1
|
Nicks R, Shah A, Stathas SA, Kirsch D, Horowitz SM, Saltiel N, Calderazzo SM, Butler MLMD, Cormier KA, Aytan N, Tu-Zahra F, Mathias R, Faheem F, Marcus S, Spurlock E, Fishbein L, Esnault CD, Boden A, Rosen G, Xia W, Daley S, Meng G, Martin BR, Daneshvar DH, Nowinski CJ, Alosco ML, Mez J, Tripodis Y, Huber BR, Alvarez VE, Cherry JD, McKee AC, Stein TD. Neurodegeneration in the cortical sulcus is a feature of chronic traumatic encephalopathy and associated with repetitive head impacts. Acta Neuropathol 2024; 148:79. [PMID: 39643767 PMCID: PMC11624223 DOI: 10.1007/s00401-024-02833-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2024] [Revised: 11/11/2024] [Accepted: 11/11/2024] [Indexed: 12/09/2024]
Abstract
Neurodegeneration is a seminal feature of many neurological disorders. Chronic traumatic encephalopathy (CTE) is caused by repetitive head impacts (RHI) and is characterized by sulcal tau pathology. However, quantitative assessments of regional neurodegeneration in CTE have not been described. In this study, we quantified three key neurodegenerative measures, including cortical thickness, neuronal density, and synaptic proteins, in contact sport athletes (n = 185) and non-athlete controls (n = 52) within the sulcal depth, middle, and gyral crest of the dorsolateral frontal cortex. Cortical thickness and neuronal density were decreased within the sulcus in CTE compared to controls (p's < 0.05). Measurements of synaptic proteins within the gyral crest showed a reduction of α-synuclein with CTE stage (p = 0.002) and variable changes in PSD-95 density. After adjusting for age, multiple linear regression models demonstrated a strong association between the duration of contact sports play and cortical thinning (p = 0.001) and neuronal loss (p = 0.032) within the sulcus. Additional regression models, adjusted for tau pathology, suggest that within the sulcus, the duration of play was associated with neuronal loss predominantly through tau pathology. In contrast, the association of duration of play with cortical thinning was minimally impacted by tau pathology. Overall, CTE is associated with cortical atrophy and a predominant sulcal neurodegeneration. Furthermore, the duration of contact sports play is associated with measures of neurodegeneration that are more severe in the cortical sulcus and may occur through tau-dependent and independent mechanisms.
Collapse
Affiliation(s)
- Raymond Nicks
- Boston University Alzheimer's Disease and CTE Center, Boston Chobanian & Avedisian University School of Medicine, Boston, MA, USA
- VA Boston Healthcare System, Boston, MA, USA
- VA Bedford Healthcare System, Bedford, MA, USA
| | - Arsal Shah
- Boston University Alzheimer's Disease and CTE Center, Boston Chobanian & Avedisian University School of Medicine, Boston, MA, USA
- VA Boston Healthcare System, Boston, MA, USA
| | - Spiro Anthony Stathas
- Boston University Alzheimer's Disease and CTE Center, Boston Chobanian & Avedisian University School of Medicine, Boston, MA, USA
- VA Bedford Healthcare System, Bedford, MA, USA
| | - Daniel Kirsch
- Boston University Alzheimer's Disease and CTE Center, Boston Chobanian & Avedisian University School of Medicine, Boston, MA, USA
- VA Boston Healthcare System, Boston, MA, USA
- Department of Pathology and Laboratory Medicine, Boston University School of Medicine, Boston, MA, USA
| | - Sarah M Horowitz
- Boston University Alzheimer's Disease and CTE Center, Boston Chobanian & Avedisian University School of Medicine, Boston, MA, USA
- VA Boston Healthcare System, Boston, MA, USA
| | - Nicole Saltiel
- Boston University Alzheimer's Disease and CTE Center, Boston Chobanian & Avedisian University School of Medicine, Boston, MA, USA
| | - Samantha M Calderazzo
- Boston University Alzheimer's Disease and CTE Center, Boston Chobanian & Avedisian University School of Medicine, Boston, MA, USA
- VA Boston Healthcare System, Boston, MA, USA
- Department of Pathology and Laboratory Medicine, Boston University School of Medicine, Boston, MA, USA
| | - Morgane L M D Butler
- Boston University Alzheimer's Disease and CTE Center, Boston Chobanian & Avedisian University School of Medicine, Boston, MA, USA
- VA Boston Healthcare System, Boston, MA, USA
- Department of Anatomy & Neurobiology, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
| | - Kerry A Cormier
- Boston University Alzheimer's Disease and CTE Center, Boston Chobanian & Avedisian University School of Medicine, Boston, MA, USA
- VA Bedford Healthcare System, Bedford, MA, USA
| | - Nurgul Aytan
- Boston University Alzheimer's Disease and CTE Center, Boston Chobanian & Avedisian University School of Medicine, Boston, MA, USA
- Department of Neurology, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
| | - Fatima Tu-Zahra
- Boston University Alzheimer's Disease and CTE Center, Boston Chobanian & Avedisian University School of Medicine, Boston, MA, USA
- Department of Biostatistics, Boston University School of Public Health, Boston, MA, USA
| | - Rebecca Mathias
- Boston University Alzheimer's Disease and CTE Center, Boston Chobanian & Avedisian University School of Medicine, Boston, MA, USA
- VA Boston Healthcare System, Boston, MA, USA
| | - Farwa Faheem
- Boston University Alzheimer's Disease and CTE Center, Boston Chobanian & Avedisian University School of Medicine, Boston, MA, USA
| | | | - Elizabeth Spurlock
- Boston University Alzheimer's Disease and CTE Center, Boston Chobanian & Avedisian University School of Medicine, Boston, MA, USA
| | - Lucas Fishbein
- Boston University Alzheimer's Disease and CTE Center, Boston Chobanian & Avedisian University School of Medicine, Boston, MA, USA
| | - Camille D Esnault
- Boston University Alzheimer's Disease and CTE Center, Boston Chobanian & Avedisian University School of Medicine, Boston, MA, USA
| | - Alexandra Boden
- Boston University Alzheimer's Disease and CTE Center, Boston Chobanian & Avedisian University School of Medicine, Boston, MA, USA
| | - Grace Rosen
- VA Boston Healthcare System, Boston, MA, USA
- Department of Neurology, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
| | - Weiming Xia
- Boston University Alzheimer's Disease and CTE Center, Boston Chobanian & Avedisian University School of Medicine, Boston, MA, USA
- VA Bedford Healthcare System, Bedford, MA, USA
- Department of Biological Sciences, Kennedy College of Science, University of Massachusetts, Lowell, MA, USA
| | - Sarah Daley
- Boston University Alzheimer's Disease and CTE Center, Boston Chobanian & Avedisian University School of Medicine, Boston, MA, USA
- VA Bedford Healthcare System, Bedford, MA, USA
| | | | - Brett R Martin
- Boston University Alzheimer's Disease and CTE Center, Boston Chobanian & Avedisian University School of Medicine, Boston, MA, USA
- Department of Biostatistics, Boston University School of Public Health, Boston, MA, USA
| | - Daniel H Daneshvar
- 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, Mass General Brigham-Spaulding Rehabilitation, Charlestown, MA, USA
| | - Christopher J Nowinski
- Boston University Alzheimer's Disease and CTE Center, Boston Chobanian & Avedisian University School of Medicine, Boston, MA, USA
- Concussion Legacy Foundation, Boston, MA, USA
| | - Michael L Alosco
- Boston University Alzheimer's Disease and CTE Center, Boston Chobanian & Avedisian University School of Medicine, Boston, MA, USA
- Department of Neurology, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
| | - Jesse Mez
- Boston University Alzheimer's Disease and CTE Center, Boston Chobanian & Avedisian University School of Medicine, Boston, MA, USA
- Department of Neurology, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
| | - Yorghos Tripodis
- Boston University Alzheimer's Disease and CTE Center, Boston Chobanian & Avedisian University School of Medicine, Boston, MA, USA
- Department of Biostatistics, Boston University School of Public Health, Boston, MA, USA
| | - Bertrand R Huber
- Boston University Alzheimer's Disease and CTE Center, Boston Chobanian & Avedisian University School of Medicine, Boston, MA, USA
- VA Boston Healthcare System, Boston, MA, USA
- Department of Neurology, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
| | - Victor E Alvarez
- Boston University Alzheimer's Disease and CTE Center, Boston Chobanian & Avedisian University School of Medicine, Boston, MA, USA
- VA Boston Healthcare System, Boston, MA, USA
- Department of Neurology, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
- VA Bedford Healthcare System, Bedford, MA, USA
| | - Jonathan D Cherry
- Boston University Alzheimer's Disease and CTE Center, Boston Chobanian & Avedisian University School of Medicine, Boston, MA, USA
- VA Boston Healthcare System, Boston, MA, USA
- Department of Neurology, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
| | - Ann C McKee
- Boston University Alzheimer's Disease and CTE Center, Boston Chobanian & Avedisian University School of Medicine, Boston, MA, USA
- VA Boston Healthcare System, Boston, MA, USA
- Department of Neurology, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
- VA Bedford Healthcare System, Bedford, MA, USA
- Department of Pathology and Laboratory Medicine, Boston University School of Medicine, Boston, MA, USA
| | - Thor D Stein
- Boston University Alzheimer's Disease and CTE Center, Boston Chobanian & Avedisian University School of Medicine, Boston, MA, USA.
- VA Boston Healthcare System, Boston, MA, USA.
- Department of Neurology, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA.
- VA Bedford Healthcare System, Bedford, MA, USA.
- Department of Pathology and Laboratory Medicine, Boston University School of Medicine, Boston, MA, USA.
| |
Collapse
|
2
|
Shanaki Bavarsad M, Spina S, Oehler A, Allen IE, Suemoto CK, Leite REP, Seeley WS, Green A, Jagust W, Rabinovici GD, Grinberg LT. Comprehensive mapping of synaptic vesicle protein 2A (SV2A) in health and neurodegenerative diseases: a comparative analysis with synaptophysin and ground truth for PET-imaging interpretation. Acta Neuropathol 2024; 148:58. [PMID: 39476256 PMCID: PMC11827533 DOI: 10.1007/s00401-024-02816-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2024] [Revised: 09/27/2024] [Accepted: 10/09/2024] [Indexed: 11/07/2024]
Abstract
Synaptic dysfunction and loss are central to neurodegenerative diseases and correlate with cognitive decline. Synaptic Vesicle Protein 2A (SV2A) is a promising PET-imaging target for assessing synaptic density in vivo, but comprehensive mapping in the human brain is needed to validate its biomarker potential. This study used quantitative immunohistochemistry and Western blotting to map SV2A and synaptophysin (SYP) densities across six cortical regions in healthy controls and patients with early-onset Alzheimer's disease (EOAD), late-onset Alzheimer's disease (LOAD), progressive supranuclear palsy (PSP), and frontotemporal lobar degeneration with TDP-43 inclusions (FTLD-GRN). We identified region in SV2A density among controls and observed disease- and region-specific reductions, with the most severe in FTLD-GRN (up to 59.5%) and EOAD. EOAD showed a 49% reduction in the middle frontal gyrus (MFG), while LOAD had over 30% declines in the inferior frontal gyrus (IFG) and hippocampus (CA1). In PSP, smaller but significant reductions were noted in the hippocampal formation, with the inferior temporal gyrus (ITG) relatively unaffected. A strong positive correlation between SV2A and SYP densities confirmed SV2A's reliability as a synaptic integrity marker. This study supports the use of SV2A PET imaging for early diagnosis and monitoring of neurodegenerative diseases, providing essential data for interpreting in vivo PET results. Further research should explore SV2A as a therapeutic target and validate these findings in larger, longitudinal studies.
Collapse
Affiliation(s)
- Mahsa Shanaki Bavarsad
- Memory and Aging Center, Department of Neurology, Weill Institute for Neurosciences, University of California, San Francisco (UCSF), 675 Nelson Rising Lane, Suite 190, San Francisco, CA, 94158, USA
| | - Salvatore Spina
- Memory and Aging Center, Department of Neurology, Weill Institute for Neurosciences, University of California, San Francisco (UCSF), 675 Nelson Rising Lane, Suite 190, San Francisco, CA, 94158, USA
| | - Abby Oehler
- Institute for Neurodegenerative Diseases, University of California, San Francisco (UCSF), San Francisco, USA
| | - Isabel E Allen
- Department of Biostatistics and Epidemiology, University of California, San Francisco (UCSF), San Francisco, USA
| | - Claudia K Suemoto
- Discipline of Geriatrics, University of Sao Paulo Medical School, Sao Paulo, Brazil
| | - Renata E P Leite
- Department of Pathology, Lim22, University of Sao Paulo Medical School, Sao Paulo, Brazil
| | - William S Seeley
- Memory and Aging Center, Department of Neurology, Weill Institute for Neurosciences, University of California, San Francisco (UCSF), 675 Nelson Rising Lane, Suite 190, San Francisco, CA, 94158, USA
| | - Ari Green
- Department of Neurology, Weill Institute for Neurosciences, University of California, San Francisco (UCSF), San Francisco, USA
| | - William Jagust
- Department of Neuroscience, University of California Berkeley, Berkeley, USA
| | - Gil D Rabinovici
- Memory and Aging Center, Department of Neurology, Weill Institute for Neurosciences, University of California, San Francisco (UCSF), 675 Nelson Rising Lane, Suite 190, San Francisco, CA, 94158, USA
| | - Lea T Grinberg
- Memory and Aging Center, Department of Neurology, Weill Institute for Neurosciences, University of California, San Francisco (UCSF), 675 Nelson Rising Lane, Suite 190, San Francisco, CA, 94158, USA.
- Department of Pathology, Lim22, University of Sao Paulo Medical School, Sao Paulo, Brazil.
| |
Collapse
|
3
|
Bai Q, Shao E, Ma D, Jiao B, Scheetz SD, Hartnett-Scott KA, Ilin VA, Aizenman E, Kofler J, Burton EA. A human Tau expressing zebrafish model of progressive supranuclear palsy identifies Brd4 as a regulator of microglial synaptic elimination. Nat Commun 2024; 15:8195. [PMID: 39294122 PMCID: PMC11410960 DOI: 10.1038/s41467-024-52173-0] [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: 07/11/2024] [Accepted: 08/28/2024] [Indexed: 09/20/2024] Open
Abstract
Progressive supranuclear palsy (PSP) is an incurable neurodegenerative disease characterized by 4-repeat (0N/4R)-Tau protein accumulation in CNS neurons. We generated transgenic zebrafish expressing human 0N/4R-Tau to investigate PSP pathophysiology. Tau zebrafish replicated multiple features of PSP, including: decreased survival; hypokinesia; impaired optokinetic responses; neurodegeneration; neuroinflammation; synapse loss; and Tau hyperphosphorylation, misfolding, mislocalization, insolubility, truncation, and oligomerization. Using automated assays, we screened 147 small molecules for activity in rescuing neurological deficits in Tau zebrafish. (+)JQ1, a bromodomain inhibitor, improved hypokinesia, survival, microgliosis, and brain synapse elimination. A heterozygous brd4+/- mutant reducing expression of the bromodomain protein Brd4 similarly rescued these phenotypes. Microglial phagocytosis of synaptic material was decreased by (+)JQ1 in both Tau zebrafish and rat primary cortical cultures. Microglia in human PSP brains expressed Brd4. Our findings implicate Brd4 as a regulator of microglial synaptic elimination in tauopathy and provide an unbiased approach for identifying mechanisms and therapeutic targets in PSP.
Collapse
Affiliation(s)
- Qing Bai
- Department of Neurology, University of Pittsburgh, Pittsburgh, PA, 15213, USA
- Pittsburgh Institute for Neurodegenerative Diseases, University of Pittsburgh, Pittsburgh, PA, 15213, USA
| | - Enhua Shao
- Department of Neurology, University of Pittsburgh, Pittsburgh, PA, 15213, USA
- Pittsburgh Institute for Neurodegenerative Diseases, University of Pittsburgh, Pittsburgh, PA, 15213, USA
- Tsinghua University School of Medicine, Beijing, China
| | - Denglei Ma
- Department of Neurology, University of Pittsburgh, Pittsburgh, PA, 15213, USA
- Pittsburgh Institute for Neurodegenerative Diseases, University of Pittsburgh, Pittsburgh, PA, 15213, USA
| | - Binxuan Jiao
- Department of Neurology, University of Pittsburgh, Pittsburgh, PA, 15213, USA
- Pittsburgh Institute for Neurodegenerative Diseases, University of Pittsburgh, Pittsburgh, PA, 15213, USA
- Tsinghua University School of Medicine, Beijing, China
| | - Seth D Scheetz
- Department of Neurology, University of Pittsburgh, Pittsburgh, PA, 15213, USA
- Pittsburgh Institute for Neurodegenerative Diseases, University of Pittsburgh, Pittsburgh, PA, 15213, USA
| | - Karen A Hartnett-Scott
- Pittsburgh Institute for Neurodegenerative Diseases, University of Pittsburgh, Pittsburgh, PA, 15213, USA
- Department of Neurobiology, University of Pittsburgh, Pittsburgh, PA, 15213, USA
| | - Vladimir A Ilin
- Department of Neurology, University of Pittsburgh, Pittsburgh, PA, 15213, USA
- Pittsburgh Institute for Neurodegenerative Diseases, University of Pittsburgh, Pittsburgh, PA, 15213, USA
| | - Elias Aizenman
- Pittsburgh Institute for Neurodegenerative Diseases, University of Pittsburgh, Pittsburgh, PA, 15213, USA
- Department of Neurobiology, University of Pittsburgh, Pittsburgh, PA, 15213, USA
| | - Julia Kofler
- Department of Pathology, University of Pittsburgh, Pittsburgh, PA, 15213, USA
- Alzheimer's Disease Research Center, University of Pittsburgh, Pittsburgh, PA, 15213, USA
| | - Edward A Burton
- Department of Neurology, University of Pittsburgh, Pittsburgh, PA, 15213, USA.
