Arts & Sciences Brown School McKelvey School of Engineering School of Medicine Weekly Publications

WashU weekly Neuroscience publications

“In-vivo design feedback and perceived utility of a genetically-informed smoking risk tool among current smokers in the community” (2021) BMC Medical Genomics

In-vivo design feedback and perceived utility of a genetically-informed smoking risk tool among current smokers in the community
(2021) BMC Medical Genomics, 14 (1), art. no. 139, . 

Bourdon, J.L.a , Dorsey, A.b , Zalik, M.b , Pietka, A.b , Salyer, P.b , Bray, M.J.b , Bierut, L.J.b , Ramsey, A.T.b

a Wellbridge Center for Addiction Treatment and Research, Center for Addiction Science, 525 Jan Way, Room 1523, Calverton, NY 11922, United States
b Department of Psychiatry, Washington University in St. Louis, St. Louis, MO, United States

Abstract
Background: The use of genetically-informed personalized risk information for behavioral disorders, namely smoking and smoking-related behaviors, is a promising yet understudied area. The Genetics and Smoking Risk Profile, or RiskProfile, leverages genetic and environmental information to communicate one’s risk for smoking-related diseases. Although prior studies have examined attitudes toward genetic results, little research has investigated these perceptions through a lens of in-vivo testing; that is, user-centered design feedback in response to personalized genetic results being returned contemporaneously. This qualitative study engaged current smokers in usability testing of the RiskProfile within the context of concurrently receiving this personalized, genetically-informed smoking cessation intervention. Methods: Eighty-nine participants who were current smokers responded to open-ended interview questions on perceptions of smoking-related genetic information and the content and format of the RiskProfile intervention that they had received moments before. Data were analyzed via the conventional content analysis approach in which themes were allowed to emerge throughout the analysis. Results: Participants were able to reference and offer design input on specific elements of the RiskProfile. Overall, current smokers perceived the RiskProfile to have high potential utility. Constructive feedback that current smokers offered about the tool centered around suggested improvements to optimize its usability and technical content. Conclusions: The detailed and constructive feedback from participants highlights that in-vivo feedback offers a useful design approach that addresses concerns of rigor and relevance when returning genetic results. This unique method demonstrated perceived utility and constructive design feedback for the RiskProfile among current smokers and can play an important role in optimizing the design and implementation of personalized genetic risk interventions moving forward. © 2021, The Author(s).

Author Keywords
Cessation;  Genetics;  Implementation;  Smoking

Funding details
National Institutes of HealthNIH5T32MH014677, R34DA052928
National Institute on Drug AbuseNIDAT32DA015035
National Cancer InstituteNCIP30CA091842
National Center for Advancing Translational SciencesNCATSUL1TR002345
University of WashingtonUWK12DA041449

Document Type: Article
Publication Stage: Final
Source: Scopus

“Within-subject reaction time variability: Role of cortical networks and underlying neurophysiological mechanisms” (2021) NeuroImage

Within-subject reaction time variability: Role of cortical networks and underlying neurophysiological mechanisms
(2021) NeuroImage, 237, art. no. 118127, . 

Paraskevopoulou, S.E.a , Coon, W.G.b , Brunner, P.a c d e , Miller, K.J.f g h , Schalk, G.a i

a National Center for Adaptive Neurotechnologies, Albany, NY, United States
b Applied Physics Laboratory, Johns Hopkins University, Baltimore, MD, United States
c Department of Neurosurgery, Washington University School of Medicine, St. Louis, MO, United States
d Department of Neurology, Albany Medical College, Albany, NY, United States
e Department of Neurosci and Experimental Therapeutics, Albany Medical College, Albany, NY, United States
f Department of Neurosurgery, Mayo Clinic, Rochester, MN, United States
g Department of Physiology, Mayo Clinic, Rochester, MN, United States
h Department of Biomedical Engineering, Mayo Clinic, Rochester, MN, United States
i Department of Biomedical Science, State University of New York at Albany, Albany, NY, United States

Abstract
Variations in reaction time are a ubiquitous characteristic of human behavior. Extensively documented, they have been successfully modeled using parameters of the subject or the task, but the neural basis of behavioral reaction time that varies within the same subject and the same task has been minimally studied. In this paper, we investigate behavioral reaction time variance using 28 datasets of direct cortical recordings in humans who engaged in four different types of simple sensory-motor reaction time tasks. Using a previously described technique that can identify the onset of population-level cortical activity and a novel functional connectivity algorithm described herein, we show that the cumulative latency difference of population-level neural activity across the task-related cortical network can explain up to 41% of the trial-by-trial variance in reaction time. Furthermore, we show that reaction time variance may primarily be due to the latencies in specific brain regions and demonstrate that behavioral latency variance is accumulated across the whole task-related cortical network. Our results suggest that population-level neural activity monotonically increases prior to movement execution, and that trial-by-trial changes in that increase are, in part, accounted for by inhibitory activity indexed by low-frequency oscillations. This pre-movement neural activity explains 19% of the measured variance in neural latencies in our data. Thus, our study provides a mechanistic explanation for a sizable fraction of behavioral reaction time when the subject’s task is the same from trial to trial. © 2021

Author Keywords
Cortical excitability;  Cortical network;  ECoG;  Electrocorticography;  Reaction time

Funding details
National Institutes of HealthNIHKL2-TR002379, P41-EB018783, P50-MH109429, R01-EB026439, U01-NS108916, U24-NS109103
Army Research OfficeAROW911NF-14-1-0440

Document Type: Article
Publication Stage: Final
Source: Scopus

“Differential classification of states of consciousness using envelope- and phase-based functional connectivity” (2021) NeuroImage

Differential classification of states of consciousness using envelope- and phase-based functional connectivity
(2021) NeuroImage, 237, art. no. 118171, . 

Duclos, C.a b , Maschke, C.a c , Mahdid, Y.a c , Berkun, K.d , Castanheira, J.D.S.b c , Tarnal, V.e , Picton, P.e , Vanini, G.e , Golmirzaie, G.e , Janke, E.e , Avidan, M.S.f , Kelz, M.B.g , Liuzzi, L.h , Brookes, M.J.i , Mashour, G.A.e , Blain-Moraes, S.a b

a Montreal General Hospital, McGill University Health Centre, 1650 Cedar Ave, Montreal, QC, Canada
b School of Physical and Occupational Therapy, McGill University, 3654 Promenade Sir-William-Osler MontrealQC H3G 1Y5, Canada
c Integrated Program in Neuroscience, McGill University, 3801 University Street, Montreal Quebec, H3A 2B4, Canada
d Section on Behavioral Neuroscience, National Institutes of Health, 9000 Rockville Pike, Bethesda, MD 20892, United States
e Center for Consciousness Science and Department of Anesthesiology, University of Michigan Medical School, 1301 Catherine Street, 4102 Medical Science 1, Ann Arbor, MI 48109, United States
f Department of Anesthesiology, Washington University School of Medicine, 660 S. Euclid Ave. St. LouisMO 63110, United States
g Department of Anesthesiology, Perelman School of Medicine, University of Pennsylvania, 3400 Spruce St, Philadelphia, PA 19104, United States
h Mood Brain and Development Unit, Emotion and Development Branch, National Institute of Mental Health, National Institutes of Health, 9000 Rockville Pike, Bethesda, MD 20892, United States
i Sir Peter Mansfield Imaging Centre, School of Physics and Astronomy, University of Nottingham, Nottingham, NG7 2RD, United Kingdom

Abstract
The development of sophisticated computational tools to quantify changes in the brain’s oscillatory dynamics across states of consciousness have included both envelope- and phase-based measures of functional connectivity (FC), but there are very few direct comparisons of these techniques using the same dataset. The goal of this study was to compare an envelope-based (i.e. Amplitude Envelope Correlation, AEC) and a phase-based (i.e. weighted Phase Lag Index, wPLI) measure of FC in their classification of states of consciousness. Nine healthy participants underwent a three-hour experimental anesthetic protocol with propofol induction and isoflurane maintenance, in which five minutes of 128-channel electroencephalography were recorded before, during, and after anesthetic-induced unconsciousness, at the following time points: Baseline; light sedation with propofol (Light Sedation); deep unconsciousness following three hours of surgical levels of anesthesia with isoflurane (Unconscious); five minutes prior to the recovery of consciousness (Pre-ROC); and three hours following the recovery of consciousness (Recovery). Support vector machine classification was applied to the source-localized EEG in the alpha (8–13 Hz) frequency band in order to investigate the ability of AEC and wPLI (separately and together) to discriminate i) the four states from Baseline; ii) Unconscious (“deep” unconsciousness) vs. Pre-ROC (“light” unconsciousness); and iii) responsiveness (Baseline, Light Sedation, Recovery) vs. unresponsiveness (Unconscious, Pre-ROC). AEC and wPLI yielded different patterns of global connectivity across states of consciousness, with AEC showing the strongest network connectivity during the Unconscious epoch, and wPLI showing the strongest connectivity during full consciousness (i.e., Baseline and Recovery). Both measures also demonstrated differential predictive contributions across participants and used different brain regions for classification. AEC showed higher classification accuracy overall, particularly for distinguishing anesthetic-induced unconsciousness from Baseline (83.7 ± 0.8%). AEC also showed stronger classification accuracy than wPLI when distinguishing Unconscious from Pre-ROC (i.e., “deep” from “light” unconsciousness) (AEC: 66.3 ± 1.2%; wPLI: 56.2 ± 1.3%), and when distinguishing between responsiveness and unresponsiveness (AEC: 76.0 ± 1.3%; wPLI: 63.6 ± 1.8%). Classification accuracy was not improved compared to AEC when both AEC and wPLI were combined. This analysis of source-localized EEG data demonstrates that envelope- and phase-based FC provide different information about states of consciousness but that, on a group level, AEC is better able to detect relative alterations in brain FC across levels of anesthetic-induced unconsciousness compared to wPLI. © 2021

Author Keywords
Anesthesia;  Connectivity;  Consciousness;  Electroencephalography;  Machine learning;  Network

Funding details
James S. McDonnell FoundationJSMF
Canadian Institutes of Health ResearchCIHRFRN 152562
Natural Sciences and Engineering Research Council of CanadaNSERCRGPIN-2016–03817
Fonds de recherche du Québec – Nature et technologiesFRQNT

Document Type: Article
Publication Stage: Final
Source: Scopus

“The role of affect in chronic pain: A systematic review of within-person symptom dynamics” (2021) Journal of Psychosomatic Research

The role of affect in chronic pain: A systematic review of within-person symptom dynamics
(2021) Journal of Psychosomatic Research, 147, art. no. 110527, . 

Frumkin, M.R., Rodebaugh, T.L.

Department of Psychological & Brain Sciences, Washington University in St. Louis, St. Louis, MO, United States

Abstract
Objective: Chronic pain is conceptualized as a biopsychosocial phenomenon that involves both physical and emotional processes. The vast majority of research regarding these facets of chronic pain characterizes differences between individuals. In this review, we describe problems with assuming that differences between persons accurately characterize within-person processes. We also provide a systematic review of studies that have examined within-person relationships between pain and affect among individuals with chronic pain. Method: Articles published by December 2020 that pertained to within-person assessment of pain and emotion, affect, or mood were identified. Data regarding study design, adherence, and concurrent and prospective relationships among pain and affect variables were extracted and summarized. Results: Of 611 abstracts, 55 studies met inclusion criteria. Results suggest that individuals with chronic pain tend to experience increased negative affect and decreased positive affect when experiencing more severe pain (rpooled =.18 and −.19, respectively). However, the size of these effects appeared smaller than between-person associations, and there was evidence of significant variability between individuals. Examination of predictive relationships between pain and affect largely suggested the tendency of symptoms to predict themselves, rather than pain predicting affect or vice versa. Conclusions: Consistent with group-level relationships, experiencing more severe pain relative to an individual’s average seems to be associated with more negative affect and less positive affect. However, individuals vary in the size and even direction of these effects. More research is necessary to understand the implications of such variability for the assessment and treatment of chronic pain. © 2021 Elsevier Inc.

