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

WashU weekly Neuroscience publications

“Contribution of animal models toward understanding resting state functional connectivity” (2021) NeuroImage

Contribution of animal models toward understanding resting state functional connectivity(2021) NeuroImage, 245, art. no. 118630, . 

Pais-Roldán, P.a , Mateo, C.b , Pan, W.-J.c , Acland, B.d , Kleinfeld, D.b e , Snyder, L.H.d , Yu, X.f , Keilholz, S.c

a Medical Imaging Physics, Institute of Neuroscience and Medicine 4, Forschungszentrum Jülich52425, Germanyb Department of Physics, University of California San Diego, La Jolla, CA 92093, United Statesc Wallace H. Coulter Department of Biomedical Engineering, Emory University/Georgia Institute of Technology, Atlanta, GA 30322, United Statesd Department of Neuroscience, Washington University School of Medicine, Saint Louis, MO 63110, United Statese Section of Neurobiology, University of California, La Jolla, San Diego, CA 92093, United Statesf Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA 02129, United States

AbstractFunctional connectivity, which reflects the spatial and temporal organization of intrinsic activity throughout the brain, is one of the most studied measures in human neuroimaging research. The noninvasive acquisition of resting state functional magnetic resonance imaging (rs-fMRI) allows the characterization of features designated as functional networks, functional connectivity gradients, and time-varying activity patterns that provide insight into the intrinsic functional organization of the brain and potential alterations related to brain dysfunction. Functional connectivity, hence, captures dimensions of the brain’s activity that have enormous potential for both clinical and preclinical research. However, the mechanisms underlying functional connectivity have yet to be fully characterized, hindering interpretation of rs-fMRI studies. As in other branches of neuroscience, the identification of the neurophysiological processes that contribute to functional connectivity largely depends on research conducted on laboratory animals, which provide a platform where specific, multi-dimensional investigations that involve invasive measurements can be carried out. These highly controlled experiments facilitate the interpretation of the temporal correlations observed across the brain. Indeed, information obtained from animal experimentation to date is the basis for our current understanding of the underlying basis for functional brain connectivity. This review presents a compendium of some of the most critical advances in the field based on the efforts made by the animal neuroimaging community. © 2021 The Author(s)

Funding detailsNational Science FoundationNSF1533260, 1822606, DK R35NS097265, R01EY012135, R01MH111438, R01NS122904, R34NS118618, RF1NS113278

Document Type: ArticlePublication Stage: FinalSource: Scopus

“Cardiovascular autonomic nervous system function and hip fracture risk: the Cardiovascular Health Study” (2021) Archives of Osteoporosis

Cardiovascular autonomic nervous system function and hip fracture risk: the Cardiovascular Health Study(2021) Archives of Osteoporosis, 16 (1), art. no. 163, . 

Stein, P.K.a , Buzkova, P.b , Fink, H.A.c d , Robbins, J.A.e , Mukamal, K.J.f , Cauley, J.A.g , Carbone, L.h i , Elam, R.h i , McMillan, D.W.j , Valderrabano, R.k , Barzilay, J.I.l

a Division of Cardiology, Washington University School of Medicine, St. Louis, MO, United Statesb Department of Biostatistics, University of Washington School of Public Health, Seattle, WA, United Statesc Geriatric Research Education and Clinical Center, VA Health Care System, Minneapolis, MN, United Statesd Department of Medicine, University of Minnesota, Minneapolis, MN, United Statese Department of Medicine, University of California, Davis, Sacramento, CA, United Statesf Department of Medicine, Beth Israel Deaconess Medical Center, Brookline, MA, United Statesg Department of Epidemiology, Graduate School of Public Health, University of Pittsburgh, Pittsburgh, PA, United Statesh Veterans Affairs Medical Center, Augusta, GA, United Statesi Department of Medicine, Division of Rheumatology, Medical College of Georgia at Augusta University, Augusta, GA, United Statesj Division of Rehabilitation Medicine, University of Miami School of Medicine, Miami, FL, United Statesk Division of Rheumatology, University of Miami School of Medicine, Miami, FL, United Statesl Division of Endocrinology, Kaiser Permanente of Georgia and Division of Endocrinology, Emory University School of Medicine, Atlanta, GA, United States

AbstractSummary: Among 1299 older adults with 24-h Holter monitoring data at baseline, followed for approximately 15 years, 190 incident hip fractures occurred. Increased heart rate variability was independently associated with reduced risk of hip fracture among female participants. Purpose: Autonomic nervous system function modulates bone remodeling in rodent osteoporosis models. We tested whether cardiovascular autonomic function is associated with hip fracture risk in humans. Methods: Participants were 1299 subjects from the Cardiovascular Health Study (mean age 72.8 years). Eight heart rate variability (HRV) measures (time and frequency domains, detrended fluctuation analysis variables, and heart rate turbulence) were derived from 24-h Holter monitor scans in sinus rhythm. Median follow-up for incident hip fracture was 14.7 years [IQR 9.1, 20.2]. Cox proportional hazards models were used to calculate hazard ratios (95% confidence intervals, CI). Results: There were 144 hip fractures among 714 women (1.31 [1.06, 1.61] per 100-person years) and 46 among 585 men (0.62 [0.43, 0.90] per 100 person-years). From among HRV variables examined, a one standard deviation (SD) higher variation between normal heart beats over 24 h (the SD of NN intervals [SDNN]) was associated with a multivariable-adjusted lower hip fracture risk (HR= 0.80; 95% CI 0.65–0.99; p = 0.04) in women. The adjusted association between very low frequency power, and hip fracture was borderline statistically significant in women (HR= 0.82; 95% CI, 0.66–1.00; p = 0.06). When the 8 HRV variables were considered conjointly and adjusted for each other’s association with hip fracture risk, a 1 SD higher SDNN value was significantly associated with reduced hip fracture risk in women (HR 0.74; 95% CI, 0.50–0.99; p = 0.05). No HRV variables were associated with hip fracture in men. Conclusions: In older women, increased heart rate variation is associated with hip fracture risk. © 2021, International Osteoporosis Foundation and National Osteoporosis Foundation.

