1
|
Zhong X, Tai W, Liu ML, Ma S, Shen T, Zou Y, Zhang CL. The Citron homology domain of MAP4Ks improves outcomes of traumatic brain injury. Neural Regen Res 2025; 20:3233-3244. [PMID: 39314140 DOI: 10.4103/nrr.nrr-d-24-00113] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2024] [Accepted: 08/30/2024] [Indexed: 09/25/2024] Open
Abstract
JOURNAL/nrgr/04.03/01300535-202511000-00027/figure1/v/2024-12-20T164640Z/r/image-tiff The mitogen-activated protein kinase kinase kinase kinases (MAP4Ks) signaling pathway plays a pivotal role in axonal regrowth and neuronal degeneration following insults. Whether targeting this pathway is beneficial to brain injury remains unclear. In this study, we showed that adeno-associated virus-delivery of the Citron homology domain of MAP4Ks effectively reduces traumatic brain injury-induced reactive gliosis, tauopathy, lesion size, and behavioral deficits. Pharmacological inhibition of MAP4Ks replicated the ameliorative effects observed with expression of the Citron homology domain. Mechanistically, the Citron homology domain acted as a dominant-negative mutant, impeding MAP4K-mediated phosphorylation of the dishevelled proteins and thereby controlling the Wnt/β-catenin pathway. These findings implicate a therapeutic potential of targeting MAP4Ks to alleviate the detrimental effects of traumatic brain injury.
Collapse
Affiliation(s)
- Xiaoling Zhong
- Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Hamon Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Wenjiao Tai
- Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Hamon Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Meng-Lu Liu
- Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Shuaipeng Ma
- Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Hamon Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Tianjin Shen
- Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Hamon Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Yuhua Zou
- Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Hamon Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Chun-Li Zhang
- Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Hamon Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Peter O'Donnell Jr. Brain Institute, University of Texas Southwestern Medical Center, Dallas, TX, USA
| |
Collapse
|
2
|
Mavroudis I, Petridis F, Ciobica A, Kamal FZ, Padurariu M, Kazis D. Advancements in diagnosing Post-concussion Syndrome: insights into epidemiology, pathophysiology, neuropathology, neuroimaging, and salivary biomarkers. Acta Neurol Belg 2025:10.1007/s13760-024-02695-7. [PMID: 39776059 DOI: 10.1007/s13760-024-02695-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2024] [Accepted: 11/23/2024] [Indexed: 01/11/2025]
Abstract
Post-Concussion Syndrome (PCS) represents a complex constellation of symptoms that persist following a concussion or mild traumatic brain injury (mTBI), with significant implications for patient care and outcomes. Despite its prevalence, diagnosing PCS presents considerable challenges due to the subjective nature of symptoms, the absence of specific diagnostic tests, and the overlap with other neurological and psychiatric conditions. This review explores the multifaceted diagnostic challenges associated with PCS, including the heterogeneity of symptom presentation, the limitations of current neuroimaging techniques, and the overlap of PCS symptoms with other disorders. We also discuss the potential of emerging biomarkers and advanced imaging modalities to enhance diagnostic accuracy and provide a more objective basis for PCS identification. Additionally, the review highlights the importance of a multidisciplinary approach in the diagnosis and management of PCS, integrating clinical evaluation with innovative diagnostic tools to improve patient outcomes. Through a comprehensive analysis of current practices and future directions, this review aims to shed light on the complexities of PCS diagnosis and pave the way for improved strategies in the identification and treatment of this condition.
Collapse
Affiliation(s)
- Ioannis Mavroudis
- Department of Neuroscience, Leeds Teaching Hospitals, NHS Trust, Leeds, UK
- Leeds University, Leeds, UK
| | - Foivos Petridis
- Third Department of Neurology, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Alin Ciobica
- Department of Biology, Faculty of Biology, Alexandru Ioan Cuza University of Iasi, Bd. Carol I nr. 20A, Iasi, 700505, Romania
- Centre of Biomedical Research, Romanian Academy, Bd. Carol I, no. 8, Iasi, 700506, Romania
- Academy of Romanian Scientists, Str. Splaiul Independentei no. 54, Sector 5, Bucharest, 050094, Romania
- Preclinical Department, Apollonia University, Păcurari Street 11, Iasi, 700511, Romania
| | - Fatima Zahra Kamal
- Laboratory of Physical Chemistry of Processes, Faculty of Sciences and Techniques, Hassan First University, B.P. 539, Settat, 26000, Morocco.
- Higher Institute of Nursing Professions and Health Technical (ISPITS), Marrakech, Morocco.
| | - Manuela Padurariu
- Socola Institute of Psychiatry, Șoseaua Bucium 36, Iași, 700282, Romania
| | - Dimitrios Kazis
- Third Department of Neurology, Aristotle University of Thessaloniki, Thessaloniki, Greece
| |
Collapse
|
3
|
Zhang H, Wang J, Qu Y, Yang Y, Guo ZN. Brain injury biomarkers and applications in neurological diseases. Chin Med J (Engl) 2025; 138:5-14. [PMID: 38915214 PMCID: PMC11717530 DOI: 10.1097/cm9.0000000000003061] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2023] [Indexed: 06/26/2024] Open
Abstract
ABSTRACT Neurological diseases are a major health concern, and brain injury is a typical pathological process in various neurological disorders. Different biomarkers in the blood or the cerebrospinal fluid are associated with specific physiological and pathological processes. They are vital in identifying, diagnosing, and treating brain injuries. In this review, we described biomarkers for neuronal cell body injury (neuron-specific enolase, ubiquitin C-terminal hydrolase-L1, αII-spectrin), axonal injury (neurofilament proteins, tau), astrocyte injury (S100β, glial fibrillary acidic protein), demyelination (myelin basic protein), autoantibodies, and other emerging biomarkers (extracellular vesicles, microRNAs). We aimed to summarize the applications of these biomarkers and their related interests and limits in the diagnosis and prognosis for neurological diseases, including traumatic brain injury, status epilepticus, stroke, Alzheimer's disease, and infection. In addition, a reasonable outlook for brain injury biomarkers as ideal detection tools for neurological diseases is presented.
Collapse
Affiliation(s)
- Han Zhang
- Stroke Center, Department of Neurology, the First Hospital of Jilin University, Changchun, Jilin 130021, China
| | - Jing Wang
- Stroke Center, Department of Neurology, the First Hospital of Jilin University, Changchun, Jilin 130021, China
| | - Yang Qu
- Stroke Center, Department of Neurology, the First Hospital of Jilin University, Changchun, Jilin 130021, China
| | - Yi Yang
- Stroke Center, Department of Neurology, the First Hospital of Jilin University, Changchun, Jilin 130021, China
| | - Zhen-Ni Guo
- Stroke Center, Department of Neurology, the First Hospital of Jilin University, Changchun, Jilin 130021, China
- Neuroscience Research Center, Department of Neurology, the First Hospital of Jilin University, Changchun, Jilin 130021, China
| |
Collapse
|
4
|
Olesen MA, Villavicencio-Tejo F, Cuevas-Espinoza V, Quintanilla RA. Unknown roles of tau pathology in neurological disorders. Challenges and new perspectives. Ageing Res Rev 2025; 103:102594. [PMID: 39577774 DOI: 10.1016/j.arr.2024.102594] [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: 10/14/2024] [Revised: 11/16/2024] [Accepted: 11/18/2024] [Indexed: 11/24/2024]
Abstract
Aging presents progressive changes that increase the susceptibility of the central nervous system (CNS) to suffer neurological disorders (NDs). Several studies have reported that an aged brain suffering from NDs shows the presence of pathological forms of tau protein, a microtubule accessory protein (MAP) critical for neuronal function. In this context, accumulative evidence has shown a pivotal contribution of pathological forms of tau to Alzheimer's disease (AD) and tauopathies. However, current investigations have implicated tau toxicity in other NDs that affect the central nervous system (CNS), including Parkinson's disease (PD), Huntington's disease (HD), Traumatic brain injury (TBI), Multiple sclerosis (MS), and Amyotrophic lateral sclerosis (ALS). These diseases are long-term acquired, affecting essential functions such as motor movement, cognition, hearing, and vision. Previous evidence indicated that toxic forms of tau do not have a critical contribution to the genesis or progression of these diseases. However, recent studies have shown that these tau forms contribute to neuronal dysfunction, inflammation, oxidative damage, and mitochondrial impairment events that contribute to the pathogenesis of these NDs. Recent studies have suggested that these neuropathologies could be associated with a prion-like behavior of tau, which induces a pathological dissemination of these toxic protein forms to different brain areas. Moreover, it has been suggested that this toxic propagation of tau from neurons into neighboring cells impairs the function of glial cells, oligodendrocytes, and endothelial cells by affecting metabolic function and mitochondrial health and inducing oxidative damage by tau pathology. Therefore, in this review, we will discuss current evidence demonstrating the critical role of toxic tau forms on NDs not related to AD and how its propagation and induced-bioenergetics failure may contribute to the pathogenic mechanism present in these NDs.
Collapse
Affiliation(s)
- Margrethe A Olesen
- Laboratory of Neurodegenerative Diseases, Instituto de Ciencias Biomédicas, Facultad de Ciencias de la Salud, Universidad Autónoma de Chile, Chile
| | - Francisca Villavicencio-Tejo
- Laboratory of Neurodegenerative Diseases, Instituto de Ciencias Biomédicas, Facultad de Ciencias de la Salud, Universidad Autónoma de Chile, Chile
| | - Víctor Cuevas-Espinoza
- Laboratory of Neurodegenerative Diseases, Instituto de Ciencias Biomédicas, Facultad de Ciencias de la Salud, Universidad Autónoma de Chile, Chile
| | - Rodrigo A Quintanilla
- Laboratory of Neurodegenerative Diseases, Instituto de Ciencias Biomédicas, Facultad de Ciencias de la Salud, Universidad Autónoma de Chile, Chile.
| |
Collapse
|
5
|
Spitz G, Hicks AJ, McDonald SJ, Dore V, Krishnadas N, O’Brien TJ, O’Brien WT, Vivash L, Law M, Ponsford JL, Rowe C, Shultz SR. Plasma biomarkers in chronic single moderate-severe traumatic brain injury. Brain 2024; 147:3690-3701. [PMID: 39315931 PMCID: PMC11531850 DOI: 10.1093/brain/awae255] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2023] [Revised: 06/06/2024] [Accepted: 07/04/2024] [Indexed: 09/25/2024] Open
Abstract
Blood biomarkers are an emerging diagnostic and prognostic tool that reflect a range of neuropathological processes following traumatic brain injury (TBI). Their effectiveness in identifying long-term neuropathological processes after TBI is unclear. Studying biomarkers in the chronic phase is vital because elevated levels in TBI might result from distinct neuropathological mechanisms during acute and chronic phases. Here, we examine plasma biomarkers in the chronic period following TBI and their association with amyloid and tau PET, white matter microarchitecture, brain age and cognition. We recruited participants ≥40 years of age who had suffered a single moderate-severe TBI ≥10 years previously between January 2018 and March 2021. We measured plasma biomarkers using single molecule array technology [ubiquitin C-terminal hydrolase L1 (UCH-L1), neurofilament light (NfL), tau, glial fibrillary acidic protein (GFAP) and phosphorylated tau (P-tau181)]; PET tracers to measure amyloid-β (18F-NAV4694) and tau neurofibrillary tangles (18F-MK6240); MRI to assess white matter microstructure and brain age; and the Rey Auditory Verbal Learning Test to measure verbal-episodic memory. A total of 90 post-TBI participants (73% male; mean = 58.2 years) were recruited on average 22 years (range = 10-33 years) post-injury, and 32 non-TBI control participants (66% male; mean = 57.9 years) were recruited. Plasma UCH-L1 levels were 67% higher {exp(b) = 1.67, P = 0.018, adjusted P = 0.044, 95% confidence interval (CI) [10% to 155%], area under the curve = 0.616} and P-tau181 were 27% higher {exp(b) = 1.24, P = 0.011, adjusted P = 0.044, 95% CI [5% to 46%], area under the curve = 0.632} in TBI participants compared with controls. Amyloid and tau PET were not elevated in TBI participants. Higher concentrations of plasma P-tau181, UCH-L1, GFAP and NfL were significantly associated with worse white matter microstructure but not brain age in TBI participants. For TBI participants, poorer verbal-episodic memory was associated with higher concentration of P-tau181 {short delay: b = -2.17, SE = 1.06, P = 0.043, 95% CI [-4.28, -0.07]; long delay: bP-tau = -2.56, SE = 1.08, P = 0.020, 95% CI [-4.71, -0.41]}, tau {immediate memory: bTau = -6.22, SE = 2.47, P = 0.014, 95% CI [-11.14, -1.30]} and UCH-L1 {immediate memory: bUCH-L1 = -2.14, SE = 1.07, P = 0.048, 95% CI [-4.26, -0.01]}, but was not associated with functional outcome. Elevated plasma markers related to neuronal damage and accumulation of phosphorylated tau suggest the presence of ongoing neuropathology in the chronic phase following a single moderate-severe TBI. Plasma biomarkers were associated with measures of microstructural brain disruption on MRI and disordered cognition, further highlighting their utility as potential objective tools to monitor evolving neuropathology post-TBI.
Collapse
Affiliation(s)
- Gershon Spitz
- Monash-Epworth Rehabilitation Research Centre, School of Psychological Sciences, Faculty of Medicine, Nursing and Health Sciences, Monash University, Clayton, VIC 3800, Australia
- Department of Neuroscience, School of Translational Medicine, Faculty of Medicine, Nursing and Health Sciences, Monash University, Clayton, VIC 3004, Australia
| | - Amelia J Hicks
- Monash-Epworth Rehabilitation Research Centre, School of Psychological Sciences, Faculty of Medicine, Nursing and Health Sciences, Monash University, Clayton, VIC 3800, Australia
| | - Stuart J McDonald
- Department of Neuroscience, School of Translational Medicine, Faculty of Medicine, Nursing and Health Sciences, Monash University, Clayton, VIC 3004, Australia
- Department of Neurology, The Alfred, Melbourne, VIC 3004, Australia
| | - Vincent Dore
- Florey Department of Neuroscience and Mental Health, University of Melbourne, Parkville, VIC 3010, Australia
- Department of Molecular Imaging and Therapy, Austin Health, Heidelberg, VIC 3084, Australia
| | - Natasha Krishnadas
- Florey Department of Neuroscience and Mental Health, University of Melbourne, Parkville, VIC 3010, Australia
- Department of Molecular Imaging and Therapy, Austin Health, Heidelberg, VIC 3084, Australia
| | - Terence J O’Brien
- Department of Neuroscience, School of Translational Medicine, Faculty of Medicine, Nursing and Health Sciences, Monash University, Clayton, VIC 3004, Australia
- Department of Neurology, The Alfred, Melbourne, VIC 3004, Australia
- Department of Medicine, Royal Melbourne Hospital, The University of Melbourne, Parkville, VIC 3010, Australia
| | - William T O’Brien
- Department of Neuroscience, School of Translational Medicine, Faculty of Medicine, Nursing and Health Sciences, Monash University, Clayton, VIC 3004, Australia
| | - Lucy Vivash
- Department of Neuroscience, School of Translational Medicine, Faculty of Medicine, Nursing and Health Sciences, Monash University, Clayton, VIC 3004, Australia
- Department of Neurology, The Alfred, Melbourne, VIC 3004, Australia
- Department of Medicine, Royal Melbourne Hospital, The University of Melbourne, Parkville, VIC 3010, Australia
| | - Meng Law
- Department of Neuroscience, School of Translational Medicine, Faculty of Medicine, Nursing and Health Sciences, Monash University, Clayton, VIC 3004, Australia
- Department of Radiology, Alfred Health, Melbourne, VIC 3004, Australia
| | - Jennie L Ponsford
- Monash-Epworth Rehabilitation Research Centre, School of Psychological Sciences, Faculty of Medicine, Nursing and Health Sciences, Monash University, Clayton, VIC 3800, Australia
| | - Christopher Rowe
- Florey Department of Neuroscience and Mental Health, University of Melbourne, Parkville, VIC 3010, Australia
- Department of Molecular Imaging and Therapy, Austin Health, Heidelberg, VIC 3084, Australia
| | - Sandy R Shultz
- Department of Neuroscience, School of Translational Medicine, Faculty of Medicine, Nursing and Health Sciences, Monash University, Clayton, VIC 3004, Australia
- Department of Neurology, The Alfred, Melbourne, VIC 3004, Australia
- The Centre for Trauma and Mental Health Research, Health Sciences and Human Services, Vancouver Island University, Nanaimo, BC V9R 5S5, Canada
| |
Collapse
|
6
|
Friberg S, Lindblad C, Zeiler FA, Zetterberg H, Granberg T, Svenningsson P, Piehl F, Thelin EP. Fluid biomarkers of chronic traumatic brain injury. Nat Rev Neurol 2024; 20:671-684. [PMID: 39363129 DOI: 10.1038/s41582-024-01024-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/10/2024] [Indexed: 10/05/2024]
Abstract
Traumatic brain injury (TBI) is a leading cause of long-term disability across the world. Evidence for the usefulness of imaging and fluid biomarkers to predict outcomes and screen for the need to monitor complications in the acute stage is steadily increasing. Still, many people experience symptoms such as fatigue and cognitive and motor dysfunction in the chronic phase of TBI, where objective assessments for brain injury are lacking. Consensus criteria for traumatic encephalopathy syndrome, a clinical syndrome possibly associated with the neurodegenerative disease chronic traumatic encephalopathy, which is commonly associated with sports concussion, have been defined only recently. However, these criteria do not fit all individuals living with chronic consequences of TBI. The pathophysiology of chronic TBI shares many similarities with other neurodegenerative and neuroinflammatory conditions, such as Alzheimer disease. As with Alzheimer disease, advancements in fluid biomarkers represent one of the most promising paths for unravelling the chain of pathophysiological events to enable discrimination between these conditions and, with time, provide prediction modelling and therapeutic end points. This Review summarizes fluid biomarker findings in the chronic phase of TBI (≥6 months after injury) that demonstrate the involvement of inflammation, glial biology and neurodegeneration in the long-term complications of TBI. We explore how the biomarkers associate with outcome and imaging findings and aim to establish mechanistic differences in biomarker patterns between types of chronic TBI and other neurodegenerative conditions. Finally, current limitations and areas of priority for future fluid biomarker research are highlighted.
Collapse
Affiliation(s)
- Susanna Friberg
- Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
- Department of Neurology, Karolinska University Hospital, Stockholm, Sweden
| | - Caroline Lindblad
- Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
- Department of Medical Sciences, Uppsala University, Uppsala, Sweden
- Department of Neurosurgery, Uppsala University Hospital, Uppsala, Sweden
| | - Frederick A Zeiler
- Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
- Section of Neurosurgery, Department of Surgery, University of Manitoba, Rady Faculty of Health Sciences, Winnipeg, Manitoba, Canada
- Department of Biomedical Engineering, Price Faculty of Engineering, University of Manitoba, Winnipeg, Manitoba, Canada
- Pan Am Clinic Foundation, Winnipeg, Manitoba, Canada
- Division of Anaesthesia, Department of Medicine, Addenbrooke's Hospital, University of Cambridge, Cambridge, UK
| | - Henrik Zetterberg
- UK Dementia Research Institute, University College London, London, UK
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
- Department of Neurodegenerative Disease, University College London, Queen Square Institute of Neurology, London, UK
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
- Hong Kong Center for Neurodegenerative Diseases, Clear Water Bay, Hong Kong, China
- Wisconsin Alzheimer's Disease Research Center, University of Wisconsin School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, USA
| | - Tobias Granberg
- Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
- Department of Neuroradiology, Karolinska University Hospital, Stockholm, Sweden
| | - Per Svenningsson
- Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
- Department of Neurology, Karolinska University Hospital, Stockholm, Sweden
- Department of Basic and Clinical Neuroscience, King's College London, London, UK
| | - Fredrik Piehl
- Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
- Department of Neurology, Karolinska University Hospital, Stockholm, Sweden
| | - Eric P Thelin
- Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden.
- Department of Neurology, Karolinska University Hospital, Stockholm, Sweden.
| |
Collapse
|
7
|
Pollaci G, Potenza A, Gorla G, Carrozzini T, Marinoni G, De Toma C, Canavero I, Rifino N, Boncoraglio GB, Difrancesco JC, Damavandi PT, Stanziano M, Erbetta A, Caroppo P, Di Fede G, Catania M, Zulueta A, Parati EA, Bersano A, Gatti L, Storti B. CSF and Plasma Biomarkers in Patients With Iatrogenic Cerebral Amyloid Angiopathy. Neurology 2024; 103:e209828. [PMID: 39284112 PMCID: PMC11399065 DOI: 10.1212/wnl.0000000000209828] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/22/2024] Open
Abstract
OBJECTIVES Recently, a subset of patients affected by cerebral amyloid angiopathy (CAA) distinguished by atypical juvenile onset and a hypothesized iatrogenic origin (iatrogenic CAA, iCAA) has emerged. β-Amyloid (Aβ) accumulation evidenced by amyloid PET positivity or CSF Aβ decrease was included in the iCAA diagnostic criteria. Conversely, diagnostic criteria for sporadic CAA (sCAA) do not involve biomarker analysis. The aim of this study was to assess CSF and plasma levels of Aβ and tau in iCAA and sCAA cohorts. METHODS Patients affected by probable or possible CAA according to established criteria (Boston 2.0) were prospectively recruited at Fondazione IRCCS Carlo Besta and San Gerardo dei Tintori from May 2021 to January 2024. Patients with probable and possible iCAA or sCAA with available plasma and/or CSF samples were included. Clinical and neurologic data were collected, and levels of Aβ40, Aβ42, total tau, and phospho-tau (p-tau) were assessed in CSF and plasma by SiMoA and Lumipulse. RESULTS 21 patients with iCAA (72% male, mean age at symptom onset 50 years [36-74]) and 32 patients with sCAA (44% male, mean age at symptom onset 68 years [52-80]) were identified. Cognitive impairment and cardiovascular risk factors in the sCAA cohort were more common compared with the iCAA cohort. Patients with sCAA and iCAA showed similar CSF levels for Aβ40 (p = 0.5 [sCAA, 95% CI 2,604-4,228; iCAA, 95% CI 1,958-3,736]), Aβ42 (p = 0.7 [sCAA, 95% CI 88-157; iCAA, 95% CI 83-155]), and total tau (p = 0.08 [sCAA, 95% CI 80-134; iCAA, 95% CI 37-99]). Plasma levels of Aβ40 (p = 0.08, 95% CI 181-222), Aβ42 (p = 0.3, 95% CI 6-8), and total tau (p = 0.4, 95% CI 3-6) were not statistically different in patients with sCAA compared with iCAA ones (Aβ40, 95% CI 153-193; Aβ42, 95% CI 6-7 and total tau, 95% CI 2-4). DISCUSSION Despite presenting with a younger age at onset, fewer cardiovascular risk factors, and lower cognitive impairment, patients with iCAA demonstrated Aβ and tau levels comparable with elderly patients with sCAA, supporting a common molecular paradigm between the 2 CAA forms.