- Pittsburgh Institute for Neurodegenerative Diseases, University of Pittsburgh, Pittsburgh, PA, 15213, USA.
- Geriatrics Research, Education and Clinical Center, Pittsburgh VA Healthcare System, Pittsburgh, PA, 15240, USA.
| |
Collapse
|
4
|
Perry A, Hughes LE, Adams NE, Naessens M, Kloosterman NA, Rouse MA, Murley AG, Street D, Jones PS, Rowe JB. Frontotemporal lobar degeneration changes neuronal beta-frequency dynamics during the mismatch negativity response. Neuroimage Clin 2024; 44:103671. [PMID: 39305652 PMCID: PMC11439566 DOI: 10.1016/j.nicl.2024.103671] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2024] [Revised: 09/07/2024] [Accepted: 09/07/2024] [Indexed: 10/04/2024]
Abstract
The consequences of frontotemporal lobar degeneration include changes in prefrontal cortical neurophysiology, with abnormalities of neural dynamics reported in the beta frequency range (14-30 Hz) that correlate with functional severity. We examined beta dynamics in two clinical syndromes associated with frontotemporal lobar degeneration: the behavioral variant of frontotemporal dementia (bvFTD) and progressive supranuclear palsy (PSP). Whilst these two syndromes are partially convergent in cognitive effects, they differ in disease mechanisms such as molecular pathologies and prefrontal atrophy. Whether bvFTD and PSP also differ in neurophysiology remains to be fully investigated. We compared magnetoencephalography from 20 controls, 23 people with bvFTD and 21 people with PSP (Richardson's syndrome) during an auditory roving oddball paradigm. We measured changes in low and high total beta power responses (14-22 and 22-30 Hz respectively) over frontotemporal cortex in the period of the mismatch negativity response (100-250 ms post-stimulus). In controls, we found increased 14-22 Hz beta power following unexpected sensory events (i.e. increased deviant versus standard response), from right prefrontal cortex. Relative to controls, PSP reversed the mismatch response in this time-frequency window, reflecting reduced responses to the deviant stimuli (relative to standard stimuli). Abnormal beta at baseline in PSP could account for the reduced task-modulation of beta. Across bvFTD and PSP groups, the beta response to deviant stimuli (relative to standard stimuli) correlated with clinical severity, but not with atrophy of the prefrontal source region. These findings confirm the proposed importance of higher-order cortical regions, and their beta-power generators, in sensory change detection and context-updating during oddball paradigms. The physiological effects are proposed to result from changes in synaptic density, cortical neurotransmitters and subcortical connections, rather than merely atrophy. Beta-power changes may assist clinical stratification and provide intermediate outcomes for experimental medicine studies of novel therapeutic strategies.
Collapse
Affiliation(s)
- Alistair Perry
- MRC Cognition and Brain Sciences Unit, University of Cambridge, Cambridge, United Kingdom; Department of Clinical Neurosciences and Cambridge University Hospitals NHS Trust, University of Cambridge, United Kingdom
| | - Laura E Hughes
- MRC Cognition and Brain Sciences Unit, University of Cambridge, Cambridge, United Kingdom; Department of Clinical Neurosciences and Cambridge University Hospitals NHS Trust, University of Cambridge, United Kingdom
| | - Natalie E Adams
- Department of Clinical Neurosciences and Cambridge University Hospitals NHS Trust, University of Cambridge, United Kingdom
| | - Michelle Naessens
- MRC Cognition and Brain Sciences Unit, University of Cambridge, Cambridge, United Kingdom; Department of Clinical Neurosciences and Cambridge University Hospitals NHS Trust, University of Cambridge, United Kingdom
| | - Niels A Kloosterman
- Institut für Psychologie I, Universität zu Lübeck, Germany; Max Planck Institute for Human Development, Berlin, Germany
| | - Matthew A Rouse
- MRC Cognition and Brain Sciences Unit, University of Cambridge, Cambridge, United Kingdom
| | - Alexander G Murley
- Department of Clinical Neurosciences and Cambridge University Hospitals NHS Trust, University of Cambridge, United Kingdom
| | - Duncan Street
- Department of Clinical Neurosciences and Cambridge University Hospitals NHS Trust, University of Cambridge, United Kingdom
| | - P Simon Jones
- Department of Clinical Neurosciences and Cambridge University Hospitals NHS Trust, University of Cambridge, United Kingdom
| | - James B Rowe
- MRC Cognition and Brain Sciences Unit, University of Cambridge, Cambridge, United Kingdom; Department of Clinical Neurosciences and Cambridge University Hospitals NHS Trust, University of Cambridge, United Kingdom.
| |
Collapse
|
5
|
Bavarsad MS, Grinberg LT. SV2A PET imaging in human neurodegenerative diseases. Front Aging Neurosci 2024; 16:1380561. [PMID: 38699560 PMCID: PMC11064927 DOI: 10.3389/fnagi.2024.1380561] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2024] [Accepted: 03/20/2024] [Indexed: 05/05/2024] Open
Abstract
This manuscript presents a thorough review of synaptic vesicle glycoprotein 2A (SV2A) as a biomarker for synaptic integrity using Positron Emission Tomography (PET) in neurodegenerative diseases. Synaptic pathology, characterized by synaptic loss, has been linked to various brain diseases. Therefore, there is a need for a minimally invasive approach to measuring synaptic density in living human patients. Several radiotracers targeting synaptic vesicle protein 2A (SV2A) have been created and effectively adapted for use in human subjects through PET scans. SV2A is an integral glycoprotein found in the membranes of synaptic vesicles in all synaptic terminals and is widely distributed throughout the brain. The review delves into the development of SV2A-specific PET radiotracers, highlighting their advancements and limitations in neurodegenerative diseases. Among these tracers, 11C-UCB-J is the most used so far. We summarize and discuss an increasing body of research that compares measurements of synaptic density using SV2A PET with other established indicators of neurodegenerative diseases, including cognitive performance and radiological findings, thus providing a comprehensive analysis of SV2A's effectiveness and reliability as a diagnostic tool in contrast to traditional markers. Although the literature overall suggests the promise of SV2A as a diagnostic and therapeutic monitoring tool, uncertainties persist regarding the superiority of SV2A as a biomarker compared to other available markers. The review also underscores the paucity of studies characterizing SV2A distribution and loss in human brain tissue from patients with neurodegenerative diseases, emphasizing the need to generate quantitative neuropathological maps of SV2A density in cases with neurodegenerative diseases to fully harness the potential of SV2A PET imaging in clinical settings. We conclude by outlining future research directions, stressing the importance of integrating SV2A PET imaging with other biomarkers and clinical assessments and the need for longitudinal studies to track SV2A changes throughout neurodegenerative disease progression, which could lead to breakthroughs in early diagnosis and the evaluation of new treatments.
Collapse
Affiliation(s)
| | - Lea T. Grinberg
- Memory and Aging Center, Weill Institute for Neurosciences, University of California, San Francisco (UCSF), San Francisco, CA, United States
| |
Collapse
|
6
|
Martin SL, Uribe C, Strafella AP. PET imaging of synaptic density in Parkinsonian disorders. J Neurosci Res 2024; 102:e25253. [PMID: 37814917 DOI: 10.1002/jnr.25253] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2023] [Revised: 08/31/2023] [Accepted: 09/21/2023] [Indexed: 10/11/2023]
Abstract
Synaptic dysfunction and altered synaptic pruning are present in people with Parkinsonian disorders. Dopamine loss and alpha-synuclein accumulation, two hallmarks of Parkinson's disease (PD) pathology, contribute to synaptic dysfunction and reduced synaptic density in PD. Atypical Parkinsonian disorders are likely to have unique spatiotemporal patterns of synaptic density, differentiating them from PD. Therefore, quantification of synaptic density has the potential to support diagnoses, monitor disease progression, and treatment efficacy. Novel radiotracers for positron emission tomography which target the presynaptic vesicle protein SV2A have been developed to quantify presynaptic density. The radiotracers have successfully investigated synaptic density in preclinical models of PD and people with Parkinsonian disorders. Therefore, this review will summarize the preclinical and clinical utilization of SV2A radiotracers in people with Parkinsonian disorders. We will evaluate how SV2A abundance is associated with other imaging modalities and the considerations for interpreting SV2A in Parkinsonian pathology.
Collapse
Affiliation(s)
- Sarah L Martin
- Brain Health Imaging Centre, Centre for Addiction and Mental Health, Toronto, Ontario, Canada
| | - Carme Uribe
- Brain Health Imaging Centre, Centre for Addiction and Mental Health, Toronto, Ontario, Canada
- Unitat de Psicologia Medica, Departament de Medicina, Institute of Neuroscience, Universitat de Barcelona, Barcelona, Spain
| | - Antonio P Strafella
- Brain Health Imaging Centre, Centre for Addiction and Mental Health, Toronto, Ontario, Canada
- Edmond J. Safra Parkinson Disease Program, Neurology Division, Toronto Western Hospital & Krembil Brain Institute, University Health Network, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
7
|
Whiteside DJ, Holland N, Tsvetanov KA, Mak E, Malpetti M, Savulich G, Jones PS, Naessens M, Rouse MA, Fryer TD, Hong YT, Aigbirhio FI, Mulroy E, Bhatia KP, Rittman T, O'Brien JT, Rowe JB. Synaptic density affects clinical severity via network dysfunction in syndromes associated with frontotemporal lobar degeneration. Nat Commun 2023; 14:8458. [PMID: 38114493 PMCID: PMC10730886 DOI: 10.1038/s41467-023-44307-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2023] [Accepted: 12/07/2023] [Indexed: 12/21/2023] Open
Abstract
There is extensive synaptic loss from frontotemporal lobar degeneration, in preclinical models and human in vivo and post mortem studies. Understanding the consequences of synaptic loss for network function is important to support translational models and guide future therapeutic strategies. To examine this relationship, we recruited 55 participants with syndromes associated with frontotemporal lobar degeneration and 24 healthy controls. We measured synaptic density with positron emission tomography using the radioligand [11C]UCB-J, which binds to the presynaptic vesicle glycoprotein SV2A, neurite dispersion with diffusion magnetic resonance imaging, and network function with task-free magnetic resonance imaging functional connectivity. Synaptic density and neurite dispersion in patients was associated with reduced connectivity beyond atrophy. Functional connectivity moderated the relationship between synaptic density and clinical severity. Our findings confirm the importance of synaptic loss in frontotemporal lobar degeneration syndromes, and the resulting effect on behaviour as a function of abnormal connectivity.
Collapse
Affiliation(s)
- David J Whiteside
- Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK.
- Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK.
| | - Negin Holland
- Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK
- Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK
| | - Kamen A Tsvetanov
- Medical Research Council Cognition and Brain Sciences Unit, University of Cambridge, Cambridge, UK
| | - Elijah Mak
- Department of Psychiatry, University of Cambridge, Cambridge, UK
| | - Maura Malpetti
- Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK
- Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK
| | - George Savulich
- Department of Psychiatry, University of Cambridge, Cambridge, UK
| | - P Simon Jones
- Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK
| | - Michelle Naessens
- Medical Research Council Cognition and Brain Sciences Unit, University of Cambridge, Cambridge, UK
| | - Matthew A Rouse
- Medical Research Council Cognition and Brain Sciences Unit, University of Cambridge, Cambridge, UK
| | - Tim D Fryer
- Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK
- Wolfson Brain Imaging Centre, University of Cambridge, Cambridge, UK
| | - Young T Hong
- Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK
- Wolfson Brain Imaging Centre, University of Cambridge, Cambridge, UK
| | - Franklin I Aigbirhio
- Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK
- Wolfson Brain Imaging Centre, University of Cambridge, Cambridge, UK
| | - Eoin Mulroy
- UCL Queen Square Institute of Neurology, University College London, London, WC1N 3BG, UK
| | - Kailash P Bhatia
- UCL Queen Square Institute of Neurology, University College London, London, WC1N 3BG, UK
| | - Timothy Rittman
- Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK
- Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK
| | - John T O'Brien
- Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK
- Department of Psychiatry, University of Cambridge, Cambridge, UK
| | - James B Rowe
- Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK
- Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK
- Medical Research Council Cognition and Brain Sciences Unit, University of Cambridge, Cambridge, UK
| |
Collapse
|
8
|
Strobel J, Müller HP, Ludolph AC, Beer AJ, Sollmann N, Kassubek J. New Perspectives in Radiological and Radiopharmaceutical Hybrid Imaging in Progressive Supranuclear Palsy: A Systematic Review. Cells 2023; 12:2776. [PMID: 38132096 PMCID: PMC10742083 DOI: 10.3390/cells12242776] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2023] [Revised: 11/28/2023] [Accepted: 12/04/2023] [Indexed: 12/23/2023] Open
Abstract
Progressive supranuclear palsy (PSP) is a neurodegenerative disease characterized by four-repeat tau deposition in various cell types and anatomical regions, and can manifest as several clinical phenotypes, including the most common phenotype, Richardson's syndrome. The limited availability of biomarkers for PSP relates to the overlap of clinical features with other neurodegenerative disorders, but identification of a growing number of biomarkers from imaging is underway. One way to increase the reliability of imaging biomarkers is to combine different modalities for multimodal imaging. This review aimed to provide an overview of the current state of PSP hybrid imaging by combinations of positron emission tomography (PET) and magnetic resonance imaging (MRI). Specifically, combined PET and MRI studies in PSP highlight the potential of [18F]AV-1451 to detect tau, but also the challenge in differentiating PSP from other neurodegenerative diseases. Studies over the last years showed a reduced synaptic density in [11C]UCB-J PET, linked [11C]PK11195 and [18F]AV-1451 markers to disease progression, and suggested the potential role of [18F]RO948 PET for identifying tau pathology in subcortical regions. The integration of quantitative global and regional gray matter analysis by MRI may further guide the assessment of reduced cortical thickness or volume alterations, and diffusion MRI could provide insight into microstructural changes and structural connectivity in PSP. Challenges in radiopharmaceutical biomarkers and hybrid imaging require further research targeting markers for comprehensive PSP diagnosis.
Collapse
Affiliation(s)
- Joachim Strobel
- Department of Nuclear Medicine, University Hospital Ulm, 89081 Ulm, Germany;
| | - Hans-Peter Müller
- Department of Neurology, University Hospital Ulm, 89081 Ulm, Germany; (H.-P.M.); (A.C.L.); (J.K.)
| | - Albert C. Ludolph
- Department of Neurology, University Hospital Ulm, 89081 Ulm, Germany; (H.-P.M.); (A.C.L.); (J.K.)
- German Center for Neurodegenerative Diseases (DZNE), Ulm University, 89081 Ulm, Germany
| | - Ambros J. Beer
- Department of Nuclear Medicine, University Hospital Ulm, 89081 Ulm, Germany;
| | - Nico Sollmann
- Department of Diagnostic and Interventional Radiology, University Hospital Ulm, 89081 Ulm, Germany;
- Department of Diagnostic and Interventional Neuroradiology, School of Medicine, Klinikum rechts der Isar, Technical University of Munich, 81675 Munich, Germany
- TUM-Neuroimaging Center, Klinikum rechts der Isar, Technical University of Munich, 81675 Munich, Germany
| | - Jan Kassubek
- Department of Neurology, University Hospital Ulm, 89081 Ulm, Germany; (H.-P.M.); (A.C.L.); (J.K.)
- German Center for Neurodegenerative Diseases (DZNE), Ulm University, 89081 Ulm, Germany
| |
Collapse
|
9
|
Vogler L, Ballweg A, Bohr B, Briel N, Wind K, Antons M, Kunze LH, Gnörich J, Lindner S, Gildehaus FJ, Baumann K, Bartenstein P, Boening G, Ziegler SI, Levin J, Zwergal A, Höglinger GU, Herms J, Brendel M. Assessment of synaptic loss in mouse models of β-amyloid and tau pathology using [ 18F]UCB-H PET imaging. Neuroimage Clin 2023; 39:103484. [PMID: 37541098 PMCID: PMC10407951 DOI: 10.1016/j.nicl.2023.103484] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2023] [Revised: 07/24/2023] [Accepted: 07/25/2023] [Indexed: 08/06/2023]
Abstract
OBJECTIVE In preclinical research, the use of [18F]Fluorodesoxyglucose (FDG) as a biomarker for neurodegeneration may induce bias due to enhanced glucose uptake by immune cells. In this study, we sought to investigate synaptic vesicle glycoprotein 2A (SV2A) PET with [18F]UCB-H as an alternative preclinical biomarker for neurodegenerative processes in two mouse models representing the pathological hallmarks of Alzheimer's disease (AD). METHODS A total of 29 PS2APP, 20 P301S and 12 wild-type mice aged 4.4 to 19.8 months received a dynamic [18F]UCB-H SV2A-PET scan (14.7 ± 1.5 MBq) 0-60 min post injection. Quantification of tracer uptake in cortical, cerebellar and brainstem target regions was implemented by calculating relative volumes of distribution (VT) from an image-derived-input-function (IDIF). [18F]UCB-H binding was compared across all target regions between transgenic and wild-type mice. Additional static scans were performed in a subset of mice to compare [18F]FDG and [18F]GE180 (18 kDa translocator protein tracer as a surrogate for microglial activation) standardized uptake values (SUV) with [18F]UCB-H binding at different ages. Following the final scan, a subset of mouse brains was immunohistochemically stained with synaptic markers for gold standard validation of the PET results. RESULTS [18F]UCB-H binding in all target regions was significantly reduced in 8-months old P301S transgenic mice when compared to wild-type controls (temporal lobe: p = 0.014; cerebellum: p = 0.0018; brainstem: p = 0.0014). Significantly lower SV2A tracer uptake was also observed in 13-months (temporal lobe: p = 0.0080; cerebellum: p = 0.006) and 19-months old (temporal lobe: p = 0.0042; cerebellum: p = 0.011) PS2APP transgenic versus wild-type mice, whereas the brainstem revealed no significantly altered [18F]UCB-H binding. Immunohistochemical analyses of post-mortem mouse brain tissue confirmed the SV2A PET findings. Correlational analyses of [18F]UCB-H and [18F]FDG using Pearson's correlation coefficient revealed a significant negative association in the PS2APP mouse model (R = -0.26, p = 0.018). Exploratory analyses further stressed microglial activation as a potential reason for this inverse relationship, since [18F]FDG and [18F]GE180 quantification were positively correlated in this cohort (R = 0.36, p = 0.0076). CONCLUSION [18F]UCB-H reliably depicts progressive synaptic loss in PS2APP and P301S transgenic mice, potentially qualifying as a more reliable alternative to [18F]FDG as a biomarker for assessment of neurodegeneration in preclinical research.
