Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 20
Filter
Add more filters










Publication year range
1.
Neuroreport ; 32(3): 188-197, 2021 02 03.
Article in English | MEDLINE | ID: mdl-33470761

ABSTRACT

Pyroptosis has been reported to contribute to the traumatic brain injury (TBI) process. Ac-FLTD-CMK is a newly synthesized pyroptosis inhibitor. However, whether Ac-FLTD-CMK inhibits pyroptosis and plays a neuroprotective role after TBI is unknown. The present study aimed to determine the effects of Ac-FLTD-CMK on TBI in a mouse model. Male C57BL/6 mice were randomly divided into sham, TBI + vehicle, and TBI + Ac-FLTD-CMK groups. TBI was induced using a weight-drop apparatus. Intraventricular injection of Ac-FLTD-CMK was performed 30 min after TBI. Caspase-1, caspase-11, gasdermin-D (GSDMD), and caspase-3 expression in the peri-contusional cortex were assessed by western blotting. Interleukin-1ß (IL-1ß) and interleukin-18 (IL-18) expression in the peri-contusional cortex were measured using ELISA. Behavioral experiments, brain water content, Evans blue extravasation, lactate dehydrogenase (LDH) release, and terminal deoxynucleotidyl transferase-mediated dUTP-biotin nick end labeling staining were also performed. The results showed that Ac-FLTD-CMK administration significantly downregulated caspase-1 p20, caspase-11 p20, GSDMD N-terminal, IL-1ß, and IL-18 expression; reduced LDH release; alleviated neuronal death; attenuated brain edema and blood-brain barrier damage; and improved neurobehavioral function. These findings indicate that Ac-FLTD-CMK treatment suppresses pyroptosis and protects mice against TBI.


Subject(s)
Brain Contusion/metabolism , Brain/drug effects , Caspase Inhibitors/pharmacology , Neuroprotective Agents/pharmacology , Pyroptosis/drug effects , Animals , Blood-Brain Barrier/drug effects , Blood-Brain Barrier/metabolism , Brain/metabolism , Brain Injuries, Traumatic/metabolism , Caspase 1/drug effects , Caspase 1/metabolism , Caspase 3/drug effects , Caspase 3/metabolism , Caspases, Initiator/drug effects , Caspases, Initiator/metabolism , Disease Models, Animal , Interleukin-18/metabolism , Interleukin-1beta/drug effects , Interleukin-1beta/metabolism , Intracellular Signaling Peptides and Proteins/drug effects , Intracellular Signaling Peptides and Proteins/metabolism , Mice , Neurons/drug effects , Neurons/metabolism , Open Field Test , Phosphate-Binding Proteins/drug effects , Phosphate-Binding Proteins/metabolism , Rotarod Performance Test
2.
World Neurosurg ; 147: e16-e24, 2021 03.
Article in English | MEDLINE | ID: mdl-33189916

ABSTRACT

OBJECTIVE: Traumatic brain injury (TBI) is a health problem worldwide, and therapeutic strategies to enhance brain tissue repair to lessen neurologic sequels are imperative. We aimed to analyze the impact of the inflammatory process in TBI through CXCR4 and CXCR7 chemokine receptors and their ligands' CXCL11 and CXCL12 expression profile in search for potential new druggable targets. METHODS: Twelve pericontusional tissues from severe TBI patients submitted to surgical treatment, and 20 control brain tissues from normal autopsy were analyzed for expression profile by real-time quantitative-polymerase chain reaction. CXCR7 and CXCR4 protein expressions were analyzed by immunohistochemistry. The findings were correlated with the clinical evolution. RESULTS: Increased gene expression of both receptors and their ligands was observed in TBI compared with controls, presenting high sensitivity and specificity to differentiate TBI from normal control (area under the curve ranging from 0.85 to 0.98, P < 0.001). In particular, CXCR7 expression highly correlated with CXCR4 and both ligands' expressions in TBI. Higher immunoreactions for CXCR7 and CXCR4 were identified in neurons and endothelial cells of TBI samples compared with controls. The patients presenting upregulated chemokine expression levels showed a trend toward favorable clinical evolution at up to 6 months of follow-up. CONCLUSIONS: The neuroprotective trend of CXCR4, CXCR7, CXCL11, and CXCL12 in TBI observed in this initial analysis warrants further studies with more patients, analyzing the involved signaling pathways for the development of new therapeutic strategies for TBI.


Subject(s)
Brain Contusion/genetics , Chemokine CXCL11/genetics , Chemokine CXCL12/genetics , Receptors, CXCR4/genetics , Receptors, CXCR/genetics , Adult , Aged , Brain Contusion/metabolism , Brain Injuries, Traumatic/genetics , Brain Injuries, Traumatic/metabolism , Case-Control Studies , Chemokine CXCL11/metabolism , Chemokine CXCL12/metabolism , Female , Gene Expression , Humans , Immunohistochemistry , Male , Middle Aged , RNA, Messenger/metabolism , Real-Time Polymerase Chain Reaction , Receptors, CXCR/metabolism , Receptors, CXCR4/metabolism , Young Adult
3.
J Neurosci Res ; 99(6): 1533-1549, 2021 06.
Article in English | MEDLINE | ID: mdl-33269491

