

ABSTRACT

Background: Patella alta and elevated tibial tubercle-trochlear groove (TT-TG) distance can predispose patients to lateral patellar dislocations and recurrent instability. Their influence on patient-reported outcomes (PROs) after medial patellofemoral ligament (MPFL) reconstruction is less clear. Hypothesis: We hypothesized that neither moderately increased TT-TG distance nor patella alta would negatively affect PROs after isolated MPFL reconstruction in patients with relatively normal patellar tracking (no large J-sign). Study Design: Cohort study; Level of evidence, 3. Methods: We identified patients who underwent isolated MPFL reconstruction at a single institution between 2008 and 2016. The decision to perform an isolated MPFL reconstruction was at the discretion of the operating surgeon but was not performed in the setting of a large J-sign. Patient characteristics and surgical details were collected, and patients completed the Norwich Patellar Instability Score, Knee injury and Osteoarthritis Outcome Score, and Marx activity score. Patellar height (Caton-Deschamps Index [CDI]) was assessed on preoperative lateral radiographs, and TT-TG distance was measured on preoperative axial magnetic resonance imaging (MRI) scans. Patients were grouped based on CDI and TT-TG distance, and outcomes were compared. Linear regression modeling was performed to determine whether patella alta or elevated TT-TG distance was associated with poorer PRO scores. Results: Of 165 knees in 152 patients who underwent isolated MPFL reconstruction, 115 patients (125 knees; 76%) with minimum 1-year follow-up were contacted at a mean of 5.2 years after surgery. Recurrent dislocation occurred in 5 of 125 knees (4%). Preoperative radiographs were available in 111 knees (89%), and preoperative MRI scans were available in 89 knees (71%). Mean CDI was 1.13, and 35% had a CDI ≥1.20. Mean TT-TG distance was 17.5 mm, and 26% had a TT-TG distance >20 mm. After adjusting for patient age, sex, body mass index, and graft choice, we observed that neither patella alta nor elevated TT-TG distance were associated with poorer PROs. Conclusion: Isolated MPFL reconstruction in the setting of moderately elevated TT-TG distance or patella alta was not associated with worse PROs in this cohort with relatively normal patellar tracking (no large J-sign).

18.
Front Bioeng Biotechnol ; 10: 821169, 2022.
Article in English | MEDLINE | ID: mdl-35392409

ABSTRACT

Explosive devices, either conventional or improvised, are common sources of injuries during combat, civil unrest, and terror attacks, resulting in trauma from exposure to blast. A blast wave (BW), a near-instantaneous rise in pressure followed by a negative pressure, propagates through the body in milliseconds and can affect physiology for days/months after exposure. Epidemiological data show that blast-related casualties result in significantly higher susceptibility to wound infections, suggesting long-lasting immune modulatory effects from blast exposure. The mechanisms involved in BW-induced immune changes are poorly understood. We evaluated the effects of BW on the immune system using an established murine model. Animals were exposed to BWs (using an Advanced Blast Simulator), followed by longitudinally sampling for 14 days. Blood, bone marrow, and spleen were analyzed for changes in the 1) complete blood count (CBC), and 2) composition of bone marrow cells (BMC) and splenocytes, and 3) concentrations of systemic cytokines/chemokines. Our data demonstrate that BW results in transient bone marrow failure and long-term changes in the frequency and profile of progenitor cell populations. Viability progressively decreased in hematopoietic stem cells and pluripotent progenitor cells. Significant decrease of CD4+ T cells in the spleen indicates reduced functionality of adaptive immune system. Dynamic changes in the concentrations of several cytokines and chemokines such as IL-1α and IL-17 occurred potentially contributing to dysregulation of immune response after trauma. This work lays the foundation for identifying the potential mechanisms behind BW's immunosuppressive effects to inform the recognition of this compromised status is crucial for the development of therapeutic interventions for infections to reduce recovery time of wounded patients injured by explosive devices.

19.
Mol Ther Nucleic Acids ; 27: 505-516, 2022 Mar 08.
Article in English | MEDLINE | ID: mdl-35036061

ABSTRACT

Gene editing utilizing homology-directed repair has advanced significantly for many monogenic diseases of the hematopoietic system in recent years but has also been hindered by decreases between in vitro and in vivo gene integration rates. Homology-directed repair occurs primarily in the S/G2 phases of the cell cycle, whereas long-term engrafting hematopoietic stem cells are typically quiescent. Alternative methods for a targeted integration have been proposed including homology-independent targeted integration and precise integration into target chromosome, which utilize non-homologous end joining and microhomology-mediated end joining, respectively. Non-homologous end joining occurs throughout the cell cycle, while microhomology-mediated end joining occurs predominantly in the S phase. We compared these pathways for the integration of a corrective DNA cassette at the Bruton's tyrosine kinase gene for the treatment of X-linked agammaglobulinemia. Homology-directed repair generated the most integration in K562 cells; however, synchronizing cells into G1 resulted in the highest integration rates with homology-independent targeted integration. Only homology-directed repair produced seamless junctions, making it optimal for targets where insertions and deletions are impermissible. Bulk CD34+ cells were best edited by homology-directed repair and precise integration into the target chromosome, while sorted hematopoietic stem cells contained similar integration rates using all corrective donors.

20.
Front Neurol ; 12: 746370, 2021.
Article in English | MEDLINE | ID: mdl-34712199

ABSTRACT

Previous findings have indicated that pain relieving medications such as opioids and non-steroidal anti-inflammatory drugs (NSAIDs) may be neuroprotective after traumatic brain injury in rodents, but only limited studies have been performed in a blast-induced traumatic brain injury (bTBI) model. In addition, many pre-clinical TBI studies performed in rodents did not use analgesics due to the possibility of neuroprotection or other changes in cognitive, behavioral, and pathology outcomes. To examine this in a pre-clinical setting, we examined the neurobehavioral changes in rats given a single pre-blast dose of meloxicam, buprenorphine, or no pain relieving medication and exposed to tightly-coupled repeated blasts in an advanced blast simulator and evaluated neurobehavioral functions up to 28 days post-blast. A 16.7% mortality rate was recorded in the rats treated with buprenorphine, which might be attributed to the physiologically depressive side effects of buprenorphine in combination with isoflurane anesthesia and acute brain injury. Rats given buprenorphine, but not meloxicam, took more time to recover from the isoflurane anesthesia given just before blast. We found that treatment with meloxicam protected repeated blast-exposed rats from vestibulomotor dysfunctions up to day 14, but by day 28 the protective effects had receded. Both pain relieving medications seemed to promote short-term memory deficits in blast-exposed animals, whereas vehicle-treated blast-exposed animals showed only a non-significant trend toward worsening short-term memory by day 27. Open field exploratory behavior results showed that blast exposed rats treated with meloxicam engaged in significantly more locomotor activities and possibly a lesser degree of responses thought to reflect anxiety and depressive-like behaviors than any of the other groups. Rats treated with analgesics to alleviate possible pain from the blast ate more than their counterparts that were not treated with analgesics, which supports that both analgesics were effective in alleviating some of the discomfort that these rats potentially experienced post-blast injury. These results suggest that meloxicam and, to a lesser extent buprenorphine alter a variety of neurobehavioral functions in a rat bTBI model and, because of their impact on these neurobehavioral changes, may be less than ideal analgesic agents for pre-clinical studies evaluating these neurobehavioral responses after TBI.

SELECTION OF CITATIONS
SEARCH DETAIL
...