1
|
Tanaka H, Martinez-Valbuena I, Forrest SL, Couto B, Reyes NG, Morales-Rivero A, Lee S, Li J, Karakani AM, Tang-Wai DF, Tator C, Khadadadi M, Sadia N, Tartaglia MC, Lang AE, Kovacs GG. Distinct involvement of the cranial and spinal nerves in progressive supranuclear palsy. Brain 2024; 147:1399-1411. [PMID: 37972275 PMCID: PMC10994524 DOI: 10.1093/brain/awad381] [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: 08/10/2023] [Revised: 10/08/2023] [Accepted: 10/26/2023] [Indexed: 11/19/2023] Open
Abstract
The most frequent neurodegenerative proteinopathies include diseases with deposition of misfolded tau or α-synuclein in the brain. Pathological protein aggregates in the PNS are well-recognized in α-synucleinopathies and have recently attracted attention as a diagnostic biomarker. However, there is a paucity of observations in tauopathies. To characterize the involvement of the PNS in tauopathies, we investigated tau pathology in cranial and spinal nerves (PNS-tau) in 54 tauopathy cases [progressive supranuclear palsy (PSP), n = 15; Alzheimer's disease (AD), n = 18; chronic traumatic encephalopathy (CTE), n = 5; and corticobasal degeneration (CBD), n = 6; Pick's disease, n = 9; limbic-predominant neuronal inclusion body 4-repeat tauopathy (LNT), n = 1] using immunohistochemistry, Gallyas silver staining, biochemistry, and seeding assays. Most PSP cases revealed phosphorylated and 4-repeat tau immunoreactive tau deposits in the PNS as follows: (number of tau-positive cases/available cases) cranial nerves III: 7/8 (88%); IX/X: 10/11 (91%); and XII: 6/6 (100%); anterior spinal roots: 10/10 (100%). The tau-positive inclusions in PSP often showed structures with fibrillary (neurofibrillary tangle-like) morphology in the axon that were also recognized with Gallyas silver staining. CBD cases rarely showed fine granular non-argyrophilic tau deposits. In contrast, tau pathology in the PNS was not evident in AD, CTE and Pick's disease cases. The single LNT case also showed tau pathology in the PNS. In PSP, the severity of PNS-tau involvement correlated with that of the corresponding nuclei, although, occasionally, p-tau deposits were present in the cranial nerves but not in the related brainstem nuclei. Not surprisingly, most of the PSP cases presented with eye movement disorder and bulbar symptoms, and some cases also showed lower-motor neuron signs. Using tau biosensor cells, for the first time we demonstrated seeding capacity of tau in the PNS. In conclusion, prominent PNS-tau distinguishes PSP from other tauopathies. The morphological differences of PNS-tau between PSP and CBD suggest that the tau pathology in PNS could reflect that in the central nervous system. The high frequency and early presence of tau lesions in PSP suggest that PNS-tau may have clinical and biomarker relevance.
Collapse
Affiliation(s)
- Hidetomo Tanaka
- Department of Laboratory Medicine and Pathobiology and Tanz Centre for Research in Neurodegenerative Disease, University of Toronto, Toronto, Ontario M5T 0S8, Canada
| | - Ivan Martinez-Valbuena
- Department of Laboratory Medicine and Pathobiology and Tanz Centre for Research in Neurodegenerative Disease, University of Toronto, Toronto, Ontario M5T 0S8, Canada
| | - Shelley L Forrest
- Department of Laboratory Medicine and Pathobiology and Tanz Centre for Research in Neurodegenerative Disease, University of Toronto, Toronto, Ontario M5T 0S8, Canada
- Laboratory Medicine Program and Krembil Brain Institute, University Health Network, Toronto, Ontario M5T 0S8, Canada
| | - Blas Couto
- Edmond J. Safra Program in Parkinson's Disease, Rossy Program for PSP Research and the Morton and Gloria Shulman Movement Disorders Clinic, Toronto Western Hospital, Toronto, Ontario M5T 2S8, Canada
| | - Nikolai Gil Reyes
- Edmond J. Safra Program in Parkinson's Disease, Rossy Program for PSP Research and the Morton and Gloria Shulman Movement Disorders Clinic, Toronto Western Hospital, Toronto, Ontario M5T 2S8, Canada
| | - Alonso Morales-Rivero
- University Health Network Memory Clinic, Toronto Western Hospital, Toronto, Ontario M5T 2S8, Canada
| | - Seojin Lee
- Department of Laboratory Medicine and Pathobiology and Tanz Centre for Research in Neurodegenerative Disease, University of Toronto, Toronto, Ontario M5T 0S8, Canada
| | - Jun Li
- Department of Laboratory Medicine and Pathobiology and Tanz Centre for Research in Neurodegenerative Disease, University of Toronto, Toronto, Ontario M5T 0S8, Canada
| | - Ali M Karakani
- Department of Laboratory Medicine and Pathobiology and Tanz Centre for Research in Neurodegenerative Disease, University of Toronto, Toronto, Ontario M5T 0S8, Canada
| | - David F Tang-Wai
- University Health Network Memory Clinic, Toronto Western Hospital, Toronto, Ontario M5T 2S8, Canada
- Department of Medicine/Division of Neurology, University of Toronto, Toronto, Ontario M5S 3H2, Canada
- Krembil Brain Institute, Toronto Western Hospital, Toronto, Ontario M5T 2S8, Canada
| | - Charles Tator
- Krembil Brain Institute, Toronto Western Hospital, Toronto, Ontario M5T 2S8, Canada
- Canadian Concussion Centre, Krembil Brain Institute, University Health Network, Toronto, Ontario M5T 0S8, Canada
| | - Mozhgan Khadadadi
- Canadian Concussion Centre, Krembil Brain Institute, University Health Network, Toronto, Ontario M5T 0S8, Canada
| | - Nusrat Sadia
- Canadian Concussion Centre, Krembil Brain Institute, University Health Network, Toronto, Ontario M5T 0S8, Canada
| | - Maria Carmela Tartaglia
- University Health Network Memory Clinic, Toronto Western Hospital, Toronto, Ontario M5T 2S8, Canada
- Department of Medicine/Division of Neurology, University of Toronto, Toronto, Ontario M5S 3H2, Canada