Collapse
Affiliation(s)
- Letizia Vogler
- Department of Nuclear Medicine, University Hospital of Munich, Ludwig-Maximilians-University (LMU) Munich, Munich, Germany
| | - Anna Ballweg
- Department of Nuclear Medicine, University Hospital of Munich, Ludwig-Maximilians-University (LMU) Munich, Munich, Germany
| | - Bernd Bohr
- Department of Nuclear Medicine, University Hospital of Munich, Ludwig-Maximilians-University (LMU) Munich, Munich, Germany
| | - Nils Briel
- Center for Neuropathology, LMU Munich, Munich, Germany
| | - Karin Wind
- Department of Nuclear Medicine, University Hospital of Munich, Ludwig-Maximilians-University (LMU) Munich, Munich, Germany
| | - Melissa Antons
- Department of Nuclear Medicine, University Hospital of Munich, Ludwig-Maximilians-University (LMU) Munich, Munich, Germany
| | - Lea H Kunze
- Department of Nuclear Medicine, University Hospital of Munich, Ludwig-Maximilians-University (LMU) Munich, Munich, Germany
| | - Johannes Gnörich
- Department of Nuclear Medicine, University Hospital of Munich, Ludwig-Maximilians-University (LMU) Munich, Munich, Germany
| | - Simon Lindner
- Department of Nuclear Medicine, University Hospital of Munich, Ludwig-Maximilians-University (LMU) Munich, Munich, Germany
| | - Franz-Josef Gildehaus
- Department of Nuclear Medicine, University Hospital of Munich, Ludwig-Maximilians-University (LMU) Munich, Munich, Germany
| | - Karlheinz Baumann
- Roche Pharma Research and Early Development, Neuroscience Discovery, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd., Basel, Switzerland
| | - Peter Bartenstein
- Department of Nuclear Medicine, University Hospital of Munich, Ludwig-Maximilians-University (LMU) Munich, Munich, Germany
| | - Guido Boening
- Department of Nuclear Medicine, University Hospital of Munich, Ludwig-Maximilians-University (LMU) Munich, Munich, Germany
| | - Sibylle I Ziegler
- Department of Nuclear Medicine, University Hospital of Munich, Ludwig-Maximilians-University (LMU) Munich, Munich, Germany
| | - Johannes Levin
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany; Munich Cluster for Systems Neurology (SyNergy), Munich, Germany; Department of Neurology, University Hospital of Munich, LMU Munich, Munich, Germany
| | - Andreas Zwergal
- Department of Neurology, University Hospital of Munich, LMU Munich, Munich, Germany; German Center for Vertigo and Balance Disorders (DSGZ), University Hospital of Munich, LMU Munich, Munich, Germany
| | - Günter U Höglinger
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany; Department of Neurology, University Hospital of Munich, LMU Munich, Munich, Germany
| | - Jochen Herms
- Center for Neuropathology, LMU Munich, Munich, Germany
| | - Matthias Brendel
- Department of Nuclear Medicine, University Hospital of Munich, Ludwig-Maximilians-University (LMU) Munich, Munich, Germany; German Center for Neurodegenerative Diseases (DZNE), Munich, Germany; Munich Cluster for Systems Neurology (SyNergy), Munich, Germany.
| |
Collapse
|
10
|
Holland N, Jones PS, Savulich G, Naessens M, Malpetti M, Whiteside DJ, Street D, Swann P, Hong YT, Fryer TD, Rittman T, Mulroy E, Aigbirhio FI, Bhatia KP, O'Brien JT, Rowe JB. Longitudinal Synaptic Loss in Primary Tauopathies: An In Vivo [ 11 C]UCB-J Positron Emission Tomography Study. Mov Disord 2023; 38:1316-1326. [PMID: 37171832 PMCID: PMC10947001 DOI: 10.1002/mds.29421] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2023] [Revised: 03/22/2023] [Accepted: 04/10/2023] [Indexed: 05/13/2023] Open
Abstract
BACKGROUND Synaptic loss is characteristic of many neurodegenerative diseases; it occurs early and is strongly related to functional deficits. OBJECTIVE In this longitudinal observational study, we determine the rate at which synaptic density is reduced in the primary tauopathies of progressive supranuclear palsy (PSP) and corticobasal degeneration (CBD), and we test the relationship with disease progression. METHODS Our cross-sectional cohort included 32 participants with probable PSP and 16 with probable CBD (all amyloid-negative corticobasal syndrome), recruited from tertiary care centers in the United Kingdom, and 33 sex- and age-matched healthy control subjects. Synaptic density was estimated by positron emission tomography imaging with the radioligand [11 C]UCB-J that binds synaptic vesicle 2A. Clinical severity and cognition were assessed by the PSP Rating Scale and the Addenbrooke's cognitive examination. Regional [11 C]UCB-J nondisplaceable binding potential was estimated in Hammersmith Atlas regions of interest. Twenty-two participants with PSP/CBD had a follow-up [11 C]UCB-J positron emission tomography scan after 1 year. We calculated the annualized change in [11 C]UCB-J nondisplaceable binding potential and correlated this with the change in clinical severity. RESULTS We found significant annual synaptic loss within the frontal lobe (-3.5%, P = 0.03) and the right caudate (-3.9%, P = 0.046). The degree of longitudinal synaptic loss within the frontal lobe correlated with the rate of change in the PSP Rating Scale (R = 0.47, P = 0.03) and cognition (Addenbrooke's Cognitive Examination-Revised, R = -0.62, P = 0.003). CONCLUSIONS We provide in vivo evidence for rapid progressive synaptic loss, correlating with clinical progression in primary tauopathies. Synaptic loss may be an important therapeutic target and outcome variable for early-phase clinical trials of disease-modifying treatments. © 2023 The Authors. Movement Disorders published by Wiley Periodicals LLC on behalf of International Parkinson and Movement Disorder Society.
Collapse
Affiliation(s)
- Negin Holland
- Department of Clinical NeurosciencesUniversity of Cambridge, Cambridge Biomedical CampusCambridgeUnited Kingdom
- Cambridge University Hospitals NHS Foundation TrustCambridgeUnited Kingdom
| | - P. Simon Jones
- Department of Clinical NeurosciencesUniversity of Cambridge, Cambridge Biomedical CampusCambridgeUnited Kingdom
| | - George Savulich
- Department of PsychiatryUniversity of Cambridge, School of Clinical Medicine, Cambridge Biomedical CampusCambridgeUnited Kingdom
| | - Michelle Naessens
- Department of Clinical NeurosciencesUniversity of Cambridge, Cambridge Biomedical CampusCambridgeUnited Kingdom
| | - Maura Malpetti
- Department of Clinical NeurosciencesUniversity of Cambridge, Cambridge Biomedical CampusCambridgeUnited Kingdom
| | - David J. Whiteside
- Department of Clinical NeurosciencesUniversity of Cambridge, Cambridge Biomedical CampusCambridgeUnited Kingdom
| | - Duncan Street
- Department of Clinical NeurosciencesUniversity of Cambridge, Cambridge Biomedical CampusCambridgeUnited Kingdom
| | - Peter Swann
- Cambridge University Hospitals NHS Foundation TrustCambridgeUnited Kingdom
- Department of PsychiatryUniversity of Cambridge, School of Clinical Medicine, Cambridge Biomedical CampusCambridgeUnited Kingdom
| | - Young T. Hong
- Department of Clinical NeurosciencesUniversity of Cambridge, Cambridge Biomedical CampusCambridgeUnited Kingdom
- Wolfson Brain Imaging CentreUniversity of CambridgeCambridgeUnited Kingdom
| | - Tim D. Fryer
- Department of Clinical NeurosciencesUniversity of Cambridge, Cambridge Biomedical CampusCambridgeUnited Kingdom
- Wolfson Brain Imaging CentreUniversity of CambridgeCambridgeUnited Kingdom
| | - Timothy Rittman
- Department of Clinical NeurosciencesUniversity of Cambridge, Cambridge Biomedical CampusCambridgeUnited Kingdom
| | - Eoin Mulroy
- Department of Clinical and Movement NeurosciencesUCL Queen Square Institute of NeurologyLondonUnited Kingdom
| | - Franklin I. Aigbirhio
- Department of Clinical NeurosciencesUniversity of Cambridge, Cambridge Biomedical CampusCambridgeUnited Kingdom
| | - Kailash P. Bhatia
- Department of Clinical and Movement NeurosciencesUCL Queen Square Institute of NeurologyLondonUnited Kingdom
| | - John T. O'Brien
- Cambridge University Hospitals NHS Foundation TrustCambridgeUnited Kingdom
- Department of PsychiatryUniversity of Cambridge, School of Clinical Medicine, Cambridge Biomedical CampusCambridgeUnited Kingdom
| | - James B. Rowe
- Department of Clinical NeurosciencesUniversity of Cambridge, Cambridge Biomedical CampusCambridgeUnited Kingdom
- Cambridge University Hospitals NHS Foundation TrustCambridgeUnited Kingdom
- Medical Research Council Cognition and Brain Sciences UnitUniversity of CambridgeCambridgeUnited Kingdom
| |
Collapse
|
11
|
Sakuwa M, Adachi T, Suzuki Y, Takigawa H, Hanajima R. Neuropathological analysis of cognitive impairment in progressive supranuclear palsy. J Neurol Sci 2023; 451:120718. [PMID: 37385026 DOI: 10.1016/j.jns.2023.120718] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2023] [Revised: 05/23/2023] [Accepted: 06/17/2023] [Indexed: 07/01/2023]
Abstract
BACKGROUND Cognitive impairment is an important symptom in progressive supranuclear palsy (PSP), but the pathological changes underlying the cognitive impairment are unclear. This study aimed to elucidate relationships between the severity of cognitive impairment and PSP-related pathology. METHODS We investigated the clinicopathological characteristics of 10 autopsy cases of PSP, including neuronal loss/gliosis and the burden of PSP-related tau pathology by using a semiquantitative score in 17 brain regions. Other concurrent pathologies such as Braak neurofibrillary tangle stage, Thal amyloid phase, Lewy-related pathology, argyrophilic grains, and TDP-43-related pathology were also assessed. We retrospectively divided the patients into a normal cognition group (PSP-NC) and cognitive impairment group (PSP-CI) based on antemortem clinical information about cognitive impairment and compared the pathological changes between these groups. RESULTS Seven patients were categorized into the PSP-CI group (men = 4) and three into the PSP-NC group (men = 3). The severity of neuronal loss/gliosis and concurrent pathologies were not different between the two groups. However, the total load of tau pretangles/neurofibrillary tangles was higher in the PSP-CI group than in the PSP-NC group. In addition, the burden of tufted astrocytes in the subthalamic nucleus and medial thalamus was higher in the PSP-CI group than in the PSP-NC group. CONCLUSION Cognitive impairment in PSP may be associated with the amount of tufted astrocyte pathology in the subthalamic nucleus and medial thalamus.
Collapse
Affiliation(s)
- Mayuko Sakuwa
- Division of Neurology, Department of Brain and Neurosciences, Faculty of Medicine, Tottori University, Yonago, Japan
| | - Tadashi Adachi
- Division of Neuropathology, Department of Brain and Neurosciences, Faculty of Medicine, Tottori University, Yonago, Japan.
| | - Yuki Suzuki
- Division of Neurology, Department of Brain and Neurosciences, Faculty of Medicine, Tottori University, Yonago, Japan
| | - Hiroshi Takigawa
- Division of Neurology, Department of Brain and Neurosciences, Faculty of Medicine, Tottori University, Yonago, Japan
| | - Ritsuko Hanajima
- Division of Neurology, Department of Brain and Neurosciences, Faculty of Medicine, Tottori University, Yonago, Japan
| |
Collapse
|
12
|
Carson RE, Naganawa M, Toyonaga T, Koohsari S, Yang Y, Chen MK, Matuskey D, Finnema SJ. Imaging of Synaptic Density in Neurodegenerative Disorders. J Nucl Med 2022; 63:60S-67S. [PMID: 35649655 DOI: 10.2967/jnumed.121.263201] [Citation(s) in RCA: 53] [Impact Index Per Article: 17.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Revised: 02/10/2022] [Indexed: 02/07/2023] Open
Abstract
PET technology has produced many radiopharmaceuticals that target specific brain proteins and other measures of brain function. Recently, a new approach has emerged to image synaptic density by targeting the synaptic vesicle protein 2A (SV2A), an integral glycoprotein in the membrane of synaptic vesicles and widely distributed throughout the brain. Multiple SV2A ligands have been developed and translated to human use. The most successful of these to date is 11C-UCB-J, because of its high uptake, moderate metabolism, and effective quantification with a 1-tissue-compartment model. Further, since SV2A is the target of the antiepileptic drug levetiracetam, human blocking studies have characterized specific binding and potential reference regions. Regional brain SV2A levels were shown to correlate with those of synaptophysin, another commonly used marker of synaptic density, providing the basis for SV2A PET imaging to have broad utility across neuropathologic diseases. In this review, we highlight the development of SV2A tracers and the evaluation of quantification methods, including compartment modeling and simple tissue ratios. Mouse and rat models of neurodegenerative diseases have been studied with small-animal PET, providing validation by comparison to direct tissue measures. Next, we review human PET imaging results in multiple neurodegenerative disorders. Studies on Parkinson disease and Alzheimer disease have progressed most rapidly at multiple centers, with generally consistent results of patterns of SV2A or synaptic loss. In Alzheimer disease, the synaptic loss patterns differ from those of amyloid, tau, and 18F-FDG, although intertracer and interregional correlations have been found. Smaller studies have been reported in other disorders, including Lewy body dementia, frontotemporal dementia, Huntington disease, progressive supranuclear palsy, and corticobasal degeneration. In conclusion, PET imaging of SV2A has rapidly developed, and qualified radioligands are available. PET studies on humans indicate that SV2A loss might be specific to disease-associated brain regions and consistent with synaptic density loss. The recent availability of new 18F tracers, 18F-SynVesT-1 and 18F-SynVesT-2, will substantially broaden the application of SV2A PET. Future studies are needed in larger patient cohorts to establish the clinical value of SV2A PET and its potential for diagnosis and progression monitoring of neurodegenerative diseases, as well as efficacy assessment of disease-modifying therapies.
Collapse
Affiliation(s)
- Richard E Carson
- Department of Radiology and Biomedical Imaging, Yale Positron Emission Tomography Center, Yale University, New Haven, Connecticut;
- Department of Biomedical Engineering, Yale University, New Haven, Connecticut
| | - Mika Naganawa
- Department of Radiology and Biomedical Imaging, Yale Positron Emission Tomography Center, Yale University, New Haven, Connecticut
| | - Takuya Toyonaga
- Department of Radiology and Biomedical Imaging, Yale Positron Emission Tomography Center, Yale University, New Haven, Connecticut
| | - Sheida Koohsari
- Department of Radiology and Biomedical Imaging, Yale Positron Emission Tomography Center, Yale University, New Haven, Connecticut
| | - Yanghong Yang
- Department of Radiology and Biomedical Imaging, Yale Positron Emission Tomography Center, Yale University, New Haven, Connecticut
| | - Ming-Kai Chen
- Department of Radiology and Biomedical Imaging, Yale Positron Emission Tomography Center, Yale University, New Haven, Connecticut
| | - David Matuskey
- Department of Radiology and Biomedical Imaging, Yale Positron Emission Tomography Center, Yale University, New Haven, Connecticut
- Department of Psychiatry, Yale School of Medicine, New Haven, Connecticut
- Department of Neurology, Yale School of Medicine, New Haven, Connecticut; and
| | - Sjoerd J Finnema
- Neuroscience Discovery Research, Translational Imaging, AbbVie, North Chicago, Illinois
| |
Collapse
|
13
|
Tau deposition patterns are associated with functional connectivity in primary tauopathies. Nat Commun 2022; 13:1362. [PMID: 35292638 PMCID: PMC8924216 DOI: 10.1038/s41467-022-28896-3] [Citation(s) in RCA: 60] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Accepted: 02/14/2022] [Indexed: 11/08/2022] Open
Abstract
Tau pathology is the main driver of neuronal dysfunction in 4-repeat tauopathies, including cortico-basal degeneration and progressive supranuclear palsy. Tau is assumed to spread prion-like across connected neurons, but the mechanisms of tau propagation are largely elusive in 4-repeat tauopathies, characterized not only by neuronal but also by astroglial and oligodendroglial tau accumulation. Here, we assess whether connectivity is associated with 4R-tau deposition patterns by combining resting-state fMRI connectomics with both 2nd generation 18F-PI-2620 tau-PET in 46 patients with clinically diagnosed 4-repeat tauopathies and post-mortem cell-type-specific regional tau assessments from two independent progressive supranuclear palsy patient samples (n = 97 and n = 96). We find that inter-regional connectivity is associated with higher inter-regional correlation of both tau-PET and post-mortem tau levels in 4-repeat tauopathies. In regional cell-type specific post-mortem tau assessments, this association is stronger for neuronal than for astroglial or oligodendroglial tau, suggesting that connectivity is primarily associated with neuronal tau accumulation. Using tau-PET we find further that patient-level tau patterns are associated with the connectivity of subcortical tau epicenters. Together, the current study provides combined in vivo tau-PET and histopathological evidence that brain connectivity is associated with tau deposition patterns in 4-repeat tauopathies.
