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

WashU weekly Neuroscience publications

“Genomic prediction of alcohol-related morbidity and mortality” (2020) Translational Psychiatry

Genomic prediction of alcohol-related morbidity and mortality
(2020) Translational Psychiatry, 10 (1), art. no. 23, . 

Kiiskinen, T.a f , Mars, N.J.f , Palviainen, T.f , Koskela, J.f , Rämö, J.T.f , Ripatti, P.f , Ruotsalainen, S.f , Palotie, A.f g h , Madden, P.A.F.b , Rose, R.J.c , Kaprio, J.d f , Salomaa, V.a , Mäkelä, P.a , Havulinna, A.S.a f , Ripatti, S.d e f , FinnGen, GSCAN Consortiumf g h

a Finnish Institute for Health and Welfare (THL), Helsinki, Finland
b Department of Psychiatry, Washington University School of Medicine in St.Louis, St.Louis, MO, United States
c Department of Psychological and Brain Sciences, Indiana University Bloomington, Bloomington, IN, United States
d Department of Public Health, Clinicum, Faculty of Medicine, University of Helsinki, Helsinki, Finland
e The Broad Institute of MIT and Harvard, Cambridge, MA, United States
f Institute for Molecular Medicine Finland (FIMM), HiLIFE, University of Helsinki, Helsinki, Finland
g Analytic and Translational Genetics Unit, Department of Medicine, Department of Neurology and Department of Psychiatry Massachusetts General Hospital, Boston, MA, United States
h The Stanley Center for Psychiatric Research and Program in Medical and Population Genetics, The Broad Institute of MIT and Harvard, Cambridge, Boston, MA, United States

Abstract
While polygenic risk scores (PRS) have been shown to predict many diseases and risk factors, the potential of genomic prediction in harm caused by alcohol use has not yet been extensively studied. Here, we built a novel polygenic risk score of 1.1 million variants for alcohol consumption and studied its predictive capacity in 96,499 participants from the FinnGen study and 39,695 participants from prospective cohorts with detailed baseline data and up to 25 years of follow-up time. A 1 SD increase in the PRS was associated with 11.2 g (=0.93 drinks) higher weekly alcohol consumption (CI = 9.85–12.58 g, p = 2.3 × 10–58). The PRS was associated with alcohol-related morbidity (4785 incident events) and the risk estimate between the highest and lowest quintiles of the PRS was 1.83 (95% CI = 1.66–2.01, p = 1.6 × 10–36). When adjusted for self-reported alcohol consumption, education, marital status, and gamma-glutamyl transferase blood levels in 28,639 participants with comprehensive baseline data from prospective cohorts, the risk estimate between the highest and lowest quintiles of the PRS was 1.58 (CI = 1.26–1.99, p = 8.2 × 10–5). The PRS was also associated with all-cause mortality with a risk estimate of 1.33 between the highest and lowest quintiles (CI = 1.20–1.47, p = 4.5 × 10–8) in the adjusted model. In conclusion, the PRS for alcohol consumption independently associates for both alcohol-related morbidity and all-cause mortality. Together, these findings underline the importance of heritable factors in alcohol-related health burden while highlighting how measured genetic risk for an important behavioral risk factor can be used to predict related health outcomes. © 2019, The Author(s).

Document Type: Article
Publication Stage: Final
Source: Scopus
Access Type: Open Access

“Medication-assisted therapies for opioid use disorders in patients with chronic pain” (2020) Journal of the Neurological Sciences

Medication-assisted therapies for opioid use disorders in patients with chronic pain
(2020) Journal of the Neurological Sciences, 411, art. no. 116728, . 

Oesterle, T.S.a , Kolla, B.P.a , Rummans, T.A.a , Gold, M.S.b

a Mayo Clinic – Rochester, Department of Psychiatry & Psychology, 200 First Street SW, Rochester, MN 55905, United States
b Washington University in St. Louis, School of Medicine, St Louis, MO, United States

Abstract
Opioids have been used to treat pain and invoke pleasure for centuries. Modern scientific advancements have led to more potent, synthetic opioids. While certainly more effective in treating pain, they can also be much more addictive. Over the years the scientific community has developed a clearer understanding of the role opioid receptors play in causing and treating opioid use disorders (OUD) and we now know that OUD can develop in individuals taking opioids for “legitimate” pain. Current guidelines suggest that all prescribers (especially those prescribing opioids) be capable treating OUD. Pharmacological advances have led to a wide array of safe and effective treatment options to address OUDs. This paper will discuss the history of opioid development, what is known about the transition from analgesic uses to addiction and modern evidenced based treatment strategies to address OUDs. © 2020 Elsevier B.V.

Document Type: Review
Publication Stage: Final
Source: Scopus

“Molecular neurological correlates of endorphinergic/dopaminergic mechanisms in reward circuitry linked to endorphinergic deficiency syndrome (EDS)” (2020) Journal of the Neurological Sciences

Molecular neurological correlates of endorphinergic/dopaminergic mechanisms in reward circuitry linked to endorphinergic deficiency syndrome (EDS)
(2020) Journal of the Neurological Sciences, 411, art. no. 116733, . 

Blum, K.a , Baron, D.a , McLaughlin, T.b , Gold, M.S.c

a Graduate School of Biomedical Sciences, Western University Health Sciences, Pomona, CA, United States
b Center for Psychiatric Medicine, Lawrence, MA, United States
c Department of Psychiatry, Washington University School of Medicine, St. Louis, MO, United States

Abstract
The consensus of the current literature strongly supports the concept that brain neurotransmitters, and second messengers involved in the net release of dopamine in the mesolimbic region, especially the Nucleus Accumbens (NAc), is directly linked to motivation, anti-stress, incentive salience (wanting), and well-being. The role of dopamine in terms of alcohol withdrawal symptomology, cocaine craving behavior, dopamine –condensation products (TIQs), and more recently, the genetic aspects of drug-seeking and pro-dopamine regulation, provide compelling evidence of the relevant molecular neurological correlates of dopaminergic /endorphinergic mechanisms in reward circuitry due to genetic polymorphisms and epigenetic insults. In the face of an Americans opioid epidemic, the clinical consensus is to treat Opioid Use Disorder (OUD) with life-long opioid substitution therapy. However, the authors suggest a paradigm shift involving novel modalities like targeting the endorphinergic system linked to dopamine release at the NAc, in terms of the induction of required “dopamine homeostasis.” Utilizing the known genetic – environmental interaction theorem P = G +E, the authors provide a clear rationale for the adoption of genetic risk testing coupled with endorphinergic/dopamine regulation to address dysfunction across the brain reward circuitry. The goal of altering resting-state, functional connectivity may require a gentle “neurotransmitter fix” vis enkephalinase inhibition to overcome or combat – self-induction of acute dopamine release via psychoactive substance misuse resulting in chronic dopamine down-regulation. As subsets of reward deficiency, we are poised to provide novel, genetically guided therapy for endorphinergic, opioidergic, and dopaminergic deficiencies and related syndromes, utilizing “Precision Addiction Management. © 2020

Author Keywords
Brain Reward Cascade (BRC);  Dopamine deficiency syndrome;  Dopamine release and homeostasis;  Endorphinergic deficiency syndrome;  Endorphinergic mechanisms;  Genetic testing of addiction liability;  Neurotransmission;  Opioid deficiency syndrome;  Opioid Use Disorder (OUD);  Precision Addiction Management (PAM);  Reward Deficiency Syndrome (RDS)

Document Type: Review
Publication Stage: Final
Source: Scopus

“TRPM3_miR-204: a complex locus for eye development and disease” (2020) Human Genomics

TRPM3_miR-204: a complex locus for eye development and disease
(2020) Human Genomics, 14 (1), p. 7. 

Shiels, A.

