1
|
Sweeney N, Kim TY, Morrison CT, Li L, Acosta D, Liang J, Datla NV, Fitzgerald JA, Huang H, Liu X, Tan GH, Wu M, Karelina K, Bray CE, Weil ZM, Scharre DW, Serrano GE, Saito T, Saido TC, Beach TG, Kokiko-Cochran ON, Godbout JP, Johnson GVW, Fu H. Neuronal BAG3 attenuates tau hyperphosphorylation, synaptic dysfunction, and cognitive deficits induced by traumatic brain injury via the regulation of autophagy-lysosome pathway. Acta Neuropathol 2024; 148:52. [PMID: 39394356 PMCID: PMC11469979 DOI: 10.1007/s00401-024-02810-1] [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: 04/16/2024] [Revised: 09/25/2024] [Accepted: 09/25/2024] [Indexed: 10/13/2024]
Abstract
Growing evidence supports that early- or middle-life traumatic brain injury (TBI) is a risk factor for developing Alzheimer's disease (AD) and AD-related dementia (ADRD). Nevertheless, the molecular mechanisms underlying TBI-induced AD-like pathology and cognitive deficits remain unclear. In this study, we found that a single TBI (induced by controlled cortical impact) reduced the expression of BCL2-associated athanogene 3 (BAG3) in neurons and oligodendrocytes, which is associated with decreased proteins related to the autophagy-lysosome pathway (ALP) and increased hyperphosphorylated tau (ptau) accumulation in excitatory neurons and oligodendrocytes, gliosis, synaptic dysfunction, and cognitive deficits in wild-type (WT) and human tau knock-in (hTKI) mice. These pathological changes were also found in human cases with a TBI history and exaggerated in human AD cases with TBI. The knockdown of BAG3 significantly inhibited autophagic flux, while overexpression of BAG3 significantly increased it in vitro. Specific overexpression of neuronal BAG3 in the hippocampus attenuated AD-like pathology and cognitive deficits induced by TBI in hTKI mice, which is associated with increased ALP-related proteins. Our data suggest that targeting neuronal BAG3 may be a therapeutic strategy for preventing or reducing AD-like pathology and cognitive deficits induced by TBI.
Collapse
Affiliation(s)
- Nicholas Sweeney
- Department of Neuroscience, College of Medicine, Ohio State University, Columbus, OH, USA
| | - Tae Yeon Kim
- Department of Neuroscience, College of Medicine, Ohio State University, Columbus, OH, USA
- Biomedical Sciences Graduate Program, College of Medicine, Ohio State University, Columbus, OH, USA
| | - Cody T Morrison
- Department of Neuroscience, College of Medicine, Ohio State University, Columbus, OH, USA
| | - Liangping Li
- Department of Neuroscience, College of Medicine, Ohio State University, Columbus, OH, USA
| | - Diana Acosta
- Department of Neuroscience, College of Medicine, Ohio State University, Columbus, OH, USA
| | - Jiawen Liang
- Department of Neuroscience, College of Medicine, Ohio State University, Columbus, OH, USA
| | - Nithin V Datla
- Department of Neuroscience, College of Medicine, Ohio State University, Columbus, OH, USA
| | - Julie A Fitzgerald
- Department of Neuroscience, College of Medicine, Ohio State University, Columbus, OH, USA
| | - Haoran Huang
- Medical Scientist Training Program, College of Medicine, Ohio State University, Columbus, OH, USA
| | - Xianglan Liu
- Department of Neuroscience, College of Medicine, Ohio State University, Columbus, OH, USA
| | - Gregory Huang Tan
- Department of Neuroscience, College of Medicine, Ohio State University, Columbus, OH, USA
| | - Min Wu
- Department of Neuroscience, College of Medicine, Ohio State University, Columbus, OH, USA
| | - Kate Karelina
- Department of Neuroscience, Rockefeller Neuroscience Institute, West Virginia University, Morgantown, WV, USA
| | - Chelsea E Bray
- Department of Neuroscience, College of Medicine, Ohio State University, Columbus, OH, USA
| | - Zachary M Weil
- Department of Neuroscience, Rockefeller Neuroscience Institute, West Virginia University, Morgantown, WV, USA
| | - Douglas W Scharre
- Department of Neurology, College of Medicine, Ohio State University, Columbus, OH, USA
| | | | - Takashi Saito
- RIKEN Center for Brain Science, Laboratory for Proteolytic Neuroscience, Saitama, 351-0198, Japan
- Department of Neurocognitive Science, Institute of Brain Science, Nagoya City University Graduate School of Medical Sciences, Nagoya, 467-8601, Japan
| | - Takaomi C Saido
- RIKEN Center for Brain Science, Laboratory for Proteolytic Neuroscience, Saitama, 351-0198, Japan
| | | | - Olga N Kokiko-Cochran
- Department of Neuroscience, College of Medicine, Ohio State University, Columbus, OH, USA
- Chronic Brain Injury Program, The Ohio State University, 175 Pomerene Hall, 1760 Neil Ave, Columbus, OH, USA
| | - Jonathan P Godbout
- Department of Neuroscience, College of Medicine, Ohio State University, Columbus, OH, USA
- Chronic Brain Injury Program, The Ohio State University, 175 Pomerene Hall, 1760 Neil Ave, Columbus, OH, USA
| | - Gail V W Johnson
- Department of Anesthesiology, University of Rochester, Rochester, NY, USA
| | - Hongjun Fu
- Department of Neuroscience, College of Medicine, Ohio State University, Columbus, OH, USA.
- Chronic Brain Injury Program, The Ohio State University, 175 Pomerene Hall, 1760 Neil Ave, Columbus, OH, USA.
| |
Collapse
|
2
|
Siciliano AM, Moro F, De Simone G, Pischiutta F, Morabito A, Pastorelli R, Brunelli L, Zanier ER, Davoli E. Mapping small metabolite changes after traumatic brain injury using AP-MALDI MSI. Anal Bioanal Chem 2024; 416:4941-4949. [PMID: 39090264 PMCID: PMC11330407 DOI: 10.1007/s00216-024-05422-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2024] [Revised: 06/26/2024] [Accepted: 06/27/2024] [Indexed: 08/04/2024]
Abstract
Traumatic brain injury (TBI) is an alteration of brain function caused by a sudden transmission of an external force to the head. The biomechanical impact induces acute and chronic metabolic changes that highly contribute to injury evolution and outcome. TBI heterogeneity calls for approaches allowing the mapping of regional molecular and metabolic changes underpinning disease progression, with mass spectrometry imaging (MSI) as an efficient tool to study the spatial distribution of small metabolites. In this study, we applied an innovative targeted atmospheric pressure-MALDI mass spectrometry imaging (AP-MALDI MSI) approach, starting from an extensive list of metabolites, representative of different metabolic pathways, individually validated on the tissue under analysis with original standards using 2,5-dihydroxybenzoic acid (DHB), to characterize the impact of TBI on regional changes to small metabolites in the brain. Brains from sham and TBI mice obtained 21 days post-injury were analyzed to examine the spatial metabolic profile of small metabolites belonging to different metabolic pathways. By a whole brain analysis, we identified four metabolites (alanine, lysine, histidine, and inosine) with higher abundance in TBI than sham mice. Within the TBI group, lysine, histidine, and inosine were higher in the hemisphere ipsilateral to the biomechanical impact vs. the contralateral one. Images showed a major involvement of the ipsilateral thalamus characterized by the increase of arginine, lysine, histidine, and inosine and a significant reduction of glutamic acid, and N-acetylaspartic acid compared to the contralateral thalamus. These findings indicate high-resolution imaging mass spectrometry as a powerful tool to identify region-specific changes after a TBI to understand the metabolic changes underlying brain injury evolution.
Collapse
Affiliation(s)
- Angela Marika Siciliano
- Mass Spectrometry Research Centre for Health and Environment and Laboratory of Mass Spectrometry, Environmental Health Sciences Department, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, 20156, Milan, Italy
| | - Federico Moro
- Laboratory of Traumatic Brain Injury and Neuroprotection, Department of Acute Brain and Cardiovascular Injury, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy
| | - Giulia De Simone
- Laboratory of Protein and Metabolites in Translational Research, Environmental Health Sciences Department, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, 20156, Milan, Italy
| | - Francesca Pischiutta
- Laboratory of Traumatic Brain Injury and Neuroprotection, Department of Acute Brain and Cardiovascular Injury, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy
| | - Aurelia Morabito
- Laboratory of Protein and Metabolites in Translational Research, Environmental Health Sciences Department, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, 20156, Milan, Italy
- Department of Electronics, Information and Bioengineering, Politecnico di Milano, 20133, Milan, Italy
| | - Roberta Pastorelli
- Laboratory of Protein and Metabolites in Translational Research, Environmental Health Sciences Department, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, 20156, Milan, Italy
| | - Laura Brunelli
- Laboratory of Protein and Metabolites in Translational Research, Environmental Health Sciences Department, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, 20156, Milan, Italy
| | - Elisa R Zanier
- Laboratory of Traumatic Brain Injury and Neuroprotection, Department of Acute Brain and Cardiovascular Injury, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy
| | - Enrico Davoli
- Mass Spectrometry Research Centre for Health and Environment and Laboratory of Mass Spectrometry, Environmental Health Sciences Department, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, 20156, Milan, Italy.
| |
Collapse
|
3
|
Parra Bravo C, Krukowski K, Barker S, Wang C, Li Y, Fan L, Vázquez-Rosa E, Shin MK, Wong MY, McCullough LD, Kitagawa RS, Choi HA, Cacace A, Sinha SC, Pieper AA, Rosi S, Chen X, Gan L. Anti-acetylated-tau immunotherapy is neuroprotective in tauopathy and brain injury. Mol Neurodegener 2024; 19:51. [PMID: 38915105 PMCID: PMC11197196 DOI: 10.1186/s13024-024-00733-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2024] [Accepted: 05/15/2024] [Indexed: 06/26/2024] Open
Abstract
BACKGROUND Tau is aberrantly acetylated in various neurodegenerative conditions, including Alzheimer's disease, frontotemporal lobar degeneration (FTLD), and traumatic brain injury (TBI). Previously, we reported that reducing acetylated tau by pharmacologically inhibiting p300-mediated tau acetylation at lysine 174 reduces tau pathology and improves cognitive function in animal models. METHODS We investigated the therapeutic efficacy of two different antibodies that specifically target acetylated lysine 174 on tau (ac-tauK174). We treated PS19 mice, which harbor the P301S tauopathy mutation that causes FTLD, with anti-ac-tauK174 and measured effects on tau pathology, neurodegeneration, and neurobehavioral outcomes. Furthermore, PS19 mice received treatment post-TBI to evaluate the ability of the immunotherapy to prevent TBI-induced exacerbation of tauopathy phenotypes. Ac-tauK174 measurements in human plasma following TBI were also collected to establish a link between trauma and acetylated tau levels, and single nuclei RNA-sequencing of post-TBI brain tissues from treated mice provided insights into the molecular mechanisms underlying the observed treatment effects. RESULTS Anti-ac-tauK174 treatment mitigates neurobehavioral impairment and reduces tau pathology in PS19 mice. Ac-tauK174 increases significantly in human plasma 24 h after TBI, and anti-ac-tauK174 treatment of PS19 mice blocked TBI-induced neurodegeneration and preserved memory functions. Anti-ac-tauK174 treatment rescues alterations of microglial and oligodendrocyte transcriptomic states following TBI in PS19 mice. CONCLUSIONS The ability of anti-ac-tauK174 treatment to rescue neurobehavioral impairment, reduce tau pathology, and rescue glial responses demonstrates that targeting tau acetylation at K174 is a promising neuroprotective therapeutic approach to human tauopathies resulting from TBI or genetic disease.
Collapse
Affiliation(s)
- Celeste Parra Bravo
- Brain and Mind Research Institute, Helen and Appel Alzheimer Disease Research Institute, Weill Cornell Medicine, New York, NY, USA
- Weill Cornell Graduate School of Medical Sciences, Weill Cornell Medicine, New York, NY, USA
| | - Karen Krukowski
- Department of Physical Therapy & Rehabilitation Science, Department of Neurological Surgery, University of California, San Francisco, San Francisco, CA, USA
| | - Sarah Barker
- Brain Health Medicines Center, Harrington Discovery Institute, University Hospitals Cleveland Medical Center, Cleveland, OH, USA
- Institute for Transformative Molecular Medicine, School of Medicine, Case Western Reserve University, Cleveland, OH, USA
- Department of Psychiatry, Case Western Reserve University, Cleveland, OH, USA
- Geriatric Psychiatry, GRECC, Louis Stokes VA Medical Center, Cleveland, OH, USA
- Department of Pathology, Case Western Reserve University, Cleveland, OH, USA
| | - Chao Wang
- Gladstone Institute of Neurological Disease, San Francisco, CA, USA
| | - Yaqiao Li
- Gladstone Institute of Neurological Disease, San Francisco, CA, USA
| | - Li Fan
- Brain and Mind Research Institute, Helen and Appel Alzheimer Disease Research Institute, Weill Cornell Medicine, New York, NY, USA
| | - Edwin Vázquez-Rosa
- Brain Health Medicines Center, Harrington Discovery Institute, University Hospitals Cleveland Medical Center, Cleveland, OH, USA
- Institute for Transformative Molecular Medicine, School of Medicine, Case Western Reserve University, Cleveland, OH, USA
- Department of Psychiatry, Case Western Reserve University, Cleveland, OH, USA
- Geriatric Psychiatry, GRECC, Louis Stokes VA Medical Center, Cleveland, OH, USA
| | - Min-Kyoo Shin
- Brain Health Medicines Center, Harrington Discovery Institute, University Hospitals Cleveland Medical Center, Cleveland, OH, USA
- Institute for Transformative Molecular Medicine, School of Medicine, Case Western Reserve University, Cleveland, OH, USA
- Department of Psychiatry, Case Western Reserve University, Cleveland, OH, USA
- Geriatric Psychiatry, GRECC, Louis Stokes VA Medical Center, Cleveland, OH, USA
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul, Republic of Korea
| | - Man Ying Wong
- Brain and Mind Research Institute, Helen and Appel Alzheimer Disease Research Institute, Weill Cornell Medicine, New York, NY, USA
| | - Louise D McCullough
- Department of Neurology, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Ryan S Kitagawa
- Department of Neurosurgery, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - H Alex Choi
- Department of Neurosurgery, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | | | - Subhash C Sinha
- Brain and Mind Research Institute, Helen and Appel Alzheimer Disease Research Institute, Weill Cornell Medicine, New York, NY, USA
| | - Andrew A Pieper
- Brain Health Medicines Center, Harrington Discovery Institute, University Hospitals Cleveland Medical Center, Cleveland, OH, USA
- Institute for Transformative Molecular Medicine, School of Medicine, Case Western Reserve University, Cleveland, OH, USA
- Department of Psychiatry, Case Western Reserve University, Cleveland, OH, USA
- Geriatric Psychiatry, GRECC, Louis Stokes VA Medical Center, Cleveland, OH, USA
- Department of Pathology, Case Western Reserve University, Cleveland, OH, USA
- Department of Neurosciences, Case Western Reserve University, Cleveland, OH, USA
| | - Susanna Rosi
- Department of Physical Therapy & Rehabilitation Science, Department of Neurological Surgery, University of California, San Francisco, San Francisco, CA, USA.
- Weill Institute for Neuroscience, University of California San Francisco, San Francisco, CA, USA.
- Department of Neurological Surgery, University of California San Francisco, San Francisco, CA, USA.
| | - Xu Chen
- Gladstone Institute of Neurological Disease, San Francisco, CA, USA.
- Department of Neurosciences, School of Medicine, University of California, San Diego, USA.
| | - Li Gan
- Brain and Mind Research Institute, Helen and Appel Alzheimer Disease Research Institute, Weill Cornell Medicine, New York, NY, USA.
- Weill Cornell Graduate School of Medical Sciences, Weill Cornell Medicine, New York, NY, USA.
| |
Collapse
|
4
|
Barbas H, Garcia-Cabezas MA, John Y, Bautista J, McKee A, Zikopoulos B. Cortical circuit principles predict patterns of trauma induced tauopathy in humans. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.02.592271. [PMID: 38746103 PMCID: PMC11092596 DOI: 10.1101/2024.05.02.592271] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/16/2024]
Abstract
Connections in the cortex of diverse mammalian species are predicted reliably by the Structural Model for direction of pathways and signal processing (reviewed in 1,2). The model is rooted in the universal principle of cortical systematic variation in laminar structure and has been supported widely for connection patterns in animals but has not yet been tested for humans. Here, in postmortem brains of individuals neuropathologically diagnosed with chronic traumatic encephalopathy (CTE) we studied whether the hyperphosphorylated tau (p-tau) pathology parallels connection sequence in time by circuit mechanisms. CTE is a progressive p-tau pathology that begins focally in perivascular sites in sulcal depths of the neocortex (stages I-II) and later involves the medial temporal lobe (MTL) in stages III-IV. We provide novel quantitative evidence that the p-tau pathology in MTL A28 and nearby sites in CTE stage III closely follows the graded laminar patterns seen in homologous cortico-cortical connections in non-human primates. The Structural Model successfully predicted the laminar distribution of the p-tau neurofibrillary tangles and neurites and their density, based on the relative laminar (dis)similarity between the cortical origin (seed) and each connection site. The findings were validated for generalizability by a computational progression model. By contrast, the early focal perivascular pathology in the sulcal depths followed local columnar connectivity rules. These findings support the general applicability of a theoretical model to unravel the direction and progression of p-tau pathology in human neurodegeneration via a cortico-cortical mechanism. Cortical pathways converging on medial MTL help explain the progressive spread of p-tau pathology from focal cortical sites in early CTE to widespread lateral MTL areas and beyond in later disease stages.
Collapse
Affiliation(s)
- Helen Barbas
- Neural Systems Laboratory, Department of Health Sciences, Boston University, Boston, MA 022152
- Graduate Program in Neuroscience, Boston Univ. and School of Medicine
- Department of Anatomy and Neurobiology, Boston University School of Medicine, Boston, MA
- Center for Systems Neuroscience, Boston University, Boston, MA
| | - Miguel Angel Garcia-Cabezas
- Departamento de Anatomía, Histología y Neurociencia, Facultad de Medicina, Universidad Autónoma de Madrid, Madrid, Spain
| | - Yohan John
- Neural Systems Laboratory, Department of Health Sciences, Boston University, Boston, MA 022152
| | - Julied Bautista
- Neural Systems Laboratory, Department of Health Sciences, Boston University, Boston, MA 022152
| | - Ann McKee
- Veterans Affairs (VA) Boston Healthcare System, US Department of Veteran Affairs, Boston, Massachusetts
- Alzheimer’s Disease Research Center and Chronic Traumatic Encephalopathy Center, Chobanian and Avedisian School of Medicine, Boston University, Boston, Massachusetts
| | - Basilis Zikopoulos
- Graduate Program in Neuroscience, Boston Univ. and School of Medicine
- Department of Anatomy and Neurobiology, Boston University School of Medicine, Boston, MA
- Center for Systems Neuroscience, Boston University, Boston, MA
- Human Systems Neuroscience Laboratory, Department of Health Sciences, Boston University
| |
Collapse
|
5
|
Fesharaki-Zadeh A, Datta D. An overview of preclinical models of traumatic brain injury (TBI): relevance to pathophysiological mechanisms. Front Cell Neurosci 2024; 18:1371213. [PMID: 38682091 PMCID: PMC11045909 DOI: 10.3389/fncel.2024.1371213] [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: 01/16/2024] [Accepted: 03/20/2024] [Indexed: 05/01/2024] Open
Abstract
Background Traumatic brain injury (TBI) is a major cause of morbidity and mortality, affecting millions annually worldwide. Although the majority of TBI patients return to premorbid baseline, a subset of patient can develop persistent and often debilitating neurocognitive and behavioral changes. The etiology of TBI within the clinical setting is inherently heterogenous, ranging from sport related injuries, fall related injuries and motor vehicle accidents in the civilian setting, to blast injuries in the military setting. Objective Animal models of TBI, offer the distinct advantage of controlling for injury modality, duration and severity. Furthermore, preclinical models of TBI have provided the necessary temporal opportunity to study the chronic neuropathological sequelae of TBI, including neurodegenerative sequelae such as tauopathy and neuroinflammation within the finite experimental timeline. Despite the high prevalence of TBI, there are currently no disease modifying regimen for TBI, and the current clinical treatments remain largely symptom based. The preclinical models have provided the necessary biological substrate to examine the disease modifying effect of various pharmacological agents and have imperative translational value. Methods The current review will include a comprehensive survey of well-established preclinical models, including classic preclinical models including weight drop, blast injury, fluid percussion injury, controlled cortical impact injury, as well as more novel injury models including closed-head impact model of engineered rotational acceleration (CHIMERA) models and closed-head projectile concussive impact model (PCI). In addition to rodent preclinical models, the review will include an overview of other species including large animal models and Drosophila. Results There are major neuropathological perturbations post TBI captured in various preclinical models, which include neuroinflammation, calcium dysregulation, tauopathy, mitochondrial dysfunction and oxidative stress, axonopathy, as well as glymphatic system disruption. Conclusion The preclinical models of TBI continue to offer valuable translational insight, as well as essential neurobiological basis to examine specific disease modifying therapeutic regimen.
