Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
1.
J Neurochem ; 168(9): 2935-2955, 2024 Sep.
Article in English | MEDLINE | ID: mdl-38943350

ABSTRACT

Astrocytes provide metabolic support to neurons, maintain ionic and water homeostasis, and uptake and recycle neurotransmitters. After exposure to the prototypical PAMP lipopolysaccharide (LPS), reactive astrocytes increase the expression of pro-inflammatory genes, facilitating neurodegeneration. In this study, we analyzed the expression of homeostatic genes in astrocytes exposed to LPS and identified the epigenetic factors contributing to the suppression of homeostatic genes in reactive astrocytes. Primary astrocytic cultures were acutely exposed to LPS and allowed to recover for 24, 72 h, and 7 days. As expected, LPS exposure induced reactive astrogliosis and increased the expression of pro-inflammatory IL-1B and IL-6. Interestingly, the acute exposure resulted in persistent hypermethylation of astroglial DNA. Similar hypermethylation was observed in highly reactive astrocytes from the traumatic brain injury (TBI) penumbra in vivo. Hypermethylation was accompanied by decreased expression of homeostatic genes including LDHA and Scl16a1 (MCT1) both involved in the lactate shuttle to neurons; glutamine synthase (GS) responsible for glutamate processing; Kcnj10 (Kir4.1) important for K+ homeostasis, and the water channel aquaporin-4 (Aqp4). Furthermore, the master regulator of DNA methylation, MAFG-1, as well as DNA methyl transferases DNMT1 and DNMT3a were overexpressed. The downregulation of homeostatic genes correlated with increased methylation of CpG islands in their promoters, as assessed by methylation-sensitive PCR and increased DNMT3a binding to the GS promoter. Treatment with decitabine, a DNMT inhibitor, prevented the LPS- and the HMGB-1-induced downregulation of homeostatic genes. Decitabine treatment also prevented the neurotoxic effects of these astrocytes in primary cortical cultures. In summary, our findings reveal that the pathological remodeling of reactive astrocytes encompasses not only the pro-inflammatory response but, significantly, also entails a long-term suppression of homeostatic gene expression with methylation of crucial CpG islands within their promoters.


Subject(s)
Astrocytes , DNA Methylation , Down-Regulation , Homeostasis , Astrocytes/metabolism , Astrocytes/drug effects , Astrocytes/pathology , DNA Methylation/drug effects , Animals , Homeostasis/drug effects , Down-Regulation/drug effects , Cells, Cultured , Lipopolysaccharides/pharmacology , Male , Mice , Brain Injuries, Traumatic/pathology , Brain Injuries, Traumatic/metabolism , Brain Injuries, Traumatic/genetics , Rats , Mice, Inbred C57BL
2.
Neurochem Int ; 176: 105742, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38641028

ABSTRACT

Traumatic brain injury (TBI) remains a major health concern which causes long-term neurological disability particularly in war veterans, athletes and young adults. In spite of intense clinical and research investigations, there is no effective therapy to cease the pathogenesis of the disease. It is believed that axonal injury during TBI is potentiated by neuroinflammation and demyelination and/or failure to remyelination. This study highlights the use of naturally available cinnamein, also chemically known as benzyl cinnamate, in inhibiting neuroinflammation, promoting remyelination and combating the disease process of controlled cortical impact (CCI)-induced TBI in mice. Oral delivery of cinnamein through gavage brought down the activation of microglia and astrocytes to decrease the expression of inducible nitric oxide synthase (iNOS), glial fibrillary acidic protein (GFAP) and ionized calcium binding adaptor molecule 1 (Iba1) in hippocampus and cortex of TBI mice. Cinnamein treatment also stimulated remyelination in TBI mice as revealed by PLP and A2B5 double-labeling, luxol fast blue (LFB) staining and axonal double-labeling for neurofilament and MBP. Furthermore, oral cinnamein reduced the size of lesion cavity in the brain, improved locomotor functions and restored memory and learning in TBI mice. These results suggest a new neuroprotective property of cinnamein that may be valuable in the treatment of TBI.


Subject(s)
Brain Injuries, Traumatic , Disease Models, Animal , Animals , Brain Injuries, Traumatic/drug therapy , Brain Injuries, Traumatic/pathology , Brain Injuries, Traumatic/metabolism , Mice , Male , Mice, Inbred C57BL , Cerebral Cortex/drug effects , Cerebral Cortex/metabolism , Cerebral Cortex/pathology , Neuroprotective Agents/pharmacology , Neuroprotective Agents/therapeutic use
3.
Mol Ther ; 31(5): 1313-1331, 2023 05 03.
Article in English | MEDLINE | ID: mdl-36739479