ABSTRACT

Traumatic brain injury is a leading cause of mortality and morbidity in the United States. Acute trauma to the brain triggers chronic secondary injury mechanisms that contribute to long-term neurological impairment. We have developed a single, unilateral contusion injury model of sensorimotor dysfunction in adult mice. By targeting a topographically defined neurological circuit with a mild impact, we are able to track sustained behavioral deficits in sensorimotor function in the absence of tissue cavitation or neuronal loss in the contused cortex of these mice. Stereological histopathology and multiplex enzyme-linked immunosorbent assay proteomic screening confirm contusion resulted in chronic gliosis and the robust expression of innate immune cytokines and monocyte attractant chemokines IL-1ß, IL-5, IL-6, TNFα, CXCL1, CXCL2, CXCL10, CCL2, and CCL3 in the contused cortex. In contrast, the expression of neuroinflammatory proteins with adaptive immune functions was not significantly modulated by injury. Our data support widespread activation of innate but not adaptive immune responses, confirming an association between sensorimotor dysfunction with innate immune activation in the absence of tissue or neuronal loss in our mice.


Subject(s)
Adaptive Immunity/immunology , Brain Contusion/pathology , Cerebral Cortex/injuries , Inflammation Mediators/metabolism , Movement Disorders/etiology , Neuroinflammatory Diseases/metabolism , Neuroinflammatory Diseases/pathology , Neurons/pathology , Sensation Disorders/etiology , Animals , Brain Contusion/immunology , Brain Contusion/metabolism , Cerebral Cortex/metabolism , Cerebral Cortex/pathology , Chemokines/metabolism , Cytokines/metabolism , Disease Models, Animal , Female , Male , Mice , Mice, Inbred C57BL , Movement Disorders/immunology , Movement Disorders/pathology , Neuroinflammatory Diseases/immunology , Neurons/immunology , Neurons/metabolism , Sensation Disorders/immunology , Sensation Disorders/pathology
4.
J Neurophysiol ; 124(2): 536-543, 2020 08 01.
Article in English | MEDLINE | ID: mdl-32697670

ABSTRACT

Traumatic brain injury (TBI) is one of the most common neurological disorders causing memory reduction, particularly short-term memory (STM). We showed that, during TBI-induced inflammation, increased blood content of fibrinogen (Fg) enhanced vascular protein transcytosis and deposition of extravasated Fg in vasculo-astrocyte interfaces. In addition, we found that deposition of cellular prion protein (PrPC) was also increased in the vasculo-astrocyte endfeet interface. However, association of Fg and PrPC was not confirmed. Presently, we aimed to define whether Fg can associate with PrPC on astrocytes and cause their activation. Cultured mouse brain astrocytes were treated with medium alone (control), Fg (2 mg/mL or 4 mg/mL), 4 mg/mL of Fg in the presence of a function-blocking anti-PrPC peptide or anti-mouse IgG, function-blocking anti-PrPC peptide, or anti-mouse IgG alone. After treatment, either cell lysates were collected and analyzed via Western blot or coimmunoprecipitation was performed, or astrocytes were fixed and their activation was assessed with immunohistochemistry. Results showed that Fg dose-dependently activated astrocytes, increased expressions of PrPC and tyrosine (tropomyosin) receptor kinase B (TrkB), and PrP gene. Blocking the function of PrPC reduced these effects. Coimmunoprecipitation demonstrated Fg and PrPC association. Since it is known that prion protein has a greater effect on memory reduction than amyloid beta, and that activation of TrkB is involved in neurodegeneration, our findings confirming the possible formation of Fg-PrPC and Fg-induced overexpression of TrkB on astrocytes suggest a possible triggering mechanism for STM reduction that was seen previously during mild-to-moderate TBI.NEW & NOTEWORTHY For the first time we showed that fibrinogen (Fg) can associate with cellular prion protein (PrPC) on the surface of cultured mouse brain astrocytes. At high levels, Fg causes upregulation of astrocyte PrPC and astrocyte activation accompanied with overexpression of tyrosine receptor kinase B (TrkB), which results in nitric oxide (NO) production and generation of reactive oxygen species (ROS). Fg/PrPC interaction can be a triggering mechanism for TrkB-NO-ROS axis activation and the resultant astrocyte-mediated neurodegeneration.


Subject(s)
Astrocytes/metabolism , Brain Contusion , Cerebral Cortex , Fibrinogen/metabolism , Membrane Glycoproteins/metabolism , Nitric Oxide/metabolism , Prion Proteins/metabolism , Protein-Tyrosine Kinases/metabolism , Reactive Oxygen Species/metabolism , Animals , Brain Contusion/metabolism , Brain Contusion/pathology , Cells, Cultured , Cerebral Cortex/injuries , Cerebral Cortex/metabolism , Cerebral Cortex/pathology , Immunoglobulin G , Mice , Up-Regulation
5.
Neurocrit Care ; 32(1): 353-356, 2020 02.
Article in English | MEDLINE | ID: mdl-31342448