Ophthalmology and Visual Sciences, Washington University School of Medicine, 660 S. Euclid Ave., Box 8096, St. Louis, MO, 63110, USA

Abstract
First discovered in a light-sensitive retinal mutant of Drosophila, the transient receptor potential (TRP) superfamily of non-selective cation channels serve as polymodal cellular sensors that participate in diverse physiological processes across the animal kingdom including the perception of light, temperature, pressure, and pain. TRPM3 belongs to the melastatin sub-family of TRP channels and has been shown to function as a spontaneous calcium channel, with permeability to other cations influenced by alternative splicing and/or non-canonical channel activity. Activators of TRPM3 channels include the neurosteroid pregnenolone sulfate, calmodulin, phosphoinositides, and heat, whereas inhibitors include certain drugs, plant-derived metabolites, and G-protein subunits. Activation of TRPM3 channels at the cell membrane elicits a signal transduction cascade of mitogen-activated kinases and stimulus response transcription factors. The mammalian TRPM3 gene hosts a non-coding microRNA gene specifying miR-204 that serves as both a tumor suppressor and a negative regulator of post-transcriptional gene expression during eye development in vertebrates. Ocular co-expression of TRPM3 and miR-204 is upregulated by the paired box 6 transcription factor (PAX6) and mutations in all three corresponding genes underlie inherited forms of eye disease in humans including early-onset cataract, retinal dystrophy, and coloboma. This review outlines the genomic and functional complexity of the TRPM3_miR-204 locus in mammalian eye development and disease.

Author Keywords
Eye development;  Eye disease;  MicroRNA;  TRP channel

Document Type: Review
Publication Stage: Final
Source: Scopus
Access Type: Open Access

“Reconfigurable nanophotonic silicon probes for sub-millisecond deep-brain optical stimulation” (2020) Nature Biomedical Engineering

Reconfigurable nanophotonic silicon probes for sub-millisecond deep-brain optical stimulation
(2020) Nature Biomedical Engineering, . 

Mohanty, A.a b , Li, Q.c d , Tadayon, M.A.a , Roberts, S.P.a , Bhatt, G.R.a , Shim, E.a , Ji, X.a b , Cardenas, J.a e , Miller, S.A.a , Kepecs, A.c d f , Lipson, M.a

a Department of Electrical Engineering, Columbia University, New York, NY, United States
b School of Electrical and Computer Engineering, Cornell University, Ithaca, NY, United States
c Cold Spring Harbor Laboratory, Cold Spring Harbor, New York, NY, United States
d Department of Neuroscience, Washington University in St. Louis, St. Louis, MO, United States
e Institute of Optics, University of Rochester, Rochester, NY, United States
f Department of Psychiatry, Washington University in St. Louis, St. Louis, MO, United States

Abstract
The use of nanophotonics to rapidly and precisely reconfigure light beams for the optical stimulation of neurons in vivo has remained elusive. Here we report the design and fabrication of an implantable silicon-based probe that can switch and route multiple optical beams to stimulate identified sets of neurons across cortical layers and simultaneously record the produced spike patterns. Each switch in the device consists of a silicon nitride waveguide structure that can be rapidly (<20 μs) reconfigured by electrically tuning the phase of light. By using an eight-beam probe, we show in anaesthetized mice that small groups of single neurons can be independently stimulated to produce multineuron spike patterns at sub-millisecond precision. We also show that a probe integrating co-fabricated electrical recording sites can simultaneously optically stimulate and electrically measure deep-brain neural activity. The technology is scalable, and it allows for beam focusing and steering and for structured illumination via beam shaping. The high-bandwidth optical-stimulation capacity of the device might facilitate the probing of the spatiotemporal neural codes underlying behaviour. © 2020, The Author(s), under exclusive licence to Springer Nature Limited.

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

“Shared genetic risk between eating disorder- and substance-use-related phenotypes: Evidence from genome-wide association studies” (2020) Addiction Biology

Shared genetic risk between eating disorder- and substance-use-related phenotypes: Evidence from genome-wide association studies
(2020) Addiction Biology, art. no. e12880, . 