Author Keywords
Affect;  Ecological momentary assessment;  Pain

Funding details
National Institutes of HealthNIH5R01DC017451-02

Document Type: Review
Publication Stage: Final
Source: Scopus

“Tracking plasticity of individual human brains” (2021) Current Opinion in Behavioral Sciences

Tracking plasticity of individual human brains
(2021) Current Opinion in Behavioral Sciences, 40, pp. 161-168. 

Newbold, D.J.a , Dosenbach, N.U.a b c d e

a Department of Neurology – Washington University School of Medicine, St. Louis, MO 63110, United States
b Department of Radiology – Washington University School of Medicine, St. Louis, MO 63110, United States
c Department of Pediatrics – Washington University School of Medicine, St. Louis, MO 63110, United States
d Program in Occupational Therapy – Washington University School of Medicine, St. Louis, MO 63110, United States
e Department of Biomedical Engineering – Washington University in St. Louis, St. Louis, MO 63130, United States

Abstract
Understanding how behavior affects human brain organization was the original motivation behind Precision Functional Mapping (PFM), a deep phenotyping approach to human neuroimaging. Here we review the original PFM studies, as well as research investigating the impact of sensory and/or motor deprivation, or disuse, on brain function. Next, we discuss precision functional mapping of brain plasticity, focusing on experiments that tracked casting of the dominant upper extremity with daily resting-state functional MRI scans. Mechanisms that shape brain circuits during early development may persist into adulthood, helping to maintain the organization of disused circuits. © 2021

Funding details
14-011
MH121276, MH122066, MH124567, MH96773, NS088590, NS110332
Child Neurology FoundationCNF
McDonnell Center for Systems Neuroscience
Hope Center for Neurological Disorders
Jacobs Foundation2016121703

Document Type: Review
Publication Stage: Final
Source: Scopus

“Psychotic-like experiences among 9,564 Kenyan adolescents and young adults” (2021) Psychiatry Research

Psychotic-like experiences among 9,564 Kenyan adolescents and young adults
(2021) Psychiatry Research, 302, art. no. 113994, . 

Mamah, D.a , Mutiso, V.N.b , Ndetei, D.M.b c

a Department of Psychiatry, Washington University Medical School, 660 S. Euclid, Saint Louis, Missouri 63110, USA, United States
b Africa Mental Health Research and Training Foundation, Nairobi, Kenya
c Department of Psychiatry, University of Nairobi, Kenya

Abstract
Objective: To investigate the prevalence and characteristics of psychotic-like experiences (PLE) in a large cohort of Kenyan adolescents and young adults. Method: The Washington Early Recognition Center Affectivity and Psychosis (WERCAP) Screen was used to the study the 12-month prevalence of PLE’s among 9,564 Kenyan youths (aged 15-25 yrs), and the rates of psychosis high-risk (HR) and medium-risk (MR) cases, based on cut-off scores. Relationships with clinical, demographic and economic profiles were investigated. Results: Across all participants, 72% reported having had at least one PLE over the last year. 4.6% and 30.6% were HR and MR based on symptom scores. There were similar PLE rates in females and males. PLE severity correlated with mood (r=0.67), stress (r=0.46), and autistic traits (r=0.18). PLE severity was also related to poverty, lower education attainment, and underemployment. Compared to controls, HR and MR youths were more likely to report lifetime substance use and to have more significant use. Conclusion: Psychosis screening can provide valuable information about individuals and may help identify those who may require clinical assessment and intervention to improve outcomes. This is particularly relevant in many parts of Africa where the resources are limited for treating more advanced illness. © 2021

Author Keywords
Africa;  Kenya;  Psychosis;  Psychotic experiences;  Risk;  Schizophrenia

Funding details
National Institute of Mental HealthNIMHR56 MH111300

Document Type: Article
Publication Stage: Final
Source: Scopus

“Time to Treatment in Pediatric Convulsive Refractory Status Epilepticus: The Weekend Effect” (2021) Pediatric Neurology

Time to Treatment in Pediatric Convulsive Refractory Status Epilepticus: The Weekend Effect
(2021) Pediatric Neurology, 120, pp. 71-79. 

Barcia Aguilar, C.a b , Amengual-Gual, M.a c , Sánchez Fernández, I.a d , Abend, N.S.e , Anderson, A.f , Appavu, B.g , Arya, R.h , Brenton, J.N.i , Carpenter, J.L.j , Chapman, K.E.k , Clark, J.a , Farias-Moeller, R.l , Gaillard, W.D.j , Gaínza-Lein, M.m n , Glauser, T.h , Goldstein, J.L.o , Goodkin, H.P.i , Guerriero, R.M.p , Huh, L.q , Lai, Y.-C.r , McDonough, T.L.s , Mikati, M.A.t , Morgan, L.A.u , Novotny, E.J.u , Ostendorf, A.v , Payne, E.T.w , Peariso, K.h , Piantino, J.x , Riviello, J.f , Sannagowdara, K.l , Sheehan, T.a , Sands, T.T.y , Tasker, R.C.z aa , Tchapyjnikov, D.t , Topjian, A.A.ab , Vasquez, A.a ac , Wainwright, M.S.u , Wilfong, A.A.g , Williams, K.g , Loddenkemper, T.a , pSERGad

a Division of Epilepsy and Clinical Neurophysiology, Department of Neurology, Boston Children’s Hospital, Harvard Medical School, Boston, MA, United States
b Department of Child Neurology, Hospital Universitario La Paz, Universidad Autónoma de Madrid, Madrid, Spain
c Pediatric Neurology Unit, Department of Pediatrics, Hospital Universitari Son Espases, Universitat de les Illes Balears, Palma, Spain
d Department of Child Neurology, Hospital Sant Joan de Déu, Universidad de Barcelona, Barcelona, Spain
e Division of Neurology, The Children’s Hospital of Philadelphia, the Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, United States
f Section of Neurology and Developmental Neuroscience, Department of Pediatrics, Baylor College of Medicine, Houston, TX, United States
g Department of Neurosciences, Barrow Neurological Institute at Phoenix Children’s Hospital, Phoenix, AZ, United States
h Division of Pediatric Neurology, Cincinnati Children’s Hospital Medical Center, Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, United States
i Department of Neurology and Pediatrics, University of Virginia Health System, Charlottesville, VA, United States
j Center for Neuroscience, Children’s National Hospital, George Washington University School of Medicine and Health Sciences, Washington, DC, United States
k Departments of Pediatrics and Neurology, Children’s Hospital Colorado, University of Colorado School of Medicine, Aurora, CO, United States
l Department of Pediatric Neurology, Children’s Hospital of Wisconsin, Medical College of Wisconsin, Milwaukee, WI, United States
m Instituto de Pediatría, Facultad de Medicina, Universidad Austral de Chile, Valdivia, Chile
n Servicio de Neuropsiquiatría Infantil, Hospital Clínico San Borja Arriarán, Universidad de Chile, Santiago, Chile
o Davee Pediatric Neurocritical Care Program, Northwestern University Feinberg School of Medicine, Chicago, IL, United States
p Division of Pediatric and Developmental Neurology, Department of Neurology, Washington University School of Medicine, St. Louis, MO, United States
q Department of Pediatrics, British Columbia Children’s Hospital, the University of British ColumbiaBC, Canada
r Section of Pediatric Critical Care Medicine, Department of Pediatrics, Baylor College of Medicine, Houston, TX, United States
s Division of Neurology and Epilepsy, Department of Pediatrics, Ann & Robert H. Lurie Children’s Hospital of Chicago, Northwestern University Feinberg School of Medicine, Chicago, IL, United States
t Division of Pediatric Neurology, Duke University Medical Center, Duke University, Durham, NC, United States
u Division of Pediatric Neurology, Department of Neurology, University of Washington, Seattle, WA, United States
v Division of Pediatric Neurology, Department of Pediatrics, Nationwide Children’s Hospital, The Ohio State University, Columbus, OH, United States
w Division of Neurology, Department of Pediatrics, Alberta Children’s Hospital, Calgary, AB, Canada
x Department of Neurology, Doernbecher Children’s Hospital, Oregon Health & Science University, Portland, OR, United States
y Department of Neurology, Columbia University Medical Center, New York, NY, United States
z Department of Neurology, Boston Children’s Hospital, Harvard Medical School, Boston, MA, United States
aa Department of Anesthesiology, Critical Care and Pain Medicine, Boston Children’s Hospital, Harvard Medical School, Boston, MA, United States
ab Division of Critical Care Medicine, The Children’s Hospital of Philadelphia, The Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, United States
ac Division of Child and Adolescent Neurology, Mayo Clinic, Mayo Clinic School of Medicine, Rochester, MN, United States

Abstract
Background: Time to treatment in pediatric refractory status epilepticus is delayed. We aimed to evaluate the influence of weekends and holidays on time to treatment of this pediatric emergency. Methods: We performed a retrospective analysis of prospectively collected observational data of pediatric patients with refractory status epilepticus. Results: We included 329 patients (56% males) with a median (p25 to p75) age of 3.8 (1.3 to 9) years. The median (p25 to p75) time to first BZD on weekdays and weekends/holidays was 20 (6.8 to 48.3) minutes versus 11 (5 to 35) minutes, P = 0.01; adjusted hazard ratio (HR) = 1.20 (95% confidence interval [CI]: 0.95 to 1.55), P = 0.12. The time to first non-BZD ASM was longer on weekdays than on weekends/holidays (68 [42.8 to 153.5] minutes versus 59 [27 to 120] minutes, P = 0.006; adjusted HR = 1.38 [95% CI: 1.08 to 1.76], P = 0.009). However, this difference was mainly driven by status epilepticus with in-hospital onset: among 108 patients, the time to first non-BZD ASM was longer during weekdays than during weekends/holidays (55.5 [28.8 to 103.5] minutes versus 28 [15.8 to 66.3] minutes, P = 0.003; adjusted HR = 1.65 [95% CI: 1.08 to 2.51], P = 0.01). Conclusions: The time to first non-BZD ASM in pediatric refractory status epilepticus is shorter on weekends/holidays than on weekdays, mainly driven by in-hospital onset status epilepticus. Data on what might be causing this difference may help tailor policies to improve medication application timing. © 2021 Elsevier Inc.

Author Keywords
Clinical neurology;  Epilepsy;  Outcome research;  Pediatric;  Status epilepticus

Funding details
American Epilepsy SocietyAES
Epilepsy FoundationEFEF-213583
Pediatric Epilepsy Research FoundationPERF

Document Type: Article
Publication Stage: Final
Source: Scopus

“The stability of visual perspective and vividness during mental time travel” (2021) Consciousness and Cognition

The stability of visual perspective and vividness during mental time travel
(2021) Consciousness and Cognition, 92, art. no. 103116, . 

Berg, J.J., Gilmore, A.W., Shaffer, R.A., McDermott, K.B.

Washington University in St. Louis, St. Louis, MO, United States

Abstract
When remembering or imagining, people can experience an event from their own eyes, or as an outside observer, with differing levels of vividness. The perspective from, and vividness with, which a person remembers or imagines has been related to numerous individual difference characteristics. These findings require that phenomenology during mental time travel be trait-like—that people consistently experience similar perspectives and levels of vividness. This assumption remains untested. Across two studies (combined N = 295), we examined the stability of visual perspective and vividness across multiple trials and timepoints. Perspective and vividness showed weak within-session stability when reported across just a few trials but showed strong within-session stability when sufficient trials were collected. Importantly, both visual perspective and vividness demonstrated good-to-excellent across-session stability across different delay intervals (two days to six weeks). Overall, our results suggest that people dependably experience similar visual phenomenology across occurrences of mental time travel. © 2021 Elsevier Inc.

