1
|
Bazarian JJ, Zetterberg H, Buki A, Dengler BA, Diaz-Arrastia R, Korley FK, Lazarus R, Meier TB, Mondello S, Moritz K, Okonkwo DO, Papa L, Phillips JB, Posti JP, Puccio AM, Sloley S, Steyerberg E, Wang KK, Awwad HO, Dams-O'Connor K, Doperalski A, Maas AIR, McCrea MA, Umoh N, Manley GT. Blood-Based Biomarkers for Improved Characterization of Traumatic Brain Injury: Recommendations from the 2024 National Institute for Neurological Disorders and Stroke Traumatic Brain Injury Classification and Nomenclature Initiative Blood-Based Biomarkers Working Group. J Neurotrauma 2025. [PMID: 40393505 DOI: 10.1089/neu.2024.0581] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/22/2025] Open
Abstract
A 2022 report by the National Academies of Sciences, Engineering, and Medicine called for a Traumatic Brain Injury (TBI) Classification Workshop by the National Institutes of Health (NIH) to develop a more precise, evidence-based classification system. The workshop aimed to revise the Glasgow Coma Scale-based system by incorporating neuroimaging and validated blood biomarker tests. In December 2022, the National Institute for Neurological Disorders and Stroke formed six working groups of TBI experts to make recommendations for this revision. This report presents the findings and recommendations from the blood-based biomarker (BBM) working group, including feedback from the workshop and subsequent public review. The application of BBMs in a TBI classification system has potential to allow for a more adaptable and nuanced approach to triage, diagnosis, prognosis, and treatment. Current evidence supports the use of glial fibrillary acidic protein (GFAP), ubiquitin C-terminal hydrolase L1, and S100B calcium-binding protein (S100B) to assist in reclassification of TBI at acute time points (0-24 h) primarily in emergency department settings, while neurofilament light chain (NfL), GFAP, and S100B have utility at subacute time points (1-30 days) in-hospital and intensive care unit settings. Blood levels of these biomarkers reflect the extent of structural brain injury in TBI and may be useful for describing the extent of structural brain injury in a classification system. While there is insufficient evidence to support a role for BBMs at chronic time points (>30 days), emerging evidence suggests that NfL and phosphorylated tau may have a potential future role in this regard. For inclusion in a revised TBI classification system, BBM assays must have appropriate age- and sex-specific reference ranges, be harmonized across platforms, and achieve high analytical precision, including accuracy, linearity, detection limits, selectivity, recovery, reproducibility, and stability. Improving transparency in BBM assay development can be achieved through large-scale data sharing of methods and results. Future research should focus on methods for promoting clinical adoption of BBM results, correlating BBMs with advanced neuroimaging, and on discovering new biomarkers for improved diagnosis and prognosis.
Collapse
Affiliation(s)
- Jeffrey J Bazarian
- Departments of Emergency Medicine and Neurology, University of Rochester School of Medicine and Dentistry, Rochester, New York, USA
| | | | | | - Bradley A Dengler
- Military Traumatic Brain Injury Initiative, Uniformed Services University of the Health Sciences, Bethesda, Maryland, 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
| | - Rachel Lazarus
- American Association of Retired Persons, Washington District of Columbia, USA
| | - Timothy B Meier
- Department of Neurosurgery, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| | | | - Kasey Moritz
- U.S. Army Medical Research and Development Command, Combat Casualty Care Research Program, Fort Detrick, Maryland, USA
| | - David O Okonkwo
- Department of Neurological Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Linda Papa
- Orlando Health Orlando Regional Medical Center, Orlando, Florida, USA
| | - James B Phillips
- U.S. Army Medical Research and Development Command, Combat Casualty Care Research Program, Fort Detrick, Maryland, USA
| | - Jussi P Posti
- Neurocenter, Department of Neurosurgery and Turku Brain Injury Center, Turku University Hospital and University of Turku, Turku, Finland
| | - Ava M Puccio
- Department of Neurological Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Stephanie Sloley
- TBI Center of Excellence, Defense Health Agency, Silver Spring, Maryland, USA
| | | | - Kevin K Wang
- Center for Neurotrauma, Multiomics & Biomarkers, Neuroscience institute, Morehouse School of Medicine, Atlanta, Georgia, USA
- Center for Visual and Neurocognitive Rehabilitation, Atlanta VA Health Care System, Decatur, Georgia, USA
| | - Hibah O Awwad
- Division of Neuroscience, National Institute of Neurological Disorders and Stroke, Bethesda, Maryland, USA
| | - Kristen Dams-O'Connor
- Department of Rehabilitation and Human Performance, Icahn School of Medicine, Mount Sinai, New York, New York, USA
- Department of Neurology, Icahn School of Medicine, Mount Sinai, New York, New York, USA
| | - Adele Doperalski
- Department of Neurology, Icahn School of Medicine, Mount Sinai, New York, New York, USA
| | - 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
| | - Michael A McCrea
- Department of Neurosurgery, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| | - Nsini Umoh
- Department of Neurology, Icahn School of Medicine, Mount Sinai, New York, New York, USA
| | - Geoffrey T Manley
- Neurological Surgery, University of California San Francisco, San Francisco, California, USA
| |
Collapse
|
2
|
Whitehouse D, Mikolić A, Czeiter E, Richter S, Buki A, Wang KK, Steyerberg E, Maas A, Menon D, Lecky F, Newcombe V. Serum Biomarkers as Adjuncts to the National Institute for Health and Care Excellence Head Injury Guidelines (NG232, 2023) When Selecting Patients with Traumatic Brain Injury for Computed Tomography: A Collaborative European NeuroTrauma Effectiveness Research in Traumatic Brain Injury Study. J Neurotrauma 2025. [PMID: 40329809 DOI: 10.1089/neu.2024.0276] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/08/2025] Open
Abstract
This article explores the diagnostic performance of a panel of six biomarkers (glial fibrillary acidic protein [GFAP], neurofilament light [NFL], neuron-specific enolase [NSE], S100 calcium-binding protein B [S100B], total tau [t-tau], and ubiquitin C-terminal hydrolase L1 [UCH-L1]) in the context of the "2023 UK National Institute for Health and Care Excellence (NICE) Head Injury: Assessment and early management (NG232)" guideline. Emphasis is placed on subjects where clinical equipoise remains concerning the decision for head computed tomography (CT), medium-risk subjects. All adult subjects from the Collaborative European NeuroTrauma Effectiveness Research in Traumatic Brain Injury (CENTER-TBI) dataset with a complete biomarker profile and interpretable CT scan within 24 h of injury were classified as high, medium, and low-risk according to the NICE NG232 Clinical Decision Rule (CDR) for CT head imaging following head injury. In subjects classified as medium-risk, the area under the receiver operating characteristic curve (AUC) was used to assess the diagnostic performance of biomarkers to identify those with (1) CT abnormality or (2) potential neurosurgical lesion, with CT considered the gold standard diagnosis. A time-to-biomarker sub-analysis was performed in subjects with a time from injury to sampling within 6 h, in keeping with current clinical usage of biomarkers. Among 1979 CENTER-TBI participants with sufficient clinical information to facilitate classification, 385 subjects were classified as medium-risk. Biomarker concentrations were significantly higher in those with traumatic CT abnormalities as compared with those without for all biomarkers aside from NSE (all p < 0.05). When sampled within 24 h of injury, GFAP demonstrated the best diagnostic performance for CT abnormality (AUC 0.81 [0.77-0.86]), with NFL, t-tau, and UCH-L1 showing moderate performance. At a threshold to provide a 95% sensitivity, GFAP, NFL, t-tau, and UCH-L1 demonstrated specificities ranging from 18% to 33% corresponding to a potential reduction of total CT images performed in these subjects by 14-23%. S100B and UCH-L1 showed improved performance when biomarker sampling time was limited to 6 h following injury. In intoxicated subjects with a persistent Glasgow Coma Score of 13-14, biomarker levels were significantly higher in subjects with CT abnormality as compared with those without. In conclusion, serum biomarkers demonstrate potential for the reduction in CT scan requirements in those classified as medium-risk in reference to the NG232 CDR criteria. These results highlight a need for further prospective studies on the use of diagnostic TBI biomarkers in current emergency medicine practice, with future consideration given to the integration of biomarkers in the NICE NG232 head injury guidelines.
Collapse
Affiliation(s)
- Daniel Whitehouse
- Division of Anaesthesia, Department of Medicine, Addenbrooke's Hospital, University of Cambridge, Cambridge, United Kingdom
| | - Ana Mikolić
- Department of Psychology, University of British Columbia, Vancouver, British Columbia, Canada
- Rehabilitation Research Program, GF Strong Rehabilitation Centre, Vancouver, British Columbia, Canada
| | - Endre Czeiter
- Department of Neurosurgery, Medical School, University of Pecs, Pecs, Hungary
- Neurotrauma Research Group, Szentagothai Research Centre, University of Pecs, Pecs, Hungary
| | - Sophie Richter
- Division of Anaesthesia, Department of Medicine, Addenbrooke's Hospital, University of Cambridge, Cambridge, United Kingdom
| | - Andras Buki
- Faculty of Medicine and Health, Department of Neurosurgery, Örebro University, Örebro, Sweden
| | - Kevin K Wang
- Department of Neurobiology, Center for Neurotrauma, Multiomics & Biomarkers (CNMB) Neuroscience institute, Morehouse School of Medicine (MSM), Atlanta, Georgia, USA
- Program for Neurotrauma, Neuroproteomics and Biomarker Research, Departments of Emergency Medicine, Psychiatry and Neuroscience, McKnight Brain Institute, University of Florida, Gainesville, Florida, USA
| | - Ewout Steyerberg
- Department of Biomedical Data Sciences, Leiden University Medical Center, Leiden, Netherlands
| | - Andrew Maas
- Department of Neurosurgery, Antwerp University Hospital, Edegem, Belgium
- Faculty of Medicine and Health Science, Department of Translational Neuroscience, University of Antwerp, Antwerp, Belgium
| | - David Menon
- Division of Anaesthesia, Department of Medicine, Addenbrooke's Hospital, University of Cambridge, Cambridge, United Kingdom
| | - Fiona Lecky
- Centre for Urgent and emergency care REsearch (CURE), Health Services Research Section, School of Health and Related Research (ScHARR), University of Sheffield, Sheffield, United Kingdom
| | - Virginia Newcombe
- Division of Anaesthesia, Department of Medicine, Addenbrooke's Hospital, University of Cambridge, Cambridge, United Kingdom
| |
Collapse
|
3
|
Ionescu C, Ghidersa M, Ciobica A, Mavroudis I, Kazis D, Petridis FE, Gorgan DL, Balmus IM. Potential Correlation Between Molecular Biomarkers and Oxidative Stress in Traumatic Brain Injury. Int J Mol Sci 2025; 26:3858. [PMID: 40332547 PMCID: PMC12027598 DOI: 10.3390/ijms26083858] [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: 03/14/2025] [Revised: 04/14/2025] [Accepted: 04/16/2025] [Indexed: 05/08/2025] Open
Abstract
Diagnosing traumatic brain injury (TBI) remains challenging due to an incomplete understanding of its neuropathological mechanisms. TBI is recognised as a complex condition involving both primary and secondary injuries. Although oxidative stress is a non-specific molecular phenomenon observed in various neuropathological conditions, it plays a crucial role in brain injury response and recovery. Due to these aspects, we aimed to evaluate the interaction between some known TBI molecular biomarkers and oxidative stress in providing evidence for its possible relevance in clinical diagnosis and outcome prediction. We found that while many of the currently validated molecular biomarkers interact with oxidative pathways, their patterns of variation could assist the diagnosis, prognosis, and outcomes prediction in TBI cases.
Collapse
Affiliation(s)
- Cătălina Ionescu
- Department of Biology, Faculty of Biology, “Alexandru Ioan Cuza” University of Iasi, 700505 Iasi, Romania; (C.I.); (M.G.); (A.C.); (D.L.G.)
| | - Madalina Ghidersa
- Department of Biology, Faculty of Biology, “Alexandru Ioan Cuza” University of Iasi, 700505 Iasi, Romania; (C.I.); (M.G.); (A.C.); (D.L.G.)
| | - Alin Ciobica
- Department of Biology, Faculty of Biology, “Alexandru Ioan Cuza” University of Iasi, 700505 Iasi, Romania; (C.I.); (M.G.); (A.C.); (D.L.G.)
- “Ioan Haulica” Institute, Apollonia University, 700511 Iasi, Romania
- Center of Biomedical Research, Romanian Academy, Iasi Branch, 2 Teodor Codrescu Street, 700481 Iasi, Romania
| | - Ioannis Mavroudis
- Academy of Romanian Scientists, 050094 Bucharest, Romania
- Department of Neurosciences, Leeds Teaching Hospitals, Leeds LS9 7TF, UK
| | - Dimitrios Kazis
- Third Department of Neurology, Aristotle University of Thessaloniki, 541 24 Thessaloniki, Greece; (D.K.); (F.E.P.)
| | - Foivos E. Petridis
- Third Department of Neurology, Aristotle University of Thessaloniki, 541 24 Thessaloniki, Greece; (D.K.); (F.E.P.)
| | - Dragoș Lucian Gorgan
- Department of Biology, Faculty of Biology, “Alexandru Ioan Cuza” University of Iasi, 700505 Iasi, Romania; (C.I.); (M.G.); (A.C.); (D.L.G.)
| | - Ioana-Miruna Balmus
- Department of Exact Sciences and Natural Sciences, Institute of Interdisciplinary Research, “Alexandru Ioan Cuza” University of Iasi, 700057 Iasi, Romania;
| |
Collapse
|
4
|
Vázquez-Mojena Y, Rodríguez-Labrada R, Córdova-Rodríguez Y, Domínguez-Barrios Y, Fernández-Herrera ME, León-Arcia K, Pavón-Fuentes N, Robinson-Agramonte MDLA, Velázquez-Pérez L. Serum S100β Levels Are Linked with Cognitive Decline and Peripheral Inflammation in Spinocerebellar Ataxia Type 2. CEREBELLUM (LONDON, ENGLAND) 2024; 23:1509-1520. [PMID: 38347269 DOI: 10.1007/s12311-024-01665-2] [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] [Accepted: 01/24/2024] [Indexed: 07/25/2024]
Abstract
Experimental and clinical studies have indicated a potential role of the protein S100β in the pathogenesis and phenotype of neurodegenerative diseases. However, its impact on spinocerebellar ataxia type 2 (SCA2) remains to be elucidated. The objective of the study is to determine the serum levels of S100β in SCA2 and its relationship with molecular, clinical, cognitive, and peripheral inflammatory markers of the disease. Serum concentrations of S100β were measured by enzyme-linked immunosorbent assay in 39 SCA2 subjects and 36 age- and gender-matched controls. Clinical scores of ataxia, non-ataxia symptoms, cognitive dysfunction, and some blood cell count-derived inflammatory indices were assessed. The SCA2 individuals manifested S100β levels similar to the control group, at low nanomolar concentrations. However, the S100β levels were directly associated with a better performance of cognitive evaluation within the SCA2 cohort. Moreover, the S100β levels were inversely correlated with most peripheral inflammatory indices. Indeed, the neutrophil-to-lymphocyte ratio significantly mediated the effect of serum S100β on cognitive performance, even after controlling for the ataxia severity in the causal mediation analysis. Our findings suggested that, within physiologic concentrations, the protein S100β exerts a neuroprotective role against cognitive dysfunction in SCA2, likely via the suppression of pro-inflammatory mechanisms.
Collapse
Affiliation(s)
- Yaimeé Vázquez-Mojena
- Department of Molecular Biology, Cuban Centre for Neuroscience, 190 St, Between 25 St & 27 St, 11300, Playa, Havana, Cuba
| | - Roberto Rodríguez-Labrada
- Department of Molecular Biology, Cuban Centre for Neuroscience, 190 St, Between 25 St & 27 St, 11300, Playa, Havana, Cuba.