Munn-Chernoff, M.A.a , Johnson, E.C.b , Chou, Y.-L.b , Coleman, J.R.I.c d , Thornton, L.M.a , Walters, R.K.e f , Yilmaz, Z.a g , Baker, J.H.a , Hübel, C.c h , Gordon, S.i , Medland, S.E.i , Watson, H.J.a j k , Gaspar, H.A.c d , Bryois, J.h , Hinney, A.l , Leppä, V.M.h , Mattheisen, M.m n o p , Ripke, S.e f q , Yao, S.h , Giusti-Rodríguez, P.g , Hanscombe, K.B.r , Adan, R.A.H.s t u , Alfredsson, L.v , Ando, T.w , Andreassen, O.A.x , Berrettini, W.H.y , Boehm, I.z , Boni, C.aa , Boraska Perica, V.ab ac , Buehren, K.ad , Burghardt, R.ae , Cassina, M.af , Cichon, S.ag ah ai , Clementi, M.af , Cone, R.D.aj , Courtet, P.ak , Crow, S.al , Crowley, J.J.g n , Danner, U.N.am , Davis, O.S.P.an ao , de Zwaan, M.ap , Dedoussis, G.aq , Degortes, D.ar , DeSocio, J.E.as , Dick, D.M.at au av , Dikeos, D.aw , Dina, C.ax , Dmitrzak-Weglarz, M.ay , Docampo, E.az ba bb , Duncan, L.E.bc , Egberts, K.bd , Ehrlich, S.z , Escaramís, G.az ba bb , Esko, T.be bf , Estivill, X.az ba bb bg , Farmer, A.c , Favaro, A.ar , Fernández-Aranda, F.bh bi , Fichter, M.M.bj bk , Fischer, K.be , Föcker, M.bl , Foretova, L.bm , Forstner, A.J.ah bn bo bp , Forzan, M.af , Franklin, C.S.ab , Gallinger, S.bq , Giegling, I.br , Giuranna, J.l , Gonidakis, F.bs , Gorwood, P.bt bu , Gratacos Mayora, M.az ba bb , Guillaume, S.ak , Guo, Y.bv , Hakonarson, H.bv bw , Hatzikotoulas, K.ab bx , Hauser, J.by , Hebebrand, J.l , Helder, S.G.c bz , Herms, S.ag ah , Herpertz-Dahlmann, B.ad , Herzog, W.ca , Huckins, L.M.ab cb , Hudson, J.I.cc , Imgart, H.cd , Inoko, H.ce , Janout, V.cf , Jiménez-Murcia, S.bh bi , Julià, A.cg , Kalsi, G.c , Kaminská, D.ch , Karhunen, L.ci , Karwautz, A.cj , Kas, M.J.H.s ck , Kennedy, J.L.cl cm cn , Keski-Rahkonen, A.co , Kiezebrink, K.cp , Kim, Y.-R.cq , Klump, K.L.cr , Knudsen, G.P.S.cs , La Via, M.C.a , Le Hellard, S.ct cu cv , Levitan, R.D.cm , Li, D.bv , Lilenfeld, L.cw , Lin, B.D.s , Lissowska, J.cx , Luykx, J.s , Magistretti, P.J.cy cz , Maj, M.da , Mannik, K.be db , Marsal, S.cg , Marshall, C.R.dc , Mattingsdal, M.dd , McDevitt, S.de df , McGuffin, P.c , Metspalu, A.be dg , Meulenbelt, I.dh , Micali, N.di dj , Mitchell, K.dk dl , Monteleone, A.M.da , Monteleone, P.dm , Nacmias, B.dn , Navratilova, M.bm , Ntalla, I.aq , O’Toole, J.K.do , Ophoff, R.A.dp dq , Padyukov, L.dr , Palotie, A.bf ds dt , Pantel, J.aa , Papezova, H.ch , Pinto, D.cb , Rabionet, R.du dv dw , Raevuori, A.co , Ramoz, N.aa , Reichborn-Kjennerud, T.cs dx , Ricca, V.dy , Ripatti, S.dz , Ritschel, F.z ea , Roberts, M.c , Rotondo, A.eb , Rujescu, D.br , Rybakowski, F.ec , Santonastaso, P.ed , Scherag, A.ee , Scherer, S.W.ef eg , Schmidt, U.eh , Schork, N.J.ei , Schosser, A.ej , Seitz, J.ad , Slachtova, L.ek , Slagboom, P.E.el , Slof-Op’t Landt, M.C.T.em en , Slopien, A.eo , Sorbi, S.dn ep , Świątkowska, B.eq , Szatkiewicz, J.P.g , Tachmazidou, I.ab , Tenconi, E.ar , Tortorella, A.er es , Tozzi, F.et , Treasure, J.eh , Tsitsika, A.eu , Tyszkiewicz-Nwafor, M.eo , Tziouvas, K.ev , van Elburg, A.A.t ew , van Furth, E.F.em en , Wagner, G.cj , Walton, E.z , Widen, E.ds , Zeggini, E.ab bx , Zerwas, S.a , Zipfel, S.ex , Bergen, A.W.ey ez , Boden, J.M.fa , Brandt, H.fb , Crawford, S.fb , Halmi, K.A.fc , Horwood, L.J.fa , Johnson, C.fd , Kaplan, A.S.cl cm cn , Kaye, W.H.fe , Mitchell, J.ff , Olsen, C.M.fg , Pearson, J.F.fh , Pedersen, N.L.h , Strober, M.fi fj , Werge, T.fk , Whiteman, D.C.fg , Woodside, D.B.cm cn fl fm , Grove, J.m fn fo fp , Henders, A.K.fq , Larsen, J.T.fn fr fs , Parker, R.i , Petersen, L.V.fn fr fs , Jordan, J.ft fu , Kennedy, M.A.fv , Birgegård, A.h n o , Lichtenstein, P.h , Norring, C.n o , Landén, M.h fw , Mortensen, P.B.fn fr fs , Polimanti, R.fx fy , McClintick, J.N.fz , Adkins, A.E.at au , Aliev, F.at ga , Bacanu, S.-A.gb gc gd , Batzler, A.ge , Bertelsen, S.gf , Biernacka, J.M.gg gh , Bigdeli, T.B.gi , Chen, L.-S.b , Clarke, T.-K.gj , Degenhardt, F.gk , Docherty, A.R.gl , Edwards, A.C.gc gd , Foo, J.C.gm , Fox, L.b , Frank, J.gm , Hack, L.M.bc , Hartmann, A.M.br , Hartz, S.M.b , Heilmann-Heimbach, S.gk , Hodgkinson, C.gn , Hoffmann, P.bo go gp , Hottenga, J.-J.gq , Konte, B.br , Lahti, J.gr , Lahti-Pulkkinen, M.gs , Lai, D.gt , Ligthart, L.gq , Loukola, A.ds , Maher, B.S.gu , Mbarek, H.gq , McIntosh, A.M.gv , McQueen, M.B.gw , Meyers, J.L.gx , Milaneschi, Y.gy , Palviainen, T.ds , Peterson, R.E.gc gd , Ryu, E.gh , Saccone, N.L.hb , Salvatore, J.E.at gc gd , Sanchez-Roige, S.fe , Schwandt, M.gz , Sherva, R.ha , Streit, F.gm , Strohmaier, J.gm , Thomas, N.at au , Wang, J.-C.gf , Webb, B.T.gb gc gd , Wedow, R.e f hb hc , Wetherill, L.gt , Wills, A.G.hd , Zhou, H.fx fy , Boardman, J.D.he hf , Chen, D.f , Choi, D.-S.hg , Copeland, W.E.hh , Culverhouse, R.C.hi , Dahmen, N.hj , Degenhardt, L.hk , Domingue, B.W.hl , Frye, M.A.gh , Gäbel, W.hm , Hayward, C.hn , Ising, M.ho , Keyes, M.hp , Kiefer, F.hq , Koller, G.hr , Kramer, J.hs , Kuperman, S.ht , Lucae, S.ho , Lynskey, M.T.ht , Maier, W.hu , Mann, K.hq , Männistö, S.hv , Müller-Myhsok, B.hw , Murray, A.D.hx , Nurnberger, J.I.gu hy , Preuss, U.hz ia , Räikkönen, K.gs , Reynolds, M.D.ib , Ridinger, M.ic , Scherbaum, N.id , Schuckit, M.A.fe , Soyka, M.ie if , Treutlein, J.gm , Witt, S.H.gm , Wodarz, N.ig , Zill, P.ih , Adkins, D.E.gl ii , Boomsma, D.I.gq , Bierut, L.J.b , Brown, S.A.fe ij , Bucholz, K.K.b , Costello, E.J.ik , de Wit, H.il , Diazgranados, N.im , Eriksson, J.G.in io , Farrer, L.A.hb ip iq ir is , Foroud, T.M.gt , Gillespie, N.A.gc gd , Goate, A.M.gf , Goldman, D.gn it , Grucza, R.A.b , Hancock, D.B.iu , Harris, K.M.iv iw , Hesselbrock, V.ix , Hewitt, J.K.iy , Hopfer, C.J.iz , Iacono, W.G.hq , Johnson, E.O.iv ja , Karpyak, V.M.gg , Kendler, K.S.gc gd , Kranzler, H.R.jb jc , Krauter, K.jd , Lind, P.A.i , McGue, M.hq , MacKillop, J.je jf , Madden, P.A.F.b , Maes, H.H.gc , Magnusson, P.K.E.h , Nelson, E.C.b , Nöthen, M.M.gk , Palmer, A.A.fe jg , Penninx, B.W.J.H.jh , Porjesz, B.gx , Rice, J.P.b , Rietschel, M.gm , Riley, B.P.gb gc gd , Rose, R.J.ji , Shen, P.-H.gn , Silberg, J.gc gd , Stallings, M.C.iz , Tarter, R.E.ic , Vanyukov, M.M.ic , Vrieze, S.hq , Wall, T.L.fe , Whitfield, J.B.i , Zhao, H.jj , Neale, B.M.e f , Wade, T.D.jk , Heath, A.C.b , Montgomery, G.W.i fq jl , Martin, N.G.i , Sullivan, P.F.a g h , Kaprio, J.co ds , Breen, G.c d , Gelernter, J.fx fy jm jn , Edenberg, H.J.fz gt , Bulik, C.M.a h jn , Agrawal, A.b