Author KeywordsDetrended fluctuation analysis;  Frequency domain;  Heart rate turbulence;  Heart rate variation;  Hip fracture;  Time domain

Funding detailsFoundation for the National Institutes of HealthFNIH

Document Type: ArticlePublication Stage: FinalSource: Scopus

“Ancestry may confound genetic machine learning: Candidate-gene prediction of opioid use disorder as an example” (2021) Drug and Alcohol Dependence

Ancestry may confound genetic machine learning: Candidate-gene prediction of opioid use disorder as an example(2021) Drug and Alcohol Dependence, 229, art. no. 109115, . 

Hatoum, A.S.a , Wendt, F.R.b , Galimberti, M.b , Polimanti, R.b c , Neale, B.d e , Kranzler, H.R.f g , Gelernter, J.b c h i , Edenberg, H.J.j k , Agrawal, A.a

a Washington University in St. Louis, School of Medicine, Department of Psychiatry, United Statesb Department of Psychiatry, Division of Human Genetics, Yale School of Medicine, New Haven, CT, United Statesc Veterans Affairs Connecticut Healthcare System, West Haven, CT, United Statesd Analytic and Translational Genetics Unit, Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA, United Statese Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, United Statesf Center for Studies of Addiction, Department of Psychiatry, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, United Statesg VISN 4 MIRECC, Crescenz VAMC, Philadelphia, PA, United Statesh Department of Genetics, Yale School of Medicine, New Haven, CT, United Statesi Department of Neuroscience, Yale School of Medicine, New Haven, CT, United Statesj Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN, United Statesk Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, IN, United States

AbstractBackground: Machine learning (ML) models are beginning to proliferate in psychiatry, however machine learning models in psychiatric genetics have not always accounted for ancestry. Using an empirical example of a proposed genetic test for OUD, and exploring a similar test for tobacco dependence and a simulated binary phenotype, we show that genetic prediction using ML is vulnerable to ancestral confounding. Methods: We utilize five ML algorithms trained with 16 brain reward-derived “candidate” SNPs proposed for commercial use and examine their ability to predict OUD vs. ancestry in an out-of-sample test set (N = 1000, stratified into equal groups of n = 250 cases and controls each of European and African ancestry). We rerun analyses with 8 random sets of allele-frequency matched SNPs. We contrast findings with 11 genome-wide significant variants for tobacco smoking. To document generalizability, we generate and test a random phenotype. Results: None of the 5 ML algorithms predict OUD better than chance when ancestry was balanced but were confounded with ancestry in an out-of-sample test. In addition, the algorithms preferentially predicted admixed subpopulations. Random sets of variants matched to the candidate SNPs by allele frequency produced similar bias. Genome-wide significant tobacco smoking variants were also confounded by ancestry. Finally, random SNPs predicting a random simulated phenotype show that the bias attributable to ancestral confounding could impact any ML-based genetic prediction. Conclusions: Researchers and clinicians are encouraged to be skeptical of claims of high prediction accuracy from ML-derived genetic algorithms for polygenic traits like addiction, particularly when using candidate variants. © 2021 Elsevier B.V.

Author KeywordsAlgorithmic bias;  Ancestry;  Candidate genes;  Machine learning;  Opioid use disorder

Funding detailsNational Institutes of HealthNIHR01 AA017535, R01 AA11330, R01 DA12690, R01 DA12849, R01 DA18432, RC2 DA028909

Document Type: ArticlePublication Stage: FinalSource: Scopus

“Socio-demographic factors related to parent engagement in the NICU and the impact of the SENSE program” (2021) Early Human Development

Socio-demographic factors related to parent engagement in the NICU and the impact of the SENSE program(2021) Early Human Development, 163, art. no. 105486, . 

Whitehill, L.a b , Smith, J.c , Colditz, G.d , Le, T.e , Kellner, P.f , Pineda, R.e f g h i

a School of Medicine, University College Dublin, Belfield, Dublin 4, Irelandb Institute for Public Health, Washington University in St. Louis, St. Louis, MO, United Statesc Department of Quality, Safety and Practice Excellence, St. Louis Children’s Hospital, St. Louis, MO, United Statesd Department of Surgery, Washington University School of Medicine, St. Louis, MO, United Statese Program in Occupational Therapy, Washington University School of Medicine, St. Louis, MO, United Statesf Chan Division of Occupational Science and Occupational Therapy, University of Southern California, Los Angeles, CA, United Statesg Department of Pediatrics, Keck School of Medicine, Los Angeles, CA, United Statesh Gehr Family Center for Health Systems Science and Innovation, University of Southern California, Los Angeles, CA, United Statesi Center for the Changing Family, University of Southern California, Los Angeles, CA, United States

AbstractBackground: Early parent engagement in the neonatal intensive care unit (NICU) is important for both parent and infant mental health and for improving developmental outcomes. It remains unclear how different programs, such as the Supporting and Enhancing NICU Sensory Experiences (SENSE) program, may empower parents from various socio-demographic groups to engage in the NICU. An improved understanding could aid in individualizing interventions for those at the highest risk for health disparities. Aims: This exploratory study, which was part of a larger study, sought to explore 1) socio-demographic factors related to parent presence and engagement in the NICU and 2) if the SENSE program related to increased parent presence and engagement among different socio-demographic groups. Methods: Seventy parent-infant dyads (born ≤ 32 weeks gestation) were randomized to SENSE programming (parent education and age-appropriate, positive sensory interventions for parents to conduct with their infants every day of hospitalization) or standard care after admission to the NICU. The amount of parent presence and participation in sensory activities was tracked using bedside logs, nursing records, and research team documentation. Results: Being married (p = 0.048; p = 0.01), having private insurance (p < 0.001; p = 0.01), and having fewer children (p = 0.004; p = 0.03) related to more parent presence and engagement respectively. Parents who were Black had less presence and engagement in the NICU (p = 0.04; p = 0.02). Participation in the SENSE program was related to more parent presence and engagement among younger mothers (p = 0.002; p ≤0.001) and among parents living farther distances from the hospital (p < 0.001; p = 0.004). Conclusion: Programming, such as the SENSE program, can improve parent engagement in the NICU among high-risk groups. © 2021 Elsevier B.V.