- Cuban Centre for Neurosciences, 190 Street, 19818, Between 25 & 27, 11600, Cubanacan, Playa, Havana, Cuba.
| | - Yanetsy Córdova-Rodríguez
- Institute of Nephrology "Abelardo Buch López", 26 Avenue & Rancho Boyeros Avenue10400, Plaza de La Revolución, Havana, Cuba
| | - Yennis Domínguez-Barrios
- Clinical & Surgical Hospital "Calixto Garcia", Universidad Avenue & J St, Vedado10400, Plaza de La Revolución, Havana, Cuba
| | - Mario E Fernández-Herrera
- Department of Human Physiology, Medical University of Havana, 146 St, 3102, 11300, Playa, Havana, Cuba
| | - Karen León-Arcia
- Department of Molecular Biology, Cuban Centre for Neuroscience, 190 St, Between 25 St & 27 St, 11300, Playa, Havana, Cuba
| | - Nancy Pavón-Fuentes
- Neuroimmunology Dept, International Centre for Neurological Restoration, 25 Avenue 15805, Between 158 St & 160 St, 11300, Playa, Havana, Cuba
| | | | - Luis Velázquez-Pérez
- Department of Human Physiology, Medical University of Havana, 146 St, 3102, 11300, Playa, Havana, Cuba
- Cuban Academy of Sciences, Cuba St 460, Between Teniente Rey & Amargura, Habana Vieja, 10100, Havana, Cuba
- Faculty of Chemistry, University of Havana, Zapata St Between G St & Carlitos Aguirre St, 10400, Plaza de La Revolución, Havana, Cuba
| |
Collapse
|
5
|
Lopes AN, Regner A, Simon D. The Role of S100b Protein Biomarker in Brain Death: A Literature Review. Cureus 2024; 16:e62707. [PMID: 39036258 PMCID: PMC11259197 DOI: 10.7759/cureus.62707] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/19/2024] [Indexed: 07/23/2024] Open
Abstract
Brain death (BD) represents the irreversible loss of all brain functions, including the brainstem, and is equivalent to clinical death established by neurological criteria. However, clinical diagnosis, mainly based on the absence of primary reflexes post-acute brain injury, remains a challenge in hospital settings. The S100 calcium-binding protein beta (S100b) is used to monitor brain injuries, as recommended by neurotrauma care guidelines in some countries. Its levels are associated with severity and mortality, particularly after traumatic brain injury (TBI) and cerebral hemorrhage. The evaluation of S100b levels in investigating brain death is promising; however, aspects such as cutoff values remain to be elucidated. This paper reviews the literature on the use of S100b as a biomarker in diagnosing brain death. It is noteworthy that there is still no defined cutoff for S100b levels in confirming brain death. Additionally, when considering the use of S100b in emergency situations, a point-of-care methodology should be established to support clinical decision-making quickly and easily in the early identification of patients who are more likely to progress to brain death. In this context, S100b levels may assist in establishing the diagnosis of brain death, complementing existing clinical evidence. This, in turn, can optimize and qualify the organ donation process, reducing costs with ineffective therapies and minimizing the suffering of the families involved.
Collapse
Affiliation(s)
| | - Andrea Regner
- Critical Care, Hospital Materno Infantil Presidente Vargas, Porto Alegre, BRA
| | - Daniel Simon
- Genetics, Universidade Luterana do Brasil, Canoas, BRA
| |
Collapse
|
6
|
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
|
7
|
Essl D, Schöchl H, Oberladstätter D, Lockie C, Islam M, Slezak C, Voelckel WG. Admission S100B fails as neuro-marker but is a good predictor for intrahospital mortality in major trauma patients. Injury 2024; 55:111187. [PMID: 37980176 DOI: 10.1016/j.injury.2023.111187] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/13/2023] [Revised: 10/08/2023] [Accepted: 11/04/2023] [Indexed: 11/20/2023]
Abstract
BACKGROUND S100 B is an extensively studied neuro-trauma marker, but its specificity and subsequently interpretation in major trauma patients might be limited, since extracerebral injuries are known to increase serum levels. Thus, we evaluated the potential role of S100B in the assessment of severe traumatic brain injury (TBI) in multiple injured patients upon emergency room (ER) admission and the first days of intensive care unit (ICU) stay. METHODS Retrospective study employing trauma registry data derived from a level 1 trauma center. Four cohorts of patients were grouped: isolated TBI (iTBI), polytrauma patients with TBI (PT + TBI), polytrauma patients without TBI (PT-TBI) and patients without polytrauma or TBI (control). S100B-serum levels were assessed immediately after admission in the emergency room and during the subsequent ICU stay. Values were correlated with injury severity score (ISS), Glasgow Coma Score (GCS) and in-hospital mortality. RESULTS 780 predominantly male patients (76 %) with a median age of 48 (30-63) and a median ISS of 24 (17-30) were enrolled in the study. Admission S100B correlated with ISS and TBI severity defined by the GCS (both p < 0.0001) but not with head abbreviated injury score (AIS) (p = 0.38). Compared with survivors, non-survivors had significantly higher median S100B levels in the ER (6.14 μg/L vs. 2.06 μg/L; p < 0.0001) and at ICU-day 1 (0.69 μg/L vs. 0.17 μg/L; p < 0.0001). S100B in the ER predicted mortality with an area under curve (AUC) of 0.77 (95 % CI 0,70-0,83, p < 0.0001), vs. 0.86 at ICU-day 1 (95 % CI 0,80-0,91, p < 0.0001). CONCLUSION In conclusion, S100B is a valid biomarker for prediction of mortality in major trauma patients with a higher accuracy when assessed at the first day of ICU stay vs. immediately after ER admission. Since S100B did not correlate with pathologic TBI findings in multiple injured patients, it failed as predictive neuro-marker because extracerebral injuries demonstrated a higher influence on admission levels than neurotrauma. Although S100B levels are indicative for injury severity they should be interpreted with caution in polytrauma patients.
Collapse
Affiliation(s)
- Daniel Essl
- Departement of Anaesthesiology and Intensive Care Medicine AUVA Trauma Centre Salzburg, Academic Teaching Hospital of the Paracelsus Medical University, Salzburg, Austria; Department of Anesthesiology and Critical Care Medicine, Medical University, Innsbruck, Austria
| | - Herbert Schöchl
- Departement of Anaesthesiology and Intensive Care Medicine AUVA Trauma Centre Salzburg, Academic Teaching Hospital of the Paracelsus Medical University, Salzburg, Austria; Ludwig Boltzmann Institute for Experimental and Clinical Traumatology, AUVA Trauma Research Centre, Vienna, Austria
| | - Daniel Oberladstätter
- Departement of Anaesthesiology and Intensive Care Medicine AUVA Trauma Centre Salzburg, Academic Teaching Hospital of the Paracelsus Medical University, Salzburg, Austria; Ludwig Boltzmann Institute for Experimental and Clinical Traumatology, AUVA Trauma Research Centre, Vienna, Austria
| | - Chris Lockie
- Departement of Anaesthesiology and Intensive Care Medicine AUVA Trauma Centre Salzburg, Academic Teaching Hospital of the Paracelsus Medical University, Salzburg, Austria
| | - Mohamed Islam
- Department of Mathematics, Utah Valley University, Orem, USA
| | - Cyrill Slezak
- Ludwig Boltzmann Institute for Experimental and Clinical Traumatology, AUVA Trauma Research Centre, Vienna, Austria; Department of Mathematics, Utah Valley University, Orem, USA
| | - Wolfgang G Voelckel
- Departement of Anaesthesiology and Intensive Care Medicine AUVA Trauma Centre Salzburg, Academic Teaching Hospital of the Paracelsus Medical University, Salzburg, Austria; University of Stavanger, Network for Medical Science, Stavanger, Norway.
| |
Collapse
|
8
|
Tefr Faridová A, Heřman H, Danačíková Š, Svoboda J, Otáhal J. Serum biomarkers of hypoxic-ischemic brain injury. Physiol Res 2023; 72:S461-S474. [PMID: 38165751 PMCID: PMC10861251 DOI: 10.33549/physiolres.935214] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2024] Open
Abstract
Brain injury is a multifaceted condition arising from nonspecific damage to nervous tissue. The resulting cognitive developmental impairments reverberate through patients' lives, affecting their families, and even the broader economic landscape. The significance of early brain injury detection lies in its potential to stave off severe consequences and enhance the effectiveness of tailored therapeutic interventions. While established methods like neuroimaging and neurophysiology serve as valuable diagnostic tools, their demanding nature restricts their accessibility, particularly in scenarios such as small hospitals, nocturnal or weekend shifts, and cases involving unstable patients. Hence, there is a pressing need for more accessible and efficient diagnostic avenues. Among the spectrum of brain injuries, hypoxic-ischemic encephalopathy stands out as a predominant affliction in the pediatric population. Diagnosing brain injuries in newborns presents challenges due to the subjective nature of assessments like Apgar scores and the inherent uncertainty in neurological examinations. In this context, methods like magnetic resonance and ultrasound hold recommendations for more accurate diagnosis. Recognizing the potential of serum biomarkers derived from blood samples, this paper underscores their promise as a more expedient and resource-efficient means of assessing brain injuries. The review compiles current insights into serum biomarkers, drawing from experiments conducted on animal models as well as human brain pathologies. The authors aim to elucidate specific characteristics, temporal profiles, and the available corpus of experimental and clinical data for serum biomarkers specific to brain injuries. These include neuron-specific enolase (NSE), ubiquitin carboxy-terminal hydrolase L1 (UCH-L1), S100 calcium-binding protein beta (S100B), glial fibrillary acidic protein (GFAP), and high-mobility-group-protein-box-1 (HMGB1). This comprehensive endeavor contributes to advancing the understanding of brain injury diagnostics and potential avenues for therapeutic intervention.
Collapse
Affiliation(s)
- A Tefr Faridová
- A. Tefr Faridová, Department of Pathophysiology, Second Faculty of Medicine, Charles University, Prague 5, Czech Republic. and
| | | | | | | | | |
Collapse
|
9
|
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
|
10
|
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
|
11
|
Harris G, Stickland CA, Lim M, Goldberg Oppenheimer P. Raman Spectroscopy Spectral Fingerprints of Biomarkers of Traumatic Brain Injury. Cells 2023; 12:2589. [PMID: 37998324 PMCID: PMC10670390 DOI: 10.3390/cells12222589] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2023] [Revised: 11/02/2023] [Accepted: 11/06/2023] [Indexed: 11/25/2023] Open
Abstract
Traumatic brain injury (TBI) affects millions of people of all ages around the globe. TBI is notoriously hard to diagnose at the point of care, resulting in incorrect patient management, avoidable death and disability, long-term neurodegenerative complications, and increased costs. It is vital to develop timely, alternative diagnostics for TBI to assist triage and clinical decision-making, complementary to current techniques such as neuroimaging and cognitive assessment. These could deliver rapid, quantitative TBI detection, by obtaining information on biochemical changes from patient's biofluids. If available, this would reduce mis-triage, save healthcare providers costs (both over- and under-triage are expensive) and improve outcomes by guiding early management. Herein, we utilize Raman spectroscopy-based detection to profile a panel of 18 raw (human, animal, and synthetically derived) TBI-indicative biomarkers (N-acetyl-aspartic acid (NAA), Ganglioside, Glutathione (GSH), Neuron Specific Enolase (NSE), Glial Fibrillary Acidic Protein (GFAP), Ubiquitin C-terminal Hydrolase L1 (UCHL1), Cholesterol, D-Serine, Sphingomyelin, Sulfatides, Cardiolipin, Interleukin-6 (IL-6), S100B, Galactocerebroside, Beta-D-(+)-Glucose, Myo-Inositol, Interleukin-18 (IL-18), Neurofilament Light Chain (NFL)) and their aqueous solution. The subsequently derived unique spectral reference library, exploiting four excitation lasers of 514, 633, 785, and 830 nm, will aid the development of rapid, non-destructive, and label-free spectroscopy-based neuro-diagnostic technologies. These biomolecules, released during cellular damage, provide additional means of diagnosing TBI and assessing the severity of injury. The spectroscopic temporal profiles of the studied biofluid neuro-markers are classed according to their acute, sub-acute, and chronic temporal injury phases and we have further generated detailed peak assignment tables for each brain-specific biomolecule within each injury phase. The intensity ratios of significant peaks, yielding the combined unique spectroscopic barcode for each brain-injury marker, are compared to assess variance between lasers, with the smallest variance found for UCHL1 (σ2 = 0.000164) and the highest for sulfatide (σ2 = 0.158). Overall, this work paves the way for defining and setting the most appropriate diagnostic time window for detection following brain injury. Further rapid and specific detection of these biomarkers, from easily accessible biofluids, would not only enable the triage of TBI, predict outcomes, indicate the progress of recovery, and save healthcare providers costs, but also cement the potential of Raman-based spectroscopy as a powerful tool for neurodiagnostics.
Collapse
Affiliation(s)
- Georgia Harris
- Advanced Nanomaterials Structures and Applications Laboratories, School of Chemical Engineering, College of Engineering and Physical Sciences, University of Birmingham, Edgbaston, Birmingham B15 2TT, UK
| | - Clarissa A. Stickland
- Advanced Nanomaterials Structures and Applications Laboratories, School of Chemical Engineering, College of Engineering and Physical Sciences, University of Birmingham, Edgbaston, Birmingham B15 2TT, UK
| | - Matthias Lim
- Advanced Nanomaterials Structures and Applications Laboratories, School of Chemical Engineering, College of Engineering and Physical Sciences, University of Birmingham, Edgbaston, Birmingham B15 2TT, UK
| | - Pola Goldberg Oppenheimer
- Advanced Nanomaterials Structures and Applications Laboratories, School of Chemical Engineering, College of Engineering and Physical Sciences, University of Birmingham, Edgbaston, Birmingham B15 2TT, UK
- Institute of Healthcare Technologies, Mindelsohn Way, Birmingham B15 2TH, UK
| |
Collapse
|
12
|
Lundsgaard CC, Gbyl K, Videbech P. Blood-brain barrier permeability and electroconvulsive therapy: a systematic review. Acta Neuropsychiatr 2023; 37:e22. [PMID: 37842858 DOI: 10.1017/neu.2023.48] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/17/2023]
Abstract
OBJECTIVE The cause of cognitive side effects after electroconvulsive therapy (ECT) is largely unknown. Alterations in the blood-brain barrier (BBB) have been considered in several recent ECT studies. We therefore found it worthwhile to perform a systematic review of the literature to examine if electrically induced seizures affect the permeability of the BBB. METHODS PubMed/MEDLINE and Embase were searched 16 November 2022. Studies with a direct measurement of BBB permeability in animals treated with modified electroconvulsive stimulation (ECS) and in humans treated with ECT were included. Synthesis of results was narrative due to the low number of studies and differences in study designs. RESULTS Four animal and two human (31 participants) studies were included. In animals, two studies found increased BBB permeability to some smaller molecules after modified ECS, while the two other studies found marginally increased or unchanged permeability to albumin after treatment. In contrast, the human studies did not find increased BBB permeability to smaller molecules or albumin after ECT. CONCLUSION Animal but not human studies support increased BBB permeability to some smaller molecules after electrically induced seizures. However, this conclusion is confined by the low number of studies and the lack of studies applying state-of-the-art methods. More studies using modern approaches to measuring of BBB permeability are warranted. FUNDING AND REGISTRATION The study was founded by Mental Health Services in the Capital Region of Denmark (grant number 61151-05) and was registered on PROSPERO before data extraction was initiated (CRD42022331385).
