1
|
Jin J, Zhang H, Lu Q, Tian L, Yao S, Lai F, Liang Y, Liu C, Lu Y, Tian S, Zhao Y, Ren W. Nanocarrier-mediated siRNA delivery: a new approach for the treatment of traumatic brain injury-related Alzheimer's disease. Neural Regen Res 2025; 20:2538-2555. [PMID: 39314170 DOI: 10.4103/nrr.nrr-d-24-00303] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2024] [Accepted: 07/17/2024] [Indexed: 09/25/2024] Open
Abstract
Traumatic brain injury and Alzheimer's disease share pathological similarities, including neuronal loss, amyloid-β deposition, tau hyperphosphorylation, blood-brain barrier dysfunction, neuroinflammation, and cognitive deficits. Furthermore, traumatic brain injury can exacerbate Alzheimer's disease-like pathologies, potentially leading to the development of Alzheimer's disease. Nanocarriers offer a potential solution by facilitating the delivery of small interfering RNAs across the blood-brain barrier for the targeted silencing of key pathological genes implicated in traumatic brain injury and Alzheimer's disease. Unlike traditional approaches to neuroregeneration, this is a molecular-targeted strategy, thus avoiding non-specific drug actions. This review focuses on the use of nanocarrier systems for the efficient and precise delivery of siRNAs, discussing the advantages, challenges, and future directions. In principle, siRNAs have the potential to target all genes and non-targetable proteins, holding significant promise for treating various diseases. Among the various therapeutic approaches currently available for neurological diseases, siRNA gene silencing can precisely "turn off" the expression of any gene at the genetic level, thus radically inhibiting disease progression; however, a significant challenge lies in delivering siRNAs across the blood-brain barrier. Nanoparticles have received increasing attention as an innovative drug delivery tool for the treatment of brain diseases. They are considered a potential therapeutic strategy with the advantages of being able to cross the blood-brain barrier, targeted drug delivery, enhanced drug stability, and multifunctional therapy. The use of nanoparticles to deliver specific modified siRNAs to the injured brain is gradually being recognized as a feasible and effective approach. Although this strategy is still in the preclinical exploration stage, it is expected to achieve clinical translation in the future, creating a new field of molecular targeted therapy and precision medicine for the treatment of Alzheimer's disease associated with traumatic brain injury.
Collapse
Affiliation(s)
- Jie Jin
- Institute of Disaster and Emergency Medicine, Tianjin University, Tianjin, China
| | - Huajing Zhang
- Institute of Disaster and Emergency Medicine, Tianjin University, Tianjin, China
- Key Laboratory for Disaster Medicine Technology, Tianjin, China
| | - Qianying Lu
- Institute of Disaster and Emergency Medicine, Tianjin University, Tianjin, China
- Key Laboratory for Disaster Medicine Technology, Tianjin, China
| | - Linqiang Tian
- Henan Medical Key Laboratory for Research of Trauma and Orthopedics, The Third Affiliated Hospital of Xinxiang Medical University, Xinxiang, Henan Province, China
- Clinical Medical Center of Tissue Engineering and Regeneration, Xinxiang Medical University, Xinxiang, Henan Province, China
| | - Sanqiao Yao
- Institutes of Health Central Plain, Xinxiang Medical University, Xinxiang, Henan Province, China
- School of Public Health, Xinxiang Medical University, Xinxiang, Henan Province, China
| | - Feng Lai
- Henan Medical Key Laboratory for Research of Trauma and Orthopedics, The Third Affiliated Hospital of Xinxiang Medical University, Xinxiang, Henan Province, China
| | - Yangfan Liang
- Institute of Disaster and Emergency Medicine, Tianjin University, Tianjin, China
| | - Chuanchuan Liu
- Institute of Disaster and Emergency Medicine, Tianjin University, Tianjin, China
| | - Yujia Lu
- Institute of Disaster and Emergency Medicine, Tianjin University, Tianjin, China
| | - Sijia Tian
- Institute of Disaster and Emergency Medicine, Tianjin University, Tianjin, China
| | - Yanmei Zhao
- Institute of Disaster and Emergency Medicine, Tianjin University, Tianjin, China
- Key Laboratory for Disaster Medicine Technology, Tianjin, China
| | - Wenjie Ren
- Henan Medical Key Laboratory for Research of Trauma and Orthopedics, The Third Affiliated Hospital of Xinxiang Medical University, Xinxiang, Henan Province, China
- Clinical Medical Center of Tissue Engineering and Regeneration, Xinxiang Medical University, Xinxiang, Henan Province, China
- Institutes of Health Central Plain, Xinxiang Medical University, Xinxiang, Henan Province, China
| |
Collapse
|
2
|
Zeng D, Basilio AV, Pichay LA, Ateshian GA, Hansen OS, Romanov A, Morrison B. Experimental Measurement and Mathematical Quantification of Fixed-Charged Density in Rat and Pig Brain Tissue. Ann Biomed Eng 2024:10.1007/s10439-024-03666-y. [PMID: 39702733 DOI: 10.1007/s10439-024-03666-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2024] [Accepted: 12/02/2024] [Indexed: 12/21/2024]
Abstract
Cerebral edema is associated with poor prognosis because brain swelling within the rigid skull raises intracranial pressure, exacerbating secondary injuries following traumatic brain injury. Brain swelling can be characterized by triphasic biomechanics, which models brain tissue as a mixture of a deformable porous solid matrix with a negative fixed-charged density (FCD), water, and monovalent counterions. When brain cells die, the intracellular FCD is exposed, attracting cations into the cells. The increase in intracellular solute concentration generates osmotic pressure via the Gibbs-Donnan effect, driving water into cells and causing swelling. This study quantifies the FCD of rat and pig brain tissue by measuring the pressure generated by tissue within a confined volume as cells died. Rat brain tissue generated an averaged swelling pressure of 52.92 ± 20.40 mmHg (mean ± one standard deviation). Variations were observed between pig cortical white matter (7.14 ± 4.79 mmHg) and cortical gray matter (33.86 ± 11.89 mmHg). The corresponding FCD values were 42.54 ± 8.14 mEq/L for rat brain tissue, and 15.18 ± 5.38 mEq/L and 34.22 ± 6.31 mEq/L for pig cortical white and gray matter, respectively. Treating the rat brain tissue with DNAse, heparinase I, heparinase III, and chondroitinase ABC to degrade FCD significantly reduced swelling pressure. Good agreement between the experimental and numerically simulated responses supported the role of the FCD in cerebral edema formation. The reported FCD values can improve the biofidelity of computational models to predict post-traumatic cerebral edema, aiding the improvement of safety systems.
Collapse
Affiliation(s)
- Delin Zeng
- Department of Biomedical Engineering, Columbia University, 351 Engineering Terrace MC 8904, 1210 Amsterdam Avenue, New York, NY, 10027, USA
| | - Andrew V Basilio
- Department of Biomedical Engineering, Columbia University, 351 Engineering Terrace MC 8904, 1210 Amsterdam Avenue, New York, NY, 10027, USA
| | - Leanne A Pichay
- Department of Biomedical Engineering, Columbia University, 351 Engineering Terrace MC 8904, 1210 Amsterdam Avenue, New York, NY, 10027, USA
| | - Gerard A Ateshian
- Department of Biomedical Engineering, Columbia University, 351 Engineering Terrace MC 8904, 1210 Amsterdam Avenue, New York, NY, 10027, USA
- Department of Mechanical Engineering, Columbia University, 220 S. W. Mudd Building, 500 West 120th Street, New York, NY, 10027, USA
| | - Olivia S Hansen
- Department of Biomedical Engineering, Columbia University, 351 Engineering Terrace MC 8904, 1210 Amsterdam Avenue, New York, NY, 10027, USA
| | - Alexander Romanov
- Institute of Comparative Medicine, Columbia University, 650 West 168th Street, BB 1912B, New York, NY, 10032, USA
| | - Barclay Morrison
- Department of Biomedical Engineering, Columbia University, 351 Engineering Terrace MC 8904, 1210 Amsterdam Avenue, New York, NY, 10027, USA.
| |
Collapse
|
3
|
Fullerton JL, Hay J, Bryant-Craig C, Atkinson J, Smith DH, Stewart W. Pediatric Traumatic Brain Injury and Microvascular Blood-Brain Barrier Pathology. JAMA Netw Open 2024; 7:e2446767. [PMID: 39585695 PMCID: PMC11589795 DOI: 10.1001/jamanetworkopen.2024.46767] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/24/2024] [Accepted: 09/27/2024] [Indexed: 11/26/2024] Open
Abstract
Importance Pediatric traumatic brain injury (TBI) is a major cause of morbidity and mortality, with an increased risk of catastrophic outcome compared with adult TBI, including diffuse brain swelling and so-called second impact syndrome. Nevertheless, the biological substrates driving adverse outcomes in pediatric TBI remain poorly described. Objective To compare neuropathological evidence of brain swelling and blood-brain barrier (BBB) disruption after moderate or severe acute TBI in adult vs pediatric case material. Design, Setting, and Participants In this retrospective case series, cases of pediatric (aged 3-18 years) and adult (aged ≥19 years) TBI were accrued from January 1, 1979, to December 31, 2005, and underwent laboratory-based assessment of autopsy material from the Glasgow TBI Archive. Data analysis was performed from January 2019 to January 2024. Exposures Single moderate or severe TBI. Main Outcomes and Measures Evaluation of representative brain tissue sections stained for markers of endothelia (CD34) and BBB integrity (fibrinogen and immunoglobin G). Results Eighty-one pediatric patients (mean [SD] age, 12.1 [4.6] years; 50 [62%] male) and 62 adult patients (mean [SD] age, 38.7 [12.9] years; 35 [56%] male) were studied. At autopsy, when present, brain swelling was more often diffuse and bilateral among pediatric patients (64 of 81 cases [83%]) when compared with adult patients (21 of 62 [34%]) (P < .001). Histologic evidence of BBB disruption was common in material from both adult (57 of 62 [91%]) and pediatric (65 of 81 [80%]) (P = .06) patients. In pediatric patients, however, this was a predominantly microvascular, capillary-level pathology, which was a less common finding in adult case material (mean [SD], 84.7% [8.6%] vs 31.2% [7.7%]; P < .001). Conclusions and Relevance This autopsy case series of patients dying in the acute phase after single moderate or severe TBI provides neuropathological evidence of age-dependent differences in vascular pathology. Specifically, although BBB disruption in pediatric material was typically confined to microvascular, capillary-level vessels, in adult case material, BBB disruption more typically involved larger-diameter vessels. This observation of distinct microvascular pathology in pediatric acute TBI requires further investigation. In the meantime, this study presents an intriguing potential candidate pathology contributing to diffuse brain swelling in this age group.
Collapse
Affiliation(s)
- Josie L. Fullerton
- School of Cardiovascular & Metabolic Health, University of Glasgow, Glasgow, United Kingdom
| | - Jennifer Hay
- The Francis Crick Institute, London, United Kingdom
| | | | - Josephine Atkinson
- Centre of Discovery Brain Sciences, University of Edinburgh, Edinburgh, United Kingdom
| | - Douglas H. Smith
- Penn Centre for Brain Injury and Repair, Department of Neurosurgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia
| | - William Stewart
- School of Psychology & Neuroscience, University of Glasgow, Glasgow, UK
- Department of Neuropathology, Queen Elizabeth University Hospital, Glasgow, United Kingdom
| |
Collapse
|
4
|
Mhaske A, Shukla S, Ahirwar K, Singh KK, Shukla R. Receptor-Assisted Nanotherapeutics for Overcoming the Blood-Brain Barrier. Mol Neurobiol 2024; 61:8702-8738. [PMID: 38558360 PMCID: PMC11496374 DOI: 10.1007/s12035-024-04015-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Accepted: 02/03/2024] [Indexed: 04/04/2024]
Abstract
Blood-brain barrier (BBB) is a distinguishing checkpoint that segregates peripheral organs from neural compartment. It protects the central nervous system from harmful ambush of antigens and pathogens. Owing to such explicit selectivity, the BBB hinders passage of various neuroprotective drug molecules that escalates into poor attainability of neuroprotective agents towards the brain. However, few molecules can surpass the BBB and gain access in the brain parenchyma by exploiting surface transporters and receptors. For successful development of brain-targeted therapy, understanding of BBB transporters and receptors is crucial. This review focuses on the transporter and receptor-based mechanistic pathway that can be manoeuvred for better comprehension of reciprocity of receptors and nanotechnological vehicle delivery. Nanotechnology has emerged as one of the expedient noninvasive approaches for brain targeting via manipulating the hurdle of the BBB. Various nanovehicles are being reported for brain-targeted delivery such as nanoparticles, nanocrystals, nanoemulsion, nanolipid carriers, liposomes and other nanovesicles. Nanotechnology-aided brain targeting can be a strategic approach to circumvent the BBB without altering the inherent nature of the BBB.
Collapse
Affiliation(s)
- Akshada Mhaske
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER)-Raebareli, Bijnor-Sisendi Road, Sarojini Nagar, Lucknow, Uttar Pradesh, 226002, India
| | - Shalini Shukla
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER)-Raebareli, Bijnor-Sisendi Road, Sarojini Nagar, Lucknow, Uttar Pradesh, 226002, India
| | - Kailash Ahirwar
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER)-Raebareli, Bijnor-Sisendi Road, Sarojini Nagar, Lucknow, Uttar Pradesh, 226002, India
| | - Kamalinder K Singh
- School of Pharmacy and Biomedical Sciences, University of Central Lancashire, Preston, PR1 2HE, UK.
- Biomedical Evidence-based Transdisciplinary Health Research Institute, University of Central Lancashire, Preston, PR1 2HE, UK.
| | - Rahul Shukla
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER)-Raebareli, Bijnor-Sisendi Road, Sarojini Nagar, Lucknow, Uttar Pradesh, 226002, India.
| |
Collapse
|
5
|
Suryavanshi P, Langton R, Fairhead K, Glykys J. Brief and Diverse Excitotoxic Insults Increase the Neuronal Nuclear Membrane Permeability in the Neonatal Brain, Resulting in Neuronal Dysfunction and Cell Death. J Neurosci 2024; 44:e0350242024. [PMID: 39214703 PMCID: PMC11466074 DOI: 10.1523/jneurosci.0350-24.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2024] [Revised: 08/16/2024] [Accepted: 08/23/2024] [Indexed: 09/04/2024] Open
Abstract
Neuronal cytotoxic edema is implicated in neuronal injury and death, yet mitigating brain edema with osmotic and surgical interventions yields poor clinical outcomes. Importantly, neuronal swelling and its downstream consequences during early brain development remain poorly investigated, and new treatment approaches are needed. We explored Ca2+-dependent downstream effects after neuronal cytotoxic edema caused by diverse injuries in mice of both sexes using multiphoton Ca2+ imaging in vivo [Postnatal Day (P)12-17] and in acute brain slices (P8-12). After different excitotoxic insults, cytosolic GCaMP6s translocated into the nucleus after a few minutes in a subpopulation of neurons, persisting for hours. We used an automated morphology-detection algorithm to detect neuronal soma and quantified the nuclear translocation of GCaMP6s as the nuclear to cytosolic intensity (N/C ratio). Elevated neuronal N/C ratios occurred concurrently with persistent elevation in Ca2+ loads and could also occur independently from neuronal swelling. Electron microscopy revealed that the nuclear translocation was associated with the increased nuclear pore size. The nuclear accumulation of GCaMP6s in neurons led to neocortical circuit dysfunction, mitochondrial pathology, and increased cell death. Inhibiting calpains, a family of Ca2+-activated proteases, prevented elevated N/C ratios and neuronal swelling. In summary, in the developing brain, we identified a calpain-dependent alteration of nuclear transport in a subpopulation of neurons after disease-relevant insults leading to long-term circuit dysfunction and cell death. The nuclear translocation of GCaMP6 and other cytosolic proteins after acute excitotoxicity can be an early biomarker of brain injury in the developing brain.
Collapse
Affiliation(s)
- Pratyush Suryavanshi
- Department of Pediatrics, The University of Iowa, Iowa City, Iowa 52242
- Iowa Neuroscience Institute, The University of Iowa, Iowa City, Iowa 52242
| | - Rachel Langton
- Department of Pediatrics, The University of Iowa, Iowa City, Iowa 52242
- Iowa Neuroscience Institute, The University of Iowa, Iowa City, Iowa 52242
| | - Kimberly Fairhead
- Biomedical Sciences, College of Liberal Arts and Sciences, The University of Iowa, Iowa City, Iowa 52242
| | - Joseph Glykys
- Department of Pediatrics, The University of Iowa, Iowa City, Iowa 52242
- Iowa Neuroscience Institute, The University of Iowa, Iowa City, Iowa 52242
- Department of Neurology, The University of Iowa, Iowa City, Iowa 52242
| |
Collapse
|
6
|
Baumer T, Higginbotham G, Hayes K, Thomas M. The Effect of Early Beta-Blockade with Esmolol on Therapy Intensity Level in Adults with Severe Traumatic Brain Injury. Neurotrauma Rep 2024; 5:982-987. [PMID: 39440150 PMCID: PMC11491576 DOI: 10.1089/neur.2024.0055] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2024] Open
Abstract
Following severe traumatic brain injury (TBI), elevated catecholamine levels are associated with worsened secondary brain injury and poorer clinical outcomes. The mechanisms are uncertain but may include cerebral ischemia and blood-brain barrier disruption, with consequent cerebral edema manifesting as intracranial hypertension. Early beta-blockade (EBB) may mitigate these detrimental hyperadrenergic effects. Therapy Intensity Level (TIL) is a validated score that quantifies intracranial pressure (ICP)-lowering interventions, with higher TIL being a surrogate for more severe intracranial hypertension. In this post hoc secondary analysis of a dose-finding study of EBB with esmolol in adults with TBI, we compared summary TIL (TIL24) and domain TIL between patients who received esmolol and those who did not. The primary outcome was TIL24 for each 24-h epoch of the esmolol intervention period of 96 h. Baseline characteristics were comparable in the esmolol (E) and non-esmolol (NE) groups. Mean TIL24 was similar in both groups up to 48 h but then diverged. The mean (standard deviation) TIL24 score between 48 and 72 h was 4.8 (1.5) in group E versus 6.6 (5.4) in group NE and at 72-96 h 4.5 (1.5) in group E versus 7.0 (4.0) in group NE. TIL domain scores were lower in group E for hyperosmolar therapy, targeted temperature management, and surgical management (cerebrospinal fluid drainage, evacuation, or decompressive craniectomy). The association between esmolol use after TBI and the reduction in ICP-directed interventions is consistent with an effect of beta-blockade on reduction of cerebral edema. Further research is necessary to determine causality and mechanism.