Collapse
Affiliation(s)
- Giuliana Pollaci
- From the Cerebrovascular Unit (G.P., A.P., G.G., T.C., G.M., C.T., I.C., N.R., G.B.B., A.B., L.G., B.S.), Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan; Department of Pharmacological and Biomolecular Sciences (G.P., A.P.), University of Milan; Department of Neurology (J.C.D., P.T.D.), Fondazione IRCCS San Gerardo dei Tintori, Monza; Neuroradiology Unit (M.S., A.E.), Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan; ALS Centre (M.S.), "Rita Levi Montalcini" Department of Neuroscience, University of Turin; Neuropathology Unit (P.C., G.D.F., M.C.), Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan; and Istituti Clinici Scientifici Maugeri IRCCS (A.Z., E.A.P.), Neurorehabilitation Unit of Milan Institute, Italy
| | - Antonella Potenza
- From the Cerebrovascular Unit (G.P., A.P., G.G., T.C., G.M., C.T., I.C., N.R., G.B.B., A.B., L.G., B.S.), Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan; Department of Pharmacological and Biomolecular Sciences (G.P., A.P.), University of Milan; Department of Neurology (J.C.D., P.T.D.), Fondazione IRCCS San Gerardo dei Tintori, Monza; Neuroradiology Unit (M.S., A.E.), Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan; ALS Centre (M.S.), "Rita Levi Montalcini" Department of Neuroscience, University of Turin; Neuropathology Unit (P.C., G.D.F., M.C.), Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan; and Istituti Clinici Scientifici Maugeri IRCCS (A.Z., E.A.P.), Neurorehabilitation Unit of Milan Institute, Italy
| | - Gemma Gorla
- From the Cerebrovascular Unit (G.P., A.P., G.G., T.C., G.M., C.T., I.C., N.R., G.B.B., A.B., L.G., B.S.), Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan; Department of Pharmacological and Biomolecular Sciences (G.P., A.P.), University of Milan; Department of Neurology (J.C.D., P.T.D.), Fondazione IRCCS San Gerardo dei Tintori, Monza; Neuroradiology Unit (M.S., A.E.), Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan; ALS Centre (M.S.), "Rita Levi Montalcini" Department of Neuroscience, University of Turin; Neuropathology Unit (P.C., G.D.F., M.C.), Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan; and Istituti Clinici Scientifici Maugeri IRCCS (A.Z., E.A.P.), Neurorehabilitation Unit of Milan Institute, Italy
| | - Tatiana Carrozzini
- From the Cerebrovascular Unit (G.P., A.P., G.G., T.C., G.M., C.T., I.C., N.R., G.B.B., A.B., L.G., B.S.), Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan; Department of Pharmacological and Biomolecular Sciences (G.P., A.P.), University of Milan; Department of Neurology (J.C.D., P.T.D.), Fondazione IRCCS San Gerardo dei Tintori, Monza; Neuroradiology Unit (M.S., A.E.), Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan; ALS Centre (M.S.), "Rita Levi Montalcini" Department of Neuroscience, University of Turin; Neuropathology Unit (P.C., G.D.F., M.C.), Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan; and Istituti Clinici Scientifici Maugeri IRCCS (A.Z., E.A.P.), Neurorehabilitation Unit of Milan Institute, Italy
| | - Giulia Marinoni
- From the Cerebrovascular Unit (G.P., A.P., G.G., T.C., G.M., C.T., I.C., N.R., G.B.B., A.B., L.G., B.S.), Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan; Department of Pharmacological and Biomolecular Sciences (G.P., A.P.), University of Milan; Department of Neurology (J.C.D., P.T.D.), Fondazione IRCCS San Gerardo dei Tintori, Monza; Neuroradiology Unit (M.S., A.E.), Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan; ALS Centre (M.S.), "Rita Levi Montalcini" Department of Neuroscience, University of Turin; Neuropathology Unit (P.C., G.D.F., M.C.), Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan; and Istituti Clinici Scientifici Maugeri IRCCS (A.Z., E.A.P.), Neurorehabilitation Unit of Milan Institute, Italy
| | - Carolina De Toma
- From the Cerebrovascular Unit (G.P., A.P., G.G., T.C., G.M., C.T., I.C., N.R., G.B.B., A.B., L.G., B.S.), Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan; Department of Pharmacological and Biomolecular Sciences (G.P., A.P.), University of Milan; Department of Neurology (J.C.D., P.T.D.), Fondazione IRCCS San Gerardo dei Tintori, Monza; Neuroradiology Unit (M.S., A.E.), Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan; ALS Centre (M.S.), "Rita Levi Montalcini" Department of Neuroscience, University of Turin; Neuropathology Unit (P.C., G.D.F., M.C.), Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan; and Istituti Clinici Scientifici Maugeri IRCCS (A.Z., E.A.P.), Neurorehabilitation Unit of Milan Institute, Italy
| | - Isabella Canavero
- From the Cerebrovascular Unit (G.P., A.P., G.G., T.C., G.M., C.T., I.C., N.R., G.B.B., A.B., L.G., B.S.), Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan; Department of Pharmacological and Biomolecular Sciences (G.P., A.P.), University of Milan; Department of Neurology (J.C.D., P.T.D.), Fondazione IRCCS San Gerardo dei Tintori, Monza; Neuroradiology Unit (M.S., A.E.), Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan; ALS Centre (M.S.), "Rita Levi Montalcini" Department of Neuroscience, University of Turin; Neuropathology Unit (P.C., G.D.F., M.C.), Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan; and Istituti Clinici Scientifici Maugeri IRCCS (A.Z., E.A.P.), Neurorehabilitation Unit of Milan Institute, Italy
| | - Nicola Rifino
- From the Cerebrovascular Unit (G.P., A.P., G.G., T.C., G.M., C.T., I.C., N.R., G.B.B., A.B., L.G., B.S.), Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan; Department of Pharmacological and Biomolecular Sciences (G.P., A.P.), University of Milan; Department of Neurology (J.C.D., P.T.D.), Fondazione IRCCS San Gerardo dei Tintori, Monza; Neuroradiology Unit (M.S., A.E.), Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan; ALS Centre (M.S.), "Rita Levi Montalcini" Department of Neuroscience, University of Turin; Neuropathology Unit (P.C., G.D.F., M.C.), Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan; and Istituti Clinici Scientifici Maugeri IRCCS (A.Z., E.A.P.), Neurorehabilitation Unit of Milan Institute, Italy
| | - Giorgio B Boncoraglio
- From the Cerebrovascular Unit (G.P., A.P., G.G., T.C., G.M., C.T., I.C., N.R., G.B.B., A.B., L.G., B.S.), Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan; Department of Pharmacological and Biomolecular Sciences (G.P., A.P.), University of Milan; Department of Neurology (J.C.D., P.T.D.), Fondazione IRCCS San Gerardo dei Tintori, Monza; Neuroradiology Unit (M.S., A.E.), Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan; ALS Centre (M.S.), "Rita Levi Montalcini" Department of Neuroscience, University of Turin; Neuropathology Unit (P.C., G.D.F., M.C.), Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan; and Istituti Clinici Scientifici Maugeri IRCCS (A.Z., E.A.P.), Neurorehabilitation Unit of Milan Institute, Italy
| | - Jacopo C Difrancesco
- From the Cerebrovascular Unit (G.P., A.P., G.G., T.C., G.M., C.T., I.C., N.R., G.B.B., A.B., L.G., B.S.), Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan; Department of Pharmacological and Biomolecular Sciences (G.P., A.P.), University of Milan; Department of Neurology (J.C.D., P.T.D.), Fondazione IRCCS San Gerardo dei Tintori, Monza; Neuroradiology Unit (M.S., A.E.), Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan; ALS Centre (M.S.), "Rita Levi Montalcini" Department of Neuroscience, University of Turin; Neuropathology Unit (P.C., G.D.F., M.C.), Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan; and Istituti Clinici Scientifici Maugeri IRCCS (A.Z., E.A.P.), Neurorehabilitation Unit of Milan Institute, Italy
| | - Payam Tabaee Damavandi
- From the Cerebrovascular Unit (G.P., A.P., G.G., T.C., G.M., C.T., I.C., N.R., G.B.B., A.B., L.G., B.S.), Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan; Department of Pharmacological and Biomolecular Sciences (G.P., A.P.), University of Milan; Department of Neurology (J.C.D., P.T.D.), Fondazione IRCCS San Gerardo dei Tintori, Monza; Neuroradiology Unit (M.S., A.E.), Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan; ALS Centre (M.S.), "Rita Levi Montalcini" Department of Neuroscience, University of Turin; Neuropathology Unit (P.C., G.D.F., M.C.), Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan; and Istituti Clinici Scientifici Maugeri IRCCS (A.Z., E.A.P.), Neurorehabilitation Unit of Milan Institute, Italy
| | - Mario Stanziano
- From the Cerebrovascular Unit (G.P., A.P., G.G., T.C., G.M., C.T., I.C., N.R., G.B.B., A.B., L.G., B.S.), Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan; Department of Pharmacological and Biomolecular Sciences (G.P., A.P.), University of Milan; Department of Neurology (J.C.D., P.T.D.), Fondazione IRCCS San Gerardo dei Tintori, Monza; Neuroradiology Unit (M.S., A.E.), Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan; ALS Centre (M.S.), "Rita Levi Montalcini" Department of Neuroscience, University of Turin; Neuropathology Unit (P.C., G.D.F., M.C.), Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan; and Istituti Clinici Scientifici Maugeri IRCCS (A.Z., E.A.P.), Neurorehabilitation Unit of Milan Institute, Italy
| | - Alessandra Erbetta
- From the Cerebrovascular Unit (G.P., A.P., G.G., T.C., G.M., C.T., I.C., N.R., G.B.B., A.B., L.G., B.S.), Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan; Department of Pharmacological and Biomolecular Sciences (G.P., A.P.), University of Milan; Department of Neurology (J.C.D., P.T.D.), Fondazione IRCCS San Gerardo dei Tintori, Monza; Neuroradiology Unit (M.S., A.E.), Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan; ALS Centre (M.S.), "Rita Levi Montalcini" Department of Neuroscience, University of Turin; Neuropathology Unit (P.C., G.D.F., M.C.), Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan; and Istituti Clinici Scientifici Maugeri IRCCS (A.Z., E.A.P.), Neurorehabilitation Unit of Milan Institute, Italy
| | - Paola Caroppo
- From the Cerebrovascular Unit (G.P., A.P., G.G., T.C., G.M., C.T., I.C., N.R., G.B.B., A.B., L.G., B.S.), Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan; Department of Pharmacological and Biomolecular Sciences (G.P., A.P.), University of Milan; Department of Neurology (J.C.D., P.T.D.), Fondazione IRCCS San Gerardo dei Tintori, Monza; Neuroradiology Unit (M.S., A.E.), Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan; ALS Centre (M.S.), "Rita Levi Montalcini" Department of Neuroscience, University of Turin; Neuropathology Unit (P.C., G.D.F., M.C.), Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan; and Istituti Clinici Scientifici Maugeri IRCCS (A.Z., E.A.P.), Neurorehabilitation Unit of Milan Institute, Italy
| | - Giuseppe Di Fede
- From the Cerebrovascular Unit (G.P., A.P., G.G., T.C., G.M., C.T., I.C., N.R., G.B.B., A.B., L.G., B.S.), Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan; Department of Pharmacological and Biomolecular Sciences (G.P., A.P.), University of Milan; Department of Neurology (J.C.D., P.T.D.), Fondazione IRCCS San Gerardo dei Tintori, Monza; Neuroradiology Unit (M.S., A.E.), Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan; ALS Centre (M.S.), "Rita Levi Montalcini" Department of Neuroscience, University of Turin; Neuropathology Unit (P.C., G.D.F., M.C.), Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan; and Istituti Clinici Scientifici Maugeri IRCCS (A.Z., E.A.P.), Neurorehabilitation Unit of Milan Institute, Italy
| | - Marcella Catania
- From the Cerebrovascular Unit (G.P., A.P., G.G., T.C., G.M., C.T., I.C., N.R., G.B.B., A.B., L.G., B.S.), Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan; Department of Pharmacological and Biomolecular Sciences (G.P., A.P.), University of Milan; Department of Neurology (J.C.D., P.T.D.), Fondazione IRCCS San Gerardo dei Tintori, Monza; Neuroradiology Unit (M.S., A.E.), Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan; ALS Centre (M.S.), "Rita Levi Montalcini" Department of Neuroscience, University of Turin; Neuropathology Unit (P.C., G.D.F., M.C.), Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan; and Istituti Clinici Scientifici Maugeri IRCCS (A.Z., E.A.P.), Neurorehabilitation Unit of Milan Institute, Italy
| | - Aida Zulueta
- From the Cerebrovascular Unit (G.P., A.P., G.G., T.C., G.M., C.T., I.C., N.R., G.B.B., A.B., L.G., B.S.), Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan; Department of Pharmacological and Biomolecular Sciences (G.P., A.P.), University of Milan; Department of Neurology (J.C.D., P.T.D.), Fondazione IRCCS San Gerardo dei Tintori, Monza; Neuroradiology Unit (M.S., A.E.), Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan; ALS Centre (M.S.), "Rita Levi Montalcini" Department of Neuroscience, University of Turin; Neuropathology Unit (P.C., G.D.F., M.C.), Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan; and Istituti Clinici Scientifici Maugeri IRCCS (A.Z., E.A.P.), Neurorehabilitation Unit of Milan Institute, Italy
| | - Eugenio Agostino Parati
- From the Cerebrovascular Unit (G.P., A.P., G.G., T.C., G.M., C.T., I.C., N.R., G.B.B., A.B., L.G., B.S.), Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan; Department of Pharmacological and Biomolecular Sciences (G.P., A.P.), University of Milan; Department of Neurology (J.C.D., P.T.D.), Fondazione IRCCS San Gerardo dei Tintori, Monza; Neuroradiology Unit (M.S., A.E.), Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan; ALS Centre (M.S.), "Rita Levi Montalcini" Department of Neuroscience, University of Turin; Neuropathology Unit (P.C., G.D.F., M.C.), Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan; and Istituti Clinici Scientifici Maugeri IRCCS (A.Z., E.A.P.), Neurorehabilitation Unit of Milan Institute, Italy
| | - Anna Bersano
- From the Cerebrovascular Unit (G.P., A.P., G.G., T.C., G.M., C.T., I.C., N.R., G.B.B., A.B., L.G., B.S.), Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan; Department of Pharmacological and Biomolecular Sciences (G.P., A.P.), University of Milan; Department of Neurology (J.C.D., P.T.D.), Fondazione IRCCS San Gerardo dei Tintori, Monza; Neuroradiology Unit (M.S., A.E.), Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan; ALS Centre (M.S.), "Rita Levi Montalcini" Department of Neuroscience, University of Turin; Neuropathology Unit (P.C., G.D.F., M.C.), Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan; and Istituti Clinici Scientifici Maugeri IRCCS (A.Z., E.A.P.), Neurorehabilitation Unit of Milan Institute, Italy
| | - Laura Gatti
- From the Cerebrovascular Unit (G.P., A.P., G.G., T.C., G.M., C.T., I.C., N.R., G.B.B., A.B., L.G., B.S.), Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan; Department of Pharmacological and Biomolecular Sciences (G.P., A.P.), University of Milan; Department of Neurology (J.C.D., P.T.D.), Fondazione IRCCS San Gerardo dei Tintori, Monza; Neuroradiology Unit (M.S., A.E.), Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan; ALS Centre (M.S.), "Rita Levi Montalcini" Department of Neuroscience, University of Turin; Neuropathology Unit (P.C., G.D.F., M.C.), Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan; and Istituti Clinici Scientifici Maugeri IRCCS (A.Z., E.A.P.), Neurorehabilitation Unit of Milan Institute, Italy
| | - Benedetta Storti
- From the Cerebrovascular Unit (G.P., A.P., G.G., T.C., G.M., C.T., I.C., N.R., G.B.B., A.B., L.G., B.S.), Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan; Department of Pharmacological and Biomolecular Sciences (G.P., A.P.), University of Milan; Department of Neurology (J.C.D., P.T.D.), Fondazione IRCCS San Gerardo dei Tintori, Monza; Neuroradiology Unit (M.S., A.E.), Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan; ALS Centre (M.S.), "Rita Levi Montalcini" Department of Neuroscience, University of Turin; Neuropathology Unit (P.C., G.D.F., M.C.), Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan; and Istituti Clinici Scientifici Maugeri IRCCS (A.Z., E.A.P.), Neurorehabilitation Unit of Milan Institute, Italy
| |
Collapse
|
8
|
Rindler RS, Robertson H, De Yampert L, Khatri V, Texakalidis P, Eshraghi S, Grey S, Schobel S, Elster EA, Boulis N, Grossberg JA. Predicting Vasospasm and Early Mortality in Severe Traumatic Brain Injury: A Model Using Serum Cytokines, Neuronal Proteins, and Clinical Data. Neurosurgery 2024:00006123-990000000-01390. [PMID: 39471078 DOI: 10.1227/neu.0000000000003224] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Accepted: 08/28/2024] [Indexed: 11/01/2024] Open
Abstract
BACKGROUND AND OBJECTIVES Prediction of patient outcomes after severe traumatic brain injury (sTBI) is limited with current clinical tools. This study aimed to improve such prognostication by combining clinical data and serum inflammatory and neuronal proteins in patients with sTBI to develop predictive models for post-traumatic vasospasm (PTV) and mortality. METHODS Fifty-three adult civilian patients were prospectively enrolled in the sTBI arm of the Surgical Critical Care Initiative (SC2i). Clinical, serum inflammatory, and neuronal protein data were combined using the parsimonious machine learning methods of least absolute shrinkage and selection operator (LASSO) and classification and regression trees (CART) to construct parsimonious models for predicting development of PTV and mortality. RESULTS Thirty-six (67.9%) patients developed vasospasm and 10 (18.9%) died. The mean age was 39.2 years; 22.6% were women. CART identified lower IL9, lower presentation pulse rate, and higher eotaxin as predictors of vasospasm development (full data area under curve (AUC) = 0.89, mean cross-validated AUC = 0.47). LASSO identified higher Rotterdam computed tomography score and lower age as risk factors for vasospasm development (full data AUC 0.94, sensitivity 0.86, and specificity 0.94; cross-validation AUC 0.87, sensitivity 0.79, and specificity 0.93). CART identified high levels of eotaxin as most predictive of mortality (AUC 0.74, cross-validation AUC 0.57). LASSO identified higher serum IL6, lower IL12, and higher glucose as predictive of mortality (full data AUC 0.9, sensitivity 1.0, and specificity 0.72; cross-validation AUC 0.8, sensitivity 0.85, and specificity 0.79). CONCLUSION Inflammatory cytokine levels after sTBI may have predictive value that exceeds conventional clinical variables for certain outcomes. IL-9, pulse rate, and eotaxin as well as Rotterdam score and age predict development of PTV. Eotaxin, IL-6, IL-12, and glucose were predictive of mortality. These results warrant validation in a prospective cohort.
Collapse
Affiliation(s)
- Rima S Rindler
- Sierra Neurosurgery Group, Reno, Nevada, USA
- Department of Neurosurgery, Emory University, Atlanta, Georgia, USA
| | - Henry Robertson
- Surgical Critical Care Initiative (SC2i), Uniformed Services University of the Health Sciences, Bethesda, Maryland, USA
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc, North Bethesda, Maryland, USA
| | | | - Vivek Khatri
- Surgical Critical Care Initiative (SC2i), Uniformed Services University of the Health Sciences, Bethesda, Maryland, USA
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc, North Bethesda, Maryland, USA
| | - Pavlos Texakalidis
- Department of Neurosurgery, Emory University, Atlanta, Georgia, USA
- Department of Neurosurgery, Northwestern University, Chicago, Illinois, USA
| | - Sheila Eshraghi
- Department of Neurosurgery, Emory University, Atlanta, Georgia, USA
- Department of Neurosurgery, University of New Mexico School of Medicine, Albuquerque, New Mexico, USA
| | - Scott Grey
- Surgical Critical Care Initiative (SC2i), Uniformed Services University of the Health Sciences, Bethesda, Maryland, USA
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc, North Bethesda, Maryland, USA
| | - Seth Schobel
- Surgical Critical Care Initiative (SC2i), Uniformed Services University of the Health Sciences, Bethesda, Maryland, USA
| | - Eric A Elster
- Surgical Critical Care Initiative (SC2i), Uniformed Services University of the Health Sciences, Bethesda, Maryland, USA
- Walter Reed National Military Medical Center, Bethesda, Maryland, USA
| | - Nicholas Boulis
- Department of Neurosurgery, Emory University, Atlanta, Georgia, USA
| | | |
Collapse
|
9
|
Bosse M, Bélik F, van Pesch V, Bayart JL. Phosphorylated tau 181 (p-tau 181) as an innovative, fast and robust biomarker for cerebrospinal fluid leaks. J Neurol 2024; 271:6724-6728. [PMID: 39162804 DOI: 10.1007/s00415-024-12624-6] [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: 07/01/2024] [Revised: 07/30/2024] [Accepted: 08/02/2024] [Indexed: 08/21/2024]
Abstract
BACKGROUND Cerebrospinal fluid (CSF) leaks can lead to serious complications if left untreated, making rapid and accurate diagnosis essential. Biomarkers such as β2-transferrin (B2TRF) and β-trace protein are used to detect CSF leaks, but their limitations warrant the exploration of alternative markers. This study investigates the potential of phosphorylated tau at threonine 181 (p-tau181) as a biomarker for CSF leaks. METHODS Samples from 56 subjects were analyzed for B2TRF and p-tau181 using immunoaffinity blotting and chemiluminescent enzyme immunoassay, respectively. Data analysis included Mann-Whitney test to assess the overall difference in median p-tau181 concentrations between B2TRF positive and negative patients and a receiver operating characteristic (ROC) curve analysis to determine optimal p-tau181 cutoff values for predicting B2TRF positivity. RESULTS p-tau181 levels were significantly higher in B2TRF positive samples compared to negative samples (p < 0.001). ROC analysis showed high diagnostic performance for p-tau181, with an optimal cutoff of 13.22 pg/mL providing 92.0% sensitivity and 93.1% specificity. Excluding hemolyzed samples improved further the diagnostic performances, maintaining high sensitivity (90.9%) and achieving perfect specificity (100.0%). CONCLUSIONS This study highlights the potential of p-tau181 as a valuable biomarker for the detection of CSF leaks due to its high diagnostic accuracy and practical advantages over the current biomarkers. The characteristics of p-tau181 assay being both quantitative and rapid, with high diagnostic accuracy, suggest that it could be a valuable tool for the detection of CSF leaks. Further research are now needed to validate these findings.
Collapse
Affiliation(s)
- Maxime Bosse
- Department of Laboratory Medicine, Clinique Saint-Pierre, 1340, Ottignies, Belgium
| | - Florian Bélik
- Department of Laboratory Medicine, Cliniques Universitaires Saint-Luc, Brussels, Belgium
| | - Vincent van Pesch
- Department of Laboratory Medicine, Cliniques Universitaires Saint-Luc, Brussels, Belgium
- Department of Neurology, Cliniques Universitaires Saint-Luc, Brussels, Belgium
- Institute of Neuroscience, Université Catholique de Louvain, Brussels, Belgium
| | - Jean-Louis Bayart
- Department of Laboratory Medicine, Clinique Saint-Pierre, 1340, Ottignies, Belgium.
- Department of Laboratory Medicine, Cliniques Universitaires Saint-Luc, Brussels, Belgium.
| |
Collapse
|
10
|
Schöll M, Verberk IMW, Del Campo M, Delaby C, Therriault J, Chong JR, Palmqvist S, Alcolea D. Challenges in the practical implementation of blood biomarkers for Alzheimer's disease. THE LANCET. HEALTHY LONGEVITY 2024; 5:100630. [PMID: 39369727 DOI: 10.1016/j.lanhl.2024.07.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2023] [Revised: 06/23/2024] [Accepted: 07/29/2024] [Indexed: 10/08/2024] Open
Abstract
Blood biomarkers have emerged as accessible, cost-effective, and highly promising tools for advancing the diagnostics of Alzheimer's disease. However, transitioning from cerebrospinal fluid biomarkers to blood biomarkers-eg, to verify amyloid β pathology-requires careful consideration. This Series paper highlights the main challenges in the implementation of blood biomarkers for Alzheimer's disease in different possible contexts of use. Despite the robustness of measuring blood biomarker concentrations, the widespread adoption of blood biomarkers requires rigorous standardisation efforts to address inherent challenges in diverse contexts of use. The challenges include understanding the effect of pre-analytical and analytical conditions, potential confounding factors, and comorbidities that could influence outcomes of blood biomarkers and their use in diverse populations. Additionally, distinct scenarios present their own specific challenges. In memory clinics, the successful integration of blood biomarkers in diagnostic tests will require well-established diagnostic accuracy and comprehensive assessments of the effect of blood biomarkers on the diagnostic confidence and patient management of clinicians. In primary care settings, and even more when implemented in population-based screening programmes for which no experience with any biomarkers for Alzheimer's disease currently exists, the implementation of blood biomarkers will be challenged by the need for education of primary care clinical staff and clear guidelines. However, despite the challenges, blood biomarkers hold great promise for substantially enhancing the diagnostic accuracy and effectively streamlining referral processes, leading to earlier diagnosis and access to treatments. The ongoing efforts that are shaping the integration of blood biomarkers across diverse clinical settings pave the way towards precision medicine in Alzheimer's disease.