Collapse
Affiliation(s)
- Christoffer C Lundsgaard
- Center for Neuropsychiatric Depression Research, Mental Health Centre Glostrup, Denmark
- Institute of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Krzysztof Gbyl
- Center for Neuropsychiatric Depression Research, Mental Health Centre Glostrup, Denmark
- Institute of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Poul Videbech
- Center for Neuropsychiatric Depression Research, Mental Health Centre Glostrup, Denmark
- Institute of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
13
|
Saletti PG, Mowrey WB, Liu W, Li Q, McCullough J, Aniceto R, Lin I, Eklund M, Casillas‐Espinosa PM, Ali I, Santana‐Gomez C, Coles L, Shultz SR, Jones N, Staba R, O'Brien TJ, Moshé SL, Agoston DV, Galanopoulou AS, for the EpiBioS4Rx Study Group. Early preclinical plasma protein biomarkers of brain trauma are influenced by early seizures and levetiracetam. Epilepsia Open 2023; 8:586-608. [PMID: 37026764 PMCID: PMC10235584 DOI: 10.1002/epi4.12738] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2022] [Accepted: 04/04/2023] [Indexed: 04/08/2023] Open
Abstract
OBJECTIVE We used the lateral fluid percussion injury (LFPI) model of moderate-to-severe traumatic brain injury (TBI) to identify early plasma biomarkers predicting injury, early post-traumatic seizures or neuromotor functional recovery (neuroscores), considering the effect of levetiracetam, which is commonly given after severe TBI. METHODS Adult male Sprague-Dawley rats underwent left parietal LFPI, received levetiracetam (200 mg/kg bolus, 200 mg/kg/day subcutaneously for 7 days [7d]) or vehicle post-LFPI, and were continuously video-EEG recorded (n = 14/group). Sham (craniotomy only, n = 6), and naïve controls (n = 10) were also used. Neuroscores and plasma collection were done at 2d or 7d post-LFPI or equivalent timepoints in sham/naïve. Plasma protein biomarker levels were determined by reverse phase protein microarray and classified according to injury severity (LFPI vs. sham/control), levetiracetam treatment, early seizures, and 2d-to-7d neuroscore recovery, using machine learning. RESULTS Low 2d plasma levels of Thr231 -phosphorylated tau protein (pTAU-Thr231 ) and S100B combined (ROC AUC = 0.7790) predicted prior craniotomy surgery (diagnostic biomarker). Levetiracetam-treated LFPI rats were differentiated from vehicle treated by the 2d-HMGB1, 2d-pTAU-Thr231 , and 2d-UCHL1 plasma levels combined (ROC AUC = 0.9394) (pharmacodynamic biomarker). Levetiracetam prevented the seizure effects on two biomarkers that predicted early seizures only among vehicle-treated LFPI rats: pTAU-Thr231 (ROC AUC = 1) and UCHL1 (ROC AUC = 0.8333) (prognostic biomarker of early seizures among vehicle-treated LFPI rats). Levetiracetam-resistant early seizures were predicted by high 2d-IFNγ plasma levels (ROC AUC = 0.8750) (response biomarker). 2d-to-7d neuroscore recovery was best predicted by higher 2d-S100B, lower 2d-HMGB1, and 2d-to-7d increase in HMGB1 or decrease in TNF (P < 0.05) (prognostic biomarkers). SIGNIFICANCE Antiseizure medications and early seizures need to be considered in the interpretation of early post-traumatic biomarkers.
Collapse
Affiliation(s)
- Patricia G. Saletti
- Saul R. Korey Department of Neurology, Laboratory of Developmental EpilepsyAlbert Einstein College of MedicineBronxNew YorkUSA
| | - Wenzhu B. Mowrey
- Department of Epidemiology & Population HealthAlbert Einstein College of MedicineBronxNew YorkUSA
| | - Wei Liu
- Saul R. Korey Department of Neurology, Laboratory of Developmental EpilepsyAlbert Einstein College of MedicineBronxNew YorkUSA
| | - Qianyun Li
- Saul R. Korey Department of Neurology, Laboratory of Developmental EpilepsyAlbert Einstein College of MedicineBronxNew YorkUSA
| | - Jesse McCullough
- Department of Anatomy, Physiology and GeneticsUniformed Services UniversityBethesdaMarylandUSA
| | - Roxanne Aniceto
- Department of Anatomy, Physiology and GeneticsUniformed Services UniversityBethesdaMarylandUSA
| | - I‐Hsuan Lin
- Department of Anatomy, Physiology and GeneticsUniformed Services UniversityBethesdaMarylandUSA
| | - Michael Eklund
- Department of Anatomy, Physiology and GeneticsUniformed Services UniversityBethesdaMarylandUSA
| | - Pablo M. Casillas‐Espinosa
- Department of NeuroscienceMonash UniversityMelbourneVictoriaAustralia
- Department of MedicineThe University of MelbourneParkvilleVictoriaAustralia
- Department of NeurologyAlfred HealthMelbourneVictoriaAustralia
| | - Idrish Ali
- Department of NeuroscienceMonash UniversityMelbourneVictoriaAustralia
- Department of MedicineThe University of MelbourneParkvilleVictoriaAustralia
- Department of NeurologyAlfred HealthMelbourneVictoriaAustralia
| | | | - Lisa Coles
- University of Minnesota Twin CitiesMinneapolisMinnesotaUSA
| | - Sandy R. Shultz
- Department of NeuroscienceMonash UniversityMelbourneVictoriaAustralia
- Department of MedicineThe University of MelbourneParkvilleVictoriaAustralia
- Department of NeurologyAlfred HealthMelbourneVictoriaAustralia
| | - Nigel Jones
- Department of NeuroscienceMonash UniversityMelbourneVictoriaAustralia
- Department of MedicineThe University of MelbourneParkvilleVictoriaAustralia
- Department of NeurologyAlfred HealthMelbourneVictoriaAustralia
| | | | - Terence J. O'Brien
- Department of NeuroscienceMonash UniversityMelbourneVictoriaAustralia
- Department of MedicineThe University of MelbourneParkvilleVictoriaAustralia
- Department of NeurologyAlfred HealthMelbourneVictoriaAustralia
| | - Solomon L. Moshé
- Saul R. Korey Department of Neurology, Laboratory of Developmental EpilepsyAlbert Einstein College of MedicineBronxNew YorkUSA
- Isabelle Rapin Division of Child NeurologyAlbert Einstein College of MedicineBronxNew YorkUSA
- Dominick P Purpura Department of NeuroscienceAlbert Einstein College of MedicineBronxNew YorkUSA
- Department of PediatricsAlbert Einstein College of MedicineBronxNew YorkUSA
| | - Denes V. Agoston
- Department of Anatomy, Physiology and GeneticsUniformed Services UniversityBethesdaMarylandUSA
| | - Aristea S. Galanopoulou
- Saul R. Korey Department of Neurology, Laboratory of Developmental EpilepsyAlbert Einstein College of MedicineBronxNew YorkUSA
- Isabelle Rapin Division of Child NeurologyAlbert Einstein College of MedicineBronxNew YorkUSA
- Dominick P Purpura Department of NeuroscienceAlbert Einstein College of MedicineBronxNew YorkUSA
| | | |
Collapse
|
14
|
Hong G, Li T, Zhao H, Zeng Z, Zhai J, Li X, Luo X. Diagnostic value and mechanism of plasma S100A1 protein in acute ischemic stroke: a prospective and observational study. PeerJ 2023; 11:e14440. [PMID: 36643631 PMCID: PMC9838205 DOI: 10.7717/peerj.14440] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2022] [Accepted: 11/01/2022] [Indexed: 01/12/2023] Open
Abstract
Background Plasma S100A1 protein is a novel inflammatory biomarker associated with acute myocardial infarction and neurodegenerative disease's pathophysiological mechanisms. This study aimed to determine the levels of this protein in patients with acute ischemic stroke early in the disease progression and to investigate its role in the pathogenesis of acute ischemic stroke. Methods A total of 192 participants from hospital stroke centers were collected for the study. Clinically pertinent data were recorded. The volume of the cerebral infarction was calculated according to the Pullicino formula. Multivariate logistic regression analysis was used to select independent influences. ROC curve was used to analyze the diagnostic value of AIS and TIA. The correlation between S100A1, NF-κB p65, and IL-6 levels and cerebral infarction volume was detected by Pearson correlation analysis. Results There were statistically significant differences in S100A1, NF-κB p65, and IL-6 among the AIS,TIA, and PE groups (S100A1, [230.96 ± 39.37] vs [185.85 ± 43.24] vs [181.47 ± 27.39], P < 0.001; NF-κB p65, [3.99 ± 0.65] vs [3.58 ± 0.74] vs [3.51 ± 0.99], P = 0.001; IL-6, [13.32 ± 1.57] vs [11.61 ± 1.67] vs [11.42 ± 2.34], P < 0.001). Multivariate logistic regression analysis showed that S100A1 might be an independent predictive factor for the diagnosis of disease (P < 0.001). The AUC of S100A1 for diagnosis of AIS was 0.818 (P < 0.001, 95% CI [0.749-0.887], cut off 181.03, Jmax 0.578, Se 95.0%, Sp 62.7%). The AUC of S100A1 for diagnosis of TIA was 0.720 (P = 0.001, 95% CI [0.592-0.848], cut off 150.14, Jmax 0.442, Se 50.0%, Sp 94.2%). There were statistically significant differences in S100A1, NF-κB p65, and IL-6 among the SCI,MCI, and LCI groups (S100A1, [223.98 ± 40.21] vs [225.42 ± 30.92] vs [254.25 ± 37.07], P = 0.001; NF-κB p65, [3.88 ± 0.66] vs [3.85 ± 0.64] vs [4.41 ± 0.45], P < 0.001; IL-6, [13.27 ± 1.65] vs [12.77 ± 1.31] vs [14.00 ± 1.40], P = 0.007). Plasma S100A1, NF-κB p65, and IL-6 were significantly different from cerebral infarction volume (S100A1, r = 0.259, P = 0.002; NF-κB p65, r = 0.316, P < 0.001; IL-6, r = 0.177, P = 0.036). There was a positive correlation between plasma S100A1 and IL-6 with statistical significance (R = 0.353, P < 0.001). There was no significant positive correlation between plasma S100A1 and NF-κB p65 (R < 0.3), but there was statistical significance (R = 0.290, P < 0.001). There was a positive correlation between IL-6 and NF-κB p65 with statistical significance (R = 0.313, P < 0.001). Conclusion S100A1 might have a better diagnostic efficacy for AIS and TIA. S100A1 was associated with infarct volume in AIS, and its level reflected the severity of acute cerebral infarction to a certain extent. There was a correlation between S100A1 and IL-6 and NF-κB p65, and it was reasonable to speculate that this protein might mediate the inflammatory response through the NF-κB pathway during the pathophysiology of AIS.
Collapse
Affiliation(s)
- Guo Hong
- Department of Neurology, Second Clinical Medical College of Jinan University, Shenzhen, China
| | - Tingting Li
- Department of Neurology, Yizheng People’s Hospital affiliated to Yangzhou University, Yangzhou, China
| | - Haina Zhao
- Department of Neurology, Institutes of Brain Science, Jiangsu Subei People’s Hospital affiliated to Yangzhou University, Yangzhou, China
| | - Zhaohao Zeng
- Department of Neurology, Second Clinical Medical College of Jinan University, Shenzhen, China
| | - Jinglei Zhai
- School of Medicine, Southern University of Science and Technology, Shenzhen, China
| | - Xiaobo Li
- Department of Neurology, Institutes of Brain Science, Jiangsu Subei People’s Hospital affiliated to Yangzhou University, Yangzhou, China
| | - Xiaoguang Luo
- Department of Neurology, Second Clinical Medical College of Jinan University, Shenzhen, China
| |
Collapse
|
15
|
Harris G, Rickard JJS, Butt G, Kelleher L, Blanch RJ, Cooper J, Oppenheimer PG. Review: Emerging Eye-Based Diagnostic Technologies for Traumatic Brain Injury. IEEE Rev Biomed Eng 2023; 16:530-559. [PMID: 35320105 PMCID: PMC9888755 DOI: 10.1109/rbme.2022.3161352] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2021] [Revised: 02/11/2022] [Accepted: 03/15/2022] [Indexed: 11/06/2022]
Abstract
The study of ocular manifestations of neurodegenerative disorders, Oculomics, is a growing field of investigation for early diagnostics, enabling structural and chemical biomarkers to be monitored overtime to predict prognosis. Traumatic brain injury (TBI) triggers a cascade of events harmful to the brain, which can lead to neurodegeneration. TBI, termed the "silent epidemic" is becoming a leading cause of death and disability worldwide. There is currently no effective diagnostic tool for TBI, and yet, early-intervention is known to considerably shorten hospital stays, improve outcomes, fasten neurological recovery and lower mortality rates, highlighting the unmet need for techniques capable of rapid and accurate point-of-care diagnostics, implemented in the earliest stages. This review focuses on the latest advances in the main neuropathophysiological responses and the achievements and shortfalls of TBI diagnostic methods. Validated and emerging TBI-indicative biomarkers are outlined and linked to ocular neuro-disorders. Methods detecting structural and chemical ocular responses to TBI are categorised along with prospective chemical and physical sensing techniques. Particular attention is drawn to the potential of Raman spectroscopy as a non-invasive sensing of neurological molecular signatures in the ocular projections of the brain, laying the platform for the first tangible path towards alternative point-of-care diagnostic technologies for TBI.
Collapse
Affiliation(s)
- Georgia Harris
- School of Chemical Engineering, Advanced Nanomaterials Structures and Applications Laboratories, College of Engineering and Physical SciencesUniversity of BirminghamB15 2TTBirminghamU.K.
| | - Jonathan James Stanley Rickard
- School of Chemical Engineering, Advanced Nanomaterials Structures and Applications Laboratories, College of Engineering and Physical SciencesUniversity of BirminghamB15 2TTBirminghamU.K.
- Department of Physics, Cavendish LaboratoryUniversity of CambridgeCB3 0HECambridgeU.K.
| | - Gibran Butt
- Ophthalmology DepartmentUniversity Hospitals Birmingham NHS Foundation TrustB15 2THBirminghamU.K.
| | - Liam Kelleher
- School of Chemical Engineering, Advanced Nanomaterials Structures and Applications Laboratories, College of Engineering and Physical SciencesUniversity of BirminghamB15 2TTBirminghamU.K.
| | - Richard James Blanch
- Department of Military Surgery and TraumaRoyal Centre for Defence MedicineB15 2THBirminghamU.K.
- Neuroscience and Ophthalmology, Department of Ophthalmology, University Hospitals Birmingham NHS Foundation TrustcBirminghamU.K.
| | - Jonathan Cooper
- School of Biomedical EngineeringUniversity of GlasgowG12 8LTGlasgowU.K.
| | - Pola Goldberg Oppenheimer
- School of Chemical Engineering, Advanced Nanomaterials Structures and Applications Laboratories, College of Engineering and Physical SciencesUniversity of BirminghamB15 2TTBirminghamU.K.
- Healthcare Technologies Institute, Institute of Translational MedicineB15 2THBirminghamU.K.
| |
Collapse
|
16
|
Hier DB, Azizi S, Thimgan MS, Wunsch DC. Tau kinetics in Alzheimer's disease. Front Aging Neurosci 2022; 14:1055170. [PMID: 36437992 PMCID: PMC9682289 DOI: 10.3389/fnagi.2022.1055170] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2022] [Accepted: 10/27/2022] [Indexed: 07/20/2023] Open
Abstract
The cytoskeletal protein tau is implicated in the pathogenesis of Alzheimer's disease which is characterized by intra-neuronal neurofibrillary tangles containing abnormally phosphorylated insoluble tau. Levels of soluble tau are elevated in the brain, the CSF, and the plasma of patients with Alzheimer's disease. To better understand the causes of these elevated levels of tau, we propose a three-compartment kinetic model (brain, CSF, and plasma). The model assumes that the synthesis of tau follows zero-order kinetics (uncorrelated with compartmental tau levels) and that the release, absorption, and clearance of tau is governed by first-order kinetics (linearly related to compartmental tau levels). Tau that is synthesized in the brain compartment can be released into the interstitial fluid, catabolized, or retained in neurofibrillary tangles. Tau released into the interstitial fluid can mix with the CSF and eventually drain to the plasma compartment. However, losses of tau in the drainage pathways may be significant. The kinetic model estimates half-life of tau in each compartment (552 h in the brain, 9.9 h in the CSF, and 10 h in the plasma). The kinetic model predicts that an increase in the neuronal tau synthesis rate or a decrease in tau catabolism rate best accounts for observed increases in tau levels in the brain, CSF, and plasma found in Alzheimer's disease. Furthermore, the model predicts that increases in brain half-life of tau in Alzheimer's disease should be attributed to decreased tau catabolism and not to increased tau synthesis. Most clearance of tau in the neuron occurs through catabolism rather than release to the CSF compartment. Additional experimental data would make ascertainment of the model parameters more precise.
