1
|
Ghosh U, Tse E, Yang H, Shi M, Caro CD, Wang F, Merz GE, Prusiner SB, Southworth DR, Condello C. Cryo-EM structures reveal tau filaments from Down syndrome adopt Alzheimer's disease fold. Acta Neuropathol Commun 2024; 12:94. [PMID: 38867338 PMCID: PMC11167798 DOI: 10.1186/s40478-024-01806-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2024] [Accepted: 05/21/2024] [Indexed: 06/14/2024] Open
Abstract
Down syndrome (DS) is a common genetic condition caused by trisomy of chromosome 21. Among their complex clinical features, including musculoskeletal, neurological, and cardiovascular disabilities, individuals with DS have an increased risk of developing progressive dementia and early-onset Alzheimer's disease (AD). This dementia is attributed to the increased gene dosage of the amyloid-β (Aβ) precursor protein gene, the formation of self-propagating Aβ and tau prion conformers, and the deposition of neurotoxic Aβ plaques and tau neurofibrillary tangles. Tau amyloid fibrils have previously been established to adopt many distinct conformations across different neurodegenerative conditions. Here, we report the characterization of brain samples from four DS cases spanning 36-63 years of age by spectral confocal imaging with conformation-specific dyes and cryo-electron microscopy (cryo-EM) to determine structures of isolated tau fibrils. High-resolution structures revealed paired helical filament (PHF) and straight filament (SF) conformations of tau that were identical to those determined from AD cases. The PHFs and SFs are made of two C-shaped protofilaments, each containing a cross-β/β-helix motif. Similar to filaments from AD cases, most filaments from the DS cases adopted the PHF form, while a minority (approximately 20%) formed SFs. Samples from the youngest individual with no documented dementia had sparse tau deposits. To isolate tau for cryo-EM from this challenging sample we used a novel affinity-grid method involving a graphene oxide surface derivatized with anti-tau antibodies. This method improved isolation and revealed that primarily tau PHFs and a minor population of chronic traumatic encephalopathy type II-like filaments were present in this youngest case. These findings expand the similarities between AD and DS to the molecular level, providing insight into their related pathologies and the potential for targeting common tau filament folds by small-molecule therapeutics and diagnostics.
Collapse
Affiliation(s)
- Ujjayini Ghosh
- Institute for Neurodegenerative Diseases, University of California San Francisco, San Francisco, CA, USA
| | - Eric Tse
- Institute for Neurodegenerative Diseases, University of California San Francisco, San Francisco, CA, USA
| | - Hyunjun Yang
- Institute for Neurodegenerative Diseases, University of California San Francisco, San Francisco, CA, USA
| | - Marie Shi
- Institute for Neurodegenerative Diseases, University of California San Francisco, San Francisco, CA, USA
| | - Christoffer D Caro
- Institute for Neurodegenerative Diseases, University of California San Francisco, San Francisco, CA, USA
| | - Feng Wang
- Department of Biochemistry and Biophysics, University of California San Francisco, San Francisco, CA, USA
| | - Gregory E Merz
- Institute for Neurodegenerative Diseases, University of California San Francisco, San Francisco, CA, USA
- Department of Neurology, University of California San Francisco, San Francisco, CA, USA
| | - Stanley B Prusiner
- Institute for Neurodegenerative Diseases, University of California San Francisco, San Francisco, CA, USA
- Department of Biochemistry and Biophysics, University of California San Francisco, San Francisco, CA, USA
- Department of Neurology, University of California San Francisco, San Francisco, CA, USA
| | - Daniel R Southworth
- Institute for Neurodegenerative Diseases, University of California San Francisco, San Francisco, CA, USA.
- Department of Biochemistry and Biophysics, University of California San Francisco, San Francisco, CA, USA.
| | - Carlo Condello
- Institute for Neurodegenerative Diseases, University of California San Francisco, San Francisco, CA, USA.
- Department of Neurology, University of California San Francisco, San Francisco, CA, USA.
| |
Collapse
|
2
|
Ghosh U, Tse E, Yang H, Shi M, Carlo CD, Wang F, Merz GE, Prusiner SB, Southworth DR, Condello C. Cryo-EM Structures Reveal Tau Filaments from Down Syndrome Adopt Alzheimer's Disease Fold. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.04.02.587507. [PMID: 38617229 PMCID: PMC11014571 DOI: 10.1101/2024.04.02.587507] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/16/2024]
Abstract
Down syndrome (DS) is a common genetic condition caused by trisomy of chromosome 21. Among the complex clinical features including musculoskeletal, neurological and cardiovascular disabilities, individuals with DS have an increased risk of developing progressive dementia and early onset Alzheimer's Disease (AD). This is attributed to the increased gene dosage of amyloid-β (Aβ) precursor protein gene, the formation of self-propagating Aβ and tau prion conformers, and the deposition of neurotoxic Aβ plaques and tau neurofibrillary tangles. Tau amyloid fibrils have previously been established to adopt many distinct conformations across different neurodegenerative conditions. Here we report the characterization of brain samples from four DS cases spanning 36 to 63 years of age by spectral confocal imaging with conformation-specific dyes and cryo-electron microscopy (cryo-EM) to determine structures of isolated tau fibrils. High-resolution structures reveal paired helical filament (PHF) and straight filament (SF) conformations of tau that are identical to those determined from AD. The PHFs and SFs are made of two C-shaped protofilaments with a cross-β/β-helix motif. Similar to filaments from AD cases, most filaments from the DS cases adopted the PHF form, while a minority (~20%) formed SFs. Samples from the youngest individual with no documented dementia had sparse tau deposits. To isolate tau for cryo-EM from this challenging sample we used a novel affinity-grid method involving a graphene-oxide surface derivatized with anti-tau antibodies. This improved isolation and revealed primarily tau PHFs and a minor population of chronic traumatic encephalopathy type II-like filaments were present in this youngest case. These findings expand the similarities between AD and DS to the molecular level, providing insight into their related pathologies and the potential for targeting common tau filament folds by small-molecule therapeutics and diagnostics.
Collapse
|
3
|
Smith M, Knight IS, Kormos RC, Pepe JG, Kunach P, Diamond MI, Shahmoradian SH, Irwin JJ, DeGrado WF, Shoichet BK. Docking for Molecules That Bind in a Symmetric Stack with SymDOCK. J Chem Inf Model 2024; 64:425-434. [PMID: 38191997 PMCID: PMC10806807 DOI: 10.1021/acs.jcim.3c01749] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2023] [Revised: 12/21/2023] [Accepted: 12/22/2023] [Indexed: 01/10/2024]
Abstract
Discovering ligands for amyloid fibrils, such as those formed by the tau protein, is an area of great current interest. In recent structures, ligands bind in stacks in the tau fibrils to reflect the rotational and translational symmetry of the fibril itself; in these structures, the ligands make few interactions with the protein but interact extensively with each other. To exploit this symmetry and stacking, we developed SymDOCK, a method to dock molecules that follow the protein's symmetry. For each prospective ligand pose, we apply the symmetry operation of the fibril to generate a self-interacting and fibril-interacting stack, checking that doing so will not cause a clash between the original molecule and its image. Absent a clash, we retain that pose and add the ligand-ligand van der Waals energy to the ligand's docking score (here using DOCK3.8). We can check these geometries and energies using an implementation of ANI, a neural-network-based quantum-mechanical evaluation of the ligand stacking energies. In retrospective calculations, symmetry docking can reproduce the poses of three tau PET tracers whose structures have been determined. More convincingly, in a prospective study, SymDOCK predicted the structure of the PET tracer MK-6240 bound in a symmetrical stack to AD PHF tau before that structure was determined; the docked pose was used to determine how MK-6240 fit the cryo-EM density. In proof-of-concept studies, SymDOCK enriched known ligands over property-matched decoys in retrospective screens without sacrificing docking speed and can address large library screens that seek new symmetrical stackers. Future applications of this approach will be considered.
Collapse
Affiliation(s)
- Matthew
S. Smith
- Department
of Pharmaceutical Chemistry, University
of California, UCSF Genentech
Hall Box 2280, 600 16th St Rm 518,San Francisco, California 94158, United States
- Program
in Biophysics, University of California, UCSF Genentech Hall MC2240, 600
16th St Rm N474D,San Francisco, California 94143, United States
| | - Ian S. Knight
- Department
of Pharmaceutical Chemistry, University
of California, UCSF Genentech
Hall Box 2280, 600 16th St Rm 518,San Francisco, California 94158, United States
| | - Rian C. Kormos
- Department
of Pharmaceutical Chemistry, University
of California, UCSF Genentech
Hall Box 2280, 600 16th St Rm 518,San Francisco, California 94158, United States
- Program
in Biophysics, University of California, UCSF Genentech Hall MC2240, 600
16th St Rm N474D,San Francisco, California 94143, United States
| | - Joseph G. Pepe
- Department
of Pharmaceutical Chemistry, University
of California, UCSF Genentech
Hall Box 2280, 600 16th St Rm 518,San Francisco, California 94158, United States
- Program
in Biophysics, University of California, UCSF Genentech Hall MC2240, 600
16th St Rm N474D,San Francisco, California 94143, United States
| | - Peter Kunach
- McGill
Research Centre for Studies in Aging, McGill
University, 6875 Boulevard LaSalle, Montreal, Quebec H4H 1R3, Canada
- Department
of Neurology and Neurosurgery, McGill University, 1033 Pine Avenue West, Room 310, Montreal, Quebec H3A 1A1, Canada
- Center
for Alzheimer’s and Neurodegenerative Diseases, Peter O’Donnell
Jr. Brain Institute, University of Texas
Southwestern Medical Center, 6124 Harry Hines Blvd. Suite NS03.200, Dallas, Texas 75390, United States
- Department
of Neurology, University of Texas Southwestern
Medical Center, 5323 Harry Hines Blvd., G2.222, Dallas, Texas 75390-9368, United States
- Department
of Neuroscience, University of Texas Southwestern
Medical Center, 5323 Harry Hines Blvd., Dallas, Texas 75390-9111, United States
| | - Marc I. Diamond
- Center
for Alzheimer’s and Neurodegenerative Diseases, Peter O’Donnell
Jr. Brain Institute, University of Texas
Southwestern Medical Center, 6124 Harry Hines Blvd. Suite NS03.200, Dallas, Texas 75390, United States
- Department
of Neurology, University of Texas Southwestern
Medical Center, 5323 Harry Hines Blvd., G2.222, Dallas, Texas 75390-9368, United States
- Department
of Neuroscience, University of Texas Southwestern
Medical Center, 5323 Harry Hines Blvd., Dallas, Texas 75390-9111, United States
| | - Sarah H. Shahmoradian
- Center
for Alzheimer’s and Neurodegenerative Diseases, Peter O’Donnell
Jr. Brain Institute, University of Texas
Southwestern Medical Center, 6124 Harry Hines Blvd. Suite NS03.200, Dallas, Texas 75390, United States
- Department
of Biophysics, University of Texas Southwestern
Medical Center, 5323 Harry Hines Blvd., Dallas, Texas 75390-8816, United States
| | - John J. Irwin
- Department
of Pharmaceutical Chemistry, University
of California, UCSF Genentech
Hall Box 2280, 600 16th St Rm 518,San Francisco, California 94158, United States
| | - William F. DeGrado
- Department
of Pharmaceutical Chemistry, University
of California, UCSF Genentech
Hall Box 2280, 600 16th St Rm 518,San Francisco, California 94158, United States
- Cardiovascular
Research Institute, University of California, 555 Mission Bay Blvd South, PO Box 589001, San Francisco, California 94158-9001, United
States
| | - Brian K. Shoichet
- Department
of Pharmaceutical Chemistry, University
of California, UCSF Genentech
Hall Box 2280, 600 16th St Rm 518,San Francisco, California 94158, United States
| |
Collapse
|
4
|
Qi C, Verheijen BM, Kokubo Y, Shi Y, Tetter S, Murzin AG, Nakahara A, Morimoto S, Vermulst M, Sasaki R, Aronica E, Hirokawa Y, Oyanagi K, Kakita A, Ryskeldi-Falcon B, Yoshida M, Hasegawa M, Scheres SHW, Goedert M. Tau filaments from amyotrophic lateral sclerosis/parkinsonism-dementia complex adopt the CTE fold. Proc Natl Acad Sci U S A 2023; 120:e2306767120. [PMID: 38100415 PMCID: PMC10743375 DOI: 10.1073/pnas.2306767120] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2023] [Accepted: 10/31/2023] [Indexed: 12/17/2023] Open
Abstract
The amyotrophic lateral sclerosis/parkinsonism-dementia complex (ALS/PDC) of the island of Guam and the Kii peninsula of Japan is a fatal neurodegenerative disease of unknown cause that is characterized by the presence of abundant filamentous tau inclusions in brains and spinal cords. Here, we used electron cryo-microscopy to determine the structures of tau filaments from the cerebral cortex of three cases of ALS/PDC from Guam and eight cases from Kii, as well as from the spinal cord of two of the Guam cases. Tau filaments had the chronic traumatic encephalopathy (CTE) fold, with variable amounts of Type I and Type II filaments. Paired helical tau filaments were also found in three Kii cases and tau filaments with the corticobasal degeneration fold in one Kii case. We identified a new Type III CTE tau filament, where protofilaments pack against each other in an antiparallel fashion. ALS/PDC is the third known tauopathy with CTE-type filaments and abundant tau inclusions in cortical layers II/III, the others being CTE and subacute sclerosing panencephalitis. Because these tauopathies are believed to have environmental causes, our findings support the hypothesis that ALS/PDC is caused by exogenous factors.
Collapse
Affiliation(s)
- Chao Qi
- Medical Research Council, Laboratory of Molecular Biology, CambridgeCB2 0QH, United Kingdom
| | - Bert M. Verheijen
- Leonard Davis School of Gerontology, University of Southern California, Los Angeles, CA90089
| | - Yasumasa Kokubo
- Graduate School of Regional Innovation Studies, Mie University, Tsu514-8507, Japan
| | - Yang Shi
- Medical Research Council, Laboratory of Molecular Biology, CambridgeCB2 0QH, United Kingdom
| | - Stephan Tetter
- Medical Research Council, Laboratory of Molecular Biology, CambridgeCB2 0QH, United Kingdom
| | - Alexey G. Murzin
- Medical Research Council, Laboratory of Molecular Biology, CambridgeCB2 0QH, United Kingdom
| | - Asa Nakahara
- Department of Pathology, Brain Research Institute, Niigata University, Niigata951-8585, Japan
| | - Satoru Morimoto
- Department of Oncologic Pathology, Graduate School of Medicine, Mie University, Tsu514-8507, Japan
| | - Marc Vermulst
- Leonard Davis School of Gerontology, University of Southern California, Los Angeles, CA90089
| | - Ryogen Sasaki
- Department of Nursing, Suzuka University of Medical Science, Suzuka513-8670, Japan
| | - Eleonora Aronica
- Department of Neuropathology, Amsterdam University Medical Centers (UMC), University of Amsterdam, Amsterdam Neuroscience, Amsterdam1105 AZ, The Netherlands
| | - Yoshifumi Hirokawa
- Department of Oncologic Pathology, Graduate School of Medicine, Mie University, Tsu514-8507, Japan
| | - Kiyomitsu Oyanagi
- Department of Brain Disease Research, Shinshu University School of Medicine, Matsumoto390-8621, Japan
| | - Akiyoshi Kakita
- Department of Pathology, Brain Research Institute, Niigata University, Niigata951-8585, Japan
| | | | - Mari Yoshida
- Department of Neuropathology, Institute for Medical Science of Aging, Aichi Medical University, Nagakute480-1195, Japan
| | - Masato Hasegawa
- Department of Brain and Neuroscience, Tokyo Metropolitan Institute of Medical Science, Tokyo156-8506, Japan
| | - Sjors H. W. Scheres
- Medical Research Council, Laboratory of Molecular Biology, CambridgeCB2 0QH, United Kingdom
| | - Michel Goedert
- Medical Research Council, Laboratory of Molecular Biology, CambridgeCB2 0QH, United Kingdom
| |
Collapse
|
5
|
Smith MS, Knight IS, Kormos RC, Pepe JG, Kunach P, Diamond MI, Shahmoradian SH, Irwin JJ, DeGrado WF, Shoichet BK. Docking for molecules that bind in a symmetric stack with SymDOCK. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.10.27.564400. [PMID: 37961414 PMCID: PMC10634874 DOI: 10.1101/2023.10.27.564400] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/15/2023]
Abstract
Discovering ligands for amyloid fibrils, such as those formed by the tau protein, is an area of much current interest. In recent structures, ligands bind in stacks in the tau fibrils to reflect the rotational and translational symmetry of the fibril itself; in these structures the ligands make few interactions with the protein but interact extensively with each other. To exploit this symmetry and stacking, we developed SymDOCK, a method to dock molecules that follow the protein's symmetry. For each prospective ligand pose, we apply the symmetry operation of the fibril to generate a self-interacting and fibril-interacting stack, checking that doing so will not cause a clash between the original molecule and its image. Absent a clash, we retain that pose and add the ligand-ligand van der Waals energy to the ligand's docking score (here using DOCK3.8). We can check these geometries and energies using an implementation of ANI, a neural network-based quantum-mechanical evaluation of the ligand stacking energies. In retrospective calculations, symmetry docking can reproduce the poses of three tau PET tracers whose structures have been determined. More convincingly, in a prospective study SymDOCK predicted the structure of the PET tracer MK-6240 bound in a symmetrical stack to AD PHF tau before that structure was determined; the docked pose was used to determine how MK-6240 fit the cryo-EM density. In proof-of-concept studies, SymDOCK enriched known ligands over property-matched decoys in retrospective screens without sacrificing docking speed, and can address large library screens that seek new symmetrical stackers. Future applications of this approach will be considered.