Collapse
Affiliation(s)
- Arman Fesharaki-Zadeh
- Department of Neurology and Psychiatry, Yale University School of Medicine, New Haven, CT, United States
| | - Dibyadeep Datta
- Division of Aging and Geriatric Psychiatry, Alzheimer’s Disease Research Unit, Department of Psychiatry, New Haven, CT, United States
| |
Collapse
|
6
|
Katchur NJ, Notterman DA. Recent insights from non-mammalian models of brain injuries: an emerging literature. Front Neurol 2024; 15:1378620. [PMID: 38566857 PMCID: PMC10985199 DOI: 10.3389/fneur.2024.1378620] [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: 01/29/2024] [Accepted: 02/26/2024] [Indexed: 04/04/2024] Open
Abstract
Traumatic brain injury (TBI) is a major global health concern and is increasingly recognized as a risk factor for neurodegenerative diseases including Alzheimer's disease (AD) and chronic traumatic encephalopathy (CTE). Repetitive TBIs (rTBIs), commonly observed in contact sports, military service, and intimate partner violence (IPV), pose a significant risk for long-term sequelae. To study the long-term consequences of TBI and rTBI, researchers have typically used mammalian models to recapitulate brain injury and neurodegenerative phenotypes. However, there are several limitations to these models, including: (1) lengthy observation periods, (2) high cost, (3) difficult genetic manipulations, and (4) ethical concerns regarding prolonged and repeated injury of a large number of mammals. Aquatic vertebrate model organisms, including Petromyzon marinus (sea lampreys), zebrafish (Danio rerio), and invertebrates, Caenorhabditis elegans (C. elegans), and Drosophila melanogaster (Drosophila), are emerging as valuable tools for investigating the mechanisms of rTBI and tauopathy. These non-mammalian models offer unique advantages, including genetic tractability, simpler nervous systems, cost-effectiveness, and quick discovery-based approaches and high-throughput screens for therapeutics, which facilitate the study of rTBI-induced neurodegeneration and tau-related pathology. Here, we explore the use of non-vertebrate and aquatic vertebrate models to study TBI and neurodegeneration. Drosophila, in particular, provides an opportunity to explore the longitudinal effects of mild rTBI and its impact on endogenous tau, thereby offering valuable insights into the complex interplay between rTBI, tauopathy, and neurodegeneration. These models provide a platform for mechanistic studies and therapeutic interventions, ultimately advancing our understanding of the long-term consequences associated with rTBI and potential avenues for intervention.
Collapse
Affiliation(s)
- Nicole J. Katchur
- Department of Molecular Biology, Princeton University, Princeton, NJ, United States
- Rutgers-Robert Wood Johnson Medical School, Piscataway, NJ, United States
| | - Daniel A. Notterman
- Department of Molecular Biology, Princeton University, Princeton, NJ, United States
| |
Collapse
|
7
|
Grandizoli Saletti P, Casillas-Espinosa PM, Panagiotis Lisgaras C, Bi Mowrey W, Li Q, Liu W, Brady RD, Ali I, Silva J, Yamakawa G, Hudson M, Li C, Braine EL, Coles L, Cloyd JC, Jones NC, Shultz SR, Moshé SL, O'Brien TJ, Galanopoulou AS. Tau Phosphorylation Patterns in the Rat Cerebral Cortex After Traumatic Brain Injury and Sodium Selenate Effects: An Epibios4rx Project 2 Study. J Neurotrauma 2024; 41:222-243. [PMID: 36950806 PMCID: PMC11079442 DOI: 10.1089/neu.2022.0219] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/24/2023] Open
Abstract
Sodium selenate (SS) activates protein phosphatase 2 (PP2A) and reduces phosphorylated tau (pTAU) and late post-traumatic seizures after lateral fluid percussion injury (LFPI). In EpiBioS4Rx Project 2, a multi-center international study for post-traumatic targets, biomarkers, and treatments, we tested the target relevance and modification by SS of pTAU forms and PP2A and in the LFPI model, at two sites: Einstein and Melbourne. In Experiment 1, adult male rats were assigned to LFPI and sham (both sites) and naïve controls (Einstein). Motor function was monitored by neuroscores. Brains were studied with immunohistochemistry (IHC), Western blots (WBs), or PP2A activity assay, from 2 days to 8 weeks post-operatively. In Experiment 2, LFPI rats received SS for 7 days (SS0.33: 0.33 mg/kg/day; SS1: 1 mg/kg/day, subcutaneously) or vehicle (Veh) post-LFPI and pTAU, PR55 expression, or PP2A activity were studied at 2 days and 1 week (on treatment), or 2 weeks (1 week off treatment). Plasma selenium and SS levels were measured. In Experiment 1 IHC, LFPI rats had higher cortical pTAU-Ser202/Thr205-immunoreactivity (AT8-ir) and pTAU-Ser199/202-ir at 2 days, and pTAU-Thr231-ir (AT180-ir) at 2 days, 2 weeks, and 8 weeks, ipsilaterally to LFPI, than controls. LFPI-2d rats also had higher AT8/total-TAU5-ir in cortical extracts ipsilateral to the lesion (WB). PP2A (PR55-ir) showed time- and region-dependent changes in IHC, but not in WB. PP2A activity was lower in LFPI-1wk than in sham rats. In Experiment 2, SS did not affect neuroscores or cellular AT8-ir, AT180-ir, or PR55-ir in IHC. In WB, total cortical AT8/total-TAU-ir was lower in SS0.33 and SS1 LFPI rats than in Veh rats (2 days, 1 week); total cortical PR55-ir (WB) and PP2A activity were higher in SS1 than Veh rats (2 days). SS dose dependently increased plasma selenium and SS levels. Concordant across-sites data confirm time and pTAU form-specific cortical increases ipsilateral to LFPI. The discordant SS effects may either suggest SS-induced reduction in the numbers of cells with increased pTAU-ir, need for longer treatment, or the involvement of other mechanisms of action.
Collapse
Affiliation(s)
- Patricia Grandizoli Saletti
- Saul R. Korey Department of Neurology, Laboratory of Developmental Epilepsy, Albert Einstein College of Medicine, Bronx New York, USA
| | - Pablo M. Casillas-Espinosa
- Department of Neuroscience, Monash University, Melbourne, Australia
- Department of Medicine, The University of Melbourne, Parkville, Australia
- Department of Neurology, Alfred Health, Melbourne, Australia
| | - Christos Panagiotis Lisgaras
- Saul R. Korey Department of Neurology, Laboratory of Developmental Epilepsy, Albert Einstein College of Medicine, Bronx New York, USA
| | - Wenzhu Bi Mowrey
- Department of Epidemiology and Population Health, Albert Einstein College of Medicine, Bronx New York, USA
| | - Qianyun Li
- Saul R. Korey Department of Neurology, Laboratory of Developmental Epilepsy, Albert Einstein College of Medicine, Bronx New York, USA
| | - Wei Liu
- Saul R. Korey Department of Neurology, Laboratory of Developmental Epilepsy, Albert Einstein College of Medicine, Bronx New York, USA
| | - Rhys D. Brady
- Department of Neuroscience, Monash University, Melbourne, Australia
- Department of Medicine, The University of Melbourne, Parkville, Australia
| | - Idrish Ali
- Department of Neuroscience, Monash University, Melbourne, Australia
- Department of Medicine, The University of Melbourne, Parkville, Australia
| | - Juliana Silva
- Department of Neuroscience, Monash University, Melbourne, Australia
| | - Glenn Yamakawa
- Department of Medicine, The University of Melbourne, Parkville, Australia
| | - Matt Hudson
- Department of Neuroscience, Monash University, Melbourne, Australia
- Department of Medicine, The University of Melbourne, Parkville, Australia
| | - Crystal Li
- Department of Neuroscience, Monash University, Melbourne, Australia
| | - Emma L. Braine
- Department of Neuroscience, Monash University, Melbourne, Australia
- Department of Medicine, The University of Melbourne, Parkville, Australia
| | - Lisa Coles
- University of Minnesota Twin Cities, Minneapolis, Minnesota, USA
| | - James C. Cloyd
- University of Minnesota Twin Cities, Minneapolis, Minnesota, USA
| | - Nigel C. Jones
- Department of Neuroscience, Monash University, Melbourne, Australia
- Department of Medicine, The University of Melbourne, Parkville, Australia
- Department of Neurology, Alfred Health, Melbourne, Australia
| | - Sandy R. Shultz
- Department of Neuroscience, Monash University, Melbourne, Australia
- Department of Medicine, The University of Melbourne, Parkville, Australia
- Department of Neurology, Alfred Health, Melbourne, Australia
| | - Solomon L. Moshé
- Saul R. Korey Department of Neurology, Laboratory of Developmental Epilepsy, Albert Einstein College of Medicine, Bronx New York, USA
- Isabelle Rapin Division of Child Neurology, Albert Einstein College of Medicine, Bronx New York, USA
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, Bronx New York, USA
- Department of Pediatrics, Albert Einstein College of Medicine, Bronx New York, USA
| | - Terence J. O'Brien
- Department of Neuroscience, Monash University, Melbourne, Australia
- Department of Medicine, The University of Melbourne, Parkville, Australia
- Department of Neurology, Alfred Health, Melbourne, Australia
| | - Aristea S. Galanopoulou
- Saul R. Korey Department of Neurology, Laboratory of Developmental Epilepsy, Albert Einstein College of Medicine, Bronx New York, USA
- Isabelle Rapin Division of Child Neurology, Albert Einstein College of Medicine, Bronx New York, USA
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, Bronx New York, USA
| |
Collapse
|
8
|
Yang C, Jiang J, Zhou J, Hitosug M, Wang Z. Traffic safety and public health in China - Past knowledge, current status, and future directions. ACCIDENT; ANALYSIS AND PREVENTION 2023; 192:107272. [PMID: 37683567 DOI: 10.1016/j.aap.2023.107272] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/08/2022] [Revised: 08/08/2023] [Accepted: 08/24/2023] [Indexed: 09/10/2023]
Abstract
Transportation-related harms have developed into a social disease, threatening public safety and health in China. We aimed to increase the global understanding of traffic safety and public health in China from past knowledge, current status, and future directions by collecting, collating, and analyzing the Chinese traffic incidents reported in the published literature. A systematic search of China National Knowledge Infrastructure, Weipu, and published articles referenced in PubMed, Web of Science and ProQuest between January 1, 1988 and April 30, 2023 was performed. China encountered the first recorded traffic accident as early as three thousand years ago in the Shang Dynasty. An increase in vehicle capacity and velocity increased the traffic risks during the transition from rickshaws and livestock to motor vehicles in varying traffic environments. Humans are not only the decisive factor of a large number of vehicles, traffic routes, and environmental variables, but also the victims at the end and starting point of traffic accidents. Injuries (mechanical force, burns) and diseases (traffic-related air pollution, noise) caused by traffic activities not only threaten public health, but also cause risks to safe driving. Analysis of traffic activities and biomarkers promotes the treatment of traffic injuries in ethology and medicine. China prepared for the construction of healthy transportation in the "decade of road safety" toward an estimation of worldwide road traffic injuries in 2030. Improvement of traffic safety concerning public health under the "Outline of the National Comprehensive Three-dimensional Transportation Network Planning" in China will propel the realization of worldwide traffic environmental advancement.
Collapse
Affiliation(s)
- Ce Yang
- State Key Laboratory of Trauma and Chemical Poisoning, Research Institute of Surgery, Research Institute of Traffic Medicine, Daping Hospital, Third Military Medical University, Chongqing 400042, PR China.
| | - Jianxin Jiang
- State Key Laboratory of Trauma and Chemical Poisoning, Research Institute of Surgery, Research Institute of Traffic Medicine, Daping Hospital, Third Military Medical University, Chongqing 400042, PR China
| | - Jihong Zhou
- State Key Laboratory of Trauma and Chemical Poisoning, Research Institute of Surgery, Research Institute of Traffic Medicine, Daping Hospital, Third Military Medical University, Chongqing 400042, PR China
| | - Masahito Hitosug
- Department of Legal Medicine, Shiga University of Medical Science, Otsu, Shiga, Japan
| | - Zhengguo Wang
- State Key Laboratory of Trauma and Chemical Poisoning, Research Institute of Surgery, Research Institute of Traffic Medicine, Daping Hospital, Third Military Medical University, Chongqing 400042, PR China; International Traffic Medicine Association, Bloomfield Hills, MI, USA.
| |
Collapse
|
9
|
Brennan DJ, Duda J, Ware JB, Whyte J, Choi JY, Gugger J, Focht K, Walter AE, Bushnik T, Gee JC, Diaz‐Arrastia R, Kim JJ. Spatiotemporal profile of atrophy in the first year following moderate-severe traumatic brain injury. Hum Brain Mapp 2023; 44:4692-4709. [PMID: 37399336 PMCID: PMC10400790 DOI: 10.1002/hbm.26410] [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: 12/12/2022] [Revised: 06/04/2023] [Accepted: 06/12/2023] [Indexed: 07/05/2023] Open
Abstract
Traumatic brain injury (TBI) triggers progressive neurodegeneration resulting in brain atrophy that continues months-to-years following injury. However, a comprehensive characterization of the spatial and temporal evolution of TBI-related brain atrophy remains incomplete. Utilizing a sensitive and unbiased morphometry analysis pipeline optimized for detecting longitudinal changes, we analyzed a sample consisting of 37 individuals with moderate-severe TBI who had primarily high-velocity and high-impact injury mechanisms. They were scanned up to three times during the first year after injury (3 months, 6 months, and 12 months post-injury) and compared with 33 demographically matched controls who were scanned once. Individuals with TBI already showed cortical thinning in frontal and temporal regions and reduced volume in the bilateral thalami at 3 months post-injury. Longitudinally, only a subset of cortical regions in the parietal and occipital lobes showed continued atrophy from 3 to 12 months post-injury. Additionally, cortical white matter volume and nearly all deep gray matter structures exhibited progressive atrophy over this period. Finally, we found that disproportionate atrophy of cortex along sulci relative to gyri, an emerging morphometric marker of chronic TBI, was present as early as 3 month post-injury. In parallel, neurocognitive functioning largely recovered during this period despite this pervasive atrophy. Our findings demonstrate msTBI results in characteristic progressive neurodegeneration patterns that are divergent across regions and scale with the severity of injury. Future clinical research using atrophy during the first year of TBI as a biomarker of neurodegeneration should consider the spatiotemporal profile of atrophy described in this study.
Collapse
Affiliation(s)
- Daniel J. Brennan
- CUNY Neuroscience Collaborative, The Graduate CenterCity University of New YorkNew YorkNew YorkUnited States
- Department of Molecular, Cellular, and Biomedical SciencesCUNY School of Medicine, The City College of New YorkNew YorkNew YorkUnited States
| | - Jeffrey Duda
- Department of RadiologyUniversity of Pennsylvania Perelman School of MedicinePhiladelphiaPennsylvaniaUnited States
- Penn Image Computing and Science LaboratoryUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUnited States
| | - Jeffrey B. Ware
- Department of RadiologyUniversity of Pennsylvania Perelman School of MedicinePhiladelphiaPennsylvaniaUnited States
| | - John Whyte
- Moss Rehabilitation Research Institute, Einstein Healthcare NetworkElkins ParkPennsylvaniaUnited States
| | - Joon Yul Choi
- Department of Molecular, Cellular, and Biomedical SciencesCUNY School of Medicine, The City College of New YorkNew YorkNew YorkUnited States
- Department of Biomedical EngineeringYonsei UniversityWonjuRepublic of Korea
| | - James Gugger
- Department of NeurologyUniversity of Pennsylvania Perelman School of MedicinePhiladelphiaPennsylvaniaUnited States
| | - Kristen Focht
- Widener University School for Graduate Clinical PsychologyChesterPennsylvaniaUnited States
| | - Alexa E. Walter
- Department of NeurologyUniversity of Pennsylvania Perelman School of MedicinePhiladelphiaPennsylvaniaUnited States
| | - Tamara Bushnik
- NYU Grossman School of MedicineNew YorkNew YorkUnited States
| | - James C. Gee
- Department of RadiologyUniversity of Pennsylvania Perelman School of MedicinePhiladelphiaPennsylvaniaUnited States
- Penn Image Computing and Science LaboratoryUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUnited States
| | - Ramon Diaz‐Arrastia
- Department of NeurologyUniversity of Pennsylvania Perelman School of MedicinePhiladelphiaPennsylvaniaUnited States
| | - Junghoon J. Kim
- CUNY Neuroscience Collaborative, The Graduate CenterCity University of New YorkNew YorkNew YorkUnited States
- Department of Molecular, Cellular, and Biomedical SciencesCUNY School of Medicine, The City College of New YorkNew YorkNew YorkUnited States
| |
Collapse
|
10
|
Sparks S, Pinto J, Hayes G, Spitschan M, Bulte DP. The impact of Alzheimer's disease risk factors on the pupillary light response. Front Neurosci 2023; 17:1248640. [PMID: 37650103 PMCID: PMC10463762 DOI: 10.3389/fnins.2023.1248640] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2023] [Accepted: 07/31/2023] [Indexed: 09/01/2023] Open
Abstract
Alzheimer's disease (AD) is the leading cause of dementia, and its prevalence is increasing and is expected to continue to increase over the next few decades. Because of this, there is an urgent requirement to determine a way to diagnose the disease, and to target interventions to delay and ideally stop the onset of symptoms, specifically those impacting cognition and daily livelihood. The pupillary light response (PLR) is controlled by the sympathetic and parasympathetic branches of the autonomic nervous system, and impairments to the pupillary light response (PLR) have been related to AD. However, most of these studies that assess the PLR occur in patients who have already been diagnosed with AD, rather than those who are at a higher risk for the disease but without a diagnosis. Determining whether the PLR is similarly impaired in subjects before an AD diagnosis is made and before cognitive symptoms of the disease begin, is an important step before using the PLR as a diagnostic tool. Specifically, identifying whether the PLR is impaired in specific at-risk groups, considering both genetic and non-genetic risk factors, is imperative. It is possible that the PLR may be impaired in association with some risk factors but not others, potentially indicating different pathways to neurodegeneration that could be distinguished using PLR. In this work, we review the most common genetic and lifestyle-based risk factors for AD and identify established relationships between these risk factors and the PLR. The evidence here shows that many AD risk factors, including traumatic brain injury, ocular and intracranial hypertension, alcohol consumption, depression, and diabetes, are directly related to changes in the PLR. Other risk factors currently lack sufficient literature to make any conclusions relating directly to the PLR but have shown links to impairments in the parasympathetic nervous system; further research should be conducted in these risk factors and their relation to the PLR.