Collapse
Affiliation(s)
- Daniel B. Hier
- Applied Computational Intelligence Laboratory, Department of Electrical & Computer Engineering, Missouri University of Science & Technology, Rolla, MO, United States
- Department of Neurology and Rehabilitation, University of Illinois at Chicago, Chicago, IL, United States
| | - Sima Azizi
- Applied Computational Intelligence Laboratory, Department of Electrical & Computer Engineering, Missouri University of Science & Technology, Rolla, MO, United States
| | - Matthew S. Thimgan
- Department of Biological Sciences, Missouri University of Science & Technology, Rolla, MO, United States
| | - Donald C. Wunsch
- Applied Computational Intelligence Laboratory, Department of Electrical & Computer Engineering, Missouri University of Science & Technology, Rolla, MO, United States
- ECCS Division, National Science Foundation, Alexandria, VA, United States
| |
Collapse
|
17
|
Abstract
OBJECTIVE S100B is a glial cell protein with bimodal function. In low concentrations, it exerts neurotrophic effects, but higher levels reflect neuronal distress. Recent research suggests that this molecule may be a biomarker of response to electroconvulsive therapy (ECT). We examined the effect of ECT on serum S100B and its utility as 1) a biomarker of a depressive state and 2) a predictor of ECT response. We also wanted to ensure that ECT does not cause a marked serum S100B elevation, indicating neural distress. METHODS We measured serum S100B in 22 in-patients treated with ECT due to depression. Depression severity was assessed using 17-item Hamilton Rating Scale for Depression (HAMD-17). The data were collected before an ECT series, within 1 week after the series (post-ECT), and at a 6-month follow-up. Changes in serum S100B and clinical outcomes were tested using a linear mixed model. A relationship between serum S100B and the clinical outcomes was examined using Spearman's and partial correlation. RESULTS Serum S100B did not change significantly immediately after an ECT series or 6 months later. The post-ECT serum S100B change was not associated with the clinical effect (rho = 0.14, n = 22, p = 0.54). The baseline serum S100B did not predict the clinical effect when controlling for age (r = 0.02, n = 22, df = 19, p = 0.92). CONCLUSION The study neither supports serum S100B as a state marker of depression nor a predictor of ECT response. No evidence for ECT-related neural distress was found.
Collapse
|
18
|
Kocheril PA, Moore SC, Lenz KD, Mukundan H, Lilley LM. Progress Toward a Multiomic Understanding of Traumatic Brain Injury: A Review. Biomark Insights 2022; 17:11772719221105145. [PMID: 35719705 PMCID: PMC9201320 DOI: 10.1177/11772719221105145] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2022] [Accepted: 05/17/2022] [Indexed: 12/11/2022] Open
Abstract
Traumatic brain injury (TBI) is not a single disease state but describes an array
of conditions associated with insult or injury to the brain. While some
individuals with TBI recover within a few days or months, others present with
persistent symptoms that can cause disability, neuropsychological trauma, and
even death. Understanding, diagnosing, and treating TBI is extremely complex for
many reasons, including the variable biomechanics of head impact, differences in
severity and location of injury, and individual patient characteristics. Because
of these confounding factors, the development of reliable diagnostics and
targeted treatments for brain injury remains elusive. We argue that the
development of effective diagnostic and therapeutic strategies for TBI requires
a deep understanding of human neurophysiology at the molecular level and that
the framework of multiomics may provide some effective solutions for the
diagnosis and treatment of this challenging condition. To this end, we present
here a comprehensive review of TBI biomarker candidates from across the
multiomic disciplines and compare them with known signatures associated with
other neuropsychological conditions, including Alzheimer’s disease and
Parkinson’s disease. We believe that this integrated view will facilitate a
deeper understanding of the pathophysiology of TBI and its potential links to
other neurological diseases.
Collapse
Affiliation(s)
- Philip A Kocheril
- Physical Chemistry and Applied Spectroscopy Group, Chemistry Division, Los Alamos National Laboratory, Los Alamos, NM, USA
| | - Shepard C Moore
- Physical Chemistry and Applied Spectroscopy Group, Chemistry Division, Los Alamos National Laboratory, Los Alamos, NM, USA
| | - Kiersten D Lenz
- Physical Chemistry and Applied Spectroscopy Group, Chemistry Division, Los Alamos National Laboratory, Los Alamos, NM, USA
| | - Harshini Mukundan
- Physical Chemistry and Applied Spectroscopy Group, Chemistry Division, Los Alamos National Laboratory, Los Alamos, NM, USA
| | - Laura M Lilley
- Physical Chemistry and Applied Spectroscopy Group, Chemistry Division, Los Alamos National Laboratory, Los Alamos, NM, USA
| |
Collapse
|
19
|
Chen H, Ding V, Zhu G, Jiang B, Li Y, Boothroyd D, Rezaii P, Bet AM, Paulino A, Weber A, Glushakova OY, Hayes RL, Wintermark M. Association between Blood and CT Imaging Biomarkers in a Cohort of Mild Traumatic Brain Injury Patients. J Neurotrauma 2022; 39:1329-1338. [PMID: 35546284 DOI: 10.1089/neu.2021.0390] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
OBJECTIVE To analyze the relationships between traumatic brain injury (TBI) on CT imaging and blood concentration of glial fibrillary acidic protein (GFAP), ubiquitin C-terminal hydrolase-L1 (UCH-L1), and S100B. METHODS This prospective cohort study involved 644 TBI patients referred to Stanford Hospital's Emergency Department between November 2015 and April 2017. Plasma and serum samples of 462 patients were analyzed for levels of GFAP, UCH-L1 and S100B. Glial neuronal ratio (GNR) was calculated as the ratio between GFAP and UCH-L1 concentrations. Admission head CT scans were reviewed for TBI imaging common data elements, and performance of biomarkers for identifying TBI was assessed via area under the receiver operating characteristic curve (ROC). We also dichotomized biomarkers at established thresholds and estimated standard measures of classification accuracy. We assessed the ability of GFAP, UCH-L1 and GNR to discriminate small and large/diffuse lesions based on CT imaging using an ROC analysis. RESULTS In our cohort of mostly mild TBI patients, GFAP was significantly more accurate in detecting all types of acute brain injuries than UCH-L1 in terms of area under the curves (AUC) values (P<0.001), and also compared to S100B (P<0.001). UCH-L1 and S100B had similar performance (comparable AUC values, P=0.342). Sensitivity exceeded 0.8 for each biomarker across all different types of TBI injuries, and no significant differences were observed by type of injury. Significant differences of GFAP and GNR distinguished between small lesions and large/diffuse lesions in all injuries (P=0.004, P=0.007). CONCLUSIONS GFAP, UCH-L1, and S100B show high sensitivity and negative predictive values for all types of TBI lesions on head CT. A combination of negative blood biomarkers (GFAP and UCH-L1) in a patient suspected of TBI may be used to safely obviate the need for a head CT scan. GFAP is a promising indicator to discriminate between small and large/diffuse TBI lesions.
Collapse
Affiliation(s)
- Hui Chen
- Stanford University School of Medicine, 10624, Radiology, Stanford, California, United States;
| | - Victoria Ding
- Stanford University, 6429, Department of Medicine, Quantitative Sciences Unit, Stanford, California, United States;
| | - Guangming Zhu
- Stanford University School of Medicine, 10624, Radiology, Stanford, California, United States;
| | - Bin Jiang
- Stanford University Department of Radiology, 209657, Radiology, Neuroradiology Division, 300 Pasteur Dr., Stanford, California, United States, 94305;
| | - Ying Li
- Stanford University School of Medicine, 10624, Stanford, California, United States;
| | - Derek Boothroyd
- Stanford University, 6429, Department of Medicine, Stanford, California, United States;
| | - Paymon Rezaii
- Stanford University, Department of Radiology, 300 Pasteur Drive, Room S047, Stanford, California, United States, 94305;
| | - Anthony Marco Bet
- Stanford University, Department of Neurosurgery, 300 Pasteur Dr, Stanford, California, United States, 94305;
| | - Amy Paulino
- Banyan Biomarkers Inc San Diego, 506046, 16470 W Bernardo Drive, Suite 100, San Diego, California, United States, 92127;
| | - Art Weber
- Banyan Biomarkers Inc San Diego, 506046, Clinical Affairs, 16470 West Bernardo Drive, Suite 100, San Diego, California, United States, 92127;
| | - Olena Y Glushakova
- Virginia Commonwealth University, Department of Neurosurgery, PO Box 980631, Richmond, Virginia, United States, 23298-0631;
| | - Ronald L Hayes
- Banyan Biomarkers, Inc., Director, Center of Innovative Research, 12085 Research Dr., Alachua, Florida, United States, 32615.,United States;
| | - Max Wintermark
- Stanford University Department of Radiology, 209657, Radiology, Neuroradiology Division, 300 Pasteur Dr, Grant Building S047, Stanford, California, United States, 94305-5105;
| |
Collapse
|
20
|
Whitehouse DP, Vile AR, Adatia K, Herlekar R, Roy AS, Mondello S, Czeiter E, Amrein K, Büki A, Maas AIR, Menon DK, Newcombe VFJ. Blood Biomarkers and Structural Imaging Correlations Post-Traumatic Brain Injury: A Systematic Review. Neurosurgery 2022; 90:170-179. [PMID: 34995235 DOI: 10.1227/neu.0000000000001776] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2021] [Accepted: 08/24/2021] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND Blood biomarkers are of increasing importance in the diagnosis and assessment of traumatic brain injury (TBI). However, the relationship between them and lesions seen on imaging remains unclear. OBJECTIVE To perform a systematic review of the relationship between blood biomarkers and intracranial lesion types, intracranial lesion injury patterns, volume/number of intracranial lesions, and imaging classification systems. METHODS We searched Medical Literature Analysis and Retrieval System Online, Excerpta Medica dataBASE, and Cumulative Index to Nursing and Allied Health Literature from inception to May 2021, and the references of included studies were also screened. Heterogeneity in study design, biomarker types, imaging modalities, and analyses inhibited quantitative analysis, with a qualitative synthesis presented. RESULTS Fifty-nine papers were included assessing one or more biomarker to imaging comparisons per paper: 30 assessed imaging classifications or injury patterns, 28 assessed lesion type, and 11 assessed lesion volume or number. Biomarker concentrations were associated with the burden of brain injury, as assessed by increasing intracranial lesion volume, increasing numbers of traumatic intracranial lesions, and positive correlations with imaging classification scores. There were inconsistent findings associating different biomarkers with specific imaging phenotypes including diffuse axonal injury, cerebral edema, and intracranial hemorrhage. CONCLUSION Blood-based biomarker concentrations after TBI are consistently demonstrated to correlate burden of intracranial disease. The relation with specific injury types is unclear suggesting a lack of diagnostic specificity and/or is the result of the complex and heterogeneous nature of TBI.
Collapse
Affiliation(s)
- Daniel P Whitehouse
- Department of Medicine, University Division of Anaesthesia, University of Cambridge, Cambridge, UK
| | | | - Krishma Adatia
- Department of Medicine, University Division of Anaesthesia, University of Cambridge, Cambridge, UK
| | - Rahul Herlekar
- School of Clinical Medicine, Addenbrooke's Hospital, University of Cambridge, Cambridge, UK
| | - Akangsha Sur Roy
- School of Clinical Medicine, Addenbrooke's Hospital, University of Cambridge, Cambridge, UK
| | - Stefania Mondello
- Department of Biomedical and Dental Sciences and Morphofunctional Imaging, University of Messina, Messina, Italy
| | - Endre Czeiter
- Department of Neurosurgery, Medical School, University of Pécs, Pécs, Hungary
- Neurotrauma Research Group, Szentágothai Research Centre, University of Pécs, Pécs, Hungary
- MTA-PTE Clinical Neuroscience MR Research Group, Pécs, Hungary
| | - Krisztina Amrein
- Department of Neurosurgery, Medical School, University of Pécs, Pécs, Hungary
- Neurotrauma Research Group, Szentágothai Research Centre, University of Pécs, Pécs, Hungary
| | - András Büki
- Department of Neurosurgery, Medical School, University of Pécs, Pécs, Hungary
- Neurotrauma Research Group, Szentágothai Research Centre, University of Pécs, Pécs, Hungary
| | - Andrew I R Maas
- Department of Neurosurgery, Antwerp University Hospital and University of Antwerp, Edegem, Belgium
| | - David K Menon
- Department of Medicine, University Division of Anaesthesia, University of Cambridge, Cambridge, UK
| | - Virginia F J Newcombe
- Department of Medicine, University Division of Anaesthesia, University of Cambridge, Cambridge, UK
| |
Collapse
|
21
|
Wilde EA, Wanner I, Kenney K, Gill J, Stone JR, Disner S, Schnakers C, Meyer R, Prager EM, Haas M, Jeromin A. A Framework to Advance Biomarker Development in the Diagnosis, Outcome Prediction, and Treatment of Traumatic Brain Injury. J Neurotrauma 2022; 39:436-457. [PMID: 35057637 PMCID: PMC8978568 DOI: 10.1089/neu.2021.0099] [Citation(s) in RCA: 36] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Affiliation(s)
- Elisabeth A. Wilde
- University of Utah, Neurology, 383 Colorow, Salt Lake City, Utah, United States, 84108
- VA Salt Lake City Health Care System, 20122, 500 Foothill Dr., Salt Lake City, Utah, United States, 84148-0002
| | - Ina Wanner
- UCLA, Semel Institute, NRB 260J, 635 Charles E. Young Drive South, Los Angeles, United States, 90095-7332, ,
| | - Kimbra Kenney
- Uniformed Services University of the Health Sciences, Neurology, Center for Neuroscience and Regenerative Medicine, 4301 Jones Bridge Road, Bethesda, Maryland, United States, 20814
| | - Jessica Gill
- National Institutes of Health, National Institute of Nursing Research, 1 cloister, Bethesda, Maryland, United States, 20892
| | - James R. Stone
- University of Virginia, Radiology and Medical Imaging, Box 801339, 480 Ray C. Hunt Dr. Rm. 185, Charlottesville, Virginia, United States, 22903, ,
| | - Seth Disner
- Minneapolis VA Health Care System, 20040, Minneapolis, Minnesota, United States
- University of Minnesota Medical School Twin Cities, 12269, 10Department of Psychiatry and Behavioral Sciences, Minneapolis, Minnesota, United States
| | - Caroline Schnakers
- Casa Colina Hospital and Centers for Healthcare, 6643, Pomona, California, United States
- Ronald Reagan UCLA Medical Center, 21767, Los Angeles, California, United States
| | - Restina Meyer
- Cohen Veterans Bioscience, 476204, New York, New York, United States
| | - Eric M Prager
- Cohen Veterans Bioscience, 476204, External Affairs, 535 8th Ave, New York, New York, United States, 10018
| | - Magali Haas
- Cohen Veterans Bioscience, 476204, 535 8th Avenue, 12th Floor, New York City, New York, United States, 10018,
| | - Andreas Jeromin
- Cohen Veterans Bioscience, 476204, Translational Sciences, Cambridge, Massachusetts, United States
| |
Collapse
|
22
|
Bassi TG, Rohrs EC, Fernandez KC, Ornowska M, Nicholas M, Gani M, Evans D, Reynolds SC. Transvenous Diaphragm Neurostimulation Mitigates Ventilation-associated Brain Injury. Am J Respir Crit Care Med 2021; 204:1391-1402. [PMID: 34491883 PMCID: PMC8865722 DOI: 10.1164/rccm.202101-0076oc] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2021] [Accepted: 09/01/2021] [Indexed: 11/16/2022] Open
Abstract
Rationale: Mechanical ventilation (MV) is associated with hippocampal apoptosis and inflammation, and it is important to study strategies to mitigate them. Objectives: To explore whether temporary transvenous diaphragm neurostimulation (TTDN) in association with MV mitigates hippocampal apoptosis and inflammation after 50 hours of MV. Methods: Normal-lung porcine study comparing apoptotic index, inflammatory markers, and neurological-damage serum markers between never-ventilated subjects, subjects undergoing 50 hours of MV plus either TTDN every other breath or every breath, and subjects undergoing 50 hours of MV (MV group). MV settings in volume control were Vt of 8 ml/kg, and positive end-expiratory pressure of 5 cm H2O. Measurements and Main Results: Apoptotic indices, microglia percentages, and reactive astrocyte percentages were greater in the MV group in comparison with the other groups (P < 0.05). Transpulmonary pressure at baseline and at study end were both lower in the group receiving TTDN every breath, but lung injury scores and systemic inflammatory markers were not different between the groups. Serum concentrations of four neurological-damage markers were lower in the group receiving TTDN every breath than in the MV group (P < 0.05). Heart rate variability declined significantly in the MV group and increased significantly in both TTDN groups over the course of the experiments. Conclusions: Our study found that mechanical ventilation is associated with hippocampal apoptosis and inflammation, independent of lung injury and systemic inflammation. Also, in a porcine model, TTDN results in neuroprotection after 50 hours, and the degree of neuroprotection increases with greater exposure to TTDN.
Collapse
Affiliation(s)
- Thiago G. Bassi
- Simon Fraser University, Burnaby, British Columbia, Canada
- Lungpacer Medical Inc., Vancouver, British Columbia, Canada; and
| | - Elizabeth C. Rohrs
- Simon Fraser University, Burnaby, British Columbia, Canada
- Fraser Health Authority, Royal Columbian Hospital, New Westminster, British Columbia, Canada
| | - Karl C. Fernandez
- Simon Fraser University, Burnaby, British Columbia, Canada
- Fraser Health Authority, Royal Columbian Hospital, New Westminster, British Columbia, Canada
| | | | - Michelle Nicholas
- Simon Fraser University, Burnaby, British Columbia, Canada
- Fraser Health Authority, Royal Columbian Hospital, New Westminster, British Columbia, Canada
| | - Matt Gani
- Lungpacer Medical Inc., Vancouver, British Columbia, Canada; and
| | - Doug Evans
- Lungpacer Medical Inc., Vancouver, British Columbia, Canada; and
| | - Steven C. Reynolds
- Simon Fraser University, Burnaby, British Columbia, Canada
- Fraser Health Authority, Royal Columbian Hospital, New Westminster, British Columbia, Canada
| |
Collapse
|
23
|
Wang KK, Munoz Pareja JC, Mondello S, Diaz-Arrastia R, Wellington C, Kenney K, Puccio AM, Hutchison J, McKinnon N, Okonkwo DO, Yang Z, Kobeissy F, Tyndall JA, Büki A, Czeiter E, Pareja Zabala MC, Gandham N, Berman R. Blood-based traumatic brain injury biomarkers - Clinical utilities and regulatory pathways in the United States, Europe and Canada. Expert Rev Mol Diagn 2021; 21:1303-1321. [PMID: 34783274 DOI: 10.1080/14737159.2021.2005583] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
INTRODUCTION Traumatic brain injury (TBI) is a major global health issue, resulting in debilitating consequences to families, communities, and health-care systems. Prior research has found that biomarkers aid in the pathophysiological characterization and diagnosis of TBI. Significantly, the FDA has recently cleared both a bench-top assay and a rapid point-of-care assays of tandem biomarker (UCH-L1/GFAP)-based blood test to aid in the diagnosis mTBI patients. With the global necessity of TBI biomarkers research, several major consortium multicenter observational studies with biosample collection and biomarker analysis have been created in the USA, Europe, and Canada. As each geographical region regulates its data and findings, the International Initiative for Traumatic Brain Injury Research (InTBIR) was formed to facilitate data integration and dissemination across these consortia. AREAS COVERED This paper covers heavily investigated TBI biomarkers and emerging non-protein markers. Finally, we analyze the regulatory pathways for converting promising TBI biomarkers into approved in-vitro diagnostic tests in the United States, European Union, and Canada. EXPERT OPINION TBI biomarker research has significantly advanced in the last decade. The recent approval of an iSTAT point of care test to detect mild TBI has paved the way for future biomarker clearance and appropriate clinical use across the globe.
Collapse
Affiliation(s)
- Kevin K Wang
- Program for Neurotrauma, Neuroprotoemics & Biomarker Research, Department of Emergency Medicine, University of Florida College of Medicine, Gainesville, Florida, USA.,Brain Rehabilitation Research Center (BRRC), Malcom Randall Veterans Affairs Medical Center, Gainesville, Florida, USA
| | - Jennifer C Munoz Pareja
- Department of Pediatric Critical Care, University of Miami Miller School of Medicine, Miami, Florida, USA
| | - Stefania Mondello
- Department of Biomedical and Dental Sciences and Morphofunctional Imaging, University of Messina, Messina, Italy
| | - Ramon Diaz-Arrastia
- Department of Neurology, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| | - Cheryl Wellington
- Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Canada
| | - Kimbra Kenney
- Department of Neurology, Uniformed Service University, Bethesda, Maryland, USA
| | - Ava M Puccio
- Department of Neurological Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Jamie Hutchison
- The Hospital for Sick Children, Department of Critical Care Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Nicole McKinnon
- The Hospital for Sick Children, Department of Critical Care Medicine, University of Toronto, Toronto, Ontario, Canada
| | - David O Okonkwo
- Department of Neurological Surgery, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania, USA
| | - Zhihui Yang
- Program for Neurotrauma, Neuroprotoemics & Biomarker Research, Department of Emergency Medicine, University of Florida College of Medicine, Gainesville, Florida, USA.,Brain Rehabilitation Research Center (BRRC), Malcom Randall Veterans Affairs Medical Center, Gainesville, Florida, USA
| | - Firas Kobeissy
- Program for Neurotrauma, Neuroprotoemics & Biomarker Research, Department of Emergency Medicine, University of Florida College of Medicine, Gainesville, Florida, USA.,Brain Rehabilitation Research Center (BRRC), Malcom Randall Veterans Affairs Medical Center, Gainesville, Florida, USA
| | - J Adrian Tyndall
- Program for Neurotrauma, Neuroprotoemics & Biomarker Research, Department of Emergency Medicine, University of Florida College of Medicine, Gainesville, Florida, USA
| | | | - Endre Czeiter
- Department of Neurosurgery, Pecs University, Pecs, Hungary
| | | | - Nithya Gandham
- Program for Neurotrauma, Neuroprotoemics & Biomarker Research, Department of Emergency Medicine, University of Florida College of Medicine, Gainesville, Florida, USA
| | - Rebecca Berman
- National Institute of Neurological Disorders and Stroke, National Institute of Health, Bethesda, MD, USA
| | | |
Collapse
|
24
|
Ahmad-Molaei L, Pourhamzeh M, Ahadi R, Khodagholi F, Hassanian-Moghaddam H, Haghparast A. Time-Dependent Changes in the Serum Levels of Neurobiochemical Factors After Acute Methadone Overdose in Adolescent Male Rat. Cell Mol Neurobiol 2021; 41:1635-1649. [PMID: 32712727 PMCID: PMC11444013 DOI: 10.1007/s10571-020-00931-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2020] [Accepted: 07/20/2020] [Indexed: 12/20/2022]
Abstract
Acute methadone toxicity is a major public health concern which has adverse effects on brain tissue and results in recurrent or delayed respiratory arrest. Our study aimed to investigate the time-dependent changes in several serum biochemical markers of brain damage, spatial working memory, and the brain tissue following acute methadone overdose. Adolescent male rats underwent an intraperitoneal (i.p.) injection of 15 mg/kg methadone. In case of apnea occurrence, resuscitation was performed by a ventilatory pump and administrating naloxone (2 mg/kg; i.p.). The animals were classified into groups of treated rats; methadone and naloxone-Apnea (M/N-Apnea), M/N-Sedate, Methadone, Naloxone, and control (saline) groups. The serum levels of S100B, neuron-specific enolase (NSE), myelin basic protein factors, and (Lactate/Pyruvate) L/P ratio were evaluated at the time-points of 6, 24, and 48 h (h). We found that the alterations of S100B and L/P ratio were considerable in the M/N-Apnea and Methadone groups from the early hours post-methadone overdose, while NSE serum levels elevation was observed only in M/N-Apnea group with a delay at 48 h. Further, we assessed the spatial working memory (Y-maze test), morphological changes, and neuronal loss. The impaired spontaneous alternation behavior was detected in the M/N-Apnea groups on days 5 and 10 post-methadone overdose. The morphological changes of neurons and the neuronal loss were detectable in the CA1, striatum, and cerebellum regions, which were pronounced in both M/N-Apnea and Methadone groups. Together, our findings suggest that alterations in the serum levels of S100B and NSE factors as well as L/P ratio could be induced by methadone overdose with the presence or absence of apnea before the memory impairment and tissue injury in adolescent male rats.
Collapse
Affiliation(s)
- Leila Ahmad-Molaei
- Neuroscience Research Center, School of Medicine, Shahid Beheshti University of Medical Sciences, P.O. Box: 19615-1178, Tehran, Iran
| | - Mahsa Pourhamzeh
- Department of Anatomy, Faculty of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Reza Ahadi
- Department of Anatomy, Faculty of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Fariba Khodagholi
- Neuroscience Research Center, School of Medicine, Shahid Beheshti University of Medical Sciences, P.O. Box: 19615-1178, Tehran, Iran
| | - Hossein Hassanian-Moghaddam
- Department of Clinical Toxicology, Loghman-Hakim Hospital, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- Social Determinants of Health Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Abbas Haghparast
- Neuroscience Research Center, School of Medicine, Shahid Beheshti University of Medical Sciences, P.O. Box: 19615-1178, Tehran, Iran.
| |
Collapse
|
25
|
McDonald SJ, Shultz SR, Agoston DV. The Known Unknowns: An Overview of the State of Blood-Based Protein Biomarkers of Mild Traumatic Brain Injury. J Neurotrauma 2021; 38:2652-2666. [PMID: 33906422 DOI: 10.1089/neu.2021.0011] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Blood-based protein biomarkers have revolutionized several fields of medicine by enabling molecular level diagnosis, as well as monitoring disease progression and treatment efficacy. Traumatic brain injury (TBI) so far has benefitted only moderately from using protein biomarkers to improve injury outcome. Because of its complexity and dynamic nature, TBI, especially its most prevalent mild form (mild TBI; mTBI), presents unique challenges toward protein biomarker discovery and validation given that blood is frequently obtained and processed outside of the clinical laboratory (e.g., athletic fields, battlefield) under variable conditions. As it stands, the field of mTBI blood biomarkers faces a number of outstanding questions. Do elevated blood levels of currently used biomarkers-ubiquitin carboxy-terminal hydrolase L1, glial fibrillary acidic protein, neurofilament light chain, and tau/p-tau-truly mirror the extent of parenchymal damage? Do these different proteins represent distinct injury mechanisms? Is the blood-brain barrier a "brick wall"? What is the relationship between intra- versus extracranial values? Does prolonged elevation of blood levels reflect de novo release or extended protein half-lives? Does biological sex affect the pathobiological responses after mTBI and thus blood levels of protein biomarkers? At the practical level, it is unknown how pre-analytical variables-sample collection, preparation, handling, and stability-affect the quality and reliability of biomarker data. The ever-increasing sensitivity of assay systems and lack of quality control of samples, combined with the almost complete reliance on antibody-based assay platforms, represent important unsolved issues given that false-negative results can lead to false clinical decision making and adverse outcomes. This article serves as a commentary on the state of mTBI biomarkers and the landscape of significant challenges. We highlight and discusses several biological and methodological "known unknowns" and close with some practical recommendations.
Collapse
Affiliation(s)
- Stuart J McDonald
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, Victoria, Australia
- Department of Physiology, Anatomy and Microbiology, School of Life Sciences, La Trobe University, Bundoora, Victoria, Australia
| | - Sandy R Shultz
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, Victoria, Australia
- Department of Medicine, Royal Melbourne Hospital, The University of Melbourne, Parkville, Victoria, Australia
| | - Denes V Agoston
- Department of Anatomy, Physiology and Genetics, Uniformed Services University of the Health Sciences, Bethesda, Maryland, USA
| |
Collapse
|
26
|
Mozaffari K, Dejam D, Duong C, Ding K, French A, Ng E, Preet K, Franks A, Kwan I, Phillips HW, Kim DY, Yang I. Systematic Review of Serum Biomarkers in Traumatic Brain Injury. Cureus 2021; 13:e17056. [PMID: 34522534 PMCID: PMC8428323 DOI: 10.7759/cureus.17056] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/10/2021] [Indexed: 12/20/2022] Open
Abstract
Traumatic brain injury (TBI) is responsible for the majority of trauma-related deaths and is a leading cause of disability. It is characterized by an inflammatory process involved in the progression of secondary brain injury. TBI is measured by the Glasgow Coma Scale (GCS) with scores ranging from 15-3, demonstrating mild to severe brain injury. Apart from this clinical assessment of TBI, compendiums of literature have been published on TBI-related serum markers.Herein we create a comprehensive appraisal of the most prominent serum biomarkers used in the assessment and care of TBI.The PubMed, Scopus, Cochrane, and Web of Science databases were queried with the terms “biomarker” and “traumatic brain injury” as search terms with only full-text, English articles within the past 10 years selected. Non-human studies were excluded, and only adult patients fell within the purview of this analysis. A total of 528 articles were analyzed in the initial search with 289 selected for screening. A further 152 were excluded for primary screening. Of the remaining 137, 54 were included in the final analysis. Serum biomarkers were listed into the following broad categories for ease of discussion: immune markers and markers of inflammation, hormones as biomarkers, coagulation and vasculature, genetic polymorphisms, antioxidants and oxidative stress, apoptosis and degradation pathways, and protein markers. Glial fibrillary acidic protein(GFAP), S100, and neurons specific enolase (NSE) were the most prominent and frequently cited markers. Amongst these three, no single serum biomarker demonstrated neither superior sensitivity nor specificity compared to the other two, therefore noninvasive panels should incorporate these three serum biomarkers to retain sensitivity and maximize specificity for TBI.
Collapse
Affiliation(s)
- Khashayar Mozaffari
- Neurosurgery, Ronald Reagan University of California Los Angeles Medical Center, Los Angeles, USA
| | - Dillon Dejam
- Neurosurgery, David Geffen School of Medicine, University of California, Los Angeles, USA
| | - Courtney Duong
- Neurosurgery, David Geffen School of Medicine, University of California, Los Angeles, USA
| | - Kevin Ding
- Neurosurgery, Ronald Reagan University of California Los Angeles Medical Center, Los Angeles, USA
| | - Alexis French
- Neurosurgery, Ronald Reagan University of California Los Angeles Medical Center, Los Angeles, USA
| | - Edwin Ng
- Neurosurgery, Ronald Reagan University of California Los Angeles Medical Center, Los Angeles, USA
| | - Komal Preet
- Neurosurgery, University of California, Los Angeles, USA
| | - Alyssa Franks
- Neurosurgery, Ronald Reagan University of California Los Angeles Medical Center, Los Angeles, USA
| | - Isabelle Kwan
- Neurosurgery, Ronald Reagan University of California Los Angeles Medical Center, Los Angeles, USA
| | - H Westley Phillips
- Neurosurgery, Ronald Reagan University of California Los Angeles Medical Center, Los Angeles, USA
| | - Dennis Y Kim
- Biomedical Sciences, Harbor University of California Los Angeles Medical Center, Los Angeles, USA
| | - Isaac Yang
- Neurosurgery, David Geffen School of Medicine, University of California, Los Angeles, USA
| |
Collapse
|
27
|
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
|
28
|
Huie JR, Mondello S, Lindsell CJ, Antiga L, Yuh EL, Zanier ER, Masson S, Rosario BL, Ferguson AR. Biomarkers for Traumatic Brain Injury: Data Standards and Statistical Considerations. J Neurotrauma 2021; 38:2514-2529. [PMID: 32046588 PMCID: PMC8403188 DOI: 10.1089/neu.2019.6762] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
Abstract
Recent biomarker innovations hold potential for transforming diagnosis, prognostic modeling, and precision therapeutic targeting of traumatic brain injury (TBI). However, many biomarkers, including brain imaging, genomics, and proteomics, involve vast quantities of high-throughput and high-content data. Management, curation, analysis, and evidence synthesis of these data are not trivial tasks. In this review, we discuss data management concepts and statistical and data sharing strategies when dealing with biomarker data in the context of TBI research. We propose that application of biomarkers involves three distinct steps-discovery, evaluation, and evidence synthesis. First, complex/big data has to be reduced to useful data elements at the stage of biomarker discovery. Second, inferential statistical approaches must be applied to these biomarker data elements for assessment of biomarker clinical utility and validity. Last, synthesis of relevant research is required to support practice guidelines and enable health decisions informed by the highest quality, up-to-date evidence available. We focus our discussion around recent experiences from the International Traumatic Brain Injury Research (InTBIR) initiative, with a specific focus on four major clinical projects (Transforming Research and Clinical Knowledge in TBI, Collaborative European NeuroTrauma Effectiveness Research in TBI, Collaborative Research on Acute Traumatic Brain Injury in Intensive Care Medicine in Europe, and Approaches and Decisions in Acute Pediatric TBI Trial), which are currently enrolling subjects in North America and Europe. We discuss common data elements, data collection efforts, data-sharing opportunities, and challenges, as well as examine the statistical techniques required to realize successful adoption and use of biomarkers in the clinic as a foundation for precision medicine in TBI.
