Publications

Hope Center Member Publications

List of publications for June 20, 2022

Randomized Phase 2 Study of ACE-083 in Patients With Charcot-Marie-Tooth Disease” (2022) Neurology

Randomized Phase 2 Study of ACE-083 in Patients With Charcot-Marie-Tooth Disease
(2022) Neurology, 98 (23), pp. E2356-E2367. 

Thomas, F.P.a , Brannagan, T.H.b , Butterfield, R.J.c , Desai, U.d , Habib, A.A.e , Herrmann, D.N.f , Eichinger, K.J.f , Johnson, N.E.g , Karam, C.h , Pestronk, A.i , Quinn, C.j , Shy, M.E.k , Statland, J.M.l , Subramony, S.H.m , Walk, D.n , Stevens-Favorite, K.o , Miller, B.p , Leneus, A.p , Fowler, M.p , Van De Rijn, M.p , Attie, K.M.p

a Hackensack University Medical Center, Hackensack Meridian School of Medicine, Nutley, NJ, United States
b Columbia University Medical Center, New York, NY, United States
c University of Utah, Salt Lake City, United States
d Carolinas Healthcare System Neurosciences Institute, Charlotte, NC, United States
e University of California Irvine, United States
f University of Rochester Medical CenterNY, United States
g Virginia Commonwealth University, Richmond, United States
h Oregon Health and Science University, Portland, United States
i Washington University School of Medicine, St. Louis, MO, United States
j University of Pennsylvania, Philadelphia, United States
k University of Iowa, Iowa City, United States
l University of Kansas Medical Center, Kansas City, United States
m University of Florida, Gainesville, United States
n University of Minnesota, Minneapolis, United States
o Cadent Medical Communications, Llc, A Syneos Health Group Company, New York, NY, United States
p Acceleron Pharma, Cambridge, MA, United States

Abstract
Background and ObjectivesThe goal of this work was to determine whether locally acting ACE-083 is safe and well tolerated and increases muscle volume, motor function, and quality of life (QoL) in adults with Charcot-Marie-Tooth disease (CMT) type 1.MethodsThis phase 2 study enrolled adults with CMT1 or CMTX (N = 63). Part 1 was open label and evaluated the safety and tolerability of different dose levels of ACE-083 for use in part 2. Part 2 was a randomized, placebo-controlled, 6-month study of 240 mg/muscle ACE-083 injected bilaterally into the tibialis anterior muscle, followed by a 6-month, open-label extension in which all patients received ACE-083. Pharmacodynamic endpoints included total muscle volume (TMV; primary endpoint), contractile muscle volume (CMV), and fat fraction. Additional secondary endpoints included 6-minute walk test, 10-m walk/run, muscle strength, and QoL. Safety was assessed with treatment-emergent adverse events (TEAEs) and clinical laboratory tests.ResultsIn part 1 (n = 18), ACE-083 was generally safe and well tolerated at all dose levels, with no serious adverse events, TEAEs of grade 3 or greater, or death reported. In part 2 (n = 45 enrolled, n = 44 treated), there was significantly greater change in TMV with ACE-083 compared with placebo (least-squares mean difference 13.5%; p = 0.0096). There was significant difference between ACE-083 and placebo for CMV and change in ankle dorsiflexion strength. Fat fraction and all other functional outcomes were not significantly improved by ACE-083. Moderate to mild injection-site reactions were the most common TEAEs.DiscussionDespite significantly increased TMV and CMV, patients with CMT receiving ACE-083 in tibialis anterior muscles did not demonstrate greater functional improvement compared with those receiving placebo.Trial Registration InformationClinical Trials Registration: NCT03124459.Classification of EvidenceThis study provides Class II evidence that intramuscular ACE-083 is safe and well tolerated and increases total muscle volume after 6 months of treatment in adults with CMT1 or CMTX. © American Academy of Neurology.

Funding details
National Institutes of HealthNIH
Centers for Disease Control and PreventionCDCDD19-002
U.S. Food and Drug AdministrationFDA7R01FD006071-02
National Institute of Neurological Disorders and StrokeNINDS4K23NS091511, R01NS104010
Friedreich’s Ataxia Research AllianceFARA
Genzyme
Muscular Dystrophy AssociationMDA
CSL Behring
FSH Society
Acceleron
AveXis

Document Type: Article
Publication Stage: Final
Source: Scopus

ecapitulation of endogenous 4R tau expression and formation of insoluble tau in directly reprogrammed human neurons” (2022) Cell Stem Cell

Recapitulation of endogenous 4R tau expression and formation of insoluble tau in directly reprogrammed human neurons
(2022) Cell Stem Cell, 29 (6), pp. 918-932.e8. 