Collapse
Affiliation(s)
- Sierra Sparks
- Department of Engineering Science, Institute of Biomedical Engineering, University of Oxford, Oxford, United Kingdom
| | - Joana Pinto
- Department of Engineering Science, Institute of Biomedical Engineering, University of Oxford, Oxford, United Kingdom
| | - Genevieve Hayes
- Department of Engineering Science, Institute of Biomedical Engineering, University of Oxford, Oxford, United Kingdom
| | - Manuel Spitschan
- TUM Department of Sport and Health Sciences (TUM SG), Chronobiology and Health, Technical University of Munich, Munich, Germany
- TUM Institute for Advanced Study (TUM-IAS), Technical University of Munich, Garching, Germany
- Max Planck Institute for Biological Cybernetics, Translational Sensory and Circadian Neuroscience, Tübingen, Germany
| | - Daniel P. Bulte
- Department of Engineering Science, Institute of Biomedical Engineering, University of Oxford, Oxford, United Kingdom
| |
Collapse
|
11
|
Flavin WP, Hosseini H, Ruberti JW, Kavehpour HP, Giza CC, Prins ML. Traumatic brain injury and the pathways to cerebral tau accumulation. Front Neurol 2023; 14:1239653. [PMID: 37638180 PMCID: PMC10450935 DOI: 10.3389/fneur.2023.1239653] [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: 06/13/2023] [Accepted: 07/24/2023] [Indexed: 08/29/2023] Open
Abstract
Tau is a protein that has received national mainstream recognition for its potential negative impact to the brain. This review succinctly provides information on the structure of tau and its normal physiological functions, including in hibernation and changes throughout the estrus cycle. There are many pathways involved in phosphorylating tau including diabetes, stroke, Alzheimer's disease (AD), brain injury, aging, and drug use. The common mechanisms for these processes are put into context with changes observed in mild and repetitive mild traumatic brain injury (TBI). The phosphorylation of tau is a part of the progression to pathology, but the ability for tau to aggregate and propagate is also addressed. Summarizing both the functional and dysfunctional roles of tau can help advance our understanding of this complex protein, improve our care for individuals with a history of TBI, and lead to development of therapeutic interventions to prevent or reverse tau-mediated neurodegeneration.
Collapse
Affiliation(s)
- William P. Flavin
- Department of Neurology, David Geffen School of Medicine, UCLA, Los Angeles, CA, United States
- Steve Tisch BrainSPORT Program, Department of Pediatrics and Neurosurgery, David Geffen School of Medicine, UCLA, Los Angeles, CA, United States
| | - Helia Hosseini
- Department of Bioengineering, UCLA, Los Angeles, CA, United States
| | - Jeffrey W. Ruberti
- Department of Bioengineering, Northeastern University, Boston, MA, United States
| | - H. Pirouz Kavehpour
- Department of Bioengineering, UCLA, Los Angeles, CA, United States
- Department of Mechanical and Aerospace Engineering, UCLA, Los Angeles, CA, United States
| | - Christopher C. Giza
- Steve Tisch BrainSPORT Program, Department of Pediatrics and Neurosurgery, David Geffen School of Medicine, UCLA, Los Angeles, CA, United States
- Department of Bioengineering, UCLA, Los Angeles, CA, United States
- Department of Neurosurgery, Brain Injury Research Center, David Geffen School of Medicine, UCLA, Los Angeles, CA, United States
| | - Mayumi L. Prins
- Steve Tisch BrainSPORT Program, Department of Pediatrics and Neurosurgery, David Geffen School of Medicine, UCLA, Los Angeles, CA, United States
- Department of Bioengineering, UCLA, Los Angeles, CA, United States
- Department of Neurosurgery, Brain Injury Research Center, David Geffen School of Medicine, UCLA, Los Angeles, CA, United States
| |
Collapse
|
12
|
Graham NS, Cole JH, Bourke NJ, Schott JM, Sharp DJ. Distinct patterns of neurodegeneration after TBI and in Alzheimer's disease. Alzheimers Dement 2023; 19:3065-3077. [PMID: 36696255 PMCID: PMC10955776 DOI: 10.1002/alz.12934] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2022] [Revised: 12/17/2022] [Accepted: 12/20/2022] [Indexed: 01/26/2023]
Abstract
INTRODUCTION Traumatic brain injury (TBI) is a dementia risk factor, with Alzheimer's disease (AD) more common following injury. Patterns of neurodegeneration produced by TBI can be compared to AD and aging using volumetric MRI. METHODS A total of 55 patients after moderate to severe TBI (median age 40), 45 with AD (median age 69), and 61 healthy volunteers underwent magnetic resonance imaging over 2 years. Atrophy patterns were compared. RESULTS AD patients had markedly lower baseline volumes. TBI was associated with increased white matter (WM) atrophy, particularly involving corticospinal tracts and callosum, whereas AD rates were increased across white and gray matter (GM). Subcortical WM loss was shared in AD/TBI, but deep WM atrophy was TBI-specific and cortical atrophy AD-specific. Post-TBI atrophy patterns were distinct from aging, which resembled AD. DISCUSSION Post-traumatic neurodegeneration 1.9-4.0 years (median) following moderate-severe TBI is distinct from aging/AD, predominantly involving central WM. This likely reflects distributions of axonal injury, a neurodegeneration trigger. HIGHLIGHTS We compared patterns of brain atrophy longitudinally after moderate to severe TBI in late-onset AD and healthy aging. Patients after TBI had abnormal brain atrophy involving the corpus callosum and other WM tracts, including corticospinal tracts, in a pattern that was specific and distinct from AD and aging. This pattern is reminiscent of axonal injury following TBI, and atrophy rates were predicted by the extent of axonal injury on diffusion tensor imaging, supporting a relationship between early axonal damage and chronic neurodegeneration.
Collapse
Affiliation(s)
- Neil S.N. Graham
- Department of Brain SciencesImperial College LondonLondonUK
- UK Dementia Research Institute Centre for Care Research and Technology at Imperial College LondonLondonUK
| | - James H. Cole
- Dementia Research CentreUCL Queen Square Institute of NeurologyLondonUK
- Centre for Medical Image ComputingUCLLondonUK
| | - Niall J. Bourke
- Department of Brain SciencesImperial College LondonLondonUK
- UK Dementia Research Institute Centre for Care Research and Technology at Imperial College LondonLondonUK
| | | | - David J. Sharp
- Department of Brain SciencesImperial College LondonLondonUK
- UK Dementia Research Institute Centre for Care Research and Technology at Imperial College LondonLondonUK
- Centre for Injury StudiesImperial College LondonLondonUK
| |
Collapse
|
13
|
Diomede L, Zanier ER, Moro F, Vegliante G, Colombo L, Russo L, Cagnotto A, Natale C, Xodo FM, De Luigi A, Mosconi M, Beeg M, Catania M, Rossi G, Tagliavini F, Di Fede G, Salmona M. Aβ1-6 A2V(D) peptide, effective on Aβ aggregation, inhibits tau misfolding and protects the brain after traumatic brain injury. Mol Psychiatry 2023; 28:2433-2444. [PMID: 37198260 PMCID: PMC10611578 DOI: 10.1038/s41380-023-02101-3] [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: 01/10/2023] [Revised: 04/21/2023] [Accepted: 05/02/2023] [Indexed: 05/19/2023]
Abstract
Alzheimer's disease (AD), the leading cause of dementia in older adults, is a double proteinopathy characterized by amyloid-β (Aβ) and tau pathology. Despite enormous efforts that have been spent in the last decades to find effective therapies, late pharmacological interventions along the course of the disease, inaccurate clinical methodologies in the enrollment of patients, and inadequate biomarkers for evaluating drug efficacy have not allowed the development of an effective therapeutic strategy. The approaches followed so far for developing drugs or antibodies focused solely on targeting Aβ or tau protein. This paper explores the potential therapeutic capacity of an all-D-isomer synthetic peptide limited to the first six amino acids of the N-terminal sequence of the A2V-mutated Aβ, Aβ1-6A2V(D), that was developed following the observation of a clinical case that provided the background for its development. We first performed an in-depth biochemical characterization documenting the capacity of Aβ1-6A2V(D) to interfere with the aggregation and stability of tau protein. To tackle Aβ1-6A2V(D) in vivo effects against a neurological decline in genetically predisposed or acquired high AD risk mice, we tested its effects in triple transgenic animals harboring human PS1(M146 V), APP(SW), and MAPT(P301L) transgenes and aged wild-type mice exposed to experimental traumatic brain injury (TBI), a recognized risk factor for AD. We found that Aβ1-6A2V(D) treatment in TBI mice improved neurological outcomes and reduced blood markers of axonal damage. Exploiting the C. elegans model as a biosensor of amyloidogenic proteins' toxicity, we observed a rescue of locomotor defects in nematodes exposed to the brain homogenates from TBI mice treated with Aβ1-6A2V(D) compared to TBI controls. By this integrated approach, we demonstrate that Aβ1-6A2V(D) not only impedes tau aggregation but also favors its degradation by tissue proteases, confirming that this peptide interferes with both Aβ and tau aggregation propensity and proteotoxicity.
Collapse
Affiliation(s)
- Luisa Diomede
- Department of Molecular Biochemistry and Pharmacology, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Via Mario Negri 2, Milan, Italy.
| | - Elisa R Zanier
- Department of Neuroscience, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Via Mario Negri 2, Milan, Italy
| | - Federico Moro
- Department of Neuroscience, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Via Mario Negri 2, Milan, Italy
| | - Gloria Vegliante
- Department of Neuroscience, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Via Mario Negri 2, Milan, Italy
| | - Laura Colombo
- Department of Molecular Biochemistry and Pharmacology, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Via Mario Negri 2, Milan, Italy
| | - Luca Russo
- Department of Molecular Biochemistry and Pharmacology, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Via Mario Negri 2, Milan, Italy
| | - Alfredo Cagnotto
- Department of Molecular Biochemistry and Pharmacology, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Via Mario Negri 2, Milan, Italy
| | - Carmina Natale
- Department of Molecular Biochemistry and Pharmacology, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Via Mario Negri 2, Milan, Italy
| | - Federica Marta Xodo
- Department of Molecular Biochemistry and Pharmacology, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Via Mario Negri 2, Milan, Italy
| | - Ada De Luigi
- Department of Molecular Biochemistry and Pharmacology, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Via Mario Negri 2, Milan, Italy
| | - Michele Mosconi
- Department of Molecular Biochemistry and Pharmacology, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Via Mario Negri 2, Milan, Italy
| | - Marten Beeg
- Department of Molecular Biochemistry and Pharmacology, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Via Mario Negri 2, Milan, Italy
| | - Marcella Catania
- Neurology V - Neuropathology Unit, Fondazione IRCCS Istituto Neurologico Carlo Besta, Via Celoria 11, Milan, Italy
| | - Giacomina Rossi
- Neurology V - Neuropathology Unit, Fondazione IRCCS Istituto Neurologico Carlo Besta, Via Celoria 11, Milan, Italy
| | - Fabrizio Tagliavini
- Neurology V - Neuropathology Unit, Fondazione IRCCS Istituto Neurologico Carlo Besta, Via Celoria 11, Milan, Italy
| | - Giuseppe Di Fede
- Neurology V - Neuropathology Unit, Fondazione IRCCS Istituto Neurologico Carlo Besta, Via Celoria 11, Milan, Italy
| | - Mario Salmona
- Department of Molecular Biochemistry and Pharmacology, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Via Mario Negri 2, Milan, Italy.
| |
Collapse
|
14
|
Gu W, Bai Y, Cai J, Mi H, Bao Y, Zhao X, Lu C, Zhang F, Li YH, Lu Q. Hypothermia impairs glymphatic drainage in traumatic brain injury as assessed by dynamic contrast-enhanced MRI with intrathecal contrast. Front Neurosci 2023; 17:1061039. [PMID: 36816105 PMCID: PMC9932501 DOI: 10.3389/fnins.2023.1061039] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2022] [Accepted: 01/16/2023] [Indexed: 02/05/2023] Open
Abstract
Introduction The impact of hypothermia on the impaired drainage function of the glymphatic system in traumatic brain injury (TBI) is not understood. Methods Male Sprague-Dawley rats undergoing controlled cortical impact injury (CCI) were subjected to hypothermia or normothermia treatment. The rats undergoing sham surgery without CCI were used as the control. Dynamic contrast-enhanced magnetic resonance imaging (DCE-MRI) with intrathecal administration of low- and high-molecular-weight contrast agents (Gd-DTPA and hyaluronic acid conjugated Gd-DTPA) was performed after TBI and head temperature management. The semiquantitative kinetic parameters characterizing the contrast infusion and cleanout in the brain, including influx rate, efflux rate, and clearance duration, were calculated from the average time-intensity curves. Results and discussion The qualitative and semiquantitative results of DCE-MRI obtained from all examined perivascular spaces and most brain tissue regions showed a significantly increased influx rate and efflux rate and decreased clearance duration among all TBI animals, demonstrating a significant impairment of glymphatic drainage function. This glymphatic drainage dysfunction was exacerbated when additional hypothermia was applied. The early glymphatic drainage reduction induced by TBI and aggravated by hypothermia was linearly related to the late increased deposition of p-tau and beta-amyloid revealed by histopathologic and biochemical analysis and cognitive impairment assessed by the Barnes maze and novel object recognition test. The glymphatic system dysfunction induced by hypothermia may be an indirect alternative pathophysiological factor indicating injury to the brain after TBI. Longitudinal studies and targeted glymphatic dysfunction management are recommended to explore the potential effect of hypothermia in TBI.
Collapse
Affiliation(s)
- Wenquan Gu
- Department of Radiology, Shanghai Punan Hospital of Pudong New Area, Shanghai, China
| | - Yingnan Bai
- Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Jianguo Cai
- Department of Radiology, Xinhua Hospital Chongming Branch, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Honglan Mi
- Department of Radiology, School of Medicine, Renji Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Yinghui Bao
- Department of Neurology, School of Medicine, Renji Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Xinxin Zhao
- Department of Radiology, School of Medicine, Renji Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Chen Lu
- Shanghai Weiyu International School, Shanghai, China
| | - Fengchen Zhang
- Department of Neurology, School of Medicine, Renji Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Yue-hua Li
- Department of Radiology, Shanghai Sixth People’s Hospital, Shanghai Jiao Tong University, Shanghai, China,*Correspondence: Yue-hua Li,
| | - Qing Lu
- School of Medicine, Shanghai East Hospital, Tongji University, Shanghai, China,Qing Lu,
| |
Collapse
|
15
|
Drieu A, Lanquetin A, Prunotto P, Gulhan Z, Pédron S, Vegliante G, Tolomeo D, Serrière S, Vercouillie J, Galineau L, Tauber C, Kuhnast B, Rubio M, Zanier ER, Levard D, Chalon S, Vivien D, Ali C. Persistent neuroinflammation and behavioural deficits after single mild traumatic brain injury. J Cereb Blood Flow Metab 2022; 42:2216-2229. [PMID: 35945692 PMCID: PMC9670002 DOI: 10.1177/0271678x221119288] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/04/2022] [Revised: 06/17/2022] [Accepted: 06/20/2022] [Indexed: 12/14/2022]
Abstract
Despite an apparently silent imaging, some patients with mild traumatic brain injury (TBI) experience cognitive dysfunctions, which may persist chronically. Brain changes responsible for these dysfunctions are unclear and commonly overlooked. It is thus crucial to increase our understanding of the mechanisms linking the initial event to the functional deficits, and to provide objective evidence of brain tissue alterations underpinning these deficits. We first set up a murine model of closed-head controlled cortical impact, which provoked persistent cognitive and sensorimotor deficits, despite no evidence of brain contusion or bleeding on MRI, thus recapitulating features of mild TBI. Molecular MRI for P-selectin, a key adhesion molecule, detected no sign of cerebrovascular inflammation after mild TBI, as confirmed by immunostainings. By contrast, in vivo PET imaging with the TSPO ligand [18F]DPA-714 demonstrated persisting signs of neuroinflammation in the ipsilateral cortex and hippocampus after mild TBI. Interestingly, immunohistochemical analyses confirmed these spatio-temporal profiles, showing a robust parenchymal astrogliosis and microgliosis, at least up to 3 weeks post-injury in both the cortex and hippocampus. In conclusion, we show that even one single mild TBI induces long-term behavioural deficits, associated with a persistent neuro-inflammatory status that can be detected by PET imaging.
Collapse
Affiliation(s)
- Antoine Drieu
- Normandie Univ, UNICAEN, INSERM, INSERM UMR-S U1237,
Physiopathology and Imaging of Neurological Disorders, Institut Blood and Brain
@ Caen-Normandie, Cyceron, France
| | - Anastasia Lanquetin
- Normandie Univ, UNICAEN, INSERM, INSERM UMR-S U1237,
Physiopathology and Imaging of Neurological Disorders, Institut Blood and Brain
@ Caen-Normandie, Cyceron, France
| | - Paul Prunotto
- Normandie Univ, UNICAEN, INSERM, INSERM UMR-S U1237,
Physiopathology and Imaging of Neurological Disorders, Institut Blood and Brain
@ Caen-Normandie, Cyceron, France
| | - Zuhal Gulhan
- UMR 1253, iBrain, Université de Tours, INSERM, Tours,
France
| | - Swannie Pédron
- Normandie Univ, UNICAEN, INSERM, INSERM UMR-S U1237,
Physiopathology and Imaging of Neurological Disorders, Institut Blood and Brain
@ Caen-Normandie, Cyceron, France
| | - Gloria Vegliante
- Department of Neuroscience, Istituto di Ricerche Farmacologiche
Mario Negri, IRCCS, Milan, Italy
| | - Daniele Tolomeo
- Department of Neuroscience, Istituto di Ricerche Farmacologiche
Mario Negri, IRCCS, Milan, Italy
| | - Sophie Serrière
- UMR 1253, iBrain, Université de Tours, INSERM, Tours,
France
| | | | | | - Clovis Tauber
- UMR 1253, iBrain, Université de Tours, INSERM, Tours,
France
| | - Bertrand Kuhnast
- IMIV, Service Hospitalier Frédéric Joliot, CEA, Inserm,
Université Paris Sud, CNRS, Université Paris-Saclay, Orsay, France
| | - Marina Rubio
- Normandie Univ, UNICAEN, INSERM, INSERM UMR-S U1237,
Physiopathology and Imaging of Neurological Disorders, Institut Blood and Brain
@ Caen-Normandie, Cyceron, France
| | - Elisa R Zanier
- Department of Neuroscience, Istituto di Ricerche Farmacologiche
Mario Negri, IRCCS, Milan, Italy
| | - Damien Levard
- Normandie Univ, UNICAEN, INSERM, INSERM UMR-S U1237,
Physiopathology and Imaging of Neurological Disorders, Institut Blood and Brain
@ Caen-Normandie, Cyceron, France
| | - Sylvie Chalon
- UMR 1253, iBrain, Université de Tours, INSERM, Tours,
France
| | - Denis Vivien
- Normandie Univ, UNICAEN, INSERM, INSERM UMR-S U1237,
Physiopathology and Imaging of Neurological Disorders, Institut Blood and Brain
@ Caen-Normandie, Cyceron, France
- Department of Clinical Research, Caen-Normandie Hospital (CHU),
Caen, France
| | - Carine Ali
- Normandie Univ, UNICAEN, INSERM, INSERM UMR-S U1237,
Physiopathology and Imaging of Neurological Disorders, Institut Blood and Brain
@ Caen-Normandie, Cyceron, France
| |
Collapse
|
16
|
Natale C, Barzago MM, Colnaghi L, De Luigi A, Orsini F, Fioriti L, Diomede L. A Combined Cell-Worm Approach to Search for Compounds Counteracting the Toxicity of Tau Oligomers In Vivo. Int J Mol Sci 2022; 23:11277. [PMID: 36232578 PMCID: PMC9569484 DOI: 10.3390/ijms231911277] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2022] [Revised: 09/21/2022] [Accepted: 09/22/2022] [Indexed: 11/16/2022] Open
Abstract
A clear relationship between the tau assemblies and toxicity has still to be established. To correlate the tau conformation with its proteotoxic effect in vivo, we developed an innovative cell-worm-based approach. HEK293 cells expressing tau P301L under a tetracycline-inducible system (HEK T-Rex) were employed to produce different tau assemblies whose proteotoxic potential was evaluated using C. elegans. Lysates from cells induced for five days significantly reduced the worm's locomotor activity. This toxic effect was not related to the total amount of tau produced by cells or to its phosphorylation state but was related to the formation of multimeric tau assemblies, particularly tetrameric ones. We investigated the applicability of this approach for testing compounds acting against oligomeric tau toxicity, using doxycycline (Doxy) as a prototype drug. Doxy affected tau solubility and promoted the disassembly of already formed toxic aggregates in lysates of cells induced for five days. These effects translated into a dose-dependent protective action in C. elegans. These findings confirm the validity of the combined HEK T-Rex cells and the C. elegans-based approach as a platform for pharmacological screening.
