Publications

Hope Center member publications

List of publications for the week of January 31, 2022

Murine roseolovirus does not accelerate amyloid-β pathology and human roseoloviruses are not over-represented in Alzheimer disease brains” (2022) Molecular Neurodegeneration

Murine roseolovirus does not accelerate amyloid-β pathology and human roseoloviruses are not over-represented in Alzheimer disease brains
(2022) Molecular Neurodegeneration, 17 (1), art. no. 10, . 

Bigley, T.M.a , Xiong, M.b c d , Ali, M.f , Chen, Y.b c e , Wang, C.b , Serrano, J.R.b , Eteleeb, A.f g , Harari, O.b f g , Yang, L.h , Patel, S.J.h , Cruchaga, C.b f g , Yokoyama, W.M.h , Holtzman, D.M.b

a Division of Rheumatology, Department of Pediatrics, Washington University School of Medicine, St. Louis, MO 63110, United States
b Department of Neurology, Hope Center for Neurological Disorders, Knight Alzheimer’s Disease Research Center, Washington University School of Medicine, St. Louis, MO 63110, United States
c Division of Biology and Biomedical Sciences (DBBS), Washington University School of Medicine, St. Louis, MO 63110, United States
d Present address: Genentech, 1 DNA Way, South San Francisco, CA 94080, United States
e Department of Pathology & Immunology, Washington University School of Medicine, St. Louis, MO 63110, United States
f Department Psychiatry, Washington University School of Medicine (WUSM), 660 S. Euclid Ave. B8134, St. Louis, MO 63110, United States
g NeuroGenomics and Informatics, Washington University School of Medicine, St. Louis, MO, United States
h Division of Rheumatology, Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110, United States

Abstract
Background: The role of viral infection in Alzheimer Disease (AD) pathogenesis is an area of great interest in recent years. Several studies have suggested an association between the human roseoloviruses, HHV-6 and HHV-7, and AD. Amyloid-β (Aβ) plaques are a hallmark neuropathological finding of AD and were recently proposed to have an antimicrobial function in response to infection. Identifying a causative and mechanistic role of human roseoloviruses in AD has been confounded by limitations in performing in vivo studies. Recent -omics based approaches have demonstrated conflicting associations between human roseoloviruses and AD. Murine roseolovirus (MRV) is a natural murine pathogen that is highly-related to the human roseoloviruses, providing an opportunity to perform well-controlled studies of the impact of roseolovirus on Aβ deposition. Methods: We utilized the 5XFAD mouse model to test whether MRV induces Aβ deposition in vivo. We also evaluated viral load and neuropathogenesis of MRV infection. To evaluate Aβ interaction with MRV, we performed electron microscopy. RNA-sequencing of a cohort of AD brains compared to control was used to investigate the association between human roseolovirus and AD. Results: We found that 5XFAD mice were susceptible to MRV infection and developed neuroinflammation. Moreover, we demonstrated that Aβ interacts with viral particles in vitro and, subsequent to this interaction, can disrupt infection. Despite this, neither peripheral nor brain infection with MRV increased or accelerated Aβ plaque formation. Moreover, −omics based approaches have demonstrated conflicting associations between human roseoloviruses and AD. Our RNA-sequencing analysis of a cohort of AD brains compared to controls did not show an association between roseolovirus infection and AD. Conclusion: Although MRV does infect the brain and cause transient neuroinflammation, our data do not support a role for murine or human roseoloviruses in the development of Aβ plaque formation and AD. © 2022, The Author(s).

Author Keywords
Alzheimer’s disease;  Amyloid-beta;  Human roseolovirus;  Murine roseolovirus;  Neuroinflammation

Funding details
CDI-CORE-2015-505, CDI-CORE-2019-813
National Institutes of HealthNIHT32AI106688–07, T32AR007279–40
National Institute on AgingNIAAG062027, P01AG003991, P30AG066444, R01AG044546, R01AG057777, RF1AG053303, RF1AG058501, U01AG058922
Foundation for Barnes-Jewish Hospital3770, 4642, R01AG047644, R01NS090934
JPB Foundation
Cure Alzheimer’s FundCAF
Washington University School of Medicine in St. LouisWUSM
Center for Cellular Imaging, Washington UniversityWUCCI
Tau Consortium

Document Type: Article
Publication Stage: Final
Source: Scopus

Variant-dependent heterogeneity in amyloid β burden in autosomal dominant Alzheimer’s disease: cross-sectional and longitudinal analyses of an observational study” (2022) The Lancet Neurology

Variant-dependent heterogeneity in amyloid β burden in autosomal dominant Alzheimer’s disease: cross-sectional and longitudinal analyses of an observational study
(2022) The Lancet Neurology, 21 (2), pp. 140-152. Cited 1 time.

Chhatwal, J.P.a b c , Schultz, S.A.a b , McDade, E.d , Schultz, A.P.a b , Liu, L.a c , Hanseeuw, B.J.a b i , Joseph-Mathurin, N.e , Feldman, R.e , Fitzpatrick, C.D.b c , Sparks, K.P.b , Levin, J.j k n , Berman, S.B.l , Renton, A.E.aa , Esposito, B.T.aa , Fernandez, M.V.f , Sung, Y.J.f , Lee, J.H.p , Klunk, W.E.l m , Hofmann, A.o , Noble, J.M.q , Graff-Radford, N.r , Mori, H.s , Salloway, S.M.u , Masters, C.L.v w , Martins, R.x , Karch, C.M.d , Xiong, C.g , Cruchaga, C.f , Perrin, R.J.h , Gordon, B.A.e , Benzinger, T.L.S.e , Fox, N.C.t , Schofield, P.R.y z , Fagan, A.M.d , Goate, A.M.aa , Morris, J.C.d , Bateman, R.J.d , Johnson, K.A.a b c , Sperling, R.A.a b c , Dominantly Inherited Alzheimer’s Network Investigatorsab