Collapse
Affiliation(s)
- Matthew S. Smith
- Department of Pharmaceutical Chemistry, University of California, San Francisco, San Francisco, CA, USA
- Program in Biophysics, University of California, San Francisco, San Francisco, CA, USA
| | - Ian S. Knight
- Department of Pharmaceutical Chemistry, University of California, San Francisco, San Francisco, CA, USA
| | - Rian C. Kormos
- Department of Pharmaceutical Chemistry, University of California, San Francisco, San Francisco, CA, USA
- Program in Biophysics, University of California, San Francisco, San Francisco, CA, USA
| | - Joseph G. Pepe
- Department of Pharmaceutical Chemistry, University of California, San Francisco, San Francisco, CA, USA
- Program in Biophysics, University of California, San Francisco, San Francisco, CA, USA
| | - Peter Kunach
- McGill Research Centre for Studies in Aging, McGill University, Montreal, QC, Canada
- Department of Neurology and Neurosurgery, McGill University, Montreal, QC, Canada
- Center for Alzheimer’s and Neurodegenerative Diseases, Peter O’Donnell Jr. Brain Institute, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Department of Neurology, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Department of Neuroscience, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Marc I. Diamond
- Center for Alzheimer’s and Neurodegenerative Diseases, Peter O’Donnell Jr. Brain Institute, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Department of Neurology, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Department of Neuroscience, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Sarah H. Shahmoradian
- Center for Alzheimer’s and Neurodegenerative Diseases, Peter O’Donnell Jr. Brain Institute, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Department of Biophysics, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - John J. Irwin
- Department of Pharmaceutical Chemistry, University of California, San Francisco, San Francisco, CA, USA
| | - William F. DeGrado
- Department of Pharmaceutical Chemistry, University of California, San Francisco, San Francisco, CA, USA
- Cardiovascular Research Institute, University of California, San Francisco, San Francisco, CA, USA
| | - Brian K. Shoichet
- Department of Pharmaceutical Chemistry, University of California, San Francisco, San Francisco, CA, USA
| |
Collapse
|
6
|
Bell-Simons M, Buchholz S, Klimek J, Zempel H. Laser-Induced Axotomy of Human iPSC-Derived and Murine Primary Neurons Decreases Somatic Tau and AT8 Tau Phosphorylation: A Single-Cell Approach to Study Effects of Acute Axonal Damage. Cell Mol Neurobiol 2023; 43:3497-3510. [PMID: 37171549 PMCID: PMC10477226 DOI: 10.1007/s10571-023-01359-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2023] [Accepted: 05/02/2023] [Indexed: 05/13/2023]
Abstract
The microtubule-associated protein Tau is highly enriched in axons of brain neurons where it regulates axonal outgrowth, plasticity, and transport. Efficient axonal Tau sorting is critical since somatodendritic Tau missorting is a major hallmark of Alzheimer's disease and other tauopathies. However, the molecular mechanisms of axonal Tau sorting are still not fully understood. In this study, we aimed to unravel to which extent anterograde protein transport contributes to axonal Tau sorting. We developed a laser-based axotomy approach with single-cell resolution and combined it with spinning disk confocal microscopy enabling multi live-cell monitoring. We cultivated human iPSC-derived cortical neurons and mouse primary forebrain neurons in specialized chambers allowing reliable post-fixation identification and Tau analysis. Using this approach, we achieved high post-axotomy survival rates and observed axonal regrowth in a subset of neurons. When we assessed somatic missorting and phosphorylation levels of endogenous human or murine Tau at different time points after axotomy, we surprisingly did not observe somatic Tau accumulation or hyperphosphorylation, regardless of their regrowing activity, consistent for both models. These results indicate that impairment of anterograde transit of Tau protein and acute axonal damage may not play a role for the development of somatic Tau pathology. In sum, we developed a laser-based axotomy model suitable for studying the impact of different Tau sorting mechanisms in a highly controllable and reproducible setting, and we provide evidence that acute axon loss does not induce somatic Tau accumulation and AT8 Tau phosphorylation. UV laser-induced axotomy of human iPSC-derived and mouse primary neurons results in decreased somatic levels of endogenous Tau and AT8 Tau phosphorylation.
Collapse
Affiliation(s)
- M Bell-Simons
- Institute of Human Genetics, University Hospital Cologne, Kerpener Str. 34, 50931, Cologne, Germany
- Center for Molecular Medicine Cologne (CMMC), Robert-Koch-Str. 21, 50931, Cologne, Germany
| | - S Buchholz
- Institute of Human Genetics, University Hospital Cologne, Kerpener Str. 34, 50931, Cologne, Germany
- Center for Molecular Medicine Cologne (CMMC), Robert-Koch-Str. 21, 50931, Cologne, Germany
| | - J Klimek
- Institute of Human Genetics, University Hospital Cologne, Kerpener Str. 34, 50931, Cologne, Germany
- Center for Molecular Medicine Cologne (CMMC), Robert-Koch-Str. 21, 50931, Cologne, Germany
| | - H Zempel
- Institute of Human Genetics, University Hospital Cologne, Kerpener Str. 34, 50931, Cologne, Germany.
- Center for Molecular Medicine Cologne (CMMC), Robert-Koch-Str. 21, 50931, Cologne, Germany.
| |
Collapse
|
7
|
Qi C, Verheijen BM, Kokubo Y, Shi Y, Tetter S, Murzin AG, Nakahara A, Morimoto S, Vermulst M, Sasaki R, Aronica E, Hirokawa Y, Oyanagi K, Kakita A, Ryskeldi-Falcon B, Yoshida M, Hasegawa M, Scheres SH, Goedert M. Tau Filaments from Amyotrophic Lateral Sclerosis/Parkinsonism-Dementia Complex (ALS/PDC) adopt the CTE Fold. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.04.26.538417. [PMID: 37162924 PMCID: PMC10168338 DOI: 10.1101/2023.04.26.538417] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/11/2023]
Abstract
The amyotrophic lateral sclerosis/parkinsonism-dementia complex (ALS/PDC) of the island of Guam and the Kii peninsula of Japan is a fatal neurodegenerative disease of unknown cause that is characterised by the presence of abundant filamentous tau inclusions in brains and spinal cords. Here we used electron cryo-microscopy (cryo-EM) to determine the structures of tau filaments from the cerebral cortex of three cases of ALS/PDC from Guam and eight cases from Kii, as well as from the spinal cord of two of the Guam cases. Tau filaments had the chronic traumatic encephalopathy (CTE) fold, with variable amounts of Type I and Type II filaments. Paired helical tau filaments were also found in two Kii cases. We also identified a novel Type III CTE tau filament, where protofilaments pack against each other in an anti-parallel fashion. ALS/PDC is the third known tauopathy with CTE-type filaments and abundant tau inclusions in cortical layers II/III, the others being CTE and subacute sclerosing panencephalitis. Because these tauopathies are believed to have environmental causes, our findings support the hypothesis that ALS/PDC is caused by exogenous factors.
Collapse
Affiliation(s)
- Chao Qi
- MRC Laboratory of Molecular Biology, Cambridge, UK
| | - Bert M. Verheijen
- Leonard Davis School of Gerontology, University of Southern California, Los Angeles, USA
| | - Yasumasa Kokubo
- Graduate School of Regional Innovation Studies, Mie University, Tsu, Japan
| | - Yang Shi
- MRC Laboratory of Molecular Biology, Cambridge, UK
- Current address: MOE Frontier Science Center for Brain Science and Brain-machine Integration, School of Brain Science and Brain Medicine, Zhejiang University, Hangzhou, China
| | | | | | - Asa Nakahara
- Department of Pathology, Brain Research Institute, Niigata University, Niigata, Japan
| | - Satoru Morimoto
- Department of Oncologic Pathology, Graduate School of Medicine, Mie University, Tsu, Japan
| | - Marc Vermulst
- Leonard Davis School of Gerontology, University of Southern California, Los Angeles, USA
| | - Ryogen Sasaki
- Department of Nursing, Suzuka University of Medical Science, Suzuka, Japan
| | - Eleonora Aronica
- Department of Neuropathology, University of Amsterdam, Amsterdam, The Netherlands
| | - Yoshifumi Hirokawa
- Department of Oncologic Pathology, Graduate School of Medicine, Mie University, Tsu, Japan
| | - Kiyomitsu Oyanagi
- Department of Brain Disease Research, Shinshu University School of Medicine, Matsumoto, Japan
| | - Akiyoshi Kakita
- Department of Pathology, Brain Research Institute, Niigata University, Niigata, Japan
| | | | - Mari Yoshida
- Department of Neuropathology, Institute for Medical Science of Aging, Aichi Medical University, Nagakute, Japan
| | - Masato Hasegawa
- Department of Brain and Neuroscience, Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan
| | | | | |
Collapse
|
8
|
McKee AC, Stein TD, Huber BR, Crary JF, Bieniek K, Dickson D, Alvarez VE, Cherry JD, Farrell K, Butler M, Uretsky M, Abdolmohammadi B, Alosco ML, Tripodis Y, Mez J, Daneshvar DH. Chronic traumatic encephalopathy (CTE): criteria for neuropathological diagnosis and relationship to repetitive head impacts. Acta Neuropathol 2023; 145:371-394. [PMID: 36759368 PMCID: PMC10020327 DOI: 10.1007/s00401-023-02540-w] [Citation(s) in RCA: 78] [Impact Index Per Article: 39.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2022] [Revised: 01/16/2023] [Accepted: 01/18/2023] [Indexed: 02/11/2023]
Abstract
Over the last 17 years, there has been a remarkable increase in scientific research concerning chronic traumatic encephalopathy (CTE). Since the publication of NINDS-NIBIB criteria for the neuropathological diagnosis of CTE in 2016, and diagnostic refinements in 2021, hundreds of contact sport athletes and others have been diagnosed at postmortem examination with CTE. CTE has been reported in amateur and professional athletes, including a bull rider, boxers, wrestlers, and American, Canadian, and Australian rules football, rugby union, rugby league, soccer, and ice hockey players. The pathology of CTE is unique, characterized by a pathognomonic lesion consisting of a perivascular accumulation of neuronal phosphorylated tau (p-tau) variably alongside astrocytic aggregates at the depths of the cortical sulci, and a distinctive molecular structural configuration of p-tau fibrils that is unlike the changes observed with aging, Alzheimer's disease, or any other tauopathy. Computational 3-D and finite element models predict the perivascular and sulcal location of p-tau pathology as these brain regions undergo the greatest mechanical deformation during head impact injury. Presently, CTE can be definitively diagnosed only by postmortem neuropathological examination; the corresponding clinical condition is known as traumatic encephalopathy syndrome (TES). Over 97% of CTE cases published have been reported in individuals with known exposure to repetitive head impacts (RHI), including concussions and nonconcussive impacts, most often experienced through participation in contact sports. While some suggest there is uncertainty whether a causal relationship exists between RHI and CTE, the preponderance of the evidence suggests a high likelihood of a causal relationship, a conclusion that is strengthened by the absence of any evidence for plausible alternative hypotheses. There is a robust dose-response relationship between CTE and years of American football play, a relationship that remains consistent even when rigorously accounting for selection bias. Furthermore, a recent study suggests that selection bias underestimates the observed risk. Here, we present the advances in the neuropathological diagnosis of CTE culminating with the development of the NINDS-NIBIB criteria, the multiple international studies that have used these criteria to report CTE in hundreds of contact sports players and others, and the evidence for a robust dose-response relationship between RHI and CTE.
Collapse
Affiliation(s)
- Ann C McKee
- VA Boston Healthcare System, U.S. Department of Veteran Affairs, Boston, MA, USA.
- Boston University Alzheimer's Disease Research Center and CTE Centers, Department of Neurology, Boston University School of Medicine, 150 S Huntington Ave, Boston, MA, 02130, USA.
- Department of Neurology, Boston University School of Medicine, Boston, MA, USA.
- Department of Pathology and Laboratory Medicine, Boston University School of Medicine, Boston, MA, USA.