a Department of Psychiatry, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
b Department of Psychiatry, Washington University School of Medicine, Saint Louis, MO, United States
c Social, Genetic and Developmental Psychiatry (SGDP) Centre, Institute of Psychiatry, Psychology and Neuroscience, King’s College London, London, United Kingdom
d National Institute for Health Research Biomedical Research Centre, King’s College London and South London and Maudsley National Health Service Trust, London, United Kingdom
e Analytic and Translational Genetics Unit, Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA, United States
f Stanley Center for Psychiatric Research, Broad Institute of the Massachusetts Institute of Technology and Harvard University, Cambridge, MA, United States
g Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
h Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
i QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia
j School of Psychology, Curtin University, Perth, WA, Australia
k School of Paediatrics and Child Health, University of Western Australia, Perth, WA, Australia
l Department of Child and Adolescent Psychiatry, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
m Department of Biomedicine, Aarhus University, Aarhus, Denmark
n Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
o Center for Psychiatry Research, Stockholm Health Care Services, Stockholm City Council, Stockholm, Sweden
p Department of Psychiatry, Psychosomatics and Psychotherapy, University of Würzburg, Germany
q Department of Psychiatry and Psychotherapy, Charité – Universitätsmedizin, Berlin, Germany
r Department of Medical and Molecular Genetics, King’s College London, Guy’s Hospital, London, United Kingdom
s Department of Translational Neuroscience, Brain Center Rudolf Magnus, University Medical Center Utrecht, Utrecht, Netherlands
t Center for Eating Disorders Rintveld, Altrecht Mental Health Institute, Zeist, Netherlands
u Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
v Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
w Department of Behavioral Medicine, National Institute of Mental Health, National Center of Neurology and Psychiatry, Kodaira, Japan
x Division of Mental Health and Addiction, NORMENT Centre, University of Oslo, Oslo University Hospital, Oslo, Norway
y Department of Psychiatry, Center for Neurobiology and Behavior, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, United States
z Division of Psychological and Social Medicine and Developmental Neurosciences, Faculty of Medicine, Technische Universität Dresden, Dresden, Germany
aa Centre of Psychiatry and Neuroscience, INSERM U894, Paris, France
ab Wellcome Sanger Institute, Wellcome Genome Campus, Cambridge, United Kingdom
ac Department of Medical Biology, School of Medicine, University of Split, Split, Croatia
ad Department of Child and Adolescent Psychiatry, Psychosomatics and Psychotherapy, RWTH Aachen University, Aachen, Germany
ae Klinikum Frankfurt/Oder, Frankfurt, Germany
af Clinical Genetics Unit, Department of Woman and Child Health, University of Padova, Italy
ag Institute of Medical Genetics and Pathology, University Hospital Basel, Basel, Switzerland
ah Department of Biomedicine, University of Basel, Basel, Switzerland
ai Institute of Neuroscience and Medicine (INM-1), Research Center Juelich, Germany
aj Department of Molecular and Integrative Physiology, Life Sciences Institute, University of Michigan, Ann Arbor, MI, United States
ak Department of Emergency Psychiatry and Post-Acute Care, CHRU Montpellier, University of Montpellier, Montpellier, France
al Department of Psychiatry, University of Minnesota, Minneapolis, MN, United States
am Altrecht Eating Disorders Rintveld, Altrecht Mental Health Institute, Zeist, Netherlands
an MRC Integrative Epidemiology Unit, University of Bristol, Bristol, United Kingdom
ao School of Social and Community Medicine, University of Bristol, Bristol, United Kingdom
ap Department of Psychosomatic Medicine and Psychotherapy, Hannover Medical School, Hannover, Germany
aq Department of Nutrition and Dietetics, Harokopio University, Athens, Greece
ar Department of Neurosciences, University of Padova, Padova, Italy
as College of Nursing, Seattle University, Seattle, WA, United States
at Department of Psychology, Virginia Commonwealth University, Richmond, VA, United States
au College Behavioral and Emotional Health Institute, Virginia Commonwealth University, Richmond, VA, United States
av Department of Human & Molecular Genetics, Virginia Commonwealth University, Richmond, VA, United States
aw Department of Psychiatry, Athens University Medical School, Athens University, Athens, Greece
ax l’institut du thorax, INSERM, CNRS, Univ Nantes, Nantes, France
ay Department of Psychiatric Genetics, Poznan University of Medical Sciences, Poznan, Poland
az Barcelona Institute of Science and Technology, Barcelona, Spain
ba Universitat Pompeu Fabra, Barcelona, Spain
bb Centro de Investigación Biomédica en Red en Epidemiología y Salud Pública (CIBERESP), Barcelona, Spain
bc Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA, United States
bd Department of Child and Adolescent Psychiatry, Psychosomatics and Psychotherapy, Centre for Mental Health, University Hospital of Würzburg, Würzburg, Germany
be Estonian Genome Center, University of Tartu, Tartu, Estonia
bf Program in Medical and Population Genetics, Broad Institute of the Massachusetts Institute of Technology and Harvard University, Cambridge, MA, United States
bg Genomics and Disease, Bioinformatics and Genomics Programme, Centre for Genomic Regulation, Barcelona, Spain
bh Department of Psychiatry, University Hospital of Bellvitge –IDIBELL and CIBERobn, Barcelona, Spain
bi Department of Clinical Sciences, School of Medicine, University of Barcelona, Barcelona, Spain
bj Department of Psychiatry and Psychotherapy, Ludwig-Maximilians-University, Munich, Germany
bk Schön Klinik Roseneck affiliated with the Medical Faculty of the University of Munich, Munich, Germany
bl Department of Child and Adolescent Psychiatry, University of Münster, Münster, Germany
bm Department of Cancer, Epidemiology and Genetics, Masaryk Memorial Cancer Institute, Brno, Czech Republic
bn Centre for Human Genetics, University of Marburg, Marburg, Germany
bo Institute of Human Genetics, School of Medicine & University Hospital Bonn, University of Bonn, Bonn, Germany
bp Department of Psychiatry (UPK), University of Basel, Basel, Switzerland
bq Department of Surgery, Faculty of Medicine, University of Toronto, Toronto, ON, Canada
br Department of Psychiatry, Psychotherapy and Psychosomatics, Martin-Luther-University Halle-Wittenberg, Halle (Saale), Germany
bs 1st Psychiatric Department, National and Kapodistrian University of Athens, Medical School, Eginition Hospital, Athens, Greece
bt Institute of Psychiatry and Neuroscience of Paris, INSERM U1266, Paris, France
bu CMME (GHU Paris Psychiatrie et Neurosciences), Paris Descartes University, Paris, France
bv Center for Applied Genomics, Children’s Hospital of Philadelphia, Philadelphia, PA, United States
bw Department of Pediatrics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, United States
bx Helmholtz Zentrum München – German Research Centre for Environmental Health, Institute of Translational Genomics, Neuherberg, Germany
by Department of Adult Psychiatry, Poznan University of Medical Sciences, Poznan, Poland
bz Zorg op Orde, Delft, Netherlands
ca Department of General Internal Medicine and Psychosomatics, Heidelberg University Hospital, Heidelberg University, Heidelberg, Germany
cb Department of Psychiatry, and Genetics and Genomics Sciences, Division of Psychiatric Genomics, Icahn School of Medicine at Mount Sinai, New York, NY, United States
cc Biological Psychiatry Laboratory, McLean Hospital/Harvard Medical School, Boston, MA, United States
cd Eating Disorders Unit, Parklandklinik, Bad Wildungen, Germany
ce Department of Molecular Life Science, Division of Basic Medical Science and Molecular Medicine, School of Medicine, Tokai University, Isehara, Japan
cf Faculty of Health Sciences, Palacky University, Olomouc, Czech Republic
cg Rheumatology Research Group, Vall d’Hebron Research Institute, Barcelona, Spain
ch Department of Psychiatry, First Faculty of Medicine, Charles University, Prague, Czech Republic
ci Department of Clinical Nutrition, Institute of Public Health and Clinical Nutrition, University of Eastern Finland, Kuopio, Finland
cj Eating Disorders Unit, Department of Child and Adolescent Psychiatry, Medical University of Vienna, Vienna, Austria
ck Groningen Institute for Evolutionary Life Sciences, University of Groningen, Groningen, Netherlands
cl Centre for Addiction and Mental Health, Toronto, ON, Canada
cm Institute of Medical Science, University of Toronto, Toronto, ON, Canada
cn Department of Psychiatry, University of Toronto, Toronto, ON, Canada
co Department of Public Health, University of Helsinki, Helsinki, Finland
cp Institute of Applied Health Sciences, School of Medicine, Medical Sciences and Nutrition, University of Aberdeen, Aberdeen, United Kingdom
cq Department of Psychiatry, Seoul Paik Hospital, Inje University, Seoul, South Korea
cr Department of Psychology, Michigan State University, East Lansing, MI, United States
cs Department of Mental Disorders, Norwegian Institute of Public Health, Oslo, Norway
ct Department of Clinical Science, Norwegian Centre for Mental Disorders Research (NORMENT), University of Bergen, Bergen, Norway
cu Dr. Einar Martens Research Group for Biological Psychiatry, Center for Medical Genetics and Molecular Medicine, Haukeland University Hospital, Bergen, Norway
cv Department of Clinical Medicine, Laboratory Building, Haukeland University Hospital, Bergen, Norway
cw The Chicago School of Professional Psychology, Washington DC Campus, Washington, District of Columbia, United States
cx Department of Cancer Epidemiology and Prevention, M Skłodowska-Curie Cancer Center – Oncology Center, Warsaw, Poland
cy BESE Division, King Abdullah University of Science and Technology, Thuwal, Saudi Arabia
cz Department of Psychiatry, University of Lausanne-University Hospital of Lausanne (UNIL-CHUV), Lausanne, Switzerland
da Department of Psychiatry, University of Campania “Luigi Vanvitelli”, Naples, Italy
db Center for Integrative Genomics, University of Lausanne, Lausanne, Switzerland
dc Department of Paediatric Laboratory Medicine, Division of Genome Diagnostics, The Hospital for Sick Children, Toronto, ON, Canada
dd NORMENT KG Jebsen Centre, Division of Mental Health and Addiction, University of Oslo, Oslo University Hospital, Oslo, Norway
de Department of Psychiatry, University College Cork, Cork, Ireland
df Eist Linn Adolescent Unit, Bessborough, Health Service Executive South, Cork, Ireland
dg Institute of Molecular and Cell Biology, University of Tartu, Tartu, Estonia
dh Molecular Epidemiology Section (Department of Biomedical Datasciences), Leiden University Medical Centre, Leiden, Netherlands
di Department of Psychiatry, Faculty of Medicine, University of Geneva, Geneva, Switzerland
dj Division of Child and Adolescent Psychiatry, Geneva University Hospital, Geneva, Switzerland
dk National Center for PTSD, VA Boston Healthcare System, Boston, MA, United States
dl Department of Psychiatry, Boston University School of Medicine, Boston, MA, United States
dm Department of Medicine, Surgery and Dentistry “Scuola Medica Salernitana”, University of Salerno, Salerno, Italy
dn Department of Neuroscience, Psychology, Drug Research and Child Health (NEUROFARBA), University of Florence, Florence, Italy
do Kartini Clinic, Portland, OR, United States
dp Center for Neurobehavioral Genetics, Semel Institute for Neuroscience and Human Behavior, University of California Los Angeles, Los Angeles, CA, United States
dq Department of Psychiatry, Erasmus MC, University Medical Center Rotterdam, Rotterdam, Netherlands
dr Department of Medicine, Center for Molecular Medicine, Division of Rheumatology, Karolinska Institutet and Karolinska University Hospital, Stockholm, Sweden
ds Institute for Molecular Medicine FIMM, HiLIFE, University of Helsinki, Helsinki, Finland
dt Center for Human Genome Research, Massachusetts General Hospital, Boston, MA, United States
du Saint Joan de Déu Research Institute, Saint Joan de Déu Barcelona Children’s Hospital, Barcelona, Spain
dv Institute of Biomedicine (IBUB), University of Barcelona, Barcelona, Spain
dw Department of Genetics, Microbiology and Statistics, University of Barcelona, Barcelona, Spain
dx Institute of Clinical Medicine, University of Oslo, Oslo, Norway
dy Department of Health Science, University of Florence, Florence, Italy
dz Department of Biometry, University of Helsinki, Helsinki, Finland
ea Department of Child and Adolescent Psychiatry, Faculty of Medicine, Eating Disorders Research and Treatment Center, Technische Universität Dresden, Dresden, Germany
eb Department of Psychiatry, Neurobiology, Pharmacology, and Biotechnologies, University of Pisa, Pisa, Italy
ec Department of Psychiatry, Poznan University of Medical Sciences, Poznan, Poland
ed Department of Neurosciences, Padua Neuroscience Center, University of Padova, Padova, Italy
ee Institute of Medical Statistics, Computer and Data Sciences, Jena University Hospital, Jena, Germany
