850 results on '"Controlled cortical impact"'
Search Results
2. Delayed-and-abbreviated environmental enrichment after traumatic brain injury confers neurobehavioral benefits similar to immediate-and-continuous exposure
- Author
-
Bittner, Rachel A., Greene, Anna M., Leary, Jacob B., Donald, Hailey M., Capeci, Haley E., Moschonas, Eleni H., Cheng, Jeffrey P., Bondi, Corina O., and Kline, Anthony E.
- Published
- 2025
- Full Text
- View/download PDF
3. Sustained attention performance deficits in the three-choice serial reaction time task in male and female rats after experimental brain trauma
- Author
-
Kutash, Lindsay A., Moschonas, Eleni H., O'Neil, Darik A., Craine, Timothy J., Iouchmanov, Anna L., Sunleaf, Carlson R., Nicholas, Melissa A., Grobengieser, Katherine O., Patel, Aarti K., Toader, Mihaela, Ranellone, Tyler S., Rennerfeldt, Piper L., Cheng, Jeffrey P., Race, Nicholas S., Kline, Anthony E., and Bondi, Corina O.
- Published
- 2023
- Full Text
- View/download PDF
4. Spatial Measurement and Inhibition of Calpain Activity in Traumatic Brain Injury with an Activity-Based Nanotheranostic Platform.
- Author
-
Madias, Marianne, Stessman, Lilyane, Warlof, Sophia, Kudryashev, Julia, and Kwon, Ester
- Subjects
TUNEL ,activity-based nanosensor ,calpastatin ,controlled cortical impact ,polyethylene glycol ,Calpain ,Brain Injuries ,Traumatic ,Animals ,Mice ,Male ,Mice ,Inbred C57BL ,Peptides ,Humans - Abstract
Traumatic brain injury (TBI) is a major public health concern that can result in long-term neurological impairments. Calpain is a calcium-dependent cysteine protease that is activated within minutes after TBI, and sustained calpain activation is known to contribute to neurodegeneration and blood-brain barrier dysregulation. Based on its role in disease progression, calpain inhibition has been identified as a promising therapeutic target. Efforts to develop therapeutics for calpain inhibition would benefit from the ability to measure calpain activity with spatial precision within the injured tissue. In this work, we designed an activity-based nanotheranostic (ABNT) that can both sense and inhibit calpain activity in TBI. To sense calpain activity, we incorporated a peptide substrate of calpain flanked by a fluorophore/quencher pair. To inhibit calpain activity, we incorporated calpastatin peptide, an endogenous inhibitor of calpain. Both sensor and inhibitor peptides were scaffolded onto a polymeric nanoscaffold to create our ABNT. We show that in the presence of recombinant calpain, our ABNT construct is able to sense and inhibit calpain activity. In a mouse model of TBI, systemically administered ABNT can access perilesional brain tissue through passive accumulation and inhibit calpain activity in the cortex and hippocampus. In an analysis of cellular calpain activity, we observe the ABNT-mediated inhibition of calpain activity in neurons, endothelial cells, and microglia of the cortex. In a comparison of neuronal calpain activity by brain structure, we observe greater ABNT-mediated inhibition of calpain activity in cortical neurons compared to that in hippocampal neurons. Furthermore, we found that apoptosis was dependent on both calpain inhibition and brain structure. We present a theranostic platform that can be used to understand the regional and cell-specific therapeutic inhibition of calpain activity to help inform drug design for TBI.
- Published
- 2024
5. Administration of monophosphoryl lipid A shortly after traumatic brain injury blocks the following spatial and avoidance memory loss and neuroinflammation.
- Author
-
Hooshmand, Maryam, Sadeghi, Mohammad Reza, Asoodeh, Ahmad, Pourbadie, Hamid Gholami, Mehni, Mahbobeh Kamrani, and Sayyah, Mohamad
- Subjects
- *
MEMORY disorders , *MEMORY loss , *SPATIAL memory , *BRAIN injuries , *CEREBRAL ventricles , *SCOPOLAMINE - Abstract
Traumatic brain injury (TBI) frequently leads to cognitive impairments. The toll-like receptor 4 (TLR4) ligand, Monophosphoryl lipid A (MPL), has shown promise in modulating neuroinflammatory responses after TBI. We investigated the effects of MPL on spatial memory, passive avoidance memory, neuronal survival, and inflammatory/anti-inflammatory cytokines in rat brain following mild-to-moderate TBI. Rats underwent a learning period in the Morris water maze and shuttle box, followed by TBI induction by controlled cortical impact. MPL was administered into the cerebral ventricle 20 min after TBI. Spatial memory was assessed 7 and 28 days later. Passive avoidance memory was assessed 2 and 6 days after TBI. MPL significantly improved the spatial memory deficit at 7 days but not 28 days after TBI. It also improved impairment of the avoidance memory at both 2 and 6 days after TBI. MPL prohibited the TBI-induced TNF-α increase and IL-10 decrease in the injured region at 7 days post-TBI period. MPL prevented the neuronal loss induced by TBI in the hippocampus. A single administration of MPL shortly after TBI alleviates short-term memory deficits, through anti-inflammatory and anti-cell loss activities. Repeated MPL administration may also inhibit the long-term memory deficits after TBI. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
6. Metabolic-driven analytics of traumatic brain injury and neuroprotection by ethyl pyruvate.
- Author
-
Golovachev, Nikita, Siebold, Lorraine, Sutton, Richard L., Ghavim, Sima, Harris, Neil G., and Bartnik-Olson, Brenda
- Subjects
- *
BRAIN injuries , *PRINCIPAL components analysis , *METABOLOMICS , *RANDOM forest algorithms ,BRAIN metabolism - Abstract
Background: Research on traumatic brain injury (TBI) highlights the significance of counteracting its metabolic impact via exogenous fuels to support metabolism and diminish cellular damage. While ethyl pyruvate (EP) treatment shows promise in normalizing cellular metabolism and providing neuroprotection, there is a gap in understanding the precise metabolic pathways involved. Metabolomic analysis of the acute post-injury metabolic effects, with and without EP treatment, aims to deepen our knowledge by identifying and comparing the metabolite profiles, thereby illuminating the injury's effects and EP's therapeutic potential. Methods: In the current study, an untargeted metabolomics approach was used to reveal brain metabolism changes in rats 24 h after a controlled cortical impact (CCI) injury, with or without EP treatment. Using principal component analysis (PCA), volcano plots, Random Forest and pathway analysis we differentiated the brain metabolomes of CCI and sham injured animals treated with saline (Veh) or EP, identifying key metabolites and pathways affected by injury. Additionally, the effect of EP on the non-injured brain was also explored. Results: PCA showed a clear separation of the four study groups (sham-Veh, CCI-Veh, sham-EP, CCI-EP) based on injury. Following CCI injury (CCI-Veh), 109 metabolites belonging to the amino acid, carbohydrate, lipid, nucleotide, and xenobiotic families exhibited a twofold change at 24 h compared to the sham-Veh group, with 93 of these significantly increasing and 16 significantly decreasing (p < 0.05). CCI animals were treated with EP (CCI-EP) showed only 5 metabolites in the carbohydrate, amino acids, peptides, nucleotides, lipids, and xenobiotics super families that exhibited a twofold change, compared to the CCI-Veh group (p < 0.05). In the non-injured brain, EP treatment (sham-EP) resulted in a twofold change in 6 metabolites within the amino acid, peptide, nucleotide, and lipid super families compared to saline treated sham animals (sham-Veh, p < 0.05). Conclusions: This study delineates the unique metabolic signatures resulting from a CCI injury and those related to EP treatment in both the injured and non-injured brain, underscoring the metabolic adaptations to brain injury and the effects of EP. Our analysis uncovers significant shifts in metabolites associated with inflammation, energy metabolism, and neuroprotection after injury, and demonstrates how EP intervention after injury alters metabolites associated with mitigating inflammation and oxidative damage. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
7. Evaluating the Efficacy of Chronic Galantamine on Sustained Attention and Cholinergic Neurotransmission in A Pre-Clinical Model of Traumatic Brain Injury.
- Author
-
Moschonas, Eleni H., Capeci, Haley E., Annas, Ellen M., Domyslawski, Veronica B., Steber, Jade A., Donald, Hailey M., Genkinger, Nicholas R., Rennerfeldt, Piper L., Bittner, Rachel A., Vozzella, Vincent J., Cheng, Jeffrey P., Kline, Anthony E., and Bondi, Corina O.
- Subjects
- *
REVERSE phase liquid chromatography , *NICOTINIC acetylcholine receptors , *BRAIN injuries , *ELECTROCHEMICAL sensors , *PREFRONTAL cortex , *CONTINUOUS performance test , *TOTAL body irradiation - Abstract
Cholinergic disruptions underlie attentional deficits following traumatic brain injury (TBI). Yet, drugs specifically targeting acetylcholinesterase (AChE) inhibition have yielded mixed outcomes. Therefore, we hypothesized that galantamine (GAL), a dual-action competitive AChE inhibitor and α7 nicotinic acetylcholine receptor (nAChR) positive allosteric modulator, provided chronically after injury, will attenuate TBI-induced deficits of sustained attention and enhance ACh efflux in the medial prefrontal cortex (mPFC), as assessed by in vivo microdialysis. In Experiment 1, adult male rats (n = 10–15/group) trained in the 3-choice serial reaction time (3-CSRT) test were randomly assigned to controlled cortical impact (CCI) or sham surgery and administered GAL (0.5, 2.0, or 5.0 mg/kg; i.p.) or saline vehicle (VEH; 1 mL/kg; i.p) beginning 24-h post-surgery and once daily thereafter for 27 days. Measures of sustained attention and distractibility were assessed on post-operative days 21–25 in the 3-CSRT, following which cortical lesion volume and basal forebrain cholinergic cells were quantified on day 27. In Experiment 2, adult male rats (n = 3–4/group) received a CCI and 24 h later administered (i.p.) one of the three doses of GAL or VEH for 21 days to quantify the dose-dependent effect of GAL on in vivo ACh efflux in the mPFC. Two weeks after the CCI, a guide cannula was implanted in the right mPFC. On post-surgery day 21, baseline and post-injection dialysate samples were collected in a temporally matched manner with the cohort undergoing behavior. ACh levels were analyzed using reverse phase high-performance liquid chromatography (HPLC) coupled to an electrochemical detector. Cortical lesion volume was quantified on day 22. The data were subjected to ANOVA, with repeated measures where appropriate, followed by Newman–Keuls post hoc analyses. All TBI groups displayed impaired sustained attention versus the pooled SHAM controls (p's < 0.05). Moreover, the highest dose of GAL (5.0 mg/kg) exacerbated attentional deficits relative to VEH and the two lower doses of GAL (p's < 0.05). TBI significantly reduced cholinergic cells in the right basal forebrain, regardless of treatment condition, versus SHAM (p < 0.05). In vivo microdialysis revealed no differences in basal ACh in the mPFC; however, GAL (5.0 mg/kg) significantly increased ACh efflux 30 min following injection compared to the VEH and the other GAL (0.5 and 2.0 mg/kg) treated groups (p's < 0.05). In both experiments, there were no differences in cortical lesion volume across treatment groups (p's > 0.05). In summary, albeit the higher dose of GAL increased ACh release, it did not improve measures of sustained attention or histopathological markers, thereby partially supporting the hypothesis and providing the impetus for further investigations into alternative cholinergic pharmacotherapies such as nAChR positive allosteric modulators. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
8. The Immune Response in Two Models of Traumatic Injury of the Immature Brain.
- Author
-
Al-Khateeb, Zahra F., Henson, Siân M., Tremoleda, Jordi L., and Michael-Titus, Adina T.
- Subjects
- *
BRAIN injuries , *MYELOID cells , *KILLER cells , *NATURAL immunity , *BLOOD cells - Abstract
Traumatic brain injury (TBI) can cause major disability and increases the risk of neurodegeneration. Post-TBI, there is infiltration of peripheral myeloid and lymphoid cells; there is limited information on the peripheral immune response post-TBI in the immature brain—where injury may interfere with neurodevelopment. We carried out two injury types in juvenile mice: invasive TBI with a controlled cortical impact (CCI) and repetitive mild TBI (rmTBI) using weight drop injury and analysed the response at 5- and 35-days post-injury. In the two models, we detected the brain infiltration of immune cells (e.g., neutrophils, monocytes, dendritic cells, CD4+ T cells, and NK cells). There were increases in macrophages, neutrophils, and dendritic cells in the spleen, increases in dendritic cells in blood, and increases in CD8+ T cells and B cells in lymph nodes. These results indicate a complex peripheral immune response post-TBI in the immature brain, with differences between an invasive injury and a repetitive mild injury. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
9. Temporal-Specific Sex and Injury-Dependent Changes on Neurogranin-Associated Synaptic Signaling After Controlled Cortical Impact in Rats.