Collapse
Affiliation(s)
- Michael Schöll
- Wallenberg Centre for Molecular and Translational Medicine, University of Gothenburg, Mölndal, Sweden; Department of Psychiatry and Neurochemistry, University of Gothenburg, Mölndal, Sweden; Dementia Research Centre, Queen Square Institute of Neurology, University College London, London, UK; Department of Clinical Physiology, Sahlgrenska University Hospital, Gothenburg, Sweden.
| | - Inge M W Verberk
- Neurochemistry Laboratory, Department of Laboratory Medicine, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam Neuroscience, Amsterdam, Netherlands
| | - Marta Del Campo
- Barcelonaβeta Brain Research Center (BBRC), Pasqual Maragall Foundation, Barcelona, Spain; Hospital del Mar Research Institute (IMIM), Barcelona, Spain; Departamento de Ciencias Farmacéuticas y de la Salud, Facultad de Farmacia, Universidad San Pablo-CEU, CEU Universities, Madrid, Spain
| | - Constance Delaby
- LBPC-PPC, University of Montpellier, CHU Montpellier, INM INSERM, Montpellier, France; Institut de Recerca Sant Pau (IR SANT PAU), Barcelona, Spain
| | - Joseph Therriault
- Translational Neuroimaging Laboratory, McGill Centre for Studies in Aging, McGill University, Montreal, QC, Canada; Montreal Neurological Institute, Department of Neurology and Neurosurgery, McGill University, Montreal, QC, Canada
| | - Joyce R Chong
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore; Memory, Aging and Cognition Centre, National University Health Systems, Singapore
| | - Sebastian Palmqvist
- Clinical Memory Research Unit, Clinical Sciences in Malmö, Lund University, Lund, Sweden; Memory Clinic, Skåne University Hospital, Malmö, Sweden
| | - Daniel Alcolea
- Sant Pau Memory Unit, Department of Neurology, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain; Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain.
| |
Collapse
|
11
|
Mastandrea P, Mengozzi S, Bernardini S. Systematic review and meta-analysis of observational studies evaluating glial fibrillary acidic protein (GFAP) and ubiquitin C-terminal hydrolase L1 (UCHL1) as blood biomarkers of mild acute traumatic brain injury (mTBI) or sport-related concussion (SRC) in adult subjects. Diagnosis (Berl) 2024:dx-2024-0078. [PMID: 39167371 DOI: 10.1515/dx-2024-0078] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2024] [Accepted: 07/14/2024] [Indexed: 08/23/2024]
Abstract
INTRODUCTION Neurotrauma is the leading cause of death in individuals <45 years old. Many of the published articles on UCHL1 and GFAP lack rigorous methods and reporting. CONTENT Due to the high heterogeneity between studies, we evaluated blood GFAP and UCHL1 levels in the same subjects. We determined the biomarker congruence among areas under the ROC curves (AUCs), sensitivities, specificities, and laboratory values in ng/L to avoid spurious results. The definitive meta-analysis included 1,880 subjects in eight studies. The items with the highest risk of bias were as follows: cut-off not prespecified and case-control design not avoided. The AUC of GFAP was greater than the AUC of UCHL1, with a lower prediction interval (PI) limit of 50.1 % for GFAP and 37.3 % for UCHL1, and a significantly greater percentage of GFAP Sp. The PI of laboratory results for GFAP and UCHL1 were 0.517-7,518 ng/L (diseased), 1.2-255 ng/L (nondiseased), and 3-4,180 vs. 3.2-1,297 ng/L, respectively. SUMMARY Only the GFAP positive cut-off (255 ng/L) appears to be reliable. The negative COs appear unreliable. OUTLOOK GFAP needs better standardization. However, the AUCs of the phospho-Tau and phospho-Tau/Tau proteins resulted not significantly lower than AUC of GFAP, but this result needs further verifications.
Collapse
Affiliation(s)
- Paolo Mastandrea
- Department of Clinical Pathology, 90384 Azienda Ospedaliera di Rilievo Nazionale e di Alta Specialità San Giuseppe Moscati , Salerno, Italy
| | - Silvia Mengozzi
- U.O. Patologia Clinica, AUSL della Romagna, Laboratorio Unico, Cesena, Forli'-Cesena, Italy
| | - Sergio Bernardini
- Department of Experimental Medicine and Surgery, "Tor Vergata" University Hospital, Rome, Rome, Italy
| |
Collapse
|
12
|
Behzadi F, Luy DD, Schaible PA, Zywiciel JF, Puccio AM, Germanwala AV. A systematic review and meta-analysis of major blood protein biomarkers that predict unfavorable outcomes in severe traumatic brain injury. Clin Neurol Neurosurg 2024; 242:108312. [PMID: 38733758 DOI: 10.1016/j.clineuro.2024.108312] [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: 03/28/2024] [Revised: 04/28/2024] [Accepted: 04/29/2024] [Indexed: 05/13/2024]
Abstract
INTRODUCTION Severe traumatic brain injury (TBI) presentation and late clinical outcomes are usually evaluated by the Glasgow Outcome Scale-Extended (GOS-E), which lacks strong prognostic predictability. Several blood biomarkers have been linked to TBI, such as Tau, GFAP, UCH-L1, S-100B, and NSE. Clinical values of TBI biomarkers have yet to be evaluated in a focused multi-study meta-analysis. We reviewed relevant articles evaluating potential relationships between TBI biomarkers and both early and 6-month outcomes. METHODS All PubMed article publications from January 2000 to November 2023 with the search criteria "Protein Biomarker" AND "Traumatic Brain Injury" were included. Amongst all comparative studies, the sensitivity means and range values of biomarkers in predicting CT Rotterdam scores, ICU admission in the early period, or predicting GOS-E < 4 at the 6-month period were calculated from confusion matrices. Sensitivity values were modeled for each biomarker across studies and compared statistically for heterogeneity and differences. RESULTS From the 65 articles that met the criteria, 13 were included in this study. Six articles involved early-period TBI outcomes and seven involved 6-month outcomes. In the early period TBI outcomes, GFAP had a superior sensitivity to UCH-L1 and S-100B, and similar sensitivity to the CT Rotterdam score. In the 6-month period TBI outcomes, total Tau and NSE both had significant interstudy heterogeneity, making them inferior to GFAP, phosphorylated Tau, UCH-L1, and S-100B, all four of which had similar sensitivities at 75 %. This sensitivity range at 6-month outcomes was still relatively inferior to the CT Rotterdam score. Total Tau did not show any prognostic advantage at six months with GOS-E < 4, and phosphorylated Tau was similar in its sensitivity to other biomarkers such as GFAP and UCH-L1 and still inferior to the CT Rotterdam score. CONCLUSION This data suggests that TBI protein biomarkers do not possess better prognostic value with regards to outcomes.
Collapse
Affiliation(s)
- Faraz Behzadi
- Loyola University Medical Center, Department of Neurological Surgery, USA
| | - Diego D Luy
- Loyola University Medical Center, Department of Neurological Surgery, USA
| | - Peter A Schaible
- Midwestern University Chicago, College of Osteopathic Medicine, USA
| | | | - Ava M Puccio
- University of Pittsburgh Medical Center, Department of Neurological Surgery, USA
| | - Anand V Germanwala
- Loyola University Medical Center, Department of Neurological Surgery, USA; Loyola University Chicago, Stritch School of Medicine, USA.
| |
Collapse
|
13
|
Lu KP, Zhou XZ. Pin1-catalyzed conformational regulation after phosphorylation: A distinct checkpoint in cell signaling and drug discovery. Sci Signal 2024; 17:eadi8743. [PMID: 38889227 PMCID: PMC11409840 DOI: 10.1126/scisignal.adi8743] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2023] [Accepted: 05/30/2024] [Indexed: 06/20/2024]
Abstract
Protein phosphorylation is one of the most common mechanisms regulating cellular signaling pathways, and many kinases and phosphatases are proven drug targets. Upon phosphorylation, protein functions can be further regulated by the distinct isomerase Pin1 through cis-trans isomerization. Numerous protein targets and many important roles have now been elucidated for Pin1. However, no tools are available to detect or target cis and trans conformation events in cells. The development of Pin1 inhibitors and stereo- and phospho-specific antibodies has revealed that cis and trans conformations have distinct and often opposing cellular functions. Aberrant conformational changes due to the dysregulation of Pin1 can drive pathogenesis but can be effectively targeted in age-related diseases, including cancers and neurodegenerative disorders. Here, we review advances in understanding the roles of Pin1 signaling in health and disease and highlight conformational regulation as a distinct signal transduction checkpoint in disease development and treatment.
Collapse
Affiliation(s)
- Kun Ping Lu
- Departments of Biochemistry and Oncology, Schulich School of Medicine & Dentistry
- Robarts Research Institute, Schulich School of Medicine & Dentistry
| | - Xiao Zhen Zhou
- Departments of Biochemistry and Oncology, Schulich School of Medicine & Dentistry
- Department of Pathology and Laboratory Medicine, Schulich School of Medicine & Dentistry
- Lawson Health Research Institute, Western University, London, ON N6G 2V4, Canada
| |
Collapse
|
14
|
Stellpflug SJ, Dalrymple KA, Stone D, Southgate S, Bachman DS, LeFevere RC, Hasan J, Zwank MD. Impact of repeated sportive chokes on carotid intima media thickness and brain injury biomarkers in grappling athletes. PHYSICIAN SPORTSMED 2024:1-9. [PMID: 38857060 DOI: 10.1080/00913847.2024.2366154] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/16/2024] [Accepted: 06/06/2024] [Indexed: 06/11/2024]
Abstract
PURPOSE Vascular neck compression techniques, referred to as 'chokes' in combat sports, reduce cerebral perfusion, causing loss of consciousness or voluntary submission by the choked athlete. Despite these chokes happening millions of times yearly around the world, there is scant research on their long-term effects. This pilot study evaluated whether repeated choking in submission grappling impacts the carotid intima media thickness (CIMT) and brain injury biomarkers (NFL, hGFAP, t-Tau, and UCH-L1). METHODS Participants (n = 39, 29 male; ages 27-60 years) were assigned to one of two study arms: Grapplers (n = 20, 15 male) and 19 age/sex/body size matched controls. Grapplers had been exposed to >500 choke events while training for >5 years in a choke-inclusive sport. Exclusion criteria were recent TBI or deficits from a past TBI or stroke. Bilateral ultrasound measurement of the CIMT was performed, and blood was collected for quantitative analysis of four brain injury markers. Subgroup analyses were performed within the Grappler group to account for blunt head trauma as a possible confounder. RESULTS There was no overall difference in CIMT measurements between Grapplers (mean 0.55 mm, SD 0.07) and Controls (mean 0.57 mm, SD 0.10) p = 0.498 [95% CI -0.04-0.08], nor were there CIMT differences between Grappler subgroups of blunt Trauma and No-Trauma. There were no significant differences in any biomarkers comparing Grapplers and Controls or comparing Grappler subgroups of Trauma and No-Trauma. CONCLUSION This study found no significant difference in CIMT and serum brain injury biomarkers between controls and grapplers with extensive transient choke experience, nor between grapplers with extensive past blunt head trauma and those without.
Collapse
Affiliation(s)
| | | | - Daniel Stone
- Department of Emergency Medicine, Regions Hospital, Saint Paul, MN, USA
| | - Samuel Southgate
- Department of Emergency Medicine, Regions Hospital, Saint Paul, MN, USA
| | - David S Bachman
- Critical Care Research Center, HealthPartners, Saint Paul, MN, USA
| | - Robert C LeFevere
- Department of Emergency Medicine, Regions Hospital, Saint Paul, MN, USA
| | - Jaan Hasan
- Department of Emergency Medicine, Wyckoff Heights Medical Center, Brooklyn, NY, USA
| | - Michael D Zwank
- Department of Emergency Medicine, Regions Hospital, Saint Paul, MN, USA
| |
Collapse
|
15
|
Singh AK, Asif S, Pandey DK, Chaudhary A, Kapoor V, Verma PK. Biomarkers in Acute Traumatic Brain Injury: A Systematic Review and Meta-Analysis. Cureus 2024; 16:e63020. [PMID: 39050316 PMCID: PMC11268976 DOI: 10.7759/cureus.63020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/24/2024] [Indexed: 07/27/2024] Open
Abstract
Traumatic brain injury (TBI) stands as a significant contributor to traumatic death and disability worldwide. In recent years, researchers have identified biomarkers to gauge useful outcomes in TBI patients. However, the enigma of timely sample collection to measure the biomarkers remains a controversial point in the case of TBI, unlike other degenerative diseases like Alzheimer's disease and Parkinson's disease, where we can collect the sample at any point in time. The purpose of this study is to evaluate the sensitivity of biomarkers in TBI concerning time of injury by analyzing recent available data on biomarkers in the medical literature. A total of 2,256 studies were initially retrieved from the search engine. After an initial screening, only 1,750 unique articles remained. After excluding review articles, animal studies, meta-analysis, and studies with children (screened by title and abstract), 30 kinds of literature were found relevant to search the required variables. Further 16 studies were excluded due to the nonavailability of complete variables or data. Finally, 14 studies remained and were included in the analysis. This study has analyzed the four most commonly described biomarkers for TBI in the literature: glial fibrillary acidic protein (GFAP), S100 calcium-binding protein B, ubiquitin carboxy-terminal hydrolase L1, and Tau. According to this statistical analysis, all biomarkers included in the study have shown their serum levels after trauma. So, all these biomarkers can be used for further study in the outcome prediction and diagnosis of TBI patients. The meta-analysis suggests that the best biomarker for TBI is Tau in cases where sample collection is done within 24 hours, while GFAP is the best biomarker to be studied for TBI if sample collection is done 24 hours after trauma.
Collapse
Affiliation(s)
- Adarsh Kumar Singh
- Department of Biotechnology, Centre of BioMedical Research (CBMR) Sanjay Gandhi Postgraduate Institute of Medical Sciences, Lucknow, IND
| | - Shafaque Asif
- Department of Molecular Medicine, Sanjay Gandhi Postgraduate Institute of Medical Sciences, Lucknow, IND
| | - Deepika Kumari Pandey
- Department of Neurosurgery, Sanjay Gandhi Postgraduate Institute of Medical Sciences, Lucknow, IND
| | - Akash Chaudhary
- Department of Neurosurgery, Sanjay Gandhi Postgraduate Institute of Medical Sciences, Lucknow, IND
| | - Vishwas Kapoor
- Department of Biostatistics and Health Informatics, Sanjay Gandhi Postgraduate Institute of Medical Sciences, Lucknow, IND
| | - Pawan Kumar Verma
- Department of Neurosurgery, Sanjay Gandhi Postgraduate Institute of Medical Sciences, Lucknow, IND
| |
Collapse
|
16
|
Clarke GJB, Follestad T, Skandsen T, Zetterberg H, Vik A, Blennow K, Olsen A, Håberg AK. Chronic immunosuppression across 12 months and high ability of acute and subacute CNS-injury biomarker concentrations to identify individuals with complicated mTBI on acute CT and MRI. J Neuroinflammation 2024; 21:109. [PMID: 38678300 PMCID: PMC11056044 DOI: 10.1186/s12974-024-03094-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2024] [Accepted: 04/05/2024] [Indexed: 04/29/2024] Open
Abstract
BACKGROUND Identifying individuals with intracranial injuries following mild traumatic brain injury (mTBI), i.e. complicated mTBI cases, is important for follow-up and prognostication. The main aims of our study were (1) to assess the temporal evolution of blood biomarkers of CNS injury and inflammation in individuals with complicated mTBI determined on computer tomography (CT) and magnetic resonance imaging (MRI); (2) to assess the corresponding discriminability of both single- and multi-biomarker panels, from acute to chronic phases after injury. METHODS Patients with mTBI (n = 207), defined as Glasgow Coma Scale score between 13 and 15, loss of consciousness < 30 min and post-traumatic amnesia < 24 h, were included. Complicated mTBI - i.e., presence of any traumatic intracranial injury on neuroimaging - was present in 8% (n = 16) on CT (CT+) and 12% (n = 25) on MRI (MRI+). Blood biomarkers were sampled at four timepoints following injury: admission (within 72 h), 2 weeks (± 3 days), 3 months (± 2 weeks) and 12 months (± 1 month). CNS biomarkers included were glial fibrillary acidic protein (GFAP), neurofilament light (NFL) and tau, along with 12 inflammation markers. RESULTS The most discriminative single biomarkers of traumatic intracranial injury were GFAP at admission (CT+: AUC = 0.78; MRI+: AUC = 0.82), and NFL at 2 weeks (CT+: AUC = 0.81; MRI+: AUC = 0.89) and 3 months (MRI+: AUC = 0.86). MIP-1β and IP-10 concentrations were significantly lower across follow-up period in individuals who were CT+ and MRI+. Eotaxin and IL-9 were significantly lower in individuals who were MRI+ only. FGF-basic concentrations increased over time in MRI- individuals and were significantly higher than MRI+ individuals at 3 and 12 months. Multi-biomarker panels improved discriminability over single biomarkers at all timepoints (AUCs > 0.85 for admission and 2-week models classifying CT+ and AUC ≈ 0.90 for admission, 2-week and 3-month models classifying MRI+). CONCLUSIONS The CNS biomarkers GFAP and NFL were useful single diagnostic biomarkers of complicated mTBI, especially in acute and subacute phases after mTBI. Several inflammation markers were suppressed in patients with complicated versus uncomplicated mTBI and remained so even after 12 months. Multi-biomarker panels improved diagnostic accuracy at all timepoints, though at acute and 2-week timepoints, the single biomarkers GFAP and NFL, respectively, displayed similar accuracy compared to multi-biomarker panels.
Collapse
Affiliation(s)
- Gerard Janez Brett Clarke
- Department of Radiology and Nuclear Medicine, St. Olavs Hospital, Trondheim University Hospital, Trondheim, Norway
- Department of Neuromedicine and Movement Sciences, NTNU, Trondheim, Norway
| | - Turid Follestad
- Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology (NTNU), Trondheim, N-7491, Norway
| | - Toril Skandsen
- Department of Neuromedicine and Movement Sciences, NTNU, Trondheim, Norway
- Clinic of Rehabilitation, St. Olavs Hospital, Trondheim University Hospital, Trondheim, Norway
| | - Henrik Zetterberg
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Mölndal, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
- Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, Queen Square, London, UK
- UK Dementia Research Institute at UCL, London, UK
- Hong Kong Center for Neurodegenerative Diseases, Clear Water Bay, Sha Tin, Hong Kong, China
- Wisconsin Alzheimer's Disease Research Center, University of Wisconsin School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, USA
| | - Anne Vik
- Department of Neuromedicine and Movement Sciences, NTNU, Trondheim, Norway
- Department of Neurosurgery, St Olavs Hospital, Trondheim University Hospital, Trondheim, Norway
| | - Kaj Blennow
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Mölndal, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
| | - Alexander Olsen
- Clinic of Rehabilitation, St. Olavs Hospital, Trondheim University Hospital, Trondheim, Norway
- Department of Psychology, Norwegian University of Science and Technology, Trondheim, Norway
- NorHEAD - Norwegian Centre for Headache Research, Trondheim, Norway
| | - Asta Kristine Håberg
- Department of Radiology and Nuclear Medicine, St. Olavs Hospital, Trondheim University Hospital, Trondheim, Norway.
- Department of Neuromedicine and Movement Sciences, NTNU, Trondheim, Norway.
| |
Collapse
|
17
|
Lai JD, Berlind JE, Fricklas G, Lie C, Urenda JP, Lam K, Sta Maria N, Jacobs R, Yu V, Zhao Z, Ichida JK. KCNJ2 inhibition mitigates mechanical injury in a human brain organoid model of traumatic brain injury. Cell Stem Cell 2024; 31:519-536.e8. [PMID: 38579683 DOI: 10.1016/j.stem.2024.03.004] [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: 04/27/2023] [Revised: 11/21/2023] [Accepted: 03/06/2024] [Indexed: 04/07/2024]
Abstract
Traumatic brain injury (TBI) strongly correlates with neurodegenerative disease. However, it remains unclear which neurodegenerative mechanisms are intrinsic to the brain and which strategies most potently mitigate these processes. We developed a high-intensity ultrasound platform to inflict mechanical injury to induced pluripotent stem cell (iPSC)-derived cortical organoids. Mechanically injured organoids elicit classic hallmarks of TBI, including neuronal death, tau phosphorylation, and TDP-43 nuclear egress. We found that deep-layer neurons were particularly vulnerable to injury and that TDP-43 proteinopathy promotes cell death. Injured organoids derived from C9ORF72 amyotrophic lateral sclerosis/frontotemporal dementia (ALS/FTD) patients displayed exacerbated TDP-43 dysfunction. Using genome-wide CRISPR interference screening, we identified a mechanosensory channel, KCNJ2, whose inhibition potently mitigated neurodegenerative processes in vitro and in vivo, including in C9ORF72 ALS/FTD organoids. Thus, targeting KCNJ2 may reduce acute neuronal death after brain injury, and we present a scalable, genetically flexible cerebral organoid model that may enable the identification of additional modifiers of mechanical stress.
Collapse
Affiliation(s)
- Jesse D Lai
- Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA; Amgen Inc., Thousand Oaks, CA, USA; Neurological & Rare Diseases, Dewpoint Therapeutics, Boston, MA, USA.
| | - Joshua E Berlind
- Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA; Eli and Edythe Broad CIRM Center for Regenerative Medicine and Stem Cell Research, University of Southern California, Los Angeles, CA, USA
| | - Gabriella Fricklas
- Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA; Eli and Edythe Broad CIRM Center for Regenerative Medicine and Stem Cell Research, University of Southern California, Los Angeles, CA, USA
| | - Cecilia Lie
- Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA; Eli and Edythe Broad CIRM Center for Regenerative Medicine and Stem Cell Research, University of Southern California, Los Angeles, CA, USA
| | - Jean-Paul Urenda
- Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA; Eli and Edythe Broad CIRM Center for Regenerative Medicine and Stem Cell Research, University of Southern California, Los Angeles, CA, USA
| | - Kelsey Lam
- Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA; Eli and Edythe Broad CIRM Center for Regenerative Medicine and Stem Cell Research, University of Southern California, Los Angeles, CA, USA
| | - Naomi Sta Maria
- Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Russell Jacobs
- Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Violeta Yu
- Amgen Inc., Thousand Oaks, CA, USA; Neurological & Rare Diseases, Dewpoint Therapeutics, Boston, MA, USA
| | - Zhen Zhao
- Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Justin K Ichida
- Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA; Eli and Edythe Broad CIRM Center for Regenerative Medicine and Stem Cell Research, University of Southern California, Los Angeles, CA, USA; Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA.
| |
Collapse
|
18
|
Qiu C, Li Z, Leigh DA, Duan B, Stucky JE, Kim N, Xie G, Lu KP, Zhou XZ. The role of the Pin1- cis P-tau axis in the development and treatment of vascular contribution to cognitive impairment and dementia and preeclampsia. Front Cell Dev Biol 2024; 12:1343962. [PMID: 38628595 PMCID: PMC11019028 DOI: 10.3389/fcell.2024.1343962] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2023] [Accepted: 03/14/2024] [Indexed: 04/19/2024] Open
Abstract
Tauopathies are neurodegenerative diseases characterized by deposits of abnormal Tau protein in the brain. Conventional tauopathies are often defined by a limited number of Tau epitopes, notably neurofibrillary tangles, but emerging evidence suggests structural heterogeneity among tauopathies. The prolyl isomerase Pin1 isomerizes cis P-tau to inhibit the development of oligomers, tangles and neurodegeneration in multiple neurodegenerative diseases such as Alzheimer's disease, traumatic brain injury, vascular contribution to cognitive impairment and dementia (VCID) and preeclampsia (PE). Thus, cis P-tau has emerged as an early etiological driver, blood marker and therapeutic target for multiple neurodegenerative diseases, with clinical trials ongoing. The discovery of cis P-tau and other tau pathologies in VCID and PE calls attention for simplistic classification of tauopathy in neurodegenerative diseases. These recent advances have revealed the exciting novel role of the Pin1-cis P-tau axis in the development and treatment of vascular contribution to cognitive impairment and dementia and preeclampsia.