a Department of Neurology, Harvard Medical School, Boston, MA, United States
b Massachusetts General Hospital, Boston, MA, United States
c Brigham and Women’s Hospital, Boston, MA, United States
d Department of Neurology, Washington University in St Louis, St Louis, MO, United States
e Mallinckrodt Institute of Radiology, Washington University in St Louis, St Louis, MO, United States
f Department of Psychiatry, Washington University in St Louis, St Louis, MO, United States
g Division of Biostatistics, Washington University in St Louis, St Louis, MO, United States
h Department of Pathology, Washington University in St Louis, St Louis, MO, United States
i Université Catholique de Louvain, Brussels, Belgium
j Department of Neurology, Ludwig-Maximilians Universität München, Munich, Germany
k Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
l Department of Neurology, University of Pittsburgh, Pittsburgh, PA, United States
m Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA, United States
n German Center for Neurodegenerative Diseases, Munich, Germany
o German Center for Neurodegenerative Disease, Tübingen, Germany
p Department of Neurology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, South Korea
q Columbia University Irving Medical Center, Department of Neurology, New York, NY, United States
r Mayo Clinic, Department of Neurology, Jacksonville, FL, United States
s Osaka City University, Sumiyoshi Ward, Osaka, Japan
t UCL Queen Square Institute of Neurology, Dementia Research Centre, London, UK, United Kingdom
u Butler Hospital, Memory and Aging Program, Brown University Alpert Medical School, Providence, RI, United States
v The University of Melbourne, Melbourne, VIC, Australia
w Florey Institute, Melbourne, VIC, Australia
x Department of Biomedical Sciences, Macquarie University, Sydney, NSW, Australia
y Neuroscience Research Australia, Sydney, NSW, Australia
z School of Medical Sciences, University of New South Wales, Sydney, NSW, Australia
aa Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, United States

Abstract
Background: Insights gained from studying individuals with autosomal dominant Alzheimer’s disease have broadly influenced mechanistic hypotheses, biomarker development, and clinical trials in both sporadic and dominantly inherited Alzheimer’s disease. Although pathogenic variants causing autosomal dominant Alzheimer’s disease are highly penetrant, there is substantial heterogeneity in levels of amyloid β (Aβ) between individuals. We aimed to examine whether this heterogeneity is related to disease progression and to investigate the association with mutation location within PSEN1, PSEN2, or APP. Methods: We did cross-sectional and longitudinal analyses of data from the Dominantly Inherited Alzheimer’s Network (DIAN) observational study, which enrols individuals from families affected by autosomal dominant Alzheimer’s disease. 340 participants in the DIAN study who were aged 18 years or older, had a history of autosomal dominant Alzheimer’s disease in their family, and who were enrolled between September, 2008, and June, 2019, were included in our analysis. 206 participants were carriers of pathogenic mutations in PSEN1, PSEN2, or APP, and 134 were non-carriers. 62 unique pathogenic variants were identified in the cohort and were grouped in two ways. First, we sorted variants in PSEN1, PSEN2, or APP by the affected protein domain. Second, we divided PSEN1 variants according to position before or after codon 200. We examined variant-dependent variability in Aβ biomarkers, specifically Pittsburgh-Compound-B PET (PiB-PET) signal, levels of CSF Aβ1-42 (Aβ42), and levels of Aβ1-40 (Aβ40). Findings: Cortical and striatal PiB-PET signal showed striking variant-dependent variability using both grouping approaches (p<0·0001), despite similar progression on the clinical dementia rating (p>0·7), and CSF Aβ42 levels (codon-based grouping: p=0·49; domain-based grouping: p=0·095). Longitudinal PiB-PET signal also varied across codon-based groups, mirroring cross-sectional analyses. Interpretation: Autosomal dominant Alzheimer’s disease pathogenic variants showed highly differential temporal and regional patterns of PiB-PET signal, despite similar functional progression. These findings suggest that although increased PiB-PET signal is generally seen in autosomal dominant Alzheimer’s disease, higher levels of PiB-PET signal at an individual level might not reflect more severe or more advanced disease. Our results have high relevance for ongoing clinical trials in autosomal dominant Alzheimer’s disease, including those using Aβ PET as a surrogate marker of disease progression. Additionally, and pertinent to both sporadic and autosomal dominant Alzheimer’s disease, our results suggest that CSF and PET measures of Aβ levels are not interchangeable and might reflect different Aβ-driven pathobiological processes. Funding: National Institute on Aging, Doris Duke Charitable Foundation, German Center for Neurodegenerative Diseases, Japanese Agency for Medical Research and Development. © 2022 Elsevier Ltd

Funding details
National Institutes of HealthNIH
National Institute on AgingNIA
Alzheimer’s AssociationAA
Biogen
Japan Agency for Medical Research and DevelopmentAMED
Deutsches Zentrum für Neurodegenerative ErkrankungenDZNE

Document Type: Article
Publication Stage: Final
Source: Scopus

Astrocytic 4R tau expression drives astrocyte reactivity and dysfunction” (2022) JCI Insight

Astrocytic 4R tau expression drives astrocyte reactivity and dysfunction
(2022) JCI Insight, 7 (1), art. no. 152012, . 

