101
|
Talbot K, Feneberg E, Scaber J, Thompson AG, Turner MR. Amyotrophic lateral sclerosis: the complex path to precision medicine. J Neurol 2018; 265:2454-2462. [PMID: 30054789 PMCID: PMC6182683 DOI: 10.1007/s00415-018-8983-8] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2018] [Accepted: 07/19/2018] [Indexed: 12/11/2022]
Abstract
Amyotrophic lateral sclerosis (ALS) is a progressive neurodegenerative disease of the corticomotorneuronal network responsible for voluntary movement. There are well-established clinical, genetic and pathological overlaps between ALS and frontotemporal dementia (FTD), which together constitute the 'TDP-43 proteinopathies'. An ever-expanding list of genes in which mutation leads to typical ALS have implicated abnormalities in RNA processing, protein homoeostasis and axonal transport. How these apparently distinct pathways converge to cause the characteristic clinical syndrome of ALS remains unclear. Although there are major gaps in our understanding of the essential nature of ALS pathophysiology, the identification of genetic causes in up to 15% of ALS patients, coupled with advances in biotechnology and biomarker research provide a foundation for approaches to treatment based on 'precision medicine', and even prevention of the disease in pre-symptomatic mutation carriers in the future. Currently, multidisciplinary care remains the bedrock of management and this is increasingly being put onto an evidence-based footing.
Collapse
Affiliation(s)
- Kevin Talbot
- Nuffield Department of Clinical Neurosciences, John Radcliffe Hospital, University of Oxford, Oxford, OX3 9DU, UK.
| | - Emily Feneberg
- Nuffield Department of Clinical Neurosciences, John Radcliffe Hospital, University of Oxford, Oxford, OX3 9DU, UK
| | - Jakub Scaber
- Nuffield Department of Clinical Neurosciences, John Radcliffe Hospital, University of Oxford, Oxford, OX3 9DU, UK
| | - Alexander G Thompson
- Nuffield Department of Clinical Neurosciences, John Radcliffe Hospital, University of Oxford, Oxford, OX3 9DU, UK
| | - Martin R Turner
- Nuffield Department of Clinical Neurosciences, John Radcliffe Hospital, University of Oxford, Oxford, OX3 9DU, UK
| |
Collapse
|
102
|
Woollacott IOC, Nicholas JM, Heslegrave A, Heller C, Foiani MS, Dick KM, Russell LL, Paterson RW, Keshavan A, Fox NC, Warren JD, Schott JM, Zetterberg H, Rohrer JD. Cerebrospinal fluid soluble TREM2 levels in frontotemporal dementia differ by genetic and pathological subgroup. Alzheimers Res Ther 2018; 10:79. [PMID: 30111356 PMCID: PMC6094471 DOI: 10.1186/s13195-018-0405-8] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2018] [Accepted: 07/12/2018] [Indexed: 12/13/2022]
Abstract
BACKGROUND Reliable biomarkers of frontotemporal dementia (FTD) are currently lacking. FTD may be associated with chronic immune dysfunction, microglial activation and raised inflammatory markers, particularly in progranulin (GRN) mutation carriers. Levels of soluble triggering receptor expressed on myeloid cells 2 (sTREM2) are elevated in Alzheimer's disease (AD), but they have not been fully explored in FTD. METHODS We investigated whether cerebrospinal fluid (CSF) sTREM2 levels differ between FTD and controls, across different clinical and genetic subtypes of FTD, or between individuals with FTD due to AD versus non-AD pathology (based on CSF neurodegenerative biomarkers). We also assessed relationships between CSF sTREM2 and other CSF biomarkers (total tau [T-tau], tau phosphorylated at position threonine-181 [P-tau] and β-amyloid 1-42 [Aβ42]) and age and disease duration. Biomarker levels were measured using immunoassays in 17 healthy controls and 64 patients with FTD (behavioural variant FTD, n = 20; primary progressive aphasia, n = 44). Ten of 64 had familial FTD, with mutations in GRN (n = 3), MAPT (n = 4), or C9orf72 (n = 3). Fifteen of 64 had neurodegenerative biomarkers consistent with AD pathology (11 of whom had logopenic variant PPA). Levels were compared using multivariable linear regressions. RESULTS CSF sTREM2 levels did not differ between FTD and controls or between clinical subgroups. However, GRN mutation carriers had higher levels than controls (mean ([SD] = 9.7 [2.9] vs. 6.8 [1.6] ng/ml; P = 0.028) and MAPT (3.9 [1.5] ng/ml; P = 0.003] or C9orf72 [4.6 [1.8] ng/ml; P = 0.006) mutation carriers. Individuals with AD-like CSF had higher sTREM2 levels than those with non-AD-like CSF (9.0 [3.6] vs. 6.9 [3.0] ng/ml; P = 0.029). CSF sTREM2 levels were associated with T-tau levels in control and FTD groups and also with P-tau in those with FTD and AD-like CSF. CSF sTREM2 levels were influenced by both age and disease duration in FTD. CONCLUSIONS Although CSF sTREM2 levels are not raised in FTD overall or in a particular clinical subtype of FTD, levels are raised in familial FTD associated with GRN mutations and in FTD syndromes due to AD pathology. Because CSF sTREM2 levels correlate with a marker of neuronal injury (T-tau), sTREM2 should be explored as a biomarker of disease intensity in future longitudinal studies of FTD.
Collapse
Affiliation(s)
- Ione O. C. Woollacott
- Dementia Research Centre, Department of Neurodegenerative Disease, UCL Institute of Neurology, Queen Square, London, WC1N 3BG UK
| | - Jennifer M. Nicholas
- Department of Medical Statistics, London School of Hygiene and Tropical Medicine, London, UK
| | - Amanda Heslegrave
- Department of Molecular Neuroscience, UCL Institute of Neurology, Queen Square, London, UK
- UK Dementia Research Institute, London, UK
| | - Carolin Heller
- Department of Molecular Neuroscience, UCL Institute of Neurology, Queen Square, London, UK
- UK Dementia Research Institute, London, UK
| | - Martha S. Foiani
- Department of Molecular Neuroscience, UCL Institute of Neurology, Queen Square, London, UK
- UK Dementia Research Institute, London, UK
| | - Katrina M. Dick
- Dementia Research Centre, Department of Neurodegenerative Disease, UCL Institute of Neurology, Queen Square, London, WC1N 3BG UK
| | - Lucy L. Russell
- Dementia Research Centre, Department of Neurodegenerative Disease, UCL Institute of Neurology, Queen Square, London, WC1N 3BG UK
| | - Ross W. Paterson
- Dementia Research Centre, Department of Neurodegenerative Disease, UCL Institute of Neurology, Queen Square, London, WC1N 3BG UK
| | - Ashvini Keshavan
- Dementia Research Centre, Department of Neurodegenerative Disease, UCL Institute of Neurology, Queen Square, London, WC1N 3BG UK
| | - Nick C. Fox
- Dementia Research Centre, Department of Neurodegenerative Disease, UCL Institute of Neurology, Queen Square, London, WC1N 3BG UK
- UK Dementia Research Institute, London, UK
| | - Jason D. Warren
- Dementia Research Centre, Department of Neurodegenerative Disease, UCL Institute of Neurology, Queen Square, London, WC1N 3BG UK
| | - Jonathan M. Schott
- Dementia Research Centre, Department of Neurodegenerative Disease, UCL Institute of Neurology, Queen Square, London, WC1N 3BG UK
| | - Henrik Zetterberg
- Department of Molecular Neuroscience, UCL Institute of Neurology, Queen Square, London, UK
- UK Dementia Research Institute, London, UK
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
| | - Jonathan D. Rohrer
- Dementia Research Centre, Department of Neurodegenerative Disease, UCL Institute of Neurology, Queen Square, London, WC1N 3BG UK
| |
Collapse
|
103
|
Nag S, Yu L, Boyle PA, Leurgans SE, Bennett DA, Schneider JA. TDP-43 pathology in anterior temporal pole cortex in aging and Alzheimer's disease. Acta Neuropathol Commun 2018; 6:33. [PMID: 29716643 PMCID: PMC5928580 DOI: 10.1186/s40478-018-0531-3] [Citation(s) in RCA: 117] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2018] [Accepted: 04/05/2018] [Indexed: 11/10/2022] Open
Abstract
TDP-43 pathology was investigated in the anterior temporal pole cortex (ATPC) and orbital frontal cortex (OFC), regions often degenerated in frontotemporal lobar degenerations (FTLD), in aging and Alzheimer's disease (AD). Diagnosis of dementia in the 1160 autopsied participants from 3 studies of community-dwelling elders was based on clinical evaluation and cognitive performance tests which were used to create summary measures of the five cognitive domains. Neuronal and glial TDP-43 cytoplasmic inclusions were quantitated in 8 brain regions by immunohistochemistry, and used in ANOVA and regression analyses. TDP-43 pathology was present in 547 (49.4%) participants in whom ATPC (41.9%) was the most frequently involved neocortical region and in 15.5% of these cases, ATPC was the only neocortical area with TDP-43 pathology suggesting not only that ATPC is involved early by TDP-43 but that ATPC may represent an intermediate stage between mesial temporal lobe involvement by TDP-43 and the last stage with involvement of other neocortical areas. To better study this intermediary neocortical stage, and to integrate with other staging schemes, our previous 3 stage distribution of TDP-43 pathology was revised to a 5 stage distribution scheme with stage 1 showing involvement of the amygdala only; stage 2 showed extension to hippocampus and/or entorhinal cortex; stage 3 showed extension to the ATPC; stage 4 - showed extension to the midtemporal cortex and/or OFC and finally in stage 5, there was extension to the midfrontal cortex. Clinically, cases in stages 2 to 5 had impaired episodic memory, however, stage 3 was distinct from stage 2 since stage 3 cases had significantly increased odds of dementia. The proportion of cases with hippocampal sclerosis increased progressively across the stages with stage 5 showing the largest proportion of hippocampal sclerosis cases. Stage 5 cases differed from other stages by having impairment of semantic memory and perceptual speed, in addition to episodic memory impairment. These data suggest that of the regions studied, TDP-43 pathology in the ATPC is an important early neocortical stage of TDP-43 progression in aging and AD while extension of TDP-43 pathology to the midfrontal cortex is a late stage associated with more severe and global cognitive impairment.
Collapse
Affiliation(s)
- Sukriti Nag
- Rush Alzheimer Disease Center, Rush University Medical Center, Suite 1000, 1750 W Harrison Street, Chicago, IL 60612 USA
- Departments of Pathology (Neuropathology), Rush University Medical Center, Chicago, IL USA
| | - Lei Yu
- Rush Alzheimer Disease Center, Rush University Medical Center, Suite 1000, 1750 W Harrison Street, Chicago, IL 60612 USA
- Departments of Neurological Sciences, Rush University Medical Center, Chicago, IL USA
| | - Patricia A. Boyle
- Rush Alzheimer Disease Center, Rush University Medical Center, Suite 1000, 1750 W Harrison Street, Chicago, IL 60612 USA
- Departments of Behavioral Sciences, Rush University Medical Center, Chicago, IL USA
| | - Sue E. Leurgans
- Rush Alzheimer Disease Center, Rush University Medical Center, Suite 1000, 1750 W Harrison Street, Chicago, IL 60612 USA
- Departments of Neurological Sciences, Rush University Medical Center, Chicago, IL USA
| | - David A. Bennett
- Rush Alzheimer Disease Center, Rush University Medical Center, Suite 1000, 1750 W Harrison Street, Chicago, IL 60612 USA
- Departments of Neurological Sciences, Rush University Medical Center, Chicago, IL USA
| | - Julie A. Schneider
- Rush Alzheimer Disease Center, Rush University Medical Center, Suite 1000, 1750 W Harrison Street, Chicago, IL 60612 USA
- Departments of Pathology (Neuropathology), Rush University Medical Center, Chicago, IL USA
- Departments of Neurological Sciences, Rush University Medical Center, Chicago, IL USA
| |
Collapse
|
104
|
Kellmeyer P, Grosse-Wentrup M, Schulze-Bonhage A, Ziemann U, Ball T. Electrophysiological correlates of neurodegeneration in motor and non-motor brain regions in amyotrophic lateral sclerosis-implications for brain-computer interfacing. J Neural Eng 2018; 15:041003. [PMID: 29676287 DOI: 10.1088/1741-2552/aabfa5] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
OBJECTIVE For patients with amyotrophic lateral sclerosis (ALS) who are suffering from severe communication or motor problems, brain-computer interfaces (BCIs) can improve the quality of life and patient autonomy. However, current BCI systems are not as widely used as their potential and patient demand would let assume. This underutilization is a result of technological as well as user-based limitations but also of the comparatively poor performance of currently existing BCIs in patients with late-stage ALS, particularly in the locked-in state. APPROACH Here we review a broad range of electrophysiological studies in ALS patients with the aim to identify electrophysiological correlates of ALS-related neurodegeneration in motor and non-motor brain regions in to better understand potential neurophysiological limitations of current BCI systems for ALS patients. To this end we analyze studies in ALS patients that investigated basic sensory evoked potentials, resting-state and task-based paradigms using electroencephalography or electrocorticography for basic research purposes as well as for brain-computer interfacing. Main results and significance. Our review underscores that, similarly to mounting evidence from neuroimaging and neuropathology, electrophysiological measures too indicate neurodegeneration in non-motor areas in ALS. Furthermore, we identify an unexpected gap of basic and advanced electrophysiological studies in late-stage ALS patients, particularly in the locked-in state. We propose a research strategy on how to fill this gap in order to improve the design and performance of future BCI systems for this patient group.
Collapse
Affiliation(s)
- Philipp Kellmeyer
- Translational Neurotechnology Lab, Department of Neurosurgery, Medical Center-University of Freiburg, Freiburg im Breisgau, Germany. Cluster of Excellence BrainLinks-BrainTools, University of Freiburg, Freiburg im Breisgau, Germany
| | | | | | | | | |
Collapse
|
105
|
Koenig AM, Nobuhara CK, Williams VJ, Arnold SE. Biomarkers in Alzheimer's, Frontotemporal, Lewy Body, and Vascular Dementias. FOCUS: JOURNAL OF LIFE LONG LEARNING IN PSYCHIATRY 2018; 16:164-172. [PMID: 31975911 DOI: 10.1176/appi.focus.20170048] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
This article reviews the current evidence base for biomarkers of the most common causes of dementia in later life: Alzheimer's disease (AD), frontotemporal lobar degenerations, Lewy body dementias, and vascular cognitive impairment and dementia. Biomarkers are objectively measurable indicators of normal physiology, pathological processes, or response to an intervention. Ideally, they are sensitive, specific, easy to obtain, and closely reflect the underlying biological processes of interest. While such markers are well established and in broad clinical use for common disorders in general medicine (e.g., thallium stress tests for coronary artery disease or serum blood urea nitrogen and creatinine for renal failure), analogous, validated markers for AD or other common dementias are limited, although biomarkers in research settings and specialty dementia clinics are progressing toward clinical use. By way of introducing current and future biomarkers for dementias of later life, this article will benefit the practicing clinician by increasing awareness of the availability and utility of current and emerging biomarkers in dementia diagnosis and prognosis and for monitoring new disease-modifying therapeutics that arrive in the clinic over the coming decade.