- Krembil Brain Institute, Toronto Western Hospital, Toronto, Ontario M5T 2S8, Canada
- Canadian Concussion Centre, Krembil Brain Institute, University Health Network, Toronto, Ontario M5T 0S8, Canada
- Tanz Centre for Research in Neurodegenerative Diseases, University of Toronto, Toronto, Ontario M5T 0S8, Canada
| | - Anthony E Lang
- Edmond J. Safra Program in Parkinson's Disease, Rossy Program for PSP Research and the Morton and Gloria Shulman Movement Disorders Clinic, Toronto Western Hospital, Toronto, Ontario M5T 2S8, Canada
- Department of Medicine/Division of Neurology, University of Toronto, Toronto, Ontario M5S 3H2, Canada
- Krembil Brain Institute, Toronto Western Hospital, Toronto, Ontario M5T 2S8, Canada
| | - Gabor G Kovacs
- Department of Laboratory Medicine and Pathobiology and Tanz Centre for Research in Neurodegenerative Disease, University of Toronto, Toronto, Ontario M5T 0S8, Canada
- Department of Medicine/Division of Neurology, University of Toronto, Toronto, Ontario M5S 3H2, Canada
- Krembil Brain Institute, Toronto Western Hospital, Toronto, Ontario M5T 2S8, Canada
- Laboratory Medicine Program, University Health Network, Toronto, Ontario M5T 0S8, Canada
| |
Collapse
|
2
|
Chang K, Barrett A, Pham K, Troncoso JC. Lateral geniculate body is spared of tau pathology in Pick disease. J Neuropathol Exp Neurol 2024; 83:238-244. [PMID: 38412343 PMCID: PMC10951972 DOI: 10.1093/jnen/nlae016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/29/2024] Open
Abstract
The pathobiology of tau is of great importance for understanding the mechanisms of neurodegeneration in aging and age-associated disorders such as Alzheimer disease (AD) and frontotemporal dementias. It is critical to identify neuronal populations and brain regions that are vulnerable or resistant to tau pathological changes. Pick disease (PiD) is a three-repeat (3R) tauopathy that belongs to the group of frontotemporal lobar degenerations. The neuropathologic changes of PiD are characterized by globular tau-positive neuronal intracytoplasmic inclusions, called Pick bodies, in the granule cells of the dentate gyrus and frontal and temporal neocortices, and ballooned neurons, named Pick neurons, in the neocortex. In the present study, we examined 13 autopsy-confirmed cases of PiD. Using immunohistochemistry for phospho-tau (AT8) and 3R tau isoform, all PiD cases demonstrated extensive lesions involving the hippocampus and neocortex. However, the lateral geniculate body (LGB) is spared of significant tau lesions in contrast to the neighboring hippocampus and other thalamic nuclei. Only 1 PiD case (7.7%) had tau-positive neurons, and 4 cases had tau-positive neurites (31%) in the LGB. By contrast, the LGB does consistently harbor tau lesions in other tauopathies including progressive supranuclear palsy, corticobasal degeneration, and AD.
Collapse
Affiliation(s)
- Koping Chang
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
- Department and Graduate Institute of Pathology, National Taiwan University Hospital, National Taiwan University College of Medicine, Taipei, Taiwan
| | - Alexander Barrett
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Khoa Pham
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Juan C Troncoso
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| |
Collapse
|
3
|
Chen Y, Wang H, Wang B, Li W, Ye P, Xu W, Liu P, Chen X, Cen Z, Ouyang Z, Wu S, Dou X, Liao Y, Zhang H, Tian M, Luo W. Retinal Thinning as a Marker of Disease Severity in Progressive Supranuclear Palsy. J Mov Disord 2024; 17:55-63. [PMID: 37748923 PMCID: PMC10846978 DOI: 10.14802/jmd.23102] [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: 05/21/2023] [Revised: 08/18/2023] [Accepted: 09/25/2023] [Indexed: 09/27/2023] Open
Abstract
OBJECTIVE Progressive supranuclear palsy (PSP) involves a variety of visual symptoms that are thought to be partially caused by structural abnormalities of the retina. However, the relationship between retinal structural changes, disease severity, and intracranial alterations remains unknown. We investigated distinct retinal thinning patterns and their relationship with clinical severity and intracranial alterations in a PSP cohort. METHODS We enrolled 19 patients with PSP (38 eyes) and 20 age-matched healthy controls (40 eyes). All of the participants underwent peripapillary and macular optical coherence tomography. Brain 11C-2β-carbomethoxy-3β-(4-fluorophenyl) tropane (11C-CFT) and 18F-fluorodeoxyglucose (18F-FDG) positron emission tomography imaging were also performed in patients with PSP. We investigated the association between retinal thickness changes and clinical features, striatal dopamine transporter availability, and cerebral glucose metabolism. RESULTS The peripapillary retinal nerve fiber layer (pRNFL) and macula were significantly thinner in patients with PSP than in controls. The thickness of the superior sector of the pRNFL demonstrated a significant negative relationship with the Movement Disorder Society-Unified Parkinson's Disease Rating Scale part III and Hoehn and Yahr staging scale scores. A significant negative correlation was found between outer inferior macular thickness and disease duration. Outer temporal macular thickness was positively correlated with Montreal Cognitive Assessment scores. In PSP, lower outer temporal macular thickness was also positively correlated with decreased dopamine transporter binding in the caudate. CONCLUSION The pRNFL and macular thinning may be candidate markers for monitoring disease severity. Additionally, macular thinning may be an in vivo indicator of nigrostriatal dopaminergic cell degeneration in PSP patients.