Author Keywords
Autobiographical memory;  Episodic future thought;  Field perspective;  Individual differences;  Mental time travel;  Observer perspective;  Phenomenology;  Visual perspective;  Vividness

Funding details
National Science FoundationNSFDGE-1143954
James S. McDonnell FoundationJSMF

Document Type: Article
Publication Stage: Final
Source: Scopus

“Epigenetic regulation during human cortical development: Seq-ing answers from the brain to the organoid” (2021) Neurochemistry International

Epigenetic regulation during human cortical development: Seq-ing answers from the brain to the organoid
(2021) Neurochemistry International, 147, art. no. 105039, . 

Lewis, E.M.A., Kaushik, K., Sandoval, L.A., Antony, I., Dietmann, S., Kroll, K.L.

Department of Developmental Biology, Washington University School of Medicine, 660 S. Euclid Avenue St, Louis, MO 63110, United States

Abstract
Epigenetic regulation plays an important role in controlling gene expression during complex processes, such as development of the human brain. Mutations in genes encoding chromatin modifying proteins and in the non-protein coding sequences of the genome can potentially alter transcription factor binding or chromatin accessibility. Such mutations can frequently cause neurodevelopmental disorders, therefore understanding how epigenetic regulation shapes brain development is of particular interest. While epigenetic regulation of neural development has been extensively studied in murine models, significant species-specific differences in both the genome sequence and in brain development necessitate human models. However, access to human fetal material is limited and these tissues cannot be grown or experimentally manipulated ex vivo. Therefore, models that recapitulate particular aspects of human fetal brain development, such as the in vitro differentiation of human pluripotent stem cells (hPSCs), are instrumental for studying the epigenetic regulation of human neural development. Here, we examine recent studies that have defined changes in the epigenomic landscape during fetal brain development. We compare these studies with analogous data derived by in vitro differentiation of hPSCs into specific neuronal cell types or as three-dimensional cerebral organoids. Such comparisons can be informative regarding which aspects of fetal brain development are faithfully recapitulated by in vitro differentiation models and provide a foundation for using experimentally tractable in vitro models of human brain development to study neural gene regulation and the basis of its disruption to cause neurodevelopmental disorders. © 2021 The Author(s)

Author Keywords
Chromatin;  Epigenetic regulation;  Human brain development;  Neuron;  Organoid;  Pluripotent stem cells

Funding details
National Institutes of HealthNIHR01MH124808, R01NS114551, T32 GM 7067, T32 GM 7067–43, U01 HG007530, U54 HD087011-S1
Institute of Clinical and Translational SciencesICTS
Children’s Discovery InstituteCDI
Colorado Clinical and Translational Sciences InstituteCCTSI
Centre de Recherches MathématiquesCRM
Center of Regenerative Medicine, Washington University in St. LouisCRM, WUSTL

Document Type: Article
Publication Stage: Final
Source: Scopus

“Novel de novo TREX1 mutation in a patient with retinal vasculopathy with cerebral leukoencephalopathy and systemic manifestations mimicking demyelinating disease” (2021) Multiple Sclerosis and Related Disorders

Novel de novo TREX1 mutation in a patient with retinal vasculopathy with cerebral leukoencephalopathy and systemic manifestations mimicking demyelinating disease
(2021) Multiple Sclerosis and Related Disorders, 52, art. no. 103015, . 

Macaron, G.a b , Khoury, J.c , Hajj-Ali, R.A.d , Prayson, R.A.e , Srivastava, S.f , Ehlers, J.P.f , Mamsa, H.g , Liszewski, M.K.h , Jen, J.C.g , Bermel, R.A.a , Ontaneda, D.a

a Mellen Center for Multiple Sclerosis Treatment and Research, Cleveland Clinic, Cleveland, OH, United States
b Faculty of Medicine, Université Saint Joseph de Beyrouth, Beirut, Lebanon
c Neurological Institute, Cleveland Clinic, Cleveland, OH, United States
d Department of Rheumatic and Immunologic Disease, Cleveland Clinic, Cleveland, OH, United States
e Department of Anatomic Pathology, Cleveland Clinic, Cleveland, OH, United States
f Cole Eye Institute, Cleveland Clinic, Cleveland, OH, United States
g Department of Neurology, University of California, Los Angeles, CA, United States
h Division of Rheumatology, Department of Medicine, Washington University, School of Medicine, Saint Louis, MO, United States

Abstract
Retinal vasculopathy with cerebral leukoencephalopathy and systemic manifestations (RVCL-S) is a rare fatal autosomal dominant vasculopathy associated with mutations in the TREX1 gene. Only one de novo case has been reported in the literature. We report the long-term clinical, radiological, and pathological presentation of a patient with a de novo and novel mutation in this gene. Description of the clinical, genetic, imaging and pathologic characteristics is important to better characterize RVCL-S and prevent unnecessary interventions. RVCL-S should be considered in patients with tumefactive brain lesions unresponsive to immunotherapy. © 2021

Author Keywords
de novo;  Leukoencephalopathy;  Novel;  Retinal vasculopathy;  TREX1

Funding details
National Institutes of HealthNIH
National Multiple Sclerosis Society
Novartis

Document Type: Letter
Publication Stage: Final
Source: Scopus

“Spatial and Frequency-specific Electrophysiological Signatures of Tonic Pain Recovery in Humans” (2021) Neuroscience

Spatial and Frequency-specific Electrophysiological Signatures of Tonic Pain Recovery in Humans
(2021) Neuroscience, 465, pp. 23-37. 

Rustamov, N.a b , Sharma, L.c , Chiang, S.N.a b , Burk, C.c d , Haroutounian, S.c d , Leuthardt, E.C.a b e f g

a Department of Neurological Surgery, Washington University School of Medicine, St. Louis, MO, United States
b Center for Innovation in Neuroscience and Technology, Washington University School of Medicine, St. Louis, MO, United States
c Department of Anesthesiology, Washington University School of Medicine, St. Louis, MO, United States
d Washington University Pain Center, St. Louis, MO, United States
e Department of Neuroscience, Washington University School of Medicine, St. Louis, MO, United States
f Department of Mechanical Engineering and Materials Science, Washington University in St. Louis, St. Louis, MO, United States
g Department of Biomedical Engineering, Washington University in St. Louis, Louis, MO, United States

Abstract
The objective of this study was to comprehensively investigate patterns of brain activities associated with pain recovery following experimental tonic pain in humans. Specific electrophysiological features of pain recovery may either be monitored or be modulated through neurofeedback (NF) as a novel chronic pain treatment. The cold pressor test was applied with simultaneous electroencephalogram (EEG) recording. EEG data were acquired, and analyzed to define: (1) EEG power topography patterns of pain recovery; (2) source generators of pain recovery at cortical level; (3) changes in functional connectivity associated with pain recovery; (4) features of phase-amplitude coupling (PAC) as it relates to pain recovery. The novel finding of this study is that recovery from pain was characterized by significant theta power rebound at the left fronto-central area. The sources of theta power over-recovery were located in the left dorsolateral prefrontal cortex (DLPFC), cingulate cortex, left insula and contralateral sensorimotor cortex. These effects were paralleled by theta band connectivity increase within hemispheres in a prefrontal–somatosensory network and interhemispherically between prefrontal and parietal areas. In addition, this study revealed significant reduction in PAC between theta/alpha and gamma oscillations during recovery period following tonic pain. These findings have largely been replicated across two identical sessions. Our study emphasizes the association between pain recovery and left lateral prefrontal theta power rebound, and significant over-recovery of functional connectivity in prefrontal-sensorimotor neural network synchronized at theta frequencies. These findings may provide basis for chronic pain treatment by modulating neural oscillations at theta frequencies in left prefrontal cortex. © 2021 IBRO

Author Keywords
EEG;  experimental tonic pain;  neurofeedback;  pain recovery;  prefrontal cortex

Document Type: Article
Publication Stage: Final
Source: Scopus

“Modular network between postrhinal visual cortex, amygdala, and entorhinal cortex” (2021) Journal of Neuroscience

Modular network between postrhinal visual cortex, amygdala, and entorhinal cortex
(2021) Journal of Neuroscience, 41 (22), pp. 4809-4825. 

Meier, A.M.a b , Wang, Q.a c , Ji, W.a , Ganachaud, J.a , Burkhalter, A.a

a Department of Neuroscience, Washington University School of Medicine in St. Louis, St. Louis, MO 63110, United States
b Boston University College of Engineering, Boston, MA 02215, United States
c Allen Institute for Brain Science, Seattle, WA 98109, United States

Abstract
The postrhinal area (POR) is a known center for integrating spatial with nonspatial visual information and a possible hub for influencing landmark navigation by affective input from the amygdala. This may involve specific circuits within muscarinic acetylcholine receptor 2 (M2)-positive (M21) or M2– modules of POR that associate inputs from the thalamus, cortex, and amygdala, and send outputs to the entorhinal cortex. Using anterograde and retrograde labeling with conventional and viral tracers in male and female mice, we found that all higher visual areas of the ventral cortical stream project to the amygdala, while such inputs are absent from primary visual cortex and dorsal stream areas. Unexpectedly for the presumed salt- and-pepper organization of mouse extrastriate cortex, tracing results show that inputs from the dorsal lateral geniculate nucleus and lateral posterior nucleus were spatially clustered in layer 1 (L1) and overlapped with M21 patches of POR. In contrast, input from the amygdala to L1 of POR terminated in M2– interpatches. Importantly, the amygdalocortical input to M2– interpatches in L1 overlapped preferentially with spatially clustered apical dendrites of POR neurons projecting to amygdala and entorhinal area lateral, medial (ENTm). The results suggest that subnetworks in POR, used to build spatial maps for navigation, do not receive direct thalamocortical M21 patch-targeting inputs. Instead, they involve local networks of M2–interpatches, which are influenced by affective information from the amygdala and project to ENTm, whose cells respond to visual landmark cues for navigation. Copyright © 2021 the authors

Author Keywords
Amygdala;  Entorhinal cortex;  Navigation;  Postrhinal cortex;  Thalamocortical connections;  Visual cortex

Funding details
National Eye InstituteNEIR21-EY-027946, RO1-EY-016184, RO1-EY-020523, RO1-EY-027383
McDonnell Center for Systems Neuroscience

Document Type: Article
Publication Stage: Final
Source: Scopus

“Endogenous erythropoietin concentrations and association with retinopathy of prematurity and brain injury in preterm infants” (2021) PLoS ONE

Endogenous erythropoietin concentrations and association with retinopathy of prematurity and brain injury in preterm infants
(2021) PLoS ONE, 16 (6 June), art. no. e0252655, . 

Fahim, N.M.a , Georgieff, M.K.b , Zhang, L.c , Naisbitt, S.b , Rao, R.B.b , Inder, T.E.d

a Department of Pediatrics, Washington University in St. Louis, St. Louis, MO, United States
b Department of Pediatrics, University of Minnesota, Minneapolis, MN, United States
c Division of Biostatistics, School of Public Health, University of Minnesota, Minneapolis, MN, United States
d Departments of Pediatrics, Neurology and Radiology, Washington University in St. Louis, St. Louis, MO, United States

Abstract
Background: Endogenous erythropoietin (EPO) concentrations vary widely in preterm infants and may be associated with perinatal risk factors and neurological outcomes. Erythropoietin is elevated in fetal hypoxia but is also a potential neuroprotectant. Methods: In a prospective study of 27 infants ≤ 30 weeks gestation, serum erythropoietin concentrations were measured during the first month of life, on day 1 and weeks 1, 2, and 4, and related to perinatal risk factors and outcomes including retinopathy of prematurity and cerebral injury evaluated near term-equivalent post menstrual age using magnetic resonance imaging with quantitative scoring. Results: Lower birth weight was associated with higher EPO concentrations throughout the first 2 weeks of life (r = -0.6, p < 0.01). Higher day 1 and week 1 EPO concentrations were associated with lower Apgar score at 1 minute (r = – 0.5) and 5 minutes (r = -0.7), respectively (p < 0.01). Higher day 1 EPO concentrations and 2-week area under the curve were associated with increased risk (p = 0.01) and severity (r = 0.5, p < 0.02) of retinopathy of prematurity. Higher EPO concentrations at 2 weeks were associated with increased total brain injury score (r = 0.5, p < 0.05). Conclusion: Elevated endogenous erythropoietin concentrations in the first two weeks of life are associated with lower birth weight and increased risk of adverse outcomes. Copyright © 2021 Fahim et al.