ABSTRACT

Astrocyte-microglial interaction plays a crucial role in brain injury-associated neuroinflammation. Our previous data illustrated that astrocytes secrete microRNA, leading to anti-inflammatory effects on microglia. Long non-coding RNAs participate in neuroinflammation regulation after traumatic brain injury. However, the effect of astrocytes on microglial phenotype via long non-coding RNAs and the underlying molecular mechanisms remain elusive. We used long non-coding RNA sequencing on murine astrocytes and found that exosomal long non-coding RNA 4933431K23Rik attenuated traumatic brain injury-induced microglial activation in vitro and in vivo and ameliorated cognitive function deficiency. Furthermore, microRNA and messenger RNA sequencing together with binding prediction illustrated that exosomal long non-coding RNA 4933431K23Rik up-regulates E2F7 and TFAP2C expression by sponging miR-10a-5p. Additionally, E2F7 and TFAP2C, as transcription factors, regulated microglial Smad7 expression. Using Cx3cr1-Smad7 overexpression of adeno-associated virus, microglia specifically overexpressed Smad7 in the attenuation of neuroinflammation, resulting in less cognitive deficiency after traumatic brain injury. Mechanically, overexpressed Smad7 physically binds to IκBα and inhibits its ubiquitination, preventing NF-κB signaling activation. The Smad7 activator asiaticoside alleviates neuroinflammation and protects neuronal function in traumatic brain injury mice. This study revealed that an exosomal long non-coding RNA from astrocytes attenuates microglial activation after traumatic brain injury by up-regulating Smad7, providing a potential therapeutic target.


Subject(s)
Brain Injuries, Traumatic , MicroRNAs , RNA, Long Noncoding , Mice , Animals , Microglia/metabolism , RNA, Long Noncoding/genetics , RNA, Long Noncoding/metabolism , Astrocytes/metabolism , Neuroinflammatory Diseases , MicroRNAs/metabolism , Brain Injuries, Traumatic/genetics , Brain Injuries, Traumatic/metabolism , Phenotype , Mice, Inbred C57BL
4.
Clinics (Sao Paulo) ; 76: e3131, 2021.
Article in English | MEDLINE | ID: mdl-34878029

ABSTRACT

OBJECTIVES: To investigate the molecular mechanism of edaravone (EDA) in improving the post-traumatic brain injury (TBI) dysfunction in learning and memory. METHODS: In vitro and in vivo TBI models were established using hydrogen peroxide (H2O2) treatment for hippocampal nerve stem cells (NSCs) and surgery for rats, followed by EDA treatment. WST 1 measurement, methylthiazol tetrazolium assay, and flow cytometry were performed to determine the activity, proliferation, and apoptosis of NSCs, and malondialdehyde (MDA), lactic dehydrogenase (LDH), and reactive oxygen species (ROS) detection kits were used to analyze the oxides in NSCs. RESULTS: Following EDA pretreatment, NSCs presented with promising resistance to H2O2-induced oxidative stress, whereas NSCs manifested significant increases in activity and proliferation and a decrease in apoptosis. Meanwhile, for NSCs, EDA pretreatment reduced the levels of MDA, LDH, and ROS, with a significant upregulation of Nrf2/antioxidant response element (ARE) signaling pathway, whereas for EDA-treated TBI rats, a significant reduction was observed in the trauma area and injury to the hippocampus, with improvement in memory and learning performance and upregulation of Nrf2/ARE signaling pathway. CONCLUSIONS: EDA, by regulating the activity of Nrf2/ARE signal pathway, can improve the TBI-induced injury to NSCs and learning and memory dysfunction in rats.


Subject(s)
Antioxidant Response Elements , Brain Injuries, Traumatic , Edaravone , Learning , Memory , NF-E2-Related Factor 2 , Animals , Apoptosis/drug effects , Brain Injuries, Traumatic/drug therapy , Brain Injuries, Traumatic/metabolism , Brain Injuries, Traumatic/physiopathology , Edaravone/pharmacology , Learning/drug effects , Memory/drug effects , NF-E2-Related Factor 2/metabolism , Oxidative Stress/drug effects , Rats , Reactive Oxygen Species/metabolism , Signal Transduction/drug effects
5.
Clinics ; Clinics;76: e3131, 2021. graf
Article in English | LILACS | ID: biblio-1350610

ABSTRACT

OBJECTIVES: To investigate the molecular mechanism of edaravone (EDA) in improving the post-traumatic brain injury (TBI) dysfunction in learning and memory. METHODS: In vitro and in vivo TBI models were established using hydrogen peroxide (H2O2) treatment for hippocampal nerve stem cells (NSCs) and surgery for rats, followed by EDA treatment. WST 1 measurement, methylthiazol tetrazolium assay, and flow cytometry were performed to determine the activity, proliferation, and apoptosis of NSCs, and malondialdehyde (MDA), lactic dehydrogenase (LDH), and reactive oxygen species (ROS) detection kits were used to analyze the oxides in NSCs. RESULTS: Following EDA pretreatment, NSCs presented with promising resistance to H2O2-induced oxidative stress, whereas NSCs manifested significant increases in activity and proliferation and a decrease in apoptosis. Meanwhile, for NSCs, EDA pretreatment reduced the levels of MDA, LDH, and ROS, with a significant upregulation of Nrf2/antioxidant response element (ARE) signaling pathway, whereas for EDA-treated TBI rats, a significant reduction was observed in the trauma area and injury to the hippocampus, with improvement in memory and learning performance and upregulation of Nrf2/ARE signaling pathway. CONCLUSIONS: EDA, by regulating the activity of Nrf2/ARE signal pathway, can improve the TBI-induced injury to NSCs and learning and memory dysfunction in rats.