Collapse
Affiliation(s)
- Chenxi Qiu
- Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, United States
| | - Zhixiong Li
- Departments of Biochemistry and Oncology, Schulich School of Medicine and Dentistry and Robarts Research Institute, Western University, London, ON, Canada
| | - David A. Leigh
- Department of Genetics, Harvard Medical School, Boston, MA, United States
| | - Bingbing Duan
- Department of Biological Sciences, University of Pittsburgh, Pittsburgh, PA, United States
| | - Joseph E. Stucky
- Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, United States
| | - Nami Kim
- Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, United States
| | - George Xie
- Departments of Biochemistry and Oncology, Schulich School of Medicine and Dentistry and Robarts Research Institute, Western University, London, ON, Canada
| | - Kun Ping Lu
- Departments of Biochemistry and Oncology, Schulich School of Medicine and Dentistry and Robarts Research Institute, Western University, London, ON, Canada
| | - Xiao Zhen Zhou
- Departments of Biochemistry and Oncology, Schulich School of Medicine and Dentistry and Robarts Research Institute, Western University, London, ON, Canada
- Departments of Pathology and Laboratory Medicine, Schulich School of Medicine and Dentistry, and Lawson Health Research Institute, Western University, London, ON, Canada
| |
Collapse
|
19
|
Clarke GJB, Skandsen T, Zetterberg H, Follestad T, Einarsen CE, Vik A, Mollnes TE, Pischke SE, Blennow K, Håberg AK. Longitudinal Associations Between Persistent Post-Concussion Symptoms and Blood Biomarkers of Inflammation and CNS-Injury After Mild Traumatic Brain Injury. J Neurotrauma 2024; 41:862-878. [PMID: 38117157 DOI: 10.1089/neu.2023.0419] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2023] Open
Abstract
The aim of our study was to investigate the biological underpinnings of persistent post-concussion symptoms (PPCS) at 3 months following mild traumatic brain injury (mTBI). Patients (n = 192, age 16-60 years) with mTBI, defined as Glasgow Coma Scale (GCS) score between 13 and 15, loss of consciousness (LOC) <30 min, and post-traumatic amnesia (PTA) <24 h were included. Blood samples were collected at admission (within 72 h), 2 weeks, and 3 months. Concentrations of blood biomarkers associated with central nervous system (CNS) damage (glial fibrillary acidic protein [GFAP], neurofilament light [NFL], and tau) and inflammation (interferon gamma [IFNγ], interleukin [IL]-8, eotaxin, macrophage inflammatory protein-1-beta [MIP]-1β, monocyte chemoattractant protein [MCP]-1, interferon-gamma-inducible protein [IP]-10, IL-17A, IL-9, tumor necrosis factor [TNF], basic fibroblast growth factor [FGF]-basic platelet-derived growth factor [PDGF], and IL-1 receptor antagonist [IL-1ra]) were obtained. Demographic and injury-related factors investigated were age, sex, GCS score, LOC, PTA duration, traumatic intracranial finding on magnetic resonance imaging (MRI; within 72 h), and extracranial injuries. Delta values, that is, time-point differences in biomarker concentrations between 2 weeks minus admission and 3 months minus admission, were also calculated. PPCS was assessed with the British Columbia Post-Concussion Symptom Inventory (BC-PSI). In single variable analyses, longer PTA duration and a higher proportion of intracranial findings on MRI were found in the PPCS group, but no single biomarker differentiated those with PPCS from those without. In multi-variable models, female sex, longer PTA duration, MRI findings, and lower GCS scores were associated with increased risk of PPCS. Inflammation markers, but not GFAP, NFL, or tau, were associated with PPCS. At admission, higher concentrations of IL-8 and IL-9 and lower concentrations of TNF, IL-17a, and MCP-1 were associated with greater likelihood of PPCS; at 2 weeks, higher IL-8 and lower IFNγ were associated with PPCS; at 3 months, higher PDGF was associated with PPCS. Higher delta values of PDGF, IL-17A, and FGF-basic at 2 weeks compared with admission, MCP-1 at 3 months compared with admission, and TNF at 2 weeks and 3 months compared with admission were associated with greater likelihood of PPCS. Higher IL-9 delta values at both time-point comparisons were negatively associated with PPCS. Discriminability of individual CNS-injury and inflammation biomarkers for PPCS was around chance level, whereas the optimal combination of biomarkers yielded areas under the curve (AUCs) between 0.62 and 0.73. We demonstrate a role of biological factors on PPCS, including both positive and negative effects of inflammation biomarkers that differed based on sampling time-point after mTBI. PPCS was associated more with acute inflammatory processes, rather than ongoing inflammation or CNS-injury biomarkers. However, the modest discriminative ability of the models suggests other factors are more important in the development of PPCS.
Collapse
Affiliation(s)
- Gerard Janez Brett Clarke
- Department of Radiology and Nuclear Medicine, St. Olavs Hospital, Trondheim University Hospital, Trondheim, Norway
- Department of Neuromedicine and Movement Sciences, Department of Clinical and Molecular Research, Norwegian University of Science and Technology (NTNU), Trondheim, Norway
| | - Toril Skandsen
- Department of Neuromedicine and Movement Sciences, Department of Clinical and Molecular Research, Norwegian University of Science and Technology (NTNU), Trondheim, Norway
- Clinic of Rehabilitation, St. Olavs Hospital, Trondheim University Hospital, Trondheim, Norway
| | - Henrik Zetterberg
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, the Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
- Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, University College London, London, United Kingdom
- UK Dementia Research Institute at UCL, University College London, London, United Kingdom
- Hong Kong Center for Neurodegenerative Diseases, Clear Water Bay, Hong Kong, China
- Wisconsin Alzheimer's Disease Research Center, University of Wisconsin School of Medicine and Public Health, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Turid Follestad
- Department of Clinical and Molecular Medicine, Department of Clinical and Molecular Research, Norwegian University of Science and Technology (NTNU), Trondheim, Norway
- Clinical Research Unit Central Norway, St. Olavs Hospital, Trondheim University Hospital, Trondheim, Norway
| | - Cathrine Elisabeth Einarsen
- Department of Neuromedicine and Movement Sciences, Department of Clinical and Molecular Research, Norwegian University of Science and Technology (NTNU), Trondheim, Norway
- Clinic of Rehabilitation, St. Olavs Hospital, Trondheim University Hospital, Trondheim, Norway
| | - Anne Vik
- Department of Neuromedicine and Movement Sciences, Department of Clinical and Molecular Research, Norwegian University of Science and Technology (NTNU), Trondheim, Norway
- Department of Neurosurgery, St. Olavs Hospital, Trondheim University Hospital, Trondheim, Norway
| | - Tom Eirik Mollnes
- Department of Immunology, Department of Anesthesiology and Intensive Care Medicine, Oslo University Hospital and University of Oslo, Oslo, Norway
- Center of Molecular Inflammation Research, Department of Clinical and Molecular Research, Norwegian University of Science and Technology (NTNU), Trondheim, Norway
| | - Søren Erik Pischke
- Department of Immunology, Department of Anesthesiology and Intensive Care Medicine, Oslo University Hospital and University of Oslo, Oslo, Norway
- Clinic for Emergencies and Critical Care, Department of Anesthesiology and Intensive Care Medicine, Oslo University Hospital and University of Oslo, Oslo, Norway
| | - Kaj Blennow
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, the Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
| | - Asta Kristine Håberg
- Department of Radiology and Nuclear Medicine, St. Olavs Hospital, Trondheim University Hospital, Trondheim, Norway
- Department of Neuromedicine and Movement Sciences, Department of Clinical and Molecular Research, Norwegian University of Science and Technology (NTNU), Trondheim, Norway
| |
Collapse
|
20
|
Graham N, Zimmerman K, Heslegrave AJ, Keshavan A, Moro F, Abed-Maillard S, Bernini A, Dunet V, Garbero E, Nattino G, Chieregato A, Fainardi E, Baciu C, Gradisek P, Magnoni S, Oddo M, Bertolini G, Schott JM, Zetterberg H, Sharp D. Alzheimer's disease marker phospho-tau181 is not elevated in the first year after moderate-to-severe TBI. J Neurol Neurosurg Psychiatry 2024; 95:356-359. [PMID: 37833041 PMCID: PMC10958285 DOI: 10.1136/jnnp-2023-331854] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/17/2023] [Accepted: 09/19/2023] [Indexed: 10/15/2023]
Abstract
BACKGROUND Traumatic brain injury (TBI) is associated with the tauopathies Alzheimer's disease and chronic traumatic encephalopathy. Advanced immunoassays show significant elevations in plasma total tau (t-tau) early post-TBI, but concentrations subsequently normalise rapidly. Tau phosphorylated at serine-181 (p-tau181) is a well-validated Alzheimer's disease marker that could potentially seed progressive neurodegeneration. We tested whether post-traumatic p-tau181 concentrations are elevated and relate to progressive brain atrophy. METHODS Plasma p-tau181 and other post-traumatic biomarkers, including total-tau (t-tau), neurofilament light (NfL), ubiquitin carboxy-terminal hydrolase L1 (UCH-L1) and glial fibrillary acidic protein (GFAP), were assessed after moderate-to-severe TBI in the BIO-AX-TBI cohort (first sample mean 2.7 days, second sample within 10 days, then 6 weeks, 6 months and 12 months, n=42). Brain atrophy rates were assessed in aligned serial MRI (n=40). Concentrations were compared patients with and without Alzheimer's disease, with healthy controls. RESULTS Plasma p-tau181 concentrations were significantly raised in patients with Alzheimer's disease but not after TBI, where concentrations were non-elevated, and remained stable over one year. P-tau181 after TBI was not predictive of brain atrophy rates in either grey or white matter. In contrast, substantial trauma-associated elevations in t-tau, NfL, GFAP and UCH-L1 were seen, with concentrations of NfL and t-tau predictive of brain atrophy rates. CONCLUSIONS Plasma p-tau181 is not significantly elevated during the first year after moderate-to-severe TBI and levels do not relate to neuroimaging measures of neurodegeneration.
Collapse
Affiliation(s)
- Neil Graham
- Brain Sciences, Imperial College London, London, UK
- UK Dementia Research Institute Centre for Care Research and Technology, Imperial College London, London, UK
| | - Karl Zimmerman
- Brain Sciences, Imperial College London, London, UK
- UK Dementia Research Institute Centre for Care Research and Technology, Imperial College London, London, UK
| | | | - Ashvini Keshavan
- Dementia Research Centre, UCL Queen Square Institute of Neurology, London, UK
| | - Federico Moro
- Laboratory of Acute Brain Injury and Neuroprotection, Department of Acute Brain and Cardiovascular Injury, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy
- Dipartimento di Anestesia e Rianimazione, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Samia Abed-Maillard
- Neuroscience Critical Care Research Group, Department of Intensive Care Medicine, CHUV Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Adriano Bernini
- Department of Clinical Neurosciences, CHUV Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Vincent Dunet
- Department of Medical Radiology, CHUV Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Elena Garbero
- Laboratory of Clinical Epidemiology, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Ranica, Italy
| | - Giovanni Nattino
- Laboratory of Clinical Epidemiology, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Ranica, Italy
| | - Arturo Chieregato
- Terapia Intensiva ad indirizzo Neurologico & Neurochirurgico, ASST Grande Ospedale Metropolitano Niguarda, Milan, Italy
| | - Enrico Fainardi
- Department of Experimental and Clinical Sciences, Careggi University Hospital and University of Firenze, Florence, Italy
| | - Camelia Baciu
- Terapia Intensiva ad indirizzo Neurologico & Neurochirurgico, ASST Grande Ospedale Metropolitano Niguarda, Milan, Italy
| | - Primoz Gradisek
- Clinical Department of Anaesthesiology and Intensive Therapy, University Medical Center, Ljubljana, Slovenia
| | - Sandra Magnoni
- Department of Anesthesia and Intensive Care, Santa Chiara Hospital, Trento, Italy
| | - Mauro Oddo
- Neuroscience Critical Care Research Group, Department of Intensive Care Medicine, CHUV Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
- Directorate for Innovation and Clinical Research, CHUV Lausanne University Hospital, Lausanne, Switzerland
| | - Guido Bertolini
- Laboratory of Clinical Epidemiology, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Ranica, Italy
| | - Jonathan M Schott
- UK Dementia Research Institute, University College London, London, UK
- Dementia Research Centre and Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, London, UK
| | - Henrik Zetterberg
- UK Dementia Research Institute, University College London, London, UK
- Department of Psychiatry and Neurochemistry, The Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
| | - David Sharp
- Brain Sciences, Imperial College London, London, UK
- UK Dementia Research Institute Centre for Care Research and Technology, Imperial College London, London, UK
| |
Collapse
|
21
|
Formisano R, D’Ippolito M, Giustini M, Catani S, Mondello S, Piccolino I, Iannuzzi F, Wang KK, Hayes RL. The Prognostic Role of Candidate Serum Biomarkers in the Post-Acute and Chronic Phases of Disorder of Consciousness: A Preliminary Study. Brain Sci 2024; 14:239. [PMID: 38539627 PMCID: PMC10968965 DOI: 10.3390/brainsci14030239] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2024] [Revised: 02/22/2024] [Accepted: 02/24/2024] [Indexed: 11/11/2024] Open
Abstract
INTRODUCTION Serum biomarkers, such as Neurofilament Light (NF-L), Glial Fibrillary Acidic Protein (GFAP), Ubiquitin C-terminal Hydrolase (UCH-L1), and Total-tau (T-Tau) have been proposed for outcome prediction in the acute phase of severe traumatic brain injury, but they have been less investigated in patients with prolonged DoC (p-DoC). METHODS We enrolled 25 p-DoC patients according to the Coma Recovery Scale-Revised (CRS-R). We identified different time points: injury onset (t0), first blood sampling at admission in Neurorehabilitation (t1), and second blood sampling at discharge (t2). Patients were split into improved (improved level of consciousness from t1 to t2) and not-improved (unchanged or worsened level of consciousness from t1 to t2). RESULTS All biomarker levels decreased over time, even though each biomarker reveals typical features. Serum GFAP showed a weak correlation between t1 and t2 (p = 0.001), while no correlation was observed for serum NF-L (p = 0.955), UCH-L1 (p = 0.693), and T-Tau (p = 0.535) between t1 and t2. Improved patients showed a significant decrease in the level of NF-L (p = 0.0001), UCH-L1 (p = 0.001), and T-Tau (p = 0.002), but not for serum GFAP (p = 0.283). No significant statistical differences were observed in the not-improved group. CONCLUSIONS A significant correlation was found between the level of consciousness improvement and decreased NF-L, UCH-L1, and T-Tau levels. Future studies on the association of serum biomarkers with neurophysiological and neuroimaging prognostic indicators are recommended.
Collapse
Affiliation(s)
- Rita Formisano
- Neurorehabilitation 2, Post-Coma Unit, IRCCS Fondazione Santa Lucia, 00179 Rome, Italy;
| | | | - Marco Giustini
- Environmental and Social Epidemiology Unit, National Institute of Health, 00161 Rome, Italy;
| | - Sheila Catani
- Multiple Sclerosis Unit, IRCCS Fondazione Santa Lucia, 00179 Rome, Italy;
| | - Stefania Mondello
- Department of Biomedical and Dental Sciences and Morphofunctional Imaging, University of Messina, 98122 Messina, Italy;
| | - Iliana Piccolino
- Experimental Neuro-Psychobiology Laboratory, IRCCS Fondazione Santa Lucia, 00179 Rome, Italy; (I.P.); (F.I.)
| | - Filomena Iannuzzi
- Experimental Neuro-Psychobiology Laboratory, IRCCS Fondazione Santa Lucia, 00179 Rome, Italy; (I.P.); (F.I.)
| | - Kevin K. Wang
- Department of Neurobiology, Center for Neurotrauma, Multiomics & Biomarkers (CNMB), Neuroscience Institute, Morehouse School of Medicine, Atlanta, GE 30310, USA;
- Brain Rehabilitation Research Center (BRRC), Malcom Randall Veterans Affairs Medical Center, Gainesville, FL 32608, USA
- Center for Visual and Neurocognitive Rehabilitation, Atlanta VA Health Care System, Decatur, GA 30033, USA
| | | |
Collapse
|
22
|
Wu YC, Bogale TA, Koistinaho J, Pizzi M, Rolova T, Bellucci A. The contribution of β-amyloid, Tau and α-synuclein to blood-brain barrier damage in neurodegenerative disorders. Acta Neuropathol 2024; 147:39. [PMID: 38347288 PMCID: PMC10861401 DOI: 10.1007/s00401-024-02696-z] [Citation(s) in RCA: 20] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2023] [Revised: 01/23/2024] [Accepted: 01/23/2024] [Indexed: 02/15/2024]
Abstract
Central nervous system (CNS) accumulation of fibrillary deposits made of Amyloid β (Aβ), hyperphosphorylated Tau or α-synuclein (α-syn), present either alone or in the form of mixed pathology, characterizes the most common neurodegenerative diseases (NDDs) as well as the aging brain. Compelling evidence supports that acute neurological disorders, such as traumatic brain injury (TBI) and stroke, are also accompanied by increased deposition of toxic Aβ, Tau and α-syn species. While the contribution of these pathological proteins to neurodegeneration has been experimentally ascertained, the cellular and molecular mechanisms driving Aβ, Tau and α-syn-related brain damage remain to be fully clarified. In the last few years, studies have shown that Aβ, Tau and α-syn may contribute to neurodegeneration also by inducing and/or promoting blood-brain barrier (BBB) disruption. These pathological proteins can affect BBB integrity either directly by affecting key BBB components such as pericytes and endothelial cells (ECs) or indirectly, by promoting brain macrophages activation and dysfunction. Here, we summarize and critically discuss key findings showing how Aβ, Tau and α-syn can contribute to BBB damage in most common NDDs, TBI and stroke. We also highlight the need for a deeper characterization of the role of these pathological proteins in the activation and dysfunction of brain macrophages, pericytes and ECs to improve diagnosis and treatment of acute and chronic neurological disorders.
Collapse
Affiliation(s)
- Ying-Chieh Wu
- Neuroscience Center, HiLIFE, University of Helsinki, Helsinki, Finland
| | - Tizibt Ashine Bogale
- Department of Molecular and Translational Medicine, University of Brescia, Viale Europa 11, 25123, Brescia, BS, Italy
- Department of Acute Brain and Cardiovascular Injury, Istituto Di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy
| | - Jari Koistinaho
- Neuroscience Center, HiLIFE, University of Helsinki, Helsinki, Finland
| | - Marina Pizzi
- Department of Molecular and Translational Medicine, University of Brescia, Viale Europa 11, 25123, Brescia, BS, Italy
| | - Taisia Rolova
- Neuroscience Center, HiLIFE, University of Helsinki, Helsinki, Finland
| | - Arianna Bellucci
- Department of Molecular and Translational Medicine, University of Brescia, Viale Europa 11, 25123, Brescia, BS, Italy.
| |
Collapse
|
23
|
Pulliam A, Gier EC, Gaul DA, Moore SG, Fernández FM, LaPlaca MC. Comparing Brain and Blood Lipidome Changes following Single and Repetitive Mild Traumatic Brain Injury in Rats. ACS Chem Neurosci 2024; 15:300-314. [PMID: 38179922 PMCID: PMC10797623 DOI: 10.1021/acschemneuro.3c00603] [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: 09/16/2023] [Revised: 12/07/2023] [Accepted: 12/07/2023] [Indexed: 01/06/2024] Open
Abstract
Traumatic brain injury (TBI) is a major health concern in the United States and globally, contributing to disability and long-term neurological problems. Lipid dysregulation after TBI is underexplored, and a better understanding of lipid turnover and degradation could point to novel biomarker candidates and therapeutic targets. Here, we investigated overlapping lipidome changes in the brain and blood using a data-driven discovery approach to understand lipid alterations in the brain and serum compartments acutely following mild TBI (mTBI) and the potential efflux of brain lipids to peripheral blood. The cortices and sera from male and female Sprague-Dawley rats were analyzed via ultra-high performance liquid chromatography-mass spectrometry (UHPLC-MS) in both positive and negative ion modes following single and repetitive closed head impacts. The overlapping lipids in the data sets were identified with an in-house data dictionary for investigating lipid class changes. MS-based lipid profiling revealed overall increased changes in the serum compartment, while the brain lipids primarily showed decreased changes. Interestingly, there were prominent alterations in the sphingolipid class in the brain and blood compartments after single and repetitive injury, which may suggest efflux of brain sphingolipids into the blood after TBI. Genetic algorithms were used for predictive panel selection to classify injured and control samples with high sensitivity and specificity. These overlapping lipid panels primarily mapped to the glycerophospholipid metabolism pathway with Benjamini-Hochberg adjusted q-values less than 0.05. Collectively, these results detail overlapping lipidome changes following mTBI in the brain and blood compartments, increasing our understanding of TBI-related lipid dysregulation while identifying novel biomarker candidates.
Collapse
Affiliation(s)
- Alexis
N. Pulliam
- Coulter
Department of Biomedical Engineering, Georgia
Institute of Technology/Emory University, Atlanta, GA 30332 USA
- Petit
Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA 30332, USA
| | - Eric C. Gier
- School
of Chemistry and Biochemistry, Georgia Institute
of Technology, Atlanta, GA 30332 USA
- Petit
Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA 30332, USA
| | - David A. Gaul
- School
of Chemistry and Biochemistry, Georgia Institute
of Technology, Atlanta, GA 30332 USA
- Petit
Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA 30332, USA
| | - Samuel G. Moore
- School
of Chemistry and Biochemistry, Georgia Institute
of Technology, Atlanta, GA 30332 USA
- Petit
Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA 30332, USA
| | - Facundo M. Fernández
- School
of Chemistry and Biochemistry, Georgia Institute
of Technology, Atlanta, GA 30332 USA
- Petit
Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA 30332, USA
| | - Michelle C. LaPlaca
- Coulter
Department of Biomedical Engineering, Georgia
Institute of Technology/Emory University, Atlanta, GA 30332 USA
- Petit
Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA 30332, USA
| |
Collapse
|
24
|
Martin SP, Leeman-Markowski BA. Proposed mechanisms of tau: relationships to traumatic brain injury, Alzheimer's disease, and epilepsy. Front Neurol 2024; 14:1287545. [PMID: 38249745 PMCID: PMC10797726 DOI: 10.3389/fneur.2023.1287545] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2023] [Accepted: 11/30/2023] [Indexed: 01/23/2024] Open
Abstract
Traumatic brain injury (TBI), Alzheimer's disease (AD), and epilepsy share proposed mechanisms of injury, including neuronal excitotoxicity, cascade signaling, and activation of protein biomarkers such as tau. Although tau is typically present intracellularly, in tauopathies, phosphorylated (p-) and hyper-phosphorylated (hp-) tau are released extracellularly, the latter leading to decreased neuronal stability and neurofibrillary tangles (NFTs). Tau cleavage at particular sites increases susceptibility to hyper-phosphorylation, NFT formation, and eventual cell death. The relationship between tau and inflammation, however, is unknown. In this review, we present evidence for an imbalanced endoplasmic reticulum (ER) stress response and inflammatory signaling pathways resulting in atypical p-tau, hp-tau and NFT formation. Further, we propose tau as a biomarker for neuronal injury severity in TBI, AD, and epilepsy. We present a hypothesis of tau phosphorylation as an initial acute neuroprotective response to seizures/TBI. However, if the underlying seizure pathology or TBI recurrence is not effectively treated, and the pathway becomes chronically activated, we propose a "tipping point" hypothesis that identifies a transition of tau phosphorylation from neuroprotective to injurious. We outline the role of amyloid beta (Aβ) as a "last ditch effort" to revert the cell to programmed death signaling, that, when fails, transitions the mechanism from injurious to neurodegenerative. Lastly, we discuss targets along these pathways for therapeutic intervention in AD, TBI, and epilepsy.
Collapse
Affiliation(s)
- Samantha P. Martin
- Comprehensive Epilepsy Center, New York University Langone Health, New York, NY, United States
- Department of Neurology, New York University Langone Health, New York, NY, United States
- New York University Grossman School of Medicine, New York, NY, United States
- VA New York Harbor Healthcare System, New York, NY, United States
| | - Beth A. Leeman-Markowski
- Comprehensive Epilepsy Center, New York University Langone Health, New York, NY, United States
- Department of Neurology, New York University Langone Health, New York, NY, United States
- VA New York Harbor Healthcare System, New York, NY, United States
| |
Collapse
|
25
|
Zhang Y, Li Z, Wang H, Pei Z, Zhao S. Molecular biomarkers of diffuse axonal injury: recent advances and future perspectives. Expert Rev Mol Diagn 2024; 24:39-47. [PMID: 38183228 DOI: 10.1080/14737159.2024.2303319] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Accepted: 12/18/2023] [Indexed: 01/07/2024]
Abstract
INTRODUCTION Diffuse axonal injury (DAI), with high mortality and morbidity both in children and adults, is one of the most severe pathological consequences of traumatic brain injury. Currently, clinical diagnosis, disease assessment, disability identification, and postmortem diagnosis of DAI is mainly limited by the absent of specific molecular biomarkers. AREAS COVERED In this review, we first introduce the pathophysiology of DAI, summarized the reported biomarkers in previous animal and human studies, and then the molecular biomarkers such as β-Amyloid precursor protein, neurofilaments, S-100β, myelin basic protein, tau protein, neuron-specific enolase, Peripherin and Hemopexin for DAI diagnosis is summarized. Finally, we put forward valuable views on the future research direction of diagnostic biomarkers of DAI. EXPERT OPINION In recent years, the advanced technology has ultimately changed the research of DAI, and the numbers of potential molecular biomarkers was introduced in related studies. We summarized the latest updated information in such studies to provide references for future research and explore the potential pathophysiological mechanism on diffuse axonal injury.
