1
|
Saha P, Skrodzki D, Aditya T, Moitra P, Alafeef M, Dighe K, Molinaro M, Hicks SD, Pan D. Tailored Anti-miR Decorated Covalent Organic Framework Enables Electrochemical Detection of Salivary miRNAs for Mild Traumatic Brain Injury. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2025; 21:e2412107. [PMID: 39961046 PMCID: PMC11983262 DOI: 10.1002/smll.202412107] [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: 12/12/2024] [Indexed: 04/11/2025]
Abstract
MicroRNAs (miRNAs) play pivotal role as biomarkers for various diseases, with salivary miRNAs offering a non-invasive diagnostic tool. For mild traumatic brain injury (mTBI), salivary miRNAs like miR-let7a, miR-21, and miR-30e show promise for early detection of subtle injuries lacking reliable indicators. To advance the detection of mTBI-related salivary miRNAs, this study integrates anti-miRNA and miRNA hybridization-based sensing with the development of a nanoscale covalent-organic framework (COF) platform. COFs, with their highly customizable structures, large surface area, and biocompatibility, serve as a versatile foundation for biosensing applications. Here, post-synthetic modification (PSM) of COFs is introduced for essential covalent conjugation of streptavidin for further immobilization of methylene blue-labeled and biotinylated Anti-miRNAs. Furthermore, the layer-by-layer assembly of conductive polymers enhanced the biosensor's electrical performance, enabling ultrasensitive and multiplexed detection of salivary miRNAs. Validated with samples from mixed martial arts participants and confirmed by polymerase chain reaction (PCR), this COF-based platform demonstrates robust accuracy and reliability. By combining COF functionalization with advanced electrode design, it offers a powerful, non-invasive solution for early mTBI detection and broader biomedical applications.
Collapse
Affiliation(s)
- Pranay Saha
- Department of Nuclear EngineeringThe Pennsylvania State UniversityUniversity ParkPA16802USA
| | - David Skrodzki
- Department of Materials Science and EngineeringThe Pennsylvania State UniversityUniversity ParkPA16802USA
| | - Teresa Aditya
- Department of Nuclear EngineeringThe Pennsylvania State UniversityUniversity ParkPA16802USA
| | - Parikshit Moitra
- Department of Nuclear EngineeringThe Pennsylvania State UniversityUniversity ParkPA16802USA
- Present address:
Department of Chemical SciencesIISER BerhampurBerhampurOdisha760010India
| | - Maha Alafeef
- Department of Nuclear EngineeringThe Pennsylvania State UniversityUniversity ParkPA16802USA
- Biomedical Engineering DepartmentJordan University of Science and TechnologyIrbid22110Jordan
| | - Ketan Dighe
- Department of Biomedical EngineeringThe Pennsylvania State UniversityUniversity ParkPA16802USA
| | - Matthew Molinaro
- Department of Engineering Science and MechanicsThe Pennsylvania State UniversityUniversity ParkPA16802USA
| | - Steven D. Hicks
- Department of PediatricsPenn State Health Children's HospitalHersheyPA17033USA
| | - Dipanjan Pan
- Department of Nuclear EngineeringThe Pennsylvania State UniversityUniversity ParkPA16802USA
- Department of Materials Science and EngineeringThe Pennsylvania State UniversityUniversity ParkPA16802USA
- Huck Institutes of the Life Sciences101 Huck Life Sciences BuildingUniversity ParkPA16802USA
- Center for Infectious Disease DynamicsThe Pennsylvania State UniversityUniversity ParkPA16802USA
| |
Collapse
|
2
|
Xie XH, Chen MM, Xu SX, Mei J, Yang Q, Wang C, Lyu H, Gong Q, Liu Z. Isolating Astrocyte-Derived Extracellular Vesicles From Urine. Int J Nanomedicine 2025; 20:2475-2484. [PMID: 40027875 PMCID: PMC11872092 DOI: 10.2147/ijn.s492381] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2024] [Accepted: 02/03/2025] [Indexed: 03/05/2025] Open
Abstract
Introduction Brain-derived extracellular vesicles (BDEVs) can cross the blood-brain barrier and enter the periphery. Therefore, quantifying and analyzing peripherally circulating BDEVs offer a promising approach to directly obtain a window into central nervous system (CNS) pathobiology in vivo. Rapidly evolving CNS diseases require high-frequency sampling, but daily venipuncture of human subjects is highly invasive and usually unfeasible. Methods To address this challenge, here we present a novel method for isolating astrocyte-derived extracellular vesicles from urine (uADEVs), combining urine concentration, ultracentrifugation to isolate total EVs, and then glutamate-aspartate transporter (GLAST) EV isolation using an anti-GLAST antibody. Results The identity of these GLAST+EVs as uADEVs was confirmed by transmission electron microscopy, nanoparticle tracking analysis, western blotting, and assessment of astrocyte-related neurotrophins. Conclusions Leveraging the convenience and availability of urine samples, the non-invasive uADEV approach provides a novel tool that allows high-frequency sampling to investigate rapidly evolving CNS diseases.
Collapse
Affiliation(s)
- Xin-hui Xie
- Department of Psychiatry, Renmin Hospital of Wuhan University, Wuhan, Hubei, People’s Republic of China
| | - Mian-mian Chen
- Department of Psychiatry, Renmin Hospital of Wuhan University, Wuhan, Hubei, People’s Republic of China
| | - Shu-xian Xu
- Department of Psychiatry, Renmin Hospital of Wuhan University, Wuhan, Hubei, People’s Republic of China
| | - Junhua Mei
- Department of Psychiatry, Renmin Hospital of Wuhan University, Wuhan, Hubei, People’s Republic of China
- Department of Neurology, Wuhan First Hospital, Wuhan, Hubei, People’s Republic of China
| | - Qing Yang
- Department of Neurology, Wuhan First Hospital, Wuhan, Hubei, People’s Republic of China
| | - Chao Wang
- Department of Psychiatry, Renmin Hospital of Wuhan University, Wuhan, Hubei, People’s Republic of China
| | - Honggang Lyu
- Department of Psychiatry, Renmin Hospital of Wuhan University, Wuhan, Hubei, People’s Republic of China
| | - Qian Gong
- Department of Psychiatry, Renmin Hospital of Wuhan University, Wuhan, Hubei, People’s Republic of China
| | - Zhongchun Liu
- Department of Psychiatry, Renmin Hospital of Wuhan University, Wuhan, Hubei, People’s Republic of China
- Taikang Center for Life and Medical Sciences, Wuhan University, Wuhan, People’s Republic of China
| |
Collapse
|
3
|
Godoy DA, Fossi F, Robba C. Neuroworsening in Moderate Traumatic Brain Injury. Neurol Clin 2025; 43:51-63. [PMID: 39547741 DOI: 10.1016/j.ncl.2024.08.002] [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: 11/17/2024]
Abstract
Patients with moderate traumatic brain injury (TBI) are at high risk for developing intracerebral complications and in particular neuroworsening (NW). NW can be unpredictable and may be an important risk factor for poor neurologic outcome and for increased mortality. NW is often a medical and surgical emergency, and it is, therefore, fundamental to identify patients at risk early because they require strict neuromonitoring and repeated neuroimaging. So far, there is no standardized and validated definition of NW. In this review, we aim to discuss the definition, risk factors, and management of patients with moderate TBI at high risk of NW.
Collapse
Affiliation(s)
- Daniel Agustin Godoy
- Neurointensive Care Unit, Sanatorio Pasteur Medical Center, Catamarca, Argentina
| | - Francesca Fossi
- Neurointensive Care Unit, Grande Ospedale Metropolitano Niguarda, Milano, Italy
| | - Chiara Robba
- Department of Surgical Sciences and Integrated Diagnostics, University of Genoa, Italy; Neurological and General Intensive Care, IRCCS Ospedale Policlinico San Martino, Largo Rosanna Benzi, 10, Genova 16100, Italy.
| |
Collapse
|
4
|
Wilson L, Newcombe VFJ, Whitehouse DP, Mondello S, Maas AIR, Menon DK. Association of early blood-based biomarkers and six-month functional outcomes in conventional severity categories of traumatic brain injury: capturing the continuous spectrum of injury. EBioMedicine 2024; 107:105298. [PMID: 39191173 PMCID: PMC11400615 DOI: 10.1016/j.ebiom.2024.105298] [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: 12/27/2023] [Revised: 08/06/2024] [Accepted: 08/07/2024] [Indexed: 08/29/2024] Open
Abstract
BACKGROUND Traumatic brain injury is conventionally categorised as mild, moderate, or severe on the Glasgow Coma Scale (GCS). Recently developed biomarkers can provide more objective and nuanced measures of the extent of brain injury. METHODS Exposure-response relationships were investigated in 2479 patients aged ≥16 enrolled in the Collaborative European NeuroTrauma Effectiveness Research in Traumatic Brain Injury (CENTER-TBI) prospective observational cohort study. Neurofilament protein-light (NFL), ubiquitin carboxy-terminal hydrolase L1 (UCH-L1), and glial fibrillary acidic protein (GFAP) were assayed from serum sampled in the first 24 h; concentrations were divided into quintiles within GCS severity groups. Relationships with the Glasgow Outcome Scale-Extended were examined using modified Poisson regression including age, sex, major extracranial injury, time to sample, and log biomarker concentration as covariates. FINDINGS Within severity groups there were associations between biomarkers and outcomes after adjustment for covariates: GCS 13-15 and negative CT imaging (relative risks [RRs] from 1.28 to 3.72), GCS 13-15 and positive CT (1.21-2.81), GCS 9-12 (1.16-2.02), GCS 3-8 (1.09-1.94). RRs were associated with clinically important differences in expectations of prognosis. In patients with GCS 3 (RRs 1.51-1.80) percentages of unfavourable outcome were 37-51% in the lowest quintiles of biomarker levels and reached 90-94% in the highest quintiles. Similarly, for GCS 15 (RRs 1.83-3.79), the percentages were 2-4% and 19-28% in the lowest and highest biomarker quintiles, respectively. INTERPRETATION Conventional TBI severity classification is inadequate and underestimates heterogeneity of brain injury and associated outcomes. The adoption of circulating biomarkers can add to clinical assessment of injury severity. FUNDING European Union 7th Framework program (EC grant 602150), Hannelore Kohl Stiftung, One Mind, Integra LifeSciences, Neuro-Trauma Sciences, NIHR Rosetrees Trust.
Collapse
Affiliation(s)
- Lindsay Wilson
- Division of Psychology, University of Stirling, Stirling, United Kingdom.
| | - Virginia F J Newcombe
- Division of Anaesthesia and PACE, Department of Medicine, University of Cambridge, Cambridge, United Kingdom
| | - Daniel P Whitehouse
- Division of Anaesthesia and PACE, Department of Medicine, University of Cambridge, Cambridge, United Kingdom
| | - Stefania Mondello
- Department of Biomedical and Dental Sciences and Morphofunctional Imaging, University of Messina, Messina, Italy
| | - Andrew I R Maas
- Department of Neurosurgery, Antwerp University Hospital, Edegem, Belgium; Department of Translational Neuroscience, Faculty of Medicine and Health Science, University of Antwerp, Antwerp, Belgium
| | - David K Menon
- Division of Anaesthesia and PACE, Department of Medicine, University of Cambridge, Cambridge, United Kingdom
| |
Collapse
|
5
|
Kocik VI, Dengler BA, Rizzo JA, Ma Moran M, Willis AM, April MD, Schauer SG. A Narrative Review of Existing and Developing Biomarkers in Acute Traumatic Brain Injury for Potential Military Deployed Use. Mil Med 2024; 189:e1374-e1380. [PMID: 37995274 DOI: 10.1093/milmed/usad433] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2023] [Revised: 08/31/2023] [Indexed: 11/25/2023] Open
Abstract
INTRODUCTION Traumatic brain injury (TBI) is a leading cause of morbidity and mortality in both adult civilian and military populations. Currently, diagnostic and prognostic methods are limited to imaging and clinical findings. Biomarker measurements offer a potential method to assess head injuries and help predict outcomes, which has a potential benefit to the military, particularly in the deployed setting where imaging modalities are limited. We determine how biomarkers such as ubiquitin C-terminal hydrolase-L1 (UCH-L1), glial fibrillary acidic protein (GFAP), S100B, neurofilament light chain (NFL), and tau proteins can offer important information to guide the diagnosis, acute management, and prognosis of TBI, specifically in military personnel. MATERIALS AND METHODS We performed a narrative review of peer-reviewed literature using online databases of Google Scholar and PubMed. We included articles published between 1988 and 2022. RESULTS We screened a total of 73 sources finding a total of 39 original research studies that met inclusion for this review. We found five studies that focused on GFAP, four studies that focused on UCH-L1, eight studies that focused on tau proteins, six studies that focused on NFL, and eight studies that focused on S100B. The remainder of the studies included more than one of the biomarkers of interest. CONCLUSIONS TBI occurs frequently in the military and civilian settings with limited methods to diagnose and prognosticate outcomes. We highlighted several promising biomarkers for these purposes including S100B, UCH-L1, NFL, GFAP, and tau proteins. S100B and UCH-L1 appear to have the strongest data to date, but further research is necessary. The robust data that explain the optimal timing and, more importantly, trending of these biomarker measurements are necessary before widespread application.
Collapse
Affiliation(s)
| | - Bradley A Dengler
- Walter Reed National Military Medical Center, Bethesda, MD, USA
- Center for Neuroscience and Regenerative Medicine, Uniformed Services University of the Health Sciences, Bethesda, MD 20814, USA
| | - Julie A Rizzo
- Brooke Army Medical Center, JBSA Fort Sam Houston, TX, USA
- Department of Military and Emergency Medicine, Uniformed Services University of the Health Sciences, Bethesda, MD 20814, USA
| | | | | | - Michael D April
- Department of Military and Emergency Medicine, Uniformed Services University of the Health Sciences, Bethesda, MD 20814, USA
- 14th Field Hospital, Fort Stewart, GA 31314, USA
| | - Steven G Schauer
- Department of Military and Emergency Medicine, Uniformed Services University of the Health Sciences, Bethesda, MD 20814, USA
- Departments of Anesthesiology and Emergency Medicine, University of Colorado School of Medicine, Aurora, CO 80045, USA
- Center for Combat and Battlefield (COMBAT) Research, University of Colorado School of Medicine, Aurora, CO 80045, USA
| |
Collapse
|
6
|
Silvestro S, Raffaele I, Quartarone A, Mazzon E. Innovative Insights into Traumatic Brain Injuries: Biomarkers and New Pharmacological Targets. Int J Mol Sci 2024; 25:2372. [PMID: 38397046 PMCID: PMC10889179 DOI: 10.3390/ijms25042372] [Citation(s) in RCA: 16] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2024] [Revised: 02/08/2024] [Accepted: 02/15/2024] [Indexed: 02/25/2024] Open
Abstract
A traumatic brain injury (TBI) is a major health issue affecting many people across the world, causing significant morbidity and mortality. TBIs often have long-lasting effects, disrupting daily life and functionality. They cause two types of damage to the brain: primary and secondary. Secondary damage is particularly critical as it involves complex processes unfolding after the initial injury. These processes can lead to cell damage and death in the brain. Understanding how these processes damage the brain is crucial for finding new treatments. This review examines a wide range of literature from 2021 to 2023, focusing on biomarkers and molecular mechanisms in TBIs to pinpoint therapeutic advancements. Baseline levels of biomarkers, including neurofilament light chain (NF-L), ubiquitin carboxy-terminal hydrolase-L1 (UCH-L1), Tau, and glial fibrillary acidic protein (GFAP) in TBI, have demonstrated prognostic value for cognitive outcomes, laying the groundwork for personalized treatment strategies. In terms of pharmacological progress, the most promising approaches currently target neuroinflammation, oxidative stress, and apoptotic mechanisms. Agents that can modulate these pathways offer the potential to reduce a TBI's impact and aid in neurological rehabilitation. Future research is poised to refine these therapeutic approaches, potentially revolutionizing TBI treatment.
Collapse
Affiliation(s)
| | | | | | - Emanuela Mazzon
- IRCCS Centro Neurolesi Bonino Pulejo, Via Provinciale Palermo, SS 113, Contrada Casazza, 98124 Messina, Italy; (S.S.); (I.R.); (A.Q.)
| |
Collapse
|
7
|
Hossain I, Marklund N, Czeiter E, Hutchinson P, Buki A. Blood biomarkers for traumatic brain injury: A narrative review of current evidence. BRAIN & SPINE 2023; 4:102735. [PMID: 38510630 PMCID: PMC10951700 DOI: 10.1016/j.bas.2023.102735] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/14/2023] [Revised: 12/05/2023] [Accepted: 12/11/2023] [Indexed: 03/22/2024]
Abstract
Introduction A blood-based biomarker (BBBM) test could help to better stratify patients with traumatic brain injury (TBI), reduce unnecessary imaging, to detect and treat secondary insults, predict outcomes, and monitor treatment effects and quality of care. Research question What evidence is available for clinical applications of BBBMs in TBI and how to advance this field? Material and methods This narrative review discusses the potential clinical applications of core BBBMs in TBI. A literature search in PubMed, Scopus, and ISI Web of Knowledge focused on articles in English with the words "traumatic brain injury" together with the words "blood biomarkers", "diagnostics", "outcome prediction", "extracranial injury" and "assay method" alone-, or in combination. Results Glial fibrillary acidic protein (GFAP) combined with Ubiquitin C-terminal hydrolase-L1(UCH-L1) has received FDA clearance to aid computed tomography (CT)-detection of brain lesions in mild (m) TBI. Application of S100B led to reduction of head CT scans. GFAP may also predict magnetic resonance imaging (MRI) abnormalities in CT-negative cases of TBI. Further, UCH-L1, S100B, Neurofilament light (NF-L), and total tau showed value for predicting mortality or unfavourable outcome. Nevertheless, biomarkers have less role in outcome prediction in mTBI. S100B could serve as a tool in the multimodality monitoring of patients in the neurointensive care unit. Discussion and conclusion Largescale systematic studies are required to explore the kinetics of BBBMs and their use in multiple clinical groups. Assay development/cross validation should advance the generalizability of those results which implicated GFAP, S100B and NF-L as most promising biomarkers in the diagnostics of TBI.
Collapse
Affiliation(s)
- Iftakher Hossain
- Neurocenter, Department of Neurosurgery, Turku University Hospital, Turku, Finland
- Department of Clinical Neurosciences, Neurosurgery Unit, University of Cambridge, Addenbrooke’s Hospital, Cambridge, United Kingdom
- Department of Neuroscience, Karolinska Institute, Stockholm, Sweden
| | - Niklas Marklund
- Department of Clinical Sciences Lund, Neurosurgery, Lund University, Department of Neurosurgery, Skåne University Hospital, Lund, Sweden
| | - Endre Czeiter
- Department of Neurosurgery, Medical School, Neurotrauma Research Group, Szentagothai Research Centre, And HUN-REN-PTE Clinical Neuroscience MR Research Group, University of Pecs, Pecs, Hungary
| | - Peter Hutchinson
- Department of Clinical Neurosciences, Neurosurgery Unit, University of Cambridge, Addenbrooke’s Hospital, Cambridge, United Kingdom
| | - Andras Buki
- Department of Neurosurgery, University of Örebro, Örebro, Sweden
| |
Collapse
|
8
|
Yuguero O, Bernal M, Farré J, Martinez-Alonso M, Vena A, Purroy F. Clinical complications after a traumatic brain injury and its relation with brain biomarkers. Sci Rep 2023; 13:20057. [PMID: 37973882 PMCID: PMC10654919 DOI: 10.1038/s41598-023-47267-6] [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: 07/02/2023] [Accepted: 11/11/2023] [Indexed: 11/19/2023] Open
Abstract
We aimed to find out which are the most frequent complications for patients who suffer a traumatic brain injury (TBI) and its relation with brain biomarker levels. We conducted a hospital cohort study with patients who attended the Hospital Emergency Department between 1 June 2018 and 31 December 2020. Different variables were collected such as biomarkers levels after 6 h and 12 h of TBI (S100, NSE, UCHL1 and GFAP), clinical and sociodemographic variables, complementary tests, and complications 48 h and 7 days after TBI. Qualitative variables were analysed with Pearson's chi-square test, and quantitative variables with the Mann-Whitney U test. A multivariate logistic regression model for the existence of complications one week after discharge was performed to assess the discriminatory capacity of the clinical variables. A total of 51 controls and 540 patients were included in this study. In the TBI group, the mean age was 83 years, and 53.9% of the patients were male. Complications at seven days were associated with the severity of TBI (p < 0.05) and the number of platelets (p = 0.016). All biomarkers except GFAP showed significant differences in their distribution of values according to gender, with significantly higher values of the three biomarkers for women with respect to men. Patients with complications presented significantly higher S100 values (p < 0.05). The patient's baseline status, the severity of the TBI and the S100 levels can be very important elements in determining whether a patient may develop complications in the few hours after TBI.
