1
|
Kitaba NT, Østergaard TM, Lønnebotn M, Accordini S, Real FG, Malinovschi A, Oudin A, Benediktsdottir B, González FJC, Gómez LP, Holm M, Jõgi NO, Dharmage SC, Skulstad SM, Schlünssen V, Svanes C, Holloway JW. Father's adolescent body silhouette is associated with offspring asthma, lung function and BMI through DNA methylation. Commun Biol 2025; 8:796. [PMID: 40410506 PMCID: PMC12102279 DOI: 10.1038/s42003-025-08121-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2024] [Accepted: 04/23/2025] [Indexed: 05/25/2025] Open
Abstract
Boys' pubertal overweight associates with future offspring's asthma and low lung function. To identify how paternal overweight is associated with offspring's DNA methylation (DNAm), we conducted an epigenome-wide association study of father's body silhouette (FBS) at three timepoints (age 8, voice break and 30) and change in FBS between these times, with offspring DNAm, in the RHINESSA cohort (N = 339). We identified 2005 differentially methylated cytosine-phosphate-guanine (dmCpG) sites (FDR < 0.05), including dmCpGs associated with offspring asthma (119), lung function (178) and BMI (291). Voice break FBS associated with dmCpGs in loci including KCNJ10, FERMT1, NCK2 and WWP1. Change in FBS across sexual maturation associated with DNAm at loci including NOP10, TRRAP, EFHD1, MRPL17 and NORD59A;ATP5B and showed strong correlation in reduced gene expression in loci NAP1L5, ATP5B, ZNF695, ZNF600, VTRNA2-1, SOAT2 and AGPAT2. We identified 24 imprinted genes including: VTRNA2-1, BLCAP, WT1, NAP1L5 and PTPRN2. Identified pathways relate to lipid and glucose metabolism and adipogenesis. Father's overweight at puberty and during reproductive maturation was strongly associated with offspring DNA, suggesting a key role for epigenetic mechanisms in intergenerational transfer from father to offspring in humans. The results support an important vulnerability window in male puberty for future offspring health.
Collapse
Grants
- We thank all the study participants, fieldworkers and scientists in RHINESSA, Co-ordination of the RHINESSA study has received funding from the Research Council of Norway (Grants No. 274767, 214123, 228174, 230827 and 273838), ERC StG project BRuSH #804199, the European Union's Horizon 2020 research and innovation programme under grant agreement No. 633212 (the ALEC Study), the Bergen Medical Research Foundation, and the Western Norwegian Regional Health Authorities (Grants No. 912011, 911892 and 911631). Study centres have further received local funding from the following: Bergen: the above grants for study establishment and co-ordination, and, in addition, World University Network (REF and Sustainability grants), Norwegian Labour Inspection, and the Norwegian Asthma and Allergy Association. Albacete and Huelva: Sociedad Española de Patología Respiratoria (SEPAR) Fondo de Investigación Sanitaria (FIS PS09). Gøteborg, Umeå and Uppsala: the Swedish Heart and Lung Foundation, the Swedish Asthma and Allergy Association. Reykjavik: Iceland University. Melbourne: National Health and Medical Research Council (NHMRC) of Australia (research grants 299901 and 1021275). Tartu: the Estonian Research Council (Grant No. PUT562). Århus: The Danish Wood Foundation (Grant No. 444508795), the Danish Working Environment Authority (Grant No. 20150067134), Aarhus University (PhD scholarship).
Collapse
Affiliation(s)
- Negusse Tadesse Kitaba
- Human Development and Health, Faculty of Medicine, University of Southampton, Southampton, UK
| | - Toril Mørkve Østergaard
- Department of Global Public Health and Primary Care, Centre for International Health, University of Bergen, Bergen, Norway
| | - Marianne Lønnebotn
- Department of Health and Caring Sciences, Western Norway University of Applied Sciences, Bergen, Norway
| | - Simone Accordini
- Unit of Epidemiology and Medical Statistics, Department of Diagnostics and Public Health, University of Verona, Verona, Italy
| | | | - Andrei Malinovschi
- Department of Medical Sciences: Clinical Physiology, Uppsala University, Uppsala, Sweden
| | - Anna Oudin
- Section of Sustainable Health, Department of Public Health and Clinical Medicine, Umeå University, Umeå, Sweden
| | - Bryndis Benediktsdottir
- Department of Allergy, Respiratory Medicine and Sleep, Landspitali University Hospital, Reykjavik, Iceland Faculty of Medicine, University of Iceland, Landspitali, Iceland
| | | | | | - Mathias Holm
- Occupational and Environmental Medicine, School of Public Health and Community Medicine, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Nils Oskar Jõgi
- Department of Medical Sciences: Clinical Physiology, Uppsala University, Uppsala, Sweden
| | - Shyamali C Dharmage
- Centre for Epidemiology and Biostatistics, Melbourne School of Population and Global Health, University of Melbourne, Melbourne, Australia
| | - Svein Magne Skulstad
- Department of Global Public Health and Primary Care, Centre for International Health, University of Bergen, Bergen, Norway
| | - Vivi Schlünssen
- Department of Public Health, Research Unit for Environment, Work and Health, Danish Ramazzini Centre, Aarhus University Denmark, Aarhus, Denmark
| | - Cecilie Svanes
- Department of Global Public Health and Primary Care, Centre for International Health, University of Bergen, Bergen, Norway.
- Department of Occupational Medicine, Haukeland University Hospital, Bergen, Norway.
| | - John W Holloway
- Human Development and Health, Faculty of Medicine, University of Southampton, Southampton, UK
- NIHR Southampton Biomedical Research Centre, University Hospitals Southampton, Southampton, UK
| |
Collapse
|
2
|
Reus LM, Boltz T, Francia M, Bot M, Ramesh N, Koromina M, Pijnenburg YAL, den Braber A, van der Flier WM, Visser PJ, van der Lee SJ, Tijms BM, Teunissen CE, Loohuis LO, Ophoff RA. Quantitative trait loci mapping of circulating metabolites in cerebrospinal fluid to uncover biological mechanisms involved in brain-related phenotypes. Mol Psychiatry 2025:10.1038/s41380-025-02934-0. [PMID: 40021830 DOI: 10.1038/s41380-025-02934-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/07/2023] [Revised: 12/16/2024] [Accepted: 02/12/2025] [Indexed: 03/03/2025]
Abstract
Genomic studies of molecular traits have provided mechanistic insights into complex disease, though these lag behind for brain-related traits due to the inaccessibility of brain tissue. We leveraged cerebrospinal fluid (CSF) to study neurobiological mechanisms in vivo, measuring 5543 CSF metabolites, the largest panel in CSF to date, in 977 individuals of European ancestry. Individuals originated from two separate cohorts including cognitively healthy subjects (n = 490) and a well-characterized memory clinic sample, the Amsterdam Dementia Cohort (ADC, n = 487). We performed metabolite quantitative trait loci (mQTL) mapping on CSF metabolomics and found 126 significant mQTLs, representing 65 unique CSF metabolites across 51 independent loci. To better understand the role of CSF mQTLs in brain-related disorders we integrated our CSF mQTL results with pre-existing summary statistics on brain traits, identifying 34 genetic associations between CSF metabolites and brain traits. Over 90% of significant mQTLs demonstrated colocalized associations with brain-specific gene expression, unveiling potential neurobiological pathways.
Collapse
Affiliation(s)
- Lianne M Reus
- Center for Neurobehavioral Genetics, Semel Institute for Neuroscience and Human Behavior, University of California, Los Angeles, CA, USA.
- Alzheimer Center Amsterdam, Neurology, Vrije Universiteit Amsterdam, Amsterdam UMC location VUmc, Amsterdam, The Netherlands.
- Amsterdam Neuroscience, Neurodegeneration, Vrije Universiteit Amsterdam, Amsterdam UMC location VUmc, Amsterdam, The Netherlands.
| | - Toni Boltz
- Center for Neurobehavioral Genetics, Semel Institute for Neuroscience and Human Behavior, University of California, Los Angeles, CA, USA.
- Department of Human Genetics, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA.