Collapse
Affiliation(s)
- Carmina Natale
- Department of Molecular Biochemistry and Pharmacology, Istituto Di Ricerche Farmacologiche Mario Negri IRCCS, Via Mario Negri 2, 20156 Milan, Italy
| | - Maria Monica Barzago
- Department of Molecular Biochemistry and Pharmacology, Istituto Di Ricerche Farmacologiche Mario Negri IRCCS, Via Mario Negri 2, 20156 Milan, Italy
| | - Luca Colnaghi
- Department of Molecular Biochemistry and Pharmacology, Istituto Di Ricerche Farmacologiche Mario Negri IRCCS, Via Mario Negri 2, 20156 Milan, Italy
| | - Ada De Luigi
- Department of Molecular Biochemistry and Pharmacology, Istituto Di Ricerche Farmacologiche Mario Negri IRCCS, Via Mario Negri 2, 20156 Milan, Italy
| | - Franca Orsini
- Dulbecco Telethon Institute and Department of Neuroscience, Istituto Di Ricerche Farmacologiche Mario Negri IRCCS, Via Mario Negri 2, 20156 Milan, Italy
| | - Luana Fioriti
- Dulbecco Telethon Institute and Department of Neuroscience, Istituto Di Ricerche Farmacologiche Mario Negri IRCCS, Via Mario Negri 2, 20156 Milan, Italy
| | - Luisa Diomede
- Department of Molecular Biochemistry and Pharmacology, Istituto Di Ricerche Farmacologiche Mario Negri IRCCS, Via Mario Negri 2, 20156 Milan, Italy
| |
Collapse
|
17
|
Hwang K, Vaknalli RN, Addo-Osafo K, Vicente M, Vossel K. Tauopathy and Epilepsy Comorbidities and Underlying Mechanisms. Front Aging Neurosci 2022; 14:903973. [PMID: 35923547 PMCID: PMC9340804 DOI: 10.3389/fnagi.2022.903973] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2022] [Accepted: 06/22/2022] [Indexed: 11/16/2022] Open
Abstract
Tau is a microtubule-associated protein known to bind and promote assembly of microtubules in neurons under physiological conditions. However, under pathological conditions, aggregation of hyperphosphorylated tau causes neuronal toxicity, neurodegeneration, and resulting tauopathies like Alzheimer's disease (AD). Clinically, patients with tauopathies present with either dementia, movement disorders, or a combination of both. The deposition of hyperphosphorylated tau in the brain is also associated with epilepsy and network hyperexcitability in a variety of neurological diseases. Furthermore, pharmacological and genetic targeting of tau-based mechanisms can have anti-seizure effects. Suppressing tau phosphorylation decreases seizure activity in acquired epilepsy models while reducing or ablating tau attenuates network hyperexcitability in both Alzheimer's and epilepsy models. However, it remains unclear whether tauopathy and epilepsy comorbidities are mediated by convergent mechanisms occurring upstream of epileptogenesis and tau aggregation, by feedforward mechanisms between the two, or simply by coincident processes. In this review, we investigate the relationship between tauopathies and seizure disorders, including temporal lobe epilepsy (TLE), post-traumatic epilepsy (PTE), autism spectrum disorder (ASD), Dravet syndrome, Nodding syndrome, Niemann-Pick type C disease (NPC), Lafora disease, focal cortical dysplasia, and tuberous sclerosis complex. We also explore potential mechanisms implicating the role of tau kinases and phosphatases as well as the mammalian target of rapamycin (mTOR) in the promotion of co-pathology. Understanding the role of these co-pathologies could lead to new insights and therapies targeting both epileptogenic mechanisms and cognitive decline.
Collapse
|
18
|
Mohamed AZ, Cumming P, Nasrallah FA. Escalation of Tau Accumulation after a Traumatic Brain Injury: Findings from Positron Emission Tomography. Brain Sci 2022; 12:876. [PMID: 35884683 PMCID: PMC9313362 DOI: 10.3390/brainsci12070876] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2022] [Revised: 06/27/2022] [Accepted: 06/28/2022] [Indexed: 01/27/2023] Open
Abstract
Traumatic brain injury (TBI) has come to be recognized as a risk factor for Alzheimer's disease (AD), with poorly understood underlying mechanisms. We hypothesized that a history of TBI would be associated with greater tau deposition in elders with high-risk for dementia. A Groups of 20 participants with self-reported history of TBI and 100 without any such history were scanned using [18F]-AV1451 positron emission tomography as part of the Alzheimer's Disease Neuroimaging Initiative (ADNI). Scans were stratified into four groups according to TBI history, and by clinical dementia rating scores into cognitively normal (CDR = 0) and those showing cognitive decline (CDR ≥ 0.5). We pursued voxel-based group comparison of [18F]-AV1451 uptake to identify the effect of TBI history on brain tau deposition, and for voxel-wise correlation analyses between [18F]-AV1451 uptake and different neuropsychological measures and cerebrospinal fluid (CSF) biomarkers. Compared to the TBI-/CDR ≥ 0.5 group, the TBI+/CDR ≥ 0.5 group showed increased tau deposition in the temporal pole, hippocampus, fusiform gyrus, and inferior and middle temporal gyri. Furthermore, the extent of tau deposition in the brain of those with TBI history positively correlated with the extent of cognitive decline, CSF-tau, and CSF-amyloid. This might suggest TBI to increase the risk for tauopathies and Alzheimer's disease later in life.
Collapse
Affiliation(s)
- Abdalla Z. Mohamed
- Thompson Institute, University of Sunshine Coast, Birtinya, QLD 4575, Australia;
- Queensland Brain Institute, The University of Queensland, Brisbane, QLD 4072, Australia
| | - Paul Cumming
- Department of Nuclear Medicine, Bern University Hospital, 3010 Bern, Switzerland;
- School of Psychology and Counselling, Queensland University of Technology, Brisbane, QLD 4059, Australia
| | - Fatima A. Nasrallah
- Queensland Brain Institute, The University of Queensland, Brisbane, QLD 4072, Australia
| | | |
Collapse
|
19
|
Ameen-Ali KE, Bretzin A, Lee EB, Folkerth R, Hazrati LN, Iacono D, Keene CD, Kofler J, Kovacs GG, Nolan A, Perl DP, Priemer DS, Smith DH, Wiebe DJ, Stewart W. Detection of astrocytic tau pathology facilitates recognition of chronic traumatic encephalopathy neuropathologic change. Acta Neuropathol Commun 2022; 10:50. [PMID: 35410438 PMCID: PMC8996534 DOI: 10.1186/s40478-022-01353-4] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2022] [Accepted: 03/25/2022] [Indexed: 02/08/2023] Open
Abstract
Traumatic brain injury (TBI) is associated with the development of a range of neurodegenerative pathologies, including chronic traumatic encephalopathy (CTE). Current consensus diagnostic criteria define the pathognomonic cortical lesion of CTE neuropathologic change (CTE-NC) as a patchy deposition of hyperphosphorylated tau in neurons, with or without glial tau in thorn-shaped astrocytes, typically towards the depths of sulci and clustered around small blood vessels. Nevertheless, although incorporated into consensus diagnostic criteria, the contribution of the individual cellular components to identification of CTE-NC has not been formally evaluated. To address this, from the Glasgow TBI Archive, cortical tissue blocks were selected from consecutive brain donations from contact sports athletes in which there was known to be either CTE-NC (n = 12) or Alzheimer's disease neuropathologic change (n = 4). From these tissue blocks, adjacent tissue sections were stained for tau antibodies selected to reveal either solely neuronal pathology (3R tau; GT-38) or mixed neuronal and astroglial pathologies (4R tau; PHF-1). These stained sections were then randomised and independently assessed by a panel of expert neuropathologists, blind to patient clinical history and primary antibody applied to each section, who were asked to record whether CTE-NC was present. Results demonstrate that, in sections stained for either 4R tau or PHF-1, consensus recognition of CTE-NC was high. In contrast, recognition of CTE-NC in sections stained for 3R tau or GT-38 was poor; in the former no better than chance. Our observations demonstrate that the presence of both neuronal and astroglial tau pathologies facilitates detection of CTE-NC, with its detection less consistent when neuronal tau pathology alone is visible. The combination of both glial and neuronal pathologies, therefore, may be required for detection of CTE-NC.
Collapse
Affiliation(s)
- Kamar E Ameen-Ali
- Institute of Neuroscience and Psychology, University of Glasgow, Queen Elizabeth University Hospital, Glasgow, UK
| | - Abigail Bretzin
- Department of Biostatistics, Epidemiology and Informatics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Edward B Lee
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Rebecca Folkerth
- Office of Chief Medical Examiner, New York, NY, USA
- Department of Forensic Medicine, New York University School of Medicine, New York, NY, USA
| | - Lili-Naz Hazrati
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada
- Department of Pathology, The Hospital for Sick Children, Toronto, ON, Canada
| | - Diego Iacono
- Department of Defense/Uniformed Services, University Brain Tissue Repository and Neuropathology Program, Uniformed Services University, Bethesda, MD, USA
- Department of Pathology, F. Edward Hébert School of Medicine, Uniformed Services University, Bethesda, MD, USA
- Department of Neurology, F. Edward Hébert School of Medicine, Uniformed Services University, Bethesda, MD, USA
- Neurodegeneration Disorders Clinic, National Institute of Neurological Disorders and Stroke, NINDS, NIH, Bethesda, MD, USA
- The Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc., Bethesda, MD, USA
| | - C Dirk Keene
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, USA
| | - Julia Kofler
- Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Gabor G Kovacs
- Tanz Centre for Research in Neurodegenerative Disease (CRND) and Department of Laboratory Medicine and Pathobiology, Krembil Discovery Tower, University of Toronto, 60 Leonard Ave, Toronto, ON, Canada
- Laboratory Medicine Program and Krembil Brain Institute, University Health Network, Toronto, ON, Canada
| | - Amber Nolan
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, USA
| | - Daniel P Perl
- Department of Defense/Uniformed Services, University Brain Tissue Repository and Neuropathology Program, Uniformed Services University, Bethesda, MD, USA
- Department of Pathology, F. Edward Hébert School of Medicine, Uniformed Services University, Bethesda, MD, USA
| | - David S Priemer
- Department of Defense/Uniformed Services, University Brain Tissue Repository and Neuropathology Program, Uniformed Services University, Bethesda, MD, USA
- Department of Pathology, F. Edward Hébert School of Medicine, Uniformed Services University, Bethesda, MD, USA
- The Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc., Bethesda, MD, USA
| | - Douglas H Smith
- Center for Brain Injury and Repair, Department of Neurosurgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Douglas J Wiebe
- Department of Biostatistics, Epidemiology and Informatics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - William Stewart
- Institute of Neuroscience and Psychology, University of Glasgow, Queen Elizabeth University Hospital, Glasgow, UK.
- Department of Neuropathology, Laboratory Medicine Building, Elizabeth University Hospital, Glasgow, Queen, UK.
| |
Collapse
|
20
|
Mavroudis I, Kazis D, Chowdhury R, Petridis F, Costa V, Balmus IM, Ciobica A, Luca AC, Radu I, Dobrin RP, Baloyannis S. Post-Concussion Syndrome and Chronic Traumatic Encephalopathy: Narrative Review on the Neuropathology, Neuroimaging and Fluid Biomarkers. Diagnostics (Basel) 2022; 12:diagnostics12030740. [PMID: 35328293 PMCID: PMC8947595 DOI: 10.3390/diagnostics12030740] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2022] [Revised: 03/14/2022] [Accepted: 03/15/2022] [Indexed: 01/08/2023] Open
Abstract
Traumatic brain injury is a significant public health issue and represents the main contributor to death and disability globally among all trauma-related injuries. Martial arts practitioners, military veterans, athletes, victims of physical abuse, and epileptic patients could be affected by the consequences of repetitive mild head injuries (RMHI) that do not resume only to short-termed traumatic brain injuries (TBI) effects but also to more complex and time-extended outcomes, such as post-concussive syndrome (PCS) and chronic traumatic encephalopathy (CTE). These effects in later life are not yet well understood; however, recent studies suggested that even mild head injuries can lead to an elevated risk of later-life cognitive impairment and neurodegenerative disease. While most of the PCS hallmarks consist in immediate consequences and only in some conditions in long-termed processes undergoing neurodegeneration and impaired brain functions, the neuropathological hallmark of CTE is the deposition of p-tau immunoreactive pre-tangles and thread-like neurites at the depths of cerebral sulci and neurofibrillary tangles in the superficial layers I and II which are also one of the main hallmarks of neurodegeneration. Despite different CTE diagnostic criteria in clinical and research approaches, their specificity and sensitivity remain unclear and CTE could only be diagnosed post-mortem. In CTE, case risk factors include RMHI exposure due to profession (athletes, military personnel), history of trauma (abuse), or pathologies (epilepsy). Numerous studies aimed to identify imaging and fluid biomarkers that could assist diagnosis and probably lead to early intervention, despite their heterogeneous outcomes. Still, the true challenge remains the prediction of neurodegeneration risk following TBI, thus in PCS and CTE. Further studies in high-risk populations are required to establish specific, preferably non-invasive diagnostic biomarkers for CTE, considering the aim of preventive medicine.
Collapse
Affiliation(s)
- Ioannis Mavroudis
- Department of Neuroscience, Leeds Teaching Hospitals, NHS Trust, Leeds LS2 9JT, UK; (I.M.); (R.C.)
- Laboratory of Neuropathology and Electron Microscopy, Aristotle University of Thessaloniki, 54634 Thessaloniki, Greece; (V.C.); (S.B.)
- Research Institute for Alzheimer’s Disease and Neurodegenerative Diseases, Heraklion Langada, 57200 Thessaloniki, Greece
| | - Dimitrios Kazis
- Third Department of Neurology, Aristotle University of Thessaloniki, 57010 Thessaloniki, Greece; (D.K.); (F.P.)
| | - Rumana Chowdhury
- Department of Neuroscience, Leeds Teaching Hospitals, NHS Trust, Leeds LS2 9JT, UK; (I.M.); (R.C.)
| | - Foivos Petridis
- Third Department of Neurology, Aristotle University of Thessaloniki, 57010 Thessaloniki, Greece; (D.K.); (F.P.)
| | - Vasiliki Costa
- Laboratory of Neuropathology and Electron Microscopy, Aristotle University of Thessaloniki, 54634 Thessaloniki, Greece; (V.C.); (S.B.)
| | - Ioana-Miruna Balmus
- Department of Exact Sciences and Natural Sciences, Institute of Interdisciplinary Research, “Alexandru Ioan Cuza” University of Iași, 700057 Iași, Romania;
| | - Alin Ciobica
- Department of Biology, Faculty of Biology, Alexandru Ioan Cuza University, 700506 Iași, Romania
- Correspondence: (A.C.); (A.-C.L.); (R.P.D.)
| | - Alina-Costina Luca
- Faculty of Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iași, Romania;
- Correspondence: (A.C.); (A.-C.L.); (R.P.D.)
| | - Iulian Radu
- Faculty of Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iași, Romania;
| | - Romeo Petru Dobrin
- Faculty of Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iași, Romania;
- Correspondence: (A.C.); (A.-C.L.); (R.P.D.)
| | - Stavros Baloyannis
- Laboratory of Neuropathology and Electron Microscopy, Aristotle University of Thessaloniki, 54634 Thessaloniki, Greece; (V.C.); (S.B.)
- Research Institute for Alzheimer’s Disease and Neurodegenerative Diseases, Heraklion Langada, 57200 Thessaloniki, Greece
| |
Collapse
|
21
|
Walker A, Chapin B, Abisambra J, DeKosky ST. Association between single moderate to severe traumatic brain injury and long-term tauopathy in humans and preclinical animal models: a systematic narrative review of the literature. Acta Neuropathol Commun 2022; 10:13. [PMID: 35101132 PMCID: PMC8805270 DOI: 10.1186/s40478-022-01311-0] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2021] [Accepted: 12/22/2021] [Indexed: 11/28/2022] Open
Abstract
BACKGROUND The initiation, anatomic pattern, and extent of tau spread in traumatic brain injury (TBI), and the mechanism by which TBI leads to long-term tau pathology, remain controversial. Some studies suggest that moderate to severe TBI is sufficient to promote tau pathology; however, others suggest that it is simply a consequence of aging. We therefore conducted a systematic narrative review of the literature addressing whether a single moderate to severe head injury leads to long-term development of tauopathy in both humans and animal models. METHODS Studies considered for inclusion in this review assessed a single moderate to severe TBI, assessed tau pathology at long-term timepoints post-injury, comprised experimental or observational studies, and were peer-reviewed and published in English. Databases searched included: PUBMED, NCBI-PMC, EMBASE, Web of Science, Academic Search Premiere, and APA Psychnet. Search results were uploaded to Covidence®, duplicates were removed, and articles underwent an abstract and full-text screening process. Data were then extracted and articles assessed for risk of bias. FINDINGS Of 4,150 studies screened, 26 were eligible for inclusion, of which 17 were human studies, 8 were preclinical animal studies, and 1 included both human and preclinical animal studies. Most studies had low to moderate risk of bias. Most human and animal studies (n = 12 and 9, respectively) suggested that a single moderate to severe TBI resulted in greater development of long-term tauopathy compared to no history of head injury. This conclusion should be interpreted with caution, however, due to several limitations: small sample sizes; inconsistencies in controlling for confounding factors that may have affected tau pathology (e.g., family history of dementia or neurological illnesses, apolipoprotein E genotype, etc.), inclusion of mostly males, and variation in reporting injury parameters. INTERPRETATION Results indicate that a single moderate to severe TBI leads to greater chronic development of tauopathy compared to no history of head injury. This implies that tau pathology induced may not be transient, but can progressively develop over time in both humans and animal models. Targeting these tau changes for therapeutic intervention should be further explored to elucidate if disease progression can be reversed or mitigated.
Collapse
Affiliation(s)
- Ariel Walker
- Center for Translational Research in Neurodegenerative Disease, University of Florida, Gainesville, FL, 32610, USA
- Department of Neuroscience, University of Florida, Gainesville, FL, 32610, USA
- McKnight Brain Institute, University of Florida, Gainesville, FL, 32610, USA
| | - Ben Chapin
- Department of Neurology, University of Florida, Gainesville, FL, 32610, USA
| | - Jose Abisambra
- Center for Translational Research in Neurodegenerative Disease, University of Florida, Gainesville, FL, 32610, USA.
- Department of Neuroscience, University of Florida, Gainesville, FL, 32610, USA.
- McKnight Brain Institute, University of Florida, Gainesville, FL, 32610, USA.
- Brain Injury, Rehabilitation, and Neuroresilience (BRAIN) Center, University of Florida, Gainesville, FL, 32610, USA.
| | - Steven T DeKosky
- Center for Translational Research in Neurodegenerative Disease, University of Florida, Gainesville, FL, 32610, USA.
- Department of Neuroscience, University of Florida, Gainesville, FL, 32610, USA.
- McKnight Brain Institute, University of Florida, Gainesville, FL, 32610, USA.
- Brain Injury, Rehabilitation, and Neuroresilience (BRAIN) Center, University of Florida, Gainesville, FL, 32610, USA.
- Department of Neurology, University of Florida, Gainesville, FL, 32610, USA.
| |
Collapse
|
22
|
Zhu D, Gao F, Chen C. Endocannabinoid Metabolism and Traumatic Brain Injury. Cells 2021; 10:cells10112979. [PMID: 34831202 PMCID: PMC8616221 DOI: 10.3390/cells10112979] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2021] [Revised: 10/27/2021] [Accepted: 10/29/2021] [Indexed: 12/23/2022] Open
Abstract
Traumatic brain injury (TBI) represents a major cause of morbidity and disability and is a risk factor for developing neurodegenerative diseases, including Alzheimer’s disease (AD). However, no effective therapies are currently available for TBI-induced AD-like disease. Endocannabinoids are endogenous lipid mediators involved in a variety of physiological and pathological processes. The compound 2-arachidonoylglycerol (2-AG) is the most abundant endocannabinoid with profound anti-inflammatory and neuroprotective properties. This molecule is predominantly metabolized by monoacylglycerol lipase (MAGL), a key enzyme degrading about 85% of 2-AG in the brain. Studies using animal models of inflammation, AD, and TBI provide evidence that inactivation of MAGL, which augments 2-AG signaling and reduces its metabolites, exerts neuroprotective effects, suggesting that MAGL is a promising therapeutic target for neurodegenerative diseases. In this short review, we provide an overview of the inhibition of 2-AG metabolism for the alleviation of neuropathology and the improvement of synaptic and cognitive functions after TBI.