Collapse
Affiliation(s)
- Aaron M Koenig
- All authors are with the MassGeneral Institute for Neurodegenerative Disease (MIND), Department of Neurology, Massachusetts General Hospital, Boston
| | - Chloe K Nobuhara
- All authors are with the MassGeneral Institute for Neurodegenerative Disease (MIND), Department of Neurology, Massachusetts General Hospital, Boston
| | - Victoria J Williams
- All authors are with the MassGeneral Institute for Neurodegenerative Disease (MIND), Department of Neurology, Massachusetts General Hospital, Boston
| | - Steven E Arnold
- All authors are with the MassGeneral Institute for Neurodegenerative Disease (MIND), Department of Neurology, Massachusetts General Hospital, Boston
| |
Collapse
|
106
|
Neuropathological Assessment as an Endpoint in Clinical Trial Design. Methods Mol Biol 2018. [PMID: 29512079 DOI: 10.1007/978-1-4939-7704-8_18] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register]
Abstract
Different neurodegenerative conditions can have complex, overlapping clinical presentations that make accurate diagnosis during life very challenging. For this reason, confirmation of the clinical diagnosis still requires postmortem verification. This is particularly relevant for clinical trials of novel therapeutics where it is important to ascertain what disease and/or pathology modifying effects the therapeutics have had. Furthermore, it is important to confirm that patients in the trial actually had the correct clinical diagnosis as this will have a major bearing on the interpretation of trial results. Here we present a simple protocol for pathological assessment of neurodegenerative changes.
Collapse
|
107
|
Meeter LHH, Vijverberg EG, Del Campo M, Rozemuller AJM, Donker Kaat L, de Jong FJ, van der Flier WM, Teunissen CE, van Swieten JC, Pijnenburg YAL. Clinical value of neurofilament and phospho-tau/tau ratio in the frontotemporal dementia spectrum. Neurology 2018. [PMID: 29514947 PMCID: PMC5890612 DOI: 10.1212/wnl.0000000000005261] [Citation(s) in RCA: 95] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Objective To examine the clinical value of neurofilament light chain (NfL) and the phospho-tau/total tau ratio (p/t-tau) across the entire frontotemporal dementia (FTD) spectrum in a large, well-defined cohort. Methods CSF NfL and p/t-tau levels were studied in 361 patients with FTD: 179 behavioral variant FTD, 17 FTD with motor neuron disease (FTD-MND), 36 semantic variant primary progressive aphasia (PPA), 19 nonfluent variant PPA, 4 logopenic variant PPA (lvPPA), 42 corticobasal syndrome, and 64 progressive supranuclear palsy. Forty-five cognitively healthy controls were also included. Definite pathology was known in 68 patients (49 frontotemporal lobar degeneration [FTLD]-TDP, 18 FTLD-tau, 1 FTLD-FUS). Results NfL was higher in all diagnoses, except lvPPA (n = 4), than in controls, equally elevated in behavioral variant FTD, semantic variant PPA, nonfluent variant PPA, and corticobasal syndrome, and highest in FTD-MND. The p/t-tau was lower in all clinical groups, except lvPPA, than in controls and lowest in FTD-MND. NfL did not discriminate between TDP and tau pathology, while the p/t-tau ratio had a good specificity (76%) and moderate sensitivity (67%). Both high NfL and low p/t-tau were associated with poor survival (hazard ratio on tertiles 1.7 for NfL, 0.7 for p/t-tau). Conclusion NfL and p/t-tau similarly discriminated FTD from controls, but not between clinical subtypes, apart from FTD-MND. Both markers predicted survival and are promising monitoring biomarkers for clinical trials. Of note, p/t-tau, but not NfL, was specific to discriminate TDP from tau pathology in vivo. Classification of evidence This study provides Class III evidence that for patients with cognitive issues, CSF NfL and p/t-tau levels discriminate between those with and without FTD spectrum disorders.
Collapse
Affiliation(s)
- Lieke H H Meeter
- From the Alzheimer Center and Department of Neurology (L.H.H.M., L.D.K., F.J.d.J., J.C.v.S.), Erasmus Medical Center, Rotterdam; Alzheimer Center and Department of Neurology (E.G.V., W.M.v.d.F., Y.A.L.P.), Neurochemistry Laboratory, Department of Clinical Chemistry (M.D.C., C.E.T.), Department of Pathology (A.J.M.R.), and Department of Clinical Genetics (J.C.v.S.), Neuroscience Campus Amsterdam, VU University Medical Center, Amsterdam, the Netherlands.
| | - Everard G Vijverberg
- From the Alzheimer Center and Department of Neurology (L.H.H.M., L.D.K., F.J.d.J., J.C.v.S.), Erasmus Medical Center, Rotterdam; Alzheimer Center and Department of Neurology (E.G.V., W.M.v.d.F., Y.A.L.P.), Neurochemistry Laboratory, Department of Clinical Chemistry (M.D.C., C.E.T.), Department of Pathology (A.J.M.R.), and Department of Clinical Genetics (J.C.v.S.), Neuroscience Campus Amsterdam, VU University Medical Center, Amsterdam, the Netherlands
| | - Marta Del Campo
- From the Alzheimer Center and Department of Neurology (L.H.H.M., L.D.K., F.J.d.J., J.C.v.S.), Erasmus Medical Center, Rotterdam; Alzheimer Center and Department of Neurology (E.G.V., W.M.v.d.F., Y.A.L.P.), Neurochemistry Laboratory, Department of Clinical Chemistry (M.D.C., C.E.T.), Department of Pathology (A.J.M.R.), and Department of Clinical Genetics (J.C.v.S.), Neuroscience Campus Amsterdam, VU University Medical Center, Amsterdam, the Netherlands
| | - Annemieke J M Rozemuller
- From the Alzheimer Center and Department of Neurology (L.H.H.M., L.D.K., F.J.d.J., J.C.v.S.), Erasmus Medical Center, Rotterdam; Alzheimer Center and Department of Neurology (E.G.V., W.M.v.d.F., Y.A.L.P.), Neurochemistry Laboratory, Department of Clinical Chemistry (M.D.C., C.E.T.), Department of Pathology (A.J.M.R.), and Department of Clinical Genetics (J.C.v.S.), Neuroscience Campus Amsterdam, VU University Medical Center, Amsterdam, the Netherlands
| | - Laura Donker Kaat
- From the Alzheimer Center and Department of Neurology (L.H.H.M., L.D.K., F.J.d.J., J.C.v.S.), Erasmus Medical Center, Rotterdam; Alzheimer Center and Department of Neurology (E.G.V., W.M.v.d.F., Y.A.L.P.), Neurochemistry Laboratory, Department of Clinical Chemistry (M.D.C., C.E.T.), Department of Pathology (A.J.M.R.), and Department of Clinical Genetics (J.C.v.S.), Neuroscience Campus Amsterdam, VU University Medical Center, Amsterdam, the Netherlands
| | - Frank Jan de Jong
- From the Alzheimer Center and Department of Neurology (L.H.H.M., L.D.K., F.J.d.J., J.C.v.S.), Erasmus Medical Center, Rotterdam; Alzheimer Center and Department of Neurology (E.G.V., W.M.v.d.F., Y.A.L.P.), Neurochemistry Laboratory, Department of Clinical Chemistry (M.D.C., C.E.T.), Department of Pathology (A.J.M.R.), and Department of Clinical Genetics (J.C.v.S.), Neuroscience Campus Amsterdam, VU University Medical Center, Amsterdam, the Netherlands
| | - Wiesje M van der Flier
- From the Alzheimer Center and Department of Neurology (L.H.H.M., L.D.K., F.J.d.J., J.C.v.S.), Erasmus Medical Center, Rotterdam; Alzheimer Center and Department of Neurology (E.G.V., W.M.v.d.F., Y.A.L.P.), Neurochemistry Laboratory, Department of Clinical Chemistry (M.D.C., C.E.T.), Department of Pathology (A.J.M.R.), and Department of Clinical Genetics (J.C.v.S.), Neuroscience Campus Amsterdam, VU University Medical Center, Amsterdam, the Netherlands
| | - Charlotte E Teunissen
- From the Alzheimer Center and Department of Neurology (L.H.H.M., L.D.K., F.J.d.J., J.C.v.S.), Erasmus Medical Center, Rotterdam; Alzheimer Center and Department of Neurology (E.G.V., W.M.v.d.F., Y.A.L.P.), Neurochemistry Laboratory, Department of Clinical Chemistry (M.D.C., C.E.T.), Department of Pathology (A.J.M.R.), and Department of Clinical Genetics (J.C.v.S.), Neuroscience Campus Amsterdam, VU University Medical Center, Amsterdam, the Netherlands
| | - John C van Swieten
- From the Alzheimer Center and Department of Neurology (L.H.H.M., L.D.K., F.J.d.J., J.C.v.S.), Erasmus Medical Center, Rotterdam; Alzheimer Center and Department of Neurology (E.G.V., W.M.v.d.F., Y.A.L.P.), Neurochemistry Laboratory, Department of Clinical Chemistry (M.D.C., C.E.T.), Department of Pathology (A.J.M.R.), and Department of Clinical Genetics (J.C.v.S.), Neuroscience Campus Amsterdam, VU University Medical Center, Amsterdam, the Netherlands
| | - Yolande A L Pijnenburg
- From the Alzheimer Center and Department of Neurology (L.H.H.M., L.D.K., F.J.d.J., J.C.v.S.), Erasmus Medical Center, Rotterdam; Alzheimer Center and Department of Neurology (E.G.V., W.M.v.d.F., Y.A.L.P.), Neurochemistry Laboratory, Department of Clinical Chemistry (M.D.C., C.E.T.), Department of Pathology (A.J.M.R.), and Department of Clinical Genetics (J.C.v.S.), Neuroscience Campus Amsterdam, VU University Medical Center, Amsterdam, the Netherlands
| |
Collapse
|
108
|
Kim WS, Jary E, Pickford R, He Y, Ahmed RM, Piguet O, Hodges JR, Halliday GM. Lipidomics Analysis of Behavioral Variant Frontotemporal Dementia: A Scope for Biomarker Development. Front Neurol 2018. [PMID: 29541056 PMCID: PMC5835505 DOI: 10.3389/fneur.2018.00104] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Behavioral variant frontotemporal dementia (bvFTD) is the most prevalent form of FTD syndromes. bvFTD is characterized clinically by changes in behavior and cognition and pathologically by focal brain atrophy and concomitant loss of lipids. bvFTD is further characterized by eating abnormalities that result in dyslipidemia. Although dyslipidemia is apparent in bvFTD, very little is known about global lipid changes in bvFTD and lipid dysregulation underlying bvFTD. Here, we undertook a comprehensive lipidomics analysis of blood plasma from patients with bvFTD, patients with Alzheimer’s disease (AD) and controls, using liquid chromatography-tandem mass spectrometry, with the aim of understanding lipid dysregulation in bvFTD. In our analysis, we detected all four major classes of lipids (glycerolipids, phospholipids, sphingolipids, sterols), 17 subclasses of lipids, and 3,225 putative individual lipid species in total, as well as a group of dietary lipids. We found that the levels of numerous lipid species were significantly altered in bvFTD compared to AD and control. We found that the total abundance of triglyceride (TG) increased significantly in bvFTD, whereas phosphatidylserine and phosphatidylglycerol decreased significantly in bvFTD. These results suggest manifestation of hypertriglyceridemia and hypoalphalipoproteinemia in bvFTD. We also identified five lipid molecules—TG (16:0/16:0/16:0), diglyceride (18:1/22:0), phosphatidylcholine (32:0), phosphatidylserine (41:5), and sphingomyelin (36:4)—that could potentially be used for developing biomarkers for bvFTD. Furthermore, an analysis of plant lipids revealed significant decreases in monogalactosyldiacylglycerol and sitosteryl ester in bvFTD, indicating altered eating behavior in bvFTD. This study represents the first lipidomics analysis of bvFTD and has provided new insights into an unrecognized perturbed pathology in bvFTD, providing evidence in support of considerable lipid dysregulation in bvFTD.
Collapse
Affiliation(s)
- Woojin Scott Kim
- Brain and Mind Centre, Sydney Medical School, The University of Sydney, Sydney, NSW, Australia.,Neuroscience Research Australia, Sydney, NSW, Australia.,School of Medical Sciences, University of New South Wales, Sydney, NSW, Australia
| | - Eve Jary
- Brain and Mind Centre, Sydney Medical School, The University of Sydney, Sydney, NSW, Australia
| | - Russell Pickford
- Bioanalytical Mass Spectrometry Facility, University of New South Wales, Sydney, NSW, Australia
| | - Ying He
- Brain and Mind Centre, Sydney Medical School, The University of Sydney, Sydney, NSW, Australia
| | - Rebekah M Ahmed
- Brain and Mind Centre, Sydney Medical School, The University of Sydney, Sydney, NSW, Australia.,ARC Centre of Excellence in Cognition and Its Disorders, Sydney, NSW, Australia
| | - Olivier Piguet
- Brain and Mind Centre, Sydney Medical School, The University of Sydney, Sydney, NSW, Australia.,Neuroscience Research Australia, Sydney, NSW, Australia.,ARC Centre of Excellence in Cognition and Its Disorders, Sydney, NSW, Australia.,School of Psychology, The University of Sydney, Sydney, NSW, Australia
| | - John R Hodges
- Brain and Mind Centre, Sydney Medical School, The University of Sydney, Sydney, NSW, Australia.,Neuroscience Research Australia, Sydney, NSW, Australia.,ARC Centre of Excellence in Cognition and Its Disorders, Sydney, NSW, Australia
| | - Glenda M Halliday
- Brain and Mind Centre, Sydney Medical School, The University of Sydney, Sydney, NSW, Australia.,Neuroscience Research Australia, Sydney, NSW, Australia.,School of Medical Sciences, University of New South Wales, Sydney, NSW, Australia
| |
Collapse
|
109
|
Ferrari R, Grassi M, Graziano F, Palluzzi F, Archetti S, Bonomi E, Bruni AC, Maletta RG, Bernardi L, Cupidi C, Colao R, Rainero I, Rubino E, Pinessi L, Galimberti D, Scarpini E, Serpente M, Nacmias B, Piaceri I, Bagnoli S, Rossi G, Giaccone G, Tagliavini F, Benussi L, Binetti G, Ghidoni R, Singleton A, Hardy J, Momeni P, Padovani A, Borroni B. Effects of Multiple Genetic Loci on Age at Onset in Frontotemporal Dementia. J Alzheimers Dis 2018; 56:1271-1278. [PMID: 28128768 DOI: 10.3233/jad-160949] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
In frontotemporal dementia (FTD), age at disease onset (AAO) is unpredictable in both early and late-onset cases; AAO variability is found even in autosomal dominant FTD. The present study was aimed at identifying genetic modifiers modulating AAO in a large cohort of Italian FTD patients. We conducted an association analysis on 411 FTD patients, belonging to 7 Italian Centers, and for whom AAO was available. Population structure was evaluated by principal component analysis to infer continuous axes of genetic variation, and single linear regression models were applied. A genetic score (GS) was calculated on the basis of suggestive single nucleotide polymorphisms (SNPs) found by association analyses. GS showed genome-wide significant slope decrease by -3.86 (95% CI: -4.64 to -3.07, p < 2×10-16) per standard deviation of the GS for 6 SNPs mapping to genes involved in neuronal development and signaling, axonal myelinization, and glutamatergic/GABA neurotransmission. An increase of the GS was associated with a decrease of the AAO. Our data indicate that there is indeed a genetic component that underpins and modulates up to 14.5% of variability of AAO in Italian FTD. Future studies on genetic modifiers in FTD are warranted.