Subject(s)
Blood Flow Velocity , Brain Edema/physiopathology , Brain Injuries, Traumatic/physiopathology , Kidney Failure, Chronic/therapy , Middle Cerebral Artery/diagnostic imaging , Renal Dialysis/adverse effects , Status Epilepticus/physiopathology , Vascular Resistance , Aged , Blood-Brain Barrier/metabolism , Brain Contusion/complications , Brain Contusion/diagnostic imaging , Brain Contusion/metabolism , Brain Contusion/physiopathology , Brain Edema/diagnostic imaging , Brain Edema/etiology , Brain Edema/metabolism , Brain Injuries, Traumatic/complications , Brain Injuries, Traumatic/diagnostic imaging , Brain Injuries, Traumatic/metabolism , Cerebral Hemorrhage, Traumatic/complications , Cerebral Hemorrhage, Traumatic/diagnostic imaging , Cerebral Hemorrhage, Traumatic/metabolism , Cerebral Hemorrhage, Traumatic/physiopathology , Consciousness Disorders/etiology , Consciousness Disorders/metabolism , Consciousness Disorders/physiopathology , Headache/etiology , Headache/metabolism , Headache/physiopathology , Hematoma, Subdural, Acute/complications , Hematoma, Subdural, Acute/diagnostic imaging , Hematoma, Subdural, Acute/metabolism , Hematoma, Subdural, Acute/physiopathology , Humans , Kidney Failure, Chronic/complications , Kidney Failure, Chronic/metabolism , Male , Middle Cerebral Artery/physiopathology , Monitoring, Physiologic , Nausea/etiology , Nausea/metabolism , Nausea/physiopathology , Pulsatile Flow , Status Epilepticus/etiology , Status Epilepticus/metabolism , Ultrasonography, Doppler, Transcranial , Vomiting/etiology , Vomiting/metabolism , Vomiting/physiopathology
6.
Cell Transplant ; 28(9-10): 1183-1196, 2019.
Article in English | MEDLINE | ID: mdl-31177840

ABSTRACT

Traumatic brain injury (TBI), a major cause of mortality and morbidity, affects 10 million people worldwide, with limited treatment options. We have previously shown that (-)-phenserine (Phen), an acetylcholinesterase inhibitor originally designed and tested in clinical phase III trials for Alzheimer's disease, can reduce neurodegeneration after TBI and reduce cognitive impairments induced by mild TBI. In this study, we used a mouse model of moderate to severe TBI by controlled cortical impact to assess the effects of Phen on post-trauma histochemical and behavioral changes. Animals were treated with Phen (2.5 mg/kg, IP, BID) for 5 days started on the day of injury and the effects were evaluated by behavioral and histological examinations at 1 and 2 weeks after injury. Phen significantly attenuated TBI-induced contusion volume, enlargement of the lateral ventricle, and behavioral impairments in motor asymmetry, sensorimotor functions, motor coordination, and balance functions. The morphology of microglia was shifted to an active from a resting form after TBI, and Phen dramatically reduced the ratio of activated to resting microglia, suggesting that Phen also mitigates neuroinflammation after TBI. While Phen has potent anti-acetylcholinesterase activity, its (+) isomer Posiphen shares many neuroprotective properties but is almost completely devoid of anti-acetylcholinesterase activity. We evaluated Posiphen at a similar dose to Phen and found similar mitigation in lateral ventricular size increase, motor asymmetry, motor coordination, and balance function, suggesting the improvement of these histological and behavioral tests by Phen treatment occur via pathways other than anti-acetylcholinesterase inhibition. However, the reduction of lesion size and improvement of sensorimotor function by Posiphen were much smaller than with equivalent doses of Phen. Taken together, these results show that post-injury treatment with Phen over 5 days significantly ameliorates severity of TBI. These data suggest a potential development of this compound for clinical use in TBI therapy.


Subject(s)
Behavior, Animal/drug effects , Brain Contusion , Neuroprotective Agents/pharmacology , Physostigmine/analogs & derivatives , Animals , Brain Contusion/drug therapy , Brain Contusion/metabolism , Brain Contusion/pathology , Brain Contusion/physiopathology , Inflammation/drug therapy , Inflammation/metabolism , Inflammation/pathology , Inflammation/physiopathology , Male , Mice , Microglia/metabolism , Microglia/pathology , Physostigmine/pharmacology , Time Factors
7.
JCI Insight ; 52019 04 30.
Article in English | MEDLINE | ID: mdl-31038473

ABSTRACT

Traumatic brain injury (TBI) causes cortical dysfunction and can lead to post-traumatic epilepsy. Multiple studies demonstrate that GABAergic inhibitory network function is compromised following TBI, which may contribute to hyperexcitability and motor, behavioral, and cognitive deficits. Preserving the function of GABAergic interneurons, therefore, is a rational therapeutic strategy to preserve cortical function after TBI and prevent long-term clinical complications. Here, we explored an approach based on the ketogenic diet, a neuroprotective and anticonvulsant dietary therapy which results in reduced glycolysis and increased ketosis. Utilizing a pharmacologic inhibitor of glycolysis (2-deoxyglucose, or 2-DG), we found that acute in vitro application of 2-DG decreased the excitability of excitatory neurons, but not inhibitory interneurons, in cortical slices from naïve mice. Employing the controlled cortical impact (CCI) model of TBI in mice, we found that in vitro 2-DG treatment rapidly attenuated epileptiform activity seen in acute cortical slices 3 to 5 weeks after TBI. One week of in vivo 2-DG treatment immediately after TBI prevented the development of epileptiform activity, restored excitatory and inhibitory synaptic activity, and attenuated the loss of parvalbumin-expressing inhibitory interneurons. In summary, 2-DG may have therapeutic potential to restore network function following TBI.