- VA Bedford Healthcare System, Bedford, MA, USA.
| | - Thor D Stein
- VA Boston Healthcare System, U.S. Department of Veteran Affairs, Boston, MA, USA
- Boston University Alzheimer's Disease Research Center and CTE Centers, Department of Neurology, Boston University School of Medicine, 150 S Huntington Ave, Boston, MA, 02130, USA
- Department of Pathology and Laboratory Medicine, Boston University School of Medicine, Boston, MA, USA
- VA Bedford Healthcare System, Bedford, MA, USA
| | - Bertrand R Huber
- VA Boston Healthcare System, U.S. Department of Veteran Affairs, Boston, MA, USA
- Boston University Alzheimer's Disease Research Center and CTE Centers, Department of Neurology, Boston University School of Medicine, 150 S Huntington Ave, Boston, MA, 02130, USA
- Department of Neurology, Boston University School of Medicine, Boston, MA, USA
- Department of Pathology and Laboratory Medicine, Boston University School of Medicine, Boston, MA, USA
| | - John F Crary
- Departments of Pathology, Neuroscience, and Artificial Intelligence and Human Health, Neuropathology Brain Bank and Research Core, Ronald M. Loeb Center for Alzheimer's Disease, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Kevin Bieniek
- Department of Pathology and Laboratory Medicine, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
- Glenn Biggs Institute for Alzheimer's and Neurodegenerative Diseases, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - Dennis Dickson
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, USA
| | - Victor E Alvarez
- VA Boston Healthcare System, U.S. Department of Veteran Affairs, Boston, MA, USA
- Boston University Alzheimer's Disease Research Center and CTE Centers, Department of Neurology, Boston University School of Medicine, 150 S Huntington Ave, Boston, MA, 02130, USA
- Department of Pathology and Laboratory Medicine, Boston University School of Medicine, Boston, MA, USA
- VA Bedford Healthcare System, Bedford, MA, USA
| | - Jonathan D Cherry
- VA Boston Healthcare System, U.S. Department of Veteran Affairs, Boston, MA, USA
- Boston University Alzheimer's Disease Research Center and CTE Centers, Department of Neurology, Boston University School of Medicine, 150 S Huntington Ave, Boston, MA, 02130, USA
- Department of Pathology and Laboratory Medicine, Boston University School of Medicine, Boston, MA, USA
| | - Kurt Farrell
- Departments of Pathology, Neuroscience, and Artificial Intelligence and Human Health, Neuropathology Brain Bank and Research Core, Ronald M. Loeb Center for Alzheimer's Disease, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Morgane Butler
- Boston University Alzheimer's Disease Research Center and CTE Centers, Department of Neurology, Boston University School of Medicine, 150 S Huntington Ave, Boston, MA, 02130, USA
| | - Madeline Uretsky
- Boston University Alzheimer's Disease Research Center and CTE Centers, Department of Neurology, Boston University School of Medicine, 150 S Huntington Ave, Boston, MA, 02130, USA
| | - Bobak Abdolmohammadi
- Boston University Alzheimer's Disease Research Center and CTE Centers, Department of Neurology, Boston University School of Medicine, 150 S Huntington Ave, Boston, MA, 02130, USA
| | - Michael L Alosco
- Boston University Alzheimer's Disease Research Center and CTE Centers, Department of Neurology, Boston University School of Medicine, 150 S Huntington Ave, Boston, MA, 02130, USA
- Department of Neurology, Boston University School of Medicine, Boston, MA, USA
| | - Yorghos Tripodis
- Boston University Alzheimer's Disease Research Center and CTE Centers, Department of Neurology, Boston University School of Medicine, 150 S Huntington Ave, Boston, MA, 02130, USA
- Department of Biostatistics, Boston University School of Public Health, Boston, MA, USA
- Department of Epidemiology, Boston University School of Public Health, Boston, MA, USA
| | - Jesse Mez
- Boston University Alzheimer's Disease Research Center and CTE Centers, Department of Neurology, Boston University School of Medicine, 150 S Huntington Ave, Boston, MA, 02130, USA
- Department of Neurology, Boston University School of Medicine, Boston, MA, USA
| | - Daniel H Daneshvar
- Boston University Alzheimer's Disease Research Center and CTE Centers, Department of Neurology, Boston University School of Medicine, 150 S Huntington Ave, Boston, MA, 02130, USA
- Department of Physical Medicine and Rehabilitation, Harvard Medical School, Boston, MA, USA
- Department of Physical Medicine and Rehabilitation, Massachusetts General Hospital, Boston, MA, USA
- Department of Physical Medicine and Rehabilitation, Spaulding Rehabilitation Hospital, Boston, MA, USA
| |
Collapse
|
9
|
Juan SMA, Daglas M, Adlard P. Tau pathology, metal dyshomeostasis and repetitive mild traumatic brain injury: an unexplored link paving the way for neurodegeneration. J Neurotrauma 2022; 39:902-922. [PMID: 35293225 DOI: 10.1089/neu.2021.0241] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Repetitive mild traumatic brain injury (r-mTBI), commonly experienced by athletes and military personnel, causes changes in multiple intracellular pathways, one of which involves the tau protein. Tau phosphorylation plays a role in several neurodegenerative conditions including chronic traumatic encephalopathy (CTE), a progressive neurodegenerative disorder linked to repeated head trauma. There is now mounting evidence suggesting that tau phosphorylation may be regulated by metal ions (such as iron, zinc and copper), which themselves are implicated in ageing and neurodegenerative disorders such as Alzheimer's disease (AD). Recent work has also shown that a single TBI can result in age-dependent and region-specific modulation of metal ions. As such, this review explores the link between TBI, CTE, ageing and neurodegeneration with a specific focus on the involvement of (and interaction between) tau pathology and metal dyshomeostasis. The authors highlight that metal dyshomeostasis has yet to be investigated in the context of repeat head trauma or CTE. Given the evidence that metal dyshomeostasis contributes to the onset and/or progression of neurodegeneration, and that CTE itself is a neurodegenerative condition, this brings to light an uncharted link that should be explored. The development of adequate models of r-mTBI and/or CTE will be crucial in deepening our understanding of the pathological mechanisms that drive the clinical manifestations in these conditions and also in the development of effective therapeutics targeted towards slowing progressive neurodegenerative disorders.
Collapse
Affiliation(s)
- Sydney M A Juan
- The Florey Institute of Neuroscience and Mental Health, 56369, 30 Royal Parade, Parkville, Melbourne, Victoria, Australia, 3052;
| | - Maria Daglas
- The Florey Institute of Neuroscience and Mental Health, 56369, Parkville, Victoria, Australia;
| | - Paul Adlard
- Florey Institute of Neuroscience and Mental Health, 56369, Parkville, Victoria, Australia;
| |
Collapse
|
10
|
Turk KW, Geada A, Alvarez VE, Xia W, Cherry JD, Nicks R, Meng G, Daley S, Tripodis Y, Huber BR, Budson AE, Dwyer B, Kowall NW, Cantu RC, Goldstein LE, Katz DI, Stern RA, Alosco ML, Mez J, McKee AC, Stein TD. A comparison between tau and amyloid-β cerebrospinal fluid biomarkers in chronic traumatic encephalopathy and Alzheimer disease. Alzheimers Res Ther 2022; 14:28. [PMID: 35139894 PMCID: PMC8830027 DOI: 10.1186/s13195-022-00976-y] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2021] [Accepted: 02/02/2022] [Indexed: 01/14/2023]
Abstract
BACKGROUND Cerebrospinal fluid (CSF) tau and beta-amyloid levels in chronic traumatic encephalopathy (CTE), a disease which can be clinically indistinguishable from Alzheimer's disease (AD), are largely unknown. We examined postmortem CSF analytes among participants with autopsy confirmed CTE and AD. METHODS In this cross-sectional study 192 participants from the Boston University AD Research Center, VA-BU-CLF Center, and Framingham Heart Study (FHS) had post-mortem CSF collected at autopsy. Participants were divided into pathological groups based on AD and CTE criteria, with 61 CTE participants (18 low, 43 high stage), 79 AD participants (23 low, 56 intermediate to high), 11 participants with CTE combined with AD, and 41 participants lacking both CTE and AD neuropathology. The Meso Scale Discovery immunoassay system was utilized to measure amyloid-beta (Aβ1-40, Aβ1-42), total tau (t-tau), and phosphorylated tau (p-tau181 and p-tau231). CSF analytes were then compared across the pathological groups: no CTE/no AD (control), Low CTE, Low AD, High CTE, Intermediate/High AD, and AD+CTE. RESULTS Among the Low disease state groups, the Low CTE group had significantly higher levels of p-tau231 versus the control group and compared to the Low AD group. The Low CTE group was also found to have significantly lower levels of Aβ1-42 compared to the control group. The high CTE group had higher levels of p-tau231 and lower levels of Aβ1-42 compared to Intermediate/High AD group. CONCLUSIONS Importantly, p-tau231 and Aβ1-42 were predictors of diagnosis of CTE vs. control and CTE vs. AD. Increased CSF p-tau231 is a promising potentially sensitive biomarker of CTE, and CSF Aβ1-42 needs further investigation in CTE.
Collapse
Affiliation(s)
- Katherine W Turk
- Boston University Alzheimer's Disease Research and CTE Center, Boston University School of Medicine, Boston, MA, 02118, USA
- VA Boston Healthcare System, 150 S. Huntington Avenue, Boston, MA, 02130, USA
- Department of Neurology, Boston University School of Medicine, Boston, MA, 20118, USA
| | - Alexandra Geada
- Boston University School of Medicine, Boston, MA, 02118, USA
| | - Victor E Alvarez
- Boston University Alzheimer's Disease Research and CTE Center, Boston University School of Medicine, Boston, MA, 02118, USA
- VA Boston Healthcare System, 150 S. Huntington Avenue, Boston, MA, 02130, USA
- Department of Neurology, Boston University School of Medicine, Boston, MA, 20118, USA
- VA Bedford Healthcare System, Bedford, MA, 01730, USA
| | - Weiming Xia
- Boston University Alzheimer's Disease Research and CTE Center, Boston University School of Medicine, Boston, MA, 02118, USA
- VA Bedford Healthcare System, Bedford, MA, 01730, USA
| | - Jonathan D Cherry
- Boston University Alzheimer's Disease Research and CTE Center, Boston University School of Medicine, Boston, MA, 02118, USA
- VA Boston Healthcare System, 150 S. Huntington Avenue, Boston, MA, 02130, USA
- Department of Neurology, Boston University School of Medicine, Boston, MA, 20118, USA
| | - Raymond Nicks
- Boston University Alzheimer's Disease Research and CTE Center, Boston University School of Medicine, Boston, MA, 02118, USA
- VA Bedford Healthcare System, Bedford, MA, 01730, USA
| | - Gaoyuan Meng
- VA Boston Healthcare System, 150 S. Huntington Avenue, Boston, MA, 02130, USA
- VA Bedford Healthcare System, Bedford, MA, 01730, USA
| | - Sarah Daley
- Boston University Alzheimer's Disease Research and CTE Center, Boston University School of Medicine, Boston, MA, 02118, USA
- VA Bedford Healthcare System, Bedford, MA, 01730, USA
| | - Yorghos Tripodis
- Department of Biostatistics, Boston University School of Public Health, Boston, MA, 20118, USA
| | - Bertrand R Huber
- Boston University Alzheimer's Disease Research and CTE Center, Boston University School of Medicine, Boston, MA, 02118, USA
- VA Boston Healthcare System, 150 S. Huntington Avenue, Boston, MA, 02130, USA
- Department of Neurology, Boston University School of Medicine, Boston, MA, 20118, USA
| | - Andrew E Budson
- Boston University Alzheimer's Disease Research and CTE Center, Boston University School of Medicine, Boston, MA, 02118, USA
- VA Boston Healthcare System, 150 S. Huntington Avenue, Boston, MA, 02130, USA
- Department of Neurology, Boston University School of Medicine, Boston, MA, 20118, USA
| | - Brigid Dwyer
- Department of Neurology, Boston University School of Medicine, Boston, MA, 20118, USA
- Braintree Rehabilitation Hospital, Braintree, MA, 02118, USA
| | - Neil W Kowall
- Boston University Alzheimer's Disease Research and CTE Center, Boston University School of Medicine, Boston, MA, 02118, USA
- VA Boston Healthcare System, 150 S. Huntington Avenue, Boston, MA, 02130, USA
- Department of Neurology, Boston University School of Medicine, Boston, MA, 20118, USA
| | - Robert C Cantu
- Boston University Alzheimer's Disease Research and CTE Center, Boston University School of Medicine, Boston, MA, 02118, USA
- Department of Anatomy and Neurobiology, Boston University School of Medicine, Boston, MA, 20119, USA
- Concussion Legacy Foundation, Boston, MA, 02115, USA
- Department of Neurosurgery, Boston University School of Medicine, Boston, MA, 02118, USA
- Department of Neurosurgery, Emerson Hospital, Concord, MA, 01742, USA
| | - Lee E Goldstein
- Boston University Alzheimer's Disease Research and CTE Center, Boston University School of Medicine, Boston, MA, 02118, USA
- Departments of Psychiatry, Ophthalmology, Boston University School of Medicine, Boston, USA
- Departments of Biomedical, Electrical & Computer Engineering, Boston University College of Engineering, Boston, USA
- Department of Pathology and Laboratory Medicine, Boston University School of Medicine, Boston, MA, 02118, USA
| | - Douglas I Katz
- Department of Neurology, Boston University School of Medicine, Boston, MA, 20118, USA
- Braintree Rehabilitation Hospital, Braintree, MA, 02118, USA
| | - Robert A Stern
- Boston University Alzheimer's Disease Research and CTE Center, Boston University School of Medicine, Boston, MA, 02118, USA
- Department of Neurology, Boston University School of Medicine, Boston, MA, 20118, USA
- Department of Anatomy and Neurobiology, Boston University School of Medicine, Boston, MA, 20119, USA
- Department of Neurosurgery, Boston University School of Medicine, Boston, MA, 02118, USA
| | - Michael L Alosco
- Boston University Alzheimer's Disease Research and CTE Center, Boston University School of Medicine, Boston, MA, 02118, USA
- Department of Neurology, Boston University School of Medicine, Boston, MA, 20118, USA
| | - Jesse Mez
- Boston University Alzheimer's Disease Research and CTE Center, Boston University School of Medicine, Boston, MA, 02118, USA
- Department of Neurology, Boston University School of Medicine, Boston, MA, 20118, USA
| | - Ann C McKee
- Boston University Alzheimer's Disease Research and CTE Center, Boston University School of Medicine, Boston, MA, 02118, USA
- VA Boston Healthcare System, 150 S. Huntington Avenue, Boston, MA, 02130, USA
- Department of Neurology, Boston University School of Medicine, Boston, MA, 20118, USA
- VA Bedford Healthcare System, Bedford, MA, 01730, USA
- Department of Pathology and Laboratory Medicine, Boston University School of Medicine, Boston, MA, 02118, USA
| | - Thor D Stein
- Boston University Alzheimer's Disease Research and CTE Center, Boston University School of Medicine, Boston, MA, 02118, USA.
- VA Boston Healthcare System, 150 S. Huntington Avenue, Boston, MA, 02130, USA.
- VA Bedford Healthcare System, Bedford, MA, 01730, USA.
- Department of Pathology and Laboratory Medicine, Boston University School of Medicine, Boston, MA, 02118, USA.
| |
Collapse
|
11
|
Bakhtiarydavijani A, Khalid G, Murphy MA, Johnson KL, Peterson LE, Jones M, Horstemeyer MF, Dobbins AC, Prabhu RK. A mesoscale finite element modeling approach for understanding brain morphology and material heterogeneity effects in chronic traumatic encephalopathy. Comput Methods Biomech Biomed Engin 2021; 24:1169-1183. [PMID: 33635182 DOI: 10.1080/10255842.2020.1867851] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
Abstract
Chronic Traumatic Encephalopathy (CTE) affects a significant portion of athletes in contact sports but is difficult to quantify using clinical examinations and modeling approaches. We use an in silico approach to quantify CTE biomechanics using mesoscale Finite Element (FE) analysis that bridges with macroscale whole head FE analysis. The sulci geometry produces complex stress waves that interact with one another to create increased shear stresses at the sulci depth that are significantly larger than in analyses without sulci (from 0.5 to 18.0 kPa). Sulci peak stress concentration regions coincide with experimentally observed CTE sites documented in the literature. HighlightsSulci introduce stress localizations at their depth in the gray matterSulci stress fields interact to produce stress concentration sites in white matterDifferentiating brain tissue properties did not significantly affect peak stresses.
Collapse
Affiliation(s)
- A Bakhtiarydavijani
- Center for Advanced Vehicular Systems, Mississippi State University, Starkville, MS, USA
| | - G Khalid
- Middle Technical University, Baghdad, Iraq
| | - M A Murphy
- Center for Advanced Vehicular Systems, Mississippi State University, Starkville, MS, USA
| | | | - L E Peterson
- Center for Advanced Vehicular Systems, Mississippi State University, Starkville, MS, USA
| | - M Jones
- Institute of Medical Engineering & Medical Physics, Cardiff University, Cardiff, Wales, UK
| | | | - A C Dobbins
- Department of Biomedical Engineering, University of Alabama at Birmingham, Birmingham, USA
| | - R K Prabhu
- Center for Advanced Vehicular Systems, Mississippi State University, Starkville, MS, USA.,Department of Agricultural and Biological Engineering, Mississippi State University, MS, USA
| |
Collapse
|
12
|
Goedert M, Spillantini MG, Falcon B, Zhang W, Newell KL, Hasegawa M, Scheres SHW, Ghetti B. Tau Protein and Frontotemporal Dementias. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1281:177-199. [PMID: 33433876 DOI: 10.1007/978-3-030-51140-1_12] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Filamentous inclusions of tau protein are found in cases of inherited and sporadic frontotemporal dementias (FTDs). Mutations in MAPT, the tau gene, cause approximately 5% of cases of FTD. They proved that dysfunction of tau protein is sufficient to cause neurodegeneration and dementia. Clinically and pathologically, cases with MAPT mutations can resemble sporadic diseases, such as Pick's disease, globular glial tauopathy, progressive supranuclear palsy and corticobasal degeneration. The structures of tau filaments from Pick's disease and corticobasal degeneration, determined by electron cryo-microscopy, revealed the presence of specific tau folds in each disease, with no inter-individual variation. The same was true of chronic traumatic encephalopathy.