- Author
-
Svirsky, Sarah E., Henchir, Jeremy, Li, Youming, Carlson, Shaun W., and Dixon, C. Edward
- Abstract
Extensive effort has been made to study the role of synaptic deficits in cognitive impairment after traumatic brain injury (TBI). Neurogranin (Ng) is a calcium-sensitive calmodulin (CaM)–binding protein essential for Ca
2+ /CaM-dependent kinase II (CaMKII) autophosphorylation which subsequently modulates synaptic plasticity. Given the loss of Ng expression after injury, additional research is warranted to discern changes in hippocampal post-synaptic signaling after TBI. Under isoflurane anesthesia, adult, male and female Sprague–Dawley rats received a sham/control or controlled cortical impact (CCI) injury. Ipsilateral hippocampal synaptosomes were isolated at 24 h and 1, 2, and 4 weeks post-injury, and western blot was used to evaluate protein expression of Ng-associated signaling proteins. Non-parametric Mann–Whitney tests were used to determine significance of injury for each sex at each time point. There were significant changes in the hippocampal synaptic expression of Ng and associated synaptic proteins such as phosphorylated Ng, CaMKII, and CaM up to 4 weeks post-CCI, demonstrating TBI alters hippocampal post-synaptic signaling. This study furthers our understanding of mechanisms of cognitive dysfunction within the synapse sub-acutely after TBI. [ABSTRACT FROM AUTHOR]- Published
- 2024
- Full Text
- View/download PDF
10. Infusible Extracellular Matrix Biomaterial Promotes Vascular Integrity and Modulates the Inflammatory Response in Acute Traumatic Brain Injury.
- Author
-
Diaz, Miranda, Kandell, Rebecca, Wu, Jason, Chen, Alexander, Christman, Karen, and Kwon, Ester
- Subjects
biomaterials ,controlled cortical impact ,decellularized extracellular matrix ,hydrogels ,traumatic brain injury ,vascular permeability ,Humans ,Mice ,Animals ,Endothelial Cells ,Brain Injuries ,Brain Injuries ,Traumatic ,Brain ,Blood-Brain Barrier ,Disease Models ,Animal - Abstract
Traumatic brain injury (TBI) affects millions of people each year and, in many cases, results in long-term disabilities. Once a TBI has occurred, there is a significant breakdown of the blood-brain barrier resulting in increased vascular permeability and progression of the injury. In this study, the use of an infusible extracellular matrix-derived biomaterial (iECM) for its ability to reduce vascular permeability and modulate gene expression in the injured brain is investigated. First, the pharmacokinetics of iECM administration in a mouse model of TBI is characterized, and the robust accumulation of iECM at the site of injury is demonstrated. Next, it is shown that iECM administration after injury can reduce the extravasation of molecules into the brain, and in vitro, iECM increases trans-endothelial electrical resistance across a monolayer of TNFα-stimulated endothelial cells. In gene expression analysis of brain tissue, iECM induces changes that are indicative of downregulation of the proinflammatory response 1-day post-injury/treatment and neuroprotection at 5 days post-injury/treatment. Therefore, iECM shows potential as a treatment for TBI.
- Published
- 2023
11. Modelling lung infection with Klebsiella pneumoniae after murine traumatic brain injury
- Author
-
Ali Shad, Sarah S. J. Rewell, Matthew Macowan, Natasha Gandasasmita, Jiping Wang, Ke Chen, Ben Marsland, Terence J. O’Brien, Jian Li, and Bridgette D. Semple
- Subjects
Bacteria ,Pneumonia ,Hospital-acquired infection ,Inflammation ,Controlled cortical impact ,Neurology. Diseases of the nervous system ,RC346-429 - Abstract
Abstract Pneumonia is a common comorbidity in patients with severe traumatic brain injury (TBI), and is associated with increased morbidity and mortality. In this study, we established a model of intratracheal Klebsiella pneumoniae administration in young adult male and female mice, at 4 days following an experimental TBI, to investigate how K. pneumoniae infection influences acute post-TBI outcomes. A dose-response curve determined the optimal dose of K. pneumoniae for inoculation (1 x 10^6 colony forming units), and administration at 4 days post-TBI resulted in transient body weight loss and sickness behaviors (hypoactivity and acute dyspnea). K. pneumoniae infection led to an increase in pro-inflammatory cytokines in serum and bronchoalveolar lavage fluid at 24 h post-infection, in both TBI and sham (uninjured) mice. By 7 days, when myeloperoxidase + neutrophil numbers had returned to baseline in all groups, lung histopathology was observed with an increase in airspace size in TBI + K. pneumoniae mice compared to TBI + vehicle mice. In the brain, increased neuroinflammatory gene expression was observed acutely in response to TBI, with an exacerbated increase in Ccl2 and Hmox1 in TBI + K. pneumoniae mice compared to either TBI or K. pneumoniae alone. However, the presence of neuroinflammatory immune cells in the injured brain, and the extent of damage to cortical and hippocampal brain tissue, was comparable between K. pneumoniae and vehicle-treated mice by 7 days. Examination of the fecal microbiome across a time course did not reveal any pronounced effects of either injury or K. pneumoniae on bacterial diversity or abundance. Together, these findings demonstrate that K. pneumoniae lung infection after TBI induces an acute and transient inflammatory response, primarily localized to the lungs with some systemic effects. However, this infection had minimal impact on secondary injury processes in the brain following TBI. Future studies are needed to evaluate the potential longer-term consequences of this dual-hit insult.
- Published
- 2024
- Full Text
- View/download PDF
12. Behavioral Interventions Can Improve Brain Injury-Induced Deficits in Behavioral Flexibility and Impulsivity Linked to Impaired Reward-Feedback Beta Oscillations.
- Author
-
Koloski, Miranda F., O'Hearn, Christopher M., Frankot, Michelle, Giesler, Lauren P., Ramanathan, Dhakshin S., and Vonder Haar, Cole
- Subjects
- *
LABORATORY rats , *NEUROPSYCHOLOGICAL rehabilitation , *IMPULSIVE personality , *BRAIN injuries , *PREFRONTAL cortex , *PHYSIOLOGY - Abstract
Traumatic brain injury (TBI) affects a large population, resulting in severe cognitive impairments. Although cognitive rehabilitation is an accepted treatment for some deficits, studies in patients are limited in ability to probe physiological and behavioral mechanisms. Therefore, animal models are needed to optimize strategies. Frontal TBI in a rat model results in robust and replicable cognitive deficits, making this an ideal candidate for investigating various behavioral interventions. In this study, we report three distinct frontal TBI experiments assessing behavior well into the chronic post-injury period using male Long-Evans rats. First, we evaluated the impact of frontal injury on local field potentials recorded simultaneously from 12 brain regions during a probabilistic reversal learning (PbR) task. Next, a set of rats were tested on a similar PbR task or an impulsivity task (differential reinforcement of low-rate behavior [DRL]) and half received salient cues associated with reinforcement contingencies to encourage engagement in the target behavior. After intervention on the PbR task, brains were stained for markers of activity. On the DRL task, cue relevance was decoupled from outcomes to determine if beneficial effects persisted on impulsive behavior. TBI decreased the ability to detect reinforced outcomes; this was evident in task performance and reward-feedback signals occurring at beta frequencies in lateral orbitofrontal cortex (OFC) and associated frontostriatal regions. The behavioral intervention improved flexibility and increased OFC activity. Intervention also reduced impulsivity, even after cues were decoupled, which was partially mediated by improvements in timing behavior. The current study established a platform to begin investigating cognitive rehabilitation in rats and identified a strong role for dysfunctional OFC signaling in probabilistic learning after frontal TBI. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
13. Spatial lipidomics maps brain alterations associated with mild traumatic brain injury.
- Author
-
Leontyev, Dmitry, Pulliam, Alexis N., Ma, Xin, Gaul, David A., LaPlaca, Michelle C., Fernández, Facundo M., Zemaitis, Kevin J., and Burnum-Johnson, Kristin E.
- Subjects
- *
LIPIDOMICS , *BRAIN mapping , *BRAIN injuries , *HIPPOCAMPUS (Brain) , *NEUROINFLAMMATION , *OXIDATIVE stress - Abstract
Traumatic brain injury (TBI) is a global public health problem with 50-60 million incidents per year, most of which are considered mild (mTBI) and many of these repetitive (rmTBI). Despite their massive implications, the pathologies of mTBI and rmTBI are not fully understood, with a paucity of information on brain lipid dysregulation following mild injury event(s). To gain more insight on mTBI and rmTBI pathology, a non-targeted spatial lipidomics workflow utilizing high resolution mass spectrometry imaging was developed to map brain region-specific lipid alterations in rats following injury. Discriminant multivariate models were created for regions of interest including the hippocampus, cortex, and corpus callosum to pinpoint lipid species that differentiated between injured and sham animals. A multivariate model focused on the hippocampus region differentiated injured brain tissues with an area under the curve of 0.99 using only four lipid species. Lipid classes that were consistently discriminant included polyunsaturated fatty acid-containing phosphatidylcholines (PC), lysophosphatidylcholines (LPC), LPC-plasmalogens (LPC-P) and PC potassium adducts. Many of the polyunsaturated fatty acid-containing PC and LPC-P selected have never been previously reported as altered in mTBI. The observed lipid alterations indicate that neuroinflammation and oxidative stress are important pathologies that could serve to explain cognitive deficits associated with rmTBI. Therapeutics which target or attenuate these pathologies may be beneficial to limit persistent damage following a mild brain injury event. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
14. Modelling lung infection with Klebsiella pneumoniae after murine traumatic brain injury.
- Author
-
Shad, Ali, Rewell, Sarah S. J., Macowan, Matthew, Gandasasmita, Natasha, Wang, Jiping, Chen, Ke, Marsland, Ben, O'Brien, Terence J., Li, Jian, and Semple, Bridgette D.
- Subjects
BRAIN injuries ,KLEBSIELLA infections ,LUNG infections ,KLEBSIELLA pneumoniae ,PULMONARY eosinophilia ,BACTERIAL diversity - Abstract
Pneumonia is a common comorbidity in patients with severe traumatic brain injury (TBI), and is associated with increased morbidity and mortality. In this study, we established a model of intratracheal Klebsiella pneumoniae administration in young adult male and female mice, at 4 days following an experimental TBI, to investigate how K. pneumoniae infection influences acute post-TBI outcomes. A dose-response curve determined the optimal dose of K. pneumoniae for inoculation (1 x 10^6 colony forming units), and administration at 4 days post-TBI resulted in transient body weight loss and sickness behaviors (hypoactivity and acute dyspnea). K. pneumoniae infection led to an increase in pro-inflammatory cytokines in serum and bronchoalveolar lavage fluid at 24 h post-infection, in both TBI and sham (uninjured) mice. By 7 days, when myeloperoxidase + neutrophil numbers had returned to baseline in all groups, lung histopathology was observed with an increase in airspace size in TBI + K. pneumoniae mice compared to TBI + vehicle mice. In the brain, increased neuroinflammatory gene expression was observed acutely in response to TBI, with an exacerbated increase in Ccl2 and Hmox1 in TBI + K. pneumoniae mice compared to either TBI or K. pneumoniae alone. However, the presence of neuroinflammatory immune cells in the injured brain, and the extent of damage to cortical and hippocampal brain tissue, was comparable between K. pneumoniae and vehicle-treated mice by 7 days. Examination of the fecal microbiome across a time course did not reveal any pronounced effects of either injury or K. pneumoniae on bacterial diversity or abundance. Together, these findings demonstrate that K. pneumoniae lung infection after TBI induces an acute and transient inflammatory response, primarily localized to the lungs with some systemic effects. However, this infection had minimal impact on secondary injury processes in the brain following TBI. Future studies are needed to evaluate the potential longer-term consequences of this dual-hit insult. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
15. Endorepellin downregulation promotes angiogenesis after experimental traumatic brain injury.
- Author
-
Qian Zhang, Yao Jing, Qiuyuan Gong, Lin Cai, Ren Wang, Dianxu Yang, Liping Wang, Meijie Qu, Hao Chen, Yaohui Tang, Hengli Tian, Jun Ding, and Zhiming Xu
- Published
- 2024
- Full Text
- View/download PDF
16. Pediatric Traumatic Brain Injury: Models, Therapeutics, and Outcomes
- Author
-
Moschonas, Eleni H., Annas, Ellen M., Zamudio-Flores, Jonathan, Jarvis, Jessica M., Lajud, Naima, Bondi, Corina O., Kline, Anthony E., Verkhratsky, Alexej, Series Editor, Noble-Haeusslein, Linda J., editor, and Schnyer, David M., editor
- Published
- 2024
- Full Text
- View/download PDF
17. Cognition and Behavior in the Aging Brain Following TBI: Surveying the Preclinical Evidence
- Author
-
Race, Nicholas S., Moschonas, Eleni H., Kline, Anthony E., Bondi, Corina O., Verkhratsky, Alexej, Series Editor, Noble-Haeusslein, Linda J., editor, and Schnyer, David M., editor
- Published
- 2024
- Full Text
- View/download PDF
18. Murine Traumatic Brain Injury Model Comparison: Closed Head Injury Versus Controlled Cortical Impact.
- Author
-
Baucom, Matthew R., Price, Adam D., England, Lisa, Schuster, Rebecca M., Pritts, Timothy A., and Goodman, Michael D.