Collapse
Affiliation(s)
- Youyou Zhang
- Department of Geriatrics Neurology, the Second Affiliated Hospital of Xi'an Jiao Tong University, Xi'an Jiaotong University, Xi'an, Shaanxi, China
- Linfen People's Hosiptal, the Seventh Clinical Medical College of Shanxi Medical University, Linfen, Shanxi, China
| | - Zhaoyang Li
- Department of Occupational and Environmental Health, School of Public Health, Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Hui Wang
- Department of Geriatrics Neurology, the Second Affiliated Hospital of Xi'an Jiao Tong University, Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Zhiyong Pei
- Linfen People's Hosiptal, the Seventh Clinical Medical College of Shanxi Medical University, Linfen, Shanxi, China
| | - Shuquan Zhao
- Department of Forensic Pathology, Zhongshan School of Medicine Sun Yat-sen University, Guangzhou, Guangdong, China
- Guangdong Province Translational Forensic Medicine Engineering Technology Research Center, Sun Yat-sen University, Guangzhou, Guangdong, China
| |
Collapse
|
26
|
Kobeissy F, Arja RD, Munoz JC, Shear DA, Gilsdorf J, Zhu J, Yadikar H, Haskins W, Tyndall JA, Wang KK. The game changer: UCH-L1 and GFAP-based blood test as the first marketed in vitro diagnostic test for mild traumatic brain injury. Expert Rev Mol Diagn 2024; 24:67-77. [PMID: 38275158 DOI: 10.1080/14737159.2024.2306876] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2023] [Accepted: 12/15/2023] [Indexed: 01/27/2024]
Abstract
INTRODUCTION Major organ-based in vitro diagnostic (IVD) tests like ALT/AST for the liver and cardiac troponins for the heart are established, but an approved IVD blood test for the brain has been missing, highlighting a gap in medical diagnostics. AREAS COVERED In response to this need, Abbott Diagnostics secured FDA clearance in 2021 for the i-STAT Alinity™, a point-of-care plasma blood test for mild traumatic brain injury (TBI). BioMerieux VIDAS, also approved in Europe, utilizes two brain-derived protein biomarkers: neuronal ubiquitin C-terminal hydrolase-L1 (UCH-L1) and glial fibrillary acidic protein (GFAP). These biomarkers, which are typically present in minimal amounts in healthy individuals, are instrumental in diagnosing mild TBI with potential brain lesions. The study explores how UCH-L1 and GFAP levels increase significantly in the bloodstream following traumatic brain injury, aiding in early and accurate diagnosis. EXPERT OPINION The introduction of the i-STAT Alinity™ and the Biomerieux VIDAS TBI blood tests mark a groundbreaking development in TBI diagnosis. It paves the way for the integration of TBI biomarker tools into clinical practice and therapeutic trials, enhancing the precision medicine approach by generating valuable data. This advancement is a critical step in addressing the long-standing gap in brain-related diagnostics and promises to revolutionize the management and treatment of mild TBI.
Collapse
Affiliation(s)
- Firas Kobeissy
- Program for Neurotrauma, Neuroproteomics & Biomarker Research, Neorobiology, Morehouse School of Medicine, Atlanta, GA, USA
| | - Rawad Daniel Arja
- Program for Neurotrauma, Neuroproteomics & Biomarker Research, Neorobiology, Morehouse School of Medicine, Atlanta, GA, USA
| | - Jennifer C Munoz
- Department of Pediatric Critical Care, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Deborah A Shear
- Brain Trauma Neuroprotection & Neurorestoration (BTNN) Branch, Center for Military Psychiatry and Neuroscience, Walter Reed Army Institute of Research, Silver Spring, MD, USA
| | - Janice Gilsdorf
- Brain Trauma Neuroprotection & Neurorestoration (BTNN) Branch, Center for Military Psychiatry and Neuroscience, Walter Reed Army Institute of Research, Silver Spring, MD, USA
| | - Jiepei Zhu
- Program for Neurotrauma, Neuroproteomics & Biomarker Research, Neorobiology, Morehouse School of Medicine, Atlanta, GA, USA
| | - Hamad Yadikar
- Program for Neurotrauma, Neuroproteomics & Biomarker Research, Neorobiology, Morehouse School of Medicine, Atlanta, GA, USA
- Department of Biological Sciences, Kuwait University, Safat, Kuwait
| | | | | | - Kevin K Wang
- Program for Neurotrauma, Neuroproteomics & Biomarker Research, Neorobiology, Morehouse School of Medicine, Atlanta, GA, USA
| |
Collapse
|
27
|
Mavroudis I, Jabeen S, Balmus IM, Ciobica A, Burlui V, Romila L, Iordache A. Exploring the Potential of Exosomal Biomarkers in Mild Traumatic Brain Injury and Post-Concussion Syndrome: A Systematic Review. J Pers Med 2023; 14:35. [PMID: 38248736 PMCID: PMC10817245 DOI: 10.3390/jpm14010035] [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: 11/20/2023] [Revised: 12/13/2023] [Accepted: 12/22/2023] [Indexed: 01/23/2024] Open
Abstract
BACKGROUND Alongside their long-term effects, post-concussion syndrome (PCS) and mild traumatic brain injuries (mTBI) are significant public health concerns. Currently, there is a lack of reliable biomarkers for diagnosing and monitoring mTBI and PCS. Exosomes are small extracellular vesicles secreted by cells that have recently emerged as a potential source of biomarkers for mTBI and PCS due to their ability to cross the blood-brain barrier and reflect the pathophysiology of brain injury. In this study, we aimed to investigate the role of salivary exosomal biomarkers in mTBI and PCS. METHODS A systematic review using the PRISMA guidelines was conducted, and studies were selected based on their relevance to the topic. RESULTS The analyzed studies have shown that exosomal tau, phosphorylated tau (p-tau), amyloid beta (Aβ), and microRNAs (miRNAs) are potential biomarkers for mTBI and PCS. Specifically, elevated levels of exosomal tau and p-tau have been associated with mTBI and PCS as well as repetitive mTBI. Dysregulated exosomal miRNAs have also been observed in individuals with mTBI and PCS. Additionally, exosomal Prion cellular protein (PRPc), coagulation factor XIII (XIIIa), synaptogyrin-3, IL-6, and aquaporins have been identified as promising biomarkers for mTBI and PCS. CONCLUSION Salivary exosomal biomarkers have the potential to serve as non-invasive and easily accessible diagnostic and prognostic tools for mTBI and PCS. Further studies are needed to validate these biomarkers and develop standardized protocols for their use in clinical settings. Salivary exosomal biomarkers can improve the diagnosis, monitoring, and treatment of mTBI and PCS, leading to improved patient outcomes.
Collapse
Affiliation(s)
- Ioannis Mavroudis
- Department of Neuroscience, Leeds Teaching Hospitals, NHS Trust, Leeds LS2 9JT, UK;
| | - Sidra Jabeen
- Liaquat National Hospital and Medical College, Karachi 74800, Pakistan
| | - Ioana Miruna Balmus
- Department of Exact Sciences and Natural Sciences, Institute of Interdisciplinary Research, “Alexandru Ioan Cuza” University of Iasi, 26th Alexandru Lapusneanu Street, 700057 Iasi, Romania
| | - Alin Ciobica
- Department of Biology, Faculty of Biology, Alexandru Ioan Cuza University of Iasi, 20th Carol I Avenue, 700506 Iași, Romania
| | - Vasile Burlui
- Preclinical Department, Apollonia University, Păcurari Street 11, 700511 Iasi, Romania
| | - Laura Romila
- Preclinical Department, Apollonia University, Păcurari Street 11, 700511 Iasi, Romania
| | - Alin Iordache
- Faculty of Medicine, University of Medicine and Pharmacy “Grigore T. Popa”, 700115 Iasi, Romania;
| |
Collapse
|
28
|
Hossain I, Marklund N, Czeiter E, Hutchinson P, Buki A. Blood biomarkers for traumatic brain injury: A narrative review of current evidence. BRAIN & SPINE 2023; 4:102735. [PMID: 38510630 PMCID: PMC10951700 DOI: 10.1016/j.bas.2023.102735] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/14/2023] [Revised: 12/05/2023] [Accepted: 12/11/2023] [Indexed: 03/22/2024]
Abstract
Introduction A blood-based biomarker (BBBM) test could help to better stratify patients with traumatic brain injury (TBI), reduce unnecessary imaging, to detect and treat secondary insults, predict outcomes, and monitor treatment effects and quality of care. Research question What evidence is available for clinical applications of BBBMs in TBI and how to advance this field? Material and methods This narrative review discusses the potential clinical applications of core BBBMs in TBI. A literature search in PubMed, Scopus, and ISI Web of Knowledge focused on articles in English with the words "traumatic brain injury" together with the words "blood biomarkers", "diagnostics", "outcome prediction", "extracranial injury" and "assay method" alone-, or in combination. Results Glial fibrillary acidic protein (GFAP) combined with Ubiquitin C-terminal hydrolase-L1(UCH-L1) has received FDA clearance to aid computed tomography (CT)-detection of brain lesions in mild (m) TBI. Application of S100B led to reduction of head CT scans. GFAP may also predict magnetic resonance imaging (MRI) abnormalities in CT-negative cases of TBI. Further, UCH-L1, S100B, Neurofilament light (NF-L), and total tau showed value for predicting mortality or unfavourable outcome. Nevertheless, biomarkers have less role in outcome prediction in mTBI. S100B could serve as a tool in the multimodality monitoring of patients in the neurointensive care unit. Discussion and conclusion Largescale systematic studies are required to explore the kinetics of BBBMs and their use in multiple clinical groups. Assay development/cross validation should advance the generalizability of those results which implicated GFAP, S100B and NF-L as most promising biomarkers in the diagnostics of TBI.
Collapse
Affiliation(s)
- Iftakher Hossain
- Neurocenter, Department of Neurosurgery, Turku University Hospital, Turku, Finland
- Department of Clinical Neurosciences, Neurosurgery Unit, University of Cambridge, Addenbrooke’s Hospital, Cambridge, United Kingdom
- Department of Neuroscience, Karolinska Institute, Stockholm, Sweden
| | - Niklas Marklund
- Department of Clinical Sciences Lund, Neurosurgery, Lund University, Department of Neurosurgery, Skåne University Hospital, Lund, Sweden
| | - Endre Czeiter
- Department of Neurosurgery, Medical School, Neurotrauma Research Group, Szentagothai Research Centre, And HUN-REN-PTE Clinical Neuroscience MR Research Group, University of Pecs, Pecs, Hungary
| | - Peter Hutchinson
- Department of Clinical Neurosciences, Neurosurgery Unit, University of Cambridge, Addenbrooke’s Hospital, Cambridge, United Kingdom
| | - Andras Buki
- Department of Neurosurgery, University of Örebro, Örebro, Sweden
| |
Collapse
|
29
|
Lantero-Rodriguez J, Tissot C, Snellman A, Servaes S, Benedet AL, Rahmouni N, Montoliu-Gaya L, Therriault J, Brum WS, Stevenson J, Lussier FZ, Bezgin G, Macedo AC, Chamoun M, Mathotaarachi SS, Pascoal TA, Ashton NJ, Zetterberg H, Neto PR, Blennow K. Plasma and CSF concentrations of N-terminal tau fragments associate with in vivo neurofibrillary tangle burden. Alzheimers Dement 2023; 19:5343-5354. [PMID: 37190913 DOI: 10.1002/alz.13119] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2023] [Revised: 03/16/2023] [Accepted: 04/11/2023] [Indexed: 05/17/2023]
Abstract
INTRODUCTION Fluid biomarkers capable of specifically tracking tau tangle pathology in vivo are greatly needed. METHODS We measured cerebrospinal fluid (CSF) and plasma concentrations of N-terminal tau fragments (NTA-tau), using a novel immunoassay (NTA) in the TRIAD cohort, consisting of 272 individuals assessed with amyloid beta (Aβ) positron emission tomography (PET), tau PET, magnetic resonance imaging (MRI) and cognitive assessments. RESULTS CSF and plasma NTA-tau concentrations were specifically increased in cognitively impaired Aβ-positive groups. CSF and plasma NTA-tau concentrations displayed stronger correlations with tau PET than with Aβ PET and MRI, both in global uptake and at the voxel level. Regression models demonstrated that both CSF and plasma NTA-tau are preferentially associated with tau pathology. Moreover, plasma NTA-tau was associated with longitudinal tau PET accumulation across the aging and Alzheimer's disease (AD) spectrum. DISCUSSION NTA-tau is a biomarker closely associated with in vivo tau deposition in the AD continuum and has potential as a tau tangle biomarker in clinical settings and trials. HIGHLIGHTS An assay for detecting N-terminal tau fragments (NTA-tau) in plasma and CSF was evaluated. NTA-tau is more closely associated with tau PET than amyloid PET or neurodegeneration. NTA-tau can successfully track in vivo tau deposition across the AD continuum. Plasma NTA-tau increased over time only in cognitively impaired amyloid-β positive individuals.
Collapse
Affiliation(s)
- Juan Lantero-Rodriguez
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, the Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
| | - Cécile Tissot
- Translational Neuroimaging Laboratory, McGill Research Centre for Studies in Aging, Douglas Mental Health Institute, Le Centre intégré universitaire de santé et de services sociaux (CIUSSS) de l'Ouest de l'Île de Montréal, Montreal, Canada
- Department of Neurology and Neurosurgery, Faculty of Medicine, McGill University, Montreal, Canada
| | - Anniina Snellman
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, the Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
- Turku PET Centre, University of Turku, Turku University Hospital, Turku, Finland
| | - Stijn Servaes
- Translational Neuroimaging Laboratory, McGill Research Centre for Studies in Aging, Douglas Mental Health Institute, Le Centre intégré universitaire de santé et de services sociaux (CIUSSS) de l'Ouest de l'Île de Montréal, Montreal, Canada
- Department of Neurology and Neurosurgery, Faculty of Medicine, McGill University, Montreal, Canada
| | - Andrea L Benedet
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, the Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
| | - Nesrine Rahmouni
- Translational Neuroimaging Laboratory, McGill Research Centre for Studies in Aging, Douglas Mental Health Institute, Le Centre intégré universitaire de santé et de services sociaux (CIUSSS) de l'Ouest de l'Île de Montréal, Montreal, Canada
- Department of Neurology and Neurosurgery, Faculty of Medicine, McGill University, Montreal, Canada
| | - Laia Montoliu-Gaya
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, the Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
| | - Joseph Therriault
- Translational Neuroimaging Laboratory, McGill Research Centre for Studies in Aging, Douglas Mental Health Institute, Le Centre intégré universitaire de santé et de services sociaux (CIUSSS) de l'Ouest de l'Île de Montréal, Montreal, Canada
- Department of Neurology and Neurosurgery, Faculty of Medicine, McGill University, Montreal, Canada
| | - Wagner S Brum
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, the Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
- Graduate Program in Biological Sciences: Biochemistry, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil
| | - Jenna Stevenson
- Translational Neuroimaging Laboratory, McGill Research Centre for Studies in Aging, Douglas Mental Health Institute, Le Centre intégré universitaire de santé et de services sociaux (CIUSSS) de l'Ouest de l'Île de Montréal, Montreal, Canada
- Department of Neurology and Neurosurgery, Faculty of Medicine, McGill University, Montreal, Canada
| | - Firoza Z Lussier
- Translational Neuroimaging Laboratory, McGill Research Centre for Studies in Aging, Douglas Mental Health Institute, Le Centre intégré universitaire de santé et de services sociaux (CIUSSS) de l'Ouest de l'Île de Montréal, Montreal, Canada
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Gleb Bezgin
- Translational Neuroimaging Laboratory, McGill Research Centre for Studies in Aging, Douglas Mental Health Institute, Le Centre intégré universitaire de santé et de services sociaux (CIUSSS) de l'Ouest de l'Île de Montréal, Montreal, Canada
- Department of Neurology and Neurosurgery, Faculty of Medicine, McGill University, Montreal, Canada
| | - Arthur C Macedo
- Translational Neuroimaging Laboratory, McGill Research Centre for Studies in Aging, Douglas Mental Health Institute, Le Centre intégré universitaire de santé et de services sociaux (CIUSSS) de l'Ouest de l'Île de Montréal, Montreal, Canada
- Department of Neurology and Neurosurgery, Faculty of Medicine, McGill University, Montreal, Canada
| | - Mira Chamoun
- Translational Neuroimaging Laboratory, McGill Research Centre for Studies in Aging, Douglas Mental Health Institute, Le Centre intégré universitaire de santé et de services sociaux (CIUSSS) de l'Ouest de l'Île de Montréal, Montreal, Canada
- Department of Neurology and Neurosurgery, Faculty of Medicine, McGill University, Montreal, Canada
| | - Sulantha S Mathotaarachi
- Translational Neuroimaging Laboratory, McGill Research Centre for Studies in Aging, Douglas Mental Health Institute, Le Centre intégré universitaire de santé et de services sociaux (CIUSSS) de l'Ouest de l'Île de Montréal, Montreal, Canada
- Department of Neurology and Neurosurgery, Faculty of Medicine, McGill University, Montreal, Canada
| | - Tharick A Pascoal
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Nicholas J Ashton
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, the Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
- Wallenberg Centre for Molecular and Translational Medicine, University of Gothenburg, Gothenburg, Sweden
- Department of Old Age Psychiatry, Maurice Wohl Clinical Neuroscience Institute, King's College London, London, UK
- NIHR Biomedical Research Centre for Mental Health & Biomedical Research Unit for Dementia at South London & Maudsley NHS Foundation, London, UK
| | - Henrik Zetterberg
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, the Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
- Department of Neurodegenerative Disease, Queen Square Institute of Neurology, University College London, London, UK
- UK Dementia Research Institute, University College London, London, UK
- Hong Kong Center for Neurodegenerative Diseases, Hong Kong, China
- Wisconsin Alzheimer's Disease Research Center, University of Wisconsin School of Medicine and Public Health, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Pedro Rosa Neto
- Translational Neuroimaging Laboratory, McGill Research Centre for Studies in Aging, Douglas Mental Health Institute, Le Centre intégré universitaire de santé et de services sociaux (CIUSSS) de l'Ouest de l'Île de Montréal, Montreal, Canada
- Department of Neurology and Neurosurgery, Faculty of Medicine, McGill University, Montreal, Canada
| | - Kaj Blennow
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, the Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
| |
Collapse
|
30
|
Tong S, Xie L, Xie X, Xu J, You Y, Sun Y, Zhou S, Ma C, Jiang G, Ma F, Wang Z, Gao X, Chen J. Nano-Plumber Reshapes Glymphatic-Lymphatic System to Sustain Microenvironment Homeostasis and Improve Long-Term Prognosis after Traumatic Brain Injury. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2304284. [PMID: 37867233 PMCID: PMC10700187 DOI: 10.1002/advs.202304284] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/27/2023] [Revised: 08/24/2023] [Indexed: 10/24/2023]
Abstract
Traumatic brain injury (TBI) is a leading cause of death and disability worldwide. Long-term changes in the microenvironment of the brain contribute to the degeneration of neurological function following TBI. However, current research focuses primarily on short-term modulation during the early phases of TBI, not on the critical significance of long-term homeostasis in the brain microenvironment. Notably, dysfunction of the glymphatic-lymphatic system results in the accumulation of danger/damage-associated molecular patterns (DAMPs) in the brain, which is regarded as the leading cause of long-term microenvironmental disturbances following TBI. Here, a nanostructure, Nano-plumber, that co-encapsulates the microenvironment regulator pro-DHA and the lymphatic-specific growth factor VEGF-C is developed, allowing for a sustainable and orderly regulation of the microenvironment to promote long-term neurological recovery. Nano-plumber reverses the injury microenvironment by suppressing microglia and astrocytes activation and maintaining reduced activation via enhanced glymphatic-lymphatic drainage, and significantly improves the neurological function of rodents with TBI. This study demonstrates that glymphatic-lymphatic system reconstruction is essential for enhancing long-term prognosis following TBI, and that the Nano-plumber developed here may serve as a clinically translatable treatment option for TBI.
Collapse
Affiliation(s)
- Shiqiang Tong
- Department of PharmaceuticsSchool of Pharmacy & Shanghai Pudong HospitalFudan UniversityShanghai201203China
- Key Laboratory of Smart Drug DeliveryMinistry of EducationSchool of PharmacyFudan UniversityShanghai201203China
| | - Laozhi Xie
- Department of PharmaceuticsSchool of Pharmacy & Shanghai Pudong HospitalFudan UniversityShanghai201203China
- Key Laboratory of Smart Drug DeliveryMinistry of EducationSchool of PharmacyFudan UniversityShanghai201203China
| | - Xiaoying Xie
- Department of PharmaceuticsSchool of Pharmacy & Shanghai Pudong HospitalFudan UniversityShanghai201203China
- Key Laboratory of Smart Drug DeliveryMinistry of EducationSchool of PharmacyFudan UniversityShanghai201203China
| | - Jianpei Xu
- Department of PharmaceuticsSchool of Pharmacy & Shanghai Pudong HospitalFudan UniversityShanghai201203China
- Key Laboratory of Smart Drug DeliveryMinistry of EducationSchool of PharmacyFudan UniversityShanghai201203China
| | - Yang You
- Department of PharmaceuticsSchool of Pharmacy & Shanghai Pudong HospitalFudan UniversityShanghai201203China
- Key Laboratory of Smart Drug DeliveryMinistry of EducationSchool of PharmacyFudan UniversityShanghai201203China
| | - Yinzhe Sun
- Department of PharmaceuticsSchool of Pharmacy & Shanghai Pudong HospitalFudan UniversityShanghai201203China
- Key Laboratory of Smart Drug DeliveryMinistry of EducationSchool of PharmacyFudan UniversityShanghai201203China
| | - Songlei Zhou
- Department of PharmaceuticsSchool of Pharmacy & Shanghai Pudong HospitalFudan UniversityShanghai201203China
- Key Laboratory of Smart Drug DeliveryMinistry of EducationSchool of PharmacyFudan UniversityShanghai201203China
| | - Chuchu Ma
- Department of PharmaceuticsSchool of Pharmacy & Shanghai Pudong HospitalFudan UniversityShanghai201203China
- Key Laboratory of Smart Drug DeliveryMinistry of EducationSchool of PharmacyFudan UniversityShanghai201203China
| | - Gan Jiang
- Department of Pharmacology and Chemical BiologyState Key Laboratory of Oncogenes and Related GenesShanghai Universities Collaborative Innovation Center for Translational MedicineShanghai Jiao Tong University School of MedicineShanghai200025China
| | - Fenfen Ma
- Department of PharmaceuticsSchool of Pharmacy & Shanghai Pudong HospitalFudan UniversityShanghai201203China
- Key Laboratory of Smart Drug DeliveryMinistry of EducationSchool of PharmacyFudan UniversityShanghai201203China
- Department of PharmacyShanghai Pudong HospitalFudan UniversityShanghai201399China
| | - Zhihua Wang
- Department of EmergencyShanghai Pudong HospitalFudan University Pudong Medical CenterShanghai201399China
| | - Xiaoling Gao
- Department of Pharmacology and Chemical BiologyState Key Laboratory of Oncogenes and Related GenesShanghai Universities Collaborative Innovation Center for Translational MedicineShanghai Jiao Tong University School of MedicineShanghai200025China
| | - Jun Chen
- Department of PharmaceuticsSchool of Pharmacy & Shanghai Pudong HospitalFudan UniversityShanghai201203China
- Key Laboratory of Smart Drug DeliveryMinistry of EducationSchool of PharmacyFudan UniversityShanghai201203China
| |
Collapse
|
31
|
Kobeissy F, Goli M, Yadikar H, Shakkour Z, Kurup M, Haidar MA, Alroumi S, Mondello S, Wang KK, Mechref Y. Advances in neuroproteomics for neurotrauma: unraveling insights for personalized medicine and future prospects. Front Neurol 2023; 14:1288740. [PMID: 38073638 PMCID: PMC10703396 DOI: 10.3389/fneur.2023.1288740] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2023] [Accepted: 11/01/2023] [Indexed: 02/12/2024] Open
Abstract
Neuroproteomics, an emerging field at the intersection of neuroscience and proteomics, has garnered significant attention in the context of neurotrauma research. Neuroproteomics involves the quantitative and qualitative analysis of nervous system components, essential for understanding the dynamic events involved in the vast areas of neuroscience, including, but not limited to, neuropsychiatric disorders, neurodegenerative disorders, mental illness, traumatic brain injury, chronic traumatic encephalopathy, and other neurodegenerative diseases. With advancements in mass spectrometry coupled with bioinformatics and systems biology, neuroproteomics has led to the development of innovative techniques such as microproteomics, single-cell proteomics, and imaging mass spectrometry, which have significantly impacted neuronal biomarker research. By analyzing the complex protein interactions and alterations that occur in the injured brain, neuroproteomics provides valuable insights into the pathophysiological mechanisms underlying neurotrauma. This review explores how such insights can be harnessed to advance personalized medicine (PM) approaches, tailoring treatments based on individual patient profiles. Additionally, we highlight the potential future prospects of neuroproteomics, such as identifying novel biomarkers and developing targeted therapies by employing artificial intelligence (AI) and machine learning (ML). By shedding light on neurotrauma's current state and future directions, this review aims to stimulate further research and collaboration in this promising and transformative field.