Collapse
Affiliation(s)
- Oriol Yuguero
- ERLab, Emergency Medicine Research Group, Institute for Biomedical Research Dr. Pifarré Foundation, IRBLLEIDA, Avda. Rovira Roure 80, 25198, Lleida, Spain.
- Faculty of Medicine, University of Lleida, Avda. Rovira Roure 80, 25198, Lleida, Spain.
| | - Maria Bernal
- Clinical Laboratory, University Hospital Arnau de Vilanova, Avda. Rovira Roure 80, 25198, Lleida, Spain
| | - Joan Farré
- Clinical Laboratory, University Hospital Arnau de Vilanova, Avda. Rovira Roure 80, 25198, Lleida, Spain
| | - Montserrat Martinez-Alonso
- Faculty of Medicine, University of Lleida, Avda. Rovira Roure 80, 25198, Lleida, Spain
- Systems Biology and Statistical Methods for Biomedical Research Group, Institute for Biomedical Research Dr. Pifarré Foundation, IRBLLEIDA, Avda. Rovira Roure 80, 25198, Lleida, Spain
| | - Ana Vena
- ERLab, Emergency Medicine Research Group, Institute for Biomedical Research Dr. Pifarré Foundation, IRBLLEIDA, Avda. Rovira Roure 80, 25198, Lleida, Spain
- Faculty of Medicine, University of Lleida, Avda. Rovira Roure 80, 25198, Lleida, Spain
| | - Francisco Purroy
- Faculty of Medicine, University of Lleida, Avda. Rovira Roure 80, 25198, Lleida, Spain
- Clinical neurosciences group, Institute for Biomedical Research Dr. Pifarré Foundation, IRBLLEIDA, Avda. Rovira Roure 80, 25198, Lleida, Spain
| |
Collapse
|
9
|
Agoston DV, Helmy A. Fluid-Based Protein Biomarkers in Traumatic Brain Injury: The View from the Bedside. Int J Mol Sci 2023; 24:16267. [PMID: 38003454 PMCID: PMC10671762 DOI: 10.3390/ijms242216267] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2023] [Revised: 11/07/2023] [Accepted: 11/09/2023] [Indexed: 11/26/2023] Open
Abstract
There has been an explosion of research into biofluid (blood, cerebrospinal fluid, CSF)-based protein biomarkers in traumatic brain injury (TBI) over the past decade. The availability of very large datasets, such as CENTRE-TBI and TRACK-TBI, allows for correlation of blood- and CSF-based molecular (protein), radiological (structural) and clinical (physiological) marker data to adverse clinical outcomes. The quality of a given biomarker has often been framed in relation to the predictive power on the outcome quantified from the area under the Receiver Operating Characteristic (ROC) curve. However, this does not in itself provide clinical utility but reflects a statistical association in any given population between one or more variables and clinical outcome. It is not currently established how to incorporate and integrate biofluid-based biomarker data into patient management because there is no standardized role for such data in clinical decision making. We review the current status of biomarker research and discuss how we can integrate existing markers into current clinical practice and what additional biomarkers do we need to improve diagnoses and to guide therapy and to assess treatment efficacy. Furthermore, we argue for employing machine learning (ML) capabilities to integrate the protein biomarker data with other established, routinely used clinical diagnostic tools, to provide the clinician with actionable information to guide medical intervention.
Collapse
Affiliation(s)
- Denes V. Agoston
- Department of Anatomy, Physiology and Genetic, School of Medicine, Uniformed Services University, Bethesda, MD 20814, USA
| | - Adel Helmy
- Division of Neurosurgery, Department of Clinical Neurosciences, University of Cambridge, Addenbrooke’s Hospital, Cambridge CB2 0QQ, UK;
| |
Collapse
|
10
|
Khosh-Fetrat M, Kosha F, Ansari-Moghaddam A, Guest PC, Vahedian-Azimi A, Barreto GE, Sahebkar A. Determining the value of early measurement of interleukin-10 in predicting the absence of brain lesions in CT scans of patients with mild traumatic brain injury. J Neurol Sci 2023; 446:120563. [PMID: 36701890 DOI: 10.1016/j.jns.2023.120563] [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: 11/08/2022] [Revised: 01/10/2023] [Accepted: 01/19/2023] [Indexed: 01/22/2023]
Abstract
Blood-based biomarkers were recently proposed as predictors of traumatic brain injury (TBI) outcomes. This would be a critical step forward since the majority of TBI events are mild and structural brain damage in this group may be missed by current brain imaging methods. We sought to determine the performance of early measurement of interleukin-10 (IL-10) to distinguish computed tomography (CT)-positive from negative patients with mild TBI. We designed a single-center prospective observational study, which enrolled consecutive patients classed with mild TBI according to Glasgow Coma Scale [GCS] scores and appearance of at least one clinical symptom. Serum IL-10 levels were measured <3 h post hospital admission. The performance of IL-10 levels in correctly classifying patients was evaluated. IL-10 levels were significantly higher in the group with positive CT scans (p < 0.001). With sensitivity set at 100%, the specificity of IL-10 was only 38.1%. However, the specificities of IL-10 for prediction of negative and positive cases increased to 59% and 49%, respectively, when both parameters were assessed within 90 min of admission. For mild TBI patients between 36 and 66 years, classification performance increased significantly at the 100% sensitivity level with a specificity of 93%. Our results suggest that IL-10 may be an easily accessible clinically useful diagnostic biomarker that can distinguish between mild TBI patients with and without structural brain damage with higher effectiveness when lower times of blood sampling are employed and patients are between 36 and 66 years of age.
Collapse
Affiliation(s)
- Masoum Khosh-Fetrat
- Department of Anesthesiology and Critical Care, Khatamolanbia Hospital, Zahedan University of Medical Sciences, Zahedan, Iran
| | - Fariba Kosha
- Department of Anesthesiology and Critical Care, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | | | - Paul C Guest
- Department of Biochemistry and Tissue Biology, University of Campinas (UNICAMP), Campinas, Brazil
| | - Amir Vahedian-Azimi
- Trauma Research Center, Nursing Faculty, Baqiyatallah University of Medical Sciences, Tehran, Iran.
| | - George E Barreto
- Department of Biological Sciences, University of Limerick, V94 T9PX Limerick, Ireland.
| | - Amirhossein Sahebkar
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran; Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran; Department of Biotechnology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
11
|
Gardner RC, Puccio AM, Korley FK, Wang KKW, Diaz-Arrastia R, Okonkwo DO, Puffer RC, Yuh EL, Yue JK, Sun X, Taylor SR, Mukherjee P, Jain S, Manley GT, the TRACK-TBI Investigators
FeeserVenkata RFergusonAdam RGaudetteEtienneGopinathShankarKeeneC DirkMaddenChristopherMartinAlastairMcCreaMichaelMerchantRandallMukherjeePratikNgwenyaLaura BRobertsonClaudiaTemkinNancyVassarMaryYueJohn KZafonteRoss, Ferguson AR, Gaudette E, Shankar GC, Keene D, Madden C, Martin A, McCrea M, Merchant R, Mukherjee P, Ngwenya LB, Robertson C, Temkin N, Vassar M, Yue JK, Zafonte R, the TRACK-TBI Investigators. Effects of age and time since injury on traumatic brain injury blood biomarkers: a TRACK-TBI study. Brain Commun 2022; 5:fcac316. [PMID: 36642999 PMCID: PMC9832515 DOI: 10.1093/braincomms/fcac316] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2022] [Revised: 09/07/2022] [Accepted: 11/30/2022] [Indexed: 12/03/2022] Open
Abstract
Older adults have the highest incidence of traumatic brain injury globally. Accurate blood-based biomarkers are needed to assist with diagnosis of patients across the spectrum of age and time post-injury. Several reports have suggested lower accuracy for blood-based biomarkers in older adults, and there is a paucity of data beyond day-1 post-injury. Our aims were to investigate age-related differences in diagnostic accuracy and 2-week evolution of four leading candidate blood-based traumatic brain injury biomarkers-plasma glial fibrillary acidic protein, ubiquitin carboxy-terminal hydrolase L1, S100 calcium binding protein B and neuron-specific enolase-among participants in the 18-site prospective cohort study Transforming Research And Clinical Knowledge in Traumatic Brain Injury. Day-1 biomarker data were available for 2602 participants including 2151 patients with traumatic brain injury, 242 orthopedic trauma controls and 209 healthy controls. Participants were stratified into 3 age categories (young: 17-39 years, middle-aged: 40-64 years, older: 65-90 years). We investigated age-stratified biomarker levels and biomarker discriminative abilities across three diagnostic groups: head CT-positive/negative; traumatic brain injury/orthopedic controls; and traumatic brain injury/healthy controls. The difference in day-1 glial fibrillary acidic protein, ubiquitin carboxy-terminal hydrolase L1 and neuron-specific enolase levels across most diagnostic groups was significantly smaller for older versus younger adults, resulting in a narrower range within which a traumatic brain injury diagnosis may be discriminated in older adults. Despite this, day-1 glial fibrillary acidic protein had good to excellent performance across all age-categories for discriminating all three diagnostic groups (area under the curve 0.84-0.96; lower limit of 95% confidence intervals all >0.78). Day-1 S100 calcium-binding protein B and ubiquitin carboxy-terminal hydrolase L1 showed good discrimination of CT-positive versus negative only among adults under age 40 years within 6 hours of injury. Longitudinal blood-based biomarker data were available for 522 hospitalized patients with traumatic brain injury and 24 hospitalized orthopaedic controls. Glial fibrillary acidic protein levels maintained good to excellent discrimination across diagnostic groups until day 3 post-injury irrespective of age, until day 5 post-injury among middle-aged or younger patients and until week 2 post-injury among young patients only. In conclusion, the blood-based glial fibrillary acidic protein assay tested here has good to excellent performance across all age-categories for discriminating key traumatic brain injury diagnostic groups to at least 3 days post-injury in this trauma centre cohort. The addition of a blood-based diagnostic to the evaluation of traumatic brain injury, including geriatric traumatic brain injury, has potential to streamline diagnosis.
Collapse
Affiliation(s)
- Raquel C Gardner
- Correspondence to: Raquel C. Gardner, MD Sheba Medical Center, Derech Sheba 2 Ramat Gan, Israel 52621 E-mail:
| | - Ava M Puccio
- Department of Neurological Surgery, University of Pittsburgh Medical Center, Pittsburgh, PA 15213, USA
| | - Frederick K Korley
- Department of Emergency Medicine, University of Michigan, Ann Arbor, MI 48109, USA
| | - Kevin K W Wang
- Departments of Emergency Medicine, Psychiatry, and Neuroscience, McKnight Brain Institute, University of Florida, Gainesville, FL 32610, USA,Brain Rehabilitation Research Center (BRRC), Malcom Randall VA Medical Center, North Florida/South Georgia Veterans Health System, 1601 SW Archer Rd., 32608, USA
| | - Ramon Diaz-Arrastia
- Department of Neurology, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - David O Okonkwo
- Department of Neurological Surgery, University of Pittsburgh Medical Center, Pittsburgh, PA 15213, USA
| | - Ross C Puffer
- Department of Neurological Surgery, University of Pittsburgh Medical Center, Pittsburgh, PA 15213, USA,Department of Neurological Surgery, Mayo Clinic, Rochester, MN 55901, USA
| | - Esther L Yuh
- Department of Radiology, University of California, San Francisco, San Francisco, CA 94143, USA
| | - John K Yue
- Department of Neurological Surgery, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Xiaoying Sun
- Herbert Wertheim School of Public Health and Human Longevity Science, University of California San Diego, San Diego, CA 92161, USA
| | - Sabrina R Taylor
- Department of Neurological Surgery, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Pratik Mukherjee
- Department of Radiology, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Sonia Jain
- Herbert Wertheim School of Public Health and Human Longevity Science, University of California San Diego, San Diego, CA 92161, USA
| | - Geoffrey T Manley
- Department of Neurological Surgery, University of California, San Francisco, San Francisco, CA 94143, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
12
|
Iverson GL, Minkkinen M, Karr JE, Berghem K, Zetterberg H, Blennow K, Posti JP, Luoto TM. Examining four blood biomarkers for the detection of acute intracranial abnormalities following mild traumatic brain injury in older adults. Front Neurol 2022; 13:960741. [PMID: 36484020 PMCID: PMC9723459 DOI: 10.3389/fneur.2022.960741] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2022] [Accepted: 10/20/2022] [Indexed: 01/25/2023] Open
Abstract
Blood-based biomarkers have been increasingly studied for diagnostic and prognostic purposes in patients with mild traumatic brain injury (MTBI). Biomarker levels in blood have been shown to vary throughout age groups. Our aim was to study four blood biomarkers, glial fibrillary acidic protein (GFAP), ubiquitin C-terminal hydrolase-L1 (UCH-L1), neurofilament light (NF-L), and total tau (t-tau), in older adult patients with MTBI. The study sample was collected in the emergency department in Tampere University Hospital, Finland, between November 2015 and November 2016. All consecutive adult patients with head injury were eligible for inclusion. Serum samples were collected from the enrolled patients, which were frozen and later sent for biomarker analyses. Patients aged 60 years or older with MTBI, head computed tomography (CT) imaging, and available biomarker levels were eligible for this study. A total of 83 patients (mean age = 79.0, SD = 9.58, range = 60-100; 41.0% men) were included in the analysis. GFAP was the only biomarker to show statistically significant differentiation between patients with and without acute head CT abnormalities [U(83) = 280, p < 0.001, r = 0.44; area under the curve (AUC) = 0.79, 95% CI = 0.67-0.91]. The median UCH-L1 values were modestly greater in the abnormal head CT group vs. normal head CT group [U (83) = 492, p = 0.065, r = 0.20; AUC = 0.63, 95% CI = 0.49-0.77]. Older age was associated with biomarker levels in the normal head CT group, with the most prominent age associations being with NF-L (r = 0.56) and GFAP (r = 0.54). The results support the use of GFAP in detecting abnormal head CT findings in older adults with MTBIs. However, small sample sizes run the risk for producing non-replicable findings that may not generalize to the population and do not translate well to clinical use. Further studies should consider the potential effect of age on biomarker levels when establishing clinical cut-off values for detecting head CT abnormalities.
Collapse
Affiliation(s)
- Grant L. Iverson
- Department of Physical Medicine and Rehabilitation, Harvard Medical School, Boston, MA, United States,Department of Physical Medicine and Rehabilitation, Spaulding Rehabilitation Hospital and the Schoen Adams Research Institute at Spaulding Rehabilitation, Charlestown, MA, United States,Home Base, A Red Sox Foundation and Massachusetts General Hospital Program, Boston, MA, United States
| | - Mira Minkkinen
- Faculty of Medicine and Health Technology, Tampere University and Tampere University Hospital, Tampere, Finland
| | - Justin E. Karr
- Department of Psychology, University of Kentucky, Lexington, KY, United States
| | - Ksenia Berghem
- Medical Imaging Centre, Department of Radiology, Tampere University Hospital, Tampere, Finland
| | - Henrik Zetterberg
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden,Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden,UK Dementia Research Institute at University College London, London, United Kingdom,Department of Neurodegenerative Disease, University College London Queen Square Institute of Neurology, London, United Kingdom,Hong Kong Center for Neurodegenerative Diseases, Hong Kong, Hong Kong SAR, China
| | - Kaj Blennow
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden,Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
| | - Jussi P. Posti
- Neurocenter, Department of Neurosurgery, Turku University Hospital and University of Turku, Turku, Finland,Turku Brain Injury Center, Turku University Hospital and University of Turku, Turku, Finland
| | - Teemu M. Luoto
- Department of Neurosurgery, Tampere University Hospital and Tampere University, Tampere, Finland,*Correspondence: Teemu M. Luoto
| |
Collapse
|
13
|
Hicks C, Dhiman A, Barrymore C, Goswami T. Traumatic Brain Injury Biomarkers, Simulations and Kinetics. Bioengineering (Basel) 2022; 9:612. [PMID: 36354523 PMCID: PMC9687153 DOI: 10.3390/bioengineering9110612] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2022] [Revised: 10/02/2022] [Accepted: 10/20/2022] [Indexed: 10/21/2023] Open
Abstract
This paper reviews the predictive capabilities of blood-based biomarkers to quantify traumatic brain injury (TBI). Biomarkers for concussive conditions also known as mild, to moderate and severe TBI identified along with post-traumatic stress disorder (PTSD) and chronic traumatic encephalopathy (CTE) that occur due to repeated blows to the head during one's lifetime. Since the pathways of these biomarkers into the blood are not fully understood whether there is disruption in the blood-brain barrier (BBB) and the time it takes after injury for the expression of the biomarkers to be able to predict the injury effectively, there is a need to understand the protein biomarker structure and other physical properties. The injury events in terms of brain and mechanics are a result of external force with or without the shrapnel, in the wake of a wave result in local tissue damage. Thus, these mechanisms express specific biomarkers kinetics of which reaches half-life within a few hours after injury to few days. Therefore, there is a need to determine the concentration levels that follow injury. Even though current diagnostics linking biomarkers with TBI severity are not fully developed, there is a need to quantify protein structures and their viability after injury. This research was conducted to fully understand the structures of 12 biomarkers by performing molecular dynamics simulations involving atomic movement and energies of forming hydrogen bonds. Molecular dynamics software, NAMD and VMD were used to determine and compare the approximate thermodynamic stabilities of the biomarkers and their bonding energies. Five biomarkers used clinically were S100B, GFAP, UCHL1, NF-L and tau, the kinetics obtained from literature show that the concentration values abruptly change with time after injury. For a given protein length, associated number of hydrogen bonds and bond energy describe a lower bound region where proteins self-dissolve and do not have long enough half-life to be detected in the fluids. However, above this lower bound, involving higher number of bonds and energy, we hypothesize that biomarkers will be viable to disrupt the BBB and stay longer to be modeled for kinetics for diagnosis and therefore may help in the discoveries of new biomarkers.
Collapse
Affiliation(s)
- Celeste Hicks
- Biomedical, Industrial and Human Factors Engineering, Wright State University, 3640 Col. Glen Hwy, Dayton, OH 45435, USA
| | - Akshima Dhiman
- Boonshoft School of Medicine, Wright State University, 3640 Col. Glen Hwy, Dayton, OH 45435, USA
| | - Chauntel Barrymore
- Boonshoft School of Medicine, Wright State University, 3640 Col. Glen Hwy, Dayton, OH 45435, USA
| | - Tarun Goswami
- Biomedical, Industrial and Human Factors Engineering, Wright State University, 3640 Col. Glen Hwy, Dayton, OH 45435, USA
| |
Collapse
|
14
|
Korley FK, Jain S, Sun X, Puccio AM, Yue JK, Gardner RC, Wang KKW, Okonkwo DO, Yuh EL, Mukherjee P, Nelson LD, Taylor SR, Markowitz AJ, Diaz-Arrastia R, Manley GT. Prognostic value of day-of-injury plasma GFAP and UCH-L1 concentrations for predicting functional recovery after traumatic brain injury in patients from the US TRACK-TBI cohort: an observational cohort study. Lancet Neurol 2022; 21:803-813. [PMID: 35963263 PMCID: PMC9462598 DOI: 10.1016/s1474-4422(22)00256-3] [Citation(s) in RCA: 93] [Impact Index Per Article: 31.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2021] [Revised: 04/26/2022] [Accepted: 05/30/2022] [Indexed: 12/21/2022]
Abstract
BACKGROUND The prognostic value of glial fibrillary acidic protein (GFAP) and ubiquitin C-terminal hydrolase L1 (UCH-L1) as day-of-injury predictors of functional outcome after traumatic brain injury is not well understood. GFAP is a protein found in glial cells and UCH-L1 is found in neurons, and these biomarkers have been cleared to aid in decision making regarding whether brain CT should be performed after traumatic brain injury. We aimed to quantify their prognostic accuracy and investigate whether these biomarkers contribute novel prognostic information to existing clinical models. METHODS We enrolled patients from the Transforming Research and Clinical Knowledge in Traumatic Brain Injury (TRACK-TBI) observational cohort study. TRACK-TBI includes patients 17 years and older who are evaluated for TBI at 18 US level 1 trauma centres. All patients receive head CT at evaluation, have adequate visual acuity and hearing preinjury, and are fluent in either English or Spanish. In our analysis, we included participants aged 17-90 years who had day-of-injury plasma samples for measurement of GFAP and UCH-L1 and completed 6-month assessments for outcome due to traumatic brain injury with the Glasgow Outcome Scale-Extended (GOSE-TBI). Biomarkers were analysed as continuous variables and in quintiles. This study is registered with ClinicalTrials.gov, NCT02119182. FINDINGS We enrolled 2552 patients from Feb 26, 2014, to Aug 8, 2018. Of the 1696 participants with brain injury and data available at baseline and at 6 months who were included in the analysis, 120 (7·1%) died (GOSE-TBI=1), 235 (13·9%) had an unfavourable outcome (ie, GOSE-TBI ≤4), 1135 (66·9%) had incomplete recovery (ie, GOSE-TBI <8), and 561 (33·1%) recovered fully (ie, GOSE-TBI=8). The area under the curve (AUC) of GFAP for predicting death at 6 months in all patients was 0·87 (95% CI 0·83-0·91), for unfavourable outcome was 0·86 (0·83-0·89), and for incomplete recovery was 0·62 (0·59-0·64). The corresponding AUCs for UCH-L1 were 0·89 (95% CI 0·86-0·92) for predicting death, 0·86 (0·84-0·89) for unfavourable outcome, and 0·61 (0·59-0·64) for incomplete recovery at 6 months. AUCs were higher for participants with traumatic brain injury and Glasgow Coma Scale (GCS) score of 3-12 than for those with GCS score of 13-15. Among participants with GCS score of 3-12 (n=353), adding GFAP and UCH-L1 (alone or combined) to each of the three International Mission for Prognosis and Analysis of Clinical Trials in traumatic brain injury models significantly increased their AUCs for predicting death (AUC range 0·90-0·94) and unfavourable outcome (AUC range 0·83-0·89). However, among participants with GCS score of 13-15 (n=1297), adding GFAP and UCH-L1 to the UPFRONT study model modestly increased the AUC for predicting incomplete recovery (AUC range 0·69-0·69, p=0·025). INTERPRETATION In addition to their known diagnostic value, day-of-injury GFAP and UCH-L1 plasma concentrations have good to excellent prognostic value for predicting death and unfavourable outcome, but not for predicting incomplete recovery at 6 months. These biomarkers contribute the most prognostic information for participants presenting with a GCS score of 3-12. FUNDING US National Institutes of Health, National Institute of Neurologic Disorders and Stroke, US Department of Defense, One Mind, US Army Medical Research and Development Command.