Funding details
National Institutes of HealthNIH
National Institute of Child Health and Human DevelopmentNICHD
National Center for Advancing Translational SciencesNCATSUL1-TR002494
Eunice Kennedy Shriver National Institute of Child Health and Human DevelopmentNICHDHD057098-01

Document Type: Article
Publication Stage: Final
Source: Scopus

“Genetic and heritable considerations in patients or families with both intracranial and extracranial aneurysms” (2021) Journal of Neurosurgery

Genetic and heritable considerations in patients or families with both intracranial and extracranial aneurysms
(2021) Journal of Neurosurgery, 134 (6), pp. 1999-2006. 

Huguenard, A.L.a , Gupta, V.P.a , Braverman, A.C.b , Dacey, R.G.a

a Department of Neurosurgery, Washington University, St. Louis, United States
b Cardiovascular Division, Department of Medicine, Washington University, St. Louis, MO, United States

Document Type: Review
Publication Stage: Final
Source: Scopus

“Cognitive Impairment in Survivors of Pediatric Extracranial Solid Tumors and Lymphomas” (2021) Journal of Clinical Oncology: Official Journal of the American Society of Clinical Oncology

Cognitive Impairment in Survivors of Pediatric Extracranial Solid Tumors and Lymphomas
(2021) Journal of Clinical Oncology: Official Journal of the American Society of Clinical Oncology, 39 (16), pp. 1727-1740. 

Foster, R.a b , Zheng, D.J.c , Netson-Amore, K.L.d , Kadan-Lottick, N.S.e

a St Louis Children’s Hospital, St Louis, MO
b Washington University, St Louis, MO
c Boston Children’s Hospital, MA, Boston
d University of Kansas School of Medicine-Wichita, Wichita, United States
e Yale University School of Medicine and Yale Cancer Center, CT, New Haven, United States

Document Type: Article
Publication Stage: Final
Source: Scopus

“MiR-124 synergism with ELAVL3 enhances target gene expression to promote neuronal maturity” (2021) Proceedings of the National Academy of Sciences of the United States of America

MiR-124 synergism with ELAVL3 enhances target gene expression to promote neuronal maturity
(2021) Proceedings of the National Academy of Sciences of the United States of America, 118 (22), art. no. e2015454118, . 

Lu, Y.-L.a b c , Liu, Y.a b , McCoy, M.J.a b d e , Yoo, A.S.a b

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 Developmental, Regenerative and Stem Cell Biology, Washington University School of Medicine, St. Louis, MO 63110, United States
d Program in Molecular Genetics and Genomics, Washington University School of Medicine, St. Louis, MO 63110, United States
e Department of Pathology, Stanford University School of Medicine, Stanford, CA 94305, United States

Abstract
Neuron-enriched microRNAs (miRNAs), miR-9/9* and miR-124 (miR-9/9*-124), direct cell fate switching of human fibroblasts to neurons when ectopically expressed by repressing antineurogenic genes. How these miRNAs function after the repression of fibroblast genes for neuronal fate remains unclear. Here, we identified targets of miR-9/9*-124 as reprogramming cells activate the neuronal program and reveal the role of miR-124 that directly promotes the expression of its target genes associated with neuronal development and function. The mode of miR-124 as a positive regulator is determined by the binding of both AGO and a neuron-enriched RNA-binding protein, ELAVL3, to target transcripts. Although existing literature indicates that miRNA–ELAVL family protein interaction can result in either target gene up-regulation or down-regulation in a context-dependent manner, we specifically identified neuronal ELAVL3 as the driver for miR-124 target gene up-regulation in neurons. In primary human neurons, repressing miR-124 and ELAVL3 led to the down-regulation of genes involved in neuronal function and process outgrowth and cellular phenotypes of reduced inward currents and neurite outgrowth. Our results highlight the synergistic role between miR-124 and RNA-binding proteins to promote target gene regulation and neuronal function. © 2021 National Academy of Sciences. All rights reserved.

Author Keywords
Direct reprogramming;  MicroRNA target;  MiR-124;  Neuronal maturity;  RNA-binding protein

Funding details
National Institutes of HealthNIHDP2NS083372, R01NS107488, RF1AG056296
University of WashingtonUWT32GM081739

Document Type: Article
Publication Stage: Final
Source: Scopus

“Exome sequencing revealed PDE11A as a novel candidate gene for early-onset Alzheimer’s disease” (2021) Human Molecular Genetics

Exome sequencing revealed PDE11A as a novel candidate gene for early-onset Alzheimer’s disease
(2021) Human Molecular Genetics, 30 (9), pp. 811-822. 

Qin, W.a , Zhou, A.a , Zuo, X.a , Jia, L.a , Li, F.a , Wang, Q.a , Li, Y.a , Wei, Y.a , Jin, H.a , Cruchaga, C.b c d , Benitez, B.A.b c , Jia, J.a e f g

a Innovation Center for Neurological Disorders and Department of Neurology, Xuanwu Hospital, Capital Medical University, National Clinical Research Center for Geriatric DiseasesBeijing 100053, China
b Department of Psychiatry, Washington University, St. Louis, MO 63110, USA
c NeuroGenomics and Informatics Center, Washington University, St. Louis, MO 63110, USA
d Department of Genetics, Washington University, St. Louis, MO 63110, USA
e Beijing Key Laboratory of Geriatric Cognitive Disorders, Capital Medical UniversityBeijing 10053, China
f Clinical Center for Neurodegenerative Disease and Memory Impairment, Capital Medical UniversityBeijing 10053, China
g Center of Alzheimer’s Disease, Beijing Institute of Brain Disorders, Collaborative Innovation Center for Brain Disorders, Capital Medical UniversityBeijing 10053, China

Abstract
To identify novel risk genes and better understand the molecular pathway underlying Alzheimer’s disease (AD), whole-exome sequencing was performed in 215 early-onset AD (EOAD) patients and 255 unrelated healthy controls of Han Chinese ethnicity. Subsequent validation, computational annotation and in vitro functional studies were performed to evaluate the role of candidate variants in EOAD. We identified two rare missense variants in the phosphodiesterase 11A (PDE11A) gene in individuals with EOAD. Both variants are located in evolutionarily highly conserved amino acids, are predicted to alter the protein conformation and are classified as pathogenic. Furthermore, we found significantly decreased protein levels of PDE11A in brain samples of AD patients. Expression of PDE11A variants and knockdown experiments with specific short hairpin RNA (shRNA) for PDE11A both resulted in an increase of AD-associated Tau hyperphosphorylation at multiple epitopes in vitro. PDE11A variants or PDE11A shRNA also caused increased cyclic adenosine monophosphate (cAMP) levels, protein kinase A (PKA) activation and cAMP response element-binding protein phosphorylation. In addition, pretreatment with a PKA inhibitor (H89) suppressed PDE11A variant-induced Tau phosphorylation formation. This study offers insight into the involvement of Tau phosphorylation via the cAMP/PKA pathway in EOAD pathogenesis and provides a potential new target for intervention. © The Author(s) 2021. Published by Oxford University Press. All rights reserved. For Permissions, please email: journals.permissions@oup.com.

Document Type: Article
Publication Stage: Final
Source: Scopus

“Long-term Intake of Gluten and Cognitive Function among US Women” (2021) JAMA Network Open

Long-term Intake of Gluten and Cognitive Function among US Women
(2021) JAMA Network Open, 4 (5), . 

Wang, Y.a b , Lebwohl, B.c d , Mehta, R.a b , Cao, Y.e f , Green, P.H.R.c , Grodstein, F.g h , Jovani, M.i , Lochhead, P.a , Okereke, O.I.j k l , Sampson, L.m , Willett, W.C.j l m , Sun, Q.j l m , Chan, A.T.a b j m

a Clinical and Translational Epidemiology Unit, Massachusetts General Hospital, Harvard Medical School, Boston, United States
b Division of Gastroenterology, Massachusetts General Hospital, Harvard Medical School, Boston, United States
c Celiac Disease Center, Department of Medicine, Columbia University College of Physicians and Surgeons, New York, NY, United States
d Department of Epidemiology, Mailman School of Public Health, Columbia University, New York, NY, United States
e Division of Public Health Sciences, Department of Surgery, Washington University in St Louis, St Louis, MO, United States
f Division of Gastroenterology, Department of Medicine, Washington University in St Louis, St Louis, MO, United States
g Rush Alzheimer’s Disease Center, Rush University Medical Center, Chicago, IL, United States
h Department of Internal Medicine, Rush Medical College, Chicago, IL, United States
i Division of Gastroenterology and Hepatology, Johns Hopkins Hospital, Baltimore, MD, United States
j Channing Division of Network Medicine, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, United States
k Department of Psychiatry, Massachusetts General Hospital, Harvard Medical School, Boston, United States
l Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, United States
m Department of Nutrition, Harvard T. H. Chan School of Public Health, Boston, MA, United States

Abstract
Importance: Gluten avoidance has been suggested as having a benefit to cognitive health among the general population, given the link between gluten and cognitive impairment in patients with celiac disease. However, data are lacking in individuals without celiac disease. Objective: To examine whether gluten intake is associated with cognitive function in women without celiac disease. Design, Setting, and Participants: This cohort study included US women who participated in the longitudinal, population-based Nurses’ Health Study II and had not previously or subsequently been diagnosed with celiac disease. Dietary data were collected from 1991 to 2015, and data on cognitive function were collected from 2014 to 2019. Data analysis was conducted from October 2020 to April 2021. Exposures: Energy-adjusted gluten intake, cumulatively averaged across questionnaire cycles prior to cognitive assessment. Main Outcomes and Measures: Three standardized cognitive scores assessed by the validated Cogstate Brief Battery: (1) psychomotor speed and attention score, (2) learning and working memory score, and (3) global cognition score. Higher scores indicated better performance. Results: The cohort included 13494 women (mean [SD] age, 60.6 [4.6] years). The mean (SD) gluten intake was 6.3 (1.6) g/d. After controlling for demographic and lifestyle risk factors in linear regression, no significant differences in standardized cognitive scores (mean [SD], 0 [1]) by quintile of gluten intake were found across highest and lowest quintiles of gluten intake (psychomotor speed and attention: -0.02; 95% CI, -0.07 to 0.03; P for trend =.22; learning and working memory: 0.02; 95% CI, -0.03 to 0.07; P for trend =.30; global cognition: -0.002; 95% CI, -0.05 to 0.05; P for trend =.78). The null associations persisted after additional adjustment for major sources of dietary gluten (ie, refined grains or whole grains), comparing decile categories of gluten intake, using gluten intake updated at each previous questionnaire cycle, or modeling changes in gluten intake. Similarly, these associations were not materially altered in sensitivity analyses that excluded women who had reported cancer or dementia diagnosis or had not completed all dietary assessments. Conclusions and Relevance: In this study, long-term gluten intake was not associated with cognitive scores in middle-aged women without celiac disease. Our results do not support recommendations to restrict dietary gluten to maintain cognitive function in the absence of celiac disease or established gluten sensitivity. © 2021 American Medical Association. All rights reserved.