Ezerskiy, L.A.a , Schoch, K.M.a , Sato, C.a , Beltcheva, M.b , Horie, K.a , Rigo, F.c , Martynowicz, R.a , Karch, C.M.d , Bateman, R.J.a , Miller, T.M.a

a Department of Neurology, Washington University, School of Medicine, St. Louis, MO, United States
b Center of Regenerative Medicine, Washington University, School of Medicine, St. Louis, MO, United States
c Ionis Pharmaceuticals, Carlsbad, CA, United States
d Department of Psychiatry, Washington University, School of Medicine, St. Louis, MO, United States

Abstract
The protein tau and its isoforms are associated with several neurodegenerative diseases, many of which are characterized by greater deposition of the 4-repeat (4R) tau isoform; however, the role of 4R tau in disease pathogenesis remains unclear. We created antisense oligonucleotides (ASOs) that alter the ratio of 3R to 4R tau to investigate the role of specific tau isoforms in disease. Preferential expression of 4R tau in human tau-expressing (hTau-expressing) mice was previously shown to increase seizure severity and phosphorylated tau deposition without neuronal or synaptic loss. In this study, we observed strong colocalization of 4R tau within reactive astrocytes and increased expression of pan-reactive and neurotoxic genes following 3R to 4R tau splicing ASO treatment in hTau mice. Increasing 4R tau levels in primary astrocytes provoked a similar response, including a neurotoxic genetic profile and diminished homeostatic function, which was replicated in human induced pluripotent stem cell-derived (iPSC-derived) astrocytes harboring a mutation that exhibits greater 4R tau. Healthy neurons cultured with 4R tau-expressing human iPSC-derived astrocytes exhibited a higher firing frequency and hypersynchrony, which could be prevented by lowering tau expression. These findings support a potentially novel pathway by which astrocytic 4R tau mediates reactivity and dysfunction and suggest that astrocyte-targeted therapeutics against 4R tau may mitigate neurodegenerative disease progression. © 2022, Ezerskiy et al.

Funding details
CDI-CORE-2015-505, CDI-CORE-2019-813
National Institutes of HealthNIHNS110890
National Institute on AgingNIAK01AG062796
Foundation for Barnes-Jewish Hospital3770, 4642
Rainwater Charitable FoundationRCF

Document Type: Article
Publication Stage: Final
Source: Scopus

Brain ventricles as windows into brain development and disease” (2022) Neuron

Brain ventricles as windows into brain development and disease
(2022) Neuron, 110 (1), pp. 12-15. 

Duy, P.Q.a , Rakic, P.b , Alper, S.L.c , Butler, W.E.d , Walsh, C.A.e , Sestan, N.b , Geschwind, D.H.f , Jin, S.C.g , Kahle, K.T.h

a Department of Neurosurgery, Yale University School of Medicine, New Haven, CT, USA; Medical Scientist Training Program, Yale University School of Medicine, New Haven, CT, USA; Department of Neuroscience, Yale University School of Medicine, New Haven, CT, USA
b Department of Neuroscience, Yale University School of Medicine, CT, New Haven, United States
c Division of Nephrology and Vascular Biology Research Center, Beth Israel Deaconess Medical Center and Department of Medicine, Harvard Medical School, MA, Boston, United States
d Department of Neurosurgery, Massachusetts General Hospital, MA, Boston, United States
e Division of Genetics and Genomics, Boston Children’s Hospital, Boston, MA, USA; Departments of Pediatrics and Neurology, Harvard Medical School, Boston, MA, USA; Broad Institute of MIT and Harvard, Cambridge, MA, USA
f Department of Human Genetics, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
g Department of Genetics, Washington University School of Medicine, St. Louis, MO, USA; Department of Pediatrics, Washington University School of Medicine, St. Louis, MO, USA
h Department of Neurosurgery, Massachusetts General Hospital, Boston, MA, USA; Division of Genetics and Genomics, Boston Children’s Hospital, Boston, MA, USA; Broad Institute of MIT and Harvard, Cambridge, MA, USA; MGH Hydrocephalus and Neurodevelopmental Disorders Program, Massachusetts General Hospital, Boston, MA, USA

Abstract
Dilation of the fluid-filled cerebral ventricles (ventriculomegaly) characterizes hydrocephalus and is frequently seen in autism and schizophrenia. Recent work suggests that the genomic study of congenital hydrocephalus may be unexpectedly fertile ground for revealing insights into neural stem cell regulation, human cerebrocortical development, and pathogenesis of neuropsychiatric disease. Copyright © 2021 Elsevier Inc. All rights reserved.

Author Keywords
brain ventricle;  cerebrospinal fluid;  CH;  congenital hydrocephalus;  CSF;  genomics;  neural development;  neural stem cell;  neurodevelopmental disorders;  NSC

Document Type: Article
Publication Stage: Final
Source: Scopus

Disrupted Association of Sensory Neurons With Enveloping Satellite Glial Cells in Fragile X Mouse Model” (2022) Frontiers in Molecular Neuroscience

Disrupted Association of Sensory Neurons With Enveloping Satellite Glial Cells in Fragile X Mouse Model
(2022) Frontiers in Molecular Neuroscience, 14, art. no. 796070, . 

Avraham, O.a , Deng, P.-Y.b , Maschi, D.b , Klyachko, V.A.b c , Cavalli, V.a c d

a Department of Neuroscience, Washington University School of Medicine, St. LouisMO, United States
b Department of Cell Biology and Physiology, Washington University School of Medicine, St. Louis, MO, United States
c Hope Center for Neurological Disorders, Washington University School of Medicine, St. Louis, MO, United States
d Center of Regenerative Medicine, Washington University School of Medicine, St. Louis, MO, United States

Abstract
Among most prevalent deficits in individuals with Fragile X syndrome (FXS) is hypersensitivity to sensory stimuli and somatosensory alterations. Whether dysfunction in peripheral sensory system contributes to these deficits remains poorly understood. Satellite glial cells (SGCs), which envelop sensory neuron soma, play critical roles in regulating neuronal function and excitability. The potential contributions of SGCs to sensory deficits in FXS remain unexplored. Here we found major structural defects in sensory neuron-SGC association in the dorsal root ganglia (DRG), manifested by aberrant covering of the neuron and gaps between SGCs and the neuron along their contact surface. Single-cell RNAseq analyses demonstrated transcriptional changes in both neurons and SGCs, indicative of defects in neuronal maturation and altered SGC vesicular secretion. We validated these changes using fluorescence microscopy, qPCR, and high-resolution transmission electron microscopy (TEM) in combination with computational analyses using deep learning networks. These results revealed a disrupted neuron-glia association at the structural and functional levels. Given the well-established role for SGCs in regulating sensory neuron function, altered neuron-glia association may contribute to sensory deficits in FXS. Copyright © 2022 Avraham, Deng, Maschi, Klyachko and Cavalli.