Collapse
Affiliation(s)
- Firas Kobeissy
- Department of Neurobiology, School of Medicine, Neuroscience Institute, Atlanta, GA, United States
| | - Mona Goli
- Department of Chemistry and Biochemistry, Texas Tech University, Lubbock, TX, United States
| | - Hamad Yadikar
- Department of Biological Sciences Faculty of Science, Kuwait University, Safat, Kuwait
| | - Zaynab Shakkour
- Department of Pathology and Anatomical Sciences, University of Missouri School of Medicine, Columbia, MO, United States
| | - Milin Kurup
- Alabama College of Osteopathic Medicine, Dothan, AL, United States
| | | | - Shahad Alroumi
- Department of Biological Sciences Faculty of Science, Kuwait University, Safat, Kuwait
| | - Stefania Mondello
- Department of Biomedical and Dental Sciences and Morphofunctional Imaging, University of Messina, Messina, Italy
| | - Kevin K. Wang
- Department of Neurobiology, School of Medicine, Neuroscience Institute, Atlanta, GA, United States
| | - Yehia Mechref
- Department of Chemistry and Biochemistry, Texas Tech University, Lubbock, TX, United States
| |
Collapse
|
32
|
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: 1] [Impact Index Per Article: 0.5] [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
|
33
|
Bernick C, Shan G, Ritter A, Ashton NJ, Blennow K, Lantero-Rodriguez J, Snellman A, Zetterberg H. Blood biomarkers and neurodegeneration in individuals exposed to repetitive head impacts. Alzheimers Res Ther 2023; 15:173. [PMID: 37828595 PMCID: PMC10571311 DOI: 10.1186/s13195-023-01310-w] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2023] [Accepted: 09/18/2023] [Indexed: 10/14/2023]
Abstract
BACKGROUND It is unknown if fluid biomarkers reflective of brain pathologies are useful in detecting and following a neurodegenerative process in individuals exposed to repetitive head impacts. This study explores the relationship between blood biomarkers and longitudinal change in cognitive function and regional brain volumes in a cohort of professional fighters. METHODS Participants are drawn from a convenience sample of active and retired professional boxers and Mixed Martial Arts fighters and a control group with no prior exposure to head impacts. 3 T MRI brain imaging, plasma samples, and computerized cognitive testing were obtained at baseline and, for a subset, annually. MRI regional volumes were extracted, along with plasma levels of neurofilament light chain (NfL), glial fibrillary acidic protein (GFAP), p-tau231, and N-terminal tau (NTA). Statistical analyses were performed to assess the relationship between plasma levels and regional brain volumes and cognitive performance at baseline and longitudinally. RESULTS One hundred forty active boxers (mean age: 31 with standard deviation (SD) of 8), 211 active MMA (mean age of 30 with SD of 5), 69 retired boxers (mean age 49 with SD of 9), and 52 control participants (mean age 36 with SD of 12) were included in the analyses. Baseline GFAP levels were highest in the retired boxers (retired boxers v. active MMA: p = 0.0191), whereas active boxers had higher levels of NfL (active boxers v. MMA: p = 0.047). GFAP showed an increase longitudinally in retired boxers that was associated with decreasing volumes of multiple cortical and subcortical structures (e.g., hippocampus: B = - 1.25, 95% CI, - 1.65 to - 0.85) and increase in lateral ventricle size (B = 1.75, 95% CI, 1.46 to 2.04). Furthermore, performance on cognitive domains including memory, processing speed, psychomotor speed, and reaction time declined over time with increasing GFAP (e.g., processing speed: B = - 0.04, 95% CI, - 0.07 to - 0.02; reaction time: B = 0.52, 95% CI, 0.28 to 0.76). Among active fighters, increasing levels of GFAP were correlated with lower thalamic (B = - 1.42, 95% CI, - 2.34 to -0.49) and corpus callosum volumes, along with worsening scores on psychomotor speed (B = 0.14, 95% CI, 0.01 to 0.27). CONCLUSION Longitudinal plasma GFAP levels may have a role in identifying individuals exposed to repetitive head impacts who are at risk of showing progressive regional atrophy and cognitive decline.
Collapse
Affiliation(s)
- Charles Bernick
- Neurological Institute, Cleveland Clinic, Las Vegas, NV, USA.
| | - Guogen Shan
- Department of Biostatistics, University of Florida, Gainesville, FL, USA
| | - Aaron Ritter
- Neurological Institute, Cleveland Clinic, Las Vegas, NV, USA
| | - Nicholas J Ashton
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, the Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
| | - Kaj Blennow
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, the Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
| | - Juan Lantero-Rodriguez
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, the Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
| | - Anniina Snellman
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, the Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
| | - Henrik Zetterberg
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, the Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
- Department of Neurodegenerative Disease, UCL Institute of Neurology, Queen Square, London, UK
- UK Dementia Research Institute at UCL, London, UK
- Hong Kong Center for Neurodegenerative Diseases, Clear Water Bay, Hong Kong, China
- Wisconsin Alzheimer's Disease Research Center, School of Medicine and Public Health, University of Wisconsin, University of Wisconsin-Madison, Madison, WI, USA
| |
Collapse
|
34
|
Tabor JB, Galarneau JM, Penner LC, Cooper J, Ghodsi M, Fraser DD, Wellington CL, Debert CT, Emery CA. Use of Biostatistical Models to Manage Replicate Error in Concussion Biomarker Research. JAMA Netw Open 2023; 6:e2339733. [PMID: 37870831 PMCID: PMC10594140 DOI: 10.1001/jamanetworkopen.2023.39733] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/16/2023] [Accepted: 09/13/2023] [Indexed: 10/24/2023] Open
Abstract
Importance Advancing research on fluid biomarkers associated with sport-related concussion (SRC) highlights the importance of detecting low concentrations using ultrasensitive platforms. However, common statistical practices may overlook replicate errors and specimen exclusion, emphasizing the need to explore robust modeling approaches that consider all available replicate data for comprehensive understanding of sample variation and statistical inferences. Objective To evaluate the impact of replicate error and different biostatistical modeling approaches on SRC biomarker interpretation. Design, Setting, and Participants This cross-sectional study within the Surveillance in High Schools to Reduce the Risk of Concussions and Their Consequences study used data from healthy youth athletes (ages 11-18 years) collected from 3 sites across Canada between September 2019 and November 2021. Data were analyzed from November 2022 to February 2023. Exposures Demographic variables included age, sex, and self-reported history of previous concussion. Main Outcomes and Measures Outcomes of interest were preinjury plasma glial fibrillary acidic protein (GFAP), ubiquitin C-terminal hydrolase-L1 (UCH-L1), neurofilament-light (NFL), total tau (t-tau) and phosphorylated-tau-181 (p-tau-181) assayed in duplicate. Bland-Altman analysis determined the 95% limits of agreement (LOAs) for each biomarker. The impact of replicate error was explored using 3 biostatistical modeling approaches assessing the associations of age, sex, and previous concussion on biomarker concentrations: multilevel regression using all available replicate data, single-level regression using the means of replicate data, and single-level regression with replicate means, excluding specimens demonstrating more than 20% coefficient variation (CV). Results The sample included 149 healthy youth athletes (78 [52%] male; mean [SD] age, 15.74 [1.41] years; 51 participants [34%] reporting ≥1 previous concussions). Wide 95% LOAs were observed for GFAP (-17.74 to 18.20 pg/mL), UCH-L1 (-13.80 to 14.77 pg/mL), and t-tau (65.27% to 150.03%). GFAP and UCH-L1 were significantly associated with sex in multilevel regression (GFAP: effect size, 15.65%; β = -0.17; 95% CI, -0.30 to -0.04]; P = .02; UCH-L1: effect size, 17.24%; β = -0.19; 95% CI, -0.36 to -0.02]; P = .03) and single-level regression using the means of replicate data (GFAP: effect size, 15.56%; β = -0.17; 95% CI, -0.30 to -0.03]; P = .02; UCH-L1: effect size, 18.02%; β = -0.20; 95% CI, -0.37 to -0.03]; P = .02); however, there was no association for UCH-L1 after excluding specimens demonstrating more than 20% CV. Excluding specimens demonstrating more than 20% CV resulted in decreased differences associated with sex in GFAP (effect size, 12.29%; β = -0.14; 95% CI, -0.273 to -0.004]; P = .04) and increased sex differences in UCH-L1 (effect size, 23.59%; β = -0.27; 95% CI, -0.55 to 0.01]; P = .06), with the widest 95% CIs (ie, least precision) found in UCH-L1. Conclusions and Relevance In this cross-sectional study of healthy youth athletes, varying levels of agreement between SRC biomarker technical replicates suggested that means of measurements may not optimize precision for population values. Multilevel regression modeling demonstrated how incorporating all available biomarker data could capture replicate variation, avoiding challenges associated with means and percentage of CV exclusion thresholds to produce more representative estimates of association.
Collapse
Affiliation(s)
- Jason B. Tabor
- Sport Injury Prevention Research Centre, Faculty of Kinesiology, University of Calgary, Calgary, Alberta, Canada
- Hotchkiss Brain Institute, University of Calgary, Calgary, Alberta, Canada
- Alberta Children’s Hospital Research Institute, University of Calgary, Calgary, Alberta, Canada
| | - Jean-Michel Galarneau
- Sport Injury Prevention Research Centre, Faculty of Kinesiology, University of Calgary, Calgary, Alberta, Canada
| | - Linden C. Penner
- Sport Injury Prevention Research Centre, Faculty of Kinesiology, University of Calgary, Calgary, Alberta, Canada
- Hotchkiss Brain Institute, University of Calgary, Calgary, Alberta, Canada
- Alberta Children’s Hospital Research Institute, University of Calgary, Calgary, Alberta, Canada
| | - Jennifer Cooper
- Department of Pathology and Laboratory Medicine, Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, British Columbia, Canada
| | - Mohammad Ghodsi
- Department of Pathology and Laboratory Medicine, Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, British Columbia, Canada
| | - Douglas D. Fraser
- Department of Pediatrics and Clinical Neurological Sciences, Western University, London, Ontario, Canada
| | - Cheryl L. Wellington
- Department of Pathology and Laboratory Medicine, Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, British Columbia, Canada
| | - Chantel T. Debert
- Sport Injury Prevention Research Centre, Faculty of Kinesiology, University of Calgary, Calgary, Alberta, Canada
- Hotchkiss Brain Institute, University of Calgary, Calgary, Alberta, Canada
- Alberta Children’s Hospital Research Institute, University of Calgary, Calgary, Alberta, Canada
- Department of Clinical Neurosciences, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Carolyn A. Emery
- Sport Injury Prevention Research Centre, Faculty of Kinesiology, University of Calgary, Calgary, Alberta, Canada
- Hotchkiss Brain Institute, University of Calgary, Calgary, Alberta, Canada
- Alberta Children’s Hospital Research Institute, University of Calgary, Calgary, Alberta, Canada
- Departments of Pediatrics and Community Health Sciences, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| |
Collapse
|
35
|
Lepinay E, Cicchetti F. Tau: a biomarker of Huntington's disease. Mol Psychiatry 2023; 28:4070-4083. [PMID: 37749233 DOI: 10.1038/s41380-023-02230-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Revised: 07/31/2023] [Accepted: 08/11/2023] [Indexed: 09/27/2023]
Abstract
Developing effective treatments for patients with Huntington's disease (HD)-a neurodegenerative disorder characterized by severe cognitive, motor and psychiatric impairments-is proving extremely challenging. While the monogenic nature of this condition enables to identify individuals at risk, robust biomarkers would still be extremely valuable to help diagnose disease onset and progression, and especially to confirm treatment efficacy. If measurements of cerebrospinal fluid neurofilament levels, for example, have demonstrated use in recent clinical trials, other proteins may prove equal, if not greater, relevance as biomarkers. In fact, proteins such as tau could specifically be used to detect/predict cognitive affectations. We have herein reviewed the literature pertaining to the association between tau levels and cognitive states, zooming in on Alzheimer's disease, Parkinson's disease and traumatic brain injury in which imaging, cerebrospinal fluid, and blood samples have been interrogated or used to unveil a strong association between tau and cognition. Collectively, these areas of research have accrued compelling evidence to suggest tau-related measurements as both diagnostic and prognostic tools for clinical practice. The abundance of information retrieved in this niche of study has laid the groundwork for further understanding whether tau-related biomarkers may be applied to HD and guide future investigations to better understand and treat this disease.
Collapse
Affiliation(s)
- Eva Lepinay
- Centre de Recherche du CHU de Québec, Axe Neurosciences, Québec, QC, Canada
- Département de Psychiatrie & Neurosciences, Université Laval, Québec, QC, Canada
| | - Francesca Cicchetti
- Centre de Recherche du CHU de Québec, Axe Neurosciences, Québec, QC, Canada.
- Département de Psychiatrie & Neurosciences, Université Laval, Québec, QC, Canada.
| |
Collapse
|
36
|
Jash S, Banerjee S, Cheng S, Wang B, Qiu C, Kondo A, Ernerudh J, Zhou XZ, Lu KP, Sharma S. Cis P-tau is a central circulating and placental etiologic driver and therapeutic target of preeclampsia. Nat Commun 2023; 14:5414. [PMID: 37669931 PMCID: PMC10480164 DOI: 10.1038/s41467-023-41144-6] [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: 07/25/2022] [Accepted: 08/24/2023] [Indexed: 09/07/2023] Open
Abstract
Preeclampsia (PE) is the leading cause of maternal and fetal mortality globally and may trigger dementia later in life in mothers and their offspring. However, the etiological drivers remain elusive. Cis P-tau is an early etiological driver and blood biomarker in pre-clinical Alzheimer's and after vascular or traumatic brain injury, which can be targeted by stereo-specific antibody, with clinical trials ongoing. Here we find significant cis P-tau in the placenta and serum of PE patients, and in primary human trophoblasts exposed to hypoxia or sera from PE patients due to Pin1 inactivation. Depletion of cis P-tau from PE patient sera by the antibody prevents their ability to disrupt trophoblast invasion and endovascular activity and to cause the PE-like pathological and clinical features in pregnant humanized tau mice. Our studies uncover that cis P-tau is a central circulating etiological driver and its stereo-specific antibody is valuable for early PE diagnosis and treatment.
Collapse
Affiliation(s)
- Sukanta Jash
- Departments of Pediatrics, Women and Infants Hospital, Warren Alpert Medical School, Brown University, Providence, RI, 02905, USA
| | - Sayani Banerjee
- Departments of Pediatrics, Women and Infants Hospital, Warren Alpert Medical School, Brown University, Providence, RI, 02905, USA
| | - Shibin Cheng
- Departments of Pediatrics, Women and Infants Hospital, Warren Alpert Medical School, Brown University, Providence, RI, 02905, USA
| | - Bin Wang
- Division of Translational Therapeutics, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, 02215, USA
| | - Chenxi Qiu
- Division of Translational Therapeutics, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, 02215, USA
| | - Asami Kondo
- Division of Translational Therapeutics, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, 02215, USA
| | - Jan Ernerudh
- Department of Biomedical and Clinical Sciences, Linköping University, SE 58183, Linköping, Sweden
- Department of Clinical Immunology and Transfusion Medicine, Linköping University, SE 58183, Linköping, Sweden
| | - Xiao Zhen Zhou
- Departments of Biochemistry, Schulich School of Medicine & Dentistry, Western University, London, ON, N6G 2V4, Canada.
- Departments of Oncology, Schulich School of Medicine & Dentistry, Western University, London, ON, N6G 2V4, Canada.
- Departments of Pathology & Laboratory Medicine, Schulich School of Medicine & Dentistry, Western University, London, ON, N6G 2V4, Canada.
- Lawson Health Research Institute, Schulich School of Medicine & Dentistry, Western University, London, ON, N6G 2V4, Canada.
| | - Kun Ping Lu
- Departments of Biochemistry, Schulich School of Medicine & Dentistry, Western University, London, ON, N6G 2V4, Canada.
- Departments of Oncology, Schulich School of Medicine & Dentistry, Western University, London, ON, N6G 2V4, Canada.
- Robarts Research Institute, Schulich School of Medicine & Dentistry Western University, London, ON, N6G 2V4, Canada.
| | - Surendra Sharma
- Departments of Pediatrics, Women and Infants Hospital, Warren Alpert Medical School, Brown University, Providence, RI, 02905, USA.
- Departments of Pathology, Women and Infants Hospital, Warren Alpert Medical School, Brown University, Providence, RI, 02905, USA.
| |
Collapse
|
37
|
Wang R, Lu KP, Zhou XZ. Function and regulation of cis P-tau in the pathogenesis and treatment of conventional and nonconventional tauopathies. J Neurochem 2023; 166:904-914. [PMID: 37638382 DOI: 10.1111/jnc.15909] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2022] [Revised: 06/02/2023] [Accepted: 06/22/2023] [Indexed: 08/29/2023]
Abstract
Conventional tauopathies are a group of disease characterized by tau inclusions in the brains, including Alzheimer's disease (AD), Pick's disease (PiD), progressive supranuclear palsy (PSP), corticobasal degeneration (CBD), and certain types of frontotemporal dementia (FTD), among which AD is the most prevalent. Extensive post-translational modifications, especially hyperphosphorylation, and abnormal aggregation of tau protein underlie tauopathy. Cis-trans isomerization of protein plays an important role in protein folding, function, and degradation, which is regulated by peptidyl-proline isomerases (PPIases). Peptidyl-prolyl cis-trans isomerase NIMA-interacting 1 (Pin1), the only PPIase found to isomerize Pro following phosphorylated Ser or Thr residues, alters phosphorylated tau protein conformation at pT231-P motif. The cis P-tau but not trans P-tau serves as an early driver of multiple neurodegenerative disease, encompassing AD, traumatic brain injury (TBI), chronic traumatic encephalopathy (CTE), and vascular contributions to cognitive impairment and dementia (VCID). Cis but not trans P-tau is resistant to protein dephosphorylation and degradation, and also prone to protein aggregation. Cis P-tau loses its ability to stabilize microtubule, causing and spreading tauopathy mainly in axons, a pathological process called cistauosis. The conformation-specific monoclonal antibody that targets only the cis P-tau serves as a very early diagnosis method and a potential treatment of not only conventional tauopathies but also nonconventional tauopathies such as VCID, with clinical trials ongoing. Notably, cis P-tau antibody is the only clinical-stage Alzheimer's therapeutic that has shown the efficacy in animal models of not only AD but also TBI and stroke, which are very early stages of dementia. Here we review the identification and pathological consequences of cis pt231-tau, the role of its regulator Pin1, as well as the clinical implication of cis pt231-tau conformation-specific antibody in conventional and nonconventional tauopathies.
Collapse
Affiliation(s)
- Ruizhi Wang
- Departments of Biochemistry and Oncology, Schulich School of Medicine & Dentistry, Western University, London, Ontario, Canada
- Robarts Research Institute, Western University, London, Ontario, Canada
| | - Kun Ping Lu
- Departments of Biochemistry and Oncology, Schulich School of Medicine & Dentistry, Western University, London, Ontario, Canada
- Robarts Research Institute, Western University, London, Ontario, Canada
| | - Xiao Zhen Zhou
- Departments of Biochemistry and Oncology, Schulich School of Medicine & Dentistry, Western University, London, Ontario, Canada
- Department of Pathology and Laboratory Medicine, and Oncology, Western University, London, Ontario, Canada
- Lawson Health Research Institute, Western University, London, Ontario, Canada
| |
Collapse
|
38
|
Buh FC, Taiwe GS, Kobeissy FH, Wang KW, Maas AIR, Motah M, Meh BK, Youm E, Hutchinson PJA, Sumbele IUN. Serum Biomarker Concentrations upon Admission in Acute Traumatic Brain Injury: Associations with TBI Severity, Toxoplasma gondii Infection, and Outcome in a Referral Hospital Setting in Cameroon. NEUROSCI 2023; 4:164-177. [PMID: 39483201 PMCID: PMC11523680 DOI: 10.3390/neurosci4030015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2023] [Revised: 06/21/2023] [Accepted: 06/26/2023] [Indexed: 11/03/2024] Open
Abstract
Despite the available literature on traumatic brain injury (TBI) biomarkers elsewhere, data are limited or non-existent in sub-Saharan Africa (SSA). The aim of the study was to analyse associations in acute TBI between the admission serum biomarker concentrations and TBI severity, CT-scan findings, and outcome, as well as to explore the influence of concurrent Toxoplasma gondii infection. The concentrations of serum biomarkers (GFAP, NFL Tau, UCH-L1, and S100B) were measured and Toxoplasma gondii were detected in the samples obtained <24 h post injury. GOSE was used to evaluate the 6-month outcome. All of the biomarker levels increased with the severity of TBI, but this increase was significant only for NFL (p = 0.01). The GFAP values significantly increased (p = 0.026) in those with an unfavourable outcome. The Tau levels were higher in those who died (p = 0.017). GFAP and NFL were sensitive to CT-scan pathology (p values of 0.004 and 0.002, respectively). The S100B levels were higher (p < 0.001) in TBI patients seropositive to Toxoplasma gondii. In conclusion, NFL was found to be sensitive to TBI severity, while NFL and GFAP were predictive of CT intracranial abnormalities. Increased levels of GFAP and Tau were associated with poorer outcomes 6 months after TBI, and the S100B levels were significantly affected by concurrent T. gondii infection in TBI patients compared with the seronegative patients.
Collapse
Affiliation(s)
- Franklin Chu Buh
- Department of Animal Biology and Conservation, Faculty of Science, University of Buea, Buea P.O. Box 63, Cameroon; (G.S.T.); (B.K.M.); (I.U.N.S.)
- Panafrican Hospital Center, Douala P.O. Box 13152, Cameroon
| | - Germain Sotoing Taiwe
- Department of Animal Biology and Conservation, Faculty of Science, University of Buea, Buea P.O. Box 63, Cameroon; (G.S.T.); (B.K.M.); (I.U.N.S.)
| | - Firas H Kobeissy
- Center for Neurotrauma, Multiomics & Biomarkers (CNMB), Department of Neurobiology, Neuroscience Institute, Morehouse School of Medicine, 720 Westview Dr SW, Atlanta, GA 30310-1458, USA; (F.H.K.); (K.W.W.)
| | - Kevin W Wang
- Center for Neurotrauma, Multiomics & Biomarkers (CNMB), Department of Neurobiology, Neuroscience Institute, Morehouse School of Medicine, 720 Westview Dr SW, Atlanta, GA 30310-1458, USA; (F.H.K.); (K.W.W.)
| | - Andrew I R Maas
- Department of Neurosurgery, Antwerp University Hospital, University of Antwerp, 2000 Edegem, Belgium;
| | - Mathieu Motah
- Department of Surgery, Faculty of Medicine and Pharmaceutical Sciences, University of Douala, Douala P.O. Box 2701, Cameroon;
| | - Basil Kum Meh
- Department of Animal Biology and Conservation, Faculty of Science, University of Buea, Buea P.O. Box 63, Cameroon; (G.S.T.); (B.K.M.); (I.U.N.S.)
| | - Eric Youm
- Holo Healthcare, Nairobi 00400, Kenya;
| | - Peter J A Hutchinson
- Department of Clinical Neuroscience, University of Cambridge, Cambridge CB2 0QQ, UK;
| | - Irene Ule Ngole Sumbele
- Department of Animal Biology and Conservation, Faculty of Science, University of Buea, Buea P.O. Box 63, Cameroon; (G.S.T.); (B.K.M.); (I.U.N.S.)
| |
Collapse
|
39
|
Devoto C, Vorn R, Mithani S, Meier TB, Lai C, Broglio SP, McAllister T, Giza CC, Huber D, Harezlak J, Cameron KL, McGinty G, Jackson J, Guskiewicz K, Mihalik JP, Brooks A, Duma S, Rowson S, Nelson LD, Pasquina P, Turtzo C, Latour L, McCrea MA, Gill JM. Plasma phosphorylated tau181 as a biomarker of mild traumatic brain injury: findings from THINC and NCAA-DoD CARE Consortium prospective cohorts. Front Neurol 2023; 14:1202967. [PMID: 37662031 PMCID: PMC10470112 DOI: 10.3389/fneur.2023.1202967] [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: 04/09/2023] [Accepted: 07/18/2023] [Indexed: 09/05/2023] Open
Abstract
Objective The aim of this study was to investigate phosphorylated tau (p-tau181) protein in plasma in a cohort of mild traumatic brain injury (mTBI) patients and a cohort of concussed athletes. Methods This pilot study comprised two independent cohorts. The first cohort-part of a Traumatic Head Injury Neuroimaging Classification (THINC) study-with a mean age of 46 years was composed of uninjured controls (UIC, n = 30) and mTBI patients (n = 288) recruited from the emergency department with clinical computed tomography (CT) and research magnetic resonance imaging (MRI) findings. The second cohort-with a mean age of 19 years-comprised 133 collegiate athletes with (n = 112) and without (n = 21) concussions. The participants enrolled in the second cohort were a part of a multicenter, prospective, case-control study conducted by the NCAA-DoD Concussion Assessment, Research and Education (CARE) Consortium at six CARE Advanced Research Core (ARC) sites between 2015 and 2019. Blood was collected within 48 h of injury for both cohorts. Plasma concentration (pg/ml) of p-tau181 was measured using the Single Molecule Array ultrasensitive assay. Results Concentrations of plasma p-tau181 in both cohorts were significantly elevated compared to controls within 48 h of injury, with the highest concentrations of p-tau181 within 18 h of injury, with an area under the curve (AUC) of 0.690-0.748, respectively, in distinguishing mTBI patients and concussed athletes from controls. Among the mTBI patients, the levels of plasma p-tau181 were significantly higher in patients with positive neuroimaging (either CT+/MRI+, n = 74 or CT-/MRI+, n = 89) compared to mTBI patients with negative neuroimaging (CT-/MRI-, n = 111) findings and UIC (P-values < 0.05). Conclusion These findings indicate that plasma p-tau181 concentrations likely relate to brain injury, with the highest levels in patients with neuroimaging evidence of injury. Future research is needed to replicate and validate this protein assay's performance as a possible early diagnostic biomarker for mTBI/concussions.