Subject(s)
Animals , Rats , Antioxidant Response Elements , Brain Injuries, Traumatic/physiopathology , Brain Injuries, Traumatic/metabolism , Brain Injuries, Traumatic/drug therapy , Edaravone/pharmacology , Learning/drug effects , Signal Transduction/drug effects , Reactive Oxygen Species/metabolism , Apoptosis/drug effects , Oxidative Stress/drug effects , NF-E2-Related Factor 2/metabolism , Memory/drug effects
6.
J Pediatr ; 227: 170-175, 2020 12.
Article in English | MEDLINE | ID: mdl-32622673

ABSTRACT

OBJECTIVE: To examine levels of plasma osteopontin (OPN), a recently described neuroinflammatory biomarker, in children with abusive head trauma (AHT) compared with children with other types of traumatic brain injury (TBI). STUDY DESIGN: The study cohort comprised children aged <4 years diagnosed with TBI and seen in the intensive care unit in a tertiary children's hospital. Patients were classified as having confirmed or suspected AHT or TBI by other mechanisms (eg, motor vehicle accidents), as identified by a Child Protection Team clinician. Serial blood samples were collected at admission and at 24, 48, and 72 hours after admission. Levels of OPN were compared across groups. RESULTS: Of 77 patients identified, 24 had confirmed AHT, 12 had suspected AHT, and 41 had TBI. There were no differences in the Glasgow Coma Scale score between the patients with confirmed AHT and those with suspected AHT and those with TBI (median score, 4.5 vs 4 and 7; P = .39). At admission to the emergency department, OPN levels were significantly higher in children with confirmed AHT compared with the other 2 groups (mean confirmed AHT, 471.5 ng/mL; median suspected AHT, 322.3 ng/mL; mean TBI, 278.0 ng/mL; P = .03). Furthermore, the adjusted mean trajectory levels of OPN were significantly higher in the confirmed AHT group compared with the other 2 groups across all subsequent time points (P = <.01). CONCLUSIONS: OPN is significantly elevated in children with confirmed AHT compared with those with suspected AHT and those with other types of TBI. OPN expression may help identify children with suspected AHT to aid resource stratification and triage of appropriate interventions for children who are potential victims of abuse.


Subject(s)
Brain Injuries, Traumatic/blood , Child Abuse , Craniocerebral Trauma/blood , Osteopontin/blood , Brain Injuries, Traumatic/diagnosis , Brain Injuries, Traumatic/metabolism , Child Abuse/diagnosis , Child, Preschool , Craniocerebral Trauma/diagnosis , Craniocerebral Trauma/metabolism , Female , Humans , Infant , Male , Osteopontin/biosynthesis , Prospective Studies
7.
Neurosci Biobehav Rev ; 102: 345-361, 2019 07.
Article in English | MEDLINE | ID: mdl-31102601

ABSTRACT

Traumatic brain injury (TBI) is a devastating condition which often initiates a sequel of neurological disorders that can last throughout lifespan. From metabolic perspective, TBI also compromises systemic physiology including the function of body organs with subsequent malfunctions in metabolism. The emerging panorama is that the effects of TBI on the periphery strike back on the brain and exacerbate the overall TBI pathogenesis. An increasing number of clinical reports are alarming to show that metabolic dysfunction is associated with incidence of long-term neurological and psychiatric disorders. The autonomic nervous system, associated hypothalamic-pituitary axis, and the immune system are at the center of the interface between brain and body and are central to the regulation of overall homeostasis and disease. We review the strong association between mechanisms that regulate cell metabolism and inflammation which has important clinical implications for the communication between body and brain. We also discuss the integrative actions of lifestyle interventions such as diet and exercise on promoting brain and body health and cognition after TBI.


Subject(s)
Autonomic Nervous System Diseases , Brain Injuries, Traumatic , Inflammation , Metabolic Syndrome , Neuronal Plasticity , Autonomic Nervous System Diseases/etiology , Autonomic Nervous System Diseases/immunology , Autonomic Nervous System Diseases/metabolism , Autonomic Nervous System Diseases/physiopathology , Brain Injuries, Traumatic/complications , Brain Injuries, Traumatic/immunology , Brain Injuries, Traumatic/metabolism , Brain Injuries, Traumatic/physiopathology , Humans , Inflammation/etiology , Inflammation/immunology , Inflammation/metabolism , Inflammation/physiopathology , Metabolic Syndrome/etiology , Metabolic Syndrome/immunology , Metabolic Syndrome/metabolism , Metabolic Syndrome/physiopathology , Neuronal Plasticity/physiology
8.
Eur J Pharmacol ; 854: 387-397, 2019 Jul 05.
Article in English | MEDLINE | ID: mdl-30807746

ABSTRACT

Traumatic brain injury (TBI) is a public health problem characterized by a combination of immediate mechanical dysfunction of the brain tissue, and secondary damage. Based on the hypothesis that selected targets, such as Na+ K+-ATPase are involved in the secondary damage after TBI and modulation of this enzyme activity by triterpene 3ß, 6ß, 16ß-trihidroxilup-20 (29)-ene (TTHL) supports the ethnomedical applications of this plant, we decided to investigate whether previous TTHL treatment interrupts the progression of pathophysiology induced by TBI. Statistical analyses revealed that percussion fluid injury (FPI) increased Na+,K+-ATPase activity in all isoform (α1 and α2/3) 15 min after neuronal injury. The FPI protocol inhibited Na+,K+-ATPase activity total and α1 isoform, increased [3H]MK-801 binding but did not alter Dichloro-dihydro-fluorescein diacetate (DCFH-DA) oxidation, carbonylated proteins and free -SH groups 60 min after injury. The increase of immunoreactivity of protein PKC and state of phosphorylation of at Ser16 of Na+,K+-ATPase 60 min after FPI suggest the involvement of PKC on Na+,K+-ATPase activity oscillations characterized by inhibition of total and α1 isoform. Our experimental data also revealed that natural product rich in compounds such as triterpenes (TTHL; 30 mg/kg) attenuates [3H]MK-801 binding increase, phosphorylation of the PKC and the Na+,K+-ATPase alpha 1 subunit (Ser16) induced by FPI. The previous TTHL treatment had not effect on motor disability but protected against spatial memory deficit, BDNF, TrKB expression decrease, protein carbonylation and hippocampal cell death 7 days after FPI. These data suggest that TTHL-induced reduction on initial damage limits the long-term secondary degeneration and supports neural repair or behavioral compensation after neuronal injury.