Collapse
Affiliation(s)
| | | | | | | | - Kathy L Newell
- Department of Pathology and Laboratory Medicine, Indiana University, Indianapolis, IN, USA
| | - Masato Hasegawa
- Department of Dementia and Higher Brain Function, Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan
| | | | - Bernardino Ghetti
- Department of Pathology and Laboratory Medicine, Indiana University, Indianapolis, IN, USA
| |
Collapse
|
13
|
Sharpe RLS, Mahmud M, Kaiser MS, Chen J. Gamma entrainment frequency affects mood, memory and cognition: an exploratory pilot study. Brain Inform 2020; 7:17. [PMID: 33226543 PMCID: PMC7683678 DOI: 10.1186/s40708-020-00119-9] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2020] [Accepted: 10/29/2020] [Indexed: 12/14/2022] Open
Abstract
Here we provide evidence with an exploratory pilot study that through the use of a Gamma 40 Hz entrainment frequency, mood, memory and cognition can be improved with respect to a 9-participant cohort. Participants constituted towards three binaural entrainment frequency groups: the 40 Hz, 25 Hz and 100 Hz. Participants attended a total of eight entrainment frequency sessions twice over the duration of a 4-week period. Additionally, participants were assessed based on their cognitive abilities, mood as well as memory, where the cognitive and memory assessments occurred before and after a 5-min binaural beat stimulation. The mood assessment scores were collected from sessions 1, 4 and 8, respectively. With respect to the Gamma 40 Hz entrainment frequency population, we observed a mean improvement in cognitive scores, elevating from 75% average to 85% average upon conclusion of the experimentation at weak statistical significance ([Formula: see text] = 0.10, p = 0.076). Similarly, memory score improvements at a greater significance ([Formula: see text] = 0.05, p = 0.0027) were noted, elevating from an average of 87% to 95%. In pertinence to the mood scores, a negative correlation across all populations were noted, inferring an overall increase in mood due to lower scores correlating with elevated mood. Finally, correlation analysis revealed a stronger R[Formula: see text] value (0.9838) within the 40 Hz group between sessions as well as mood score when compared across the entire frequency group cohort.
Collapse
Affiliation(s)
- Ryan L. S. Sharpe
- Department of Computer Science, Nottingham Trent University, Nottingham, NG11 8NS UK
| | - Mufti Mahmud
- Department of Computer Science, Nottingham Trent University, Nottingham, NG11 8NS UK
- Medical Technology Innovation Facility, Nottingham Trent University, Nottingham, NG11 8NS UK
| | - M. Shamim Kaiser
- Institute of Information Engineering, Jahangirnagar University, Savar, 1342 Dhaka, Bangladesh
| | - Jianhui Chen
- Beijing Key Laboratory of MRI and Brain Informatics, Beijing University of Technology, Beijing, 100124 China
- Beijing International Collaboration Base on Brain Informatics and Wisdom Services, Beijing, China
| |
Collapse
|
14
|
Abstract
With age, the presence of multiple neuropathologies in a single individual becomes increasingly common. Given that traumatic brain injury and the repetitive head impacts (RHIs) that occur in contact sports have been associated with the development of many neurodegenerative diseases, including chronic traumatic encephalopathy (CTE), Alzheimer's disease, Lewy body disease, and amyotrophic lateral sclerosis, it is becoming critical to understand the relationship and interactions between these pathologies. In fact, comorbid pathology is common in CTE and likely influenced by both age and the severity and type of exposure to RHI as well as underlying genetic predisposition. Here, we review the major comorbid pathologies seen with CTE and in former contact sports athletes and discuss what is known about the associations between RHI, age, and the development of neuropathologies. In addition, we examine the distinction between CTE and age-related pathology including primary age-related tauopathy and age-related tau astrogliopathy.
Collapse
Affiliation(s)
- Thor D. Stein
- Department of Pathology and Laboratory Medicine, Boston University School of Medicine, Boston, Massachusetts,Boston University Alzheimer’s Disease and CTE Center, Boston University School of Medicine, Boston, Massachusetts,Departments of Research and Pathology & Laboratory Medicine, VA Boston Healthcare System, Boston, Massachusetts,Department of Veterans Affairs Medical Center, Bedford, Massachusetts
| | - John F. Crary
- Department of Pathology, Neuropathology Brain Bank & Research Core, Ronald M. Loeb Center for Alzheimer’s Disease, Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York
| |
Collapse
|
15
|
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: 58] [Impact Index Per Article: 11.6] [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
|
16
|
Abrahamson EE, Ikonomovic MD. Brain injury-induced dysfunction of the blood brain barrier as a risk for dementia. Exp Neurol 2020; 328:113257. [PMID: 32092298 DOI: 10.1016/j.expneurol.2020.113257] [Citation(s) in RCA: 58] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2019] [Revised: 01/31/2020] [Accepted: 02/20/2020] [Indexed: 02/06/2023]
Abstract
The blood-brain barrier (BBB) is a complex and dynamic physiological interface between brain parenchyma and cerebral vasculature. It is composed of closely interacting cells and signaling molecules that regulate movement of solutes, ions, nutrients, macromolecules, and immune cells into the brain and removal of products of normal and abnormal brain cell metabolism. Dysfunction of multiple components of the BBB occurs in aging, inflammatory diseases, traumatic brain injury (TBI, severe or mild repetitive), and in chronic degenerative dementing disorders for which aging, inflammation, and TBI are considered risk factors. BBB permeability changes after TBI result in leakage of serum proteins, influx of immune cells, perivascular inflammation, as well as impairment of efflux transporter systems and accumulation of aggregation-prone molecules involved in hallmark pathologies of neurodegenerative diseases with dementia. In addition, cerebral vascular dysfunction with persistent alterations in cerebral blood flow and neurovascular coupling contribute to brain ischemia, neuronal degeneration, and synaptic dysfunction. While the idea of TBI as a risk factor for dementia is supported by many shared pathological features, it remains a hypothesis that needs further testing in experimental models and in human studies. The current review focusses on pathological mechanisms shared between TBI and neurodegenerative disorders characterized by accumulation of pathological protein aggregates, such as Alzheimer's disease and chronic traumatic encephalopathy. We discuss critical knowledge gaps in the field that need to be explored to clarify the relationship between TBI and risk for dementia and emphasize the need for longitudinal in vivo studies using imaging and biomarkers of BBB dysfunction in people with single or multiple TBI.
Collapse
Affiliation(s)
- Eric E Abrahamson
- Geriatric Research Education and Clinical Center, VA Pittsburgh Healthcare System, University of Pittsburgh, Pittsburgh, PA, United States; Department of Neurology, University of Pittsburgh, Pittsburgh, PA, United States
| | - Milos D Ikonomovic
- Geriatric Research Education and Clinical Center, VA Pittsburgh Healthcare System, University of Pittsburgh, Pittsburgh, PA, United States; Department of Neurology, University of Pittsburgh, Pittsburgh, PA, United States; Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA, United States.
| |
Collapse
|
17
|
Effect of the Gamma Entrainment Frequency in Pertinence to Mood, Memory and Cognition. Brain Inform 2020. [DOI: 10.1007/978-3-030-59277-6_5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022] Open
|
18
|
Wu Z, Wang ZH, Liu X, Zhang Z, Gu X, Yu SP, Keene CD, Cheng L, Ye K. Traumatic brain injury triggers APP and Tau cleavage by delta-secretase, mediating Alzheimer's disease pathology. Prog Neurobiol 2019; 185:101730. [PMID: 31778772 DOI: 10.1016/j.pneurobio.2019.101730] [Citation(s) in RCA: 57] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2019] [Revised: 10/17/2019] [Accepted: 11/18/2019] [Indexed: 10/25/2022]
Abstract
Traumatic brain injury (TBI) is associated in some studies with clinical dementia, and neuropathological features, including amyloid plaque deposition and Tau neurofibrillary degeneration commonly identified in Alzheimer's disease (AD). However, the molecular mechanisms linking TBI to AD remain unclear. Here we show that TBI activates transcription factor CCAAT/Enhancer Binding Protein Beta (C/EBPβ), increasing delta-secretase (AEP) expression. Activated AEP cleaves both APP and Tau at APP N585 and Tau N368 sites, respectively, which mediate AD pathogenesis by promoting Aβ production and Tau hyperphosphorylation and inducing neuroinflammation and neurotoxicity. Knockout of AEP or C/EBPβ diminishes TBI-induced AD-like pathology and cognitive impairment in the 3xTg AD mouse model. Remarkably, viral expression of AEP-resistant Tau N368A in the hippocampus of 3xTg mice also ameliorates the pathological and cognitive consequences of TBI. Finally, clinical TBI activates C/EBPβ and escalates AEP expression, leading to APP N585 and Tau N368 proteolytic cleavage in TBI patient brains. Hence, our findings support a potential role for AEP in linking TBI exposure with AD pathogenesis.
Collapse
Affiliation(s)
- Zhourui Wu
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, GA 30322, USA; Division of Spine, Department of Orthopedics, Tongji Hospital Affiliated to Tongji University School of Medicine, Shanghai, 200065, China; Key Laboratory of Spine and Spinal Cord Injury Repair and Regeneration, Ministry of Education of the People's Republic of China, Shanghai, 200072, China
| | - Zhi-Hao Wang
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Xia Liu
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Zhentao Zhang
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Xiaohuan Gu
- Department of Anesthesiology, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Shan Ping Yu
- Department of Anesthesiology, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - C Dirk Keene
- Department of Pathology, University of Washington School of Medicine, Seattle, WA, 98104, USA
| | - Liming Cheng
- Division of Spine, Department of Orthopedics, Tongji Hospital Affiliated to Tongji University School of Medicine, Shanghai, 200065, China; Key Laboratory of Spine and Spinal Cord Injury Repair and Regeneration, Ministry of Education of the People's Republic of China, Shanghai, 200072, China.
| | - Keqiang Ye
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, GA 30322, USA.
| |
Collapse
|
19
|
Van Ommeren R, Hazrati LN. Pathological Assessment of Chronic Traumatic Encephalopathy: Review of Concepts and Methodology. Acad Forensic Pathol 2019; 8:555-564. [PMID: 31240059 DOI: 10.1177/1925362118797729] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2018] [Accepted: 06/01/2018] [Indexed: 11/17/2022]
Abstract
Chronic traumatic encephalopathy (CTE) has become a topic of considerable interest in recent years, with wide-ranging implications for athletes, military members, and other groups exposed to frequent concussive or subconcussive head trauma. The condition has been subject to intensive neuropathological characterization by various groups, with assessment methodologies and staging criteria proposed. Clinical characterization of symptoms has also been performed, but has not yet been definitively formalized. While efforts are underway to develop in vivo markers of tauopathies including CTE, these remain experimental at this time, necessitating postmortem analysis for definitive diagnosis. The putative link between development of cognitive and behavioral dysfunction and neuropathological findings of CTE may prompt requests for postmortem assessment in the forensic setting. Here, we review current concepts in CTE research, describe histopathological findings in CTE, and describe methodologies for pathological assessment of CTE which may be useful to the forensic pathologist.
Collapse
|
20
|
Takahashi M, Yasuno F, Yamamuro K, Matsuoka K, Kitamura S, Yoshikawa H, Yamamoto A, Iida H, Fukuda T, Ihara M, Nagatsuka K, Kishimoto T. Detection of brain amyloid-β deposits due to the repetitive head trauma in a former karate player. Psychogeriatrics 2019; 19:276-281. [PMID: 30565811 DOI: 10.1111/psyg.12383] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/05/2018] [Revised: 08/30/2018] [Accepted: 09/24/2018] [Indexed: 11/30/2022]
Abstract
Head trauma is a well-established epidemiological risk factor for Alzheimer's disease, but a study of early detection of its pathology has not yet been performed in human patients in vivo. To address this issue, we performed 11 C-labelled Pittsburgh compound B-positron emission tomography on a right-handed 30-year-old man with cognitive deterioration after repetitive head trauma during karate matches. Structural magnetic resonance imaging was also performed on this patient. The same positron emission tomography analysis was performed on elderly healthy controls (15 men, mean age: 70.7 ± 6.2 years). To analyze grey matter volume, structural magnetic resonance imaging was performed on age-matched healthy controls (15 men, mean age: 28.5 ± 3.6 years). The cognitive deterioration in our patient was fixed and partially improved in the 10 years after the repetitive head trauma. However, Pittsburgh compound B-non-displaceable binding potential was significantly elevated in the patient. Volume reduction was shown in the medial temporal region, cerebellum, and the basal frontal cortex, while amyloid-β increase was shown in the bilateral prefrontal cortex. This is the first study to show an early degenerative process due to head trauma in the prefrontal cortex, where structural damage is not yet visible. Early recognition of the degenerative pathology due to repetitive head trauma by amyloid and possibly tau imaging would help clinicians determine how to treat those with early symptoms.
Collapse
Affiliation(s)
- Masato Takahashi
- Department of Psychiatry, Nara Medical University, Kashihara, Japan
| | - Fumihiko Yasuno
- Department of Psychiatry, Nara Medical University, Kashihara, Japan.,Department of Neurology, National Cerebral and Cardiovascular Center, Suita, Japan.,Department of Psychiatry, National Center for Geriatrics and Gerontology, Obu, Japan
| | | | - Kiwamu Matsuoka
- Department of Psychiatry, Nara Medical University, Kashihara, Japan
| | | | | | - Akihide Yamamoto
- Department of Investigative Radiology, National Cerebral and Cardiovascular Center, Suita, Japan
| | - Hidehiro Iida
- Department of Investigative Radiology, National Cerebral and Cardiovascular Center, Suita, Japan
| | - Tetsuya Fukuda
- Department of Radiology, National Cerebral and Cardiovascular Center, Suita, Japan
| | - Masafumi Ihara
- Department of Psychiatry, National Center for Geriatrics and Gerontology, Obu, Japan
| | - Kazuyuki Nagatsuka
- Department of Psychiatry, National Center for Geriatrics and Gerontology, Obu, Japan
| | | |
Collapse
|
21
|
Novel tau filament fold in chronic traumatic encephalopathy encloses hydrophobic molecules. Nature 2019; 568:420-423. [PMID: 30894745 PMCID: PMC6472968 DOI: 10.1038/s41586-019-1026-5] [Citation(s) in RCA: 510] [Impact Index Per Article: 85.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2018] [Accepted: 02/19/2019] [Indexed: 12/14/2022]
Abstract
Chronic traumatic encephalopathy (CTE) is a neurodegenerative tauopathy that is associated with repetitive head impacts or exposure to blast waves. First described as punch-drunk syndrome and dementia pugilistica in retired boxers1-3, CTE has since been identified in former participants of other contact sports, ex-military personnel and after physical abuse4-7. No disease-modifying therapies currently exist, and diagnosis requires an autopsy. CTE is defined by an abundance of hyperphosphorylated tau protein in neurons, astrocytes and cell processes around blood vessels8,9. This, together with the accumulation of tau inclusions in cortical layers II and III, distinguishes CTE from Alzheimer's disease and other tauopathies10,11. However, the morphologies of tau filaments in CTE and the mechanisms by which brain trauma can lead to their formation are unknown. Here we determine the structures of tau filaments from the brains of three individuals with CTE at resolutions down to 2.3 Å, using cryo-electron microscopy. We show that filament structures are identical in the three cases but are distinct from those of Alzheimer's and Pick's diseases, and from those formed in vitro12-15. Similar to Alzheimer's disease12,14,16-18, all six brain tau isoforms assemble into filaments in CTE, and residues K274-R379 of three-repeat tau and S305-R379 of four-repeat tau form the ordered core of two identical C-shaped protofilaments. However, a different conformation of the β-helix region creates a hydrophobic cavity that is absent in tau filaments from the brains of patients with Alzheimer's disease. This cavity encloses an additional density that is not connected to tau, which suggests that the incorporation of cofactors may have a role in tau aggregation in CTE. Moreover, filaments in CTE have distinct protofilament interfaces to those of Alzheimer's disease. Our structures provide a unifying neuropathological criterion for CTE, and support the hypothesis that the formation and propagation of distinct conformers of assembled tau underlie different neurodegenerative diseases.