Collapse
Affiliation(s)
- Frederick K Korley
- Department of Emergency Medicine, University of Michigan, Ann Arbor, MI, USA.
| | - Sonia Jain
- Herbert Wertheim School of Public Health and Human Longevity Science, University of California at San Diego, La Jolla, CA, USA
| | - Xiaoying Sun
- Herbert Wertheim School of Public Health and Human Longevity Science, University of California at San Diego, La Jolla, CA, USA
| | - Ava M Puccio
- Department of Neurological Surgery, University of Pittsburgh Medical Center, Pittsburgh, PA, USA
| | - John K Yue
- Department of Neurological Surgery, University of California at San Francisco, San Francisco, CA, USA
| | - Raquel C Gardner
- Department of Neurology, Memory and Aging Center, University of California at San Francisco, San Francisco, CA, USA; Weill Institute for Neurosciences, University of California at San Francisco, San Francisco, CA, USA
| | - Kevin K W Wang
- Program for Neurotrauma, Neuroproteomics and Biomarkers Research, Department of Emergency Medicine, McKnight Brain Institute, University of Florida, Gainesville, FL, USA
| | - David O Okonkwo
- Department of Neurological Surgery, University of Pittsburgh Medical Center, Pittsburgh, PA, USA
| | - Esther L Yuh
- Department of Radiology, University of California at San Francisco, San Francisco, CA, USA
| | - Pratik Mukherjee
- Department of Radiology, University of California at San Francisco, San Francisco, CA, USA
| | - Lindsay D Nelson
- Department of Neurosurgery and Department of Neurology, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Sabrina R Taylor
- Brain and Spinal Injury Center, University of California at San Francisco, San Francisco, CA, USA
| | - Amy J Markowitz
- Department of Neurological Surgery, University of California at San Francisco, San Francisco, CA, USA
| | - Ramon Diaz-Arrastia
- Department of Neurology, University of Pennsylvania, Philadelphia, PA, USA; Traumatic Brain Injury Clinical Research Center, Penn Presbyterian Medical Center, Philadelphia, PA, USA
| | - Geoffrey T Manley
- Department of Neurological Surgery, University of California at San Francisco, San Francisco, CA, USA
| |
Collapse
|
15
|
Murcko R, Marchi N, Bailey D, Janigro D. Diagnostic biomarker kinetics: how brain-derived biomarkers distribute through the human body, and how this affects their diagnostic significance: the case of S100B. Fluids Barriers CNS 2022; 19:32. [PMID: 35546671 PMCID: PMC9092835 DOI: 10.1186/s12987-022-00329-9] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2022] [Accepted: 04/19/2022] [Indexed: 11/22/2022] Open
Abstract
Blood biomarkers of neurological diseases are often employed to rule out or confirm the presence of significant intracranial or cerebrovascular pathology or for the differential diagnosis of conditions with similar presentations (e.g., hemorrhagic vs. embolic stroke). More widespread utilization of biomarkers related to brain health is hampered by our incomplete understanding of the kinetic properties, release patterns, and excretion of molecules derived from the brain. This is, in particular, true for S100B, an astrocyte-derived protein released across the blood–brain barrier (BBB). We developed an open-source pharmacokinetic computer model that allows investigations of biomarker’s movement across the body, the sources of biomarker’s release, and its elimination. This model was derived from a general in silico model of drug pharmacokinetics adapted for protein biomarkers. We improved the model’s predictive value by adding realistic blood flow values, organ levels of S100B, lymphatic and glymphatic circulation, and glomerular filtration for excretion in urine. Three key variables control biomarker levels in blood or saliva: blood–brain barrier permeability, the S100B partition into peripheral organs, and the cellular levels of S100B in astrocytes. A small contribution to steady-state levels of glymphatic drainage was also observed; this mechanism also contributed to the uptake of organs of circulating S100B. This open-source model can also mimic the kinetic behavior of other markers, such as GFAP or NF-L. Our results show that S100B, after uptake by various organs from the systemic circulation, can be released back into systemic fluids at levels that do not significantly affect the clinical significance of venous blood or salivary levels after an episode of BBB disruption.
Collapse
Affiliation(s)
| | - Nicola Marchi
- Laboratory of Cerebrovascular and Glia Research, Department of Neuroscience, Institute of Functional Genomics (UMR 5203 CNRS - U 1191 INSERM), University of Montpellier, Montpellier, France
| | - Damian Bailey
- Neurovascular Research Laboratory, Faculty of Life Sciences and Education, University of South Wales, Newport, UK
| | - Damir Janigro
- FloTBI Inc., Cleveland, OH, USA. .,Department of Physiology and Biophysics, Case Western Reserve University, Cleveland, OH, USA.
| |
Collapse
|
16
|
Janigro D, Mondello S, Posti JP, Unden J. GFAP and S100B: What You Always Wanted to Know and Never Dared to Ask. Front Neurol 2022; 13:835597. [PMID: 35386417 PMCID: PMC8977512 DOI: 10.3389/fneur.2022.835597] [Citation(s) in RCA: 42] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Accepted: 02/03/2022] [Indexed: 12/19/2022] Open
Abstract
Traumatic brain injury (TBI) is a major global health issue, with outcomes spanning from intracranial bleeding, debilitating sequelae, and invalidity with consequences for individuals, families, and healthcare systems. Early diagnosis of TBI by testing peripheral fluids such as blood or saliva has been the focus of many research efforts, leading to FDA approval for a bench-top assay for blood GFAP and UCH-L1 and a plasma point-of-care test for GFAP. The biomarker S100B has been included in clinical guidelines for mTBI (mTBI) in Europe. Despite these successes, several unresolved issues have been recognized, including the robustness of prior data, the presence of biomarkers in tissues beyond the central nervous system, and the time course of biomarkers in peripheral body fluids. In this review article, we present some of these issues and provide a viewpoint derived from an analysis of existing literature. We focus on two astrocytic proteins, S100B and GFAP, the most commonly employed biomarkers used in mTBI. We also offer recommendations that may translate into a broader acceptance of these clinical tools.
Collapse
Affiliation(s)
- Damir Janigro
- Department of Physiology and Biophysics, Case Western Reserve University, Cleveland, OH, United States.,FloTBI, Cleveland, OH, United States
| | - Stefania Mondello
- Department of Biomedical and Dental Sciences and Morphofunctional Imaging, University of Messina, Messina, Italy
| | - Jussi P Posti
- Department of Neurosurgery, Neurocenter, Turku Brain Injury Center, Turku University Hospital, University of Turku, Turku, Finland
| | - Johan Unden
- Department of Operation and Intensive Care, Hallands Hospital Halmstad, Lund University, Lund, Sweden
| |
Collapse
|
17
|
Posti JP, Tenovuo O. Blood-based biomarkers and traumatic brain injury-A clinical perspective. Acta Neurol Scand 2022; 146:389-399. [PMID: 35383879 DOI: 10.1111/ane.13620] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2022] [Revised: 03/08/2022] [Accepted: 03/27/2022] [Indexed: 12/19/2022]
Abstract
Blood-based biomarkers are promising tools to complement clinical variables and imaging findings in the diagnosis, monitoring and outcome prediction of traumatic brain injury (TBI). Several promising biomarker candidates have been found for various clinical questions, but the translation of TBI biomarkers into clinical applications has been negligible. Measured biomarker levels are influenced by patient-related variables such as age, blood-brain barrier integrity and renal and liver function. It is not yet fully understood how biomarkers enter the bloodstream from the interstitial fluid of the brain. In addition, the diagnostic performance of TBI biomarkers is affected by sampling timing and analytical methods. In this focused review, the clinical aspects of glial fibrillary acidic protein, neurofilament light, S100 calcium-binding protein B, tau and ubiquitin C-terminal hydrolase-L1 are examined. Current findings and clinical caveats are addressed.
Collapse
Affiliation(s)
- Jussi P. Posti
- Neurocenter Department of Neurosurgery and Turku Brain Injury Center Turku University Hospital and University of Turku Turku Finland
| | - Olli Tenovuo
- Neurocenter Turku Brain Injury Center Turku University Hospital and University of Turku Turku Finland
| |
Collapse
|
18
|
Nishimura K, Cordeiro JG, Ahmed AI, Yokobori S, Gajavelli S. Advances in Traumatic Brain Injury Biomarkers. Cureus 2022; 14:e23804. [PMID: 35392277 PMCID: PMC8978594 DOI: 10.7759/cureus.23804] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/04/2022] [Indexed: 11/05/2022] Open
Abstract
Traumatic brain injury (TBI) is increasingly a major cause of disability across the globe. The current methods of diagnosis are inadequate at classifying patients and prognosis. TBI is a diagnostic and therapeutic challenge. There is no Food and Drug Administration (FDA)-approved treatment for TBI yet. It took about 16 years of preclinical research to develop accurate and objective diagnostic measures for TBI. Two brain-specific protein biomarkers, namely, ubiquitin C-terminal hydrolase-L1 and glial fibrillary acidic protein, have been extensively characterized. Recently, the two biomarkers were approved by the FDA as the first blood-based biomarker, Brain Trauma Indicator™ (BTI™), via the Breakthrough Devices Program. This scoping review presents (i) TBI diagnosis challenges, (ii) the process behind the FDA approval of biomarkers, and (iii) known unknowns in TBI biomarker biology. The current lag in TBI incidence and hospitalization can be reduced if digital biomarkers such as hard fall detection are standardized and used as a mechanism to alert paramedics to an unresponsive trauma patient.
Collapse
|
19
|
Dong M, Tang Z, Hicks S, Guan W. Rolling Circle Amplification-Coupled Glass Nanopore Counting of Mild Traumatic Brain Injury-Related Salivary miRNAs. Anal Chem 2022; 94:3865-3871. [PMID: 35192325 DOI: 10.1021/acs.analchem.1c04781] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Mild traumatic brain injury (mTBI) could be underdiagnosed and underreported due to the delayed onset of symptoms and the conventional subjective assessment. Recent studies have suggested that salivary microRNAs (miRNAs) could be reliable biomarkers for objective mTBI diagnosis. In this work, we demonstrated a rolling circle amplification (RCA)-coupled resistive pulse-counting platform for profiling mTBI-related miRNAs, using easy-to-fabricate large glass nanopores (200 nm diameter). The method relies on the linear and specific elongation of the miRNA to a much larger RCA product, which the large glass nanopore can digitally count with a high signal-to-noise ratio. We developed and validated the RCA assay against let-7a, miR-30e, and miR-21. We demonstrated the quantification capability of this large glass nanopore counting platform for purified miRNAs as well as miRNAs in salivary total RNA background. Finally, we quantitatively evaluated the performance of profiling each individual miRNAs in a mixed analyte. Our results showed that the RCA-coupled large glass nanopore counting provides a promising and accessible alternative toward the clinical diagnosis of mTBI using salivary miRNAs.
Collapse
Affiliation(s)
- Ming Dong
- Department of Electrical Engineering, Pennsylvania State University, University Park, Pennsylvania 16802, United States
| | - Zifan Tang
- Department of Electrical Engineering, Pennsylvania State University, University Park, Pennsylvania 16802, United States
| | - Steven Hicks
- Department of Pediatrics, Penn State College of Medicine, Hershey, Pennsylvania 17033, United States
| | - Weihua Guan
- Department of Electrical Engineering, Pennsylvania State University, University Park, Pennsylvania 16802, United States.,Department of Biomedical Engineering, Pennsylvania State University, University Park, Pennsylvania 16802, United States
| |
Collapse
|
20
|
Blood GFAP as an emerging biomarker in brain and spinal cord disorders. Nat Rev Neurol 2022; 18:158-172. [PMID: 35115728 DOI: 10.1038/s41582-021-00616-3] [Citation(s) in RCA: 369] [Impact Index Per Article: 123.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/21/2021] [Indexed: 12/14/2022]
Abstract
Blood-derived biomarkers for brain and spinal cord diseases are urgently needed. The introduction of highly sensitive immunoassays led to a rapid increase in the number of potential blood-derived biomarkers for diagnosis and monitoring of neurological disorders. In 2018, the FDA authorized a blood test for clinical use in the evaluation of mild traumatic brain injury (TBI). The test measures levels of the astrocytic intermediate filament glial fibrillary acidic protein (GFAP) and neuroaxonal marker ubiquitin carboxy-terminal hydrolase L1. In TBI, blood GFAP levels are correlated with clinical severity and extent of intracranial pathology. Evidence also indicates that blood GFAP levels hold the potential to reflect, and might enable prediction of, worsening of disability in individuals with progressive multiple sclerosis. A growing body of evidence suggests that blood GFAP levels can be used to detect even subtle injury to the CNS. Most importantly, the successful completion of the ongoing validation of point-of-care platforms for blood GFAP might ameliorate the decision algorithms for acute neurological diseases, such as TBI and stroke, with important economic implications. In this Review, we provide a systematic overview of the evidence regarding the utility of blood GFAP as a biomarker in neurological diseases. We propose a model for GFAP concentration dynamics in different conditions and discuss the limitations that hamper the widespread use of GFAP in the clinical setting. In our opinion, the clinical use of blood GFAP measurements has the potential to contribute to accelerated diagnosis and improved prognostication, and represents an important step forward in the era of precision medicine.
Collapse
|
21
|
Whitehouse DP, Monteiro M, Czeiter E, Vyvere TV, Valerio F, Ye Z, Amrein K, Kamnitsas K, Xu H, Yang Z, Verheyden J, Das T, Kornaropoulos EN, Steyerberg E, Maas AIR, Wang KKW, Büki A, Glocker B, Menon DK, Newcombe VFJ. Relationship of admission blood proteomic biomarkers levels to lesion type and lesion burden in traumatic brain injury: A CENTER-TBI study. EBioMedicine 2022; 75:103777. [PMID: 34959133 PMCID: PMC8718895 DOI: 10.1016/j.ebiom.2021.103777] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2021] [Revised: 11/12/2021] [Accepted: 12/10/2021] [Indexed: 11/26/2022] Open
Abstract
BACKGROUND We aimed to understand the relationship between serum biomarker concentration and lesion type and volume found on computed tomography (CT) following all severities of TBI. METHODS Concentrations of six serum biomarkers (GFAP, NFL, NSE, S100B, t-tau and UCH-L1) were measured in samples obtained <24 hours post-injury from 2869 patients with all severities of TBI, enrolled in the CENTER-TBI prospective cohort study (NCT02210221). Imaging phenotypes were defined as intraparenchymal haemorrhage (IPH), oedema, subdural haematoma (SDH), extradural haematoma (EDH), traumatic subarachnoid haemorrhage (tSAH), diffuse axonal injury (DAI), and intraventricular haemorrhage (IVH). Multivariable polynomial regression was performed to examine the association between biomarker levels and both distinct lesion types and lesion volumes. Hierarchical clustering was used to explore imaging phenotypes; and principal component analysis and k-means clustering of acute biomarker concentrations to explore patterns of biomarker clustering. FINDINGS 2869 patient were included, 68% (n=1946) male with a median age of 49 years (range 2-96). All severities of TBI (mild, moderate and severe) were included for analysis with majority (n=1946, 68%) having a mild injury (GCS 13-15). Patients with severe diffuse injury (Marshall III/IV) showed significantly higher levels of all measured biomarkers, with the exception of NFL, than patients with focal mass lesions (Marshall grades V/VI). Patients with either DAI+IVH or SDH+IPH+tSAH, had significantly higher biomarker concentrations than patients with EDH. Higher biomarker concentrations were associated with greater volume of IPH (GFAP, S100B, t-tau;adj r2 range:0·48-0·49; p<0·05), oedema (GFAP, NFL, NSE, t-tau, UCH-L1;adj r2 range:0·44-0·44; p<0·01), IVH (S100B;adj r2 range:0.48-0.49; p<0.05), Unsupervised k-means biomarker clustering revealed two clusters explaining 83·9% of variance, with phenotyping characteristics related to clinical injury severity. INTERPRETATION Interpretation: Biomarker concentration within 24 hours of TBI is primarily related to severity of injury and intracranial disease burden, rather than pathoanatomical type of injury. FUNDING CENTER-TBI is funded by the European Union 7th Framework programme (EC grant 602150).