Collapse
Affiliation(s)
- Yueting Chen
- Department of Neurology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Department of Ultrasound, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang, China
| | - Haotian Wang
- Department of Neurology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Bo Wang
- Department of Neurology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Wenbo Li
- Department of Eye Center, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Zhejiang Provincial Key Lab of Ophthalmology, Hangzhou, Zhejiang, China
| | - Panpan Ye
- Department of Eye Center, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Zhejiang Provincial Key Lab of Ophthalmology, Hangzhou, Zhejiang, China
| | - Wen Xu
- Department of Eye Center, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Zhejiang Provincial Key Lab of Ophthalmology, Hangzhou, Zhejiang, China
| | - Peng Liu
- Department of Neurology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Department of Neurology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Xinhui Chen
- Department of Neurology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Zhidong Cen
- Department of Neurology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Zhiyuan Ouyang
- Department of Neurology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Sheng Wu
- Department of Neurology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Xiaofeng Dou
- Department of Neurology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Yi Liao
- Department of Nuclear Medicine and PET-CT Center, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Hong Zhang
- Department of Nuclear Medicine and PET-CT Center, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Key Laboratory for Biomedical Engineering of Ministry of Education, Zhejiang University, Hangzhou, Zhejiang, China
- The College of Biomedical Engineering and Instrument Science, Zhejiang University, Hangzhou, Zhejiang, China
| | - Mei Tian
- Department of Nuclear Medicine and PET-CT Center, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Huashan Hospital and Human Phenome Institute, Fudan University, Shanghai, China
| | - Wei Luo
- Department of Neurology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| |
Collapse
|
4
|
Cheng X, Tang Y, Vidyadhara D, Li BZ, Zimmerman M, Pak A, Nareddula S, Edens PA, Chandra SS, Chubykin AA. Impaired pre-synaptic plasticity and visual responses in auxilin-knockout mice. iScience 2023; 26:107842. [PMID: 37766983 PMCID: PMC10520332 DOI: 10.1016/j.isci.2023.107842] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2023] [Revised: 08/06/2023] [Accepted: 09/04/2023] [Indexed: 09/29/2023] Open
Abstract
Auxilin (DNAJC6/PARK19), an endocytic co-chaperone, is essential for maintaining homeostasis in the readily releasable pool (RRP) by aiding clathrin-mediated uncoating of synaptic vesicles. Its loss-of-function mutations, observed in familial Parkinson's disease (PD), lead to basal ganglia motor deficits and cortical dysfunction. We discovered that auxilin-knockout (Aux-KO) mice exhibited impaired pre-synaptic plasticity in layer 4 to layer 2/3 pyramidal cell synapses in the primary visual cortex (V1), including reduced short-term facilitation and depression. Computational modeling revealed increased RRP refilling during short repetitive stimulation, which diminished during prolonged stimulation. Silicon probe recordings in V1 of Aux-KO mice demonstrated disrupted visual cortical circuit responses, including reduced orientation selectivity, compromised visual mismatch negativity, and shorter visual familiarity-evoked theta oscillations. Pupillometry analysis revealed an impaired optokinetic response. Auxilin-dependent pre-synaptic endocytosis dysfunction was associated with deficits in pre-synaptic plasticity, visual cortical functions, and eye movement prodromally or at the early stage of motor symptoms.
Collapse
Affiliation(s)
- Xi Cheng
- Department of Biological Sciences, Purdue Institute for Integrative Neuroscience, Purdue Autism Research Center, Purdue University, West Lafayette, IN 47907, USA
| | - Yu Tang
- Department of Biological Sciences, Purdue Institute for Integrative Neuroscience, Purdue Autism Research Center, Purdue University, West Lafayette, IN 47907, USA
| | - D.J. Vidyadhara
- Department of Neurology, Yale University, CT, USA
- Department of Neuroscience, Yale University, CT, USA
| | - Ben-Zheng Li
- Department of Physiology and Biophysics, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
- Department of Electrical Engineering, University of Colorado, Denver, Denver, CO, USA
| | - Michael Zimmerman
- Department of Biological Sciences, Purdue Institute for Integrative Neuroscience, Purdue Autism Research Center, Purdue University, West Lafayette, IN 47907, USA
- Department of Biomedical Engineering, Purdue University, West Lafayette, IN 47907, USA
| | - Alexandr Pak
- Department of Biological Sciences, Purdue Institute for Integrative Neuroscience, Purdue Autism Research Center, Purdue University, West Lafayette, IN 47907, USA
| | - Sanghamitra Nareddula
- Department of Biological Sciences, Purdue Institute for Integrative Neuroscience, Purdue Autism Research Center, Purdue University, West Lafayette, IN 47907, USA
| | - Paige Alyssa Edens
- Department of Biological Sciences, Purdue Institute for Integrative Neuroscience, Purdue Autism Research Center, Purdue University, West Lafayette, IN 47907, USA
| | - Sreeganga S. Chandra
- Department of Neurology, Yale University, CT, USA
- Department of Neuroscience, Yale University, CT, USA