Collapse
Affiliation(s)
- Thomas Baumer
- Specialist Registrar in Intensive Care Medicine, Southmead Hospital, North Bristol NHS Trust, Bristol, UK
| | - George Higginbotham
- Academic Foundation Doctor, Southmead Hospital, North Bristol NHS Trust, Bristol, UK
| | - Kati Hayes
- Research Nurse, Southmead Hospital, North Bristol NHS Trust, Bristol, UK
| | - Matt Thomas
- Consultant in Intensive Care Medicine, Southmead Hospital, North Bristol NHS Trust, Bristol, UK
| | - for the EBB-TBI Investigators
- Specialist Registrar in Intensive Care Medicine, Southmead Hospital, North Bristol NHS Trust, Bristol, UK
- Academic Foundation Doctor, Southmead Hospital, North Bristol NHS Trust, Bristol, UK
- Research Nurse, Southmead Hospital, North Bristol NHS Trust, Bristol, UK
- Consultant in Intensive Care Medicine, Southmead Hospital, North Bristol NHS Trust, Bristol, UK
| |
Collapse
|
7
|
Vande Vyvere T, Pisică D, Wilms G, Claes L, Van Dyck P, Snoeckx A, van den Hauwe L, Pullens P, Verheyden J, Wintermark M, Dekeyzer S, Mac Donald CL, Maas AIR, Parizel PM. Imaging Findings in Acute Traumatic Brain Injury: a National Institute of Neurological Disorders and Stroke Common Data Element-Based Pictorial Review and Analysis of Over 4000 Admission Brain Computed Tomography Scans from the Collaborative European NeuroTrauma Effectiveness Research in Traumatic Brain Injury (CENTER-TBI) Study. J Neurotrauma 2024; 41:2248-2297. [PMID: 38482818 DOI: 10.1089/neu.2023.0553] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/20/2024] Open
Abstract
In 2010, the National Institute of Neurological Disorders and Stroke (NINDS) created a set of common data elements (CDEs) to help standardize the assessment and reporting of imaging findings in traumatic brain injury (TBI). However, as opposed to other standardized radiology reporting systems, a visual overview and data to support the proposed standardized lexicon are lacking. We used over 4000 admission computed tomography (CT) scans of patients with TBI from the Collaborative European NeuroTrauma Effectiveness Research in Traumatic Brain Injury (CENTER-TBI) study to develop an extensive pictorial overview of the NINDS TBI CDEs, with visual examples and background information on individual pathoanatomical lesion types, up to the level of supplemental and emerging information (e.g., location and estimated volumes). We documented the frequency of lesion occurrence, aiming to quantify the relative importance of different CDEs for characterizing TBI, and performed a critical appraisal of our experience with the intent to inform updating of the CDEs. In addition, we investigated the co-occurrence and clustering of lesion types and the distribution of six CT classification systems. The median age of the 4087 patients in our dataset was 50 years (interquartile range, 29-66; range, 0-96), including 238 patients under 18 years old (5.8%). Traumatic subarachnoid hemorrhage (45.3%), skull fractures (37.4%), contusions (31.3%), and acute subdural hematoma (28.9%) were the most frequently occurring CT findings in acute TBI. The ranking of these lesions was the same in patients with mild TBI (baseline Glasgow Coma Scale [GCS] score 13-15) compared with those with moderate-severe TBI (baseline GCS score 3-12), but the frequency of occurrence was up to three times higher in moderate-severe TBI. In most TBI patients with CT abnormalities, there was co-occurrence and clustering of different lesion types, with significant differences between mild and moderate-severe TBI patients. More specifically, lesion patterns were more complex in moderate-severe TBI patients, with more co-existing lesions and more frequent signs of mass effect. These patients also had higher and more heterogeneous CT score distributions, associated with worse predicted outcomes. The critical appraisal of the NINDS CDEs was highly positive, but revealed that full assessment can be time consuming, that some CDEs had very low frequencies, and identified a few redundancies and ambiguity in some definitions. Whilst primarily developed for research, implementation of CDE templates for use in clinical practice is advocated, but this will require development of an abbreviated version. In conclusion, with this study, we provide an educational resource for clinicians and researchers to help assess, characterize, and report the vast and complex spectrum of imaging findings in patients with TBI. Our data provides a comprehensive overview of the contemporary landscape of TBI imaging pathology in Europe, and the findings can serve as empirical evidence for updating the current NINDS radiologic CDEs to version 3.0.
Collapse
Affiliation(s)
- Thijs Vande Vyvere
- Department of Radiology, Antwerp University Hospital, Antwerp, Belgium
- Department of Molecular Imaging and Radiology (MIRA), Faculty of Medicine and Health Science, University of Antwerp, Antwerp, Belgium
| | - Dana Pisică
- Department of Neurosurgery, Erasmus MC - University Medical Center Rotterdam, Rotterdam, the Netherlands
- Department of Public Health, Erasmus MC - University Medical Center Rotterdam, Rotterdam, the Netherlands
| | - Guido Wilms
- Department of Radiology, University Hospitals Leuven, Leuven, Belgium
| | - Lene Claes
- icometrix, Research and Development, Leuven, Belgium
| | - Pieter Van Dyck
- Department of Radiology, Antwerp University Hospital, Antwerp, Belgium
- Department of Molecular Imaging and Radiology (MIRA), Faculty of Medicine and Health Science, University of Antwerp, Antwerp, Belgium
| | - Annemiek Snoeckx
- Department of Radiology, Antwerp University Hospital, Antwerp, Belgium
- Department of Molecular Imaging and Radiology (MIRA), Faculty of Medicine and Health Science, University of Antwerp, Antwerp, Belgium
| | - Luc van den Hauwe
- Department of Radiology, Antwerp University Hospital, Antwerp, Belgium
| | - Pim Pullens
- Department of Imaging, University Hospital Ghent; IBITech/MEDISIP, Engineering and Architecture, Ghent University; Ghent Institute for Functional and Metabolic Imaging, Ghent University, Belgium
| | - Jan Verheyden
- icometrix, Research and Development, Leuven, Belgium
| | - Max Wintermark
- Department of Neuroradiology, University of Texas MD Anderson Center, Houston, Texas, USA
| | - Sven Dekeyzer
- Department of Radiology, Antwerp University Hospital, Antwerp, Belgium
- Department of Radiology, University Hospital Ghent, Belgium
| | - Christine L Mac Donald
- Department of Neurological Surgery, School of Medicine, Harborview Medical Center, Seattle, Washington, USA
- Department of Neurological Surgery, School of Medicine, University of Washington, Seattle, Washington, USA
| | - Andrew I R Maas
- Department of Neurosurgery, Antwerp University Hospital, Antwerp, Belgium
- Department of Translational Neuroscience, Faculty of Medicine and Health Science, University of Antwerp, Antwerp, Belgium
| | - Paul M Parizel
- Department of Radiology, Royal Perth Hospital (RPH) and University of Western Australia (UWA), Perth, Australia; Western Australia National Imaging Facility (WA NIF) node, Australia
| |
Collapse
|
8
|
Balleste AF, Alvarez JC, Placeres-Uray F, Mastromatteo-Alberga P, Torres MD, Dallera CA, Dietrich WD, Parry TJ, Verdoorn TA, Billing CB, Buller B, Atkins CM. Improvement in edema and cognitive recovery after moderate traumatic brain injury with the neurosteroid prodrug NTS-104. Neurotherapeutics 2024; 21:e00456. [PMID: 39366874 PMCID: PMC11585901 DOI: 10.1016/j.neurot.2024.e00456] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2024] [Revised: 09/20/2024] [Accepted: 09/20/2024] [Indexed: 10/06/2024] Open
Abstract
Neuroactive steroids reduce mortality, decrease edema, and improve functional outcomes in preclinical and clinical traumatic brain injury (TBI) studies. In this study, we tested the efficacy of two related novel neuroactive steroids, NTS-104 and NTS-105, in a rat model of TBI. NTS-104 is a water-soluble prodrug of NTS-105, a partial progesterone receptor agonist. To investigate the effects of NTS-104 on TBI recovery, adult male Sprague Dawley rats received moderate parasagittal fluid-percussion injury or sham surgery and were treated with vehicle or NTS-104 (10 mg/kg, intramuscularly) at 4, 10, 24, and 48 h post-TBI. The therapeutic time window was also assessed using the neuroactive steroid NTS-105 (3 mg/kg, intramuscularly). Edema in the parietal cortex and hippocampus, measured at 3 days post-injury (DPI), was reduced by NTS-104 and NTS-105. NTS-105 was effective in reducing edema when given at 4, 10, or 24 h post-injury. Sensorimotor deficits in the cylinder test at 3 DPI were ameliorated by NTS-104 and NTS-105 treatment. Cognitive recovery, assessed with cue and contextual fear conditioning and retention of the water maze task assessed subacutely 1-3 weeks post-injury, also improved with NTS-104 treatment. Cortical and hippocampal atrophy at 22 DPI did not improve, indicating that NTS-104/NTS-105 may promote post-TBI cognitive recovery by controlling edema and other processes. These results demonstrate that NTS-104/NTS-105 is a promising therapeutic approach to improve motor and cognitive recovery after moderate TBI.
Collapse
Affiliation(s)
- Alyssa F Balleste
- The Miami Project to Cure Paralysis, Department of Neurological Surgery, University of Miami Miller School of Medicine, 1095 NW 14th Terrace, Miami, FL, USA
| | - Jacqueline C Alvarez
- The Miami Project to Cure Paralysis, Department of Neurological Surgery, University of Miami Miller School of Medicine, 1095 NW 14th Terrace, Miami, FL, USA
| | - Fabiola Placeres-Uray
- The Miami Project to Cure Paralysis, Department of Neurological Surgery, University of Miami Miller School of Medicine, 1095 NW 14th Terrace, Miami, FL, USA
| | - Patrizzia Mastromatteo-Alberga
- The Miami Project to Cure Paralysis, Department of Neurological Surgery, University of Miami Miller School of Medicine, 1095 NW 14th Terrace, Miami, FL, USA
| | - Maria Dominguez Torres
- The Miami Project to Cure Paralysis, Department of Neurological Surgery, University of Miami Miller School of Medicine, 1095 NW 14th Terrace, Miami, FL, USA
| | - Carlos A Dallera
- The Miami Project to Cure Paralysis, Department of Neurological Surgery, University of Miami Miller School of Medicine, 1095 NW 14th Terrace, Miami, FL, USA
| | - W Dalton Dietrich
- The Miami Project to Cure Paralysis, Department of Neurological Surgery, University of Miami Miller School of Medicine, 1095 NW 14th Terrace, Miami, FL, USA
| | | | | | | | | | - Coleen M Atkins
- The Miami Project to Cure Paralysis, Department of Neurological Surgery, University of Miami Miller School of Medicine, 1095 NW 14th Terrace, Miami, FL, USA.
| |
Collapse
|
9
|
Li S, Xu J, Qian Y, Zhang R. Hydrogel in the Treatment of Traumatic Brain Injury. Biomater Res 2024; 28:0085. [PMID: 39328790 PMCID: PMC11425593 DOI: 10.34133/bmr.0085] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2024] [Revised: 08/16/2024] [Accepted: 08/30/2024] [Indexed: 09/28/2024] Open
Abstract
The high prevalence of traumatic brain injury (TBI) poses an important global public health challenge. Current treatment modalities for TBI primarily involve pharmaceutical interventions and surgical procedures; however, the efficacy of these approaches remains limited. In the field of regenerative medicine, hydrogels have garnered significant attention and research efforts. This review provides an overview of the existing landscape and pathological manifestations of TBI, with a specific emphasis on delineating the therapeutic potential of hydrogels incorporated with various bioactive agents for TBI management. Particularly, the review delves into the utilization and efficacy of hydrogels based on extracellular matrix (ECM), stem cell-loaded, drug-loaded, self-assembled peptide structures or conductive in the context of TBI treatment. These applications are shown to yield favorable outcomes such as tissue damage mitigation, anti-inflammatory effects, attenuation of oxidative stress, anti-apoptotic properties, promotion of neurogenesis, and facilitation of angiogenesis. Lastly, a comprehensive analysis of the merits and constraints associated with hydrogel utilization in TBI treatment is presented, aiming to steer and advance future research endeavors in this domain.
Collapse
Affiliation(s)
- Shanhe Li
- Institute of Medical Technology, Shanxi Medical University, Taiyuan 030001, China
| | - Jiajun Xu
- Institute of Medical Technology, Shanxi Medical University, Taiyuan 030001, China
| | - Yuqing Qian
- Institute of Medical Technology, Shanxi Medical University, Taiyuan 030001, China
| | - Ruiping Zhang
- The Radiology Department of Shanxi Provincial People' Hospital Affiliated to Shanxi Medical University, Taiyuan 030001, China
| |
Collapse
|
10
|
Vidyadharan S, Rao BVVSNP, Yogeeswari P, Kesavadas C, Rajagopalan V. Accurate low and high grade glioma classification using free water eliminated diffusion tensor metrics and ensemble machine learning. Sci Rep 2024; 14:19844. [PMID: 39191905 PMCID: PMC11350135 DOI: 10.1038/s41598-024-70627-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2024] [Accepted: 08/19/2024] [Indexed: 08/29/2024] Open
Abstract
Glioma, a predominant type of brain tumor, can be fatal. This necessitates an early diagnosis and effective treatment strategies. Current diagnosis is based on biopsy, prompting the need for non invasive neuroimaging alternatives. Diffusion tensor imaging (DTI) is a promising method for studying the pathophysiological impact of tumors on white matter (WM) tissue. Single-shell DTI studies in brain glioma patients have not accounted for free water (FW) contamination due to tumors. This study aimed to (a) assess the efficacy of a two-compartment DTI model that accounts for FW contamination and (b) identify DTI-based biomarkers to classify low-grade glioma (LGG) and high-grade glioma (HGG) patients. DTI data from 86 patients (LGG n = 39, HGG n = 47) were obtained using a routine clinical imaging protocol. DTI metrics of tumorous regions and normal-appearing white matter (NAWM) were evaluated. Advanced stacked-based ensemble learning was employed to classify LGG and HGG patients using both single- and two-compartment DTI model measures. The DTI metrics of the two-compartment model outperformed those of the standard single-compartment DTI model in terms of sensitivity, specificity, and area under the curve of receiver operating characteristic (AUC-ROC) score in classifying LGG and HGG patients. Four features (out of 16 features), namely fractional anisotropy (FA) of the edema and core region and FA and mean diffusivity of the NAWM region, showed superior performance (sensitivity = 92%, specificity = 90%, and AUC-ROC = 90%) in classifying LGG and HGG. This demonstrates that both tumorous and NAWM regions may be differentially affected in LGG and HGG patients. Our results demonstrate the significance of using a two-compartment DTI model that accounts for FW contamination by improving diagnostic accuracy. This improvement may eventually aid in planning treatment strategies for glioma patients.
Collapse
Affiliation(s)
- Sreejith Vidyadharan
- Department of Electrical and Electronics Engineering, Birla Institute of Technology and Science Pilani, Hyderabad Campus, Hyderabad, 500078, India
| | - B V V S N Prabhakar Rao
- Department of Electrical and Electronics Engineering, Birla Institute of Technology and Science Pilani, Hyderabad Campus, Hyderabad, 500078, India
| | - P Yogeeswari
- Department of Pharmacy, Birla Institute of Technology and Science Pilani, Hyderabad Campus, Hyderabad, 500078, India
| | - C Kesavadas
- Department of Imaging Sciences and Interventional Radiology, Sree Chitra Tirunal Institute for Medical Sciences and Technology, Trivandrum, 695011, India
| | - Venkateswaran Rajagopalan
- Department of Electrical and Electronics Engineering, Birla Institute of Technology and Science Pilani, Hyderabad Campus, Hyderabad, 500078, India.
| |
Collapse
|
11
|
El-Menyar A, Asim M, Khan N, Rizoli S, Mahmood I, Al-Ani M, Kanbar A, Alaieb A, Hakim S, Younis B, Taha I, Jogol H, Siddiqui T, Hammo AA, Abdurraheim N, Alabdallat M, Bahey AAA, Ahmed K, Atique S, Chaudry IH, Prabhu KS, Uddin S, Al-Thani H. Systemic and cerebro-cardiac biomarkers following traumatic brain injury: an interim analysis of randomized controlled clinical trial of early administration of beta blockers. Sci Rep 2024; 14:19574. [PMID: 39179700 PMCID: PMC11343837 DOI: 10.1038/s41598-024-70470-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2024] [Accepted: 08/16/2024] [Indexed: 08/26/2024] Open
Abstract
This is an interim analysis of the Beta-blocker (Propranolol) use in traumatic brain injury (TBI) based on the high-sensitive troponin status (BBTBBT) study. The BBTBBT is an ongoing double-blind placebo-controlled randomized clinical trial with a target sample size of 771 patients with TBI. We sought, after attaining 50% of the sample size, to explore the impact of early administration of beta-blockers (BBs) on the adrenergic surge, pro-inflammatory cytokines, and the TBI biomarkers linked to the status of high-sensitivity troponin T (HsTnT). Patients were stratified based on the severity of TBI using the Glasgow coma scale (GCS) and HsTnT status (positive vs negative) before randomization. Patients with positive HsTnT (non-randomized) received propranolol (Group-1; n = 110), and those with negative test were randomized to receive propranolol (Group-2; n = 129) or placebo (Group-3; n = 111). Propranolol was administered within 24 h of injury for 6 days, guided by the heart rate (> 60 bpm), systolic blood pressure (≥ 100 mmHg), or mean arterial pressure (> 70 mmHg). Luminex and ELISA-based immunoassays were used to quantify the serum levels of pro-inflammatory cytokines (Interleukin (IL)-1β, IL-6, IL-8, and IL-18), TBI biomarkers [S100B, Neuron-Specific Enolase (NSE), and epinephrine]. Three hundred and fifty patients with comparable age (mean 34.8 ± 9.9 years) and gender were enrolled in the interim analysis. Group 1 had significantly higher baseline levels of IL-6, IL-1B, S100B, lactate, and base deficit than the randomized groups (p = 0.001). Group 1 showed a significant temporal reduction in serum IL-6, IL-1β, epinephrine, and NSE levels from baseline to 48 h post-injury (p = 0.001). Patients with severe head injuries had higher baseline levels of IL-6, IL-1B, S100B, and HsTnT than mild and moderate TBI (p = 0.01). HsTnT levels significantly correlated with the Injury Severity Score (ISS) (r = 0.275, p = 0.001), GCS (r = - 0.125, p = 0.02), and serum S100B (r = 0.205, p = 0.001). Early Propranolol administration showed a significant reduction in cytokine levels and TBI biomarkers from baseline to 48 h post-injury, particularly among patients with positive HsTnT, indicating the potential role in modulating inflammation post-TBI.Trial registration: ClinicalTrials.gov NCT04508244. It was registered first on 11/08/2020. Recruitment started on 29 December 2020 and is ongoing. The study was partly presented at the 23rd European Congress of Trauma and Emergency Surgery (ECTES), April 28-30, 2024, in Estoril, Lisbon, Portugal.