Collapse
|
23
|
Byrns CN, Saikumar J, Bonini NM. Glial AP1 is activated with aging and accelerated by traumatic brain injury. NATURE AGING 2021; 1:585-597. [PMID: 34723199 PMCID: PMC8553014 DOI: 10.1038/s43587-021-00072-0] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/23/2020] [Accepted: 04/30/2021] [Indexed: 01/05/2023]
Abstract
The emergence of degenerative disease after traumatic brain injury is often described as an acceleration of normal age-related processes. Whether similar molecular processes occur after injury and in age is unclear. Here we identify a functionally dynamic and lasting transcriptional response in glia, mediated by the conserved transcription factor AP1. In the early post-TBI period, glial AP1 is essential for recovery, ensuring brain integrity and animal survival. In sharp contrast, chronic AP1 activation promotes human tau pathology, tissue loss, and mortality. We show a similar process activates in healthy fly brains with age. In humans, AP1 activity is detected after moderate TBI and correlates with microglial activation and tau pathology. Our data provide key molecular insight into glia, highlighting that the same molecular process drives dynamic and contradictory glia behavior in TBI, and possibly age, first acting to protect but chronically promoting disease.
Collapse
Affiliation(s)
- China N Byrns
- Medical Scientist Training Program, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Neuroscience Graduate Group, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Janani Saikumar
- Department of Biology, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Nancy M Bonini
- Neuroscience Graduate Group, University of Pennsylvania, Philadelphia, PA 19104, USA
- Department of Biology, University of Pennsylvania, Philadelphia, PA 19104, USA
| |
Collapse
|
24
|
Zanier ER, Barzago MM, Vegliante G, Romeo M, Restelli E, Bertani I, Natale C, Colnaghi L, Colombo L, Russo L, Micotti E, Fioriti L, Chiesa R, Diomede L. C. elegans detects toxicity of traumatic brain injury generated tau. Neurobiol Dis 2021; 153:105330. [PMID: 33711491 PMCID: PMC8039186 DOI: 10.1016/j.nbd.2021.105330] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2020] [Revised: 03/02/2021] [Accepted: 03/04/2021] [Indexed: 11/30/2022] Open
Abstract
Traumatic brain injury (TBI) is associated with widespread tau pathology in about 30% of patients surviving late after injury. We previously found that TBI in mice induces the formation of an abnormal form of tau (tauTBI) which progressively spreads from the site of injury to remote brain regions. Intracerebral inoculation of TBI brain homogenates into naïve mice induced progressive tau pathology, synaptic loss and late cognitive decline, suggesting a pivotal role of tauTBI in post-TBI neurodegeneration. However, the possibility that tauTBI was a marker of TBI-associated neurodegeneration rather than a toxic driver of functional decline could not be excluded. Here we employed the nematode C. elegans as a biosensor to test the pathogenic role of TBI generated tau. The motility of this nematode depends on efficient neuromuscular transmission and is exceptionally sensitive to the toxicity of amyloidogenic proteins, providing a tractable model for our tests. We found that worms exposed to brain homogenates from chronic but not acute TBI mice, or from mice in which tauTBI had been transmitted by intracerebral inoculation, had impaired motility and neuromuscular synaptic transmission. Results were similar when worms were given brain homogenates from transgenic mice overexpressing tau P301L, a tauopathy mouse model, suggesting that TBI-induced and mutant tau have similar toxic properties. P301L brain homogenate toxicity was similar in wild-type and ptl-1 knock-out worms, indicating that the nematode tau homolog protein PTL-1 was not required to mediate the toxic effect. Harsh protease digestion to eliminate the protein component of the homogenates, pre-incubation with anti-tau antibodies or tau depletion by immunoprecipitation, abolished the toxicity. Homogenates of chronic TBI brains from tau knock-out mice were not toxic to C. elegans, whereas oligomeric recombinant tau was sufficient to impair their motility. This study indicates that tauTBI impairs motor activity and synaptic transmission in C. elegans and supports a pathogenic role of tauTBI in the long-term consequences of TBI. It also sets the groundwork for the development of a C. elegans-based platform for screening anti-tau compounds. Traumatic brain injury (TBI) in mice induces a progressive tau pathology. Brain-injured tissue from chronic but not acute TBI mice impairs C. elegans motility. TBI tissue immunodepleted of tau or from tau knock-out mice has no toxic effect. Brain-injured tissue from TBI mice impairs neuromuscular transmission in worms. C. elegans is a tractable model for investigating tau toxicity generated by TBI.
Collapse
Affiliation(s)
- Elisa R Zanier
- Department of Neuroscience, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy.
| | - Maria Monica Barzago
- Department of Molecular Biochemistry and Pharmacology, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy
| | - Gloria Vegliante
- Department of Neuroscience, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy
| | - Margherita Romeo
- Department of Molecular Biochemistry and Pharmacology, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy
| | - Elena Restelli
- Department of Neuroscience, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy
| | - Ilaria Bertani
- Department of Neuroscience, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy
| | - Carmina Natale
- Department of Neuroscience, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy; Department of Molecular Biochemistry and Pharmacology, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy
| | - Luca Colnaghi
- Department of Neuroscience, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy; Department of Molecular Biochemistry and Pharmacology, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy
| | - Laura Colombo
- Department of Molecular Biochemistry and Pharmacology, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy
| | - Luca Russo
- Department of Molecular Biochemistry and Pharmacology, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy
| | - Edoardo Micotti
- Department of Neuroscience, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy
| | - Luana Fioriti
- Department of Neuroscience, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy
| | - Roberto Chiesa
- Department of Neuroscience, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy.
| | - Luisa Diomede
- Department of Molecular Biochemistry and Pharmacology, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy.
| |
Collapse
|
25
|
Soni N, Medeiros R, Alateeq K, To XV, Nasrallah FA. Diffusion Tensor Imaging Detects Acute Pathology-Specific Changes in the P301L Tauopathy Mouse Model Following Traumatic Brain Injury. Front Neurosci 2021; 15:611451. [PMID: 33716645 PMCID: PMC7943881 DOI: 10.3389/fnins.2021.611451] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2020] [Accepted: 01/25/2021] [Indexed: 11/18/2022] Open
Abstract
Traumatic brain injury (TBI) has been linked with tauopathy. However, imaging methods that can non-invasively detect tau-protein abnormalities following TBI need further investigation. This study aimed to investigate the potential of diffusion tensor imaging (DTI) to detect tauopathy following TBI in P301L mutant-tau-transgenic-pR5-mice. A total of 24 9-month-old pR5 mice were randomly assigned to sham and TBI groups. Controlled cortical injuries/craniotomies were performed for TBI/sham groups followed by DTI data acquisition on days 1 and 7 post-injury. DTI data were analyzed by using voxelwise analysis and track-based spatial statistics for gray matter and white matter. Further, immunohistochemistry was performed for total-tau and phosphorylated-tau, astrocytes, and microglia. To detect the association of DTI with these pathological markers, a correlation analysis was performed between DTI and histology findings. At day 1 post-TBI, DTI revealed a widespread reduction in fractional anisotropy (FA) and axial diffusivity (AxD) in the TBI group compared to shams. On day 7, further reduction in FA, AxD, and mean diffusivity and increased radial diffusivity were observed. FA was significantly increased in the amygdala and cortex. Correlation results showed that in the ipsilateral hemisphere FA reduction was associated with increased phosphorylated-tau and glial-immunoreactivity, whereas in the contralateral regions, the FA increase was associated with increased immunostaining for astrocytes. This study is the first to exploit DTI to investigate the effect of TBI in tau-transgenic mice. We show that alterations in the DTI signal were associated with glial activity following TBI and would most likely reflect changes that co-occur with/without phosphorylated-tau. In addition, FA may be a promising measure to identify discrete pathological processes such as increased astroglia activation, tau-hyperphosphorylation or both in the brain following TBI.
Collapse
Affiliation(s)
- Neha Soni
- Queensland Brain Institute, The University of Queensland, St. Lucia, QLD, Australia
| | - Rodrigo Medeiros
- Queensland Brain Institute, The University of Queensland, St. Lucia, QLD, Australia
| | - Khawlah Alateeq
- Queensland Brain Institute, The University of Queensland, St. Lucia, QLD, Australia
| | - Xuan Vinh To
- Queensland Brain Institute, The University of Queensland, St. Lucia, QLD, Australia
| | - Fatima A Nasrallah
- Queensland Brain Institute, The University of Queensland, St. Lucia, QLD, Australia
| |
Collapse
|
26
|
Bieniek KF, Cairns NJ, Crary JF, Dickson DW, Folkerth RD, Keene CD, Litvan I, Perl DP, Stein TD, Vonsattel JP, Stewart W, Dams-O’Connor K, Gordon WA, Tripodis Y, Alvarez VE, Mez J, Alosco ML, McKee AC. The Second NINDS/NIBIB Consensus Meeting to Define Neuropathological Criteria for the Diagnosis of Chronic Traumatic Encephalopathy. J Neuropathol Exp Neurol 2021; 80:210-219. [PMID: 33611507 PMCID: PMC7899277 DOI: 10.1093/jnen/nlab001] [Citation(s) in RCA: 121] [Impact Index Per Article: 40.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Chronic traumatic encephalopathy (CTE) is a neurodegenerative disorder associated with exposure to head trauma. In 2015, a panel of neuropathologists funded by the NINDS/NIBIB defined preliminary consensus neuropathological criteria for CTE, including the pathognomonic lesion of CTE as "an accumulation of abnormal hyperphosphorylated tau (p-tau) in neurons and astroglia distributed around small blood vessels at the depths of cortical sulci and in an irregular pattern," based on review of 25 tauopathy cases. In 2016, the consensus panel met again to review and refine the preliminary criteria, with consideration around the minimum threshold for diagnosis and the reproducibility of a proposed pathological staging scheme. Eight neuropathologists evaluated 27 cases of tauopathies (17 CTE cases), blinded to clinical and demographic information. Generalized estimating equation analyses showed a statistically significant association between the raters and CTE diagnosis for both the blinded (OR = 72.11, 95% CI = 19.5-267.0) and unblinded rounds (OR = 256.91, 95% CI = 63.6-1558.6). Based on the challenges in assigning CTE stage, the panel proposed a working protocol including a minimum threshold for CTE diagnosis and an algorithm for the assessment of CTE severity as "Low CTE" or "High CTE" for use in future clinical, pathological, and molecular studies.
Collapse
Affiliation(s)
- Kevin F Bieniek
- Department of Pathology, Glenn Biggs Institute for Alzheimer’s and Neurodegenerative Diseases, UT Health San Antonio, San Antonio, Texas
| | - Nigel J Cairns
- Department of Neurology, Washington University School of Medicine, Saint Louis, Missouri
| | - John F Crary
- Departments of Pathology & Neuroscience, Ronald M. Loeb Center for Alzheimer’s Disease, Freidman Brain Institute, Icahn School of Medicine at Mount Sinai School, New York, New York
| | | | - Rebecca D Folkerth
- New York City Office of Chief Medical Examiner and Department of Forensic Medicine, New York University School of Medicine, New York, New York
| | - C Dirk Keene
- Department of Pathology, University of Washington School of Medicine, Seattle, Washington
| | - Irene Litvan
- Department of Neurosciences, University of California San Diego School of Medicine, La Jolla, California
| | - Daniel P Perl
- Department of Pathology, Uniformed Services University of the Health Sciences, Bethesda, Maryland
| | - Thor D Stein
- VA Boston Healthcare System, Boston, Massachusetts
- Department of Pathology, Boston University School of Medicine, Boston, Massachusetts; Department of Veteran Affairs Medical Center, Bedford, Massachusetts
- Alzheimer's Disease Center, CTE Program, Boston University School of Medicine, Boston, Massachusetts; Department of Veteran Affairs Medical Center, Bedford, Massachusetts
| | - Jean-Paul Vonsattel
- Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University Medical Center, New York, New York
| | - William Stewart
- Department of Neuropathology, University of Glasgow Institute of Neuroscience and Psychology and Queen Elizabeth University Hospital, Glasgow, United Kingdom
| | - Kristen Dams-O’Connor
- Department of Rehabilitation Medicine, Icahn School of Medicine at Mount Sinai, New York, New York
- Department of Neurology (KD-O), Icahn School of Medicine at Mount Sinai, New York, New York
| | - Wayne A Gordon
- Department of Rehabilitation Medicine, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Yorghos Tripodis
- Alzheimer's Disease Center, CTE Program, Boston University School of Medicine, Boston, Massachusetts; Department of Veteran Affairs Medical Center, Bedford, Massachusetts
| | - Victor E Alvarez
- VA Boston Healthcare System, Boston, Massachusetts
- Department of Neurology, Boston University School of Medicine, Boston, Massachusetts; Department of Veteran Affairs Medical Center, Bedford, Massachusetts
- Alzheimer's Disease Center, CTE Program, Boston University School of Medicine, Boston, Massachusetts; Department of Veteran Affairs Medical Center, Bedford, Massachusetts
| | - Jesse Mez
- Department of Neurology, Boston University School of Medicine, Boston, Massachusetts; Department of Veteran Affairs Medical Center, Bedford, Massachusetts
- Alzheimer's Disease Center, CTE Program, Boston University School of Medicine, Boston, Massachusetts; Department of Veteran Affairs Medical Center, Bedford, Massachusetts
| | - Michael L Alosco
- Department of Neurology, Boston University School of Medicine, Boston, Massachusetts; Department of Veteran Affairs Medical Center, Bedford, Massachusetts
- Alzheimer's Disease Center, CTE Program, Boston University School of Medicine, Boston, Massachusetts; Department of Veteran Affairs Medical Center, Bedford, Massachusetts
| | - Ann C McKee
- VA Boston Healthcare System, Boston, Massachusetts
- Department of Pathology, Boston University School of Medicine, Boston, Massachusetts; Department of Veteran Affairs Medical Center, Bedford, Massachusetts
- Department of Neurology, Boston University School of Medicine, Boston, Massachusetts; Department of Veteran Affairs Medical Center, Bedford, Massachusetts
- Alzheimer's Disease Center, CTE Program, Boston University School of Medicine, Boston, Massachusetts; Department of Veteran Affairs Medical Center, Bedford, Massachusetts
| |
Collapse
|
27
|
Ekker M. Visualizing traumatic brain injuries. eLife 2021; 10:e65676. [PMID: 33527900 PMCID: PMC7853714 DOI: 10.7554/elife.65676] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2021] [Accepted: 01/25/2021] [Indexed: 11/23/2022] Open
Abstract
Zebrafish larvae models can be used to study the link between seizures and the neurodegeneration that follows brain trauma.
Collapse
Affiliation(s)
- Marc Ekker
- Department of Biology, University of OttawaOttawaCanada
| |
Collapse
|
28
|
Alyenbaawi H, Kanyo R, Locskai LF, Kamali-Jamil R, DuVal MG, Bai Q, Wille H, Burton EA, Allison WT. Seizures are a druggable mechanistic link between TBI and subsequent tauopathy. eLife 2021; 10:e58744. [PMID: 33527898 PMCID: PMC7853719 DOI: 10.7554/elife.58744] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2020] [Accepted: 12/07/2020] [Indexed: 12/18/2022] Open
Abstract
Traumatic brain injury (TBI) is a prominent risk factor for dementias including tauopathies like chronic traumatic encephalopathy (CTE). The mechanisms that promote prion-like spreading of Tau aggregates after TBI are not fully understood, in part due to lack of tractable animal models. Here, we test the putative role of seizures in promoting the spread of tauopathy. We introduce 'tauopathy reporter' zebrafish expressing a genetically encoded fluorescent Tau biosensor that reliably reports accumulation of human Tau species when seeded via intraventricular brain injections. Subjecting zebrafish larvae to a novel TBI paradigm produced various TBI features including cell death, post-traumatic seizures, and Tau inclusions. Bath application of dynamin inhibitors or anticonvulsant drugs rescued TBI-induced tauopathy and cell death. These data suggest a role for seizure activity in the prion-like seeding and spreading of tauopathy following TBI. Further work is warranted regarding anti-convulsants that dampen post-traumatic seizures as a route to moderating subsequent tauopathy.
Collapse
Affiliation(s)
- Hadeel Alyenbaawi
- Centre for Prions & Protein Folding Disease, University of AlbertaEdmontonCanada
- Department of Medical Genetics, University of AlbertaEdmontonCanada
- Majmaah UniversityMajmaahSaudi Arabia
| | - Richard Kanyo
- Centre for Prions & Protein Folding Disease, University of AlbertaEdmontonCanada
- Department of Biological Sciences, University of AlbertaEdmontonCanada
| | - Laszlo F Locskai
- Centre for Prions & Protein Folding Disease, University of AlbertaEdmontonCanada
- Department of Biological Sciences, University of AlbertaEdmontonCanada
| | - Razieh Kamali-Jamil
- Centre for Prions & Protein Folding Disease, University of AlbertaEdmontonCanada
- Department of Biochemistry, University of AlbertaEdmontonCanada
| | - Michèle G DuVal
- Department of Biological Sciences, University of AlbertaEdmontonCanada
| | - Qing Bai
- Department of Neurology, University of PittsburghPittsburghUnited States
| | - Holger Wille
- Centre for Prions & Protein Folding Disease, University of AlbertaEdmontonCanada
- Department of Biochemistry, University of AlbertaEdmontonCanada
| | - Edward A Burton
- Department of Neurology, University of PittsburghPittsburghUnited States
- Geriatric Research, Education and Clinical Center, Pittsburgh VA Healthcare SystemPittsburghUnited States
| | - W Ted Allison
- Centre for Prions & Protein Folding Disease, University of AlbertaEdmontonCanada
- Department of Medical Genetics, University of AlbertaEdmontonCanada
- Department of Biological Sciences, University of AlbertaEdmontonCanada
| |
Collapse
|
29
|
Andrews M, Ross R, Malhotra A, Ancoli-Israel S, Brewer JB, Banks SJ. Sleep and Tau Pathology in Vietnam War Veterans with Preclinical and Prodromal Alzheimer's Disease. J Alzheimers Dis Rep 2021; 5:41-48. [PMID: 33681715 PMCID: PMC7903001 DOI: 10.3233/adr-200245] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
Background The increasing prevalence of Alzheimer's disease (AD) and lack of effective medications has led to a need to identify modifiable risk factors as targets for interventions. Objective In this cross-sectional study, we sought to determine whether worse sleep quality is associated with increased pathological tau, and whether this relationship is affected by amyloid pathology. Methods 66 male participants underwent Florbetapir (AV45) positron emission tomography (PET) and Flortaucipir (FTP) PET and completed the Pittsburgh Sleep Quality Index questionnaire (PSQI) as part of the Department of Defense Alzheimer's Disease Neuroimaging Initiative, a multicenter study collecting data from Vietnam War veterans, some of whom have a history of post-traumatic stress disorder, or non-penetrating traumatic brain injury. AV45 PET was used to determine the presence of significant amyloid pathology. We used regression models to determine the effects of amyloid pathology and PSQI on tau deposition in brain regions associated with Braak stages. Results Among the 66 participants, 14 individuals were amyloid positive (21%) and 52 were amyloid negative (79%). In regions associated with Braak stages III-IV, there was a significant interaction of amyloid status on PSQI (β= 0.04, p = 0.003) with higher PSQI correlating with higher FTP SUVr in amyloid-positive individuals only (β= 0.031, p = 0.005). Conclusion Our study found that an AD profile of tau deposition was associated with an interaction between self-reported sleep quality and amyloid pathology such that worse self-reported sleep was related to higher tau in regions usually associated with AD progression, but only in individuals with high cerebral amyloid deposition.
Collapse
Affiliation(s)
- Murray Andrews
- Department of Neuroscience, University of California, San Diego, La Jolla, CA, USA
| | - Ryan Ross
- Department of Neuroscience, University of California, San Diego, La Jolla, CA, USA
| | - Atul Malhotra
- Department of Health Sciences, University of California, San Diego, La Jolla, CA, USA
| | - Sonia Ancoli-Israel
- Department of Psychiatry, University of California, San Diego, La Jolla, CA, USA
| | - James B Brewer
- Department of Neuroscience, University of California, San Diego, La Jolla, CA, USA
| | - Sarah J Banks
- Department of Neuroscience, University of California, San Diego, La Jolla, CA, USA
| | | |
Collapse
|
30
|
Arena JD, Johnson VE, Lee EB, Gibbons GS, Smith DH, Trojanowski JQ, Stewart W. Astroglial tau pathology alone preferentially concentrates at sulcal depths in chronic traumatic encephalopathy neuropathologic change. Brain Commun 2020; 2:fcaa210. [PMID: 33426528 PMCID: PMC7784042 DOI: 10.1093/braincomms/fcaa210] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Revised: 10/06/2020] [Accepted: 11/02/2020] [Indexed: 12/14/2022] Open
Abstract
Current diagnostic criteria for the neuropathological evaluation of the traumatic brain injury-associated neurodegeneration, chronic traumatic encephalopathy, define the pathognomonic lesion as hyperphosphorylated tau-immunoreactive neuronal and astroglial profiles in a patchy cortical distribution, clustered around small vessels and showing preferential localization to the depths of sulci. However, despite adoption into diagnostic criteria, there has been no formal assessment of the cortical distribution of the specific cellular components defining chronic traumatic encephalopathy neuropathologic change. To address this, we performed comprehensive mapping of hyperphosphorylated tau-immunoreactive neurofibrillary tangles and thorn-shaped astrocytes contributing to chronic traumatic encephalopathy neuropathologic change. From the Glasgow Traumatic Brain Injury Archive and the University of Pennsylvania Center for Neurodegenerative Disease Research Brain Bank, material was selected from patients with known chronic traumatic encephalopathy neuropathologic change, either following exposure to repetitive mild (athletes n = 17; non-athletes n = 1) or to single moderate or severe traumatic brain injury (n = 4), together with material from patients with previously confirmed Alzheimer's disease neuropathologic changes (n = 6) and no known exposure to traumatic brain injury. Representative sections were stained for hyperphosphorylated or Alzheimer's disease conformation-selective tau, after which stereotypical neurofibrillary tangles and thorn-shaped astrocytes were identified and mapped. Thorn-shaped astrocytes in chronic traumatic encephalopathy neuropathologic change were preferentially distributed towards sulcal depths [sulcal depth to gyral crest ratio of thorn-shaped astrocytes 12.84 ± 15.47 (mean ± standard deviation)], with this pathology more evident in material from patients with a history of survival from non-sport injury than those exposed to sport-associated traumatic brain injury (P = 0.009). In contrast, neurofibrillary tangles in chronic traumatic encephalopathy neuropathologic change showed a more uniform distribution across the cortex in sections stained for either hyperphosphorylated (sulcal depth to gyral crest ratio of neurofibrillary tangles 1.40 ± 0.74) or Alzheimer's disease conformation tau (sulcal depth to gyral crest ratio 1.64 ± 1.05), which was comparable to that seen in material from patients with known Alzheimer's disease neuropathologic changes (P = 0.82 and P = 0.91, respectively). Our data demonstrate that in chronic traumatic encephalopathy neuropathologic change the astroglial component alone shows preferential distribution to the depths of cortical sulci. In contrast, the neuronal pathology of chronic traumatic encephalopathy neuropathologic change is distributed more uniformly from gyral crest to sulcal depth and echoes that of Alzheimer's disease. These observations provide new insight into the neuropathological features of chronic traumatic encephalopathy that distinguish it from other tau pathologies and suggest that current diagnostic criteria should perhaps be reviewed and refined.
Collapse
Affiliation(s)
- John D Arena
- Department of Neurosurgery, Penn Center for Brain Injury and Repair, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Victoria E Johnson
- Department of Neurosurgery, Penn Center for Brain Injury and Repair, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Edward B Lee
- Department of Pathology and Laboratory Medicine, Center for Neurodegenerative Disease Research, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA.,Translational Neuropathology Research Laboratory, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Garrett S Gibbons
- Department of Pathology and Laboratory Medicine, Center for Neurodegenerative Disease Research, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Douglas H Smith
- Department of Neurosurgery, Penn Center for Brain Injury and Repair, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - John Q Trojanowski
- Department of Pathology and Laboratory Medicine, Center for Neurodegenerative Disease Research, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - William Stewart
- Department of Neuropathology, Queen Elizabeth University Hospital, Glasgow G51 4TF, UK.,Institute of Neuroscience and Psychology, University of Glasgow, Glasgow G12 8QQ, UK
| |
Collapse
|
31
|
Kochanek PM, Jackson TC, Jha RM, Clark RS, Okonkwo DO, Bayır H, Poloyac SM, Wagner AK, Empey PE, Conley YP, Bell MJ, Kline AE, Bondi CO, Simon DW, Carlson SW, Puccio AM, Horvat CM, Au AK, Elmer J, Treble-Barna A, Ikonomovic MD, Shutter LA, Taylor DL, Stern AM, Graham SH, Kagan VE, Jackson EK, Wisniewski SR, Dixon CE. Paths to Successful Translation of New Therapies for Severe Traumatic Brain Injury in the Golden Age of Traumatic Brain Injury Research: A Pittsburgh Vision. J Neurotrauma 2020; 37:2353-2371. [PMID: 30520681 PMCID: PMC7698994 DOI: 10.1089/neu.2018.6203] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
New neuroprotective therapies for severe traumatic brain injury (TBI) have not translated from pre-clinical to clinical success. Numerous explanations have been suggested in both the pre-clinical and clinical arenas. Coverage of TBI in the lay press has reinvigorated interest, creating a golden age of TBI research with innovative strategies to circumvent roadblocks. We discuss the need for more robust therapies. We present concepts for traditional and novel approaches to defining therapeutic targets. We review lessons learned from the ongoing work of the pre-clinical drug and biomarker screening consortium Operation Brain Trauma Therapy and suggest ways to further enhance pre-clinical consortia. Biomarkers have emerged that empower choice and assessment of target engagement by candidate therapies. Drug combinations may be needed, and it may require moving beyond conventional drug therapies. Precision medicine may also link the right therapy to the right patient, including new approaches to TBI classification beyond the Glasgow Coma Scale or anatomical phenotyping-incorporating new genetic and physiologic approaches. Therapeutic breakthroughs may also come from alternative approaches in clinical investigation (comparative effectiveness, adaptive trial design, use of the electronic medical record, and big data). The full continuum of care must also be represented in translational studies, given the important clinical role of pre-hospital events, extracerebral insults in the intensive care unit, and rehabilitation. TBI research from concussion to coma can cross-pollinate and further advancement of new therapies. Misconceptions can stifle/misdirect TBI research and deserve special attention. Finally, we synthesize an approach to deliver therapeutic breakthroughs in this golden age of TBI research.
Collapse
Affiliation(s)
- Patrick M. Kochanek
- Safar Center for Resuscitation Research, UPMC Children's Hospital of Pittsburgh, Pittsburgh, Pennsylvania, USA
- Department of Critical Care Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Travis C. Jackson
- Safar Center for Resuscitation Research, UPMC Children's Hospital of Pittsburgh, Pittsburgh, Pennsylvania, USA
- Department of Critical Care Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Ruchira M. Jha
- Safar Center for Resuscitation Research, UPMC Children's Hospital of Pittsburgh, Pittsburgh, Pennsylvania, USA
- Department of Critical Care Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Robert S.B. Clark
- Safar Center for Resuscitation Research, UPMC Children's Hospital of Pittsburgh, Pittsburgh, Pennsylvania, USA
- Department of Critical Care Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - David O. Okonkwo
- Department of Neurological Surgery, UPMC Presbyterian Hospital, Pittsburgh, Pennsylvania, USA
| | - Hülya Bayır
- Safar Center for Resuscitation Research, UPMC Children's Hospital of Pittsburgh, Pittsburgh, Pennsylvania, USA
- Department of Critical Care Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
- Department of Environmental and Occupational Health, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Samuel M. Poloyac
- Safar Center for Resuscitation Research, UPMC Children's Hospital of Pittsburgh, Pittsburgh, Pennsylvania, USA
- University of Pittsburgh School of Pharmacy, Pittsburgh, Pennsylvania, USA
| | - Amy K. Wagner
- Safar Center for Resuscitation Research, UPMC Children's Hospital of Pittsburgh, Pittsburgh, Pennsylvania, USA
- Department of Physical Medicine and Rehabilitation, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Philip E. Empey
- Safar Center for Resuscitation Research, UPMC Children's Hospital of Pittsburgh, Pittsburgh, Pennsylvania, USA
- Pharmacy and Therapeutics, University of Pittsburgh School of Pharmacy, Pittsburgh, Pennsylvania, USA
| | - Yvette P. Conley
- Health Promotion and Development, University of Pittsburgh School of Nursing, Pittsburgh, Pennsylvania, USA
| | - Michael J. Bell
- Department of Critical Care Medicine, Children's National Medical Center, Washington, DC, USA
| | - Anthony E. Kline
- Safar Center for Resuscitation Research, UPMC Children's Hospital of Pittsburgh, Pittsburgh, Pennsylvania, USA
- Department of Physical Medicine and Rehabilitation, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Corina O. Bondi
- Safar Center for Resuscitation Research, UPMC Children's Hospital of Pittsburgh, Pittsburgh, Pennsylvania, USA
- Department of Physical Medicine and Rehabilitation, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Dennis W. Simon
- Safar Center for Resuscitation Research, UPMC Children's Hospital of Pittsburgh, Pittsburgh, Pennsylvania, USA
- Department of Critical Care Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Shaun W. Carlson
- Safar Center for Resuscitation Research, UPMC Children's Hospital of Pittsburgh, Pittsburgh, Pennsylvania, USA
- Department of Neurological Surgery, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Ava M. Puccio
- Department of Neurological Surgery, UPMC Presbyterian Hospital, Pittsburgh, Pennsylvania, USA
| | - Christopher M. Horvat
- Safar Center for Resuscitation Research, UPMC Children's Hospital of Pittsburgh, Pittsburgh, Pennsylvania, USA
- Department of Critical Care Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Alicia K. Au
- Safar Center for Resuscitation Research, UPMC Children's Hospital of Pittsburgh, Pittsburgh, Pennsylvania, USA
- Department of Critical Care Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Jonathan Elmer
- Departments of Emergency Medicine and Critical Care Medicine, University of Pittsburgh School of Medicine, UPMC Presbyterian Hospital, Pittsburgh, Pennsylvania, USA
| | - Amery Treble-Barna
- Safar Center for Resuscitation Research, UPMC Children's Hospital of Pittsburgh, Pittsburgh, Pennsylvania, USA
- Department of Physical Medicine and Rehabilitation, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Milos D. Ikonomovic
- Department of Psychiatry, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
- Department of Neurology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Lori A. Shutter
- Department of Critical Care Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - D. Lansing Taylor
- University of Pittsburgh Drug Discovery Institute, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Andrew M. Stern
- Drug Discovery Institute, Department of Computational and Systems Biology, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Steven H. Graham
- Geriatric Research Education and Clinical Center, VA Pittsburgh Healthcare System, Pittsburgh, Pennsylvania, USA
- Department of Neurology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Valerian E. Kagan
- Department of Environmental and Occupational Health, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Edwin K. Jackson
- Department of Pharmacology and Chemical Biology, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Stephen R. Wisniewski
- University of Pittsburgh Graduate School of Public Health, Pittsburgh, Pennsylvania, USA
| | - C. Edward Dixon
- Department of Neurological Surgery, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
- Geriatric Research Education and Clinical Center, VA Pittsburgh Healthcare System, Pittsburgh, Pennsylvania, USA
| |
Collapse
|
32
|
Banbury C, Styles I, Eisenstein N, Zanier ER, Vegliante G, Belli A, Logan A, Goldberg Oppenheimer P. Spectroscopic detection of traumatic brain injury severity and biochemistry from the retina. BIOMEDICAL OPTICS EXPRESS 2020; 11:6249-6261. [PMID: 33282487 PMCID: PMC7687955 DOI: 10.1364/boe.399473] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/04/2020] [Revised: 08/19/2020] [Accepted: 09/15/2020] [Indexed: 05/11/2023]
Abstract
Traumatic brain injury (TBI) is a major burden on healthcare services worldwide, where scientific and clinical innovation is needed to provide better understanding of biochemical damage to improve both pre-hospital assessment and intensive care monitoring. Here, we present an unconventional concept of using Raman spectroscopy to measure the biochemical response to the retina in an ex-vivo murine model of TBI. Through comparison to spectra from the brain and retina following injury, we elicit subtle spectral changes through the use of multivariate analysis, linked to a decrease in cardiolipin and indicating metabolic disruption. The ability to classify injury severity via spectra of the retina is demonstrated for severe TBI (82.0 %), moderate TBI (75.1 %) and sham groups (69.4 %). By showing that optical spectroscopy can be used to explore the eye as the window to the brain, we lay the groundwork for further exploitation of Raman spectroscopy for indirect, non-invasive assessment of brain chemistry.
Collapse
Affiliation(s)
- Carl Banbury
- School of Chemical Engineering, The University of Birmingham, Edgbaston, Birmingham, B15 2TT, UK
| | - Iain Styles
- Computer Science, The University of Birmingham, Edgbaston, Birmingham, B15 2TT, UK
| | - Neil Eisenstein
- School of Chemical Engineering, The University of Birmingham, Edgbaston, Birmingham, B15 2TT, UK
| | - Elisa R. Zanier
- Department of Neuroscience, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy
| | - Gloria Vegliante
- Department of Neuroscience, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy
| | - Antonio Belli
- Institute of Inflammation and Ageing, The University of Birmingham, Edgbaston, Birmingham, B15 2TT, UK
| | - Ann Logan
- Institute of Inflammation and Ageing, The University of Birmingham, Edgbaston, Birmingham, B15 2TT, UK
| | | |
Collapse
|
33
|
Mercurio D, Oggioni M, Fumagalli S, Lynch NJ, Roscher S, Minuta D, Perego C, Ippati S, Wallis R, Schwaeble WJ, De Simoni MG. Targeted deletions of complement lectin pathway genes improve outcome in traumatic brain injury, with MASP-2 playing a major role. Acta Neuropathol Commun 2020; 8:174. [PMID: 33115535 PMCID: PMC7592565 DOI: 10.1186/s40478-020-01041-1] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2020] [Accepted: 09/24/2020] [Indexed: 12/13/2022] Open
Abstract
The lectin pathway (LP) of complement activation is believed to contribute to brain inflammation. The study aims to identify the key components of the LP contributing to TBI outcome as possible novel pharmacological targets. We compared the long-term neurological deficits and neuropathology of wild-type mice (WT) to that of mice carrying gene deletions of key LP components after experimental TBI. WT or MASP-2 (Masp2-/-), ficolin-A (Fcna-/-), CL-11 (Colec11-/-), MASP-1/3 (Masp1-/-), MBL-C (Mbl2-/-), MBL-A (Mbl1-/-) or MBL-/- (Mbl1-/-/Mbl2-/-) deficient male C57BL/6J mice were used. Mice underwent sham surgery or TBI by controlled cortical impact. The sensorimotor response was evaluated by neuroscore and beam walk tests weekly for 4 weeks. To obtain a comparative analysis of the functional outcome each transgenic line was rated according to a health score calculated on sensorimotor performance. For selected genotypes, brains were harvested 6 weeks after injury for histopathological analysis. MASP-2-/-, MBL-/- and FCN-A-/- mice had better outcome scores compared to WT. Of these, MASP-2-/- mice had the best recovery after TBI, showing reduced sensorimotor deficits (by 33% at 3 weeks and by 36% at 4 weeks). They also showed higher neuronal density in the lesioned cortex with a 31.5% increase compared to WT. Measurement of LP functional activity in plasma from MASP-2-/- mice revealed the absence of LP functional activity using a C4b deposition assay. The LP critically contributes to the post-traumatic inflammatory pathology following TBI with the highest degree of protection achieved through the absence of the LP key enzyme MASP-2, underlining a therapeutic utility of MASP-2 targeting in TBI.
Collapse
Affiliation(s)
- D Mercurio
- Department of Neuroscience, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, via Mario Negri 2, 20156, Milan, Italy
| | - M Oggioni
- Department of Neuroscience, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, via Mario Negri 2, 20156, Milan, Italy
| | - S Fumagalli
- Department of Neuroscience, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, via Mario Negri 2, 20156, Milan, Italy
| | - N J Lynch
- Department of Neuroscience, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, via Mario Negri 2, 20156, Milan, Italy
- Department of Veterinary Medicine, University of Cambridge, Madingley Road, CB3 0ES, Cambridge, UK
| | - S Roscher
- Department of Respiratory Sciences, University of Leicester, University Road, LE1 9HN, Leicester, UK
| | - D Minuta
- Department of Neuroscience, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, via Mario Negri 2, 20156, Milan, Italy
- San Raffaele Scientific Institute, San Raffaele Hospital, 20132, Milan, Italy
| | - C Perego
- Department of Neuroscience, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, via Mario Negri 2, 20156, Milan, Italy
| | - S Ippati
- Department of Neuroscience, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, via Mario Negri 2, 20156, Milan, Italy
- National Research Council (CNR), Institute of Neuroscience, 20129, Milan, Italy
| | - R Wallis
- Department of Respiratory Sciences, University of Leicester, University Road, LE1 9HN, Leicester, UK
| | - W J Schwaeble
- Department of Veterinary Medicine, University of Cambridge, Madingley Road, CB3 0ES, Cambridge, UK
| | - M-G De Simoni
- Department of Neuroscience, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, via Mario Negri 2, 20156, Milan, Italy.
| |
Collapse
|
34
|
Alyenbaawi H, Allison WT, Mok SA. Prion-Like Propagation Mechanisms in Tauopathies and Traumatic Brain Injury: Challenges and Prospects. Biomolecules 2020; 10:E1487. [PMID: 33121065 PMCID: PMC7692808 DOI: 10.3390/biom10111487] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2020] [Revised: 10/22/2020] [Accepted: 10/23/2020] [Indexed: 12/23/2022] Open
Abstract
The accumulation of tau protein in the form of filamentous aggregates is a hallmark of many neurodegenerative diseases such as Alzheimer's disease (AD) and chronic traumatic encephalopathy (CTE). These dementias share traumatic brain injury (TBI) as a prominent risk factor. Tau aggregates can transfer between cells and tissues in a "prion-like" manner, where they initiate the templated misfolding of normal tau molecules. This enables the spread of tau pathology to distinct parts of the brain. The evidence that tauopathies spread via prion-like mechanisms is considerable, but work detailing the mechanisms of spread has mostly used in vitro platforms that cannot fully reveal the tissue-level vectors or etiology of progression. We review these issues and then briefly use TBI and CTE as a case study to illustrate aspects of tauopathy that warrant further attention in vivo. These include seizures and sleep/wake disturbances, emphasizing the urgent need for improved animal models. Dissecting these mechanisms of tauopathy progression continues to provide fresh inspiration for the design of diagnostic and therapeutic approaches.
Collapse
Affiliation(s)
- Hadeel Alyenbaawi
- Centre for Prions & Protein Folding Disease, University of Alberta, Edmonton, AB T6G 2M8, Canada; (H.A.); (W.T.A.)
- Department of Medical Genetics, University of Alberta, Edmonton, AB T6G 2H7, Canada
- Department of Medical Laboratories, Majmaah University, Majmaah 11952, Saudi Arabia
| | - W. Ted Allison
- Centre for Prions & Protein Folding Disease, University of Alberta, Edmonton, AB T6G 2M8, Canada; (H.A.); (W.T.A.)
- Department of Medical Genetics, University of Alberta, Edmonton, AB T6G 2H7, Canada
- Department of Biological Sciences, University of Alberta, Edmonton, AB T6G 2E9, Canada
| | - Sue-Ann Mok
- Centre for Prions & Protein Folding Disease, University of Alberta, Edmonton, AB T6G 2M8, Canada; (H.A.); (W.T.A.)
- Department of Biochemistry, University of Alberta, Edmonton, AB T6G 2H7, Canada
| |
Collapse
|
35
|
C. elegans Models to Study the Propagation of Prions and Prion-Like Proteins. Biomolecules 2020; 10:biom10081188. [PMID: 32824215 PMCID: PMC7464663 DOI: 10.3390/biom10081188] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Revised: 08/10/2020] [Accepted: 08/13/2020] [Indexed: 12/14/2022] Open
Abstract
A hallmark common to many age-related neurodegenerative diseases, such as Alzheimer’s disease (AD), Parkinson’s disease (PD), and amyotrophic lateral sclerosis (ALS), is that patients develop proteinaceous deposits in their central nervous system (CNS). The progressive spreading of these inclusions from initially affected sites to interconnected brain areas is reminiscent of the behavior of bona fide prions in transmissible spongiform encephalopathies (TSEs), hence the term prion-like proteins has been coined. Despite intensive research, the exact mechanisms that facilitate the spreading of protein aggregation between cells, and the associated loss of neurons, remain poorly understood. As population demographics in many countries continue to shift to higher life expectancy, the incidence of neurodegenerative diseases is also rising. This represents a major challenge for healthcare systems and patients’ families, since patients require extensive support over several years and there is still no therapy to cure or stop these diseases. The model organism Caenorhabditis elegans offers unique opportunities to accelerate research and drug development due to its genetic amenability, its transparency, and the high degree of conservation of molecular pathways. Here, we will review how recent studies that utilize this soil dwelling nematode have proceeded to investigate the propagation and intercellular transmission of prions and prion-like proteins and discuss their relevance by comparing their findings to observations in other model systems and patients.
Collapse
|
36
|
Livingston G, Huntley J, Sommerlad A, Ames D, Ballard C, Banerjee S, Brayne C, Burns A, Cohen-Mansfield J, Cooper C, Costafreda SG, Dias A, Fox N, Gitlin LN, Howard R, Kales HC, Kivimäki M, Larson EB, Ogunniyi A, Orgeta V, Ritchie K, Rockwood K, Sampson EL, Samus Q, Schneider LS, Selbæk G, Teri L, Mukadam N. Dementia prevention, intervention, and care: 2020 report of the Lancet Commission. Lancet 2020; 396:413-446. [PMID: 32738937 PMCID: PMC7392084 DOI: 10.1016/s0140-6736(20)30367-6] [Citation(s) in RCA: 4852] [Impact Index Per Article: 1213.0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/03/2019] [Revised: 01/31/2020] [Accepted: 02/07/2020] [Indexed: 02/06/2023]
Affiliation(s)
- Gill Livingston
- Division of Psychiatry, University College London, London, UK; Camden and Islington NHS Foundation Trust, London, UK.
| | - Jonathan Huntley
- Division of Psychiatry, University College London, London, UK; Camden and Islington NHS Foundation Trust, London, UK
| | - Andrew Sommerlad
- Division of Psychiatry, University College London, London, UK; Camden and Islington NHS Foundation Trust, London, UK
| | - David Ames
- National Ageing Research Institute and Academic Unit for Psychiatry of Old Age, University of Melbourne, Royal Melbourne Hospital, Parkville, VIC, Australia
| | | | - Sube Banerjee
- Faculty of Health: Medicine, Dentistry and Human Sciences, University of Plymouth, Plymouth, UK
| | - Carol Brayne
- Cambridge Institute of Public Health, University of Cambridge, Cambridge, UK
| | - Alistair Burns
- Department of Old Age Psychiatry, University of Manchester, Manchester, UK
| | - Jiska Cohen-Mansfield
- Department of Health Promotion, School of Public Health, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel; Heczeg Institute on Aging, Tel Aviv University, Tel Aviv, Israel; Minerva Center for Interdisciplinary Study of End of Life, Tel Aviv University, Tel Aviv, Israel
| | - Claudia Cooper
- Division of Psychiatry, University College London, London, UK; Camden and Islington NHS Foundation Trust, London, UK
| | - Sergi G Costafreda
- Division of Psychiatry, University College London, London, UK; Camden and Islington NHS Foundation Trust, London, UK
| | - Amit Dias
- Department of Preventive and Social Medicine, Goa Medical College, Goa, India
| | - Nick Fox
- Dementia Research Centre, UK Dementia Research Institute, University College London, London, UK; Institute of Neurology, National Hospital for Neurology and Neurosurgery, University College London Hospitals NHS Foundation Trust, London, UK
| | - Laura N Gitlin
- Center for Innovative Care in Aging, Johns Hopkins University, Baltimore, MA, USA
| | - Robert Howard
- Division of Psychiatry, University College London, London, UK; Camden and Islington NHS Foundation Trust, London, UK
| | - Helen C Kales
- Department of Psychiatry and Behavioral Sciences, UC Davis School of Medicine, University of California, Sacramento, CA, USA
| | - Mika Kivimäki
- Department of Epidemiology and Public Health, University College London, London, UK
| | - Eric B Larson
- Kaiser Permanente Washington Health Research Institute, Seattle, WA, USA
| | | | - Vasiliki Orgeta
- Division of Psychiatry, University College London, London, UK
| | - Karen Ritchie
- Inserm, Unit 1061, Neuropsychiatry: Epidemiological and Clinical Research, La Colombière Hospital, University of Montpellier, Montpellier, France; Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, UK
| | - Kenneth Rockwood
- Centre for the Health Care of Elderly People, Geriatric Medicine Dalhousie University, Halifax, NS, Canada
| | - Elizabeth L Sampson
- Division of Psychiatry, University College London, London, UK; Barnet, Enfield, and Haringey Mental Health Trust, London, UK
| | - Quincy Samus
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University, Baltimore, MA, USA
| | - Lon S Schneider
- Department of Psychiatry and the Behavioural Sciences and Department of Neurology, Keck School of Medicine, Leonard Davis School of Gerontology of the University of Southern California, Los Angeles, CA, USA
| | - Geir Selbæk
- Norwegian National Advisory Unit on Ageing and Health, Vestfold Hospital Trust, Tønsberg, Norway; Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway; Geriatric Department, Oslo University Hospital, Oslo, Norway
| | - Linda Teri
- Department Psychosocial and Community Health, School of Nursing, University of Washington, Seattle, WA, USA
| | - Naaheed Mukadam
- Division of Psychiatry, University College London, London, UK; Camden and Islington NHS Foundation Trust, London, UK
| |
Collapse
|
37
|
Iverson GL, Luoto TM, Karhunen PJ, Castellani RJ. Mild Chronic Traumatic Encephalopathy Neuropathology in People With No Known Participation in Contact Sports or History of Repetitive Neurotrauma. J Neuropathol Exp Neurol 2020; 78:615-625. [PMID: 31169877 DOI: 10.1093/jnen/nlz045] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
It has been asserted that chronic traumatic encephalopathy (CTE) pathology is only present in former athletes and others who have been exposed to repetitive concussions, subconcussive blows, or both. We hypothesized that CTE pathology would be present in men who had no known history of repetitive neurotrauma. Comprehensive medical record reviews and health surveys completed by a family member were available for the 8 men in this case series, none of whom had known exposure to repetitive neurotrauma but 2 of whom had a history of traumatic brain injury (TBI). Postmortem tissue was immunostained for hyperphosphorylated tau (p-tau) to assess for CTE pathology, Braak stage, and aging-related p-tau. The neuropathologist was blind to age, personal history, and clinical history. Six of the 8 cases (75%) showed p-tau in neurons, astrocytes, and cell processes around small blood vessels in an irregular pattern at the depths of the cortical sulci. The changes were focal and limited in terms of overall extent, and some of the cases had a clearer pattern of pathology and some could be considered equivocal. Two of the 8 cases had a history of TBI and one of them showed CTE pathology. Five of the 6 cases with no known history of neurotrauma appeared to meet consensus criteria for CTE. This study adds to the emerging literature indicating that CTE pathology is present in people not known to have experienced multiple concussions or subconcussive blows to the head.
Collapse
Affiliation(s)
- Grant L Iverson
- Department of Physical Medicine and Rehabilitation, Harvard Medical School.,Spaulding Rehabilitation Hospital, Spaulding Research Institute.,MassGeneral Hospital for Children™ Sports Concussion Program.,Home Base, A Red Sox Foundation and Massachusetts General Hospital Program, Boston, Massachusetts
| | - Teemu M Luoto
- Department of Neurosurgery, Tampere University Hospital and University of Tampere, Tampere, Finland
| | - Pekka J Karhunen
- Department of Forensic Medicine, Faculty of Medicine and Life Sciences, University of Tampere.,Fimlab Laboratories and Finnish Cardiovascular Research Center Tampere, Tampere, Finland
| | - Rudolph J Castellani
- Department of Pathology, Anatomy and Laboratory Medicine.,Department of Neuroscience, Rockefeller Neuroscience Institute.,West Virginia University School of Medicine, Morgantown, West Virginia
| |
Collapse
|
38
|
Zhou Y, Chen Q, Wang Y, Wu H, Xu W, Pan Y, Gao S, Dong X, Zhang JH, Shao A. Persistent Neurovascular Unit Dysfunction: Pathophysiological Substrate and Trigger for Late-Onset Neurodegeneration After Traumatic Brain Injury. Front Neurosci 2020; 14:581. [PMID: 32581697 PMCID: PMC7296179 DOI: 10.3389/fnins.2020.00581] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2020] [Accepted: 05/12/2020] [Indexed: 12/14/2022] Open
Abstract
Traumatic brain injury (TBI) represents one of the major causes of death worldwide and leads to persisting neurological deficits in many of the survivors. One of the most significant long-term sequelae deriving from TBI is neurodegenerative disease, which is a group of incurable diseases that impose a heavy socio-economic burden. However, mechanisms underlying the increased susceptibility of TBI to neurodegenerative disease remain elusive. The neurovascular unit (NVU) is a functional unit composed of neurons, neuroglia, vascular cells, and the basal lamina matrix. The key role of NVU dysfunction in many central nervous system diseases has been revealed. Studies have proved the presence of prolonged structural and functional abnormalities of the NVU after TBI. Moreover, growing evidence suggests impaired NVU function is also implicated in neurodegenerative diseases. Therefore, we propose the Neurovascular Unit Dysfunction (NVUD) Hypothesis, in which the persistent NVU dysfunction is thought to underlie the development of post-TBI neurodegeneration. We deduce NVUD Hypothesis through relational inference and supporting evidence, and suggest continued NVU abnormalities following TBI serve as the pathophysiological substrate and trigger yielding chronic neuroinflammation, proteinopathies and oxidative stress, consequently leading to the progression of neurodegenerative diseases. The NVUD Hypothesis may provide potential treatment and prevention strategies for TBI and late-onset neurodegenerative diseases.
Collapse
Affiliation(s)
- Yunxiang Zhou
- Department of Surgical Oncology, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Qiang Chen
- Department of Surgical Oncology, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Yali Wang
- Department of Surgical Oncology, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Haijian Wu
- Department of Neurosurgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Weilin Xu
- Department of Neurosurgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Yuanbo Pan
- Department of Neurosurgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Shiqi Gao
- Department of Neurosurgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Xiao Dong
- Department of Neurosurgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - John H. Zhang
- Department of Physiology and Pharmacology, Basic Sciences, School of Medicine, Loma Linda University, Loma Linda, CA, United States
- Department of Anesthesiology, Neurosurgery and Neurology, School of Medicine, Loma Linda University, Loma Linda, CA, United States
| | - Anwen Shao
- Department of Neurosurgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| |
Collapse
|
39
|
Iverson GL, Gardner AJ, Shultz SR, Solomon GS, McCrory P, Zafonte R, Perry G, Hazrati LN, Keene CD, Castellani RJ. Chronic traumatic encephalopathy neuropathology might not be inexorably progressive or unique to repetitive neurotrauma. Brain 2020; 142:3672-3693. [PMID: 31670780 PMCID: PMC6906593 DOI: 10.1093/brain/awz286] [Citation(s) in RCA: 51] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2019] [Revised: 07/01/2019] [Accepted: 07/15/2019] [Indexed: 12/11/2022] Open
Abstract
In the 20th century, chronic traumatic encephalopathy (CTE) was conceptualized as a neurological disorder affecting some active and retired boxers who had tremendous exposure to neurotrauma. In recent years, the two research groups in the USA who have led the field have asserted definitively that CTE is a delayed-onset and progressive neurodegenerative disease, with symptoms appearing in midlife or decades after exposure. Between 2005 and 2012 autopsy cases of former boxers and American football players described neuropathology attributed to CTE that was broad and diverse. This pathology, resulting from multiple causes, was aggregated and referred to, in toto, as the pathology ‘characteristic’ of CTE. Preliminary consensus criteria for defining the neuropathology of CTE were forged in 2015 and published in 2016. Most of the macroscopic and microscopic neuropathological findings described as characteristic of CTE, in studies published before 2016, were not included in the new criteria for defining the pathology. In the past few years, there has been steadily emerging evidence that the neuropathology described as unique to CTE may not be unique. CTE pathology has been described in individuals with no known participation in collision or contact sports and no known exposure to repetitive neurotrauma. This pathology has been reported in individuals with substance abuse, temporal lobe epilepsy, amyotrophic lateral sclerosis, multiple system atrophy, and other neurodegenerative diseases. Moreover, throughout history, some clinical cases have been described as not being progressive, and there is now evidence that CTE neuropathology might not be progressive in some individuals. Considering the current state of knowledge, including the absence of a series of validated sensitive and specific biomarkers, CTE pathology might not be inexorably progressive or specific to those who have experienced repetitive neurotrauma.
Collapse
Affiliation(s)
- Grant L Iverson
- Department of Physical Medicine and Rehabilitation, Harvard Medical School, Boston, Massachusetts, USA.,Spaulding Rehabilitation Hospital and Spaulding Research Institute, Boston, Massachusetts, USA.,MassGeneral Hospital for Children™ Sports Concussion Program, Boston, Massachusetts, USA.,Home Base, A Red Sox Foundation and Massachusetts General Hospital Program, Boston, Massachusetts, USA
| | - Andrew J Gardner
- Hunter New England Local Health District, Sports Concussion Program, University of Newcastle, Callaghan, NSW, Australia.,Centre for Stroke and Brain Injury, School of Medicine and Public Health, University of Newcastle, Callaghan, NSW, Australia
| | - Sandy R Shultz
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, VIC, Australia
| | - Gary S Solomon
- Department of Neurological Surgery, Orthopaedic Surgery and Rehabilitation, Vanderbilt University School of Medicine, Nashville, Tennessee, USA.,Department of Psychiatry and Behavioral Sciences, Vanderbilt University School of Medicine, Nashville, Tennessee, USA.,Vanderbilt Sports Concussion Center, Vanderbilt University School of Medicine, Nashville, Tennessee, USA
| | - Paul McCrory
- The Florey Institute of Neuroscience and Mental Health, Melbourne Brain Centre - Austin Campus, Heidelberg, Victoria Australia
| | - Ross Zafonte
- Department of Physical Medicine and Rehabilitation, Harvard Medical School, Boston, Massachusetts, USA.,Spaulding Rehabilitation Hospital and Spaulding Research Institute, Boston, Massachusetts, USA.,Home Base, A Red Sox Foundation and Massachusetts General Hospital Program, Boston, Massachusetts, USA
| | - George Perry
- College of Sciences, University of Texas, San Antonio; San Antonio, Texas, USA
| | - Lili-Naz Hazrati
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada
| | - C Dirk Keene
- Department of Pathology, Division of Neuropathology, University of Washington School of Medicine, Seattle, Washington, USA
| | - Rudolph J Castellani
- Department of Pathology, Anatomy and Laboratory Medicine, West Virginia University School of Medicine, Morgantown, USA.,Department of Neuroscience, Rockefeller Neuroscience Institute, West Virginia University School of Medicine, Morgantown, USA
| |
Collapse
|
40
|
Arena JD, Smith DH, Lee EB, Gibbons GS, Irwin DJ, Robinson JL, Lee VMY, Trojanowski JQ, Stewart W, Johnson VE. Tau immunophenotypes in chronic traumatic encephalopathy recapitulate those of ageing and Alzheimer's disease. Brain 2020; 143:1572-1587. [PMID: 32390044 PMCID: PMC7241956 DOI: 10.1093/brain/awaa071] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2019] [Revised: 01/21/2020] [Accepted: 02/02/2020] [Indexed: 12/14/2022] Open
Abstract
Traumatic brain injury (TBI) is a risk factor for neurodegenerative disease, including chronic traumatic encephalopathy (CTE). Preliminary consensus criteria define the pathognomonic lesion of CTE as patchy tau pathology within neurons and astrocytes at the depths of cortical sulci. However, the specific tau isoform composition and post-translational modifications in CTE remain largely unexplored. Using immunohistochemistry, we performed tau phenotyping of CTE neuropathologies and compared this to a range of tau pathologies, including Alzheimer's disease, primary age-related tauopathy, ageing-related tau astrogliopathy and multiple subtypes of frontotemporal lobar degeneration with tau inclusions. Cases satisfying preliminary consensus diagnostic criteria for CTE neuropathological change (CTE-NC) were identified (athletes, n = 10; long-term survivors of moderate or severe TBI, n = 4) from the Glasgow TBI Archive and Penn Neurodegenerative Disease Brain Bank. In addition, material from a range of autopsy-proven ageing-associated and primary tauopathies in which there was no known history of exposure to TBI was selected as non-injured controls (n = 32). Each case was then stained with a panel of tau antibodies specific for phospho-epitopes (PHF1, CP13, AT100, pS262), microtubule-binding repeat domains (3R, 4R), truncation (Tau-C3) or conformation (GT-7, GT-38) and the extent and distribution of staining assessed. Cell types were confirmed with double immunofluorescent labelling. Results demonstrate that astroglial tau pathology in CTE is composed of 4R-immunoreactive thorn-shaped astrocytes, echoing the morphology and immunophenotype of astrocytes encountered in ageing-related tau astrogliopathy. In contrast, neurofibrillary tangles of CTE contain both 3R and 4R tau, with post-translational modifications and conformations consistent with Alzheimer's disease and primary age-related tauopathy. Our observations establish that the astroglial and neurofibrillary tau pathologies of CTE are phenotypically distinct from each other and recapitulate the tau immunophenotypes encountered in ageing and Alzheimer's disease. As such, the immunohistochemical distinction of CTE neuropathology from other mixed 3R/4R tauopathies of Alzheimer's disease and ageing may rest solely on the pattern and distribution of pathology.
Collapse
Affiliation(s)
- John D Arena
- Department of Neurosurgery, Penn Center for Brain Injury and Repair, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Douglas H Smith
- Department of Neurosurgery, Penn Center for Brain Injury and Repair, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Edward B Lee
- Department of Pathology and Laboratory Medicine, Center for Neurodegenerative Disease Research, Perelman School of Medicine, University of Pennsylvania, Philadelphia, USA
- Translational Neuropathology Research Laboratory, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Garrett S Gibbons
- Department of Pathology and Laboratory Medicine, Center for Neurodegenerative Disease Research, Perelman School of Medicine, University of Pennsylvania, Philadelphia, USA
| | - David J Irwin
- Department of Neurology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - John L Robinson
- Department of Pathology and Laboratory Medicine, Center for Neurodegenerative Disease Research, Perelman School of Medicine, University of Pennsylvania, Philadelphia, USA
| | - Virginia M -Y Lee
- Department of Pathology and Laboratory Medicine, Center for Neurodegenerative Disease Research, Perelman School of Medicine, University of Pennsylvania, Philadelphia, USA
| | - John Q Trojanowski
- Department of Pathology and Laboratory Medicine, Center for Neurodegenerative Disease Research, Perelman School of Medicine, University of Pennsylvania, Philadelphia, USA
| | - William Stewart
- Department of Neuropathology, Queen Elizabeth University Hospital, Glasgow G51 4TF, UK
- Institute of Neuroscience and Psychology, University of Glasgow, Glasgow, G12 8QQ, UK
| | - Victoria E Johnson
- Department of Neurosurgery, Penn Center for Brain Injury and Repair, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| |
Collapse
|
41
|
Brunello CA, Merezhko M, Uronen RL, Huttunen HJ. Mechanisms of secretion and spreading of pathological tau protein. Cell Mol Life Sci 2020; 77:1721-1744. [PMID: 31667556 PMCID: PMC7190606 DOI: 10.1007/s00018-019-03349-1] [Citation(s) in RCA: 163] [Impact Index Per Article: 40.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2019] [Revised: 10/10/2019] [Accepted: 10/16/2019] [Indexed: 12/12/2022]
Abstract
Accumulation of misfolded and aggregated forms of tau protein in the brain is a neuropathological hallmark of tauopathies, such as Alzheimer's disease and frontotemporal lobar degeneration. Tau aggregates have the ability to transfer from one cell to another and to induce templated misfolding and aggregation of healthy tau molecules in previously healthy cells, thereby propagating tau pathology across different brain areas in a prion-like manner. The molecular mechanisms involved in cell-to-cell transfer of tau aggregates are diverse, not mutually exclusive and only partially understood. Intracellular accumulation of misfolded tau induces several mechanisms that aim to reduce the cellular burden of aggregated proteins and also promote secretion of tau aggregates. However, tau may also be released from cells physiologically unrelated to protein aggregation. Tau secretion involves multiple vesicular and non-vesicle-mediated pathways, including secretion directly through the plasma membrane. Consequently, extracellular tau can be found in various forms, both as a free protein and in vesicles, such as exosomes and ectosomes. Once in the extracellular space, tau aggregates can be internalized by neighboring cells, both neurons and glial cells, via endocytic, pinocytic and phagocytic mechanisms. Importantly, accumulating evidence suggests that prion-like propagation of misfolding protein pathology could provide a general mechanism for disease progression in tauopathies and other related neurodegenerative diseases. Here, we review the recent literature on cellular mechanisms involved in cell-to-cell transfer of tau, with a particular focus in tau secretion.