Author KeywordsDevelopment;  Health disparities;  Neonatal intensive care unit;  Parent engagement;  Sensory interventions;  Socio-demographic

Funding detailsNational Institutes of HealthNIHNational Institute of Child Health and Human DevelopmentNICHDP30 HD062171Gordon and Betty Moore FoundationGBMFInstitute of Clinical and Translational SciencesICTSUL1TR002345Institute for Public Health, Washington University in St. Louis

Document Type: ArticlePublication Stage: FinalSource: Scopus

“Reward deficiency syndrome (Rds): A cytoarchitectural common neurobiological trait of all addictions” (2021) International Journal of Environmental Research and Public Health

Reward deficiency syndrome (Rds): A cytoarchitectural common neurobiological trait of all addictions(2021) International Journal of Environmental Research and Public Health, 18 (21), art. no. 11529, . 

Blum, K.a b c d e f g , Bowirrat, A.h , Braverman, E.R.g , Baron, D.a , Cadet, J.L.i , Kazmi, S.a , Elman, I.j , Thanos, P.K.k , Badgaiyan, R.D.l m , Downs, W.B.g , Bagchi, D.g n , Llanos-Gomez, L.g , Gold, M.S.o p

a Division of Addiction Research & Education, Center for Psychiatry, Medicine, & Primary Care (Office of the Provost), Western University Health Sciences, Pomona, CA 91766, United Statesb Institute of Psychology, Eotvos Loránd University, Budapest, 1053, Hungaryc Division of Nutrigenomics, Synaptamine Inc., Austin, TX 78701, United Statesd Department of Psychiatry, Wright State University Boonshoft School of Medicine and Dayton VA Medical Center, Dayton, OH 45435, United Statese Department of Psychiatry, University of Vermont, Burlington, VA 05405, United Statesf Division of Precision Addiction Management, Geneus Health (Division of Ivitalize Inc.), San Antonio, TX 78249, United Statesg Division of Nutrigenomics, The Kenneth Blum Behavioral & Neurogenetic Institute (Division of Ivitalize, Inc.), Austin, TX 78701, United Statesh Department of Molecular Biology, Adelson School of Medicine, Ariel University, Ariel, 40700, Israeli Molecular Neuropsychiatry Research Branch, NIH National Institute on Drug Abuse, Bethesda, MD 20892, United Statesj Department of Psychiatry, Harvard School of Medicine, Cambridge, MA 02115, United Statesk Behavioral Neuropharmacology and Neuroimaging Laboratory on Addictions, Clinical Research Institute on Addictions, Department of Pharmacology and Toxicology, Jacobs School of Medicine and Biosciences, State University of New York at Buffalo, Buffalo, NY 14260, United Statesl Department of Psychiatry, South Texas Veteran Health Care System, Audie L. Murphy Memorial VA Hospital, San Antonio, TX 78229, United Statesm Long School of Medicine, University of Texas Medical Center, San Antonio, TX 78229, United Statesn Department of Pharmaceutical Sciences, College of Pharmacy, Southern University, Houston, TX 77004, United Stateso Department of Psychiatry, Tulane University School of Medicine, New Orleans, LA 70118, United Statesp Department of Psychiatry, Washington University School of Medicine, St. Louis, MO 63130, United States

AbstractAlcohol and other substance use disorders share comorbidity with other RDS disorders, i.e. a reduction in dopamine signaling within the reward pathway. RDS is a term that connects addictive, obsessive, compulsive, and impulsive behavioral disorders. An estimated 2 million individuals in the United States have opioid use disorder related to prescription opioids. It is estimated that the overall cost of the illegal and legally prescribed opioid crisis exceeds one trillion dollars. Opioid Replacement Therapy is the most common treatment for addictions and other RDS disorders. Even after repeated relapses, patients are repeatedly prescribed the same opioid replacement treatments. A recent JAMA report indicates that non-opioid treatments fare better than chronic opioid treatments. Research demonstrates that over 50 percent of all suicides are related to alcohol or other drug use. In addition to effective fellowship programs and spirituality acceptance, nutrigenomic therapies (e.g., KB220Z) optimize gene expression, rebalance neurotransmitters, and restore neurotransmitter functional connectivity. KB220Z was shown to increase functional connectivity across specific brain regions involved in dopaminergic function. KB220/Z significantly reduces RDS behavioral disorders and relapse in human DUI offenders. Taking a Genetic Addiction Risk Severity (GARS) test combined with a the KB220Z semi-customized nutrigenomic supplement effectively restores dopamine homeostasis. (WC 199). © 2021 by the authors. Licensee MDPI, Basel, Switzerland.

Author KeywordsDopamine homeostasis;  GARS;  KB220;  Neuroimaging;  Precision addiction management;  Reward deficiency syndrome (RDS)

Document Type: ArticlePublication Stage: FinalSource: Scopus

“Physical Activity Patterns and Relationships With Cognitive Function in Patients With Breast Cancer Before, During, and After Chemotherapy in a Prospective, Nationwide Study” (2021) Journal of Clinical Oncology: Official Journal of the American Society of Clinical Oncology

Physical Activity Patterns and Relationships With Cognitive Function in Patients With Breast Cancer Before, During, and After Chemotherapy in a Prospective, Nationwide Study(2021) Journal of Clinical Oncology: Official Journal of the American Society of Clinical Oncology, 39 (29), pp. 3283-3292. 