| | - Marcelo Francia
- Center for Neurobehavioral Genetics, Semel Institute for Neuroscience and Human Behavior, University of California, Los Angeles, CA, USA
| | - Merel Bot
- Center for Neurobehavioral Genetics, Semel Institute for Neuroscience and Human Behavior, University of California, Los Angeles, CA, USA
| | - Naren Ramesh
- Center for Neurobehavioral Genetics, Semel Institute for Neuroscience and Human Behavior, University of California, Los Angeles, CA, USA
| | - Maria Koromina
- Charles Bronfman Institute for Personalized Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, New York, NY, USA
| | - Yolande A L Pijnenburg
- Alzheimer Center Amsterdam, Neurology, Vrije Universiteit Amsterdam, Amsterdam UMC location VUmc, Amsterdam, The Netherlands
- Amsterdam Neuroscience, Neurodegeneration, Vrije Universiteit Amsterdam, Amsterdam UMC location VUmc, Amsterdam, The Netherlands
| | - Anouk den Braber
- Alzheimer Center Amsterdam, Neurology, Vrije Universiteit Amsterdam, Amsterdam UMC location VUmc, Amsterdam, The Netherlands
- Amsterdam Neuroscience, Neurodegeneration, Vrije Universiteit Amsterdam, Amsterdam UMC location VUmc, Amsterdam, The Netherlands
- Department of Biological Psychology, Amsterdam Public Health Research Institute, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Wiesje M van der Flier
- Alzheimer Center Amsterdam, Neurology, Vrije Universiteit Amsterdam, Amsterdam UMC location VUmc, Amsterdam, The Netherlands
- Amsterdam Neuroscience, Neurodegeneration, Vrije Universiteit Amsterdam, Amsterdam UMC location VUmc, Amsterdam, The Netherlands
- Epidemiology and Data Science, Vrije Universiteit Amsterdam, Amsterdam UMC location VUmc, Amsterdam, The Netherlands
| | - Pieter Jelle Visser
- Alzheimer Center Amsterdam, Neurology, Vrije Universiteit Amsterdam, Amsterdam UMC location VUmc, Amsterdam, The Netherlands
- Amsterdam Neuroscience, Neurodegeneration, Vrije Universiteit Amsterdam, Amsterdam UMC location VUmc, Amsterdam, The Netherlands
- Department of Psychiatry, Maastricht University, Maastricht, The Netherlands
- Department of Neurobiology, Care Sciences and Society, Division of Neurogeriatrics, Karolinska Institutet, Stockholm, Sweden
| | - Sven J van der Lee
- Alzheimer Center Amsterdam, Neurology, Vrije Universiteit Amsterdam, Amsterdam UMC location VUmc, Amsterdam, The Netherlands
- Amsterdam Neuroscience, Neurodegeneration, Vrije Universiteit Amsterdam, Amsterdam UMC location VUmc, Amsterdam, The Netherlands
- Section Genomics of Neurodegenerative Diseases and Aging, Department of Human Genetics, Vrije Universiteit Amsterdam, Amsterdam UMC, Amsterdam, the Netherlands
| | - Betty M Tijms
- Alzheimer Center Amsterdam, Neurology, Vrije Universiteit Amsterdam, Amsterdam UMC location VUmc, Amsterdam, The Netherlands
- Amsterdam Neuroscience, Neurodegeneration, Vrije Universiteit Amsterdam, Amsterdam UMC location VUmc, Amsterdam, The Netherlands
| | - Charlotte E Teunissen
- Neurochemistry Lab, Department of Laboratory Medicine, Amsterdam Neuroscience, Neurodegeneration, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Loes Olde Loohuis
- Center for Neurobehavioral Genetics, Semel Institute for Neuroscience and Human Behavior, University of California, Los Angeles, CA, USA
- Department of Human Genetics, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
- Department of Computational Medicine, University of California, Los Angeles, CA, USA
| | - Roel A Ophoff
- Center for Neurobehavioral Genetics, Semel Institute for Neuroscience and Human Behavior, University of California, Los Angeles, CA, USA.
- Department of Human Genetics, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA.
- Department of Psychiatry, Erasmus University Medical Center, Rotterdam, The Netherlands.
| |
Collapse
|
3
|
Gal Z, Torok D, Gonda X, Eszlari N, Anderson IM, Deakin B, Petschner P, Juhasz G, Bagdy G. New Evidence for the Role of the Blood-Brain Barrier and Inflammation in Stress-Associated Depression: A Gene-Environment Analysis Covering 19,296 Genes in 109,360 Humans. Int J Mol Sci 2024; 25:11332. [PMID: 39457114 PMCID: PMC11508422 DOI: 10.3390/ijms252011332] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2024] [Revised: 10/16/2024] [Accepted: 10/18/2024] [Indexed: 10/28/2024] Open
Abstract
Mounting evidence supports the key role of the disrupted integrity of the blood-brain barrier (BBB) in stress- and inflammation-associated depression. We assumed that variations in genes regulating the expression and coding proteins constructing and maintaining this barrier, along with those involved in inflammation, have a predisposing or protecting role in the development of depressive symptoms after experiencing severe stress. To prove this, genome-by-environment (GxE) interaction analyses were conducted on 6.26 M SNPS covering 19,296 genes on PHQ9 depression in interaction with adult traumatic events scores in the UK Biobank (n = 109,360) in a hypothesis-free setup. Among the 63 genes that were significant in stress-connected depression, 17 were associated with BBB, 23 with inflammatory processes, and 4 with neuroticism. Compared to all genes, the enrichment of significant BBB-associated hits was 3.82, and those of inflammation-associated hits were 1.59. Besides some sex differences, CSMD1 and PTPRD, encoding proteins taking part in BBB integrity, were the most significant hits in both males and females. In conclusion, the identified risk genes and their encoded proteins could provide biomarkers or new drug targets to promote BBB integrity and thus prevent or decrease stress- and inflammation-associated depressive symptoms, and possibly infection, e.g., COVID-19-associated mental and neurological symptoms.
Collapse
Affiliation(s)
- Zsofia Gal
- Department of Pharmacodynamics, Faculty of Pharmaceutical Sciences, Semmelweis University, 1089 Budapest, Hungary; (Z.G.); (D.T.); (N.E.); (P.P.); (G.J.)
- NAP3.0-SE Neuropsychopharmacology Research Group, Hungarian Brain Research Program, Semmelweis University, 1089 Budapest, Hungary;
| | - Dora Torok
- Department of Pharmacodynamics, Faculty of Pharmaceutical Sciences, Semmelweis University, 1089 Budapest, Hungary; (Z.G.); (D.T.); (N.E.); (P.P.); (G.J.)
- NAP3.0-SE Neuropsychopharmacology Research Group, Hungarian Brain Research Program, Semmelweis University, 1089 Budapest, Hungary;
| | - Xenia Gonda
- NAP3.0-SE Neuropsychopharmacology Research Group, Hungarian Brain Research Program, Semmelweis University, 1089 Budapest, Hungary;
- Department of Psychiatry and Psychotherapy, Semmelweis University, 1083 Budapest, Hungary
| | - Nora Eszlari
- Department of Pharmacodynamics, Faculty of Pharmaceutical Sciences, Semmelweis University, 1089 Budapest, Hungary; (Z.G.); (D.T.); (N.E.); (P.P.); (G.J.)
- NAP3.0-SE Neuropsychopharmacology Research Group, Hungarian Brain Research Program, Semmelweis University, 1089 Budapest, Hungary;
| | - Ian Muir Anderson
- Division of Neuroscience, School of Biological Sciences, Faculty of Biology, Medicine and Health, The University of Manchester and Manchester Academic Health Sciences Centre, Manchester M13 9NT, UK; (I.M.A.); (B.D.)
| | - Bill Deakin
- Division of Neuroscience, School of Biological Sciences, Faculty of Biology, Medicine and Health, The University of Manchester and Manchester Academic Health Sciences Centre, Manchester M13 9NT, UK; (I.M.A.); (B.D.)
| | - Peter Petschner
- Department of Pharmacodynamics, Faculty of Pharmaceutical Sciences, Semmelweis University, 1089 Budapest, Hungary; (Z.G.); (D.T.); (N.E.); (P.P.); (G.J.)
- NAP3.0-SE Neuropsychopharmacology Research Group, Hungarian Brain Research Program, Semmelweis University, 1089 Budapest, Hungary;
- Bioinformatics Center, Institute of Chemical Research, Kyoto University, Uji, Kyoto 611-0011, Japan
- Research Unit for Realization of Sustainable Society, Kyoto University, Gokasho, Uji, Kyoto 611-0011, Japan
| | - Gabriella Juhasz
- Department of Pharmacodynamics, Faculty of Pharmaceutical Sciences, Semmelweis University, 1089 Budapest, Hungary; (Z.G.); (D.T.); (N.E.); (P.P.); (G.J.)
- NAP3.0-SE Neuropsychopharmacology Research Group, Hungarian Brain Research Program, Semmelweis University, 1089 Budapest, Hungary;
| | - Gyorgy Bagdy
- Department of Pharmacodynamics, Faculty of Pharmaceutical Sciences, Semmelweis University, 1089 Budapest, Hungary; (Z.G.); (D.T.); (N.E.); (P.P.); (G.J.)
- NAP3.0-SE Neuropsychopharmacology Research Group, Hungarian Brain Research Program, Semmelweis University, 1089 Budapest, Hungary;
| |
Collapse
|
4
|
Reus LM, Boltz T, Francia M, Bot M, Ramesh N, Koromina M, Pijnenburg YAL, den Braber A, van der Flier WM, Visser PJ, van der Lee SJ, Tijms BM, Teunissen CE, Loohuis LO, Ophoff RA. Quantitative trait loci mapping of circulating metabolites in cerebrospinal fluid to uncover biological mechanisms involved in brain-related phenotypes. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.09.26.559021. [PMID: 37808647 PMCID: PMC10557608 DOI: 10.1101/2023.09.26.559021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/10/2023]
Abstract
Genomic studies of molecular traits have provided mechanistic insights into complex disease, though these lag behind for brain-related traits due to the inaccessibility of brain tissue. We leveraged cerebrospinal fluid (CSF) to study neurobiological mechanisms in vivo , measuring 5,543 CSF metabolites, the largest panel in CSF to date, in 977 individuals of European ancestry. Individuals originated from two separate cohorts including cognitively healthy subjects (n=490) and a well-characterized memory clinic sample, the Amsterdam Dementia Cohort (ADC, n=487). We performed metabolite quantitative trait loci (mQTL) mapping on CSF metabolomics and found 126 significant mQTLs, representing 65 unique CSF metabolites across 51 independent loci. To better understand the role of CSF mQTLs in brain-related disorders, we performed a metabolome-wide association study (MWAS), identifying 40 associations between CSF metabolites and brain traits. Similarly, over 90% of significant mQTLs demonstrated colocalized associations with brain-specific gene expression, unveiling potential neurobiological pathways.