Collapse
Affiliation(s)
- Raffaele Ferrari
- Department of Molecular Neuroscience, Institute of Neurology, UCL, London, UK
| | - Mario Grassi
- Department of Brain and Behavioural Sciences, Medical and Genomic Statistics Unit, University of Pavia, Pavia, Italy
| | - Francesca Graziano
- Department of Brain and Behavioural Sciences, Medical and Genomic Statistics Unit, University of Pavia, Pavia, Italy
| | - Fernando Palluzzi
- Department of Brain and Behavioural Sciences, Medical and Genomic Statistics Unit, University of Pavia, Pavia, Italy
| | - Silvana Archetti
- Department of Laboratories, III Laboratory of Analysis, Brescia Hospital, Brescia, Italy
| | - Elisa Bonomi
- Department of Clinical and Experimental Science, Neurology Unit, University of Brescia, Italy
| | - Amalia C Bruni
- Neurogenetic Regional Centre ASPCZ Lamezia Terme, Lamezia Terme, Italy
| | | | - Livia Bernardi
- Neurogenetic Regional Centre ASPCZ Lamezia Terme, Lamezia Terme, Italy
| | - Chiara Cupidi
- Neurogenetic Regional Centre ASPCZ Lamezia Terme, Lamezia Terme, Italy
| | - Rosanna Colao
- Neurogenetic Regional Centre ASPCZ Lamezia Terme, Lamezia Terme, Italy
| | - Innocenzo Rainero
- Department of Neuroscience, Neurology I, University of Torino and Cittá della Salute e della Scienza di Torino, Turin, Italy
| | - Elisa Rubino
- Department of Neuroscience, Neurology I, University of Torino and Cittá della Salute e della Scienza di Torino, Turin, Italy
| | - Lorenzo Pinessi
- Department of Neuroscience, Neurology I, University of Torino and Cittá della Salute e della Scienza di Torino, Turin, Italy
| | - Daniela Galimberti
- Department of Pathophysiology and Transplantation, Neurology Unit, University of Milan, Fondazione Cà Granda, IRCCS Ospedale Policlinico, Milan, Italy
| | - Elio Scarpini
- Department of Pathophysiology and Transplantation, Neurology Unit, University of Milan, Fondazione Cà Granda, IRCCS Ospedale Policlinico, Milan, Italy
| | - Maria Serpente
- Department of Pathophysiology and Transplantation, Neurology Unit, University of Milan, Fondazione Cà Granda, IRCCS Ospedale Policlinico, Milan, Italy
| | - Benedetta Nacmias
- Department of Neuroscience, Psychology, Drug Research and Child Health, University of Florence, Florence, Italy
| | - Irene Piaceri
- Department of Neuroscience, Psychology, Drug Research and Child Health, University of Florence, Florence, Italy
| | - Silvia Bagnoli
- Department of Neuroscience, Psychology, Drug Research and Child Health, University of Florence, Florence, Italy
| | - Giacomina Rossi
- Division of Neurology V and Neuropathology, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milano, Italy
| | - Giorgio Giaccone
- Division of Neurology V and Neuropathology, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milano, Italy
| | - Fabrizio Tagliavini
- Division of Neurology V and Neuropathology, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milano, Italy
| | - Luisa Benussi
- Molecular Markers Laboratory, IRCCS Istituto Centro San Giovanni di Dio Fatebenefratelli, Brescia, Italy
| | - Giuliano Binetti
- Molecular Markers Laboratory, IRCCS Istituto Centro San Giovanni di Dio Fatebenefratelli, Brescia, Italy.,MAC Memory Center, IRCCS Istituto Centro San Giovanni di Dio-Fatebenefratelli, Brescia, Italy
| | - Roberta Ghidoni
- Molecular Markers Laboratory, IRCCS Istituto Centro San Giovanni di Dio Fatebenefratelli, Brescia, Italy
| | - Andrew Singleton
- Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, MD, USA
| | - John Hardy
- Department of Molecular Neuroscience, Institute of Neurology, UCL, London, UK
| | - Parastoo Momeni
- Department of Internal Medicine, Laboratory of Neurogenetics, Texas Tech University Health Science Center, Lubbock, TX, USA
| | - Alessandro Padovani
- Department of Clinical and Experimental Science, Neurology Unit, University of Brescia, Italy
| | - Barbara Borroni
- Department of Clinical and Experimental Science, Neurology Unit, University of Brescia, Italy
| |
Collapse
|
110
|
Kuuluvainen L, Pöyhönen M, Pasanen P, Siitonen M, Rummukainen J, Tienari PJ, Paetau A, Myllykangas L. A Novel Loss-of-Function GRN Mutation p.(Tyr229*): Clinical and Neuropathological Features. J Alzheimers Dis 2018; 55:1167-1174. [PMID: 27767988 DOI: 10.3233/jad-160647] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Mutations in the progranulin (GRN) gene represent about 5-10% of frontotemporal lobar degeneration (FTLD). We describe a proband with a novel GRN mutation c.687T>A, p.(Tyr229*), presenting with dyspraxia, dysgraphia, and dysphasia at the age of 60 and a very severe FTLD neuropathological phenotype with TDP43 inclusions. The nephew of the proband had signs of dementia and personality changes at the age of 60 and showed similar but milder FTLD pathology. Three other family members had had early-onset dementia. Gene expression studies showed decreased GRN gene expression in mutation carriers' blood samples. In conclusion, we describe a novel GRN, p.(Tyr229*) mutation, resulting in haploinsufficiency of GRN and a severe neuropathologic FTLD phenotype.
Collapse
Affiliation(s)
- Liina Kuuluvainen
- Department of Clinical Genetics, Helsinki University Central Hospital and Department of Medical Genetics, University of Helsinki, Helsinki, Finland
| | - Minna Pöyhönen
- Department of Clinical Genetics, Helsinki University Central Hospital and Department of Medical Genetics, University of Helsinki, Helsinki, Finland
| | - Petra Pasanen
- Department of Medical Biochemistry and Genetics, Institute of Biomedicine, University of Turku, and Tyks Microbiology and Genetics, Department of Medical Genetics, Turku University Hospital, Turku, Finland
| | - Maija Siitonen
- Department of Medical Biochemistry and Genetics, Institute of Biomedicine, University of Turku, and Tyks Microbiology and Genetics, Department of Medical Genetics, Turku University Hospital, Turku, Finland
| | - Jaana Rummukainen
- Department of Pathology, Kuopio University Hospital, Kuopio, Finland
| | - Pentti J Tienari
- Department of Neurology, Helsinki University Hospital, and Molecular Neurology, Research Programs Unit, Biomedicum, University of Helsinki, Helsinki, Finland
| | - Anders Paetau
- Department of Pathology, University of Helsinki and HUSLAB, Helsinki, Finland
| | - Liisa Myllykangas
- Department of Pathology, University of Helsinki and HUSLAB, Helsinki, Finland
| |
Collapse
|
111
|
Forrest SL, Kril JJ, Stevens CH, Kwok JB, Hallupp M, Kim WS, Huang Y, McGinley CV, Werka H, Kiernan MC, Götz J, Spillantini MG, Hodges JR, Ittner LM, Halliday GM. Retiring the term FTDP-17 as MAPT mutations are genetic forms of sporadic frontotemporal tauopathies. Brain 2018; 141:521-534. [PMID: 29253099 PMCID: PMC5888940 DOI: 10.1093/brain/awx328] [Citation(s) in RCA: 108] [Impact Index Per Article: 15.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2017] [Revised: 09/18/2017] [Accepted: 10/18/2017] [Indexed: 01/08/2023] Open
Abstract
See Josephs (doi:10.1093/brain/awx367) for a scientific commentary on this article.In many neurodegenerative disorders, familial forms have provided important insights into the pathogenesis of their corresponding sporadic forms. The first mutations associated with frontotemporal lobar degeneration (FTLD) were found in the microtubule-associated protein tau (MAPT) gene on chromosome 17 in families with frontotemporal degeneration and parkinsonism (FTDP-17). However, it was soon discovered that 50% of these families had a nearby mutation in progranulin. Regardless, the original FTDP-17 nomenclature has been retained for patients with MAPT mutations, with such patients currently classified independently from the different sporadic forms of FTLD with tau-immunoreactive inclusions (FTLD-tau). The separate classification of familial FTLD with MAPT mutations implies that familial forms cannot inform on the pathogenesis of the different sporadic forms of FTLD-tau. To test this assumption, this study pathologically assessed all FTLD-tau cases with a known MAPT mutation held by the Sydney and Cambridge Brain Banks, and compared them to four cases of four subtypes of sporadic FTLD-tau, in addition to published case reports. Ten FTLD-tau cases with a MAPT mutation (K257T, S305S, P301L, IVS10+16, R406W) were screened for the core differentiating neuropathological features used to diagnose the different sporadic FTLD-tau subtypes to determine whether the categorical separation of MAPT mutations from sporadic FTLD-tau is valid. Compared with sporadic cases, FTLD-tau cases with MAPT mutations had similar mean disease duration but were younger at age of symptom onset (55 ± 4 years versus 70 ± 6 years). Interestingly, FTLD-tau cases with MAPT mutations had similar patterns and severity of neuropathological features to sporadic FTLD-tau subtypes and could be classified into: Pick's disease (K257T), corticobasal degeneration (S305S, IVS10+16, R406W), progressive supranuclear palsy (S305S) or globular glial tauopathy (P301L, IVS10+16). The finding that the S305S mutation could be classified into two tauopathies suggests additional modifying factors. Assessment of our cases and previous reports suggests that distinct MAPT mutations result in particular FTLD-tau subtypes, supporting the concept that they are likely to inform on the varied cellular mechanisms involved in distinctive forms of sporadic FTLD-tau. As such, FTLD-tau cases with MAPT mutations should be considered familial forms of FTLD-tau subtypes rather than a separate FTDP-17 category, and continued research on the effects of different mutations more focused on modelling their impact to produce the very different sporadic FTLD-tau pathologies in animal and cellular models.
Collapse
Affiliation(s)
- Shelley L Forrest
- Charles Perkins Centre and Discipline of Pathology, Sydney Medical School, University of Sydney, Australia
| | - Jillian J Kril
- Charles Perkins Centre and Discipline of Pathology, Sydney Medical School, University of Sydney, Australia
| | - Claire H Stevens
- Dementia Research Unit, School of Medical Sciences, University of New South Wales, Australia
| | - John B Kwok
- Brain and Mind Centre and Central Clinical School, Sydney Medical School, University of Sydney, Australia
- Neuroscience Research Australia, Sydney, Australia
- School of Medical Sciences, University of New South Wales, Australia
| | - Marianne Hallupp
- Brain and Mind Centre and Central Clinical School, Sydney Medical School, University of Sydney, Australia
| | - Woojin S Kim
- Brain and Mind Centre and Central Clinical School, Sydney Medical School, University of Sydney, Australia
- Neuroscience Research Australia, Sydney, Australia
- School of Medical Sciences, University of New South Wales, Australia
| | - Yue Huang
- School of Medical Sciences, University of New South Wales, Australia
| | - Ciara V McGinley
- Charles Perkins Centre and Discipline of Pathology, Sydney Medical School, University of Sydney, Australia
| | - Hellen Werka
- Charles Perkins Centre and Discipline of Pathology, Sydney Medical School, University of Sydney, Australia
| | - Matthew C Kiernan
- Brain and Mind Centre and Central Clinical School, Sydney Medical School, University of Sydney, Australia
| | - Jürgen Götz
- Clem Jones Centre for Ageing Dementia Research, Queensland Brain Institute, The University of Queensland, Australia
| | | | - John R Hodges
- Brain and Mind Centre and Central Clinical School, Sydney Medical School, University of Sydney, Australia
- Neuroscience Research Australia, Sydney, Australia
- School of Medical Sciences, University of New South Wales, Australia
| | - Lars M Ittner
- Dementia Research Unit, School of Medical Sciences, University of New South Wales, Australia
- Neuroscience Research Australia, Sydney, Australia
| | - Glenda M Halliday
- Brain and Mind Centre and Central Clinical School, Sydney Medical School, University of Sydney, Australia
- Neuroscience Research Australia, Sydney, Australia
- School of Medical Sciences, University of New South Wales, Australia
| |
Collapse
|
112
|
López-González I, Palmeira A, Aso E, Carmona M, Fernandez L, Ferrer I. FOXP2 Expression in Frontotemporal Lobar Degeneration-Tau. J Alzheimers Dis 2018; 54:471-5. [PMID: 27497476 DOI: 10.3233/jad-160274] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
FOXP2 is altered in a variety of language disorders. We found reduced mRNA and protein expression of FOXP2 in frontal cortex area 8 in Pick's disease, and frontotemporal lobar degeneration-tau linked to P301L mutation presenting with language impairment in comparison with age-matched controls and cases with parkinsonian variant progressive supranuclear palsy. Foxp2 mRNA and protein are also reduced with disease progression in the somatosensory cortex in transgenic mice bearing the P301S mutation in MAPT when compared with wild-type littermates. Our findings support the presence of FOXP2 expression abnormalities in sporadic and familial frontotemporal degeneration tauopathies.
Collapse
Affiliation(s)
- Irene López-González
- Institute of Neuropathology, Bellvitge University Hospital-IDIBELL, L'Hospitalet de Llobregat, Barcelona, Spain
| | - Andre Palmeira
- Universidade Federal de Ciências da Saúde de Porto Alegre (UFCSPA), Porto Alegre, Brazil
| | - Ester Aso
- Institute of Neuropathology, Bellvitge University Hospital-IDIBELL, L'Hospitalet de Llobregat, Barcelona, Spain
| | - Margarita Carmona
- Institute of Neuropathology, Bellvitge University Hospital-IDIBELL, L'Hospitalet de Llobregat, Barcelona, Spain
| | - Liana Fernandez
- Universidade Federal de Ciências da Saúde de Porto Alegre (UFCSPA), Porto Alegre, Brazil
| | - Isidro Ferrer
- Institute of Neuropathology, Bellvitge University Hospital-IDIBELL, L'Hospitalet de Llobregat, Barcelona, Spain.,University of Barcelona, L'Hospitalet de Llobregat, Barcelona, Spain.,CIBERNED, Instituto Carlos III, Madrid, Spain
| |
Collapse
|
113
|
Cui R, Tuo M, Li P, Zhou C. Association between TBK1 mutations and risk of amyotrophic lateral sclerosis/frontotemporal dementia spectrum: a meta-analysis. Neurol Sci 2018; 39:811-820. [PMID: 29349657 DOI: 10.1007/s10072-018-3246-0] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2017] [Accepted: 01/05/2018] [Indexed: 12/13/2022]
Abstract
Recently, mutations in TBK1 (TANK-binding kinase 1) have been reported to be a cause of amyotrophic lateral sclerosis/frontotemporal dementia (ALS/FTD) spectrum, but the relationship between them remains unclear owing to the small sample size and low mutation rate. Therefore, we performed a two-stage meta-analysis to investigate the frequency of TBK1 mutations in ALS/FTD patients and the association between the mutations and risk of ALS/FTD spectrum. In the first stage, 12 studies involving 4173 ALS/FTD patients were included. The frequencies of loss of function (LoF) and missense mutations were 1.0% (95% CI 0.6-1.7%) and 1.8% (95% CI 0.9-3.4%) in ALS/FTD patients respectively. Subgroup analysis suggested a higher prevalence of TBK1 mutations in European patients than that in Asian patients. In the second stage, 7 studies involving 3146 cases and 4856 controls were enrolled. Results showed that TBK1 LoF mutations were associated with a significant increased risk for ALS/FTD spectrum (OR 11.78; 95% CI 4.21-33.00; p < 0.0001), while TBK1 missense mutations were associated with a moderately increased susceptibility for ALS/FTD spectrum (OR 1.62; 95% CI 1.19-2.19; p = 0.002). In conclusion, TBK1 LoF and missense mutations are not frequently found in ALS/FTD patients, and both of them are associated with an increased risk for ALS/FTD spectrum.