Author Keywords
fragile X syndrome;  hyperexcitability;  neuron-glia communication;  satellite glial cells;  sensory neuron

Funding details
CDI-CORE-2015-505, CDI-CORE-2019-813
National Institutes of HealthNIHNS111596, R01 NS111719, R35 NS122260
Foundation for Barnes-Jewish HospitalFBJH
Washington University School of Medicine in St. LouisWUSM
Center for Cellular Imaging, Washington UniversityWUCCI

Document Type: Article
Publication Stage: Final
Source: Scopus

Quantitative Gradient Echo MRI Identifies Dark Matter as a New Imaging Biomarker of Neurodegeneration that Precedes Tisssue Atrophy in Early Alzheimer’s Disease” (2022) Journal of Alzheimer’s Disease

Quantitative Gradient Echo MRI Identifies Dark Matter as a New Imaging Biomarker of Neurodegeneration that Precedes Tisssue Atrophy in Early Alzheimer’s Disease
(2022) Journal of Alzheimer’s Disease, 85 (2), pp. 901-920. 

Kothapalli, S.V.V.N.a , Benzinger, T.L.a b , Aschenbrenner, A.J.b c , Perrin, R.J.b c d e , Hildebolt, C.F.a , Goyal, M.S.a c , Fagan, A.M.b c e , Raichle, M.E.a c e , Morris, J.C.b c , Yablonskiy, D.A.a b e

a Department of Radiology, Washington University in St. Louis, St. Louis, MO, United States
b Knight Alzheimer Disease Research Center, Washington University in St. Louis, St. Louis, MO, United States
c Department of Neurology, Washington University in St. Louis, St. Louis, MO, United States
d Department of Pathology and Immunology, Washington University in St. Louis, St. Louis, MO, United States
e The Hope Center for Neurological Disorders, Washington University in St. Louis, St. Louis, MO, United States

Abstract
Background: Currently, brain tissue atrophy serves as an in vivo MRI biomarker of neurodegeneration in Alzheimer’s disease (AD). However, postmortem histopathological studies show that neuronal loss in AD exceeds volumetric loss of tissue and that loss of memory in AD begins when neurons and synapses are lost. Therefore, in vivo detection of neuronal loss prior to detectable atrophy in MRI is essential for early AD diagnosis. Objective: To apply a recently developed quantitative Gradient Recalled Echo (qGRE) MRI technique for in vivo evaluation of neuronal loss in human hippocampus. Methods: Seventy participants were recruited from the Knight Alzheimer Disease Research Center, representing three groups: Healthy controls [Clinical Dementia Rating® (CDR®) = 0, amyloid β (Aβ)-negative, n = 34]; Preclinical AD (CDR = 0, Aβ-positive, n = 19); and mild AD (CDR = 0.5 or 1, Aβ-positive, n = 17). Results: In hippocampal tissue, qGRE identified two types of regions: one, practically devoid of neurons, we designate as ‘Dark Matter’, and the other, with relatively preserved neurons, ‘Viable Tissue’. Data showed a greater loss of neurons than defined by atrophy in the mild AD group compared with the healthy control group; neuronal loss ranged between 31% and 43%, while volume loss ranged only between 10% and 19%. The concept of Dark Matter was confirmed with histopathological study of one participant who underwent in vivo qGRE 14 months prior to expiration. Conclusion: In vivo qGRE method identifies neuronal loss that is associated with impaired AD-related cognition but is not recognized by MRI measurements of tissue atrophy, therefore providing new biomarkers for early AD detection. © 2022 – The authors. Published by IOS Press.

Author Keywords
Alzheimer’s disease;  brain atrophy;  cognitive impairment;  hippocampal subfields;  hippocampus;  magnetic resonance imaging;  neurodegeneration;  quantitative Gradient Recalled Echo

Funding details
National Institutes of HealthNIHP50 AG0581, R01 AG054513

Document Type: Article
Publication Stage: Final
Source: Scopus

Quantifying regional α -synuclein, amyloid β, and tau accumulation in lewy body dementia” (2022) Annals of Clinical and Translational Neurology

Quantifying regional α -synuclein, amyloid β, and tau accumulation in lewy body dementia
(2022) Annals of Clinical and Translational Neurology, . 

Miller, R.L.a b , Dhavale, D.D.a b , O’Shea, J.Y.a b , Andruska, K.M.a b , Liu, J.a b , Franklin, E.E.b c , Buddhala, C.a b , Loftin, S.K.a d , Cirrito, J.R.a b , Perrin, R.J.a b c , Cairns, N.J.a b c e , Campbell, M.C.a b d , Perlmutter, J.S.a d f g h , Kotzbauer, P.T.a b i

a Department of Neurology, Washington University School of Medicine, St. Louis, MO, United States
b Hope Center for Neurological Disorders, Washington University School of Medicine, St. Louis, MO, United States
c Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO, United States
d Department of Radiology, Washington University School of Medicine, St. Louis, MO, United States
e College of Medicine and Health, University of Exeter, Exeter, United Kingdom
f Department of Neuroscience, Washington University School of Medicine, St. Louis, MO, United States
g Program in Occupational Therapy, Washington University School of Medicine, St. Louis, MO, United States
h Program in Physical Therapy, Washington University School of Medicine, St. Louis, MO, United States
i Developmental Biology, Washington University School of Medicine, St. Louis, MO, United States