Collapse
|
22
|
Subacute to chronic Alzheimer-like alterations after controlled cortical impact in human tau transgenic mice. Sci Rep 2019; 9:3789. [PMID: 30846870 PMCID: PMC6405988 DOI: 10.1038/s41598-019-40678-4] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2018] [Accepted: 02/21/2019] [Indexed: 12/14/2022] Open
Abstract
Repetitive traumatic brain injury (TBI) has been linked to late life development of chronic traumatic encephalopathy (CTE), a neurodegenerative disorder histopathologically characterized by perivascular tangles of hyperphosphorylated tau at the depth of sulci to later widespread neurofibrillary pathology. Although tau hyperphosphorylation and neurofibrillary-like pathology have been observed in the brain of transgenic mice overexpressing human tau with aggregation-prone mutation after TBI, they have not been consistently recapitulated in rodents expressing wild-type tau only. Here, we characterized Alzheimer-like alterations behaviorally, biochemically and immunohistochemically 6 weeks and 7 months after unilateral mild-to-moderate controlled cortical impact (CCI) in 5–7-month-old Tg/htau mice, which express all six isoforms of non-mutated human tau in a mouse tau null background. We detected hyperphosphorylation of tau at multiple sites in ipsilateral hippocampus 6 weeks but not 7 months after CCI. However, neuronal accumulation of AT8 positive phospho-tau was sustained in the chronic phase, in parallel to prolonged astrogliosis, and decreased neural and synaptic markers. The mice with CCI also exhibited cognitive and locomotor impairment. These results indicate subacute to chronic Alzheimer-like alterations after CCI in Tg/htau mice. This is the first known study providing insight into the role of CCI in Alzheimer-like brain alterations in young adult mice expressing only non-mutated human tau.
Collapse
|
23
|
Cheng Y, Pereira M, Raukar N, Reagan JL, Queseneberry M, Goldberg L, Borgovan T, LaFrance WC, Dooner M, Deregibus M, Camussi G, Ramratnam B, Quesenberry P. Potential biomarkers to detect traumatic brain injury by the profiling of salivary extracellular vesicles. J Cell Physiol 2019; 234:14377-14388. [PMID: 30644102 PMCID: PMC6478516 DOI: 10.1002/jcp.28139] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2018] [Accepted: 12/13/2018] [Indexed: 12/11/2022]
Abstract
Traumatic brain injury (TBI) is a common cause of death and acquired disability in adults and children. Identifying biomarkers for mild TBI (mTBI) that can predict functional impairments on neuropsychiatric and neurocognitive testing after head trauma is yet to be firmly established. Extracellular vesicles (EVs) are known to traffic from the brain to the oral cavity and can be detected in saliva. We hypothesize the genetic profile of salivary EVs in patients who have suffered head trauma will differ from normal healthy controls, thus constituting a unique expression signature for mTBI. We enrolled a total of 54 subjects including for saliva sampling, 23 controls with no history of head traumas, 16 patients enrolled from an outpatient concussion clinic, and 15 patients from the emergency department who had sustained a head trauma within 24 hr. We performed real‐time PCR of the salivary EVs of the 54 subjects profiling 96 genes from the TaqMan Human Alzheimer's disease array. Real‐time PCR analysis revealed 57 (15 genes, p < 0.05) upregulated genes in emergency department patients and 56 (14 genes,
p < 0.05) upregulated genes in concussion clinic patients when compared with controls. Three genes were upregulated in both the emergency department patients and concussion clinic patients: CDC2, CSNK1A1, and CTSD (
p < 0.05). Our results demonstrate that salivary EVs gene expression can serve as a viable source of biomarkers for mTBI. This study shows multiple Alzheimer's disease genes present after an mTBI.
Collapse
Affiliation(s)
- Yan Cheng
- Department of Medicine Division of Hematology/Oncology, Rhode Island Hospital, Providence, Rhode Island
| | - Mandy Pereira
- Department of Medicine Division of Hematology/Oncology, Rhode Island Hospital, Providence, Rhode Island
| | - Neha Raukar
- Department of Emergency Medicine, Rhode Island Hospital, Providence, Rhode Island
| | - John L Reagan
- Department of Medicine Division of Hematology/Oncology, Rhode Island Hospital, Providence, Rhode Island
| | - Mathew Queseneberry
- Department of Medicine Division of Hematology/Oncology, Rhode Island Hospital, Providence, Rhode Island
| | - Laura Goldberg
- Department of Medicine Division of Hematology/Oncology, Rhode Island Hospital, Providence, Rhode Island
| | - Theodor Borgovan
- Department of Medicine Division of Hematology/Oncology, Rhode Island Hospital, Providence, Rhode Island
| | - W Curt LaFrance
- Department of Psychiatry/Neurology, Rhode Island Hospital, Providence, Rhode Island
| | - Mark Dooner
- Department of Medicine Division of Hematology/Oncology, Rhode Island Hospital, Providence, Rhode Island
| | - Maria Deregibus
- Department of Medical Sciences, University of Turin, Turin, Italy
| | - Giovanni Camussi
- Department of Medical Sciences, University of Turin, Turin, Italy
| | - Bharat Ramratnam
- Department of Medicine Division of Infectious Diseases, Rhode Island Hospital, Providence, Rhode Island
| | - Peter Quesenberry
- Department of Medicine Division of Hematology/Oncology, Rhode Island Hospital, Providence, Rhode Island
| |
Collapse
|
24
|
Castellani RJ, Perry G. Tau Biology, Tauopathy, Traumatic Brain Injury, and Diagnostic Challenges. J Alzheimers Dis 2019; 67:447-467. [PMID: 30584140 PMCID: PMC6398540 DOI: 10.3233/jad-180721] [Citation(s) in RCA: 61] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/08/2018] [Indexed: 12/12/2022]
Abstract
There is considerable interest in the pathobiology of tau protein, given its potential role in neurodegenerative diseases and aging. Tau is an important microtubule associated protein, required for the assembly of tubulin into microtubules and maintaining structural integrity of axons. Tau has other diverse cellular functions involving signal transduction, cellular proliferation, developmental neurobiology, neuroplasticity, and synaptic activity. Alternative splicing results in tau isoforms with differing microtubule binding affinity, differing representation in pathological inclusions in certain disease states, and differing roles in developmental biology and homeostasis. Tau haplotypes confer differing susceptibility to neurodegeneration. Tau phosphorylation is a normal metabolic process, critical in controlling tau's binding to microtubules, and is ongoing within the brain at all times. Tau may be hyperphosphorylated, and may aggregate as detectable fibrillar deposits in tissues, in both aging and neurodegenerative disease. The hypothesis that p-tau is neurotoxic has prompted constructs related to isomers, low-n assembly intermediates or oligomers, and the "tau prion". Human postmortem studies have elucidated broad patterns of tauopathy, with tendencies for those patterns to differ as a function of disease phenotype. However, there is extensive overlap, not only between genuine neurodegenerative diseases, but also between aging and disease. Recent studies highlight uniqueness to pathological patterns, including a pattern attributed to repetitive head trauma, although clinical correlations have been elusive. The diagnostic process for tauopathies and neurodegenerative diseases in general is challenging in many respects, and may be particularly problematic for postmortem evaluation of former athletes and military service members.
Collapse
Affiliation(s)
- Rudy J. Castellani
- Departments of Pathology and Neuroscience, West Virginia University School of Medicine, Morgantown, WV, USA
| | - George Perry
- College of Sciences, University of Texas, San Antonio, San Antonio, TX, USA
| |
Collapse
|
25
|
DeLeo AM, Ikezu T. Extracellular Vesicle Biology in Alzheimer's Disease and Related Tauopathy. J Neuroimmune Pharmacol 2018; 13:292-308. [PMID: 29185187 PMCID: PMC5972041 DOI: 10.1007/s11481-017-9768-z] [Citation(s) in RCA: 77] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2017] [Accepted: 10/23/2017] [Indexed: 12/14/2022]
Abstract
Extracellular vesicles (EVs) are physiological vesicles secreted from most eukaryotes and contain cargos of their cell of origin. EVs, and particularly a subset of EV known as exosomes, are emerging as key mediators of cell to cell communication and waste management for cells both during normal organismal function and in disease. In this review, we investigate the rapidly growing field of exosome biology, their biogenesis, cargo loading, and uptake by other cells. We particularly consider the role of exosomes in Alzheimer's disease, both as a pathogenic agent and as a disease biomarker. We also explore the emerging role of exosomes in chronic traumatic encephalopathy. Finally, we highlight open questions in these fields and the possible use of exosomes as therapeutic targets and agents.
Collapse
Affiliation(s)
- Annina M DeLeo
- Laboratory of Molecular NeuroTherapeutics, Department of Pharmacology & Experimental Therapeutics, Boston University School of Medicine, 72 East Concord St, L-606, Boston, MA, 02118, USA.
| | - Tsuneya Ikezu
- Laboratory of Molecular NeuroTherapeutics, Department of Pharmacology & Experimental Therapeutics, Boston University School of Medicine, 72 East Concord St, L-606, Boston, MA, 02118, USA.
- Department of Neurology, Boston University School of Medicine, Boston, MA, 02118, USA.
| |
Collapse
|
26
|
Chauderlier A, Gilles M, Spolcova A, Caillierez R, Chwastyniak M, Kress M, Drobecq H, Bonnefoy E, Pinet F, Weil D, Buée L, Galas MC, Lefebvre B. Tau/DDX6 interaction increases microRNA activity. BIOCHIMICA ET BIOPHYSICA ACTA-GENE REGULATORY MECHANISMS 2018; 1861:762-772. [DOI: 10.1016/j.bbagrm.2018.06.006] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/11/2018] [Revised: 06/27/2018] [Accepted: 06/27/2018] [Indexed: 12/17/2022]
|
27
|
Abstract
Extensive exposure of boxers to neurotrauma in the early 20th century led to the so-called punch drunk syndrome, which was formally recognized in the medical literature in 1928. "Punch drunk" terminology was replaced by the less derisive 'dementia pugilistica' in 1937. In the early case material, the diagnosis of dementia pugilistica required neurological deficits, including slurring dysarthria, ataxia, pyramidal signs, extrapyramidal signs, memory impairment, and personality changes, although the specific clinical substrate has assumed lesser importance in recent years with a shift in focus on molecular pathogenesis. The postmortem neuropathology of dementia pugilistica has also evolved substantially over the past 90 years, from suspected concussion-related hemorrhages to diverse structural and neurofibrillary changes to geographic tauopathy. Progressive neurodegenerative tauopathy is among the prevailing theories for disease pathogenesis currently, although this may be overly simplistic. Careful examination of historical cases reveals both misdiagnoses and a likelihood that dementia pugilistica at that time was caused by cumulative structural brain injury. More recent neuropathological studies indicate subclinical and possibly static tauopathy in some athletes and non-athletes. Indeed, it is unclear from the literature whether retired boxers reach the inflection point that tends toward progressive neurodegeneration in the manner of Alzheimer's disease due to boxing. Even among historical cases with extreme levels of exposure, progressive disease was exceptional.
Collapse
Affiliation(s)
- Rudy J Castellani
- Center for Neuropathology, Western Michigan University School of Medicine, Kalamazoo, MI, USA
| | - George Perry
- College of Sciences, University of Texas, San Antonio, San Antonio, TX, USA
| |
Collapse
|
28
|
Ling H, Neal JW, Revesz T. Evolving concepts of chronic traumatic encephalopathy as a neuropathological entity. Neuropathol Appl Neurobiol 2018; 43:467-476. [PMID: 28664614 DOI: 10.1111/nan.12425] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2017] [Revised: 06/20/2017] [Accepted: 06/30/2017] [Indexed: 12/14/2022]
Abstract
Chronic traumatic encephalopathy (CTE) is a long-term neurodegenerative consequence of repetitive head impacts which can only be definitively diagnosed in post-mortem. Recently, the consensus neuropathological criteria for the diagnosis of CTE was published requiring the presence of the accumulation of abnormal tau in neurons and astroglia distributed around small blood vessels at the depths of cortical sulci in an irregular pattern as the mandatory features. The clinical diagnosis and antemortem prediction of CTE pathology remain challenging if not impossible due to the common co-existing underlying neurodegenerative pathologies and the lack of specific clinical pointers and reliable biomarkers. This review summarizes the historical evolution of CTE as a neuropathological entity and highlights the latest advances and future directions of research studies on the topic of CTE.
Collapse
Affiliation(s)
- H Ling
- Queen Square Brain Bank for Neurological Disorders, UCL Institute of Neurology, University College London, London, UK.,Reta Lila Weston Institute for Neurological Studies, UCL Institute of Neurology, London, UK.,Department of Molecular Neuroscience, UCL Institute of Neurology, University College London, London, UK
| | - J W Neal
- Department of Cellular Pathology, Cardiff University, Wales, UK
| | - T Revesz
- Queen Square Brain Bank for Neurological Disorders, UCL Institute of Neurology, University College London, London, UK.,Reta Lila Weston Institute for Neurological Studies, UCL Institute of Neurology, London, UK.,Department of Molecular Neuroscience, UCL Institute of Neurology, University College London, London, UK
| |
Collapse
|
29
|
Kneynsberg A, Combs B, Christensen K, Morfini G, Kanaan NM. Axonal Degeneration in Tauopathies: Disease Relevance and Underlying Mechanisms. Front Neurosci 2017; 11:572. [PMID: 29089864 PMCID: PMC5651019 DOI: 10.3389/fnins.2017.00572] [Citation(s) in RCA: 75] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2017] [Accepted: 09/29/2017] [Indexed: 12/14/2022] Open
Abstract
Tauopathies are a diverse group of diseases featuring progressive dying-back neurodegeneration of specific neuronal populations in association with accumulation of abnormal forms of the microtubule-associated protein tau. It is well-established that the clinical symptoms characteristic of tauopathies correlate with deficits in synaptic function and neuritic connectivity early in the course of disease, but mechanisms underlying these critical pathogenic events are not fully understood. Biochemical in vitro evidence fueled the widespread notion that microtubule stabilization represents tau's primary biological role and that the marked atrophy of neurites observed in tauopathies results from loss of microtubule stability. However, this notion contrasts with the mild phenotype associated with tau deletion. Instead, an analysis of cellular hallmarks common to different tauopathies, including aberrant patterns of protein phosphorylation and early degeneration of axons, suggests that alterations in kinase-based signaling pathways and deficits in axonal transport (AT) associated with such alterations contribute to the loss of neuronal connectivity triggered by pathogenic forms of tau. Here, we review a body of literature providing evidence that axonal pathology represents an early and common pathogenic event among human tauopathies. Observations of axonal degeneration in animal models of specific tauopathies are discussed and similarities to human disease highlighted. Finally, we discuss potential mechanistic pathways other than microtubule destabilization by which disease-related forms of tau may promote axonopathy.