- Subjects
- *
BRAIN injuries , *TOTAL body irradiation , *GLIAL fibrillary acidic protein , *HEAD injuries , *SPECKLE interference , *CEREBRAL circulation - Abstract
Various murine models have been utilized to study TBI, including closed head injury (CHI) and controlled cortical impact (CCI), without direct comparison. The aim of our study was to evaluate these models to determine differences in neurological and behavioral outcomes postinjury. Male C57B/6 mice (9-10 wk) were separated into six groups including: untouched, sham craniotomy (4 mm), CCI 0.9 mm depth of impact, CCI 1.6 mm, CCI 2.2 mm, and CHI. CCI was performed using a 3 mm impact tip at a velocity of 5 m/s, dwell time of 250 ms, and depth as noted above. CHI was completed with a centered 400 g weight drop from 1 cm height. Mice were survived to 14-d (n = 5 per group) and 30-d (n = 5 per group) respectively for histological analysis of p-tau within the hippocampus. These mice underwent Morris Water Maze memory testing and Rotarod motor testing. Serum was collected from a separate cohort of mice (n = 5 per group) including untouched, isoflurane only, CCI 1.6 mm, CHI at 1, 4, 6, and 24 h for analysis of neuron specific enolase and glial fibrillary acidic protein (GFAP) via ELISA. Laser speckle contrast imaging was analyzed prior to and after impact in the CHI and CCI 1.6 mm groups. There were no significant differences in Morris Water Maze or Rotarod testing times between groups at 14- or 30-d. P-tau was significantly elevated in all groups except CCI 1.6 mm contralateral and CCI 2.2 mm ipsilateral compared to untouched mice at 30-d. P-tau was also significantly elevated in the CHI group at 30 d compared to CCI 1.6 mm contralateral and CCI 2.2 mm on both sides. GFAP was significantly increased in mice undergoing CHI (9959 ± 91 pg/mL) compared to CCI (2299 ± 1288 pg/mL), isoflurane only (133 ± 75 pg/mL), and sham (86 ± 58 pg/mL) at 1-h post TBI (P < 0.0001). There were no differences in serum neuron specific enolase levels between groups. Laser doppler imaging demonstrated similar decreases in cerebral blood flow between CHI and CCI; however, CCI mice had a reduction in blood flow with craniotomy only that did not significantly decrease further with impact. Based on our findings, CHI leads to increased serum GFAP levels and increased p-tau within the hippocampus at 30-d postinjury. While CCI allows the comparison of one cerebral hemisphere to the other, CHI may be a better model of TBI as it requires less technical expertise and has similar neurological outcomes in these murine models. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
19. ROCK2 regulates microglia proliferation and neuronal survival after traumatic brain injury.
- Author
-
Willis, Emily F., Kim, Seung Jae, Chen, Wei, Nyuydzefe, Melanie, MacDonald, Kelli P.A., Zanin-Zhorov, Alexandra, Ruitenberg, Marc J., and Vukovic, Jana
- Subjects
- *
BRAIN injuries , *MICROGLIA , *POPULATION dynamics - Abstract
• ROCK2 signalling mediates microglial proliferation in vitro and in vivo after TBI. • With resident microglia, ROCK2 inhibition modestly improves TBI outcomes. • Acute ROCK2 inhibition negatively interferes with microglial repopulation. • Acute ROCK2 inhibition annuls the beneficial effects of repopulating microglia. Traumatic brain injury (TBI) results in prolonged and non-resolving activation of microglia. Forced turnover of these cells during the acute phase of TBI aids recovery, but the cell-intrinsic pathways that underpin the pro-repair phenotype of these repopulating microglia remain unclear. Here, we show that selective targeting of ROCK2 with the small molecule inhibitor KD025 impairs the proliferative response of microglia after TBI as well as during genetically induced turnover of microglia. KD025 treatment abolished the substantial neuroprotective and cognitive benefits conferred by repopulating microglia, preventing these cells from replenishing the depleted niche during the early critical time window post-injury. Delaying KD025 treatment to the subacute phase of TBI allowed microglial repopulation to occur, but this did not enhance the benefits conferred by repopulating microglia. Taken together, our data indicate that ROCK2 mediates neuronal survival and microglial population dynamics after TBI, including the emergence of repopulating microglia with a pro-repair phenotype. ROCK2 is a novel mediatior of microglial proliferation with implications for neuroprotection after brain injury. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
20. Fecal microbiota transplantation unveils sex-specific differences in a controlled cortical impact injury mouse model.
- Author
-
Pasam, Tulasi and Dandekar, Manoj P.
- Subjects
FECAL microbiota transplantation ,CRASH injuries ,LABORATORY mice ,ANIMAL disease models ,BRAIN injuries ,REPERFUSION ,ALLOCATION of organs, tissues, etc. ,BACTEROIDES fragilis - Abstract
Introduction: Contusion type of traumatic brain injury (TBI) is a major cause of locomotor disability and mortality worldwide. While post-TBI deleterious consequences are influenced by gender and gut dysbiosis, the sex-specific importance of commensal gut microbiota is underexplored after TBI. In this study, we investigated the impact of controlled cortical impact (CCI) injury on gut microbiota signature in a sex-specific manner in mice. Methods: We depleted the gut microflora of male and female C57BL/6 mice using antibiotic treatment. Thereafter, male mice were colonized by the gut microbiota of female mice and vice versa, employing the fecal microbiota transplantation (FMT) method. CCI surgery was executed using a stereotaxic impactor (Impact One™). For the 16S rRNA gene amplicon study, fecal boli of mice were collected at 3 days post-CCI (dpi). Results and discussion: CCI-operated male and female mice exhibited a significant alteration in the genera of Akkermansia, Alistipes, Bacteroides, Clostridium, Lactobacillus, Prevotella, and Ruminococcus. At the species level, less abundance of Lactobacillus helveticus and Lactobacillus hamsteri was observed in female mice, implicating the importance of sex-specific bacteriotherapy in CCI-induced neurological deficits. FMT from female donor mice to male mice displayed an increase in genera of Alistipes, Lactobacillus, and Ruminococcus and species of Bacteroides acidifaciens and Ruminococcus gnavus. Female FMT-recipient mice from male donors showed an upsurge in the genus Lactobacillus and species of Lactobacillus helveticus, Lactobacillus hamsteri, and Prevotella copri. These results suggest that the post-CCI neurological complications may be influenced by the differential gut microbiota perturbation in male and female mice. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
21. Early Life Stress Negatively Impacts Spatial Learning Acquisition and Increases Hippocampal CA1 Microglial Activation After a Mild Traumatic Brain Injury in Adult Male Rats.
- Author
-
Salinas-García, Ana Fernanda, Roque, Angélica, Zamudio-Flores, Jonathan, Meléndez-Herrera, Esperanza, Kline, Anthony E., and Lajud, Naima
- Abstract
Early life stress (ELS) affects neurogenesis and spatial learning, and increases neuroinflammation after a pediatric mild traumatic brain injury (mTBI). Previous studies have shown that ELS has minimal effects in juveniles but shows age-dependent effects in adults. Hence, we aimed to evaluate the effects of ELS in adult male rats after an mTBI. Maternal separation for 180 min per day (MS180) during the first 21 post-natal (P) days was used as the ELS model. At P110, the rats were subjected to a mild controlled cortical impact injury (2.6 mm) or sham surgery. Spatial learning was evaluated in the Morris water maze (MWM) 14 days after surgery and both microglial activation and neurogenesis were quantified. The results indicate that MS180 + mTBI, but not control (CONT) + mTBI, rats show deficiencies in the acquisition of spatial learning. mTBI led to comparable increases in microglial activation in both the hilus and cortical regions for both groups. However, MS180 + mTBI rats exhibited a greater increase in microglial activation in the ipsilateral CA1 hippocampus subfield compared with CONT + mTBI. Interestingly, for the contralateral CA1 region, this effect was observed exclusively in MS180 + mTBI. ELS and mTBI independently caused a decrease in hippocampal neurogenesis and this effect was not increased further in MS180 + mTBI rats. The findings demonstrate that ELS and mTBI synergistically affect cognitive performance and neuroinflammation, thus supporting the hypothesis that increased inflammation resulting from the combination of ELS and mTBI could underlie the observed effects on learning. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
22. Traumatic Brain Injury Induces Nociceptin/Orphanin FQ and Nociceptin Opioid Peptide Receptor Expression within 24 Hours.
- Author
-
Al Yacoub, Omar N., Zhang, Yong, Patankar, Panini S., and Standifer, Kelly M.
- Subjects
- *
NOCICEPTIN , *BRAIN injuries , *PEPTIDE receptors , *OPIOID receptors , *GENE expression , *PEPTIDES - Abstract
Traumatic brain injury (TBI) is a major cause of mortality and disability around the world, for which no treatment has been found. Nociceptin/Orphanin FQ (N/OFQ) and the nociceptin opioid peptide (NOP) receptor are rapidly increased in response to fluid percussion, stab injury, and controlled cortical impact (CCI) TBI. TBI-induced upregulation of N/OFQ contributes to cerebrovascular impairment, increased excitotoxicity, and neurobehavioral deficits. Our objective was to identify changes in N/OFQ and NOP receptor peptide, protein, and mRNA relative to the expression of injury markers and extracellular regulated kinase (ERK) 24 h following mild (mTBI) and moderate TBI (ModTBI) in wildtype (WT) and NOP receptor-knockout (KO) rats. N/OFQ was quantified by radioimmunoassay, mRNA expression was assessed using real-time PCR and protein levels were determined by immunoblot analysis. This study revealed increased N/OFQ mRNA and peptide levels in the CSF and ipsilateral tissue of WT, but not KO, rats 24 h post-TBI; NOP receptor mRNA increased after ModTBI. Cofilin-1 activation increased in the brain tissue of WT but not KO rats, ERK activation increased in all rats following ModTBI; no changes in injury marker levels were noted in brain tissue at this time. In conclusion, this study elucidates transcriptional and translational changes in the N/OFQ-NOP receptor system relative to TBI-induced neurological deficits and initiation of signaling cascades that support the investigation of the NOP receptor as a therapeutic target for TBI. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
23. Targeting the Extracellular Matrix in Traumatic Brain Injury Increases Signal Generation from an Activity-Based Nanosensor
- Author
-
Kandell, Rebecca M, Kudryashev, Julia A, and Kwon, Ester J
- Subjects
Brain Disorders ,Physical Injury - Accidents and Adverse Effects ,Traumatic Brain Injury (TBI) ,Neurosciences ,Traumatic Head and Spine Injury ,Neurological ,Injuries and accidents ,Brain ,Brain Injuries ,Brain Injuries ,Traumatic ,Calpain ,Extracellular Matrix ,Humans ,hyaluronic acid ,controlled cortical impact ,calpain-1 ,peptides ,protease activity ,light sheet microscopy ,CUBIC ,Nanoscience & Nanotechnology - Abstract
Traumatic brain injury (TBI) is a critical public health concern and major contributor to death and long-term disability. After the initial trauma, a sustained secondary injury involving a complex continuum of pathophysiology unfolds, ultimately leading to the destruction of nervous tissue. One disease hallmark of TBI is ectopic protease activity, which can mediate cell death, extracellular matrix breakdown, and inflammation. We previously engineered a fluorogenic activity-based nanosensor for TBI (TBI-ABN) that passively accumulates in the injured brain across the disrupted vasculature and generates fluorescent signal in response to calpain-1 cleavage, thus enabling in situ visualization of TBI-associated calpain-1 protease activity. In this work, we hypothesized that actively targeting the extracellular matrix (ECM) of the injured brain would improve nanosensor accumulation in the injured brain beyond passive delivery alone and lead to increased nanosensor activation. We evaluated several peptides that bind exposed/enriched ECM constituents in the brain and discovered that nanomaterials modified with peptides that target hyaluronic acid (HA) displayed widespread distribution across the injury lesion, in particular colocalizing with perilesional and hippocampal neurons. Modifying TBI-ABN with HA-targeting peptide led to increases in activation in a ligand-valency-dependent manner, up to 6.6-fold in the injured cortex compared to a nontargeted nanosensor. This robust nanosensor activation enabled 3D visualization of injury-specific protease activity in a cleared and intact brain. In our work, we establish that targeting brain ECM with peptide ligands can be leveraged to improve the distribution and function of a bioresponsive imaging nanomaterial.