Abstract
Objective: Parkinson disease (PD) is defined by the accumulation of misfolded α-synuclein (α-syn) in Lewy bodies and Lewy neurites. It affects multiple cortical and subcortical neuronal populations. The majority of people with PD develop dementia, which is associated with Lewy bodies in neocortex and referred to as Lewy body dementia (LBD). Other neuropathologic changes, including amyloid β (Aβ) and tau accumulation, occur in some LBD cases. We sought to quantify α-syn, Aβ, and tau accumulation in neocortical, limbic, and basal ganglia regions. Methods: We isolated insoluble protein from fresh frozen postmortem brain tissue samples for eight brains regions from 15 LBD, seven Alzheimer disease (AD), and six control cases. We measured insoluble α-syn, Aβ, and tau with recently developed sandwich ELISAs. Results: We detected a wide range of insoluble α-syn accumulation in LBD cases. The majority had substantial α-syn accumulation in most regions, and dementia severity correlated with neocortical α-syn. However, three cases had low neocortical levels that were indistinguishable from controls. Eight LBD cases had substantial Aβ accumulation, although the mean Aβ level in LBD was lower than in AD. The presence of Aβ was associated with greater α-syn accumulation. Tau accumulation accompanied Aβ in only one LBD case. Interpretation: LBD is associated with insoluble α-syn accumulation in neocortical regions, but the relatively low neocortical levels in some cases suggest that other changes contribute to impaired function, such as loss of neocortical innervation from subcortical regions. The correlation between Aβ and α-syn accumulation suggests a pathophysiologic relationship between these two processes. © 2021 The Authors. Annals of Clinical and Translational Neurology published by Wiley Periodicals LLC on behalf of American Neurological Association

Funding details
National Institutes of HealthNIHNS075321, NS097437, NS097799, NS110436
National Institute on AgingNIA
National Institute of Neurological Disorders and StrokeNINDSAG003991, AG026276, AG066444
Michael J. Fox Foundation for Parkinson’s ResearchMJFF
American Parkinson Disease AssociationAPDA
Washington University in St. LouisWUSTL
Foundation for Barnes-Jewish Hospital

Document Type: Article
Publication Stage: Article in Press
Source: Scopus

What is Operative? Conceptualizing Neuralgia: Neuroma, Compression Neuropathy, Painful Hyperalgesia, and Phantom Nerve Pain” (2022) Journal of Hand Surgery Global Online

What is Operative? Conceptualizing Neuralgia: Neuroma, Compression Neuropathy, Painful Hyperalgesia, and Phantom Nerve Pain
(2022) Journal of Hand Surgery Global Online, . 

Hill, E.J.R.a , Patterson, J.M.M.b , Yee, A.c , Crock, L.W.d , Mackinnon, S.E.c

a Department of Orthopedic Surgery, Division of Hand and Microsurgery, Washington University in St. Louis School of Medicine, St. Louis, MO
b Department of Orthopedic Surgery, University of North Carolina, Chapel Hill, NC, United States
c Division of Plastic and Reconstructive Surgery, Washington University in St. Louis School of Medicine, St. Louis, MO
d Division of Pain Management, Washington University in St. Louis School of Medicine, St. Louis, MO

Abstract
Neuralgia, or nerve pain, is a common presenting complaint for the hand surgeon. When the nerve at play is easily localized, and the cause of the pain is clear (eg, carpal tunnel syndrome), the patient may be easily treated with excellent results. However, in more complex cases, the underlying pathophysiology and cause of neuralgia can be more difficult to interpret; if incorrectly managed, this leads to frustration for both the patient and surgeon. Here we offer a way to conceptualize neuralgia into 4 categories—compression neuropathy, neuroma, painful hyperalgesia, and phantom nerve pain—and offer an illustrative clinical vignette and strategies for optimal management of each. Further, we delineate the reasons why compression neuropathy and neuroma are amenable to surgery, while painful hyperalgesia and phantom nerve pain are not. © 2022 The Authors

Author Keywords
Nerve pain;  Nerve surgery;  Neuralgia;  Neuroma;  Neuropathy

Document Type: Article
Publication Stage: Article in Press
Source: Scopus

Functional characterization of hiPSCs-derived glial cells and neurons from patients harboring a TREM2 loss of function mutation” (2021) Alzheimer’s & Dementia: The Journal of the Alzheimer’s Association

Functional characterization of hiPSCs-derived glial cells and neurons from patients harboring a TREM2 loss of function mutation
(2021) Alzheimer’s & Dementia: The Journal of the Alzheimer’s Association, 17, p. e058712. 

Filipello, F.a , You, S.-F.b , Martinez, R.a , Korvatska, O.c , Raskind, W.H.c , Mahali, S.b , Ghezzi, L.b , Wandy, B.b , Cella, M.d , Piccio, L.e f , Karch, C.M.g h i

a Washington University, MO, Saint Louis, United States
b Washington University, St Louis, MO, USA
c University of Washington, Seattle, WA, USA
d Washington University in St. Louis, St. Louis, MO, USA
e Hope Center for Neurological Disorders, Washington University School of Medicine, St. Louis, MO, USA
f Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA
g Hope Center for Neurological Disorders, St. Louis, MO, USA
h Washington University in St. Louis, St Louis, MO, USA
i Washington University School of Medicine, St. Louis, MO, USA

Abstract
BACKGROUND: Triggering receptor expressed on myeloid cells 2 (TREM2) is an innate immune receptor expressed by microglia in the adult brain. Homozygous loss of function TREM2 variants cause a rare leukodystrophy characterized by bone cysts and early-onset dementia, Nasu-Hakola disease (NHD). However, despite intense investigation, the role of TREM2 in NHD pathogenesis remains poorly understood. METHOD: Here, we investigated the mechanisms by which a homozygous stop-gain TREM2 variant (Q33X), which leads to a truncated TREM2 transcript, contributes to disease pathology in NHD. Human induced pluripotent stem cells (hiPSCs)-derived microglia (iMGLs), neurons and astrocytes were obtained from two siblings homozygous for the TREM2 Q33X mutation and one non-carrier sibling. RESULT: Transcriptomic analysis and biochemical assays revealed that iMGLs from NHD patients display decreased activation, reduced lipid droplet content and defects in lysosomal function compared to related and unrelated controls. These in vitro findings were validated in brain tissues from NHD patients carrying loss of function TREM2 mutations. Strikingly, we observed defects beyond iMGLs. iPSCs-derived neurons and astrocytes from NHD siblings displayed downregulation of pathways involved in synaptic activation, neuronal development and interferon response compared to the unaffected sibling. These pathways were also observed to be similarly altered in NHD brains. This could be due to the presence of low levels of mutant TREM2 transcripts in iPSC and neural progenitor cells, which may initiate a cascade of events that drives cellular dysfunction beyond microglia. CONCLUSION: These finding open a new scenario on TREM2 function and reveal that NHD is a complex pathology affecting glial cells and neurons at multiple levels. © 2021 the Alzheimer’s Association.