Collapse
Affiliation(s)
- Rongrong Cui
- Department of Neurology, The Affiliated Hospital of Qingdao University, Number 16 Jiangsu Road, Qingdao, Shandong Province, 266003, China
| | - Miao Tuo
- Department of Neurology, The Affiliated Hospital of Qingdao University, Number 16 Jiangsu Road, Qingdao, Shandong Province, 266003, China
| | - Pengfei Li
- Department of Neurology, The Affiliated Hospital of Qingdao University, Number 16 Jiangsu Road, Qingdao, Shandong Province, 266003, China
| | - Chang Zhou
- Department of Neurology, The Affiliated Hospital of Qingdao University, Number 16 Jiangsu Road, Qingdao, Shandong Province, 266003, China.
| |
Collapse
|
114
|
Abstract
Alzheimer's disease, the commonest cause of dementia, is a growing global health concern with huge implications for individuals and society. In this review, current understanding of the epidemiology, genetics, pathology and pathogenesis of Alzheimer's disease is outlined, before its clinical presentation and current treatment strategies are discussed. Finally, the review discusses how our enhanced understanding of Alzheimer pathogenesis, including the recognition of a protracted preclinical phase, is informing new therapeutic strategies with the aim of moving from treatment to prevention.
Collapse
Affiliation(s)
- C A Lane
- Dementia Research Centre, UCL Institute of Neurology, London, UK
| | - J Hardy
- Reta Lila Weston Research Laboratories, Department of Molecular Neuroscience, UCL Institute of Neurology, London, UK
| | - J M Schott
- Dementia Research Centre, UCL Institute of Neurology, London, UK
| |
Collapse
|
115
|
Lo Piccolo L. Drosophila as a Model to Gain Insight into the Role of lncRNAs in Neurological Disorders. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2018; 1076:119-146. [PMID: 29951818 DOI: 10.1007/978-981-13-0529-0_8] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
It is now clear that the majority of transcription in humans results in the production of long non-protein-coding RNAs (lncRNAs) with a variable length spanning from 200 bp up to several kilobases. To date, we have a limited understanding of the lncRNA function, but a huge number of evidences have suggested that lncRNAs represent an outstanding asset for cells. In particular, temporal and spatial expression of lncRNAs appears to be important for proper neurological functioning. Stunningly, abnormal lncRNA function has been found as being critical for the onset of neurological disorders. This chapter focus on the lncRNAs with a role in diseases affecting the central nervous system with particular regard for the lncRNAs causing those neurodegenerative diseases that exhibit dementia and/or motor dysfunctions. A specific section will be dedicated to the human neuronal lncRNAs that have been modelled in Drosophila. Finally, even if only few examples have been reported so far, an overview of the Drosophila lncRNAs with neurological functions will be also included in this chapter.
Collapse
Affiliation(s)
- Luca Lo Piccolo
- Department of Neurotherapeutics, Osaka University Graduate School of Medicine 2-2 Yamadaoka, Suita Osaka, 565-0871, Japan.
| |
Collapse
|
116
|
Young JJ, Lavakumar M, Tampi D, Balachandran S, Tampi RR. Frontotemporal dementia: latest evidence and clinical implications. Ther Adv Psychopharmacol 2018; 8:33-48. [PMID: 29344342 PMCID: PMC5761910 DOI: 10.1177/2045125317739818] [Citation(s) in RCA: 95] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/21/2017] [Accepted: 09/26/2017] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Frontotemporal dementia (FTD) describes a cluster of neurocognitive syndromes that present with impairment of executive functioning, changes in behavior, and a decrease in language proficiency. FTD is the second most common form of dementia in those younger than 65 years and is expected to increase in prevalence as the population ages. This goal in our review is to describe advances in the understanding of neurobiological pathology, classification, assessment, and treatment of FTD syndromes. METHODS PubMed was searched to obtain reviews and studies that pertain to advancements in genetics, neurobiology, neuroimaging, classification, and treatment of FTD syndromes. Articles were chosen with a predilection to more recent preclinical/clinical trials and systematic reviews. RESULTS Recent reviews and trials indicate a significant advancement in the understanding of molecular and neurobiological clinical correlates to variants of FTD. Genetic and histopathologic markers have only recently been discovered in the past decade. Current therapeutic modalities are limited, with most studies reporting improvement in symptoms with nonpharmacological interventions. However, a small number of studies have reported improvement of behavioral symptoms with selective serotonin reuptake inhibitor (SSRI) treatment. Stimulants may help with disinhibition, apathy, and risk-taking behavior. Memantine and cholinesterase inhibitors have not demonstrated efficacy in ameliorating FTD symptoms. Antipsychotics have been used to treat agitation and psychosis, but safety concerns and side effect profiles limit utilization in the general FTD population. Nevertheless, recent breakthroughs in the understanding of FTD pathology have led to developments in pharmacological interventions that focus on producing treatments with autoimmune, genetic, and molecular targets. CONCLUSION FTD is an underdiagnosed group of neurological syndromes comprising multiple variants with distinct neurobiological profiles and presentations. Recent advances suggest there is an array of potential novel therapeutic targets, although data concerning their effectiveness are still preliminary or preclinical. Further studies are required to develop pharmacological interventions, as there are currently no US Food and Drug administration approved treatments to manage FTD syndromes.
Collapse
Affiliation(s)
- Juan Joseph Young
- Department of Psychiatry, MetroHealth Medical Center, Cleveland, OH, USA Case Western Reserve University, Cleveland, OH, USA
| | - Mallika Lavakumar
- Department of Psychiatry, MetroHealth Medical Center, Cleveland, OH, USA Case Western Reserve University, Cleveland, OH, USA
| | - Deena Tampi
- Mercy Regional Medical Center, 3700 Kolbe Rd, Lorain, OH 44053, USA
| | - Silpa Balachandran
- Department of Psychiatry, MetroHealth Medical Center, Cleveland, OH, USA Case Western Reserve University, Cleveland, OH, USA
| | - Rajesh R Tampi
- MetroHealth Medical Center, Case Western Reserve University School of Medicine, 2500 MetroHealth Drive, Cleveland, OH 44109, USA
| |
Collapse
|
117
|
Guo W, Fumagalli L, Prior R, Van Den Bosch L. Current Advances and Limitations in Modeling ALS/FTD in a Dish Using Induced Pluripotent Stem Cells. Front Neurosci 2017; 11:671. [PMID: 29326542 PMCID: PMC5733489 DOI: 10.3389/fnins.2017.00671] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2017] [Accepted: 11/20/2017] [Indexed: 12/11/2022] Open
Abstract
Amyotrophic lateral sclerosis (ALS) and frontotemporal dementia (FTD) are two age-dependent multifactorial neurodegenerative disorders, which are typically characterized by the selective death of motor neurons and cerebral cortex neurons, respectively. These two diseases share many clinical, genetic and pathological aspects. During the past decade, cell reprogramming technologies enabled researchers to generate human induced pluripotent stem cells (iPSCs) from somatic cells. This resulted in the unique opportunity to obtain specific neuronal and non-neuronal cell types from patients which could be used for basic research. Moreover, these in vitro models can mimic not only the familial forms of ALS/FTD, but also sporadic cases without known genetic cause. At present, there have been extensive technical advances in the generation of iPSCs, as well as in the differentiation procedures to obtain iPSC-derived motor neurons, cortical neurons and non-neuronal cells. The major challenge at this moment is to determine whether these iPSC-derived cells show relevant phenotypes that recapitulate complex diseases. In this review, we will summarize the work related to iPSC models of ALS and FTD. In addition, we will discuss potential drawbacks and solutions for establishing more trustworthy iPSC models for both ALS and FTD.
Collapse
Affiliation(s)
- Wenting Guo
- KU Leuven-Department of Neurosciences, Experimental Neurology and Leuven Institute for Neuroscience and Disease, Leuven, Belgium.,Laboratory of Neurobiology, VIB & KU Leuven Center for Brain & Disease Research, Leuven, Belgium
| | - Laura Fumagalli
- KU Leuven-Department of Neurosciences, Experimental Neurology and Leuven Institute for Neuroscience and Disease, Leuven, Belgium.,Laboratory of Neurobiology, VIB & KU Leuven Center for Brain & Disease Research, Leuven, Belgium
| | - Robert Prior
- KU Leuven-Department of Neurosciences, Experimental Neurology and Leuven Institute for Neuroscience and Disease, Leuven, Belgium.,Laboratory of Neurobiology, VIB & KU Leuven Center for Brain & Disease Research, Leuven, Belgium
| | - Ludo Van Den Bosch
- KU Leuven-Department of Neurosciences, Experimental Neurology and Leuven Institute for Neuroscience and Disease, Leuven, Belgium.,Laboratory of Neurobiology, VIB & KU Leuven Center for Brain & Disease Research, Leuven, Belgium
| |
Collapse
|
118
|
Alonso R, Pisa D, Fernández-Fernández AM, Rábano A, Carrasco L. Fungal infection in neural tissue of patients with amyotrophic lateral sclerosis. Neurobiol Dis 2017; 108:249-260. [DOI: 10.1016/j.nbd.2017.09.001] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2017] [Revised: 08/24/2017] [Accepted: 09/03/2017] [Indexed: 12/11/2022] Open
|
119
|
Clinical and biological phenotypes of frontotemporal dementia: Perspectives for disease modifying therapies. Eur J Pharmacol 2017; 817:76-85. [DOI: 10.1016/j.ejphar.2017.05.056] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2016] [Revised: 03/28/2017] [Accepted: 05/30/2017] [Indexed: 12/12/2022]
|
120
|
Ishigaki S, Fujioka Y, Okada Y, Riku Y, Udagawa T, Honda D, Yokoi S, Endo K, Ikenaka K, Takagi S, Iguchi Y, Sahara N, Takashima A, Okano H, Yoshida M, Warita H, Aoki M, Watanabe H, Okado H, Katsuno M, Sobue G. Altered Tau Isoform Ratio Caused by Loss of FUS and SFPQ Function Leads to FTLD-like Phenotypes. Cell Rep 2017; 18:1118-1131. [PMID: 28147269 DOI: 10.1016/j.celrep.2017.01.013] [Citation(s) in RCA: 78] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2015] [Revised: 12/16/2016] [Accepted: 01/05/2017] [Indexed: 12/13/2022] Open
Abstract
Fused in sarcoma (FUS) and splicing factor, proline- and glutamine-rich (SFPQ) are RNA binding proteins that regulate RNA metabolism. We found that alternative splicing of the Mapt gene at exon 10, which generates 4-repeat tau (4R-T) and 3-repeat tau (3R-T), is regulated by interactions between FUS and SFPQ in the nuclei of neurons. Hippocampus-specific FUS- or SFPQ-knockdown mice exhibit frontotemporal lobar degeneration (FTLD)-like behaviors, reduced adult neurogenesis, accumulation of phosphorylated tau, and hippocampal atrophy with neuronal loss through an increased 4R-T/3R-T ratio. Normalization of this increased ratio by 4R-T-specific silencing results in recovery of the normal phenotype. These findings suggest a biological link among FUS/SFPQ, tau isoform alteration, and phenotypic expression, which may function in the early pathomechanism of FTLD.
Collapse
Affiliation(s)
- Shinsuke Ishigaki
- Department of Neurology, Nagoya University Graduate School of Medicine, Nagoya, Aichi 466-8550, Japan; Department of Therapeutics for Intractable Neurological Disorders, Nagoya University Graduate School of Medicine, Nagoya, Aichi 466-8550, Japan.
| | - Yusuke Fujioka
- Department of Neurology, Nagoya University Graduate School of Medicine, Nagoya, Aichi 466-8550, Japan
| | - Yohei Okada
- Department of Neurology, Aichi Medical University, School of Medicine, Aichi 480-1195, Japan
| | - Yuichi Riku
- Department of Neurology, Nagoya University Graduate School of Medicine, Nagoya, Aichi 466-8550, Japan; Department of Neuropathology, Institute for Medical Science of Aging, Aichi Medical University, Aichi 480-1195, Japan
| | - Tsuyoshi Udagawa
- Department of Neurology, Nagoya University Graduate School of Medicine, Nagoya, Aichi 466-8550, Japan
| | - Daiyu Honda
- Department of Neurology, Nagoya University Graduate School of Medicine, Nagoya, Aichi 466-8550, Japan
| | - Satoshi Yokoi
- Department of Neurology, Nagoya University Graduate School of Medicine, Nagoya, Aichi 466-8550, Japan
| | - Kuniyuki Endo
- Department of Neurology, Nagoya University Graduate School of Medicine, Nagoya, Aichi 466-8550, Japan
| | - Kensuke Ikenaka
- Department of Neurology, Nagoya University Graduate School of Medicine, Nagoya, Aichi 466-8550, Japan
| | - Shinnosuke Takagi
- Department of Neurology, Nagoya University Graduate School of Medicine, Nagoya, Aichi 466-8550, Japan
| | - Yohei Iguchi
- Department of Neurology, Nagoya University Graduate School of Medicine, Nagoya, Aichi 466-8550, Japan
| | - Naruhiko Sahara
- Department of Functional Brain Imaging Research, National Institute of Radiological Sciences, Chiba 263-8555, Japan
| | - Akihiko Takashima
- Faculty of Science, Gakushuin University, Toshima-ku, Tokyo 171-8588, Japan
| | - Hideyuki Okano
- Department of Physiology, Keio University School of Medicine, Shinjuku-ku, Tokyo 160-8582, Japan
| | - Mari Yoshida
- Department of Neuropathology, Institute for Medical Science of Aging, Aichi Medical University, Aichi 480-1195, Japan
| | - Hitoshi Warita
- Department of Neurology, Tohoku University Graduate School of Medicine, Sendai, Miyagi 980-8575, Japan
| | - Masashi Aoki
- Department of Neurology, Tohoku University Graduate School of Medicine, Sendai, Miyagi 980-8575, Japan
| | - Hirohisa Watanabe
- Department of Neurology, Nagoya University Graduate School of Medicine, Nagoya, Aichi 466-8550, Japan; Brain and Mind Research Center, Nagoya University, Nagoya, Aichi 466-8550, Japan
| | - Haruo Okado
- Department of Brain Development and Neural Regeneration, Tokyo Metropolitan Institute of Medical Science, Setagaya-ku, Tokyo 156-8506, Japan
| | - Masahisa Katsuno
- Department of Neurology, Nagoya University Graduate School of Medicine, Nagoya, Aichi 466-8550, Japan
| | - Gen Sobue
- Department of Neurology, Nagoya University Graduate School of Medicine, Nagoya, Aichi 466-8550, Japan; Brain and Mind Research Center, Nagoya University, Nagoya, Aichi 466-8550, Japan; Research Division of Dementia and Neurodegenerative Disease, Nagoya University Graduate School of Medicine, Nagoya, Aichi 466-8550, Japan.
| |
Collapse
|
121
|
Preza E, Hardy J, Warner T, Wray S. Review: Induced pluripotent stem cell models of frontotemporal dementia. Neuropathol Appl Neurobiol 2017; 42:497-520. [PMID: 27291591 DOI: 10.1111/nan.12334] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2016] [Revised: 06/09/2016] [Accepted: 06/13/2016] [Indexed: 12/11/2022]
Abstract
The increasing prevalence of dementia in the ageing population combined with the lack of treatments and the burden on national health care systems globally make dementia a public health priority. Despite the plethora of important research findings published over the past two decades, the mechanisms underlying dementia are still poorly understood and the progress in pharmacological interventions is limited. Recent advances in cellular reprogramming and genome engineering technologies offer an unprecedented new paradigm in disease modeling. Induced pluripotent stem cells (iPSCs) have enabled the study of patient-derived neurons in vitro, a significant progress in the field of dementia research. The first studies using iPSCs to model dementia have recently emerged, holding promise for elucidating disease pathogenic mechanisms and accelerating drug discovery. In this review, we summarize the major findings of iPSC-based studies in frontotemporal dementia (FTD) and FTD overlapping with amyotrophic lateral sclerosis (FTD/ALS). We also discuss some of the main challenges in the use of iPSCs to model complex, late-onset neurodegenerative diseases such as dementias.