Capano, L.S.a b c , Sato, C.d , Ficulle, E.g , Yu, A.h , Horie, K.d , Kwon, J.-S.a i , Burbach, K.F.a j , Barthélemy, N.R.d , Fox, S.G.h , Karch, C.M.e f k , Bateman, R.J.d e f , Houlden, H.g , Morimoto, R.I.h , Holtzman, D.M.d e f , Duff, K.E.g , Yoo, A.S.a b f

a Department of Developmental Biology, Washington University School of Medicine, St. Louis, MO 63110, United States
b Center for Regenerative Medicine, Washington University School of Medicine, St. Louis, MO 63110, United States
c Program in Molecular and Cell Biology, Washington University School of Medicine, St. Louis, MO, United States
d Department of Neurology, Washington University School of Medicine, St. Louis, MO 63110, United States
e Knight Alzheimer’s Disease Research Center, Washington University School of Medicine, St. Louis, MO 63110, United States
f Hope Center for Neurological Disorders, Knight ADRC, St. Louis, MO 63110, United States
g UK Dementia Research Institute at University College London, London, WC1E 6BT, United Kingdom
h Department of Molecular Biosciences, Rice Institute for Biomedical Research, Northwestern University, Evanston, IL 60208, United States
i Program in Computational and Systems Biology, Washington University School of Medicine, St. Louis, MO, United States
j Program in Molecular Genetics and Genomics, Washington University School of Medicine, St. Louis, MO, United States
k Department of Psychiatry, Washington University School of Medicine, St. Louis, MO 63110, United States

Abstract
Tau is a microtubule-binding protein expressed in neurons, and the equal ratios between 4-repeat (4R) and 3-repeat (3R) isoforms are maintained in normal adult brain function. Dysregulation of 3R:4R ratio causes tauopathy, and human neurons that recapitulate tau isoforms in health and disease will provide a platform for elucidating pathogenic processes involving tau pathology. We carried out extensive characterizations of tau isoforms expressed in human neurons derived by microRNA-induced neuronal reprogramming of adult fibroblasts. Transcript and protein analyses showed that miR neurons expressed all six isoforms with the 3R:4R isoform ratio equivalent to that detected in human adult brains. Also, miR neurons derived from familial tauopathy patients with a 3R:4R ratio altering mutation showed increased 4R tau and the formation of insoluble tau with seeding activity. Our results collectively demonstrate the utility of miRNA-induced neuronal reprogramming to recapitulate endogenous tau regulation comparable with the adult brain in health and disease. © 2022 Elsevier Inc.

Author Keywords
4R tau;  adult human neurons;  insoluble tau;  microRNA-induced neurons;  neuronal reprogramming;  tau isoform ratio;  tau isoforms;  tau seeding;  tauopathy

Funding details
P01 AG026276
P01 AG03991
JIT659H
National Institutes of HealthNIH
National Institute on AgingNIARF1AG056296
National Institute of Neurological Disorders and StrokeNINDSAG063521, K01AG062796, R01 NS095773, R01NS107488
Eli Lilly and Company
Roche
Columbia University
Desert Research InstituteDRI
Cure Alzheimer’s FundCAF
University of WashingtonUW
Institute of Clinical and Translational SciencesICTST32 GM007067
Wellcome TrustWTR56 NS110890
Alzheimer’s Disease Research Center, University of WashingtonADRC, UW
Alzheimer’s Disease Research Center, University of PittsburghADRCP30 AG066444, P50 AG05691
Medical Research CouncilMRC
Alzheimer’s Society
Alzheimer’s Research UKARUK
Eisai

Document Type: Article
Publication Stage: Final
Source: Scopus

Interactions Between Weight Loss and Plasma Neurodegenerative Markers for Determining Cognitive Decline Among Community-Dwelling Older Adults” (2022) The Journals of Gerontology. Series A, Biological Sciences and Medical Sciences

Interactions Between Weight Loss and Plasma Neurodegenerative Markers for Determining Cognitive Decline Among Community-Dwelling Older Adults
(2022) The Journals of Gerontology. Series A, Biological Sciences and Medical Sciences, 77 (6), pp. 1159-1168. 