- Program in Cellular Neuroscience, Neurodegeneration and Repair, Yale University, CT, USA
| | - Alexander A. Chubykin
- Department of Biological Sciences, Purdue Institute for Integrative Neuroscience, Purdue Autism Research Center, Purdue University, West Lafayette, IN 47907, USA
| |
Collapse
|
5
|
Maran JJ, Adesina MM, Green CR, Kwakowsky A, Mugisho OO. The central role of the NLRP3 inflammasome pathway in the pathogenesis of age-related diseases in the eye and the brain. Ageing Res Rev 2023; 88:101954. [PMID: 37187367 DOI: 10.1016/j.arr.2023.101954] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2023] [Revised: 05/08/2023] [Accepted: 05/12/2023] [Indexed: 05/17/2023]
Abstract
With increasing age, structural changes occur in the eye and brain. Neuronal death, inflammation, vascular disruption, and microglial activation are among many of the pathological changes that can occur during ageing. Furthermore, ageing individuals are at increased risk of developing neurodegenerative diseases in these organs, including Alzheimer's disease (AD), Parkinson's disease (PD), glaucoma and age-related macular degeneration (AMD). Although these diseases pose a significant global public health burden, current treatment options focus on slowing disease progression and symptomatic control rather than targeting underlying causes. Interestingly, recent investigations have proposed an analogous aetiology between age-related diseases in the eye and brain, where a process of chronic low-grade inflammation is implicated. Studies have suggested that patients with AD or PD are also associated with an increased risk of AMD, glaucoma, and cataracts. Moreover, pathognomonic amyloid-β and α-synuclein aggregates, which accumulate in AD and PD, respectively, can be found in ocular parenchyma. In terms of a common molecular pathway that underpins these diseases, the nucleotide-binding domain, leucine-rich-containing family, and pyrin domain-containing-3 (NLRP3) inflammasome is thought to play a vital role in the manifestation of all these diseases. This review summarises the current evidence regarding cellular and molecular changes in the brain and eye with age, similarities between ocular and cerebral age-related diseases, and the role of the NLRP3 inflammasome as a critical mediator of disease propagation in the eye and the brain during ageing.
Collapse
Affiliation(s)
- Jack J Maran
- Buchanan Ocular Therapeutics Unit, Department of Ophthalmology and the New Zealand National Eye Centre, University of Auckland, New Zealand
| | - Moradeke M Adesina
- Buchanan Ocular Therapeutics Unit, Department of Ophthalmology and the New Zealand National Eye Centre, University of Auckland, New Zealand
| | - Colin R Green
- Department of Ophthalmology and the New Zealand National Eye Centre, Faculty of Medical and Health Sciences, University of Auckland, New Zealand
| | - Andrea Kwakowsky
- Pharmacology and Therapeutics, School of Medicine, Galway Neuroscience Centre, University of Galway, Galway, Ireland
| | - Odunayo O Mugisho
- Buchanan Ocular Therapeutics Unit, Department of Ophthalmology and the New Zealand National Eye Centre, University of Auckland, New Zealand.
| |
Collapse
|
6
|
Veys L, Devroye J, Lefevere E, Cools L, Vandenabeele M, De Groef L. Characterizing the Retinal Phenotype of the Thy1-h[A30P]α-syn Mouse Model of Parkinson's Disease. Front Neurosci 2021; 15:726476. [PMID: 34557068 PMCID: PMC8452874 DOI: 10.3389/fnins.2021.726476] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2021] [Accepted: 08/19/2021] [Indexed: 12/30/2022] Open
Abstract
Despite decades of research, disease-modifying treatments of Parkinson’s disease (PD), the second most common neurodegenerative disease worldwide, remain out of reach. One of the reasons for this treatment gap is the incomplete understanding of how misfolded alpha-synuclein (α-syn) contributes to PD pathology. The retina, as an integral part of the central nervous system, recapitulates the PD disease processes that are typically seen in the brain, and retinal manifestations have emerged as prodromal symptoms of the disease. The timeline of PD manifestations in the visual system, however, is not fully elucidated and the underlying mechanisms are obscure. This highlights the need for new studies investigating retinal pathology, in order to propel its use as PD biomarker, and to develop validated research models to investigate PD pathogenesis. The present study pioneers in characterizing the retina of the Thy1-h[A30P]α-syn PD transgenic mouse model. We demonstrate widespread α-syn accumulation in the inner retina of these mice, of which a proportion is phosphorylated yet not aggregated. This α-syn expression coincides with inner retinal atrophy due to postsynaptic degeneration. We also reveal abnormal retinal electrophysiological responses. Absence of selective loss of melanopsin retinal ganglion cells or dopaminergic amacrine cells and inflammation indicates that the retinal manifestations in these transgenic mice diverge from their brain phenotype, and questions the specific cellular or molecular alterations that underlie retinal pathology in this PD mouse model. Nevertheless, the observed α-syn accumulation, synapse loss and functional deficits suggest that the Thy1-h[A30P]α-syn retina mimics some of the features of prodromal PD, and thus may provide a window to monitor and study the preclinical/prodromal stages of PD, PD-associated retinal disease processes, as well as aid in retinal biomarker discovery and validation.