Subject(s)
Antimetabolites/pharmacology , Brain Injuries, Traumatic/metabolism , Cerebral Cortex/drug effects , Cortical Excitability/drug effects , Deoxyglucose/pharmacology , Epilepsy, Post-Traumatic/metabolism , GABAergic Neurons/drug effects , Glycolysis/drug effects , Animals , Brain Contusion/metabolism , Cerebral Cortex/metabolism , Diet, Ketogenic , Disease Models, Animal , GABAergic Neurons/metabolism , In Vitro Techniques , Interneurons/drug effects , Interneurons/metabolism , Mice , Neural Inhibition/drug effects , Parvalbumins/metabolism
8.
Fa Yi Xue Za Zhi ; 35(2): 136-142, 2019 Apr.
Article in English, Chinese | MEDLINE | ID: mdl-31135105

ABSTRACT

ABSTRACT: Objective To investigate the expression of cannabinoid type 2 receptor (CB2R) at different time points after brain contusion and its relationship with wound age of mice. Methods A mouse brain contusion model was established with PCI3000 Precision Cortical Impactor. Expression changes of CB2R around the injured area were detected with immunohistochemical staining, immunofluorescent staining and Western blotting at different time points. Results Immunohistochemical staining results showed that only a few cells in the cerebral cortex of the sham operated group had CB2R positive expression. The ratio of CB2R positive cells gradually increased after injury and reached the peak twice at 12 h and 7 d post-injury, followed by a decrease to the normal level 28 d post-injury. The results of Western blotting were consistent with the immunohistochemical staining results. Immunofluorescent staining demonstrated that the changes of the ratio of CB2R positive cells in neurons, CB2R positive cells in monocytes and CB2R positive cells in astrocytes to the total cell number showed a single peak pattern, which peaked at 12 h, 1 d and 7 d post-injury, respectively. Conclusion The expression of CB2R after brain contusion in neurons, monocytes and astrocytes in mice suggests that it is likely to be involved in the regulation of the biological functions of those cells. The changes in CB2R are time-dependent, which suggests its potential applicability as a biological indicator for wound age estimation of brain contusion in forensic practice.


Subject(s)
Brain Contusion/metabolism , Brain Injuries , Muscle, Skeletal/metabolism , Receptor, Cannabinoid, CB2/metabolism , Wound Healing/physiology , Animals , Blotting, Western , Forensic Pathology , Mice , Muscle, Skeletal/pathology , Receptors, Cannabinoid , Time Factors
9.
Eur J Neurosci ; 50(2): 1981-1993, 2019 07.
Article in English | MEDLINE | ID: mdl-30828870

ABSTRACT

Nrf2 plays a pivotal role in antioxidant response and anti-inflammation after traumatic brain injury (TBI), and its deletion aggravates TBI-induced brain damage. Previous studies have demonstrated that Nrf2 is activated post TBI, but dynamic changes in expression and cell type-specific characteristics remain unclear. In this study, the Feeney weight-drop contusion model was conducted to mimic TBI, and the ipsilateral cerebral cortex was collected at 1, 3, 7 and 14 days post TBI (dpi). Nrf2 protein levels were observed by western blot. Cell type-specific localization of Nrf2 after TBI was detected at different time intervals by double immunofluorescence staining. NeuN, GFAP, IBA1 and NG2 were used as cell type-specific markers to neurons, astrocytes, microglia and NG2 glia, respectively. After TBI, Nrf2 protein levels peaked at 1 dpi. Robust transient Nrf2 accumulation was co-localized with neurons, which was predominant at 1 dpi. Continuous weak Nrf2 expression was detected in activated astrocytes, and the number of double positive cells peaked at 7 dpi. Inducible widespread immunostaining of Nrf2 was observed in the nucleus of the microglia, and the number of Nrf2+ microglia peaked at 7 dpi. In addition, we also explored colocalization of Nrf2 in NG2 glia, in which the percentage of Nrf2+ in NG2 glia reached a climax at 3 dpi. This study reveals that the accumulation of endogenous Nrf2 might mediate different pathophysical roles in neurons and glias after TBI, the cell-type specific and time-dependent expression provide insights to explain the roles of Nrf2 in different neural cells.


Subject(s)
Brain Contusion/metabolism , Cerebral Cortex/metabolism , NF-E2-Related Factor 2/metabolism , Neuroglia/metabolism , Neurons/metabolism , Animals , Disease Models, Animal , Male , Mice , Mice, Inbred C57BL
10.
J Neurotrauma ; 36(2): 370-379, 2019 01 15.
Article in English | MEDLINE | ID: mdl-29768967

ABSTRACT

Important differences in the biology of focal and diffuse traumatic brain injury (TBI) subtypes may result in unique pathophysiological responses to shared molecular mechanisms. Interleukin-1 (IL-1) signaling has been tested as a potential therapeutic target in preclinical models of cerebral contusion and diffuse TBI, and in a phase II clinical trial, but no published studies have examined IL-1 signaling in an impact/acceleration closed head injury (CHI) model. We hypothesized that genetic deletion of IL-1 receptor-1 (IL-1R1 KO) would be beneficial in focal (contusion) and CHI in mice. Wild type and IL-1R1 KO mice were subjected to controlled cortical impact (CCI), or to CHI. CCI produced brain leukocyte infiltration, HMGB1 translocation and release, edema, cell death, and cognitive deficits. CHI induced peak rotational acceleration of 9.7 × 105 ± 8.1 × 104 rad/s2, delayed time to righting reflex, and robust Morris water maze deficits without deficits in tests of anxiety, locomotion, sensorimotor function, or depression. CHI produced no discernable acute plasmalemma damage or cell death, blood-brain barrier permeability to IgG, or brain edema and only a modest increase in brain leukocyte infiltration at 72 h. In both models, mature (17 kDa) interleukin-1 beta (IL-1ß) was induced by 24 h in CD31+ endothelial cells isolated from injured brain but was not induced in CD11b+ cells in either model. High mobility group box protein-1 was released from injured brain cells in CCI but not CHI. Surprisingly, cognitive outcome in mice with global deletion of IL-1R1 was improved in CHI, but worse after CCI without affecting lesion size, edema, or infiltration of CD11b+/CD45+ leukocytes in CCI. IL-1R1 may induce unique biological responses, beneficial or detrimental to cognitive outcome, after TBI depending on the pathoanatomical subtype. Brain endothelium is a hitherto unrecognized source of mature IL-1ß in both models.