Subject(s)
Brain Injuries, Traumatic/pathology , Brain Injuries, Traumatic/physiopathology , Sodium-Potassium-Exchanging ATPase/metabolism , Triterpenes/pharmacology , Animals , Brain Injuries, Traumatic/drug therapy , Brain Injuries, Traumatic/metabolism , Cell Count , Cognition/drug effects , Male , Mice , Motor Activity/drug effects , Neuronal Plasticity/drug effects , Neurons/drug effects , Neurons/pathology , Oxidative Stress/drug effects , Time Factors , Triterpenes/therapeutic use
9.
Neuroendocrinology ; 108(2): 142-160, 2019.
Article in English | MEDLINE | ID: mdl-30391959

ABSTRACT

Traumatic brain injury (TBI) is a serious public health problem. It may result in severe neurological disabilities and in a variety of cellular metabolic alterations for which available therapeutic strategies are limited. In the last decade, the use of estrogenic compounds, which activate protective mechanisms in astrocytes, has been explored as a potential experimental therapeutic approach. Previous works have suggested estradiol (E2) as a neuroprotective hormone that acts in the brain by binding to estrogen receptors (ERs). Several steroidal and nonsteroidal estrogenic compounds can imitate the effects of estradiol on ERs. These include hormonal estrogens, phytoestrogens and synthetic estrogens, such as selective ER modulators or tibolone. Current evidence of the role of astrocytes in mediating protective actions of estrogenic compounds after TBI is reviewed in this paper. We conclude that the use of estrogenic compounds to modulate astrocytic properties is a promising therapeutic approach for the treatment of TBI.


Subject(s)
Astrocytes/drug effects , Brain Injuries, Traumatic/drug therapy , Estrogens/therapeutic use , Neuroprotective Agents/therapeutic use , Receptors, Estrogen/metabolism , Animals , Astrocytes/metabolism , Brain Injuries, Traumatic/metabolism , Estrogens/pharmacology , Humans , Neuroprotective Agents/pharmacology
10.
Crit Care Med ; 46(9): 1471-1479, 2018 09.
Article in English | MEDLINE | ID: mdl-29742587

ABSTRACT

OBJECTIVES: To employ metabolomics-based pathway and network analyses to evaluate the cerebrospinal fluid metabolome after severe traumatic brain injury in children and the capacity of combination therapy with probenecid and N-acetylcysteine to impact glutathione-related and other pathways and networks, relative to placebo treatment. DESIGN: Analysis of cerebrospinal fluid obtained from children enrolled in an Institutional Review Board-approved, randomized, placebo-controlled trial of a combination of probenecid and N-acetylcysteine after severe traumatic brain injury (Trial Registration NCT01322009). SETTING: Thirty-six-bed PICU in a university-affiliated children's hospital. PATIENTS AND SUBJECTS: Twelve children 2-18 years old after severe traumatic brain injury and five age-matched control subjects. INTERVENTION: Probenecid (25 mg/kg) and N-acetylcysteine (140 mg/kg) or placebo administered via naso/orogastric tube. MEASUREMENTS AND MAIN RESULTS: The cerebrospinal fluid metabolome was analyzed in samples from traumatic brain injury patients 24 hours after the first dose of drugs or placebo and control subjects. Feature detection, retention time, alignment, annotation, and principal component analysis and statistical analysis were conducted using XCMS-online. The software "mummichog" was used for pathway and network analyses. A two-component principal component analysis revealed clustering of each of the groups, with distinct metabolomics signatures. Several novel pathways with plausible mechanistic involvement in traumatic brain injury were identified. A combination of metabolomics and pathway/network analyses showed that seven glutathione-centered pathways and two networks were enriched in the cerebrospinal fluid of traumatic brain injury patients treated with probenecid and N-acetylcysteine versus placebo-treated patients. Several additional pathways/networks consisting of components that are known substrates of probenecid-inhibitable transporters were also identified, providing additional mechanistic validation. CONCLUSIONS: This proof-of-concept neuropharmacometabolomics assessment reveals alterations in known and previously unidentified metabolic pathways and supports therapeutic target engagement of the combination of probenecid and N-acetylcysteine treatment after severe traumatic brain injury in children.