Salerno, E.A.a b , Culakova, E.c , Kleckner, A.S.c d , Heckler, C.E.c d , Lin, P.-J.c d , Matthews, C.E.b , Conlin, A.e , Weiselberg, L.f , Mitchell, J.g , Mustian, K.M.c d , Janelsins, M.C.c d

a Division of Public Health Sciences, Department of Surgery, Washington University School of Medicine in St Louis, St Louis, MOb Division of Cancer Epidemiology and Genetics, Metabolic Epidemiology Branch, National Cancer Institute, MD, Rockville, United Statesc Department of Surgery, University of Rochester Medical Center, Rochester, NYd Wilmot Cancer Institute, University of Rochester Medical Center, Rochester, NYe Pacific Cancer Research Consortium-National Cancer Institute Community Clinical Oncology Research Program (NCORP), Seattle, WAf North Shore LIJ Health System NCORP ,Lake Success, NYg Columbus NCORP, Columbus, OH

AbstractPURPOSE: Physical activity (PA) is a promising intervention for cancer-related cognitive decline, yet research assessing its use during chemotherapy is limited. This study evaluated patterns of PA before, during, and after chemotherapy in patients with breast cancer and the association between PA and cognitive function. METHODS: In a nationwide, prospective cohort study, we assessed PA (Aerobics Center Longitudinal Study PA measure) and perceived and objectively measured cognitive functioning (Functional Assessment of Cancer Therapy-Cognitive, Delayed Match to Sample, and Rapid Visual Processing measures) at prechemotherapy (T1), postchemotherapy (T2), and 6 months postchemotherapy (T3) in patients with breast cancer and cancer-free, age-matched controls at equivalent time points. Longitudinal linear mixed-effects models (LMMs) characterized PA changes over time between patients and controls, adjusting for demographic and clinical factors. LMMs further estimated the role of prechemotherapy PA and changes in PA during chemotherapy on cognitive changes over time. RESULTS: Patients with stage I-IIIC breast cancer (n = 580; age M [standard deviation] = 53.4 [10.6] years) and controls (n = 363; age M [standard deviation] = 52.6 [10.3] years) were included. One third of patients met national PA guidelines at T1, dropping to 21% at T2 before rising to 37% at T3. LMMs revealed declines in PA from T1 to T2 in patients compared with controls (all P < .001). Patients meeting guidelines at T1 demonstrated better cognitive scores over time on the Functional Assessment of Cancer Therapy-Cognitive and Rapid Visual Processing (all P < .05), with similar patterns of objectively-measured cognitive function as controls. In patients, greater moderate-to-vigorous PA at the previous time point was significantly associated with better cognitive trajectories (all P < .05), and adherence to PA guidelines throughout chemotherapy was associated with better self-reported cognition (P < .01). CONCLUSION: This nationwide study demonstrates that PA maintenance before and during chemotherapy is associated with better cognitive function immediately and 6 months after chemotherapy completion.

Document Type: ArticlePublication Stage: FinalSource: Scopus

“Genome-wide association study and functional validation implicates JADE1 in tauopathy” (2021) Acta Neuropathologica

Genome-wide association study and functional validation implicates JADE1 in tauopathy(2021) Acta Neuropathologica, . 

Farrell, K.a b c , Kim, S.H.a b c , Han, N.a b c , Iida, M.A.a b c , Gonzalez, E.M.d , Otero-Garcia, M.e , Walker, J.M.f , Richardson, T.E.f , Renton, A.E.b g , Andrews, S.J.b g , Fulton-Howard, B.b g , Humphrey, J.b g , Vialle, R.A.b g , Bowles, K.R.g , de Paiva Lopes, K.b g , Whitney, K.a b c , Dangoor, D.K.a b c , Walsh, H.a b c , Marcora, E.b g , Hefti, M.M.h , Casella, A.a b c , Sissoko, C.T.a b c , Kapoor, M.b g , Novikova, G.b g , Udine, E.b g , Wong, G.b g , Tang, W.ae , Bhangale, T.i , Hunkapiller, J.i , Ayalon, G.j , Graham, R.R.k , Cherry, J.D.l , Cortes, E.P.a b , Borukov, V.Y.a b , McKee, A.C.l , Stein, T.D.l , Vonsattel, J.-P.m , Teich, A.F.m , Gearing, M.n , Glass, J.n , Troncoso, J.C.o , Frosch, M.P.p , Hyman, B.T.p , Dickson, D.W.q , Murray, M.E.q , Attems, J.r , Flanagan, M.E.s , Mao, Q.s , Mesulam, M.-M.s , Weintraub, S.s , Woltjer, R.L.t , Pham, T.t , Kofler, J.u , Schneider, J.A.v , Yu, L.v , Purohit, D.P.a w , Haroutunian, V.b w , Hof, P.R.b , Gandy, S.w am , Sano, M.w , Beach, T.G.x , Poon, W.y , Kawas, C.H.an , Corrada, M.M.y , Rissman, R.A.z , Metcalf, J.z , Shuldberg, S.z , Salehi, B.z , Nelson, P.T.aa , Trojanowski, J.Q.ab , Lee, E.B.ab , Wolk, D.A.ac , McMillan, C.T.ac , Keene, C.D.ad , Latimer, C.S.ad , Montine, T.J.ad ae , Kovacs, G.G.af ag ah , Lutz, M.I.ah , Fischer, P.ao , Perrin, R.J.ai , Cairns, N.J.al , Franklin, E.E.ai , Cohen, H.T.aj , Raj, T.b g , Cobos, I.ae , Frost, B.d , Goate, A.b g , White III, C.L.ak , Crary, J.F.a b c