Collapse
Affiliation(s)
- Andrew Kneynsberg
- Neuroscience Program, Michigan State University, East Lansing, MI, United States.,Department of Translational Science and Molecular Medicine, College of Human Medicine, Michigan State University, Grand Rapids, MI, United States
| | - Benjamin Combs
- Department of Translational Science and Molecular Medicine, College of Human Medicine, Michigan State University, Grand Rapids, MI, United States
| | - Kyle Christensen
- Neuroscience Program, Michigan State University, East Lansing, MI, United States.,Department of Translational Science and Molecular Medicine, College of Human Medicine, Michigan State University, Grand Rapids, MI, United States
| | - Gerardo Morfini
- Department of Anatomy and Cell Biology, University of Illinois at Chicago, Chicago, IL, United States
| | - Nicholas M Kanaan
- Neuroscience Program, Michigan State University, East Lansing, MI, United States.,Department of Translational Science and Molecular Medicine, College of Human Medicine, Michigan State University, Grand Rapids, MI, United States.,Hauenstein Neuroscience Center, Mercy Health Saint Mary's, Grand Rapids, MI, United States
| |
Collapse
|
30
|
Wang ML, Wei XE, Yu MM, Li PY, Li WB. Self-reported traumatic brain injury and in vivo measure of AD-vulnerable cortical thickness and AD-related biomarkers in the ADNI cohort. Neurosci Lett 2017; 655:115-120. [PMID: 28689050 DOI: 10.1016/j.neulet.2017.06.055] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2017] [Revised: 06/24/2017] [Accepted: 06/28/2017] [Indexed: 11/27/2022]
Abstract
In this study, we aimed to investigate whether self-reported mild traumatic brain injury (mTBI) was associated with decreased AD-vulnerable cortical thickness, and to assess the relationship between AD-vulnerable cortical thickness and AD-related biomarker in the Alzheimer's Disease Neuroimaging Initiative subjects. We identified 45 self-reported mTBI subjects, who had structural MRI, 18F-AV45 PET, and cerebrospinal fluid (CSF) data. Of them, eight subjects were normal; ten were preclinical AD; seventeen were MCI due to AD; ten were AD. Additional demographics-controlled 45 subjects were included. Cortical thickness of eight AD-vulnerable regions, mean AD-vulnerable cortical thickness, 18F-AV45 PET mean amyloid SUVR, CSF Aβ42, CSF total tau (T-tau), and CSF phosphorylated tau (P-tau) were compared between mTBI and non-TBI groups. Correlational analysis was done to investigate the relationship between mean AD-vulnerable cortical thickness and mean amyloid SUVR, CSF Aβ42, CSF T-Tau, CSF P-Tau. Our study revealed that preclinical AD subjects with self-reported mTBI had smaller cortical thickness in mean and three AD-vulnerable cortical regions than non-TBI subjects (P<0.05). The mean AD-vulnerable cortical thickness was correlated with CSF T-tau (r=-0.81, P=0.001). There was no statistical difference in the comparison of normal, MCI due to AD, and AD groups. Our study indicated that among individuals with preclinical AD, but not normal, MCI due to AD and AD subjects, self-reported mTBI was associated with more decreased AD-vulnerable cortical thickness which was related to CSF tau pathology, suggesting the possible early involvement of tau pathology in the decreased AD-vulnerable cortical thickness of self-reported TBI subjects.
Collapse
Affiliation(s)
- Ming-Liang Wang
- Department of Radiology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200233, China
| | - Xiao-Er Wei
- Department of Radiology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200233, China
| | - Meng-Meng Yu
- Department of Radiology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200233, China
| | - Peng-Yang Li
- Department of Cardiology, Peking University Aerospace School of Clinical Medicine, Peking University Health Science Center, Beijing 100049, China
| | - Wen-Bin Li
- Department of Radiology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200233, China; Imaging Center, Kashgar Prefecture Second People's Hospital, Kashgar 844000, China.
| | | |
Collapse
|
31
|
Shively SB, Edgerton SL, Iacono D, Purohit DP, Qu BX, Haroutunian V, Davis KL, Diaz-Arrastia R, Perl DP. Localized cortical chronic traumatic encephalopathy pathology after single, severe axonal injury in human brain. Acta Neuropathol 2017; 133:353-366. [PMID: 27885490 PMCID: PMC5325841 DOI: 10.1007/s00401-016-1649-7] [Citation(s) in RCA: 44] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2016] [Revised: 11/17/2016] [Accepted: 11/19/2016] [Indexed: 12/14/2022]
Abstract
Chronic traumatic encephalopathy (CTE) is a neurodegenerative disease associated with repetitive mild impact traumatic brain injury from contact sports. Recently, a consensus panel defined the pathognomonic lesion for CTE as accumulations of abnormally hyperphosphorylated tau (p-tau) in neurons (neurofibrillary tangles), astrocytes and cell processes distributed around small blood vessels at sulcal depths in irregular patterns within the cortex. The pathophysiological mechanism for this lesion is unknown. Moreover, a subset of CTE cases harbors cortical β-amyloid plaques. In this study, we analyzed postmortem brain tissues from five institutionalized patients with schizophrenia and history of surgical leucotomy with subsequent survival of at least another 40 years. Because leucotomy involves severing axons bilaterally in prefrontal cortex, this surgical procedure represents a human model of single traumatic brain injury with severe axonal damage and no external impact. We examined cortical tissues at the leucotomy site and at both prefrontal cortex rostral and frontal cortex caudal to the leucotomy site. For comparison, we analyzed brain tissues at equivalent neuroanatomical sites from non-leucotomized patients with schizophrenia, matched in age and gender. All five leucotomy cases revealed severe white matter damage with dense astrogliosis at the axotomy site and also neurofibrillary tangles and p-tau immunoreactive neurites in the overlying gray matter. Four cases displayed p-tau immunoreactivity in neurons, astrocytes and cell processes encompassing blood vessels at cortical sulcal depths in irregular patterns, similar to CTE. The three cases with apolipoprotein E ε4 haplotype showed scattered β-amyloid plaques in the overlying gray matter, but not the two cases with apolipoprotein E ε3/3 genotype. Brain tissue samples from prefrontal cortex rostral and frontal cortex caudal to the leucotomy site, and all cortical samples from the non-leucotomized patients, showed minimal p-tau and β-amyloid pathology. These findings suggest that chronic axonal damage contributes to the unique pathology of CTE over time.
Collapse
Affiliation(s)
- Sharon B Shively
- Department of Pathology, F. Edward Hébert School of Medicine, Uniformed Services University of the Health Sciences, 4301 Jones Bridge Road, Bethesda, MD, 20814, USA
- Center for Neuroscience and Regenerative Medicine, 4301 Jones Bridge Road, Bethesda, MD, 20814, USA
- Henry M. Jackson Foundation for the Advancement of Military Medicine, 6720 Rockledge Drive, Bethesda, MD, 20817, USA
| | - Sarah L Edgerton
- Center for Neuroscience and Regenerative Medicine, 4301 Jones Bridge Road, Bethesda, MD, 20814, USA
- Henry M. Jackson Foundation for the Advancement of Military Medicine, 6720 Rockledge Drive, Bethesda, MD, 20817, USA
| | - Diego Iacono
- Department of Pathology, F. Edward Hébert School of Medicine, Uniformed Services University of the Health Sciences, 4301 Jones Bridge Road, Bethesda, MD, 20814, USA
- Center for Neuroscience and Regenerative Medicine, 4301 Jones Bridge Road, Bethesda, MD, 20814, USA
- Henry M. Jackson Foundation for the Advancement of Military Medicine, 6720 Rockledge Drive, Bethesda, MD, 20817, USA
| | - Dushyant P Purohit
- Department of Pathology, Icahn School of Medicine at Mount Sinai, 1468 Madison Avenue, New York, NY, 10029, USA
| | - Bao-Xi Qu
- Center for Neuroscience and Regenerative Medicine, 4301 Jones Bridge Road, Bethesda, MD, 20814, USA
- Department of Neurology, F. Edward Hébert School of Medicine, Uniformed Services University of the Health Sciences, 4301 Jones Bridge Road, Bethesda, MD, 20814, USA
| | - Vahram Haroutunian
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, 1 Gustave L. Levy Place, New York, NY, 10029, USA
- Department of Neuroscience, Icahn School of Medicine at Mount Sinai, 1 Gustave L. Levy Place, New York, NY, 10029, USA
- Mental Illness Research Education and Clinical Center, James J. Peters Veterans Affairs Medical Center, 130 West Kingsbridge Road, Bronx, NY, 10468, USA
| | - Kenneth L Davis
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, 1 Gustave L. Levy Place, New York, NY, 10029, USA
- Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, 1 Gustave L. Levy Place, New York, NY, 10029, USA
| | - Ramon Diaz-Arrastia
- Center for Neuroscience and Regenerative Medicine, 4301 Jones Bridge Road, Bethesda, MD, 20814, USA
- Department of Neurology, F. Edward Hébert School of Medicine, Uniformed Services University of the Health Sciences, 4301 Jones Bridge Road, Bethesda, MD, 20814, USA
| | - Daniel P Perl
- Department of Pathology, F. Edward Hébert School of Medicine, Uniformed Services University of the Health Sciences, 4301 Jones Bridge Road, Bethesda, MD, 20814, USA.
- Center for Neuroscience and Regenerative Medicine, 4301 Jones Bridge Road, Bethesda, MD, 20814, USA.
| |
Collapse
|
32
|
Bangen KJ, Clark AL, Werhane M, Edmonds EC, Nation DA, Evangelista N, Libon DJ, Bondi MW, Delano-Wood L. Cortical Amyloid Burden Differences Across Empirically-Derived Mild Cognitive Impairment Subtypes and Interaction with APOE ɛ4 Genotype. J Alzheimers Dis 2017; 52:849-61. [PMID: 27031472 DOI: 10.3233/jad-150900] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
We examined cortical amyloid-β (Aβ) levels and interactions with apolipoprotein (APOE) ɛ4 genotype status across empirically-derived mild cognitive impairment (MCI) subgroups and cognitively normal older adults. Participants were 583 ADNI participants (444 MCI, 139 normal controls [NC]) with baseline florbetapir positron emission tomography (PET) amyloid imaging and neuropsychological testing. Of those with ADNI-defined MCI, a previous cluster analysis [1] classified 51% (n = 227) of the current sample as amnestic MCI, 8% (n = 37) as dysexecutive/mixed MCI, and 41% (n = 180) as cluster-derived normal (cognitively normal). Results demonstrated that the dysexecutive/mixed and amnestic MCI groups showed significantly greater levels of amyloid relative to the cluster-derived normal and NC groups who did not differ from each other. Additionally, 78% of the dysexecutive/mixed, 63% of the amnestic MCI, 42% of the cluster-derived normal, and 34% of the NC group exceeded the amyloid positivity threshold. Finally, a group by APOE genotype interaction demonstrated that APOE ɛ4 carriers within the amnestic MCI, cluster-derived normal, and NC groups showed significantly greater amyloid accumulation compared to non-carriers of their respective group. Such an interaction was not revealed within the dysexecutive/mixed MCI group which was characterized by both greater cognitive impairment and amyloid accumulation compared to the other participant groups. Our results from the ADNI cohort show considerable heterogeneity in Aβ across all groups studied, even within a group of robust NC participants. Findings suggest that conventional criteria for MCI may be susceptible to false positive diagnostic errors, and that onset of Aβ accumulation may occur earlier in APOE ɛ4 carriers compared to non-carriers.
Collapse
Affiliation(s)
- Katherine J Bangen
- Veterans Affairs San Diego Healthcare System, San Diego, CA, USA.,Department of Psychiatry, University of California, San Diego, La Jolla, CA, USA
| | - Alexandra L Clark
- Veterans Affairs San Diego Healthcare System, San Diego, CA, USA.,San Diego State University/University of California, San Diego (SDSU/UCSD) Joint Doctoral Program in Clinical Psychology, San Diego, CA, USA
| | - Madeline Werhane
- San Diego State University/University of California, San Diego (SDSU/UCSD) Joint Doctoral Program in Clinical Psychology, San Diego, CA, USA
| | - Emily C Edmonds
- Veterans Affairs San Diego Healthcare System, San Diego, CA, USA.,Department of Psychiatry, University of California, San Diego, La Jolla, CA, USA
| | - Daniel A Nation
- Department of Psychology, University of Southern California, Los Angeles, CA, USA
| | - Nicole Evangelista
- Veterans Affairs San Diego Healthcare System, San Diego, CA, USA.,Department of Psychiatry, University of California, San Diego, La Jolla, CA, USA
| | - David J Libon
- Department of Neurology, Drexel University College of Medicine, Philadelphia, PA, USA
| | - Mark W Bondi
- Veterans Affairs San Diego Healthcare System, San Diego, CA, USA.,Department of Psychiatry, University of California, San Diego, La Jolla, CA, USA
| | - Lisa Delano-Wood
- Veterans Affairs San Diego Healthcare System, San Diego, CA, USA.,Department of Psychiatry, University of California, San Diego, La Jolla, CA, USA
| | | |
Collapse
|
33
|
Johnson VE, Stewart W, Arena JD, Smith DH. Traumatic Brain Injury as a Trigger of Neurodegeneration. ADVANCES IN NEUROBIOLOGY 2017; 15:383-400. [PMID: 28674990 DOI: 10.1007/978-3-319-57193-5_15] [Citation(s) in RCA: 70] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Although millions of individuals suffer a traumatic brain injury (TBI) worldwide each year, it is only recently that TBI has been recognized as a major public health problem. Beyond the acute clinical manifestations, there is growing recognition that a single severe TBI (sTBI) or repeated mild TBIs (rTBI) can also induce insidious neurodegenerative processes, which may be associated with early dementia, in particular chronic traumatic encephalopathy (CTE). Identified at autopsy examination in individuals with histories of exposure to sTBI or rTBI, CTE is recognized as a complex pathology featuring both macroscopic and microscopic abnormalities. These include cavum septum pellucidum, brain atrophy and ventricular dilation, together with pathologies in tau, TDP-43, and amyloid-β. However, the establishment and characterization of CTE as a distinct disease entity is in its infancy. Moreover, the relative "dose" of TBI, such as the frequency and severity of injury, associated with risk of CTE remains unknown. As such, there is a clear and pressing need to improve the recognition and diagnosis of CTE and to identify mechanistic links between TBI and chronic neurodegeneration.
Collapse
Affiliation(s)
- Victoria E Johnson
- Department of Neurosurgery, Penn Center for Brain Injury and Repair, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - William Stewart
- Department of Neurosurgery, Penn Center for Brain Injury and Repair, University of Pennsylvania, Philadelphia, PA, 19104, USA.,Department of Neuropathology, Queen Elizabeth University Hospital, Glasgow, G51 4TF, UK.,University of Glasgow, Glasgow, G12 8QQ, UK
| | - John D Arena
- Department of Neurosurgery, Penn Center for Brain Injury and Repair, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Douglas H Smith
- Department of Neurosurgery, Penn Center for Brain Injury and Repair, University of Pennsylvania, Philadelphia, PA, 19104, USA.
| |
Collapse
|
34
|
Lee HH, Lee WH, Seo HG, Han D, Kim Y, Oh BM. Current State and Prospects of Development of Blood-based Biomarkers for Mild Traumatic Brain Injury. BRAIN & NEUROREHABILITATION 2017. [DOI: 10.12786/bn.2017.10.e3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022] Open
Affiliation(s)
- Hyun Haeng Lee
- Department of Rehabilitation Medicine, Seoul National University College of Medicine, Seoul National University Hospital, Seoul, Korea
| | - Woo Hyung Lee
- Department of Rehabilitation Medicine, Seoul National University College of Medicine, Seoul National University Hospital, Seoul, Korea
| | - Han Gil Seo
- Department of Rehabilitation Medicine, Seoul National University College of Medicine, Seoul National University Hospital, Seoul, Korea
| | - Dohyun Han
- Proteomics Core Facility, Biomedical Research Institute, Seoul National University Hospital, Seoul, Korea
| | - Youngsoo Kim
- Department of Biomedical Engineering, Seoul National University College of Medicine, Seoul, Korea
| | - Byung-Mo Oh
- Department of Rehabilitation Medicine, Seoul National University College of Medicine, Seoul National University Hospital, Seoul, Korea
| |
Collapse
|
35
|
Role of neurotoxicants and traumatic brain injury in α-synuclein protein misfolding and aggregation. Brain Res Bull 2016; 133:60-70. [PMID: 27993598 DOI: 10.1016/j.brainresbull.2016.12.003] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2016] [Revised: 11/23/2016] [Accepted: 12/15/2016] [Indexed: 12/14/2022]
Abstract
Protein misfolding and aggregation are key pathological features of many neurodegenerative diseases including Parkinson's disease (PD) and other forms of human Parkinsonism. PD is a complex and multifaceted disorder whose etiology is not fully understood. However, several lines of evidence support the multiple hit hypothesis that genetic vulnerability and environmental toxicants converge to trigger PD pathology. Alpha-synuclein (α-Syn) aggregation in the brain is an important pathophysiological characteristic of synucleinopathies including PD. Epidemiological and experimental studies have shown that metals and pesticides play a crucial role in α-Syn aggregation leading to the onset of various neurodegenerative diseases including PD. In this review, we will emphasize key findings of several epidemiological as well as experimental studies of metal- and pesticide-induced α-Syn aggregation and neurodegeneration. We will also discuss other factors such as traumatic brain injury and oxidative insult in the context of α-Syn-related neurodegenerative processes.