ef Department of Genetics and Genomic Biology, The Hospital for Sick Children, Toronto, ON, Canada
eg McLaughlin Centre, University of Toronto, Toronto, ON, Canada
eh Department of Psychological Medicine, Institute of Psychiatry, Psychology and Neuroscience, King’s College London, London, United Kingdom
ei J. Craig Venter Institute (JCVI), La Jolla, CA, United States
ej Department of Psychiatry and Psychotherapy, Medical University of Vienna, Vienna, Austria
ek Department of Pediatrics and Center of Applied Genomics, First Faculty of Medicine, Charles University, Prague, Czech Republic
el Molecular Epidemiology Section (Department of Medical Statistics), Leiden University Medical Centre, Leiden, Netherlands
em Center for Eating Disorders Ursula, Rivierduinen, Leiden, Netherlands
en Department of Psychiatry, Leiden University Medical Centre, Leiden, Netherlands
eo Department of Child and Adolescent Psychiatry, Poznan University of Medical Sciences, Poznan, Poland
ep IRCCS Fondazione Don Carlo Gnocchi, Florence, Italy
eq Department of Environmental Epidemiology, Nofer Institute of Occupational Medicine, Lodz, Poland
er Department of Psychiatry, University of Naples SUN, Naples, Italy
es Department of Psychiatry, University of Perugia, Perugia, Italy
et Brain Sciences Department, Stremble Ventures, Limassol, Cyprus
eu Adolescent Health Unit, Second Department of Pediatrics, “P. & A. Kyriakou” Children’s Hospital, University of Athens, Athens, Greece
ev Pediatric Intensive Care Unit, “P. & A. Kyriakou” Children’s Hospital, University of Athens, Athens, Greece
ew Faculty of Social and Behavioral Sciences, Utrecht University, Utrecht, Netherlands
ex Department of Internal Medicine VI, Psychosomatic Medicine and Psychotherapy, University Medical Hospital Tuebingen, Tuebingen, Germany
ey BioRealm, LLC, Walnut, CA, United States
ez Oregon Research Institute, Eugene, OR, United States
fa Christchurch Health and Development Study, University of Otago, Christchurch, New Zealand
fb The Center for Eating Disorders at Sheppard Pratt, Baltimore, MD, United States
fc Department of Psychiatry, Weill Cornell Medical College, New York, NY, United States
fd Eating Recovery Center, Denver, CO, United States
fe Department of Psychiatry, University of California San Diego, La Jolla, CA, United States
ff Department of Psychiatry and Behavioral Science, University of North Dakota School of Medicine and Health Sciences, Fargo, ND, United States
fg Population Health Department, QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia
fh Biostatistics and Computational Biology Unit, University of Otago, Christchurch, New Zealand
fi Department of Psychiatry and Biobehavioral Science, Semel Institute for Neuroscience and Human Behavior, University of California Los Angeles, California, Los Angeles, United States
fj David Geffen School of Medicine, University of California Los Angeles, California, Los Angeles, United States
fk Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
fl Centre for Mental Health, University Health Network, Toronto, ON, Canada
fm Program for Eating Disorders, University Health Network, Toronto, ON, Canada
fn The Lundbeck Foundation Initiative for Integrative Psychiatric Research (iPSYCH), Aarhus, Denmark
fo Centre for Integrative Sequencing, iSEQ, Aarhus University, Aarhus, Denmark
fp Bioinformatics Research Centre, Aarhus University, Aarhus, Denmark
fq Institute for Molecular Bioscience, University of Queensland, Brisbane, QLD, Australia
fr National Centre for Register-Based Research, Aarhus BSS, Aarhus University, Aarhus, Denmark
fs Centre for Integrated Register-based Research (CIRRAU), Aarhus University, Aarhus, Denmark
ft Department of Psychological Medicine, University of Otago, Christchurch, New Zealand
fu Canterbury District Health Board, Christchurch, New Zealand
fv Department of Pathology and Biomedical Science, University of Otago, Christchurch, New Zealand
fw Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
fx Department of Psychiatry, Division of Human Genetics, Yale School of Medicine, New Haven, CT, United States
fy Veterans Affairs Connecticut Healthcare System, West Haven, CT, United States
fz Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, IN, United States
ga Faculty of Business, Karabuk University, Karabuk, Turkey
gb Virginia Commonwealth University Alcohol Research Center, Virginia Commonwealth University, Richmond, VA, United States
gc Virginia Institute for Psychiatric and Behavioral Genetics, Virginia Commonwealth University, Richmond, VA, United States
gd Department of Psychiatry, Virginia Commonwealth University, Richmond, VA, United States
ge Psychiatric Genomics and Pharmacogenomics Program, Mayo Clinic, Rochester, MN, United States
gf Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, United States
gg Department of Health Sciences Research, Mayo Clinic, Rochester, MN, United States
gh Department of Psychiatry and Psychology, Mayo Clinic, Rochester, MN, United States
gi Department of Psychiatry and Behavioral Sciences, State University of New York Downstate Medical Center, Brooklyn, NY, United States
gj Division of Psychiatry, University of Edinburgh, Edinburgh, United Kingdom
gk Institute of Human Genetics, University of Bonn School of Medicine & University Hospital Bonn, Bonn, Germany
gl Department of Psychiatry, University of Utah, Salt Lake City, UT, United States
gm Department of Genetic Epidemiology in Psychiatry, Central Institute of Mental Health, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
gn Laboratory of Neurogenetics, NIH/NIAAA, Bethesda, MD, United States
go Human Genomics Research Group, Department of Biomedicine, University of Basel, Basel, Switzerland
gp Institute of Medical Genetics and Pathology, University Hospital Basel, University Hospital Basel, Basel, Switzerland
gq Department of Biological Psychology, Amsterdam Public Health Research Institute, Vrije Universiteit Amsterdam, Amsterdam, Netherlands
gr Turku Institute for Advanced Studies, University of Turku, Turku, Finland
gs Department of Psychology and Logopedics, University of Helsinki, Helsinki, Finland
gt Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN, United States
gu Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, United States
gv Division of Psychiatry, Centre for Cognitive Ageing and Cognitive Epidemiology, University of Edinburgh, Edinburgh, United Kingdom
gw Department of Integrative Physiology, University of Colorado Boulder, Boulder, CO, United States
gx Department of Psychiatry and Behavioral Sciences, Henri Begleiter Neurodynamics Laboratory, SUNY Downstate Medical Center, Brooklyn, NY, United States
gy Department of Psychiatry, Amsterdam Public Health Research Institute, VU University Medical Center/GGz inGeest, Amsterdam, Netherlands
gz Department of Medicine (Biomedical Genetics), Boston University School of Medicine, Boston, MA, United States
ha Department of Epidemiology, Harvard T.H. Chan School of Public Health, Harvard University, Cambridge, MA, United States
hb Department of Sociology, Harvard University, Cambridge, MA, United States
hc Department of Pharmacology, University of Colorado School of Medicine, Aurora, CO, United States
hd Institute of Behavioral Science, University of Colorado, Boulder, CO, United States
he Department of Sociology, University of Colorado, Boulder, CO, United States
hf Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, MN, United States
hg Department of Psychiatry, University of Vermont Medical Center, Burlington, VT, United States
hh Department of Medicine, Division of Biostatistics, Washington University School of Medicine, Saint Louis, MO, United States
hi Department of Psychiatry, University of Mainz, Mainz, Germany
hj National Drug and Alcohol Research Centre, University of New South Wales, Sydney, NSW, Australia
hk Stanford University Graduate School of Education, Stanford University, Stanford, CA, United States
hl Department of Psychiatry and Psychotherapy, University of Düsseldorf, Duesseldorf, Germany
hm MRC Human Genetics Unit, Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh, United Kingdom
hn Max-Planck-Institute of Psychiatry, Munich, Germany
ho Department of Psychology, University of Minnesota, Minneapolis, MN, United States
hp Department of Addictive Behavior and Addiction Medicine, Central Institute of Mental Health, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
hq Department of Psychiatry and Psychotherapy, University Hospital, LMU Munich, Munich, Germany
hr Department of Psychiatry, University of Iowa Roy J and Lucille A Carver College of Medicine, Iowa City, IA, United States
hs Addictions Department, Institute of Psychiatry, Psychology & Neuroscience, King’s College London, London, United Kingdom
ht Department of Psychiatry, University of Bonn, Bonn, Germany
hu Department of Public Health Solutions, National Institute for Health and Welfare, Helsinki, Finland
hv Department of Statistical Genetics, Max-Planck-Institute of Psychiatry, München, Germany
hw Aberdeen Biomedical Imaging Centre, School of Medicine, Medical Sciences & Nutrition, University of Aberdeen, Foresterhill, Aberdeen, United Kingdom
hx Department of Psychiatry, Indiana University School of Medicine, Indianapolis, IN, United States
hy Department of Psychiatry, Psychotherapy and Psychosomatics, Martin-Luther-University Halle-Wittenberg, Herborn, Germany
hz Department of Psychiatry and Psychotherapy, Vitos Hospital Herborn, Herborn, Germany
ia School of Pharmacy, University of Pittsburgh, Pittsburgh, PA, United States
ib Department of Psychiatry and Psychotherapy, University of Regensburg Psychiatric Health Care Aargau, Regensburg, Germany
ic Department of Psychiatry and Psychotherapy and Department of Addictive Behaviour and Addiction Medicine, Medical Faculty, LVR-Hospital Essen, University of Duisburg-Essen, Essen, Germany
id Medical Park Chiemseeblick in Bernau-Felden, Ludwig-Maximilians-University, Bernau am Chiemsee, Germany
ie Psychiatric Hospital, Ludwig-Maximilians-University, Bernau am Chiemsee, Germany
if Department of Psychiatry and Psychotherapy, University of Regensburg, Regensburg, Germany
ig Department of Psychiatry, Psychiatric Hospital, Ludwig-Maximilians-University, Munich, Germany
ih Department of Sociology, University of Utah, Salt Lake City, UT, United States
ii Department of Psychology, University of California San Diego, San Diego, CA, United States
ij Department of Psychiatry and Behavioral Sciences, Duke University Medical Center, Durham, NC, United States
ik Department of Psychiatry and Behavioral Neuroscience, University of Chicago, Chicago, IL, United States
il NIAAA Intramural Research Program, Bethesda, MD, United States
im Department of General Practice and Primary Health Care, University of Helsinki, Helsinki, Finland
in National Institute for Health and Welfare, Helsinki, Finland
io Department of Neurology, Boston University School of Medicine, Boston, MA, United States
ip Department of Ophthalmology, Boston University School of Medicine, Boston, MA, United States
iq Department of Epidemiology, School of Public Health, Boston University, Boston, MA, United States
ir Department of Biostatistics, School of Public Health, Boston University, Boston, MA, United States
is Office of the Clinical Director, NIH/NIAAA, Besthesda, MD, United States
it Center for Omics Discovery and Epidemiology, Behavioral Health Research Division, RTI International, Research Triangle Park, NC, United States
iu Department of Sociology, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
iv Carolina Population Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
iw Department of Psychiatry, University of Connecticut School of Medicine, Farmington, CT, United States
ix Institute for Behavioral Genetics, University of Colorado Boulder, Boulder, CO, United States
iy Department of Psychiatry, University of Colorado Denver, Aurora, CO, United States
iz Fellow Program, RTI International, Research Triangle Park, NC, United States
ja Center for Studies of Addiction, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, United States
jb VISN 4 MIRECC, Crescenz VAMC, Philadelphia, PA, United States
jc Department of Molecular, Cellular, and Developmental Biology, University of Colorado Boulder, Boulder, CO, United States
jd Peter Boris Centre for Addictions Research, McMaster University/St. Joseph’s Healthcare Hamilton, Hamilton, ON, Canada
je Michael G. DeGroote Centre for Medicinal Cannabis Research, McMaster University/St. Joseph’s Healthcare Hamilton, Hamilton, ON, Canada
jf Institute for Genomic Medicine, University of California San Diego, La Jolla, CA, United States
jg Department of Psychiatry, Amsterdam UMC, VU University and GGZinGeest, Amsterdam, Netherlands
jh Department of Psychological & Brain Sciences, Indiana University Bloomington, Bloomington, IN, United States
ji Department of Biostatistics, Yale School of Public Health, Yale University, New Haven, CT, United States
jj School of Psychology, Flinders University, Adelaide, SA, Australia
jk Queensland Brain Institute, University of Queensland, Brisbane, QLD, Australia
jl Department of Genetics, Yale School of Medicine, New Haven, CT, United States
jm Department of Neuroscience, Yale School of Medicine, New Haven, CT, United States
jn Department of Nutrition, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States