Subject(s)
Acetylcysteine/therapeutic use , Brain Injuries, Traumatic/cerebrospinal fluid , Brain Injuries, Traumatic/drug therapy , Probenecid/therapeutic use , Adjuvants, Pharmaceutic , Adolescent , Brain Injuries, Traumatic/metabolism , Child , Child, Preschool , Double-Blind Method , Drug Therapy, Combination , Humans , Injury Severity Score , Metabolomics
11.
BMC Res Notes ; 11(1): 150, 2018 Feb 21.
Article in English | MEDLINE | ID: mdl-29467028

ABSTRACT

OBJECTIVE: Data from our laboratory suggest that recovery from a traumatic brain injury depends on the time of day at which it occurred. In this study, we examined whether traumatic brain injury -induced damage is related to circadian variation in N-methyl-D-aspartate receptor expression in rat cortex. RESULTS: We confirmed that traumatic brain injury recovery depended on the time of day at which the damage occurred. We also found that motor cortex N-methyl-D-aspartate receptor subunit NR1 expression exhibited diurnal variation in both control and traumatic brain injury-subjected rats. However, this rhythm is more pronounced in traumatic brain injury-subjected rats, with minimum expression in those injured during nighttime hours. These findings suggest that traumatic brain injury occurrence times should be considered in future clinical studies and when designing neuroprotective strategies for patients.


Subject(s)
Brain Injuries, Traumatic/metabolism , Brain Injuries, Traumatic/physiopathology , Circadian Rhythm/physiology , Motor Cortex/injuries , Motor Cortex/metabolism , Motor Cortex/physiopathology , Receptors, N-Methyl-D-Aspartate/metabolism , Animals , Disease Models, Animal , Male , Rats , Rats, Wistar , Time Factors
12.
Behav Brain Res ; 336: 145-150, 2018 01 15.
Article in English | MEDLINE | ID: mdl-28842271

ABSTRACT

The sensorimotor cortex and the striatum are interconnected by the corticostriatal pathway, suggesting that cortical injury alters the striatal function that is associated with skilled movements and motor learning, which are functions that may be modulated by dopamine (DA). In this study, we explored motor coordination and balance in order to investigate whether the activation of D1 receptors (D1Rs) modulates functional recovery after cortical injury. The results of the beam-walking test showed motor deficit in the injured group at 24, 48 and 96h post-injury, and the recovery time was observed at 192h after cortical injury. In the sham and injured rats, systemic administration of the D1R antagonist SCH-23390 (1mg/kg) alone at 24, 48, 96 and 192h significantly (P<0.01) increased the motor deficit, while administration of the D1R agonist SKF-38393 alone (2, 3 and 4mg/kg) at 24, 48, 96 and 192h post-injury did not produce a significant difference; however, the co-administration of SKF-38393 and SCH-23390 prevented the antagonist-induced increase in the motor deficit. The cortical+striatal injury showed significantly increased the motor deficit at 24, 48, 96 and 192h post-injury (P<0.01) but did not show recovery at 192h. In conclusion, the administration of the D1R agonist did not accelerate the motor recovery, but the activation of D1Rs maintained motor coordination, confirming that an intact striatum may be necessary for achieving recovery.


Subject(s)
Receptors, Dopamine D1/metabolism , Receptors, Dopamine D1/physiology , Sensorimotor Cortex/physiology , 2,3,4,5-Tetrahydro-7,8-dihydroxy-1-phenyl-1H-3-benzazepine/metabolism , 2,3,4,5-Tetrahydro-7,8-dihydroxy-1-phenyl-1H-3-benzazepine/pharmacology , Animals , Benzazepines/metabolism , Benzazepines/pharmacology , Brain Injuries, Traumatic/metabolism , Brain Injuries, Traumatic/physiopathology , Corpus Striatum/metabolism , Disease Models, Animal , Dopamine/metabolism , Dopamine Antagonists/pharmacology , Male , Motor Cortex/physiopathology , Neostriatum/metabolism , Rats , Rats, Wistar , Receptors, Dopamine D1/agonists , Receptors, Dopamine D2/metabolism , Sensorimotor Cortex/metabolism
13.
Brain Res ; 1663: 78-86, 2017 05 15.
Article in English | MEDLINE | ID: mdl-28288867

ABSTRACT

Moderate traumatic brain injury (TBI) might increase the vulnerability to neuronal neurodegeneration, but the basis of such selective neuronal susceptibility has remained elusive. In keeping with the disruption of the blood-brain barrier (BBB) caused by TBI, changes in BBB permeability following brain injury could facilitate the access of xenobiotics into the brain. To test this hypothesis, here we evaluated whether TBI would increase the susceptibility of nigrostriatal dopaminergic fibers to the systemic administration of 6-hydroxydopamine (6-OHDA), a classic neurotoxin used to trigger a PD-like phenotype in mice, but that in normal conditions is unable to cross the BBB. Adult Swiss mice were submitted to a moderate TBI using a free weight-drop device and, 5h later, they were injected intraperitoneally with a single dose of 6-OHDA (100mg/kg). Afterwards, during a period of 4weeks, the mice were submitted to a battery of behavioral tests, including the neurological severity score (NSS), the open field and the rotarod. Animals from the TBI plus 6-OHDA group displayed significant motor and neurological impairments that were improved by acute l-DOPA administration (25mg/kg, i.p.). Moreover, the observation of the motor deficits correlates with (i) a significant decrease in the tyrosine hydroxylase levels mainly in the rostral striatum and (ii) a significant increase in the levels of striatal glial fibrillary acidic protein (GFAP) levels. On the whole, the present findings demonstrate that a previous moderate TBI event increases the susceptibility to motor, neurological and neurochemical alterations induced by systemic administration of the dopaminergic neurotoxin 6-OHDA in mice.