Document Type: Article
Publication Stage: Final
Source: Scopus

Defining the role of PLD3 in Alzheimer’s disease pathology” (2021) Alzheimer’s & Dementia: The Journal of the Alzheimer’s Association

Defining the role of PLD3 in Alzheimer’s disease pathology
(2021) Alzheimer’s & Dementia: The Journal of the Alzheimer’s Association, 17, p. e058730. 

Rosene, M.J.a , Hsu, S.b , Martinez, R.c , Norton, J.a b d e , Yan, P.a , Cirrito, J.R.f , Lee, J.-M.g , Cuervo, A.M.h , Goate, A.M.i j , Cruchaga, C.d k l m n , Karch, C.M.c d k o

a Washington University School of Medicine, St. Louis, MO, USA
b Washington University in St Louis, St Louis, MO, USA
c Washington University, MO, Saint Louis, United States
d Hope Center for Neurological Disorders, St. Louis, MO, USA
e Knight Alzheimer Disease Research Center, St Louis, MO, USA
f Washington University School of Medicine, MO, Saint Louis, United States
g Washington University School of Medicine, St Louis, MO, USA
h Albert Einstein College of Medicine, Bronx, NY, USA
i Ronald M. Loeb Center for Alzheimer’s disease, NY, NY, United States
j Icahn School of Medicine at Mount Sinai, NY, NY, United States
k NeuroGenomics and Informatics Center, St. Louis, MO, USA
l Washington University, St. Louis, MO, USA
m Knight Alzheimer Disease Research Center, MO, Saint Louis, United States
n Washington University in St. Louis, MO, Saint Louis, United States
o Washington University in St. Louis, St Louis, MO, USA

Abstract
BACKGROUND: Alzheimer’s disease (AD) is characterized by the accumulation of amyloid-β (Aβ) in the brain. We recently identified coding variants in the phospholipase D3 (PLD3) gene that double the risk for late onset AD. METHOD: We examined the impact of PLD3 risk variants on PLD3 and Aβ metabolism using CRISPR/Cas9 in induced pluripotent stem cells (iPSC). We then modeled the PLD3 expression patterns observed in AD brains in immortalized cell and AD mouse models. Lysosomal function was assessed in human brain tissue. RESULT: PLD3 A442A disrupts a splicing enhancer binding site and reduces PLD3 splicing in human brains. Differentiation of PLD3 A442A and isogenic control iPSCs into cortical neurons produced cells that were morphologically similar. At the molecular level, PLD3 A442A neurons displayed a similar defect in PLD3 splicing as was observed in human brains and a significant increase in Aβ42/Aβ40 compared with isogenic control lines. Thus, PLD3 A442A is sufficient to alter PLD3 splicing and Aβ metabolism. PLD3 expression was significantly lower in AD brains compared with controls, and PLD3 expression was highly correlated with expression of lysosomal genes. Thus, we sought to determine whether PLD3 contributes to Aβ accumulation in AD via disrupted Aβ metabolism. We found that overexpression of PLD3 in immortalized cells decreased Aβ levels while shRNA silencing of Pld3 increased Aβ levels. In an AD mouse model, overexpression of PLD3 in hippocampal neurons produced decreased interstitial fluid (ISF) Aβ levels and accelerated Aβ turnover. Conversely, knocking out Pld3 increased ISF Aβ, reduced Aβ turnover, and increased APP protein levels. Knocking out Pld3 overtime lead to altered amyloid morphology. To begin to determine whether PLD3 influences Aβ turnover via the lysosome, we isolated lysosomal fractions from human AD and control brains. PLD3 was enriched in lysosomal subfractions and PLD3 distribution in these subfractions was altered in AD. Furthermore, PLD3 stability in the lysosomal fractions was disrupted in AD brains. CONCLUSION: Together, our findings demonstrate that PLD3 promotes Aβ clearance through pathways involving lysosomal degradation. © 2021 the Alzheimer’s Association.

Document Type: Article
Publication Stage: Final
Source: Scopus

Astrocytic BMAL1 regulates protein aggregation in mouse models of alpha-synuclein and tau pathology” (2021) Alzheimer’s & Dementia: The Journal of the Alzheimer’s Association

Astrocytic BMAL1 regulates protein aggregation in mouse models of alpha-synuclein and tau pathology
(2021) Alzheimer’s & Dementia: The Journal of the Alzheimer’s Association, 17, p. e058631. 