Collapse
Affiliation(s)
- Ayman El-Menyar
- Department of Surgery, Clinical Research, Trauma and Vascular Surgery, Hamad Medical Corporation, Doha, Qatar.
- Department of Clinical Medicine, Weill Cornell Medicine, P.O. Box 24144, Doha, Qatar.
| | - Mohammad Asim
- Department of Surgery, Clinical Research, Trauma and Vascular Surgery, Hamad Medical Corporation, Doha, Qatar
| | - Naushad Khan
- Department of Surgery, Clinical Research, Trauma and Vascular Surgery, Hamad Medical Corporation, Doha, Qatar
| | - Sandro Rizoli
- Department of Surgery, Trauma Surgery, Hamad Medical Corporation, Doha, Qatar
| | - Ismail Mahmood
- Department of Surgery, Trauma Surgery, Hamad Medical Corporation, Doha, Qatar
| | - Mushreq Al-Ani
- Department of Surgery, Trauma Surgery, Hamad Medical Corporation, Doha, Qatar
| | - Ahad Kanbar
- Department of Surgery, Trauma Surgery, Hamad Medical Corporation, Doha, Qatar
| | - Abubaker Alaieb
- Department of Surgery, Trauma Surgery, Hamad Medical Corporation, Doha, Qatar
| | - Suhail Hakim
- Department of Surgery, Trauma Surgery, Hamad Medical Corporation, Doha, Qatar
| | - Basil Younis
- Department of Surgery, Trauma Surgery, Hamad Medical Corporation, Doha, Qatar
| | - Ibrahim Taha
- Department of Surgery, Trauma Surgery, Hamad Medical Corporation, Doha, Qatar
| | - Hisham Jogol
- Department of Surgery, Trauma Surgery, Hamad Medical Corporation, Doha, Qatar
| | - Tariq Siddiqui
- Department of Surgery, Trauma Surgery, Hamad Medical Corporation, Doha, Qatar
| | - Abdel Aziz Hammo
- Department of Surgery, Trauma Surgery, Hamad Medical Corporation, Doha, Qatar
| | - Nuri Abdurraheim
- Department of Surgery, Trauma Surgery, Hamad Medical Corporation, Doha, Qatar
| | - Mohammad Alabdallat
- Department of Surgery, Trauma Surgery, Hamad Medical Corporation, Doha, Qatar
| | | | - Khalid Ahmed
- Department of Surgery, Trauma Surgery, Hamad Medical Corporation, Doha, Qatar
| | - Sajid Atique
- Department of Surgery, Trauma Surgery, Hamad Medical Corporation, Doha, Qatar
| | - Irshad H Chaudry
- Department of Surgery, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Kirti S Prabhu
- Translational Research Institute, Academic Health System, Hamad Medical Corporation, Doha, Qatar
| | - Shahab Uddin
- Translational Research Institute, Academic Health System, Hamad Medical Corporation, Doha, Qatar
| | - Hassan Al-Thani
- Department of Surgery, Trauma Surgery, Hamad Medical Corporation, Doha, Qatar
| |
Collapse
|
12
|
Singh A, Gong S, Vu A, Li S, Obenaus A. Social deficits mirror delayed cerebrovascular dysfunction after traumatic brain injury. Acta Neuropathol Commun 2024; 12:126. [PMID: 39107831 PMCID: PMC11304659 DOI: 10.1186/s40478-024-01840-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2024] [Accepted: 07/28/2024] [Indexed: 08/10/2024] Open
Abstract
Traumatic brain injury (TBI) survivors face debilitating long-term psychosocial consequences, including social isolation and depression. TBI modifies neurovascular physiology and behavior but the chronic physiological implications of altered brain perfusion on social interactions are unknown. Adult C57/BL6 male mice received a moderate cortical TBI, and social behaviors were assessed at baseline, 3-, 7-, 14-, 30-, and 60-days post injury (dpi). Magnetic resonance imaging (MRI, 9.4T) using dynamic susceptibility contrast perfusion weighted MRI were acquired. At 60dpi mice underwent histological angioarchitectural mapping. Analysis utilized standardized protocols followed by cross-correlation metrics. Social behavior deficits at 60dpi emerged as reduced interactions with a familiar cage-mate (partner) that mirrored significant reductions in cerebral blood flow (CBF) at 60dpi. CBF perturbations were dynamic temporally and across brain regions including regions known to regulate social behavior such as hippocampus, hypothalamus, and rhinal cortex. Social isolation in TBI-mice emerged with a significant decline in preference to spend time with a cage mate. Cortical vascular density was also reduced corroborating the decline in brain perfusion and social interactions. Thus, the late emergence of social interaction deficits mirrored the reduced vascular density and CBF in regions known to be involved in social behaviors. Vascular morphology and function improved prior to the late decrements in social function and our correlations strongly implicate a linkage between vascular density, cerebral perfusion, and social interactions. Our study provides a clinically relevant timeline of alterations in social deficits alongside functional vascular recovery that can guide future therapeutics.
Collapse
Affiliation(s)
- Aditya Singh
- Department of Pediatrics, School of Medicine, University of California Irvine, Hewitt Hall Rm. 2066, Irvine, CA, 92697, USA
- Department of Neurology, The Lundquist Institute for Biomedical Innovation at Harbor-UCLA, 120 Walter P Martin Research Center, Torrance, California, 90502, USA
- Department of Neurosurgery, University of California Los Angeles, Los Angeles, California, 90095, USA
| | - Steven Gong
- Department of Pediatrics, School of Medicine, University of California Irvine, Hewitt Hall Rm. 2066, Irvine, CA, 92697, USA
| | - Anh Vu
- Department of Pediatrics, School of Medicine, University of California Irvine, Hewitt Hall Rm. 2066, Irvine, CA, 92697, USA
| | - Scott Li
- Department of Pediatrics, School of Medicine, University of California Irvine, Hewitt Hall Rm. 2066, Irvine, CA, 92697, USA
| | - Andre Obenaus
- Department of Pediatrics, School of Medicine, University of California Irvine, Hewitt Hall Rm. 2066, Irvine, CA, 92697, USA.
- Division of Biomedical Sciences, 206 SOM Research Bldg, University of California Riverside, Riverside, CA, 92521, USA.
| |
Collapse
|
13
|
Dekundy A, Pichler G, El Badry R, Scheschonka A, Danysz W. Amantadine for Traumatic Brain Injury-Supporting Evidence and Mode of Action. Biomedicines 2024; 12:1558. [PMID: 39062131 PMCID: PMC11274811 DOI: 10.3390/biomedicines12071558] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2024] [Accepted: 07/10/2024] [Indexed: 07/28/2024] Open
Abstract
Traumatic brain injury (TBI) is an important global clinical issue, requiring not only prevention but also effective treatment. Following TBI, diverse parallel and intertwined pathological mechanisms affecting biochemical, neurochemical, and inflammatory pathways can have a severe impact on the patient's quality of life. The current review summarizes the evidence for the utility of amantadine in TBI in connection to its mechanism of action. Amantadine, the drug combining multiple mechanisms of action, may offer both neuroprotective and neuroactivating effects in TBI patients. Indeed, the use of amantadine in TBI has been encouraged by several clinical practice guidelines/recommendations. Amantadine is also available as an infusion, which may be of particular benefit in unconscious patients with TBI due to immediate delivery to the central nervous system and the possibility of precise dosing. In other situations, orally administered amantadine may be used. There are several questions that remain to be addressed: can amantadine be effective in disorders of consciousness requiring long-term treatment and in combination with drugs approved for the treatment of TBI? Do the observed beneficial effects of amantadine extend to disorders of consciousness due to factors other than TBI? Well-controlled clinical studies are warranted to ultimately confirm its utility in the TBI and provide answers to these questions.
Collapse
Affiliation(s)
- Andrzej Dekundy
- Merz Therapeutics GmbH, Eckenheimer Landstraße 100, 60318 Frankfurt am Main, Germany; (A.D.); (A.S.)
| | - Gerald Pichler
- Department of Neurology, Albert-Schweitzer-Hospital Graz, Albert-Schweitzer-Gasse 36, 8020 Graz, Austria;
| | - Reda El Badry
- Department of Neurology and Psychiatry, Faculty of Medicine, Assiut University Hospital, Assiut University, Assiut 71526, Egypt;
| | - Astrid Scheschonka
- Merz Therapeutics GmbH, Eckenheimer Landstraße 100, 60318 Frankfurt am Main, Germany; (A.D.); (A.S.)
| | - Wojciech Danysz
- Danysz Pharmacology Consulting, Vor den Gärten 16, 61130 Nidderau, Germany
| |
Collapse
|
14
|
Tan H, Wang J, Li F, Peng Y, Lan J, Zhang Y, Zhao D, Bao Y. Prediction Value of Initial Serum Levels of SERPINA3 in Intracranial Pressure and Long-Term Neurological Outcomes in Traumatic Brain Injury. Diagnostics (Basel) 2024; 14:1245. [PMID: 38928660 PMCID: PMC11202773 DOI: 10.3390/diagnostics14121245] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2024] [Revised: 06/04/2024] [Accepted: 06/11/2024] [Indexed: 06/28/2024] Open
Abstract
Traumatic brain injury (TBI) is a severe neurological condition characterized by inflammation in the central nervous system. SERPINA3 has garnered attention as a potential biomarker for assessing this inflammation. Our study aimed to explore the predictive value of postoperative serum SERPINA3 levels in identifying the risk of cerebral edema and its prognostic implications in TBI. This study is a prospective observational study, including 37 patients with TBI who finally met our criteria. The Glasgow Outcome Scale (GOS), Levels of Cognitive Functioning (LCF), Disability Rating Scale (DRS), and Early Rehabilitation Barthel Index (ERBI) scores at six months after trauma were defined as the main study endpoint. We further calculated the ventricle-to-intracranial-volume ratio (VBR) at 6 months from CT scans. The study included patients with Glasgow Coma Scale (GCS) scores ranging from 3 to 8, who were subsequently categorized into two groups: the critical TBI group (GCS 3-5 points) and the severe TBI group (GCS 6-8 points). Within the critical TBI group, SERPINA3 levels were notably lower. However, among patients with elevated SERPINA3 levels, both the peak intracranial pressure (ICP) and average mannitol consumption were significantly reduced compared with those of patients with lower SERPINA3 levels. In terms of the 6-month outcomes measured via the GOS, LCF, DRS, and ERBI, lower levels of SERPINA3 were indicative of poorer prognosis. Furthermore, we found a negative correlation between serum SERPINA3 levels and the VBR. The receiver operating characteristic (ROC) curve and decision curve analysis (DCA) demonstrated the predictive performance of SERPINA3. In conclusion, incorporating the novel biomarker SERPINA3 alongside traditional assessment tools offers neurosurgeons an effective and easily accessible means, which is readily accessible early on, to predict the risk of intracranial pressure elevation and long-term prognosis in TBI patients.
Collapse
Affiliation(s)
- Haoyuan Tan
- Department of Neurosurgery, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200127, China; (H.T.); (J.W.); (J.L.)
| | - Jiamian Wang
- Department of Neurosurgery, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200127, China; (H.T.); (J.W.); (J.L.)
| | - Fengshi Li
- Neurologic Surgery Department, Huashan Hospital, Fudan University, Shanghai 200437, China;
| | - Yidong Peng
- Brain Injury Center, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai Institute of Head Trauma, Shanghai 200127, China;
| | - Jin Lan
- Department of Neurosurgery, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200127, China; (H.T.); (J.W.); (J.L.)
| | - Yuanda Zhang
- Minhang Hospital, Fudan University, Shanghai 200437, China;
| | - Dongxu Zhao
- Department of Neurosurgery, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200127, China; (H.T.); (J.W.); (J.L.)
| | - Yinghui Bao
- Department of Neurosurgery, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200127, China; (H.T.); (J.W.); (J.L.)
| |
Collapse
|
15
|
Nguyen P, Albayram M, Tuna I. Intramyelinic edema manifesting as central white matter diffusion restriction associated with brain contusion in pediatric patients. Neuroradiol J 2024:19714009241260796. [PMID: 38856642 PMCID: PMC11571443 DOI: 10.1177/19714009241260796] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/11/2024] Open
Abstract
In traumatic brain injury, white matter diffusion restriction can be an imaging manifestation of non-hemorrhagic axonal injury. In this article, a different pattern of widespread white matter diffusion restriction associated with ipsilateral cortical damage, all noted in pediatric and young adult TBI patients, is presented. Its atypical pattern of distribution and extensive scope on imaging suggest excitotoxicity and intramyelinic edema as possible underlying mechanisms.
Collapse
Affiliation(s)
- Phuong Nguyen
- Department of Radiology, College of Medicine, University of Florida, Gainesville, FL, USA
| | - Mehmet Albayram
- Department of Radiology, College of Medicine, University of Florida, Gainesville, FL, USA
| | - Ibrahim Tuna
- Department of Radiology, College of Medicine, University of Florida, Gainesville, FL, USA
| |
Collapse
|
16
|
Sun F, Zhou J, Chen X, Yang T, Wang G, Ge J, Zhang Z, Mei Z. No-reflow after recanalization in ischemic stroke: From pathomechanisms to therapeutic strategies. J Cereb Blood Flow Metab 2024; 44:857-880. [PMID: 38420850 PMCID: PMC11318407 DOI: 10.1177/0271678x241237159] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/10/2023] [Revised: 01/07/2024] [Accepted: 02/18/2024] [Indexed: 03/02/2024]
Abstract
Endovascular reperfusion therapy is the primary strategy for acute ischemic stroke. No-reflow is a common phenomenon, which is defined as the failure of microcirculatory reperfusion despite clot removal by thrombolysis or mechanical embolization. It has been reported that up to 25% of ischemic strokes suffer from no-reflow, which strongly contributes to an increased risk of poor clinical outcomes. No-reflow is associated with functional and structural alterations of cerebrovascular microcirculation, and the injury to the microcirculation seriously hinders the neural functional recovery following macrovascular reperfusion. Accumulated evidence indicates that pathology of no-reflow is linked to adhesion, aggregation, and rolling of blood components along the endothelium, capillary stagnation with neutrophils, astrocytes end-feet, and endothelial cell edema, pericyte contraction, and vasoconstriction. Prevention or treatment strategies aim to alleviate or reverse these pathological changes, including targeted therapies such as cilostazol, adhesion molecule blocking antibodies, peroxisome proliferator-activated receptors (PPARs) activator, adenosine, pericyte regulators, as well as adjunctive therapies, such as extracorporeal counterpulsation, ischemic preconditioning, and alternative or complementary therapies. Herein, we provide an overview of pathomechanisms, predictive factors, diagnosis, and intervention strategies for no-reflow, and attempt to convey a new perspective on the clinical management of no-reflow post-ischemic stroke.
Collapse
Affiliation(s)
- Feiyue Sun
- Key Laboratory of Hunan Province for Integrated Traditional Chinese and Western Medicine on Prevention and Treatment of Cardio-Cerebral Diseases, College of Integrated Traditional Chinese Medicine and Western Medicine, Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Jing Zhou
- Key Laboratory of Hunan Province for Integrated Traditional Chinese and Western Medicine on Prevention and Treatment of Cardio-Cerebral Diseases, College of Integrated Traditional Chinese Medicine and Western Medicine, Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Xiangyu Chen
- Key Laboratory of Hunan Province for Integrated Traditional Chinese and Western Medicine on Prevention and Treatment of Cardio-Cerebral Diseases, College of Integrated Traditional Chinese Medicine and Western Medicine, Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Tong Yang
- Key Laboratory of Hunan Province for Integrated Traditional Chinese and Western Medicine on Prevention and Treatment of Cardio-Cerebral Diseases, College of Integrated Traditional Chinese Medicine and Western Medicine, Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Guozuo Wang
- Key Laboratory of Hunan Province for Integrated Traditional Chinese and Western Medicine on Prevention and Treatment of Cardio-Cerebral Diseases, College of Integrated Traditional Chinese Medicine and Western Medicine, Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Jinwen Ge
- Key Laboratory of Hunan Province for Integrated Traditional Chinese and Western Medicine on Prevention and Treatment of Cardio-Cerebral Diseases, College of Integrated Traditional Chinese Medicine and Western Medicine, Hunan University of Chinese Medicine, Changsha, Hunan, China
- Hunan Academy of Chinese Medicine, Changsha, Hunan, China
| | - Zhanwei Zhang
- The First Affiliated Hospital of Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Zhigang Mei
- Key Laboratory of Hunan Province for Integrated Traditional Chinese and Western Medicine on Prevention and Treatment of Cardio-Cerebral Diseases, College of Integrated Traditional Chinese Medicine and Western Medicine, Hunan University of Chinese Medicine, Changsha, Hunan, China
- Third-Grade Pharmacological Laboratory on Chinese Medicine Approved by State Administration of Traditional Chinese Medicine, College of Medicine and Health Sciences, China Three Gorges University, Yichang, Hubei, China
| |
Collapse
|
17
|
El Baassiri MG, Raouf Z, Badin S, Escobosa A, Sodhi CP, Nasr IW. Dysregulated brain-gut axis in the setting of traumatic brain injury: review of mechanisms and anti-inflammatory pharmacotherapies. J Neuroinflammation 2024; 21:124. [PMID: 38730498 PMCID: PMC11083845 DOI: 10.1186/s12974-024-03118-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2024] [Accepted: 04/30/2024] [Indexed: 05/13/2024] Open
Abstract
Traumatic brain injury (TBI) is a chronic and debilitating disease, associated with a high risk of psychiatric and neurodegenerative diseases. Despite significant advancements in improving outcomes, the lack of effective treatments underscore the urgent need for innovative therapeutic strategies. The brain-gut axis has emerged as a crucial bidirectional pathway connecting the brain and the gastrointestinal (GI) system through an intricate network of neuronal, hormonal, and immunological pathways. Four main pathways are primarily implicated in this crosstalk, including the systemic immune system, autonomic and enteric nervous systems, neuroendocrine system, and microbiome. TBI induces profound changes in the gut, initiating an unrestrained vicious cycle that exacerbates brain injury through the brain-gut axis. Alterations in the gut include mucosal damage associated with the malabsorption of nutrients/electrolytes, disintegration of the intestinal barrier, increased infiltration of systemic immune cells, dysmotility, dysbiosis, enteroendocrine cell (EEC) dysfunction and disruption in the enteric nervous system (ENS) and autonomic nervous system (ANS). Collectively, these changes further contribute to brain neuroinflammation and neurodegeneration via the gut-brain axis. In this review article, we elucidate the roles of various anti-inflammatory pharmacotherapies capable of attenuating the dysregulated inflammatory response along the brain-gut axis in TBI. These agents include hormones such as serotonin, ghrelin, and progesterone, ANS regulators such as beta-blockers, lipid-lowering drugs like statins, and intestinal flora modulators such as probiotics and antibiotics. They attenuate neuroinflammation by targeting distinct inflammatory pathways in both the brain and the gut post-TBI. These therapeutic agents exhibit promising potential in mitigating inflammation along the brain-gut axis and enhancing neurocognitive outcomes for TBI patients.