Collapse
|
5
|
Luykx JJ, Gonzalez-Diaz JM, Guu TW, van der Horst MZ, van Dellen E, Boks MP, Guloksuz S, DeLisi LE, Sommer IE, Cummins R, Shiers D, Lee J, Every-Palmer S, Mhalla A, Chadly Z, Chan SKW, Cotes RO, Takahashi S, Benros ME, Wagner E, Correll CU, Hasan A, Siskind D, Endres D, MacCabe J, Tiihonen J. An international research agenda for clozapine-resistant schizophrenia. Lancet Psychiatry 2023:S2215-0366(23)00109-8. [PMID: 37329895 DOI: 10.1016/s2215-0366(23)00109-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/20/2022] [Revised: 02/24/2023] [Accepted: 03/17/2023] [Indexed: 06/19/2023]
Abstract
Treatment-resistant symptoms occur in about a third of patients with schizophrenia and are associated with a substantial reduction in their quality of life. The development of new treatment options for clozapine-resistant schizophrenia constitutes a crucial, unmet need in psychiatry. Additionally, an overview of past and possible future research avenues to optimise the early detection, diagnosis, and management of clozapine-resistant schizophrenia is unavailable. In this Health Policy, we discuss the ongoing challenges associated with clozapine-resistant schizophrenia faced by patients and health-care providers worldwide to improve the understanding of this condition. We then revisit several clozapine guidelines, the diagnostic tests and treatment options for clozapine-resistant schizophrenia, and currently applied research approaches in clozapine-resistant schizophrenia. We also suggest methodologies and targets for future research, divided into innovative nosology-oriented field trials (eg, examining dimensional symptom staging), translational approaches (eg, genetics), epidemiological research (eg, real-world studies), and interventional studies (eg, non-traditional trial designs incorporating lived experiences and caregivers' perspectives). Finally, we note that low-income and middle-income countries are under-represented in studies on clozapine-resistant schizophrenia and propose an agenda to guide multinational research on the cause and treatment of clozapine-resistant schizophrenia. We hope that this research agenda will empower better global representation of patients living with clozapine-resistant schizophrenia and ultimately improve their functional outcomes and quality of life.
Collapse
Affiliation(s)
- Jurjen J Luykx
- Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience, Maastricht University Medical Centre, Maastricht, Netherlands; Department of Psychiatry, UMC Utrecht Brain Center, University Medical Center Utrecht, Utrecht University, Utrecht, Netherlands; GGNet Mental Health, Warnsveld, Netherlands.
| | - Jairo M Gonzalez-Diaz
- Barcelona Clínic Schizophrenia Unit, Neurosciences Institute, Hospital Clinic, Universitat de Barcelona, Barcelona, Spain; UR Center for Mental Health, School of Medicine and Health Sciences, Universidad del Rosario, Bogotá, Colombia; Clínica Nuestra Señora de la Paz, Orden Hospitalaria de San Juan de Dios, Bogotá, Colombia
| | - Ta-Wei Guu
- Department of Old Age Psychiatry, Institute of Psychiatry Psychology and Neuroscience, King's College London, London, UK; Division of Psychiatry, Department of Internal Medicine, China Medical University Beigang Hospital, Yunlin, Taiwan
| | - Marte Z van der Horst
- Department of Psychiatry, UMC Utrecht Brain Center, University Medical Center Utrecht, Utrecht University, Utrecht, Netherlands; GGNet Mental Health, Warnsveld, Netherlands
| | - Edwin van Dellen
- Department of Psychiatry, UMC Utrecht Brain Center, University Medical Center Utrecht, Utrecht University, Utrecht, Netherlands; Department of Intensive Care Medicine, UMC Utrecht Brain Center, University Medical Center Utrecht, Utrecht University, Utrecht, Netherlands; Department of Neurology, UZ Brussel and Vrije Universiteit Brussel, Jette, Belgium
| | - Marco P Boks
- Department of Psychiatry, UMC Utrecht Brain Center, University Medical Center Utrecht, Utrecht University, Utrecht, Netherlands
| | - Sinan Guloksuz
- Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience, Maastricht University Medical Centre, Maastricht, Netherlands; Department of Psychiatry, Yale University School of Medicine, New Haven, CT, USA
| | - Lynn E DeLisi
- Department of Psychiatry, Cambridge Health Alliance, Harvard Medical School, Cambridge, MA, USA
| | - Iris E Sommer
- Department of Biomedical Sciences of Cells and Systems, University of Groningen, University Medical Center Groningen, Groningen, Netherlands
| | | | - David Shiers
- Psychosis Research Unit, Greater Manchester Mental Health NHS Trust, Manchester, UK
| | - Jimmy Lee
- Department of Psychosis, Institute of Mental Health, Singapore; Neuroscience and Mental Health, Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore
| | - Susanna Every-Palmer
- Department of Psychological Medicine, University of Otago Wellington, Wellington, New Zealand
| | - Ahmed Mhalla
- Department of Psychiatry, Fattouma Bourguiba Hospital, Faculty of Medicine of Monastir, University of Monastir, Monastir, Tunisia
| | - Zohra Chadly
- Department of Pharmacology, Fattouma Bourguiba Hospital, Faculty of Medicine of Monastir, University of Monastir, Monastir, Tunisia
| | - Sherry K W Chan
- Department of Psychiatry, School of Clinical Medicine, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong Special Administrative Region, China; State Key Laboratory of Brain and Cognitive Sciences, The University of Hong Kong, Hong Kong Special Administrative Region, China
| | - Robert O Cotes
- Department of Psychiatry and Behavioral Sciences, Emory University School of Medicine, Atlanta, GA, USA
| | - Shun Takahashi
- Department of Psychiatry, Osaka University Graduate School of Medicine, Suita, Japan; Graduate School of Rehabilitation Science, Osaka Metropolitan University, Habikino, Japan; Clinical Research and Education Center, Asakayama General Hospital, Sakai, Japan; Department of Neuropsychiatry, Wakayama Medical University, Wakayama, Japan
| | - Michael E Benros
- Biological and Precision Psychiatry, Copenhagen Research Center for Mental Health, Mental Health Center Copenhagen, Copenhagen University Hospital, Copenhagen, Denmark; Department of Immunology and Microbiology, Faculty of Health and Medical Science, University of Copenhagen, Copenhagen, Denmark
| | - Elias Wagner
- Department of Psychiatry and Psychotherapy, University Hospital, LMU Munich, Munich, Germany
| | - Christoph U Correll
- Department of Child and Adolescent Psychiatry, Charité Universitaetsmedizin Berlin, Berlin, Germany; Department of Psychiatry and Molecular Medicine, Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Hempstead, NY, USA; Department of Psychiatry, The Zucker Hillside Hospital, Northwell Health, Glen Oaks, NY, USA
| | - Alkomiet Hasan
- Department of Psychiatry, Psychotherapy and Psychosomatics, University of Augsburg, Medical Faculty, Augsburg, Germany
| | - Dan Siskind
- Faculty of Medicine, University of Queensland, Brisbane, QLD, Australia; Metro South Addiction and Mental Health Service, Brisbane, QLD, Australia
| | - Dominique Endres
- Department of Psychiatry and Psychotherapy, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - James MacCabe
- Department of Psychosis Studies, Institute of Psychiatry Psychology and Neuroscience, King's College London, London, UK
| | - Jari Tiihonen
- Department of Forensic Psychiatry, University of Eastern Finland, Niuvanniemi Hospital, Kuopio, Finland; Department of Clinical Neuroscience, Karolinska Institutet, and Center for Psychiatry Research, Stockholm City Council, Stockholm, Sweden
| |
Collapse
|
6
|
Wang X, Chen X, Liu Y, Huang S, Ding J, Wang B, Dong P, Sun Z, Chen L. CSMD1 suppresses cancer progression by inhibiting proliferation, epithelial-mesenchymal transition, chemotherapy-resistance and inducing immunosuppression in esophageal squamous cell carcinoma. Exp Cell Res 2022; 417:113220. [PMID: 35623420 DOI: 10.1016/j.yexcr.2022.113220] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2022] [Revised: 05/15/2022] [Accepted: 05/19/2022] [Indexed: 11/17/2022]
Abstract
Human CUB and Sushi multiple domains (CSMD1) is considered a crucial role in cancer progression, but the specific function in esophageal squamous cell carcinoma (ESCC) is not clear. Understanding the role of CSMD1 in ESCC progression may lead to a novel strategy for ESCC treatment. Here, we found that both CSMD1 mRNA and protein levels were downregulated in ESCC tissues. Reduced CSMD1 expression was correlated with a poor prognosis in ESCC patients. CSMD1 expression inhibited proliferation, migration and invasion in ESCC cell lines in vitro. CSMD1 deficiency in established xenografted tumors increases tumor size and weight. We further found that CSMD1-overexpression cells are more sensitive to chemotherapy. Moreover, we addressed the role of CSMD1 in the CD8+ T cell immune response. An in vitro killing assay showed that the cytotoxicity of CD8+ T cells was inhibited in CSMD1-overexpression tumor cells. In vivo, in CSMD1 deficiency tumor-bearing mice activation and expansion of CD8+ T cells were increased. Further investigation showed that CSMD1 expression on tumor cells was positively correlated with CD8+ T cells infiltration and cytokines secretion. These findings highlight that CSMD1 is a tumor suppressor gene in ESCC patients and a positive regulator of CD8+ T cells expansion and activation, and could increase cytokines secretion, indicating that tumor cell-associated CSMD1 might be a target for ESCC.