Collapse
Affiliation(s)
- J. Russell Huie
- Brain and Spinal Injury Center, Department of Neurological Surgery, University of California San Francisco, San Francisco, California, USA
| | - Stefania Mondello
- Department of Biomedical and Dental Sciences and Morphofunctional Imaging, University of Messina, Messina, Italy
| | - Christopher J. Lindsell
- Department of Biostatistics, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | | | - Esther L. Yuh
- Brain and Spinal Injury Center, Department of Neurological Surgery, University of California San Francisco, San Francisco, California, USA
| | - Elisa R. Zanier
- Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milano, Italy
| | - Serge Masson
- Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milano, Italy
| | - Bedda L. Rosario
- Department of Epidemiology, University of Pittsburgh Graduate School of Public Health, Pittsburgh, Pennsylvania, USA
| | - Adam R. Ferguson
- Brain and Spinal Injury Center, Department of Neurological Surgery, University of California San Francisco, San Francisco, California, USA
- San Francisco Veterans Affairs Medical Center (SFVAMC), San Francisco, California, USA
| |
Collapse
|
29
|
Kearns J, Ross AM, Walsh DR, Cahalane RM, Hinchion R, Ryan MC, Conway E, Comyns TM, Kenny IC, O'Connor EM, McGourty KD, Mulvihill JJE. A blood biomarker and clinical correlation cohort study protocol to diagnose sports-related concussion and monitor recovery in elite rugby. BMJ Open Sport Exerc Med 2021; 6:e000948. [PMID: 34422289 PMCID: PMC8323462 DOI: 10.1136/bmjsem-2020-000948] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Revised: 10/14/2020] [Accepted: 10/30/2020] [Indexed: 11/04/2022] Open
Abstract
Introduction In professional rugby, sports-related concussion (SRC) remains the most frequent time loss injury. Therefore, accurately diagnosing SRC and monitoring player recovery, through a multi-modal assessment process, is critical to SRC management. In this protocol study, we aim to assess SRC over multiple time points post-injury to determine the value of multi-modal assessments to monitor player recovery. This is of significance to minimise premature return-to-play and, ultimately, to reduce the long-term effects associated with SRC. The study will also establish the logistics of implementing such a study in a professional setting to monitor a player's SRC recovery. Methods and analysis All players from the participating professional rugby club within the Irish Rugby Football Union are invited to participate in the current study. Player assessment includes head injury assessment (HIA), neuropsychometric assessment (ImPACT), targeted biomarker analysis and untargeted biomarker analysis. Baseline HIA, ImPACT, and blood draws are performed prior to the start of playing season. During the baseline tests, player's complete consent forms and an SRC history questionnaire. Subsequently, any participant that enters the HIA process over the playing season due to a suspected SRC will be clinically assessed (HIA and ImPACT) and their blood will be drawn within 3 days of injury, 6 days post-injury, and 13 days post-injury. Ethics and dissemination Ethical approval was attained from the Science and Engineering Research Ethics Committee, University of Limerick (Approval Code: 2018_06_11_S&E). On completion of the study, further manuscripts will be published to present the results of the tests and their ability to measure player recovery from SRC. Trial registration number NCT04485494.
Collapse
Affiliation(s)
- Jamie Kearns
- Munster Rugby Club, High Performance Centre, Limerick, Ireland
| | - Aisling M Ross
- School of Engineering, University of Limerick, Limerick, Ireland
| | - Darragh R Walsh
- School of Engineering, University of Limerick, Limerick, Ireland
| | | | - Rita Hinchion
- Clinical Research Support Unit, University Hospital Limerick, Limerick, Ireland.,Health Research Institute, University of Limerick, Limerick, Ireland
| | - Maria C Ryan
- Clinical Research Support Unit, University Hospital Limerick, Limerick, Ireland.,Health Research Institute, University of Limerick, Limerick, Ireland
| | - Elaine Conway
- Clinical Research Support Unit, University Hospital Limerick, Limerick, Ireland.,Health Research Institute, University of Limerick, Limerick, Ireland
| | - Tom M Comyns
- Health Research Institute, University of Limerick, Limerick, Ireland
| | - Ian C Kenny
- Health Research Institute, University of Limerick, Limerick, Ireland.,Physical Education and Sport Sciences, University of Limerick, Limerick, Ireland
| | - Eibhlís M O'Connor
- Health Research Institute, University of Limerick, Limerick, Ireland.,Biological Sciences, University of Limerick, Limerick, Ireland.,Bernal Institute, University of Limerick, Limerick, Ireland
| | - Kieran D McGourty
- Health Research Institute, University of Limerick, Limerick, Ireland.,Bernal Institute, University of Limerick, Limerick, Ireland.,Chemical Sciences, University of Limerick, Limerick, Ireland
| | - John Joseph Eugene Mulvihill
- School of Engineering, University of Limerick, Limerick, Ireland.,Health Research Institute, University of Limerick, Limerick, Ireland.,Bernal Institute, University of Limerick, Limerick, Ireland
| |
Collapse
|
30
|
Sayeed I, Stein DG. Response to Korley et al.: Progesterone Treatment Does Not Decrease Serum Levels of Biomarkers of Glial and Neuronal Cell Injury in Moderate and Severe TBI Subjects: A Secondary Analysis of the Progesterone for Traumatic Brain Injury, Experimental Clinical Treatment (ProTECT) III Trial (DOI: 10.1089/neu.2020.7072). J Neurotrauma 2021; 38:2923-2926. [PMID: 34130480 DOI: 10.1089/neu.2021.0064] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023] Open
Affiliation(s)
- Iqbal Sayeed
- Department of Emergency Medicine, Emory University, Atlanta, Georgia, USA
| | - Donald G Stein
- Department of Emergency Medicine, Emory University, Atlanta, Georgia, USA
| |
Collapse
|
31
|
Korley F, Pauls Q, Yeatts SD, Jones CMC, Corbett-Valade E, Silbergleit R, Frankel M, Barsan W, Cahill ND, Bazarian JJ, Wright DW. Progesterone Treatment Does Not Decrease Serum Levels of Biomarkers of Glial and Neuronal Cell Injury in Moderate and Severe Traumatic Brain Injury Subjects: A Secondary Analysis of the Progesterone for Traumatic Brain Injury, Experimental Clinical Treatment (ProTECT) III Trial. J Neurotrauma 2021; 38:1953-1960. [PMID: 33319651 PMCID: PMC8260894 DOI: 10.1089/neu.2020.7072] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Early treatment of moderate/severe traumatic brain injury (TBI) with progesterone does not improve clinical outcomes. This is in contrast with findings from pre-clinical studies of progesterone in TBI. To understand the reasons for the negative clinical trial, we investigated whether progesterone treatment has the desired biological effect of decreasing brain cell death. We quantified brain cell death using serum levels of biomarkers of glial and neuronal cell death (glial fibrillary acidic protein [GFAP], ubiquitin carboxy-terminal hydrolase-L1 [UCH-L1], S100 calcium-binding protein B [S100B], and Alpha II Spectrin Breakdown Product 150 [SBDP]) in the Biomarkers of Injury and Outcome-Progesterone for Traumatic Brain Injury, Experimental Clinical Treatment (BIO-ProTECT) study. Serum levels of GFAP, UCHL1, S100B, and SBDP were measured at baseline (≤4 h post-injury and before administration of study drug) and at 24 and 48 h post-injury. Serum progesterone levels were measured at 24 and 48 h post-injury. The primary outcome of ProTECT was based on the Glasgow Outcome Scale-Extended assessed at 6 months post-randomization. We found that at baseline, there were no differences in biomarker levels between subjects randomized to progesterone treatment and those randomized to placebo (p > 0.10). Similarly, at 24 and 48 h post-injury, there were no differences in biomarker levels in the progesterone versus placebo groups (p > 0.15). There was no statistically significant correlation between serum progesterone concentrations and biomarker values obtained at 24 and 48 h. When examined as a continuous variable, baseline biomarker levels did not modify the association between progesterone treatment and neurological outcome (p of interaction term >0.39 for all biomarkers). We conclude that progesterone treatment does not decrease levels of biomarkers of glial and neuronal cell death during the first 48 h post-injury.
Collapse
Affiliation(s)
- Frederick Korley
- Department of Emergency Medicine, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - Qi Pauls
- Department of Public Health Sciences, Medical University of South Carolina, Charleston, South Carolina, USA
| | - Sharon D. Yeatts
- Department of Public Health Sciences, Medical University of South Carolina, Charleston, South Carolina, USA
| | - Courtney Marie Cora Jones
- Departments of Emergency Medicine, Neurosurgery, and Physical Medicine and Rehabilitation, School of Medicine and Dentistry, University of Rochester, Rochester, New York, USA
- Public Health Sciences, Neurosurgery, and Physical Medicine and Rehabilitation, School of Medicine and Dentistry, University of Rochester, Rochester, New York, USA
| | - Emily Corbett-Valade
- Departments of Emergency Medicine, Neurosurgery, and Physical Medicine and Rehabilitation, School of Medicine and Dentistry, University of Rochester, Rochester, New York, USA
| | - Robert Silbergleit
- Department of Emergency Medicine, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - Michael Frankel
- Department of Neurology, Grady Memorial Hospital, Marcus Stroke and Neuroscience Center, School of Medicine, Emory University, Atlanta, Georgia, USA
| | - William Barsan
- Department of Emergency Medicine, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - Nathan D. Cahill
- School of Mathematical Sciences, Rochester Institute of Technology, Rochester, New York, USA
| | - Jeffrey J. Bazarian
- Departments of Emergency Medicine, Neurosurgery, and Physical Medicine and Rehabilitation, School of Medicine and Dentistry, University of Rochester, Rochester, New York, USA
- Public Health Sciences, Neurosurgery, and Physical Medicine and Rehabilitation, School of Medicine and Dentistry, University of Rochester, Rochester, New York, USA
- Departments of Neurology, Neurosurgery, and Physical Medicine and Rehabilitation, School of Medicine and Dentistry, University of Rochester, Rochester, New York, USA
| | - David W. Wright
- Department of Emergency Medicine, Grady Memorial Hospital, Marcus Stroke and Neuroscience Center, School of Medicine, Emory University, Atlanta, Georgia, USA
| |
Collapse
|
32
|
Azizi S, Hier DB, Allen B, Obafemi-Ajayi T, Olbricht GR, Thimgan MS, Wunsch DC. A Kinetic Model for Blood Biomarker Levels After Mild Traumatic Brain Injury. Front Neurol 2021; 12:668606. [PMID: 34295300 PMCID: PMC8289906 DOI: 10.3389/fneur.2021.668606] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2021] [Accepted: 06/09/2021] [Indexed: 01/23/2023] Open
Abstract
Traumatic brain injury (TBI) imposes a significant economic and social burden. The diagnosis and prognosis of mild TBI, also called concussion, is challenging. Concussions are common among contact sport athletes. After a blow to the head, it is often difficult to determine who has had a concussion, who should be withheld from play, if a concussed athlete is ready to return to the field, and which concussed athlete will develop a post-concussion syndrome. Biomarkers can be detected in the cerebrospinal fluid and blood after traumatic brain injury and their levels may have prognostic value. Despite significant investigation, questions remain as to the trajectories of blood biomarker levels over time after mild TBI. Modeling the kinetic behavior of these biomarkers could be informative. We propose a one-compartment kinetic model for S100B, UCH-L1, NF-L, GFAP, and tau biomarker levels after mild TBI based on accepted pharmacokinetic models for oral drug absorption. We approximated model parameters using previously published studies. Since parameter estimates were approximate, we did uncertainty and sensitivity analyses. Using estimated kinetic parameters for each biomarker, we applied the model to an available post-concussion biomarker dataset of UCH-L1, GFAP, tau, and NF-L biomarkers levels. We have demonstrated the feasibility of modeling blood biomarker levels after mild TBI with a one compartment kinetic model. More work is needed to better establish model parameters and to understand the implications of the model for diagnostic use of these blood biomarkers for mild TBI.
Collapse
Affiliation(s)
- Sima Azizi
- Applied Computational Intelligence Laboratory, Department of Electrical and Computer Engineering, Missouri University of Science and Technology, Rolla, MO, United States
| | - Daniel B Hier
- Applied Computational Intelligence Laboratory, Department of Electrical and Computer Engineering, Missouri University of Science and Technology, Rolla, MO, United States
| | - Blaine Allen
- Applied Computational Intelligence Laboratory, Department of Electrical and Computer Engineering, Missouri University of Science and Technology, Rolla, MO, United States
| | - Tayo Obafemi-Ajayi
- Engineering Program, Missouri State University, Springfield, MO, United States
| | - Gayla R Olbricht
- Department of Mathematics and Statistics, Missouri University of Science and Technology, Rolla, MO, United States
| | - Matthew S Thimgan
- Department of Biological Sciences, Missouri University of Science and Technology, Rolla, MO, United States
| | - Donald C Wunsch
- Applied Computational Intelligence Laboratory, Department of Electrical and Computer Engineering, Missouri University of Science and Technology, Rolla, MO, United States.,ECCS Division, National Science Foundation, Alexandria, VA, United States
| |
Collapse
|
33
|
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
|
34
|
Brain injury after 50 h of lung-protective mechanical ventilation in a preclinical model. Sci Rep 2021; 11:5105. [PMID: 33658581 PMCID: PMC7930247 DOI: 10.1038/s41598-021-84440-1] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2020] [Accepted: 02/15/2021] [Indexed: 01/12/2023] Open
Abstract
Mechanical ventilation is the cornerstone of the Intensive Care Unit. However, it has been associated with many negative consequences. Recently, ventilator-induced brain injury has been reported in rodents under injurious ventilation settings. Our group wanted to explore the extent of brain injury after 50 h of mechanical ventilation, sedation and physical immobility, quantifying hippocampal apoptosis and inflammation, in a normal-lung porcine study. After 50 h of lung-protective mechanical ventilation, sedation and immobility, greater levels of hippocampal apoptosis and neuroinflammation were clearly observed in the mechanically ventilated group, in comparison to a never-ventilated group. Markers in the serum for astrocyte damage and neuronal damage were also higher in the mechanically ventilated group. Therefore, our study demonstrated that considerable hippocampal insult can be observed after 50 h of lung-protective mechanical ventilation, sedation and physical immobility.
Collapse
|
35
|
Muñoz ER, Caccese JB, Wilson BE, Shuler KT, Santos FV, Cabán CT, Jeka JJ, Langford D, Hudson MB. Effects of purposeful soccer heading on circulating small extracellular vesicle concentration and cargo. JOURNAL OF SPORT AND HEALTH SCIENCE 2021; 10:122-130. [PMID: 33189894 PMCID: PMC7987560 DOI: 10.1016/j.jshs.2020.11.006] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/01/2020] [Revised: 08/29/2020] [Accepted: 09/22/2020] [Indexed: 05/09/2023]
Abstract
BACKGROUND Considering the potential cumulative effects of repetitive head impact (HI) exposure, we need sensitive biomarkers to track short- and long-term effects. Circulating small extracellular vesicles (sEVs) (<200 nm) traffic biological molecules throughout the body and may have diagnostic value as biomarkers for disease. The purpose of this study was to identify the microRNA (miRNA) profile in circulating sEVs derived from human plasma following repetitive HI exposure. METHODS Healthy adult (aged 18-35 years) soccer players were randomly assigned to one of 3 groups: the HI group performed 10 standing headers, the leg impact group performed 10 soccer ball trapping maneuvers over 10 min, and the control group did not participate in any soccer drills. Plasma was collected before testing and 24 h afterward, and sEVs were isolated and characterized via nanoparticle tracking analysis. Next-generation sequencing was utilized to identify candidate miRNAs isolated from sEVs, and candidate microRNAs were analyzed via quantitative polymerase chain reaction. In silico target prediction was performed using TargetScan (Version 7.0; targetscan.org) and miRWalk (http://mirwalk.umm.uni-heidelberg.de/) programs, and target validation was performed using luciferase reporter vectors with a miR-7844-5p mimic in human embryonic kidney (HEK) 293T/17 cells. RESULTS Plasma sEV concentration and size were not affected across time and group following repetitive HI exposure. After 24 h, the HI read count from next-generation sequencing showed a 4-fold or greater increase in miR-92b-5p, miR-423-5p, and miR-24-3p and a 3-fold or greater decrease in miR-7844-5p, miR-144-5p, miR-221-5p, and miR-22-3p. Analysis of quantitative polymerase chain reaction revealed that leg impact did not alter the candidate miRNA levels. To our knowledge, miR-7844-5p is a previously unknown miRNA. We identified 8 miR-7844-5p mRNA targets: protein phosphatase 1 regulatory inhibitor subunit 1B (PPP1R1B), LIM and senescent cell antigen-like domains 1 (LIMS1), autophagy-related 12 (ATG12), microtubule-associated protein 1 light chain 3 beta (MAP1LC3B), integrin subunit alpha-1 (ITGA1), mitogen-activated protein kinase 1 (MAPK1), glycogen synthase kinase 3β (GSK3β), and mitogen-activated protein kinase 8 (MAPK8). CONCLUSION Collectively, these data indicate repetitive HI exposure alters plasma sEV miRNA content, but not sEV size or number. Furthermore, for the first time we demonstrate that previously unknown miR-7844-5p targets mRNAs known to be involved in mitochondrial apoptosis, autophagy regulation, mood disorders, and neurodegenerative disease.