Collapse
Affiliation(s)
- Mahmoud G El Baassiri
- Pediatric Surgery, Department of Surgery, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA
| | - Zachariah Raouf
- Pediatric Surgery, Department of Surgery, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA
| | - Sarah Badin
- Pediatric Surgery, Department of Surgery, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA
| | - Alejandro Escobosa
- Pediatric Surgery, Department of Surgery, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA
| | - Chhinder P Sodhi
- Pediatric Surgery, Department of Surgery, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA
| | - Isam W Nasr
- Pediatric Surgery, Department of Surgery, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA.
| |
Collapse
|
18
|
Caldwell DJ, Scheer JK, Umbach G, Aghi MK. Acute hyponatremia post craniotomy resulting in a unilateral fixed and dilated pupil: A case study on diagnosis and management. Surg Neurol Int 2024; 15:160. [PMID: 38840620 PMCID: PMC11152517 DOI: 10.25259/sni_105_2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2024] [Accepted: 04/16/2024] [Indexed: 06/07/2024] Open
Abstract
Background Postoperative hyponatremia is a known complication of intracranial surgery, which can present with depressed mental status. Hyponatremia resulting in focal neurologic deficits is less frequently described. Case Description We describe a patient who, after a bifrontal craniotomy for olfactory groove meningioma, developed acute hyponatremia overnight with a decline in mental status from Glasgow coma scale (GCS) score 15 to GCS 7 and a unilateral fixed dilated pupil. Head computed tomography showed expected postoperative changes without new acute or localizing findings, such as unilateral uncal herniation. The patient's mental status and pupil immediately improved with the administration of mannitol; however, there was a subsequent decline in mental status with a preserved pupil later that morning. Hypertonic saline reversed the neurologic change, and the patient was eventually discharged without a neurologic deficit. Focal neurologic deficits need not always arise following a craniotomy from a postoperative hematoma, stroke, or other finding with radiographic correlate. Conclusion Post-craniotomy hyponatremia should now be seen as a postoperative complication that can result in both a general neurologic decline in mental status, as well as with focal neurologic signs such as a fixed, dilated pupil, which can be reversed with hyperosmolar therapy and correction of the hyponatremia.
Collapse
Affiliation(s)
- David J. Caldwell
- Department of Neurological Surgery, University of California San Francisco, San Francisco, California, United States
| | | | | | | |
Collapse
|
19
|
Chen X, Józsa TI, Cardim D, Robba C, Czosnyka M, Payne SJ. Modelling midline shift and ventricle collapse in cerebral oedema following acute ischaemic stroke. PLoS Comput Biol 2024; 20:e1012145. [PMID: 38805558 PMCID: PMC11161059 DOI: 10.1371/journal.pcbi.1012145] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2023] [Revised: 06/07/2024] [Accepted: 05/08/2024] [Indexed: 05/30/2024] Open
Abstract
In ischaemic stroke, a large reduction in blood supply can lead to the breakdown of the blood-brain barrier and to cerebral oedema after reperfusion therapy. The resulting fluid accumulation in the brain may contribute to a significant rise in intracranial pressure (ICP) and tissue deformation. Changes in the level of ICP are essential for clinical decision-making and therapeutic strategies. However, the measurement of ICP is constrained by clinical techniques and obtaining the exact values of the ICP has proven challenging. In this study, we propose the first computational model for the simulation of cerebral oedema following acute ischaemic stroke for the investigation of ICP and midline shift (MLS) relationship. The model consists of three components for the simulation of healthy blood flow, occluded blood flow and oedema, respectively. The healthy and occluded blood flow components are utilized to obtain oedema core geometry and then imported into the oedema model for the simulation of oedema growth. The simulation results of the model are compared with clinical data from 97 traumatic brain injury patients for the validation of major model parameters. Midline shift has been widely used for the diagnosis, clinical decision-making, and prognosis of oedema patients. Therefore, we focus on quantifying the relationship between ICP and midline shift (MLS) and identify the factors that can affect the ICP-MLS relationship. Three major factors are investigated, including the brain geometry, blood-brain barrier damage severity and the types of oedema (including rare types of oedema). Meanwhile, the two major types (stress and tension/compression) of mechanical brain damage are also presented and the differences in the stress, tension, and compression between the intraparenchymal and periventricular regions are discussed. This work helps to predict ICP precisely and therefore provides improved clinical guidance for the treatment of brain oedema.
Collapse
Affiliation(s)
- Xi Chen
- Institute of Biomedical Engineering, Department of Engineering Science, University of Oxford, Oxford, United Kingdom
| | - Tamás I. Józsa
- School of Aerospace, Transport and Manufacturing Cranfield University, Cranfield, United Kingdom
| | - Danilo Cardim
- Department of Neurology, University of Texas Southwestern Medical Centre, Dallas, Texas, United States of America
- Institute for Exercise and Environmental Medicine, Texas Health Presbyterian Hospital, Dallas, Texas, United States of America
| | - Chiara Robba
- Department of Anesthesia and Intensive Care, IRCCS Ospedale Policlinico San Martino, Genova, Italy
| | - Marek Czosnyka
- Division of Neurosurgery, Department of Clinical Neurosciences, University of Cambridge, Cambridge, United Kingdom
- Institute of Electronic Systems, Warsaw University of Technology, Warsaw, Poland
| | - Stephen J. Payne
- Institute of Applied Mechanics, National Taiwan University, Taiwan
| |
Collapse
|
20
|
Yan A, Torpey A, Morrisroe E, Andraous W, Costa A, Bergese S. Clinical Management in Traumatic Brain Injury. Biomedicines 2024; 12:781. [PMID: 38672137 PMCID: PMC11048642 DOI: 10.3390/biomedicines12040781] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Revised: 03/06/2024] [Accepted: 03/14/2024] [Indexed: 04/28/2024] Open
Abstract
Traumatic brain injury is one of the leading causes of morbidity and mortality worldwide and is one of the major public healthcare burdens in the US, with millions of patients suffering from the traumatic brain injury itself (approximately 1.6 million/year) or its repercussions (2-6 million patients with disabilities). The severity of traumatic brain injury can range from mild transient neurological dysfunction or impairment to severe profound disability that leaves patients completely non-functional. Indications for treatment differ based on the injury's severity, but one of the goals of early treatment is to prevent secondary brain injury. Hemodynamic stability, monitoring and treatment of intracranial pressure, maintenance of cerebral perfusion pressure, support of adequate oxygenation and ventilation, administration of hyperosmolar agents and/or sedatives, nutritional support, and seizure prophylaxis are the mainstays of medical treatment for severe traumatic brain injury. Surgical management options include decompressive craniectomy or cerebrospinal fluid drainage via the insertion of an external ventricular drain. Several emerging treatment modalities are being investigated, such as anti-excitotoxic agents, anti-ischemic and cerebral dysregulation agents, S100B protein, erythropoietin, endogenous neuroprotectors, anti-inflammatory agents, and stem cell and neuronal restoration agents, among others.
Collapse
Affiliation(s)
- Amy Yan
- Department of Anesthesiology, Renaissance School of Medicine, Stony Brook University, Stony Brook, NY 11794, USA; (A.Y.); (A.T.); (W.A.); (A.C.)
| | - Andrew Torpey
- Department of Anesthesiology, Renaissance School of Medicine, Stony Brook University, Stony Brook, NY 11794, USA; (A.Y.); (A.T.); (W.A.); (A.C.)
| | - Erin Morrisroe
- Renaissance School of Medicine, Stony Brook University, Stony Brook, NY 11794, USA;
| | - Wesam Andraous
- Department of Anesthesiology, Renaissance School of Medicine, Stony Brook University, Stony Brook, NY 11794, USA; (A.Y.); (A.T.); (W.A.); (A.C.)
| | - Ana Costa
- Department of Anesthesiology, Renaissance School of Medicine, Stony Brook University, Stony Brook, NY 11794, USA; (A.Y.); (A.T.); (W.A.); (A.C.)
| | - Sergio Bergese
- Department of Anesthesiology and Neurological Surgery, Renaissance School of Medicine, Stony Brook University, Stony Brook, NY 11794, USA
| |
Collapse
|
21
|
Thomas M, Hayes K, White P, Ramesh A, Culliford L, Ackland G, Pickering A. Early Intravenous Beta-Blockade with Esmolol in Adults with Severe Traumatic Brain Injury (EBB-TBI): Protocol for a Phase 2a Intervention Design Study. Neurocrit Care 2024; 40:795-806. [PMID: 37308729 PMCID: PMC10959800 DOI: 10.1007/s12028-023-01755-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2023] [Accepted: 05/10/2023] [Indexed: 06/14/2023]
Abstract
Traumatic brain injury is a leading cause of death and disability worldwide. Interventions that mitigate secondary brain injury have the potential to improve outcomes for patients and reduce the impact on communities and society. Increased circulating catecholamines are associated with worse outcomes and there are supportive animal data and indications in human studies of benefit from beta-blockade after severe traumatic brain injury. Here, we present the protocol for a dose-finding study using esmolol in adults commenced within 24 h of severe traumatic brain injury. Esmolol has practical advantages and theoretical benefits as a neuroprotective agent in this setting, but these must be balanced against the known risk of secondary injury from hypotension. The aim of this study is to determine a dose schedule for esmolol, using the continual reassessment method, that combines a clinically significant reduction in heart rate as a surrogate for catecholamine drive with maintenance of cerebral perfusion pressure. The maximum tolerated dosing schedule for esmolol can then be tested for patient benefit in subsequent randomized controlled trials.Trial registration ISRCTN, ISRCTN11038397, registered retrospectively 07/01/2021 https://www.isrctn.com/ISRCTN11038397.
Collapse
Affiliation(s)
- Matt Thomas
- Intensive Care Unit, North Bristol NHS Trust, Bristol, UK.
| | - Kati Hayes
- Intensive Care Unit, North Bristol NHS Trust, Bristol, UK
| | - Paul White
- School of Data Science and Mathematics, University of the West of England, Bristol, UK
| | - Aravind Ramesh
- GW4 Clinical Academic Training Programme for Health Professionals, Faculty of Health Sciences, University of Bristol, Bristol, UK
| | - Lucy Culliford
- Bristol Trials Centre, Bristol Medical School (PHS), University of Bristol, Bristol, UK
| | - Gareth Ackland
- Translational Medicine and Therapeutics, William Harvey Research Institute, Queen Mary University of London, London, UK
| | - Anthony Pickering
- School of Physiology, Pharmacology and Neuroscience, University of Bristol, Bristol, UK
| |
Collapse
|
22
|
Mishra S, Grewal J, Wal P, Bhivshet GU, Tripathi AK, Walia V. Therapeutic potential of vasopressin in the treatment of neurological disorders. Peptides 2024; 174:171166. [PMID: 38309582 DOI: 10.1016/j.peptides.2024.171166] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/19/2023] [Revised: 01/18/2024] [Accepted: 01/29/2024] [Indexed: 02/05/2024]
Abstract
Vasopressin (VP) is a nonapeptide made of nine amino acids synthesized by the hypothalamus and released by the pituitary gland. VP acts as a neurohormone, neuropeptide and neuromodulator and plays an important role in the regulation of water balance, osmolarity, blood pressure, body temperature, stress response, emotional challenges, etc. Traditionally VP is known to regulate the osmolarity and tonicity. VP and its receptors are widely expressed in the various region of the brain including cortex, hippocampus, basal forebrain, amygdala, etc. VP has been shown to modulate the behavior, stress response, circadian rhythm, cerebral blood flow, learning and memory, etc. The potential role of VP in the regulation of these neurological functions have suggested the therapeutic importance of VP and its analogues in the management of neurological disorders. Further, different VP analogues have been developed across the world with different pharmacotherapeutic potential. In the present work authors highlighted the therapeutic potential of VP and its analogues in the treatment and management of various neurological disorders.
Collapse
Affiliation(s)
- Shweta Mishra
- SGT College of Pharmacy, SGT University, Gurugram, India
| | - Jyoti Grewal
- Maharisi Markandeshwar University, Sadopur, India
| | - Pranay Wal
- Pranveer Singh Institute of Pharmacy, Kanpur, India
| | | | | | - Vaibhav Walia
- SGT College of Pharmacy, SGT University, Gurugram, India.
| |
Collapse
|
23
|
Yang H, Song P, Li B, Li S, Yang J. Design, synthesis and biological evaluation of Nrf2 modulators for the treatment of glioblastoma multiforme. Bioorg Med Chem 2024; 103:117684. [PMID: 38493731 DOI: 10.1016/j.bmc.2024.117684] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2024] [Revised: 02/29/2024] [Accepted: 03/13/2024] [Indexed: 03/19/2024]
Abstract
Glioblastoma multiforme (GBM) is a prevalent primary brain tumor. However, no specific therapeutic drug has been developed for it. Nuclear factor erythroid 2-related factor 2 (Nrf2) is a crucial transcription factor involved in the cellular response to oxidative stress. Numerous studies have demonstrated that Nrf2 plays a pivotal role in GBM angiogenesis, and inhibiting Nrf2 can significantly enhance patient prognosis. Using virtual screening technology, we examined our in-house library and identified pinosylvin as a potential compound with high activity. Pinosylvin exhibited robust hydrogen bond and Π-Π interaction with Nrf2. Cell experiments revealed that pinosylvin effectively reduced the proliferation of U87 tumor cells by regulating Nrf2 and demonstrated greater inhibitory activity than temozolomide. Consequently, we believe that this study will offer valuable guidance for the future development of highly efficient therapeutic drugs for GBM.
Collapse
Affiliation(s)
- Huihui Yang
- School of Health and Life Sciences, University of Health and Rehabilitation Sciences, Qingdao 266001, China
| | - Peilu Song
- School of Health and Life Sciences, University of Health and Rehabilitation Sciences, Qingdao 266001, China
| | - Baohu Li
- School of Health and Life Sciences, University of Health and Rehabilitation Sciences, Qingdao 266001, China; School of Pharmaceutical Sciences, Shandong University, Jinan 250012, China
| | - Shutang Li
- School of Health and Life Sciences, University of Health and Rehabilitation Sciences, Qingdao 266001, China
| | - Jinfei Yang
- School of Health and Life Sciences, University of Health and Rehabilitation Sciences, Qingdao 266001, China; School of Pharmaceutical Sciences, Shandong University, Jinan 250012, China.
| |
Collapse
|
24
|
Doron O, Patel AB, Hawryluk GWJ. Neurovascular Interventions for Neurotrauma: From Treatment of Injured Vessels to Treatment of the Injured Brain? Oper Neurosurg (Hagerstown) 2024; 26:247-255. [PMID: 37976141 DOI: 10.1227/ons.0000000000000980] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2023] [Accepted: 09/17/2023] [Indexed: 11/19/2023] Open
Abstract
Traumatic brain injury is often associated with a direct or secondary neurovascular pathology. In this review, we present recent advancements in endovascular neurosurgery that enable accurate and effective vessel reconstruction with emphasis on its role in early diagnosis, the expanding use of flow diversion in pseudoaneurysms, and traumatic arteriovenous fistulas. In addition, future directions in which catheter-based interventions could potentially affect traumatic brain injury are described: targeting blood brain barrier integrity using the advantages of intra-arterial drug delivery of blood brain barrier stabilizers to prevent secondary brain edema, exploring the impact of endovascular venous access as a means to modulate venous outflow in an attempt to reduce intracranial pressure and augment brain perfusion, applying selective intra-arterial hypothermia as a neuroprotection method mitigating some of the risks conferred by systemic cooling, trans-vessel wall delivery of regenerative therapy agents, and shifting attention using multimodal neuromonitoring to post-traumatic vasospasm to further characterize the role it plays in secondary brain injury. Thus, we believe that the potential of endovascular tools can be expanded because they enable access to the "highways" governing perfusion and flow and call for further research focused on exploring these routes because it may contribute to novel endovascular approaches currently used for treating injured vessels, harnessing them for treatment of the injured brain.