Sheehan, P.W.a , Kanan, M.a , Benitez, B.A.a b c d e , Harari, O.b c d e f , Davis, A.A.c , Musiek, E.S.a e

a Washington University in St. Louis, St. Louis, MO, USA
b Hope Center for Neurological Disorders, St. Louis, MO, USA
c Washington University School of Medicine, St. Louis, MO, USA
d NeuroGenomics and Informatics Center, St. Louis, MO, USA
e Knight Alzheimer Disease Research Center, St. Louis, MO, USA
f Washington University, MO, Saint Louis, United States

Abstract
BACKGROUND: The circadian clock regulates inflammatory responses in the peripheral immune system, though its function in neuroinflammation is poorly understood. Deletion of the master circadian clock transcription factor BMAL1 abrogates cellular circadian clock function and can be used to probe cell-type specific functions of the clock. Our lab has previously shown that Bmal1 deletion induces oxidative stress, astrocyte activation, and increased β-amyloid plaque deposition in mice. We hypothesized that deletion of Bmal1 would increase pathology in other protein aggregation models. METHODS: We generated global inducible Bmal1 knock out mice expressing human P301S mutant tau. Pathology, aggregation state, and gliosis were quantified at nine months of age. We induced synuclein aggregation in global Bmal1 knock out mice using an alpha-synuclein preformed fibril injection (PFF) model. Three months post-PFF injection, mice were analyzed for synuclein pathology and gliosis. To determine a cell-type specific effect, we generated astrocyte- and microglia-specific Bmal1 knock out mice and used these for PFF injections. Pathology and gliosis were analyzed from these mice three months post-PFF injection. We then generated astrocyte-specific Bmal1 knock out mice expressing human P301S mutant tau to test the effect of Bmal1 deletion in astrocytes on tau aggregation. RESULTS: Global Bmal1 deletion in the P301S model resulted in a significant decrease in aggregated tau and microglia activation. Global Bmal1 knock out mice injected with PFFs had significantly decreased synuclein pathology and a decrease in microgliosis. Bmal1 deletion specifically in astrocytes was sufficient to reduce both tau and synuclein pathology. Using bioinformatics methods, we discovered an astrocytic candidate gene, Bag3, which may play a role in reducing pathology in our models. We used an in vitro PFF uptake assay and found that Bmal1 knockdown resulted in increased PFF uptake, which was reduced to baseline after simultaneous knockdown of Bag3. CONCLUSION: Bmal1 deletion in astrocytes is sufficient to reduce tau and synuclein pathologies and attenuate microglia activation. This effect may be driven by the upregulation of the macroautophagy chaperone protein, Bag3. Our data suggests that targeting astrocytic protein degradation machinery may be a therapeutic strategy for reducing intra-neuronal protein aggregates, such as tau and synuclein. © 2021 the Alzheimer’s Association.

Document Type: Article
Publication Stage: Final
Source: Scopus

Impact of MAPT mutations on transcriptomic signatures of FTLD brains and patient-derived pluripotent cell models”(2021) Alzheimer’s & Dementia: The Journal of the Alzheimer’s Association

Impact of MAPT mutations on transcriptomic signatures of FTLD brains and patient-derived pluripotent cell models
(2021) Alzheimer’s & Dementia: The Journal of the Alzheimer’s Association, 17, p. e058313. 

Minaya, M.a , Martinez, R.b , Eteleeb, A.c , Cruchaga, C.d , Harari, O.b , Karch, C.M.d

a Washington University, MO, Saint Louis, United States
b Washington University, MO, Saint Louis, United States
c Washington University School of Medicine, MO, Saint Louis, United States
d Washington University School of Medicine, St. Louis, MO, USA

Abstract
BACKGROUND: Mutations in the microtubule-associated protein tau (MAPT) cause heterogeneous forms of frontotemporal lobar dementia with tau inclusions (FTLD-tau). Yet the pathogenic events linked to disease remain poorly understood. This study aimed to identify genes and pathways that lead to FTLD-tau. METHOD: To identify the earliest genes and pathways that are dysregulated in FTLD-tau, we identified differentially expressed genes in RNA-seq data generated from induced pluripotent stem cell (iPSC)-derived cortical neurons carrying MAPT R406W, MAPT P301L, and MAPT IVS10+16 and isogenic, controls and brain tissue samples from progressive supranuclear palsy (PSP) and control brains. We then identified pathological pathways and drug targets that were enriched among the differentially expressed genes. RESULTS: We identified 275 genes that were differentially expressed in iPSC-derived cortical neurons from MAPT R406W carriers compared to isogenic controls, MAPT IVS10+16 carriers compared to isogenic controls, and MAPT P301L carriers compared with isogenic controls. These commonly dysregulated genes were enriched for pathways involving synaptic function, neuronal development, and endolysosomal function. A subset of these genes were also changed in brains from human subjects with PSP compared to normal control brains. Finally, a subset of genes, enriched in glutamate receptor signaling, were altered across the mutant neurons and significantly changed with tau accumulation in a mouse model of tauopathy (Tau-P301L mice). CONCLUSION: The results from this study demonstrate that iPSC-derived neurons capture molecular processes that occur in human brains and can be used to model disease and point to common molecular pathways driven by 3 distinct MAPT mutations. © 2021 the Alzheimer’s Association.

Document Type: Article
Publication Stage: Final
Source: Scopus

Variants in Mitochondrial ATP Synthase Cause Variable Neurologic Phenotypes” (2021) Annals of Neurology

Variants in Mitochondrial ATP Synthase Cause Variable Neurologic Phenotypes
(2021) Annals of Neurology, . 

Zech, M.a b , Kopajtich, R.a b , Steinbrücker, K.c , Bris, C.d e , Gueguen, N.d e , Feichtinger, R.G.c , Achleitner, M.T.c , Duzkale, N.f , Périvier, M.g , Koch, J.c , Engelhardt, H.h , Freisinger, P.i , Wagner, M.a b , Brunet, T.a b , Berutti, R.a b , Smirnov, D.a b , Navaratnarajah, T.j , Rodenburg, R.J.T.k , Pais, L.S.l , Austin-Tse, C.m , O’Leary, M.n , Boesch, S.o , Jech, R.p , Bakhtiari, S.q r , Jin, S.C.s t , Wilbert, F.u , Kruer, M.C.q r , Wortmann, S.B.c v , Eckenweiler, M.u , Mayr, J.A.c , Distelmaier, F.j , Steinfeld, R.w , Winkelmann, J.a b x y , Prokisch, H.a b