Collapse
|
14
|
Briel N, Ruf VC, Pratsch K, Roeber S, Widmann J, Mielke J, Dorostkar MM, Windl O, Arzberger T, Herms J, Struebing FL. Single-nucleus chromatin accessibility profiling highlights distinct astrocyte signatures in progressive supranuclear palsy and corticobasal degeneration. Acta Neuropathol 2022; 144:615-635. [PMID: 35976433 PMCID: PMC9468099 DOI: 10.1007/s00401-022-02483-8] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2022] [Revised: 08/03/2022] [Accepted: 08/08/2022] [Indexed: 01/31/2023]
Abstract
Tauopathies such as progressive supranuclear palsy (PSP) and corticobasal degeneration (CBD) exhibit characteristic neuronal and glial inclusions of hyperphosphorylated Tau (pTau). Although the astrocytic pTau phenotype upon neuropathological examination is the most guiding feature in distinguishing both diseases, regulatory mechanisms controlling their transitions into disease-specific states are poorly understood to date. Here, we provide accessible chromatin data of more than 45,000 single nuclei isolated from the frontal cortex of PSP, CBD, and control individuals. We found a strong association of disease-relevant molecular changes with astrocytes and demonstrate that tauopathy-relevant genetic risk variants are tightly linked to astrocytic chromatin accessibility profiles in the brains of PSP and CBD patients. Unlike the established pathogenesis in the secondary tauopathy Alzheimer disease, microglial alterations were relatively sparse. Transcription factor (TF) motif enrichments in pseudotime as well as modeling of the astrocytic TF interplay suggested a common pTau signature for CBD and PSP that is reminiscent of an inflammatory immediate-early response. Nonetheless, machine learning models also predicted discriminatory features, and we observed marked differences in molecular entities related to protein homeostasis between both diseases. Predicted TF involvement was supported by immunofluorescence analyses in postmortem brain tissue for their highly correlated target genes. Collectively, our data expand the current knowledge on risk gene involvement (e.g., MAPT, MAPK8, and NFE2L2) and molecular pathways leading to the phenotypic changes associated with CBD and PSP.
Collapse
Affiliation(s)
- Nils Briel
- Center for Neuropathology and Prion Research, University Hospital Munich, Ludwig–Maximilians-University, Feodor-Lynen-Str. 23, 81377 Munich, Germany ,German Center for Neurodegenerative Diseases, Feodor-Lynen-Str. 17, 81377 Munich, Germany ,Munich Medical Research School, Faculty of Medicine, Ludwig-Maximilians-University, Bavariaring 19, 80336 Munich, Germany
| | - Viktoria C. Ruf
- Center for Neuropathology and Prion Research, University Hospital Munich, Ludwig–Maximilians-University, Feodor-Lynen-Str. 23, 81377 Munich, Germany
| | - Katrin Pratsch
- Center for Neuropathology and Prion Research, University Hospital Munich, Ludwig–Maximilians-University, Feodor-Lynen-Str. 23, 81377 Munich, Germany ,German Center for Neurodegenerative Diseases, Feodor-Lynen-Str. 17, 81377 Munich, Germany
| | - Sigrun Roeber
- Center for Neuropathology and Prion Research, University Hospital Munich, Ludwig–Maximilians-University, Feodor-Lynen-Str. 23, 81377 Munich, Germany
| | - Jeannine Widmann
- Center for Neuropathology and Prion Research, University Hospital Munich, Ludwig–Maximilians-University, Feodor-Lynen-Str. 23, 81377 Munich, Germany
| | - Janina Mielke
- Center for Neuropathology and Prion Research, University Hospital Munich, Ludwig–Maximilians-University, Feodor-Lynen-Str. 23, 81377 Munich, Germany
| | - Mario M. Dorostkar
- Center for Neuropathology and Prion Research, University Hospital Munich, Ludwig–Maximilians-University, Feodor-Lynen-Str. 23, 81377 Munich, Germany
| | - Otto Windl
- Center for Neuropathology and Prion Research, University Hospital Munich, Ludwig–Maximilians-University, Feodor-Lynen-Str. 23, 81377 Munich, Germany
| | - Thomas Arzberger
- Center for Neuropathology and Prion Research, University Hospital Munich, Ludwig–Maximilians-University, Feodor-Lynen-Str. 23, 81377 Munich, Germany ,German Center for Neurodegenerative Diseases, Feodor-Lynen-Str. 17, 81377 Munich, Germany ,Department of Psychiatry and Psychotherapy, University Hospital Munich, Ludwig-Maximilians-University, Nussbaumstr. 7, 80336 Munich, Germany
| | - Jochen Herms
- Center for Neuropathology and Prion Research, University Hospital Munich, Ludwig–Maximilians-University, Feodor-Lynen-Str. 23, 81377 Munich, Germany ,German Center for Neurodegenerative Diseases, Feodor-Lynen-Str. 17, 81377 Munich, Germany ,Munich Cluster of Systems Neurology (SyNergy), Feodor-Lynen-Str. 17, 81377 Munich, Germany
| | - Felix L. Struebing
- Center for Neuropathology and Prion Research, University Hospital Munich, Ludwig–Maximilians-University, Feodor-Lynen-Str. 23, 81377 Munich, Germany ,German Center for Neurodegenerative Diseases, Feodor-Lynen-Str. 17, 81377 Munich, Germany
| |
Collapse
|
15
|
Wu M, Zhang M, Yin X, Chen K, Hu Z, Zhou Q, Cao X, Chen Z, Liu D. The role of pathological tau in synaptic dysfunction in Alzheimer's diseases. Transl Neurodegener 2021; 10:45. [PMID: 34753506 PMCID: PMC8579533 DOI: 10.1186/s40035-021-00270-1] [Citation(s) in RCA: 59] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2021] [Accepted: 10/25/2021] [Indexed: 12/12/2022] Open
Abstract
Alzheimer’s disease (AD) is a neurodegenerative disease characterized by progressive cognitive decline, accompanied by amyloid-β (Aβ) overload and hyperphosphorylated tau accumulation in the brain. Synaptic dysfunction, an important pathological hallmark in AD, is recognized as the main cause of the cognitive impairments. Accumulating evidence suggests that synaptic dysfunction could be an early pathological event in AD. Pathological tau, which is detached from axonal microtubules and mislocalized into pre- and postsynaptic neuronal compartments, is suggested to induce synaptic dysfunction in several ways, including reducing mobility and release of presynaptic vesicles, decreasing glutamatergic receptors, impairing the maturation of dendritic spines at postsynaptic terminals, disrupting mitochondrial transport and function in synapses, and promoting the phagocytosis of synapses by microglia. Here, we review the current understanding of how pathological tau mediates synaptic dysfunction and contributes to cognitive decline in AD. We propose that elucidating the mechanism by which pathological tau impairs synaptic function is essential for exploring novel therapeutic strategies for AD.
Collapse
Affiliation(s)
- Moxin Wu
- Department of Medical Laboratory, Affiliated Hospital of Jiujiang University, Jiujiang, 332000, China.,Jiujiang Clinical Precision Medicine Research Center, Jiujiang, 332000, China
| | - Manqing Zhang
- Medical College of Jiujiang University, Jiujiang, 332000, China
| | - Xiaoping Yin
- Jiujiang Clinical Precision Medicine Research Center, Jiujiang, 332000, China.,Department of Neurology, Affiliated Hospital of Jiujiang University, Jiujiang, 332000, China
| | - Kai Chen
- Department of Dermatology, Wuhan No. 1 Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Zhijian Hu
- Department of Medical Laboratory, Affiliated Hospital of Jiujiang University, Jiujiang, 332000, China
| | - Qin Zhou
- Jiujiang Clinical Precision Medicine Research Center, Jiujiang, 332000, China
| | - Xianming Cao
- Jiujiang Clinical Precision Medicine Research Center, Jiujiang, 332000, China.,Department of Neurology, Affiliated Hospital of Jiujiang University, Jiujiang, 332000, China
| | - Zhiying Chen
- Jiujiang Clinical Precision Medicine Research Center, Jiujiang, 332000, China. .,Department of Neurology, Affiliated Hospital of Jiujiang University, Jiujiang, 332000, China.
| | - Dan Liu
- Department of Medical Genetics, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.
| |
Collapse
|
16
|
Holland N, Malpetti M, Rittman T, Mak EE, Passamonti L, Kaalund SS, Hezemans FH, Jones PS, Savulich G, Hong YT, Fryer TD, Aigbirhio FI, O'Brien JT, Rowe JB. Molecular pathology and synaptic loss in primary tauopathies: an 18F-AV-1451 and 11C-UCB-J PET study. Brain 2021; 145:340-348. [PMID: 34398211 PMCID: PMC8967099 DOI: 10.1093/brain/awab282] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2021] [Revised: 07/02/2021] [Accepted: 07/10/2021] [Indexed: 12/02/2022] Open
Abstract
The relationship between in vivo synaptic density and molecular pathology in primary tauopathies is key to understanding the impact of tauopathy on functional decline and in informing new early therapeutic strategies. In this cross-sectional observational study, we determine the in vivo relationship between synaptic density and molecular pathology in the primary tauopathies of progressive supranuclear palsy and corticobasal degeneration as a function of disease severity. Twenty-three patients with progressive supranuclear palsy and 12 patients with corticobasal syndrome were recruited from a tertiary referral centre. Nineteen education-, sex- and gender-matched control participants were recruited from the National Institute for Health Research ‘Join Dementia Research’ platform. Cerebral synaptic density and molecular pathology, in all participants, were estimated using PET imaging with the radioligands 11C-UCB-J and 18F-AV-1451, respectively. Patients with corticobasal syndrome also underwent amyloid PET imaging with 11C-PiB to exclude those with likely Alzheimer’s pathology—we refer to the amyloid-negative cohort as having corticobasal degeneration, although we acknowledge other underlying pathologies exist. Disease severity was assessed with the progressive supranuclear palsy rating scale; regional non-displaceable binding potentials of 11C-UCB-J and 18F-AV-1451 were estimated in regions of interest from the Hammersmith Atlas, excluding those with known off-target binding for 18F-AV-1451. As an exploratory analysis, we also investigated the relationship between molecular pathology in cortical brain regions and synaptic density in subcortical areas. Across brain regions, there was a positive correlation between 11C-UCB-J and 18F-AV-1451 non-displaceable binding potentials (β = 0.4, t = 3.6, P = 0.001), independent of age or time between PET scans. However, this correlation became less positive as a function of disease severity in patients (β = −0.02, t = −2.9, P = 0.007, R = −0.41). Between regions, cortical 18F-AV-1451 binding was negatively correlated with synaptic density in subcortical areas (caudate nucleus, putamen). Brain regions with higher synaptic density are associated with a higher 18F-AV-1451 binding in progressive supranuclear palsy/corticobasal degeneration, but this association diminishes with disease severity. Moreover, higher cortical 18F-AV-1451 binding correlates with lower subcortical synaptic density. Longitudinal imaging is required to confirm the mediation of synaptic loss by molecular pathology. However, the effect of disease severity suggests a biphasic relationship between synaptic density and molecular pathology with synapse-rich regions vulnerable to accrual of pathological aggregates, followed by a loss of synapses in response to the molecular pathology. Given the importance of synaptic function for cognition and action, our study elucidates the pathophysiology of primary tauopathies and may inform the design of future clinical trials.
Collapse
Affiliation(s)
- Negin Holland
- Department of Clinical Neurosciences, University of Cambridge, Cambridge Biomedical Campus, Cambridge, CB2 0SZ, UK.,Cambridge University Hospitals NHS Foundation Trust, Cambridge, CB2 0QQ, UK
| | - Maura Malpetti
- Department of Clinical Neurosciences, University of Cambridge, Cambridge Biomedical Campus, Cambridge, CB2 0SZ, UK
| | - Timothy Rittman
- Department of Clinical Neurosciences, University of Cambridge, Cambridge Biomedical Campus, Cambridge, CB2 0SZ, UK.,Cambridge University Hospitals NHS Foundation Trust, Cambridge, CB2 0QQ, UK
| | - Elijah E Mak
- Department of Psychiatry, University of Cambridge, School of Clinical Medicine, Cambridge Biomedical Campus, CB2 0QQ, UK
| | - Luca Passamonti
- Department of Clinical Neurosciences, University of Cambridge, Cambridge Biomedical Campus, Cambridge, CB2 0SZ, UK.,Istituto di Bioimmagini e Fisiologia Molecolare (IBFM), Consiglio Nazionale delle Ricerche (CNR), 20090, Milano, Italy
| | - Sanne S Kaalund
- Department of Clinical Neurosciences, University of Cambridge, Cambridge Biomedical Campus, Cambridge, CB2 0SZ, UK
| | - Frank H Hezemans
- Department of Clinical Neurosciences, University of Cambridge, Cambridge Biomedical Campus, Cambridge, CB2 0SZ, UK.,Medical Research Council Cognition and Brain Sciences Unit, University of Cambridge, CB2 7EF, UK
| | - P Simon Jones
- Department of Clinical Neurosciences, University of Cambridge, Cambridge Biomedical Campus, Cambridge, CB2 0SZ, UK
| | - George Savulich
- Department of Psychiatry, University of Cambridge, School of Clinical Medicine, Cambridge Biomedical Campus, CB2 0QQ, UK
| | - Young T Hong
- Wolfson Brain Imaging Centre, University of Cambridge, CB2 0QQ, UK
| | - Tim D Fryer
- Department of Clinical Neurosciences, University of Cambridge, Cambridge Biomedical Campus, Cambridge, CB2 0SZ, UK.,Wolfson Brain Imaging Centre, University of Cambridge, CB2 0QQ, UK
| | - Franklin I Aigbirhio
- Department of Clinical Neurosciences, University of Cambridge, Cambridge Biomedical Campus, Cambridge, CB2 0SZ, UK
| | - John T O'Brien
- Cambridge University Hospitals NHS Foundation Trust, Cambridge, CB2 0QQ, UK.,Department of Psychiatry, University of Cambridge, School of Clinical Medicine, Cambridge Biomedical Campus, CB2 0QQ, UK
| | - James B Rowe
- Department of Clinical Neurosciences, University of Cambridge, Cambridge Biomedical Campus, Cambridge, CB2 0SZ, UK.,Cambridge University Hospitals NHS Foundation Trust, Cambridge, CB2 0QQ, UK.,Medical Research Council Cognition and Brain Sciences Unit, University of Cambridge, CB2 7EF, UK
| |
Collapse
|
17
|
Adams NE, Hughes LE, Rouse MA, Phillips HN, Shaw AD, Murley AG, Cope TE, Bevan-Jones WR, Passamonti L, Street D, Holland N, Nesbitt D, Friston K, Rowe JB. GABAergic cortical network physiology in frontotemporal lobar degeneration. Brain 2021; 144:2135-2145. [PMID: 33710299 PMCID: PMC8370432 DOI: 10.1093/brain/awab097] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2020] [Revised: 12/31/2020] [Accepted: 01/03/2021] [Indexed: 11/23/2022] Open
Abstract
The clinical syndromes caused by frontotemporal lobar degeneration are heterogeneous, including the behavioural variant frontotemporal dementia (bvFTD) and progressive supranuclear palsy. Although pathologically distinct, they share many behavioural, cognitive and physiological features, which may in part arise from common deficits of major neurotransmitters such as γ-aminobutyric acid (GABA). Here, we quantify the GABAergic impairment and its restoration with dynamic causal modelling of a double-blind placebo-controlled crossover pharmaco-magnetoencephalography study. We analysed 17 patients with bvFTD, 15 patients with progressive supranuclear palsy, and 20 healthy age- and gender-matched controls. In addition to neuropsychological assessment and structural MRI, participants undertook two magnetoencephalography sessions using a roving auditory oddball paradigm: once on placebo and once on 10 mg of the oral GABA reuptake inhibitor tiagabine. A subgroup underwent ultrahigh-field magnetic resonance spectroscopy measurement of GABA concentration, which was reduced among patients. We identified deficits in frontotemporal processing using conductance-based biophysical models of local and global neuronal networks. The clinical relevance of this physiological deficit is indicated by the correlation between top-down connectivity from frontal to temporal cortex and clinical measures of cognitive and behavioural change. A critical validation of the biophysical modelling approach was evidence from parametric empirical Bayes analysis that GABA levels in patients, measured by spectroscopy, were related to posterior estimates of patients’ GABAergic synaptic connectivity. Further evidence for the role of GABA in frontotemporal lobar degeneration came from confirmation that the effects of tiagabine on local circuits depended not only on participant group, but also on individual baseline GABA levels. Specifically, the phasic inhibition of deep cortico-cortical pyramidal neurons following tiagabine, but not placebo, was a function of GABA concentration. The study provides proof-of-concept for the potential of dynamic causal modelling to elucidate mechanisms of human neurodegenerative disease, and explains the variation in response to candidate therapies among patients. The laminar- and neurotransmitter-specific features of the modelling framework, can be used to study other treatment approaches and disorders. In the context of frontotemporal lobar degeneration, we suggest that neurophysiological restoration in selected patients, by targeting neurotransmitter deficits, could be used to bridge between clinical and preclinical models of disease, and inform the personalized selection of drugs and stratification of patients for future clinical trials.