Collapse
Affiliation(s)
- Cecilia A Brunello
- Neuroscience Center, HiLIFE, University of Helsinki, P.O. Box 63, Haartmaninkatu 8, 00014, Helsinki, Finland
| | - Maria Merezhko
- Neuroscience Center, HiLIFE, University of Helsinki, P.O. Box 63, Haartmaninkatu 8, 00014, Helsinki, Finland
| | - Riikka-Liisa Uronen
- Neuroscience Center, HiLIFE, University of Helsinki, P.O. Box 63, Haartmaninkatu 8, 00014, Helsinki, Finland
| | - Henri J Huttunen
- Neuroscience Center, HiLIFE, University of Helsinki, P.O. Box 63, Haartmaninkatu 8, 00014, Helsinki, Finland.
| |
Collapse
|
42
|
Cell-to-Cell Transmission of Tau and α-Synuclein. Trends Mol Med 2020; 26:936-952. [PMID: 32371172 DOI: 10.1016/j.molmed.2020.03.012] [Citation(s) in RCA: 87] [Impact Index Per Article: 21.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2020] [Revised: 03/27/2020] [Accepted: 03/31/2020] [Indexed: 12/20/2022]
Abstract
The stereotypical spread of pathological protein inclusions and clinicopathological heterogeneity are well described in neurodegenerative diseases. Accumulating evidence suggests that the former can be attributed to consecutive cell-to-cell transmission of pathological proteins between anatomically connected brain regions, while the latter has been hypothesized to result from the spread of conformationally distinct pathological protein aggregates, or strains. These emerging concepts have dramatically changed our understanding of neurodegenerative diseases. In this review, we first summarize the background and recent findings underpinning these concepts with a focus on two major pathological proteins: tau and α-synuclein. We then discuss their clinical implications for tauopathies and synucleinopathies and propose a working hypothesis for future research.
Collapse
|
43
|
Li M, Reisman J, Morris-Eppolito B, Qian SX, Kazis LE, Wolozin B, Goldstein LE, Xia W. Beneficial association of angiotensin-converting enzyme inhibitors and statins on the occurrence of possible Alzheimer's disease after traumatic brain injury. ALZHEIMERS RESEARCH & THERAPY 2020; 12:33. [PMID: 32220235 PMCID: PMC7102441 DOI: 10.1186/s13195-020-00589-3] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/13/2019] [Accepted: 02/25/2020] [Indexed: 12/31/2022]
Abstract
BACKGROUND Pathological analysis of brain tissue from animals and humans with a history of traumatic brain injury (TBI) suggests that TBI could be one of the risk factors facilitating onset of dementia with possible Alzheimer's disease (AD), but medications to prevent or delay AD onset are not yet available. METHODS This study explores four medication classes (angiotensin-converting enzyme inhibitors (ACEI), beta blockers, metformin, and statins) approved by the Food and Drug Administration (FDA) for other indications and evaluates their influence when used in combination on the risk of possible AD development for patients with a history of TBI. We identified patients with history of TBI from an existing Department of Veterans Affairs (VA) national database. Among 1,660,151 veterans who used VA services between the ages of 50 to 89 years old, we analyzed 733,920 patients, including 15,450 patients with a history of TBI and 718,470 non-TBI patients. The TBI patients were followed for up to 18.5 years, with an average of 7.7 ± 4.7 years, and onset of dementia with possible AD was recorded based on International Statistical Classification of Diseases (ICD) 9 or 10 codes. The effect of TBI on possible AD development was evaluated by multivariable logistic regression models adjusted by age, gender, race, and other comorbidities. The association of ACEI, beta blockers, metformin, statins, and combinations of these agents over time from the first occurrence of TBI to possible AD onset was assessed using Cox proportional hazard models adjusted for demographics and comorbidities. RESULTS Veterans with at least two TBI occurrences by claims data were 25% (odds ratio (OR) = 1.25, 95% confidence intervals (CI) (1.13, 1.37)) more likely to develop dementia with possible AD, compared to those with no record of TBI. In multivariable logistic regression models (propensity score weighted or adjusted), veterans taking a combination of ACEI and statins had reduced risk in developing possible AD after suffering TBI, and use of this medication class combination was associated with a longer period between TBI occurring and dementia with possible AD onset, compared to patients who took statins alone or did not take any of the four target drugs after TBI. CONCLUSIONS The combination of ACEI and statins significantly lowered the risk of development of dementia with possible AD in a national cohort of people with a history of TBI, thus supporting a clinical approach to lowering the risk of dementia with possible AD.
Collapse
Affiliation(s)
- Mingfei Li
- Center for Healthcare Organization and Implementation Research, Edith Nourse Rogers Memorial Hospital, Bedford, MA, USA.,Department of Mathematical Sciences, Bentley University, Waltham, MA, USA
| | - Joel Reisman
- Center for Healthcare Organization and Implementation Research, Edith Nourse Rogers Memorial Hospital, Bedford, MA, USA
| | - Benjamin Morris-Eppolito
- Geriatric Research Education Clinical Center, Edith Nourse Rogers Memorial Veterans Hospital, Bedford, MA, 01730, USA
| | - Shirley X Qian
- Center for Healthcare Organization and Implementation Research, Edith Nourse Rogers Memorial Hospital, Bedford, MA, USA.,Department of Health Law, Policy and Management, Boston University School of Public Health, Boston, MA, USA
| | - Lewis E Kazis
- Center for Healthcare Organization and Implementation Research, Edith Nourse Rogers Memorial Hospital, Bedford, MA, USA.,Department of Health Law, Policy and Management, Boston University School of Public Health, Boston, MA, USA
| | - Benjamin Wolozin
- Department of Pharmacology and Experimental Therapeutics, Boston University School of Medicine, Boston, MA, USA
| | - Lee E Goldstein
- Departments of Radiology, Psychiatry, Neurology, and Pathology, Boston University School of Medicine, Boston, MA, USA.,Departments of Biomedical, Electrical, and Computer Engineering, Boston University College of Engineering & Photonics Center, Boston, MA, USA.,Boston University Alzheimer's Disease Center, Boston, MA, USA
| | - Weiming Xia
- Geriatric Research Education Clinical Center, Edith Nourse Rogers Memorial Veterans Hospital, Bedford, MA, 01730, USA. .,Department of Pharmacology and Experimental Therapeutics, Boston University School of Medicine, Boston, MA, USA.
| |
Collapse
|
44
|
Graham NS, Sharp DJ. Understanding neurodegeneration after traumatic brain injury: from mechanisms to clinical trials in dementia. J Neurol Neurosurg Psychiatry 2019; 90:1221-1233. [PMID: 31542723 PMCID: PMC6860906 DOI: 10.1136/jnnp-2017-317557] [Citation(s) in RCA: 180] [Impact Index Per Article: 36.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/13/2019] [Revised: 07/31/2019] [Accepted: 07/31/2019] [Indexed: 12/12/2022]
Abstract
Traumatic brain injury (TBI) leads to increased rates of dementia, including Alzheimer's disease. The mechanisms by which trauma can trigger neurodegeneration are increasingly understood. For example, diffuse axonal injury is implicated in disrupting microtubule function, providing the potential context for pathologies of tau and amyloid to develop. The neuropathology of post-traumatic dementias is increasingly well characterised, with recent work focusing on chronic traumatic encephalopathy (CTE). However, clinical diagnosis of post-traumatic dementia is problematic. It is often difficult to disentangle the direct effects of TBI from those produced by progressive neurodegeneration or other post-traumatic sequelae such as psychiatric impairment. CTE can only be confidently identified at postmortem and patients are often confused and anxious about the most likely cause of their post-traumatic problems. A new approach to the assessment of the long-term effects of TBI is needed. Accurate methods are available for the investigation of other neurodegenerative conditions. These should be systematically employed in TBI. MRI and positron emission tomography neuroimaging provide biomarkers of neurodegeneration which may be of particular use in the postinjury setting. Brain atrophy is a key measure of disease progression and can be used to accurately quantify neuronal loss. Fluid biomarkers such as neurofilament light can complement neuroimaging, representing sensitive potential methods to track neurodegenerative processes that develop after TBI. These biomarkers could characterise endophenotypes associated with distinct types of post-traumatic neurodegeneration. In addition, they might profitably be used in clinical trials of neuroprotective and disease-modifying treatments, improving trial design by providing precise and sensitive measures of neuronal loss.
Collapse
Affiliation(s)
- Neil Sn Graham
- Brain Sciences, Imperial College London, London, UK.,UK DRI Care Research & Technology Centre, Imperial College London, London, United Kingdom
| | - David J Sharp
- Brain Sciences, Imperial College London, London, UK .,UK DRI Care Research & Technology Centre, Imperial College London, London, United Kingdom.,The Royal British Legion Centre for Blast Injury Studies, Department of Bioengineering, Imperial College London, London, United Kingdom
| |
Collapse
|
45
|
Katsumoto A, Takeuchi H, Tanaka F. Tau Pathology in Chronic Traumatic Encephalopathy and Alzheimer's Disease: Similarities and Differences. Front Neurol 2019; 10:980. [PMID: 31551922 PMCID: PMC6748163 DOI: 10.3389/fneur.2019.00980] [Citation(s) in RCA: 84] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2019] [Accepted: 08/28/2019] [Indexed: 12/14/2022] Open
Abstract
Traumatic brain injury (TBI) has been associated with the development of Alzheimer's disease (AD) because these conditions share common pathological hallmarks: amyloid-β and hyperphosphorylated tau accumulation. However, given recent data it is uncertain if a history of TBI leads to the development of AD. Moreover, chronic traumatic encephalopathy (CTE), caused by repetitive mild TBI and characterized by progressive neurodegeneration with hyperphosphorylated tau, has come to be recognized as distinct from AD. Therefore, it is important to elucidate the clinical outcomes and molecular mechanisms underlying tau pathology following TBI. We summarize the histopathological features and clinical course of TBI in CTE, comparing the tau pathology with that in AD. Following brain injury, diffuse axonal injury, and hyperphosphorylated tau aggregates are observed within a shorter period than in AD. Hyperphosphorylated tau deposition usually begins in the perivascular area of the sulci in the cerebral cortex, then spreads unevenly in the cortex in CTE, while AD shows diffuse distribution of hyperphosphorylated tau in the cortical areas. We also highlight the molecular profile of tau and the implications of tau progression throughout the brain in both diseases. Tau contains phosphorylation sites common to both conditions. In particular, phosphorylation at Thr231 triggers a conformational change to the toxic cis form of tau, which is suggested to drive neurodegeneration. Although the mechanism of rapid tau accumulation remains unknown, the structural diversity of tau might result in these different outcomes. Finally, future perspectives on CTE in terms of tau reduction are discussed.
Collapse
Affiliation(s)
- Atsuko Katsumoto
- Department of Neurology and Stroke Medicine, Graduate School of Medicine, Yokohama City University, Yokohama, Japan
| | - Hideyuki Takeuchi
- Department of Neurology and Stroke Medicine, Graduate School of Medicine, Yokohama City University, Yokohama, Japan
| | - Fumiaki Tanaka
- Department of Neurology and Stroke Medicine, Graduate School of Medicine, Yokohama City University, Yokohama, Japan
| |
Collapse
|
46
|
Lee EB, Kinch K, Johnson VE, Trojanowski JQ, Smith DH, Stewart W. Chronic traumatic encephalopathy is a common co-morbidity, but less frequent primary dementia in former soccer and rugby players. Acta Neuropathol 2019; 138:389-399. [PMID: 31152201 PMCID: PMC6689293 DOI: 10.1007/s00401-019-02030-y] [Citation(s) in RCA: 80] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2019] [Accepted: 05/25/2019] [Indexed: 02/07/2023]
Abstract
Chronic traumatic encephalopathy (CTE) is reported at high prevalence in selected autopsy case series of former contact sports athletes. Nevertheless, the contribution of CTE pathology to clinical presentation and its interaction with co-morbid neurodegenerative pathologies remain unclear. To address these issues, we performed comprehensive neuropathology assessments on the brains of former athletes with dementia and considered these findings together with detailed clinical histories to derive an integrated clinicopathological diagnosis for each case. Consecutive, autopsy-acquired brains from former soccer and rugby players with dementia were assessed for neurodegenerative pathologies using established and preliminary consensus protocols. Thereafter, next of kin interviews were conducted to obtain detailed accounts of the patient’s clinical presentation and course of disease to inform a final, integrated clinicopathological diagnosis. Neuropathologic change consistent with CTE (CTE-NC) was confirmed in five of seven former soccer and three of four former rugby players’ brains, invariably in combination with mixed, often multiple neurodegenerative pathologies. However, in just three cases was the integrated dementia diagnosis consistent with CTE, the remainder having alternate diagnoses, with the most frequent integrated diagnosis Alzheimer’s disease (AD) (four cases; one as mixed AD and vascular dementia). This consecutive autopsy series identifies neuropathologic change consistent with preliminary diagnostic criteria for CTE (CTE-NC) in a high proportion of former soccer and rugby players dying with dementia. However, in the majority, CTE-NC appears as a co-morbidity rather than the primary, dementia causing pathology. As such, we suggest that while CTE-NC might be common in former athletes with dementia, in many cases its clinical significance remains uncertain.
Collapse
Affiliation(s)
- Edward B Lee
- Translational Neuropathology Research Laboratory, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Kevin Kinch
- Department of Neuropathology, Queen Elizabeth University Hospital, 1345 Govan Rd, Glasgow, G51 4TF, UK
| | - Victoria E Johnson
- Department of Neurosurgery, Penn Center for Brain Injury and Repair, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - John Q Trojanowski
- Department of Pathology and Laboratory Medicine, Center for Neurodegenerative Disease Research, Institute on Aging, Perelman School of Medicine, University of Pennsylvania, Philadelphia, USA
| | - Douglas H Smith
- Department of Neurosurgery, Penn Center for Brain Injury and Repair, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - William Stewart
- Department of Neuropathology, Queen Elizabeth University Hospital, 1345 Govan Rd, Glasgow, G51 4TF, UK.
- Institute of Neuroscience and Psychology, University of Glasgow, Glasgow, G12 8QQ, UK.
| |
Collapse
|
47
|
|
48
|
Erdener ŞE, Dalkara T. Small Vessels Are a Big Problem in Neurodegeneration and Neuroprotection. Front Neurol 2019; 10:889. [PMID: 31474933 PMCID: PMC6707104 DOI: 10.3389/fneur.2019.00889] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2019] [Accepted: 08/01/2019] [Indexed: 12/11/2022] Open
Abstract
The cerebral microcirculation holds a critical position to match the high metabolic demand by neuronal activity. Functionally, microcirculation is virtually inseparable from other nervous system cells under both physiological and pathological conditions. For successful bench-to-bedside translation of neuroprotection research, the role of microcirculation in acute and chronic neurodegenerative disorders appears to be under-recognized, which may have contributed to clinical trial failures with some neuroprotectants. Increasing data over the last decade suggest that microcirculatory impairments such as endothelial or pericyte dysfunction, morphological irregularities in capillaries or frequent dynamic stalls in blood cell flux resulting in excessive heterogeneity in capillary transit may significantly compromise tissue oxygen availability. We now know that ischemia-induced persistent abnormalities in capillary flow negatively impact restoration of reperfusion after recanalization of occluded cerebral arteries. Similarly, microcirculatory impairments can accompany or even precede neural loss in animal models of several neurodegenerative disorders including Alzheimer's disease. Macrovessels are relatively easy to evaluate with radiological or experimental imaging methods but they cannot faithfully reflect the downstream microcirculatory disturbances, which may be quite heterogeneous across the tissue at microscopic scale and/or happen fast and transiently. The complexity and size of the elements of microcirculation, therefore, require utilization of cutting-edge imaging techniques with high spatiotemporal resolution as well as multidisciplinary team effort to disclose microvascular-neurodegenerative connection and to test treatment approaches to advance the field. Developments in two photon microscopy, ultrafast ultrasound, and optical coherence tomography provide valuable experimental tools to reveal those microscopic events with high resolution. Here, we review the up-to-date advances in understanding of the primary microcirculatory abnormalities that can result in neurodegenerative processes and the combined neurovascular protection approaches that can prevent acute as well as chronic neurodegeneration.
Collapse
Affiliation(s)
- Şefik Evren Erdener
- Institute of Neurological Sciences and Psychiatry, Hacettepe University, Ankara, Turkey
| | - Turgay Dalkara
- Institute of Neurological Sciences and Psychiatry, Hacettepe University, Ankara, Turkey.,Department of Neurology, Faculty of Medicine, Hacettepe University, Ankara, Turkey
| |
Collapse
|
49
|
Vogels T, Murgoci AN, Hromádka T. Intersection of pathological tau and microglia at the synapse. Acta Neuropathol Commun 2019; 7:109. [PMID: 31277708 PMCID: PMC6612163 DOI: 10.1186/s40478-019-0754-y] [Citation(s) in RCA: 116] [Impact Index Per Article: 23.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2019] [Accepted: 06/19/2019] [Indexed: 02/07/2023] Open
Abstract
Tauopathies are a heterogenous class of diseases characterized by cellular accumulation of aggregated tau and include diseases such as Alzheimer’s disease (AD), progressive supranuclear palsy and chronic traumatic encephalopathy. Tau pathology is strongly linked to neurodegeneration and clinical symptoms in tauopathy patients. Furthermore, synapse loss is an early pathological event in tauopathies and is the strongest correlate of cognitive decline. Tau pathology is additionally associated with chronic neuroinflammatory processes, such as reactive microglia, astrocytes, and increased levels of pro-inflammatory molecules (e.g. complement proteins, cytokines). Recent studies show that as the principal immune cells of the brain, microglia play a particularly important role in the initiation and progression of tau pathology and associated neurodegeneration. Furthermore, AD risk genes such as Triggering receptor expressed on myeloid cells 2 (TREM2) and Apolipoprotein E (APOE) are enriched in the innate immune system and modulate the neuroinflammatory response of microglia to tau pathology. Microglia can play an active role in synaptic dysfunction by abnormally phagocytosing synaptic compartments of neurons with tau pathology. Furthermore, microglia are involved in synaptic spreading of tau – a process which is thought to underlie the progressive nature of tau pathology propagation through the brain. Spreading of pathological tau is also the predominant target for tau-based immunotherapy. Active tau vaccines, therapeutic tau antibodies and other approaches targeting the immune system are actively explored as treatment options for AD and other tauopathies. This review describes the role of microglia in the pathobiology of tauopathies and the mechanism of action of potential therapeutics targeting the immune system in tauopathies.
Collapse
|
50
|
Sastre M, Van Leuven F, Gentleman SM. TauBI or not TauBI: what was the question? Brain 2018; 141:2536-2539. [PMID: 30169590 DOI: 10.1093/brain/awy225] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Affiliation(s)
| | - Fred Van Leuven
- Experimental Genetics Group-LEGTEGG, Department of Human Genetics, KU Leuven, Leuven, Belgium
| | | |
Collapse
|