Collapse
Affiliation(s)
- Daniel P Whitehouse
- University Division of Anaesthesia, Department of Medicine, University of Cambridge, UK
| | - Miguel Monteiro
- Biomedical Image Analysis Group, Department of Computing, Imperial College, London, UK
| | - Endre Czeiter
- Department of Neurosurgery, Medical School, University of Pécs, Rét u. 2, H-7623 Pécs, Hungary; Neurotrauma Research Group, Szentágothai Research Centre, University of Pécs, Ifjúság útja 20, H-7624 Pécs, Hungary; MTA-PTE Clinical Neuroscience MR Research Group; Pécs, Hungary
| | - Thijs Vande Vyvere
- Research and Development, Icometrix, Leuven, Belgium; Department of Radiology, Antwerp University Hospital and University of Antwerp, Wilrijkstraat 10, 2650, Edegem, Belgium
| | - Fernanda Valerio
- University Division of Anaesthesia, Department of Medicine, University of Cambridge, UK
| | - Zheng Ye
- University Division of Anaesthesia, Department of Medicine, University of Cambridge, UK
| | - Krisztina Amrein
- Department of Neurosurgery, Medical School, University of Pécs, Rét u. 2, H-7623 Pécs, Hungary; Neurotrauma Research Group, Szentágothai Research Centre, University of Pécs, Ifjúság útja 20, H-7624 Pécs, Hungary
| | | | - Haiyan Xu
- Program for Neurotrauma, Neuroproteomics and Biomarker Research, Departments of Emergency Medicine, Psychiatry and Neuroscience, University of Florida, McKnight Brain Institute, L4-100L 1149 South Newell Drive, Gainesville, FL 32611, USA
| | - Zhihui Yang
- Program for Neurotrauma, Neuroproteomics and Biomarker Research, Departments of Emergency Medicine, Psychiatry and Neuroscience, University of Florida, McKnight Brain Institute, L4-100L 1149 South Newell Drive, Gainesville, FL 32611, USA
| | - Jan Verheyden
- Research and Development, Icometrix, Leuven, Belgium
| | - Tilak Das
- Department of Radiology, Addenbrooke's Hospital, Cambridge, UK
| | | | - Ewout Steyerberg
- Center for Medical Decision Making, Department of Public Health, Erasmus University Medical Center, Dr. Molewaterplein 40, 3015 GD Rotterdam, Netherlands; Department of Biomedical Data Sciences, Leiden University Medical Center, Leiden, Netherlands
| | - Andrew I R Maas
- Department of Neurosurgery, Antwerp University Hospital and University of Antwerp, Wijlrijkstraat 10, 2650 Edegem, Belgium
| | - Kevin K W Wang
- Program for Neurotrauma, Neuroproteomics and Biomarker Research, Departments of Emergency Medicine, Psychiatry and Neuroscience, University of Florida, McKnight Brain Institute, L4-100L 1149 South Newell Drive, Gainesville, FL 32611, USA; Brain Rehabilitation Research Center, Malcom Randall Veterans Affairs Medical Center (VAMC), 1601 SW, Archer Rd. Gainesville FL 32608, USA
| | - András Büki
- Department of Neurosurgery, Medical School, University of Pécs, Rét u. 2, H-7623 Pécs, Hungary; Neurotrauma Research Group, Szentágothai Research Centre, University of Pécs, Ifjúság útja 20, H-7624 Pécs, Hungary
| | - Ben Glocker
- Biomedical Image Analysis Group, Department of Computing, Imperial College, London, UK
| | - David K Menon
- University Division of Anaesthesia, Department of Medicine, University of Cambridge, UK
| | - Virginia F J Newcombe
- University Division of Anaesthesia, Department of Medicine, University of Cambridge, UK.
| |
Collapse
|
22
|
Meshkini A, Ghorbani Haghjo A, Hasanpour Segherlou Z, Nouri-Vaskeh M. S100 Calcium-Binding Protein B and Glial Fibrillary Acidic Protein in Patients with Mild Traumatic Brain Injury. Bull Emerg Trauma 2021; 9:183-187. [PMID: 34692869 PMCID: PMC8525696 DOI: 10.30476/beat.2021.89355.1231] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2020] [Revised: 06/13/2021] [Accepted: 06/19/2021] [Indexed: 11/22/2022] Open
Abstract
Objective: To examine the GFAP and S100B ability in prevention unnecessary brain Computed tomography (CT) scan in mild traumatic brain injury (mTBI) and compare them with the single extremity fracture in orthopedic patients. Methods: In this prospective cohort study, two orthopedics patients’ groups and mTBI patients were studied to assess the biomarkers’ ability in prevention unnecessary brain CT scan at the emergency setting. There were 40 orthopedics’ patients with single extremity fracture and 41 mTBI patients. Brain CT scans were done for all mTBI patients. Results: Brain CT scans showed no intracranial traumatic lesions. The median levels for S100B in the mTBI group was 14.8 (4.4-335.9) ng/L, and in orthopedic patients’ group was 13.3 (5-353.10) ng/L. Statistically significant differences were observed between both groups in S100B levels (p=0.006). The median Glial Fibrillary Acidic Protein (GFAP) levels in the mTBI patients’ group were 600 (400-16300) and in the orthopedic patients’ groups was 60 ng/L (300-14900). Statistically significant differences were observed between groups in GFAP (p=0.041). Conclusion: Our results showed that S100B and GFAP serum levels were significantly higher in patients with mTBI than in patients with a single limb fracture.
Collapse
Affiliation(s)
- Ali Meshkini
- Road Injuries Prevention Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Amir Ghorbani Haghjo
- Biotechnology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | | | - Masoud Nouri-Vaskeh
- Medical Philosophy and History Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
23
|
Hier DB, Obafemi-Ajayi T, Thimgan MS, Olbricht GR, Azizi S, Allen B, Hadi BA, Wunsch DC. Blood biomarkers for mild traumatic brain injury: a selective review of unresolved issues. Biomark Res 2021; 9:70. [PMID: 34530937 PMCID: PMC8447604 DOI: 10.1186/s40364-021-00325-5] [Citation(s) in RCA: 71] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2021] [Accepted: 08/26/2021] [Indexed: 01/03/2023] Open
Abstract
Background The use of blood biomarkers after mild traumatic brain injury (mTBI) has been widely studied. We have identified eight unresolved issues related to the use of five commonly investigated blood biomarkers: neurofilament light chain, ubiquitin carboxy-terminal hydrolase-L1, tau, S100B, and glial acidic fibrillary protein. We conducted a focused literature review of unresolved issues in three areas: mode of entry into and exit from the blood, kinetics of blood biomarkers in the blood, and predictive capacity of the blood biomarkers after mTBI. Findings Although a disruption of the blood brain barrier has been demonstrated in mild and severe traumatic brain injury, biomarkers can enter the blood through pathways that do not require a breach in this barrier. A definitive accounting for the pathways that biomarkers follow from the brain to the blood after mTBI has not been performed. Although preliminary investigations of blood biomarkers kinetics after TBI are available, our current knowledge is incomplete and definitive studies are needed. Optimal sampling times for biomarkers after mTBI have not been established. Kinetic models of blood biomarkers can be informative, but more precise estimates of kinetic parameters are needed. Confounding factors for blood biomarker levels have been identified, but corrections for these factors are not routinely made. Little evidence has emerged to date to suggest that blood biomarker levels correlate with clinical measures of mTBI severity. The significance of elevated biomarker levels thirty or more days following mTBI is uncertain. Blood biomarkers have shown a modest but not definitive ability to distinguish concussed from non-concussed subjects, to detect sub-concussive hits to the head, and to predict recovery from mTBI. Blood biomarkers have performed best at distinguishing CT scan positive from CT scan negative subjects after mTBI.
Collapse
Affiliation(s)
- Daniel B Hier
- Department of Electrical and Computer Engineering, Missouri University of Science and Technology, Rolla, MO 65401, USA.
| | - Tayo Obafemi-Ajayi
- Cooperative Engineering Program, Missouri State University, Springfield, MO 65897, United States
| | - Matthew S Thimgan
- Department of Biological Sciences, Missouri University of Science and Technology, Rolla, MO 65409, United States
| | - Gayla R Olbricht
- Department of Mathematics and Statistics, Missouri University of Science and Technology, Rolla, MO 65409, United States
| | - Sima Azizi
- Department of Electrical and Computer Engineering, Missouri University of Science and Technology, Rolla, MO 65401, USA
| | - Blaine Allen
- Department of Electrical and Computer Engineering, Missouri University of Science and Technology, Rolla, MO 65401, USA
| | - Bassam A Hadi
- Department of Surgery, Mercy Hospital, St. Louis MO, Missouri, MO 63141, United States
| | - Donald C Wunsch
- Department of Electrical and Computer Engineering, Missouri University of Science and Technology, Rolla, MO 65401, USA.,National Science Foundation, ECCS Division, Virginia, 22314, USA
| |
Collapse
|
24
|
Shin MK, Vázquez-Rosa E, Koh Y, Dhar M, Chaubey K, Cintrón-Pérez CJ, Barker S, Miller E, Franke K, Noterman MF, Seth D, Allen RS, Motz CT, Rao SR, Skelton LA, Pardue MT, Fliesler SJ, Wang C, Tracy TE, Gan L, Liebl DJ, Savarraj JPJ, Torres GL, Ahnstedt H, McCullough LD, Kitagawa RS, Choi HA, Zhang P, Hou Y, Chiang CW, Li L, Ortiz F, Kilgore JA, Williams NS, Whitehair VC, Gefen T, Flanagan ME, Stamler JS, Jain MK, Kraus A, Cheng F, Reynolds JD, Pieper AA. Reducing acetylated tau is neuroprotective in brain injury. Cell 2021; 184:2715-2732.e23. [PMID: 33852912 PMCID: PMC8491234 DOI: 10.1016/j.cell.2021.03.032] [Citation(s) in RCA: 129] [Impact Index Per Article: 32.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2020] [Revised: 01/21/2021] [Accepted: 03/15/2021] [Indexed: 10/21/2022]
Abstract
Traumatic brain injury (TBI) is the largest non-genetic, non-aging related risk factor for Alzheimer's disease (AD). We report here that TBI induces tau acetylation (ac-tau) at sites acetylated also in human AD brain. This is mediated by S-nitrosylated-GAPDH, which simultaneously inactivates Sirtuin1 deacetylase and activates p300/CBP acetyltransferase, increasing neuronal ac-tau. Subsequent tau mislocalization causes neurodegeneration and neurobehavioral impairment, and ac-tau accumulates in the blood. Blocking GAPDH S-nitrosylation, inhibiting p300/CBP, or stimulating Sirtuin1 all protect mice from neurodegeneration, neurobehavioral impairment, and blood and brain accumulation of ac-tau after TBI. Ac-tau is thus a therapeutic target and potential blood biomarker of TBI that may represent pathologic convergence between TBI and AD. Increased ac-tau in human AD brain is further augmented in AD patients with history of TBI, and patients receiving the p300/CBP inhibitors salsalate or diflunisal exhibit decreased incidence of AD and clinically diagnosed TBI.
Collapse
Affiliation(s)
- Min-Kyoo Shin
- Harrington Discovery Institute, University Hospitals Cleveland Medical Center, Cleveland, OH, USA; Department of Psychiatry, Case Western Reserve University, Cleveland, OH, USA; Geriatric Psychiatry, GRECC, Louis Stokes Cleveland VA Medical Center; Cleveland, OH, USA; Institute for Transformative Molecular Medicine, School of Medicine, Case Western Reserve University, Cleveland, OH, USA
| | - Edwin Vázquez-Rosa
- Harrington Discovery Institute, University Hospitals Cleveland Medical Center, Cleveland, OH, USA; Department of Psychiatry, Case Western Reserve University, Cleveland, OH, USA; Geriatric Psychiatry, GRECC, Louis Stokes Cleveland VA Medical Center; Cleveland, OH, USA; Institute for Transformative Molecular Medicine, School of Medicine, Case Western Reserve University, Cleveland, OH, USA
| | - Yeojung Koh
- Harrington Discovery Institute, University Hospitals Cleveland Medical Center, Cleveland, OH, USA; Department of Psychiatry, Case Western Reserve University, Cleveland, OH, USA; Geriatric Psychiatry, GRECC, Louis Stokes Cleveland VA Medical Center; Cleveland, OH, USA; Institute for Transformative Molecular Medicine, School of Medicine, Case Western Reserve University, Cleveland, OH, USA
| | - Matasha Dhar
- Harrington Discovery Institute, University Hospitals Cleveland Medical Center, Cleveland, OH, USA; Department of Psychiatry, Case Western Reserve University, Cleveland, OH, USA; Geriatric Psychiatry, GRECC, Louis Stokes Cleveland VA Medical Center; Cleveland, OH, USA; Institute for Transformative Molecular Medicine, School of Medicine, Case Western Reserve University, Cleveland, OH, USA
| | - Kalyani Chaubey
- Harrington Discovery Institute, University Hospitals Cleveland Medical Center, Cleveland, OH, USA; Department of Psychiatry, Case Western Reserve University, Cleveland, OH, USA; Geriatric Psychiatry, GRECC, Louis Stokes Cleveland VA Medical Center; Cleveland, OH, USA; Institute for Transformative Molecular Medicine, School of Medicine, Case Western Reserve University, Cleveland, OH, USA
| | - Coral J Cintrón-Pérez
- Harrington Discovery Institute, University Hospitals Cleveland Medical Center, Cleveland, OH, USA; Department of Psychiatry, Case Western Reserve University, Cleveland, OH, USA; Geriatric Psychiatry, GRECC, Louis Stokes Cleveland VA Medical Center; Cleveland, OH, USA; Institute for Transformative Molecular Medicine, School of Medicine, Case Western Reserve University, Cleveland, OH, USA
| | - Sarah Barker
- Harrington Discovery Institute, University Hospitals Cleveland Medical Center, Cleveland, OH, USA; Department of Psychiatry, Case Western Reserve University, Cleveland, OH, USA; Geriatric Psychiatry, GRECC, Louis Stokes Cleveland VA Medical Center; Cleveland, OH, USA; Institute for Transformative Molecular Medicine, School of Medicine, Case Western Reserve University, Cleveland, OH, USA
| | - Emiko Miller
- Harrington Discovery Institute, University Hospitals Cleveland Medical Center, Cleveland, OH, USA; Department of Psychiatry, Case Western Reserve University, Cleveland, OH, USA; Geriatric Psychiatry, GRECC, Louis Stokes Cleveland VA Medical Center; Cleveland, OH, USA; Institute for Transformative Molecular Medicine, School of Medicine, Case Western Reserve University, Cleveland, OH, USA
| | - Kathryn Franke
- Harrington Discovery Institute, University Hospitals Cleveland Medical Center, Cleveland, OH, USA; Department of Psychiatry, Case Western Reserve University, Cleveland, OH, USA; Geriatric Psychiatry, GRECC, Louis Stokes Cleveland VA Medical Center; Cleveland, OH, USA; Institute for Transformative Molecular Medicine, School of Medicine, Case Western Reserve University, Cleveland, OH, USA
| | - Maria F Noterman
- Department of Psychiatry, University of Iowa Carver College of Medicine, Iowa City, IA, USA
| | - Divya Seth
- Institute for Transformative Molecular Medicine, School of Medicine, Case Western Reserve University, Cleveland, OH, USA; Department of Medicine, University Hospitals Cleveland Medical Center, Cleveland, OH, USA
| | - Rachael S Allen
- Center for Visual and Neurocognitive Rehabilitation, Atlanta VA Healthcare System, Atlanta, GA, USA; Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, GA, US
| | - Cara T Motz
- Center for Visual and Neurocognitive Rehabilitation, Atlanta VA Healthcare System, Atlanta, GA, USA; Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, GA, US
| | - Sriganesh Ramachandra Rao
- Departments of Ophthalmology and Biochemistry, and the Neuroscience Graduate Program, SUNY-University at Buffalo, Buffalo, NY, USA; Research Service, VA Western NY Healthcare System, Buffalo, NY, USA
| | - Lara A Skelton
- Departments of Ophthalmology and Biochemistry, and the Neuroscience Graduate Program, SUNY-University at Buffalo, Buffalo, NY, USA; Research Service, VA Western NY Healthcare System, Buffalo, NY, USA
| | - Machelle T Pardue
- Center for Visual and Neurocognitive Rehabilitation, Atlanta VA Healthcare System, Atlanta, GA, USA; Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, GA, US
| | - Steven J Fliesler
- Departments of Ophthalmology and Biochemistry, and the Neuroscience Graduate Program, SUNY-University at Buffalo, Buffalo, NY, USA; Research Service, VA Western NY Healthcare System, Buffalo, NY, USA
| | - Chao Wang
- Gladstone Institute of Neurological Disease, San Francisco, CA, USA
| | | | - Li Gan
- Helen and Robert Appel Alzheimer's Disease Research Institute, Weill Cornell Medicine, New York, NY, USA
| | - Daniel J Liebl
- The Miami Project to Cure Paralysis, Department of Neurological Surgery, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Jude P J Savarraj
- Department of Neurosurgery, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Glenda L Torres
- Department of Neurosurgery, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Hilda Ahnstedt
- Department of Neurology, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Louise D McCullough
- Department of Neurology, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Ryan S Kitagawa
- Department of Neurosurgery, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - H Alex Choi
- Department of Neurosurgery, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Pengyue Zhang
- Department of Biomedical Informatics, College of Medicine, Ohio State University, Columbus, OH, USA
| | - Yuan Hou
- Genomic Medicine Institute, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Chien-Wei Chiang
- Department of Biomedical Informatics, College of Medicine, Ohio State University, Columbus, OH, USA
| | - Lang Li
- Department of Biomedical Informatics, College of Medicine, Ohio State University, Columbus, OH, USA
| | - Francisco Ortiz
- Department of Biochemistry, UT Southwestern Medical Center, Dallas, TX, USA
| | - Jessica A Kilgore
- Department of Biochemistry, UT Southwestern Medical Center, Dallas, TX, USA
| | - Noelle S Williams
- Department of Biochemistry, UT Southwestern Medical Center, Dallas, TX, USA
| | - Victoria C Whitehair
- MetroHealth Rehabilitation Institute, The MetroHealth System, Cleveland, OH; Department of Physical Medicine and Rehabilitation, Case Western Reserve University, School of Medicine, Cleveland, OH USA
| | - Tamar Gefen
- Department of Psychiatry and Behavioral Sciences, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA; Mesulam Center for Cognitive Neurology and Alzheimer's Disease, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Margaret E Flanagan
- Mesulam Center for Cognitive Neurology and Alzheimer's Disease, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA; Department of Pathology, Northwestern University, Chicago, IL, USA
| | - Jonathan S Stamler
- Harrington Discovery Institute, University Hospitals Cleveland Medical Center, Cleveland, OH, USA; Institute for Transformative Molecular Medicine, School of Medicine, Case Western Reserve University, Cleveland, OH, USA; Department of Medicine, University Hospitals Cleveland Medical Center, Cleveland, OH, USA
| | - Mukesh K Jain
- Harrington Discovery Institute, University Hospitals Cleveland Medical Center, Cleveland, OH, USA; Department of Medicine, University Hospitals Cleveland Medical Center, Cleveland, OH, USA
| | - Allison Kraus
- Department of Pathology, Case Western Reserve University, School of Medicine, Cleveland, OH, USA
| | - Feixiong Cheng
- Genomic Medicine Institute, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA; Case Comprehensive Cancer Center, Case Western Reserve University School of Medicine, Cleveland, OH, USA; Department of Molecular Medicine, Cleveland Clinic Lerner College of Medicine, Case Western Reserve University, Cleveland, OH, USA
| | - James D Reynolds
- Harrington Discovery Institute, University Hospitals Cleveland Medical Center, Cleveland, OH, USA; Institute for Transformative Molecular Medicine, School of Medicine, Case Western Reserve University, Cleveland, OH, USA; Departments of Anesthesiology & Perioperative Medicine, University Hospitals Cleveland Medical Center, Cleveland, OH, USA
| | - Andrew A Pieper
- Harrington Discovery Institute, University Hospitals Cleveland Medical Center, Cleveland, OH, USA; Department of Psychiatry, Case Western Reserve University, Cleveland, OH, USA; Geriatric Psychiatry, GRECC, Louis Stokes Cleveland VA Medical Center; Cleveland, OH, USA; Institute for Transformative Molecular Medicine, School of Medicine, Case Western Reserve University, Cleveland, OH, USA; Weill Cornell Autism Research Program, Weill Cornell Medicine of Cornell University, New York, NY, USA; Department of Neuroscience, Case Western Reserve University, School of Medicine, Cleveland, OH, USA.
| |
Collapse
|
25
|
Turner S, Lazarus R, Marion D, Main KL. Molecular and Diffusion Tensor Imaging Biomarkers of Traumatic Brain Injury: Principles for Investigation and Integration. J Neurotrauma 2021; 38:1762-1782. [PMID: 33446015 DOI: 10.1089/neu.2020.7259] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
The last 20 years have seen the advent of new technologies that enhance the diagnosis and prognosis of traumatic brain injury (TBI). There is recognition that TBI affects the brain beyond initial injury, in some cases inciting a progressive neuropathology that leads to chronic impairments. Medical researchers are now searching for biomarkers to detect and monitor this condition. Perhaps the most promising developments are in the biomolecular and neuroimaging domains. Molecular assays can identify proteins indicative of neuronal injury and/or degeneration. Diffusion imaging now allows sensitive evaluations of the brain's cellular microstructure. As the pace of discovery accelerates, it is important to survey the research landscape and identify promising avenues of investigation. In this review, we discuss the potential of molecular and diffusion tensor imaging (DTI) biomarkers in TBI research. Integration of these technologies could advance models of disease prognosis, ultimately improving care. To date, however, few studies have explored relationships between molecular and DTI variables in patients with TBI. Here, we provide a short primer on each technology, review the latest research, and discuss how these biomarkers may be incorporated in future studies.