Collapse
Affiliation(s)
- Christina Devoto
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, MD, United States
| | - Rany Vorn
- National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, United States
- School of Nursing, Johns Hopkins University, Baltimore, MD, United States
| | - Sara Mithani
- School of Nursing, University of Texas Health at San Antonio, San Antonio, TX, United States
| | - Timothy B. Meier
- Department of Neurosurgery, Medical College of Wisconsin, Milwaukee, WI, United States
| | - Chen Lai
- Center for Neuroscience and Regenerative Medicine, Uniformed Services University and Health Science, Bethesda, MD, United States
| | - Steven P. Broglio
- Michigan Concussion Center, University of Michigan, Ann Arbor, MI, United States
| | - Thomas McAllister
- Department of Psychiatry, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Christopher C. Giza
- Departments of Pediatrics and Neurosurgery, UCLA Steve Tisch BrainSPORT Program, University of California, Los Angeles, Los Angeles, CA, United States
| | - Daniel Huber
- Department of Neurosurgery, Medical College of Wisconsin, Milwaukee, WI, United States
| | - Jaroslaw Harezlak
- Department of Epidemiology and Biostatistics School of Public Health-Bloomington, Indiana University, Bloomington, IN, United States
| | - Kenneth L. Cameron
- John A. Feagin Sports Medicine Fellowship, Keller Army Hospital, West Point, NY, United States
| | - Gerald McGinty
- United States Air Force Academy, Colorado Springs, CO, United States
| | - Jonathan Jackson
- United States Air Force Academy, Colorado Springs, CO, United States
| | - Kevin Guskiewicz
- Matthew Gfeller Center, Department of Exercise and Sport Science, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Jason P. Mihalik
- Matthew Gfeller Center, Department of Exercise and Sport Science, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Alison Brooks
- Department of Orthopedics and Sports Medicine, University of Wisconsin, Madison, WI, United States
| | - Stefan Duma
- Department of Biomedical Engineering, Virginia Tech, Blacksburg, VA, United States
| | - Steven Rowson
- Department of Biomedical Engineering, Virginia Tech, Blacksburg, VA, United States
| | - Lindsay D. Nelson
- Department of Neurosurgery, Medical College of Wisconsin, Milwaukee, WI, United States
| | - Paul Pasquina
- Center for Neuroscience and Regenerative Medicine, Uniformed Services University and Health Science, Bethesda, MD, United States
| | - Christine Turtzo
- National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, United States
| | - Lawrence Latour
- National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, United States
| | - Michael A. McCrea
- Department of Neurosurgery, Medical College of Wisconsin, Milwaukee, WI, United States
| | - Jessica M. Gill
- School of Nursing, Johns Hopkins University, Baltimore, MD, United States
- Department of Neurology, Johns Hopkins University, Baltimore, MD, United States
| |
Collapse
|
40
|
Halicki MJ, Hind K, Chazot PL. Blood-Based Biomarkers in the Diagnosis of Chronic Traumatic Encephalopathy: Research to Date and Future Directions. Int J Mol Sci 2023; 24:12556. [PMID: 37628736 PMCID: PMC10454393 DOI: 10.3390/ijms241612556] [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: 07/10/2023] [Revised: 07/27/2023] [Accepted: 07/31/2023] [Indexed: 08/27/2023] Open
Abstract
Chronic Traumatic Encephalopathy (CTE) is a neurodegenerative disease consistently associated with repetitive traumatic brain injuries (TBIs), which makes multiple professions, such as contact sports athletes and the military, especially susceptible to its onset. There are currently no approved biomarkers to diagnose CTE, thus it can only be confirmed through a post-mortem brain autopsy. Several imaging and cerebrospinal fluid biomarkers have shown promise in the diagnosis. However, blood-based biomarkers can be more easily obtained and quantified, increasing their clinical feasibility and potential for prophylactic use. This article aimed to comprehensively review the studies into potential blood-based biomarkers of CTE, discussing common themes and limitations, as well as suggesting future research directions. While the interest in blood-based biomarkers of CTE has recently increased, the research is still in its early stages. The main issue for many proposed biomarkers is their lack of selectivity for CTE. However, several molecules, such as different phosphorylated tau isoforms, were able to discern CTE from different neurodegenerative diseases. Further, the results from studies on exosomal biomarkers suggest that exosomes are a promising source of biomarkers, reflective of the internal environment of the brain. Nonetheless, more longitudinal studies combining imaging, neurobehavioral, and biochemical approaches are warranted to establish robust biomarkers for CTE.
Collapse
Affiliation(s)
| | - Karen Hind
- Durham Wolfson Research Institute for Health and Wellbeing, Stockton-on-Tees TS17 6BH, UK;
| | - Paul L. Chazot
- Department of Biosciences, Wolfson Research Institute for Health and Wellbeing, Durham University, Durham DH1 3LE, UK
| |
Collapse
|
41
|
Gonzalez-Ortiz F, Dulewicz M, Ashton NJ, Kac PR, Zetterberg H, Andersson E, Yakoub Y, Hanrieder J, Turton M, Harrison P, Nellgård B, Karikari TK, Blennow K. Association of Serum Brain-Derived Tau With Clinical Outcome and Longitudinal Change in Patients With Severe Traumatic Brain Injury. JAMA Netw Open 2023; 6:e2321554. [PMID: 37399012 PMCID: PMC10318474 DOI: 10.1001/jamanetworkopen.2023.21554] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/20/2023] [Accepted: 05/10/2023] [Indexed: 07/04/2023] Open
Abstract
Importance Blood-based measurements of total tau (T-tau) are commonly used to examine neuronal injury in patients with traumatic brain injury (TBI), but current assays do not differentiate between brain-derived tau (BD-tau) and tau produced in peripheral tissues. A novel assay for BD-tau has recently been reported that selectively quantifies nonphosphorylated tau of central nervous system origin in blood samples. Objectives To examine the association of serum BD-tau with clinical outcomes in patients with severe TBI (sTBI) and its longitudinal changes over 1 year. Design, Setting, and Participants This prospective cohort study was conducted at the neurointensive unit at the Sahlgrenska University Hospital, Gothenburg, Sweden, between September 1, 2006, and July 1, 2015. The study included 39 patients with sTBI followed up for up to 1 year. Statistical analysis was performed between October and November 2021. Exposures Serum BD-tau, T-tau, phosphorylated tau231 (p-tau231), and neurofilament light chain (NfL) measured on days 0, 7, and 365 after injury. Main Outcomes and Measures Associations of serum biomarkers with clinical outcome and longitudinal change in sTBI. Severity of sTBI was evaluated using the Glasgow Coma Scale at hospital admission, while clinical outcome was assessed with the Glasgow Outcome Scale (GOS) at 1-year follow-up. Participants were classified as having a favorable outcome (GOS score, 4-5) or unfavorable outcome (GOS score, 1-3). Results Among the 39 patients (median age at admission, 36 years [IQR, 22-54 years]; 26 men [66.7%]) in the study on day 0, the mean (SD) serum BD-tau level was higher among patients with unfavorable outcomes vs those with favorable outcomes (191.4 [190.8] pg/mL vs 75.6 [60.3] pg/mL; mean difference, 115.9 pg/mL [95% CI, 25.7-206.1 pg/mL]), while the other markers had smaller between-group mean differences (serum T-tau, 60.3 pg/mL [95% CI, -22.0 to 142.7 pg/mL]; serum p-tau231, 8.3 pg/mL [95% CI, -6.4 to 23.0 pg/mL]; serum NfL, -5.4 pg/mL [95% CI, -99.0 to 88.3 pg/mL]). Similar results were recorded on day 7. Longitudinally, baseline serum BD-tau concentrations showed slower decreases in the whole cohort (42.2% on day 7 [from 138.6 to 80.1 pg/mL] and 93.0% on day 365 [from 138.6 to 9.7 pg/mL]) compared with serum T-tau (81.5% on day 7 [from 57.3 to 10.6 pg/mL] and 99.0% on day 365 [from 57.3 to 0.6 pg/mL]) and p-tau231 (92.5% on day 7 [from 20.1 to 1.5 pg/mL] and 95.0% on day 365 [from 20.1 to 1.0 pg/mL]). These results did not change when considering clinical outcome, where T-tau decreased twice as fast as BD-tau in both groups. Similar results were obtained for p-tau231. Furthermore, the biomarker levels on day 365 were lower, compared with day 7, for BD-tau but not T-tau or p-tau231. Serum NfL had a different trajectory to the tau biomarkers, with levels increasing by 255.9% on day 7 compared with day 0 (from 86.8 to 308.9 pg/mL) but decreasing by 97.0% by day 365 vs day 7 (from 308.9 to 9.2 pg/mL). Conclusions and Relevance This study suggests that serum BD-tau, T-tau, and p-tau231 have differential associations with clinical outcome and 1-year longitudinal change in patients with sTBI. Serum BD-tau demonstrated utility as a biomarker to monitor outcomes in sTBI and can provide valuable information regarding acute neuronal damage.
Collapse
Affiliation(s)
- Fernando Gonzalez-Ortiz
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, the Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
| | - Maciej Dulewicz
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, the Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
| | - Nicholas J. Ashton
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, the Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
- King’s College London, Institute of Psychiatry, Psychology and Neuroscience, Maurice Wohl Clinical Neuroscience Institute London, London, United Kingdom
- NIHR Biomedical Research Centre for Mental Health and Biomedical Research Unit for Dementia at South London and Maudsley NHS Foundation London, London, United Kingdom
- Centre for Age-Related Medicine, Stavanger University Hospital, Stavanger, Norway
| | - Przemysław R. Kac
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, the Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
| | - Henrik Zetterberg
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, the Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
- Department of Neurodegenerative Disease, UCL Institute of Neurology, Queen Square, London, United Kingdom
- UK Dementia Research Institute at UCL, London, United Kingdom
- Hong Kong Center for Neurodegenerative Diseases, Clear Water Bay, Hong Kong, China
- Wisconsin Alzheimer’s Disease Research Center, University of Wisconsin School of Medicine and Public Health, University of Wisconsin–Madison, Madison
| | - Emma Andersson
- Department of Anaesthesiology and Intensive Care, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Sweden
| | - Yara Yakoub
- Douglas Mental Health University Institute, Centre for Studies on the Prevention of Alzheimer’s Disease, Montreal, Quebec, Canada
| | - Jörg Hanrieder
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, the Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
| | | | | | - Bengt Nellgård
- Department of Anaesthesiology and Intensive Care, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Sweden
- Department of Anesthesiology, Surgery and Intensive Care; Sahlgrenska University Hospital, Mölndal, Sweden
| | - Thomas K. Karikari
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, the Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
- Department of Psychiatry, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Kaj Blennow
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, the Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
| |
Collapse
|
42
|
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. 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: 5] [Impact Index Per Article: 2.5] [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
|
43
|
Nasrallah F, Bellapart J, Walsham J, Jacobson E, To XV, Manzanero S, Brown N, Meyer J, Stuart J, Evans T, Chandra SS, Ross J, Campbell L, Senthuran S, Newcombe V, McCullough J, Fleming J, Pollard C, Reade M. PREdiction and Diagnosis using Imaging and Clinical biomarkers Trial in Traumatic Brain Injury (PREDICT-TBI) study protocol: an observational, prospective, multicentre cohort study for the prediction of outcome in moderate-to-severe TBI. BMJ Open 2023; 13:e067740. [PMID: 37094888 PMCID: PMC10151972 DOI: 10.1136/bmjopen-2022-067740] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Accepted: 03/31/2023] [Indexed: 04/26/2023] Open
Abstract
INTRODUCTION Traumatic brain injury (TBI) is a heterogeneous condition with a broad spectrum of injury severity, pathophysiological processes and variable outcomes. For moderate-to-severe TBI survivors, recovery is often protracted and outcomes can range from total dependence to full recovery. Despite advances in medical treatment options, prognosis remains largely unchanged. The objective of this study is to develop a machine learning predictive model for neurological outcomes at 6 months in patients with a moderate-to-severe TBI, incorporating longitudinal clinical, multimodal neuroimaging and blood biomarker predictor variables. METHODS AND ANALYSIS A prospective, observational, cohort study will enrol 300 patients with moderate-to-severe TBI from seven Australian hospitals over 3 years. Candidate predictors including demographic and general health variables, and longitudinal clinical, neuroimaging (CT and MRI), blood biomarker and patient-reported outcome measures will be collected at multiple time points within the acute phase of injury. The predictor variables will populate novel machine learning models to predict the Glasgow Outcome Scale Extended 6 months after injury. The study will also expand on current prognostic models by including novel blood biomarkers (circulating cell-free DNA), and the results of quantitative neuroimaging such as Quantitative Susceptibility Mapping and Dynamic Contrast Enhanced MRI as predictor variables. ETHICS AND DISSEMINATION Ethical approval has been obtained by the Royal Brisbane and Women's Hospital Human Research Ethics Committee, Queensland. Participants or their substitute decision-maker/s will receive oral and written information about the study before providing written informed consent. Study findings will be disseminated by peer-review publications and presented at national and international conferences and clinical networks. TRIAL REGISTRATION NUMBER ACTRN12620001360909.
Collapse
Affiliation(s)
- Fatima Nasrallah
- The Queensland Brain Institute, The University of Queensland, Saint Lucia, Queensland, Australia
| | - Judith Bellapart
- Intensive Care Unit, Royal Brisbane and Women's Hospital, Metro North Health Service District, Herston, Queensland, Australia
| | - James Walsham
- Intensive Care Unit, Princess Alexandra Hospital, Woolloongabba, Queensland, Australia
- Faculty of Medicine, The University of Queensland, Saint Lucia, Queensland, Australia
| | - Esther Jacobson
- Jamieson Trauma Institute, Metro North Health Service District, Herston, Queensland, Australia
| | - Xuan Vinh To
- The Queensland Brain Institute, The University of Queensland, Saint Lucia, Queensland, Australia
| | - Silvia Manzanero
- Jamieson Trauma Institute, Metro North Health Service District, Herston, Queensland, Australia
| | - Nathan Brown
- Intensive Care Unit, Royal Brisbane and Women's Hospital, Metro North Health Service District, Herston, Queensland, Australia
- Faculty of Medicine, The University of Queensland, Saint Lucia, Queensland, Australia
| | - Jason Meyer
- Intensive Care Unit, Princess Alexandra Hospital, Woolloongabba, Queensland, Australia
| | - Janine Stuart
- Intensive Care Unit, Royal Brisbane and Women's Hospital, Metro North Health Service District, Herston, Queensland, Australia
| | - Tracey Evans
- The Queensland Brain Institute, The University of Queensland, Saint Lucia, Queensland, Australia
| | - Shekhar S Chandra
- School of Information Technology and Electrical Engineering, Architecture and Information Technology, The University of Queensland, Saint Lucia, Queensland, Australia
| | - Jason Ross
- Health and Biosecurity, CSIRO, Westmead, New South Wales, Australia
| | - Lewis Campbell
- Intensive Care Unit, Royal Darwin Hospital, Casuarina, Darwin, Australia
- College of Medicine and Public Health, Flinders University, Adelaide, South Australia, Australia
| | - Siva Senthuran
- Intensive Care Unit, Townsville Hospital and Health Service, Townsville, Queensland, Australia
| | - Virginia Newcombe
- University Division of Anaesthesia, University of Cambridge, Cambridge, UK
| | - James McCullough
- Intensive Care Unit, Gold Coast Hospital and Health Service, Gold Coast, Queensland, Australia
| | - Jennifer Fleming
- School of Health and Rehabilitation Sciences, The University of Queensland, Saint Lucia, Queensland, Australia
| | - Clifford Pollard
- School of Information Technology and Electrical Engineering, Architecture and Information Technology, The University of Queensland, Saint Lucia, Queensland, Australia
| | - Michael Reade
- Intensive Care Unit, Royal Brisbane and Women's Hospital, Metro North Health Service District, Herston, Queensland, Australia
- Faculty of Medicine, The University of Queensland, Saint Lucia, Queensland, Australia
| |
Collapse
|
44
|
Ritter A, Shan G, Montes A, Randall R, Bernick C. Traumatic encephalopathy syndrome: application of new criteria to a cohort exposed to repetitive head impacts. Br J Sports Med 2023; 57:389-394. [PMID: 36517216 PMCID: PMC10086298 DOI: 10.1136/bjsports-2022-105819] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/21/2022] [Indexed: 12/15/2022]
Abstract
OBJECTIVE To examine the characteristics of those who fulfil the recent National Institute of Neurological Disease and Stroke (NINDS) Consensus Diagnostic Criteria for Traumatic Encephalopathy Syndrome (TES) and test whether they show differences in MRI-based regional brain volumes, cognitive domains, and certain plasma biomarkers. METHODS Professional fighters 35 years of age or older and/or retired were included. Participants were categorised as either having TES (TES+) or not (non-TES). TES+ participants were further subtyped by their cognitive profile. Multiple linear regression models were used to compare MRI-based regional brain volumes, cognitive performance, plasma tau and neurofilament light levels between TES- and TES+ groups. RESULTS 176 participants (110 boxers and 66 MMA) were included in the analysis. 72 (41%)/176 were categorised as having TES, the likelihood of TES increasing with age. TES+ participants tended to be boxers, started fighting at a younger age, had more professional fights and knocked out more frequently. The TES+ group had lower regional brain volumes including both grey and white matter structures. TES+ also had lower scores on simple and choice reaction time, psychomotor speed and Trails A . CONCLUSION The new TES criteria does distinguish a group of fighters with differences in regional brain volumes and reduced cognitive function. Our findings support the use of the NINDS criteria for TES in further research of the long-term effects of repetitive head impacts.
Collapse
Affiliation(s)
- Aaron Ritter
- Neurological Institute, Cleveland Clinic, Las Vegas, Nevada, USA
| | - Guogen Shan
- Biostatistics, University of Florida, Gainesville, Florida, USA
| | - Arturo Montes
- Medicine, University of Las Vegas, Las Vegas, Nevada, USA
| | - Rebekah Randall
- Neurological Institute, Cleveland Clinic, Las Vegas, Nevada, USA
| | - Charles Bernick
- Department of Neurology, University of Washington, Seattle, Washington, USA
| |
Collapse
|
45
|
Yadikar H, Johnson C, Pafundi N, Nguyen L, Kurup M, Torres I, Al-Enezy A, Yang Z, Yost R, Kobeissy FH, Wang KKW. Neurobiochemical, Peptidomic, and Bioinformatic Approaches to Characterize Tauopathy Peptidome Biomarker Candidates in Experimental Mouse Model of Traumatic Brain Injury. Mol Neurobiol 2023; 60:2295-2319. [PMID: 36635478 DOI: 10.1007/s12035-022-03165-y] [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: 03/21/2022] [Accepted: 12/10/2022] [Indexed: 01/14/2023]
Abstract
Traumatic brain injury (TBI) is a multidimensional damage, and currently, no FDA-approved medicine is available. Multiple pathways in the cell are triggered through a head injury (e.g., calpain and caspase activation), which truncate tau and generate variable fragment sizes (MW 400-45,000 K). In this study, we used an open-head TBI mouse model generated by controlled cortical impact (CCI) and collected ipsilateral (IC) and contralateral (CC) mice htau brain cortices at one (D1) three (D3), and seven (D7) days post-injury. We implemented immunological (antibody-based detection) and peptidomic approaches (nano-reversed-phase liquid chromatography/tandem mass spectrometry) to investigate proteolytic tau peptidome (low molecular weight (LMW) < 10 K)) and pathological phosphorylation sites (high-molecular-weight (HMW); > 10 K) derived from CCI-TBI animal models. Our immunoblotting analysis verified tau hyperphosphorylation, HMW, and HMW breakdown products (HMW-BDP) formation of tau (e.g., pSer202, pThr181, pThr231, pSer396, and pSer404), following CCI-TBI. Peptidomic data revealed unique sequences of injury-dependent proteolytic peptides generated from human tau protein. Among the N-terminal tau peptides, EIPEGTTAEEAGIGDTPSLEDEAAGHVTQA (a.a. 96-125) and AQPHTEIPEGTTAEEAGIGDTPSLEDEAAGHVTQARM (a.a. 91-127). Examples of tau C-terminal peptides identified include NVSSTGSIDMVDSPQLATLADEVSASLAKQGL (a.a. 410-441) and QLATLADEVSASLAKQGL (a.a. 424-441). Our peptidomic bioinformatic tools showed the association of proteases, such as CAPN1, CAPN2, and CTSL; CASP1, MMP7, and MMP9; and ELANE, GZMA, and MEP1A, in CCI-TBI tau peptidome. In clinical trials for novel TBI treatments, it might be useful to monitor a subset of tau peptidome as targets for biomarker utility and use them for a "theranostic" approach.
Collapse
Affiliation(s)
- Hamad Yadikar
- Department of Biological Sciences, Faculty of Science, Kuwait University, Kuwait, Kuwait.
| | - Connor Johnson
- Department of Biological Sciences, Faculty of Science, Kuwait University, Kuwait, Kuwait
| | - Niko Pafundi
- Department of Biological Sciences, Faculty of Science, Kuwait University, Kuwait, Kuwait
| | - Lynn Nguyen
- Department of Biological Sciences, Faculty of Science, Kuwait University, Kuwait, Kuwait
| | - Milin Kurup
- Department of Biological Sciences, Faculty of Science, Kuwait University, Kuwait, Kuwait
| | - Isabel Torres
- Department of Biological Sciences, Faculty of Science, Kuwait University, Kuwait, Kuwait
| | - Albandery Al-Enezy
- Department of Biological Sciences, Faculty of Science, Kuwait University, Kuwait, Kuwait
| | - Zhihui Yang
- Department of Biological Sciences, Faculty of Science, Kuwait University, Kuwait, Kuwait
| | - Richard Yost
- Department of Chemistry, Chemistry Laboratory Building, University of Florida, Gainesville, FL, 32611, USA
| | - Firas H Kobeissy
- Program for Neurotrauma, Neuroproteomics & Biomarkers Research, Departments of Emergency Medicine, Psychiatry, Neuroscience and Chemistry, University of Florida, Gainesville, FL, USA. .,Department of Biochemistry and Molecular Genetics, American University of Beirut, Beirut, Lebanon. .,Morehouse School of Medicine, Department of Neurobiology, Center for Neurotrauma, Multiomics & Biomarkers (CNMB), 720 Westview Dr. SW, Atlanta, GA, 30310, USA.
| | - Kevin K W Wang
- Program for Neurotrauma, Neuroproteomics & Biomarkers Research, Departments of Emergency Medicine, Psychiatry, Neuroscience and Chemistry, University of Florida, Gainesville, FL, USA. .,Morehouse School of Medicine, Department of Neurobiology, Center for Neurotrauma, Multiomics & Biomarkers (CNMB), 720 Westview Dr. SW, Atlanta, GA, 30310, USA. .,Brain Rehabilitation Research Center, Malcom Randall VA Medical Center, Gainesville, FL, 32608, USA.
| |
Collapse
|
46
|
Kagusa H, Yamaguchi I, Shono K, Mizobuchi Y, Shikata E, Matsuda T, Miyamoto T, Hara K, Kitazato KT, Uto Y, Kanematsu Y, Takagi Y. Differences in amyloid-β and tau/p-tau deposition in blood-injected mouse brains using micro-syringe to mimic traumatic brain microhemorrhages. J Chem Neuroanat 2023; 130:102258. [PMID: 36925083 DOI: 10.1016/j.jchemneu.2023.102258] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2022] [Revised: 03/13/2023] [Accepted: 03/13/2023] [Indexed: 03/17/2023]
Abstract
BACKGROUND Cerebral microbleeds (CMBs) due to traumatic brain injuries (TBI) have been shown to lead to cognitive decline and impairment. CMBs caused by TBI may be associated with pathophysiological mechanisms involving inflammation and the accumulation of amyloid-β (Aβ), tau, and phosphorylated tau (p-tau), contributing to cognitive abnormalities. However, their relationships remain unclear. OBJECTIVES To test our hypothesis that Aβ, tau, and p-tau are accumulated and regulated separately in mice with injuries imitating CMBs from TBI, we studied. METHODS Seven-week-old C57BL/6 male mice were injected with 15 μL of heparinized autologous blood or saline by micro-syringe into the front lobe. Expression profiles and regulation of Aβ, tau, and p-tau were assessed immunohistochemically over time. RESULTS On day 7 after blood injection, Iba-1+ and S100B+ cells in damaged cortex adjacent to the injection site were higher than saline injection group and non-injected sham. On days 3-14, Aβ deposition were gradually increased but normalized by day 28. In contrast, tau/p-tau deposition gradually increased during days 14-28 and dispersed along the corticomedullary junction adjacent to hem deposits, indicating different expression profiles from Aβ. Deposits of Aβ, but not tau/p-tau, were phagocytosed by CD163+ macrophages increased by Gc-protein macrophage-activating factor during days 7-28, suggesting different mechanisms of deposition and regulation between Aβ and tau/p-tau. CONCLUSION Deposition and regulation differ between Aβ and tau/p-tau in mice with injuries mimicking CMBs from TBI. Further clarification of relationships between the pathologies of cognitive impairment and their neurodegenerative consequences is needed.
Collapse
Affiliation(s)
- Hiroshi Kagusa
- Department of Neurosurgery, Graduate School of Biomedical Sciences, Tokushima University, Tokushima, Japan.
| | - Izumi Yamaguchi
- Department of Neurosurgery, Graduate School of Biomedical Sciences, Tokushima University, Tokushima, Japan
| | - Kenji Shono
- Department of Neurosurgery, Graduate School of Biomedical Sciences, Tokushima University, Tokushima, Japan
| | - Yoshifumi Mizobuchi
- Department of Neurosurgery, Graduate School of Biomedical Sciences, Tokushima University, Tokushima, Japan
| | - Eiji Shikata
- Department of Neurosurgery, Graduate School of Biomedical Sciences, Tokushima University, Tokushima, Japan
| | - Taku Matsuda
- Department of Neurosurgery, Graduate School of Biomedical Sciences, Tokushima University, Tokushima, Japan
| | - Takeshi Miyamoto
- Department of Neurosurgery, Graduate School of Biomedical Sciences, Tokushima University, Tokushima, Japan
| | - Keijiro Hara
- Department of Neurosurgery, Graduate School of Biomedical Sciences, Tokushima University, Tokushima, Japan
| | - Keiko T Kitazato
- Department of Neurosurgery, Graduate School of Biomedical Sciences, Tokushima University, Tokushima, Japan
| | - Yoshihiro Uto
- Graduate School of Technology, Industrial and Social Science, Tokushima University, Tokushima, Japan
| | - Yasuhisa Kanematsu
- Department of Neurosurgery, Graduate School of Biomedical Sciences, Tokushima University, Tokushima, Japan
| | - Yasushi Takagi
- Department of Neurosurgery, Graduate School of Biomedical Sciences, Tokushima University, Tokushima, Japan
| |
Collapse
|
47
|
Ashton NJ, Moseby-Knappe M, Benedet AL, Grötschel L, Lantero-Rodriguez J, Karikari TK, Hassager C, Wise MP, Stammet P, Kjaergaard J, Friberg H, Nielsen N, Cronberg T, Zetterberg H, Blennow K. Alzheimer Disease Blood Biomarkers in Patients With Out-of-Hospital Cardiac Arrest. JAMA Neurol 2023; 80:388-396. [PMID: 36877496 PMCID: PMC9989959 DOI: 10.1001/jamaneurol.2023.0050] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/07/2023]
Abstract
Importance Blood phosphorylated tau (p-tau) and amyloid-β peptides (Aβ) are promising peripheral biomarkers of Alzheimer disease (AD) pathology. However, their potential alterations due to alternative mechanisms, such as hypoxia in patients resuscitated from cardiac arrest, are not known. Objective To evaluate whether the levels and trajectories of blood p-tau, Aβ42, and Aβ40 following cardiac arrest, in comparison with neural injury markers neurofilament light (NfL) and total tau (t-tau), can be used for neurological prognostication following cardiac arrest. Design, Setting, and Participants This prospective clinical biobank study used data from the randomized Target Temperature Management After Out-of-Hospital Cardiac Arrest (TTM) trial. Unconscious patients with cardiac arrest of presumed cardiac origin were included between November 11, 2010, and January 10, 2013, from 29 international sites. Serum analysis for serum NfL and t-tau were performed between August 1 and August 23, 2017. Serum p-tau, Aβ42, and Aβ40 were analyzed between July 1 and July 15, 2021, and between May 13 and May 25, 2022. A total of 717 participants from the TTM cohort were examined: an initial discovery subset (n = 80) and a validation subset. Both subsets were evenly distributed for good and poor neurological outcome after cardiac arrest. Exposures Serum p-tau, Aβ42, and Aβ40 concentrations using single molecule array technology. Serum levels of NfL and t-tau were included as comparators. Main Outcomes and Measures Blood biomarker levels at 24 hours, 48 hours, and 72 hours after cardiac arrest. Poor neurologic outcome at 6-month follow-up, defined according to the cerebral performance category scale as category 3 (severe cerebral disability), 4 (coma), or 5 (brain death). Results This study included 717 participants (137 [19.1%] female and 580 male [80.9%]; mean [SD] age, 63.9 [13.5] years) who experienced out-of-hospital cardiac arrest. Significantly elevated serum p-tau levels were observed at 24 hours, 48 hours, and 72 hours in cardiac arrest patients with poor neurological outcome. The magnitude and prognostication of the change was greater at 24 hours (area under the receiver operating characteristic curve [AUC], 0.96; 95% CI, 0.95-0.97), which was similar to NfL (AUC, 0.94; 95% CI, 0.92-0.96). However, at later time points, p-tau levels decreased and were weakly associated with neurological outcome. In contrast, NfL and t-tau maintained high diagnostic accuracies, even 72 hours after cardiac arrest. Serum Aβ42 and Aβ40 concentrations increased over time in most patients but were only weakly associated with neurological outcome. Conclusions and Relevance In this case-control study, blood biomarkers indicative of AD pathology demonstrated different dynamics of change after cardiac arrest. The increase of p-tau at 24 hours after cardiac arrest suggests a rapid secretion from the interstitial fluid following hypoxic-ischemic brain injury rather than ongoing neuronal injury like NfL or t-tau. In contrast, delayed increases of Aβ peptides after cardiac arrest indicate activation of amyloidogenic processing in response to ischemia.
Collapse
Affiliation(s)
- Nicholas J Ashton
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience & Physiology, The Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden.,Wallenberg Centre for Molecular and Translational Medicine, University of Gothenburg, Gothenburg, Sweden.,Institute of Psychiatry, Psychology and Neuroscience, Maurice Wohl Clinical Neuroscience Institute, King's College London, London, United Kingdom.,National Institute for Health and Care Research Biomedical Research Centre for Mental Health and Biomedical Research Unit for Dementia, South London and Maudsley National Health Service Foundation, London, United Kingdom
| | - Marion Moseby-Knappe
- Skåne University Hospital, Department of Clinical Sciences, Neurology, Lund University, Lund, Sweden
| | - Andrea L Benedet
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience & Physiology, The Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
| | - Lana Grötschel
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience & Physiology, The Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
| | - Juan Lantero-Rodriguez
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience & Physiology, The Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
| | - Thomas K Karikari
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience & Physiology, The Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden.,McGill Centre for Studies in Aging, Translational Neuroimaging Laboratory, McGill University, Montreal, Quebec, Canada
| | - Christian Hassager
- Department of Cardiology, The Heart Centre, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark
| | - Matt P Wise
- Adult Critical Care, University Hospital of Wales, Heath Park, Cardiff, United Kingdom
| | - Pascal Stammet
- Department of Anesthesia and Intensive Care, Centre Hospitalier de Luxembourg, Luxembourg, Luxembourg.,Faculty of Science, Technology and Medicine, Department of Life Sciences and Medicine, University of Luxembourg, Esch-sur-Alzette, Luxembourg
| | - Jesper Kjaergaard
- Department of Cardiology, The Heart Centre, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark
| | - Hans Friberg
- Department of Clinical Sciences Lund, Anesthesia & Intensive Care Section, Helsingborg Hospital, Lund University, Lund, Sweden
| | - Niklas Nielsen
- Department of Clinical Sciences Lund, Anesthesia & Intensive Care Section, Helsingborg Hospital, Lund University, Lund, Sweden
| | - Tobias Cronberg
- Skåne University Hospital, Department of Clinical Sciences, Neurology, Lund University, Lund, Sweden
| | - Henrik Zetterberg
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience & Physiology, The Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden.,Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden.,Department of Neurodegenerative Disease, University College London Queen Square Institute of Neurology, London, United Kingdom.,United Kingdom Dementia Research Institute at University College London, University College London, London, United Kingdom.,Hong Kong Center for Neurodegenerative Diseases, Hong Kong, China
| | - Kaj Blennow
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience & Physiology, The Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden.,Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
| |
Collapse
|
48
|
Gonzalez-Ortiz F, Turton M, Kac PR, Smirnov D, Premi E, Ghidoni R, Benussi L, Cantoni V, Saraceno C, Rivolta J, Ashton NJ, Borroni B, Galasko D, Harrison P, Zetterberg H, Blennow K, Karikari TK. Brain-derived tau: a novel blood-based biomarker for Alzheimer's disease-type neurodegeneration. Brain 2023; 146:1152-1165. [PMID: 36572122 PMCID: PMC9976981 DOI: 10.1093/brain/awac407] [Citation(s) in RCA: 90] [Impact Index Per Article: 45.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2022] [Revised: 09/23/2022] [Accepted: 09/25/2022] [Indexed: 12/28/2022] Open
Abstract
Blood-based biomarkers for amyloid beta and phosphorylated tau show good diagnostic accuracies and agreements with their corresponding CSF and neuroimaging biomarkers in the amyloid/tau/neurodegeneration [A/T/(N)] framework for Alzheimer's disease. However, the blood-based neurodegeneration marker neurofilament light is not specific to Alzheimer's disease while total-tau shows lack of correlation with CSF total-tau. Recent studies suggest that blood total-tau originates principally from peripheral, non-brain sources. We sought to address this challenge by generating an anti-tau antibody that selectively binds brain-derived tau and avoids the peripherally expressed 'big tau' isoform. We applied this antibody to develop an ultrasensitive blood-based assay for brain-derived tau, and validated it in five independent cohorts (n = 609) including a blood-to-autopsy cohort, CSF biomarker-classified cohorts and memory clinic cohorts. In paired samples, serum and CSF brain-derived tau were significantly correlated (rho = 0.85, P < 0.0001), while serum and CSF total-tau were not (rho = 0.23, P = 0.3364). Blood-based brain-derived tau showed equivalent diagnostic performance as CSF total-tau and CSF brain-derived tau to separate biomarker-positive Alzheimer's disease participants from biomarker-negative controls. Furthermore, plasma brain-derived tau accurately distinguished autopsy-confirmed Alzheimer's disease from other neurodegenerative diseases (area under the curve = 86.4%) while neurofilament light did not (area under the curve = 54.3%). These performances were independent of the presence of concomitant pathologies. Plasma brain-derived tau (rho = 0.52-0.67, P = 0.003), but not neurofilament light (rho = -0.14-0.17, P = 0.501), was associated with global and regional amyloid plaque and neurofibrillary tangle counts. These results were further verified in two memory clinic cohorts where serum brain-derived tau differentiated Alzheimer's disease from a range of other neurodegenerative disorders, including frontotemporal lobar degeneration and atypical parkinsonian disorders (area under the curve up to 99.6%). Notably, plasma/serum brain-derived tau correlated with neurofilament light only in Alzheimer's disease but not in the other neurodegenerative diseases. Across cohorts, plasma/serum brain-derived tau was associated with CSF and plasma AT(N) biomarkers and cognitive function. Brain-derived tau is a new blood-based biomarker that outperforms plasma total-tau and, unlike neurofilament light, shows specificity to Alzheimer's disease-type neurodegeneration. Thus, brain-derived tau demonstrates potential to complete the AT(N) scheme in blood, and will be useful to evaluate Alzheimer's disease-dependent neurodegenerative processes for clinical and research purposes.
Collapse
Affiliation(s)
- Fernando Gonzalez-Ortiz
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, Gothenburg 405 30, Sweden
| | - Michael Turton
- Bioventix Plc, Romans Business Park, Farnham, Surrey GU9 7SX, UK
| | - Przemysław R Kac
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, Gothenburg 405 30, Sweden
| | - Denis Smirnov
- University of California, San Diego and Shiely-Marcos Alzheimer’s Disease Research Center, La Jolla, CA 92037, USA
| | - Enrico Premi
- Neurology Unit, Department of Clinical and Experimental Sciences, University of Brescia, Brescia, BS 25121, Italy
| | - Roberta Ghidoni
- Molecular Markers Laboratory, IRCCS Istituto Centro San Giovanni di Dio Fatebenefratelli, Brescia 25121, Italy
| | - Luisa Benussi
- Molecular Markers Laboratory, IRCCS Istituto Centro San Giovanni di Dio Fatebenefratelli, Brescia 25121, Italy
| | - Valentina Cantoni
- Neurology Unit, Department of Clinical and Experimental Sciences, University of Brescia, Brescia, BS 25121, Italy
| | - Claudia Saraceno
- Molecular Markers Laboratory, IRCCS Istituto Centro San Giovanni di Dio Fatebenefratelli, Brescia 25121, Italy
| | - Jasmine Rivolta
- Neurology Unit, Department of Clinical and Experimental Sciences, University of Brescia, Brescia, BS 25121, Italy
| | - Nicholas J Ashton
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, Gothenburg 405 30, Sweden
- Wallenberg Centre for Molecular and Translational Medicine, University of Gothenburg, Gothenburg 405 30, Sweden
- King’s College London, Institute of Psychiatry, Psychology and Neuroscience, Maurice Wohl Clinical Neuroscience Institute, London, SE5 8AF, UK
- NIHR Biomedical Research Centre for Mental Health and Biomedical Research Unit for Dementia at South London and Maudsley NHS Foundation, London, SE5 8AF, UK
| | - Barbara Borroni
- Neurology Unit, Department of Clinical and Experimental Sciences, University of Brescia, Brescia, BS 25121, Italy
| | - Douglas Galasko
- University of California, San Diego and Shiely-Marcos Alzheimer’s Disease Research Center, La Jolla, CA 92037, USA
| | - Peter Harrison
- Bioventix Plc, Romans Business Park, Farnham, Surrey GU9 7SX, UK
| | - Henrik Zetterberg
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, Gothenburg 405 30, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, 431 80, Sweden
- Department of Neurodegenerative Disease, UCL Institute of Neurology, London, WC1N 3BG, UK
- UK Dementia Research Institute at UCL, London, WC1E 6BT, UK
- Hong Kong Center for Neurodegenerative Diseases, Shatin, N.T., Hong Kong, China
| | - Kaj Blennow
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, Gothenburg 405 30, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, 431 80, Sweden
| | - Thomas K Karikari
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, Gothenburg 405 30, Sweden
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA 15213, USA
| |
Collapse
|
49
|
Ni M, Zhu ZH, Gao F, Dai LB, Lv XY, Wang Q, Zhu XX, Xie JK, Shen Y, Wang SC, Xie Q. Plasma Core Alzheimer's Disease Biomarkers Predict Amyloid Deposition Burden by Positron Emission Tomography in Chinese Individuals with Cognitive Decline. ACS Chem Neurosci 2023; 14:170-179. [PMID: 36547971 DOI: 10.1021/acschemneuro.2c00636] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Blood-based biomarkers have been considered as a promising method for the diagnosis of Alzheimer's disease (AD). The reliability and accuracy of plasma core AD biomarkers, including phosphorylated tau (P-tau181), total tau (T-tau), Aβ42, and Aβ40, have also been confirmed in diagnosing AD and predicting cerebral β-amyloid (Aβ) deposition in Western populations, while fewer research studies have ever been conducted in China's Han population. In this study, we investigated the capability of plasma core AD biomarkers in predicting cerebral Aβ deposition burden among the China Aging and Neurodegenerative Disorder Initiative (CANDI) cohort consisting of cognitively normal (CN), mild cognitive impairment (MCI), AD dementia, and non-Alzheimer's dementia disease (Non-ADD). Body fluid (plasma and CSF) AD core biomarkers were measured via single-molecule array (Simoa) immunoassay. The global standard uptake value ratio (SUVR) was then calculated by 18F-florbetapir PET, which was divided into positive (+) and negative (-). The most significant correlation between plasma and CSF was plasma P-tau181 (r = 0.526, P < 0.0001). Plasma P-tau181 and P-tau181/T-tau ratio were positively correlated with global SUVR (r = 0.257, P < 0.0001; r = 0.263, P < 0.0001, respectively), while Aβ42 and Aβ42/Aβ40 ratio were negatively correlated with global SUVR (r = -0.346, P < 0.0001; r = -0.407, P < 0.0001, respectively). Interestingly, voxel-wise analysis showed that plasma P-tau181 and P-tau181/T-tau ratio were negatively related to 18F-florbetapir PET in the hippocampus and parahippocampal cortex. The optimal predictive capability in distinguishing all Aβ+ participants from Aβ- participants and MCI+ from MCI- subgroups was the plasma P-tau181/T-tau ratio (AUC = 0.825 and 0.834, respectively). Our study suggested that plasma P-tau181 and P-tau181/T-tau ratio possessed better diagnostic and predictive values than plasma Aβ42 and Aβ42/Aβ40 in this cohort, a finding that may be useful in clinical practices and trials in China.
Collapse
Affiliation(s)
- Ming Ni
- Department of Nuclear Medicine, Division of Life Sciences and Medicine, The First Affiliated Hospital of USTC, University of Science and Technology of China, Hefei, Anhui 230001, China
| | - Ze-Hua Zhu
- Department of Nuclear Medicine, Division of Life Sciences and Medicine, The First Affiliated Hospital of USTC, University of Science and Technology of China, Hefei, Anhui 230001, China
| | - Feng Gao
- Division of Life Sciences and Medicine, Institute on Aging and Brain Disorders, The First Affiliated Hospital of USTC, University of Science and Technology of China, Hefei, Anhui 230001, China
| | - Lin-Bin Dai
- Division of Life Sciences and Medicine, Institute on Aging and Brain Disorders, The First Affiliated Hospital of USTC, University of Science and Technology of China, Hefei, Anhui 230001, China
| | - Xin-Yi Lv
- Department of Neurology, Division of Life Sciences and Medicine, The First Affiliated Hospital of USTC, University of Science and Technology of China, Hefei, Anhui 230001, China
| | - Qiong Wang
- Department of Neurology, Division of Life Sciences and Medicine, The First Affiliated Hospital of USTC, University of Science and Technology of China, Hefei, Anhui 230001, China
| | - Xing-Xing Zhu
- Department of Nuclear Medicine, Division of Life Sciences and Medicine, The First Affiliated Hospital of USTC, University of Science and Technology of China, Hefei, Anhui 230001, China
| | - Ji-Kui Xie
- Department of Nuclear Medicine, Division of Life Sciences and Medicine, The First Affiliated Hospital of USTC, University of Science and Technology of China, Hefei, Anhui 230001, China
| | - Yong Shen
- Division of Life Sciences and Medicine, Institute on Aging and Brain Disorders, The First Affiliated Hospital of USTC, University of Science and Technology of China, Hefei, Anhui 230001, China.,Neurodegenerative Disorder Research Center, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui 230026, China.,Anhui Province Key Laboratory of Biomedical Aging Research, Hefei, Anhui 230001, China
| | - Shi-Cun Wang
- Department of Nuclear Medicine, Division of Life Sciences and Medicine, The First Affiliated Hospital of USTC, University of Science and Technology of China, Hefei, Anhui 230001, China
| | - Qiang Xie
- Department of Nuclear Medicine, Division of Life Sciences and Medicine, The First Affiliated Hospital of USTC, University of Science and Technology of China, Hefei, Anhui 230001, China
| | | |
Collapse
|
50
|
Cheng Y, Gao Y, Li J, Rui T, Li Q, Chen H, Jia B, Song Y, Gu Z, Wang T, Gao C, Wang Y, Wang Z, Wang F, Tao L, Luo C. TrkB agonist N-acetyl serotonin promotes functional recovery after traumatic brain injury by suppressing ferroptosis via the PI3K/Akt/Nrf2/Ferritin H pathway. Free Radic Biol Med 2023; 194:184-198. [PMID: 36493983 DOI: 10.1016/j.freeradbiomed.2022.12.002] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/16/2022] [Revised: 11/19/2022] [Accepted: 12/02/2022] [Indexed: 12/12/2022]
Abstract
Ferroptosis is a form of regulated cell death that is mainly triggered by iron-dependent lipid peroxidation. A growing body of evidence suggests that ferroptosis is involved in the pathophysiology of traumatic brain injury (TBI), and tropomyosin-related kinase B (TrkB) deficiency would mediate TBI pathologies. As an agonist of TrkB and an immediate precursor of melatonin, N-acetyl serotonin (NAS) exerts several beneficial effects on TBI, but there is no information regarding the role of NAS in ferroptosis after TBI. Here, we examined the effect of NAS treatment on TBI-induced functional outcomes and ferroptosis. Remarkably, the administration of NAS alleviated TBI-induced neurobehavioral deficits, lesion volume, and neurodegeneration. NAS also rescued TBI-induced mitochondrial shrinkage, the changes in ferroptosis-related molecule expression, and iron accumulation in the ipsilateral cortex. Similar results were obtained with a well-established ferroptosis inhibitor, liproxstatin-1. Furthermore, NAS activated the TrkB/PI3K/Akt/Nrf2 pathway in the mouse model of TBI, while inhibition of PI3K and Nrf2 weakened the protection of NAS against ferroptosis both in vitro and in vivo, suggesting that a possible pathway linking NAS to the action of anti-ferroptosis was TrkB/PI3K/Akt/Nrf2. Given that ferritin H (Fth) is a known transcription target of Nrf2, we then investigated the effects of NAS on neuron-specific Fth knockout (Fth-KO) mice. Strikingly, Fth deletion almost abolished the protective effects of NAS against TBI-induced ferroptosis and synaptic damage, although Fth deletion-induced susceptibility toward ferroptosis after TBI was reversed by an iron chelator, deferoxamine. Taken together, these data indicate that the TrkB agonist NAS treatment appears to improve brain function after TBI by suppressing ferroptosis, at least in part, through activation of the PI3K/Akt/Nrf2/Fth pathway, providing evidence that NAS is likely to be a promising anti-ferroptosis agent for further treatment for TBI.
Collapse
Affiliation(s)
- Ying Cheng
- Department of Forensic Medicine, School of Basic Medicine and Biological Sciences, Soochow University, Suzhou, 215123, China
| | - Yuan Gao
- Department of Forensic Medicine, School of Basic Medicine and Biological Sciences, Soochow University, Suzhou, 215123, China
| | - Jing Li
- Department of Forensic Medicine, School of Basic Medicine and Biological Sciences, Soochow University, Suzhou, 215123, China
| | - Tongyu Rui
- Department of Forensic Medicine, School of Basic Medicine and Biological Sciences, Soochow University, Suzhou, 215123, China
| | - Qianqian Li
- School of Forensic Medicine, Wannan Medical College, Wuhu, 241002, China
| | - Huan Chen
- Department of Forensic Medicine, School of Basic Medicine and Biological Sciences, Soochow University, Suzhou, 215123, China
| | - Bowen Jia
- Department of Forensic Medicine, School of Basic Medicine and Biological Sciences, Soochow University, Suzhou, 215123, China
| | - Yiting Song
- Department of Forensic Medicine, School of Basic Medicine and Biological Sciences, Soochow University, Suzhou, 215123, China
| | - Zhiya Gu
- Department of Forensic Medicine, School of Basic Medicine and Biological Sciences, Soochow University, Suzhou, 215123, China
| | - Tao Wang
- Department of Forensic Medicine, School of Basic Medicine and Biological Sciences, Soochow University, Suzhou, 215123, China
| | - Cheng Gao
- Department of Forensic Medicine, School of Basic Medicine and Biological Sciences, Soochow University, Suzhou, 215123, China
| | - Ying Wang
- Department of Forensic Medicine, School of Basic Medicine and Biological Sciences, Soochow University, Suzhou, 215123, China
| | - Zufeng Wang
- Department of Forensic Medicine, School of Basic Medicine and Biological Sciences, Soochow University, Suzhou, 215123, China
| | - Fudi Wang
- The First Affiliated Hospital, School of Public Health, Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou, 310058, China.
| | - Luyang Tao
- Department of Forensic Medicine, School of Basic Medicine and Biological Sciences, Soochow University, Suzhou, 215123, China.
| | - Chengliang Luo
- Department of Forensic Medicine, School of Basic Medicine and Biological Sciences, Soochow University, Suzhou, 215123, China.
| |
Collapse
|