Collapse
Affiliation(s)
- Xing Wang
- Translational Medicine Center, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, 201620, Shanghai, China
| | - Xinwei Chen
- Department of Otolaryngology: Head and Neck Surgery, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200080, China
| | - Yuanyuan Liu
- Department of Otolaryngology: Head and Neck Surgery, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200080, China
| | - Shan Huang
- National Engineering Research Center for Nanomedicine, Key Laboratory of Molecular Biophysics of Ministry of Education, College of Life Science and Technology, Tongji Hospital, Huazhong University of Science and Technology, 430074, Wuhan, China
| | - Jian Ding
- Department of Otolaryngology: Head and Neck Surgery, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200080, China
| | - Baoxin Wang
- Department of Otolaryngology: Head and Neck Surgery, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200080, China
| | - Pin Dong
- Department of Otolaryngology: Head and Neck Surgery, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200080, China
| | - Zhenfeng Sun
- Department of Otolaryngology: Head and Neck Surgery, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200080, China
| | - Lixiao Chen
- Department of Otolaryngology: Head and Neck Surgery, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200080, China.
| |
Collapse
|
7
|
Hu J, Johnson TA, Zhang H, Cheng HW. The Microbiota-Gut-Brain Axis: Gut Microbiota Modulates Conspecific Aggression in Diversely Selected Laying Hens. Microorganisms 2022; 10:microorganisms10061081. [PMID: 35744601 PMCID: PMC9230770 DOI: 10.3390/microorganisms10061081] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2022] [Revised: 05/18/2022] [Accepted: 05/19/2022] [Indexed: 12/22/2022] Open
Abstract
The gut microbiota plays an important role in regulating brain function, influencing psychological and emotional stability. The correlations between conspecific aggression, gut microbiota, and physiological homeostasis were further studied in inbred laying chicken lines, 63 and 72, which were diversely selected for Marek’s disease, and they also behave differently in aggression. Ten sixty-week-old hens from each line were sampled for blood, brain, and cecal content. Neurotransmitters, cytokines, corticosterone, and heterophil/lymphocyte ratios were determined. Cecal microbiota compositions were determined by bacterial 16s rRNA sequencing, and functional predictions were performed. Our data showed that the central serotonin and tryptophan levels were higher in line 63 compared to line 72 (p < 0.05). Plasma corticosterone, heterophil/lymphocyte ratios, and central norepinephrine were lower in line 63 (p < 0.05). The level of tumor necrosis factor α tended to be higher in line 63. Faecalibacterium, Oscillibacter, Butyricicoccus, and Bacteriodes were enriched in line 63 birds, while Clostridiales vadin BB60, Alistipes, Mollicutes RF39 were dominated in line 72. From the predicted bacterial functional genes, the kynurenine pathway was upregulated in line 72. These results suggested a functional linkage of the line differences in serotonergic activity, stress response, innate immunity, and gut microbiota populations.
Collapse
Affiliation(s)
- Jiaying Hu
- Department of Animal Sciences, Purdue University, West Lafayette, IN 47907, USA;
| | - Timothy A. Johnson
- Department of Animal Sciences, Purdue University, West Lafayette, IN 47907, USA;
- Correspondence: (T.A.J.); (H.-W.C.)
| | - Huanmin Zhang
- USDA-Agricultural Research Service, Avian Disease and Oncology Laboratory, East Lansing, MI 48823, USA;
| | - Heng-Wei Cheng
- USDA-Agricultural Research Service, Livestock Behavior Research Unit, West Lafayette, IN 47907, USA
- Correspondence: (T.A.J.); (H.-W.C.)
| |
Collapse
|
8
|
Okhuijsen-Pfeifer C, van der Horst MZ, Bousman CA, Lin B, van Eijk KR, Ripke S, Ayhan Y, Babaoglu MO, Bak M, Alink W, van Beek H, Beld E, Bouhuis A, Edlinger M, Erdogan IM, Ertuğrul A, Yoca G, Everall IP, Görlitz T, Grootens KP, Gutwinski S, Hallikainen T, Jeger-Land E, de Koning M, Lähteenvuo M, Legge SE, Leucht S, Morgenroth C, Müderrisoğlu A, Narang A, Pantelis C, Pardiñas AF, Oviedo-Salcedo T, Schneider-Thoma J, Schreiter S, Repo-Tiihonen E, Tuppurainen H, Veereschild M, Veerman S, de Vos M, Wagner E, Cohen D, Bogers JPAM, Walters JTR, Yağcıoğlu AEA, Tiihonen J, Hasan A, Luykx JJ. Genome-wide association analyses of symptom severity among clozapine-treated patients with schizophrenia spectrum disorders. Transl Psychiatry 2022; 12:145. [PMID: 35393395 PMCID: PMC8989876 DOI: 10.1038/s41398-022-01884-3] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/27/2021] [Revised: 02/24/2022] [Accepted: 03/07/2022] [Indexed: 12/26/2022] Open
Abstract
Clozapine is the most effective antipsychotic for patients with treatment-resistant schizophrenia. However, response is highly variable and possible genetic underpinnings of this variability remain unknown. Here, we performed polygenic risk score (PRS) analyses to estimate the amount of variance in symptom severity among clozapine-treated patients explained by PRSs (R2) and examined the association between symptom severity and genotype-predicted CYP1A2, CYP2D6, and CYP2C19 enzyme activity. Genome-wide association (GWA) analyses were performed to explore loci associated with symptom severity. A multicenter cohort of 804 patients (after quality control N = 684) with schizophrenia spectrum disorder treated with clozapine were cross-sectionally assessed using the Positive and Negative Syndrome Scale and/or the Clinical Global Impression-Severity (CGI-S) scale. GWA and PRS regression analyses were conducted. Genotype-predicted CYP1A2, CYP2D6, and CYP2C19 enzyme activities were calculated. Schizophrenia-PRS was most significantly and positively associated with low symptom severity (p = 1.03 × 10-3; R2 = 1.85). Cross-disorder-PRS was also positively associated with lower CGI-S score (p = 0.01; R2 = 0.81). Compared to the lowest tertile, patients in the highest schizophrenia-PRS tertile had 1.94 times (p = 6.84×10-4) increased probability of low symptom severity. Higher genotype-predicted CYP2C19 enzyme activity was independently associated with lower symptom severity (p = 8.44×10-3). While no locus surpassed the genome-wide significance threshold, rs1923778 within NFIB showed a suggestive association (p = 3.78×10-7) with symptom severity. We show that high schizophrenia-PRS and genotype-predicted CYP2C19 enzyme activity are independently associated with lower symptom severity among individuals treated with clozapine. Our findings open avenues for future pharmacogenomic projects investigating the potential of PRS and genotype-predicted CYP-activity in schizophrenia.
Collapse
Affiliation(s)
- C Okhuijsen-Pfeifer
- Department of Psychiatry, University Medical Center Utrecht, Utrecht University, Brain Center, Utrecht, The Netherlands
- Department of Translational Neuroscience, University Medical Center Utrecht, Utrecht University, Brain Center, Utrecht, The Netherlands
| | - M Z van der Horst
- Department of Psychiatry, University Medical Center Utrecht, Utrecht University, Brain Center, Utrecht, The Netherlands
- Department of Translational Neuroscience, University Medical Center Utrecht, Utrecht University, Brain Center, Utrecht, The Netherlands
- GGNet Mental Health, Warnsveld, The Netherlands
| | - C A Bousman
- Department of Medical Genetics, University of Calgary, Calgary, Canada
- Department of Psychiatry, University of Calgary, Calgary, Canada
- Department of Physiology & Pharmacology, University of Calgary, Calgary, Canada
- Department of Psychiatry, University of Melbourne, Melbourne Neuropsychiatry Centre, Melbourne, Australia
| | - B Lin
- Department of Psychiatry, University Medical Center Utrecht, Utrecht University, Brain Center, Utrecht, The Netherlands
- Department of Translational Neuroscience, University Medical Center Utrecht, Utrecht University, Brain Center, Utrecht, The Netherlands
| | - K R van Eijk
- Department of Translational Neuroscience, University Medical Center Utrecht, Utrecht University, Brain Center, Utrecht, The Netherlands
| | - S Ripke
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Department of Psychiatry and Psychotherapy, Berlin, Germany
| | - Y Ayhan
- Department of Psychiatry, Faculty of Medicine, Hacettepe University, Ankara, Turkey
| | - M O Babaoglu
- Department of Pharmacology, Faculty of Medicine, Hacettepe University, Ankara, Turkey
| | - M Bak
- Department of Psychiatry and Neuropsychology, Maastricht University, Maastricht, The Netherlands
- Mondriaan, Mental Health Institute, Maastricht, The Netherlands
| | - W Alink
- Multicomplexe Zorg, Pro Persona, Wolfheze, The Netherlands
| | - H van Beek
- Clinical Recovery Clinic, Mental Health Services Rivierduinen, Leiden, The Netherlands
| | - E Beld
- Mental Health Organization North-Holland North location Den Helder, Den Helder, The Netherlands
| | - A Bouhuis
- Program for early psychosis & severe mental illness, Pro Persona Mental Healthcare, Wolfheze, The Netherlands
| | - M Edlinger
- Department of Psychiatry, Psychotherapy and Psychosomatics, Division for Psychiatry I, Medical University Innsbruck, Innsbruck, Austria
| | - I M Erdogan
- Department of Psychiatry, Faculty of Medicine, Hacettepe University, Ankara, Turkey
| | - A Ertuğrul
- Department of Psychiatry, Faculty of Medicine, Hacettepe University, Ankara, Turkey
| | - G Yoca
- Department of Psychiatry, Faculty of Medicine, Hacettepe University, Ankara, Turkey
- Şarkışla State Hospital, Ministry of Health, Sivas, Turkey
| | - I P Everall
- Department of Psychiatry, University of Melbourne, Melbourne Neuropsychiatry Centre, Melbourne, Australia
- Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, United Kingdom
| | - T Görlitz
- Department of Psychiatry and Psychotherapy, University Hospital, LMU Munich, Munich, Germany
- Department of Psychiatry, Psychotherapy and Psychosomatics, Medical Faculty University Augsburg, Bezirkskrankenhaus Augsburg, Augsburg, Germany
| | - K P Grootens
- Reinier van Arkel, s-Hertogenbosch, The Netherlands
- Unit for Clinical Psychopharmacology and Neuropsychiatry, Radboud University Medical Centre, Nijmegen, The Netherlands
| | - S Gutwinski
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Department of Psychiatry and Psychotherapy, Berlin, Germany