Collapse
Affiliation(s)
- E Preza
- Department of Molecular Neuroscience, UCL Institute of Neurology, London, WC1N 1PJ, UK.
| | - J Hardy
- Department of Molecular Neuroscience, UCL Institute of Neurology, London, WC1N 1PJ, UK
| | - T Warner
- Reta Lila Weston Institute of Neurological Studies, UCL Institute of Neurology, London, WC1N 1PJ, UK
| | - S Wray
- Department of Molecular Neuroscience, UCL Institute of Neurology, London, WC1N 1PJ, UK
| |
Collapse
|
122
|
Polymenidou M, Cleveland DW. Biological Spectrum of Amyotrophic Lateral Sclerosis Prions. Cold Spring Harb Perspect Med 2017; 7:cshperspect.a024133. [PMID: 28062558 DOI: 10.1101/cshperspect.a024133] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Amyotrophic lateral sclerosis (ALS) and frontotemporal lobar dementia (FTLD) are two neurodegenerative diseases with distinct clinical features but common genetic causes and neuropathological signatures. Ten years after the RNA-binding protein TDP-43 was discovered as the main protein in the cytoplasmic inclusions that characterize ALS and FTLD, their pathogenic mechanisms have never seemed more complex. Indeed, discoveries of the past decade have revolutionized our understanding of these diseases, highlighting their genetic heterogeneity and the involvement of protein-RNA assemblies in their pathogenesis. Importantly, these assemblies serve as the foci of protein misfolding and mature into insoluble structures, which further recruit native proteins, turning them into misfolded forms. This self-perpetuating mechanism is a twisted version of classical prion replication that leads to amplification of pathological protein complexes that spread throughout the neuraxis, offering a pathogenic principle that underlies the rapid disease progression that characterizes ALS and FTLD.
Collapse
Affiliation(s)
- Magdalini Polymenidou
- Institute of Molecular Life Sciences, University of Zurich, Winterthurerstrasse 190, CH-8057 Zurich, Switzerland
| | - Don W Cleveland
- Ludwig Institute for Cancer Research and Department of Cellular and Molecular Medicine, University of California, 9500 Gilman Drive, San Diego, La Jolla, California 92093-0670
| |
Collapse
|
123
|
Bajo Peñas L, Romero Mas MT, Aragonés Pasqual JM. [Patients with cognitive impairment and falls in a psychogeriatric clinic]. Rev Esp Geriatr Gerontol 2017; 52:348-349. [PMID: 28341219 DOI: 10.1016/j.regg.2017.01.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2017] [Accepted: 01/31/2017] [Indexed: 06/06/2023]
Affiliation(s)
- Lorena Bajo Peñas
- Hospital Universitari de la Santa Creu de Vic, Vic, Barcelona, España.
| | | | | |
Collapse
|
124
|
Tang SS, Li J, Tan L, Yu JT. Genetics of Frontotemporal Lobar Degeneration: From the Bench to the Clinic. J Alzheimers Dis 2017; 52:1157-76. [PMID: 27104909 DOI: 10.3233/jad-160236] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Frontotemporal lobar degeneration (FTLD) is a clinically heterogeneous neurodegenerative disease with a strong genetic component. In this review, we summarize most common mutations in MAPT, GRN, and C90RF72, as well as less common mutations in VCP, CHMP2B, TARDBP, FUS gene and so on. Several guidelines have been developed to help gene testing based on genotype-phenotype correlation, the underlying histopathological subtypes, and the neuroanatomic associations. Furthermore, we also summarize molecular pathways implicated by genes and novel targets for FTLD prevention and management in recent years.
Collapse
|
125
|
Can Astrocytes Be a Target for Precision Medicine? ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2017; 1007:111-128. [DOI: 10.1007/978-3-319-60733-7_7] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
126
|
Abstract
Sleep disorders appear to be frequent comorbidities in patients with frontotemporal dementia (FTD). Insomnia and excessive daytime sleepiness commonly occur in patients with FTD and significantly contribute to caregiver burden and burnout. Sleep is severely fragmented in FTD patients, likely secondary to behavioral disturbances, other primary sleep disorders such as sleep disordered breathing and restless leg syndrome, and neurodegeneration of nuclei involved in sleep and wakefulness. Treatment of primary sleep disorders may improve excessive daytime sleepiness and sleep quality and may improve daytime cognitive functioning. Rapid eye movement (REM) sleep behavior disorder is rare in FTD and may be confused with excessive nocturnal activity due to disturbed circadian rhythm. The relationship between FTD, sleep quality, and sleep disorders requires further study to better understand the contribution of disturbed sleep to daytime neurocognitive functioning and quality of life in FTD. Further, future studies should focus on comparing sleep disturbances between different FTD syndromes, especially behavioral variant FTD and primary progressive aphasia. Comorbid sleep disorders should be promptly sought and treated in patients with FTD to improve patient and caregiver quality of life.
Collapse
Affiliation(s)
- Stuart J McCarter
- Mayo Center for Sleep Medicine, Mayo Clinic and Foundation, 200 First Street Southwest, Rochester, MN, 55905, USA.
| | - Erik K St Louis
- Mayo Center for Sleep Medicine, Mayo Clinic and Foundation, 200 First Street Southwest, Rochester, MN, 55905, USA
- Department of Medicine, Mayo Clinic and Foundation, Rochester, MN, USA
- Department of Neurology, Mayo Clinic and Foundation, Rochester, MN, USA
| | - Bradley F Boeve
- Mayo Center for Sleep Medicine, Mayo Clinic and Foundation, 200 First Street Southwest, Rochester, MN, 55905, USA
- Department of Neurology, Mayo Clinic and Foundation, Rochester, MN, USA
| |
Collapse
|
127
|
The wide genetic landscape of clinical frontotemporal dementia: systematic combined sequencing of 121 consecutive subjects. Genet Med 2017; 20:240-249. [PMID: 28749476 PMCID: PMC5846812 DOI: 10.1038/gim.2017.102] [Citation(s) in RCA: 52] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2017] [Accepted: 05/23/2017] [Indexed: 12/12/2022] Open
Abstract
Purpose To define the genetic spectrum and relative gene frequencies underlying clinical frontotemporal dementia (FTD). Methods We investigated the frequencies and mutations in neurodegenerative disease genes in 121 consecutive FTD subjects using an unbiased, combined sequencing approach, complemented by cerebrospinal fluid Aβ1-42 and serum progranulin measurements. Subjects were screened for C9orf72 repeat expansions, GRN and MAPT mutations, and, if negative, mutations in other neurodegenerative disease genes, by whole-exome sequencing (WES) (n = 108), including WES-based copy-number variant (CNV) analysis. Results Pathogenic and likely pathogenic mutations were identified in 19% of the subjects, including mutations in C9orf72 (n = 8), GRN (n = 7, one 11-exon macro-deletion) and, more rarely, CHCHD10, TARDBP, SQSTM1 and UBQLN2 (each n = 1), but not in MAPT or TBK1. WES also unraveled pathogenic mutations in genes not commonly linked to FTD, including mutations in Alzheimer (PSEN1, PSEN2), lysosomal (CTSF, 7-exon macro-deletion) and cholesterol homeostasis pathways (CYP27A1). Conclusion Our unbiased approach reveals a wide genetic spectrum underlying clinical FTD, including 11% of seemingly sporadic FTD. It unravels several mutations and CNVs in genes and pathways hitherto not linked to FTD. This suggests that clinical FTD might be the converging downstream result of a delicate susceptibility of frontotemporal brain networks to insults in various pathways.
Collapse
|
128
|
REM Sleep Behavior Disorder and Other Sleep Disturbances in Non-Alzheimer Dementias. CURRENT SLEEP MEDICINE REPORTS 2017. [DOI: 10.1007/s40675-017-0078-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
|
129
|
Meeter LH, Kaat LD, Rohrer JD, van Swieten JC. Imaging and fluid biomarkers in frontotemporal dementia. Nat Rev Neurol 2017. [PMID: 28621768 DOI: 10.1038/nrneurol.2017.75] [Citation(s) in RCA: 144] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Frontotemporal dementia (FTD), the second most common type of presenile dementia, is a heterogeneous neurodegenerative disease characterized by progressive behavioural and/or language problems, and includes a range of clinical, genetic and pathological subtypes. The diagnostic process is hampered by this heterogeneity, and correct diagnosis is becoming increasingly important to enable future clinical trials of disease-modifying treatments. Reliable biomarkers will enable us to better discriminate between FTD and other forms of dementia and to predict disease progression in the clinical setting. Given that different underlying pathologies probably require specific pharmacological interventions, robust biomarkers are essential for the selection of patients with specific FTD subtypes. This Review emphasizes the increasing availability and potential applications of structural and functional imaging biomarkers, and cerebrospinal fluid and blood fluid biomarkers in sporadic and genetic FTD. The relevance of new MRI modalities - such as voxel-based morphometry, diffusion tensor imaging and arterial spin labelling - in the early stages of FTD is discussed, together with the ability of these modalities to classify FTD subtypes. We highlight promising new fluid biomarkers for staging and monitoring of FTD, and underline the importance of large, multicentre studies of individuals with presymptomatic FTD. Harmonization in the collection and analysis of data across different centres is crucial for the implementation of new biomarkers in clinical practice, and will become a great challenge in the next few years.
Collapse
Affiliation(s)
- Lieke H Meeter
- Department of Neurology, Erasmus Medical Center, 's Gravendijkwal 230, 3015 CE Rotterdam, Netherlands
| | - Laura Donker Kaat
- Department of Neurology, Erasmus Medical Center, 's Gravendijkwal 230, 3015 CE Rotterdam, Netherlands.,Department of Clinical Genetics, Leiden University Medical Center, Albinusdreef 2, 2333 ZA Leiden, Netherlands
| | - Jonathan D Rohrer
- Dementia Research Centre, Department of Neurodegenerative diseases, Institute of Neurology, Queen Square, University College London, London WC1N 3BG, UK
| | - John C van Swieten
- Department of Neurology, Erasmus Medical Center, 's Gravendijkwal 230, 3015 CE Rotterdam, Netherlands.,Department of Clinical Genetics, VU University Medical Center, De Boelelaan 1118, 1081 HZ Amsterdam, Netherlands
| |
Collapse
|
130
|
Hock EM, Polymenidou M. Prion-like propagation as a pathogenic principle in frontotemporal dementia. J Neurochem 2017; 138 Suppl 1:163-83. [PMID: 27502124 PMCID: PMC6680357 DOI: 10.1111/jnc.13668] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2015] [Revised: 04/22/2016] [Accepted: 05/11/2016] [Indexed: 12/12/2022]
Abstract
Frontotemporal dementia is a devastating neurodegenerative disease causing stark alterations in personality and language. Characterized by severe atrophy of the frontal and temporal brain lobes, frontotemporal dementia (FTD) shows extreme heterogeneity in clinical presentation, genetic causes, and pathological findings. Like most neurodegenerative diseases, the initial symptoms of FTD are subtle, but increase in severity over time, as the disease progresses. Clinical progression is paralleled by exacerbation of pathological findings and the involvement of broader brain regions, which currently lack mechanistic explanation. Yet, a flurry of studies indicate that protein aggregates accumulating in neurodegenerative diseases can act as propagating entities, amplifying their pathogenic conformation, in a way similar to infectious prions. In this prion‐centric view, FTD can be divided into three subtypes, TDP‐43 or FUS proteinopathy and tauopathy. Here, we review the current evidence that FTD‐linked pathology propagates in a prion‐like manner and discuss the implications of these findings for disease progression and heterogeneity.
Frontotemporal dementia (FTD) is a progressive neurodegenerative disease causing severe personality dysfunctions, characterized by profound heterogeneity. Accumulation of tau, TDP‐43 or FUS cytoplasmic aggregates characterize molecularly distinct and non‐overlapping FTD subtypes. Here, we discuss the current evidence suggesting that prion‐like propagation and cell‐to‐cell spread of each of these cytoplasmic aggregates may underlie disease progression and heterogeneity.
This article is part of the Frontotemporal Dementia special issue.
Collapse
Affiliation(s)
- Eva-Maria Hock
- Institute of Molecular Life Sciences, University of Zurich, Zurich, Switzerland
| | | |
Collapse
|
131
|
Rainero I, Rubino E, Michelerio A, D’Agata F, Gentile S, Pinessi L. Recent advances in the molecular genetics of frontotemporal lobar degeneration. FUNCTIONAL NEUROLOGY 2017; 32:7-16. [PMID: 28380318 PMCID: PMC5505533 DOI: 10.11138/fneur/2017.32.1.007] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The term frontotemporal lobar degeneration (FTLD) describes a spectrum of neurodegenerative disorders associated with deposition of misfolded proteins in the frontal and temporal lobes. Up to 40% of FTLD patients reports a family history of neurodegeneration, and approximately 1/3 of familial cases shows an autosomal dominant pattern of inheritance of the phenotype. Over the past two decades, several causative and susceptibility genes for FTLD have been discovered, supporting the notion that genetic factors are important contributors to the disease processes. Genetic variants in three genes, MAPT, GRN and C9orf72, account for about half of familial FTLD cases. In addition, rare defects in the CHMP2B, VCP, TARDBP, SQSTM1, FUS, UBQLN, OPTN, TREM2, CHCHD10 and TBK1 genes have been described. Additional genes are expected to be found in near future. The purpose of this review is to describe recent advances in the molecular genetics of the FTLD spectrum and to discuss implications for genetic counseling.