Subject(s)
Brain Concussion/metabolism , Brain Concussion/pathology , Brain Contusion/metabolism , Brain Contusion/pathology , Receptors, Interleukin-1/metabolism , Animals , Disease Models, Animal , Mice , Mice, Inbred C57BL , Mice, Knockout , Receptors, Interleukin-1/deficiency
11.
J Neurotrauma ; 36(7): 1060-1079, 2019 04 01.
Article in English | MEDLINE | ID: mdl-30160201

ABSTRACT

In severe traumatic brain injury (TBI), contusions often are worsened by contusion expansion or hemorrhagic progression of contusion (HPC), which may double the original contusion volume and worsen outcome. In humans and rodents with contusion-TBI, sulfonylurea receptor 1 (SUR1) is upregulated in microvessels and astrocytes, and in rodent models, blockade of SUR1 with glibenclamide reduces HPC. SUR1 does not function by itself, but must co-assemble with either KIR6.2 or transient receptor potential cation channel subfamily M member 4 (TRPM4) to form KATP (SUR1-KIR6.2) or SUR1-TRPM4 channels, with the two having opposite effects on membrane potential. Both KIR6.2 and TRPM4 are reportedly upregulated in TBI, especially in astrocytes, but the identity and function of SUR1-regulated channels post-TBI is unknown. Here, we analyzed human and rat brain tissues after contusion-TBI to characterize SUR1, TRPM4, and KIR6.2 expression, and in the rat model, to examine the effects on HPC of inhibiting expression of the three subunits using intravenous antisense oligodeoxynucleotides (AS-ODN). Glial fibrillary acidic protein (GFAP) immunoreactivity was used to operationally define core versus penumbral tissues. In humans and rats, GFAP-negative core tissues contained microvessels that expressed SUR1 and TRPM4, whereas GFAP-positive penumbral tissues contained astrocytes that expressed all three subunits. Förster resonance energy transfer imaging demonstrated SUR1-TRPM4 heteromers in endothelium, and SUR1-TRPM4 and SUR1-KIR6.2 heteromers in astrocytes. In rats, glibenclamide as well as AS-ODN targeting SUR1 and TRPM4, but not KIR6.2, reduced HPC at 24 h post-TBI. Our findings demonstrate upregulation of SUR1-TRPM4 and KATP after contusion-TBI, identify SUR1-TRPM4 as the primary molecular mechanism that accounts for HPC, and indicate that SUR1-TRPM4 is a crucial target of glibenclamide.


Subject(s)
Brain Contusion/metabolism , Intracranial Hemorrhages/metabolism , Potassium Channels, Inwardly Rectifying/metabolism , Sulfonylurea Receptors/metabolism , TRPM Cation Channels/metabolism , Adult , Aged , Animals , Brain/metabolism , Brain Contusion/complications , Disease Progression , Female , Glial Fibrillary Acidic Protein/metabolism , Humans , Intracranial Hemorrhages/etiology , Male , Middle Aged , Rats , Up-Regulation
12.
Journal of Forensic Medicine ; (6): 136-142, 2019.
Article in English | WPRIM (Western Pacific) | ID: wpr-984988

ABSTRACT

Objective To investigate the expression of cannabinoid type 2 receptor (CB2R) at different time points after brain contusion and its relationship with wound age of mice. Methods A mouse brain contusion model was established with PCI3000 Precision Cortical Impactor. Expression changes of CB2R around the injured area were detected with immunohistochemical staining, immunofluorescent staining and Western blotting at different time points. Results Immunohistochemical staining results showed that only a few cells in the cerebral cortex of the sham operated group had CB2R positive expression. The ratio of CB2R positive cells gradually increased after injury and reached the peak twice at 12 h and 7 d post-injury, followed by a decrease to the normal level 28 d post-injury. The results of Western blotting were consistent with the immunohistochemical staining results. Immunofluorescent staining demonstrated that the changes of the ratio of CB2R positive cells in neurons, CB2R positive cells in monocytes and CB2R positive cells in astrocytes to the total cell number showed a single peak pattern, which peaked at 12 h, 1 d and 7 d post-injury, respectively. Conclusion The expression of CB2R after brain contusion in neurons, monocytes and astrocytes in mice suggests that it is likely to be involved in the regulation of the biological functions of those cells. The changes in CB2R are time-dependent, which suggests its potential applicability as a biological indicator for wound age estimation of brain contusion in forensic practice.