Collapse
Affiliation(s)
- Lien Veys
- Research Group of Neural Circuit Development and Regeneration, Department of Biology, KU Leuven, Leuven, Belgium.,Department of Biomedical Sciences, Leuven Brain Institute, Leuven, Belgium
| | - Joyce Devroye
- Research Group of Neural Circuit Development and Regeneration, Department of Biology, KU Leuven, Leuven, Belgium.,Department of Biomedical Sciences, Leuven Brain Institute, Leuven, Belgium
| | - Evy Lefevere
- Research Group of Neural Circuit Development and Regeneration, Department of Biology, KU Leuven, Leuven, Belgium.,Department of Biomedical Sciences, Leuven Brain Institute, Leuven, Belgium
| | - Lien Cools
- Research Group of Neural Circuit Development and Regeneration, Department of Biology, KU Leuven, Leuven, Belgium.,Department of Biomedical Sciences, Leuven Brain Institute, Leuven, Belgium
| | - Marjan Vandenabeele
- Research Group of Neural Circuit Development and Regeneration, Department of Biology, KU Leuven, Leuven, Belgium.,Department of Biomedical Sciences, Leuven Brain Institute, Leuven, Belgium
| | - Lies De Groef
- Research Group of Neural Circuit Development and Regeneration, Department of Biology, KU Leuven, Leuven, Belgium.,Department of Biomedical Sciences, Leuven Brain Institute, Leuven, Belgium
| |
Collapse
|
7
|
Veys L, Van Houcke J, Aerts J, Van Pottelberge S, Mahieu M, Coens A, Melki R, Moechars D, De Muynck L, De Groef L. Absence of Uptake and Prion-Like Spreading of Alpha-Synuclein and Tau After Intravitreal Injection of Preformed Fibrils. Front Aging Neurosci 2021; 12:614587. [PMID: 33519421 PMCID: PMC7843377 DOI: 10.3389/fnagi.2020.614587] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2020] [Accepted: 12/24/2020] [Indexed: 12/26/2022] Open
Abstract
Although very different in etiology and symptoms, numerous neurodegenerative diseases can be classified as proteinopathies. More so, evidence indicates that the key misfolded proteins at the basis of different neuropathies might share common mechanisms of propagation. As such, the prion-like spreading of protein aggregates through the neural network is subject of intensive research focus and requires adequate models. Here, we made use of the well-defined architecture and large accessibility of the visual system, of which the retinotopic connections represent a simple route of anterograde signaling and an elegant model to investigate transsynaptic, prion-like spreading. In two independent studies, uptake and seeding of alpha-synuclein and tau were examined after intravitreal injection of preformed fibrils. However, extracellular matrix components in the vitreous space and at the vitreoretinal surface appeared to act as a barrier for the entry of both fibrils into the retina. These results show that further experimental refinement is needed to fully realize the potential of the visual system as a model for studying the molecular and cellular mechanisms of anterograde, transsynaptic spreading of prion-like proteins.
Collapse
Affiliation(s)
- Lien Veys
- Laboratory Neural Circuit Development and Regeneration, Department of Biology, KU Leuven, Leuven, Belgium
| | - Jessie Van Houcke
- Department of Neuroscience, Janssen Research and Development, Division of Janssen Pharmaceutica NV, Beerse, Belgium
| | - Jeroen Aerts
- Department of Neuroscience, Janssen Research and Development, Division of Janssen Pharmaceutica NV, Beerse, Belgium
| | - Sophie Van Pottelberge
- Department of Neuroscience, Janssen Research and Development, Division of Janssen Pharmaceutica NV, Beerse, Belgium
| | - Michel Mahieu
- Department of Neuroscience, Janssen Research and Development, Division of Janssen Pharmaceutica NV, Beerse, Belgium
| | - Audrey Coens
- Laboratory of Neurodegenerative Disease, Institute François Jacob, MIRCen, CEA-CNRS, Fontenay aux Roses, France
| | - Ronald Melki
- Laboratory of Neurodegenerative Disease, Institute François Jacob, MIRCen, CEA-CNRS, Fontenay aux Roses, France
| | - Dieder Moechars
- Department of Neuroscience, Janssen Research and Development, Division of Janssen Pharmaceutica NV, Beerse, Belgium
| | - Louis De Muynck
- Department of Neuroscience, Janssen Research and Development, Division of Janssen Pharmaceutica NV, Beerse, Belgium
| | - Lies De Groef
- Laboratory Neural Circuit Development and Regeneration, Department of Biology, KU Leuven, Leuven, Belgium
| |
Collapse
|
8
|
Moro E, Bellot E, Meoni S, Pelissier P, Hera R, Dojat M, Coizet V. Visual Dysfunction of the Superior Colliculus in De Novo Parkinsonian Patients. Ann Neurol 2020; 87:533-546. [DOI: 10.1002/ana.25696] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2019] [Revised: 02/05/2020] [Accepted: 02/05/2020] [Indexed: 02/02/2023]
Affiliation(s)
- Elena Moro
- Division of Neurology Grenoble University Hospital Center Grenoble France
| | - Emmanuelle Bellot
- Grenoble Alpes University Grenoble Institute of Neurosciences Grenoble France
| | - Sara Meoni
- Division of Neurology Grenoble University Hospital Center Grenoble France
| | - Pierre Pelissier
- Division of Neurology Grenoble University Hospital Center Grenoble France
| | | | - Michel Dojat
- Grenoble Alpes University Grenoble Institute of Neurosciences Grenoble France
| | - Veronique Coizet
- Grenoble Alpes University Grenoble Institute of Neurosciences Grenoble France
| | | |
Collapse
|
9
|
Kim EJ, Hwang JHL, Gaus SE, Nana AL, Deng J, Brown JA, Spina S, Lee MJ, Ramos EM, Grinberg LT, Kramer JH, Boxer AL, Gorno-Tempini ML, Rosen HJ, Miller BL, Seeley WW. Evidence of corticofugal tau spreading in patients with frontotemporal dementia. Acta Neuropathol 2020; 139:27-43. [PMID: 31542807 DOI: 10.1007/s00401-019-02075-z] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2019] [Revised: 08/10/2019] [Accepted: 08/11/2019] [Indexed: 01/09/2023]
Abstract
Common neurodegenerative diseases feature progressive accumulation of disease-specific protein aggregates in selectively vulnerable brain regions. Increasing experimental evidence suggests that misfolded disease proteins exhibit prion-like properties, including the ability to seed corruptive templating and self-propagation along axons. Direct evidence for transneuronal spread in patients, however, remains limited. To test predictions made by the transneuronal spread hypothesis in human tissues, we asked whether tau deposition within axons of the corticospinal and corticopontine pathways can be predicted based on clinical syndromes and cortical atrophy patterns seen in frontotemporal lobar degeneration (FTLD). Sixteen patients with Pick's disease, 21 with corticobasal degeneration, and 3 with FTLD-MAPT were included, spanning a range of clinical syndromes across the frontotemporal dementia (FTD) spectrum. Cortical involvement was measured using a neurodegeneration score, a tau score, and a composite score based on semiquantitative ratings and complemented by an MRI-based cortical atrophy W-map based on antemortem imaging. Midbrain cerebral peduncle and pontine base descending fibers were divided into three subregions, representing prefrontopontine, corticospinal, and parieto-temporo-occipital fiber pathways. Tau area fraction was calculated in each subregion and related to clinical syndrome and cortical measures. Within each clinical syndrome, there were predicted relationships between cortical atrophy patterns and axonal tau deposition in midbrain cerebral peduncle and pontine base. Between syndromes, contrasting and predictable patterns of brainstem axonal tau deposition emerged, with, for example, greater tau in prefrontopontine fibers in behavioral variant FTD and in corticospinal fibers in corticobasal syndrome. Finally, semiquantitative and quantitative cortical degeneration scores predicted brainstem axonal tau deposition based on anatomical principles. Taken together, these findings provide important human evidence in support of axonal tau spreading in patients with specific forms of tau-related neurodegeneration.