Funding details
National Institutes of HealthNIHK07 CA218377, K24 DK098311, R01 CA202704, R21 AA027608, R35 CA253185, R37 CA246175, U01 CA176726, UM1 CA167552, UM1 CA186107
Massachusetts General HospitalMGH

Document Type: Article
Publication Stage: Final
Source: Scopus

“A Rapid Motor Task-Based Screening Tool for Parkinsonism in Community-Based Studies” (2021) Frontiers in Neurology

A Rapid Motor Task-Based Screening Tool for Parkinsonism in Community-Based Studies
(2021) Frontiers in Neurology, 12, art. no. 653066, . 

Dlamini, W.W.a , Nielsen, S.a , Ushe, M.a , Nelson, G.b c , Racette, B.A.a b

a Department of Neurology, Washington University School of Medicine, St. Louis, MO, United States
b Faculty of Health Sciences, School of Public Health, University of the Witwatersrand, Parktown, Johannesburg, South Africa
c Institute for Global Health, University College London, London, United Kingdom

Abstract
Background: The prevalence of parkinsonism in developing countries is largely unknown due to difficulty in ascertainment because access to neurologists is often limited. Objective: Develop and validate a parkinsonism screening tool using objective motor task-based tests that can be administered by non-clinicians. Methods: In a cross-sectional population-based sample from South Africa, we evaluated 315 adults, age >40, from an Mn-exposed (smelter) community, using the Unified Parkinson Disease Rating Scale motor subsection 3 (UPDRS3), Purdue grooved pegboard, and kinematic-UPDRS3-based motor tasks. In 275 participants (training dataset), we constructed a linear regression model to predict UPDRS3. We selected motor task summary measures independently associated with UPDRS3 (p < 0.05). We validated the model internally in the remaining 40 participants from the manganese-exposed community (test dataset) using the area under the receiver operating characteristic curve (AUC), and externally in another population-based sample of 90 participants from another South African community with only background levels of environmental Mn exposure. Results: The mean UPDRS3 score in participants from the Mn-exposed community was 9.1 in both the training and test datasets (standard deviation = 6.4 and 6.1, respectively). Together, 57 (18.1%) participants in this community had a UPDRS3 ≥ 15, including three with Parkinson’s disease. In the non-exposed community, the mean UPDRS3 was 3.9 (standard deviation = 4.3). Three (3.3%) had a UPDRS3 ≥ 15. Grooved pegboard time and mean velocity for hand rotation and finger tapping tasks were strongly associated with UPDRS3. Using these motor task summary measures and age, the UPDRS3 predictive model performed very well. In the test dataset, AUCs were 0.81 (95% CI 0.68, 0.94) and 0.91 (95% CI 0.81, 1.00) for cut points for neurologist-assessed UPDRS3 ≥ 10 and UPDRS3 ≥ 15, respectively. In the external validation dataset, the AUC was 0.85 (95% CI 0.73, 0.97) for UPDRS3 ≥ 10. AUCs were 0.76–0.82 when excluding age. Conclusion: A predictive model based on a series of objective motor tasks performs very well in assessing severity of parkinsonism in both Mn-exposed and non-exposed population-based cohorts. © Copyright © 2021 Dlamini, Nielsen, Ushe, Nelson and Racette.

Author Keywords
kinematics;  manganese;  parkinsonism;  predictive model;  receiver operating characteristic

Funding details
K01ES028295, R01ES025991, R01ES026891-S1

Document Type: Article
Publication Stage: Final
Source: Scopus

“Recovery of consciousness and cognition after general anesthesia in humans” (2021) eLife

Recovery of consciousness and cognition after general anesthesia in humans
(2021) eLife, 10, . 

Mashour, G.A.a , Palanca, B.J.b , Basner, M.c , Li, D.a , Wang, W.d , Blain-Moraes, S.a , Lin, N.d , Maier, K.c , Muench, M.b , Tarnal, V.a , Vanini, G.a , Ochroch, E.A.c , Hogg, R.c , Schwartz, M.c , Maybrier, H.b , Hardie, R.c , Janke, E.a , Golmirzaie, G.a , Picton, P.a , McKinstry-Wu, A.R.c , Avidan, M.S.b , Kelz, M.B.c

a Center for Consciousness Science, Department of Anesthesiology, University of Michigan Medical School, Ann Arbor, United States
b Department of Anesthesiology, Washington University School of Medicine, St. Louis, United States
c Department of Anesthesiology and Critical Care, Perelman School of Medicine, University of Pennsylvania, Philadelphia, United States
d Department of Mathematics and Statistics, Washington University, St. Louis, United States

Abstract
Understanding how the brain recovers from unconsciousness can inform neurobiological theories of consciousness and guide clinical investigation. To address this question, we conducted a multicenter study of 60 healthy humans, half of whom received general anesthesia for 3 hr and half of whom served as awake controls. We administered a battery of neurocognitive tests and recorded electroencephalography to assess cortical dynamics. We hypothesized that recovery of consciousness and cognition is an extended process, with differential recovery of cognitive functions that would commence with return of responsiveness and end with return of executive function, mediated by prefrontal cortex. We found that, just prior to the recovery of consciousness, frontal-parietal dynamics returned to baseline. Consistent with our hypothesis, cognitive reconstitution after anesthesia evolved over time. Contrary to our hypothesis, executive function returned first. Early engagement of prefrontal cortex in recovery of consciousness and cognition is consistent with global neuronal workspace theory. © 2021, Mashour et al.

Anesthesia is a state of reversable, controlled unconsciousness. It has enabled countless medical procedures. But it also serves as a tool for scientists to study how the brain regains consciousness after disruptions such as sleep, coma or medical procedures requiring general anesthesia. It is still unclear how exactly the brain regains consciousness, and less so, why some patients do not recover normally after general anesthesia or fail to recover from brain injury. To find out more, Mashour et al. studied the patterns of reemerging consciousness and cognitive function in 30 healthy adults who underwent general anesthesia for three hours. While the volunteers were under anesthesia, their brain activity was measured with an EEG; and their sleep-wake activity was measured before and after the experiment. Each participant took part in a series of cognitive tests designed to measure the reaction speed, memory and other functions before receiving anesthesia, right after the return of consciousness, and then every 30 minutes thereafter. Thirty healthy volunteers who did not have anesthesia also completed the scans and tests as a comparison group. The experiments showed that certain normal EEG patterns resumed just before a person wakes up from anesthesia. The return of thinking abilities was an extended, multistep process, but volunteers recovered their cognitive abilities to nearly the same level as the volunteers within three hours of being deeply anesthetized. Mashour et al. also unexpectedly found that abstract problem-solving resumes early in the process, while other functions such as reaction time and attention took longer to recover. This makes sense from an evolutionary perspective. Sleep leaves individuals vulnerable. Quick evaluation and decision-making skills would be key to respond to a threat upon waking. The experiments confirm that the front of the brain, which handles thinking and decision-making, was especially active around the time of recovery. This suggests that therapies targeting this part of the brain may help people who experience loss of consciousness after a brain injury or have difficulties waking up after anesthesia. Moreover, disorders of cognition, such as delirium, in the days following surgery may be caused by factors other than the lingering effects of anesthetic drugs on the brain.

Author Keywords
anesthesia;  cognition;  consciousness;  frontal cortex;  human;  medicine;  neuroscience

Document Type: Article
Publication Stage: Final
Source: Scopus

“Plasma Neurofilament Light for Prediction of Disease Progression in Familial Frontotemporal Lobar Degeneration” (2021) Neurology

Plasma Neurofilament Light for Prediction of Disease Progression in Familial Frontotemporal Lobar Degeneration
(2021) Neurology, 96 (18), pp. e2296-e2312. 

Rojas, J.C., Wang, P., Staffaroni, A.M., Heller, C., Cobigo, Y., Wolf, A., Goh, S.-Y.M., Ljubenkov, P.A., Heuer, H.W., Fong, J.C., Taylor, J.B., Veras, E., Song, L., Jeromin, A., Hanlon, D., Yu, L., Khinikar, A., Sivasankaran, R., Kieloch, A., Valentin, M.-A., Karydas, A.M., Mitic, L.L., Pearlman, R., Kornak, J., Kramer, J.H., Miller, B.L., Kantarci, K., Knopman, D.S., Graff-Radford, N., Petrucelli, L., Rademakers, R., Irwin, D.J., Grossman, M., Ramos, E.M., Coppola, G., Mendez, M.F., Bordelon, Y., Dickerson, B.C., Ghoshal, N., Huey, E.D., Mackenzie, I.R., Appleby, B.S., Domoto-Reilly, K., Hsiung, G.-Y.R., Toga, A.W., Weintraub, S., Kaufer, D.I., Kerwin, D., Litvan, I., Onyike, C.U., Pantelyat, A., Roberson, E.D., Tartaglia, M.C., Foroud, T., Chen, W., Czerkowicz, J., Graham, D.L., van Swieten, J.C., Borroni, B., Sanchez-Valle, R., Moreno, F., Laforce, R., Graff, C., Synofzik, M., Galimberti, D., Rowe, J.B., Masellis, M., Finger, E., Vandenberghe, R., de Mendonça, A., Tagliavini, F., Santana, I., Ducharme, S., Butler, C.R., Gerhard, A., Levin, J., Danek, A., Otto, M., Sorbi, S., Cash, D.M., Convery, R.S., Bocchetta, M., Foiani, M., Greaves, C.V., Peakman, G., Russell, L., Swift, I., Todd, E., Rohrer, J.D., Boeve, B.F., Rosen, H.J., Boxer, A.L., ALLFTD and GENFI consortia

From the University of California, San Francisco (J.C.R., P.W., A.M.S., Y.C., A.W., S.-Y.M.G., P.A.L., H.W.H., J.C.F., J.B.T., A.M.K., L.L.M., J.K., J.H.K., B.L.M., H.J.S., A.L.B.); UK Dementia Research Centre (C.H., D.M.C., R.S.C., M.B., M.F., C.V.G., G.P., L.R., I.S., E.T., J.D.R.), UCL Institute of Neurology, Queen Square, London; Quanterix Corp (E.V., L.S., A.J., D.H.), Lexington; Novartis Institutes for Biomedical Research Inc (L.Y., A. Khinikar, R.S.), Cambridge, MA; Novartis Pharma AG (A. Kieloch, M.-A.V.), Basel, Switzerland; Bluefield Project to Cure Frontotemporal Dementia (L.L.M., R.P.), San Francisco, CA; Mayo Clinic (K.K., D.S.K., B.F.B.), Rochester, MN; Mayo Clinic (N.G.-R., L.P., R.R.), Jacksonville, FL; University of Pennsylvania (D.J.I., M.G.), Philadelphia; University of California, Los Angeles (E.M.R., G.C., M.F.M., Y.B.); Harvard University/Massachusetts General Hospital (B.D.C.), Boston, MA; Washington University (N.G.), St. Louis, MO; Columbia University (E.D.H.), New York, NY; University of British Columbia (I.R.M., G.-Y.R.H.), Vancouver, Canada; Case Western Reserve University (B.S.A.), Cleveland, OH; University of Washington (K.D.-R.), Seattle; Laboratory of Neuroimaging (A.W.T.), University of Southern California, Los Angeles; Northwestern University (S.W.), Chicago, IL; University of North Carolina (D.I.K.)