Subject(s)
Animals , Mice , Blotting, Western , Brain Contusion/metabolism , Brain Injuries , Forensic Pathology , Muscle, Skeletal/pathology , Receptor, Cannabinoid, CB2/metabolism , Receptors, Cannabinoid , Time Factors , Wound Healing/physiology
13.
Sci Rep ; 8(1): 11201, 2018 07 25.
Article in English | MEDLINE | ID: mdl-30046063

ABSTRACT

There is a need for pharmaceutical agents that can reduce neuronal loss and improve functional deficits following traumatic brain injury (TBI). Previous research suggests that oxidative stress and mitochondrial dysfunction play a major role in neuronal damage after TBI. Therefore, this study aimed to investigate two drugs known to have antioxidant effects, L-carnitine and exendin-4, in rats with moderate contusive TBI. L-carnitine (1.5 mM in drinking water) or exendin-4 (15 µg/kg/day, ip) were given immediately after the injury for 2 weeks. Neurological function and brain histology were examined (24 h and 6 weeks post injury). The rats with TBI showed slight sensory, motor and memory functional deficits at 24 h, but recovered by 6 weeks. Both treatments improved sensory and motor functions at 24 h, while only exendin-4 improved memory. Both treatments reduced cortical contusion at 24 h and 6 weeks, however neither affected gliosis and inflammatory cell activation. Oxidative stress was alleviated and mitochondrial reactive oxygen species was reduced by both treatments, however only mitochondrial functional marker protein transporter translocase of outer membrane 20 was increased at 24 h post injury. In conclusion, L-carnitine and exendin-4 treatments immediately after TBI can improve neurological functional outcome and tissue integrity by reducing oxidative stress.


Subject(s)
Brain Contusion/drug therapy , Brain Injuries, Traumatic/drug therapy , Carnitine/administration & dosage , Exenatide/administration & dosage , Oxidative Stress/drug effects , Animals , Antioxidants/administration & dosage , Brain/drug effects , Brain/physiopathology , Brain Contusion/metabolism , Brain Contusion/physiopathology , Brain Injuries, Traumatic/metabolism , Brain Injuries, Traumatic/physiopathology , Disease Models, Animal , Mitochondria/drug effects , Mitochondria/genetics , Neurons/drug effects , Neurons/pathology , Rats , Rats, Sprague-Dawley , Reactive Oxygen Species/metabolism
14.
Mol Neurobiol ; 55(11): 8602-8611, 2018 Nov.
Article in English | MEDLINE | ID: mdl-29574629

ABSTRACT

Physical exercise (PE) is an effective method for improving cognitive function among patients with traumatic brain injury (TBI). We previously demonstrated that PE with an infrared-sensing running wheel (ISRW) system provides strong neuroprotection in an experimental animal model of stroke. In this study, we used fluid percussion injury in rats to simulate mild TBI. For rats, we used both passive avoidance learning and the Y-maze tests to evaluate cognitive function. We investigated whether PE rehabilitation attenuated cognitive deficits in rats with TBI and determined the contribution of hippocampal and cortical expression of heat shock protein 20 (HSP20) to PE-mediated cognitive recovery. In addition to increasing hippocampal and cortical expression of HSP20, brain-derived neurotrophic factor (BDNF), and the tropomyosin receptor kinase B (TrkB) ratio, PE rehabilitation significantly attenuated brain contusion and improved cognitive deficits in the rat model. Furthermore, reducing hippocampal and cortical expression of HSP20 with an intracerebral injection of pSUPER hsp20 small interfering RNA significantly diminished the PE-induced overexpression of hippocampal and cortical BDNF and the TrkB ratio and also reversed the beneficial effect of PE in reducing neurotrauma and the cognitive deficits. A positive Pearson correlation was found between HSP20 and BDNF, as well as between HSP20 and TrkB, in the hippocampal and cortical tissues. We thus conclude that post-ischaemic ISRW exercise rehabilitation attenuates cognitive deficits, as well as brain contusions, in TBI rats by stimulating the cerebral HSP20/BDNF/TrkB signalling axis.


Subject(s)
Brain Injuries, Traumatic/complications , Brain Injuries, Traumatic/rehabilitation , Brain-Derived Neurotrophic Factor/metabolism , Cognition Disorders/metabolism , Cognition Disorders/rehabilitation , HSP20 Heat-Shock Proteins/metabolism , Physical Conditioning, Animal , Receptor, trkB/metabolism , Animals , Brain Contusion/metabolism , Brain Contusion/pathology , Hippocampus/metabolism , Hippocampus/pathology , Male , RNA Interference , Rats, Sprague-Dawley , Signal Transduction
15.
Fa Yi Xue Za Zhi ; 33(3): 221-224, 2017 Jun.
Article in Chinese | MEDLINE | ID: mdl-29230982

ABSTRACT

OBJECTIVES: To observe the changes of cystathionine ß-synthase (CBS) expression in the cerebral cortex after brain contusion at different times. METHODS: An experimental model of traumatic brain injury (TBI) in mice was established by an improved weight-drop device. Then Western blotting and immunohistochemical examination were used to detect the CBS expression in cerebral cortex around injury at different time points (1 h, 6 h, 12 h, 1 d, 2 d, 3 d, 7 d). RESULTS: The results of Western blotting revealed that the expression level of CBS was down-regulated and reached its lowest level at the 3rd days after injury, and then restored to normal level after 7 days. The results of immunohistochemistry showed that CBS was present in the normal brain cortex. CBS expression gradually decreased at the 3rd days after injury, and then restored to normal level after 7 days. CONCLUSIONS: CBS has the potential to be a reference index for time estimation after brain contusion in forensic practice.