Collapse
Affiliation(s)
- Stephanie Turner
- Defense and Veterans Brain Injury Center, Silver Spring, Maryland, USA.,General Dynamics Information Technology, Falls Church, Virginia, USA
| | - Rachel Lazarus
- Defense and Veterans Brain Injury Center, Silver Spring, Maryland, USA.,General Dynamics Information Technology, Falls Church, Virginia, USA
| | - Donald Marion
- Defense and Veterans Brain Injury Center, Silver Spring, Maryland, USA.,General Dynamics Information Technology, Falls Church, Virginia, USA
| | - Keith L Main
- Defense and Veterans Brain Injury Center, Silver Spring, Maryland, USA.,General Dynamics Information Technology, Falls Church, Virginia, USA
| |
Collapse
|
26
|
Development of a novel, sensitive translational immunoassay to detect plasma glial fibrillary acidic protein (GFAP) after murine traumatic brain injury. ALZHEIMERS RESEARCH & THERAPY 2021; 13:58. [PMID: 33678186 PMCID: PMC7938597 DOI: 10.1186/s13195-021-00793-9] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/04/2020] [Accepted: 02/17/2021] [Indexed: 12/21/2022]
Abstract
Background Glial fibrillary acidic protein (GFAP) has emerged as a promising fluid biomarker for several neurological indications including traumatic brain injury (TBI), a leading cause of death and disability worldwide. In humans, serum or plasma GFAP levels can predict brain abnormalities including hemorrhage on computed tomography (CT) scans and magnetic resonance imaging (MRI). However, assays to quantify plasma or serum GFAP in preclinical models are not yet available. Methods We developed and validated a novel sensitive GFAP immunoassay assay for mouse plasma on the Meso Scale Discovery immunoassay platform and validated assay performance for robustness, precision, limits of quantification, dilutional linearity, parallelism, recovery, stability, selectivity, and pre-analytical factors. To provide proof-of-concept data for this assay as a translational research tool for TBI and Alzheimer’s disease (AD), plasma GFAP was measured in mice exposed to TBI using the Closed Head Impact Model of Engineered Rotational Acceleration (CHIMERA) model and in APP/PS1 mice with normal or reduced levels of plasma high-density lipoprotein (HDL). Results We performed a partial validation of our novel assay and found its performance by the parameters studied was similar to assays used to quantify human GFAP in clinical neurotrauma blood specimens and to assays used to measure murine GFAP in tissues. Specifically, we demonstrated an intra-assay CV of 5.0%, an inter-assay CV of 7.2%, a lower limit of detection (LLOD) of 9.0 pg/mL, a lower limit of quantification (LLOQ) of 24.8 pg/mL, an upper limit of quantification (ULOQ) of at least 16,533.9 pg/mL, dilution linearity of calibrators from 20 to 200,000 pg/mL with 90–123% recovery, dilution linearity of plasma specimens up to 32-fold with 96–112% recovery, spike recovery of 67–100%, and excellent analyte stability in specimens exposed to up to 7 freeze-thaw cycles, 168 h at 4 °C, 24 h at room temperature (RT), or 30 days at − 20 °C. We also observed elevated plasma GFAP in mice 6 h after TBI and in aged APP/PS1 mice with plasma HDL deficiency. This assay also detects GFAP in serum. Conclusions This novel assay is a valuable translational tool that may help to provide insights into the mechanistic pathophysiology of TBI and AD. Supplementary Information The online version contains supplementary material available at 10.1186/s13195-021-00793-9.
Collapse
|
27
|
Janigro D, Bailey DM, Lehmann S, Badaut J, O'Flynn R, Hirtz C, Marchi N. Peripheral Blood and Salivary Biomarkers of Blood-Brain Barrier Permeability and Neuronal Damage: Clinical and Applied Concepts. Front Neurol 2021; 11:577312. [PMID: 33613412 PMCID: PMC7890078 DOI: 10.3389/fneur.2020.577312] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Accepted: 12/01/2020] [Indexed: 12/12/2022] Open
Abstract
Within the neurovascular unit (NVU), the blood–brain barrier (BBB) operates as a key cerebrovascular interface, dynamically insulating the brain parenchyma from peripheral blood and compartments. Increased BBB permeability is clinically relevant for at least two reasons: it actively participates to the etiology of central nervous system (CNS) diseases, and it enables the diagnosis of neurological disorders based on the detection of CNS molecules in peripheral body fluids. In pathological conditions, a suite of glial, neuronal, and pericyte biomarkers can exit the brain reaching the peripheral blood and, after a process of filtration, may also appear in saliva or urine according to varying temporal trajectories. Here, we specifically examine the evidence in favor of or against the use of protein biomarkers of NVU damage and BBB permeability in traumatic head injury, including sport (sub)concussive impacts, seizure disorders, and neurodegenerative processes such as Alzheimer's disease. We further extend this analysis by focusing on the correlates of human extreme physiology applied to the NVU and its biomarkers. To this end, we report NVU changes after prolonged exercise, freediving, and gravitational stress, focusing on the presence of peripheral biomarkers in these conditions. The development of a biomarker toolkit will enable minimally invasive routines for the assessment of brain health in a broad spectrum of clinical, emergency, and sport settings.
Collapse
Affiliation(s)
- Damir Janigro
- Department of Physiology Case Western Reserve University, Cleveland, OH, United States.,FloTBI Inc., Cleveland, OH, United States
| | - Damian M Bailey
- Neurovascular Research Laboratory, Faculty of Life Sciences and Education, University of South Wales, Wales, United Kingdom
| | - Sylvain Lehmann
- IRMB, INM, UFR Odontology, University Montpellier, INSERM, CHU Montpellier, CNRS, Montpellier, France
| | - Jerome Badaut
- Brain Molecular Imaging Lab, CNRS UMR 5287, INCIA, University of Bordeaux, Bordeaux, France
| | - Robin O'Flynn
- IRMB, INM, UFR Odontology, University Montpellier, INSERM, CHU Montpellier, CNRS, Montpellier, France
| | - Christophe Hirtz
- IRMB, INM, UFR Odontology, University Montpellier, INSERM, CHU Montpellier, CNRS, Montpellier, France
| | - Nicola Marchi
- Cerebrovascular and Glia Research, Department of Neuroscience, Institute of Functional Genomics (UMR 5203 CNRS-U 1191 INSERM, University of Montpellier), Montpellier, France
| |
Collapse
|
28
|
Okonkwo DO, Puffer RC, Puccio AM, Yuh EL, Yue JK, Diaz-Arrastia R, Korley FK, Wang KKW, Sun X, Taylor SR, Mukherjee P, Markowitz AJ, Jain S, Manley GT. Point-of-Care Platform Blood Biomarker Testing of Glial Fibrillary Acidic Protein versus S100 Calcium-Binding Protein B for Prediction of Traumatic Brain Injuries: A Transforming Research and Clinical Knowledge in Traumatic Brain Injury Study. J Neurotrauma 2020; 37:2460-2467. [PMID: 32854584 PMCID: PMC7698990 DOI: 10.1089/neu.2020.7140] [Citation(s) in RCA: 91] [Impact Index Per Article: 18.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Glial fibrillary acidic protein (GFAP) is cleared by the Food and Drug Administration (FDA) to determine need for head computed tomography (CT) within 12 h after mild traumatic brain injury (TBI) (Glasgow Coma Score [GCS] 13-15); S100 calcium-binding protein B (S100B) serves this function in Europe. This phase 1 biomarker cohort analysis of the multi-center, observational Transforming Research and Clinical Knowledge in Traumatic Brain Injury (TRACK-TBI) study compares GFAP's diagnostic performance, measured on a rapid point-of-care platform, against protein S100B to predict intracranial abnormalities on CT within 24 h post-injury across the spectrum of TBI (GCS 3-15). Head CT scan performed in TBI subjects and blood was collected for all consenting subjects presenting to 18 United States level 1 trauma centers. Plasma was analyzed on a point-of-care device prototype assay for GFAP and serum was analyzed for S100B. In 1359 patients with TBI (GCS 3-15), mean (standard deviation [SD]) age = 40.1 (17.0) years; 68% were male. Plasma GFAP levels were significantly higher in CT+ TBI subjects (median = 1358 pg/mL, interquartile range [IQR]: 472-3803) than in CT- TBI subjects (median = 116 pg/mL, IQR: 26-397) or orthopedic trauma controls (n = 122; median = 13 pg/mL, IQR: 7-20), p < 0.001. Serum S100B levels were likewise higher in CT+ TBI subjects (median = 0.17 μg/L, IQR: 0.09-0.38) than in CT- TBI subjects (median = 0.10 μg/L, IQR: 0.06-0.18), p < 0.001. Receiver operating characteristic curves were generated for prediction of intracranial injury on admission CT scan; area under the curve (AUC) for GFAP was significantly higher than for S100B in the same cohort (GFAP AUC - 0.85, 95% confidence interval [CI] 0.83-0.87; S100B AUC - 0.67, 95% CI 0.64-0.70; p < 0.001). GFAP, measured on a point-of-care platform prototype assay, has high discriminative ability to predict intracranial abnormalities on CT scan in patients with TBI across the full injury spectrum of GCS 3-15 through 24 h post-injury. GFAP substantially outperforms S100B.
Collapse
Affiliation(s)
- David O. Okonkwo
- Department of Neurological Surgery, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania, USA
| | - Ross C. Puffer
- Department of Neurological Surgery, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania, USA
- Department of Neurological Surgery, Mayo Clinic, Rochester, Minnesota, USA
| | - Ava M. Puccio
- Department of Neurological Surgery, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania, USA
| | - Esther L. Yuh
- Departments of Radiology and University of California, San Francisco, San Francisco, California, USA
- Brain and Spinal Injury Center, University of California, San Francisco, San Francisco, California, USA
- Zuckerberg San Francisco General Hospital and Trauma Center, San Francisco, California, USA
| | - John K. Yue
- Brain and Spinal Injury Center, University of California, San Francisco, San Francisco, California, USA
- Zuckerberg San Francisco General Hospital and Trauma Center, San Francisco, California, USA
- Neurological Surgery, University of California, San Francisco, San Francisco, California, USA
| | - Ramon Diaz-Arrastia
- Department of Neurology, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Frederick K. Korley
- Department of Emergency Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Kevin K. W. Wang
- Departments of Psychiatry and Neuroscience, McKnight Brain Institute, University of Florida, Gainesville, Florida, USA
| | - Xiaoying Sun
- Department of Family Medicine and Public Health, University of California San Diego, San Diego, California, USA
| | - Sabrina R. Taylor
- Brain and Spinal Injury Center, University of California, San Francisco, San Francisco, California, USA
- Zuckerberg San Francisco General Hospital and Trauma Center, San Francisco, California, USA
- Neurological Surgery, University of California, San Francisco, San Francisco, California, USA
| | - Pratik Mukherjee
- Departments of Radiology and University of California, San Francisco, San Francisco, California, USA
- Brain and Spinal Injury Center, University of California, San Francisco, San Francisco, California, USA
- Zuckerberg San Francisco General Hospital and Trauma Center, San Francisco, California, USA
| | - Amy J. Markowitz
- Brain and Spinal Injury Center, University of California, San Francisco, San Francisco, California, USA
- Zuckerberg San Francisco General Hospital and Trauma Center, San Francisco, California, USA
- Neurological Surgery, University of California, San Francisco, San Francisco, California, USA
| | - Sonia Jain
- Department of Family Medicine and Public Health, University of California San Diego, San Diego, California, USA
| | - Geoffrey T. Manley
- Brain and Spinal Injury Center, University of California, San Francisco, San Francisco, California, USA
- Zuckerberg San Francisco General Hospital and Trauma Center, San Francisco, California, USA
- Neurological Surgery, University of California, San Francisco, San Francisco, California, USA
| |
Collapse
|
29
|
Hicks SD, Onks C, Kim RY, Zhen KJ, Loeffert J, Loeffert AC, Olympia RP, Fedorchak G, DeVita S, Rangnekar A, Leddy J, Haider MN, Gagnon Z, McLoughlin CD, Badia M, Randall J, Madeira M, Yengo‐Kahn AM, Wenzel J, Heller M, Zwibel H, Roberts A, Johnson S, Monteith C, Dretsch MN, Campbell TR, Mannix R, Neville C, Middleton F. Diagnosing mild traumatic brain injury using saliva RNA compared to cognitive and balance testing. Clin Transl Med 2020; 10:e197. [PMID: 33135344 PMCID: PMC7533415 DOI: 10.1002/ctm2.197] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2020] [Revised: 09/14/2020] [Accepted: 09/23/2020] [Indexed: 01/01/2023] Open
Abstract
BACKGROUND Early, accurate diagnosis of mild traumatic brain injury (mTBI) can improve clinical outcomes for patients, but mTBI remains difficult to diagnose because of reliance on subjective symptom reports. An objective biomarker could increase diagnostic accuracy and improve clinical outcomes. The aim of this study was to assess the ability of salivary noncoding RNA (ncRNA) to serve as a diagnostic adjunct to current clinical tools. We hypothesized that saliva ncRNA levels would demonstrate comparable accuracy for identifying mTBI as measures of symptom burden, neurocognition, and balance. METHODS This case-control study involved 538 individuals. Participants included 251 individuals with mTBI, enrolled ≤14 days postinjury, from 11 clinical sites. Saliva samples (n = 679) were collected at five time points (≤3, 4-7, 8-14, 15-30, and 31-60 days post-mTBI). Levels of ncRNAs (microRNAs, small nucleolar RNAs, and piwi-interacting RNAs) were quantified within each sample using RNA sequencing. The first sample from each mTBI participant was compared to saliva samples from 287 controls. Samples were divided into testing (n = 430; mTBI = 201 and control = 239) and training sets (n = 108; mTBI = 50 and control = 58). The test set was used to identify ncRNA diagnostic candidates and create a diagnostic model. Model accuracy was assessed in the naïve test set. RESULTS A model utilizing seven ncRNA ratios, along with participant age and chronic headache status, differentiated mTBI and control participants with a cross-validated area under the curve (AUC) of .857 in the training set (95% CI, .816-.903) and .823 in the naïve test set. In a subset of participants (n = 321; mTBI = 176 and control = 145) assessed for symptom burden (Post-Concussion Symptom Scale), as well as neurocognition and balance (ClearEdge System), these clinical measures yielded cross-validated AUC of .835 (95% CI, .782-.880) and .853 (95% CI, .803-.899), respectively. A model employing symptom burden and four neurocognitive measures identified mTBI participants with similar AUC (.888; CI, .845-.925) as symptom burden and four ncRNAs (.932; 95% CI, .890-.965). CONCLUSION Salivary ncRNA levels represent a noninvasive, biologic measure that can aid objective, accurate diagnosis of mTBI.
Collapse
Affiliation(s)
- Steven D. Hicks
- Department of PediatricsPenn State College of MedicineHersheyPennsylvania
| | - Cayce Onks
- Department of Family MedicinePenn State College of MedicineHersheyPennsylvania
| | - Raymond Y. Kim
- Department of Orthopedics and RehabilitationPenn State College of MedicineHersheyPennsylvania
| | - Kevin J. Zhen
- Department of PediatricsPenn State College of MedicineHersheyPennsylvania
| | - Jayson Loeffert
- Department of Family MedicinePenn State College of MedicineHersheyPennsylvania
| | - Andrea C. Loeffert
- Department of PediatricsPenn State College of MedicineHersheyPennsylvania
| | - Robert P. Olympia
- Department of Emergency MedicinePenn State College of MedicineHersheyPennsylvania
| | | | | | | | - John Leddy
- UBMD Orthopedics and Sports Medicine, Jacobs School of Medicine and Biomedical SciencesState University of New YorkBuffaloNew York
| | - Mohammad N. Haider
- UBMD Orthopedics and Sports Medicine, Jacobs School of Medicine and Biomedical SciencesState University of New YorkBuffaloNew York
| | - Zofia Gagnon
- Department of Biomedical ScienceMarist CollegePoughkeepsieNew York
| | | | - Matthew Badia
- Department of Biomedical ScienceMarist CollegePoughkeepsieNew York
| | - Jason Randall
- Department of Environmental ScienceSchool of ScienceMarist CollegePoughkeepsieNew York
| | - Miguel Madeira
- Department of Biology, School of ScienceMarist CollegePoughkeepsieNew York
| | - Aaron M. Yengo‐Kahn
- Vanderbilt Sports Concussion CenterVanderbilt University Medical CenterNashvilleTennessee
| | - Justin Wenzel
- Vanderbilt Sports Concussion CenterVanderbilt University Medical CenterNashvilleTennessee
| | - Matthew Heller
- Department of Family MedicineNew York Institute of Technology College of Osteopathic MedicineOld WestburyNew York
| | - Hallie Zwibel
- Department of Family MedicineNew York Institute of Technology College of Osteopathic MedicineOld WestburyNew York
| | - Aaron Roberts
- Adena Bone and Joint CenterAdena Regional Medical CenterChillicotheOhio
| | - Samantha Johnson
- Adena Bone and Joint CenterAdena Regional Medical CenterChillicotheOhio
| | - Chuck Monteith
- Athletic Training DepartmentColgate UniversityHamiltonNew York
| | - Michael N. Dretsch
- US Army Medical Research Directorate‐WestWalter Reed Army Institute of ResearchJoint Base Lewis–McChordWashington
| | | | - Rebekah Mannix
- Division of Emergency Medicine, Boston Children's HospitalHarvard Medical SchoolBostonMassachusetts
| | - Christopher Neville
- Department of PT Education, Orthopedics, and NeuroscienceSUNY Upstate Medical UniversitySyracuseNew York
| | - Frank Middleton
- Department of Neuroscience and PhysiologySUNY Upstate Medical UniversitySyracuseNew York
| |
Collapse
|
30
|
Siman R, Cui H, Wewerka SS, Hamel L, Smith DH, Zwank MD. Serum SNTF, a Surrogate Marker of Axonal Injury, Is Prognostic for Lasting Brain Dysfunction in Mild TBI Treated in the Emergency Department. Front Neurol 2020; 11:249. [PMID: 32322237 PMCID: PMC7156622 DOI: 10.3389/fneur.2020.00249] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2019] [Accepted: 03/13/2020] [Indexed: 11/30/2022] Open
Abstract
Mild traumatic brain injury (mTBI) causes persisting post-concussion syndrome for many patients without abnormalities on conventional neuroimaging. Currently, there is no method for identifying at-risk cases at an early stage for directing concussion management and treatment. SNTF is a calpain-derived N-terminal proteolytic fragment of spectrin (αII-spectrin1-1176) generated in damaged axons following mTBI. Preliminary human studies suggest that elevated blood SNTF on the day of mTBI correlates with white matter disruption and lasting brain dysfunction. Here, we further evaluated serum SNTF as a prognostic marker for persistent brain dysfunction in uncomplicated mTBI patients treated in a Level I trauma center emergency department. Compared with healthy controls (n = 40), serum SNTF increased by 92% within 24 h of mTBI (n = 95; p < 0.0001), and as a diagnostic marker exhibited 100% specificity and 37% sensitivity (AUC = 0.87). To determine whether the subset of mTBI cases positive for SNTF preferentially developed lasting brain dysfunction, serum levels on the day of mTBI were compared with multiple measures of brain performance at 90 days post-injury. Elevated serum SNTF correlated significantly with persistent impairments in cognition and sensory-motor integration, and predicted worse performance in each test on a case by case basis (AUC = 0.68 and 0.76, respectively). SNTF also predicted poorer recovery of cognitive stress function from 30 to 90 days (AUC = 0.79–0.90). These results suggest that serum SNTF, a surrogate marker for axonal injury after mTBI, may have potential for the rapid prognosis of lasting post-concussion syndrome and impaired functional recovery following CT-negative mTBI. They provide further evidence linking axonal injury to persisting brain dysfunction after uncomplicated mTBI. A SNTF blood test, either alone or combined with other markers of axonal injury, may have important utilities for research, prognosis, management and treatment of concussion.
Collapse
Affiliation(s)
- Robert Siman
- Department of Neurosurgery, Center for Brain Injury and Repair, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Hongmei Cui
- Department of Neurosurgery, Center for Brain Injury and Repair, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Sandi S Wewerka
- Department of Emergency Medicine, Regions Hospital, St. Paul, MN, United States
| | - Lydia Hamel
- Department of Emergency Medicine, Regions Hospital, St. Paul, MN, United States
| | - Douglas H Smith
- Department of Neurosurgery, Center for Brain Injury and Repair, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Michael D Zwank
- Department of Emergency Medicine, Regions Hospital, St. Paul, MN, United States
| |
Collapse
|
31
|
Hiskens MI, Schneiders AG, Angoa-Pérez M, Vella RK, Fenning AS. Blood biomarkers for assessment of mild traumatic brain injury and chronic traumatic encephalopathy. Biomarkers 2020; 25:213-227. [PMID: 32096416 DOI: 10.1080/1354750x.2020.1735521] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Mild traumatic brain injuries (mTBI) are prevalent and can result in significant debilitation. Current diagnostic methods have implicit limitations, with clinical assessment tools reliant on subjective self-reported symptoms or non-specific clinical observations, and commonly available imaging techniques lacking sufficient sensitivity to detect mTBI. A blood biomarker would provide a readily accessible detector of mTBI to meet the current measurement gap. Suitable options would provide objective and quantifiable information in diagnosing mTBI, in monitoring recovery, and in establishing a prognosis of resultant neurodegenerative disease, such as chronic traumatic encephalopathy (CTE). A biomarker would also assist in progressing research, providing suitable endpoints for testing therapeutic modalities and for further exploring mTBI pathophysiology. This review highlights the most promising blood-based protein candidates that are expressed in the central nervous system (CNS) and released into systemic circulation following mTBI. To date, neurofilament light (NF-L) may be the most suitable candidate for assessing neuronal damage, and glial fibrillary acidic protein (GFAP) for assessing astrocyte activation, although further work is required. Ultimately, the heterogeneity of cells in the brain and each marker's limitations may require a combination of biomarkers, and recent developments in microRNA (miRNA) markers of mTBI show promise and warrant further exploration.