Abstract
OBJECTIVE: We tested the hypothesis that plasma neurofilament light chain (NfL) identifies asymptomatic carriers of familial frontotemporal lobar degeneration (FTLD)-causing mutations at risk of disease progression. METHODS: Baseline plasma NfL concentrations were measured with single-molecule array in original (n = 277) and validation (n = 297) cohorts. C9orf72, GRN, and MAPT mutation carriers and noncarriers from the same families were classified by disease severity (asymptomatic, prodromal, and full phenotype) using the CDR Dementia Staging Instrument plus behavior and language domains from the National Alzheimer’s Disease Coordinating Center FTLD module (CDR+NACC-FTLD). Linear mixed-effect models related NfL to clinical variables. RESULTS: In both cohorts, baseline NfL was higher in asymptomatic mutation carriers who showed phenoconversion or disease progression compared to nonprogressors (original: 11.4 ± 7 pg/mL vs 6.7 ± 5 pg/mL, p = 0.002; validation: 14.1 ± 12 pg/mL vs 8.7 ± 6 pg/mL, p = 0.035). Plasma NfL discriminated symptomatic from asymptomatic mutation carriers or those with prodromal disease (original cutoff: 13.6 pg/mL, 87.5% sensitivity, 82.7% specificity; validation cutoff: 19.8 pg/mL, 87.4% sensitivity, 84.3% specificity). Higher baseline NfL correlated with worse longitudinal CDR+NACC-FTLD sum of boxes scores, neuropsychological function, and atrophy, regardless of genotype or disease severity, including asymptomatic mutation carriers. CONCLUSIONS: Plasma NfL identifies asymptomatic carriers of FTLD-causing mutations at short-term risk of disease progression and is a potential tool to select participants for prevention clinical trials. TRIAL REGISTRATION INFORMATION: ClinicalTrials.gov Identifier: NCT02372773 and NCT02365922. CLASSIFICATION OF EVIDENCE: This study provides Class I evidence that in carriers of FTLD-causing mutations, elevation of plasma NfL predicts short-term risk of clinical progression. Copyright © 2021 The Author(s). Published by Wolters Kluwer Health, Inc. on behalf of the American Academy of Neurology.

Document Type: Article
Publication Stage: Final
Source: Scopus

“Deficits in the Skeletal Muscle Transcriptome and Mitochondrial Coupling in Progressive Diabetes-Induced CKD Relate to Functional Decline” (2021) Diabetes

Deficits in the Skeletal Muscle Transcriptome and Mitochondrial Coupling in Progressive Diabetes-Induced CKD Relate to Functional Decline
(2021) Diabetes, 70 (5), pp. 1130-1144. Cited 1 time.

Bittel, D.C.a , Bittel, A.J.a , Varadhachary, A.S.b , Pietka, T.c , Sinacore, D.R.a d

a Program in Physical Therapy, Washington University School of Medicine, St. Louis, MO
b Department of Neurology, Washington University School of Medicine, St. Louis, MO
c Center for Human Nutrition, Washington University School of Medicine, St. Louis, MO
d Department of Physical Therapy, Congdon School of Health Sciences, High Point University, High Point

Abstract
Two-thirds of people with type 2 diabetes mellitus (T2DM) have or will develop chronic kidney disease (CKD), which is characterized by rapid renal decline that, together with superimposed T2DM-related metabolic sequelae, synergistically promotes early frailty and mobility deficits that increase the risk of mortality. Distinguishing the mechanisms linking renal decline to mobility deficits in CKD progression and/or increasing severity in T2DM is instrumental both in identifying those at high risk for functional decline and in formulating effective treatment strategies to prevent renal failure. While evidence suggests that skeletal muscle energetics may relate to the development of these comorbidities in advanced CKD, this has never been assessed across the spectrum of CKD progression, especially in T2DM-induced CKD. Here, using next-generation sequencing, we first report significant downregulation in transcriptional networks governing oxidative phosphorylation, coupled electron transport, electron transport chain (ETC) complex assembly, and mitochondrial organization in both middle- and late-stage CKD in T2DM. Furthermore, muscle mitochondrial coupling is impaired as early as stage 3 CKD, with additional deficits in ETC respiration, enzymatic activity, and increased redox leak. Moreover, mitochondrial ETC function and coupling strongly relate to muscle performance and physical function. Our results indicate that T2DM-induced CKD progression impairs physical function, with implications for altered metabolic transcriptional networks and mitochondrial functional deficits as primary mechanistic factors early in CKD progression in T2DM. © 2021 by the American Diabetes Association.

Document Type: Article
Publication Stage: Final
Source: Scopus

“Medial olivocochlear reflex effects on amplitude growth functions of long- and short-latency components of click-evoked otoacoustic emissions in humans” (2021) Journal of Neurophysiology

Medial olivocochlear reflex effects on amplitude growth functions of long- and short-latency components of click-evoked otoacoustic emissions in humans
(2021) Journal of Neurophysiology, 125 (5), pp. 1938-1953. 

Goodman, S.S.a , Boothalingam, S.b , Lichtenhan, J.T.c

a Department of Communication Sciences and Disorders, University of Iowa, Iowa City, IA, United States
b Department of Communication Sciences and Disorders, Waisman Center, University of Wisconsin-Madison, Madison, WI, United States
c Department of Otolaryngology, Washington University School of Medicine in St. Louis, St. Louis, MO, United States

Abstract
Functional outcomes of medial olivocochlear reflex (MOCR) activation, such as improved hearing in background noise and protection from noise damage, involve moderate to high sound levels. Previous noninvasive measurements of MOCR in humans focused primarily on otoacoustic emissions (OAEs) evoked at low sound levels. Interpreting MOCR effects on OAEs at higher levels is complicated by the possibility of the middle-ear muscle reflex and by components of OAEs arising from different locations along the length of the cochlear spiral. We overcame these issues by presenting click stimuli at a very slow rate and by time-frequency windowing the resulting click-evoked (CE)OAEs into short-latency (SL) and long-latency (LL) components. We characterized the effects of MOCR on CEOAE components using multiple measures to more comprehensively assess these effects throughout much of the dynamic range of hearing. These measures included CEOAE amplitude attenuation, equivalent input attenuation, phase, and slope of growth functions. Results show that MOCR effects are smaller on SL components than LL components, consistent with SL components being generated slightly basal of the characteristic frequency region. Amplitude attenuation measures showed the largest effects at the lowest stimulus levels, but slope change and equivalent input attenuation measures did not decrease at higher stimulus levels. These latter measures are less commonly reported and may provide insight into the variability in listening performance and noise susceptibility seen across individuals.NEW & NOTEWORTHY The auditory efferent system, operating at moderate to high sound levels, may improve hearing in background noise and provide protection from noise damage. We used otoacoustic emissions to measure these efferent effects across a wide range of sound levels and identified level-dependent and independent effects. Previous reports have focused on level-dependent measures. The level-independent effects identified here may provide new insights into the functional relevance of auditory efferent activity in humans.

Author Keywords
auditory;  efferent;  medial olivocochlear;  otoacoustic emissions

Document Type: Article
Publication Stage: Final
Source: Scopus

“Protein kinase D1 variant associated with human epilepsy and peripheral nerve hypermyelination” (2021) Clinical Genetics

Protein kinase D1 variant associated with human epilepsy and peripheral nerve hypermyelination
(2021) Clinical Genetics, . 

Omer, S.a b , Jin, S.C.c d , Koumangoye, R.a , Robert, S.M.e , Duran, D.e , Nelson-Williams, C.c , Huttner, A.f , DiLuna, M.e , Kahle, K.T.e , Delpire, E.a b

a Department of Anesthesiology, Vanderbilt University School of Medicine, Nashville, TN, United States
b Neuroscience Graduate Program, Vanderbilt University Nashville, Nashville, TN, United States
c Department of Genetics, Yale University School of Medicine, New Haven, CT, United States
d Department of Genetics and the McDonnell Genome Institute, Washington University School of Medicine, St. Louis, MO, United States
e Department of Neurosurgery, Yale University School of Medicine, New Haven, CT, United States
f Department of Pathology, Yale University School of Medicine, New Haven, CT, United States

Abstract
We report the case of a patient with severe progressive epilepsy and peripheral neuropathy and a novel de novo inactivating variant (p.E79X) in Protein Kinase D1 (PKD1). Using CRISPR/Cas9, we engineered the homologous variant in mice and showed that in the homozygote mouse, it recapitulated the patient peripheral nerve hypermyelination pathology. The lethality of the homozygote mouse prevented us from performing an assessment of locomotor behavior. The mutant heterozygote mouse; however, exhibited a significant increase in kainate-induced seizure activity over wild-type mice, supporting the hypothesis that the PKD1 variant is a candidate for the cause of the patient epilepsy. Because PKD1 was previously identified in a kinomic screen as an interacting partner of the K-Cl cotransporter 3 (KCC3), and since KCC3 is involved in peripheral nerve disease and brain hyperexcitability, one possible mechanism of action of PKD1 in disease is through KCC3. We show that catalytically inactive PKD1 stimulates KCC3 activity, consistent with tonic relief of inhibitory phosphorylation. Our findings implicate a novel role for PKD1 in the human nervous system, and uncover a mechanism that could serve as a potential target to promote nervous system myelination. © 2021 John Wiley & Sons A/S. Published by John Wiley & Sons Ltd

Author Keywords
biotinylation;  CRISPR/Cas9;  hypermyelination;  kanaic acid;  mouse model;  myelin thickness;  seizure susceptibility;  Xenopus oocytes

Funding details
National Institutes of HealthNIHDK093501
Foundation for Cardiovascular ResearchFCR17CVDO5, CA68485, DK20593, DK58404, DK59637, EY08126, U54HD083211

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

“Are efficient learners of verbal stimuli also efficient and precise learners of visuospatial stimuli?” (2021) Memory

Are efficient learners of verbal stimuli also efficient and precise learners of visuospatial stimuli?
(2021) Memory, . 

Zerr, C.L.a , Spaventa, T.a , McDermott, K.B.a b

a Department of Psychological & Brain Sciences, Washington University in St. Louis, St. Louis, United States
b Department of Radiology, Washington University in St. Louis, St. Louis, United States

Abstract
People differ in how quickly they learn information and how long they remember it, and these two variables are correlated such that people who learn more quickly tend to retain more of the newly learned information. Zerr and colleagues [2018. Learning efficiency: Identifying individual differences in learning rate and retention in healthy adults. Psychological Science, 29(9), 1436–1450] termed the relation between learning rate and retention as learning efficiency, with more efficient learners having both a faster acquisition rate and better memory performance after a delay. Zerr et al. also demonstrated in separate experiments that how efficiently someone learns is stable across a range of days and years with the same kind of stimuli. The current experiments (combined N = 231) replicate the finding that quicker learning coincides with better retention and demonstrate that the correlation extends to multiple types of materials. We also address the generalisability of learning efficiency: A person’s efficiency with learning Lithuanian-English (verbal-verbal) pairs predicts their efficiency with Chinese-English (visuospatial-verbal) and (to a lesser extent) object-location (visuospatial-visuospatial) paired associates. Finally, we examine whether quicker learners also remember material more precisely by using a continuous measure of recall accuracy with object-location pairs. © 2021 Informa UK Limited, trading as Taylor & Francis Group.

Author Keywords
generalisability;  individual differences;  Learning efficiency;  learning rate;  memory

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

“Childhood amyotrophic lateral sclerosis caused by excess sphingolipid synthesis” (2021) Nature Medicine

Childhood amyotrophic lateral sclerosis caused by excess sphingolipid synthesis
(2021) Nature Medicine, . Cited 1 time.