| | - T Hallikainen
- Department of Forensic Psychiatry, University of Kuopio, Niuvanniemi Hospital, Kuopio, Finland
| | - E Jeger-Land
- Arkin, Institute for Mental Health, Amsterdam, The Netherlands
| | - M de Koning
- Arkin, Institute for Mental Health, Amsterdam, The Netherlands
- Amsterdam UMC, University of Amsterdam, Department of Psychiatry, Amsterdam, The Netherlands
| | - M Lähteenvuo
- Department of Forensic Psychiatry, University of Kuopio, Niuvanniemi Hospital, Kuopio, Finland
| | - S E Legge
- Division of Psychological Medicine and Clinical Neurosciences, MRC Centre for Neuropsychiatric Genetics and Genomics, School of Medicine, Cardiff University, Cardiff, United Kingdom
| | - S Leucht
- Department of Psychiatry and Psychotherapy, School of Medicine, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
| | - C Morgenroth
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Department of Psychiatry and Psychotherapy, Berlin, Germany
| | - A Müderrisoğlu
- Department of Pharmacology, Faculty of Medicine, Kırıkkale University, Kırıkkale, Turkey
| | - A Narang
- Department of Medical Genetics, University of Calgary, Calgary, Canada
| | - C Pantelis
- Department of Psychiatry, University of Melbourne, Melbourne Neuropsychiatry Centre, Melbourne, Australia
| | - A F Pardiñas
- Division of Psychological Medicine and Clinical Neurosciences, MRC Centre for Neuropsychiatric Genetics and Genomics, School of Medicine, Cardiff University, Cardiff, United Kingdom
| | - T Oviedo-Salcedo
- Department of Psychiatry and Psychotherapy, University Hospital, LMU Munich, Munich, Germany
| | - J Schneider-Thoma
- Department of Psychiatry and Psychotherapy, School of Medicine, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
| | - S Schreiter
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Department of Psychiatry and Psychotherapy, Berlin, Germany
- Berlin Institute of Health (BIH), BIH Biomedical Innovation Academy, Berlin, Germany
| | - E Repo-Tiihonen
- Department of Forensic Psychiatry, University of Kuopio, Niuvanniemi Hospital, Kuopio, Finland
| | - H Tuppurainen
- Department of Forensic Psychiatry, University of Kuopio, Niuvanniemi Hospital, Kuopio, Finland
| | | | - S Veerman
- Mental Health Organization North-Holland North location Alkmaar, Alkmaar, The Netherlands
| | - M de Vos
- GGNet Mental Health, Warnsveld, The Netherlands
| | - E Wagner
- Department of Psychiatry and Psychotherapy, University Hospital, LMU Munich, Munich, Germany
| | - D Cohen
- Mental Health Organization North-Holland North location Heerhugowaard, Heerhugowaard, The Netherlands
| | - J P A M Bogers
- High Care Clinics, Mental Health Services Rivierduinen, Leiden, The Netherlands
| | - J T R Walters
- Division of Psychological Medicine and Clinical Neurosciences, MRC Centre for Neuropsychiatric Genetics and Genomics, School of Medicine, Cardiff University, Cardiff, United Kingdom
| | - A E Anil Yağcıoğlu
- Department of Psychiatry, Faculty of Medicine, Hacettepe University, Ankara, Turkey
| | - J Tiihonen
- Department of Forensic Psychiatry, University of Kuopio, Niuvanniemi Hospital, Kuopio, Finland
- Department of Clinical Neuroscience, Karolinska Institutet, Solna, Sweden
- Center for Psychiatric Research, Stockholm City Council, Stockholm, Sweden
| | - A Hasan
- Department of Psychiatry and Psychotherapy, University Hospital, LMU Munich, Munich, Germany
- Department of Psychiatry, Psychotherapy and Psychosomatics, Medical Faculty University Augsburg, Bezirkskrankenhaus Augsburg, Augsburg, Germany
| | - J J Luykx
- Department of Psychiatry, University Medical Center Utrecht, Utrecht University, Brain Center, Utrecht, The Netherlands.
- Department of Translational Neuroscience, University Medical Center Utrecht, Utrecht University, Brain Center, Utrecht, The Netherlands.
- GGNet Mental Health, Warnsveld, The Netherlands.
| |
Collapse
|
9
|
Costanzo F, Zanni G, Fucà E, Di Paola M, Barresi S, Travaglini L, Colafati GS, Gambardella A, Bellacchio E, Bertini E, Menghini D, Vicari S. Cerebellar Agenesis and Bilateral Polimicrogyria Associated with Rare Variants of CUB and Sushi Multiple Domains 1 Gene (CSMD1): A Longitudinal Neuropsychological and Neuroradiological Case Study. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2022; 19:ijerph19031224. [PMID: 35162247 PMCID: PMC8835405 DOI: 10.3390/ijerph19031224] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/11/2021] [Revised: 01/12/2022] [Accepted: 01/17/2022] [Indexed: 12/04/2022]
Abstract
Cerebellar agenesis is an extremely rare condition characterized by a near complete absence of the cerebellum. The pathogenesis and molecular basis remain mostly unknown. We report the neuroradiological, molecular, neuropsychological and behavioral characterization of a 5-year-old girl, with cerebellar agenesis associated with parietal and peri-Sylvian polymicrogyria, followed-up for 10 years at four time points. Whole exome sequencing identified two rare variants in CSMD1, a gene associated with neurocognitive and psychiatric alterations. Mild intellectual impairment, cerebellar ataxia and deficits in language, memory and executive functions, with relatively preserved adaptive and psychopathological domains, were initially showed. Phonological awareness and verbal memory declined at 11 years of age, and social and anxiety problems emerged. Adaptive and psychopathological characteristics dramatically worsened at 15 years. In summary, the developmental clinical outcome showed impairment in multiple cognitive functions in childhood, with a progressive decline in cognitive and adaptive abilities and the emergence of psychopathological symptoms in adolescence. The observed phenotype could be the result of a complex interplay between cerebellar abnormality, brain malformation and the relations with CSMD1 variants. These findings may provide insights into the developmental clinical outcomes of a co-occurrence between rare brain malformation and rare genetic variants associated to neurodevelopmental disorders.
Collapse
Affiliation(s)
- Floriana Costanzo
- Child and Adolescent Neuropsychiatry Unit, Department of Neurosciences, Bambino Gesù Children’s Hospital IRCCS, Via Ferdinando Baldelli 41, I-00146 Rome, Italy; (F.C.); (E.F.); (S.V.)
| | - Ginevra Zanni
- Unit of Neuromuscular and Neurodegenerative Disorders, Department of Neurosciences, Bambino Gesù Children’s Hospital, IRCCS, Viale di San Paolo 15, I-00146 Rome, Italy; (G.Z.); (L.T.); (E.B.)
| | - Elisa Fucà
- Child and Adolescent Neuropsychiatry Unit, Department of Neurosciences, Bambino Gesù Children’s Hospital IRCCS, Via Ferdinando Baldelli 41, I-00146 Rome, Italy; (F.C.); (E.F.); (S.V.)
| | - Margherita Di Paola
- Department of Clinical and Behavioral Neurology, IRCCS Santa Lucia Foundation, Via Ardeatina 306, I-00179 Rome, Italy;
- Department of Mental Health, King Faisal Specialist Hospital & Research Center, Riyadh 12713, Saudi Arabia
| | - Sabina Barresi
- Pathology Unit, Department of Laboratories, Bambino Gesù Children’s Hospital, IRCCS, Viale di San Paolo 15, I-00146 Rome, Italy;
| | - Lorena Travaglini
- Unit of Neuromuscular and Neurodegenerative Disorders, Department of Neurosciences, Bambino Gesù Children’s Hospital, IRCCS, Viale di San Paolo 15, I-00146 Rome, Italy; (G.Z.); (L.T.); (E.B.)
| | - Giovanna Stefania Colafati
- Oncological Neuroradiology Unit, Department of Imaging, Bambino Gesù Children’s Hospital, IRCCS, Piazza Sant’Onofrio 4, I-00100 Rome, Italy;
| | - Antonio Gambardella
- Institute of Neurology, University Magna Græcia, I-88100 Catanzaro, Italy;
- Institute of Molecular Bioimaging and Physiology, National Research Council, I-88100 Catanzaro, Italy
| | - Emanuele Bellacchio
- Genetics and Rare Diseases Research Division, Bambino Gesù Children’s Hospital, Viale di San Paolo 15, I-00146 Rome, Italy;
| | - Enrico Bertini
- Unit of Neuromuscular and Neurodegenerative Disorders, Department of Neurosciences, Bambino Gesù Children’s Hospital, IRCCS, Viale di San Paolo 15, I-00146 Rome, Italy; (G.Z.); (L.T.); (E.B.)
| | - Deny Menghini
- Child and Adolescent Neuropsychiatry Unit, Department of Neurosciences, Bambino Gesù Children’s Hospital IRCCS, Via Ferdinando Baldelli 41, I-00146 Rome, Italy; (F.C.); (E.F.); (S.V.)
- Correspondence: ; Tel.: +39-0668597091
| | - Stefano Vicari
- Child and Adolescent Neuropsychiatry Unit, Department of Neurosciences, Bambino Gesù Children’s Hospital IRCCS, Via Ferdinando Baldelli 41, I-00146 Rome, Italy; (F.C.); (E.F.); (S.V.)
- Department of Life Science and Public Health, Catholic University of the Sacred Heart, Largo Agostino Gemelli 1, I-00168 Rome, Italy
| |
Collapse
|
10
|
Ciobanu AM, Geza L, David IG, Popa DE, Buleandra M, Ciucu AA, Dehelean L. Actualities in immunological markers and electrochemical sensors for determination of dopamine and its metabolites in psychotic disorders (Review). Exp Ther Med 2021; 22:888. [PMID: 34194566 PMCID: PMC8237259 DOI: 10.3892/etm.2021.10320] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2021] [Accepted: 05/26/2021] [Indexed: 12/03/2022] Open
Abstract
Psychotic disorders represent a serious health concern. At this moment, anamnestic data, international criteria for diagnosis/classification from the Diagnostic and Statistical Manual of Mental Disorders-5 and the International Classification of Diseases-10 and diagnostic scales are used to establish a diagnosis. The most commonly used biomarkers in psychotic illnesses are those regarding the neuroimmune system, metabolic abnormalities, neurotrophins and neurotransmitter systems and proteomics. A current issue faced by clinicians is the lack of biomarkers to help develop a more accurate diagnosis, with the possibility of initiating the most effective treatment. The detection of biological markers for psychosis has the potential to contribute to improvements in its diagnosis, prognosis and treatment effectiveness. The mixture of multiple biomarkers may improve the ability to differentiate and classify these patients. In this sense, the aim of this study was to analyze the literature concerning the potential biomarkers that could be used in medical practice and to review the newest developments in electrochemical sensors used for dopamine detection, one of the most important exploited biomarkers.