Collapse
Affiliation(s)
- Matthew I Hiskens
- School of Health, Medical and Applied Sciences, Central Queensland University, Rockhampton, Australia
| | - Anthony G Schneiders
- School of Health, Medical and Applied Sciences, Central Queensland University, Rockhampton, Australia
| | - Mariana Angoa-Pérez
- Research and Development Service, John D. Dingell VA Medical Center, Detroit, MI, USA.,Department of Psychiatry and Behavioral Neurosciences, Wayne State University School of Medicine, Detroit, MI, USA
| | - Rebecca K Vella
- School of Health, Medical and Applied Sciences, Central Queensland University, Rockhampton, Australia
| | - Andrew S Fenning
- School of Health, Medical and Applied Sciences, Central Queensland University, Rockhampton, Australia
| |
Collapse
|
32
|
Prognostic Value of Glial Fibrillary Acidic Protein in Patients With Moderate and Severe Traumatic Brain Injury: A Systematic Review and Meta-Analysis. Crit Care Med 2020; 47:e522-e529. [PMID: 30889029 DOI: 10.1097/ccm.0000000000003728] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
OBJECTIVES Biomarkers have been suggested as potential prognostic predictors following a moderate or severe traumatic brain injury but their prognostic accuracy is still uncertain. The objective of this systematic review is to assess the ability of the glial fibrillary acidic protein to predict prognosis in patients with moderate or severe traumatic brain injury. DATA SOURCES MEDLINE, Embase, CENTRAL, and BIOSIS electronic databases and conference abstracts, bibliographies of selected studies, and narrative reviews were searched. STUDY SELECTION Pairs of reviewers identified eligible studies. Cohort studies including greater than or equal to four patients with moderate or severe traumatic brain injury and reporting glial fibrillary acidic protein levels according to the outcomes of interest, namely Glasgow Outcome Scale or Extended Glasgow Outcome Scale, and mortality, were eligible. DATA EXTRACTION Pairs of reviewers independently extracted data from the selected studies using a standardized case report form. Mean levels were log-transformed, and their differences were pooled with random effect models. Results are presented as geometric mean ratios. Methodologic quality, risk of bias, and applicability concerns of the included studies were assessed. DATA SYNTHESIS Seven-thousand seven-hundred sixty-five citations were retrieved of which 15 studies were included in the systematic review (n = 1,070), and nine were included in the meta-analysis (n = 701). We found significant associations between glial fibrillary acidic protein serum levels and Glasgow Outcome Scale score less than or equal to 3 or Extended Glasgow Outcome Scale score less than or equal to 4 (six studies: geometric mean ratio 4.98 [95% CI, 2.19-11.13]; I = 94%) and between mortality (seven studies: geometric mean ratio 8.13 [95% CI, 3.89-17.00]; I = 99%). CONCLUSIONS Serum glial fibrillary acidic protein levels were significantly higher in patients with an unfavorable prognosis. Glial fibrillary acidic protein has a potential for clinical bedside use in helping for prognostic assessment. Further research should focus on multimodal approaches including tissue biomarkers for prognostic evaluation in critically ill patients with traumatic brain injury.
Collapse
|
33
|
Yue JK, Upadhyayula PS, Avalos LN, Deng H, Wang KKW. The Role of Blood Biomarkers for Magnetic Resonance Imaging Diagnosis of Traumatic Brain Injury. MEDICINA (KAUNAS, LITHUANIA) 2020; 56:E87. [PMID: 32098419 PMCID: PMC7074393 DOI: 10.3390/medicina56020087] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/20/2019] [Revised: 02/07/2020] [Accepted: 02/19/2020] [Indexed: 01/21/2023]
Abstract
Background and Objectives: The annual global incidence of traumatic brain injury (TBI) is over 10 million. An estimated 29% of TBI patients with negative computed tomography (CT-) have positive magnetic resonance imaging (MRI+) findings. Judicious use of serum biomarkers with MRI may aid in diagnosis of CT-occult TBI. The current manuscript aimed to evaluate the diagnostic, therapeutic and risk-stratification utility of known biomarkers and intracranial MRI pathology. Materials and Methods: The PubMed database was queried with keywords (plasma OR serum) AND (biomarker OR marker OR protein) AND (brain injury/trauma OR head injury/trauma OR concussion) AND (magnetic resonance imaging/MRI) (title/abstract) in English. Seventeen articles on TBI biomarkers and MRI were included: S100 calcium-binding protein B (S100B; N = 6), glial fibrillary acidic protein (GFAP; N = 3), GFAP/ubiquitin carboxyl-terminal hydrolase-L1 (UCH-L1; N = 2), Tau (N = 2), neurofilament-light (NF-L; N = 2), alpha-synuclein (N = 1), and alpha-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid receptor peptide (AMPAR; N = 1). Results: Acute GFAP distinguished CT-/MRI+ from CT-/MRI- (AUC = 0.777, 0.852 at 9-16 h). GFAP discriminated CT-/diffuse axonal injury (DAI+) from controls (AUC = 0.903). Tau correlated directly with number of head strikes and inversely with white matter fractional anisotropy (FA), and a cutoff > 1.5 pg/mL discriminated between DAI+ and DAI- (sensitivity = 74%/specificity = 69%). NF-L had 100% discrimination of DAI in severe TBI and correlated with FA. Low alpha-synuclein was associated with poorer functional connectivity. AMPAR cutoff > 0.4 ng/mL had a sensitivity of 91% and a specificity of 92% for concussion and was associated with minor MRI findings. Low/undetectable S100B had a high negative predictive value for CT/MRI pathology. UCH-L1 showed no notable correlations with MRI. Conclusions: An acute circulating biomarker capable of discriminating intracranial MRI abnormalities is critical to establishing diagnosis for CT-occult TBI and can triage patients who may benefit from outpatient MRI, surveillance and/or follow up with TBI specialists. GFAP has shown diagnostic potential for MRI findings such as DAI and awaits further validation. Tau shows promise in detecting DAI and disrupted functional connectivity. Candidate biomarkers should be evaluated within the context of analytical performance of the assays used, as well as the post-injury timeframe for blood collection relative to MRI abnormalities.
Collapse
Affiliation(s)
- John K. Yue
- Department of Neurological Surgery, University of California San Francisco, San Francisco, CA 94143, USA;
- Brain and Spinal Injury Center, Zuckerberg San Francisco General Hospital, San Francisco, CA 94110, USA
| | - Pavan S. Upadhyayula
- Department of Neurological Surgery, Columbia University Medical Center, New York, NY 10027, USA;
- Department of Neurological Surgery, University of California Diego, San Diego, CA 92093, USA
| | - Lauro N. Avalos
- Department of Neurological Surgery, University of California San Francisco, San Francisco, CA 94143, USA;
| | - Hansen Deng
- Department of Neurological Surgery, University of Pittsburgh Medical Center, Pittsburgh, PA 15213, USA;
| | - Kevin K. W. Wang
- Brain Rehabilitation Research Center (BRRC), Malcom Randall Veterans Affairs Medical Center, Gainesville, FL 32608, USA;
| |
Collapse
|
34
|
Boutté AM, Thangavelu B, LaValle CR, Nemes J, Gilsdorf J, Shear DA, Kamimori GH. Brain-related proteins as serum biomarkers of acute, subconcussive blast overpressure exposure: A cohort study of military personnel. PLoS One 2019; 14:e0221036. [PMID: 31408492 PMCID: PMC6692016 DOI: 10.1371/journal.pone.0221036] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2019] [Accepted: 07/29/2019] [Indexed: 12/13/2022] Open
Abstract
Repeated exposure to blast overpressure remains a major cause of adverse health for military personnel who, as a consequence, are at a higher risk for neurodegenerative disease and suicide. Acute, early tracking of blast related effects holds the promise of rapid health assessment prior to onset of chronic problems. Current techniques used to determine blast-related effects rely upon reporting of symptomology similar to that of concussion and neurocognitive assessment relevant to operational decrement. Here, we describe the results of a cross sectional study with pared observations. The concentration of multiple TBI-related proteins was tested in serum collected within one hour of blast exposure as a quantitative and minimally invasive strategy to augment assessment of blast-exposure effects that are associated with concussion-like symptomology and reaction time decrements. We determined that median simple reaction time (SRT) was slowed in accordance with serum Nf-L, tau, Aβ-40, and Aβ-42 elevation after overpressure exposure. In contrast, median levels of serum GFAP decreased. Individual, inter-subject analysis revealed positive correlations between changes in Nf-L and GFAP, and in Aβ-40 compared to Aβ-42. The change in Nf-L was negatively associated with tau, Aβ-40, and Aβ-42. Participants reported experiencing headaches, dizziness and taking longer to think. Dizziness was associated with reaction time decrements, GFAP or NfL suppression, as well as Aβ peptide elevation. UCH-L1 elevation had a weak association with mTBI/concussion history. Multiplexed serum biomarker quantitation, coupled with reaction time assessment and symptomology determined before and after blast exposure, may serve as a platform for tracking adverse effects in the absence of a head wound or diagnosed concussion. We propose further evaluation of serum biomarkers, which are often associated with TBI, in the context of acute operational blast exposures.
Collapse
Affiliation(s)
- Angela M. Boutté
- Brain Trauma Neuroprotection Branch, Center for Military Psychiatry and Neuroscience, Walter Reed Army Institute of Research, Silver Spring, Maryland, United States of America
| | - Bharani Thangavelu
- Brain Trauma Neuroprotection Branch, Center for Military Psychiatry and Neuroscience, Walter Reed Army Institute of Research, Silver Spring, Maryland, United States of America
| | - Christina R. LaValle
- Blast Induced Neurotrauma Branch, Center for Military Psychiatry and Neuroscience, Walter Reed Army Institute of Research, Silver Spring, Maryland, United States of America
| | - Jeffrey Nemes
- Blast Induced Neurotrauma Branch, Center for Military Psychiatry and Neuroscience, Walter Reed Army Institute of Research, Silver Spring, Maryland, United States of America
| | - Janice Gilsdorf
- Brain Trauma Neuroprotection Branch, Center for Military Psychiatry and Neuroscience, Walter Reed Army Institute of Research, Silver Spring, Maryland, United States of America
| | - Deborah A. Shear
- Brain Trauma Neuroprotection Branch, Center for Military Psychiatry and Neuroscience, Walter Reed Army Institute of Research, Silver Spring, Maryland, United States of America
| | - Gary H. Kamimori
- Blast Induced Neurotrauma Branch, Center for Military Psychiatry and Neuroscience, Walter Reed Army Institute of Research, Silver Spring, Maryland, United States of America
| |
Collapse
|
35
|
Morris MC, Bercz A, Niziolek GM, Kassam F, Veile R, Friend LA, Pritts TA, Makley AT, Goodman MD. UCH-L1 is a Poor Serum Biomarker of Murine Traumatic Brain Injury After Polytrauma. J Surg Res 2019; 244:63-68. [PMID: 31279265 DOI: 10.1016/j.jss.2019.06.023] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2019] [Revised: 04/23/2019] [Accepted: 06/06/2019] [Indexed: 12/22/2022]
Abstract
BACKGROUND Several serum biomarkers have been studied to diagnose incidence and severity of traumatic brain injury (TBI), but a reliable biomarker in TBI has yet to be identified. Ubiquitin carboxy-terminal hydrolase L1 (UCH-L1) has been proposed as a biomarker in clinical and preclinical studies, largely in the setting of isolated TBI or concussion. The aim of this study was to evaluate the performance of UCH-L1 as a serum biomarker in the setting of polytrauma and TBI. METHODS Multiple variations of murine TBI and polytrauma models were used to evaluate serum biomarkers. The different models included TBI with and without hemorrhagic shock and resuscitation, isolated extremity vascular ligation, extremity ischemia/reperfusion, and blunt tail injury. Blood was drawn at intervals after injury, and serum levels of neuron-specific enolase, UCH-L1, creatine kinase, and syndecan-1 were evaluated by enzyme-linked immunosorbent assay. RESULTS UCH-L1 levels were not significantly different between TBI, tail injury, and sham TBI. By contrast, neuron-specific enolase levels were increased in TBI mice compared with tail injury and sham TBI mice. UCH-L1 levels increased regardless of TBI status at 30 min and 4 h after hemorrhagic shock and resuscitation. In mice that underwent femoral artery cannulation followed by hemorrhagic shock/resuscitation, UCH-L1 levels were significantly elevated compared with shock sham mice at 4 h (3158 ± 2168 pg/mL, 4 h shock versus 0 ± 0 pg/mL, 4 h shock sham; P < 0.01) and at 24 h (3253 ± 2954 pg/mL, 24 h shock versus 324 ± 482 pg/mL, 24 h shock sham; P = 0.03). No differences were observed in UCH-L1 levels between the sham shock and the arterial ligation, vein ligation, or extremity ischemia/reperfusion groups at any time point. Similar to UCH-L1, creatine kinase was elevated only after shock compared with sham mice at 4, 24, and 72 h after injury. CONCLUSIONS Our study demonstrates that UCH-L1 is not a specific marker for TBI but is elevated in models that induce central and peripheral nerve ischemia. Given the increase in UCH-L1 levels observed after hemorrhagic shock, we propose that UCH-L1 may be a useful adjunct in quantifying severity of shock or global ischemia rather than as a specific marker of TBI.
Collapse
Affiliation(s)
| | - Aron Bercz
- Department of Surgery, University of Cincinnati, Cincinnati, Ohio
| | - Grace M Niziolek
- Department of Surgery, University of Cincinnati, Cincinnati, Ohio
| | - Farzaan Kassam
- Department of Surgery, University of Cincinnati, Cincinnati, Ohio
| | - Rose Veile
- Department of Surgery, University of Cincinnati, Cincinnati, Ohio
| | - Lou Ann Friend
- Department of Surgery, University of Cincinnati, Cincinnati, Ohio
| | - Timothy A Pritts
- Department of Surgery, University of Cincinnati, Cincinnati, Ohio
| | - Amy T Makley
- Department of Surgery, University of Cincinnati, Cincinnati, Ohio
| | | |
Collapse
|
36
|
Thelin E, Al Nimer F, Frostell A, Zetterberg H, Blennow K, Nyström H, Svensson M, Bellander BM, Piehl F, Nelson DW. A Serum Protein Biomarker Panel Improves Outcome Prediction in Human Traumatic Brain Injury. J Neurotrauma 2019; 36:2850-2862. [PMID: 31072225 PMCID: PMC6761606 DOI: 10.1089/neu.2019.6375] [Citation(s) in RCA: 127] [Impact Index Per Article: 21.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
Brain-enriched protein biomarkers of tissue fate are being introduced clinically to aid in traumatic brain injury (TBI) management. The aim of this study was to determine how concentrations of six different protein biomarkers, measured in samples collected during the first weeks after TBI, relate to injury severity and outcome. We included neurocritical care TBI patients that were prospectively enrolled from 2007 to 2013, all having one to three blood samples drawn during the first 2 weeks. The biomarkers analyzed were S100 calcium-binding protein B (S100B), neuron-specific enolase (NSE), glial fibrillary acidic protein (GFAP), ubiquitin carboxy-terminal hydrolase-L1 (UCH-L1), tau, and neurofilament-light (NF-L). Glasgow Outcome Score (GOS) was assessed at 12 months. In total, 172 patients were included. All serum markers were associated with injury severity as classified on computed tomography scans at admission. Almost all biomarkers outperformed other known outcome predictors with higher levels the first 5 days, correlating with unfavorable outcomes, and UCH-L1 (0.260, pseduo-R2) displaying the best discrimination in univariate analyses. After adjusting for acknowledged TBI outcome predictors, GFAP and NF-L added most independent information to predict favorable/unfavorable GOS, improving the model from 0.38 to 0.51 pseudo-R2. A correlation matrix indicated substantial covariance, with the strongest correlation between UCH-L1, GFAP, and tau (r = 0.827-0.880). Additionally, the principal component analysis exhibited clustering of UCH-L1 and tau, as well as GFAP, S100B, and NSE, which was separate from NF-L. In summary, a panel of several different protein biomarkers, all associated with injury severity, with different cellular origin and temporal trajectories, improve outcome prediction models.
Collapse
Affiliation(s)
- Eric Thelin
- Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden.,Department of Neurology, Karolinska University Hospital, Stockholm, Sweden.,Division of Neurosurgery, Department of Clinical Neurosciences, University of Cambridge, Cambridge, United Kingdom
| | - Faiez Al Nimer
- Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden.,Department of Neurology, Karolinska University Hospital, Stockholm, Sweden
| | - Arvid Frostell
- Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Henrik Zetterberg
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden.,Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, the Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden.,Department of Neurodegenerative Disease, UCL Institute of Neurology, Queen Square, London, United Kingdom.,UK Dementia Research Institute, UCL, London, United Kingdom
| | - Kaj Blennow
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden.,Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, the Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
| | - Harriet Nyström
- Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden.,Department of Neuroradiology, Karolinska University Hospital, Stockholm, Sweden
| | - Mikael Svensson
- Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden.,Department of Neurosurgery, Karolinska University Hospital, Stockholm, Sweden
| | - Bo-Michael Bellander
- Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden.,Department of Neurosurgery, Karolinska University Hospital, Stockholm, Sweden
| | - Fredrik Piehl
- Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden.,Department of Neurology, Karolinska University Hospital, Stockholm, Sweden
| | - David W Nelson
- Department of Physiology and Pharmacology, Section of Perioperative Medicine and Intensive Care, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
37
|
Posti JP, Takala RSK, Tenovuo O. TBIcare Investigators' Response to Papa and Wang (doi: 10:1089/neu.2017.5030): Raising the Bar for Traumatic Brain Injury Biomarker Research: Methods Make a Difference. J Neurotrauma 2019; 36:1680-1681. [PMID: 30569817 DOI: 10.1089/neu.2017.5209] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Affiliation(s)
- Jussi P Posti
- 1 Division of Clinical Neurosciences, Department of Neurosurgery, and Turku University Hospital, Turku, Finland.,2 Turku Brain Injury Centre, Turku University Hospital, Turku, Finland.,3 Department of Neurology, University of Turku, Turku, Finland
| | - Riikka S K Takala
- 4 Perioperative Services, Intensive Care Medicine and Pain Management, Turku University Hospital and University of Turku, Turku, Finland
| | - Olli Tenovuo
- 2 Turku Brain Injury Centre, Turku University Hospital, Turku, Finland.,3 Department of Neurology, University of Turku, Turku, Finland
| |
Collapse
|
38
|
Gan ZS, Stein SC, Swanson R, Guan S, Garcia L, Mehta D, Smith DH. Blood Biomarkers for Traumatic Brain Injury: A Quantitative Assessment of Diagnostic and Prognostic Accuracy. Front Neurol 2019; 10:446. [PMID: 31105646 PMCID: PMC6498532 DOI: 10.3389/fneur.2019.00446] [Citation(s) in RCA: 105] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2019] [Accepted: 04/12/2019] [Indexed: 12/18/2022] Open
Abstract
Blood biomarkers have been explored for their potential to provide objective measures in the assessment of traumatic brain injury (TBI). However, it is not clear which biomarkers are best for diagnosis and prognosis in different severities of TBI. Here, we compare existing studies on the discriminative abilities of serum biomarkers for four commonly studied clinical situations: detecting concussion, predicting intracranial damage after mild TBI (mTBI), predicting delayed recovery after mTBI, and predicting adverse outcome after severe TBI (sTBI). We conducted a literature search of publications on biomarkers in TBI published up until July 2018. Operating characteristics were pooled for each biomarker for comparison. For detecting concussion, 4 biomarker panels and creatine kinase B type had excellent discriminative ability. For detecting intracranial injury and the need for a head CT scan after mTBI, 2 biomarker panels, and hyperphosphorylated tau had excellent operating characteristics. For predicting delayed recovery after mTBI, top candidates included calpain-derived αII-spectrin N-terminal fragment, tau A, neurofilament light, and ghrelin. For predicting adverse outcome following sTBI, no biomarker had excellent performance, but several had good performance, including markers of coagulation and inflammation, structural proteins in the brain, and proteins involved in homeostasis. The highest-performing biomarkers in each of these categories may provide insight into the pathophysiologies underlying mild and severe TBI. With further study, these biomarkers have the potential to be used alongside clinical and radiological data to improve TBI diagnostics, prognostics, and evidence-based medical management.