Collapse
Affiliation(s)
- Innocenzo Rainero
- Neurology I, Department of Neuroscience “Rita Levi Montalcini”, University of Torino, Italy
- Department of Neuroscience and Mental Health, AOU Città della Salute e della Scienza, Torino, Italy
| | - Elisa Rubino
- Neurology I, Department of Neuroscience “Rita Levi Montalcini”, University of Torino, Italy
| | - Andrea Michelerio
- Neurology I, Department of Neuroscience “Rita Levi Montalcini”, University of Torino, Italy
| | - Federico D’Agata
- Neurology I, Department of Neuroscience “Rita Levi Montalcini”, University of Torino, Italy
| | - Salvatore Gentile
- Department of Neuroscience and Mental Health, AOU Città della Salute e della Scienza, Torino, Italy
| | - Lorenzo Pinessi
- Neurology I, Department of Neuroscience “Rita Levi Montalcini”, University of Torino, Italy
- Department of Neuroscience and Mental Health, AOU Città della Salute e della Scienza, Torino, Italy
| |
Collapse
|
132
|
Sudre CH, Bocchetta M, Cash D, Thomas DL, Woollacott I, Dick KM, van Swieten J, Borroni B, Galimberti D, Masellis M, Tartaglia MC, Rowe JB, Graff C, Tagliavini F, Frisoni G, Laforce R, Finger E, de Mendonça A, Sorbi S, Ourselin S, Cardoso MJ, Rohrer JD. White matter hyperintensities are seen only in GRN mutation carriers in the GENFI cohort. NEUROIMAGE-CLINICAL 2017; 15:171-180. [PMID: 28529873 PMCID: PMC5429247 DOI: 10.1016/j.nicl.2017.04.015] [Citation(s) in RCA: 61] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/22/2017] [Revised: 03/18/2017] [Accepted: 04/15/2017] [Indexed: 12/25/2022]
Abstract
Genetic frontotemporal dementia is most commonly caused by mutations in the progranulin (GRN), microtubule-associated protein tau (MAPT) and chromosome 9 open reading frame 72 (C9orf72) genes. Previous small studies have reported the presence of cerebral white matter hyperintensities (WMH) in genetic FTD but this has not been systematically studied across the different mutations. In this study WMH were assessed in 180 participants from the Genetic FTD Initiative (GENFI) with 3D T1- and T2-weighed magnetic resonance images: 43 symptomatic (7 GRN, 13 MAPT and 23 C9orf72), 61 presymptomatic mutation carriers (25 GRN, 8 MAPT and 28 C9orf72) and 76 mutation negative non-carrier family members. An automatic detection and quantification algorithm was developed for determining load, location and appearance of WMH. Significant differences were seen only in the symptomatic GRN group compared with the other groups with no differences in the MAPT or C9orf72 groups: increased global load of WMH was seen, with WMH located in the frontal and occipital lobes more so than the parietal lobes, and nearer to the ventricles rather than juxtacortical. Although no differences were seen in the presymptomatic group as a whole, in the GRN cohort only there was an association of increased WMH volume with expected years from symptom onset. The appearance of the WMH was also different in the GRN group compared with the other groups, with the lesions in the GRN group being more similar to each other. The presence of WMH in those with progranulin deficiency may be related to the known role of progranulin in neuroinflammation, although other roles are also proposed including an effect on blood-brain barrier permeability and the cerebral vasculature. Future studies will be useful to investigate the longitudinal evolution of WMH and their potential use as a biomarker as well as post-mortem studies investigating the histopathological nature of the lesions. In genetic FTD white matter hyperintensities (WMH) are found most prominently in symptomatic patients with GRN mutations. Frontal and occipital lobes are the most affected regions. WMH are more likely to occur close to the ventricles. WMH have a more homogenous appearance possibly suggestive of inflammatory rather than vascular lesions.
Collapse
Affiliation(s)
- Carole H Sudre
- Dementia Research Centre, Department of Neurodegenerative Disease, UCL Institute of Neurology, Queen Square, London, UK; Centre for Medical Image Computing, University College London, UK
| | - Martina Bocchetta
- Dementia Research Centre, Department of Neurodegenerative Disease, UCL Institute of Neurology, Queen Square, London, UK
| | - David Cash
- Dementia Research Centre, Department of Neurodegenerative Disease, UCL Institute of Neurology, Queen Square, London, UK; Centre for Medical Image Computing, University College London, UK
| | - David L Thomas
- Dementia Research Centre, Department of Neurodegenerative Disease, UCL Institute of Neurology, Queen Square, London, UK; Centre for Medical Image Computing, University College London, UK
| | - Ione Woollacott
- Dementia Research Centre, Department of Neurodegenerative Disease, UCL Institute of Neurology, Queen Square, London, UK
| | - Katrina M Dick
- Dementia Research Centre, Department of Neurodegenerative Disease, UCL Institute of Neurology, Queen Square, London, UK
| | | | | | - Daniela Galimberti
- Dept. of Pathophysiology and Transplantation, "Dino Ferrari" Center, University of Milan, Fondazione Cà Granda, IRCCS Ospedale Maggiore Policlinico, Milan, Italy
| | - Mario Masellis
- Cognitive Neurology Research Unit, Sunnybrook Health Sciences Centre, Hurvitz Brain Sciences Research Program, Sunnybrook Research Institute; Department of Medicine, University of Toronto, Canada
| | | | | | - Caroline Graff
- Karolinska Institutet, Stockholm, Sweden; Karolinska Institutet, Department NVS, Center for Alzheimer Research, Division of Neurogeriatrics, Sweden; Department of Geriatric Medicine, Karolinska University Hospital, Stockholm, Sweden
| | | | | | | | | | | | - Sandro Sorbi
- Department of Neurosciences, Psychology, Drug Research and Child Health (NEUROFARBA), University of Florence, Florence, Italy; IRCCS Don Gnocchi, Firenze, Italy
| | - Sébastien Ourselin
- Dementia Research Centre, Department of Neurodegenerative Disease, UCL Institute of Neurology, Queen Square, London, UK; Centre for Medical Image Computing, University College London, UK
| | - M Jorge Cardoso
- Dementia Research Centre, Department of Neurodegenerative Disease, UCL Institute of Neurology, Queen Square, London, UK; Centre for Medical Image Computing, University College London, UK
| | - Jonathan D Rohrer
- Dementia Research Centre, Department of Neurodegenerative Disease, UCL Institute of Neurology, Queen Square, London, UK.
| | | |
Collapse
|
133
|
Pan C, Jiao B, Xiao T, Hou L, Zhang W, Liu X, Xu J, Tang B, Shen L. Mutations of CCNF gene is rare in patients with amyotrophic lateral sclerosis and frontotemporal dementia from Mainland China. Amyotroph Lateral Scler Frontotemporal Degener 2017; 18:265-268. [PMID: 28281833 DOI: 10.1080/21678421.2017.1293111] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
OBJECTIVE Mutations of the cyclin F (CCNF) gene were recently identified to be associated with amyotrophic lateral sclerosis (ALS) and frontotemporal dementia (FTD) in Western and Japanese populations. The rare protein-altering variants frequency is 0.6 to 3.3% in ALS and FTD from these diverse geographic populations while no systematic analysis of CCNF variants were conducted in the Chinese population. METHODS We screened all exons of CCNF in a cohort of 269 cases (including 181 ALS and 88 FTD) from Mainland China using Sanger sequencing. RESULTS A rare heterozygous variant (c.481G > A, p.G161R) was detected in a sporadic ALS case with a frequency of 0.6%, while no mutation was identified in patients with FTD. The same variant was also found in a sporadic ALS patient from America. CONCLUSIONS Our result indicates that the mutation of CCNF is rare in patients with ALS and FTD from Mainland China.
Collapse
Affiliation(s)
- Chuzheng Pan
- a Department of Neurology , Xiangya Hospital, Central South University , Changsha , China
| | - Bin Jiao
- a Department of Neurology , Xiangya Hospital, Central South University , Changsha , China
| | - Tingting Xiao
- a Department of Neurology , Xiangya Hospital, Central South University , Changsha , China
| | - Lihua Hou
- a Department of Neurology , Xiangya Hospital, Central South University , Changsha , China
| | - Weiwei Zhang
- a Department of Neurology , Xiangya Hospital, Central South University , Changsha , China
| | - Xi Liu
- a Department of Neurology , Xiangya Hospital, Central South University , Changsha , China
| | - Jun Xu
- d Department of Neurology , Brain Centre, Neurological Institute, Northern Jiangsu Province Hospital , Yangzhou , China , and.,e Jiangsu Key Laboratory of Integrated Traditional Chinese and Western Medicine for Prevention and Treatment of Senile Diseases, School of Medicine, Yangzhou University , Yangzhou , 225001 , China
| | - Beisha Tang
- a Department of Neurology , Xiangya Hospital, Central South University , Changsha , China.,b Key Laboratory of Hunan Province in Neurodegenerative Disorders , Central South University , Changsha , China.,c State Key Laboratory of Medical Genetics , Changsha , China
| | - Lu Shen
- a Department of Neurology , Xiangya Hospital, Central South University , Changsha , China.,b Key Laboratory of Hunan Province in Neurodegenerative Disorders , Central South University , Changsha , China.,c State Key Laboratory of Medical Genetics , Changsha , China
| |
Collapse
|
134
|
Abstract
TDP-43 is a dimeric nuclear protein that plays a central role in RNA metabolism. In recent years, this protein has become a focal point of research in the amyotrophic lateral sclerosis and frontotemporal dementia (ALS/FTD) disease spectrum, as pathognomonic inclusions within affected neurons contain post-translationally modified TDP-43. A key question in TDP-43 research involves determining the mechanisms and triggers that cause TDP-43 to form pathological aggregates. This review gives a brief overview of the physiological and pathological roles of TDP-43 and focuses on the structural features of its protein domains and how they may contribute to normal protein function and to disease. A special emphasis is placed on the C-terminal prion-like region thought to be implicated in pathology, as it is where nearly all ALS/FTD-associated mutations reside. Recent structural studies of this domain revealed its crucial role in the formation of phase-separated liquid droplets through a partially populated α-helix. This new discovery provides further support for the theory that liquid droplets such as stress granules may be precursors to pathological aggregates, linking environmental effects such as stress to the potential etiology of the disease. The transition of TDP-43 among soluble, droplet, and aggregate phases and the implications of these transitions for pathological aggregation are summarized and discussed.
Collapse
Affiliation(s)
- Yulong Sun
- Department of Medical Biophysics, University of Toronto , Toronto, Ontario M5G1L7, Canada
| | - Avijit Chakrabartty
- Department of Medical Biophysics, University of Toronto , Toronto, Ontario M5G1L7, Canada.,Department of Biochemistry, University of Toronto , Toronto, Ontario M5G1L7, Canada
| |
Collapse
|
135
|
Kovacs GG, Kwong LK, Grossman M, Irwin DJ, Lee EB, Robinson JL, Suh E, Van Deerlin VM, Lee VM, Trojanowski JQ. Tauopathy with hippocampal 4-repeat tau immunoreactive spherical inclusions: a report of three cases. Brain Pathol 2017; 28:274-283. [PMID: 28019685 DOI: 10.1111/bpa.12482] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2016] [Accepted: 12/12/2016] [Indexed: 11/29/2022] Open
Abstract
Tauopathies are a major group of neurodegenerative proteinopathies characterized by the accumulation of abnormal and hyperphosphorylated tau proteins in the brain. Tau pathology is characterized as 3R (repeat) or 4R predominant or mixed 3R and 4R type. Here we report three cases lacking mutations in the microtubule associated protein tau (MAPT) gene with unusual tau pathology. The age at onset and duration of illness, respectively, were 63 and 20 years (male), 67 and 5 years (female) and 72 and 20 years (female). The clinical presentation was compatible with a diagnosis of progressive supranuclear palsy (PSP) in two subjects and with cognitive decline in all three subjects. Common neuropathological features included neuronal loss in the hippocampus and dentate gyrus associated with spherical basophilic Pick body-like inclusions showing 4R tau immunoreactivity, which was supported by the detection of predominantly 4R tau species by Western blot examination. In addition, accumulation of tau immunoreactive argyrophilic astrocytes in the hippocampus and amygdala and oligodendroglial coiled bodies in the hippocampal white matter were observed. These morphologies appeared in combination with Alzheimer disease-related pathology and subcortical tau pathology compatible with PSP. Together with a single case report in the literature, our observations on these three cases expand the spectrum of previously described tauopathies. We suggest that this tauopathy variant with hippocampal 4R tau immunoreactive spherical inclusions might contribute to the cognitive deficits in the patients reported here. The precise definition of the clinicopathological relevance of these unusual tau pathologies merits further study.
Collapse
Affiliation(s)
- Gabor G Kovacs
- Center for Neurodegenerative Disease Research, Institute on Aging and Department of Pathology & Laboratory Medicine, Philadelphia, PA.,Institute of Neurology, Medical University of Vienna, Vienna, Austria
| | - Linda K Kwong
- Center for Neurodegenerative Disease Research, Institute on Aging and Department of Pathology & Laboratory Medicine, Philadelphia, PA
| | - Murray Grossman
- Department of Neurology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - David J Irwin
- Department of Neurology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Edward B Lee
- Center for Neurodegenerative Disease Research, Institute on Aging and Department of Pathology & Laboratory Medicine, Philadelphia, PA
| | - John L Robinson
- Center for Neurodegenerative Disease Research, Institute on Aging and Department of Pathology & Laboratory Medicine, Philadelphia, PA
| | - Eunran Suh
- Center for Neurodegenerative Disease Research, Institute on Aging and Department of Pathology & Laboratory Medicine, Philadelphia, PA
| | - Vivianna M Van Deerlin
- Center for Neurodegenerative Disease Research, Institute on Aging and Department of Pathology & Laboratory Medicine, Philadelphia, PA
| | - Virginia M Lee
- Center for Neurodegenerative Disease Research, Institute on Aging and Department of Pathology & Laboratory Medicine, Philadelphia, PA
| | - John Q Trojanowski
- Center for Neurodegenerative Disease Research, Institute on Aging and Department of Pathology & Laboratory Medicine, Philadelphia, PA
| |
Collapse
|
136
|
Nilson AN, English KC, Gerson JE, Barton Whittle T, Nicolas Crain C, Xue J, Sengupta U, Castillo-Carranza DL, Zhang W, Gupta P, Kayed R. Tau Oligomers Associate with Inflammation in the Brain and Retina of Tauopathy Mice and in Neurodegenerative Diseases. J Alzheimers Dis 2017; 55:1083-1099. [PMID: 27716675 PMCID: PMC5147514 DOI: 10.3233/jad-160912] [Citation(s) in RCA: 121] [Impact Index Per Article: 15.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/12/2016] [Indexed: 12/13/2022]
Abstract
It is well-established that inflammation plays an important role in Alzheimer's disease (AD) and frontotemporal lobar dementia (FTLD). Inflammation and synapse loss occur in disease prior to the formation of larger aggregates, but the contribution of tau to inflammation has not yet been thoroughly investigated. Tau pathologically aggregates to form large fibrillar structures known as tangles. However, evidence suggests that smaller soluble aggregates, called oligomers, are the most toxic species and form prior to tangles. Furthermore, tau oligomers can spread to neighboring cells and between anatomically connected brain regions. In addition, recent evidence suggests that inspecting the retina may be a window to brain pathology. We hypothesized that there is a relationship between tau oligomers and inflammation, which are hallmarks of early disease. We conducted immunofluorescence and biochemical analyses on tauopathy mice, FTLD, and AD subjects. We showed that oligomers co-localize with astrocytes, microglia, and HMGB1, a pro-inflammatory cytokine. Additionally, we show that tau oligomers are present in the retina and are associated with inflammatory cells suggesting that the retina may be a valid non-invasive biomarker for brain pathology. These results suggest that there may be a toxic relationship between tau oligomers and inflammation. Therefore, the ability of tau oligomers to spread may initiate a feed-forward cycle in which tau oligomers induce inflammation, leading to neuronal damage, and thus more inflammation. Further mechanistic studies are warranted in order to understand this relationship, which may have critical implications for improving the treatment of tauopathies.