Collapse
Affiliation(s)
- Eun-Joo Kim
- Department of Neurology, Memory and Aging Center, University of California, 675 Nelson Rising Lane, San Francisco, CA, 94158, USA
- Department of Neurology, Pusan National University Hospital, Pusan National University School of Medicine and Medical Research Institute, Busan, Republic of Korea
| | - Ji-Hye L Hwang
- Department of Neurology, Memory and Aging Center, University of California, 675 Nelson Rising Lane, San Francisco, CA, 94158, USA
| | - Stephanie E Gaus
- Department of Neurology, Memory and Aging Center, University of California, 675 Nelson Rising Lane, San Francisco, CA, 94158, USA
| | - Alissa L Nana
- Department of Neurology, Memory and Aging Center, University of California, 675 Nelson Rising Lane, San Francisco, CA, 94158, USA
| | - Jersey Deng
- Department of Neurology, Memory and Aging Center, University of California, 675 Nelson Rising Lane, San Francisco, CA, 94158, USA
| | - Jesse A Brown
- Department of Neurology, Memory and Aging Center, University of California, 675 Nelson Rising Lane, San Francisco, CA, 94158, USA
| | - Salvatore Spina
- Department of Neurology, Memory and Aging Center, University of California, 675 Nelson Rising Lane, San Francisco, CA, 94158, USA
| | - Myung Jun Lee
- Department of Neurology, Pusan National University Hospital, Pusan National University School of Medicine and Medical Research Institute, Busan, Republic of Korea
| | - Eliana Marisa Ramos
- Department of Psychiatry, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
| | - Lea T Grinberg
- Department of Neurology, Memory and Aging Center, University of California, 675 Nelson Rising Lane, San Francisco, CA, 94158, USA
- Department of Pathology, University of California, San Francisco, USA
| | - Joel H Kramer
- Department of Neurology, Memory and Aging Center, University of California, 675 Nelson Rising Lane, San Francisco, CA, 94158, USA
| | - Adam L Boxer
- Department of Neurology, Memory and Aging Center, University of California, 675 Nelson Rising Lane, San Francisco, CA, 94158, USA
| | - Maria Luisa Gorno-Tempini
- Department of Neurology, Memory and Aging Center, University of California, 675 Nelson Rising Lane, San Francisco, CA, 94158, USA
| | - Howard J Rosen
- Department of Neurology, Memory and Aging Center, University of California, 675 Nelson Rising Lane, San Francisco, CA, 94158, USA
| | - Bruce L Miller
- Department of Neurology, Memory and Aging Center, University of California, 675 Nelson Rising Lane, San Francisco, CA, 94158, USA
| | - William W Seeley
- Department of Neurology, Memory and Aging Center, University of California, 675 Nelson Rising Lane, San Francisco, CA, 94158, USA.
- Department of Pathology, University of California, San Francisco, USA.
| |
Collapse
|
10
|
Kovacs GG. Molecular pathology of neurodegenerative diseases: principles and practice. J Clin Pathol 2019; 72:725-735. [PMID: 31395625 DOI: 10.1136/jclinpath-2019-205952] [Citation(s) in RCA: 126] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2019] [Revised: 07/25/2019] [Accepted: 07/26/2019] [Indexed: 12/12/2022]
Abstract
Neurodegenerative diseases are characterised by selective dysfunction and progressive loss of synapses and neurons associated with pathologically altered proteins that deposit primarily in the human brain and spinal cord. Recent discoveries have identified a spectrum of distinct immunohistochemically and biochemically detectable proteins, which serve as a basis for protein-based disease classification. Diagnostic criteria have been updated and disease staging procedures have been proposed. These are based on novel concepts which recognise that (1) most of these proteins follow a sequential distribution pattern in the brain suggesting a seeding mechanism and cell-to-cell propagation; (2) some of the neurodegeneration-associated proteins can be detected in peripheral organs; and (3) concomitant presence of neurodegeneration-associated proteins is more the rule than the exception. These concepts, together with the fact that the clinical symptoms do not unequivocally reflect the molecular pathological background, place the neuropathological examination at the centre of requirements for an accurate diagnosis. The need for quality control in biomarker development, clinical and neuroimaging studies, and evaluation of therapy trials, as well as an increasing demand for the general public to better understand human brain disorders, underlines the importance for a renaissance of postmortem neuropathological studies at this time. This review summarises recent advances in neuropathological diagnosis and reports novel aspects of relevance for general pathological practice.