Collapse
Affiliation(s)
- Omer Doron
- Department of Neurosurgery, Massachusetts General Hospital, Harvard Medical School, Boston , Massachusetts , USA
- Department of Biomedical Engineering, The Aldar and Iby Fleischman Faculty of Engineering, Tel Aviv University, Tel Aviv , Israel
| | - Aman B Patel
- Department of Neurosurgery, Massachusetts General Hospital, Harvard Medical School, Boston , Massachusetts , USA
| | - Gregory W J Hawryluk
- Department of Neurosurgery, Akron General Neuroscience Institute, Cleveland Clinic, Akron , Ohio , USA
| |
Collapse
|
25
|
Zhang Y, Qi X, Li W, Wan M, Ning X, Hu J. Research on the classification of early-stage brain edema by combining intrinsic optical signal imaging and laser speckle contrast imaging. JOURNAL OF BIOPHOTONICS 2024; 17:e202300394. [PMID: 38169143 DOI: 10.1002/jbio.202300394] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/22/2023] [Revised: 11/24/2023] [Accepted: 12/11/2023] [Indexed: 01/05/2024]
Abstract
The early detection and pathological classification of brain edema are very important for symptomatic treatment. The dual-optical imaging system (DOIS) consists of intrinsic optical signal imaging (IOSI) and laser speckle contrast imaging (LSCI), which can acquire cerebral hemodynamic parameters of mice in real-time, including changes of oxygenated hemoglobin concentration ( Δ C HbO 2 ), deoxyhemoglobin concentration (ΔCHbR) and relative cerebral blood flow (rCBF) within the field of view. The slope sum of Δ C HbO 2 , ΔCHbR and rCBF was proposed to classify vasogenic edema (VE) and cytotoxic edema (CE). The slope sum values in the VE and CE group remain statistically different and the classification results provide higher accuracy of more than 93% for early brain edema detection. In conclusion, the differences of hemodynamic parameters between VE and CE in the early stage were revealed and the method helps in the classification of early brain edema.
Collapse
Affiliation(s)
- Yameng Zhang
- Nanjing University of Aeronautics and Astronautics, Nanjing, China
- Nanjing Institute of Technology, Nanjing, China
| | - Xinping Qi
- Nanjing University of Aeronautics and Astronautics, Nanjing, China
| | - Weitao Li
- Nanjing University of Aeronautics and Astronautics, Nanjing, China
| | - Min Wan
- Nanjing University of Aeronautics and Astronautics, Nanjing, China
| | - Xue Ning
- Nanjing University of Aeronautics and Astronautics, Nanjing, China
| | - Jin Hu
- Department of Neurosurgery, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, China
| |
Collapse
|
26
|
Alfieri L, Montana A, Frisoni P, D'Errico S, Neri M. Application of Aquaporins as Markers in Forensic Pathology: A Systematic Review of the Literature. Int J Mol Sci 2024; 25:2664. [PMID: 38473914 DOI: 10.3390/ijms25052664] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2024] [Revised: 02/21/2024] [Accepted: 02/23/2024] [Indexed: 03/14/2024] Open
Abstract
The study of aquaporins (AQPs) in various forensic fields has offered a promising horizon in response to the need to have reliable elements for the identification of the manner of death and for the individuation of forensic markers for the timing of lesions and vitality of injury. In the literature, various tissues have been studied; the most investigated are the lungs, brain, kidneys, skin, and blood vessels. A systematic literature review on PubMed following PRISMA 2020 guidelines enabled the identification of 96 articles. In all, 34 of these were enrolled to identify Aquaporin-like (AQP-like) forensic markers. The analysis of the literature demonstrated that the most significant markers among the AQPs are as follows: for the brain, AQP4, which is very important in brain trauma and hypoxic damage; AQP3 in the skin lesions caused by various mechanisms; and AQP5 in the diagnosis of drowning. Other applications are in organ damage due to drug abuse and thrombus dating. The focus of this review is to collect all the data present in the literature about the forensic application of AQPs as forensic markers in the most important fields of application. In the current use, the individuation, validation, and application of markers in forensic investigation are very useful in real forensic applications in cases evaluated in court.
Collapse
Affiliation(s)
- Letizia Alfieri
- Department of Medical Sciences, University of Ferrara, 44121 Ferrara, Italy
| | - Angelo Montana
- Department of Biomedical Sciences and Public Health, University Politecnica delle Marche, 60126 Ancona, Italy
| | - Paolo Frisoni
- Unit of Legal Medicine, AUSL Romagna, G.B. Morgagni-L. Pierantoni Hospital, 47100 Forlì, Italy
| | - Stefano D'Errico
- Department of Medical Surgical and Health Sciences, University of Trieste, 34149 Trieste, Italy
| | - Margherita Neri
- Department of Medical Sciences, University of Ferrara, 44121 Ferrara, Italy
| |
Collapse
|
27
|
Wang S, Liu A, Xu C, Hou J, Hong J. GLP-1(7-36) protected against oxidative damage and neuronal apoptosis in the hippocampal CA region after traumatic brain injury by regulating ERK5/CREB. Mol Biol Rep 2024; 51:313. [PMID: 38374452 PMCID: PMC10876747 DOI: 10.1007/s11033-024-09244-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2023] [Accepted: 01/11/2024] [Indexed: 02/21/2024]
Abstract
BACKGROUND Glucagon-like peptide-1 (GLP-1) (7-36) amide, an endogenous active form of GLP-1, has been shown to modulate oxidative stress and neuronal cell survival in various neurological diseases. OBJECTIVE This study investigated the potential effects of GLP-1(7-36) on oxidative stress and apoptosis in neuronal cells following traumatic brain injury (TBI) and explored the underlying mechanisms. METHODS Traumatic brain injury (TBI) models were established in male SD rats for in vivo experiments. The extent of cerebral oedema was assessed using wet-to-dry weight ratios following GLP-1(7-36) intervention. Neurological dysfunction and cognitive impairment were evaluated through behavioural experiments. Histopathological changes in the brain were observed using haematoxylin and eosin staining. Oxidative stress levels in hippocampal tissues were measured. TUNEL staining and Western blotting were employed to examine cell apoptosis. In vitro experiments evaluated the extent of oxidative stress and neural apoptosis following ERK5 phosphorylation activation. Immunofluorescence colocalization of p-ERK5 and NeuN was analysed using immunofluorescence cytochemistry. RESULTS Rats with TBI exhibited neurological deterioration, increased oxidative stress, and enhanced apoptosis, which were ameliorated by GLP-1(7-36) treatment. Notably, GLP-1(7-36) induced ERK5 phosphorylation in TBI rats. However, upon ERK5 inhibition, oxidative stress and neuronal apoptosis levels were elevated, even in the presence of GLP-1(7-36). CONCLUSION In summary, this study suggested that GLP-1(7-36) suppressed oxidative damage and neuronal apoptosis after TBI by activating ERK5/CREB.
Collapse
Affiliation(s)
- Shuwei Wang
- Department of Neurosurgery, Tangshan Gongren Hospital, Tangshan, 063000, Hebei, China
| | - Aijun Liu
- Department of Neurosurgery, Tangshan Gongren Hospital, Tangshan, 063000, Hebei, China
| | - Chaopeng Xu
- Department of Neurosurgery, Tangshan Gongren Hospital, Tangshan, 063000, Hebei, China
| | - Jingxuan Hou
- Department of Neurosurgery, Tangshan Gongren Hospital, Tangshan, 063000, Hebei, China
| | - Jun Hong
- Department of Neurosurgery, Tangshan Gongren Hospital, Tangshan, 063000, Hebei, China.
| |
Collapse
|
28
|
Krzyściak W, Bystrowska B, Karcz P, Chrzan R, Bryll A, Turek A, Mazur P, Śmierciak N, Szwajca M, Donicz P, Furman K, Pilato F, Kozicz T, Popiela T, Pilecki M. Association of Blood Metabolomics Biomarkers with Brain Metabolites and Patient-Reported Outcomes as a New Approach in Individualized Diagnosis of Schizophrenia. Int J Mol Sci 2024; 25:2294. [PMID: 38396971 PMCID: PMC10888632 DOI: 10.3390/ijms25042294] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2024] [Revised: 02/06/2024] [Accepted: 02/10/2024] [Indexed: 02/25/2024] Open
Abstract
Given its polygenic nature, there is a need for a personalized approach to schizophrenia. The aim of the study was to select laboratory biomarkers from blood, brain imaging, and clinical assessment, with an emphasis on patients' self-report questionnaires. Metabolomics studies of serum samples from 51 patients and 45 healthy volunteers, based on the liquid chromatography-electrospray ionization-mass spectrometry (LC-ESI-MS/MS), led to the identification of 3 biochemical indicators (cortisol, glutamate, lactate) of schizophrenia. These metabolites were sequentially correlated with laboratory tests results, imaging results, and clinical assessment outcomes, including patient self-report outcomes. The hierarchical cluster analysis on the principal components (HCPC) was performed to identify the most homogeneous clinical groups. Significant correlations were noted between blood lactates and 11 clinical and 10 neuroimaging parameters. The increase in lactate and cortisol were significantly associated with a decrease in immunological parameters, especially with the level of reactive lymphocytes. The strongest correlations with the level of blood lactate and cortisol were demonstrated by brain glutamate, N-acetylaspartate and the concentrations of glutamate and glutamine, creatine and phosphocreatine in the prefrontal cortex. Metabolomics studies and the search for associations with brain parameters and self-reported outcomes may provide new diagnostic evidence to specific schizophrenia phenotypes.
Collapse
Affiliation(s)
- Wirginia Krzyściak
- Department of Medical Diagnostics, Jagiellonian University Medical College, Faculty of Pharmacy, 30-688 Krakow, Poland;
| | - Beata Bystrowska
- Department of Biochemical Toxicology, Jagiellonian University Medical College, Faculty of Pharmacy, 30-688 Krakow, Poland;
| | - Paulina Karcz
- Department of Electroradiology, Jagiellonian University Medical College, Faculty of Health Sciences, 31-126 Krakow, Poland;
| | - Robert Chrzan
- Department of Radiology, Jagiellonian University Medical College, Faculty of Medicine, 31-503 Krakow, Poland; (R.C.); (A.B.); (T.P.)
| | - Amira Bryll
- Department of Radiology, Jagiellonian University Medical College, Faculty of Medicine, 31-503 Krakow, Poland; (R.C.); (A.B.); (T.P.)
| | - Aleksander Turek
- Department of Child and Adolescent Psychiatry and Psychotherapy, Faculty of Medicine, Jagiellonian University Medical College, 31-501 Krakow, Poland; (A.T.); (N.Ś.); (M.S.); (P.D.); (K.F.); (M.P.)
| | - Paulina Mazur
- Department of Medical Diagnostics, Jagiellonian University Medical College, Faculty of Pharmacy, 30-688 Krakow, Poland;
| | - Natalia Śmierciak
- Department of Child and Adolescent Psychiatry and Psychotherapy, Faculty of Medicine, Jagiellonian University Medical College, 31-501 Krakow, Poland; (A.T.); (N.Ś.); (M.S.); (P.D.); (K.F.); (M.P.)
| | - Marta Szwajca
- Department of Child and Adolescent Psychiatry and Psychotherapy, Faculty of Medicine, Jagiellonian University Medical College, 31-501 Krakow, Poland; (A.T.); (N.Ś.); (M.S.); (P.D.); (K.F.); (M.P.)
| | - Paulina Donicz
- Department of Child and Adolescent Psychiatry and Psychotherapy, Faculty of Medicine, Jagiellonian University Medical College, 31-501 Krakow, Poland; (A.T.); (N.Ś.); (M.S.); (P.D.); (K.F.); (M.P.)
| | - Katarzyna Furman
- Department of Child and Adolescent Psychiatry and Psychotherapy, Faculty of Medicine, Jagiellonian University Medical College, 31-501 Krakow, Poland; (A.T.); (N.Ś.); (M.S.); (P.D.); (K.F.); (M.P.)
| | - Fabio Pilato
- Neurology, Neurophysiology and Neurobiology Unit, Department of Medicine, Università Campus Bio-Medico di Roma, 00128 Rome, Italy;
| | - Tamas Kozicz
- Department of Clinical Genomics, Center for Individualized Medicine, Mayo Clinic, Rochester, MN 55905, USA;
| | - Tadeusz Popiela
- Department of Radiology, Jagiellonian University Medical College, Faculty of Medicine, 31-503 Krakow, Poland; (R.C.); (A.B.); (T.P.)
| | - Maciej Pilecki
- Department of Child and Adolescent Psychiatry and Psychotherapy, Faculty of Medicine, Jagiellonian University Medical College, 31-501 Krakow, Poland; (A.T.); (N.Ś.); (M.S.); (P.D.); (K.F.); (M.P.)
| |
Collapse
|
29
|
Seblani M, Ertlen C, Coyle T, Decherchi P, Brezun JM. Combined effect of trifluoperazine and sodium cromoglycate on reducing acute edema and limiting lasting functional impairments after spinal cord injury in rats. Exp Neurol 2024; 372:114612. [PMID: 37993080 DOI: 10.1016/j.expneurol.2023.114612] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2023] [Revised: 11/10/2023] [Accepted: 11/15/2023] [Indexed: 11/24/2023]
Abstract
Edema formation is one of the very first events to occur after spinal cord injury (SCI) leading to an increase of the intrathecal pressure and consequently to serious spinal tissue and functional impairments. Current edema treatments are still symptomatic and/or non-specific. Since edema formation mechanisms are mainly described as vasogenic and cytotoxic, it becomes crucial to understand the interplay between these two subtypes. Acting on key targets to inhibit edema formation may reduce secondary damage and related functional impairments. In this study, we characterize the edema kinetic after T9-10 spinal contusion. We use trifluoperazine (TFP) to block the expression and the functional subcellular localization of aquaporin-4 supposed to be implicated in the cytotoxic edema formation. We also use sodium cromoglycate (SCG) to deactivate mast cell degranulation known to be implicated in the vasogenic edema formation. Our results show a significant reduction of edema after TFP treatment and after TFP-SCG combined treatment compared to control. This reduction is correlated with limited onset of initial sensorimotor impairments particularly after combined treatment. Our results highlight the importance of potential synergetic targets in early edema therapy after SCI as part of tissue sparing strategies.
Collapse
Affiliation(s)
- Mostafa Seblani
- Aix Marseille Univ, CNRS, ISM, UMR7287, Institut des Sciences du Mouvement: Etienne-Jules MAREY, Team "Plasticité des Systèmes Nerveux et Musculaire" (PSNM), Parc Scientifique et Technologique de Luminy, CC910-163, Avenue de Luminy, F-13288 Marseille, Cedex 09, France
| | - Céline Ertlen
- Aix Marseille Univ, CNRS, ISM, UMR7287, Institut des Sciences du Mouvement: Etienne-Jules MAREY, Team "Plasticité des Systèmes Nerveux et Musculaire" (PSNM), Parc Scientifique et Technologique de Luminy, CC910-163, Avenue de Luminy, F-13288 Marseille, Cedex 09, France
| | - Thelma Coyle
- Aix Marseille Univ, CNRS, ISM, UMR7287, Institut des Sciences du Mouvement: Etienne-Jules MAREY, Team "Plasticité des Systèmes Nerveux et Musculaire" (PSNM), Parc Scientifique et Technologique de Luminy, CC910-163, Avenue de Luminy, F-13288 Marseille, Cedex 09, France
| | - Patrick Decherchi
- Aix Marseille Univ, CNRS, ISM, UMR7287, Institut des Sciences du Mouvement: Etienne-Jules MAREY, Team "Plasticité des Systèmes Nerveux et Musculaire" (PSNM), Parc Scientifique et Technologique de Luminy, CC910-163, Avenue de Luminy, F-13288 Marseille, Cedex 09, France
| | - Jean-Michel Brezun
- Aix Marseille Univ, CNRS, ISM, UMR7287, Institut des Sciences du Mouvement: Etienne-Jules MAREY, Team "Plasticité des Systèmes Nerveux et Musculaire" (PSNM), Parc Scientifique et Technologique de Luminy, CC910-163, Avenue de Luminy, F-13288 Marseille, Cedex 09, France.
| |
Collapse
|
30
|
Suryavanshi P, Langton R, Fairhead K, Glykys J. Brief and diverse excitotoxic insults cause an increase in neuronal nuclear membrane permeability in the neonatal brain. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.08.22.554167. [PMID: 37662276 PMCID: PMC10473591 DOI: 10.1101/2023.08.22.554167] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/05/2023]
Abstract
Neuronal swelling after excitotoxic insults is implicated in neuronal injury and death in the developing brain, yet mitigating brain edema with osmotic and surgical interventions yields poor clinical outcomes. Importantly, neuronal swelling and its downstream consequences during early brain development remain poorly investigated. Using multiphoton Ca2+ imaging in vivo (P12-17) and in acute brain slices (P8-12), we explored Ca2+-dependent downstream effects after neuronal cytotoxic edema. We observed the translocation of cytosolic GCaMP6s into the nucleus of a subpopulation of neurons minutes after various excitotoxic insults. We used automated morphology-detection algorithms for neuronal segmentation and quantified the nuclear translocation of GCaMP6s as the ratio of nuclear and cytosolic intensity (N/C ratio). Elevated neuronal N/C ratios were correlated to higher Ca2+ loads and could occur independently of neuronal swelling. Electron microscopy revealed that the nuclear translocation was associated with increased nuclear pore size. Inhibiting calpains prevented elevated N/C ratios and neuronal swelling. Thus, our results indicate altered nuclear transport in a subpopulation of neurons shortly after injury in the developing brain, which can be used as an early biomarker of acute neuronal injury.
Collapse
Affiliation(s)
- P Suryavanshi
- Department of Pediatrics, University of Iowa, Iowa City, IA
- Iowa Neuroscience Institute, University of Iowa, Iowa City, IA
| | - R Langton
- Department of Pediatrics, University of Iowa, Iowa City, IA
- Iowa Neuroscience Institute, University of Iowa, Iowa City, IA
| | - K Fairhead
- Biomedical Sciences, College of Liberal Arts and Sciences, University of Iowa, Iowa City, IA
| | - J Glykys
- Department of Pediatrics, University of Iowa, Iowa City, IA
- Iowa Neuroscience Institute, University of Iowa, Iowa City, IA
- Department of Neurology, University of Iowa, Iowa City, IA
| |
Collapse
|
31
|
Hasan GM, Anwar S, Shamsi A, Sohal SS, Hassan MI. The neuroprotective potential of phytochemicals in traumatic brain injury: mechanistic insights and pharmacological implications. Front Pharmacol 2024; 14:1330098. [PMID: 38239205 PMCID: PMC10794744 DOI: 10.3389/fphar.2023.1330098] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2023] [Accepted: 12/15/2023] [Indexed: 01/22/2024] Open
Abstract
Traumatic brain injury (TBI) leads to brain damage, comprising both immediate primary damage and a subsequent cascade of secondary injury mechanisms. The primary injury results in localized brain damage, while the secondary damage initiates inflammatory responses, followed by the disruption of the blood-brain barrier, infiltration of peripheral blood cells, brain edema, and the release of various immune mediators, including chemotactic factors and interleukins. TBI disrupts molecular signaling, cell structures, and functions. In addition to physical tissue damage, such as axonal injuries, contusions, and haemorrhages, TBI interferes with brain functioning, impacting cognition, decision-making, memory, attention, and speech capabilities. Despite a deep understanding of the pathophysiology of TBI, an intensive effort to evaluate the underlying mechanisms with effective therapeutic interventions is imperative to manage the repercussions of TBI. Studies have commenced to explore the potential of employing natural compounds as therapeutic interventions for TBI. These compounds are characterized by their low toxicity and limited interactions with conventional drugs. Moreover, many natural compounds demonstrate the capacity to target various aspects of the secondary injury process. While our understanding of the pathophysiology of TBI, there is an urgent need for effective therapeutic interventions to mitigate its consequences. Here, we aimed to summarize the mechanism of action and the role of phytochemicals against TBI progression. This review discusses the therapeutic implications of various phytonutrients and addresses primary and secondary consequences of TBI. In addition, we highlighted the roles of emerging phytochemicals as promising candidates for therapeutic intervention of TBI. The review highlights the neuroprotective roles of phytochemicals against TBI and the mechanistic approach. Furthermore, our efforts focused on the underlying mechanisms, providing a better understanding of the therapeutic potential of phytochemicals in TBI therapeutics.