- Published
- 2021
24. Antipsychotic Drugs: The Antithesis to Neurorehabilitation in Models of Pre-Clinical Traumatic Brain Injury
- Author
-
Nicholas S. Race, Eleni H. Moschonas, Jeffrey P. Cheng, Corina O. Bondi, and Anthony E. Kline
- Subjects
antipsychotic drugs ,behavior ,cognition ,controlled cortical impact ,Morris water maze ,recovery ,Medical emergencies. Critical care. Intensive care. First aid ,RC86-88.9 - Abstract
Sixty-nine million traumatic brain injuries (TBIs) are reported worldwide each year, and, of those, close to 3 million occur in the United States. In addition to neurobehavioral and cognitive deficits, TBI induces other maladaptive behaviors, such as agitation and aggression, which must be managed for safe, accurate assessment and effective treatment of the patient. The use of antipsychotic drugs (APDs) in TBI is supported by some expert guidelines, which suggests that they are an important part of the pharmacological armamentarium to be used in the management of agitation. Despite the advantages of APDs after TBI, there are significant disadvantages that may not be fully appreciated clinically during decision making because of the lack of a readily available updated compendium. Hence, the aim of this review is to integrate the existing findings and present the current state of APD use in pre-clinical models of TBI. The studies discussed were identified through PubMed and the University of Pittsburgh Library System search strategies and reveal that APDs, particularly those with dopamine2 (D2) receptor antagonism, generally impair the recovery process in rodents of both sexes and, in some instances, attenuate the potential benefits of neurorehabilitation. We believe that the compilation of findings represented by this exhaustive review of pre-clinical TBI + APD models can serve as a convenient source for guiding informed decisions by critical care clinicians and physiatrists contemplating APD use for patients exhibiting agitation.
- Published
- 2023
- Full Text
- View/download PDF
25. Fecal microbiota transplantation unveils sex-specific differences in a controlled cortical impact injury mouse model
- Author
-
Tulasi Pasam and Manoj P. Dandekar
- Subjects
gut microbiome ,fecal microbiota transplantation ,traumatic brain injury ,probiotics ,controlled cortical impact ,Microbiology ,QR1-502 - Abstract
IntroductionContusion type of traumatic brain injury (TBI) is a major cause of locomotor disability and mortality worldwide. While post-TBI deleterious consequences are influenced by gender and gut dysbiosis, the sex-specific importance of commensal gut microbiota is underexplored after TBI. In this study, we investigated the impact of controlled cortical impact (CCI) injury on gut microbiota signature in a sex-specific manner in mice.MethodsWe depleted the gut microflora of male and female C57BL/6 mice using antibiotic treatment. Thereafter, male mice were colonized by the gut microbiota of female mice and vice versa, employing the fecal microbiota transplantation (FMT) method. CCI surgery was executed using a stereotaxic impactor (Impact One™). For the 16S rRNA gene amplicon study, fecal boli of mice were collected at 3 days post-CCI (dpi).Results and discussionCCI-operated male and female mice exhibited a significant alteration in the genera of Akkermansia, Alistipes, Bacteroides, Clostridium, Lactobacillus, Prevotella, and Ruminococcus. At the species level, less abundance of Lactobacillus helveticus and Lactobacillus hamsteri was observed in female mice, implicating the importance of sex-specific bacteriotherapy in CCI-induced neurological deficits. FMT from female donor mice to male mice displayed an increase in genera of Alistipes, Lactobacillus, and Ruminococcus and species of Bacteroides acidifaciens and Ruminococcus gnavus. Female FMT-recipient mice from male donors showed an upsurge in the genus Lactobacillus and species of Lactobacillus helveticus, Lactobacillus hamsteri, and Prevotella copri. These results suggest that the post-CCI neurological complications may be influenced by the differential gut microbiota perturbation in male and female mice.
- Published
- 2024
- Full Text
- View/download PDF
26. The Nociceptin/Orphanin FQ peptide receptor antagonist, SB-612111, improves cerebral blood flow in a rat model of traumatic brain injury.
- Author
-
Al Yacoub, Omar N., Tarantini, Stefano, Yong Zhang, Csiszar, Anna, and Standifer, Kelly M.
- Subjects
CEREBRAL circulation ,BRAIN injuries ,NOCICEPTIN ,PEPTIDE receptors ,SPECKLE interference ,SPECKLE interferometry ,NATALIZUMAB - Abstract
Traumatic brain injury (TBI) affects more than 2.5 million people in the U.S. each year and is the leading cause of death and disability in children and adults ages 1 to 44. Approximately 90% of TBI cases are classified as mild but may still lead to acute detrimental effects such as impaired cerebral blood flow (CBF) that result in prolonged impacts on brain function and quality of life in up to 15% of patients. We previously reported that nociceptin/orphanin FQ (N/OFQ) peptide (NOP) receptor antagonism reversed mild blast TBI-induced vestibulomotor deficits and prevented hypoxia. To explore mechanisms by which the NOP receptor-N/OFQ pathway modulates hypoxia and other TBI sequelae, the ability of the NOP antagonist, SB-612111 (SB), to reverse TBI-induced CBF and associated injury marker changes were tested in this study. Male Wistar rats randomly received sham craniotomy or craniotomy + TBI via controlled cortical impact. Injury severity was assessed after 1 h (modified neurological severity score (mNSS). Changes in CBF were assessed 2 h post-injury above the exposed cortex using laser speckle contrast imaging in response to the direct application of increasing concentrations of vehicle or SB (1, 10, and 100 µM) to the brain surface. TBI increased mNSS scores compared to baseline and confirmed mild TBI (mTBI) severity. CBF was significantly impaired on the ipsilateral side of the brain following mTBI, compared to contralateral side and to sham rats. SB dose-dependently improved CBF on the ipsilateral side after mTBI compared to SB effects on the respective ipsilateral side of sham rats but had no effect on contralateral CBF or in uninjured rats. N/OFQ levels increased in the cerebral spinal fluid (CSF) following mTBI, which correlated with the percent decrease in ipsilateral CBF. TBI also activated ERK and cofilin within 3 h post-TBI; ERK activation correlated with increased CSF N/OFQ. In conclusion, this study reveals a significant contribution of the N/OFQ-NOP receptor system to TBIinduced dysregulation of cerebral vasculature and suggests that the NOP receptor should be considered as a potential therapeutic target for TBI. [ABSTRACT FROM AUTHOR]
- Published
- 2023
- Full Text
- View/download PDF
27. Toll-Like Receptor 1/2 Postconditioning by the Ligand Pam3cys Tempers Posttraumatic Hyperexcitability, Neuroinflammation, and Microglial Response: A Potential Candidate for Posttraumatic Epilepsy
- Author
-
Khoshkroodian, Bahar, Javid, Hanieh, Pourbadie, Hamid Gholami, and Sayyah, Mohammad
- Published
- 2024
- Full Text
- View/download PDF
28. Differential Fecal Microbiome Dysbiosis after Equivalent Traumatic Brain Injury in Aged Versus Young Adult Mice
- Author
-
Davis, Booker T, Islam, Mecca BAR, Das, Promi, Gilbert, Jack A, Ho, Karen J, and Schwulst, Steven J
- Subjects
Microbiology ,Biological Sciences ,Traumatic Head and Spine Injury ,Aging ,Neurosciences ,Traumatic Brain Injury (TBI) ,Physical Injury - Accidents and Adverse Effects ,Brain Disorders ,Neurological ,Age ,Controlled Cortical Impact ,Dysbiosis ,Microbiome ,Trauma ,Traumatic Brain Injury - Abstract
Traumatic brain injury (TBI) has a bimodal age distribution with peak incidence at age 24 and age 65 with worse outcomes developing in aged populations. Few studies have specifically addressed age at the time of injury as an independent biologic variable in TBI-associated secondary pathology. Within the framework of our published work, identifying age related effects of TBI on neuropathology, cognition, memory and motor function we analyzed fecal pellets collected from young and aged TBI animals to assess for age-induced effects in TBI induced dysbiosis. In this follow up, work we hypothesized increased dysbiosis after TBI in aged (80-week-old, N=10) versus young (14-week-old, N=10) mice. C57BL/6 males received a sham incision or TBI via open-head controlled cortical impact. Fresh stool pellets were collected 1-day pre-TBI, then 1, 7, and 28-days post-TBI for 16S rRNA gene sequencing and taxonomic analysis. Data revealed an age induced increase in disease associated microbial species which were exacerbated by injury. Consistent with our hypothesis, aged mice demonstrated a high number of disease associated changes to the gut microbiome pre- and post-injury. Our data suggest divergent microbiome phenotypes in injury between young and aged reflecting a previously unknown interaction between age, TBI, and the gut-brain axis implying the need for different treatment strategies.
- Published
- 2021
29. HIF-1α participates in secondary brain injury through regulating neuroinflammation
- Author
-
Xu Xiaojian, Yang Mengshi, Zhang Bin, Dong Jinqian, Zhuang Yuan, Ge Qianqian, Niu Fei, and Liu Baiyun
- Subjects
traumatic brain injury ,hif-1α ,hypoxia ,neuroinflammation ,apoptosis ,innate immune ,2-methoxyestradiol ,secondary brain injury ,gsea ,controlled cortical impact ,Neurosciences. Biological psychiatry. Neuropsychiatry ,RC321-571 - Abstract
A deeper understanding of the underlying biological mechanisms of secondary brain injury induced by traumatic brain injury (TBI) will greatly advance the development of effective treatments for patients with TBI. Hypoxia-inducible factor-1 alpha (HIF-1α) is a central regulator of cellular response to hypoxia. In addition, growing evidence shows that HIF-1α plays the important role in TBI-induced changes in biological processes; however, detailed functional mechanisms are not completely known. The aim of the present work was to further explore HIF-1α-mediated events after TBI. To this end, next-generation sequencing, coupled with cellular and molecular analysis, was adopted to interrogate vulnerable events in a rat controlled cortical impact model of TBI. The results demonstrated that TBI induced accumulation of HIF-1α at the peri-injury site at 24 h post-injury, which was associated with neuronal loss. Moreover, gene set enrichment analysis unveiled that neuroinflammation, especially an innate inflammatory response, was significantly evoked by TBI, which could be attenuated by the inhibition of HIF-1α. Furthermore, the inhibition of HIF-1α could mitigate the activation of microglia and astrocytes. Taken together, all these data implied that HIF-1α might contribute to secondary brain injury through regulating neuroinflammation.