Abstract
Eating disorders and substance use disorders frequently co-occur. Twin studies reveal shared genetic variance between liabilities to eating disorders and substance use, with the strongest associations between symptoms of bulimia nervosa and problem alcohol use (genetic correlation [rg], twin-based = 0.23-0.53). We estimated the genetic correlation between eating disorder and substance use and disorder phenotypes using data from genome-wide association studies (GWAS). Four eating disorder phenotypes (anorexia nervosa [AN], AN with binge eating, AN without binge eating, and a bulimia nervosa factor score), and eight substance-use-related phenotypes (drinks per week, alcohol use disorder [AUD], smoking initiation, current smoking, cigarettes per day, nicotine dependence, cannabis initiation, and cannabis use disorder) from eight studies were included. Significant genetic correlations were adjusted for variants associated with major depressive disorder and schizophrenia. Total study sample sizes per phenotype ranged from ~2400 to ~537 000 individuals. We used linkage disequilibrium score regression to calculate single nucleotide polymorphism-based genetic correlations between eating disorder- and substance-use-related phenotypes. Significant positive genetic associations emerged between AUD and AN (rg = 0.18; false discovery rate q = 0.0006), cannabis initiation and AN (rg = 0.23; q &lt; 0.0001), and cannabis initiation and AN with binge eating (rg = 0.27; q = 0.0016). Conversely, significant negative genetic correlations were observed between three nondiagnostic smoking phenotypes (smoking initiation, current smoking, and cigarettes per day) and AN without binge eating (rgs = −0.19 to −0.23; qs &lt; 0.04). The genetic correlation between AUD and AN was no longer significant after co-varying for major depressive disorder loci. The patterns of association between eating disorder- and substance-use-related phenotypes highlights the potentially complex and substance-specific relationships among these behaviors. © 2020 Society for the Study of Addiction