Collapse
|
36
|
Luczkowski M. “No screams and cries will convince us that white is white and black is black”, an ode to the defenders of amyloid cascade hypothesis of Alzheimer's disease. Coord Chem Rev 2016. [DOI: 10.1016/j.ccr.2016.03.001] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
|
37
|
Abstract
Axonal damage is one of the most common and important pathologic features of traumatic brain injury. Severe diffuse axonal injury, resulting from inertial forces applied to the head, is associated with prolonged unconsciousness and poor outcome. The susceptibility of axons to mechanical injury appears to be due to both their viscoelastic properties and their highly organized structure in white matter tracts. Although axons are supple under normal conditions, they become brittle when exposed to rapid deformations associated with brain trauma. Accordingly, rapid stretch of axons can damage the axonal cytoskeleton, resulting in a loss of elasticity and impairment of axoplasmic transport. Subsequent swelling of the axon occurs in discrete bulb formations or in elongated varicosities that accumulate organelles. Calcium entry into damaged axons is thought to initiate further damage by the activation of proteases and the induction of mitochondrial swelling and dysfunction. Ultimately, swollen axons may become disconnected and contribute to additional neuropathologic changes in brain tissue. However, promising new therapies that reduce proteolytic activity or maintain mitochondrial integrity may attenuate progressive damage of injured axons following experimental brain trauma. Future advancements in the prevention and treatment of traumatic axonal injury will depend on our collective understanding of the relationship between the biomechanics and pathophysiology of various phases of axonal trauma.
Collapse
Affiliation(s)
- Douglas H. Smith
- Department of Neurosurgery, University of Pennsylvania, Philadelphia, Pennsylvania,
| | - David F. Meaney
- Department of Bioengineering, University of Pennsylvania, Philadelphia, Pennsylvania
| |
Collapse
|
38
|
Kanaan NM, Cox K, Alvarez VE, Stein TD, Poncil S, McKee AC. Characterization of Early Pathological Tau Conformations and Phosphorylation in Chronic Traumatic Encephalopathy. J Neuropathol Exp Neurol 2016; 75:19-34. [PMID: 26671985 DOI: 10.1093/jnen/nlv001] [Citation(s) in RCA: 76] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Chronic traumatic encephalopathy (CTE) is a neurodegenerative tauopathy that develops after repetitive head injury. Several lines of evidence in other tauopathies suggest that tau oligomer formation induces neurotoxicity and that tau oligomer-mediated neurotoxicity involves induction of axonal dysfunction through exposure of an N-terminal motif in tau, the phosphatase-activating domain (PAD). Additionally, phosphorylation at serine 422 in tau occurs early and correlates with cognitive decline in patients with Alzheimer disease (AD). We performed immunohistochemistry and immunofluorescence on fixed brain sections and biochemical analysis of fresh brain extracts to characterize the presence of PAD-exposed tau (TNT1 antibody), tau oligomers (TOC1 antibody), tau phosphorylated at S422 (pS422 antibody), and tau truncated at D421 (TauC3 antibody) in the brains of 9-11 cases with CTE and cases of nondemented aged controls and AD (Braak VI) (n = 6, each). All 3 early tau markers (ie, TNT1, TOC1, and pS422) were present in CTE and displayed extensive colocalization in perivascular tau lesions that are considered diagnostic for CTE. Notably, the TauC3 epitope, which is abundant in AD, was relatively sparse in CTE. Together, these results provide the first description of PAD exposure, TOC1 reactive oligomers, phosphorylation of S422, and TauC3 truncation in the tau pathology of CTE.
Collapse
|
39
|
Kovacs GG, Lutz MI, Ricken G, Ströbel T, Höftberger R, Preusser M, Regelsberger G, Hönigschnabl S, Reiner A, Fischer P, Budka H, Hainfellner JA. Dura mater is a potential source of Aβ seeds. Acta Neuropathol 2016; 131:911-23. [PMID: 27016065 PMCID: PMC4865536 DOI: 10.1007/s00401-016-1565-x] [Citation(s) in RCA: 74] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2016] [Revised: 03/14/2016] [Accepted: 03/15/2016] [Indexed: 12/14/2022]
Abstract
Deposition of amyloid-β (Aβ) in the brain parenchyma and vessels is one of the hallmarks of Alzheimer disease (AD). Recent observations of Aβ deposition in iatrogenic Creutzfeldt-Jakob disease (iCJD) after dural grafting or treatment with pituitary extracts raised concerns whether Aβ is capable of transmitting disease as seen in prion diseases by the disease-associated prion protein. To address this issue, we re-sampled and re-evaluated archival material, including the grafted dura mater of two cases with iCJD (28 and 33-years-old) without mutations in the AβPP, PSEN1 and PSEN2 genes, and carrying ε3/ε3 alleles of the APOE gene. In addition, we evaluated 84 dura mater samples obtained at autopsy (mean age 84.9 ± 0.3) in the community-based VITA study for the presence of Aβ deposition. We show that the dura mater may harbor Aβ deposits (13 %) in the form of cerebral amyloid angiopathy or amorphous aggregates. In both iCJD cases, the grafted dura mater had accumulated Aβ. The morphology and distribution pattern of cerebral Aβ deposition together with the lack of tau pathology distinguishes the Aβ proteinopathy in iCJD from AD, from that seen in young individuals without cognitive decline carrying one or two APOE4 alleles, and from that related to traumatic brain injury. Our novel findings of Aβ deposits in the dura mater, including the grafted dura, and the distinct cerebral Aβ distribution in iCJD support the seeding properties of Aβ. However, in contrast to prion diseases, our study suggests that such Aβ seeding is unable to reproduce the full clinicopathological phenotype of AD.
Collapse
Affiliation(s)
- Gabor G Kovacs
- Institute of Neurology, Medical University Vienna, AKH 4J, Währinger Gürtel 18-20, 1097, Vienna, Austria.
| | - Mirjam I Lutz
- Institute of Neurology, Medical University Vienna, AKH 4J, Währinger Gürtel 18-20, 1097, Vienna, Austria
| | - Gerda Ricken
- Institute of Neurology, Medical University Vienna, AKH 4J, Währinger Gürtel 18-20, 1097, Vienna, Austria
| | - Thomas Ströbel
- Institute of Neurology, Medical University Vienna, AKH 4J, Währinger Gürtel 18-20, 1097, Vienna, Austria
| | - Romana Höftberger
- Institute of Neurology, Medical University Vienna, AKH 4J, Währinger Gürtel 18-20, 1097, Vienna, Austria
| | - Matthias Preusser
- Department of Medicine I and Comprehensive Cancer Center CNS Unit, Medical University Vienna, Vienna, Austria
| | - Günther Regelsberger
- Institute of Neurology, Medical University Vienna, AKH 4J, Währinger Gürtel 18-20, 1097, Vienna, Austria
| | | | - Angelika Reiner
- Institute of Pathology, Danube Hospital Vienna, Vienna, Austria
| | - Peter Fischer
- Psychiatric Department, Medical Research Society Vienna, D.C., Danube Hospital, Vienna, Austria
| | - Herbert Budka
- Institute of Neurology, Medical University Vienna, AKH 4J, Währinger Gürtel 18-20, 1097, Vienna, Austria
- Institute of Neuropathology, University Hospital Zurich, Zurich, Switzerland
| | - Johannes A Hainfellner
- Institute of Neurology, Medical University Vienna, AKH 4J, Währinger Gürtel 18-20, 1097, Vienna, Austria
| |
Collapse
|
40
|
Abstract
Chronic traumatic encephalopathy (CTE) is a progressive neurodegenerative disease that occurs in association with repetitive mild traumatic brain injury. It is associated with a variety of clinical symptoms in multiple domains, and there is a distinct pattern of pathological changes. The abnormal tau pathology in CTE occurs uniquely in those regions of the brain that are likely most susceptible to stress concentration during trauma. CTE has been associated with a variety of types of repetitive head trauma, most frequently contact sports. In cases published to date, the mean length of exposure to repetitive head trauma was 15.4 years. The clinical symptoms of the disease began after a mean latency of 14.5 years with a mean age of death of 59.3 years. Most subjects had a reported history of concussions with a mean of 20.3. However, 16 % of published CTE subjects did not have a history of concussion suggesting that subconcussive hits are sufficient to lead to the development of CTE. Overall, the number of years of exposure, not the number of concussions, was significantly associated with worse tau pathology in CTE. This suggests that it is the chronic and repetitive nature of head trauma, irrespective of concussive symptoms, that is the most important driver of disease. CTE and exposure to repetitive head trauma is also associated with a variety of other neurodegenerations, including Alzheimer disease. In fact, amyloid β peptide deposition is altered and accelerated in CTE and is associated with worse disease. Here, we review the current exposure, clinical, and pathological associations of CTE.
Collapse
Affiliation(s)
- Thor D Stein
- VA Boston Healthcare System, Boston, MA, 02130, USA,
| | | | | |
Collapse
|
41
|
Zou J, Wang M, Uchiumi O, Shui Y, Ishigaki Y, Liu X, Tajima N, Akai T, Iizuka H, Kato N. Learning impairment by minimal cortical injury in a mouse model of Alzheimer׳s disease. Brain Res 2016; 1637:56-63. [DOI: 10.1016/j.brainres.2016.02.017] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2015] [Revised: 02/04/2016] [Accepted: 02/07/2016] [Indexed: 11/17/2022]
|
42
|
McKee AC, Cairns NJ, Dickson DW, Folkerth RD, Keene CD, Litvan I, Perl DP, Stein TD, Vonsattel JP, Stewart W, Tripodis Y, Crary JF, Bieniek KF, Dams-O'Connor K, Alvarez VE, Gordon WA. The first NINDS/NIBIB consensus meeting to define neuropathological criteria for the diagnosis of chronic traumatic encephalopathy. Acta Neuropathol 2016; 131:75-86. [PMID: 26667418 PMCID: PMC4698281 DOI: 10.1007/s00401-015-1515-z] [Citation(s) in RCA: 624] [Impact Index Per Article: 69.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2015] [Revised: 11/29/2015] [Accepted: 11/29/2015] [Indexed: 12/14/2022]
Abstract
Chronic traumatic encephalopathy (CTE) is a neurodegeneration characterized by the abnormal accumulation of hyperphosphorylated tau protein within the brain. Like many other neurodegenerative conditions, at present, CTE can only be definitively diagnosed by post-mortem examination of brain tissue. As the first part of a series of consensus panels funded by the NINDS/NIBIB to define the neuropathological criteria for CTE, preliminary neuropathological criteria were used by 7 neuropathologists to blindly evaluate 25 cases of various tauopathies, including CTE, Alzheimer's disease, progressive supranuclear palsy, argyrophilic grain disease, corticobasal degeneration, primary age-related tauopathy, and parkinsonism dementia complex of Guam. The results demonstrated that there was good agreement among the neuropathologists who reviewed the cases (Cohen's kappa, 0.67) and even better agreement between reviewers and the diagnosis of CTE (Cohen's kappa, 0.78). Based on these results, the panel defined 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. The group also defined supportive but non-specific p-tau-immunoreactive features of CTE as: pretangles and NFTs affecting superficial layers (layers II-III) of cerebral cortex; pretangles, NFTs or extracellular tangles in CA2 and pretangles and proximal dendritic swellings in CA4 of the hippocampus; neuronal and astrocytic aggregates in subcortical nuclei; thorn-shaped astrocytes at the glial limitans of the subpial and periventricular regions; and large grain-like and dot-like structures. Supportive non-p-tau pathologies include TDP-43 immunoreactive neuronal cytoplasmic inclusions and dot-like structures in the hippocampus, anteromedial temporal cortex and amygdala. The panel also recommended a minimum blocking and staining scheme for pathological evaluation and made recommendations for future study. This study provides the first step towards the development of validated neuropathological criteria for CTE and will pave the way towards future clinical and mechanistic studies.
Collapse
Affiliation(s)
- Ann C McKee
- Department of Neurology, Boston University School of Medicine, 72 East Concord Street, Boston, MA, 02118, USA.
- Department of Pathology, Boston University School of Medicine, 72 East Concord Street, Boston, MA, 02118, USA.
- Alzheimer's Disease Center, CTE Program, Boston University School of Medicine, 72 East Concord Street, Boston, MA, 02118, USA.
- VA Boston Healthcare System, 150 South Huntington Avenue, Boston, 02130, MA, USA.
- Department of Veteran Affairs Medical Center, 200 Springs Road, Bedford, MA, 01730, USA.
| | - Nigel J Cairns
- Department of Neurology, Washington University School of Medicine, 660 South Euclid Avenue, Saint Louis, MO, 63110, USA
| | - Dennis W Dickson
- Department of Neuroscience, Mayo Clinic, 4500 San Pablo Road, Jacksonville, FL, 32224, USA
| | - Rebecca D Folkerth
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, 75 Francis Street, Boston, MA, 02115, USA
| | - C Dirk Keene
- Department of Pathology, University of Washington School of Medicine, 325 Ninth Avenue, Seattle, WA, 98104, USA
| | - Irene Litvan
- Department of Neurosciences, University of California San Diego School of Medicine, 9500 Gilman Drive, La Jolla, CA, 92093, USA
| | - Daniel P Perl
- Department of Pathology, Center for Neuroscience and Regenerative Medicine, Uniformed Services University of the Health Sciences, 4301 Jones Bridge Road, Bethesda, MD, 20814, USA
| | - Thor D Stein
- Department of Pathology, Boston University School of Medicine, 72 East Concord Street, Boston, MA, 02118, USA
- Alzheimer's Disease Center, CTE Program, Boston University School of Medicine, 72 East Concord Street, Boston, MA, 02118, USA
- VA Boston Healthcare System, 150 South Huntington Avenue, Boston, 02130, MA, USA
- Department of Veteran Affairs Medical Center, 200 Springs Road, Bedford, MA, 01730, USA
| | - Jean-Paul Vonsattel
- Taub Institute for Research on Alzheimer's disease and the Aging Brain, Columbia University Medical Center, 710 West 168th Street, New York, NY, 10032, USA
| | - William Stewart
- Department of Neuropathology, University of Glasgow Institute of Neuroscience and Psychology and Queen Elizabeth University Hospital, 1345 Govan Road, Glasgow, G51 4TF, UK
| | - Yorghos Tripodis
- Alzheimer's Disease Center, CTE Program, Boston University School of Medicine, 72 East Concord Street, Boston, MA, 02118, USA
- Department of Biostatistics, Boston University School of Public Health, 801 Massachusetts Avenue, Boston, MA, 02118, USA
| | - John F Crary
- Department of Pathology, Fishberg Department of Neuroscience, Friedman Brain Institute, Ronald M. Loeb Center for Alzheimer's Disease, Icahn School of Medicine at Mount Sinai School, One Gustave L. Levy Place, New York, NY, 10029, USA
| | - Kevin F Bieniek
- Department of Neuroscience, Mayo Clinic, 4500 San Pablo Road, Jacksonville, FL, 32224, USA
| | - Kristen Dams-O'Connor
- Department of Rehabilitation Medicine, Icahn School of Medicine at Mount Sinai, 3 East 101st Street, New York, NY, 10029, USA
| | - Victor E Alvarez
- Department of Neurology, Boston University School of Medicine, 72 East Concord Street, Boston, MA, 02118, USA
- Department of Pathology, Boston University School of Medicine, 72 East Concord Street, Boston, MA, 02118, USA
- Alzheimer's Disease Center, CTE Program, Boston University School of Medicine, 72 East Concord Street, Boston, MA, 02118, USA
- VA Boston Healthcare System, 150 South Huntington Avenue, Boston, 02130, MA, USA
| | - Wayne A Gordon
- Department of Rehabilitation Medicine, Icahn School of Medicine at Mount Sinai, 3 East 101st Street, New York, NY, 10029, USA
| |
Collapse
|
43
|
Abstract
The recent interest in concussion in sport has resulted in significant media focus about chronic traumatic encephalopathy (CTE), although a direct causative link(s) between concussion and CTE is not established. Typically, sport-related CTE occurs in a retired athlete with or without a history of concussion(s) who presents with a constellation of cognitive, mood, and/or behavioral symptoms and who has postmortem findings of tau deposition within the brain. There are many confounding variables, however, that can account for brain tau deposition, including genetic mutations, drugs, normal aging, environmental factors, postmortem brain processing, and toxins. To understand the roles of such factors in neurodegenerative diseases that may occur in athletes, this article reviews some neurodegenerative diseases that may present with similar findings in nonathletes. The article also reviews pathological changes identified with normal aging, and reviews the pathological findings of CTE in light of all these factors. While many of these athletes have a history of exposure to head impacts as a part of contact sport, there is insufficient evidence to establish causation between sports concussion and CTE. It is likely that many of the cases with neuropathological findings represent the normal aging process, the effects of opiate abuse, or a variant of frontotemporal lobar degeneration. Whether particular genetic causes may place athletes at greater risk of neurodegenerative disease is yet to be determined.