Collapse
Affiliation(s)
- Eric R Muñoz
- Department of Kinesiology and Applied Physiology, University of Delaware, Newark, DE 19713, USA
| | - Jaclyn B Caccese
- School of Health and Rehabilitation Sciences, The Ohio State University College of Medicine, Columbus, OH 43210, USA
| | - Brittany E Wilson
- Department of Kinesiology and Applied Physiology, University of Delaware, Newark, DE 19713, USA
| | - Kyle T Shuler
- Department of Kinesiology and Applied Physiology, University of Delaware, Newark, DE 19713, USA
| | - Fernando V Santos
- Department of Kinesiology and Applied Physiology, University of Delaware, Newark, DE 19713, USA
| | - Carolina T Cabán
- Department of Neuroscience, Lewis Katz School of Medicine, Temple University, Philadelphia, PA 19140, USA
| | - John J Jeka
- Department of Kinesiology and Applied Physiology, University of Delaware, Newark, DE 19713, USA
| | - Dianne Langford
- Department of Neuroscience, Lewis Katz School of Medicine, Temple University, Philadelphia, PA 19140, USA
| | - Matthew B Hudson
- Department of Kinesiology and Applied Physiology, University of Delaware, Newark, DE 19713, USA.
| |
Collapse
|
36
|
Snowden T, Reid H, Kennedy S, Kenny R, McQuarrie A, Stuart-Hill L, Garcia-Barrera MA, Gawryluk J, Christie BR. Heading in the Right Direction: A Critical Review of Studies Examining the Effects of Heading in Soccer Players. J Neurotrauma 2020; 38:169-188. [PMID: 32883162 DOI: 10.1089/neu.2020.7130] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
The practice of heading in soccer has become a public concern because of the potential for subconcussive impacts to cause cumulative concussive-like effects; however, experimental evidence for this hypothesis has been mixed. This systematic review used pre-defined search parameters to assess primary literature that examined changes in cognitive, behavioral, structural, and/or biological processes after acute heading exposure in youth and young adult soccer players. The findings were synthesized into a concise and comprehensive summary of the research following the preferred reporting items for systematic reviews and meta-analyses (PRISMA) format, and suggestions for standardization of acute heading protocols are described. A total of 1189 articles were considered for this review, with 19 articles meeting all of the inclusion criteria for full analysis. An attempt was made to identify methods with significant sensitivity and reliability by grouping studies based on their outcome measures. Because of lack of standardization across intervention types and data collection protocols, no sensitive and reliable methods could be identified conclusively to assess the effects of acute heading exposure in soccer players. Based on this review, there is not enough evidence to either support or refute the potential of effects of subconcussive events from acute soccer heading exposure. Recommendations for standardization of acute heading exposure studies based on the included literature are discussed. Standardization is required to better understand the impact of acute heading exposure in soccer players, while allowing for the development of guidelines that mitigate any potential risks and allowing athletes to remain active and develop their skills.
Collapse
Affiliation(s)
- Taylor Snowden
- Division of Medical Sciences, University of Victoria, Victoria, British Columbia, Canada
| | - Hannah Reid
- Division of Medical Sciences, University of Victoria, Victoria, British Columbia, Canada
| | - Samantha Kennedy
- Division of Medical Sciences, University of Victoria, Victoria, British Columbia, Canada.,Island Medical Program, University of British Columbia, Victoria, British Columbia, Canada
| | - Rebecca Kenny
- Division of Medical Sciences, University of Victoria, Victoria, British Columbia, Canada
| | - Amanda McQuarrie
- Division of Medical Sciences, University of Victoria, Victoria, British Columbia, Canada
| | - Lynneth Stuart-Hill
- School of Exercise Science, Physical and Health Education, and University of Victoria, Victoria, British Columbia, Canada
| | | | - Jodie Gawryluk
- Department of Psychology, University of Victoria, Victoria, British Columbia, Canada
| | - Brian R Christie
- Division of Medical Sciences, University of Victoria, Victoria, British Columbia, Canada.,Island Medical Program, University of British Columbia, Victoria, British Columbia, Canada.,School of Exercise Science, Physical and Health Education, and University of Victoria, Victoria, British Columbia, Canada
| |
Collapse
|
37
|
A general model for cell death and biomarker release from injured tissues. J Pharmacokinet Pharmacodyn 2020; 48:69-82. [PMID: 32996046 DOI: 10.1007/s10928-020-09720-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2020] [Accepted: 09/18/2020] [Indexed: 10/23/2022]
Abstract
Cellular response to insults may result in the initiation of different cell death processes. For many cases the cell death process will result in an acute release of cellular material that in some circumstances provides valuable information about the process (i.e. may represent a biomarker). The characteristics of the biomarker release is often informative and plays critical roles in clinical practice and toxicology research. The aim of this study is to develop a general, semi-mechanistic model to describe cell turnover and biomarker release by injured tissue that can be used for estimation in pharmacokinetic and (toxicokinetic)-pharmacodynamic studies. The model included three components: (1) natural tissue turnover, (2) biomarker release from cell death and its movement from the cell through the tissue into the blood, (3) different target insult mechanisms of cell death. We applied the general model to biomarker release profiles for four different cell insult causes. Our model simulations showed good agreements with reported data under both delayed release and rapid release cases. Additionally, we illustrate the use of the model to provide different biomarker profiles. We also provided details on interpreting parameters and their values for other researchers to customize its use. In conclusion, our general model provides a basic structure to study the kinetic behaviour of biomarker release and disposition after cellular insult.
Collapse
|
38
|
Kjerulff JL, Seidenfaden SC, Juul N, Møller MF, Munster AMB, Bøtker MT. Influence of Simulated Pre-Hospital Transport, Time to Analysis, and Storage Temperature on S100 Calcium-Binding Protein B Values. J Neurotrauma 2020; 37:1864-1869. [DOI: 10.1089/neu.2019.6907] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Affiliation(s)
- Julie Linding Kjerulff
- Prehospital Emergency Medical Services, Research and Development, Central Denmark Region, Aarhus, Denmark
| | | | - Niels Juul
- Department of Anesthesiology, Section North, Aarhus University Hospital, Central Denmark Region, Aarhus, Denmark
| | - Mette Fogh Møller
- Department of Clinical Biochemistry, Regional Hospital Herning, Central Denmark Region, Herning, Denmark
| | | | - Morten Thingemann Bøtker
- Prehospital Emergency Medical Services, Research and Development, Central Denmark Region, Aarhus, Denmark
| |
Collapse
|
39
|
A multi-staged neuropeptide response to traumatic brain injury. Eur J Trauma Emerg Surg 2020; 48:507-517. [DOI: 10.1007/s00068-020-01431-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2020] [Accepted: 06/28/2020] [Indexed: 01/05/2023]
|
40
|
Schindler CR, Lustenberger T, Woschek M, Störmann P, Henrich D, Radermacher P, Marzi I. Severe Traumatic Brain Injury (TBI) Modulates the Kinetic Profile of the Inflammatory Response of Markers for Neuronal Damage. J Clin Med 2020; 9:jcm9061667. [PMID: 32492963 PMCID: PMC7356222 DOI: 10.3390/jcm9061667] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2020] [Revised: 05/22/2020] [Accepted: 05/26/2020] [Indexed: 02/06/2023] Open
Abstract
The inflammatory response plays an important role in the pathophysiology of multiple injuries. This study examines the effects of severe trauma and inflammatory response on markers of neuronal damage. A retrospective analysis of prospectively collected data in 445 trauma patients (Injury Severity Score (ISS) ≥ 16) is provided. Levels of neuronal biomarkers (calcium-binding Protein B (S100b), Enolase2 (NSE), glial fibrillary acidic protein (GFAP)) and Interleukins (IL-6, IL-10) in severely injured patients (with polytrauma (PT)) without traumatic brain injury (TBI) or with severe TBI (PT+TBI) and patients with isolated TBI (isTBI) were measured upon arrival until day 5. S100b, NSE, GFAP levels showed a time-dependent decrease in all cohorts. Their expression was higher after multiple injuries (p = 0.038) comparing isTBI. Positive correlation of marker level after concomitant TBI and isTBI (p = 0.001) was noted, while marker expression after PT appears to be independent. Highest levels of IL-6 and -10 were associated to PT und lowest to isTBI (p < 0.001). In all groups pro-inflammatory response (IL-6/-10 ratio) peaked on day 2 and at a lower level on day 4. Severe TBI modulates kinetic profile of inflammatory response by reducing interleukin expression following trauma. Potential markers for neuronal damage have a limited diagnostic value after severe trauma because undifferentiated increase.
Collapse
Affiliation(s)
- Cora Rebecca Schindler
- Department of Trauma, Hand and Reconstructive Surgery, University Hospital Frankfurt, 60596 Frankfurt, Germany; (T.L.); (M.W.); (P.S.); (D.H.); (I.M.)
- Correspondence: ; Tel./Fax: +49-69-6301-83304
| | - Thomas Lustenberger
- Department of Trauma, Hand and Reconstructive Surgery, University Hospital Frankfurt, 60596 Frankfurt, Germany; (T.L.); (M.W.); (P.S.); (D.H.); (I.M.)
| | - Mathias Woschek
- Department of Trauma, Hand and Reconstructive Surgery, University Hospital Frankfurt, 60596 Frankfurt, Germany; (T.L.); (M.W.); (P.S.); (D.H.); (I.M.)
| | - Philipp Störmann
- Department of Trauma, Hand and Reconstructive Surgery, University Hospital Frankfurt, 60596 Frankfurt, Germany; (T.L.); (M.W.); (P.S.); (D.H.); (I.M.)
| | - Dirk Henrich
- Department of Trauma, Hand and Reconstructive Surgery, University Hospital Frankfurt, 60596 Frankfurt, Germany; (T.L.); (M.W.); (P.S.); (D.H.); (I.M.)
| | - Peter Radermacher
- Institute of Anesthesiological Pathophysiology and Process Engineering, University Medical School, 89070 Ulm, Germany;
| | - Ingo Marzi
- Department of Trauma, Hand and Reconstructive Surgery, University Hospital Frankfurt, 60596 Frankfurt, Germany; (T.L.); (M.W.); (P.S.); (D.H.); (I.M.)
| |
Collapse
|
41
|
The dynamic change of serum S100B levels from day 1 to day 3 is more associated with sepsis-associated encephalopathy. Sci Rep 2020; 10:7718. [PMID: 32382007 PMCID: PMC7206038 DOI: 10.1038/s41598-020-64200-3] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2019] [Accepted: 03/24/2020] [Indexed: 01/08/2023] Open
Abstract
We investigated the role of dynamic changes of serum levels S100B protein in brain injury and poor outcome of sepsis. This is a prospective cohort study designed to include 104 adult patients with sepsis who are admitted to ICU from Jan 2015 to Aug 2016. Sepsis was defined as sepsis 3.0. Patients with a GCS score of <15, or at least one positive CAM-ICU score were thought to have brain dysfunction. 59 patients were diagnosed with SAE and the rest 45 patients were diagnosed with non-SAE. Serum S100B was measured on day 1 and 3 after ICU admission. Primary outcomes included brain dysfunction and 28-day/180-day mortality. The SAE group showed a significantly higher APACHE II score, SOFA scores, length of ICU stay, 28-day and 180-day mortality, serum S100B levels on day 1 and day 3. S100B levels on day 1 of 0.226 μg/L were diagnostic for SAE with 80.0% specificity and 66.1% sensitivity, and the area under (AUC) the curve was 0.728, S100B levels on day 3 of 0.144 μg/L were diagnostic for SAE with 84.44% specificity and 69.49% sensitivity, and the AUC was 0.819. In addition, the AUC for S100B on day 3 for predicting 180-day mortality was larger than for S100B on day 1 (0.731 vs. 0.611). Multiple logistic regression analysis showed that S100B3 (p = 0.001) but not S100B1 (p = 0.927) were independently correlated with SAE. Kaplan-Meier survival analysis showed that patients with S100B levels higher than 0.144 μg/L had a lower probability of survival at day 180. There were more patients with encephalopathy and a higher 28-day or 180-day mortality in the ΔS100B + group than in the ΔS100B- group. Multiple logistic regression analysis showed that SAE and IL-6 on day 3 were independently correlated with S100B dynamic increase. These findings suggest that elevated serum S100B levels on day 3 and the dynamic changes of serum S100B levels from day three to one were more associated with brain dysfunction and mortality than that on day 1 in patients with sepsis.
Collapse
|
42
|
Integrative Analysis of Circulating Metabolite Profiles and Magnetic Resonance Imaging Metrics in Patients with Traumatic Brain Injury. Int J Mol Sci 2020; 21:ijms21041395. [PMID: 32092929 PMCID: PMC7073036 DOI: 10.3390/ijms21041395] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2019] [Revised: 02/13/2020] [Accepted: 02/15/2020] [Indexed: 12/19/2022] Open
Abstract
Recent evidence suggests that patients with traumatic brain injuries (TBIs) have a distinct circulating metabolic profile. However, it is unclear if this metabolomic profile corresponds to changes in brain morphology as observed by magnetic resonance imaging (MRI). The aim of this study was to explore how circulating serum metabolites, following TBI, relate to structural MRI (sMRI) findings. Serum samples were collected upon admission to the emergency department from patients suffering from acute TBI and metabolites were measured using mass spectrometry-based metabolomics. Most of these patients sustained a mild TBI. In the same patients, sMRIs were taken and volumetric data were extracted (138 metrics). From a pool of 203 eligible screened patients, 96 met the inclusion criteria for this study. Metabolites were summarized as eight clusters and sMRI data were reduced to 15 independent components (ICs). Partial correlation analysis showed that four metabolite clusters had significant associations with specific ICs, reflecting both the grey and white matter brain injury. Multiple machine learning approaches were then applied in order to investigate if circulating metabolites could distinguish between positive and negative sMRI findings. A logistic regression model was developed, comprised of two metabolic predictors (erythronic acid and myo-inositol), which, together with neurofilament light polypeptide (NF-L), discriminated positive and negative sMRI findings with an area under the curve of the receiver-operating characteristic of 0.85 (specificity = 0.89, sensitivity = 0.65). The results of this study show that metabolomic analysis of blood samples upon admission, either alone or in combination with protein biomarkers, can provide valuable information about the impact of TBI on brain structural changes.
Collapse
|
43
|
Vojdani A, Vojdani E, Herbert M, Kharrazian D. Correlation between Antibodies to Bacterial Lipopolysaccharides and Barrier Proteins in Sera Positive for ASCA and ANCA. Int J Mol Sci 2020; 21:ijms21041381. [PMID: 32085663 PMCID: PMC7073094 DOI: 10.3390/ijms21041381] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2019] [Revised: 02/10/2020] [Accepted: 02/16/2020] [Indexed: 12/17/2022] Open
Abstract
Individuals with intestinal barrier dysfunction are more prone to autoimmunity. Lipopolysaccharides (LPS) from gut bacteria have been shown to play a role in systemic inflammation, leading to the opening of the gut and blood-brain barrier (BBB). This study aims to measure antibodies against LPS and barrier proteins in samples positive for anti-Saccharomyces cerevisiae antibodies (ASCA) and anti-neutrophil cytoplasmic antibodies (ANCA) and compare them with these same antibodies in controls to determine whether a correlation between LPS and barrier proteins could be found. We obtained 94 ASCA- and 94 ANCA-positive blood samples, as well as 188 blood samples from healthy controls. Samples were assessed for antibodies to LPS, zonulin+occludin, S100B, and aquaporin-4 (AQP4). Results show significant elevation in antibodies in about 30% of ASCA- and ANCA-positive sera and demonstrate positive linear relationships between these antibodies. The findings suggest that individuals positive for ASCA and ANCA have increased odds of developing intestinal and BBB permeability compared to healthy subjects. The levels of LPS antibodies in both ASCA- and ANCA-positive and negative specimens showed from low and moderate to high correlation with antibodies to barrier proteins. This study shows that LPS, by damaging the gut and BBBs, contribute to the extra-intestinal manifestation of IBD. We conclude that IBD patients should be screened for LPS antibodies in an effort to detect or prevent possible barrier damage at the earliest stage possible to abrogate disease symptoms in IBS and associated disorders.