Collapse
Affiliation(s)
- Zoe S Gan
- University of North Carolina School of Medicine, Chapel Hill, NC, United States
| | - Sherman C Stein
- Department of Neurosurgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Randel Swanson
- Department of Physical Medicine and Rehabilitation, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States.,Rehabilitation Medicine Service, Corporal Michael J. Crescenz VA Medical Center, Philadelphia, PA, United States.,Center for Neurotrauma, Neurodegeneration and Restoration, Corporal Michael J. Crescenz VA Medical Center, Philadelphia, PA, United States.,Department of Neurosurgery, Perelman School of Medicine, Center for Brain Injury and Repair, University of Pennsylvania, Philadelphia, PA, United States
| | - Shaobo Guan
- Department of Neurosurgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Lizette Garcia
- Department of Neurosurgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Devanshi Mehta
- Department of Neurosurgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Douglas H Smith
- Department of Neurosurgery, Perelman School of Medicine, Center for Brain Injury and Repair, University of Pennsylvania, Philadelphia, PA, United States
| |
Collapse
|
39
|
Iverson GL, Reddi PJ, Posti JP, Kotilainen AK, Tenovuo O, Öhman J, Zetterberg H, Blennow K, Luoto TM. Serum Neurofilament Light Is Elevated Differentially in Older Adults with Uncomplicated Mild Traumatic Brain Injuries. J Neurotrauma 2019; 36:2400-2406. [PMID: 30843469 DOI: 10.1089/neu.2018.6341] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Neurofilament light (NF-L) might have diagnostic and prognostic potential as a blood biomarker for mild traumatic brain injury (mTBI). However, elevated NF-L is associated with several neurological disorders associated with older age, which could confound its usefulness as a traumatic brain injury biomarker. We examined whether NF-L is elevated differentially following uncomplicated mTBI in older adults with pre-injury neurological disorders. In a case-control study, a sample of 118 adults (mean age = 62.3 years, standard deviation [SD] = 22.5, range = 18-100; 52.5% women) presenting to the emergency department (ED) with an uncomplicated mTBI were enrolled. All participants underwent head computed tomography in the ED and showed no macroscopic evidence of injury. The mean time between injury and blood sampling was 8.3 h (median [Md] = 3.5; SD = 13.5; interquartile range [IQR] = 1.9-6.0, range = 0.8-67.4, and 90% collected within 19 h). A sample of 40 orthopedically-injured trauma control subjects recruited from a second ED also were examined. Serum NF-L levels were measured and analyzed using Human Neurology 4-Plex A assay on a HD-1 Single Molecule Array (Simoa) instrument. A high correlation was found between age and NF-L levels in the total mTBI sample (r = 0.80), within the subgroups without pre-injury neurological diseases (r = 0.76) and with pre-injury neurological diseases (r = 0.68), and in the trauma control subjects (r = 0.76). Those with mTBIs and pre-injury neurological conditions had higher NF-L levels than those with no pre-injury neurological conditions (p < 0.001, Cohen's d = 1.01). Older age and pre-injury neurological diseases are associated with elevated serum NF-L levels in patients with head trauma and in orthopedically-injured control subjects.
Collapse
Affiliation(s)
- Grant L Iverson
- 1Department of Physical Medicine and Rehabilitation, Harvard Medical School, Spaulding Rehabilitation Hospital, and Home Base, a Red Sox Foundation and Massachusetts General Hospital Program, Boston, Massachusetts
| | | | - Jussi P Posti
- 3Department of Neurosurgery and Turku University Hospital and University of Turku, Turku, Finland.,4Turku Brain Injury Centre, Turku University Hospital and University of Turku, Turku, Finland
| | | | - Olli Tenovuo
- 4Turku Brain Injury Centre, Turku University Hospital and University of Turku, Turku, Finland
| | - Juha Öhman
- 6Department of Neurosurgery, Tampere University Hospital and University of Tampere, Tampere, Finland
| | - Henrik Zetterberg
- 7Department of Psychiatry and Neurochemistry, the Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden.,8Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden.,9U.K. Dementia Research Institute at University College London, London, United Kingdom.,10Department of Neurodegenerative Disease, University College London Institute of Neurology, Queen Square, London, United Kingdom
| | - Kaj Blennow
- 7Department of Psychiatry and Neurochemistry, the Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden.,8Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
| | - Teemu M Luoto
- 6Department of Neurosurgery, Tampere University Hospital and University of Tampere, Tampere, Finland
| |
Collapse
|
40
|
Posti JP, Takala RSK, Lagerstedt L, Dickens AM, Hossain I, Mohammadian M, Ala-Seppälä H, Frantzén J, van Gils M, Hutchinson PJ, Katila AJ, Maanpää HR, Menon DK, Newcombe VF, Tallus J, Hrusovsky K, Wilson DH, Gill J, Sanchez JC, Tenovuo O, Zetterberg H, Blennow K. Correlation of Blood Biomarkers and Biomarker Panels with Traumatic Findings on Computed Tomography after Traumatic Brain Injury. J Neurotrauma 2019; 36:2178-2189. [PMID: 30760178 DOI: 10.1089/neu.2018.6254] [Citation(s) in RCA: 57] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
The aim of the study was to examine the ability of eight protein biomarkers and their combinations in discriminating computed tomography (CT)-negative and CT-positive patients with traumatic brain injury (TBI), utilizing highly sensitive immunoassays in a well-characterized cohort. Blood samples were obtained from 160 patients with acute TBI within 24 h of admission. Levels of β-amyloid isoforms 1-40 (Aβ40) and 1-42 (Aβ42), glial fibrillary acidic protein (GFAP), heart fatty-acid binding protein (H-FABP), interleukin 10 (IL-10), neurofilament light (NF-L), S100 calcium-binding protein B (S100B), and tau were measured. Patients were divided into CT-negative (n = 65) and CT-positive (n = 95), and analyses were conducted separately for TBIs of all severities (Glasgow Coma Scale [GCS] score 3-15) and mild TBIs (mTBIs; GCS 13-15). NF-L, GFAP, and tau were the best in discriminating CT-negative and CT-positive patients, both in patients with mTBI and with all severities. In patients with all severities, area under the curve of the receiver operating characteristic (AUC) was 0.822, 0.817, and 0.781 for GFAP, NF-L, and tau, respectively. In patients with mTBI, AUC was 0.720, 0.689, and 0.676, for GFAP, tau, and NF-L, respectively. The best panel of three biomarkers for discriminating CT-negative and CT-positive patients in the group of all severities was a combination of GFAP+H-FABP+IL-10, with a sensitivity of 100% and specificity of 38.5%. In patients with mTBI, the best panel of three biomarkers was H-FABP+S100B+tau, with a sensitivity of 100% and specificity of 46.4%. Panels of biomarkers outperform individual biomarkers in separating CT-negative and CT-positive patients. Panels consisted mainly of different biomarkers than those that performed best as an individual biomarker.
Collapse
Affiliation(s)
- Jussi P Posti
- 1 Department of Neurosurgery, Turku University Hospital, Turku, Finland.,2 Turku Brain Injury Center, Turku University Hospital, Turku, Finland.,3 Department of Neurology, University of Turku, Turku, Finland
| | - Riikka S K Takala
- 2 Turku Brain Injury Center, Turku University Hospital, Turku, Finland.,4 Perioperative Services, Intensive Care Medicine and Pain Management, Turku University Hospital and University of Turku, Finland
| | - Linnéa Lagerstedt
- 5 Department of Specialities of Internal Medicine, University of Geneva, Geneva, Switzerland
| | - Alex M Dickens
- 6 Turku Center for Biotechnology, University of Turku, Turku, Finland
| | - Iftakher Hossain
- 1 Department of Neurosurgery, Turku University Hospital, Turku, Finland.,2 Turku Brain Injury Center, Turku University Hospital, Turku, Finland.,3 Department of Neurology, University of Turku, Turku, Finland
| | - Mehrbod Mohammadian
- 2 Turku Brain Injury Center, Turku University Hospital, Turku, Finland.,3 Department of Neurology, University of Turku, Turku, Finland
| | - Henna Ala-Seppälä
- 2 Turku Brain Injury Center, Turku University Hospital, Turku, Finland.,3 Department of Neurology, University of Turku, Turku, Finland
| | - Janek Frantzén
- 1 Department of Neurosurgery, Turku University Hospital, Turku, Finland.,3 Department of Neurology, University of Turku, Turku, Finland
| | - Mark van Gils
- 7 VTT Technical Research Center of Finland Ltd., Tampere, Finland
| | - Peter J Hutchinson
- 8 Department of Clinical Neurosciences, University of Cambridge, Addenbrooke's Hospital, Cambridge, United Kingdom
| | - Ari J Katila
- 2 Turku Brain Injury Center, Turku University Hospital, Turku, Finland.,5 Department of Specialities of Internal Medicine, University of Geneva, Geneva, Switzerland
| | - Henna-Riikka Maanpää
- 1 Department of Neurosurgery, Turku University Hospital, Turku, Finland.,2 Turku Brain Injury Center, Turku University Hospital, Turku, Finland.,3 Department of Neurology, University of Turku, Turku, Finland
| | - David K Menon
- 9 Division of Anesthesia, University of Cambridge, Addenbrooke's Hospital, Cambridge, United Kingdom
| | - Virginia F Newcombe
- 9 Division of Anesthesia, University of Cambridge, Addenbrooke's Hospital, Cambridge, United Kingdom
| | - Jussi Tallus
- 2 Turku Brain Injury Center, Turku University Hospital, Turku, Finland.,3 Department of Neurology, University of Turku, Turku, Finland.,10 Department of Radiology, Turku University Hospital, Turku, Finland
| | | | | | - Jessica Gill
- 12 National Institute of Nursing Research, National Institutes of Health, Bethesda, Maryland
| | - Jean-Charles Sanchez
- 5 Department of Specialities of Internal Medicine, University of Geneva, Geneva, Switzerland
| | - Olli Tenovuo
- 2 Turku Brain Injury Center, Turku University Hospital, Turku, Finland.,3 Department of Neurology, University of Turku, Turku, Finland
| | - Henrik Zetterberg
- 13 Department of Psychiatry and Neurochemistry, Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden.,14 Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden.,15 Department of Neurodegenerative Disease, University College London, London, United Kingdom.,16 UK Dementia Research Institute at UCL, University College London, London, United Kingdom
| | - Kaj Blennow
- 13 Department of Psychiatry and Neurochemistry, Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden.,14 Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
| |
Collapse
|
41
|
Rubenstein R, Sharma DR, Chang B, Oumata N, Cam M, Vaucelle L, Lindberg MF, Chiu A, Wisniewski T, Wang KKW, Meijer L. Novel Mouse Tauopathy Model for Repetitive Mild Traumatic Brain Injury: Evaluation of Long-Term Effects on Cognition and Biomarker Levels After Therapeutic Inhibition of Tau Phosphorylation. Front Neurol 2019; 10:124. [PMID: 30915013 PMCID: PMC6421297 DOI: 10.3389/fneur.2019.00124] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2018] [Accepted: 01/30/2019] [Indexed: 12/20/2022] Open
Abstract
Traumatic brain injury (TBI) is a risk factor for a group of neurodegenerative diseases termed tauopathies, which includes Alzheimer's disease and chronic traumatic encephalopathy (CTE). Although TBI is stratified by impact severity as either mild (m), moderate or severe, mTBI is the most common and the most difficult to diagnose. Tauopathies are pathologically related by the accumulation of hyperphosphorylated tau (P-tau) and increased total tau (T-tau). Here we describe: (i) a novel human tau-expressing transgenic mouse model, TghTau/PS1, to study repetitive mild closed head injury (rmCHI), (ii) quantitative comparison of T-tau and P-tau from brain and plasma in TghTau/PS1 mice over a 12 month period following rmCHI (and sham), (iii) the usefulness of P-tau as an early- and late-stage blood-based biochemical biomarker for rmCHI, (iii) the influence of kinase-targeted therapeutic intervention on rmCHI-associated cognitive deficits using a combination of lithium chloride (LiCl) and R-roscovitine (ros), and (iv) correlation of behavioral and cognitive changes with concentrations of the brain and blood-based T-tau and P-tau. Compared to sham-treated mice, behavior changes and cognitive deficits of rmCHI-treated TghTau/PS1 mice correlated with increases in both cortex and plasma T-tau and P-tau levels over 12 months. In addition, T-tau, but more predominantly P-tau, levels were significantly reduced in the cortex and plasma by LiCl + ros approaching the biomarker levels in sham and drug-treated sham mice (the drugs had only modest effects on the T-tau and P-tau levels in sham mice) throughout the 12 month study period. Furthermore, although we also observed a reversal of the abnormal behavior and cognitive deficits in the drug-treated rmCHI mice (compared to the untreated rmCHI mice) throughout the time course, these drug-treated effects were most pronounced up until 10 and 12 months where the abnormal behavior and cognition deficits began to gradually increase. These studies describe: (a) a translational relevant animal model for TBI-linked tauopathies, and (b) utilization of T-tau and P-tau as rmCHI biomarkers in plasma to monitor novel therapeutic strategies and treatment regimens for these neurodegenerative diseases.
Collapse
Affiliation(s)
- Richard Rubenstein
- Laboratory of Neurodegenerative Diseases and CNS Biomarker Discovery, Departments of Neurology and Physiology/Pharmacology, SUNY Downstate Medical Center, Brooklyn, NY, United States
| | - Deep R Sharma
- Laboratory of Neurodegenerative Diseases and CNS Biomarker Discovery, Departments of Neurology and Physiology/Pharmacology, SUNY Downstate Medical Center, Brooklyn, NY, United States
| | - Binggong Chang
- Laboratory of Neurodegenerative Diseases and CNS Biomarker Discovery, Departments of Neurology and Physiology/Pharmacology, SUNY Downstate Medical Center, Brooklyn, NY, United States
| | - Nassima Oumata
- ManRos Therapeutics, Centre de Perharidy, Roscoff, France
| | - Morgane Cam
- ManRos Therapeutics, Centre de Perharidy, Roscoff, France
| | - Lise Vaucelle
- ManRos Therapeutics, Centre de Perharidy, Roscoff, France
| | | | - Allen Chiu
- Laboratory of Neurodegenerative Diseases and CNS Biomarker Discovery, Departments of Neurology and Physiology/Pharmacology, SUNY Downstate Medical Center, Brooklyn, NY, United States
| | - Thomas Wisniewski
- Center for Cognitive Neurology and Departments of Neurology, Pathology and Psychiatry, New York University School of Medicine, New York, NY, United States
| | - Kevin K W Wang
- Program for Neurotrauma, Neuroproteomics and Biomarker Research, Departments of Emergency Medicine, Psychiatry and Neuroscience, University of Florida, Gainesville, FL, United States
| | - Laurent Meijer
- ManRos Therapeutics, Centre de Perharidy, Roscoff, France
| |
Collapse
|
42
|
Martinez BI, Stabenfeldt SE. Current trends in biomarker discovery and analysis tools for traumatic brain injury. J Biol Eng 2019; 13:16. [PMID: 30828380 PMCID: PMC6381710 DOI: 10.1186/s13036-019-0145-8] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2018] [Accepted: 02/06/2019] [Indexed: 12/13/2022] Open
Abstract
Traumatic brain injury (TBI) affects 1.7 million people in the United States each year, causing lifelong functional deficits in cognition and behavior. The complex pathophysiology of neural injury is a primary barrier to developing sensitive and specific diagnostic tools, which consequentially has a detrimental effect on treatment regimens. Biomarkers of other diseases (e.g. cancer) have provided critical insight into disease emergence and progression that lend to developing powerful clinical tools for intervention. Therefore, the biomarker discovery field has recently focused on TBI and made substantial advancements to characterize markers with promise of transforming TBI patient diagnostics and care. This review focuses on these key advances in neural injury biomarkers discovery, including novel approaches spanning from omics-based approaches to imaging and machine learning as well as the evolution of established techniques.
Collapse
Affiliation(s)
- Briana I. Martinez
- School of Life Sciences, Arizona State University, Tempe, AZ USA
- School of Biological and Health Systems Engineering, Ira A. Fulton School of Engineering, Arizona State University, PO Box 879709, Tempe, AZ 85287-9709 USA
| | - Sarah E. Stabenfeldt
- School of Biological and Health Systems Engineering, Ira A. Fulton School of Engineering, Arizona State University, PO Box 879709, Tempe, AZ 85287-9709 USA
| |
Collapse
|
43
|
Ojo JO, Crynen G, Reed JM, Ajoy R, Vallabhaneni P, Algamal M, Leary P, Rafi NG, Mouzon B, Mullan M, Crawford F. Unbiased Proteomic Approach Identifies Unique and Coincidental Plasma Biomarkers in Repetitive mTBI and AD Pathogenesis. Front Aging Neurosci 2018; 10:405. [PMID: 30618712 PMCID: PMC6305374 DOI: 10.3389/fnagi.2018.00405] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2018] [Accepted: 11/26/2018] [Indexed: 12/14/2022] Open
Abstract
The relationship between repetitive mild traumatic brain injury (r-mTBI) and Alzheimer's disease (AD) is well-recognized. However, the precise nature of how r-mTBI leads to or precipitates AD pathogenesis is currently not understood. Plasma biomarkers potentially provide non-invasive tools for detecting neurological changes in the brain, and can reveal overlaps between long-term consequences of r-mTBI and AD. In this study we address this by generating time-dependent molecular profiles of response to r-mTBI and AD pathogenesis in mouse models using unbiased proteomic analyses. To model AD, we used the well-validated hTau and PSAPP(APP/PS1) mouse models that develop age-related tau and amyloid pathological features, respectively, and our well-established model of r-mTBI in C57BL/6 mice. Plasma were collected at different ages (3, 9, and 15 months-old for hTau and PSAPP mice), encompassing pre-, peri- and post-"onset" of the cognitive and neuropathological phenotypes, or at different timepoints after r-mTBI (24 h, 3, 6, 9, and 12 months post-injury). Liquid chromatography/mass spectrometry (LC-MS) approaches coupled with Tandem Mass Tag labeling technology were applied to develop molecular profiles of protein species that were significantly differentially expressed as a consequence of mTBI or AD. Mixed model ANOVA after Benjamini-Hochberg correction, and a stringent cut-off identified 31 proteins significantly changing in r-mTBI groups over time and, when compared with changes over time in sham mice, 13 of these were unique to the injured mice. The canonical pathways predicted to be modulated by these changes were LXR/RXR activation, production of nitric oxide and reactive oxygen species and complement systems. We identified 18 proteins significantly changing in PSAPP mice and 19 proteins in hTau mice compared to their wild-type littermates with aging. Six proteins were found to be significantly regulated in all three models, i.e., r-mTBI, hTau, and PSAPP mice compared to their controls. The top canonical pathways coincidently changing in all three models were LXR/RXR activation, and production of nitric oxide and reactive oxygen species. This work suggests potential biomarkers for TBI and AD pathogenesis and for the overlap between these two, and warrant targeted investigation in human populations. Data are available via ProteomeXchange with identifier PXD010664.