Collapse
Affiliation(s)
- Gavin A Davis
- *Department of Neurosurgery, Cabrini Medical Centre, Malvern, Victoria, Australia; ‡Florey Institute of Neuroscience and Mental Health, Heidelberg, Victoria, Australia; §Department of Pathology, University of Maryland, Baltimore, Maryland
| | | | | |
Collapse
|
44
|
Abstract
American football remains one of the most popular sports for young athletes. The injuries sustained during football, especially those to the head and neck, have been a topic of intense interest recently in both the public media and medical literature. The recognition of these injuries and the potential for long-term sequelae have led some physicians to call for a reduction in the number of contact practices, a postponement of tackling until a certain age, and even a ban on high school football. This statement reviews the literature regarding injuries in football, particularly those of the head and neck, the relationship between tackling and football-related injuries, and the potential effects of limiting or delaying tackling on injury risk.
Collapse
|
45
|
Meehan W, Mannix R, Zafonte R, Pascual-Leone A. Chronic traumatic encephalopathy and athletes. Neurology 2015; 85:1504-11. [PMID: 26253448 DOI: 10.1212/wnl.0000000000001893] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2015] [Accepted: 05/20/2015] [Indexed: 12/13/2022] Open
Abstract
Recent case reports have described athletes previously exposed to repetitive head trauma while participating in contact sports who later in life developed mood disorders, headaches, cognitive difficulties, suicidal ideation, difficulties with speech, and aggressive behavior. Postmortem discoveries show that some of these athletes have pathologic findings that are collectively termed chronic traumatic encephalopathy (CTE). Current hypotheses suggest that concussions or perhaps blows to the head that do not cause the signs and symptoms necessary for making the diagnosis of concussion, so-called subconcussive blows, cause both the clinical and pathologic findings. There are, however, some athletes who participate in contact sports who do not develop the findings ascribed to CTE. Furthermore, there are people who have headaches, mood disorders, cognitive difficulties, suicidal ideation, and other clinical problems who have neither been exposed to repeated head trauma nor possessed the pathologic postmortem findings of those currently diagnosed with CTE. The current lack of prospective data and properly designed case-control studies limits the current understanding of CTE, leading to debate about the causes of the neuropathologic findings and the clinical observations. Given the potential for referral and recall bias in available studies, it remains unclear whether or not the pathologic findings made postmortem cause the presumed neurobehavioral sequela and whether the presumed risk factors, such as sports activity, cerebral concussions, and subconcussive blows, are solely causative of the clinical signs and symptoms. This article discusses the current evidence and the associated limitations.
Collapse
Affiliation(s)
- William Meehan
- From the Micheli Center for Sports Injury Prevention (W.M.), Waltham; the Brain Injury Center (W.M., R.M.), Sports Concussion Clinic, Division of Sports Medicine (W.M.), and Division of Emergency Medicine (W.M., R.M.), Boston Children's Hospital; the Department of Physical Medicine and Rehabilitation, Spaulding Rehabilitation Hospital (R.Z.), Massachusetts General Hospital, Brigham and Women's Hospital; and the Berenson-Allen Center and Division of Interventional Cognitive Neurosciences, Department of Neurology (A.P.-L.), Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA.
| | - Rebekah Mannix
- From the Micheli Center for Sports Injury Prevention (W.M.), Waltham; the Brain Injury Center (W.M., R.M.), Sports Concussion Clinic, Division of Sports Medicine (W.M.), and Division of Emergency Medicine (W.M., R.M.), Boston Children's Hospital; the Department of Physical Medicine and Rehabilitation, Spaulding Rehabilitation Hospital (R.Z.), Massachusetts General Hospital, Brigham and Women's Hospital; and the Berenson-Allen Center and Division of Interventional Cognitive Neurosciences, Department of Neurology (A.P.-L.), Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA
| | - Ross Zafonte
- From the Micheli Center for Sports Injury Prevention (W.M.), Waltham; the Brain Injury Center (W.M., R.M.), Sports Concussion Clinic, Division of Sports Medicine (W.M.), and Division of Emergency Medicine (W.M., R.M.), Boston Children's Hospital; the Department of Physical Medicine and Rehabilitation, Spaulding Rehabilitation Hospital (R.Z.), Massachusetts General Hospital, Brigham and Women's Hospital; and the Berenson-Allen Center and Division of Interventional Cognitive Neurosciences, Department of Neurology (A.P.-L.), Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA
| | - Alvaro Pascual-Leone
- From the Micheli Center for Sports Injury Prevention (W.M.), Waltham; the Brain Injury Center (W.M., R.M.), Sports Concussion Clinic, Division of Sports Medicine (W.M.), and Division of Emergency Medicine (W.M., R.M.), Boston Children's Hospital; the Department of Physical Medicine and Rehabilitation, Spaulding Rehabilitation Hospital (R.Z.), Massachusetts General Hospital, Brigham and Women's Hospital; and the Berenson-Allen Center and Division of Interventional Cognitive Neurosciences, Department of Neurology (A.P.-L.), Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA
| |
Collapse
|
46
|
Abstract
Accumulation of phosphorylated tau (p-tau) is accepted by many as a long-term consequence of repetitive mild neurotrauma based largely on brain findings in boxers (dementia pugilistica) and, more recently, former professional athletes, military service members, and others exposed to repetitive head trauma. The pathogenic construct is also largely accepted and suggests that repetitive head trauma (typically concussions or subconcussive forces) acts on brain parenchyma to produce a deleterious neuroinflammatory cascade, encompassing p-tau templating, transsynaptic neurotoxicity, progressive neurodegenerative disease, and associated clinical features. Some caution before accepting these concepts and assumptions is warranted, however. The association between the history of concussion and findings of p-tau at autopsy is unclear. Concussions and subconcussive head trauma exposure are poorly defined in available cases, and the clinical features reported in chronic traumatic encephalopathy are not at present distinguishable from other disorders. Because control groups are limited, the idea that p-tau drives the disease process via protein templating or some other mechanism is preliminary. Much additional research in chronic traumatic encephalopathy is needed to determine if it has unique neuropathology and clinical features, the extent to which the neuropathologic alterations cause the clinical features, and whether it can be identified accurately in a living person.
Collapse
|
47
|
Iverson GL, Gardner AJ, McCrory P, Zafonte R, Castellani RJ. A critical review of chronic traumatic encephalopathy. Neurosci Biobehav Rev 2015; 56:276-93. [PMID: 26183075 DOI: 10.1016/j.neubiorev.2015.05.008] [Citation(s) in RCA: 77] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2014] [Revised: 04/14/2015] [Accepted: 05/08/2015] [Indexed: 12/14/2022]
Abstract
Chronic traumatic encephalopathy (CTE) has been described in the literature as a neurodegenerative disease with: (i) localized neuronal and glial accumulations of phosphorylated tau (p-tau) involving perivascular areas of the cerebral cortex, sulcal depths, and with a preference for neurons within superficial cortical laminae; (ii) multifocal axonal varicosities and axonal loss involving deep cortex and subcortical white matter; (iii) relative absence of beta-amyloid deposits; (iv) TDP-43 immunoreactive inclusions and neurites; and (v) broad and diverse clinical features. Some of the pathological findings reported in the literature may be encountered with age and other neurodegenerative diseases. However, the focality of the p-tau cortical findings in particular, and the regional distribution, are believed to be unique to CTE. The described clinical features in recent cases are very similar to how depression manifests in middle-aged men and with frontotemporal dementia as the disease progresses. It has not been established that the described tau pathology, especially in small amounts, can cause complex changes in behavior such as depression, substance abuse, suicidality, personality changes, or cognitive impairment. Future studies will help determine the extent to which the neuropathology is causally related to the diverse clinical features.
Collapse
Affiliation(s)
- Grant L Iverson
- Department of Physical Medicine and Rehabilitation, Harvard Medical School, Spaulding Rehabilitation Hospital, MassGeneral Hospital for Children Sports Concussion Program, & Red Sox Foundation and Massachusetts General Hospital Home Base Program, Boston, MA, USA.
| | - Andrew J Gardner
- Hunter New England Local Health District Sports Concussion Program; & Centre for Translational Neuroscience and Mental Health, School of Medicine and Public Health, University of Newcastle, Callaghan, NSW, Australia
| | - 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; Spaulding Rehabilitation Hospital; Brigham and Women's Hospital; & Red Sox Foundation and Massachusetts General Hospital Home Base Program, Boston, MA, USA
| | - Rudy J Castellani
- Division of Neuropathology, University of Maryland School of Medicine, USA
| |
Collapse
|
48
|
Caillet-Boudin ML, Buée L, Sergeant N, Lefebvre B. Regulation of human MAPT gene expression. Mol Neurodegener 2015; 10:28. [PMID: 26170022 PMCID: PMC4499907 DOI: 10.1186/s13024-015-0025-8] [Citation(s) in RCA: 127] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2015] [Accepted: 06/30/2015] [Indexed: 12/12/2022] Open
Abstract
The number of known pathologies involving deregulated Tau expression/metabolism is increasing. Indeed, in addition to tauopathies, which comprise approximately 30 diseases characterized by neuronal aggregation of hyperphosphorylated Tau in brain neurons, this protein has also been associated with various other pathologies such as cancer, inclusion body myositis, and microdeletion/microduplication syndromes, suggesting its possible function in peripheral tissues. In addition to Tau aggregation, Tau deregulation can occur at the expression and/or splicing levels, as has been clearly demonstrated in some of these pathologies. Here, we aim to review current knowledge regarding the regulation of human MAPT gene expression at the DNA and RNA levels to provide a better understanding of its possible deregulation. Several aspects, including repeated motifs, CpG island/methylation, and haplotypes at the DNA level, as well as the key regions involved in mRNA expression and stability and the splicing patterns of different mRNA isoforms at the RNA level, will be discussed.
Collapse
Affiliation(s)
| | - Luc Buée
- Univ. Lille, UMR-S 1172, Inserm, CHU, 59000, Lille, France
| | | | - Bruno Lefebvre
- Univ. Lille, UMR-S 1172, Inserm, CHU, 59000, Lille, France
| |
Collapse
|
49
|
Beta-amyloid deposition in chronic traumatic encephalopathy. Acta Neuropathol 2015; 130:21-34. [PMID: 25943889 DOI: 10.1007/s00401-015-1435-y] [Citation(s) in RCA: 204] [Impact Index Per Article: 20.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2015] [Revised: 04/22/2015] [Accepted: 04/23/2015] [Indexed: 12/14/2022]
Abstract
Chronic traumatic encephalopathy (CTE) is a neurodegenerative disease associated with repetitive mild traumatic brain injury. It is defined pathologically by the abnormal accumulation of tau in a unique pattern that is distinct from other tauopathies, including Alzheimer's disease (AD). Although trauma has been suggested to increase amyloid β peptide (Aβ) levels, the extent of Aβ deposition in CTE has not been thoroughly characterized. We studied a heterogeneous cohort of deceased athletes and military veterans with neuropathologically diagnosed CTE (n = 114, mean age at death = 60) to test the hypothesis that Aβ deposition is altered in CTE and associated with more severe pathology and worse clinical outcomes. We found that Aβ deposition, either as diffuse or neuritic plaques, was present in 52 % of CTE subjects. Moreover, Aβ deposition in CTE occurred at an accelerated rate and with altered dynamics in CTE compared to a normal aging population (OR = 3.8, p < 0.001). We also found a clear pathological and clinical dichotomy between those CTE cases with Aβ plaques and those without. Aβ deposition was significantly associated with the presence of the APOE ε4 allele (p = 0.035), older age at symptom onset (p < 0.001), and older age at death (p < 0.001). In addition, when controlling for age, neuritic plaques were significantly associated with increased CTE tauopathy stage (β = 2.43, p = 0.018), co-morbid Lewy body disease (OR = 5.01, p = 0.009), and dementia (OR = 4.45, p = 0.012). A subset of subjects met the diagnostic criteria for both CTE and AD, and in these subjects both Aβ plaques and total levels of Aβ1-40 were increased at the depths of the cortical sulcus compared to the gyral crests. Overall, these findings suggest that Aβ deposition is altered and accelerated in a cohort of CTE subjects compared to normal aging and that Aβ is associated with both pathological and clinical progression of CTE independent of age.
Collapse
|
50
|
Washington PM, Villapol S, Burns MP. Polypathology and dementia after brain trauma: Does brain injury trigger distinct neurodegenerative diseases, or should they be classified together as traumatic encephalopathy? Exp Neurol 2015; 275 Pt 3:381-388. [PMID: 26091850 DOI: 10.1016/j.expneurol.2015.06.015] [Citation(s) in RCA: 129] [Impact Index Per Article: 12.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2015] [Revised: 06/06/2015] [Accepted: 06/15/2015] [Indexed: 12/14/2022]
Abstract
Neuropathological studies of human traumatic brain injury (TBI) cases have described amyloid plaques acutely after a single severe TBI, and tau pathology after repeat mild TBI (mTBI). This has helped drive the hypothesis that a single moderate to severe TBI increases the risk of developing late-onset Alzheimer's disease (AD), while repeat mTBI increases the risk of developing chronic traumatic encephalopathy (CTE). In this review we critically assess this position-examining epidemiological and case control human studies, neuropathological evidence, and preclinical data. Epidemiological studies emphasize that TBI is associated with the increased risk of developing multiple types of dementia, not just AD-type dementia, and that TBI can also trigger other neurodegenerative conditions such as Parkinson's disease. Further, human post-mortem studies on both single TBI and repeat mTBI can show combinations of amyloid, tau, TDP-43, and Lewy body pathology indicating that the neuropathology of TBI is best described as a 'polypathology'. Preclinical studies confirm that multiple proteins associated with the development of neurodegenerative disease accumulate in the brain after TBI. The chronic sequelae of both single TBI and repeat mTBI share common neuropathological features and clinical symptoms of classically defined neurodegenerative disorders. However, while the spectrum of chronic cognitive and neurobehavioral disorders that occur following repeat mTBI is viewed as the symptoms of CTE, the spectrum of chronic cognitive and neurobehavioral symptoms that occur after a single TBI is considered to represent distinct neurodegenerative diseases such as AD. These data support the suggestion that the multiple manifestations of TBI-induced neurodegenerative disorders be classified together as traumatic encephalopathy or trauma-induced neurodegeneration, regardless of the nature or frequency of the precipitating TBI.
Collapse
Affiliation(s)
- Patricia M Washington
- Department of Pediatrics and Critical Care Medicine, Columbia University Medical Center, New York, NY, USA; Neurotrauma and Repair Laboratory, Department of Biomedical Engineering, Columbia University, New York, NY, USA
| | - Sonia Villapol
- Laboratory for Brain Injury and Dementia, Department of Neuroscience, Georgetown University Medical Center, Washington, DC, USA
| | - Mark P Burns
- Laboratory for Brain Injury and Dementia, Department of Neuroscience, Georgetown University Medical Center, Washington, DC, USA.
| |
Collapse
|