Subject(s)
Brain Injuries, Traumatic/metabolism , Oxidopamine/toxicity , Animals , Behavior, Animal , Blood-Brain Barrier/metabolism , Brain/metabolism , Brain Injuries/metabolism , Brain Injuries, Traumatic/complications , Brain Injuries, Traumatic/pathology , Corpus Striatum/metabolism , Disease Models, Animal , Dopamine/metabolism , Dopaminergic Neurons/drug effects , Glial Fibrillary Acidic Protein/metabolism , Levodopa/metabolism , Mice , Neurodegenerative Diseases , Neurotoxicity Syndromes/metabolism , Oxidopamine/metabolism , Tyrosine 3-Monooxygenase/metabolism
14.
J Neurotrauma ; 34(7): 1318-1328, 2017 04 01.
Article in English | MEDLINE | ID: mdl-27931151

ABSTRACT

Traumatic brain injury (TBI) is a highly complex multi-factorial disorder. Experimental trauma involves primary and secondary injury cascades that underlie delayed neuronal dysfunction and death. Mitochondrial dysfunction and glutamatergic excitotoxicity are the hallmark mechanisms of damage. Accordingly, a successful pharmacological intervention requires a multi-faceted approach. Guanosine (GUO) is known for its neuromodulator effects in various models of brain pathology, specifically those that involve the glutamatergic system. The aim of the study was to investigate the GUO effects against mitochondrial damage in hippocampus and cortex of rats subjected to TBI, as well as the relationship of this effect with the glutamatergic system. Adult male Wistar rats were subjected to a unilateral moderate fluid percussion brain injury (FPI) and treated 15 min later with GUO (7.5 mg/kg) or vehicle (saline 0.9%). Analyses were performed in hippocampus and cortex 3 h post-trauma and revealed significant mitochondrial dysfunction, characterized by a disrupted membrane potential, unbalanced redox system, decreased mitochondrial viability, and complex I inhibition. Further, disruption of Ca2+ homeostasis and increased mitochondrial swelling was also noted. Our results showed that mitochondrial dysfunction contributed to decreased glutamate uptake and levels of glial glutamate transporters (glutamate transporter 1 and glutamate aspartate transporter), which leads to excitotoxicity. GUO treatment ameliorated mitochondrial damage and glutamatergic dyshomeostasis. Thus, GUO might provide a new efficacious strategy for the treatment acute physiological alterations secondary to TBI.


Subject(s)
Amino Acid Transport System X-AG/metabolism , Brain Injuries, Traumatic , Glutamic Acid/metabolism , Guanosine/pharmacology , Mitochondrial Diseases , Animals , Brain Injuries, Traumatic/complications , Brain Injuries, Traumatic/drug therapy , Brain Injuries, Traumatic/metabolism , Disease Models, Animal , Guanosine/administration & dosage , Male , Mitochondrial Diseases/drug therapy , Mitochondrial Diseases/etiology , Mitochondrial Diseases/metabolism , Mitochondrial Diseases/physiopathology , Oxidation-Reduction/drug effects , Rats , Rats, Wistar
15.
Mol Neurobiol ; 54(10): 7585-7596, 2017 12.
Article in English | MEDLINE | ID: mdl-27830534

ABSTRACT

Traumatic brain injury (TBI) is one of the most common types of brain injuries that cause death or persistent neurological disturbances in survivors. Most of the promising experimental drugs were not effective in clinical trials; therefore, the development of TBI drugs represents a huge unmet need. Guanosine, an endogenous neuroprotective nucleoside, has not been evaluated in TBI to the best of our knowledge. Therefore, the present study evaluated the effect of guanosine on TBI-induced neurological damage. Our findings showed that a single dose of guanosine (7.5 mg/kg, intraperitoneally (i.p.) injected 40 min after fluid percussion injury (FPI) in rats protected against locomotor and exploratory impairments 8 h after injury. The treatment also protected against neurochemical damage to the ipsilateral cortex, glutamate uptake, Na+/K+-ATPase, glutamine synthetase activity, and alterations in mitochondrial function. The inflammatory response and brain edema were also reduced by this nucleoside. In addition, guanosine protected against neuronal death and caspase 3 activation. Therefore, this study suggests that guanosine plays a neuroprotective role in TBI and can be exploited as a new pharmacological strategy.


Subject(s)
Brain Injuries, Traumatic/prevention & control , Guanosine/therapeutic use , Inflammation Mediators/antagonists & inhibitors , Mitochondria/drug effects , Neurons/drug effects , Neuroprotective Agents/therapeutic use , Animals , Brain Injuries, Traumatic/metabolism , Brain Injuries, Traumatic/pathology , Cell Count/methods , Cerebral Cortex/drug effects , Cerebral Cortex/metabolism , Cerebral Cortex/pathology , Guanosine/pharmacology , Inflammation/metabolism , Inflammation/pathology , Inflammation/prevention & control , Inflammation Mediators/metabolism , Male , Mitochondria/metabolism , Mitochondria/pathology , Neurons/metabolism , Neurons/pathology , Neuroprotective Agents/pharmacology , Rats , Rats, Wistar
16.
Genet Mol Res ; 15(1)2016 Jan 29.
Article in English | MEDLINE | ID: mdl-26909929