a Department of Pathology, Neuropathology Brain Bank and Research CoRE, Icahn School of Medicine at Mount Sinai, 1 Gustave L. Levy Place Box 1194, New York, NY 10029, United Statesb Nash Department of Neuroscience, Ronald M. Loeb Center for Alzheimer’s Disease, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, United Statesc Department of Artificial Intelligence and Human Health, Icahn School of Medicine at Mount Sinai, New York, NY, United Statesd Department of Cell Systems and Anatomy, Glenn Biggs Institute for Alzheimer’s and Neurodegenerative Diseases, the Sam and Ann Barshop Institute for Longevity and Aging Studies, University of Texas Health San Antonio, San Antonio, TX 78229, United Statese Department of Pathology and Laboratory Medicine, Division of Neuropathology, University of California, Los Angeles, CA, United Statesf Department of Pathology and Glenn Biggs Institute for Alzheimer’s and Neurodegenerative Diseases, UT Health San Antonio, San Antonio, TX, United Statesg Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, United Statesh Department of Pathology, University of Iowa, Iowa City, IA, United Statesi Department of Human Genetics, Genentech, South San Francisco, CA, United Statesj Neumora Therapeutics, South San Francisco, CA, United Statesk Maze Therapeutics, San Francisco, CA, United Statesl Department of Pathology (Neuropathology), VA Medical Center, Boston University School of Medicine, Boston, MA, United Statesm Department of Pathology and Cell Biology, Department of Neurology, and the Taub Institute for Research on Alzheimer’s Disease and the Aging Brain, Columbia University Medical Center, New York, NY, United Statesn Department of Pathology and Laboratory Medicine (Neuropathology) and Neurology, Emory University School of Medicine, Atlanta, GA, United Stateso Department of Pathology, Division of Neuropathology, Johns Hopkins University School of Medicine, Baltimore, MD, United Statesp Department of Neurology and Pathology, Harvard Medical School and Massachusetts General Hospital, Charlestown, MA, United Statesq Department of Neuroscience, Mayo Clinic, Jacksonville, FL, United Statesr Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne, United Kingdoms Department of Pathology (Neuropathology), Northwestern Cognitive Neurology and Alzheimer Disease Center, Northwestern University Feinberg School of Medicine, Chicago, IL, United Statest Department of Pathology, Oregon Health Sciences University, Portland, OR, United Statesu Department of Pathology (Neuropathology), University of Pittsburgh Medical Center, Pittsburgh, PA, United Statesv Departments of Pathology (Neuropathology) and Neurological Sciences, Rush University Medical Center, Chicago, IL, United Statesw Department of Psychiatry, Alzheimer’s Disease Research Center, James J. Peters VA Medical Center, Icahn School of Medicine at Mount Sinai, New York, NY, United Statesx Department of Neuropathology, Banner Sun Health Research Institute, Sun City, AZ, United Statesy Department of Neurology, Department of Epidemiology, Institute for Memory Impairments and Neurological Disorders, UC Irvine, Irvine, CA, United Statesz Department of Neurosciences University of California and the Veterans Affairs San Diego Healthcare System, La Jolla, San Diego, CA, United Statesaa Department of Pathology (Neuropathology) and Sanders-Brown Center on Aging, University of Kentucky, Lexington, KY, United Statesab Center for Neurodegenerative Disease Research, Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United Statesac Department of Neurology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United Statesad Department of Laboratory Medicine and Pathology, University of f Medicine, Seattle, WA, United Statesae Department of Pathology, Stanford University, Palo Alto, United Statesaf Laboratory Medicine Program, Krembil Brain Institute, University Health Network, Toronto, ON, Canadaag Tanz Centre for Research in Neurodegenerative Disease and Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canadaah Institute of Neurology, Medical University of Vienna, Vienna, Austriaai Department of Pathology and Immunology, Department of Neurology, Knight Alzheimer Disease Research Center, Washington University School of Medicine, St. Louis, MO, United Statesaj Departments of Medicine, Pathology, and Pharmacology, Boston University School of Medicine and Boston Medical Center, Boston, MA, United Statesak Department of Pathology (Neuropathology), University of Texas Southwestern Medical School, Dallas, TX, United Statesal College of Medicine and Health, University of Exeter, Exeter, United Kingdomam Department of Neurology, Center for Cognitive Health, Icahn School of Medicine at Mount Sinai, New York, NY, United Statesan Department of Neurology, Department of Neurobiology and Behavior, Institute for Memory Impairments and Neurological Disorders, UC Irvine, Irvine, CA, United Statesao Department of Psychiatry, Danube Hospital, Vienna, Austria

AbstractPrimary age-related tauopathy (PART) is a neurodegenerative pathology with features distinct from but also overlapping with Alzheimer disease (AD). While both exhibit Alzheimer-type temporal lobe neurofibrillary degeneration alongside amnestic cognitive impairment, PART develops independently of amyloid-β (Aβ) plaques. The pathogenesis of PART is not known, but evidence suggests an association with genes that promote tau pathology and others that protect from Aβ toxicity. Here, we performed a genetic association study in an autopsy cohort of individuals with PART (n = 647) using Braak neurofibrillary tangle stage as a quantitative trait. We found some significant associations with candidate loci associated with AD (SLC24A4, MS4A6A, HS3ST1) and progressive supranuclear palsy (MAPT and EIF2AK3). Genome-wide association analysis revealed a novel significant association with a single nucleotide polymorphism on chromosome 4 (rs56405341) in a locus containing three genes, including JADE1 which was significantly upregulated in tangle-bearing neurons by single-soma RNA-seq. Immunohistochemical studies using antisera targeting JADE1 protein revealed localization within tau aggregates in autopsy brains with four microtubule-binding domain repeats (4R) isoforms and mixed 3R/4R, but not with 3R exclusively. Co-immunoprecipitation in post-mortem human PART brain tissue revealed a specific binding of JADE1 protein to four repeat tau lacking N-terminal inserts (0N4R). Finally, knockdown of the Drosophila JADE1 homolog rhinoceros (rno) enhanced tau-induced toxicity and apoptosis in vivo in a humanized 0N4R mutant tau knock-in model, as quantified by rough eye phenotype and terminal deoxynucleotidyl transferase dUTP nick end-labeling (TUNEL) in the fly brain. Together, these findings indicate that PART has a genetic architecture that partially overlaps with AD and other tauopathies and suggests a novel role for JADE1 as a modifier of neurofibrillary degeneration. © 2021, The Author(s), under exclusive licence to Springer-Verlag GmbH Germany, part of Springer Nature.