Subject(s)
Brain Contusion/metabolism , Cerebral Cortex/metabolism , Cystathionine beta-Synthase/metabolism , Animals , Blotting, Western , Brain , Brain Contusion/pathology , Brain Injuries/metabolism , Brain Injuries/pathology , Cerebral Cortex/pathology , Cystathionine beta-Synthase/genetics , Down-Regulation , Immunohistochemistry , Male , Mice , Time Factors
16.
J Neurotrauma ; 34(8): 1636-1644, 2017 04 15.
Article in English | MEDLINE | ID: mdl-27923323

ABSTRACT

Mild therapeutic hypothermia is a candidate for the treatment of traumatic brain injury (TBI). However, the role of mild hypothermia in neuronal sprouting after TBI remains obscure. We used a fluid percussion injury (FPI) model to assess the effect of mild hypothermia on pericontusion neuronal sprouting after TBI in rats. Male Sprague-Dawley rats underwent FPI or sham surgery, followed by mild hypothermia treatment (33°C) or normothermia treatment (37°C) for 3 h. All the rats were euthanized at 7 days after FPI. Neuronal sprouting that was confirmed by an increase in growth associated protein-43 (GAP-43) expression was evaluated using immunofluorescence and Western blot assays. The expression levels of several intrinsic and extrinsic sprouting-associated genes such as neurite outgrowth inhibitor A (NogoA), phosphatase and tensin homolog (PTEN), and suppressor of cytokine signaling 3 (SOCS3) were analyzed by quantitative real-time polymerase chain reaction (RT-PCR). Our results revealed that mild hypothermia significantly increased the expression level of GAP-43 and dramatically suppressed the expression level of interleukin-6 (IL-6) and SOCS3 at 7 days after FPI in the ipsilateral cortex compared with that of the normothermia TBI group. These data suggest that post-traumatic mild hypothermia promotes pericontusion neuronal sprouting after TBI. Moreover, the mechanism of hypothermia-induced neuronal sprouting might be partially associated with decreased levels of SOCS3.


Subject(s)
Brain Injuries, Traumatic/metabolism , Brain Injuries, Traumatic/therapy , Cerebral Cortex/metabolism , GAP-43 Protein/metabolism , Hypothermia, Induced/methods , Interleukin-6/metabolism , Neurons/metabolism , Suppressor of Cytokine Signaling 3 Protein/metabolism , Animals , Brain Contusion/metabolism , Brain Contusion/therapy , Disease Models, Animal , Male , Nogo Proteins/metabolism , PTEN Phosphohydrolase/metabolism , Rats , Rats, Sprague-Dawley
17.
Eur J Clin Invest ; 46(12): 1063-1069, 2016 Dec.
Article in English | MEDLINE | ID: mdl-27759956

ABSTRACT

BACKGROUND: Transforming growth factor-beta 1 (TGF-ß1) regulates many processes after traumatic brain injury (TBI). Both Neuro AiD™ (MLC601) and astragaloside (AST) attenuate microglia activation in rats with TBI. The purpose of this study was to investigate whether MLC601 or AST improves output of TBI by affecting microglial expression of TGF-ß1. MATERIALS AND METHODS: Adult male Sprague-Dawley rats (120 in number) were used to investigate the contribution of TGF-ß1-containing microglia in the MLC601-mediated or the AST-mediated neuroprotection in the brain trauma condition using lateral fluid percussion injury. RESULTS: Pearson correlation analysis revealed that there was a positive correlation between brain injury (evidenced by both brain contused volume and neurological severity score) and the cortical numbers of TGF-ß1-containing microglia for the rats (n = 12) 4 days post-TBI. MLC601 or AST significantly (P < 0·05) attenuated TBI-induced brain contused volume (119 ± 14 mm3 or 108 ± 11 mm3 vs. 160 ± 21 mm3 ), neurological severity score (7·8 ± 0·3 or 8·1 ± 0·4 vs. 10·2 ± 0·5) and numbers of TGF-ß1-containing microglia (6% ± 2% or 11% ± 3% vs. 79% ± 7%) for the rats 4 days post-TBI. CONCLUSIONS: There was a positive correlation between TBI and cortical numbers of TGF-ß1-containing microglia which could be significantly attenuated by astragaloside or NeuroAiD™ (MLC601) in rats.


Subject(s)
Brain Contusion/metabolism , Brain Injuries, Traumatic/metabolism , Brain/drug effects , Drugs, Chinese Herbal/pharmacology , Microglia/drug effects , Saponins/pharmacology , Transforming Growth Factor beta1/drug effects , Triterpenes/pharmacology , Animals , Brain/metabolism , Brain/pathology , Brain Contusion/pathology , Brain Contusion/physiopathology , Brain Injuries, Traumatic/pathology , Brain Injuries, Traumatic/physiopathology , Disease Models, Animal , Immunohistochemistry , Male , Microglia/metabolism , Microglia/pathology , Rats , Rats, Sprague-Dawley , Transforming Growth Factor beta1/metabolism
18.
J Neurochem ; 139(4): 659-675, 2016 11.
Article in English | MEDLINE | ID: mdl-27591733