Collapse
Affiliation(s)
- Aristo Vojdani
- Immunosciences Lab, Inc. 822 S. Robertson Blvd, Ste 312, Los Angeles, CA 90035, USA
- Department of Preventive Medicine, Loma Linda University, Loma Linda, CA 92350, USA;
- Correspondence: ; Tel.: +1-310-657-1077
| | - Elroy Vojdani
- Regenera Medical, 11860 Wilshire Blvd., Ste. 301, Los Angeles, CA 90025, USA;
| | - Martha Herbert
- Martha Herbert, Pediatric Neurology, Massachusetts General Hospital, Rm CNY149-2nd Floor, Boston, MA 02114, USA;
| | - Datis Kharrazian
- Department of Preventive Medicine, Loma Linda University, Loma Linda, CA 92350, USA;
- Department of Neurology, Harvard Medical, Boston, MA 02115, USA
- Department of Neurology, Massachusetts General Hospital, Boston, MA 02129, USA
| |
Collapse
|
44
|
Forssten MP, Thelin EP, Nelson DW, Bellander BM. The Role of Glycerol-Containing Drugs in Cerebral Microdialysis: A Retrospective Study on the Effects of Intravenously Administered Glycerol. Neurocrit Care 2020; 30:590-600. [PMID: 30430381 PMCID: PMC6513829 DOI: 10.1007/s12028-018-0643-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
BACKGROUND Cerebral microdialysis (CMD) is a valuable tool for monitoring compounds in the cerebral extracellular fluid (ECF). Glycerol is one such compound which is regarded as a marker of cell membrane decomposition. Notably, in some acutely brain-injured patients, CMD-glycerol levels rise without any other apparent indication of cerebral deterioration. The aim of this study was to investigate whether this could be due to an association between CMD-glycerol levels and the administration of glycerol-containing drugs. METHODS Microdialysis data were retrospectively retrieved from the hospital's intensive care unit patient data management system (PDMS). All patients who were monitored with CMD for ≥ 96 h were included. Administered drug doses were retrieved from the PDMS and converted to exact doses of glycerol. Cross-correlation analyses were performed between the free, metabolized as well as total administered dose of glycerol and the detrended and differenced CMD-glycerol concentration. These analyses were repeated for two sets of subgroups based upon the individual catheter's graphical trend and its location in relation to the lesion. RESULTS There was no significant correlation between the differenced CMD-glycerol levels and drug-administered glycerol. Furthermore, there was no significant correlation between CMD-glycerol and catheter location or graphical trend. However, if the CMD-glycerol levels were detrended, significant but clinically non-relevant correlations were identified (maximum correlation coefficient of 0.1 (0.04-0.15, 95% CI) at a lag of 7 h using the total administered dose of glycerol). CONCLUSIONS Glycerol-containing drugs routinely administered intravenously in the clinical setting appear to have a minimal and clinically insignificant effect on levels of glycerol in the cerebral ECF.
Collapse
Affiliation(s)
- Maximilian Peter Forssten
- Department of Clinical Neuroscience, Section for Neurosurgery, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden.
| | - Eric Peter Thelin
- Department of Clinical Neuroscience, Section for Neurosurgery, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden.,Division of Neurosurgery, Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK
| | - David W Nelson
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
| | - Bo-Michael Bellander
- Department of Clinical Neuroscience, Section for Neurosurgery, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| |
Collapse
|
45
|
Alves JL, Rato J, Silva V. Why Does Brain Trauma Research Fail? World Neurosurg 2019; 130:115-121. [PMID: 31284053 DOI: 10.1016/j.wneu.2019.06.212] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2019] [Revised: 06/26/2019] [Accepted: 06/27/2019] [Indexed: 02/06/2023]
Abstract
Traumatic brain injury (TBI) represents a major health care problem and a significant social and economic issue worldwide. Considering the generalized failure in introducing effective drugs and clinical protocols, there is an urgent need for efficient treatment modalities, able to improve devastating posttraumatic morbidity and mortality. In this work, the status of brain trauma research is analyzed in all its aspects, including basic and translational science and clinical trials. Implicit and explicit challenges to different lines of research are discussed and clinical trial structures and outcomes are scrutinized, along with possible explanations for systematic therapeutic failures and their implications for future development of drug and clinical trials. Despite significant advances in basic and clinical research in recent years, no specific therapeutic protocols for TBI have been shown to be effective. New potential therapeutic targets have been identified, following a better understanding of pathophysiologic mechanisms underlying TBI, although with disappointing results. Several reasons can be pinpointed at different levels, from inaccurate animal models of disease to faulty preclinical and clinical trials, with poor design and subjective outcome measures. Distinct strategies can be delineated to overcome specific shortcomings of research studies. Identifying and contextualizing the failures that have dominated TBI research is mandatory. This review analyzes current approaches and discusses possible strategies for improving outcomes.
Collapse
Affiliation(s)
- José Luís Alves
- Department of Neurosurgery, Centro Hospitalar e Universitário de Coimbra, Coimbra, Portugal.
| | - Joana Rato
- Department of Neurosurgery, Centro Hospitalar e Universitário de Coimbra, Coimbra, Portugal
| | - Vitor Silva
- Department of Neurosurgery, Centro Hospitalar e Universitário de Coimbra, Coimbra, Portugal
| |
Collapse
|
46
|
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
|
47
|
Mahan MY, Thorpe M, Ahmadi A, Abdallah T, Casey H, Sturtevant D, Judge-Yoakam S, Hoover C, Rafter D, Miner J, Richardson C, Samadani U. Glial Fibrillary Acidic Protein (GFAP) Outperforms S100 Calcium-Binding Protein B (S100B) and Ubiquitin C-Terminal Hydrolase L1 (UCH-L1) as Predictor for Positive Computed Tomography of the Head in Trauma Subjects. World Neurosurg 2019; 128:e434-e444. [PMID: 31051301 DOI: 10.1016/j.wneu.2019.04.170] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2019] [Revised: 04/18/2019] [Accepted: 04/19/2019] [Indexed: 10/26/2022]
Abstract
OBJECTIVE Traumatic brain injuries (TBIs) are largely underdiagnosed and may have persistent refractory consequences. Current assessments for acute TBI are limited to physical examination and imaging. Biomarkers such as glial fibrillary acidic protein (GFAP), ubiquitin C-terminal hydrolase L1 (UCH-L1), and S100 calcium-binding protein B (S100B) have shown predictive value as indicators of TBI and potential screening tools. METHODS In total, 37 controls and 118 unique trauma subjects who received a clinically ordered head computed tomography (CT) in the emergency department of a level 1 trauma center were evaluated. Blood samples collected at 0-8 hours (initial) and 12-32 hours (delayed) postinjury were analyzed for GFAP, UCH-L1, and S100B concentrations. These were then compared in CT-negative and CT-positive subjects. RESULTS Median GFAP, UCH-L1, and S100B concentrations were greater in CT-positive subjects at both timepoints compared with CT-negative subjects. In addition, median UCH-L1 and S100B concentrations were lower at the delayed timepoint, whereas median GFAP concentrations were increased. As predictors of a positive CT of the head, GFAP outperformed UCH-L1 and S100B at both timepoints (initial: 0.89 sensitivity, 0.62 specificity; delayed: 0.94 sensitivity, 0.67 specificity). GFAP alone also outperformed all possible combinations of biomarkers. CONCLUSIONS GFAP, UCH-L1, and S100B demonstrated utility for rapid prediction of a CT-positive TBI within 0-8 hours of injury. GFAP exhibited the greatest predictive power at 12-32 hours. Furthermore, these results suggest that GFAP alone has greater utility for predicting a positive CT of the head than UCH-L1, S100B, or any combination of the 3.
Collapse
Affiliation(s)
- Margaret Y Mahan
- Department of Neurosurgery, Hennepin Healthcare, Minneapolis, Minnesota, USA; Department of Biomedical Informatics and Computational Biology, University of Minnesota, Minneapolis, Minnesota, USA
| | - Maxwell Thorpe
- Department of Neurosurgery, Hennepin Healthcare, Minneapolis, Minnesota, USA
| | - Aliya Ahmadi
- Department of Neurosurgery, Hennepin Healthcare, Minneapolis, Minnesota, USA
| | - Tessneem Abdallah
- Department of Neurosurgery, Hennepin Healthcare, Minneapolis, Minnesota, USA
| | - Hannah Casey
- Department of Neurosurgery, Hennepin Healthcare, Minneapolis, Minnesota, USA
| | - Dylan Sturtevant
- Department of Neurosurgery, Hennepin Healthcare, Minneapolis, Minnesota, USA
| | - Sénait Judge-Yoakam
- Department of Neurosurgery, Hennepin Healthcare, Minneapolis, Minnesota, USA
| | - Caleb Hoover
- Department of Neurosurgery, Hennepin Healthcare, Minneapolis, Minnesota, USA
| | - Daniel Rafter
- Department of Neurosurgery, Hennepin Healthcare, Minneapolis, Minnesota, USA
| | - James Miner
- Department of Emergency Medicine, Hennepin Healthcare, Minneapolis, Minnesota, USA
| | - Chad Richardson
- Department of General Surgery, Hennepin Healthcare, Minneapolis, Minnesota, USA
| | - Uzma Samadani
- Department of Neurosurgery, Hennepin Healthcare, Minneapolis, Minnesota, USA; Department of Biomedical Informatics and Computational Biology, University of Minnesota, Minneapolis, Minnesota, USA; Department of Neurosurgery, University of Minnesota, Minneapolis, Minnesota, USA.
| |
Collapse
|
48
|
Kornguth S, Rutledge N. Integration of Biomarkers Into a Signature Profile of Persistent Traumatic Brain Injury Involving Autoimmune Processes Following Water Hammer Injury From Repetitive Head Impacts. Biomark Insights 2018; 13:1177271918808216. [PMID: 30397383 PMCID: PMC6207974 DOI: 10.1177/1177271918808216] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2018] [Accepted: 09/28/2018] [Indexed: 01/07/2023] Open
Abstract
OBJECTIVES To assemble an algorithm that will describe a "Signature" predictive of an individual's vulnerability to persistent traumatic brain injury (TBI). SUBJECTS AND METHODS Studies of athletes and warriors who are subjected to repeated head impacts with rapid acceleration/deceleration forces are used to assist in the diagnosis and management of TBI-affected individuals. Data from multiple areas, including clinical, anatomical, magnetic resonance imaging, cognitive function, and biochemical analyses, are integrated to provide a Signature of persistent TBI. RESULTS Studies to date indicate that susceptibility to TBI results from an interaction between host genetic and structural vulnerability factors and force and torque of impact on the head and torso. The host factors include molecular markers affecting immune and inflammatory responses to stress/insult as well as anatomical features such as the degree of transcortical fiber projections and vascular malformations. The host response to forceful impact includes the release of intracellular neural proteins and nucleic acids into the cerebrospinal fluid and vascular compartment as well as mobilization of cytokines and macrophages into the central nervous system with subsequent activation of microglia and inflammatory responses including autoimmune processes. Maximum impact to the base of the sulci via a "water hammer effect" is consistent with the localization of microvascular and inflammatory responses in the affected brain region. CONCLUSIONS An assessment of an individuals' predisposition to persistent TBI with delayed cognitive deficits and behavioral changes requires an understanding of host vulnerability (genetic factors and brain structure) and external stressors (force and torque of impact as well as repetitive head injury and time interval between impacts). An algorithm that has utility in predicting vulnerability to TBI will include qualitative and quantitative measures of the host factors weighted against post impact markers of neural injury. Implementation of the resulting "Signature" of vulnerability at early stages of injury will help inform athletes and warriors, along with commanders and management, of the risk/benefit approaches that will markedly diminish health care costs to the nation and suffering to this population. This report attempts to define a strategy to create such an algorithm.
Collapse
Affiliation(s)
- Steven Kornguth
- Department of Kinesiology and Health Education, The University of Texas at Austin, Austin, TX, USA
- Department of Neurology, Dell Medical School, The University of Texas at Austin
| | - Neal Rutledge
- Department of Psychology, The University of Texas at Austin and Austin Radiological Association, Austin, TX, USA
| |
Collapse
|
49
|
The 3rd Joint Symposium of the International and National Neurotrauma Societies and AANS/CNS Section on Neurotrauma and Critical Care August 11–16, 2018 Toronto, Canada. J Neurotrauma 2018. [DOI: 10.1089/neu.2018.29013.abstracts] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
|
50
|
Medkova A, Srovnal J, Potomkova J, Volejnikova J, Mihal V. Multifarious diagnostic possibilities of the S100 protein family: predominantly in pediatrics and neonatology. World J Pediatr 2018; 14:315-321. [PMID: 29858979 DOI: 10.1007/s12519-018-0163-5] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/26/2017] [Accepted: 05/11/2018] [Indexed: 01/05/2023]
Abstract
BACKGROUND Numerous articles related to S100 proteins have been recently published. This review aims to introduce this large protein family and its importance in the diagnostics of many pathological conditions in children and adults. DATA SOURCES Based on original publications found in database systems, we summarize the current knowledge about the S100 protein group and highlight the most important proteins with focus on pediatric use. RESULTS The S100 family is composed of Ca2+ and Zn2+ binding proteins, which are present only in vertebrates. Some of these proteins can be used as diagnostic markers in cardiology (S100A1, S100A12), oncology (S100A2, S100A5, S100A6, S100A14, S100A16, S100P, S100B), neurology (S100B), rheumatology (S100A8/A9, S100A4, S100A6, and S100A12), nephrology and infections (S100A8, S100A9, S100A8/A9, S100A12). The most useful S100 proteins in pediatrics are S100A8, S100A9, heterodimers S100A8/A9, S100B and S100A12. CONCLUSIONS The S100 family members are promising biomarkers and provide numerous possibilities for implementation into clinical practice to optimize the differential diagnostic process.
Collapse
Affiliation(s)
- Anna Medkova
- Department of Pediatrics, Faculty of Medicine and Dentistry, Palacky University and University Hospital Olomouc, I. P. Pavlova 6, 779 00, Olomouc, Czech Republic.
| | - Josef Srovnal
- Department of Pediatrics, Faculty of Medicine and Dentistry, Palacky University and University Hospital Olomouc, I. P. Pavlova 6, 779 00, Olomouc, Czech Republic
- Faculty of Medicine and Dentistry, Institute of Molecular and Translational Medicine, Palacky University Olomouc, Hněvotínská, 1333/5, 779 00, Olomouc, Czech Republic
| | - Jarmila Potomkova
- Department of Pediatrics, Faculty of Medicine and Dentistry, Palacky University and University Hospital Olomouc, I. P. Pavlova 6, 779 00, Olomouc, Czech Republic
- Department of Science and Research, University Hospital Olomouc, I. P. Pavlova 6, 779 00, Olomouc, Czech Republic
| | - Jana Volejnikova
- Department of Pediatrics, Faculty of Medicine and Dentistry, Palacky University and University Hospital Olomouc, I. P. Pavlova 6, 779 00, Olomouc, Czech Republic
- Faculty of Medicine and Dentistry, Institute of Molecular and Translational Medicine, Palacky University Olomouc, Hněvotínská, 1333/5, 779 00, Olomouc, Czech Republic
| | - Vladimir Mihal
- Department of Pediatrics, Faculty of Medicine and Dentistry, Palacky University and University Hospital Olomouc, I. P. Pavlova 6, 779 00, Olomouc, Czech Republic
- Faculty of Medicine and Dentistry, Institute of Molecular and Translational Medicine, Palacky University Olomouc, Hněvotínská, 1333/5, 779 00, Olomouc, Czech Republic
| |
Collapse
|