Collapse
Affiliation(s)
- Ashley N. Nilson
- Mitchell Center for Neurodegenerative Diseases, University of Texas Medical Branch, Galveston, TX, USA
- Departments of Neurology, Neuroscience and Cell Biology, University of Texas Medical Branch, Galveston, TX, USA
| | - Kelsey C. English
- Mitchell Center for Neurodegenerative Diseases, University of Texas Medical Branch, Galveston, TX, USA
- Departments of Neurology, Neuroscience and Cell Biology, University of Texas Medical Branch, Galveston, TX, USA
| | - Julia E. Gerson
- Mitchell Center for Neurodegenerative Diseases, University of Texas Medical Branch, Galveston, TX, USA
- Departments of Neurology, Neuroscience and Cell Biology, University of Texas Medical Branch, Galveston, TX, USA
| | - T. Barton Whittle
- Department of Ophthalmology and Visual Sciences, University of Texas Medical Branch, Galveston, TX, USA
| | - C. Nicolas Crain
- Department of Ophthalmology and Visual Sciences, University of Texas Medical Branch, Galveston, TX, USA
| | - Judy Xue
- Mitchell Center for Neurodegenerative Diseases, University of Texas Medical Branch, Galveston, TX, USA
- Departments of Neurology, Neuroscience and Cell Biology, University of Texas Medical Branch, Galveston, TX, USA
| | - Urmi Sengupta
- Mitchell Center for Neurodegenerative Diseases, University of Texas Medical Branch, Galveston, TX, USA
- Departments of Neurology, Neuroscience and Cell Biology, University of Texas Medical Branch, Galveston, TX, USA
| | - Diana L. Castillo-Carranza
- Mitchell Center for Neurodegenerative Diseases, University of Texas Medical Branch, Galveston, TX, USA
- Departments of Neurology, Neuroscience and Cell Biology, University of Texas Medical Branch, Galveston, TX, USA
| | - Wenbo Zhang
- Department of Ophthalmology and Visual Sciences, University of Texas Medical Branch, Galveston, TX, USA
| | - Praveena Gupta
- Department of Ophthalmology and Visual Sciences, University of Texas Medical Branch, Galveston, TX, USA
| | - Rakez Kayed
- Mitchell Center for Neurodegenerative Diseases, University of Texas Medical Branch, Galveston, TX, USA
- Departments of Neurology, Neuroscience and Cell Biology, University of Texas Medical Branch, Galveston, TX, USA
| |
Collapse
|
137
|
Domoto-Reilly K, Davis MY, Keene CD, Bird TD. Unusually long duration and delayed penetrance in a family with FTD and mutation in MAPT (V337M). Am J Med Genet B Neuropsychiatr Genet 2017; 174:70-74. [PMID: 26990251 PMCID: PMC5297469 DOI: 10.1002/ajmg.b.32443] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/16/2015] [Accepted: 02/25/2016] [Indexed: 11/09/2022]
Abstract
Mutations in the MAPT gene coding for the tau protein are one of the most common causes of familial frontotemporal dementia (FTD). In a previously described family with the V337M mutation in MAPT, we now report an affected woman who died at age 92 with a >40 year duration of symptoms, more than three times the mean disease duration in her family (13.8 years). Neuropathology showed the typical findings of a diffuse tauopathy. Conversely, her 67-year-old son with the same mutation remains asymptomatic more than 15 years beyond the mean age of onset in the family (51.5 years). These two cases demonstrate the marked variability in onset and duration of familial FTD and underscore the difficulties of discussing these issues with patients and families. The presumed genetic and environmental factors influencing these parameters remain largely unknown. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
| | - Marie Y. Davis
- Department of Neurology, University of Washington, Seattle, Washington
| | - C. Dirk Keene
- Department of Pathology, University of Washington, Seattle, Washington
| | - Thomas D. Bird
- Department of Neurology, University of Washington, Seattle, Washington,Department of Medicine, University of Washington, Seattle, Washington,VA Puget Sound Health Care System, Geriatric Research Education and Clinical Center, Seattle, Washington,Correspondence to: Thomas D. Bird, M.D., 1660 SColumbian Way, S-182-GRECC, Seattle, WA 98108.
| |
Collapse
|
138
|
Clinical characteristics of parkinsonism in frontotemporal dementia according to subtypes. J Neurol Sci 2016; 372:51-56. [PMID: 28017247 DOI: 10.1016/j.jns.2016.11.033] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2015] [Revised: 10/31/2016] [Accepted: 11/14/2016] [Indexed: 12/12/2022]
Abstract
BACKGROUND We investigated the prevalence of parkinsonism in frontotemporal dementia (FTD) subtypes and the cognitive and behavioral differences between FTD with and without parkinsonism in a well-structured, prospective cohort. METHODS One hundred and ninety-one FTD patients were enrolled and all patients underwent comprehensive neurological evaluations, neuropsychological tests, and the Unified Parkinson's Disease Rating Scale. RESULTS The prevalence of parkinsonism was 38.7% (74 patients), and included 33 (46.5%) behavioral variant FTD (bvFTD), 16 (24.2%) semantic dementia (SD), 19 (45.2%) progressive nonfluent aphasia (PNFA), and 6 (50%) FTD associated with motor neuron disease (FTD-MND). SD patients with parkinsonism had higher CDR sum of boxes scores (9.7±4.5 vs 6.2±4.5, p=0.024), frontal behavioral inventory total score (33.7±20.5 vs 24.3±14.5, p=0.045), and executive function score of frontal executive dysfunction, disinhibition, and apathy (28.9±13.7 vs 19.2±12.9, p=0.021) than those without parkinsonism. Seoul Instrumental Activities of Daily Living score (bvFTD: 23.5±11.7 vs 17.3±11.3, p=0.031, SD: 23.1±11.1 vs 11.3±9.3, p=0.005) was higher for bvFTD and SD with parkinsonism than for those without parkinsonism. CONCLUSIONS Parkinsonism is found to be more common in patients with bvFTD, PNFA, and FTD-MND patients than those with SD. Behavioral disturbances were more prominent in SD with parkinsonism than without. Additional studies are needed to determine the pathomechanism and optimal treatment of parkinsonism in different FTD subtypes.
Collapse
|
139
|
Lopez G, Bayulkem K, Hallett M. Progressive supranuclear palsy (PSP): Richardson syndrome and other PSP variants. Acta Neurol Scand 2016; 134:242-9. [PMID: 27070344 DOI: 10.1111/ane.12546] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/24/2015] [Indexed: 01/05/2023]
Abstract
Phenotypic heterogeneity of progressive supranuclear palsy (PSP) has been increasingly reported in the literature and can be the source of incorrect clinical diagnosis particularly in the early stages of the disease when the classically associated symptoms of early falls and supranuclear gaze palsy may not be apparent. In addition to Richardson syndrome (RS), several atypical clinical phenotypes have been described. Advances in genetic, neuroimaging, and biochemical/molecular technologies contribute to the identification of these clinical subtypes in the context of typical PSP pathological findings. Our goal is to review the phenomenology reported in the literature that is associated with confirmed histopathological changes consistent with a PSP diagnosis and to highlight the clinical spectrum of PSP. A systematic review of the literature in PubMed through July 2015 using MeSH terms and key words related to PSP was conducted. Articles describing PSP classifications, diagnostic criteria, and case reports were reviewed and summarized. Additional PSP phenotypes not seen in recent clinicopathological studies are included. These include primary lateral sclerosis, pallido-nigro-luysian degeneration, axonal dystrophy, and multiple system atrophy in the spectrum of atypical PSP variants beyond the traditionally classified PSP subtypes. This review is intended to help with the diagnostic challenges of atypical PSP variants. We believe that large multicenter clinicopathological studies will help expand our understanding of etiology and specific mechanisms of neurodegeneration and will aid in the appropriate interpretation of outcomes when conducting clinical and basic science research.
Collapse
Affiliation(s)
- G. Lopez
- Section on Molecular Neurogenetics; Medical Genetics Branch; NHGRI Intramural Research Program; National Institutes of Health; Bethesda MD USA
| | - K. Bayulkem
- Human Motor Control Section; National Institute of Neurological Disorders and Stroke; National Institutes of Health; Bethesda MD USA
| | - M. Hallett
- Human Motor Control Section; National Institute of Neurological Disorders and Stroke; National Institutes of Health; Bethesda MD USA
| |
Collapse
|
140
|
Wen X, Westergard T, Pasinelli P, Trotti D. Pathogenic determinants and mechanisms of ALS/FTD linked to hexanucleotide repeat expansions in the C9orf72 gene. Neurosci Lett 2016; 636:16-26. [PMID: 27619540 DOI: 10.1016/j.neulet.2016.09.007] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2016] [Revised: 07/12/2016] [Accepted: 09/06/2016] [Indexed: 12/12/2022]
Abstract
Amyotrophic lateral sclerosis (ALS) and frontotemporal dementia (FTD) are two apparently distinct neurodegenerative diseases, the former characterized by selective loss of motor neurons in the brain and spinal cord and the latter characterized by selective atrophy of frontal and temporal lobes. Over the years, however, growing evidence from clinical, pathological and genetic findings has suggested that ALS and FTD belong to the same clinic-pathological spectrum disorder. This concept has been further supported by the identification of the most common genetic cause for both diseases, an aberrantly expanded hexanucleotide repeat GGGGCC/ CCCCGG sequence located in a non-coding region of the gene C9orf72. Three hypotheses have been proposed to explain how this repeats expansion causes diseases: 1) C9orf72 haploinsufficiency-expanded repeats interfere with transcription or translation of the gene, leading to decreased expression of the C9orf72 protein; 2) RNA gain of function-RNA foci formed by sense and antisense transcripts of expanded repeats interact and sequester essential RNA binding proteins, causing neurotoxicity; 3) Repeat associated non-ATG initiated (RAN) translation of expanded sense GGGGCC and antisense CCCCGG repeats produces potential toxic dipeptide repeat protein (DPR). In this review, we assess current evidence supporting or arguing against each proposed mechanism in C9 ALS/FTD disease pathogenesis. Additionally, controversial findings are also discussed. Lastly, we discuss the possibility that the three pathogenic mechanisms are not mutually exclusive and all three might be involved in disease.
Collapse
Affiliation(s)
- Xinmei Wen
- Jefferson Weinberg ALS Center, Vickie and Jack Farber Institute for Neuroscience, Department of Neuroscience, Thomas Jefferson University, Philadelphia, PA 19107, USA.
| | - Thomas Westergard
- Jefferson Weinberg ALS Center, Vickie and Jack Farber Institute for Neuroscience, Department of Neuroscience, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - Piera Pasinelli
- Jefferson Weinberg ALS Center, Vickie and Jack Farber Institute for Neuroscience, Department of Neuroscience, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - Davide Trotti
- Jefferson Weinberg ALS Center, Vickie and Jack Farber Institute for Neuroscience, Department of Neuroscience, Thomas Jefferson University, Philadelphia, PA 19107, USA
| |
Collapse
|
141
|
Li W, Tong HI, Gorantla S, Poluektova LY, Gendelman HE, Lu Y. Neuropharmacologic Approaches to Restore the Brain's Microenvironment. J Neuroimmune Pharmacol 2016; 11:484-94. [PMID: 27352074 PMCID: PMC4985494 DOI: 10.1007/s11481-016-9686-5] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2016] [Accepted: 05/25/2016] [Indexed: 12/15/2022]
Abstract
Maintaining the central nervous system microenvironment after injury, infection, inflammatory and degenerative diseases is contingent upon adequate control of glial homeostatic functions. Disease is caused by microbial, environmental and endogenous factors that compromise ongoing nervous system functions. The final result is neuronal injury, dropout and nerve connection loss, and these underlie the pathobiology of Alzheimer's and Parkinson's disease, amyotrophic lateral sclerosis, stroke, and bacterial, parasitic and viral infections. However, what promotes disease are homeostatic changes in the brain's microenvironment affected by innate glial immune pro-inflammatory and adaptive immune responses. These events disturb the brain's metabolic activities and communication abilities. How the process affects the brain's regulatory functions that can be harnessed for therapeutic gain is the subject at hand. Specific examples are provided that serve to modulate inflammation and improve disease outcomes specifically for HIV-associated neurocognitive disorders.
Collapse
Affiliation(s)
- Weizhe Li
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Hsin-I Tong
- Department of Public Health Sciences, Environmental Health Laboratory, University of Hawaii at Manoa, Honolulu, HI, 96822, USA
| | - Santhi Gorantla
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Larisa Y Poluektova
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Howard E Gendelman
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE, 68198, USA.
| | - Yuanan Lu
- Department of Public Health Sciences, Environmental Health Laboratory, University of Hawaii at Manoa, Honolulu, HI, 96822, USA.
| |
Collapse
|
142
|
A rapidly progressive dementia case with pathological diagnosis of FTLD-UPS. Acta Neuropathol 2016; 132:309-311. [PMID: 27271575 DOI: 10.1007/s00401-016-1584-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2016] [Revised: 05/24/2016] [Accepted: 05/25/2016] [Indexed: 10/21/2022]
|
143
|
Woollacott IOC, Rohrer JD. The clinical spectrum of sporadic and familial forms of frontotemporal dementia. J Neurochem 2016; 138 Suppl 1:6-31. [PMID: 27144467 DOI: 10.1111/jnc.13654] [Citation(s) in RCA: 89] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2016] [Revised: 04/10/2016] [Accepted: 04/27/2016] [Indexed: 12/11/2022]
Abstract
The term frontotemporal dementia (FTD) describes a clinically, genetically and pathologically diverse group of neurodegenerative disorders. Symptoms of FTD can present in individuals in their 20s through to their 90s, but the mean age at onset is in the sixth decade. The most common presentation is with a change in personality and impaired social conduct (behavioural variant FTD). Less frequently patients present with language problems (primary progressive aphasia). Both of these groups of patients can develop motor features consistent with either motor neuron disease (usually the amyotrophic lateral sclerosis variant) or parkinsonism (most commonly a progressive supranuclear palsy or corticobasal syndrome). In about a third of cases FTD is familial, with mutations in the progranulin, microtubule-associated protein tau and chromosome 9 open reading frame 72 genes being the major causes. Mutations in a number of other genes including TANK-binding kinase 1 are rare causes of familial FTD. This review aims to clarify the often confusing terminology of FTD, and outline the various clinical features and diagnostic criteria of sporadic and familial FTD syndromes. It will also discuss the current major challenges in FTD research and clinical practice, and potential areas for future research. This review clarifies the terminology of frontotemporal dementia (FTD) and summarizes the various clinical features and most recent diagnostic criteria of sporadic and familial FTD syndromes. It also discusses the current major challenges in FTD research and clinical practice, and highlights potential areas for future research.
Collapse
Affiliation(s)
- Ione O C Woollacott
- Dementia Research Centre, Department of Neurodegenerative Disease, UCL Institute of Neurology, Queen Square, London, UK
| | - Jonathan D Rohrer
- Dementia Research Centre, Department of Neurodegenerative Disease, UCL Institute of Neurology, Queen Square, London, UK
| |
Collapse
|
144
|
C9orf72 BAC Mouse Model with Motor Deficits and Neurodegenerative Features of ALS/FTD. Neuron 2016; 90:521-34. [DOI: 10.1016/j.neuron.2016.04.005] [Citation(s) in RCA: 245] [Impact Index Per Article: 27.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2015] [Revised: 03/01/2016] [Accepted: 03/29/2016] [Indexed: 12/12/2022]
|
145
|
Lapalme-Remis S, Oboudiyat C, Warman Chardon J, Bourque PR. Diffuse leukoencephalopathy with spheroids presenting as primary progressive aphasiaAuthor Response. Neurology 2016; 86:1464-1465. [DOI: 10.1212/01.wnl.0000482697.58240.eb] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
|
146
|
Coyle-Gilchrist ITS, Dick KM, Patterson K, Vázquez Rodríquez P, Wehmann E, Wilcox A, Lansdall CJ, Dawson KE, Wiggins J, Mead S, Brayne C, Rowe JB. Prevalence, characteristics, and survival of frontotemporal lobar degeneration syndromes. Neurology 2016; 86:1736-43. [PMID: 27037234 PMCID: PMC4854589 DOI: 10.1212/wnl.0000000000002638] [Citation(s) in RCA: 352] [Impact Index Per Article: 39.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2015] [Accepted: 01/21/2016] [Indexed: 12/13/2022] Open
Abstract
Objectives: To estimate the lifetime risk, prevalence, incidence, and mortality of the principal clinical syndromes associated with frontotemporal lobar degeneration (FTLD) using revised diagnostic criteria and including intermediate clinical phenotypes. Methods: Multisource referral over 2 years to identify all diagnosed or suspected cases of frontotemporal dementia (FTD), progressive supranuclear palsy (PSP), or corticobasal syndrome (CBS) in 2 UK counties (population 1.69 million). Diagnostic confirmation used current consensus diagnostic criteria after interview and reexamination. Results were adjusted to the 2013 European standard population. Results: The prevalence of FTD, PSP, and CBS was 10.8/100,000. The incidence and mortality were very similar, at 1.61/100,000 and 1.56/100,000 person-years, respectively. The estimated lifetime risk is 1 in 742. Survival following diagnosis varied widely: from PSP 2.9 years to semantic variant FTD 9.1 years. Age-adjusted prevalence peaked between 65 and 69 years at 42.6/100,000: the age-adjusted prevalence for persons older than 65 years is double the prevalence for those between 40 and 64 years. Fifteen percent of those screened had a relevant genetic mutation. Conclusions: Key features of this study include the revised diagnostic criteria with improved specificity and sensitivity, an unrestricted age range, and simultaneous assessment of multiple FTLD syndromes. The prevalence of FTD, PSP, and CBS increases beyond 65 years, with frequent genetic causes. The time from onset to diagnosis and from diagnosis to death varies widely among syndromes, emphasizing the challenge and importance of accurate and timely diagnosis. A high index of suspicion for FTLD syndromes is required by clinicians, even for older patients.