Giudici, K.V.a , Guyonnet, S.a b , Morley, J.E.c , Nguyen, A.D.c , Aggarwal, G.c , Parini, A.d , Li, Y.e f , Bateman, R.J.e , Vellas, B.a b , de Souto Barreto, P.a b , MAPT/DSA Groupg

a Gerontopole of Toulouse, Institute of Ageing, Toulouse University Hospital (CHU Toulouse), Toulouse, France
b University of Toulouse III, INSERM, UPS, Toulouse, France
c Division of Geriatric Medicine, School of Medicine, Saint Louis University, St. Louis, MO, United States
d Institute of Metabolic and Cardiovascular Diseases (I2MC), INSERM UMR 1048, University of Toulouse III Paul Sabatier, Toulouse, France
e Department of Neurology, Washington University School of Medicine, St. Louis, MO, United States
f Division of Biostatistics, Washington University School of Medicine, St. Louis, MO, United States

Abstract
This study aimed to investigate the interaction between weight loss (WL) and plasma amyloid-β 42/40 (Aβ 42/40), neurofilament light chain (NfL), progranulin, and their association with cognitive decline over time among older adults. This 5-year observational approach included 470 participants from the Multidomain Alzheimer Preventive Trial, mean age 76.8 years (SD = 4.5), 59.4% women. WL was defined as ≥5% decrease over the first year. Biomarkers were measured at 12 months. Cognitive function was assessed yearly from 12 months onward by Mini-Mental State Examination (MMSE); Clinical Dementia Rating sum of boxes (CDR-SB); a composite score based on Category Naming Test; Digit Symbol Substitution Test; 10 MMSE orientation items (MMSEO) and free and total recall of the Free and Cued Selective Reminding test; and these tests individually. Twenty-seven participants (5.7%) presented WL. In adjusted analyses, combined WL + lower Aβ 42/40 (≤0.103, lowest quartile) was related with more pronounced 4-year cognitive decline according to CDR-SB (p < .0001) and MMSEO (p = .021), compared with non-WL + higher Aβ 42/40. WL + higher NfL (>94.55 pg/mL, highest quartile) or progranulin (>38.4 ng/mL, 3 higher quartiles) were related with higher cognitive decline according to CDR-SB, MMSE, MMSEO, and composite score (all p < .03), compared with non-WL + lower NfL or higher progranulin. Regrouping progranulin quartiles (Q1-Q3 vs Q4) revealed higher cognitive decline among the WL + lower progranulin group compared with non-WL + lower progranulin. In conclusion, 1-year WL was associated with subsequent higher 4-year cognitive decline among older adults presenting low Aβ 42/40 or high NfL. Future studies combining plasma biomarker assessments and body weight surveillance may be useful for identifying people at risk of cognitive impairment. Clinical trial number: NCT00672685. © The Author(s) 2022. Published by Oxford University Press on behalf of The Gerontological Society of America. All rights reserved. For permissions, please e-mail: journals.permissions@oup.com.

Author Keywords
Amyloid-β;  Cognition;  Neurofilament light chain;  Progranulin

Document Type: Article
Publication Stage: Final
Source: Scopus

Synthetic gene circuits for preventing disruption of the circadian clock due to interleukin-1-induced inflammation” (2022) Science Advances

Synthetic gene circuits for preventing disruption of the circadian clock due to interleukin-1-induced inflammation
(2022) Science Advances, 8 (21), p. eabj8892. 

Pferdehirt, L.a b c d , Damato, A.R.e , Dudek, M.f , Meng, Q.-J.f , Herzog, E.D.e , Guilak, F.a b c d

a Department of Orthopedic Surgery, Washington University School of Medicine, St. Louis, MO 63110, USA
b Shriners Hospitals for Children-St. Louis, St. Louis, MO 63110, USA
c Center of Regenerative Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
d Department of Biomedical Engineering, Washington University, St. Louis, MO 63105, USA
e Department of Biology, Washington University, St. Louis, MO 63130, USA
f Wellcome Centre for Cell Matrix Research, Division of Cell Matrix Biology and Regenerative Medicine, Faculty of Biology, Medicine and Health, University of Manchester, Oxford RoadManchester M13 9PT, United Kingdom