Collapse
Affiliation(s)
- Adela Magdalena Ciobanu
- Department of Psychiatry, 'Prof. Dr. Alexandru Obregia' Clinical Psychiatric Hospital, 041914 Bucharest, Romania.,Discipline of Psychiatry, Neurosciences Department, 'Carol Davila' University of Medicine and Pharmacy, 020021 Bucharest, Romania
| | - Luana Geza
- Department of Psychiatry, 'Prof. Dr. Alexandru Obregia' Clinical Psychiatric Hospital, 041914 Bucharest, Romania.,Discipline of Psychiatry, Neurosciences Department, 'Carol Davila' University of Medicine and Pharmacy, 020021 Bucharest, Romania
| | - Iulia Gabriela David
- Department of Analytical Chemistry, Faculty of Chemistry, University of Bucharest, 050663 Bucharest, Romania
| | - Dana Elena Popa
- Department of Analytical Chemistry, Faculty of Chemistry, University of Bucharest, 050663 Bucharest, Romania
| | - Mihaela Buleandra
- Department of Analytical Chemistry, Faculty of Chemistry, University of Bucharest, 050663 Bucharest, Romania
| | - Anton Alexandru Ciucu
- Department of Analytical Chemistry, Faculty of Chemistry, University of Bucharest, 050663 Bucharest, Romania
| | - Liana Dehelean
- Department of Neurosciences-Psychiatry, Centre for Cognitive Research in Neuropsychiatric Pathology, 'Victor Babes' University of Medicine and Pharmacy of Timisoara, 300041 Timisoara, Romania
| |
Collapse
|
11
|
Sun Y, Chang S, Liu Z, Zhang L, Wang F, Yue W, Sun H, Ni Z, Chang X, Zhang Y, Chen Y, Liu J, Lu L, Shi J. Identification of novel risk loci with shared effects on alcoholism, heroin, and methamphetamine dependence. Mol Psychiatry 2021; 26:1152-1161. [PMID: 31462767 DOI: 10.1038/s41380-019-0497-y] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/25/2019] [Revised: 05/04/2019] [Accepted: 05/31/2019] [Indexed: 12/18/2022]
Abstract
Different substance dependences have common effects on reward pathway and molecular adaptations, however little is known regarding their shared genetic factors. We aimed to identify the risk genetic variants that are shared for substance dependence (SD). First, promising genome-wide significant loci were identified from 3296 patients (521 alcoholic/1026 heroin/1749 methamphetamine) vs 2859 healthy controls and independently replicated using 1954 patients vs 1904 controls. Second, the functional effects of promising variants on gene expression, addiction characteristics, brain structure (gray and white matter), and addiction behaviors in addiction animal models (chronic administration and self-administration) were assessed. In addition, we assessed the genetic correlation among the three SDs using LD score regression. We identified and replicated three novel loci that were associated with the common risk of heroin, methamphetamine addiction, and alcoholism: ANKS1B rs2133896 (Pmeta = 3.60 × 10-9), AGBL4 rs147247472 (Pmeta = 3.40 × 10-12), and CTNNA2 rs10196867 (Pmeta = 4.73 × 10-9). Rs2133896 in ANKS1B was associated with ANKS1B gene expression and had effects on gray matter of the left calcarine and white matter of the right superior longitudinal fasciculus in heroin dependence. Overexpression of anks1b gene in the ventral tegmental area decreased addiction vulnerability for heroin and methamphetamine in self-administration rat models. Our findings could shed light on the root cause for substance dependence and will be helpful for the development of cost-effective prevention strategies for general addiction disorders.
Collapse
Affiliation(s)
- Yan Sun
- National Institute on Drug Dependence, Peking University, 100191, Beijing, China
| | - Suhua Chang
- Peking University Sixth Hospital, Peking University Institute of Mental Health, NHC Key Laboratory of Mental Health (Peking University), National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital), Peking University, 100191, Beijing, China
| | - Zhen Liu
- National Institute on Drug Dependence, Peking University, 100191, Beijing, China
| | - Libo Zhang
- National Institute on Drug Dependence, Peking University, 100191, Beijing, China
| | - Fan Wang
- The Second Affiliated Hospital, Xinjiang Medical University, 830063, Urumqi, China
- Beijing Hui Long Guan Hospital, 100096, Beijing, China
| | - Weihua Yue
- Peking University Sixth Hospital, Peking University Institute of Mental Health, NHC Key Laboratory of Mental Health (Peking University), National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital), Peking University, 100191, Beijing, China
| | - Hongqiang Sun
- Peking University Sixth Hospital, Peking University Institute of Mental Health, NHC Key Laboratory of Mental Health (Peking University), National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital), Peking University, 100191, Beijing, China
| | - Zhaojun Ni
- Peking University Sixth Hospital, Peking University Institute of Mental Health, NHC Key Laboratory of Mental Health (Peking University), National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital), Peking University, 100191, Beijing, China
| | - Xiangwen Chang
- National Institute on Drug Dependence, Peking University, 100191, Beijing, China
| | - Yibing Zhang
- National Institute on Drug Dependence, Peking University, 100191, Beijing, China
| | - Yang Chen
- National Institute on Drug Dependence, Peking University, 100191, Beijing, China
| | - Jiqiang Liu
- Beijing Compass Biotechnology Company, 102206, Beijing, China
| | - Lin Lu
- National Institute on Drug Dependence, Peking University, 100191, Beijing, China
- Peking University Sixth Hospital, Peking University Institute of Mental Health, NHC Key Laboratory of Mental Health (Peking University), National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital), Peking University, 100191, Beijing, China
| | - Jie Shi
- National Institute on Drug Dependence, Peking University, 100191, Beijing, China.
- Beijing Key Laboratory of Drug Dependence Research, Peking University, 100191, Beijing, China.
- The State Key Laboratory of Natural and Biomimetic Drugs, Peking University, 100191, Beijing, China.
- The Key Laboratory for Neuroscience of the Ministry of Education and Health, Peking University, 100191, Beijing, China.
| |
Collapse
|
12
|
Panyard DJ, Kim KM, Darst BF, Deming YK, Zhong X, Wu Y, Kang H, Carlsson CM, Johnson SC, Asthana S, Engelman CD, Lu Q. Cerebrospinal fluid metabolomics identifies 19 brain-related phenotype associations. Commun Biol 2021; 4:63. [PMID: 33437055 PMCID: PMC7803963 DOI: 10.1038/s42003-020-01583-z] [Citation(s) in RCA: 72] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Accepted: 12/09/2020] [Indexed: 02/07/2023] Open
Abstract
The study of metabolomics and disease has enabled the discovery of new risk factors, diagnostic markers, and drug targets. For neurological and psychiatric phenotypes, the cerebrospinal fluid (CSF) is of particular importance. However, the CSF metabolome is difficult to study on a large scale due to the relative complexity of the procedure needed to collect the fluid. Here, we present a metabolome-wide association study (MWAS), which uses genetic and metabolomic data to impute metabolites into large samples with genome-wide association summary statistics. We conduct a metabolome-wide, genome-wide association analysis with 338 CSF metabolites, identifying 16 genotype-metabolite associations (metabolite quantitative trait loci, or mQTLs). We then build prediction models for all available CSF metabolites and test for associations with 27 neurological and psychiatric phenotypes, identifying 19 significant CSF metabolite-phenotype associations. Our results demonstrate the feasibility of MWAS to study omic data in scarce sample types.
Collapse
Grants
- R01 AG037639 NIA NIH HHS
- UL1 TR000427 NCATS NIH HHS
- T15 LM007359 NLM NIH HHS
- T32 LM012413 NLM NIH HHS
- RF1 AG027161 NIA NIH HHS
- T32 AG000213 NIA NIH HHS
- P2C HD047873 NICHD NIH HHS
- UL1 TR002373 NCATS NIH HHS
- P30 AG062715 NIA NIH HHS
- P50 AG033514 NIA NIH HHS
- R01 AG027161 NIA NIH HHS
- R01 AG054047 NIA NIH HHS
- P30 AG017266 NIA NIH HHS
- R21 AG067092 NIA NIH HHS
- U.S. Department of Health & Human Services | NIH | National Institute on Aging (U.S. National Institute on Aging)
- U.S. Department of Health & Human Services | NIH | Eunice Kennedy Shriver National Institute of Child Health and Human Development (NICHD)
- U.S. Department of Health & Human Services | NIH | U.S. National Library of Medicine (NLM)
- NSF | Directorate for Mathematical & Physical Sciences | Division of Mathematical Sciences (DMS)
- U.S. Department of Health & Human Services | NIH | National Center for Advancing Translational Sciences (NCATS)
- This research is supported by National Institutes of Health (NIH) grants R01AG27161 (Wisconsin Registry for Alzheimer Prevention: Biomarkers of Preclinical AD), R01AG054047 (Genomic and Metabolomic Data Integration in a Longitudinal Cohort at Risk for Alzheimer’s Disease), R21AG067092 (Identifying Metabolomic Risk Factors in Plasma and Cerebrospinal Fluid for Alzheimer’s Disease), R01AG037639 (White Matter Degeneration: Biomarkers in Preclinical Alzheimer’s Disease), P30AG017266 (Center for Demography of Health and Aging), and P50AG033514 and P30AG062715 (Wisconsin Alzheimer’s Disease Research Center Grant), the Helen Bader Foundation, Northwestern Mutual Foundation, Extendicare Foundation, State of Wisconsin, the Clinical and Translational Science Award (CTSA) program through the NIH National Center for Advancing Translational Sciences (NCATS) grant UL1TR000427, and the University of Wisconsin-Madison Office of the Vice Chancellor for Research and Graduate Education with funding from the Wisconsin Alumni Research Foundation. This research was supported in part by the Intramural Research Program of the National Institute on Aging. Computational resources were supported by a core grant to the Center for Demography and Ecology at the University of Wisconsin-Madison (P2CHD047873). Author DJP was supported by an NLM training grant to the Bio-Data Science Training Program (T32LM012413). Author BFD was supported by an NLM training grant to the Computation and Informatics in Biology and Medicine Training Program (NLM 5T15LM007359). Author YKD was supported by a training grant from the National Institute on Aging (T32AG000213). Author HK was supported by National Science Foundation (NSF) grant DMS-1811414 (Theory and Methods for Inferring Causal Effects with Mendelian Randomization).