Document Type: ArticlePublication Stage: Article in PressSource: Scopus

“HSP90 inhibitors reduce cholesterol storage in Niemann-Pick type C1 mutant fibroblasts” (2021) Journal of Lipid Research

HSP90 inhibitors reduce cholesterol storage in Niemann-Pick type C1 mutant fibroblasts(2021) Journal of Lipid Research, 63, art. no. 100114, . 

Pipalia, N.H.a , Saad, S.Z.a , Subramanian, K.b , Cross, A.c , al-Motawa, A.a , Garg, K.a , Blagg, B.S.J.d , Neckers, L.e , Helquist, P.d , Wiest, O.d , Ory, D.S.b , Maxfield, F.R.a

a Department of Biochemistry, Weill Cornell Medical College, New York, NY, United Statesb Department of Internal Medicine, Washington University in St. Louis, St. Louis, MO, United Statesc Natural Sciences Department, Fordham University, New York, NY, United Statesd Department of Chemistry and Biochemistry, University of Notre Dame, South Bend, IN, United Statese Urologic Oncology Branch, National Cancer Institute, Bethesda, MD, United States

AbstractNiemann-Pick type C1 (NPC1) disease is a lysosomal lipid storage disorder caused by mutations of the NPC1 gene. More than 300 disease-associated mutations are reported in patients, resulting in abnormal accumulation of unesterified cholesterol, glycosphingolipids, and other lipids in late endosomes and lysosomes (LE/Ly) of many cell types. Previously, we showed that treatment of many different NPC1 mutant fibroblasts with histone deacetylase inhibitors resulted in reduction of cholesterol storage, and we found that this was associated with enhanced exit of the NPC1 protein from the endoplasmic reticulum and delivery to LE/Ly. This suggested that histone deacetylase inhibitors may work through changes in protein chaperones to enhance the folding of NPC1 mutants, allowing them to be delivered to LE/Ly. In this study, we evaluated the effect of several HSP90 inhibitors on NPC1I1061T skin fibroblasts. We found that HSP90 inhibition resulted in clearance of cholesterol from LE/Ly, and this was associated with enhanced delivery of the mutant NPC1I1061T protein to LE/Ly. We also observed that inhibition of HSP90 increased the expression of HSP70, and overexpression of HSP70 also reduced cholesterol storage in NPC1I1061T fibroblasts. However, we did not see correction of cholesterol storage by arimoclomol, a drug that is reported to increase HSP70 expression, at doses up to 0.5 mM. The increase in other chaperones as a consequence of HSP90 improves folding of NPC1 protein and relieves cholesterol accumulation in NPC1 mutant fibroblasts. © 2021 THE AUTHORS. Published by Elsevier Inc on behalf of American Society for Biochemistry and Molecular Biology.

Author KeywordsArimoclomol;  Chaperone;  Cholesterol;  Drug therapy;  Endocytosis;  Fluorescence microscopy;  HSP70;  HSP90;  Lysosomal storage;  Niemann-Pick disease

Document Type: ArticlePublication Stage: FinalSource: Scopus

“Glycine-Rich Peptides from FUS Have an Intrinsic Ability to Self-Assemble into Fibers and Networked Fibrils” (2021) Biochemistry

Glycine-Rich Peptides from FUS Have an Intrinsic Ability to Self-Assemble into Fibers and Networked Fibrils(2021) Biochemistry, . 

Kar, M.a , Posey, A.E.b , Dar, F.c , Hyman, A.A.a , Pappu, R.V.b

a Max Planck Institute of Cell Biology and Genetics (MPI-CBG), Dresden, 01307, Germanyb Dept. of Biomedical Engineering and Center for Science and Engineering of Living Systems (CSELS), Washington University in St. Louis, St. Louis, MO 63130, United Statesc Department of Physics, Washington University in St. Louis, St. Louis, MO 63130, United States

AbstractGlycine-rich regions feature prominently in intrinsically disordered regions (IDRs) of proteins that drive phase separation and the regulated formation of membraneless biomolecular condensates. Interestingly, the Gly-rich IDRs seldom feature poly-Gly tracts. The protein fused in sarcoma (FUS) is an exception. This protein includes two 10-residue poly-Gly tracts within the prion-like domain (PLD) and at the interface between the PLD and the RNA binding domain. Poly-Gly tracts are known to be highly insoluble, being potent drivers of self-assembly into solid-like fibrils. Given that the internal concentrations of FUS and FUS-like molecules cross the high micromolar and even millimolar range within condensates, we reasoned that the intrinsic insolubility of poly-Gly tracts might be germane to emergent fluid-to-solid transitions within condensates. To assess this possibility, we characterized the concentration-dependent self-assembly for three non-overlapping 25-residue Gly-rich peptides derived from FUS. Two of the three peptides feature 10-residue poly-Gly tracts. These peptides form either long fibrils based on twisted ribbon-like structures or self-supporting gels based on physical cross-links of fibrils. Conversely, the peptide with similar Gly contents but lacking a poly-Gly tract does not form fibrils or gels. Instead, it remains soluble across a wide range of concentrations. Our findings highlight the ability of poly-Gly tracts within IDRs that drive phase separation to undergo self-assembly. We propose that these tracts are likely to contribute to nucleation of fibrillar solids within dense condensates formed by FUS. © 2021 American Chemical Society.

Funding detailsNational Institutes of HealthNIH5R01056114Air Force Office of Scientific ResearchAFOSRFA9550-20-1-0241Human Frontier Science ProgramHFSPRGP0034/2017Max-Planck-GesellschaftMPGNOMIS Stiftung

Document Type: ArticlePublication Stage: Article in PressSource: Scopus

“ITSN1: a novel candidate gene involved in autosomal dominant neurodevelopmental disorder spectrum” (2021) European Journal of Human Genetics

ITSN1: a novel candidate gene involved in autosomal dominant neurodevelopmental disorder spectrum(2021) European Journal of Human Genetics, . 