Collapse
Affiliation(s)
- Joseph O. Ojo
- Experimental Neuropathology and Proteomic Laboratory, Roskamp Institute, Sarasota, FL, United States
- James A. Haley Veterans’ Hospital, Tampa, FL, United States
- Life, Health and Chemical Sciences, The Open University, Milton Keynes, United Kingdom
| | - Gogce Crynen
- Experimental Neuropathology and Proteomic Laboratory, Roskamp Institute, Sarasota, FL, United States
- Life, Health and Chemical Sciences, The Open University, Milton Keynes, United Kingdom
| | - Jon M. Reed
- Experimental Neuropathology and Proteomic Laboratory, Roskamp Institute, Sarasota, FL, United States
- Boehringer Ingelheim Pharmaceuticals, Inc., Ridgefield, CT, United States
| | - Rosa Ajoy
- Experimental Neuropathology and Proteomic Laboratory, Roskamp Institute, Sarasota, FL, United States
| | - Prashanthi Vallabhaneni
- Experimental Neuropathology and Proteomic Laboratory, Roskamp Institute, Sarasota, FL, United States
| | - Moustafa Algamal
- Experimental Neuropathology and Proteomic Laboratory, Roskamp Institute, Sarasota, FL, United States
- Life, Health and Chemical Sciences, The Open University, Milton Keynes, United Kingdom
| | - Paige Leary
- Experimental Neuropathology and Proteomic Laboratory, Roskamp Institute, Sarasota, FL, United States
| | - Naomi G. Rafi
- Experimental Neuropathology and Proteomic Laboratory, Roskamp Institute, Sarasota, FL, United States
| | - Benoit Mouzon
- Experimental Neuropathology and Proteomic Laboratory, Roskamp Institute, Sarasota, FL, United States
- James A. Haley Veterans’ Hospital, Tampa, FL, United States
- Life, Health and Chemical Sciences, The Open University, Milton Keynes, United Kingdom
| | - Michael Mullan
- Experimental Neuropathology and Proteomic Laboratory, Roskamp Institute, Sarasota, FL, United States
- Life, Health and Chemical Sciences, The Open University, Milton Keynes, United Kingdom
| | - Fiona Crawford
- Experimental Neuropathology and Proteomic Laboratory, Roskamp Institute, Sarasota, FL, United States
- James A. Haley Veterans’ Hospital, Tampa, FL, United States
- Life, Health and Chemical Sciences, The Open University, Milton Keynes, United Kingdom
| |
Collapse
|
44
|
Metzger RR, Sheng X, Niedzwecki CM, Bennett KS, Morita DC, Zielinski B, Schober ME. Temporal response profiles of serum ubiquitin C-terminal hydrolase-L1 and the 145-kDa alpha II-spectrin breakdown product after severe traumatic brain injury in children. J Neurosurg Pediatr 2018; 22:369-374. [PMID: 29957142 DOI: 10.3171/2018.4.peds17593] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
OBJECTIVE Traumatic brain injury (TBI) is the leading cause of acquired disability among children. Brain injury biomarkers may serve as useful diagnostic and prognostic indicators for TBI. Levels of ubiquitin C-terminal hydrolase-L1 (UCH-L1) and the 145-kDa alpha II-spectrin breakdown product (SBDP-145) correlate with outcome in adults after severe TBI. The authors conducted a pilot study of these biomarkers in children after severe TBI to inform future research exploring their utility in this population. METHODS The levels of UCH-L1 and SBDP-145 were measured in serum, and UCH-L1 in CSF from pediatric patients after severe TBI over 5 days after injury. Both biomarkers were also measured in age-matched control serum and CSF. RESULTS Adequate numbers of samples were obtained in serum, but not CSF, to assess biomarker temporal response profiles. Using patients with samples from all time points, UCH-L1 levels increased rapidly and transiently, peaking at 12 hours after injury. SBDP-145 levels showed a more gradual and sustained response, peaking at 48 hours. The median serum UCH-L1 concentration was greater in patients with TBI than in controls (median [IQR] = 361 [187, 1330] vs 147 [50, 241] pg/ml, respectively; p < 0.001). Receiver operating characteristic (ROC) analysis revealed an AUC of 0.77. Similarly, serum SBDP-145 was greater in children with TBI than in controls (median [IQR] = 172 [124, 257] vs 69 [40, 99] pg/ml, respectively; p < 0.001), with an ROC AUC of 0.85. When only time points of peak levels were used for ROC analysis, the discriminability of each serum biomarker increased (AUC for UCH-L1 at 12 hours = 1.0 and for SBDP-145 at 48 hours = 0.91). Serum and CSF UCH-L1 levels correlated well in patients with TBI (r = 0.70, p < 0.001). CONCLUSIONS Findings from this exploratory study reveal robust increases of UCH-L1 and SBDP-145 in serum and UCH-L1 in CSF obtained from children after severe TBI. In addition, important temporal profile differences were found between these biomarkers that can help guide optimal time point selection for future investigations of their potential to characterize injury or predict outcomes after pediatric TBI.
Collapse
Affiliation(s)
| | | | - Christian M Niedzwecki
- 3Department of Physical Medicine and Rehabilitation, University of Utah, Salt Lake City, Utah
| | | | | | | | | |
Collapse
|
45
|
Dickens AM, Posti JP, Takala RSK, Ala-Seppälä H, Mattila I, Coles JP, Frantzén J, Hutchinson PJ, Katila AJ, Kyllönen A, Maanpää HR, Newcombe V, Outtrim J, Tallus J, Carpenter KLH, Menon DK, Hyötyläinen T, Tenovuo O, Orešic M. Serum Metabolites Associated with Computed Tomography Findings after Traumatic Brain Injury. J Neurotrauma 2018; 35:2673-2683. [PMID: 29947291 DOI: 10.1089/neu.2017.5272] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
There is a need to rapidly detect patients with traumatic brain injury (TBI) who require head computed tomography (CT). Given the energy crisis in the brain following TBI, we hypothesized that serum metabolomics would be a useful tool for developing a set of biomarkers to determine the need for CT and to distinguish among different types of injuries observed. Logistical regression models using metabolite data from the discovery cohort (n = 144, Turku, Finland) were used to distinguish between patients with traumatic intracranial findings and those with negative findings on head CT. The resultant models were then tested in the validation cohort (n = 66, Cambridge, United Kingdom). The levels of glial fibrillary acidic protein and ubiquitin C-terminal hydrolase-L1 were also quantified in the serum from the same patients. Despite there being significant differences in the protein biomarkers in patients with TBI, the model that determined the need for a CT scan validated poorly (area under the curve [AUC] = 0.64: Cambridge patients). However, using a combination of six metabolites (two amino acids, three sugar derivatives, and one ketoacid) it was possible to discriminate patients with intracranial abnormalities on CT and patients with a normal CT (AUC = 0.77 in Turku patients and AUC = 0.73 in Cambridge patients). Further, a combination of three metabolites could distinguish between diffuse brain injuries and mass lesions (AUC = 0.87 in Turku patients and AUC = 0.68 in Cambridge patients). This study identifies a set of validated serum polar metabolites, which associate with the need for a CT scan. Additionally, serum metabolites can also predict the nature of the brain injury. These metabolite markers may prevent unnecessary CT scans, thus reducing the cost of diagnostics and radiation load.
Collapse
Affiliation(s)
- Alex M Dickens
- 1 Turku Centre for Biotechnology, University of Turku , Turku, Finland
| | - Jussi P Posti
- 2 Turku Brain Injury Centre, Turku University Hospital , Turku, Finland .,3 Department of Neurology, University of Turku , Turku, Finland .,4 Division of Clinical Neurosciences, Department of Neurosurgery, Turku University Hospital , Turku, Finland
| | - Riikka S K Takala
- 5 Perioperative Services, Intensive Care Medicine and Pain Management, Turku University Hospital and University of Turku , Turku, Finland
| | | | - Ismo Mattila
- 6 Steno Diabetes Center Copenhagen , Gentofte, Denmark
| | - Jonathan P Coles
- 7 Division of Anaesthesia, Department of Medicine, University of Cambridge , Addenbrooke's Hospital, Cambridge, United Kingdom
| | - Janek Frantzén
- 2 Turku Brain Injury Centre, Turku University Hospital , Turku, Finland .,3 Department of Neurology, University of Turku , Turku, Finland .,4 Division of Clinical Neurosciences, Department of Neurosurgery, Turku University Hospital , Turku, Finland
| | - Peter J Hutchinson
- 8 Division of Neurosurgery, Department of Clinical Neurosciences, University of Cambridge , Addenbrooke's Hospital, Cambridge, United Kingdom
| | - Ari J Katila
- 5 Perioperative Services, Intensive Care Medicine and Pain Management, Turku University Hospital and University of Turku , Turku, Finland
| | - Anna Kyllönen
- 3 Department of Neurology, University of Turku , Turku, Finland
| | | | - Virginia Newcombe
- 7 Division of Anaesthesia, Department of Medicine, University of Cambridge , Addenbrooke's Hospital, Cambridge, United Kingdom
| | - Joanne Outtrim
- 7 Division of Anaesthesia, Department of Medicine, University of Cambridge , Addenbrooke's Hospital, Cambridge, United Kingdom
| | - Jussi Tallus
- 3 Department of Neurology, University of Turku , Turku, Finland
| | - Keri L H Carpenter
- 8 Division of Neurosurgery, Department of Clinical Neurosciences, University of Cambridge , Addenbrooke's Hospital, Cambridge, United Kingdom
| | - David K Menon
- 7 Division of Anaesthesia, Department of Medicine, University of Cambridge , Addenbrooke's Hospital, Cambridge, United Kingdom
| | | | - Olli Tenovuo
- 2 Turku Brain Injury Centre, Turku University Hospital , Turku, Finland .,3 Department of Neurology, University of Turku , Turku, Finland
| | - Matej Orešic
- 1 Turku Centre for Biotechnology, University of Turku , Turku, Finland .,10 Schools of Medical Science, Örebro University , Örebro, Sweden
| |
Collapse
|
46
|
Combining H-FABP and GFAP increases the capacity to differentiate between CT-positive and CT-negative patients with mild traumatic brain injury. PLoS One 2018; 13:e0200394. [PMID: 29985933 PMCID: PMC6037378 DOI: 10.1371/journal.pone.0200394] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2018] [Accepted: 06/25/2018] [Indexed: 12/21/2022] Open
Abstract
Mild traumatic brain injury (mTBI) patients may have trauma-induced brain lesions detectable using CT scans. However, most patients will be CT-negative. There is thus a need for an additional tool to detect patients at risk. Single blood biomarkers, such as S100B and GFAP, have been widely studied in mTBI patients, but to date, none seems to perform well enough. In many different diseases, combining several biomarkers into panels has become increasingly interesting for diagnoses and to enhance classification performance. The present study evaluated 13 proteins individually-H-FABP, MMP-1, MMP-3, MMP-9, VCAM, ICAM, SAA, CRP, GSTP, NKDA, PRDX1, DJ-1 and IL-10-for their capacity to differentiate between patients with and without a brain lesion according to CT results. The best performing proteins were then compared and combined with the S100B and GFAP proteins into a CT-scan triage panel. Patients diagnosed with mTBI, with a Glasgow Coma Scale score of 15 and one additional clinical symptom were enrolled at three different European sites. A blood sample was collected at hospital admission, and a CT scan was performed. Patients were divided into two two-centre cohorts and further dichotomised into CT-positive and CT-negative groups for statistical analysis. Single markers and panels were evaluated using Cohort 1. Four proteins-H-FABP, IL-10, S100B and GFAP-showed significantly higher levels in CT-positive patients. The best-performing biomarker was H-FABP, with a specificity of 32% (95% CI 23-40) and sensitivity reaching 100%. The best-performing two-marker panel for Cohort 1, subsequently validated in Cohort 2, was a combination of H-FABP and GFAP, enhancing specificity to 46% (95% CI 36-55). When adding IL-10 to this panel, specificity reached 52% (95% CI 43-61) with 100% sensitivity. These results showed that proteins combined into panels could be used to efficiently classify CT-positive and CT-negative mTBI patients.
Collapse
|
47
|
Singh GP, Nigam R, Tomar GS, Monisha M, Bhoi SK, S A, Sengar K, Akula D, Panta P, Anindya R. Early and rapid detection of UCHL1 in the serum of brain-trauma patients: a novel gold nanoparticle-based method for diagnosing the severity of brain injury. Analyst 2018; 143:3366-3373. [PMID: 29893758 DOI: 10.1039/c8an00533h] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Abstract
The clinical diagnosis of traumatic brain injury (TBI) is based on neurological examination and neuro-imaging tools such as CT scanning and MRI. However, neurological examination at times may be confounded by consumption of alcohol or drugs and neuroimaging facilities may not be available at all centers. Human ubiquitin C-terminal hydrolase (UCHL1) is a well-accepted serum biomarker for severe TBI and can be used to detect the severity of a head injury. A reliable, rapid, cost effective, bedside and easy to perform method for the detection of UCHL1 is a pre-requisite for wide clinical applications of UCHL1 as a TBI biomarker. We developed a rapid detection method for UCHL1 using surface plasmon resonance of gold nanoparticles with a limit of detection (LOD) of 0.5 ng mL-1. It has a sensitivity and specificity of 100% each and meets an analytical precision similar to that of conventional sandwich ELISA but can be performed rapidly. Using this method we successfully detected UCHL1 in a cohort of 66 patients with TBI and were reliably able to distinguish mild TBI from moderate to severe TBI.
Collapse
Affiliation(s)
- Gyaninder Pal Singh
- Department of Neuroanaesthesiology and Critical Care, JPN Apex Trauma Centre, All India Institute of Medical Sciences (AIIMS), New Delhi-110029, India
| | | | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Ondruschka B, Sieber M, Kirsten H, Franke H, Dreßler J. Measurement of Cerebral Biomarkers Proving Traumatic Brain Injuries in Post-Mortem Body Fluids. J Neurotrauma 2018; 35:2044-2055. [PMID: 29732941 DOI: 10.1089/neu.2017.5441] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Until now, it is impossible to identify a fatal traumatic brain injury (TBI) before post-mortem radiological investigations or an autopsy take place. It would be preferable to have an additional diagnostic tool such as post-mortem biochemistry to get greater insight into the pathological pathways and survival times after sustaining TBI. Cerebrospinal fluid (CSF) and serum samples of 84 autopsy cases were collected from forensic autopsies with post-mortem intervals (PMI) of up to 148 h. The cases were categorized into a fatal TBI case group (n = 42) and non-TBI controls (n = 42). The values of glial fibrillary acidic protein (GFAP), brain-derived neurotrophic factor (BDNF), and neutrophil gelatinase-associated lipocalin (NGAL) were analyzed by means of quantitative chemiluminescent multiplex immunoassays. The main results indicate that the usage of liquid samples with good macroscopic quality is more relevant for meaningful biomarker analyses than the length of the PMI. All three proteins were shown to differentiate TBI fatalities from the controls in CSF. In serum, only GFAP could be shown to be able to identify TBI cases. This study is the first approach to measure the three proteins together in CSF and serum in autopsy cases. Determined threshold values may differentiate between fatal TBI and control cases. The presented results emphasize the possible use of post-mortem biochemistry as a supplemental tool in everyday forensic routine.
Collapse
Affiliation(s)
- Benjamin Ondruschka
- 1 Institute of Legal Medicine, Medical Faculty, University of Leipzig , Leipzig, Germany
| | - Monique Sieber
- 1 Institute of Legal Medicine, Medical Faculty, University of Leipzig , Leipzig, Germany
| | - Holger Kirsten
- 2 Institute for Medical Informatics, Statistics, and Epidemiology, Medical Faculty, University of Leipzig , Leipzig, Germany
| | - Heike Franke
- 3 Rudolf Boehm Institute of Pharmacology and Toxicology, Medical Faculty, University of Leipzig , Leipzig, Germany
| | - Jan Dreßler
- 1 Institute of Legal Medicine, Medical Faculty, University of Leipzig , Leipzig, Germany
| |
Collapse
|
49
|
Korley FK, Nikolian VC, Williams AM, Dennahy IS, Weykamp M, Alam HB. Valproic Acid Treatment Decreases Serum Glial Fibrillary Acidic Protein and Neurofilament Light Chain Levels in Swine Subjected to Traumatic Brain Injury. J Neurotrauma 2018; 35:1185-1191. [PMID: 29415612 DOI: 10.1089/neu.2017.5581] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
The primary aim of this study was to examine the effects of valproic acid (VPA) treatment on serum glial fibrillary acidic protein (GFAP) and neurofilament light chain (NF-L) levels. To achieve this aim, we obtained serum samples from: 1) 10 Yorkshire swine subjected to controlled cortical impact traumatic brain injury (CCI TBI) + polytrauma and randomized to receive either normal saline (NS) + VPA (n = 5) or NS alone (n = 5) and 2) five additional swine subjected to CCI TBI without polytrauma and treated with VPA. GFAP and NF-L levels were measured in samples obtained from baseline until 10 days post-injury using a digital immunoassay from Quanterix Corporation. We found that elevated GFAP and NF-L levels were first detected at 2 h post-injury; and peaked at 24 h and 72 h respectively. GFAP levels returned to baseline levels by Day 10, while NF-L remained elevated at Day 10. In TBI + polytrauma swine, the magnitude and duration of biomarker elevation, quantified by the area under the biomarker-concentration-versus-time curve during the first 10 days (AUC0-10days), was higher in the NS group, compared with the VPA group. For GFAP, the AUC0-10days was 45,535 (IQR: 35,741-105,711) and 22,837 (IQR: 8,082-46,627) for the NS and NS+VPA groups, respectively. For NF-L, the AUC0-10days was 43,073 (IQR: 18,739-120,794) and 4,475 (2,868-11,157) for the NS and NS+VPA groups, respectively. Twenty-four hour GFAP and NF-L levels had the strongest correlation with lesion size and time to normalization of behavior. Accordingly, we conclude that treatment with VPA results in significantly lower serum GFAP and NF-L levels. The time-point at which GFAP and NF-L levels have the strongest correlation with outcome is 24 h post-injury.
Collapse
Affiliation(s)
- Frederick K Korley
- 1 Department of Emergency Medicine, University of Michigan Medical School , Ann Arbor, Michigan
| | - Vahagn C Nikolian
- 2 Department of Surgery, University of Michigan Medical School , Ann Arbor, Michigan
| | - Aaron M Williams
- 2 Department of Surgery, University of Michigan Medical School , Ann Arbor, Michigan
| | - Isabel S Dennahy
- 2 Department of Surgery, University of Michigan Medical School , Ann Arbor, Michigan
| | | | - Hasan B Alam
- 2 Department of Surgery, University of Michigan Medical School , Ann Arbor, Michigan
| |
Collapse
|
50
|
Lagerstedt L, Egea-Guerrero JJ, Rodríguez-Rodríguez A, Bustamante A, Montaner J, El Rahal A, Andereggen E, Rinaldi L, Sarrafzadeh A, Schaller K, Sanchez JC. Early measurement of interleukin-10 predicts the absence of CT scan lesions in mild traumatic brain injury. PLoS One 2018; 13:e0193278. [PMID: 29466474 PMCID: PMC5821397 DOI: 10.1371/journal.pone.0193278] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2017] [Accepted: 02/07/2018] [Indexed: 12/26/2022] Open
Abstract
Traumatic brain injury is a common event where 70%-90% will be classified as mild TBI (mTBI). Among these, only 10% will have a brain lesion visible via CT scan. A triage biomarker would help clinicians to identify patients with mTBI who are at risk of developing a brain lesion and require a CT scan. The brain cells damaged by the shearing, tearing and stretching of a TBI event set off inflammation cascades. These cause altered concentrations of a high number of both pro-inflammatory and anti-inflammatory proteins. This study aimed to discover a novel diagnostic biomarker of mTBI by investigating a broad panel of inflammation biomarkers and their capacity to correctly identify CT-positive and CT-negative patients. Patients enrolled in this study had been diagnosed with mTBI, had a GCS score of 15 and suffered from at least one clinical symptom. There were nine patients in the discovery group, 45 for verification, and 133 mTBI patients from two different European sites in the validation cohort. All patients gave blood samples, underwent a CT scan and were dichotomised into CT-positive and CT-negative groups for statistical analyses. The ability of each protein to classify patients was evaluated with sensitivity set at 100%. Three of the 92 inflammation proteins screened-MCP-1, MIP-1alpha and IL-10 -were further investigated in the verification group, and at 100% sensitivity their specificities reached 7%, 0% and 31%, respectively. IL-10 was validated on a larger cohort in comparison to the most studied mTBI diagnostic triage protein to date, S100B. Levels of both proteins were significantly higher in CT-positive than in CT-negative patients (p < 0.001). S100B's specificity at 100% sensitivity was 18% (95% CI 10.8-25.2), whereas IL-10 reached a specificity of 27% (95% CI 18.9-35.1). These results showed that IL-10 might be an interesting and clinically useful diagnostic tool, capable of differentiating between CT-positive and CT-negative mTBI patients.
Collapse
Affiliation(s)
- Linnéa Lagerstedt
- Department of Human Protein Sciences, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | | | | | - Alejandro Bustamante
- Neurovascular Research Laboratory, Vall d’Hebron Institute of Research (VHIR), Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Joan Montaner
- Neurovascular Research Laboratory, Vall d’Hebron Institute of Research (VHIR), Universitat Autònoma de Barcelona, Barcelona, Spain
- Stroke Research Programme, IBiS/Hospital Universitario Virgen del Rocío/CSIC/University of Seville, Seville, Spain
- Department of Neurology, Hospital Universitario Virgen Macarena, Seville, Spain
| | - Amir El Rahal
- Division of Neurosurgery, Geneva Neuroscience Center, Department of Clinical Neurosciences, Geneva University Hospitals, Geneva, Switzerland
| | - Elisabeth Andereggen
- Emergency Center, Geneva University Hospitals, Geneva, Switzerland
- Department of Surgery, Geneva University Hospitals, Geneva, Switzerland
| | - Lara Rinaldi
- Emergency Center, Geneva University Hospitals, Geneva, Switzerland
| | - Asita Sarrafzadeh
- Division of Neurosurgery, Geneva Neuroscience Center, Department of Clinical Neurosciences, Geneva University Hospitals, Geneva, Switzerland
| | - Karl Schaller
- Division of Neurosurgery, Geneva Neuroscience Center, Department of Clinical Neurosciences, Geneva University Hospitals, Geneva, Switzerland
| | - Jean-Charles Sanchez
- Department of Human Protein Sciences, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| |
Collapse
|