ABSTRACT

We investigated the effects of hyperbaric oxygen treatment on the Nrf2 signaling pathway in secondary injury following traumatic brain injury, using a rat model. An improved Feeney freefall method was used to establish the rat traumatic brain injury model. Sixty rats were randomly divided into three groups: a sham surgery group, a traumatic brain injury group, and a group receiving hyperbaric oxygen treatment after traumatic brain injury. Neurological function scores were assessed at 12 and 24 h after injury. The expression levels of Nrf2, heme oxygenase 1 (HO-1), and quinine oxidoreductase 1 (NQO-1) in the cortex surrounding the brain lesion were detected by western blotting 24 h after the injury. Additionally, the TUNEL method was used to detect apoptosis of nerve cells 24 h after traumatic injury and Nissl staining was used to detect the number of whole neurons. Hyperbaric oxygen treatment significantly increased the expression of nuclear Nrf2 protein (P < 0.05), HO-1, and NQO-1 in the brain tissues surrounding the lesion after a traumatic brain injury (P < 0.05) and also significantly reduced the number of apoptotic and injured nerve cells. The neurological function scores also improved with hyperbaric oxygen treatment (P < 0.05). Therefore, hyperbaric oxygen has a neuroprotective role in traumatic brain injury, which is mediated by up-regulation of the Nrf2 signaling pathway.


Subject(s)
Brain Injuries, Traumatic , Cerebral Cortex , Hyperbaric Oxygenation , NF-E2-Related Factor 2 , Signal Transduction , Up-Regulation , Animals , Male , Rats , Apoptosis , Brain Injuries, Traumatic/metabolism , Brain Injuries, Traumatic/therapy , Cerebral Cortex/metabolism , Cerebral Cortex/physiopathology , Disease Models, Animal , Heme Oxygenase (Decyclizing)/genetics , NAD(P)H Dehydrogenase (Quinone)/genetics , Neurons/metabolism , Neurons/physiology , NF-E2-Related Factor 2/genetics
17.
J Neurotrauma ; 33(14): 1317-30, 2016 07 15.
Article in English | MEDLINE | ID: mdl-26651029

ABSTRACT

Throughout the world, traumatic brain injury (TBI) is one of the major causes of disability, which can include deficits in motor function and memory, as well as acquired epilepsy. Although some studies have shown the beneficial effects of physical exercise after TBI, the prophylactic effects are poorly understood. In the current study, we demonstrated that TBI induced by fluid percussion injury (FPI) in adult male Wistar rats caused early motor impairment (24 h), learning deficit (15 days), spontaneous epileptiform events (SEE), and hilar cell loss in the hippocampus (35 days) after TBI. The hippocampal alterations in the redox status, which were characterized by dichlorofluorescein diacetate oxidation and superoxide dismutase (SOD) activity inhibition, led to the impairment of protein function (Na(+), K(+)-adenosine triphosphatase [ATPase] activity inhibition) and glutamate uptake inhibition 24 h after neuronal injury. The molecular adaptations elicited by previous swim training protected against the glutamate uptake inhibition, oxidative stress, and inhibition of selected targets for free radicals (e.g., Na(+), K(+)-ATPase) 24 h after neuronal injury. Our data indicate that this protocol of exercise protected against FPI-induced motor impairment, learning deficits, and SEE. In addition, the enhancement of the hippocampal phosphorylated nuclear factor erythroid 2-related factor (P-Nrf2)/Nrf2, heat shock protein 70, and brain-derived neurotrophic factor immune content in the trained injured rats suggests that protein expression modulation associated with an antioxidant defense elicited by previous physical exercise can prevent toxicity induced by TBI, which is characterized by cell loss in the dentate gyrus hilus at 35 days after TBI. Therefore, this report suggests that previous physical exercise can decrease lesion progression in this model of brain damage.


Subject(s)
Behavior, Animal/physiology , Brain Injuries, Traumatic/metabolism , Cognitive Dysfunction/metabolism , Dentate Gyrus/metabolism , Epilepsy/metabolism , Movement Disorders/metabolism , Oxidation-Reduction , Physical Conditioning, Animal/physiology , Signal Transduction/physiology , Animals , Brain Injuries, Traumatic/complications , Cognitive Dysfunction/etiology , Cognitive Dysfunction/prevention & control , Dentate Gyrus/pathology , Disease Models, Animal , Epilepsy/etiology , Epilepsy/prevention & control , Learning/physiology , Male , Movement Disorders/etiology , Movement Disorders/prevention & control , Rats , Rats, Wistar
18.
Mol Neurobiol ; 53(4): 2297-311, 2016 May.
Article in English | MEDLINE | ID: mdl-25976365

ABSTRACT

The central nervous system (CNS) is highly sensitive to external mechanical damage, presenting a limited capacity for regeneration explained in part by its inability to restore either damaged neurons or the synaptic network. The CNS may suffer different types of external injuries affecting its function and/or structure, including stroke, spinal cord injury, and traumatic brain injury. These pathologies critically affect the quality of life of a large number of patients worldwide and are often fatal because available therapeutics are ineffective and produce limited results. Common effects of the mentioned pathologies involves the triggering of several cellular and metabolic responses against injury, including infiltration of blood cells, inflammation, glial activation, and neuronal death. Although some of the underlying molecular mechanisms of those responses have been elucidated, the mechanisms driving these processes are poorly understood in the context of CNS injury. In the last few years, it has been suggested that the activation of the Wnt signaling pathway could be important in the regenerative response after CNS injury, activating diverse protective mechanisms including the stimulation of neurogenesis, blood brain structure consolidation and the recovery of cognitive brain functions. Because Wnt signaling is involved in several physiological processes, the putative positive role of its activation after injury could be the basis for novel therapeutic approaches to CNS injury.