Collapse
Affiliation(s)
- Gulam Mustafa Hasan
- Department of Basic Medical Science, College of Medicine, Prince Sattam Bin Abdulaziz University, Al-Kharj, Saudi Arabia
| | - Saleha Anwar
- Centre for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia, New Delhi, India
| | - Anas Shamsi
- Centre of Medical and Bio-Allied Health Sciences Research, Ajman University, Ajman, United Arab Emirates
| | - Sukhwinder Singh Sohal
- Respiratory Translational Research Group, Department of Laboratory Medicine, School of Health Sciences, College of Health and Medicine, University of Tasmania, Launceston, TAS, Australia
| | - Md. Imtaiyaz Hassan
- Centre for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia, New Delhi, India
| |
Collapse
|
32
|
Waithe OY, Shaji CA, Childs EW, Tharakan B. Determination of Blood-Brain Barrier Hyperpermeability Using Intravital Microscopy. Methods Mol Biol 2024; 2711:117-127. [PMID: 37776453 DOI: 10.1007/978-1-0716-3429-5_10] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/02/2023]
Abstract
The blood vessels that vascularize the central nervous system (CNS) exhibit unique properties, termed the blood-brain barrier (BBB). The BBB allows these blood vessels to tightly regulate the movement of ions, molecules, and cells between the blood and the brain. The BBB is held together by tight junctions of the neighboring endothelial cells of the barrier, more specifically by tight junction proteins (TJPs) which can take the form of either integral transmembrane proteins or accessory cytoplasmic membrane proteins. BBB permeability can furthermore be affected by various factors, including but not limited to TJP expression, size, shape, charge, and type of extravascular molecules, as well as the nature of the vascular beds. The BBB is essential for the proper maintenance of CNS function, and its structural integrity has been implicated in several disorders and conditions. For instance, it has been shown that in the cases of traumatic brain injury (TBI), TBI-associated edema, and increased intracranial pressure are primarily caused by cases of hyperpermeability seen because of BBB dysfunction. Intravital microscopy is one of the most reliable methods for measuring BBB hyperpermeability in rodent models of BBB dysfunction in vivo. Here, we describe the surgical and imaging methods to determine the changes in BBB permeability at the level of the pial microvasculature in a mouse model of TBI using intravital microscopy.
Collapse
Affiliation(s)
- O'lisa Yaa Waithe
- Department of Surgery, Morehouse School of Medicine, Atlanta, GA, USA
| | | | - Ed W Childs
- Department of Surgery, Morehouse School of Medicine, Atlanta, GA, USA
| | - Binu Tharakan
- Department of Surgery, Morehouse School of Medicine, Atlanta, GA, USA.
| |
Collapse
|
33
|
Sunny A, James RR, Menon SR, Rayaroth S, Daniel A, Thompson NA, Tharakan B. Matrix Metalloproteinase-9 inhibitors as therapeutic drugs for traumatic brain injury. Neurochem Int 2024; 172:105642. [PMID: 38008261 DOI: 10.1016/j.neuint.2023.105642] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2023] [Revised: 11/04/2023] [Accepted: 11/09/2023] [Indexed: 11/28/2023]
Abstract
Traumatic brain injury (TBI) is one of the leading causes of morbidity and mortality among young adults and the elderly. In the United States, TBI is responsible for around 30 percent of all injuries brought on by injuries in general. Vasogenic cerebral edema due to blood-brain barrier (BBB) dysfunction and the associated elevation of intracranial pressure (ICP) are some of the major causes of secondary injuries following traumatic brain injury. Matrix metalloproteinase-9 (MMP-9) is a therapeutic target for being an enzyme that degrades the proteins that make up a part of the microvascular basal lamina as well as inter-endothelial tight junctions of the blood-brain barrier. MMP-9-mediated BBB dysfunctions and the compromise of the BBB is a major pathway that leads the development of vasogenic cerebral edema, elevation of ICP, poor cerebral perfusion and brain herniation following traumatic brain injury. That makes MMP-9 an effective therapeutic target and endogenous or exogenous MMP-9 inhibitors as therapeutic drugs for preventing secondary brain damage after traumatic brain injury. Although our understanding of the mechanisms that underlie the primary and secondary stages of damage following a TBI has significantly improved in recent years, such information has not yet resulted in the successful development of novel pharmacological treatment options for traumatic brain injury. Recent pre-clinical and/or clinical studies have demonstrated that there are several compounds with specific or non-specific MMP-9 inhibitory properties either directly binding and inhibiting MMP-9 or by indirectly inhibiting MMP-9, with potential as therapeutic agents for traumatic brain injury. This article reviews the efficacy of several such medications and potential agents that include endogenous and exogeneous compounds that are at various levels of research and development. MMP-9-based therapeutic drug development has enormous potential in the pharmacological treatment of cerebral edema and/or neuronal injury resulting from traumatic brain injury.
Collapse
Affiliation(s)
- Angel Sunny
- Icahn School of Medicine at Mount Sinai, Elmhurst, NY, USA
| | | | | | | | - Abhijith Daniel
- Pushpagiri Institute of Medical Sciences and Research Centre, Thiruvalla, India
| | - Namita Ann Thompson
- Pushpagiri Institute of Medical Sciences and Research Centre, Thiruvalla, India
| | - Binu Tharakan
- Department of Surgery, Morehouse School of Medicine, Atlanta, GA, USA.
| |
Collapse
|
34
|
Waithe OY, Peng X, Childs EW, Tharakan B. Measurement of Blood-Brain Barrier Hyperpermeability Using Evans Blue Extravasation Assay. Methods Mol Biol 2024; 2711:177-184. [PMID: 37776457 DOI: 10.1007/978-1-0716-3429-5_14] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/02/2023]
Abstract
Blood-brain barrier (BBB) dysfunction and hyperpermeability have been implicated in a myriad of brain pathologies. The Evans Blue assay is one of the most popular methods for studying BBB integrity and permeability in rodent models of brain disorders. Under normal physiological conditions, the BBB is impermeable to albumin, so Evans Blue when injected intravenously binds to serum albumin and remains restricted within blood vessels. In traumatic and ischemic injuries, and other brain pathologies that result in BBB hyperpermeability, neighboring endothelial cells partially lose their close contacts to each other, and the BBB becomes permeable to proteins such as albumin. This paracellular leak of Evans blue-bound albumin is considered a reliable indicator of BBB dysfunction and hyperpermeability. Here, we describe the procedures for the evaluation of BBB integrity and hyperpermeability using Evans Blue extravasation assay in a mouse model of traumatic brain injury. The method described here focuses on intravenous injection of Evans Blue followed by Evans Blue dye extraction. This is followed by the measurement of fluorescence intensity of Evans Blue to determine the dye extravasation as a direct indicator of BBB hyperpermeability.
Collapse
Affiliation(s)
- O'lisa Yaa Waithe
- Department of Surgery, Morehouse School of Medicine, Atlanta, GA, USA
| | - Xu Peng
- Department of Medical Physiology, Texas A&M University College of Medicine, Bryan, TX, USA
| | - Ed W Childs
- Department of Surgery, Morehouse School of Medicine, Atlanta, GA, USA
| | - Binu Tharakan
- Department of Surgery, Morehouse School of Medicine, Atlanta, GA, USA.
| |
Collapse
|
35
|
Patil R, Patil AS, Chougule K, Gaude Y, Masareddy RS. Intranasal administration of innovative triamcinolone acetonide encapsulated cubosomal in situ gel: formulation and characterization. Drug Dev Ind Pharm 2024; 50:68-77. [PMID: 38148515 DOI: 10.1080/03639045.2023.2297275] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2023] [Accepted: 12/15/2023] [Indexed: 12/28/2023]
Abstract
AIM The primary objective of the research was to develop a cubosomal in situ gel encapsulated with Triamcinolone acetonide (TCA) in order to enhance its penetration through the blood-brain barrier (BBB) when administered via the intranasal route, thus enabling efficient and rapid action. METHOD Cubosomes were formulated by top-down approach using glyceryl monooleate (GMO), using pluronics127 (PF127) and polyvinyl alcohol (PVA) in varying proportions based on the Box-Behnken design. High resolution transmission electron microscopy (HR-TEM) analysis confirmed the morphology of the cubosomes. The in situ gel was formulated and optimized. Experiments involving ex vivo permeation and histopathology analyses were undertaken to evaluate drug permeation and tissue effects. RESULTS The cubosomes exhibited a particle size (PS) of 197.9 nm, zeta potential (ZP) of -31.11 mV, and entrapment efficacy (EE) of 84.31%, with low deviation. Batch F4 (19% PF127) showed favorable results. In vitro and ex vivo permeation studies revealed drug release of 78.59% and 76.65%, respectively, after 8 h. Drug release followed the Hixson Crowell model of release kinetics. The histopathological examination revealed no signs of toxicity or adverse effects on the nasal mucosa of the sheep. The formulation exhibited short-term stability, maintaining its integrity and properties when stored at room temperature. CONCLUSION The utilization of an intranasal cubosomal in situ gel encapsulated with TCA was anticipated to lower intracranial pressure and improve patient adherence by offering effective relief for individuals suffering from Brain edema. This efficacy is attributed to its rapid onset of action and its safe and well-tolerated dosage form.
Collapse
Affiliation(s)
- Ruturaj Patil
- Department of Pharmaceutics, KLE College of Pharmacy, KLE Academy of Higher Education and Research, Belagavi, Karnataka, India
| | - Archana S Patil
- Department of Pharmaceutics, KLE College of Pharmacy, KLE Academy of Higher Education and Research, Belagavi, Karnataka, India
| | - Krutuja Chougule
- Department of Pharmaceutics, KLE College of Pharmacy, KLE Academy of Higher Education and Research, Belagavi, Karnataka, India
| | - Yadishma Gaude
- Department of Pharmaceutics, KLE College of Pharmacy, KLE Academy of Higher Education and Research, Belagavi, Karnataka, India
| | - Rajashree S Masareddy
- Department of Pharmaceutics, KLE College of Pharmacy, KLE Academy of Higher Education and Research, Belagavi, Karnataka, India
| |
Collapse
|
36
|
Michinaga S. Drug Discovery Research for Traumatic Brain Injury Focused on Functional Molecules in Astrocytes. Biol Pharm Bull 2024; 47:350-360. [PMID: 38296549 DOI: 10.1248/bpb.b23-00731] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/15/2024]
Abstract
Traumatic brain injury (TBI) is severe damage to the head caused by traffic accidents, falls, and sports. Because TBI-induced disruption of the blood-brain barrier (BBB) causes brain edema and neuroinflammation, which are major causes of death or serious disabilities, protection and recovery of BBB function may be beneficial therapeutic strategies for TBI. Astrocytes are key components of BBB integrity, and astrocyte-derived bioactive factors promote and suppress BBB disruption in TBI. Therefore, the regulation of astrocyte function is essential for BBB protection. In the injured cerebrum of TBI model mice, we found that the endothelin ETB receptor, histamine H2 receptor, and transient receptor potential vanilloid 4 (TRPV4) were predominantly expressed in reactive astrocytes. We also showed that repeated administration of an ETB receptor antagonist, H2 receptor agonist, and TRPV4 antagonist alleviated BBB disruption and brain edema in a TBI mouse model. Furthermore, these drugs decreased the expression levels of astrocyte-derived factors promoting BBB disruption and increased the expression levels of astrocyte-derived protective factors in the injured cerebrum after TBI. These results suggest that the ETB receptor, H2 receptor, and TRPV4 are molecules that regulate astrocyte function, and might be attractive candidates for the development of therapeutic drugs for TBI.
Collapse
|
37
|
Yang HC, Lavadi RS, Sauerbeck AD, Wallendorf M, Kummer TT, Song SK, Lin TH. Diffusion basis spectrum imaging detects subclinical traumatic optic neuropathy in a closed-head impact mouse model of traumatic brain injury. Front Neurol 2023; 14:1269817. [PMID: 38152638 PMCID: PMC10752006 DOI: 10.3389/fneur.2023.1269817] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2023] [Accepted: 10/12/2023] [Indexed: 12/29/2023] Open
Abstract
Introduction Traumatic optic neuropathy (TON) is the optic nerve injury secondary to brain trauma leading to visual impairment and vision loss. Current clinical visual function assessments often fail to detect TON due to slow disease progression and clinically silent lesions resulting in potentially delayed or missed treatment in patients with traumatic brain injury (TBI). Methods Diffusion basis spectrum imaging (DBSI) is a novel imaging modality that can potentially fill this diagnostic gap. Twenty-two, 16-week-old, male mice were equally divided into a sham or TBI (induced by moderate Closed-Head Impact Model of Engineered Rotational Acceleration device) group. Briefly, mice were anesthetized with isoflurane (5% for 2.5 min followed by 2.5% maintenance during injury induction), had a helmet placed over the head, and were placed in a holder prior to a 2.1-joule impact. Serial visual acuity (VA) assessments, using the Virtual Optometry System, and DBSI scans were performed in both groups of mice. Immunohistochemistry (IHC) and histological analysis of optic nerves was also performed after in vivo MRI. Results VA of the TBI mice showed unilateral or bilateral impairment. DBSI of the optic nerves exhibited bilateral involvement. IHC results of the optic nerves revealed axonal loss, myelin injury, axonal injury, and increased cellularity in the optic nerves of the TBI mice. Increased DBSI axon volume, decreased DBSI λ||, and elevated DBSI restricted fraction correlated with decreased SMI-312, decreased SMI-31, and increased DAPI density, respectively, suggesting that DBSI can detect coexisting pathologies in the optic nerves of TBI mice. Conclusion DBSI provides an imaging modality capable of detecting subclinical changes of indirect TON in TBI mice.
Collapse
Affiliation(s)
- Hsin-Chieh Yang
- Department of Radiology, Washington University School of Medicine, St. Louis, MO, United States
| | - Raj Swaroop Lavadi
- Department of Radiology, Washington University School of Medicine, St. Louis, MO, United States
| | - Andrew D. Sauerbeck
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, United States
- Hope Center for Neurological Disorders, Washington University School of Medicine, St. Louis, MO, United States
| | - Michael Wallendorf
- Department of Biostatistics, Washington University School of Medicine, St. Louis, MO, United States
| | - Terrance T. Kummer
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, United States
- Hope Center for Neurological Disorders, Washington University School of Medicine, St. Louis, MO, United States
- VA Medical Center, St. Louis, MO, United States
| | - Sheng-Kwei Song
- Department of Radiology, Washington University School of Medicine, St. Louis, MO, United States
- Hope Center for Neurological Disorders, Washington University School of Medicine, St. Louis, MO, United States
| | - Tsen-Hsuan Lin
- Department of Radiology, Washington University School of Medicine, St. Louis, MO, United States
| |
Collapse
|
38
|
Szczygielski J, Hubertus V, Kruchten E, Müller A, Albrecht LF, Schwerdtfeger K, Oertel J. Prolonged course of brain edema and neurological recovery in a translational model of decompressive craniectomy after closed head injury in mice. Front Neurol 2023; 14:1308683. [PMID: 38053795 PMCID: PMC10694459 DOI: 10.3389/fneur.2023.1308683] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2023] [Accepted: 11/01/2023] [Indexed: 12/07/2023] Open
Abstract
Background The use of decompressive craniectomy in traumatic brain injury (TBI) remains a matter of debate. According to the DECRA trial, craniectomy may have a negative impact on functional outcome, while the RescueICP trial revealed a positive effect of surgical decompression, which is evolving over time. This ambivalence of craniectomy has not been studied extensively in controlled laboratory experiments. Objective The goal of the current study was to investigate the prolonged effects of decompressive craniectomy (both positive and negative) in an animal model. Methods Male mice were assigned to the following groups: sham, decompressive craniectomy, TBI and TBI followed by craniectomy. The analysis of functional outcome was performed at time points 3d, 7d, 14d and 28d post trauma according to the Neurological Severity Score and Beam Balance Score. At the same time points, magnetic resonance imaging was performed, and brain edema was analyzed. Results Animals subjected to both trauma and craniectomy presented the exacerbation of the neurological impairment that was apparent mostly in the early course (up to 7d) after injury. Decompressive craniectomy also caused a significant increase in brain edema volume (initially cytotoxic with a secondary shift to vasogenic edema and gliosis). Notably, delayed edema plus gliosis appeared also after decompression even without preceding trauma. Conclusion In prolonged outcomes, craniectomy applied after closed head injury in mice aggravates posttraumatic brain edema, leading to additional functional impairment. This effect is, however, transient. Treatment options that reduce brain swelling after decompression may accelerate neurological recovery and should be explored in future experiments.
Collapse
Affiliation(s)
- Jacek Szczygielski
- Department of Neurosurgery, Saarland University Medical Center and Saarland University Faculty of Medicine, Homburg, Germany
- Instutute of Neuropathology, Saarland University Medical Center and Saarland University Faculty of Medicine, Homburg, Germany
- Institute of Medical Sciences, University of Rzeszów, Rzeszow, Poland
| | - Vanessa Hubertus
- Department of Neurosurgery, Saarland University Medical Center and Saarland University Faculty of Medicine, Homburg, Germany
- Department of Neurosurgery, Charité University Medicine, Berlin, Germany
- Berlin Institute of Health at Charité, Berlin, Germany
| | - Eduard Kruchten
- Department of Neurosurgery, Saarland University Medical Center and Saarland University Faculty of Medicine, Homburg, Germany
- Institute of Interventional and Diagnostic Radiology, Karlsruhe, Germany
| | - Andreas Müller
- Department of Radiology, Saarland University Medical Center and Saarland University Faculty of Medicine, Homburg, Germany
| | - Lisa Franziska Albrecht
- Department of Neurosurgery, Saarland University Medical Center and Saarland University Faculty of Medicine, Homburg, Germany
| | - Karsten Schwerdtfeger
- Department of Neurosurgery, Saarland University Medical Center and Saarland University Faculty of Medicine, Homburg, Germany
| | - Joachim Oertel
- Department of Neurosurgery, Saarland University Medical Center and Saarland University Faculty of Medicine, Homburg, Germany
| |
Collapse
|
39
|
Villapol S, Janatpour ZC, Affram KO, Symes AJ. The Renin Angiotensin System as a Therapeutic Target in Traumatic Brain Injury. Neurotherapeutics 2023; 20:1565-1591. [PMID: 37759139 PMCID: PMC10684482 DOI: 10.1007/s13311-023-01435-8] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/31/2023] [Indexed: 09/29/2023] Open
Abstract
Traumatic brain injury (TBI) is a major public health problem, with limited pharmacological options available beyond symptomatic relief. The renin angiotensin system (RAS) is primarily known as a systemic endocrine regulatory system, with major roles controlling blood pressure and fluid homeostasis. Drugs that target the RAS are used to treat hypertension, heart failure and kidney disorders. They have now been used chronically by millions of people and have a favorable safety profile. In addition to the systemic RAS, it is now appreciated that many different organ systems, including the brain, have their own local RAS. The major ligand of the classic RAS, Angiotensin II (Ang II) acts predominantly through the Ang II Type 1 receptor (AT1R), leading to vasoconstriction, inflammation, and heightened oxidative stress. These processes can exacerbate brain injuries. Ang II receptor blockers (ARBs) are AT1R antagonists. They have been shown in several preclinical studies to enhance recovery from TBI in rodents through improvements in molecular, cellular and behavioral correlates of injury. ARBs are now under consideration for clinical trials in TBI. Several different RAS peptides that signal through receptors distinct from the AT1R, are also potential therapeutic targets for TBI. The counter regulatory RAS pathway has actions that oppose those stimulated by AT1R signaling. This alternative pathway has many beneficial effects on cells in the central nervous system, bringing about vasodilation, and having anti-inflammatory and anti-oxidative stress actions. Stimulation of this pathway also has potential therapeutic value for the treatment of TBI. This comprehensive review will provide an overview of the various components of the RAS, with a focus on their direct relevance to TBI pathology. It will explore different therapeutic agents that modulate this system and assess their potential efficacy in treating TBI patients.
Collapse
Affiliation(s)
- Sonia Villapol
- Department of Neurosurgery, Houston Methodist Hospital, Houston, TX, USA
| | - Zachary C Janatpour
- Department of Pharmacology and Molecular Therapeutics, Uniformed Services University, 4301 Jones Bridge Road, Bethesda, MD, 20814, USA
| | - Kwame O Affram
- Department of Pharmacology and Molecular Therapeutics, Uniformed Services University, 4301 Jones Bridge Road, Bethesda, MD, 20814, USA
| | - Aviva J Symes
- Department of Pharmacology and Molecular Therapeutics, Uniformed Services University, 4301 Jones Bridge Road, Bethesda, MD, 20814, USA.
| |
Collapse
|
40
|
Wang E, Kim S, Wang A, Jiang W, Shah A. Peritoneal dialysis in the setting of acute brain injury: an underappreciated modality. Hosp Pract (1995) 2023; 51:175-183. [PMID: 37491156 DOI: 10.1080/21548331.2023.2241340] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2023] [Revised: 07/12/2023] [Accepted: 07/24/2023] [Indexed: 07/27/2023]
Abstract
INTRODUCTION Dialysis is complicated in the setting of acute brain injury (ABI) due to several factors including acute solute shifts, acid base changes, need for anticoagulation, and changes in intracranial pressure. For these reasons, continuous renal replacement therapy (CRRT) is often the chosen modality for renal replacement needs in these patients. Peritoneal dialysis (PD) is less discussed but shares many of the benefits often attributed to CRRT. We describe, from both nephrology and neurosurgical perspectives, a case successfully managed with PD. CASE A 25-year-old male with history of end-stage kidney disease (ESKD) secondary to focal segmental glomerulosclerosis on continuous cycling PD for 5 years presented to the hospital with headache and altered mental status. Initial imaging revealed a large intraventricular hemorrhage extending to the fourth ventricle. He underwent an emergent right depressive hemicraniectomy and clot evacuation. Post-operative imaging revealed worsening cerebral edema, intraventricular hemorrhage, and hydrocephalus. The decision was made to continue PD, noting that it retains many of the benefits of CRRT (which it is in fact, a form of) which he tolerated well until the need for a percutaneous gastrostomy tube arose. He was transiently transitioned to hemodialysis but returned to PD once his gastrostomy healed. He continued PD for 1 year without complication and eventually received a kidney transplant. DISCUSSION In managing patients with ABI undergoing dialysis, a number of considerations must be undertaken including avoidance of hypotension to maintain cerebral perfusion pressure and minimize ischemia reperfusion injury, avoidance of anticoagulants that can precipitate or worsen bleeding, the potential for cerebral edema due to rapid solute clearance and osmotic dissipation of therapeutic hypernatremia, and the mitigation of intracellular acidosis from bicarbonate delivery. Although underutilized, PD may potentially serve as a viable option for dialysis in the setting of ABI as demonstrated by the case presented.
Collapse
Affiliation(s)
- Elaina Wang
- Department of Neurosurgery, Warren Alpert Medical School, Brown University, Providence, RI, USA
| | - Steven Kim
- Department of Medicine, Warren Alpert Medical School, Brown University, Providence, RI, USA
| | - Aaron Wang
- Warren Alpert Medical School, Brown University, Providence, RI, USA
| | - Winston Jiang
- Warren Alpert Medical School, Brown University, Providence, RI, USA
| | - Ankur Shah
- Warren Alpert Medical School, Brown University, Providence, RI, USA
- Division of Kidney Disease and Hypertension, Rhode Island Hospital, Providence, RI, USA
| |
Collapse
|
41
|
Balzi APDCC, Otsuki DA, Andrade L, Paiva W, Souza FL, Aureliano LGC, Malbouisson LMS. Can a Therapeutic Strategy for Hypotension Improve Cerebral Perfusion and Oxygenation in an Experimental Model of Hemorrhagic Shock and Severe Traumatic Brain Injury? Neurocrit Care 2023; 39:320-330. [PMID: 37535176 DOI: 10.1007/s12028-023-01802-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2023] [Accepted: 07/03/2023] [Indexed: 08/04/2023]
Abstract
BACKGROUND Restoration of brain tissue perfusion is a determining factor in the neurological evolution of patients with traumatic brain injury (TBI) and hemorrhagic shock (HS). In a porcine model of HS without neurological damage, it was observed that the use of fluids or vasoactive drugs was effective in restoring brain perfusion; however, only terlipressin promoted restoration of cerebral oxygenation and lower expression of edema and apoptosis markers. It is unclear whether the use of vasopressor drugs is effective and beneficial during situations of TBI. The objective of this study is to compare the effects of resuscitation with saline solution and terlipressin on cerebral perfusion and oxygenation in a model of TBI and HS. METHODS Thirty-two pigs weighing 20-30 kg were randomly allocated into four groups: control (no treatment), saline (60 ml/kg of 0.9% NaCl), terlipressin (2 mg of terlipressin), and saline plus terlipressin (20 ml/kg of 0.9% NaCl + 2 mg of terlipressin). Brain injury was induced by lateral fluid percussion, and HS was induced through pressure-controlled bleeding, aiming at a mean arterial pressure (MAP) of 40 mmHg. After 30 min of circulatory shock, resuscitation strategies were initiated according to the group. The systemic and cerebral hemodynamic and oxygenation parameters, lactate levels, and hemoglobin levels were evaluated. The data were subjected to analysis of variance for repeated measures. The significance level established for statistical analysis was p < 0.05. RESULTS The terlipressin and saline plus terlipressin groups showed an increase in MAP that lasted until the end of the experiment (p < 0.05). There was a notable increase in intracranial pressure in all groups after starting treatment for shock. Cerebral perfusion pressure and cerebral oximetry showed no improvement after hemodynamic recovery in any group. The groups that received saline at resuscitation had the lowest hemoglobin concentrations after treatment. CONCLUSIONS The treatment of hypotension in HS with saline and/or terlipressin cannot restore cerebral perfusion or oxygenation in experimental models of HS and severe TBI. Elevated MAP raises intracranial pressure owing to brain autoregulation dysfunction caused by TBI.
Collapse
Affiliation(s)
- Ana Paula de Carvalho Canela Balzi
- Anesthesiology Department, Hospital das Clinicas SP, School of Medicine, University of São Paulo, Av. Dr. Enéas de Carvalho Aguiar, 255, Cerqueira César, São Paulo, SP, 05403-000, Brazil.
- Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil.
- Divisão de Anestesia do ICHC, UTI Cirúrgica Pediátrica, Av. Enéas Carvalho de Aguiar, 255 - 8° Andar, Cerqueira César, São Paulo, SP, 05403-900, Brazil.
| | - Denise Aya Otsuki
- Medical Research Laboratory -LIM-08, Anesthesiology Department, School of Medicine, University of São Paulo, São Paulo, Brazil
- Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil
| | - Lucia Andrade
- Nephrology Department, Hospital das Clinicas SP, School of Medicine, University of São Paulo, São Paulo, Brazil
- Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil
| | - Wellingson Paiva
- Neurosurgery Department, Hospital das Clinicas SP, School of Medicine, University of São Paulo, São Paulo, Brazil
- Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil
| | - Felipe Lima Souza
- Medical Research Laboratory, Nephrology Department, School of Medicine, University of São Paulo, São Paulo, Brazil
- Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil
| | - Luiz Guilherme Cernaglia Aureliano
- Pathology Department, Hospital das Clinicas SP, School of Medicine, University of São Paulo, São Paulo, Brazil
- Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil
| | - Luiz Marcelo Sá Malbouisson
- Anesthesiology Department, Hospital das Clinicas SP, School of Medicine, University of São Paulo, Av. Dr. Enéas de Carvalho Aguiar, 255, Cerqueira César, São Paulo, SP, 05403-000, Brazil
- Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil
| |
Collapse
|
42
|
Buzoianu AD, Sharma A, Muresanu DF, Feng L, Huang H, Chen L, Tian ZR, Nozari A, Lafuente JV, Sjöqvist PO, Wiklund L, Sharma HS. Nanodelivery of histamine H3 receptor inverse agonist BF-2649 with H3 receptor antagonist and H4 receptor agonist clobenpropit induced neuroprotection is potentiated by antioxidant compound H-290/51 in spinal cord injury. INTERNATIONAL REVIEW OF NEUROBIOLOGY 2023; 172:37-77. [PMID: 37833018 DOI: 10.1016/bs.irn.2023.06.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/15/2023]
Abstract
Military personnel are often victims of spinal cord injury resulting in lifetime disability and decrease in quality of life. However, no suitable therapeutic measures are still available to restore functional disability or arresting the pathophysiological progression of disease in victims for leading a better quality of life. Thus, further research in spinal cord injury using novel strategies or combination of available neuroprotective drugs is urgently needed for superior neuroprotection. In this regard, our laboratory is engaged in developing TiO2 nanowired delivery of drugs, antibodies and enzymes in combination to attenuate spinal cord injury induced pathophysiology and functional disability in experimental rodent model. Previous observations show that histamine antagonists or antioxidant compounds when given alone in spinal cord injury are able to induce neuroprotection for short periods after trauma. In this investigation we used a combination of histaminergic drugs with antioxidant compound H-290/51 using their nanowired delivery for neuroprotection in spinal cord injury of longer duration. Our observations show that a combination of H3 receptor inverse agonist BF-2549 with H3 receptor antagonist and H4 receptor agonist clobenpropit induced neuroprotection is potentiated by antioxidant compound H-290/51 in spinal cord injury. These observations suggests that histamine receptors are involved in the pathophysiology of spinal cord injury and induce superior neuroprotection in combination with an inhibitor of lipid peroxidation H-290/51, not reported earlier. The possible mechanisms and significance of our findings in relation to future clinical approaches in spinal cord injury is discussed.
Collapse
Affiliation(s)
- Anca D Buzoianu
- Department of Clinical Pharmacology and Toxicology, "Iuliu Hatieganu" University of Medicine and Pharmacy, Cluj-Napoca, Romania
| | - Aruna Sharma
- International Experimental Central Nervous System Injury & Repair (IECNSIR), Dept. of Surgical Sciences, Anesthesiology & Intensive Care Medicine, Uppsala University Hospital, Uppsala University, Uppsala, Sweden.
| | - Dafin F Muresanu
- Dept. Clinical Neurosciences, University of Medicine & Pharmacy, Cluj-Napoca, Romania; ''RoNeuro'' Institute for Neurological Research and Diagnostic, Mircea Eliade Street, Cluj-Napoca, Romania
| | - Lianyuan Feng
- Department of Neurology, Bethune International Peace Hospital, Zhongshan Road (West), Shijiazhuang, Hebei Province, P.R. China
| | - Hongyun Huang
- Beijing Hongtianji Neuroscience Academy, Beijing, P.R. China
| | - Lin Chen
- Department of Neurosurgery, Dongzhimen Hospital, Beijing University of Traditional Chinese Medicine, Beijing, P.R. China
| | - Z Ryan Tian
- Dept. Chemistry & Biochemistry, University of Arkansas, Fayetteville, AR, United States
| | - Ala Nozari
- Department of Anesthesiology, Boston University, Albany str, Boston MA, United States
| | - José Vicente Lafuente
- LaNCE, Dept. Neuroscience, University of the Basque Country (UPV/EHU), Leioa, Bizkaia, Spain
| | - Per-Ove Sjöqvist
- Division of Cardiology, Department of Medicine, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - Lars Wiklund
- International Experimental Central Nervous System Injury & Repair (IECNSIR), Dept. of Surgical Sciences, Anesthesiology & Intensive Care Medicine, Uppsala University Hospital, Uppsala University, Uppsala, Sweden
| | - Hari Shanker Sharma
- International Experimental Central Nervous System Injury & Repair (IECNSIR), Dept. of Surgical Sciences, Anesthesiology & Intensive Care Medicine, Uppsala University Hospital, Uppsala University, Uppsala, Sweden; LaNCE, Dept. Neuroscience, University of the Basque Country (UPV/EHU), Leioa, Bizkaia, Spain.
| |
Collapse
|
43
|
Saei AA, Gharibi H, Lyu H, Nilsson B, Jafari M, Von Holst H, Zubarev RA. Massive Solubility Changes in Neuronal Proteins upon Simulated Traumatic Brain Injury Reveal the Role of Shockwaves in Irreversible Damage. Molecules 2023; 28:6768. [PMID: 37836614 PMCID: PMC10574794 DOI: 10.3390/molecules28196768] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2023] [Revised: 09/13/2023] [Accepted: 09/20/2023] [Indexed: 10/15/2023] Open
Abstract
We investigated the immediate molecular consequences of traumatic brain injuries (TBIs) using a novel proteomics approach. We simulated TBIs using an innovative laboratory apparatus that employed a 5.1 kg dummy head that held neuronal cells and generated a ≤4000 g-force acceleration upon impact. A Proteome Integral Solubility Alteration (PISA) assay was then employed to monitor protein solubility changes in a system-wide manner. Dynamic impacts led to both a reduction in neuron viability and massive solubility changes in the proteome. The affected proteins mapped not only to the expected pathways, such as those of cell adhesion, collagen, and laminin structures, as well as the response to stress, but also to other dense protein networks, such as immune response, complement, and coagulation cascades. The cellular effects were found to be mainly due to the shockwave rather than the g-force acceleration. Soft materials could reduce the impact's severity only until they were fully compressed. This study shows a way of developing a proteome-based meter for measuring irreversible shockwave-induced cell damage and provides a resource for identifying protein biomarkers of TBIs and potential drug targets for the development of products aimed at primary prevention and intervention.
Collapse
Affiliation(s)
- Amir Ata Saei
- Division of Physiological Chemistry I, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, 171 65 Stockholm, Sweden; (A.A.S.); (H.G.); (H.L.); (B.N.); (M.J.)
- Department of Cell Biology, Harvard Medical School, Boston, MA 02115, USA
| | - Hassan Gharibi
- Division of Physiological Chemistry I, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, 171 65 Stockholm, Sweden; (A.A.S.); (H.G.); (H.L.); (B.N.); (M.J.)
| | - Hezheng Lyu
- Division of Physiological Chemistry I, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, 171 65 Stockholm, Sweden; (A.A.S.); (H.G.); (H.L.); (B.N.); (M.J.)
| | - Brady Nilsson
- Division of Physiological Chemistry I, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, 171 65 Stockholm, Sweden; (A.A.S.); (H.G.); (H.L.); (B.N.); (M.J.)
| | - Maryam Jafari
- Division of Physiological Chemistry I, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, 171 65 Stockholm, Sweden; (A.A.S.); (H.G.); (H.L.); (B.N.); (M.J.)
| | - Hans Von Holst
- Division of Physiological Chemistry I, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, 171 65 Stockholm, Sweden; (A.A.S.); (H.G.); (H.L.); (B.N.); (M.J.)
- Division of Clinical Neuroscience, Section of Neurosurgery, Karolinska Institutet, 171 65 Stockholm, Sweden
| | - Roman A. Zubarev
- Division of Physiological Chemistry I, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, 171 65 Stockholm, Sweden; (A.A.S.); (H.G.); (H.L.); (B.N.); (M.J.)