- Published
- 2023
- Full Text
- View/download PDF
30. Chronic juvenile stress exacerbates neurobehavioral dysfunction and neuroinflammation following traumatic brain injury in adult mice
- Author
-
Sung-Jin Park, Hyun-Jeong Park, Backyoun Kim, Young-Min Kim, Suk-Woo Lee, and Hoon Kim
- Subjects
unpredictable chronic mild stress ,controlled cortical impact ,neuroinflammation ,neurobehavior ,Medical emergencies. Critical care. Intensive care. First aid ,RC86-88.9 - Abstract
Objective Chronic stress in adolescence may affect brain maturation and predispose individuals to psychiatric disorders in adulthood. However, whether chronic juvenile stress influences vulnerability to nonpsychiatric brain injuries, such as traumatic brain injury (TBI), remains unclear. Therefore, we hypothesized that juvenile stress-related neuronal circuit disturbances could aggravate brain damage following TBI in adulthood. Methods For chronic stress, we used an unpredictable chronic mild stress (UCMS) procedure for 5 weeks in adolescent mice. This was followed by a controlled cortical impact (CCI) injury to evaluate the influence of chronic juvenile stress on brain damage progression following TBI in adult mice. Mice underwent UCMS alone, UCMS followed by CCI, CCI alone, or sham operation. We characterized neurobehavioral deficits (Barnes maze, open field, and light-dark tests), neuroinflammation (ionized calcium-binding adapter molecule 1 [Iba-1], glial fibrillary acidic protein [GFAP], and neuron-specific nuclear protein [NeuN] immunoreactivity), and apoptosis (B-cell lymp [Bcl-2], Bcl-2-associated X protein [Bax], and procaspase-3 immunoreactivity). Results Following CCI, mice exposed to UCMS showed decreased spatial learning and memory in the Barnes maze test compared with unstressed mice. A significant increase in Iba-1, GFAP, and Bax/Bcl-2 immunostaining levels was observed in the mice exposed to UCMS followed by CCI compared with the CCI-only mice. In contrast, a significant decrease in NeuN immunostaining levels was observed in the UCMS with CCI group compared with the CCI alone group. Conclusion Chronic stress in a juvenile mouse model aggravates neurobehavioral impairments and potentiates glial reactivity, neuronal injury, and apoptosis following moderate-to-severe TBI that occurs in adulthood. The present study suggests that juvenile chronic stress may influence poor outcomes following TBI in later adulthood.
- Published
- 2023
- Full Text
- View/download PDF
31. The Nociceptin/Orphanin FQ peptide receptor antagonist, SB-612111, improves cerebral blood flow in a rat model of traumatic brain injury
- Author
-
Omar N. Al Yacoub, Stefano Tarantini, Yong Zhang, Anna Csiszar, and Kelly M. Standifer
- Subjects
mild traumatic brain injury ,Nociceptin/orphanin FQ (N/OFQ) ,cofilin-1 ,cerebral blood flow ,Mitogen-activated protein kinases ,controlled cortical impact ,Therapeutics. Pharmacology ,RM1-950 - Abstract
Traumatic brain injury (TBI) affects more than 2.5 million people in the U.S. each year and is the leading cause of death and disability in children and adults ages 1 to 44. Approximately 90% of TBI cases are classified as mild but may still lead to acute detrimental effects such as impaired cerebral blood flow (CBF) that result in prolonged impacts on brain function and quality of life in up to 15% of patients. We previously reported that nociceptin/orphanin FQ (N/OFQ) peptide (NOP) receptor antagonism reversed mild blast TBI-induced vestibulomotor deficits and prevented hypoxia. To explore mechanisms by which the NOP receptor-N/OFQ pathway modulates hypoxia and other TBI sequelae, the ability of the NOP antagonist, SB-612111 (SB), to reverse TBI-induced CBF and associated injury marker changes were tested in this study. Male Wistar rats randomly received sham craniotomy or craniotomy + TBI via controlled cortical impact. Injury severity was assessed after 1 h (modified neurological severity score (mNSS). Changes in CBF were assessed 2 h post-injury above the exposed cortex using laser speckle contrast imaging in response to the direct application of increasing concentrations of vehicle or SB (1, 10, and 100 µM) to the brain surface. TBI increased mNSS scores compared to baseline and confirmed mild TBI (mTBI) severity. CBF was significantly impaired on the ipsilateral side of the brain following mTBI, compared to contralateral side and to sham rats. SB dose-dependently improved CBF on the ipsilateral side after mTBI compared to SB effects on the respective ipsilateral side of sham rats but had no effect on contralateral CBF or in uninjured rats. N/OFQ levels increased in the cerebral spinal fluid (CSF) following mTBI, which correlated with the percent decrease in ipsilateral CBF. TBI also activated ERK and cofilin within 3 h post-TBI; ERK activation correlated with increased CSF N/OFQ. In conclusion, this study reveals a significant contribution of the N/OFQ-NOP receptor system to TBI-induced dysregulation of cerebral vasculature and suggests that the NOP receptor should be considered as a potential therapeutic target for TBI.
- Published
- 2023
- Full Text
- View/download PDF
32. ECoG spiking activity and signal dimension are early predictive measures of epileptogenesis in a translational mouse model of traumatic brain injury
- Author
-
Rossella Di Sapia, Massimo Rizzi, Federico Moro, Ilaria Lisi, Alessia Caccamo, Teresa Ravizza, Annamaria Vezzani, and Elisa R. Zanier
- Subjects
Biomarker ,EEG ,Epileptiform activity ,Controlled cortical impact ,Mouse ,Seizures ,Neurosciences. Biological psychiatry. Neuropsychiatry ,RC321-571 - Abstract
The latency between traumatic brain injury (TBI) and the onset of epilepsy (PTE) represents an opportunity for counteracting epileptogenesis. Antiepileptogenesis trials are hampered by the lack of sensitive biomarkers that allow to enrich patient's population at-risk for PTE. We aimed to assess whether specific ECoG signals predict PTE in a clinically relevant mouse model with ∼60% epilepsy incidence. TBI was provoked in adult CD1 male mice by controlled cortical impact on the left parieto-temporal cortex, then mice were implanted with two perilesional cortical screw electrodes and two similar electrodes in the hemisphere contralateral to the lesion site. Acute seizures and spikes/sharp waves were ECoG-recorded during 1 week post-TBI. These early ECoG events were analyzed according to PTE incidence as assessed by measuring spontaneous recurrent seizures (SRS) at 5 months post-TBI. We found that incidence, number and duration of acute seizures during 3 days post-TBI were similar in PTE mice and mice not developing epilepsy (No SRS mice). Control mice with cortical electrodes (naïve, n = 5) or with electrodes and craniotomy (sham, n = 5) exhibited acute seizures but did not develop epilepsy. The daily number of spikes/sharp waves at the perilesional electrodes was increased similarly in PTE (n = 15) and No SRS (n = 8) mice vs controls (p
- Published
- 2023
- Full Text
- View/download PDF
33. Acute intranasal osteopontin treatment in male rats following TBI increases the number of activated microglia but does not alter lesion characteristics.
- Author
-
Jullienne, Amandine, Hamer, Mary, Haddad, Elizabeth, Morita, Alexander, Gifford, Peter, Hartman, Richard, Pearce, William J, Tang, Jiping, Zhang, John H, and Obenaus, Andre
- Subjects
Blood-Brain Barrier ,Brain ,Microglia ,Animals ,Rats ,Rats ,Sprague-Dawley ,Brain Edema ,Disease Models ,Animal ,Neuroprotective Agents ,Signal Transduction ,Male ,Osteopontin ,Brain Injuries ,Traumatic ,Heme oxygenase-1 ,T2 mapping ,controlled cortical impact ,edema ,extracellular matrix protein ,microglial activation ,Brain Disorders ,Traumatic Head and Spine Injury ,Traumatic Brain Injury (TBI) ,Neurosciences ,Physical Injury - Accidents and Adverse Effects ,Neurological ,Psychology ,Neurology & Neurosurgery - Abstract
Intranasal recombinant osteopontin (OPN) has been shown to be neuroprotective in different models of acquired brain injury but has never been tested after traumatic brain injury (TBI). We used a model of moderate-to-severe controlled cortical impact in male adult Sprague Dawley rats and tested our hypothesis that OPN treatment would improve neurological outcomes, lesion and brain tissue characteristics, neuroinflammation, and vascular characteristics at 1 day post-injury. Intranasal OPN administered 1 hr after the TBI did not improve neurological score, lesion volumes, blood-brain barrier, or vascular characteristics. When assessing neuroinflammation, we did not observe any effect of OPN on the astrocyte reactivity but discovered an increased number of activated microglia within the ipsilateral hemisphere. Moreover, we found a correlation between edema and heme oxygenase-1 (HO-1) expression which was decreased in OPN-treated animals, suggesting an effect of OPN on the HO-1 response to injury. Thus, OPN may increase or accelerate the microglial response after TBI, and early response of HO-1 in modulating edema formation may limit the secondary consequences of TBI at later time points. Additional experiments and at longer time points are needed to determine if intranasal OPN could potentially be used as a treatment after TBI where it might be beneficial by activating protective signaling pathways.
- Published
- 2020
34. Microglia-specific deletion of histone deacetylase 3 promotes inflammation resolution, white matter integrity, and functional recovery in a mouse model of traumatic brain injury
- Author
-
Yongfang Zhao, Hongfeng Mu, Yichen Huang, Sicheng Li, Yangfan Wang, R. Anne Stetler, Michael V. L. Bennett, C. Edward Dixon, Jun Chen, and Yejie Shi
- Subjects
Conditional gene knockout ,Controlled cortical impact ,HDAC3 ,Neuroinflammation ,Neurology. Diseases of the nervous system ,RC346-429 - Abstract
Abstract Background Histone deacetylases (HDACs) are believed to exacerbate traumatic brain injury (TBI) based on studies using pan-HDAC inhibitors. However, the HDAC isoform responsible for the detrimental effects and the cell types involved remain unknown, which may hinder the development of specific targeting strategies that boost therapeutic efficacy while minimizing side effects. Microglia are important mediators of post-TBI neuroinflammation and critically impact TBI outcome. HDAC3 was reported to be essential to the inflammatory program of in vitro cultured macrophages, but its role in microglia and in the post-TBI brain has not been investigated in vivo. Methods We generated HDAC3LoxP mice and crossed them with CX3CR1CreER mice, enabling in vivo conditional deletion of HDAC3. Microglia-specific HDAC3 knockout (HDAC3 miKO) was induced in CX3CR1CreER:HDAC3LoxP mice with 5 days of tamoxifen treatment followed by a 30-day development interval. The effects of HDAC3 miKO on microglial phenotype and neuroinflammation were examined 3–5 days after TBI induced by controlled cortical impact. Neurological deficits and the integrity of white matter were assessed for 6 weeks after TBI by neurobehavioral tests, immunohistochemistry, electron microscopy, and electrophysiology. Results HDAC3 miKO mice harbored specific deletion of HDAC3 in microglia but not in peripheral monocytes. HDAC3 miKO reduced the number of microglia by 26%, but did not alter the inflammation level in the homeostatic brain. After TBI, proinflammatory microglial responses and brain inflammation were markedly alleviated by HDAC3 miKO, whereas the infiltration of blood immune cells was unchanged, suggesting a primary effect of HDAC3 miKO on modulating microglial phenotype. Importantly, HDAC3 miKO was sufficient to facilitate functional recovery for 6 weeks after TBI. TBI-induced injury to axons and myelin was ameliorated, and signal conduction by white matter fiber tracts was significantly enhanced in HDAC3 miKO mice. Conclusion Using a novel microglia-specific conditional knockout mouse model, we delineated for the first time the role of microglial HDAC3 after TBI in vivo. HDAC3 miKO not only reduced proinflammatory microglial responses, but also elicited long-lasting improvement of white matter integrity and functional recovery after TBI. Microglial HDAC3 is therefore a promising therapeutic target to improve long-term outcomes after TBI.
- Published
- 2022
- Full Text
- View/download PDF
35. An end-end deep learning framework for lesion segmentation on multi-contrast MR images—an exploratory study in a rat model of traumatic brain injury.
- Author
-
KN, Bhanu Prakash, CS, Arvind, Mohammed, Abdalla, Chitta, Krishna Kanth, To, Xuan Vinh, Srour, Hussein, and Nasrallah, Fatima
- Subjects
- *
BRAIN injuries , *NECROSIS , *DEEP learning , *HAUSDORFF measures , *PENUMBRA (Radiotherapy) - Abstract
Traumatic brain injury (TBI) engenders traumatic necrosis and penumbra—areas of secondary neural injury which are crucial targets for therapeutic interventions. Segmenting manually areas of ongoing changes like necrosis, edema, hematoma, and inflammation is tedious, error-prone, and biased. Using the multi-parametric MR data from a rodent model study, we demonstrate the effectiveness of an end-end deep learning global-attention-based UNet (GA-UNet) framework for automatic segmentation and quantification of TBI lesions. Longitudinal MR scans (2 h, 1, 3, 7, 14, 30, and 60 days) were performed on eight Sprague–Dawley rats after controlled cortical injury was performed. TBI lesion and sub-regions segmentation was performed using 3D-UNet and GA-UNet. Dice statistics (DSI) and Hausdorff distance were calculated to assess the performance. MR scan variations-based (bias, noise, blur, ghosting) data augmentation was performed to develop a robust model. Training/validation median DSI for U-Net was 0.9368 with T2w and MPRAGE inputs, whereas GA-UNet had 0.9537 for the same. Testing accuracies were higher for GA-UNet than U-Net with a DSI of 0.8232 for the T2w-MPRAGE inputs. Longitudinally, necrosis remained constant while oligemia and penumbra decreased, and edema appearing around day 3 which increased with time. GA-UNet shows promise for multi-contrast MR image-based segmentation/quantification of TBI in large cohort studies. [ABSTRACT FROM AUTHOR]
- Published
- 2023
- Full Text
- View/download PDF
36. In-vivo time course of organ uptake and blood-brain-barrier permeation of poly(L-lactide) and poly(perfluorodecyl acrylate) nanoparticles with different surface properties in unharmed and brain-traumatized rats.
- Author
-
Bechinger, Patrick, Serrano Sponton, Lucas, Grützner, Verena, Musyanovych, Anna, Jussen, Daniel, Krenzlin, Harald, Eldahaby, Daniela, Riede, Nicole, Kempski, Oliver, Ringel, Florian, and Alessandri, Beat
- Subjects
SURFACE properties ,BRAIN injuries ,NANOPARTICLES ,INTRAVENOUS therapy ,BLOOD-brain barrier - Abstract
Background: Traumatic brain injury (TBI) has a dramatic impact on mortality and quality of life and the development of effective treatment strategies is of great socio-economic relevance. A growing interest exists in using polymeric nanoparticles (NPs) as carriers across the blood-brain barrier (BBB) for potentially effective drugs in TBI. However, the effect of NP material and type of surfactant on their distribution within organs, the amount of the administrated dose that reaches the brain parenchyma in areas with intact and opened BBB after trauma, and a possible elicited inflammatory response are still to be clarified. Methods: The organ distribution, BBB permeation and eventual inflammatory activation of polysorbate-80 (Tw80) and sodiumdodecylsulfate (SDS) stabilized poly(L-lactide) (PLLA) and poly(perfluorodecyl acrylate) (PFDL) nanoparticles were evaluated in rats after intravenous administration. The NP uptake into the brain was assessed under intact conditions and after controlled cortical impact (CCI). Results: A significantly higher NP uptake at 4 and 24 h after injection was observed in the liver and spleen, followed by the brain and kidney, with minimal concentrations in the lungs and heart for all NPs. A significant increase of NP uptake at 4 and 24 h after CCI was observed within the traumatized hemisphere, especially in the perilesional area, but NPs were still found in areas away from the injury site and the contralateral hemisphere. NPs were internalized in brain capillary endothelial cells, neurons, astrocytes, and microglia. Immunohistochemical staining against GFAP, Iba1, TNFα, and IL1β demonstrated no glial activation or neuroinflammatory changes. Conclusions: Tw80 and SDS coated biodegradable PLLA and non-biodegradable PFDL NPs reach the brain parenchyma with and without compromised BBB by TBI, even though a high amount of NPs are retained in the liver and spleen. No inflammatory reaction is elicited by these NPs within 24 h after injection. Thus, these NPs could be considered as potentially effective carriers or markers of newly developed drugs with low or even no BBB permeation. [ABSTRACT FROM AUTHOR]
- Published
- 2023
- Full Text
- View/download PDF
37. Pre-operative environmental enrichment does not yield a prophylactic effect against traumatic brain injury-induced neurobehavioral deficits.
- Author
-
Moschonas, Eleni H., Steber, Jade A., Capeci, Haley E., Donald, Hailey M., Vozzella, Vincent J., Bittner, Rachel A., Annas, Ellen M., Rennerfeldt, Piper L., Cheng, Jeffrey P., Bondi, Corina O., and Kline, Anthony E.
- Subjects
- *
ENVIRONMENTAL enrichment , *MAZE tests , *BRAIN injuries , *MOTOR ability , *WATER testing , *TOTAL body irradiation - Abstract
The robustness of environmental enrichment (EE) in ameliorating neurobehavioral and cognitive deficits after experimental traumatic brain injury (TBI) is unequivocal. What is equivocal is whether EE can function as a prophylactic to afford resiliency and neuroprotection against TBI. We hypothesized that pre-operative EE would yield a protective effect against TBI-induced motor, cognitive, and coping deficits, and that further improvements would be conferred when EE is provided before and after TBI. To test the hypotheses, adult male rats received either 4 weeks of EE or standard (STD) housing prior to undergoing a controlled cortical impact of moderate severity (2.8 mm deformation at 4 m/s) or sham injury while under anesthesia. After injury, the rats were randomly assigned to post-operative EE or STD housing. Motor ability, spatial learning, and memory retention were assessed by beam-walk and water maze tests, respectively. Active and passive behavioral coping strategies were evaluated with the shock probe defensive burying (SPDB) test. c-Fos and cortical lesion volume were also quantified. The post-TBI enrichment groups (EE + TBI + EE and STD + TBI + EE) did not differ (p > 0.05) and performed better than the post-TBI STD-housed groups (EE + TBI + STD and STD + TBI + STD) on motor and cognition (p < 0.05). The post-TBI STD groups did not differ, regardless of whether in EE or STD living conditions before injury (p > 0.05). Moreover, both post-TBI enrichment groups performed better in the SPDB test relative to the STD + TBI + STD group (p < 0.05). c-Fos + cells were upregulated in the ipsilateral CA 1 in both pre-injury EE groups relative to the pre-injury STD groups (p < 0.05). No statistical differences were observed in cortical lesion volume among the groups. Overall, these data do not support the hypothesis as no neuroprotective effect was observed with 4 weeks of pre-operative EE and no additional benefit was achieved in the TBI group receiving both pre-and-post EE relative to the TBI group receiving only post-EE. However, the data do reinforce the consistency of post-TBI EE in producing robust neurobehavioral benefits, which further supports this paradigm as a relevant preclinical model of neurorehabilitation. • Pre-operative environmental enrichment (EE) does not yield a prophylactic effect against TBI. • Post-operative EE robustly benefits neurobehavior after TBI. • Increased c-Fos did not correlate with neurobehavioral improvements. • The data support EE as an efficacious rehabilitative therapy for TBI. [ABSTRACT FROM AUTHOR]
- Published
- 2025
- Full Text
- View/download PDF
38. Transcriptional Profiling in a Novel Swine Model of Traumatic Brain Injury
- Author
-
Samuel S. Shin, Amy C. Gottschalk, Vanessa M. Mazandi, Todd J. Kilbaugh, and Marco M. Hefti
- Subjects
controlled cortical impact ,pig ,RNA ,swine ,traumatic brain injury ,Medical emergencies. Critical care. Intensive care. First aid ,RC86-88.9 - Abstract
Transcriptomic investigations of traumatic brain injury (TBI) can give us deep insights into the pathological and compensatory processes post-injury. Thus far, transcriptomic studies in TBI have mostly used microarrays and have focused on rodent models. However, a large animal model of TBI bears a much stronger resemblance to human TBI with regard to the anatomical details, mechanics of injury, genetics, and, possibly, molecular response. Because of the advantages of a large animal TBI model, we investigated the gene expression changes between injured and uninjured sides of pig cerebral cortex after TBI. Given acute inflammation that follows after TBI and the important role that immune response plays in neuroplasticity and recovery, we hypothesized that transcriptional changes involving immune function will be upregulated. Eight female 4-week-old piglets were injured on the right hemisphere with controlled cortical impact (CCI). At 5 days after TBI, pericontusional cortex tissues from the injured side and contralateral cortical tissues were collected. After RNA extraction, library preparation and sequencing as well as gene expression changes between the ipsi- and contralateral sides were compared. There were 6642 genes that were differentially expressed between the ipsi- and contralateral sides, and 1993 genes among them had at least 3-fold differences. Differentially expressed genes were enriched for biological processes related to immune system activation, regulation of immune response, and leukocyte activation. Many of the differentially expressed genes, such as CD4, CD86, IL1A, IL23R, and IL1R1, were major regulators of immune function. This study demonstrated some of the major transcriptional changes between the pericontusional and contralateral tissue at an acute time point after TBI in pigs.
- Published
- 2022
- Full Text
- View/download PDF
39. Localization of Multi-Lamellar Vesicle Nanoparticles to Injured Brain Tissue in a Controlled Cortical Impact Injury Model of Traumatic Brain Injury in Rodents
- Author
-
Ricky Whitener, Jeremy J. Henchir, Thomas A. Miller, Emily Levy, Aubrienne Krysiewicz-Bell, Eliza S. LaRovere Abrams, Shaun W. Carlson, Naresh Menon, C. Edward Dixon, Michael J. Whalen, and Claude J. Rogers
- Subjects
blood?brain barrier ,controlled cortical impact ,in vivo studies ,traumatic brain injury ,Medical emergencies. Critical care. Intensive care. First aid ,RC86-88.9 - Abstract
Severe traumatic brain injury (TBI), such as that suffered by patients with cerebral contusion, is a major cause of death and disability in young persons. Effective therapeutics to treat or mitigate the effects of severe TBI are lacking, in part because drug delivery to the injured brain remains a challenge. Promising therapeutics targeting secondary injury mechanisms may have poor pharmacokinetics/pharmacodynamics, unwanted side effects, or high hydrophobicity. To address these challenges, we have developed a multi-lamellar vesicle nanoparticle (MLV-NP) formulation with a narrow size distribution (243?nm in diameter, 0.09 polydispersity index) and the capability of encapsulating hydrophobic small molecule drugs for delivery to the injured brain. To demonstrate the utility of these particles, we produced dual-fluorescent labeled nanoparticles containing the organic dyes, coumarin 153 and rhodamine B, that were delivered intravenously to Sprague-Dawley rats and C57Bl6/J mice at 1, 1 and 4, 24, or 48?h after controlled cortical impact injury. Distribution of particles was measured at 5, 25, 48, or 49?h post-injury by fluorescence microscopy of coronal brain sections. In all cases of MLV administration, a 1.2- to 1.9-fold enhancement of ipsilateral fluorescence signal was observed compared to the contralateral cortex. Enhanced fluorescence was also observed in the injured hippocampal tissue in these animals. MLV-NPs administered at 1?h were observed intracellularly in the injured hemisphere at 48?h, suggesting the possibility of concentrated drug delivery to injured cells. These results suggest that MLV-NP delivery of therapeutic agents may be a viable strategy for treating cerebral contusion TBI.
- Published
- 2022
- Full Text
- View/download PDF
40. In-vivo time course of organ uptake and blood-brain-barrier permeation of poly(L-lactide) and poly(perfluorodecyl acrylate) nanoparticles with different surface properties in unharmed and brain-traumatized rats
- Author
-
Patrick Bechinger, Lucas Serrano Sponton, Verena Grützner, Anna Musyanovych, Daniel Jussen, Harald Krenzlin, Daniela Eldahaby, Nicole Riede, Oliver Kempski, Florian Ringel, and Beat Alessandri
- Subjects
nanoparticles ,controlled cortical impact ,blood–brain barrier ,traumatic brain injury ,inflammation ,Neurology. Diseases of the nervous system ,RC346-429 - Abstract
BackgroundTraumatic brain injury (TBI) has a dramatic impact on mortality and quality of life and the development of effective treatment strategies is of great socio-economic relevance. A growing interest exists in using polymeric nanoparticles (NPs) as carriers across the blood-brain barrier (BBB) for potentially effective drugs in TBI. However, the effect of NP material and type of surfactant on their distribution within organs, the amount of the administrated dose that reaches the brain parenchyma in areas with intact and opened BBB after trauma, and a possible elicited inflammatory response are still to be clarified.MethodsThe organ distribution, BBB permeation and eventual inflammatory activation of polysorbate-80 (Tw80) and sodiumdodecylsulfate (SDS) stabilized poly(L-lactide) (PLLA) and poly(perfluorodecyl acrylate) (PFDL) nanoparticles were evaluated in rats after intravenous administration. The NP uptake into the brain was assessed under intact conditions and after controlled cortical impact (CCI).ResultsA significantly higher NP uptake at 4 and 24 h after injection was observed in the liver and spleen, followed by the brain and kidney, with minimal concentrations in the lungs and heart for all NPs. A significant increase of NP uptake at 4 and 24 h after CCI was observed within the traumatized hemisphere, especially in the perilesional area, but NPs were still found in areas away from the injury site and the contralateral hemisphere. NPs were internalized in brain capillary endothelial cells, neurons, astrocytes, and microglia. Immunohistochemical staining against GFAP, Iba1, TNFα, and IL1β demonstrated no glial activation or neuroinflammatory changes.ConclusionsTw80 and SDS coated biodegradable PLLA and non-biodegradable PFDL NPs reach the brain parenchyma with and without compromised BBB by TBI, even though a high amount of NPs are retained in the liver and spleen. No inflammatory reaction is elicited by these NPs within 24 h after injection. Thus, these NPs could be considered as potentially effective carriers or markers of newly developed drugs with low or even no BBB permeation.
- Published
- 2023
- Full Text
- View/download PDF
41. Polynitroxylated PEGylated hemoglobin protects pig brain neocortical gray and white matter after traumatic brain injury and hemorrhagic shock
- Author
-
Jun Wang, Yanrong Shi, Suyi Cao, Xiuyun Liu, Lee J. Martin, Jan Simoni, Bohdan J. Soltys, Carleton J. C. Hsia, and Raymond C. Koehler
- Subjects
controlled cortical impact ,frontal lobe damage ,hemoglobin-based oxygen carrier ,dendrite/axon rescue ,neuroprotection ,Medical technology ,R855-855.5 - Abstract
Polynitroxylated PEGylated hemoglobin (PNPH, aka SanFlow) possesses superoxide dismutase/catalase mimetic activities that may directly protect the brain from oxidative stress. Stabilization of PNPH with bound carbon monoxide prevents methemoglobin formation during storage and permits it to serve as an anti-inflammatory carbon monoxide donor. We determined whether small volume transfusion of hyperoncotic PNPH is neuroprotective in a porcine model of traumatic brain injury (TBI) with and without accompanying hemorrhagic shock (HS). TBI was produced by controlled cortical impact over the frontal lobe of anesthetized juvenile pigs. Hemorrhagic shock was induced starting 5 min after TBI by 30 ml/kg blood withdrawal. At 120 min after TBI, pigs were resuscitated with 60 ml/kg lactated Ringer's (LR) or 10 or 20 ml/kg PNPH. Mean arterial pressure recovered to approximately 100 mmHg in all groups. A significant amount of PNPH was retained in the plasma over the first day of recovery. At 4 days of recovery in the LR-resuscitated group, the volume of frontal lobe subcortical white matter ipsilateral to the injury was 26.2 ± 7.6% smaller than homotypic contralateral volume, whereas this white matter loss was only 8.6 ± 12.0% with 20-ml/kg PNPH resuscitation. Amyloid precursor protein punctate accumulation, a marker of axonopathy, increased in ipsilateral subcortical white matter by 132 ± 71% after LR resuscitation, whereas the changes after 10 ml/kg (36 ± 41%) and 20 ml/kg (26 ± 15%) PNPH resuscitation were not significantly different from controls. The number of cortical neuron long dendrites enriched in microtubules (length >50 microns) decreased in neocortex by 41 ± 24% after LR resuscitation but was not significantly changed after PNPH resuscitation. The perilesion microglia density increased by 45 ± 24% after LR resuscitation but was unchanged after 20 ml/kg PNPH resuscitation (4 ± 18%). Furthermore, the number with an activated morphology was attenuated by 30 ± 10%. In TBI pigs without HS followed 2 h later by infusion of 10 ml/kg LR or PNPH, PNPH remained neuroprotective. These results in a gyrencephalic brain show that resuscitation from TBI + HS with PNPH protects neocortical gray matter, including dendritic microstructure, and white matter axons and myelin. This neuroprotective effect persists with TBI alone, indicating brain-targeting benefits independent of blood pressure restoration.
- Published
- 2023
- Full Text
- View/download PDF
42. Male and Female Mice Exhibit Divergent Responses of the Cortical Vasculature to Traumatic Brain Injury
- Author
-
Jullienne, Amandine, Salehi, Arjang, Affeldt, Bethann, Baghchechi, Mohsen, Haddad, Elizabeth, Avitua, Angela, Walsworth, Mark, Enjalric, Isabelle, Hamer, Mary, Bhakta, Sonali, Tang, Jiping, Zhang, John H, Pearce, William J, and Obenaus, André
- Subjects
Physical Injury - Accidents and Adverse Effects ,Biomedical Imaging ,Neurosciences ,Traumatic Head and Spine Injury ,Brain Disorders ,Traumatic Brain Injury (TBI) ,Animals ,Brain Injuries ,Traumatic ,Cerebral Cortex ,Female ,Male ,Mice ,Mice ,Inbred C57BL ,Sex Characteristics ,controlled cortical impact ,heme oxygenase-1 ,sex difference ,vasculature ,Wnt/beta-catenin ,Wnt/β-catenin ,Clinical Sciences ,Neurology & Neurosurgery - Abstract
We previously reported that traumatic brain injuries (TBI) alter the cerebrovasculature near the injury site in rats, followed by revascularization over a 2-week period. Here, we tested our hypothesis that male and female adult mice have differential cerebrovascular responses following a moderate controlled cortical impact (CCI). Using in vivo magnetic resonance imaging (MRI), a new technique called vessel painting, and immunohistochemistry, we found no differences between males and females in lesion volume, neurodegeneration, blood-brain barrier (BBB) alteration, and microglia activation. However, females exhibited more astrocytic hypertrophy and heme-oxygenase-1 (HO-1) induction at 1 day post-injury (dpi), whereas males presented with increased endothelial activation and expression of β-catenin, shown to be involved in angiogenesis. At 7 dpi, we observed an increase in the number of vessels and an enhancement in vessel complexity in the injured cortex of males compared with females. Cerebrovasculature recovers differently after CCI, suggesting biological sex should be considered when designing new therapeutic agents.
- Published
- 2018
43. Training of the impaired forelimb after traumatic brain injury enhances hippocampal neurogenesis in the Emx1 null mice lacking a corpus callosum
- Author
-
Neumann, Melanie, Liu, Wei, Sun, Chongran, Yang, Shih Yen, Noble-Haeusslein, Linda J, and Liu, Jialing
- Subjects
Biological Psychology ,Psychology ,Traumatic Brain Injury (TBI) ,Physical Injury - Accidents and Adverse Effects ,Rehabilitation ,Neurosciences ,Brain Disorders ,Traumatic Head and Spine Injury ,Neurological ,Injuries and accidents ,Agenesis of Corpus Callosum ,Animals ,Brain Injuries ,Traumatic ,Disease Models ,Animal ,Doublecortin Protein ,Forelimb ,Hippocampus ,Homeodomain Proteins ,Male ,Mice ,Knockout ,Motor Skills ,Neurogenesis ,Neurological Rehabilitation ,Neuronal Plasticity ,Random Allocation ,Transcription Factors ,Controlled cortical impact ,DCX ,Emx1 ,Neuroplasticity ,Skill reaching ,Medical and Health Sciences ,Psychology and Cognitive Sciences ,Neurology & Neurosurgery ,Biological psychology - Abstract
Unilateral brain injury is known to disrupt the balance between the two cortices, as evidenced by an abnormally high interhemispheric inhibitory drive from motor cortex M1intact to M1lesioned transmitted transcallosally. Our previous work has shown that the deletion of homeobox gene Emx1 not only led to the agenesis of the corpus callosum (cc), but also to reduced hippocampal neurogenesis. The current study sought to determine whether lacking the cc affected the recovery of forelimb function and hippocampal plasticity following training of the affected limb in mice with unilateral traumatic brain injuries (TBI). One week after TBI, produced by a controlled cortical impact to impair the preferred limb, Emx1 wild type (WT) and knock out (KO) mice were subjected to the single-pellet reaching task with the affected limb for 4 weeks. Both TBI and Emx1 deletion had overall adverse effects on the successful rate of reaching. However, TBI significantly affected reaching performance only in the WT mice and not in the KO mice. Both TBI and Emx1 gene deletion also negatively affected hippocampal neurogenesis, demonstrated by a reduction in doublecortin (DCX)-expressing immature neurons, while limb training enhanced DCX expression. However, limb training increased DCX cells in KO mice only in the TBI-treated group, whereas it induced neurogenesis in both WT mice groups regardless of the treatment. Our finding also suggests that limb training enhances neuroplasticity after brain injury at functionally remote regions including the hippocampus, which may have implications for promoting overall recovery of function after TBI.
- Published
- 2018
44. Unravelling the impact of QRICH1 modulation on endoplasmic reticulum stress and neuronal apoptosis in traumatic brain injury.
- Author
-
Wang S, Ren Y, Duan A, Lu D, Liu G, Meng L, Zhang Y, Shou R, Li H, Wang Z, Wang Z, and Sun X
- Abstract
Background: Traumatic brain injury (TBI) is a major public health concern with high morbidity and mortality rates. Secondary brain injury, marked by inflammatory responses and apoptosis, worsens TBI outcomes. The endoplasmic reticulum stress (ERS) response has been implicated in secondary brain injury, with Glutamine Rich 1 Gene (QRICH1) emerging as a potential mediator. However, the precise role of QRICH1 in TBI pathogenesis and its therapeutic implications remain unclear., Methods: Controlled cortical impact mouse and Lipopolysaccharide-stimulated primary neuron models were used. Behavioral assessments, including the modified Garcia score, Y-maze test, and open-field test, were used to evaluate postoperative recovery in mice. QRICH1 neuron conditional knockout (cKO) mice were used to assess QRICH1 function, whereas adeno-associated virus (AAV)-mediated gene manipulation was used to modulate QRICH1 expression in cortical neurons., Results: QRICH1 expression was upregulated in the brain tissue of TBI mice, particularly 24 h post-injury, as shown by western blot analysis and immunofluorescence staining. QRICH1 is localized within neuronal nuclei, suggesting a role in cellular stress responses. QRICH1 cKO improved behavioral outcomes post-TBI, whereas AAV-mediated QRICH1 overexpression exacerbated secondary brain injury, characterized by increased ERS-related protein expression and neuronal death. Conversely, AAV-mediated QRICH1 knockdown reduced secondary brain injury as evidenced by decreased ERS-related protein expression and neuronal death., Conclusion: QRICH1 plays a critical role in exacerbating ERS and apoptosis, and influences neuronal fate in secondary brain injury. Its involvement in the ERS pathway and in the induction of neuronal apoptosis post-TBI highlights QRICH1 as a potential therapeutic target for TBI treatment., Competing Interests: Declaration of competing interest The authors declare that they have no known competing financial interests or personal relationships that could have appeared to influence the work reported in this paper., (Copyright © 2024 Elsevier B.V. All rights reserved.)
- Published
- 2024
- Full Text
- View/download PDF
45. Environmental enrichment-induced cognitive recovery after a moderate pediatric traumatic brain injury is associated with the gut microbiota and neuroinflammation.