Collapse
Affiliation(s)
- Ian T S Coyle-Gilchrist
- From the Department of Clinical Neurosciences (I.T.S.C.-G., K.P., P.V.R., A.W., C.J.L., K.E.D., J.W., J.B.R.) and Cambridge Institute of Public Health (C.B.), University of Cambridge; Dementia Research Centre (K.M.D.) and MRC Prion Unit and Department of Neurodegenerative Disease (S.M.), University College London, UK; and University Medical Center Eppendorf (E.W.), University Hamburg, Germany.
| | - Katrina M Dick
- From the Department of Clinical Neurosciences (I.T.S.C.-G., K.P., P.V.R., A.W., C.J.L., K.E.D., J.W., J.B.R.) and Cambridge Institute of Public Health (C.B.), University of Cambridge; Dementia Research Centre (K.M.D.) and MRC Prion Unit and Department of Neurodegenerative Disease (S.M.), University College London, UK; and University Medical Center Eppendorf (E.W.), University Hamburg, Germany
| | - Karalyn Patterson
- From the Department of Clinical Neurosciences (I.T.S.C.-G., K.P., P.V.R., A.W., C.J.L., K.E.D., J.W., J.B.R.) and Cambridge Institute of Public Health (C.B.), University of Cambridge; Dementia Research Centre (K.M.D.) and MRC Prion Unit and Department of Neurodegenerative Disease (S.M.), University College London, UK; and University Medical Center Eppendorf (E.W.), University Hamburg, Germany
| | - Patricia Vázquez Rodríquez
- From the Department of Clinical Neurosciences (I.T.S.C.-G., K.P., P.V.R., A.W., C.J.L., K.E.D., J.W., J.B.R.) and Cambridge Institute of Public Health (C.B.), University of Cambridge; Dementia Research Centre (K.M.D.) and MRC Prion Unit and Department of Neurodegenerative Disease (S.M.), University College London, UK; and University Medical Center Eppendorf (E.W.), University Hamburg, Germany
| | - Eileen Wehmann
- From the Department of Clinical Neurosciences (I.T.S.C.-G., K.P., P.V.R., A.W., C.J.L., K.E.D., J.W., J.B.R.) and Cambridge Institute of Public Health (C.B.), University of Cambridge; Dementia Research Centre (K.M.D.) and MRC Prion Unit and Department of Neurodegenerative Disease (S.M.), University College London, UK; and University Medical Center Eppendorf (E.W.), University Hamburg, Germany
| | - Alicia Wilcox
- From the Department of Clinical Neurosciences (I.T.S.C.-G., K.P., P.V.R., A.W., C.J.L., K.E.D., J.W., J.B.R.) and Cambridge Institute of Public Health (C.B.), University of Cambridge; Dementia Research Centre (K.M.D.) and MRC Prion Unit and Department of Neurodegenerative Disease (S.M.), University College London, UK; and University Medical Center Eppendorf (E.W.), University Hamburg, Germany
| | - Claire J Lansdall
- From the Department of Clinical Neurosciences (I.T.S.C.-G., K.P., P.V.R., A.W., C.J.L., K.E.D., J.W., J.B.R.) and Cambridge Institute of Public Health (C.B.), University of Cambridge; Dementia Research Centre (K.M.D.) and MRC Prion Unit and Department of Neurodegenerative Disease (S.M.), University College London, UK; and University Medical Center Eppendorf (E.W.), University Hamburg, Germany
| | - Kate E Dawson
- From the Department of Clinical Neurosciences (I.T.S.C.-G., K.P., P.V.R., A.W., C.J.L., K.E.D., J.W., J.B.R.) and Cambridge Institute of Public Health (C.B.), University of Cambridge; Dementia Research Centre (K.M.D.) and MRC Prion Unit and Department of Neurodegenerative Disease (S.M.), University College London, UK; and University Medical Center Eppendorf (E.W.), University Hamburg, Germany
| | - Julie Wiggins
- From the Department of Clinical Neurosciences (I.T.S.C.-G., K.P., P.V.R., A.W., C.J.L., K.E.D., J.W., J.B.R.) and Cambridge Institute of Public Health (C.B.), University of Cambridge; Dementia Research Centre (K.M.D.) and MRC Prion Unit and Department of Neurodegenerative Disease (S.M.), University College London, UK; and University Medical Center Eppendorf (E.W.), University Hamburg, Germany
| | - Simon Mead
- From the Department of Clinical Neurosciences (I.T.S.C.-G., K.P., P.V.R., A.W., C.J.L., K.E.D., J.W., J.B.R.) and Cambridge Institute of Public Health (C.B.), University of Cambridge; Dementia Research Centre (K.M.D.) and MRC Prion Unit and Department of Neurodegenerative Disease (S.M.), University College London, UK; and University Medical Center Eppendorf (E.W.), University Hamburg, Germany
| | - Carol Brayne
- From the Department of Clinical Neurosciences (I.T.S.C.-G., K.P., P.V.R., A.W., C.J.L., K.E.D., J.W., J.B.R.) and Cambridge Institute of Public Health (C.B.), University of Cambridge; Dementia Research Centre (K.M.D.) and MRC Prion Unit and Department of Neurodegenerative Disease (S.M.), University College London, UK; and University Medical Center Eppendorf (E.W.), University Hamburg, Germany
| | - James B Rowe
- From the Department of Clinical Neurosciences (I.T.S.C.-G., K.P., P.V.R., A.W., C.J.L., K.E.D., J.W., J.B.R.) and Cambridge Institute of Public Health (C.B.), University of Cambridge; Dementia Research Centre (K.M.D.) and MRC Prion Unit and Department of Neurodegenerative Disease (S.M.), University College London, UK; and University Medical Center Eppendorf (E.W.), University Hamburg, Germany
| |
Collapse
|
147
|
Schroeder SK, Joly-Amado A, Gordon MN, Morgan D. Tau-Directed Immunotherapy: A Promising Strategy for Treating Alzheimer's Disease and Other Tauopathies. J Neuroimmune Pharmacol 2016; 11:9-25. [PMID: 26538351 PMCID: PMC4746105 DOI: 10.1007/s11481-015-9637-6] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2015] [Accepted: 10/16/2015] [Indexed: 12/14/2022]
Abstract
Immunotherapy directed against tau is a promising treatment strategy for Alzheimer's Disease (AD) and tauopathies. We review initial studies on tau-directed immunotherapy, and present data from our laboratory testing antibodies using the rTg4510 mouse model, which deposits tau in forebrain neurons. Numerous antibodies have been tested for their efficacy in treating both pathology and cognitive function, in different mouse models, by different routes of administration, and at different ages or durations. We report, here, that the conformation-specific antibody MC-1 produces some degree of improvement to both cognition and pathology in rTg4510. Pathological improvements as measured by Gallyas staining for fully formed tangles and phosphorylated tau appeared 4 days after intracranial injection into the hippocampus. We also examined markers for microglial activation, which did not appear impacted from treatment. Behavioral effects were noted after continuous infusion of antibodies into the lateral ventricle for approximately 2 weeks. We examined basic motor skills, which were not impacted by treatment, but did note cognitive improvements with both novel object and radial arm water maze testing. Our results support earlier reports in the initial review presented here, and collectively show promise for this strategy of treatment. The general absence of extracellular tau deposits may avoid the opsonization and phagocytosis mechanisms activated by antibodies against amyloid, and make anti tau approaches a safer method of immunotherapy for Alzheimer's disease.
Collapse
Affiliation(s)
- Sulana K Schroeder
- Department of Molecular Pharmacology and Physiology, Morsani College of Medicine, University of South Florida, Byrd Alzheimer's Institute, 4001 E. Fletcher Avenue, Tampa, FL, 33613, USA
| | - Aurelie Joly-Amado
- Department of Molecular Pharmacology and Physiology, Morsani College of Medicine, University of South Florida, Byrd Alzheimer's Institute, 4001 E. Fletcher Avenue, Tampa, FL, 33613, USA
| | - Marcia N Gordon
- Department of Molecular Pharmacology and Physiology, Morsani College of Medicine, University of South Florida, Byrd Alzheimer's Institute, 4001 E. Fletcher Avenue, Tampa, FL, 33613, USA
| | - Dave Morgan
- Department of Molecular Pharmacology and Physiology, Morsani College of Medicine, University of South Florida, Byrd Alzheimer's Institute, 4001 E. Fletcher Avenue, Tampa, FL, 33613, USA.
| |
Collapse
|
148
|
Moretti DV, Benussi L, Fostinelli S, Ciani M, Binetti G, Ghidoni R. Progranulin Mutations Affects Brain Oscillatory Activity in Fronto-Temporal Dementia. Front Aging Neurosci 2016; 8:35. [PMID: 26973510 PMCID: PMC4770190 DOI: 10.3389/fnagi.2016.00035] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2015] [Accepted: 02/10/2016] [Indexed: 12/12/2022] Open
Abstract
Background: Mild cognitive impairment (MCI) is a clinical stage indicating a prodromal phase of dementia. This practical concept could be used also for fronto-temporal dementia (FTD). Progranulin (PGRN) has been recently recognized as a useful diagnostic biomarker for fronto-temporal lobe degeneration (FTLD) due to GRN null mutations. Electroencephalography (EEG) is a reliable tool in detecting brain networks changes. The working hypothesis of the present study is that EEG oscillations could detect different modifications among FTLD stages (FTD-MCI versus overt FTD) as well as differences between GRN mutation carriers versus non-carriers in patients with overt FTD. Materials and Methods: EEG in all patients and PGRN dosage in patients with a clear FTD were detected. The cognitive state has been investigated through mini mental state examination (MMSE). Results: MCI-FTD showed a significant lower spectral power in both alpha and theta oscillations as compared to overt FTD. GRN mutations carriers affected by FTLD show an increase in high alpha and decrease in theta oscillations as compared to non-carriers. Conclusion: EEG frequency rhythms are sensible to different stage of FTD and could detect changes in brain oscillatory activity affected by GRN mutations.
Collapse
Affiliation(s)
- Davide V Moretti
- Alzheimer Rehabilitation Research Unit, IRCCS Istituto Centro San Giovanni di Dio Fatebenefratelli Brescia, Italy
| | - Luisa Benussi
- Molecular Markers Laboratory, IRCCS Istituto Centro San Giovanni di Dio Fatebenefratelli Brescia, Italy
| | - Silvia Fostinelli
- Molecular Markers Laboratory, IRCCS Istituto Centro San Giovanni di Dio Fatebenefratelli Brescia, Italy
| | - Miriam Ciani
- Molecular Markers Laboratory, IRCCS Istituto Centro San Giovanni di Dio Fatebenefratelli Brescia, Italy
| | - Giuliano Binetti
- Memory Clinic, IRCCS Istituto Centro San Giovanni di Dio Fatebenefratelli Brescia, Italy
| | - Roberta Ghidoni
- Molecular Markers Laboratory, IRCCS Istituto Centro San Giovanni di Dio Fatebenefratelli Brescia, Italy
| |
Collapse
|
149
|
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
|
150
|
Jun MH, Han JH, Lee YK, Jang DJ, Kaang BK, Lee JA. TMEM106B, a frontotemporal lobar dementia (FTLD) modifier, associates with FTD-3-linked CHMP2B, a complex of ESCRT-III. Mol Brain 2015; 8:85. [PMID: 26651479 PMCID: PMC4676093 DOI: 10.1186/s13041-015-0177-z] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2015] [Accepted: 12/04/2015] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Transmembrane protein 106B (TMEM106B) has been identified as a risk factor for frontotemporal lobar degeneration, which is the second most common form of progressive dementia in people under 65 years of age. Mutations in charged multivesicular body protein 2B (CHMP2B), which is involved in endosomal protein trafficking, have been found in chromosome 3-linked frontotemporal dementia. Despite the number of studies on both CHMP2B and TMEM106B in the endolysosomal pathway, little is known about the relationship between CHMP2B and TMEM106B in the endosomal/autophagy pathway. RESULTS This study found that endogenous TMEM106B was partially sequestered in CHMP2B-positive structures, suggesting its possible involvement in endosomal sorting complexes required for transport (ESCRT)-associated pathways. The role of single nucleotide polymorphisms of TMEM106B (T185, S185, or S134N) in the ESCRT-associated pathways were characterized. The T185 and S185 variants were more localized to Rab5-/Rab7-positive endosomes compared with S134N, while all of the variants were more localized to Rab7-positive endosomes compared to Rab5-positive endosomes. T185 was more associated with CHMP2B compared to S185. Autophagic flux was slightly reduced in the T185-expressing cells compared to the control or S185-expressing cells. Moreover, T185 slightly enhanced the accumulation of EGFR, impairments in autophagic flux, and neurotoxicity that were caused by CHMP2B(Intron5) compared to S185-expressing cells. CONCLUSIONS These findings suggest that the T185 variant functions as a risk factor in neurodegeneration with endolysosomal defects. This study provides a better understanding of pathogenic functions of TMEM106B, which is a risk factor for the progression of neurodegenerative diseases that are associated with endosomal defects in the aged brain.
Collapse
Affiliation(s)
- Mi-Hee Jun
- Department of Biotechnology and Biological Sciences, Hannam University, 1646 Yuseongdaero, Yu-seong-gu, Daejeon, 305-811, South Korea.
| | - Jeong-Ho Han
- Department of Biotechnology and Biological Sciences, Hannam University, 1646 Yuseongdaero, Yu-seong-gu, Daejeon, 305-811, South Korea.
| | - Yu-Kyung Lee
- Department of Biotechnology and Biological Sciences, Hannam University, 1646 Yuseongdaero, Yu-seong-gu, Daejeon, 305-811, South Korea.
| | - Deok-Jin Jang
- Applied Biology, College of Ecological Environment, Kyungpook National University, 386, Gajang-dong, Sang-Ju, South Korea.
| | - Bong-Kiun Kaang
- Department of Biological Sciences, College of Natural Sciences, Seoul National University, 1 Gwanangno, Gwanak-gu, Seoul, 08826, South Korea.
| | - Jin-A Lee
- Department of Biotechnology and Biological Sciences, Hannam University, 1646 Yuseongdaero, Yu-seong-gu, Daejeon, 305-811, South Korea.
| |
Collapse
|