- Department of Pharmacological & Technological Chemistry, Sechenov First Moscow State Medical University, 119146 Moscow, Russia
- The National Medical Research Center for Endocrinology, 115478 Moscow, Russia
| |
Collapse
|
44
|
Björk S, Hånell A, Ronne-Engström E, Stenwall A, Velle F, Lewén A, Enblad P, Svedung Wettervik T. Thiopental and decompressive craniectomy as last-tier ICP-treatments in aneurysmal subarachnoid hemorrhage: is functional recovery within reach? Neurosurg Rev 2023; 46:231. [PMID: 37676578 PMCID: PMC10485091 DOI: 10.1007/s10143-023-02138-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2023] [Revised: 08/17/2023] [Accepted: 09/01/2023] [Indexed: 09/08/2023]
Abstract
The study aimed to investigate the indication and functional outcome after barbiturates and decompressive craniectomy (DC) as last-tier treatments for elevated intracranial pressure (ICP) in aneurysmal subarachnoid hemorrhage (aSAH). This observational study included 891 aSAH patients treated at a single center between 2008 and 2018. Data on demography, admission status, radiology, ICP, clinical course, and outcome 1-year post-ictus were collected. Patients treated with thiopental (barbiturate) and DC were the main target group.Thirty-nine patients (4%) were treated with thiopental alone and 52 (6%) with DC. These patients were younger and had a worse neurological status than those who did not require these treatments. Before thiopental, the median midline shift was 0 mm, whereas basal cisterns were compressed/obliterated in 66%. The median percentage of monitoring time with ICP > 20 mmHg immediately before treatment was 38%, which did not improve after 6 h of infusion. Before DC, the median midline shift was 10 mm, and the median percentage of monitoring time with ICP > 20 mmHg before DC was 56%, which both significantly improved postoperatively. At follow-up, 52% of the patients not given thiopental or operated with DC reached favorable outcome, whereas this occurred in 10% of the thiopental and DC patients.In summary, 10% of the aSAH cohort required thiopental, DC, or both. Thiopental and DC are important integrated last-tier treatment options, but careful patient selection is needed due to the risk of saving many patients into a state of suffering.
Collapse
Affiliation(s)
- Sofie Björk
- Department of Medical Sciences, Section of Neurosurgery, Uppsala University, 751 85, Uppsala, Sweden
| | - Anders Hånell
- Department of Medical Sciences, Section of Neurosurgery, Uppsala University, 751 85, Uppsala, Sweden
| | - Elisabeth Ronne-Engström
- Department of Medical Sciences, Section of Neurosurgery, Uppsala University, 751 85, Uppsala, Sweden
| | - Anton Stenwall
- Department of Medical Sciences, Section of Neurosurgery, Uppsala University, 751 85, Uppsala, Sweden
| | - Fartein Velle
- Department of Medical Sciences, Section of Neurosurgery, Uppsala University, 751 85, Uppsala, Sweden
| | - Anders Lewén
- Department of Medical Sciences, Section of Neurosurgery, Uppsala University, 751 85, Uppsala, Sweden
| | - Per Enblad
- Department of Medical Sciences, Section of Neurosurgery, Uppsala University, 751 85, Uppsala, Sweden
| | - Teodor Svedung Wettervik
- Department of Medical Sciences, Section of Neurosurgery, Uppsala University, 751 85, Uppsala, Sweden.
| |
Collapse
|
45
|
Walter J, Mende J, Hutagalung S, Alhalabi OT, Grutza M, Zheng G, Skutella T, Unterberg A, Zweckberger K, Younsi A. The Single-Dose Application of Interleukin-4 Ameliorates Secondary Brain Damage in the Early Phase after Moderate Experimental Traumatic Brain Injury in Mice. Int J Mol Sci 2023; 24:12756. [PMID: 37628939 PMCID: PMC10454634 DOI: 10.3390/ijms241612756] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2023] [Revised: 08/03/2023] [Accepted: 08/09/2023] [Indexed: 08/27/2023] Open
Abstract
Activation of the interleukin-4 (IL-4) pathway ameliorates secondary injury mechanisms after experimental traumatic brain injury (TBI); therefore, we assessed the effect of a therapeutic IL-4 administration on secondary brain damage after experimental TBI. We subjected 100 C57/Bl6 wildtype mice to controlled cortical impact (CCI) and administered IL-4 or a placebo control subcutaneously 15 min thereafter. Contusion volume (Nissl staining), neurological function (hole board, video open field, and CatWalkXT®), and the immune response (immunofluorescent staining) were analyzed up to 28 days post injury (dpi). Contusion volumes were significantly reduced after IL-4 treatment up to 14 dpi (e.g., 6.47 ± 0.41 mm3 vs. 3.80 ± 0.85 mm3, p = 0.011 3 dpi). Macrophage invasion and microglial response were significantly attenuated in the IL-4 group in the acute phase after CCI (e.g., 1.79 ± 0.15 Iba-1+/CD86+ cells/sROI vs. 1.06 ± 0.21 Iba-1/CD86+ cells/sROI, p = 0.030 in the penumbra 3 dpi), whereas we observed an increased neuroinflammation thereafter (e.g., mean GFAP intensity of 3296.04 ± 354.21 U vs. 6408.65 ± 999.54 U, p = 0.026 in the ipsilateral hippocampus 7 dpi). In terms of functional outcome, several gait parameters were improved in the acute phase following IL-4 treatment (e.g., a difference in max intensity of -7.58 ± 2.00 U vs. -2.71 ± 2.44 U, p = 0.041 3 dpi). In conclusion, the early single-dose administration of IL-4 significantly reduces secondary brain damage in the acute phase after experimental TBI in mice, which seems to be mediated by attenuation of macrophage and microglial invasion.
Collapse
Affiliation(s)
- Johannes Walter
- Department of Neurosurgery, Heidelberg University Hospital, Im Neuenheimer Feld 400, 69120 Heidelberg, Germany; (J.M.); (S.H.); (O.T.A.); (M.G.); (G.Z.); (A.U.); (K.Z.)
| | - Jannis Mende
- Department of Neurosurgery, Heidelberg University Hospital, Im Neuenheimer Feld 400, 69120 Heidelberg, Germany; (J.M.); (S.H.); (O.T.A.); (M.G.); (G.Z.); (A.U.); (K.Z.)
| | - Samuel Hutagalung
- Department of Neurosurgery, Heidelberg University Hospital, Im Neuenheimer Feld 400, 69120 Heidelberg, Germany; (J.M.); (S.H.); (O.T.A.); (M.G.); (G.Z.); (A.U.); (K.Z.)
| | - Obada T. Alhalabi
- Department of Neurosurgery, Heidelberg University Hospital, Im Neuenheimer Feld 400, 69120 Heidelberg, Germany; (J.M.); (S.H.); (O.T.A.); (M.G.); (G.Z.); (A.U.); (K.Z.)
| | - Martin Grutza
- Department of Neurosurgery, Heidelberg University Hospital, Im Neuenheimer Feld 400, 69120 Heidelberg, Germany; (J.M.); (S.H.); (O.T.A.); (M.G.); (G.Z.); (A.U.); (K.Z.)
| | - Guoli Zheng
- Department of Neurosurgery, Heidelberg University Hospital, Im Neuenheimer Feld 400, 69120 Heidelberg, Germany; (J.M.); (S.H.); (O.T.A.); (M.G.); (G.Z.); (A.U.); (K.Z.)
| | - Thomas Skutella
- Institute for Anatomy and Cell Biology, Heidelberg University, Im Neuenheimer Feld 307, 69120 Heidelberg, Germany;
| | - Andreas Unterberg
- Department of Neurosurgery, Heidelberg University Hospital, Im Neuenheimer Feld 400, 69120 Heidelberg, Germany; (J.M.); (S.H.); (O.T.A.); (M.G.); (G.Z.); (A.U.); (K.Z.)
| | - Klaus Zweckberger
- Department of Neurosurgery, Heidelberg University Hospital, Im Neuenheimer Feld 400, 69120 Heidelberg, Germany; (J.M.); (S.H.); (O.T.A.); (M.G.); (G.Z.); (A.U.); (K.Z.)
| | - Alexander Younsi
- Department of Neurosurgery, Heidelberg University Hospital, Im Neuenheimer Feld 400, 69120 Heidelberg, Germany; (J.M.); (S.H.); (O.T.A.); (M.G.); (G.Z.); (A.U.); (K.Z.)
| |
Collapse
|
46
|
Zhang C, Zhu J, Zhang M, Yuan Z, Wang X, Ye C, Jiang H, Ye X. Prognostic nomogram for predicting lower extremity deep venous thrombosis in ruptured intracranial aneurysm patients who underwent endovascular treatment. Front Neurol 2023; 14:1202076. [PMID: 37609653 PMCID: PMC10440693 DOI: 10.3389/fneur.2023.1202076] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2023] [Accepted: 06/30/2023] [Indexed: 08/24/2023] Open
Abstract
Background Lower extremity deep vein thrombosis (DVT) is one of the major postoperative complications in patients with ruptured intracranial aneurysms (RIA) who underwent endovascular treatment (EVT). However, patient-specific predictive models are still lacking. This study aimed to construct and validate a nomogram model for estimating the risk of lower extremity DVT for RIA patients who underwent EVT. Methods This cohort study enrolled 471 RIA patients who received EVT in our institution between 1 January 2020 to 4 February 2022. Perioperative information on participants is collected to develop and validate a nomogram for predicting lower extremity DVT in RIA patients after EVT. Predictive accuracy, discriminatory capability, and clinical effectiveness were evaluated by concordance index (C-index), calibration curves, and decision curve analysis. Result Multivariate logistic regression analysis showed that age, albumin, D-dimer, GCS score, middle cerebral artery aneurysm, and delayed cerebral ischemia were independent predictors for lower extremity DVT. The nomogram for assessing individual risk of lower extremity DVT indicated good predictive accuracy in the primary cohort (c-index, 0.92) and the validation cohort (c-index, 0.85), with a wide threshold probability range (4-82%) and superior net benefit. Conclusion The present study provided a reliable and convenient nomogram model developed with six optimal predictors to assess postoperative lower extremity DVT in RIA patients, which may benefit to strengthen the awareness of lower extremity DVT control and supply appropriate resources to forecast patients at high risk of RIA-related lower extremity DVT.
Collapse
Affiliation(s)
- Chengwei Zhang
- Department of Neurosurgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
- Wenzhou Medical University, Wenzhou, China
| | - Jiaqian Zhu
- Department of Neurosurgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
- Wenzhou Medical University, Wenzhou, China
| | - Minghong Zhang
- Department of Neurosurgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
- Wenzhou Medical University, Wenzhou, China
| | - Ziru Yuan
- Department of Neurosurgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
- Wenzhou Medical University, Wenzhou, China
| | - Xiaoxiong Wang
- Department of Neurosurgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
- Wenzhou Medical University, Wenzhou, China
| | - Chengxing Ye
- Department of Neurosurgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
- Wenzhou Medical University, Wenzhou, China
| | - Haojie Jiang
- Department of Neurosurgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
- Wenzhou Medical University, Wenzhou, China
| | - Xiong Ye
- Department of Neurosurgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
- Wenzhou Medical University, Wenzhou, China
| |
Collapse
|
47
|
Safwat A, Helmy A, Gupta A. The Role of Substance P Within Traumatic Brain Injury and Implications for Therapy. J Neurotrauma 2023; 40:1567-1583. [PMID: 37132595 DOI: 10.1089/neu.2022.0510] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/04/2023] Open
Abstract
This review examines the role of the neuropeptide substance P within the neuroinflammation that follows traumatic brain injury. It examines it in reference to its preferential receptor, the neurokinin-1 receptor, and explores the evidence for antagonism of this receptor in traumatic brain injury with therapeutic intent. Expression of substance P increases following traumatic brain injury. Subsequent binding to the neurokinin-1 receptor results in neurogenic inflammation, a cause of deleterious secondary effects that include an increased intracranial pressure and poor clinical outcome. In several animal models of TBI, neurokinin-1 receptor antagonism has been shown to reduce brain edema and the resultant rise in intracranial pressure. A brief overview of the history of substance P is presented, alongside an exploration into the chemistry of the neuropeptide with a relevance to its functions within the central nervous system. This review summarizes the scientific and clinical rationale for substance P antagonism as a promising therapy for human TBI.
Collapse
Affiliation(s)
- Adam Safwat
- Division of Anaesthesia, Department of Medicine, University of Cambridge, Cambridge, United Kingdom
- Department of Clinical Neurosciences, University of Cambridge, Cambridge, United Kingdom
| | - Adel Helmy
- Department of Clinical Neurosciences, University of Cambridge, Cambridge, United Kingdom
| | - Arun Gupta
- Neurosciences Critical Care Unit, Addenbrooke's Hospital, Cambridge, United Kingdom
| |
Collapse
|
48
|
Grasso G, Sallì M, Kim HS, Torregrossa F. Possible Role of the New Identified "Subarachnoid Lymphatic-Like Membrane" in Traumatic Brain Injury. World Neurosurg 2023; 174:1-2. [PMID: 36868406 DOI: 10.1016/j.wneu.2023.02.105] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2023] [Accepted: 02/22/2023] [Indexed: 03/05/2023]
Affiliation(s)
- Giovanni Grasso
- Neurosurgical Unit, Department of Biomedicine, Neurosciences and Advanced Diagnostics, University of Palermo, Palermo, Italy.
| | - Marcello Sallì
- Rehabilitation Medicine Outpatient Department, A.S.P. Palermo, Palermo, Italy
| | - Hyeun-Sung Kim
- Department of Spine Surgery, Nanoori Gangnam Hospital, Seoul, South Korea
| | - Fabio Torregrossa
- Neurosurgical Unit, Department of Biomedicine, Neurosciences and Advanced Diagnostics, University of Palermo, Palermo, Italy
| |
Collapse
|
49
|
Abstract
Traumatic brain injury is one of the most common causes of morbidity and mortality and significantly impacts the patients' quality of life and socioeconomic status. It can be classified into primary and secondary injuries. Primary injury occurs at the time of the initial head trauma, such as skull fracture, extra-axial hemorrhage, brain contusion, and diffuse axonal injury. Secondary injury develops later as complications such as diffuse cerebral edema, brain herniation, and chronic traumatic encephalopathy. This article describes the indication for imaging, imaging modalities, recommended imaging protocols, and imaging findings of primary and secondary injuries, including pitfalls of each pathology.
Collapse
Affiliation(s)
- Aniwat Sriyook
- Department of Radiology, King Chulalongkorn Memorial Hospital, The Thai Red Cross Society, and Faculty of Medicine, Chulalongkorn University, Bangkok 10330, Thailand; Department of Radiology, Massachusetts General Hospital and Harvard Medical School, 55 Fruit Street, Boston, MA 02114, USA.
| | - Rajiv Gupta
- Department of Radiology, Massachusetts General Hospital and Harvard Medical School, 55 Fruit Street, Boston, MA 02114, USA
| |
Collapse
|
50
|
Ziesel D, Nowakowska M, Scheruebel S, Kornmueller K, Schäfer U, Schindl R, Baumgartner C, Üçal M, Rienmüller T. Electrical stimulation methods and protocols for the treatment of traumatic brain injury: a critical review of preclinical research. J Neuroeng Rehabil 2023; 20:51. [PMID: 37098582 PMCID: PMC10131365 DOI: 10.1186/s12984-023-01159-y] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2022] [Accepted: 03/13/2023] [Indexed: 04/27/2023] Open
Abstract
BACKGROUND Traumatic brain injury (TBI) is a leading cause of disabilities resulting from cognitive and neurological deficits, as well as psychological disorders. Only recently, preclinical research on electrical stimulation methods as a potential treatment of TBI sequelae has gained more traction. However, the underlying mechanisms of the anticipated improvements induced by these methods are still not fully understood. It remains unclear in which stage after TBI they are best applied to optimize the therapeutic outcome, preferably with persisting effects. Studies with animal models address these questions and investigate beneficial long- and short-term changes mediated by these novel modalities. METHODS In this review, we present the state-of-the-art in preclinical research on electrical stimulation methods used to treat TBI sequelae. We analyze publications on the most commonly used electrical stimulation methods, namely transcranial magnetic stimulation (TMS), transcranial direct current stimulation (tDCS), deep brain stimulation (DBS) and vagus nerve stimulation (VNS), that aim to treat disabilities caused by TBI. We discuss applied stimulation parameters, such as the amplitude, frequency, and length of stimulation, as well as stimulation time frames, specifically the onset of stimulation, how often stimulation sessions were repeated and the total length of the treatment. These parameters are then analyzed in the context of injury severity, the disability under investigation and the stimulated location, and the resulting therapeutic effects are compared. We provide a comprehensive and critical review and discuss directions for future research. RESULTS AND CONCLUSION: We find that the parameters used in studies on each of these stimulation methods vary widely, making it difficult to draw direct comparisons between stimulation protocols and therapeutic outcome. Persisting beneficial effects and adverse consequences of electrical simulation are rarely investigated, leaving many questions about their suitability for clinical applications. Nevertheless, we conclude that the stimulation methods discussed here show promising results that could be further supported by additional research in this field.
Collapse
Affiliation(s)
- D Ziesel
- Institute of Health Care Engineering with European Testing Center of Medical Devices, Graz University of Technology, Graz, Austria
| | - M Nowakowska
- Research Unit of Experimental Neurotraumatology, Department of Neurosurgery, Medical University of Graz, Graz, Austria
| | - S Scheruebel
- Gottfried Schatz Research Center for Cell Signaling, Metabolism and Aging, Biophysics Division, Medical University of Graz, Graz, Austria
| | - K Kornmueller
- Gottfried Schatz Research Center for Cell Signaling, Metabolism and Aging, Biophysics Division, Medical University of Graz, Graz, Austria
| | - U Schäfer
- Research Unit of Experimental Neurotraumatology, Department of Neurosurgery, Medical University of Graz, Graz, Austria
- BioTechMed-Graz, Graz, Austria
| | - R Schindl
- Gottfried Schatz Research Center for Cell Signaling, Metabolism and Aging, Biophysics Division, Medical University of Graz, Graz, Austria
- BioTechMed-Graz, Graz, Austria
| | - C Baumgartner
- Institute of Health Care Engineering with European Testing Center of Medical Devices, Graz University of Technology, Graz, Austria
- BioTechMed-Graz, Graz, Austria
| | - M Üçal
- Research Unit of Experimental Neurotraumatology, Department of Neurosurgery, Medical University of Graz, Graz, Austria
- BioTechMed-Graz, Graz, Austria
| | - T Rienmüller
- Institute of Health Care Engineering with European Testing Center of Medical Devices, Graz University of Technology, Graz, Austria.
- BioTechMed-Graz, Graz, Austria.
| |
Collapse
|