- Author
-
Zamudio-Flores J, Cerqueda D, Phillips-Farfán B, Guerrero-Flores S, Salinas-García AF, Meléndez-Herrera E, Sélem-Mojica N, Kline AE, and Lajud N
- Abstract
Pediatric traumatic brain injury (TBI) is a significant health concern, yet access to rehabilitation therapies for children remains limited. Environmental enrichment (EE) is a preclinical model of neurorehabilitation that promotes behavioral recovery and reduces neuroinflammation after TBI. While the gut microbiota has recently emerged as a potential therapeutic target for treating TBI sequelae in adults, its role in recovery after pediatric TBI remains unclear. Therefore, our aim was to assess the effect of EE on gut microbiota and its correlation with cognition as well as microglial morphology in a preclinical model of pediatric TBI. Male rats underwent a controlled cortical impact of moderate severity or sham injury at postnatal day 21 and were then randomly assigned to either EE or standard (STD) housing. Cognition was evaluated using the Morris water maze (MWM) on post-injury days 14-19. Microglial morphology and caecum microbiota was characterized on post-injury day 21. Cognitive deficits and increased microglial activation in the ipsilateral cortex were observed in the STD-housed TBI rats but not those in EE. TBI decreased microbiota α-diversity, while PERMANOVA analysis showed that both TBI and EE modified microbiota β-diversity. Furthermore, regression models indicated that microglial morphology in the ipsilateral cortex and Lactobacillus reuteri predicted behavioral outcomes, while Prevotellaceae NK3B31 was associated with microglial morphology. The data suggest that EE mitigates TBI-induced alterations in gut microbiota and that there is a complex interplay between EE, microbiota and microglial morphology that predicts behavioral recovery in pediatric rats., Competing Interests: Declaration of competing interest None., (Copyright © 2024. Published by Elsevier Inc.)
- Published
- 2024
- Full Text
- View/download PDF
46. Essential role of MALAT1 in reducing traumatic brain injury
- Author
-
Na Wu, Chong-Jie Cheng, Jian-Jun Zhong, Jun-Chi He, Zhao-Si Zhang, Zhi-Gang Wang, Xiao-Chuan Sun, and Han Liu
- Subjects
angiogenesis ,controlled cortical impact ,ezh2 ,jagged-1 ,lncrna ,malat1 ,notch1 ,oxygen-glucose deprivation ,traumatic brain injury ,vascular remodeling ,Neurology. Diseases of the nervous system ,RC346-429 - Abstract
As a highly evolutionary conserved long non-coding RNA, metastasis associated lung adenocarcinoma transcript 1 (MALAT1) was first demonstrated to be related to lung tumor metastasis by promoting angiogenesis. To investigate the role of MALAT1 in traumatic brain injury, we established mouse models of controlled cortical impact and cell models of oxygen-glucose deprivation to mimic traumatic brain injury in vitro and in vivo. The results revealed that MALAT1 silencing in vitro inhibited endothelial cell viability and tube formation but increased migration. In MALAT1-deficient mice, endothelial cell proliferation in the injured cortex, functional vessel density and cerebral blood flow were reduced. Bioinformatic analyses and RNA pull-down assays validated enhancer of zeste homolog 2 (EZH2) as a downstream factor of MALAT1 in endothelial cells. Jagged-1, the Notch homolog 1 (NOTCH1) agonist, reversed the MALAT1 deficiency-mediated impairment of angiogenesis. Taken together, our results suggest that MALAT1 controls the key processes of angiogenesis following traumatic brain injury in an EZH2/NOTCH1-dependent manner.
- Published
- 2022
- Full Text
- View/download PDF
47. STAT1 Contributes to Microglial/Macrophage Inflammation and Neurological Dysfunction in a Mouse Model of Traumatic Brain Injury.
- Author
-
Yongfang Zhao, Cheng Ma, Caixia Chen, Sicheng Li, Yangfan Wang, Tuo Yang, Stetler, R. Anne, Bennett, Michael V. L., Dixon, C. Edward, Jun Chen, and Yejie Shi
- Subjects
- *
BRAIN injuries , *STAT proteins , *LABORATORY mice , *ANIMAL disease models , *SIZE of brain , *BLAST injuries - Abstract
Traumatic brain injury (TBI) triggers a plethora of inflammatory events in the brain that aggravate secondary injury and impede tissue repair. Resident microglia (Mi) and blood-borne infiltrating macrophages (MΦ) are major players of inflammatory responses in the post-TBI brain and possess high functional heterogeneity. However, the plasticity of these cells has yet to be exploited to develop therapies that can mitigate brain inflammation and improve the outcome after TBI. This study investigated the transcription factor STAT1 as a key determinant of proinflammatory Mi/MΦ responses and aimed to develop STAT1 as a novel therapeutic target for TBI using a controlled cortical impact model of TBI on adult male mice. TBI induced robust upregulation of STAT1 in the brain at the subacute injury stage, which occurred primarily in Mi/MΦ. Intraperitoneal administration of fludarabine, a selective STAT1 inhibitor, markedly alleviated proinflammatory Mi/MΦ responses and brain inflammation burden after TBI. Such phenotype-modulating effects of fludarabine on post-TBI Mi/MΦ were reproduced by tamoxifen-induced, selective KO of STAT1 in Mi/MΦ (STAT1 mKO). By propelling Mi/MΦ away from a detrimental proinflammatory phenotype, STAT1 mKO was sufficient to reduce long-term neurologic deficits and brain lesion size after TBI. Importantly, short-term fludarabine treatment after TBI elicited long-lasting improvement of TBI outcomes, but this effect was lost on STAT1 mKO mice. Together, our study provided the first line of evidence that STAT1 causatively determines the proinflammatory phenotype of brain Mi/MU after TBI. We also showed promising preclinical data supporting the use of fludarabine as a novel immunomodulating therapy to TBI. [ABSTRACT FROM AUTHOR]
- Published
- 2022
- Full Text
- View/download PDF
48. Interleukin-4 Reduces Lesion Volume and Improves Neurological Function in the Acute Phase after Experimental Traumatic Brain Injury in Mice.
- Author
-
Walter, Johannes, Kovalenko, Olga, Younsi, Alexander, Grutza, Martin, Unterberg, Andreas, and Zweckberger, Klaus
- Subjects
- *
BRAIN injuries , *INTERLEUKIN-4 , *TOTAL body irradiation , *BRAIN damage , *MICE - Abstract
Little is known about the impact of interleukin-4 (IL-4) on secondary brain damage in the acute phase after experimental traumatic brain injury (TBI). Therefore, we evaluated the effect of IL-4-Knockout (IL-4-KO) on structural damage, as well as functional impairment, in the acute phase after experimental TBI in mice. A total of 28 C57Bl/6 wildtype and 20 C57BL/6-Il4tm1Nnt/J IL-4-KO mice were subjected to controlled cortical impact (CCI). Contusion volumes, body weight and functional outcome (Video Open Field Test [VOF], Hole Board Test [HB], CatWalkXT®) were determined on post-operative Days 1 (D1), 3 (D3), and 7 (D7). Contusion volume (13.45 ± 0.88 mm3 vs. 9.50 ± 0.97 mm3, p = 0.015) and weight loss (-2.92 ± 0.52% vs. -0.85 ± 0.67%, p = 0.027) were significantly higher and exploration behavior significantly more impaired (e.g., 150.44 ± 18.71 fields explored vs. 211.56 ± 18.90 fields explored, p = 0.028 in the VOF; 23.31 ± 2.03 holes explored vs. 35.65 ± 1.93 holes explored, p < 0.001 in the HB) in IL-4-KO mice on D1. Gait impairment was significantly more pronounced in IL-4-KO mice throughout the first week after CCI (e.g., 0.07 ± 0.01 sec vs. 0.00 ± 0.01 sec, p = 0.047 for right hindpaw Swing on D1; -1.76 ± 1.34 U vs. 2.53 ± 0.90 U, p = 0.01 for right forepaw mean intensity on D3; -0.01 ± 0.01 cm2 vs. 0.05 ± 0.01 cm2, p = 0.015 for left forepaw mean area on D7). In conclusion, IL-4 reduces structural damage and improves functional outcome in the acute phase after CCI. Neurobehavioral outcome assessment in IL-4-related studies should focus on motor function on the first 3 days after trauma induction. [ABSTRACT FROM AUTHOR]
- Published
- 2022
- Full Text
- View/download PDF
49. Microglia-specific deletion of histone deacetylase 3 promotes inflammation resolution, white matter integrity, and functional recovery in a mouse model of traumatic brain injury.
- Author
-
Zhao, Yongfang, Mu, Hongfeng, Huang, Yichen, Li, Sicheng, Wang, Yangfan, Stetler, R. Anne, Bennett, Michael V. L., Dixon, C. Edward, Chen, Jun, and Shi, Yejie
- Subjects
WHITE matter (Nerve tissue) ,BRAIN injuries ,HISTONE deacetylase ,LABORATORY mice ,ANIMAL disease models - Abstract
Background: Histone deacetylases (HDACs) are believed to exacerbate traumatic brain injury (TBI) based on studies using pan-HDAC inhibitors. However, the HDAC isoform responsible for the detrimental effects and the cell types involved remain unknown, which may hinder the development of specific targeting strategies that boost therapeutic efficacy while minimizing side effects. Microglia are important mediators of post-TBI neuroinflammation and critically impact TBI outcome. HDAC3 was reported to be essential to the inflammatory program of in vitro cultured macrophages, but its role in microglia and in the post-TBI brain has not been investigated in vivo.Methods: We generated HDAC3LoxP mice and crossed them with CX3CR1CreER mice, enabling in vivo conditional deletion of HDAC3. Microglia-specific HDAC3 knockout (HDAC3 miKO) was induced in CX3CR1CreER:HDAC3LoxP mice with 5 days of tamoxifen treatment followed by a 30-day development interval. The effects of HDAC3 miKO on microglial phenotype and neuroinflammation were examined 3-5 days after TBI induced by controlled cortical impact. Neurological deficits and the integrity of white matter were assessed for 6 weeks after TBI by neurobehavioral tests, immunohistochemistry, electron microscopy, and electrophysiology.Results: HDAC3 miKO mice harbored specific deletion of HDAC3 in microglia but not in peripheral monocytes. HDAC3 miKO reduced the number of microglia by 26%, but did not alter the inflammation level in the homeostatic brain. After TBI, proinflammatory microglial responses and brain inflammation were markedly alleviated by HDAC3 miKO, whereas the infiltration of blood immune cells was unchanged, suggesting a primary effect of HDAC3 miKO on modulating microglial phenotype. Importantly, HDAC3 miKO was sufficient to facilitate functional recovery for 6 weeks after TBI. TBI-induced injury to axons and myelin was ameliorated, and signal conduction by white matter fiber tracts was significantly enhanced in HDAC3 miKO mice.Conclusion: Using a novel microglia-specific conditional knockout mouse model, we delineated for the first time the role of microglial HDAC3 after TBI in vivo. HDAC3 miKO not only reduced proinflammatory microglial responses, but also elicited long-lasting improvement of white matter integrity and functional recovery after TBI. Microglial HDAC3 is therefore a promising therapeutic target to improve long-term outcomes after TBI. [ABSTRACT FROM AUTHOR]- Published
- 2022
- Full Text
- View/download PDF
50. Skeletal Muscle Atrophy and Degeneration in a Mouse Model of Traumatic Brain Injury
- Author
-
Shahidi, Bahar, Shah, Sameer B, Esparza, Mary, Head, Brian P, and Ward, Samuel R
- Subjects
Neurosciences ,Physical Injury - Accidents and Adverse Effects ,Traumatic Brain Injury (TBI) ,Traumatic Head and Spine Injury ,Regenerative Medicine ,Brain Disorders ,Injuries and accidents ,Neurological ,Musculoskeletal ,Animals ,Brain Injuries ,Traumatic ,Disease Models ,Animal ,Male ,Mice ,Mice ,Inbred C57BL ,Muscle ,Skeletal ,Muscular Atrophy ,atrophy ,controlled cortical impact ,degeneration ,regeneration ,skeletal muscle ,traumatic brain injury ,Clinical Sciences ,Neurology & Neurosurgery - Abstract
Atrophy is thought to be a primary mode of muscle loss in neuromuscular injuries. The differential effects of central and peripheral injuries on atrophy and degeneration/regeneration in skeletal muscle tissue have not been well described. This study investigated skeletal muscle atrophy and degeneration/regeneration in an animal model of traumatic brain injury (TBI). Eight 8-month-old wild-type C57BL6 mice underwent either a sham craniotomy or TBI targeting the motor cortex. Atrophy (fiber area; FA) and degeneration/regeneration (centralized nuclei proportions; CN) of the soleus and tibialis anterior (TA) muscles were measured 2 months post-injury. Injured soleus FAs were smaller than sham soleus (p = 0.02) and injured TA (p
- Published
- 2018
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.