Bruel, A.-L.a b , Vitobello, A.a b , Thiffault, I.c , Manwaring, L.d , Willing, M.d , Agrawal, P.B.e f , Bayat, A.g , Kitzler, T.M.h i j , Brownstein, C.A.e , Genetti, C.A.e , Gonzalez-Heydrich, J.k , Jayakar, P.l , Zyskind, J.W.m , Zhu, Z.m , Vachet, C.n , Wilson, G.R.o , Pruniski, B.o , Goyette, A.-M.p , Duffourd, Y.a b , Thauvin-Robinet, C.a b q , Philippe, C.a b q , Faivre, L.a b q

a UMR1231 GAD, Inserm – Université Bourgogne-Franche Comté, Dijon, Franceb Unité Fonctionnelle Innovation en Diagnostic génomique des maladies rares, FHU-TRANSLAD, CHU Dijon Bourgogne, Dijon, Francec Center for Pediatric Genomic Medicine, Children’s Mercy Hospital, Kansas City, MO, United Statesd Department of Pediatrics, Division of Genetics and Genomic Medicine, Washington University School of Medicine, St. Louis, MO, United Statese Divisions of Newborn Medicine, Genetics and Genomics, The Manton Center for Orphan Disease Research, Boston Children’s Hospital, Boston, MA, United Statesf Department of Pediatrics, Harvard Medical School, Boston, MA, United Statesg Department of Genetics and Precision Medicine, Danish Epilepsy Centre, Dianalund, Denmarkh Research Institute, McGill University Health Centre, Montreal, QC, Canadai Division of Medical Genetics, Department of Medicine, McGill University Health Centre, Montreal, QC, Canadaj Department of Human Genetics, McGill University, Montreal, QC, Canadak Department of Psychiatry, Boston Children’s Hospital, Manton Center for Orphan Disease Research, Division of Genetics and Genomics, Boston Children’s Hospital, Department of Pediatrics, Harvard Medical School, Boston, MA, United Statesl Division of Genetics and Metabolism, Nicklaus Children’s Hospital, Miami, FL, United Statesm GeneDX, Gaitherburg, MD, United Statesn Service de néphrologie pédiatrique, Centre Hospitalier Régional Universitaire Besançon, Besançon, Franceo Division of Genetics and Metabolism, Phoenix Children’s Medical Group, Phoenix, AZ, United Statesp FRCPC, Developmental Pediatrician, Montreal Children’s Hospital, McGill University Health Center, Montreal, QC, Canadaq Fédération Hospitalo-Universitaire Médecine Translationnelle et Anomalies du Développement (TRANSLAD), Centre Hospitalier Universitaire Dijon, Dijon, France

AbstractITSN1 plays an important role in brain development. Recent studies in large cohorts of subjects with neurodevelopmental disorders have identified de novo variants in ITSN1 gene thereby suggesting that this gene is involved in the development of such disorders. The aim of this study is to provide further proof of such a link. We performed trio exome sequencing in a patient presenting autism, intellectual disability, and severe behavioral difficulties. Additional affected patients with a neurodevelopmental disorder harboring a heterozygous variant in ITSN1 (NM_003024.2) were collected through a worldwide collaboration. All patients underwent detailed phenotypic and genetic assessment and data was collected and shared by healthcare givers. We identified ten novel patients from eight families with heterozygous truncating or missense variants in ITSN1 gene. In addition, four previously published patients from large meta-analysis studies were included. In total, 7/14 patients presented a de novo variant in ITSN1. All patients showed neurodevelopmental disorders from autism spectrum disorders (90%), intellectual disability (86%), and epilepsy (30%). We demonstrated that truncating variants are in the first half of ITSN1 whereas missense variants are clustered in C-terminal region. We suggest ITSN1 gene is involved in development of an autism spectrum disorder with variable additional neurodevelopmental deficiency, thus confirming the hypothesis that ITSN1 is important for brain development. © 2021, The Author(s), under exclusive licence to European Society of Human Genetics.

Funding detailsEuropean CommissionECAgence Nationale de la RechercheANRMinistère des Affaires Sociales et de la SantéEuropean Regional Development FundERDFConseil régional de Bourgogne-Franche-Comté

Document Type: ArticlePublication Stage: Article in PressSource: Scopus

“Mental health in the UK Biobank: A roadmap to self-report measures and neuroimaging correlates” (2021) Human Brain Mapping

Mental health in the UK Biobank: A roadmap to self-report measures and neuroimaging correlates(2021) Human Brain Mapping, . 

Dutt, R.K., Hannon, K., Easley, T.O., Griffis, J.C., Zhang, W., Bijsterbosch, J.D.

Department of Radiology, Washington University School of Medicine, Saint Louis, MO, United States

AbstractThe UK Biobank (UKB) is a highly promising dataset for brain biomarker research into population mental health due to its unprecedented sample size and extensive phenotypic, imaging, and biological measurements. In this study, we aimed to provide a shared foundation for UKB neuroimaging research into mental health with a focus on anxiety and depression. We compared UKB self-report measures and revealed important timing effects between scan acquisition and separate online acquisition of some mental health measures. To overcome these timing effects, we introduced and validated the Recent Depressive Symptoms (RDS-4) score which we recommend for state-dependent and longitudinal research in the UKB. We furthermore tested univariate and multivariate associations between brain imaging-derived phenotypes (IDPs) and mental health. Our results showed a significant multivariate relationship between IDPs and mental health, which was replicable. Conversely, effect sizes for individual IDPs were small. Test–retest reliability of IDPs was stronger for measures of brain structure than for measures of brain function. Taken together, these results provide benchmarks and guidelines for future UKB research into brain biomarkers of mental health. © 2021 The Authors. Human Brain Mapping published by Wiley Periodicals LLC.