Author Keywords
eating disorders;  genetic correlation;  substance use

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

“Density and Dichotomous Family History Measures of Alcohol Use Disorder as Predictors of Behavioral and Neural Phenotypes: A Comparative Study Across Gender and Race/Ethnicity” (2020) Alcoholism: Clinical and Experimental Research

Density and Dichotomous Family History Measures of Alcohol Use Disorder as Predictors of Behavioral and Neural Phenotypes: A Comparative Study Across Gender and Race/Ethnicity
(2020) Alcoholism: Clinical and Experimental Research, . 

Pandey, G.a , Seay, M.J.b , Meyers, J.L.a , Chorlian, D.B.a , Pandey, A.K.a , Kamarajan, C.a , Ehrenberg, M.a , Pitti, D.a , Kinreich, S.a , Subbie-Saenz de Viteri, S.a , Acion, L.c , Anokhin, A.d , Bauer, L.e , Chan, G.e , Edenberg, H.f , Hesselbrock, V.e , Kuperman, S.g , McCutcheon, V.V.d , Bucholz, K.K.d , Schuckit, M.h , Porjesz, B.a

a Department of Psychiatry and Behavioral Sciences, Downstate Medical Center, State University of New York, Brooklyn, NY, United States
b Department of Psychology, University of California, Los Angeles, CA, United States
c Iowa Consortium for Substance Abuse Research and Evaluation, University of Iowa, Iowa City, IA, United States
d Department of Psychiatry, Washington University School of Medicine in St. Louis, St. Louis, MO, United States
e Department of Psychiatry, University of Connecticut School of Medicine, Farmington, CT, United States
f Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, IN, United States
g Department of Psychiatry, University of Iowa Carver College of Medicine, Iowa City, IA, United States
h Department of Psychiatry, University of California San Diego, La Jolla, CA, United States

Abstract
Background: Family history (FH) is an important risk factor for the development of alcohol use disorder (AUD). A variety of dichotomous and density measures of FH have been used to predict alcohol outcomes; yet, a systematic comparison of these FH measures is lacking. We compared 4 density and 4 commonly used dichotomous FH measures and examined variations by gender and race/ethnicity in their associations with age of onset of regular drinking, parietal P3 amplitude to visual target, and likelihood of developing AUD. Methods: Data from the Collaborative Study on the Genetics of Alcoholism (COGA) were utilized to compute the density and dichotomous measures. Only subjects and their family members with DSM-5 AUD diagnostic information obtained through direct interviews using the Semi-Structured Assessment for the Genetics of Alcoholism (SSAGA) were included in the study. Area under receiver operating characteristic curves were used to compare the diagnostic accuracy of FH measures at classifying DSM-5 AUD diagnosis. Logistic and linear regression models were used to examine associations of FH measures with alcohol outcomes. Results: Density measures had greater diagnostic accuracy at classifying AUD diagnosis, whereas dichotomous measures presented diagnostic accuracy closer to random chance. Both dichotomous and density measures were significantly associated with likelihood of AUD, early onset of regular drinking, and low parietal P3 amplitude, but density measures presented consistently more robust associations. Further, variations in these associations were observed such that among males (vs. females) and Whites (vs. Blacks), associations of alcohol outcomes with density (vs. dichotomous) measures were greater in magnitude. Conclusions: Density (vs. dichotomous) measures seem to present more robust associations with alcohol outcomes. However, associations of dichotomous and density FH measures with different alcohol outcomes (behavioral vs. neural) varied across gender and race/ethnicity. These findings have great applicability for alcohol research examining FH of AUD. © 2020 by the Research Society on Alcoholism

Author Keywords
Alcohol Use Disorder;  Endophenotype;  Family History;  P300;  Risk and Development

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

“Neural stem cell therapy of foetal onset hydrocephalus using the HTx rat as experimental model” (2020) Cell and Tissue Research

Neural stem cell therapy of foetal onset hydrocephalus using the HTx rat as experimental model
(2020) Cell and Tissue Research, . 

Henzi, R.a b , Vío, K.a , Jara, C.a , Johanson, C.E.c , McAllister, J.P.d , Rodríguez, E.M.a , Guerra, M.a

a Instituto de Anatomía, Histología y Patología, Facultad de Medicina, Universidad Austral de Chile, Valdivia, Chile
b Centro de Investigaciones Biomédicas, Facultad de Medicina, Universidad de los Andes, Santiago, Chile
c Department of Neurosurgery, Alpert Medical School at Brown University, Providence, RI, United States
d Department of Neurosurgery Division of Pediatric Neurosurgery, Washington University and the Saint Louis Children’s Hospital, St. Louis, MO, United States

Abstract
Foetal onset hydrocephalus is a disease starting early in embryonic life; in many cases it results from a cell junction pathology of neural stem (NSC) and neural progenitor (NPC) cells forming the ventricular zone (VZ) and sub-ventricular zone (SVZ) of the developing brain. This pathology results in disassembling of VZ and loss of NSC/NPC, a phenomenon known as VZ disruption. At the cerebral aqueduct, VZ disruption triggers hydrocephalus while in the telencephalon, it results in abnormal neurogenesis. This may explain why derivative surgery does not cure hydrocephalus. NSC grafting appears as a therapeutic opportunity. The present investigation was designed to find out whether this is a likely possibility. HTx rats develop hereditary hydrocephalus; 30–40% of newborns are hydrocephalic (hyHTx) while their littermates are not (nHTx). NSC/NPC from the VZ/SVZ of nHTx rats were cultured into neurospheres that were then grafted into a lateral ventricle of 1-, 2- or 7-day-old hyHTx. Once in the cerebrospinal fluid, neurospheres disassembled and the freed NSC homed at the areas of VZ disruption. A population of homed cells generated new multiciliated ependyma at the sites where the ependyma was missing due to the inherited pathology. Another population of NSC homed at the disrupted VZ differentiated into βIII-tubulin+ spherical cells likely corresponding to neuroblasts that progressed into the parenchyma. The final fate of these cells could not be established due to the protocol used to label the grafted cells. The functional outcomes of NSC grafting in hydrocephalus remain open. The present study establishes an experimental paradigm of NSC/NPC therapy of foetal onset hydrocephalus, at the etiologic level that needs to be further explored with more analytical methodologies. © 2020, Springer-Verlag GmbH Germany, part of Springer Nature.

Author Keywords
Congenital hydrocephalus;  Ependymogenesis;  Homing;  Neural stem cells;  Neurospheres;  Repair;  Transplantation;  Ventricular zone disruption

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

“Complex interactions underlie racial disparity in the risk of developing Alzheimer’s disease dementia” (2020) Alzheimer’s and Dementia

Complex interactions underlie racial disparity in the risk of developing Alzheimer’s disease dementia
(2020) Alzheimer’s and Dementia, . 