Collapse
Affiliation(s)
- Daniel J Panyard
- Department of Population Health Sciences, University of Wisconsin-Madison, 610 Walnut Street, 707 WARF Building, Madison, WI, 53726, USA
| | - Kyeong Mo Kim
- Department of Biotechnology, Yonsei University, 50 Yonsei-ro Seodaemun-gu, Seoul, 03722, Republic of Korea
| | - Burcu F Darst
- Center for Genetic Epidemiology, Keck School of Medicine, University of Southern California, 1450 Biggy Street, Los Angeles, CA, 90033, USA
| | - Yuetiva K Deming
- Department of Population Health Sciences, University of Wisconsin-Madison, 610 Walnut Street, 707 WARF Building, Madison, WI, 53726, USA
- Wisconsin Alzheimer's Disease Research Center, University of Wisconsin-Madison, 600 Highland Avenue, J5/1 Mezzanine, Madison, WI, 53792, USA
- Department of Medicine, University of Wisconsin-Madison, 1685 Highland Avenue, 5158 Medical Foundation Centennial Building, Madison, WI, 53705, USA
| | - Xiaoyuan Zhong
- Department of Biostatistics and Medical Informatics, University of Wisconsin-Madison, WARF Room 201, 610 Walnut Street, Madison, WI, 53726, USA
| | - Yuchang Wu
- Department of Biostatistics and Medical Informatics, University of Wisconsin-Madison, WARF Room 201, 610 Walnut Street, Madison, WI, 53726, USA
| | - Hyunseung Kang
- Department of Statistics, University of Wisconsin-Madison, 1300 University Avenue, Madison, WI, 53706, USA
| | - Cynthia M Carlsson
- Wisconsin Alzheimer's Disease Research Center, University of Wisconsin-Madison, 600 Highland Avenue, J5/1 Mezzanine, Madison, WI, 53792, USA
- Department of Medicine, University of Wisconsin-Madison, 1685 Highland Avenue, 5158 Medical Foundation Centennial Building, Madison, WI, 53705, USA
- William S. Middleton Memorial Veterans Hospital, 2500 Overlook Terrace, Madison, WI, 53705, USA
| | - Sterling C Johnson
- Wisconsin Alzheimer's Disease Research Center, University of Wisconsin-Madison, 600 Highland Avenue, J5/1 Mezzanine, Madison, WI, 53792, USA
- Department of Medicine, University of Wisconsin-Madison, 1685 Highland Avenue, 5158 Medical Foundation Centennial Building, Madison, WI, 53705, USA
- William S. Middleton Memorial Veterans Hospital, 2500 Overlook Terrace, Madison, WI, 53705, USA
| | - Sanjay Asthana
- Wisconsin Alzheimer's Disease Research Center, University of Wisconsin-Madison, 600 Highland Avenue, J5/1 Mezzanine, Madison, WI, 53792, USA
- Department of Medicine, University of Wisconsin-Madison, 1685 Highland Avenue, 5158 Medical Foundation Centennial Building, Madison, WI, 53705, USA
- William S. Middleton Memorial Veterans Hospital, 2500 Overlook Terrace, Madison, WI, 53705, USA
| | - Corinne D Engelman
- Department of Population Health Sciences, University of Wisconsin-Madison, 610 Walnut Street, 707 WARF Building, Madison, WI, 53726, USA
| | - Qiongshi Lu
- Department of Biostatistics and Medical Informatics, University of Wisconsin-Madison, WARF Room 201, 610 Walnut Street, Madison, WI, 53726, USA.
- Department of Statistics, University of Wisconsin-Madison, 1300 University Avenue, Madison, WI, 53706, USA.
| |
Collapse
|
13
|
De novo deleterious variants that may alter the dopaminergic reward pathway are associated with anorexia nervosa. Eat Weight Disord 2020; 25:1643-1650. [PMID: 31664672 DOI: 10.1007/s40519-019-00802-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/21/2019] [Accepted: 10/17/2019] [Indexed: 10/25/2022] Open
Abstract
PURPOSE Anorexia nervosa (AN) is a complex neuropsychiatric disorder presenting with dangerously low body weight, and a deep and persistent fear of gaining weight. Up to now, four genome-wide association studies of AN have been conducted to date and identified only few significant loci. However, both previous studies focused on common variation and on rare exonic variants. Currently, de novo variants are one of the most significant risk factors for neurodevelopmental disorders and psychiatric disorders. METHODS We analyzed by whole exome sequencing a cohort of nine female AN individuals and their parents (mother and father), and focused our analysis on de novo variants. RESULTS Here, we found seven de novo missense variants in potential genes in nine studied AN patients. Four of these genes (CSMD1, CREB3, PTPRD and GAB1) belong to a same signaling pathway involving neuron differentiation and dopamine pathway. CONCLUSIONS This study provides a list of interesting genes such as CSDM1 and CREB3 that are candidates to be involved in the etiology of anorexia nervosa. LEVEL OF EVIDENCE basic research.
Collapse
|
14
|
Krebs CE, Ori APS, Vreeker A, Wu T, Cantor RM, Boks MPM, Kahn RS, Olde Loohuis LM, Ophoff RA. Whole blood transcriptome analysis in bipolar disorder reveals strong lithium effect. Psychol Med 2020; 50:2575-2586. [PMID: 31589133 DOI: 10.1017/s0033291719002745] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
BACKGROUND Bipolar disorder (BD) is a highly heritable mood disorder with complex genetic architecture and poorly understood etiology. Previous transcriptomic BD studies have had inconsistent findings due to issues such as small sample sizes and difficulty in adequately accounting for confounders like medication use. METHODS We performed a differential expression analysis in a well-characterized BD case-control sample (Nsubjects = 480) by RNA sequencing of whole blood. We further performed co-expression network analysis, functional enrichment, and cell type decomposition, and integrated differentially expressed genes with genetic risk. RESULTS While we observed widespread differential gene expression patterns between affected and unaffected individuals, these effects were largely linked to lithium treatment at the time of blood draw (FDR < 0.05, Ngenes = 976) rather than BD diagnosis itself (FDR < 0.05, Ngenes = 6). These lithium-associated genes were enriched for cell signaling and immune response functional annotations, among others, and were associated with neutrophil cell-type proportions, which were elevated in lithium users. Neither genes with altered expression in cases nor in lithium users were enriched for BD, schizophrenia, and depression genetic risk based on information from genome-wide association studies, nor was gene expression associated with polygenic risk scores for BD. CONCLUSIONS These findings suggest that BD is associated with minimal changes in whole blood gene expression independent of medication use but emphasize the importance of accounting for medication use and cell type heterogeneity in psychiatric transcriptomic studies. The results of this study add to mounting evidence of lithium's cell signaling and immune-related mechanisms.