ABSTRACT

Traumatic brain injury (TBI) leads to changes in ion fluxes, alterations in mitochondrial function, and increased generation of reactive oxygen species, resulting in secondary tissue damage. Mitochondria play important signaling roles in coordination of multiple metabolic platforms in addition to their well-known role in bioenergetics. Mitochondrial signaling strongly depends on cardiolipin (CL), a mitochondria-specific structurally unusual anionic phospholipid containing four fatty acyl chains. While our previous reports indicated that CL is selectively oxidized and presents itself as a target for the redox therapy following TBI, the topography of changes of CL in the injured brain remained to be defined. Here, we present a matrix-assisted laser desorption/ionization imaging study which reports regio-specific changes in CL, in a controlled cortical impact model of TBI in rats. Matrix-assisted laser desorption/ionization imaging revealed that TBI caused early decreases in CL in the contusional cortex, ipsilateral hippocampus, and thalamus with the most highly unsaturated CL species being most susceptible to loss. Phosphatidylinositol was the only other lipid species that exhibited a significant decrease, albeit to a lesser extent than CL. Signals for other lipids remained unchanged. This is the first study evaluating the spatial distribution of CL loss after acute brain injury. We propose that the CL loss may constitute an upstream mechanism for CL-driven signaling in different brain regions as an early response mechanism and may also underlie the bioenergetic changes that occur in hippocampal, cortical, and thalamic mitochondria after TBI.


Subject(s)
Brain Injuries, Traumatic/diagnostic imaging , Cardiolipins , Cerebral Cortex/diagnostic imaging , Hippocampus/diagnostic imaging , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization , Thalamus/diagnostic imaging , Animals , Brain Contusion/diagnostic imaging , Brain Contusion/metabolism , Brain Injuries, Traumatic/metabolism , Cardiolipins/metabolism , Cerebral Cortex/metabolism , Hippocampus/metabolism , Male , Rats , Rats, Sprague-Dawley , Thalamus/metabolism
19.
Acta Neurochir Suppl ; 122: 133-5, 2016.
Article in English | MEDLINE | ID: mdl-27165893

ABSTRACT

Controversy exists regarding the brain tissue oxygen (PbtO2) monitor's optimal tip location and what it actually measures. Recent work [2] identified a "PbtO2 change point" (CPPbt), below which PbtO2 displays pressure-passive behavior, showing significant correlation with optimal cerebral perfusion pressure (CPPopt) as defined by the pressure reactivity index (PRx). This would further support the concept of CPPopt [1] as an individualized target. We endeavored to validate these findings and further explore the relationship between PbtO2 and suboptimal CPP. CPPopt can be determined 55 % of the time [1]. It is undetermined whether PbtO2 can be an adjunctive modality for determining CPPopt.


Subject(s)
Brain Contusion/metabolism , Brain Injuries, Diffuse/metabolism , Brain Injuries, Traumatic/metabolism , Brain/metabolism , Cerebrovascular Circulation/physiology , Oxygen/metabolism , Adult , Arterial Pressure , Brain/blood supply , Brain Contusion/diagnostic imaging , Brain Injuries, Diffuse/diagnostic imaging , Brain Injuries, Traumatic/diagnostic imaging , Brain Injuries, Traumatic/physiopathology , Humans , Intracranial Pressure , Male , Middle Aged , Monitoring, Physiologic , Tomography, X-Ray Computed , Young Adult
20.
J Neurotrauma ; 33(8): 713-20, 2016 Apr 15.
Article in English | MEDLINE | ID: mdl-26426744

ABSTRACT

MicroRNAs (miRNAs) were recently identified as important regulators of gene expression under a wide range of physiological and pathophysiological conditions. Thus, they may represent a novel class of molecular targets for the management of traumatic brain injury (TBI). In this study, we investigated the temporal profile of miRNA expression during the development of secondary brain damage after experimental TBI. For this purpose, we used a controlled cortical impact model in C57Bl/6 mice (n = 6) to induce a cortical contusion and analyzed miRNA expression in the traumatized cortex by microarray analysis during the development of secondary contusion expansion-i.e., at 1, 6, and 12 h after TBI. Of a total 780 mature miRNA sequences analyzed, 410 were detected in all experimental groups. Of these, 158 miRNAs were significantly upregulated or downregulated in TBI compared with sham-operated animals, and 52 miRNAs increased more than twofold. We validated the upregulation of five of the most differentially expressed miRNAs (miR-21*, miR-144, miR-184, miR-451, miR-2137) and the downregulation of four of the most differentially expressed miRNAs (miR-107, miR-137, miR-190, miR-541) by quantitative polymerase chain reaction (qPCR). miR-2137, the most differentially expressed miRNA after TBI, was further investigated by in situ hybridization and was found to be upregulated in neurons within the traumatic penumbra. This study gives a comprehensive picture of miRNA expression levels during secondary contusion expansion after TBI and may pave the way for the identification of novel targets for the management of brain trauma.


Subject(s)
Brain Contusion/genetics , Brain Contusion/metabolism , MicroRNAs/biosynthesis , MicroRNAs/genetics , Parietal Lobe/metabolism , Animals , Brain Contusion/pathology , Brain Injuries, Traumatic/genetics , Brain Injuries, Traumatic/metabolism , Brain Injuries, Traumatic/pathology , Gene Expression , Male , Mice , Mice, Inbred C57BL , Parietal Lobe/pathology , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...