Subject(s)
Central Nervous System/injuries , Central Nervous System/metabolism , Wnt Signaling Pathway , Animals , Brain Injuries, Traumatic/metabolism , Brain Injuries, Traumatic/pathology , Humans , Neurodegenerative Diseases/metabolism , Neurodegenerative Diseases/pathology , Spinal Cord Injuries/pathology
19.
Rev. chil. pediatr ; 82(3): 175-190, jun. 2011. ilus, tab
Article in Spanish | LILACS | ID: lil-608818

ABSTRACT

Traumatic Brain Injury (TBI) is an important Public Health issue in Chile and the world. It represents a frequent cause of consultation, constituting a significant cause of morbidity and mortality in the population under 45 years of age. Accidents are the main reason for TBI among the pediatric population, but child abuse is an important cause in children below 2 y.o. A proper evaluation is essential to develop timely and efficient treatment that avoids or decreases brain damage and eventual complications. For this purpose, it is essential that brain physiology and physiopathological changes triggered by TBI are clear and well known. Current concepts are presented in this paper, emphasizing brain hemodynamics, metabolism, and brain self-regulation.


El traumatismo encefalocraneano (TEC) es un importante problema de salud pública tanto en Chile como en el mundo. Representa un motivo de consulta frecuente constituyendo una de las mayores causas de morbi-mortalidad en la población menor de 45 años. Los accidentes son la principal causa de TEC en la población pediátrica, pero el maltrato infantil es una causa etiológica importante a considerar en los menores de 2 años. Realizar una correcta evaluación al paciente con TEC es fundamental para instaurar un tratamiento oportuno y eficiente con el fin de evitar y/o disminuir el daño cerebral y así prevenir eventuales complicaciones. Para ello es imprescindible el conocimiento de la fisiología cerebral y los cambios fisiopatológicos que se desencadenan posterior al TEC, conceptos que son revisados en este artículo con énfasis en la hemodinamia cerebral, metabolismo y autorregulación cerebral.


Subject(s)
Humans , Child , Pediatrics , Craniocerebral Trauma/physiopathology , Craniocerebral Trauma/metabolism , Brain Edema , Brain/blood supply , Regional Blood Flow/physiology , Homeostasis , Brain Injuries, Traumatic/physiopathology , Brain Injuries, Traumatic/metabolism
20.
Rev. chil. pediatr ; 78(4): 398-402, ago. 2007. ilus, graf
Article in Spanish | LILACS | ID: lil-477413

ABSTRACT

Traumatic brain injury (TBI) is one of the most frequent causes of mortality in childhood. The treatment of patients with severe TBI is directed to prevention, early detection and treatment of secondary injuries due to extra and/or intracranial etiologies. Brain ischemia is a central cause of brain damage and its prevention has become a primary goal for intensivists. New techniques have developed to detect brain ischemia directly bedside the patient, through the monitoring of oxygen tissue pressure (PtiO2). Our objective is to report 2 pediatric patients with severe TBI and PtiO2 monitoring, in order to ilustrate the possibility that offers this technique in early detection of brain ischemia and review the current literature. We analyzed the clinical records of the patients with coma Glasgow score below 8. The intracranial pressure and PtiO2 monitoring was made through a catheter implanted in the encephalic matter. The device allowed early detection of secondary injuries and an optimal therapeutic approach in the patients, both with good outcome at discharge. The PtiO2 monitoring showed high safety and reliability.


Introducción: El traumatismo encéfalo craneal (TEC) grave es una de las principales causas de morbilidad y mortalidad en la infancia. La misión primaria del intensivista está dirigida a prevenir, detectar y tratar precozmente injurias secundarias debido a causas extra y/o intracraneales, siendo una de las más relevantes la isquemia cerebral. Nuevas técnicas han sido desarrolladas para diagnosticar directamente y junto a la cama del paciente la presencia de ella, siendo una de las más promisorias la monitorización de la presión de oxigenación tisular cerebral (PtiO2). Objetivo: Reportar 2 casos pediátricos de TEC grave en quienes monitorizamos la PtiO2, e ilustrar las posibilidades que ofrece esta técnica en la detección de isquemia cerebral, la conducta derivada de la información obtenida y explorar el estado actual de la literatura. Caso y Método: Revisión de fichas clínicas de dos pacientes, en quienes se instaló un dispositivo para monitorizar presión intracraneana (PIC) y PtiO2. La información obtenida del monitoreo permitió pesquisar precozmente segundas noxas, derivando en cambios de conducta terapéutica con resultado funcional satisfactorio al alta. Conclusión: La información obtenida permitió guiar una terapia óptima al comprender mejor la fisiopatología de lo que estaba ocurriendo con la PIC y PtiO2. El método demostró ser seguro y confiable.


Subject(s)
Humans , Male , Female , Adolescent , Brain Ischemia/prevention & control , Monitoring, Physiologic/methods , Oxygen/metabolism , Brain Injuries, Traumatic/physiopathology , Brain Injuries, Traumatic/metabolism , Critical Care/methods , Intracranial Pressure , Oximetry/methods , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL