Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 4.508
Filter
3.
J Prev Alzheimers Dis ; 11(5): 1348-1354, 2024.
Article in English | MEDLINE | ID: mdl-39350380

ABSTRACT

BACKGROUND: There has been little direct examination of how traumatic brain injury (TBI) affects the rate of neurodegeneration for individuals with Alzheimer's disease (AD). METHODS: The study examined 89 cognitively normal adults (65 with and 24 without prior TBI) and 65 with AD (16 with and 49 without prior TBI). Cortical thickness was quantified from T1-weighted MRI scans at baseline and follow-up (mean interval 33.4 months). Partial least squares analysis was used to evaluate the effects of AD and TBI history on the longitudinal change in cortical thickness. RESULTS: Significant group effects were identified throughout the frontal and temporal cortices. Comparison of the AD groups to their control cohorts showed greater relative atrophy for the AD cohort with prior TBI. CONCLUSION: These results indicate that a history of TBI exacerbates longitudinal declines in cortical thickness among AD patients, providing new insights into the shared pathomechanisms between these neurological conditions.


Subject(s)
Alzheimer Disease , Brain Injuries, Traumatic , Magnetic Resonance Imaging , Humans , Alzheimer Disease/pathology , Alzheimer Disease/diagnostic imaging , Brain Injuries, Traumatic/diagnostic imaging , Brain Injuries, Traumatic/complications , Brain Injuries, Traumatic/pathology , Male , Female , Aged , Longitudinal Studies , Cerebral Cortex/pathology , Cerebral Cortex/diagnostic imaging , Atrophy , Middle Aged , Brain Cortical Thickness , Aged, 80 and over
4.
Mymensingh Med J ; 33(4): 1088-1096, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39351730

ABSTRACT

Traumatic brain injury (TBI) is a major cause of morbidity and mortality in Bangladesh and also worldwide. Secondary brain injury from progressive intracerebral hematoma, increasing cerebral edema, raised intracranial pressure and subsequent cerebral ischemia is the main cause for morbidity and mortality following TBI. Secondary brain injury is worsened by post-traumatic coagulopathy, which occurs in brain injured patients and is associated with increase in risk of death and morbidity. The antifibrinolytic agent tranexamic acid (TXA) reduces the hematoma expansion and demonstrated improved clinical outcome also reduced the mortality and morbidity. This was a randomized controlled trial (RCT) done in the Department of Neurosurgery, Dhaka Medical College and Hospital. Included patients were randomized to get either the intravenous tranexamic acid (Group A) or placebo (Group B) treatment based on a computer-generated code list (50 patients in each group) along with usual medical management for traumatic brain injury. The extent of contusion expansion (hematoma plus perihematomal oedema) as the primary outcome at 48 hour after admission and was measured by brain CT scan. The contusion and oedema volume were calculated both the times (on admission and after 48 hours). Glasgow coma scale (GCS) after 48 hours and Glasgow outcome scale (GOS) after 7 days were observed. In this study showed increase in hematoma volume in both groups (p<0.05). But the increased hematoma volume in the Group A was significantly less than that in the control group. The mean total hemorrhage expansion was (1.5±1.1) ml and (4.6±1.9) ml in the Group A and Group B, respectively. In Group A- 02(4.0%) patients required operation, whereas in Group B- 11(22.0%) patients required operation. The result was significant (p=0.023) between groups. Therefore use of tranexamic acid is associated with lesser hematoma volume progression. Mean GCS (after 48 hours), mean GOS (after 7 days) result were significantly better in Group A (p<0.001). This study concluded that tranexamic acid has beneficial effect on the patient with significant traumatic brain injury. Tranexamic acid helps in reduction of intracerebral progression of contusion and improvement of clinical outcomes in patients with TBI.


Subject(s)
Antifibrinolytic Agents , Brain Injuries, Traumatic , Hematoma , Tranexamic Acid , Humans , Tranexamic Acid/therapeutic use , Tranexamic Acid/administration & dosage , Brain Injuries, Traumatic/complications , Brain Injuries, Traumatic/drug therapy , Male , Antifibrinolytic Agents/therapeutic use , Antifibrinolytic Agents/administration & dosage , Adult , Female , Hematoma/drug therapy , Hematoma/etiology , Middle Aged , Glasgow Coma Scale , Disease Progression , Young Adult , Adolescent , Treatment Outcome
5.
Eur Rev Med Pharmacol Sci ; 28(18): 4302-4312, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39359202