Collapse
Affiliation(s)
- Gabor G Kovacs
- Laboratory Medicine Program, University Health Network, Toronto, Ontario, Canada
| |
Collapse
|
11
|
Kresl P, Rahimi J, Gelpi E, Aldecoa I, Ricken G, Danics K, Keller E, Kovacs GG. Accumulation of prion protein in the vagus nerve in creutzfeldt-jakob disease. Ann Neurol 2019; 85:782-787. [PMID: 30801763 PMCID: PMC6593447 DOI: 10.1002/ana.25451] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2018] [Revised: 02/19/2019] [Accepted: 02/19/2019] [Indexed: 12/20/2022]
Abstract
Disease‐associated proteins are thought to propagate along neuronal processes in neurodegenerative diseases. To detect disease‐associated prion protein (PrPSc) in the vagus nerve in different forms and molecular subtypes of Creutzfeldt–Jakob disease (CJD), we applied 3 different anti‐PrP antibodies. We screened the vagus nerve in 162 sporadic and 30 genetic CJD cases. Four of 31 VV‐2 type sporadic CJD and 7 of 30 genetic CJD cases showed vagal PrPSc immunodeposits with distinct morphology. Thus, PrPSc in CJD affects the vagus nerve analogously to α‐synuclein in Parkinson disease. The morphologically diverse deposition of PrPSc in genetic and sporadic CJD argues against uniform mechanisms of propagation of PrPSc. Ann Neurol 2019;85:782–787
Collapse
Affiliation(s)
- Philip Kresl
- Institute of Neurology, Medical University of Vienna, Vienna, Austria.,Austrian Reference Center for Human Prion Diseases, Medical University of Vienna, Vienna, Austria
| | - Jasmin Rahimi
- Department of Neurology and Karl Landsteiner Institute for Neuroimmunological and Neurodegenerative Disorders, Danube Hospital, Vienna, Austria
| | - Ellen Gelpi
- Institute of Neurology, Medical University of Vienna, Vienna, Austria.,Austrian Reference Center for Human Prion Diseases, Medical University of Vienna, Vienna, Austria.,Neurological Tissue Bank, Biobanc Hospital Clinic, August Pi i Sunyer Biomedical Research Institute, and Pathology Department, Biomedical Diagnostic Center, Barcelona Hospital Clinic, Barcelona, Spain
| | - Iban Aldecoa
- Neurological Tissue Bank, Biobanc Hospital Clinic, August Pi i Sunyer Biomedical Research Institute, and Pathology Department, Biomedical Diagnostic Center, Barcelona Hospital Clinic, Barcelona, Spain
| | - Gerda Ricken
- Institute of Neurology, Medical University of Vienna, Vienna, Austria
| | - Krisztina Danics
- Neuropathology and Prion Disease Reference Center, Department of Forensic and Insurance Medicine, Semmelweis University, Budapest, Hungary
| | - Eva Keller
- Neuropathology and Prion Disease Reference Center, Department of Forensic and Insurance Medicine, Semmelweis University, Budapest, Hungary
| | - Gabor G Kovacs
- Institute of Neurology, Medical University of Vienna, Vienna, Austria.,Austrian Reference Center for Human Prion Diseases, Medical University of Vienna, Vienna, Austria.,Neuropathology and Prion Disease Reference Center, Department of Forensic and Insurance Medicine, Semmelweis University, Budapest, Hungary
| |
Collapse
|
12
|
Veys L, Vandenabeele M, Ortuño-Lizarán I, Baekelandt V, Cuenca N, Moons L, De Groef L. Retinal α-synuclein deposits in Parkinson's disease patients and animal models. Acta Neuropathol 2019; 137:379-395. [PMID: 30721408 DOI: 10.1007/s00401-018-01956-z] [Citation(s) in RCA: 72] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2018] [Revised: 12/22/2018] [Accepted: 12/22/2018] [Indexed: 12/21/2022]
Abstract
Despite decades of research, accurate diagnosis of Parkinson's disease remains a challenge, and disease-modifying treatments are still lacking. Research into the early (presymptomatic) stages of Parkinson's disease and the discovery of novel biomarkers is of utmost importance to reduce this burden and to come to a more accurate diagnosis at the very onset of the disease. Many have speculated that non-motor symptoms could provide a breakthrough in the quest for early biomarkers of Parkinson's disease, including the visual disturbances and retinal abnormalities that are seen in the majority of Parkinson's disease patients. An expanding number of clinical studies have investigated the use of in vivo assessments of retinal structure, electrophysiological function, and vision-driven tasks as novel means for identifying patients at risk that need further neurological examination and for longitudinal follow-up of disease progression in Parkinson's disease patients. Often, the results of these studies have been interpreted in relation to α-synuclein deposits and dopamine deficiency in the retina, mirroring the defining pathological features of Parkinson's disease in the brain. To better understand the visual defects seen in Parkinson's disease patients and to propel the use of retinal changes as biomarkers for Parkinson's disease, however, more conclusive neuropathological evidence for the presence of retinal α-synuclein aggregates, and its relation to the cerebral α-synuclein burden, is urgently needed. This review provides a comprehensive and critical overview of the research conducted to unveil α-synuclein aggregates in the retina of Parkinson's disease patients and animal models, and thereby aims to aid the ongoing discussion about the potential use of the retinal changes and/or visual symptoms as biomarkers for Parkinson's disease.