Collapse
Affiliation(s)
- Natalie E Adams
- Department of Clinical Neurosciences, Cambridge Biomedical Campus, University of Cambridge, Cambridge CB2 0QQ, UK
| | - Laura E Hughes
- Department of Clinical Neurosciences, Cambridge Biomedical Campus, University of Cambridge, Cambridge CB2 0QQ, UK.,MMRC Cognition and Brain Sciences Unit, Cambridge CB2 7EF, UK
| | - Matthew A Rouse
- Department of Clinical Neurosciences, Cambridge Biomedical Campus, University of Cambridge, Cambridge CB2 0QQ, UK
| | - Holly N Phillips
- Department of Clinical Neurosciences, Cambridge Biomedical Campus, University of Cambridge, Cambridge CB2 0QQ, UK
| | | | - Alexander G Murley
- Department of Clinical Neurosciences, Cambridge Biomedical Campus, University of Cambridge, Cambridge CB2 0QQ, UK.,Cambridge University Hospitals, Cambridge, CB2 0QQ, UK
| | - Thomas E Cope
- Department of Clinical Neurosciences, Cambridge Biomedical Campus, University of Cambridge, Cambridge CB2 0QQ, UK.,MMRC Cognition and Brain Sciences Unit, Cambridge CB2 7EF, UK.,Cambridge University Hospitals, Cambridge, CB2 0QQ, UK
| | - W Richard Bevan-Jones
- Department of Clinical Neurosciences, Cambridge Biomedical Campus, University of Cambridge, Cambridge CB2 0QQ, UK.,Cambridge University Hospitals, Cambridge, CB2 0QQ, UK
| | - Luca Passamonti
- Department of Clinical Neurosciences, Cambridge Biomedical Campus, University of Cambridge, Cambridge CB2 0QQ, UK.,Cambridge University Hospitals, Cambridge, CB2 0QQ, UK
| | - Duncan Street
- Department of Clinical Neurosciences, Cambridge Biomedical Campus, University of Cambridge, Cambridge CB2 0QQ, UK.,Cambridge University Hospitals, Cambridge, CB2 0QQ, UK
| | - Negin Holland
- Department of Clinical Neurosciences, Cambridge Biomedical Campus, University of Cambridge, Cambridge CB2 0QQ, UK.,Cambridge University Hospitals, Cambridge, CB2 0QQ, UK
| | - David Nesbitt
- Department of Clinical Neurosciences, Cambridge Biomedical Campus, University of Cambridge, Cambridge CB2 0QQ, UK.,MMRC Cognition and Brain Sciences Unit, Cambridge CB2 7EF, UK.,Cambridge University Hospitals, Cambridge, CB2 0QQ, UK
| | - Karl Friston
- Wellcome Centre for Human Neuroimaging, University College London, London WC1N 3AR, UK
| | - James B Rowe
- Department of Clinical Neurosciences, Cambridge Biomedical Campus, University of Cambridge, Cambridge CB2 0QQ, UK.,MMRC Cognition and Brain Sciences Unit, Cambridge CB2 7EF, UK.,Cambridge University Hospitals, Cambridge, CB2 0QQ, UK
| |
Collapse
|
18
|
Mak E, Holland N, Jones PS, Savulich G, Low A, Malpetti M, Kaalund SS, Passamonti L, Rittman T, Romero-Garcia R, Manavaki R, Williams GB, Hong YT, Fryer TD, Aigbirhio FI, O'Brien JT, Rowe JB. In vivo coupling of dendritic complexity with presynaptic density in primary tauopathies. Neurobiol Aging 2021; 101:187-198. [PMID: 33631470 PMCID: PMC8209289 DOI: 10.1016/j.neurobiolaging.2021.01.016] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2020] [Revised: 01/12/2021] [Accepted: 01/13/2021] [Indexed: 01/03/2023]
Abstract
Understanding the cellular underpinnings of neurodegeneration remains a challenge; loss of synapses and dendritic arborization are characteristic and can be quantified in vivo, with [11C]UCB-J PET and MRI-based Orientation Dispersion Imaging (ODI), respectively. We aimed to assess how both measures are correlated, in 4R-tauopathies of progressive supranuclear palsy - Richardson's Syndrome (PSP-RS; n = 22) and amyloid-negative (determined by [11C]PiB PET) Corticobasal Syndrome (Cortiobasal degeneration, CBD; n =14), as neurodegenerative disease models, in this proof-of-concept study. Compared to controls (n = 27), PSP-RS and CBD patients had widespread reductions in cortical ODI, and [11C]UCB-J non-displaceable binding potential (BPND) in excess of atrophy. In PSP-RS and CBD separately, regional cortical ODI was significantly associated with [11C]UCB-J BPND in disease-associated regions (p < 0.05, FDR corrected). Our findings indicate that reductions in synaptic density and dendritic complexity in PSP-RS and CBD are more severe and extensive than atrophy. Furthermore, both measures are tightly coupled in vivo, furthering our understanding of the pathophysiology of neurodegeneration, and applicable to studies of early neurodegeneration with a safe and widely available MRI platform.
Collapse
Affiliation(s)
- Elijah Mak
- Department of Psychiatry, University of Cambridge, School of Clinical Medicine, Cambridge Biomedical Campus, Cambridge, UK
| | - Negin Holland
- Department of Clinical Neurosciences, University of Cambridge, Cambridge Biomedical Campus, Cambridge, UK; Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK.
| | - P Simon Jones
- Department of Clinical Neurosciences, University of Cambridge, Cambridge Biomedical Campus, Cambridge, UK
| | - George Savulich
- Department of Psychiatry, University of Cambridge, School of Clinical Medicine, Cambridge Biomedical Campus, Cambridge, UK
| | - Audrey Low
- Department of Psychiatry, University of Cambridge, School of Clinical Medicine, Cambridge Biomedical Campus, Cambridge, UK
| | - Maura Malpetti
- Department of Clinical Neurosciences, University of Cambridge, Cambridge Biomedical Campus, Cambridge, UK
| | - Sanne S Kaalund
- Department of Clinical Neurosciences, University of Cambridge, Cambridge Biomedical Campus, Cambridge, UK
| | - Luca Passamonti
- Department of Clinical Neurosciences, University of Cambridge, Cambridge Biomedical Campus, Cambridge, UK
| | - Timothy Rittman
- Department of Clinical Neurosciences, University of Cambridge, Cambridge Biomedical Campus, Cambridge, UK
| | - Rafael Romero-Garcia
- Department of Psychiatry, University of Cambridge, School of Clinical Medicine, Cambridge Biomedical Campus, Cambridge, UK
| | - Roido Manavaki
- Wolfson Brain Imaging Centre, University of Cambridge, Cambridge, UK
| | - Guy B Williams
- Wolfson Brain Imaging Centre, University of Cambridge, Cambridge, UK
| | - Young T Hong
- Wolfson Brain Imaging Centre, University of Cambridge, Cambridge, UK
| | - Tim D Fryer
- Department of Clinical Neurosciences, University of Cambridge, Cambridge Biomedical Campus, Cambridge, UK; Wolfson Brain Imaging Centre, University of Cambridge, Cambridge, UK
| | - Franklin I Aigbirhio
- Department of Clinical Neurosciences, University of Cambridge, Cambridge Biomedical Campus, Cambridge, UK
| | - John T O'Brien
- Department of Psychiatry, University of Cambridge, School of Clinical Medicine, Cambridge Biomedical Campus, Cambridge, UK; Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK
| | - James B Rowe
- Department of Clinical Neurosciences, University of Cambridge, Cambridge Biomedical Campus, Cambridge, UK; Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK; Medical Research Council Cognition and Brain Sciences Unit, University of Cambridge, Cambridge, UK
| |
Collapse
|
19
|
Briel N, Pratsch K, Roeber S, Arzberger T, Herms J. Contribution of the astrocytic tau pathology to synapse loss in progressive supranuclear palsy and corticobasal degeneration. Brain Pathol 2020; 31:e12914. [PMID: 33089580 PMCID: PMC8412068 DOI: 10.1111/bpa.12914] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2020] [Accepted: 10/12/2020] [Indexed: 12/15/2022] Open
Abstract
Primary 4‐repeat tauopathies with frontotemporal lobar degeneration (FTLD) like Progressive Supranuclear Palsy (PSP) or Corticobasal Degeneration (CBD) show diverse cellular pathology in various brain regions. Besides shared characteristics of neuronal and oligodendroglial cytoplasmic inclusions of accumulated hyperphosphorylated tau protein (pTau), astrocytes in PSP and CBD contain pathognomonic pTau aggregates — hence, lending the designation tufted astrocytes (TA) or astrocytic plaques (AP), respectively. pTau toxicity is most commonly assigned to neurons, whereas the implications of astrocytic pTau for maintaining neurotransmission within the tripartite synapse of human brains is not well understood. We performed immunofluorescent synapse labeling and automated puncta quantification in the medial frontal gyrus (MFG) and striatal regions from PSP and CBD postmortem samples to capture morphometric synaptic alterations. This approach indicated general synaptic losses of both, excitatory and inhibitory bipartite synapses in the frontal cortex of PSP cases, whereas in CBD lower synapse densities were only related to astrocytic plaques. In contrast to tufted astrocytes in PSP, affected astrocytes in CBD could not preserve synaptic integrity within their spatial domains, when compared to non‐affected internal astrocytes or astrocytes in healthy controls. These findings suggest a pTau pathology‐associated role of astrocytes in maintaining connections within neuronal circuits, considered as the microscopic substrate of cognitive dysfunction in CBD. By contrasting astrocytic‐synaptic associations in both diseases, we hereby highlight astrocytic pTau as an important subject of prospective research and as a potential cellular target for therapeutic approaches in the primary tauopathies PSP and CBD.
Collapse
Affiliation(s)
- Nils Briel
- German Center for Neurodegenerative Diseases (DZNE) e.V., Site Munich, Munich, Germany.,Center for Neuropathology and Prion Research, University Hospital Munich, Ludwig-Maximilians-University, Munich, Germany.,Munich Medical Research School, Faculty of Medicine, Ludwig-Maximilians-University, Munich, Germany
| | - Katrin Pratsch
- German Center for Neurodegenerative Diseases (DZNE) e.V., Site Munich, Munich, Germany.,Center for Neuropathology and Prion Research, University Hospital Munich, Ludwig-Maximilians-University, Munich, Germany
| | - Sigrun Roeber
- Center for Neuropathology and Prion Research, University Hospital Munich, Ludwig-Maximilians-University, Munich, Germany
| | - Thomas Arzberger
- German Center for Neurodegenerative Diseases (DZNE) e.V., Site Munich, Munich, Germany.,Center for Neuropathology and Prion Research, University Hospital Munich, Ludwig-Maximilians-University, Munich, Germany.,Department of Psychiatry and Psychotherapy, University Hospital Munich, Ludwig-Maximilians-University, Munich, Germany
| | - Jochen Herms
- German Center for Neurodegenerative Diseases (DZNE) e.V., Site Munich, Munich, Germany.,Center for Neuropathology and Prion Research, University Hospital Munich, Ludwig-Maximilians-University, Munich, Germany.,Munich Cluster of Systems Neurology (SyNergy), Ludwig-Maximilians-University, Munich, Germany
| |
Collapse
|
20
|
Holland N, Jones PS, Savulich G, Wiggins JK, Hong YT, Fryer TD, Manavaki R, Sephton SM, Boros I, Malpetti M, Hezemans FH, Aigbirhio FI, Coles JP, O’Brien J, Rowe JB. Synaptic Loss in Primary Tauopathies Revealed by [ 11 C]UCB-J Positron Emission Tomography. Mov Disord 2020; 35:1834-1842. [PMID: 32652635 PMCID: PMC7611123 DOI: 10.1002/mds.28188] [Citation(s) in RCA: 61] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2020] [Revised: 05/26/2020] [Accepted: 06/08/2020] [Indexed: 01/08/2023] Open
Abstract
BACKGROUND Synaptic loss is a prominent and early feature of many neurodegenerative diseases. OBJECTIVES We tested the hypothesis that synaptic density is reduced in the primary tauopathies of progressive supranuclear palsy (PSP) (Richardson's syndrome) and amyloid-negative corticobasal syndrome (CBS). METHODS Forty-four participants (15 CBS, 14 PSP, and 15 age-/sex-/education-matched controls) underwent PET with the radioligand [11 C]UCB-J, which binds to synaptic vesicle glycoprotein 2A, a marker of synaptic density; participants also had 3 Tesla MRI and clinical and neuropsychological assessment. RESULTS Nine CBS patients had negative amyloid biomarkers determined by [11 C]PiB PET and hence were deemed likely to have corticobasal degeneration (CBD). Patients with PSP-Richardson's syndrome and amyloid-negative CBS were impaired in executive, memory, and visuospatial tasks. [11 C]UCB-J binding was reduced across frontal, temporal, parietal, and occipital lobes, cingulate, hippocampus, insula, amygdala, and subcortical structures in both PSP and CBD patients compared to controls (P < 0.01), with median reductions up to 50%, consistent with postmortem data. Reductions of 20% to 30% were widespread even in areas of the brain with minimal atrophy. There was a negative correlation between global [11 C]UCB-J binding and the PSP and CBD rating scales (R = -0.61, P < 0.002; R = -0.72, P < 0.001, respectively) and a positive correlation with the revised Addenbrooke's Cognitive Examination (R = 0.52; P = 0.01). CONCLUSIONS We confirm severe synaptic loss in PSP and CBD in proportion to disease severity, providing critical insight into the pathophysiology of primary degenerative tauopathies. [11 C]UCB-J may facilitate treatment strategies for disease-modification, synaptic maintenance, or restoration. © 2020 The Authors. Movement Disorders published by Wiley Periodicals LLC on behalf of International Parkinson and Movement Disorder Society.
Collapse
Affiliation(s)
- Negin Holland
- Department of Clinical Neurosciences, University of Cambridge
| | - P. Simon Jones
- Department of Clinical Neurosciences, University of Cambridge
| | | | | | - Young T. Hong
- Department of Clinical Neurosciences, University of Cambridge
- Wolfson Brain Imaging Centre, University of Cambridge
| | - Tim D. Fryer
- Department of Clinical Neurosciences, University of Cambridge
- Wolfson Brain Imaging Centre, University of Cambridge
| | | | - Selena Milicevic Sephton
- Department of Clinical Neurosciences, University of Cambridge
- Wolfson Brain Imaging Centre, University of Cambridge
| | - Istvan Boros
- Department of Clinical Neurosciences, University of Cambridge
- Wolfson Brain Imaging Centre, University of Cambridge
| | - Maura Malpetti
- Department of Clinical Neurosciences, University of Cambridge
| | - Frank H. Hezemans
- Department of Clinical Neurosciences, University of Cambridge
- Medical Research Council Cognition and Brain Sciences Unit, University of Cambridge
| | | | - Jonathan P. Coles
- Division of Anaesthesia, Department of Medicine, University of Cambridge
- Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK
| | - John O’Brien
- Department of Psychiatry, University of Cambridge
- Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK
| | - James B. Rowe
- Department of Clinical Neurosciences, University of Cambridge
- Medical Research Council Cognition and Brain Sciences Unit, University of Cambridge
- Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK
| |
Collapse
|
21
|
Whitwell JL, Tosakulwong N, Botha H, Ali F, Clark HM, Duffy JR, Utianski RL, Stevens CA, Weigand SD, Schwarz CG, Senjem ML, Jack CR, Lowe VJ, Ahlskog JE, Dickson DW, Josephs KA. Brain volume and flortaucipir analysis of progressive supranuclear palsy clinical variants. NEUROIMAGE-CLINICAL 2019; 25:102152. [PMID: 31935638 PMCID: PMC6961761 DOI: 10.1016/j.nicl.2019.102152] [Citation(s) in RCA: 54] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/05/2019] [Revised: 11/25/2019] [Accepted: 12/26/2019] [Indexed: 12/12/2022]
Abstract
All PSP variants showed atrophy or flortaucipir uptake in subcortical structures. Speech/language, frontal and corticobasal variants showed cortical involvement. Dentatorubrothalamic tract involvement was only seen in some variants. PSP variants show different patterns of damage to subcortical-cortical circuitry.