ABSTRACT

BACKGROUND: Behavioral and neuropsychological functions are frequent long-term sequelae of severe traumatic brain injury (TBI). Neuropeptides, such as nerve growth factor (NGF), can enhance neurogenesis and improve cognitive functions after TBI, playing a pivotal role in neuroplasticity. A limited number of studies documented the safety and efficacy of intranasal NGF administration in children with severe TBI. CASE REPORT: A fourteen-year-old boy with a diffuse axonal injury secondary to severe TBI was treated with human-recombinant NGF administration. This patient underwent treatment with intranasal hr-NGF administration at a total dose of 50 gamma/kg, three times a day for seven consecutive days. The treatment schedule was performed for 4 cycles, at one month distance each. NGF administration improved radiologic functional assessment evaluated with positron emission tomography scan (PET) and single photon emission computed tomography (SPECT), with an important improvement in clinical conditions. Significant improvements were also observed, mainly in cognitive processes, memory, the planning of a communication strategy, execution skills, attention, and verbal expression. No side effects were reported. CONCLUSIONS: Additional studies are required to gain a deeper insight into this neurotrophin's neuroprotective function, but our findings reveal a potential efficacy of intranasal hr-NGF administration in enhancing cognitive and clinical outcomes among children with diffuse axonal injury after severe TBI.


Subject(s)
Administration, Intranasal , Brain Injuries, Traumatic , Cognition , Nerve Growth Factor , Humans , Male , Nerve Growth Factor/administration & dosage , Nerve Growth Factor/therapeutic use , Brain Injuries, Traumatic/drug therapy , Brain Injuries, Traumatic/complications , Adolescent , Cognition/drug effects , Recombinant Proteins/administration & dosage , Recombinant Proteins/therapeutic use , Positron-Emission Tomography , Tomography, Emission-Computed, Single-Photon
7.
Neurology ; 103(7): e209797, 2024 Oct 08.
Article in English | MEDLINE | ID: mdl-39231380

ABSTRACT

BACKGROUND AND OBJECTIVES: Traumatic brain injury (TBI) is frequently characterized by chronic motor deficits. Therefore, this clinical trial assessed whether intracranial implantation of allogeneic modified mesenchymal stromal (SB623) cells can improve chronic motor deficits after TBI. METHODS: Post hoc analysis of the double-blind, randomized, prospective, surgical sham-controlled, phase 2, STEMTRA clinical trial (June 2016 and March 2019) with 48 weeks of follow-up was conducted. In this international, multicenter clinical trial, eligible participants had moderate-to-severe TBI, were ≥12 months postinjury, and had chronic motor deficits. Participants were randomized in a 1:1:1:1 ratio to stereotactic surgical intracranial implantation of SB623 cells (2.5 × 106, 5.0 × 106, 10 × 106) or surgical sham-controlled procedure. The prespecified primary efficacy end point was significantly greater change from baseline of the Fugl-Meyer Motor Scale (FMMS) score, a measure of motor status, for the SB623 pooled vs control arm at 24 weeks. RESULTS: A total of 211 participants were screened, 148 were excluded, and 63 underwent randomization, of which 61 (97%; mean age, 34 [SD, 12] years; 43 men [70.5%]) completed the trial. Single participants in the SB623 2.5 × 106 and 5.0 × 106 cell dose groups discontinued before surgery. Safety and efficacy (modified intent-to-treat) were assessed in participants who underwent surgery (N = 61; SB623 = 46, controls = 15). The primary efficacy end point (FMMS) was achieved (least squares mean [SE] SB623: +8.3 [1.4]; 95% CI 5.5-11.2 vs control: +2.3 [2.5]; 95% CI -2.7 to 7.3; p = 0.04), with faster improvement of the FMMS score in SB623-treated groups than in controls at 24 weeks and sustained improvement at 48 weeks. At 48 weeks, improvement of function and activities of daily living (ADL) was greater, but not significantly different in SB623-treated groups vs controls. The incidence of adverse events was equivalent in SB623-treated groups and controls. There were no deaths or withdrawals due to adverse events. DISCUSSION: Intraparenchymal implantation of SB623 cells was safe and significantly improved motor status at 24 weeks in participants with chronic motor deficits after TBI, with continued improvement of function and ADL at 48 weeks. Cell therapy can modify chronic neurologic deficits after TBI. TRIAL REGISTRATION INFORMATION: ClinicalTrials.gov Identifier: NCT02416492. Submitted to registry: April 15, 2015. First participant enrolled: July 6, 2016. Available at: classic.clinicaltrials.gov/ct2/show/NCT02416492. CLASSIFICATION OF EVIDENCE: This study provides Class I evidence that intracranial implantation of allogeneic stem (SB623) cells in adults with motor deficits from chronic TBI improves motor function at 24 weeks.


Subject(s)
Brain Injuries, Traumatic , Mesenchymal Stem Cell Transplantation , Humans , Brain Injuries, Traumatic/complications , Brain Injuries, Traumatic/surgery , Brain Injuries, Traumatic/therapy , Male , Adult , Female , Double-Blind Method , Mesenchymal Stem Cell Transplantation/methods , Middle Aged , Prospective Studies , Treatment Outcome , Young Adult
9.
Neurosurg Rev ; 47(1): 538, 2024 Sep 05.
Article in English | MEDLINE | ID: mdl-39231815

ABSTRACT

Traumatic brain injury (TBI) presents complex management scenarios, particularly in patients requiring anticoagulation for concurrent conditions such as venous thromboembolism (VTE) or atrial fibrillation (AF). A systematic search of PubMed/MEDLINE, Embase, and the Cochrane Library databases was conducted to identify relevant studies. Inclusion criteria encompassed studies assessing the effects of anticoagulation therapy on outcomes such as re-hemorrhage, hematoma expansion, thrombotic events, and hemorrhagic events in TBI patients with subdural hematomas (SDH). This systematic review critically addresses two key questions: the optimal timing for initiating anticoagulation therapy and the differential impact of this timing based on the type of intracranial bleed, with a specific focus on subdural hematomas (SDH) compared to other types. Initially screening 508 articles, 7 studies met inclusion criteria, which varied in design and quality, precluding meta-analysis. The review highlights a significant knowledge gap, underscoring the lack of consensus on when to initiate anticoagulation therapy in TBI patients, exacerbated by the need for anticoagulation in the presence of VTE or AF. Early anticoagulation, particularly in patients with SDH, may elevate the risk of re-hemorrhage, posing a clinical dilemma. Evidence on whether the type of intracranial hemorrhage influences outcomes with early anticoagulation remains inconclusive, indicating a need for further research to tailor management strategies effectively. This review underscores the scarcity of high-quality evidence regarding anticoagulation therapy in TBI patients with concurrent conditions, emphasizing the necessity for well-designed prospective studies to elucidate optimal management strategies for this complex patient population.


Subject(s)
Anticoagulants , Brain Injuries, Traumatic , Adult , Humans , Anticoagulants/therapeutic use , Anticoagulants/administration & dosage , Atrial Fibrillation/drug therapy , Atrial Fibrillation/complications , Brain Injuries, Traumatic/complications , Brain Injuries, Traumatic/drug therapy , Observational Studies as Topic , Venous Thromboembolism/complications , Venous Thromboembolism/drug therapy
10.
Int Rev Psychiatry ; 36(3): 243-253, 2024 May.
Article in English | MEDLINE | ID: mdl-39255021

ABSTRACT

BACKGROUND: Boxing exposes fighters to head impacts and potential traumatic brain injury (TBI). Though research has explored the neuropsychiatric consequences of contact sports, there is limited research into Excessive Daytime Sleepiness (EDS) and its relationship to other outcomes, such as impulsiveness and depression. Therefore, this study aimed to describe EDS in retired boxers using the Epworth Sleepiness Scale (ESS) and to examine how boxing and sleepiness relate to impulsiveness and depression symptomatology. METHODS: 86 male retired professional boxers from the Professional Fighters Brain Health Study (PFBHS) met the inclusion criteria. Adjusted multivariable models analyzed relationships between professional boxing bouts, EDS (ESS), impulsiveness (Barratt Impulsiveness Scale Version 11 (BIS-11)), and/or depression (Patient Health Questionnaire-9 (PHQ-9)). A causal mediation analysis was performed to assess whether boxing bouts and ESS scores predicted BIS-11 and PHQ-9 scores. RESULTS: Mean age was ∼51 years, fighters averaged ∼36 professional bouts, and ESS mean(SD) was 7.5(5.3). ESS scores were significantly associated with raw BIS-11 (Beta = 1.26, 95%CI = 0.77-1.75, p < 0.001) and ordinal PHQ-9 (OR = 1.20, 95%CI = 1.11-1.31, p < 0.001) scores in adjusted models, and the significant relationship between boxing bouts and BIS-11/PHQ-9 was mediated by ESS. CONCLUSIONS: EDS in retired male professional boxers may be strongly associated with other neuropsychiatric sequelae of TBI (impulsiveness and depression).Sleepiness; sleep; boxing; contact sports; impulsiveness; impulsivity; depression; Epworth sleepiness scale box.


Subject(s)
Boxing , Depression , Disorders of Excessive Somnolence , Impulsive Behavior , Retirement , Humans , Male , Middle Aged , Disorders of Excessive Somnolence/etiology , Boxing/injuries , Brain Injuries, Traumatic/complications
11.
Sci Rep ; 14(1): 20737, 2024 09 05.
Article in English | MEDLINE | ID: mdl-39237683

ABSTRACT

Global outcomes have been reported to be associated with cerebrovascular reactivity (CVR) in the acute phase following moderate and severe traumatic brain injury (TBI). The association of CVR in the acute and chronic phase of injury with patient-reported health-related quality of life metrics (HRQOL) metrics has never been explored. The aim of this study is to examine the association of CVR, as measured by the cerebral oxygen indices (COx and COx_a), in the acute and chronic phase following moderate and severe TBI, with patient reported HRQOL. In this prospective cohort study, performed in a Canadian quaternary care center, the association between continuous acute and chronic phase CVR with patient reported HRQOL outcomes following moderate and severe TBI was examined. The main outcomes of interest of this study were validated measures of patient-reported HRQOL over various domains as measured by both the 12-Item Short-Form Health Survey (SF-12) and a Quality of Life after Brain Injury (QOLIBRI) questionnaire. In the 29 subjects of this cohort, acute phase CVR was found to be significantly more active in those with a favorable Mental Component Summary (MCS) scores of the SF-12 at early follow-up when measured by COx (-0.015 [IQR: -0.067 to 0.032] vs 0.040 [IQR: 0.019 to 0.137] for Favorable first MCS vs Unfavorable respectively; Mann-Whitney U test p-value = 0.046) and COx_a (0.038 [IQR: 0.009 to 0.062] vs 0.112 [IQR: 0.065 to 0.167] for Favorable first MCS vs Unfavorable respectively; Mann-Whitney U test p-value = 0.014). Further, multivariable logistic regression analysis found acute phase COx and COx_a to improve model performance when predicting favorable versus unfavorable early MCS scores over established parameters such as age and measures of injury severity. Associations between outcomes and chronic phase CVR were limited, potentially due to short recording periods. This is the first ever pilot study to identify a relationship between acute phase CVR following moderate-to-severe TBI with mental and cognitive outcomes as experienced by patients. Given the small cohort, these findings will need to be confirmed in a larger multicenter study. This highlights the need for additional examination of the role dysfunctional CVR may play in mental and cognitive outcomes, as well as patient-reported outcomes more generally following TBI.


Subject(s)
Brain Injuries, Traumatic , Quality of Life , Humans , Brain Injuries, Traumatic/psychology , Brain Injuries, Traumatic/physiopathology , Brain Injuries, Traumatic/complications , Male , Female , Adult , Middle Aged , Prospective Studies , Cerebrovascular Circulation , Surveys and Questionnaires , Canada
12.
Biomed Pharmacother ; 179: 117405, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39236478

ABSTRACT

Traumatic brain injury (TBI) is a significant contributor to global mortality and disability, and there is still no specific drug available to treat cognitive deficits in survivors. Vanillic acid (VA), a bioactive phenolic compound, has shown protective effects in various models of neurodegeneration; however, its impact on TBI outcomes remains elusive. Therefore, this study aimed to elucidate the possible role of VA in ameliorating TBI-induced cognitive decline and to reveal the mechanisms involved. TBI was induced using the Marmarou impact acceleration model to deliver an impact force of 300 g, and treatment with VA (50 mg/kg; P.O.) was initiated 30 minutes post-TBI. The cognitive performance, hippocampal long-term potentiation (LTP), oxidative stress markers, neurological function, cerebral edema, and morphological changes were assessed at scheduled points in time. TBI resulted in cognitive decline in the passive avoidance task, impaired LTP in the perforant path-dentate gyrus (PP-DG) pathway, increased hippocampal oxidative stress, cerebral edema, neurological deficits, and neuronal loss in the rat hippocampus. In contrast, acute VA administration mitigated all the aforementioned TBI outcomes. The data suggest that reducing synaptic plasticity impairment, regulating oxidative and antioxidant defense, alleviating cerebral edema, and preventing neuronal loss by VA can be at least partially attributed to its protection against TBI-induced cognitive decline.


Subject(s)
Brain Injuries, Traumatic , Cognitive Dysfunction , Hippocampus , Long-Term Potentiation , Oxidative Stress , Vanillic Acid , Animals , Vanillic Acid/pharmacology , Male , Cognitive Dysfunction/drug therapy , Cognitive Dysfunction/etiology , Cognitive Dysfunction/prevention & control , Brain Injuries, Traumatic/drug therapy , Brain Injuries, Traumatic/complications , Brain Injuries, Traumatic/psychology , Oxidative Stress/drug effects , Rats , Long-Term Potentiation/drug effects , Hippocampus/drug effects , Hippocampus/pathology , Hippocampus/metabolism , Neuroprotective Agents/pharmacology , Brain Edema/drug therapy , Rats, Wistar , Disease Models, Animal , Antioxidants/pharmacology , Cognition/drug effects , Neuronal Plasticity/drug effects
13.
J Head Trauma Rehabil ; 39(5): 329-334, 2024.
Article in English | MEDLINE | ID: mdl-39256154

ABSTRACT

OBJECTIVE: To describe experiences and challenges when updating a living evidence-based review database of randomized controlled trials (RCTs) on mental health and behavioral disorders in moderate to severe traumatic brain injury (MSTBI). METHOD: This commentary derives from our experience developing an extensive database of RCTs on MSTBI that has been conceptualized as a living evidence-based review. Our working group focused on mental health and behavior RCTs and reflected upon their experiences and challenges using the living systematic approach. We discuss challenges associated with metrics of study quality, injury etiology and severity, time post-injury, country of origin, and variability in outcome measures. RESULTS: RCTs were conducted almost solely in high income countries, with smaller sample sizes, and most conducted in the chronic phase post-TBI. Issues related to lack of transparency, unclear and incomplete reporting of injury severity, etiology, and time post-injury remain a concern and can lead to challenges associated with interpretation of results, validity, and reliability of the data. There was significant heterogeneity regarding the use of outcome measures and constructs, underscoring the need for standardization. CONCLUSION: Lack of standardization and incomplete reporting of injury characteristics makes it difficult to compare data between RCTs of MSTBI, perform meta-analyses, and generate evidence-based clinical recommendations.


Subject(s)
Brain Injuries, Traumatic , Mental Disorders , Humans , Brain Injuries, Traumatic/complications , Brain Injuries, Traumatic/diagnosis , Brain Injuries, Traumatic/therapy , Evidence-Based Medicine , Mental Disorders/etiology , Mental Disorders/therapy , Randomized Controlled Trials as Topic , Trauma Severity Indices
14.
Injury ; 55 Suppl 3: 111337, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39300616

ABSTRACT

INTRODUCTION: Early treatment of elevated intracranial pressure (ICP) is a cornerstone of the therapy in severe traumatic brain injury (TBI) patients. Treatment of refractory high ICP however, remain challenging as only limited and risky third-tier therapeutic interventions are available. Controlled lumbar cerebrospinal fluid (CSF) drainage has been known as an efficient method of ICP reduction after TBI for decades, but it is not recommended in international guidelines because of low evidence background and safety issues. Our centre has a long-standing experience using this intervention for more than 15 years. Here we present our data about the safety and efficacy of controlled lumbar drainage to avoid further second- and third tier ICP lowering therapies and beneficially influence functional outcome. METHODS: Observational (retrospective and prospective) analysis was performed using demographic, clinical and outcome data of severe TBI patients admitted to our centre. Analysis was retrospective between 2008 and 2013 and prospective from 2014 to 2019. Only severe TBI patients (GCS<9) with ICP monitoring were enrolled. Lumbar drainage (LD) was used as a second-tier therapy to control intracranial hypertension in salvageable patients with normal haemostasis and discernible basal cisterns on pre-interventional CT scan. RESULTS: Data of 45 patients were analysed. Patients were young, comatose and with severe injuries (median age: 29, GMS: 4, ISS: 25). Lumbar drain was inserted mainly on the first week and maintained for further 5 days. Episodes of intracranial hypertension (ICP>20 Hgmm) within one day (10 vs 2) were reduced. The need of additional second- and third-line therapies (deep sedation, hyperventilation, barbiturate administration, decompressive craniectomy) also significantly decreased (60 vs 25 interventions, p<0.001). The in-hospital mortality and 6-month functional outcome were more favourable than the whole TBI population and as predicted by prognostic calculations (mortality: 16% vs. 48 %; GOSE 1-4: 49% vs. 65% vs CRASH: 87% vs. IMPACT: 51 %) in this period. CONCLUSIONS: Our results support the view that controlled lumbar drainage is a highly efficient method to manage intracranial hypertension and significantly decreases the need of further harmful ICP lowering therapies without altering functional outcome of severe TBI patients.


Subject(s)
Brain Injuries, Traumatic , Drainage , Intracranial Hypertension , Humans , Intracranial Hypertension/therapy , Intracranial Hypertension/etiology , Male , Drainage/methods , Female , Brain Injuries, Traumatic/complications , Brain Injuries, Traumatic/therapy , Adult , Retrospective Studies , Treatment Outcome , Prospective Studies , Spinal Puncture , Young Adult , Intracranial Pressure/physiology , Middle Aged , Glasgow Coma Scale
15.
Medicina (Kaunas) ; 60(9)2024 Sep 18.
Article in English | MEDLINE | ID: mdl-39336560

ABSTRACT

Background and Objectives: Pediatric traumatic brain injury (pTBI) remains a major pediatric public health problem, despite well-developed injury prevention programs. The purpose of this study is to analyze the emergency surgical outcomes of pTBI in a single institute ten-year retrospective study to offer a real-world clinical result. Materials and Methods: Our institute presented a clinical retrospective, single-institute research study of 150 pediatric TBI cases that were diagnosed and underwent emergency surgical treatment from 2010 to 2019. Results: The incidence of radiological findings is detailed as follows: brain edema (30%, 45/150), followed by acute subdural hematoma (27.3%, 41/150), epidural hematoma (21.3%, 32/150), chronic subdural hemorrhage (10%, 15/150), skull fracture (6.7%, 10/150), and traumatic subarachnoid hemorrhage (4.7%, 7/150). Surgical intervention data revealed that decompressive craniectomy was still the main effective surgical method. The results showed longer hospital stays and higher morbidity rates in the brain edema, acute subdural hematoma, and chronic subdural hemorrhage groups, which were viewed as poor surgical outcome groups. Epidural hematoma, skull fracture and traumatic subarachnoid hemorrhage were categorized into good surgical outcome groups. Notably, the data revealed gross improvement in Glasgow Coma Scale/Score (GCS) evolution after surgical interventions, and the time to cranioplasty was a significant factor in the development of post-traumatic hydrocephalus (PTH). Conclusions: Our study provided real-world data for the distribution of etiology in pTBI and also categorized it into six groups, indicating disease-orientated treatment. In addition, our data supported that decompressive craniectomy (DC) remains a mainstay surgical treatment in pTBI and early cranioplasty could decrease the incidence of PTH.


Subject(s)
Brain Injuries, Traumatic , Humans , Retrospective Studies , Female , Male , Brain Injuries, Traumatic/surgery , Brain Injuries, Traumatic/complications , Child , Taiwan/epidemiology , Child, Preschool , Adolescent , Infant , Treatment Outcome , Decompressive Craniectomy/methods , Decompressive Craniectomy/statistics & numerical data , Brain Edema/surgery , Brain Edema/etiology , Hematoma, Epidural, Cranial/surgery , Skull Fractures/surgery , Skull Fractures/complications
16.
Acta Neurochir (Wien) ; 166(1): 385, 2024 Sep 27.
Article in English | MEDLINE | ID: mdl-39331162

ABSTRACT

PURPOSE: Post-traumatic cerebral venous sinus thrombosis (ptCVT) is a rare but serious complication of traumatic brain injury (TBI). Managing ptCVT is challenging due to the concurrent risk of traumatic intracranial hematoma (ICH) expansion. Limited data exists on the safety and efficacy of anticoagulation therapy (ACT) in these cases. METHODS: This single-center observational cohort study included adult TBI patients with concurrent ICH and ptCVT. Low-molecular-weight heparin (LMWH) or heparin infusion was used to treat all ptCVTs based on institutional protocols. The outcomes of interest were hemorrhagic and thrombotic complications. RESULTS: Out of 1,039 TBI-patients admitted between 2006 and 2020, 32 met the inclusion criteria. The median time from injury to ptCVT diagnosis was 24 h. ACT was initiated at a median of 9 h after ptCVT diagnosis. Patients were administered either heparin infusion (n = 8) or LMWH at dosages ranging from 28 to 72% of the therapeutic level (n = 24). There were no hemorrhagic complications, even in patients receiving LMWH at ≥ 50% of the therapeutic dose. Thrombotic complications occurred in 3 patients (9.4%) - two cases of thrombus progression and one venous infarct. The patients who developed thrombotic complications differed from those who did not by having a 17-h delay in ACT initiation after diagnosis or by receiving an initial LMWH dose at 28% of the therapeutic level. CONCLUSION: LMWH at approximately 50% of the therapeutic level was effective for managing ptCVT associated with TBI in our retrospective dataset, with no risk of hematoma expansion. Prospective trials are warranted to confirm these results.


Subject(s)
Anticoagulants , Heparin, Low-Molecular-Weight , Sinus Thrombosis, Intracranial , Humans , Sinus Thrombosis, Intracranial/drug therapy , Anticoagulants/therapeutic use , Anticoagulants/adverse effects , Male , Female , Middle Aged , Adult , Heparin, Low-Molecular-Weight/therapeutic use , Heparin, Low-Molecular-Weight/adverse effects , Cohort Studies , Aged , Intracranial Hemorrhage, Traumatic/drug therapy , Heparin/therapeutic use , Heparin/adverse effects , Brain Injuries, Traumatic/complications , Brain Injuries, Traumatic/drug therapy , Retrospective Studies
17.
Front Biosci (Landmark Ed) ; 29(9): 322, 2024 Sep 20.
Article in English | MEDLINE | ID: mdl-39344324

ABSTRACT

Neurotrauma plays a significant role in secondary injuries by intensifying the neuroinflammatory response in the brain. High Mobility Group Box-1 (HMGB1) protein is a crucial neuroinflammatory mediator involved in this process. Numerous studies have hypothesized about the underlying pathophysiology of HMGB1 and its role in cognition, but a definitive link has yet to be established. Elevated levels of HMGB1 in the hippocampus and serum have been associated with declines in cognitive performance, particularly in spatial memory and learning. This review also found that inhibiting HMGB1 can improve cognitive deficits following neurotrauma. Interestingly, HMGB1 levels are linked to the modulation of neuroplasticity and may offer neuroprotective effects in the later stages of neurotraumatic events. Consequently, administering HMGB1 during the acute phase may help reduce neuroinflammatory effects that lead to cognitive deficits in the later stages of neurotrauma. However, further research is needed to understand the time-dependent regulation of HMGB1 and the clinical implications of treatments targeting HMGB1 after neurotrauma.


Subject(s)
Cognitive Dysfunction , HMGB1 Protein , HMGB1 Protein/metabolism , HMGB1 Protein/antagonists & inhibitors , Humans , Animals , Cognitive Dysfunction/etiology , Cognitive Dysfunction/metabolism , Cognitive Dysfunction/drug therapy , Cognitive Dysfunction/physiopathology , Brain Injuries, Traumatic/complications , Brain Injuries, Traumatic/metabolism , Brain Injuries, Traumatic/drug therapy , Brain Injuries, Traumatic/physiopathology , Molecular Targeted Therapy/methods , Hippocampus/metabolism , Neuronal Plasticity/drug effects
18.
BMC Ophthalmol ; 24(1): 422, 2024 Sep 27.
Article in English | MEDLINE | ID: mdl-39334049

ABSTRACT

BACKGROUND: Traumatic brain injury (TBI) often precipitates a cascade of neurophysiological alterations, impacting structures such as the optic nerve and ocular motor system. However, the literature lacks expansive investigations into the longitudinal changes in the optic chiasm and its relationship with the clinical recovery of visual processing. This study aimed to scrutinize longitudinal changes in optic chiasm volume (OCV) and establish the relationship of OCV with process speed index at 12 months post-injury. Process speed index is derived from Wechsler Adult Intelligence Scale IV. METHODS: Thorough cross-sectional and longitudinal analyses were executed, involving 42 patients with moderate to severe TBI and 35 healthy controls. OCV was acquired at 3, 6, and 12 months post-injury using T1-weighted images. OCV of healthy controls and that of patients with TBI at 3, 6, and 12 months post-injury were compared using a Mann-Whitney U test. A multiple linear regression model was constructed to assess the association between OCV and PSI and to predict PSI at 12 months post-injury using OCV at 3 months post-injury. RESULTS: OCV of patients with TBI was significantly larger compared to healthy controls, persisting from 3 to 12 months post-injury (p < 0.05). This increased OCV negatively correlated with PSI at 12 months post-injury, indicating that larger OCV sizes were associated with decreased PSI (p = 0.031). Furthermore, the multiple linear regression model was significant in predicting PSI at 12 months post-injury utilizing OCV at 3 months post-injury (p = 0.024). CONCLUSION: For the first time, this study elucidates the increased OCV and the significant association between OCV in sub-chronic stage and PSI at 12 months post-injury, potentially providing clinicians with a tool for anticipatory cognitive rehabilitation strategies following TBI.


Subject(s)
Brain Injuries, Traumatic , Magnetic Resonance Imaging , Optic Chiasm , Humans , Brain Injuries, Traumatic/physiopathology , Brain Injuries, Traumatic/complications , Male , Female , Adult , Optic Chiasm/diagnostic imaging , Optic Chiasm/pathology , Cross-Sectional Studies , Middle Aged , Young Adult , Longitudinal Studies , Follow-Up Studies
19.
Crit Care Explor ; 6(10): e1160, 2024 Oct 01.
Article in English | MEDLINE | ID: mdl-39324956

ABSTRACT

OBJECTIVES: We sought to evaluate the effectiveness of any antiseizure medication on the incidence of early post-traumatic seizures among adult patients with traumatic brain injury. DATA SOURCES: MEDLINE, Embase, PubMed, Cochrane Central Register of Controlled Trials, and LILACS were searched from inception to October 2023. STUDY SELECTION: We included randomized trials of adult patients with traumatic brain injury evaluating any antiseizure medication compared with either placebo or another agent. DATA EXTRACTION: Two reviewers independently extracted individual study data and evaluated studies for risk of bias using the Cochrane Risk of Bias tool. Our main outcome of interest was the occurrence of early seizures (i.e., within 7 d); secondary outcomes included late-seizures and all-cause mortality. DATA SYNTHESIS: Bayesian network meta-analyses were used to derive risk ratios (RRs) alongside 95% credible intervals (CrIs). We used Grading of Recommendations Assessment, Development, and Evaluation methodology to rate the certainty in our findings. Overall, ten individual randomized controlled trials (1851 participants) were included. Compared with placebo, phenytoin (RR, 0.28; 95% CrI, 0.13-0.57; moderate certainty) and levetiracetam (RR, 0.20; 95% CrI, 0.07-0.60; moderate certainty) were associated with a reduction in the risk of early seizures. Carbamazepine may be associated with a reduced risk of early seizures, but the evidence is very uncertain (RR, 0.41; 95% CrI, 0.12-1.27; very low certainty). Valproic acid may result in little to no difference in the risk of early seizures, but the evidence is very uncertain (RR, 0.97; 95% CrI, 0.16-9.00; very low certainty). The evidence is very uncertain about the impact of any antiseizure medication on the risk of late seizures or all-cause mortality at longest reported follow-up time. CONCLUSIONS: Phenytoin or levetiracetam reduce the risk of early seizures among adult patients with traumatic brain injury. Further research is needed to evaluate required duration of therapy and long-term safety profiles.


Subject(s)
Anticonvulsants , Bayes Theorem , Brain Injuries, Traumatic , Network Meta-Analysis , Seizures , Humans , Brain Injuries, Traumatic/drug therapy , Brain Injuries, Traumatic/mortality , Brain Injuries, Traumatic/complications , Anticonvulsants/therapeutic use , Seizures/drug therapy , Adult , Levetiracetam/therapeutic use , Phenytoin/therapeutic use , Randomized Controlled Trials as Topic
20.
Int J Med Sci ; 21(12): 2272-2284, 2024.
Article in English | MEDLINE | ID: mdl-39310263

ABSTRACT

Hyperbaric oxygen (HBO) therapy can attenuate neurological impairment after traumatic brain injury (TBI) and alleviate intestinal dysfunction. However, the role and mechanism of HBO therapy in intestinal dysfunction following TBI remain unclear. Herein, by establishing a mouse model of controlled cortical impact (CCI), we found that HBO therapy reduced histopathological lesions and decreased the levels of inflammatory and oedema proteins in the intestinal tissues of mice 10 days after TBI. We also showed that HBO therapy improved microbiome abundance and probiotic (particularly g_Bifidobacterium) colonisation in mice post-CCI. Then, we identified that the m6A level imcreased notably in injured cortical tissue of CCI+HBO group compared with the CCI group following CCI. Thus, our results suggested that HBO therapy could alleviate TBI-induced intestinal dysfunction and m6A might participate in this regulation process, which provides new insights for exploring the specific mechanism and targets of HBO in the treatment of intestinal dysfunction after TBI, thereby improving the therapeutic effect of HBO.


Subject(s)
Brain Injuries, Traumatic , Disease Models, Animal , Gastrointestinal Microbiome , Hyperbaric Oxygenation , Hyperbaric Oxygenation/methods , Animals , Brain Injuries, Traumatic/therapy , Brain Injuries, Traumatic/complications , Mice , Male , Humans , Intestines , Intestinal Diseases/therapy , Intestinal Diseases/etiology , Mice, Inbred C57BL
SELECTION OF CITATIONS
SEARCH DETAIL