a Technical University of Munich, School of Medicine, Institute of Human Genetics, Munich, Germany
b Institute of Neurogenomics, Helmholtz Zentrum München, Munich, Germany
c University Children’s Hospital, Paracelsus Medical University (PMU), Salzburg, Austria
d Unité Mixte de Recherche MITOVASC, CNRS 6015/INSERM 1083, Université d’Angers, Angers, France
e Département de Biochimie et Génétique, Centre Hospitalier Universitaire d’Angers, Angers, France
f Department of Medical Genetic, Diskapi Yildirim Beyazit Training and Research Hospital, Ankara, Turkey
g Pediatric Neurology Department, CHU Clocheville, Tours, France
h Kinderkrankenhaus St. Marien gGmbH, Zentrum für Kinder- und Jugendmedizin, Landshut, Germany
i Children’s Hospital Kreiskliniken, Reutlingen, Germany
j Department of General Pediatrics, Neonatology and Pediatric Cardiology, University Children’s Hospital, Medical Faculty, Heinrich-Heine-University, Düsseldorf, Germany
k Radboud Centre for Mitochondrial Medicine, Department of Paediatrics Radboud Institute for Molecular Life Sciences, Radboud University Nijmegen Medical Centre Nijmegen, Nijmegen, Netherlands
l Program in Medical and Population Genetics, Broad Institute, Cambridge, MA, United States
m Harvard Medical School & Center for Genomic Medicine, Massachusetts General Hospital, Boston & Laboratory for Molecular Medicine, Partners Healthcare Personalized Medicine, Cambridge, MA, United States
n Broad Center for Mendelian Genomics, Broad Institute of MIT and Harvard, Cambridge, MA, United States
o Department of Neurology, Medical University of Innsbruck, Innsbruck, Austria
p Department of Neurology, Charles University, 1st Faculty of Medicine and General University Hospital in Prague, Prague, Czech Republic
q Pediatric Movement Disorders Program, Division of Pediatric Neurology, Barrow Neurological Institute, Phoenix Children’s Hospital, Phoenix, AZ, United States
r Departments of Child Health, Neurology, and Cellular & Molecular Medicine, and Program in Genetics, University of Arizona College of Medicine-Phoenix, Phoenix, AZ, United States
s Department of Genetics, Washington University School of Medicine, St. Louis, MO, United States
t Department of Pediatrics, Washington University School of Medicine, St Louis, MO, United States
u Department of Neuropediatrics and Muscle Disorders, University Medical Center, Faculty of Medicine, University of Freiburg, Freiburg, Germany
v Radboud Center for Mitochondrial Medicine, Department of Pediatrics, Amalia Children’s Hospital, Radboud UMC, Nijmegen, Netherlands
w Department of Pediatric Neurology, University Children’s Hospital Zurich, University of Zurich, Zurich, Switzerland
x Lehrstuhl für Neurogenetik, Technische Universität München, Munich, Germany
y Munich Cluster for Systems Neurology (SyNergy), Munich, Germany

Abstract
Objective: ATP synthase (ATPase) is responsible for the majority of ATP production. Nevertheless, disease phenotypes associated with mutations in ATPase subunits are extremely rare. We aimed at expanding the spectrum of ATPase-related diseases. Methods: Whole-exome sequencing in cohorts with 2,962 patients diagnosed with mitochondrial disease and/or dystonia and international collaboration were used to identify deleterious variants in ATPase-encoding genes. Findings were complemented by transcriptional and proteomic profiling of patient fibroblasts. ATPase integrity and activity were assayed using cells and tissues from 5 patients. Results: We present 10 total individuals with biallelic or de novo monoallelic variants in nuclear ATPase subunit genes. Three unrelated patients showed the same homozygous missense ATP5F1E mutation (including one published case). An intronic splice-disrupting alteration in compound heterozygosity with a nonsense variant in ATP5PO was found in one patient. Three patients had de novo heterozygous missense variants in ATP5F1A, whereas another 3 were heterozygous for ATP5MC3 de novo missense changes. Bioinformatics methods and populational data supported the variants’ pathogenicity. Immunohistochemistry, proteomics, and/or immunoblotting revealed significantly reduced ATPase amounts in association to ATP5F1E and ATP5PO mutations. Diminished activity and/or defective assembly of ATPase was demonstrated by enzymatic assays and/or immunoblotting in patient samples bearing ATP5F1A-p.Arg207His, ATP5MC3-p.Gly79Val, and ATP5MC3-p.Asn106Lys. The associated clinical profiles were heterogeneous, ranging from hypotonia with spontaneous resolution (1/10) to epilepsy with early death (1/10) or variable persistent abnormalities, including movement disorders, developmental delay, intellectual disability, hyperlactatemia, and other neurologic and systemic features. Although potentially reflecting an ascertainment bias, dystonia was common (7/10). Interpretation: Our results establish evidence for a previously unrecognized role of ATPase nuclear-gene defects in phenotypes characterized by neurodevelopmental and neurodegenerative features. ANN NEUROL 2022. © 2021 The Authors. Annals of Neurology published by Wiley Periodicals LLC on behalf of American Neurological Association.

Funding details
01GM1920A, 01KU2016A
National Heart, Lung, and Blood InstituteNHLBI2020‐224274, R01 HG009141, U01 HG0011755
National Human Genome Research InstituteNHGRIUM1 HG008900
National Eye InstituteNEI
National Institute of Neurological Disorders and StrokeNINDS1R01 NS106298
Silicon Valley Community FoundationSVCF701900167, DI 1731/2‐2
Univerzita Karlova v PrazeUK
Broad Institute
Chan Zuckerberg InitiativeCZI
Deutsche ForschungsgemeinschaftDFGDFG 458949627, WI 1820/14‐1, ZE 1213/2‐1
Ministerstvo Školství, Mládeže a TělovýchovyMŠMT825575, NV19‐04‐00233
Bundesministerium für Bildung und ForschungBMBF01GM1906A
Austrian Science FundFWF
Else Kröner-Fresenius-StiftungEKFS
Technische Universität MünchenTUM

Document Type: Article
Publication Stage: Article in Press
Source: Scopus