Mohassel, P.a , Donkervoort, S.a , Lone, M.A.b , Nalls, M.a , Gable, K.c , Gupta, S.D.c , Foley, A.R.a , Hu, Y.a , Saute, J.A.M.d , Moreira, A.L.e , Kok, F.f , Introna, A.g , Logroscino, G.g h , Grunseich, C.i , Nickolls, A.R.a , Pourshafie, N.i , Neuhaus, S.B.a , Saade, D.a , Gangfuß, A.j , Kölbel, H.j , Piccus, Z.k , Le Pichon, C.E.k , Fiorillo, C.l , Ly, C.V.m , Töpf, A.n , Brady, L.o , Specht, S.n , Zidell, A.p , Pedro, H.q , Mittelmann, E.r , Thomas, F.P.r , Chao, K.R.s , Konersman, C.G.t , Cho, M.T.u , Brandt, T.u , Straub, V.n , Connolly, A.M.v , Schara, U.j , Roos, A.j , Tarnopolsky, M.o , Höke, A.w , Brown, R.H.x , Lee, C.-H.y , Hornemann, T.b , Dunn, T.M.c , Bönnemann, C.G.a

a Neuromuscular and Neurogenetic Disorders of Childhood Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, United States
b Institute of Clinical Chemistry, University Hospital Zurich, University of Zurich, Zurich, Switzerland
c Department of Biochemistry and Molecular Biology, Uniformed Services University of Health Sciences, Bethesda, MD, United States
d Medical Genetics division and Neurology division, Hospital de Clínicas de Porto Alegre, Porto Alegre, Brazil; Graduate Program in Medicine: Medical Sciences, and Internal Medicine Department; Faculdade de Medicina, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
e Neurology Department, Faculdade de Medicina, Universidade de São Paulo, São Paulo, Brazil
f Neurogenetics Outpatient Service, Neurology Department, Hospital das Clínicas da Universidade de São Paulo, São Paulo, Brazil and Mendelics, São Paulo, Brazil
g Neurology Unit, Department of Basic Medical Sciences, Neurosciences and Sense Organs, University of Bari ‘Aldo Moro’, Bari, Italy
h Department of Clinical Research in Neurology, Center for Neurodegenerative Diseases and the Aging Brain, University of Bari at ‘Pia Fondazione Card G. Panico’ Hospital Tricase (Le), Bari, Italy
i Neurogenetics Branch, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, United States
j Department of Paediatric Neurology, Center for Neuromuscular Disorders in Children and Adolescents, University Clinic Essen, University of Duisburg-Essen, Duisburg-Essen, Germany
k Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, United States
l Paediatric Neurology and Muscular Diseases Unit, G. Gaslini Institute and Department of Neurosciences, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health University of Genoa, Genoa, Italy
m Department of Neurology, Washington University in Saint Louis School of Medicine, Saint Louis, MO, United States
n John Walton Muscular Dystrophy Research Centre, Translational and Clinical Research Institute, Newcastle University and Newcastle Hospitals NHS Foundation Trust, Newcastle upon Tyne, United Kingdom
o Division of Neuromuscular & Neurometabolic Disorders, Department of Paediatrics, McMaster University, Hamilton Health Sciences Centre, Hamilton, ON, Canada
p Center for Genetic and Genomic Medicine, Hackensack University Medical Center, Hackensack, NJ, United States
q Center for Genetic and Genomic Medicine, Hackensack University Medical Center, Hackensack Meridian School of Medicine, Hackensack, NJ, United States
r Department of Neurology, Hereditary Neuropathy Foundation Center of Excellence, Neuroscience Institute, Hackensack University Medical Center, Hackensack Meridian School of Medicine, Hackensack, NJ, United States
s Center for Mendelian Genomics, Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA, United States
t Department of Neurosciences, University of California, San Diego, San Diego, CA, United States
u GeneDx, Gaithersburg, MD, United States
v Department of Paediatrics, Neurology Division, Nationwide Children’s Hospital, Ohio State University, Columbus, OH, United States
w Department of Neurology, Neuromuscular Division, Johns Hopkins School of Medicine, Baltimore, MD, United States
x Department of Neurology, University of Massachusetts Medical School, Worcester, MA, United States
y Department of Structural Biology, St. Jude Children’s Research Hospital, Memphis, TN, United States

Abstract
Amyotrophic lateral sclerosis (ALS) is a progressive, neurodegenerative disease of the lower and upper motor neurons with sporadic or hereditary occurrence. Age of onset, pattern of motor neuron degeneration and disease progression vary widely among individuals with ALS. Various cellular processes may drive ALS pathomechanisms, but a monogenic direct metabolic disturbance has not been causally linked to ALS. Here we show SPTLC1 variants that result in unrestrained sphingoid base synthesis cause a monogenic form of ALS. We identified four specific, dominantly acting SPTLC1 variants in seven families manifesting as childhood-onset ALS. These variants disrupt the normal homeostatic regulation of serine palmitoyltransferase (SPT) by ORMDL proteins, resulting in unregulated SPT activity and elevated levels of canonical SPT products. Notably, this is in contrast with SPTLC1 variants that shift SPT amino acid usage from serine to alanine, result in elevated levels of deoxysphingolipids and manifest with the alternate phenotype of hereditary sensory and autonomic neuropathy. We custom designed small interfering RNAs that selectively target the SPTLC1 ALS allele for degradation, leave the normal allele intact and normalize sphingolipid levels in vitro. The role of primary metabolic disturbances in ALS has been elusive; this study defines excess sphingolipid biosynthesis as a fundamental metabolic mechanism for motor neuron disease. © 2021, This is a U.S. government work and not under copyright protection in the U.S.; foreign copyright protection may apply.

Funding details
National Institutes of HealthNIH
National Heart, Lung, and Blood InstituteNHLBIR01 HG009141, UM1 HG008900
National Human Genome Research InstituteNHGRI
National Eye InstituteNEI
National Institute of Neurological Disorders and StrokeNINDS
Congressionally Directed Medical Research ProgramsCDMRPK08 NS10762, W81XWH-20-1-0219
Stiftung für die Erforschung der MuskelkrankheitenSSEMR01 NS072446
Broad Institute
Schweizerischer Nationalfonds zur Förderung der Wissenschaftlichen ForschungSNF31003A_179371

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

“Maternal Depression, Child Temperament, and Early-Life Stress Predict Never-Depressed Preadolescents’ Functional Connectivity During a Negative-Mood Induction” (2021) Clinical Psychological Science

Maternal Depression, Child Temperament, and Early-Life Stress Predict Never-Depressed Preadolescents’ Functional Connectivity During a Negative-Mood Induction
(2021) Clinical Psychological Science, . 

Liu, P.a , Vandermeer, M.R.J.a , Mohamed Ali, O.a , Daoust, A.R.a , Joanisse, M.F.a , Barch, D.M.b , Hayden, E.P.a

a Department of Psychology, Brain & Mind Institute, Western University, Canada
b Psychological and Brain Sciences, Washington University in St. Louis, United States

Abstract
Understanding the development of depression can inform etiology and prevention/intervention. Maternal depression and maladaptive patterns of temperament (e.g., low positive emotionality [PE] or high negative emotionality, especially sadness) are known to predict depression. Although it is unclear how these risks cause depression, altered functional connectivity (FC) during negative-emotion processing may play an important role. We investigated whether maternal depression and age-3 emotionality predicted FC during negative mood reactivity in never-depressed preadolescents and whether these relationships were augmented by early-life stress. Maternal depression predicted decreased medial prefrontal cortex (mPFC)–amygdala and mPFC–insula FC but increased mPFC–posterior cingulate cortex (PCC) FC. PE predicted increased dorsolateral prefrontal cortex–amygdala FC, whereas sadness predicted increased PCC-based FC in insula, orbitofrontal cortex, and anterior cingulate cortex (ACC). Sadness was more strongly associated with PCC–insula and PCC–ACC FC as early stress increased. Findings indicate that early depression risks may be mediated by FC underlying negative-emotion processing. © The Author(s) 2021.

Author Keywords
functional connectivity;  maternal depression;  negative mood reactivity;  stress;  temperamental risk

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

“Drivers of Hsp104 potentiation revealed by scanning mutagenesis of the middle domain” (2021) Protein Science

Drivers of Hsp104 potentiation revealed by scanning mutagenesis of the middle domain
(2021) Protein Science, . 

Ryan, J.J., Bao, A., Bell, B., Ling, C., Jackrel, M.E.

Department of Chemistry, Washington University, St. Louis, MO, United States

Abstract
Hsp104, a yeast protein disaggregase, can be potentiated via numerous missense mutations at disparate locations throughout the coiled-coil middle domain (MD). Potentiated Hsp104 variants can counter the toxicity and misfolding of TDP-43, FUS, and α-synuclein, proteins which are implicated in neurodegenerative disorders. However, potentiated MD variants typically exhibit off-target toxicity. Further, it has remained confounding how numerous degenerate mutations confer potentiation, hampering engineering of therapeutic Hsp104 variants. Here, we sought to comprehensively define the key drivers of Hsp104 potentiation. Using scanning mutagenesis, we iteratively studied the effects of modulation at each position in the Hsp104 MD. Screening this library to identify enhanced variants reveals that missense mutations at 26% of positions in the MD yield variants that counter FUS toxicity. Modulation of the helix 2–helix 3/4 MD interface potentiates Hsp104, whereas mutations in the analogous helix 1–2 interface do not. Surprisingly, we find that there is a higher likelihood of enhancing Hsp104 activity against human disease substrates than impairing Hsp104 native function. We find that single mutations can broadly destabilize the MD structure and lead to functional potentiation, suggesting this may be a common mechanism conferring Hsp104 potentiation. Using this approach, we have demonstrated that modulation of the MD can yield engineered variants with decreased off-target effects. © 2021 The Protein Society.

Author Keywords
alpha-synuclein;  amyloid;  fused in sarcoma;  Hsp104;  protein disaggregase;  protein misfolding;  TAR DNA-binding protein 43 (TDP-43)

Funding details
National Institutes of HealthNIH
National Institute of General Medical SciencesNIGMSR35GM128772

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

“A Critical Review of Network-Based and Distributional Approaches to Semantic Memory Structure and Processes” (2021) Topics in Cognitive Science

A Critical Review of Network-Based and Distributional Approaches to Semantic Memory Structure and Processes
(2021) Topics in Cognitive Science, . 

Kumar, A.A.a , Steyvers, M.b , Balota, D.A.a

a Psychological & Brain Sciences, Washington University in St. Louis, United States
b Department of Cognitive Sciences, University of California, Irvine, United States

Abstract
Some of the earliest work on understanding how concepts are organized in memory used a network-based approach, where words or concepts are represented as nodes, and relationships between words are represented by links between nodes. Over the past two decades, advances in network science and graph theoretical methods have led to the development of computational semantic networks. This review provides a modern perspective on how computational semantic networks have proven to be useful tools to investigate the structure of semantic memory as well as search and retrieval processes within semantic memory, to ultimately model performance in a wide variety of cognitive tasks. Regarding representation, the review focuses on the distinctions and similarities between network-based (based on behavioral norms) approaches and more recent distributional (based on natural language corpora) semantic models, and the potential overlap between the two approaches. Capturing the type of relation between concepts appears to be particularly important in this modeling endeavor. Regarding processes, the review focuses on random walk models and the degree to which retrieval processes demand attention in pursuit of given task goals, which dovetails with the type of relation retrieved during tasks. Ultimately, this review provides a critical assessment of how the network perspective can be reconciled with distributional and machine-learning-based perspectives to meaning representation, and describes how cognitive network science provides a useful conceptual toolkit to probe both the structure and retrieval processes within semantic memory. © 2021 Cognitive Science Society LLC

Author Keywords
Cognitive network science;  Distributional semantic models;  Semantic memory;  Semantic networks

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

“Behavioral and cognitive functioning in individuals with Cantú syndrome” (2021) American Journal of Medical Genetics, Part A

Behavioral and cognitive functioning in individuals with Cantú syndrome
(2021) American Journal of Medical Genetics, Part A, . 