Background and purpose Progressive supranuclear palsy (PSP) is a neurodegenerative tauopathy that is associated with different clinical variants, including PSP-Richardson's syndrome (PSP-RS), PSP-parkinsonism (PSP-P), PSP-corticobasal syndrome (PSP-CBS), PSP-frontal (PSP-F), PSP-progressive gait freezing (PSP-PGF) and PSP-speech/language (PSP-SL). While PSP-RS has been well-characterized on neuroimaging, the characteristics of the other atypical variants are less well defined and it is unknown how they compare to each other or relate to neuropathology. We aimed to assess and compare regional atrophy on MRI and [18F]flortaucipir uptake on PET across PSP variants. Materials and methods 105 PSP patients (53 PSP-RS, 23 PSP-SL, 12 PSP-P, 8 PSP-CBS, 5 PSP-F and 4 PSP-PGF) underwent volumetric MRI, with 59 of these also undergoing flortaucipir PET. Voxel-level and region-level analyses were performed comparing PSP variants to 30 controls and to each other. Semi-quantitative tau burden measurements were also performed in 21 patients with autopsy-confirmed PSP. Results All variants showed evidence for atrophy or increased flortaucipir uptake in striatum, globus pallidus and thalamus. Superior cerebellar peduncle volume loss was only observed in PSP-RS, PSP-CBS and PSP-F. Volume loss in the frontal lobes was observed in PSP-SL, PSP-CBS and PSP-F, with these variants also showing highest cortical tau burden at autopsy. The PSP-P and PSP-PGF variants showed more restricted patterns of neurodegeneration predominantly involving striatum, globus pallidus, subthalamic nucleus and thalamus. The PSP-SL variant showed greater volume loss and flortaucipir uptake in supplementary motor area and motor cortex compared to all other variants, but showed less involvement of subthalamic nucleus and midbrain. Compared to PSP-RS, PSP-P had larger midbrain volume and greater flortaucipir uptake in putamen. Conclusion The PSP variants have different patterns of involvement of subcortical circuitry, perhaps suggesting different patterns of disease spread through the brain. These findings will be important in the development of appropriate neuroimaging biomarkers for the different PSP variants.
Collapse
Affiliation(s)
| | - Nirubol Tosakulwong
- Department of Health Sciences Research, Mayo Clinic, Rochester, MN, United States
| | - Hugo Botha
- Department of Neurology, Mayo Clinic, Rochester, MN, United States
| | - Farwa Ali
- Department of Neurology, Mayo Clinic, Rochester, MN, United States
| | - Heather M Clark
- Department of Neurology, Mayo Clinic, Rochester, MN, United States
| | - Joseph R Duffy
- Department of Neurology, Mayo Clinic, Rochester, MN, United States
| | - Rene L Utianski
- Department of Neurology, Mayo Clinic, Rochester, MN, United States
| | - Chase A Stevens
- Department of Neurology, Mayo Clinic, Rochester, MN, United States
| | - Stephen D Weigand
- Department of Health Sciences Research, Mayo Clinic, Rochester, MN, United States
| | | | - Matthew L Senjem
- Department of Radiology, Mayo Clinic, Rochester, MN, United States; Department of Information Technology, Mayo Clinic, Rochester, MN, United States
| | - Clifford R Jack
- Department of Radiology, Mayo Clinic, Rochester, MN, United States
| | - Val J Lowe
- Department of Radiology, Mayo Clinic, Rochester, MN, United States
| | - J Eric Ahlskog
- Department of Neurology, Mayo Clinic, Rochester, MN, United States
| | - Dennis W Dickson
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, United States
| | - Keith A Josephs
- Department of Neurology, Mayo Clinic, Rochester, MN, United States
| |
Collapse
|
22
|
Lenka A, Pasha SA, Mangalore S, George L, Jhunjhunwala KR, Bagepally BS, Naduthota RM, Saini J, Yadav R, Pal PK. Role of Corpus Callosum Volumetry in Differentiating the Subtypes of Progressive Supranuclear Palsy and Early Parkinson's Disease. Mov Disord Clin Pract 2017; 4:552-558. [PMID: 30363434 DOI: 10.1002/mdc3.12473] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2016] [Revised: 12/28/2016] [Accepted: 01/04/2017] [Indexed: 11/09/2022] Open
Abstract
Background and Objective Progressive supranuclear palsy (PSP) is a progressive neurodegenerative disorder. Classic PSP or Richardson-Steele phenotype (PSP-RS) and parkinsonian phenotype (PSP-P) are the common subtypes of PSP. At the early stage, differentiating the subtypes of PSP as well as differentiating PSP from other parkinsonian disorders, especially Parkinson's disease (PD) is challenging. Microstructural abnormalities of corpus callosum (CC) have been reported both in PSP and PD. The objective of this study was to compare the volumes of various segments of CC between patients with PSP-P, PSP-RS, and early PD. Methodology This study included 32 patients with PSP (RS: 18, P: 14), 20 patients with early PD, and 25 controls. All subjects underwent 3-Tesla MRI. An automated surface-based analysis package (FreeSurfer) was used to divide CC into five segments: anterior (CC1), midanterior (CC2), central (CC3), midposterior (CC4), and posterior (CC5). Volumes of these segments were compared among the four groups. Results The PSP-RS group had significantly lower CC volume in all segments except in CC1 and CC5, whereas the volumes of the five segments of CC were comparable among PSP-P, PD and controls. The PSP-RS group had lower CC3 volume compared to the PSP-P group, and the PSP-RS group had lower volume of both CC2 and CC3 compared to the PD group. Conclusions The lower volume of the central segment of CC (CC3) might help in differentiating PSP-RS from PSP-P. There is no significant difference in the pattern of CC atrophy in PSP-P and early PD. Studies with higher sample sizes are warranted to confirm the results of our study.
Collapse
Affiliation(s)
- Abhishek Lenka
- Department of Clinical Neurosciences National Institute of Mental Health and Neurosciences Bangalore Karnataka India.,Department of Neurology National Institute of Mental Health and Neurosciences Bangalore Karnataka India
| | - Shaik Afsar Pasha
- Department of Neurology National Institute of Mental Health and Neurosciences Bangalore Karnataka India
| | - Sandhya Mangalore
- Department of Neuroimaging and Interventional Radiology National Institute of Mental Health and Neurosciences Bangalore Karnataka India
| | - Lija George
- Department of Neurology National Institute of Mental Health and Neurosciences Bangalore Karnataka India
| | - Ketan Ramakant Jhunjhunwala
- Department of Clinical Neurosciences National Institute of Mental Health and Neurosciences Bangalore Karnataka India.,Department of Neurology National Institute of Mental Health and Neurosciences Bangalore Karnataka India
| | - Bhawani Shankar Bagepally
- Department of Clinical Neurosciences National Institute of Mental Health and Neurosciences Bangalore Karnataka India
| | - Rajini M Naduthota
- Department of Neurology National Institute of Mental Health and Neurosciences Bangalore Karnataka India
| | - Jitender Saini
- Department of Neuroimaging and Interventional Radiology National Institute of Mental Health and Neurosciences Bangalore Karnataka India
| | - Ravi Yadav
- Department of Neurology National Institute of Mental Health and Neurosciences Bangalore Karnataka India
| | - Pramod Kumar Pal
- Department of Neurology National Institute of Mental Health and Neurosciences Bangalore Karnataka India
| |
Collapse
|
23
|
de Wilde MC, Overk CR, Sijben JW, Masliah E. Meta-analysis of synaptic pathology in Alzheimer's disease reveals selective molecular vesicular machinery vulnerability. Alzheimers Dement 2016; 12:633-44. [PMID: 26776762 DOI: 10.1016/j.jalz.2015.12.005] [Citation(s) in RCA: 192] [Impact Index Per Article: 21.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2015] [Revised: 11/02/2015] [Accepted: 12/04/2015] [Indexed: 12/21/2022]
Abstract
INTRODUCTION Loss of synapses best correlates to cognitive deficits in Alzheimer's disease (AD) in which oligomeric neurotoxic species of amyloid-β appears to contribute synaptic pathology. Although a number of clinical pathologic studies have been performed with limited sample size, there are no systematic studies encompassing large samples. Therefore, we performed a meta-analysis study. METHODS We identified 417 publications reporting postmortem synapse and synaptic marker loss from AD patients. Two meta-analyses were performed using a single database of subselected publications and calculating the standard mean differences. RESULTS Meta-analysis confirmed synaptic loss in selected brain regions is an early event in AD pathogenesis. The second meta-analysis of 57 synaptic markers revealed that presynaptic makers were affected more than postsynaptic markers. DISCUSSION The present meta-analysis study showed a consistent synaptic loss across brain regions and that molecular machinery including endosomal pathways, vesicular assembly mechanisms, glutamate receptors, and axonal transport are often affected.
Collapse
Affiliation(s)
- Martijn C de Wilde
- Nutricia Advanced Medical Nutrition, Nutricia Research, Utrecht, The Netherlands
| | - Cassia R Overk
- Department of Neurosciences, University of California, San Diego, La Jolla, CA, USA
| | - John W Sijben
- Nutricia Advanced Medical Nutrition, Nutricia Research, Utrecht, The Netherlands
| | - Eliezer Masliah
- Department of Neurosciences, University of California, San Diego, La Jolla, CA, USA; Department of Pathology, University of California, San Diego, La Jolla, CA, USA.
| |
Collapse
|
24
|
Jadhav S, Cubinkova V, Zimova I, Brezovakova V, Madari A, Cigankova V, Zilka N. Tau-mediated synaptic damage in Alzheimer's disease. Transl Neurosci 2015; 6:214-226. [PMID: 28123806 PMCID: PMC4936631 DOI: 10.1515/tnsci-2015-0023] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2015] [Accepted: 10/04/2015] [Indexed: 12/16/2022] Open
Abstract
Synapses are the principal sites for chemical communication between neurons and are essential for performing the dynamic functions of the brain. In Alzheimer’s disease and related tauopathies, synapses are exposed to disease modified protein tau, which may cause the loss of synaptic contacts that culminate in dementia. In recent decades, structural, transcriptomic and proteomic studies suggest that Alzheimer’s disease represents a synaptic disorder. Tau neurofibrillary pathology and synaptic loss correlate well with cognitive impairment in these disorders. Moreover, regional distribution and the load of neurofibrillary lesions parallel the distribution of the synaptic loss. Several transgenic models of tauopathy expressing various forms of tau protein exhibit structural synaptic deficits. The pathological tau proteins cause the dysregulation of synaptic proteome and lead to the functional abnormalities of synaptic transmission. A large body of evidence suggests that tau protein plays a key role in the synaptic impairment of human tauopathies.
Collapse
Affiliation(s)
- Santosh Jadhav
- Institute of Neuroimmunology, Slovak Academy of Sciences, Centre of Excellence for Alzheimer's Disease and Related Disorders, Dubravska 9, 845 10 Bratislava, Slovak Republic
| | - Veronika Cubinkova
- Institute of Neuroimmunology, Slovak Academy of Sciences, Centre of Excellence for Alzheimer's Disease and Related Disorders, Dubravska 9, 845 10 Bratislava, Slovak Republic; Axon Neuroscience SE, Grosslingova 45, Bratislava, Slovak Republic
| | - Ivana Zimova
- Institute of Neuroimmunology, Slovak Academy of Sciences, Centre of Excellence for Alzheimer's Disease and Related Disorders, Dubravska 9, 845 10 Bratislava, Slovak Republic; Axon Neuroscience SE, Grosslingova 45, Bratislava, Slovak Republic
| | - Veronika Brezovakova
- Institute of Neuroimmunology, Slovak Academy of Sciences, Centre of Excellence for Alzheimer's Disease and Related Disorders, Dubravska 9, 845 10 Bratislava, Slovak Republic
| | - Aladar Madari
- Small animal clinic, University of Veterinary Medicine and Pharmacy, Komenskeho 73, Kosice, Slovak Republic
| | - Viera Cigankova
- Department of Anatomy, Histology and Physiology, University of Veterinary Medicine and Pharmacy, Komenskeho 73, 041 81 Kosice, Slovak Republic
| | - Norbert Zilka
- Institute of Neuroimmunology, Slovak Academy of Sciences, Centre of Excellence for Alzheimer's Disease and Related Disorders, Dubravska 9, 845 10 Bratislava, Slovak Republic; Axon Neuroscience SE, Grosslingova 45, Bratislava, Slovak Republic
| |
Collapse
|
25
|
Jadhav S, Katina S, Kovac A, Kazmerova Z, Novak M, Zilka N. Truncated tau deregulates synaptic markers in rat model for human tauopathy. Front Cell Neurosci 2015; 9:24. [PMID: 25755633 PMCID: PMC4337338 DOI: 10.3389/fncel.2015.00024] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2014] [Accepted: 01/14/2015] [Indexed: 01/04/2023] Open
Abstract
Synaptic failure and neurofibrillary degeneration are two major neuropathological substrates of cognitive dysfunction in Alzheimer’s disease (AD). Only a few studies have demonstrated a direct relationship between these two AD hallmarks. To investigate tau mediated synaptic injury we used rat model of tauopathy that develops extensive neurofibrillary pathology in the cortex. Using fractionation of cortical synapses, we identified an increase in endogenous rat tau isoforms in presynaptic compartment, and their mis-sorting to the postsynaptic density (PSD). Truncated transgenic tau was distributed in both compartments exhibiting specific phospho-pattern that was characteristic for each synaptic compartment. In the presynaptic compartment, truncated tau was associated with impairment of dynamic stability of microtubules which could be responsible for reduction of synaptic vesicles. In the PSD, truncated tau lowered the levels of neurofilaments. Truncated tau also significantly decreased the synaptic levels of Aβ40 but not Aβ42. These data show that truncated tau differentially deregulates synaptic proteome in pre- and postsynaptic compartments. Importantly, we show that alteration of Aβ can arise downstream of truncated tau pathology.
Collapse
Affiliation(s)
- Santosh Jadhav
- Institute of Neuroimmunology, Slovak Academy of Sciences Bratislava, Slovak Republic
| | - Stanislav Katina
- Axon Neuroscience GmbH Bratislava, Slovak Republic ; Institute of Mathematics and Statistics, Masaryk University Brno, Czech Republic
| | - Andrej Kovac
- Axon Neuroscience GmbH Bratislava, Slovak Republic
| | - Zuzana Kazmerova
- Institute of Neuroimmunology, Slovak Academy of Sciences Bratislava, Slovak Republic
| | - Michal Novak
- Institute of Neuroimmunology, Slovak Academy of Sciences Bratislava, Slovak Republic ; Axon Neuroscience GmbH Bratislava, Slovak Republic
| | - Norbert Zilka
- Institute of Neuroimmunology, Slovak Academy of Sciences Bratislava, Slovak Republic ; Axon Neuroscience GmbH Bratislava, Slovak Republic
| |
Collapse
|
26
|
Höllerhage M, Deck R, De Andrade A, Respondek G, Xu H, Rösler TW, Salama M, Carlsson T, Yamada ES, Gad El Hak SA, Goedert M, Oertel WH, Höglinger GU. Piericidin A aggravates Tau pathology in P301S transgenic mice. PLoS One 2014; 9:e113557. [PMID: 25437199 PMCID: PMC4249965 DOI: 10.1371/journal.pone.0113557] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2014] [Accepted: 10/29/2014] [Indexed: 11/30/2022] Open
Abstract
Objective The P301S mutation in exon 10 of the tau gene causes a hereditary tauopathy. While mitochondrial complex I inhibition has been linked to sporadic tauopathies. Piericidin A is a prototypical member of the group of the piericidins, a class of biologically active natural complex I inhibitors, isolated from streptomyces spp. with global distribution in marine and agricultural habitats. The aim of this study was to determine whether there is a pathogenic interaction of the environmental toxin piericidin A and the P301S mutation. Methods Transgenic mice expressing human tau with the P301S-mutation (P301S+/+) and wild-type mice at 12 weeks of age were treated subcutaneously with vehicle (N = 10 P301S+/+, N = 7 wild-type) or piericidin A (N = 9 P301S+/+, N = 9 wild-type mice) at a dose of 0.5 mg/kg/d for a period of 28 days via osmotic minipumps. Tau pathology was measured by stereological counts of cells immunoreative with antibodies against phosphorylated tau (AD2, AT8, AT180, and AT100) and corresponding Western blot analysis. Results Piericidin A significantly increased the number of phospho-tau immunoreactive cells in the cerebral cortex in P301S+/+ mice, but only to a variable and mild extent in wild-type mice. Furthermore, piericidin A led to increased levels of pathologically phosphorylated tau only in P301S+/+ mice. While we observed no apparent cell loss in the frontal cortex, the synaptic density was reduced by piericidin A treatment in P301S+/+ mice. Discussion This study shows that exposure to piericidin A aggravates the course of genetically determined tau pathology, providing experimental support for the concept of gene-environment interaction in the etiology of tauopathies.