Collapse
|
13
|
Ocular and Visual Manifestation of Alzheimer’s Disease: A Literature Review II Part: Clinical Studies. ARCHIVES OF NEUROSCIENCE 2019. [DOI: 10.5812/ans.74239] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
14
|
Ocular and Visual Manifestation of the Alzheimer’s Disease: A Literature Review, Part I: Animal Models and Human Pathology. ARCHIVES OF NEUROSCIENCE 2018. [DOI: 10.5812/ans.74225] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
15
|
West Greenlee MH, Lind M, Kokemuller R, Mammadova N, Kondru N, Manne S, Smith J, Kanthasamy A, Greenlee J. Temporal Resolution of Misfolded Prion Protein Transport, Accumulation, Glial Activation, and Neuronal Death in the Retinas of Mice Inoculated with Scrapie. THE AMERICAN JOURNAL OF PATHOLOGY 2016; 186:2302-9. [PMID: 27521336 PMCID: PMC5012505 DOI: 10.1016/j.ajpath.2016.05.018] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/12/2016] [Revised: 05/17/2016] [Accepted: 05/23/2016] [Indexed: 12/14/2022]
Abstract
Currently, there is a lack of pathological landmarks to describe the progression of prion disease in vivo. Our goal was to use an experimental model to determine the temporal relationship between the transport of misfolded prion protein (PrP(Sc)) from the brain to the retina, the accumulation of PrP(Sc) in the retina, the response of the surrounding retinal tissue, and loss of neurons. Retinal samples from mice inoculated with RML scrapie were collected at 30, 60, 90, 105, and 120 days post inoculation (dpi) or at the onset of clinical signs of disease (153 dpi). Retinal homogenates were tested for prion seeding activity. Antibody staining was used to assess accumulation of PrP(Sc) and the resulting response of retinal tissue. Loss of photoreceptors was used as a measure of neuronal death. PrP(Sc) seeding activity was first detected in all samples at 60 dpi. Accumulation of PrP(Sc) and coincident activation of retinal glia were first detected at 90 dpi. Activation of microglia was first detected at 105 dpi, but neuronal death was not detectable until 120 dpi. Our results demonstrate that by using the retina we can resolve the temporal separation between several key events in the pathogenesis of prion disease.
Collapse
Affiliation(s)
- M Heather West Greenlee
- Department of Biomedical Sciences, Iowa State University College of Veterinary Medicine, Ames, Iowa; Virus and Prion Disease Unit, National Animal Disease Center, US Department of Agriculture, Ames, Iowa.
| | - Melissa Lind
- Department of Biomedical Sciences, Iowa State University College of Veterinary Medicine, Ames, Iowa
| | - Robyn Kokemuller
- Virus and Prion Disease Unit, National Animal Disease Center, US Department of Agriculture, Ames, Iowa
| | - Najiba Mammadova
- Virus and Prion Disease Unit, National Animal Disease Center, US Department of Agriculture, Ames, Iowa
| | - Naveen Kondru
- Department of Biomedical Sciences, Iowa State University College of Veterinary Medicine, Ames, Iowa
| | - Sireesha Manne
- Department of Biomedical Sciences, Iowa State University College of Veterinary Medicine, Ames, Iowa
| | - Jodi Smith
- Virus and Prion Disease Unit, National Animal Disease Center, US Department of Agriculture, Ames, Iowa
| | - Anumantha Kanthasamy
- Department of Biomedical Sciences, Iowa State University College of Veterinary Medicine, Ames, Iowa
| | - Justin Greenlee
- Virus and Prion Disease Unit, National Animal Disease Center, US Department of Agriculture, Ames, Iowa
| |
Collapse
|
16
|
Kovacs GG. Molecular Pathological Classification of Neurodegenerative Diseases: Turning towards Precision Medicine. Int J Mol Sci 2016; 17:ijms17020189. [PMID: 26848654 PMCID: PMC4783923 DOI: 10.3390/ijms17020189] [Citation(s) in RCA: 176] [Impact Index Per Article: 19.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2015] [Revised: 01/21/2016] [Accepted: 01/26/2016] [Indexed: 02/06/2023] Open
Abstract
Neurodegenerative diseases (NDDs) are characterized by selective dysfunction and loss of neurons associated with pathologically altered proteins that deposit in the human brain but also in peripheral organs. These proteins and their biochemical modifications can be potentially targeted for therapy or used as biomarkers. Despite a plethora of modifications demonstrated for different neurodegeneration-related proteins, such as amyloid-β, prion protein, tau, α-synuclein, TAR DNA-binding protein 43 (TDP-43), or fused in sarcoma protein (FUS), molecular classification of NDDs relies on detailed morphological evaluation of protein deposits, their distribution in the brain, and their correlation to clinical symptoms together with specific genetic alterations. A further facet of the neuropathology-based classification is the fact that many protein deposits show a hierarchical involvement of brain regions. This has been shown for Alzheimer and Parkinson disease and some forms of tauopathies and TDP-43 proteinopathies. The present paper aims to summarize current molecular classification of NDDs, focusing on the most relevant biochemical and morphological aspects. Since the combination of proteinopathies is frequent, definition of novel clusters of patients with NDDs needs to be considered in the era of precision medicine. Optimally, neuropathological categorizing of NDDs should be translated into in vivo detectable biomarkers to support better prediction of prognosis and stratification of patients for therapy trials.
Collapse
Affiliation(s)
- Gabor G Kovacs
- Institute of Neurology, Medical University of Vienna, AKH 4J, Währinger Gürtel 18-20, A-1090 Vienna, Austria.
| |
Collapse
|