Abstract
The circadian clock regulates tissue homeostasis through temporal control of tissue-specific clock-controlled genes. In articular cartilage, disruptions in the circadian clock are linked to a procatabolic state. In the presence of inflammation, the cartilage circadian clock is disrupted, which further contributes to the pathogenesis of diseases such as osteoarthritis. Using synthetic biology and tissue engineering, we developed and tested genetically engineered cartilage from murine induced pluripotent stem cells (miPSCs) capable of preserving the circadian clock in the presence of inflammation. We found that circadian rhythms arise following chondrogenic differentiation of miPSCs. Exposure of tissue-engineered cartilage to the inflammatory cytokine interleukin-1 (IL-1) disrupted circadian rhythms and degraded the cartilage matrix. All three inflammation-resistant approaches showed protection against IL-1-induced degradation and loss of circadian rhythms. These synthetic gene circuits reveal a unique approach to support daily rhythms in cartilage and provide a strategy for creating cell-based therapies to preserve the circadian clock.

Document Type: Article
Publication Stage: Final
Source: Scopus

Development of a Comprehensive Battery to Collect Social and Structural Determinants of Health (SSDOH) in Cognitively Normal or Very Mildly Impaired Persons” (2022) Alzheimer Disease and Associated Disorders

Development of a Comprehensive Battery to Collect Social and Structural Determinants of Health (SSDOH) in Cognitively Normal or Very Mildly Impaired Persons
(2022) Alzheimer Disease and Associated Disorders, 36 (2), pp. 97-102. 

Streitz, M.L.a , Denny, A.a , Xiong, C.b , McCue, L.b , Stites, S.D.c , Midgett, S.d , Mechanic-Hamilton, D.e , Moulder, K.L.a , Morris, J.C.a , Balls-Berry, J.a

a Department of Neurology, Knight Alzheimer Disease Research Center, Washington University School of Medicine, 4488 Forest Park Avenue, St. Louis, MO 63108, United States
b Department of Biostatistics, Knight Alzheimer Disease Research Center, Washington University School of Medicine, St. Louis, MI, United States
c Departments of Psychiatry, Perelman School of Medicine, University of Pennsylvania Alzheimer Disease Research Center, Philadelphia, PA, United States
d Department of Medicine, Perelman School of Medicine, University of Pennsylvania Alzheimer Disease Research Center, Philadelphia, PA, United States
e Department of Neurology, Perelman School of Medicine, University of Pennsylvania Alzheimer Disease Research Center, Philadelphia, PA, United States

Abstract
Introduction: Research addressing Alzheimer disease and related dementias must examine nonbiological factors influencing the risk for and expression of Alzheimer disease and related dementias. These factors address the interplay of cognition with lived experiences and social and structural determinants of health (SSDOH). However, coordinated measures of SSDOH are limited. Methods: The Knight Alzheimer Disease Research Center (ADRC) at Washington University in St. Louis developed and piloted a comprehensive battery to measure SSDOH. One hundred and twelve participants, very mildly cognitively impaired or unimpaired, enrolled in memory studies completed the electronic SSDOH battery. The Clinical Dementia Rating (CDR) determined the presence or absence of cognitive impairment. Results: Four domains demonstrated above acceptable intraclass correlation scores for test-retest reliability (≥0.70), including adverse childhood events, discrimination, social status, and early education. Twenty very mildly impaired participants completed the electronic pilot study. Conclusion: Our findings indicate that participants with early-stage symptomatic Alzheimer disease are able to participate in electronic SSDOH data collection. In collaboration with the University of Pennsylvania ADRC, we replaced/modified certain assessments to increase intraclass correlation. The resulting battery, Social and Structural Life-courses Influencing Aging and Dementia (SS-DIAD), can serve as a SSDOH collection tool and is currently utilized in cognitively impaired and unimpaired research participants at both ADRCs. © 2022 Lippincott Williams and Wilkins. All rights reserved.

Author Keywords
Alzheimer disease;  cognition;  older adults;  social and structural determinants of health

Funding details
National Institutes of HealthNIHK23 AG065499
National Institute on AgingNIAP01 AG026276, P01 AG03991, P30 AG010124, P30 AG066444, R01 AG053550, R01 AG067505
Alzheimer’s AssociationAAAACSF-19-617940, AARF-17-528934, K23 AG065442

Document Type: Article
Publication Stage: Final
Source: Scopus