Collapse
Affiliation(s)
- Matthias Höllerhage
- Dept. of Neurology, Philipps-Universität, Marburg, Germany
- German Center for Neurodegenerative Diseases, Dept. for Translational Neurodegeneration, Munich, Germany
- Department of Neurology, Technische Universität München, Munich, Germany
| | - Roman Deck
- Dept. of Neurology, Philipps-Universität, Marburg, Germany
| | - Anderson De Andrade
- Dept. of Neurology, Philipps-Universität, Marburg, Germany
- German Center for Neurodegenerative Diseases, Dept. for Translational Neurodegeneration, Munich, Germany
| | - Gesine Respondek
- Dept. of Neurology, Philipps-Universität, Marburg, Germany
- German Center for Neurodegenerative Diseases, Dept. for Translational Neurodegeneration, Munich, Germany
- Department of Neurology, Technische Universität München, Munich, Germany
| | - Hong Xu
- Dept. of Neurology, Philipps-Universität, Marburg, Germany
- German Center for Neurodegenerative Diseases, Dept. for Translational Neurodegeneration, Munich, Germany
| | - Thomas W. Rösler
- Dept. of Neurology, Philipps-Universität, Marburg, Germany
- German Center for Neurodegenerative Diseases, Dept. for Translational Neurodegeneration, Munich, Germany
| | - Mohamed Salama
- Dept. of Neurology, Philipps-Universität, Marburg, Germany
- Department of Toxicology, Mansoura University, Mansoura, Egypt
| | - Thomas Carlsson
- Dept. of Neurology, Philipps-Universität, Marburg, Germany
- Department of Pharmacology, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Elizabeth S. Yamada
- Dept. of Neurology, Philipps-Universität, Marburg, Germany
- Experimental Neuropathology Laboratory, Federal University of Pará, Belém, Brazil
| | | | - Michel Goedert
- Division of Neurobiology, University of Cambridge, Cambridge, United Kingdom
| | | | - Günter U. Höglinger
- Dept. of Neurology, Philipps-Universität, Marburg, Germany
- German Center for Neurodegenerative Diseases, Dept. for Translational Neurodegeneration, Munich, Germany
- Department of Neurology, Technische Universität München, Munich, Germany
- * E-mail:
| |
Collapse
|
27
|
Magerova H, Vyhnalek M, Laczo J, Andel R, Rektorova I, Kadlecova A, Bojar M, Hort J. Odor identification in frontotemporal lobar degeneration subtypes. Am J Alzheimers Dis Other Demen 2014; 29:762-8. [PMID: 24939002 PMCID: PMC10852957 DOI: 10.1177/1533317514539033] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/03/2024]
Abstract
Odor identification impairment is a feature of several neurodegenerative disorders. Although neurodegenerative changes in the frontotemporal lobar degeneration (FTLD) subtypes involve areas important for olfactory processing, data on olfactory function in these patients are limited. An 18-item, multiple-choice odor identification test developed at our memory clinic, the Motol Hospital smell test, was administered to 9 patients with behavioral variant frontotemporal dementia, 13 patients with the language variants, primary nonfluent aphasia (n = 7) and semantic dementia (n = 6), and 8 patients with progressive supranuclear palsy. Compared to the control group (n = 15), all FTLD subgroups showed significant impairment of odor identification (P < .05). The differences between the FTLD subgroups were not significant. No correlation between odor identification and neuropsychological tests results was found. Our data suggest that odor identification impairment is a symptom common to FTLD syndromes, and it seems to be based on olfactory structure damage rather than cognitive decline.
Collapse
Affiliation(s)
- Hana Magerova
- Department of Neurology, Memory Clinic, 2nd Faculty of Medicine and Motol University Hospital, Charles University in Prague, Prague, Czech Republic
| | - Martin Vyhnalek
- Department of Neurology, Memory Clinic, 2nd Faculty of Medicine and Motol University Hospital, Charles University in Prague, Prague, Czech Republic International Clinical Research Center, St Anne's University Hospital Brno, Brno, Czech Republic
| | - Jan Laczo
- Department of Neurology, Memory Clinic, 2nd Faculty of Medicine and Motol University Hospital, Charles University in Prague, Prague, Czech Republic International Clinical Research Center, St Anne's University Hospital Brno, Brno, Czech Republic
| | - Ross Andel
- University of South Florida, School of Aging Studies, Tampa, FL, USA
| | - Irena Rektorova
- First Department of Neurology, School of Medicine and St Anne's Hospital, Masaryk University, Brno, Czech Republic Applied Neurosciences Research Group, CEITEC, Masaryk University, Brno, Czech Republic
| | - Alexandra Kadlecova
- Department of Neurology, Memory Clinic, 2nd Faculty of Medicine and Motol University Hospital, Charles University in Prague, Prague, Czech Republic
| | - Martin Bojar
- Department of Neurology, Memory Clinic, 2nd Faculty of Medicine and Motol University Hospital, Charles University in Prague, Prague, Czech Republic
| | - Jakub Hort
- Department of Neurology, Memory Clinic, 2nd Faculty of Medicine and Motol University Hospital, Charles University in Prague, Prague, Czech Republic International Clinical Research Center, St Anne's University Hospital Brno, Brno, Czech Republic
| |
Collapse
|
28
|
Ling H, Ling H, de Silva R, Massey LA, Courtney R, Hondhamuni G, Bajaj N, Lowe J, Holton JL, Lees A, Revesz T. Characteristics of progressive supranuclear palsy presenting with corticobasal syndrome: a cortical variant. Neuropathol Appl Neurobiol 2014; 40:149-63. [PMID: 23432126 PMCID: PMC4260147 DOI: 10.1111/nan.12037] [Citation(s) in RCA: 67] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2013] [Accepted: 02/15/2013] [Indexed: 12/11/2022]
Abstract
Aims Since the first description of the classical presentation of progressive supranuclear palsy (PSP) in 1963, now known as Richardson's syndrome (PSP-RS), several distinct clinical syndromes have been associated with PSP-tau pathology. Like other neurodegenerative disorders, the severity and distribution of phosphorylated tau pathology are closely associated with the clinical heterogeneity of PSP variants. PSP with corticobasal syndrome presentation (PSP-CBS) was reported to have more tau load in the mid-frontal and inferior-parietal cortices than in PSP-RS. However, it is uncertain if differences exist in the distribution of tau pathology in other brain regions or if the overall tau load is increased in the brains of PSP-CBS. Methods We sought to compare the clinical and pathological features of PSP-CBS and PSP-RS including quantitative assessment of tau load in 15 cortical, basal ganglia and cerebellar regions. Results In addition to the similar age of onset and disease duration, we demonstrated that the overall severity of tau pathology was the same between PSP-CBS and PSP-RS. We identified that there was a shift of tau burden towards the cortical regions away from the basal ganglia; supporting the notion that PSP-CBS is a ‘cortical’ PSP variant. PSP-CBS also had less severe neuronal loss in the dorsolateral and ventrolateral subregions of the substantia nigra and more severe microglial response in the corticospinal tract than in PSP-RS; however, neuronal loss in subthalamic nucleus was equally severe in both groups. Conclusions A better understanding of the factors that influence the selective pathological vulnerability in different PSP variants will provide further insights into the neurodegenerative process underlying tauopathies.
Collapse
Affiliation(s)
| | - H Ling
- Reta Lila Weston Institute of Neurological Studies, Institute of Neurology, University College London, London, UK; Queen Square Brain Bank for Neurological Disorders, Institute of Neurology, University College London, London, UK; Sara Koe PSP Research Centre, Institute of Neurology, University College London, London, UK
| | | | | | | | | | | | | | | | | | | |
Collapse
|
29
|
Scheff SW, Neltner JH, Nelson PT. Is synaptic loss a unique hallmark of Alzheimer's disease? Biochem Pharmacol 2014; 88:517-28. [PMID: 24412275 DOI: 10.1016/j.bcp.2013.12.028] [Citation(s) in RCA: 91] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2013] [Revised: 12/29/2013] [Accepted: 12/30/2013] [Indexed: 12/13/2022]
Abstract
Synapses may represent a key nidus for dementia including Alzheimer's disease (AD) pathogenesis. Here we review published studies and present new ideas related to the question of the specificity of synapse loss in AD. Currently, AD is defined by the regional presence of neuritic plaques and neurofibrillary tangles in the brain. The severity of involvement by those pathological hallmarks tends to correlate both with antemortem cognitive status, and also with synapse loss in multiple brain areas. Recent studies from large autopsy series have led to a new standard of excellence with regard to clinical-pathological correlation and to improved comprehension of the numerous brain diseases of the elderly. These studies have provided evidence that it is the rule rather than the exception for brains of aged individuals to demonstrate pathologies (often multiple) other than AD plaques and tangles. For many of these comorbid pathologies, the extent of synapse loss is imperfectly understood but could be substantial. These findings indicate that synapse loss is probably not a hallmark specific to AD but rather a change common to many diseases associated with dementia.
Collapse
Affiliation(s)
- Stephen W Scheff
- Department of Anatomy and Neurobiology, University of Kentucky Medical Center, University of Kentucky, Lexington, KY 40536, United States; Sanders-Brown Center on Aging and Alzheimer's Disease Center, University of Kentucky Medical Center, University of Kentucky, Lexington, KY 40536, United States.
| | - Janna H Neltner
- Department of Pathology and Division of Neuropathology, University of Kentucky Medical Center, University of Kentucky, Lexington, KY 40536, United States
| | - Peter T Nelson
- Sanders-Brown Center on Aging and Alzheimer's Disease Center, University of Kentucky Medical Center, University of Kentucky, Lexington, KY 40536, United States; Department of Pathology and Division of Neuropathology, University of Kentucky Medical Center, University of Kentucky, Lexington, KY 40536, United States.
| |
Collapse
|
30
|
Chapados C, Petrides M. Impairment only on the fluency subtest of the Frontal Assessment Battery after prefrontal lesions. Brain 2013; 136:2966-78. [DOI: 10.1093/brain/awt228] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023] Open
|
31
|
Nagao S, Yokota O, Nanba R, Takata H, Haraguchi T, Ishizu H, Ikeda C, Takeda N, Oshima E, Sakane K, Terada S, Ihara Y, Uchitomi Y. Progressive supranuclear palsy presenting as primary lateral sclerosis but lacking parkinsonism, gaze palsy, aphasia, or dementia. J Neurol Sci 2012; 323:147-53. [DOI: 10.1016/j.jns.2012.09.005] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2012] [Revised: 09/10/2012] [Accepted: 09/11/2012] [Indexed: 11/30/2022]
|
32
|
Lasagna-Reeves CA, Castillo-Carranza DL, Sengupta U, Clos AL, Jackson GR, Kayed R. Tau oligomers impair memory and induce synaptic and mitochondrial dysfunction in wild-type mice. Mol Neurodegener 2011; 6:39. [PMID: 21645391 PMCID: PMC3224595 DOI: 10.1186/1750-1326-6-39] [Citation(s) in RCA: 451] [Impact Index Per Article: 32.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2011] [Accepted: 06/06/2011] [Indexed: 01/22/2023] Open
Abstract
Background The correlation between neurofibrillary tangles of tau and disease progression in the brains of Alzheimer's disease (AD) patients remains an area of contention. Innovative data are emerging from biochemical, cell-based and transgenic mouse studies that suggest that tau oligomers, a pre-filament form of tau, may be the most toxic and pathologically significant tau aggregate. Results Here we report that oligomers of recombinant full-length human tau protein are neurotoxic in vivo after subcortical stereotaxic injection into mice. Tau oligomers impaired memory consolidation, whereas tau fibrils and monomers did not. Additionally, tau oligomers induced synaptic dysfunction by reducing the levels of synaptic vesicle-associated proteins synaptophysin and septin-11. Tau oligomers produced mitochondrial dysfunction by decreasing the levels of NADH-ubiquinone oxidoreductase (electron transport chain complex I), and activated caspase-9, which is related to the apoptotic mitochondrial pathway. Conclusions This study identifies tau oligomers as an acutely toxic tau species in vivo, and suggests that tau oligomers induce neurodegeneration by affecting mitochondrial and synaptic function, both of which are early hallmarks in AD and other tauopathies. These results open new avenues for neuroprotective intervention strategies of tauopathies by targeting tau oligomers.
Collapse
Affiliation(s)
- Cristian A Lasagna-Reeves
- George P, and Cynthia Woods Mitchell Center for Neurodegenerative Diseases, Departments of Neurology, and Neuroscience and Cell Biology, The University of Texas Medical Branch, Galveston, TX 77555-1045, USA.
| | | | | | | | | | | |
Collapse
|
33
|
De Vos A, Anandhakumar J, Van den Brande J, Verduyckt M, Franssens V, Winderickx J, Swinnen E. Yeast as a model system to study tau biology. Int J Alzheimers Dis 2011; 2011:428970. [PMID: 21559193 PMCID: PMC3090044 DOI: 10.4061/2011/428970] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2010] [Accepted: 01/21/2011] [Indexed: 11/20/2022] Open
Abstract
Hyperphosphorylated and aggregated human protein tau constitutes a hallmark of a multitude of neurodegenerative diseases called tauopathies, exemplified by Alzheimer's disease. In spite of an enormous amount of research performed on tau biology, several crucial questions concerning the mechanisms of tau toxicity remain unanswered. In this paper we will highlight some of the processes involved in tau biology and pathology, focusing on tau phosphorylation and the interplay with oxidative stress. In addition, we will introduce the development of a human tau-expressing yeast model, and discuss some crucial results obtained in this model, highlighting its potential in the elucidation of cellular processes leading to tau toxicity.
Collapse
Affiliation(s)
- Ann De Vos
- Laboratory of Functional Biology, Catholic University of Leuven, Kasteelpark Arenberg 31, 3001 Heverlee, Belgium
| | - Jayamani Anandhakumar
- Laboratory of Functional Biology, Catholic University of Leuven, Kasteelpark Arenberg 31, 3001 Heverlee, Belgium
| | - Jeff Van den Brande
- Laboratory of Functional Biology, Catholic University of Leuven, Kasteelpark Arenberg 31, 3001 Heverlee, Belgium
| | - Mathias Verduyckt
- Laboratory of Functional Biology, Catholic University of Leuven, Kasteelpark Arenberg 31, 3001 Heverlee, Belgium
| | - Vanessa Franssens
- Laboratory of Functional Biology, Catholic University of Leuven, Kasteelpark Arenberg 31, 3001 Heverlee, Belgium
| | - Joris Winderickx
- Laboratory of Functional Biology, Catholic University of Leuven, Kasteelpark Arenberg 31, 3001 Heverlee, Belgium
| | - Erwin Swinnen
- Laboratory of Functional Biology, Catholic University of Leuven, Kasteelpark Arenberg 31, 3001 Heverlee, Belgium
| |
Collapse
|
34
|
|
35
|
Tong M, Longato L, de la Monte SM. Early limited nitrosamine exposures exacerbate high fat diet-mediated type 2 diabetes and neurodegeneration. BMC Endocr Disord 2010; 10:4. [PMID: 20302640 PMCID: PMC3161394 DOI: 10.1186/1472-6823-10-4] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/13/2009] [Accepted: 03/19/2010] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND Type 2 diabetes mellitus (T2DM) and several types of neurodegeneration, including Alzheimer's, are linked to insulin-resistance, and chronic high dietary fat intake causes T2DM with mild neurodegeneration. Intra-cerebral Streptozotocin, a nitrosamine-related compound, causes neurodegeneration, whereas peripheral treatment causes DM. HYPOTHESIS Limited early exposures to nitrosamines that are widely present in the environment, enhance the deleterious effects of high fat intake in promoting T2DM and neurodegeneration. METHODS Long Evans rat pups were treated with N-nitrosodiethylamine (NDEA) by i.p. injection, and upon weaning, they were fed with high fat (60%; HFD) or low fat (5%; LFD) chow for 8 weeks. Cerebella were harvested to assess gene expression, and insulin and insulin-like growth factor (IGF) deficiency and resistance in the context of neurodegeneration. RESULTS HFD +/- NDEA caused T2DM, neurodegeneration with impairments in brain insulin, insulin receptor, IGF-2 receptor, or insulin receptor substrate gene expression, and reduced expression of tau and choline acetyltransferase (ChAT), which are regulated by insulin and IGF-1. In addition, increased levels of 4-hydroxynonenal and nitrotyrosine were measured in cerebella of HFD +/- NDEA treated rats, and overall, NDEA+HFD treatment reduced brain levels of Tau, phospho-GSK-3beta (reflecting increased GSK-3beta activity), glial fibrillary acidic protein, and ChAT to greater degrees than either treatment alone. Finally, pro-ceramide genes, examined because ceramides cause insulin resistance, oxidative stress, and neurodegeneration, were significantly up-regulated by HFD and/or NDEA exposure, but the highest levels were generally present in brains of HFD+NDEA treated rats. CONCLUSIONS Early limited exposure to nitrosamines exacerbates the adverse effects of later chronic high dietary fat intake in promoting T2DM and neurodegeneration. The mechanism involves increased generation of ceramides and probably other toxic lipids in brain.