Xiong, C.a b , Luo, J.a c d , Coble, D.a b , Agboola, F.a b , Kukull, W.e f , Morris, J.C.b g h

a Division of Biostatistics, Washington University School of Medicine, St. Louis, MO, United States
b Knight Alzheimer Disease Research Center, Washington University School of Medicine, St. Louis, MO, United States
c Division of Public Health Sciences, Department of Surgery, Washington University School of Medicine, St. Louis, MO, United States
d Siteman Cancer Center Biostatistics Core, Washington University School of Medicine, St. Louis, MO, United States
e Department of Epidemiology, University of Washington, Seattle, WA, United States
f National Alzheimer’s Coordinating Center, University of Washington, Seattle, WA, United States
g Department of Neurology, Washington University School of Medicine, St. Louis, MO, United States
h Departments of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO, United States

Abstract
Introduction: We aim to determine racial disparities and their modifying factors in risk for Alzheimer’s disease (AD) dementia among cognitively normal individuals 65 years or older. Methods: Longitudinal data from the National Alzheimer’s Coordinating Center Uniform Data Set on 1229 African Americans (AAs) and 6679 whites were analyzed for the risk of AD using competing risk models with death as a competing event. Results: Major AD risk factors modified racial differences which, when statistically significant, occurred only with older age among APOE ε4 negative individuals, but also with younger age among APOE ε4 positive individuals. The racial differences favored AAs among individuals with body mass index (BMI) < 30, but whites among individuals with a high BMI (≥ 30), and were additionally modified by sex, education, hypertension, and smoking status. Conclusions: The presence, direction, and relative magnitude of racial disparity for AD represent an interactive function of major AD and cerebrovascular risk factors. © 2020 the Alzheimer’s Association

Author Keywords
competing risk survival model;  interactions;  racial disparity;  risk of Alzheimer’s disease

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

“Skeletal Muscle Regeneration in Advanced Diabetic Peripheral Neuropathy” (2020) Foot and Ankle International

Skeletal Muscle Regeneration in Advanced Diabetic Peripheral Neuropathy
(2020) Foot and Ankle International, . 

Bohnert, K.L.a , Hastings, M.K.a b , Sinacore, D.R.c , Johnson, J.E.b , Klein, S.E.b , McCormick, J.J.b , Gontarz, P.d , Meyer, G.A.a b e

a Program in Physical Therapy, Washington University, St. Louis, MO, United States
b Department of Orthopaedic Surgery, Washington University, St. Louis, MO, United States
c Department of Physical Therapy, High Point University, High Point, NC, United States
d Department of Developmental Biology, Washington University, St. Louis, MO, United States
e Departments of Neurology and Biomedical Engineering, Washington University, St. Louis, MO, United States

Abstract
Background: Decreased lean muscle mass in the lower extremity in diabetic peripheral neuropathy (DPN) is thought to contribute to altered joint loading, immobility, and disability. However, the mechanism behind this loss is unknown and could derive from a reduction in size of myofibers (atrophy), destruction of myofibers (degeneration), or both. Degenerative changes require participation of muscle stem (satellite) cells to regenerate lost myofibers and restore lean mass. Determining the degenerative state and residual regenerative capacity of DPN muscle will inform the utility of regeneration-targeted therapeutic strategies. Methods: Biopsies were acquired from 2 muscles in 12 individuals with and without diabetic neuropathy undergoing below-knee amputation surgery. Biopsies were subdivided for histological analysis, transcriptional profiling, and satellite cell isolation and culture. Results: Histological analysis revealed evidence of ongoing degeneration and regeneration in DPN muscles. Transcriptional profiling supports these findings, indicating significant upregulation of regeneration-related pathways. However, regeneration appeared to be limited in samples exhibiting the most severe structural pathology as only extremely small, immature regenerated myofibers were found. Immunostaining for satellite cells revealed a significant decrease in their relative frequency only in the subset with severe pathology. Similarly, a reduction in fusion in cultured satellite cells in this group suggests impairment in regenerative capacity in severe DPN pathology. Conclusion: DPN muscle exhibited features of degeneration with attempted regeneration. In the most severely pathological muscle samples, regeneration appeared to be stymied and our data suggest that this may be partly due to intrinsic dysfunction of the satellite cell pool in addition to extrinsic structural pathology (eg, nerve damage). Clinical Relevance: Restoration of DPN muscle function for improved mobility and physical activity is a goal of surgical and rehabilitation clinicians. Identifying myofiber degeneration and compromised regeneration as contributors to dysfunction suggests that adjuvant cell-based therapies may improve clinical outcomes. © The Author(s) 2020.

Author Keywords
degeneration;  diabetes;  satellite cells;  transcriptional analysis

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

“Concern of photosensitive seizures evoked by 3D video displays or virtual reality headsets in children: Current perspective” (2020) Eye and Brain

Concern of photosensitive seizures evoked by 3D video displays or virtual reality headsets in children: Current perspective
(2020) Eye and Brain, 12, pp. 45-48. 

Tychsen, L.a b c , Thio, L.L.b c d

a Department of Ophthalmology and Visual Sciences, St. Louis Children’s Hospital, Washington University in St. Louis School of Medicine, St. Louis, MO, United States
b Department of Pediatrics, St. Louis Children’s Hospital, Washington University in St. Louis School of Medicine, St. Louis, MO, United States
c Department of Neuroscience, St. Louis Children’s Hospital, Washington University in St. Louis School of Medicine, St. Louis, MO, United States
d Department of Neurology, St. Louis Children’s Hospital, Washington University in St. Louis School of Medicine, St. Louis, MO, United States

Abstract
This review assesses the risk of a photic-induced seizure in a child during viewing of 3D (binocular 3 dimensional, stereoscopic) movies or games, either on standard video displays or when wearing a virtual reality (VR) headset. Studies published by pediatric epilepsy experts emphasize the low risk of 3D viewing even for children with known photosensitive epilepsy (PSE). The low incidence of PSE is noteworthy because the number of hours devoted to 2D or 3D screen viewing and/or VR headset use by children worldwide has increased markedly over the last decade. The medical literature does not support the notion that VR headset use poses a risk for PSE. © 2020 Tychsen and Thio.

Author Keywords
Children;  Epilepsy;  Stereoscopic;  Virtual reality

Document Type: Review
Publication Stage: Final
Source: Scopus
Access Type: Open Access

“Using practice effects for targeted trials or sub-group analysis in Alzheimer’s disease: How practice effects predict change over time” (2020) PLoS ONE

Using practice effects for targeted trials or sub-group analysis in Alzheimer’s disease: How practice effects predict change over time
(2020) PLoS ONE, 15 (2), art. no. e0228064, . 

Wang, G.a , Kennedy, R.E.b , Goldberg, T.E.c , Fowler, M.E.d , Cutter, G.R.e , Schneider, L.S.f

a Division of Biostatistics, Washington University in St. Louis, St. Louis, MO, United States
b Comprehensive Center of Aging Health, University of Alabama at Birmingham, Birmingham, AL, United States
c Department of Psychiatry, Columbia University, New York, NY, United States
d Department of Epidemiology, University of Alabama at Birmingham, Birmingham, AL, United States
e Department of Biostatistics, University of Alabama at Birmingham, Birmingham, AL, United States
f Department of Psychiatry and The Behavioral Sciences, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States

Abstract
Objective To describe the presence of practice effects in persons with Alzheimer disease (AD) or mild cognitive impairment (MCI) and to evaluate how practice effects affect cognitive progression and the outcome of clinical trials. Methods Using data from a meta-database consisting of 18 studies including participants from the Alzheimer disease Cooperative Study (ADCS) and the Alzheimer Disease Neuroimaging Initiative (ADNI) with ADAS-Cog11 as the primary outcome, we defined practice effects based on the improvement in the first two ADAS-Cog11 scores and then estimated the presence of practice effects and compared the cognitive progression between participants with and without practice effects. The robustness of practice effects was investigated using CDR SB, an outcome independent the definition itself. Furthermore, we evaluated how practice effects can affect sample size estimation. Results The overall percent of practice effects for AD participants was 39.0% and 53.3% for MCI participants. For AD studies, the mean change from baseline to 2 years was 12.8 points for the non-practice effects group vs 7.4 for the practice effects group; whereas for MCI studies, it was 4.1 for non-practice effects group vs 0.2 for the practice effects group. AD participants without practice effects progressed 0.9 points faster than those with practice effects over a period of 2 years in CDR-SB; whereas for MCI participants, the difference is 0.7 points. The sample sizes can be different by over 35% when estimated based on participants with/without practice effects. Conclusion Practice effects were prevalent and robust in persons with AD or MCI and affected the cognitive progression and sample size estimation. Planning of future AD or MCI clinical trials should account for practice effects to avoid underpower or considers target trials or stratification analysis based on practice effects. © 2020 Wang et al. This is an open access article distributed under the terms of the Creative Commons Attribution License, which permits unrestricted use, distribution, and reproduction in any medium, provided the original author and source are credited.

Document Type: Article
Publication Stage: Final
Source: Scopus