Roessler, H.I.a , van der Heuvel, L.M.b , Shields, K.c , Guilliams, K.P.d e , Knoers, N.V.A.M.f , van Haaften, G.a , Grange, D.K.c g , van Haelst, M.M.b h

a Department of Genetics, Center for Molecular Medicine, University Medical Center Utrecht, Utrecht University, Utrecht, Netherlands
b Department of Clinical Genetics, Amsterdam University Medical Centers, Amsterdam, Netherlands
c Division of Genetics and Genomic Medicine, Department of Pediatrics, Washington University School of Medicine, St. Louis, MO, United States
d Division of Pediatric Neurology, Department of Neurology, Washington University School of Medicine, St. Louis, MO, United States
e Division of Pediatric Critical Care, Department of Pediatrics, Washington University School of Medicine, St. Louis, MO, United States
f Department of Genetics, University Medical Center Groningen, Groningen, Netherlands
g Center for the Investigation of Membrane Excitability Diseases (CIMED), St. Louis, MO, United States
h Department of Clinical Genetics, VU Medical Center, Amsterdam University Medical Centers, Amsterdam, Netherlands

Abstract
Cantú syndrome (CS) is caused by pathogenic variants in ABCC9 and KCNJ8 encoding the regulatory and pore-forming subunits of ATP-sensitive potassium (KATP) channels. CS is characterized by congenital hypertrichosis, distinctive facial features, peripheral edema, and cardiac and neurodevelopmental abnormalities. Behavioral and cognitive issues have been self-reported by some CS individuals, but results of formal standardized investigations have not been published. To assess the cognitive profile, social functioning, and psychiatric symptoms in a large group of CS subjects systematically in a cross-sectional manner, we invited 35 individuals (1–69 years) with confirmed ABCC9 variants and their relatives to complete various commonly applied standardized age-related questionnaires, including the Kaufman brief intelligence test 2, the social responsiveness scale-2, and the Achenbach system of empirically based assessment. The majority of CS individuals demonstrated average verbal and nonverbal intelligence compared to the general population. Fifteen percent of cases showed social functioning strongly associated with a clinical diagnosis of autism spectrum disorder. Both externalizing and internalizing problems were also present in this cohort. In particular, anxiety, anxiety or attention deficit hyperactivity disorder, and autism spectrum behaviors were predominantly observed in the younger subjects in the cohort (≥25%), but this percentage decreased markedly in adults. © 2021 Wiley Periodicals LLC.

Author Keywords
autism;  behavior;  Cantú syndrome;  DSM V;  intelligence;  psychiatric symptoms

Funding details
National Institutes of HealthNIHHD103347

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

“An Update from the American Stroke Association and the Stroke Council” (2021) Stroke

An Update from the American Stroke Association and the Stroke Council
(2021) Stroke, pp. E269-E271.

Schwamm, L.H.a , Panagos, P.D.b , Mohl, S.M.c

a Massachusetts General Hospital, Wang Ambulatory Care Center, 55 Fruit St, Boston, MA 02114, United States
b Washington University of St. LouisMO, United States
c American Stroke Association Division, American Heart Association, Dallas, TX, United States

Author Keywords
brain;  COVID-19;  goal;  racism;  registries

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

“Characterizing Family Contextual Factors and Relationships with Child Behavior and Sleep Across the Buffering Toxic Stress Consortium” (2021) Prevention Science

Characterizing Family Contextual Factors and Relationships with Child Behavior and Sleep Across the Buffering Toxic Stress Consortium
(2021) Prevention Science, .

Phu, T.a , Miles, E.a , Dominguez, A.a , Hustedt, J.b , Watamura, S.E.a , Berlin, L.c , Harden, B.J.c , Blair, C.d , Constantino, J.N.e , Hallam, R.A.f , Han, M.f , Hustedt, J.T.f , Vu, J.A.f , Sarche, M.g , BTS Consortium Principal Investigatorsh

a Department of Psychology, University of Denver, Denver, CO, United States
b Department of Human Development and Family Sciences, University of Delaware, Newark, DE, United States
c University of Maryland, United States
d New York University, United States
e Washington University in St. Louis, United States
f University of Delaware, United States
g University of Colorado Anschutz, United States

Abstract
The Buffering Toxic Stress (BTS) consortium included six sites in locations that varied widely in racial/ethnic composition and population density. Each site tested a promising parent–child intervention designed to supplement Early Head Start (EHS) services and prevent “toxic stress.” To better understand family risk in a large and diverse EHS sample, studies gathered extensive data on family risk exposure, including demographic risk factors (single mother, unemployed, less than high school education or its equivalent, and neighborhood safety), income-to-needs ratio, household resource constraints, perceptions of economic hardship and pressure, caregiver mental health, and caregiver-reported dysfunctional parent–child interactions. Results presented here for all six sites offer context for the more targeted studies in this special issue. Average levels of family characteristics and child behavior varied by site. We also characterized associations between family characteristics, observer-rated child temperament, and child outcomes (i.e., caregiver-reported child behavior problems and behavioral sleep quality), controlling for child age; these relationships were similar across sites. Demographic risk and caregiver mental health problems were positively associated with child behavior problems, with low income-to-needs ratio and increased financial strain relating to behavioral problems in infancy and toddlerhood. Caregiver mental health problems, financial strain, and social and affect temperament dimensions were related to increased behavioral sleep problems. Dysfunctional parent–child interactions and household resource constraints did not demonstrate statistically significant associations. Findings suggest helpful targets to increase effectiveness of parent–child interventions in early childhood on behavior and sleep outcomes. © 2021, Society for Prevention Research.

Author Keywords
Child behavior;  Child sleep;  Early Head Start;  Family risk and protective factors

Funding details
90YR0058
Administration for Children and FamiliesACF
University of DelawareUD90YR0055
New York UniversityNYU90YR0057
Washington University in St. LouisWUSTL90YR0054
University of DenverDU90YR0056
Office of Planning, Research and EvaluationOPRE
School of Social Work, University of MarylandSSW, UMB90YR0059

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

“Endovascular reperfusion of M2 occlusions in acute ischemic stroke reduced disability and mortality: ETIS Registry results” (2021) Journal of NeuroInterventional Surgery

Endovascular reperfusion of M2 occlusions in acute ischemic stroke reduced disability and mortality: ETIS Registry results
(2021) Journal of NeuroInterventional Surgery, . 

Muszynski, P.a , Anadani, M.b c , Richard, S.d , Marnat, G.e , Bourcier, R.f , Sibon, I.g , Dargazanli, C.h , Arquizan, C.i , Maïer, B.j , Blanc, R.j , Lapergue, B.k , Consoli, A.l , Eugene, F.m , Vannier, S.n , Spelle, L.o , Denier, C.p , Boulanger, M.q , Gauberti, M.r , Saleme, S.s , Macian, F.t , Clarençon, F.u v , Rosso, C.w , Naggara, O.x , Turc, G.y , Ozkul-Wermester, O.z , Papagiannaki, C.aa , Viguier, A.ab , Cognard, C.ac , Le Bras, A.ad , Evain, S.ae , Wolff, V.af , Pop, R.ag , Timsit, S.ah , Gentric, J.-C.ai , Bourdain, F.aj , Veunac, L.ak , Gory, B.a al , Finitsis, S.N.am an

a Diagnostic and Therapeutic Neuroradiology, Université de Lorraine, CHRU-Nancy, Nancy, France
b Washington University, School of Medicine in St Louis, St Louis, MO, United States
c Neurology, Medical University of South Carolina, College of Medicine, Charleston, SC, United States
d Neurology, Stroke Unit, Université de Lorraine, CHRU-Nancy, Nancy, France
e Interventional and Diagnostic Neuroradiology, CHU Bordeaux, Bordeaux, France
f Neuroradiology, University Hospital of Nantes, Nantes, France
g Neurology, Stroke Unit, CHU Bordeaux, Bordeaux, France
h Neuroradiology, CHRU Gui de Chauliac, Montpellier, France
i Neurology, CHRU Gui de Chauliac, Montpellier, France
j Interventional Neuroradiology, Rothschild Foundation, Paris, France
k Neurology, Stroke Unit, Foch Hospital, Suresnes, France
l Diagnostic and Therapeutic Neuroradiology, Foch Hospital, Suresnes, France
m Neuroradiology, CHU Rennes, Rennes, France
n Neurology, Stroke Unit, CHU Rennes, Rennes, France
o Interventional Neuroradiolology, CHU Bicêtre, Le Kremlin-Bicêtre, France
p Neurology, Stroke Unit, CHU Bicêtre, Le Kremlin-Bicêtre, France
q Neurology, Stroke Unit, CHU Caen, Caen, France
r Neuroradiolology, CHU Caen, Caen, France
s Neuroradiology, CHU Limoges, Limoges, France
t Neurology, Stroke Unit, CHU Limoges, Limoges, France
u Sorbonne Université, Paris, France
v Neuroradiology, CHU Pitié-Salpêtrière, Paris, France
w Neurology, Stroke Unit, CHU Pitié-Salpêtrière, Paris, France
x Neuroradiology, Hôpital St Anne, Paris, France
y Neurology, Stroke Unit, Hôpital Saint Anne, Paris, France
z Neurology, Stroke Unit, CHU Rouen, Rouen, France
aa Interventional Neuroradiolology, CHU Rouen, Rouen, France
ab Neurology, Stroke Unit, CHU Toulouse, Toulouse, France
ac Diagnostic and Therapeutic Neuroradiology, CHU Toulouse, Toulouse, France
ad Radiology, CH Bretagne Atlantique, Vannes, France
ae Neurology, Stroke Unit, CH Bretagne Atlantique, Vannes, France
af Stroke Unit, CHU Strasbourg, Strasbourg, France
ag Interventional Neuroradiolology, CHU Strasbourg, Strasbourg, France
ah Neurology, Stroke Unit, CHU Brest, Brest, France
ai Interventionl Neuroradiolology, CHU Brest, Brest, France
aj Neurology, Stroke Unit, CH Côte Basque, Bayonne, France
ak Radiolology, CH Côte Basque, Bayonne, France
al IADI, INSERM U1254, Université de Lorraine, Nancy, France
am Neuroradiolology, University General Hospital of Thessaloniki AHEPA, Thessaloniki, Greece
an Radiology, Centre Hospitalier de l’Universite de Montreal, Montreal, QC, Canada

Abstract
Background: The predictors of successful reperfusion and the effect of reperfusion after endovascular treatment (EVT) for M2 occlusions have not been well studied. We aimed to identify predictors of successful reperfusion and the effect of reperfusion on outcomes of EVT for M2 occlusions in current practice. Methods: Patients with acute ischemic stroke due to isolated M2 occlusions who were enrolled in the prospective multicenter Endovascular Treatment in Ischemic Stroke (ETIS) Registry in France between January 2015 and March 2020 were included. The primary outcome was a favorable outcome, defined as modified Rankin Scale (mRS) score of 0-2 at 90 days. Successful reperfusion was defined as an improvement of ≥1 points in the modified Thrombolysis In Cerebral Infarction score between the first and the last intracranial angiogram. Results: A total of 458 patients were included (median National Institutes of Health Stroke Scale (NIHSS) score 14; 61.4% received prior intravenous thrombolysis). Compared with the non-reperfused patients, reperfused patients had an increased rate of excellent outcome (OR 2.3, 95% CI 0.98 to 5.36; p=0.053), favorable outcome (OR 2.79, 95% CI 1.31 to 5.93; p=0.007), and reduced 90-day mortality (OR 0.39, 95% CI 0.19 to 0.79; p<0.01). Admission NIHSS score was the only predictor of successful reperfusion. First-line strategy was not a predictor of successful reperfusion or favorable outcome, but the use of a stent retriever, alone or with an aspiration catheter, was associated with higher rates of procedural complications and 90-day mortality. Conclusions: Successful reperfusion of M2 occlusions reduced disability and mortality. However, safety is a concern, especially if the procedure failed. © Author(s) (or their employer(s)) 2021. No commercial re-use. See rights and permissions. Published by BMJ.

Author Keywords
stroke;  thrombectomy

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

“Blood Pressure Management after Endovascular Therapy: An Ongoing Debate” (2021) Stroke

Blood Pressure Management after Endovascular Therapy: An Ongoing Debate
(2021) Stroke, pp. E263-E265.

Anadani, M.a , De Havenon, A.b , Mistry, E.c , Anderson, C.S.d

a Department of Neurology, Washington University in St. Louis, School of Medicine, St Louis, MO 63110, United States
b University of Utah, Salt Lake City, United States
c Vanderbilt University Medical CenterTN, United States
d George Institute for Global Health, Faculty of Medicine, University of New South Wales, Sydney, Australia

Author Keywords
acute ischemic stroke;  blood pressure;  clinical trial;  meta-analysis;  thrombectomy

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