Author Keywordsbrain correlates;  depression;  mental health;  replication;  test–retest;  UK Biobank

Funding detailsNational Institutes of HealthNIH1 R34 NS11861801McDonnell Center for Systems Neuroscience

Document Type: ArticlePublication Stage: Article in PressSource: Scopus

“Detection of VOR dysfunction during the gaze stabilization test: Does target size matter?” (2021) Journal of Vestibular Research: Equilibrium and Orientation

Detection of VOR dysfunction during the gaze stabilization test: Does target size matter?(2021) Journal of Vestibular Research: Equilibrium and Orientation, 31 (6), pp. 495-504. 

Thompson-Harvey, A.a , Dutcher, C.E.b , Monroe, H.A.c , Sinks, B.C.d , Goebel, J.A.d

a Department of Otolaryngology and Communication Sciences, Medical College of Wisconsin, Milwaukee, WI, United Statesb Starkey Hearing Technologies, Eden Prairie, MN, United Statesc St. Louis Children’s Hospital, St. Louis, MO, United Statesd Dizziness and Balance Center, Department of Otolaryngology – Head and Neck Surgery, Washington University, School of Medicine, Saint Louis, MO, United States

AbstractBACKGROUND: The Gaze Stabilization Test (GST) identifies vestibulo-ocular reflex (VOR) dysfunction using a decline in target recognition with increasing head velocity, but there is no consensus on target (optotype) size above static visual acuity. OBJECTIVE: To determine the optimal optotype size above static visual acuity to be used during the GST in subjects with unilateral vestibular dysfunction and healthy individuals. METHODS: Eight subjects with unilateral vestibular dysfunction (UVD) and 19 age-matched, healthy control subjects were studied with the standard GST protocol using two optotype sizes, 0.2 and 0.3 logMAR above static visual acuity (ΔlogMAR). Maximal head velocity achieved while maintaining fixation on both optotypes was measured. Sensitivity, specificity and receiver-operator characteristic area under the curve (ROC AUC) analyses were performed to determine the optimal head velocity cut off point for each optotype, based on ability to identify the lesioned side of the UVD group from the control group. RESULTS: There was a significant difference in maximal head velocity between the UVD group and control group using 0.2 ΔlogMAR (p = 0.032) but not 0.3 ΔlogMAR (p = 0.061). While both targets produced similar specificities (90%) for distinguishing normal from subjects with UVD, 0.2 ΔlogMAR targets yielded higher sensitivity (75%) than 0.3 logMAR (63%) and accuracy (86% vs 80%, respectively) in detecting the lesioned side in subjects with UVD versus controls with maximal head velocities≤105 deg/s (p = 0.017). Furthermore, positive likelihood ratios were nearly twice as high when using 0.2 ΔlogMAR targets (+ LR 10) compared to 0.3 ΔlogMAR (+ LR 6.3). CONCLUSION: The 0.2 ΔlogMAR optotype demonstrated significantly superior identification of subjects with UVD, better sensitivity and positive likelihood ratios than 0.3 ΔlogMAR for detection of VOR dysfunction. Using a target size 0.2logMAR above static visual acuity (ΔlogMAR) during GST may yield better detection of VOR dysfunction to serve as a baseline for gaze stabilization rehabilitation therapy. © 2021 – IOS Press. All rights reserved.

Author KeywordsGaze stabilization test;  vestibular rehabilitation;  vestibular testing;  vestibulo-ocular reflex (VOR);  visual acuity

Document Type: ArticlePublication Stage: FinalSource: Scopus

“Association of bleeding with serotonergic antidepressants in patients receiving left ventricular assist device support” (2021) Pharmacotherapy

Association of bleeding with serotonergic antidepressants in patients receiving left ventricular assist device support(2021) Pharmacotherapy, . 

Robinson, A.C.a , January, S.E.a , Botkin, K.W.a , Vader, J.M.b , Hartupee, J.C.b , Tellor Pennington, B.R.a

a Department of Pharmacy, Barnes-Jewish Hospital, St. Louis, MO, United Statesb Division of Cardiovascular Diseases, Washington University School of Medicine in Saint Louis, St. Louis, MO, United States

AbstractStudy Objective: This study sought to determine whether SA use is associated with bleeding in patients receiving CF-LVAD support. Design: A retrospective cohort analysis was conducted of all adult patients who received CF-LVAD implantation at our institution. Setting: Barnes-Jewish Hospital between July 1, 2009, and October 1, 2018. Patients: Patients at least 18 years of age who received a HVAD™ (HeartWare Corp.), HeartMate II™ (St. Jude Medical), or HeartMate 3™ (St. Jude Medical) CF-LVAD and survived for at least 30 days postoperatively were included. Intervention: Patients who received SAs (n = 203) were compared to those who did not (n = 391) from 30 days to 18 months following implantation. The primary outcome was the incidence of first bleeding events including gastrointestinal bleed (GIB), epistaxis, or intracerebral hemorrhage (ICH). Measurements and Main Results: During follow-up, 219 patients had bleeding events: 93 of 203 (45.8%) in the SA group versus 126 of 391 (32.2%) in the control group (p = 0.001). After adjustment for age, angiotensin-converting enzyme (ACE) inhibitor and angiotensin receptor blocker (ARB) use, history of bleeding events, history of smoking, and CF-LVAD type, SA use remained associated with bleeding (adjusted odds ratio: 1.75, 95% confidence interval: 1.22–2.51, p = 0.002). HeartMate 3™ patients experienced less bleeding than HeartMate II™ patients (adjusted odds ratio 0.46, 95% confidence interval: 0.23–0.90, p = 0.024). Conclusions: In this single-center, retrospective cohort of patients supported with CF-LVADs, SA use was associated with the incidence of first bleeding events, primarily driven by GIB. Further studies are needed to assess any differential risk of bleeding among SA agents and to assess the utility of altering antithrombotic strategies. © 2021 Pharmacotherapy Publications, Inc.

Author Keywordsantidepressant;  assist;  bleeding;  device;  gastrointestinal;  serotonergic;  ventricular

Document Type: ArticlePublication Stage: Article in PressSource: Scopus