Collapse
Affiliation(s)
- Ming Tong
- Liver Research Center, Rhode Island Hospital, 55 Claverick Street, Providence, RI 02903, USA
- Warren Alpert Medical School of Brown University, Box G, 97 Waterman Street, Providence, RI 02912, USA
| | - Lisa Longato
- Liver Research Center, Rhode Island Hospital, 55 Claverick Street, Providence, RI 02903, USA
- Pathobiology Program, Brown University, Box G, 222 Richmond Street, Providence, RI 02903, USA
| | - Suzanne M de la Monte
- Department of Pathology (Neuropathology), Rhode Island Hospital, 593 Eddy Street, Providence, RI 02903, USA
- Department of Neurology, Rhode Island Hospital, 593 Eddy Street, Providence, RI 02903, USA
- Liver Research Center, Rhode Island Hospital, 55 Claverick Street, Providence, RI 02903, USA
- Pathobiology Program, Brown University, Box G, 222 Richmond Street, Providence, RI 02903, USA
- Warren Alpert Medical School of Brown University, Box G, 97 Waterman Street, Providence, RI 02912, USA
| |
Collapse
|
36
|
Gendron TF, Petrucelli L. The role of tau in neurodegeneration. Mol Neurodegener 2009; 4:13. [PMID: 19284597 PMCID: PMC2663562 DOI: 10.1186/1750-1326-4-13] [Citation(s) in RCA: 333] [Impact Index Per Article: 20.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2009] [Accepted: 03/11/2009] [Indexed: 01/31/2023] Open
Abstract
Since the identification of tau as the main component of neurofibrillary tangles in Alzheimer's disease and related tauopathies, and the discovery that mutations in the tau gene cause frontotemporal dementia, much effort has been directed towards determining how the aggregation of tau into fibrillar inclusions causes neuronal death. As evidence emerges that tau-mediated neuronal death can occur even in the absence of tangle formation, a growing number of studies are focusing on understanding how abnormalities in tau (e.g. aberrant phosphorylation, glycosylation or truncation) confer toxicity. Though data obtained from experimental models of tauopathies strongly support the involvement of pathologically modified tau and tau aggregates in neurodegeneration, the exact neurotoxic species remain unclear, as do the mechanism(s) by which they cause neuronal death. Nonetheless, it is believed that tau-mediated neurodegeneration is likely to result from a combination of toxic gains of function as well as from the loss of normal tau function. To truly appreciate the detrimental consequences of aberrant tau function, a better understanding of all functions carried out by tau, including but not limited to the role of tau in microtubule assembly and stabilization, is required. This review will summarize what is currently known regarding the involvement of tau in the initiation and development of neurodegeneration in tauopathies, and will also highlight some of the remaining questions in need of further investigation.
Collapse
Affiliation(s)
- Tania F Gendron
- Department of Neuroscience, Mayo Clinic College of Medicine, Jacksonville, Florida, USA.
| | | |
Collapse
|
37
|
Morita Y, Osaki Y, Doi Y. Transcranial magnetic stimulation for differential diagnostics in patients with parkinsonism. Acta Neurol Scand 2008; 118:159-63. [PMID: 18279482 DOI: 10.1111/j.1600-0404.2007.00988.x] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
OBJECTIVE We investigated transcranial magnetic stimulation (TMS) parameters in patients with parkinsonism, particularly in the early stages of the disease. SUBJECTS AND METHODS We performed TMS in 48 patients with PD, progressive supranuclear palsy (PSP) and multiple system atrophy (MSA). We measured motor threshold (MT), latency (L), motor-evoked potential amplitude and central motor conduction time (CMCT) and cortical silent period (CSP). Furthermore, we selected and compared 27 patients with a disease duration of less than 3 years. RESULTS CMCT, MT, L and CSP were different among the three groups. Post hoc analyses revealed that CMCT and CSP were the shortest in PD, and that MT was significantly lower in PD than in MSA. In patients whose disease duration was less than 3 years, CMCT and CSP were different among the three groups. Post hoc analyses showed significantly shorter CMCT in PD. CONCLUSIONS TMS can detect the pathophysiological difference among the groups in the early stages of the disease.
Collapse
Affiliation(s)
- Y Morita
- Department of Geriatrics, Cardiology and Neurology, Kochi Medical School, Nankoku, Japan.
| | | | | |
Collapse
|
38
|
Yoshiyama Y, Higuchi M, Zhang B, Huang SM, Iwata N, Saido TC, Maeda J, Suhara T, Trojanowski JQ, Lee VMY. Synapse loss and microglial activation precede tangles in a P301S tauopathy mouse model. Neuron 2007; 53:337-51. [PMID: 17270732 DOI: 10.1016/j.neuron.2007.01.010] [Citation(s) in RCA: 1594] [Impact Index Per Article: 88.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2006] [Revised: 12/06/2006] [Accepted: 01/10/2007] [Indexed: 11/21/2022]
Abstract
Filamentous tau inclusions are hallmarks of Alzheimer's disease (AD) and related tauopathies, but earlier pathologies may herald disease onset. To investigate this, we studied wild-type and P301S mutant human tau transgenic (Tg) mice. Filamentous tau lesions developed in P301S Tg mice at 6 months of age, and progressively accumulated in association with striking neuron loss as well as hippocampal and entorhinal cortical atrophy by 9-12 months of age. Remarkably, hippocampal synapse loss and impaired synaptic function were detected in 3 month old P301S Tg mice before fibrillary tau tangles emerged. Prominent microglial activation also preceded tangle formation. Importantly, immunosuppression of young P301S Tg mice with FK506 attenuated tau pathology and increased lifespan, thereby linking neuroinflammation to early progression of tauopathies. Thus, hippocampal synaptic pathology and microgliosis may be the earliest manifestations of neurodegenerative tauopathies, and abrogation of tau-induced microglial activation could retard progression of these disorders.
Collapse
Affiliation(s)
- Yasumasa Yoshiyama
- The Center for Neurodegenerative Disease Research, Department of Pathology and Laboratory Medicine, University of Pennsylvania School of Medicine, Philadelphia, PA 19104, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Katsuse O, Lin WL, Lewis J, Hutton ML, Dickson DW. Neurofibrillary tangle-related synaptic alterations of spinal motor neurons of P301L tau transgenic mice. Neurosci Lett 2006; 409:95-9. [PMID: 17010516 DOI: 10.1016/j.neulet.2006.09.021] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2006] [Revised: 09/08/2006] [Accepted: 09/09/2006] [Indexed: 11/30/2022]
Abstract
We investigated axosomatic synapses of anterior horn cells of transgenic (TG) mice expressing mutant P301L human tau and non-transgenic (NTG) mice using electron microscopic methods to demonstrate the relationship between neurofibrillary tangles (NFTs) and synaptic alterations. Animals aged 3.5-8.5 months were used because at this age many motor neurons in TG mice have NFTs. We measured the perimeter of anterior horn cell perikarya, the number of boutons and total length of boutons in contact with the neuronal perikarya from the micrographs of NFT and non-NFT-bearing neurons. We also calculated the proportion of the perimeter covered by boutons, density of boutons and mean size of boutons. The density of synaptic boutons in contact with NFT-bearing neurons was significantly decreased compared to non-NFT-bearing neurons. These findings suggest that synaptic reduction occurs during neurofibrillary degeneration and is probably associated with NFT. In addition, synaptic boutons were detached from NFT-bearing neurons with the resulting space occupied by astrocytic processes, suggesting that astrocytes may be involved in the observed synaptic alterations.
Collapse
Affiliation(s)
- Omi Katsuse
- Department of Neuroscience, Mayo Clinic College of Medicine, Jacksonville, FL 32224, USA
| | | | | | | | | |
Collapse
|
40
|
Halliday GM, Macdonald V, Henderson JM. A comparison of degeneration in motor thalamus and cortex between progressive supranuclear palsy and Parkinson's disease. ACTA ACUST UNITED AC 2005; 128:2272-80. [PMID: 16014651 DOI: 10.1093/brain/awh596] [Citation(s) in RCA: 86] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
Changes in motor cortical activation are associated with the major symptoms observed in both Parkinson's disease and progressive supranuclear palsy (PSP). While research has concentrated on basal ganglia abnormalities as central to these cortical changes, several studies in both disorders have shown pathology in the thalamus and motor cortices. In particular, we recently reported an 88% loss of corticocortical projection neurones in the pre-supplementary motor (pre-SMA) cortex in Parkinson's disease. Further analysis of the degree of neuronal loss and pathology in motor cortices and their thalamocortical relays in Parkinson's disease and PSP is warranted. Six cases with PSP, nine cases with Parkinson's disease and nine controls were selected from a prospectively studied brain donor cohort. alpha-Synuclein, ubiquitin and tau immunohistochemistry were used to identify pathological lesions. Unbiased stereological methods were used to analyse atrophy and neuronal loss in the motor thalamus [ventral anterior, ventrolateral anterior and ventrolateral posterior (VLp) nuclei] and motor cortices (primary motor, dorsolateral premotor and pre-SMA cortices). Analysis of variance and post hoc testing was used to determine differences between groups. In Parkinson's disease, the motor thalamus and motor cortices (apart from the pre-SMA) were preserved containing only rare alpha-synuclein-positive and ubiquitin-positive Lewy bodies. In contrast, patients with PSP had significant atrophy and neuronal loss in VLp (22 and 30%, respectively), pre-SMA (21 and 51%, respectively) and primary motor cortices (33 and 54%, respectively). In the primary motor cortex of PSP cases, neuronal loss was confined to inhibitory interneurones, whereas in the pre-SMA both interneurones (reduced by 26%) and corticocortical projection neurones (reduced by 82%) were affected. Tau-positive neurofibrillary and glial tangles were observed throughout the motor thalamus and motor cortices in PSP. These non-dopaminergic lesions in motor circuits are likely to contribute to the pathogenesis of both PSP and Parkinson's disease. The selective involvement of the VLp and primary motor cortex in PSP implicates these cerebellothalamocortical pathways as differentiating this disease, possibly contributing to the early falls.
Collapse
Affiliation(s)
- Glenda M Halliday
- Prince of Wales Medical Research Institute and University of New South Wales, Sydney, Australia.
| | | | | |
Collapse
|
41
|
Rippon GA, Boeve BF, Parisi JE, Dickson DW, Ivnik RI, Jack CR, Hutton M, Baker M, Josephs KA, Knopman DS, Petersen RC. Late-onset frontotemporal dementia associated with progressive supranuclear palsy/argyrophilic grain disease/Alzheimer's disease pathology. Neurocase 2005; 11:204-11. [PMID: 16006341 DOI: 10.1080/13554790590944753] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Abstract
Progressive supranuclear palsy (PSP) is typically manifested by vertical supranuclear gaze palsy, frequent falls early in the disease course, axial rigidity and poor response to levodopa. Prominent anterograde memory dysfunction with subsequent impairment in other cognitive domains is characteristic of Alzheimer's disease (AD). No clear clinical syndrome has been identified in argyrophilic grain disease (AGD). Frontotemporal dementia (FTD) is characterized by apathy, emotional blunting, disinhibition, and impairment in executive functioning despite relatively preserved memory and visuospatial abilities. Cognitive deficits are known to occur in PSP; however, overt clinical FTD without parkinsonism or supranuclear gaze palsy associated with PSP pathology has rarely been documented. We report an elderly patient with the typical clinical, neuropsychometric, and neuroimaging features of FTD who had autopsy findings most consistent with PSP plus AGD and AD in limbic structures. We suggest that PSP with or without coexisting AD and AGD be included in the differential diagnosis of patients presenting with FTD.
Collapse
Affiliation(s)
- G A Rippon
- Department of Neurology, Mayo Clinic, Rochester, Minnesota 55905, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Kühn AA, Grosse P, Holtz K, Brown P, Meyer BU, Kupsch A. Patterns of abnormal motor cortex excitability in atypical parkinsonian syndromes. Clin Neurophysiol 2004; 115:1786-95. [PMID: 15261857 DOI: 10.1016/j.clinph.2004.03.020] [Citation(s) in RCA: 66] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/22/2004] [Indexed: 11/20/2022]
Abstract
OBJECTIVE Multiple system atrophy (MSA), progressive supranuclear palsy (PSP), and corticobasal-ganglionic degeneration (CBGD) are all clinically characterized by an akinetic-rigid syndrome together with a variety of additional signs. We hypothesised that these atypical parkinsonian syndromes (APS) will show distinctive patterns in their motor output upon transcranial magnetic stimulation (TMS) due to their different underlying anatomico-functional deficits. METHODS We performed single and paired-pulse TMS and assessed inhibitory and excitatory response parameters from the first dorsal interosseus muscles in 13 patients with MSA, 18 with PSP, 13 with CBGD, 15 patients with Parkinson's disease and 17 healthy subjects. RESULTS PSP and MSA patients had significantly enlarged response amplitudes at rest, reduced intracortical inhibition (ICI) and prolonged ipsi- and contralateral silent periods, whereas CBGD patients showed significantly increased motor thresholds, smaller response amplitudes at rest, shortened contralateral silent period, reduced transcallosal inhibition and a reduced ICI. In 22% of APS patients ipsilateral motor responses occurred in upper limb muscles irrespective of the underlying disease. CONCLUSIONS Our results indicate that motor cortex disinhibition is predominant in patients with PSP and MSA. In CBGD more severe neuronal cell loss in the motor cortex itself may lead to hypoexcitability of corticospinal and transcallosal pathways.
Collapse
Affiliation(s)
- A A Kühn
- Department of Neurology, Charité, Humboldt University Berlin, 13353 Berlin, Augustenburger Platz 1, Germany.
| | | | | | | | | | | |
Collapse
|
43
|
Siew LK, Love S, Dawbarn D, Wilcock GK, Allen SJ. Measurement of pre- and post-synaptic proteins in cerebral cortex: effects of post-mortem delay. J Neurosci Methods 2004; 139:153-9. [PMID: 15488227 DOI: 10.1016/j.jneumeth.2004.04.020] [Citation(s) in RCA: 39] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2004] [Revised: 04/20/2004] [Accepted: 04/22/2004] [Indexed: 10/26/2022]
Abstract
Assessments of synaptic density in human brain are often based on measurements of synaptic proteins. Little information is available on their post-mortem stability. We have investigated this by ELISAs of the pre-synaptic proteins syntaxin and synaptophysin, and the post-synaptic protein PSD-95, in rat and human cortex. The rat brains were cooled in situ from 37 to 20 or 4 degrees C over 3 h, and then kept at 20 or 4 degrees C for a further 24-72 h, to simulate post-mortem storage at room temperature or in a mortuary refrigerator. Synaptophysin and PSD-95 levels in rat cerebral cortex were not significantly decreased after 72 h of incubation at 20 degrees C. Syntaxin was stable for 24 h but decreased by 39-44% at 48-72 h. Storage at 4 degrees C resulted in a similar reduction of syntaxin levels over 72 h. In human brain tissue from 160 people aged 24-102 years, post-mortem delay had little effect on synaptic protein levels in superior temporal cortex, but was associated with a decline in PSD-95 and syntaxin in mid-frontal cortex after 24 h. The more robust stability of synaptophysin may be related to its multi-transmembrane structure.
Collapse
Affiliation(s)
- Lai Khai Siew
- Henry Wellcome Laboratories for Integrative Neuroscience and Endocrinology (Care of the Elderly), University of Bristol, Dorothy Hodgkin Building, Whitson Street, Bristol BS1 3NY, UK
| | | | | | | | | |
Collapse
|
44
|
Bigio EH, Hynan LS, Sontag E, Satumtira S, White CL. Synapse loss is greater in presenile than senile onset Alzheimer disease: implications for the cognitive reserve hypothesis. Neuropathol Appl Neurobiol 2002; 28:218-27. [PMID: 12060346 DOI: 10.1046/j.1365-2990.2002.00385.x] [Citation(s) in RCA: 58] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
In the past, 'Alzheimer disease' (AD) referred to pathologic AD with clinical onset of dementia in the presenium, while 'senile dementia of the Alzheimer type' (SDAT) referred to senile onset AD. Because AD appears clinically homogeneous regardless of age of onset, the two subtypes in more recent years have not been distinguished. Pathologic differences have been noted, but synapse loss has not previously been compared between the two groups. Hypothesizing that synapse loss would be greater in presenile onset than senile onset AD, we compared synapse loss, as well as Alzheimer pathology in presenile and senile onset AD, using an ELISA method to quantify synaptophysin. Synaptophysin was significantly lower in presenile than senile AD in right frontal and bilateral parietal lobes. Neuritic plaque counts were significantly higher in presenile than senile AD in bilateral frontal and parietal lobes. Semi-quantitative evaluation of neurofibrillary tangles revealed significantly more tangles in bilateral frontal and parietal lobes in presenile than senile AD. Brain weight was significantly lower in presenile than senile AD. The differences in synapse loss and Alzheimer-type pathology in presenile and senile onset AD support the hypothesis that 'cognitive reserve' protects the human brain from neurodegenerative disease.
Collapse
Affiliation(s)
- Eileen H Bigio
- Department of Pathology, Northwestern University Medical School, Chicago, Illinois 60611, USA.
| | | | | | | | | |
Collapse
|
45
|
Callahan LM, Vaules WA, Coleman PD. Progressive reduction of synaptophysin message in single neurons in Alzheimer disease. J Neuropathol Exp Neurol 2002; 61:384-95. [PMID: 12025941 DOI: 10.1093/jnen/61.5.384] [Citation(s) in RCA: 42] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
The data presented here examine 2 hypotheses: 1) that viable but vulnerable single neurons remaining in the Alzheimer brain lose synaptic markers, and 2) that the extent of this loss is related to the disease state of these single neurons when disease state is defined by immunoreactivity. We used double immunohistochemistry (IHC) to define neurofibrillary tangle (NFT) and phosphorylation status of tau at selected defined epitopes. This double IHC was combined with quantitative in situ hybridization for message for the synaptic marker, synaptophysin, in 1,127 single hippocampal CA1 pyramidal neurons from 15 Alzheimer disease (AD) and 4 control cases. We found that there is a graded, progressive, decrease of synaptophysin message expressed by single neurons related to immunohistochemical markers of tau status, and that neurons in similar immunohistochemically defined classes show similar losses of synaptophysin message regardless of whether they were sampled from clinical control brains or advanced AD. The resulting conclusions are consistent with a suggestion that differences among clinically defined AD and control status are defined by the numbers of neurons in various disease states.
Collapse
Affiliation(s)
- Linda M Callahan
- Pathology and Laboratory Medicine in the Center for Aging and Developmental Biology, University of Rochester, New York 14642, USA
| | | | | |
Collapse
|