Collapse
Affiliation(s)
- Catharine E Krebs
- Center for Neurobehavioral Genetics, Semel Institute for Neuroscience and Human Behavior, University California Los Angeles, Los Angeles, CA, USA
- Department of Human Genetics, University California Los Angeles, Los Angeles, CA, USA
| | - Anil P S Ori
- Center for Neurobehavioral Genetics, Semel Institute for Neuroscience and Human Behavior, University California Los Angeles, Los Angeles, CA, USA
| | - Annabel Vreeker
- Center for Neurobehavioral Genetics, Semel Institute for Neuroscience and Human Behavior, University California Los Angeles, Los Angeles, CA, USA
- Department of Medical Genetics, University Medical Center Utrecht, Utrecht, Netherlands
| | - Timothy Wu
- Center for Neurobehavioral Genetics, Semel Institute for Neuroscience and Human Behavior, University California Los Angeles, Los Angeles, CA, USA
| | - Rita M Cantor
- Center for Neurobehavioral Genetics, Semel Institute for Neuroscience and Human Behavior, University California Los Angeles, Los Angeles, CA, USA
- Department of Human Genetics, University California Los Angeles, Los Angeles, CA, USA
| | - Marco P M Boks
- Department of Psychiatry, Brain Center Rudolf Magnus, University Medical Center Utrecht, Utrecht, Netherlands
| | - Rene S Kahn
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Loes M Olde Loohuis
- Center for Neurobehavioral Genetics, Semel Institute for Neuroscience and Human Behavior, University California Los Angeles, Los Angeles, CA, USA
| | - Roel A Ophoff
- Center for Neurobehavioral Genetics, Semel Institute for Neuroscience and Human Behavior, University California Los Angeles, Los Angeles, CA, USA
- Department of Human Genetics, University California Los Angeles, Los Angeles, CA, USA
- Department of Psychiatry, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
| |
Collapse
|
15
|
Bovo S, Ribani A, Muñoz M, Alves E, Araujo JP, Bozzi R, Čandek-Potokar M, Charneca R, Di Palma F, Etherington G, Fernandez AI, García F, García-Casco J, Karolyi D, Gallo M, Margeta V, Martins JM, Mercat MJ, Moscatelli G, Núñez Y, Quintanilla R, Radović Č, Razmaite V, Riquet J, Savić R, Schiavo G, Usai G, Utzeri VJ, Zimmer C, Ovilo C, Fontanesi L. Whole-genome sequencing of European autochthonous and commercial pig breeds allows the detection of signatures of selection for adaptation of genetic resources to different breeding and production systems. Genet Sel Evol 2020; 52:33. [PMID: 32591011 PMCID: PMC7318759 DOI: 10.1186/s12711-020-00553-7] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2019] [Accepted: 06/17/2020] [Indexed: 12/21/2022] Open
Abstract
Background Natural and artificial directional selection in cosmopolitan and autochthonous pig breeds and wild boars have shaped their genomes and resulted in a reservoir of animal genetic diversity. Signatures of selection are the result of these selection events that have contributed to the adaptation of breeds to different environments and production systems. In this study, we analysed the genome variability of 19 European autochthonous pig breeds (Alentejana, Bísara, Majorcan Black, Basque, Gascon, Apulo-Calabrese, Casertana, Cinta Senese, Mora Romagnola, Nero Siciliano, Sarda, Krškopolje pig, Black Slavonian, Turopolje, Moravka, Swallow-Bellied Mangalitsa, Schwäbisch-Hällisches Schwein, Lithuanian indigenous wattle and Lithuanian White old type) from nine countries, three European commercial breeds (Italian Large White, Italian Landrace and Italian Duroc), and European wild boars, by mining whole-genome sequencing data obtained by using a DNA-pool sequencing approach. Signatures of selection were identified by using a single-breed approach with two statistics [within-breed pooled heterozygosity (HP) and fixation index (FST)] and group-based FST approaches, which compare groups of breeds defined according to external traits and use/specialization/type. Results We detected more than 22 million single nucleotide polymorphisms (SNPs) across the 23 compared populations and identified 359 chromosome regions showing signatures of selection. These regions harbour genes that are already known or new genes that are under selection and relevant for the domestication process in this species, and that affect several morphological and physiological traits (e.g. coat colours and patterns, body size, number of vertebrae and teats, ear size and conformation, reproductive traits, growth and fat deposition traits). Wild boar related signatures of selection were detected across all the genome of several autochthonous breeds, which suggests that crossbreeding (accidental or deliberate) occurred with wild boars. Conclusions Our findings provide a catalogue of genetic variants of many European pig populations and identify genome regions that can explain, at least in part, the phenotypic diversity of these genetic resources.
Collapse
Affiliation(s)
- Samuele Bovo
- Department of Agricultural and Food Sciences, Division of Animal Sciences, University of Bologna, Viale Fanin 46, 40127, Bologna, Italy
| | - Anisa Ribani
- Department of Agricultural and Food Sciences, Division of Animal Sciences, University of Bologna, Viale Fanin 46, 40127, Bologna, Italy
| | - Maria Muñoz
- Departamento Mejora Genética Animal, INIA, Crta. de la Coruña km. 7,5, 28040, Madrid, Spain
| | - Estefania Alves
- Departamento Mejora Genética Animal, INIA, Crta. de la Coruña km. 7,5, 28040, Madrid, Spain
| | - Jose P Araujo
- Centro de Investigação de Montanha (CIMO), Instituto Politécnico de Viana do Castelo, Escola Superior Agrária, Refóios do Lima, 4990-706, Ponte de Lima, Portugal
| | - Riccardo Bozzi
- DAGRI - Animal Science Section, Università di Firenze, Via delle Cascine 5, 50144, Florence, Italy
| | | | - Rui Charneca
- Instituto de Ciências Agrárias e Ambientais Mediterrânicas (ICAAM), Universidade de Évora, Polo da Mitra, Apartado 94, 7006-554, Évora, Portugal
| | - Federica Di Palma
- Earlham Institute, Norwich Research Park, Colney Lane, Norwich, NR47UZ, UK
| | - Graham Etherington
- Earlham Institute, Norwich Research Park, Colney Lane, Norwich, NR47UZ, UK
| | - Ana I Fernandez
- Departamento Mejora Genética Animal, INIA, Crta. de la Coruña km. 7,5, 28040, Madrid, Spain
| | - Fabián García
- Departamento Mejora Genética Animal, INIA, Crta. de la Coruña km. 7,5, 28040, Madrid, Spain
| | - Juan García-Casco
- Departamento Mejora Genética Animal, INIA, Crta. de la Coruña km. 7,5, 28040, Madrid, Spain
| | - Danijel Karolyi
- Department of Animal Science, Faculty of Agriculture, University of Zagreb, Svetošimunska c. 25, 10000, Zagreb, Croatia
| | - Maurizio Gallo
- Associazione Nazionale Allevatori Suini (ANAS), Via Nizza 53, 00198, Rome, Italy
| | - Vladimir Margeta
- Faculty of Agrobiotechnical Sciences, University of Osijek, Vladimira Preloga 1, 31000, Osijek, Croatia
| | - José Manuel Martins
- Instituto de Ciências Agrárias e Ambientais Mediterrânicas (ICAAM), Universidade de Évora, Polo da Mitra, Apartado 94, 7006-554, Évora, Portugal
| | - Marie J Mercat
- IFIP Institut du porc, La Motte au Vicomte, BP 35104, 35651, Le Rheu Cedex, France
| | - Giulia Moscatelli
- Department of Agricultural and Food Sciences, Division of Animal Sciences, University of Bologna, Viale Fanin 46, 40127, Bologna, Italy
| | - Yolanda Núñez
- Departamento Mejora Genética Animal, INIA, Crta. de la Coruña km. 7,5, 28040, Madrid, Spain
| | - Raquel Quintanilla
- Programa de Genética y Mejora Animal, IRTA, Torre Marimon, 08140, Caldes de Montbui, Barcelona, Spain
| | - Čedomir Radović
- Department of Pig Breeding and Genetics, Institute for Animal Husbandry, Belgrade-Zemun, 11080, Serbia
| | - Violeta Razmaite
- Animal Science Institute, Lithuanian University of Health Sciences, Baisogala, Lithuania
| | - Juliette Riquet
- GenPhySE, INRAE, Université de Toulouse, Chemin de Borde-Rouge 24, Auzeville Tolosane, 31326, Castanet Tolosan, France
| | - Radomir Savić
- Faculty of Agriculture, University of Belgrade, Nemanjina 6, Belgrade-Zemun, 11080, Serbia
| | - Giuseppina Schiavo
- Department of Agricultural and Food Sciences, Division of Animal Sciences, University of Bologna, Viale Fanin 46, 40127, Bologna, Italy
| | - Graziano Usai
- AGRIS SARDEGNA, Loc. Bonassai, 07100, Sassari, Italy
| | - Valerio J Utzeri
- Department of Agricultural and Food Sciences, Division of Animal Sciences, University of Bologna, Viale Fanin 46, 40127, Bologna, Italy
| | - Christoph Zimmer
- Bäuerliche Erzeugergemeinschaft Schwäbisch Hall, Schwäbisch Hall, Germany
| | - Cristina Ovilo
- Departamento Mejora Genética Animal, INIA, Crta. de la Coruña km. 7,5, 28040, Madrid, Spain
| | - Luca Fontanesi
- Department of Agricultural and Food Sciences, Division of Animal Sciences, University of Bologna, Viale Fanin 46, 40127, Bologna, Italy.
| |
Collapse
|
16
|
Didikoglu A, Maharani A, Canal MM, Pendleton N, Payton A. Interactions between season of birth, chronological age and genetic polymorphisms in determining later-life chronotype. Mech Ageing Dev 2020; 188:111253. [PMID: 32371234 DOI: 10.1016/j.mad.2020.111253] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2020] [Revised: 03/30/2020] [Accepted: 04/17/2020] [Indexed: 01/19/2023]
Abstract
Human chronotype, the temporal pattern of daily behaviors, is influenced by postnatal seasonal programming and ageing. The aim of this study was to investigate genetic variants that are associated with season of birth programming and longitudinal chronotype change. Longitudinal sleep timing and genotype data from 1449 participants were collected for up to 27 years. Gene-environment interaction analysis was performed for 445 candidate single nucleotide polymorphisms that have previously been associated with chronotype. Associations were tested using linear mixed model. We identified 67 suggestively significant genomic loci that have genotype-ageing interaction and 25 genomic loci that may have genotype-season of birth interaction in determining chronotype. We attempted to replicate the results using longitudinal data of the UK Biobank from approximately 30,000 participants. Biological functions of these genes suggest that epigenetic regulation of gene expression and neural development may have roles in these processes. The strongest associated gene for sleep trajectories was ALKBH5, which has functions of DNA repair and epigenetic regulation. A potential candidate gene for postnatal seasonal programming was SIRT1, which has previously been implicated in postnatal programming, ageing and longevity. Genetic diversity may explain the heterogeneity in ageing-related change of sleep timing and postnatal environmental programming of later-life chronotype.
Collapse
Affiliation(s)
- Altug Didikoglu
- Division of Neuroscience & Experimental Psychology, School of Biological Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, UK(2).
| | - Asri Maharani
- Division of Nursing, Midwifery & Social Work, School of Health Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, UK
| | - Maria Mercè Canal
- Division of Neuroscience & Experimental Psychology, School of Biological Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, UK(2)
| | - Neil Pendleton
- Division of Neuroscience & Experimental Psychology, School of Biological Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, UK(2)
| | - Antony Payton
- Division of Informatics, Imaging & Data Sciences, School of Health Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, UK
| |
Collapse
|
17
|
Nag A, Kurushima Y, Bowyer RCE, Wells PM, Weiss S, Pietzner M, Kocher T, Raffler J, Völker U, Mangino M, Spector TD, Milburn MV, Kastenmüller G, Mohney RP, Suhre K, Menni C, Steves CJ. Genome-wide scan identifies novel genetic loci regulating salivary metabolite levels. Hum Mol Genet 2020; 29:864-875. [PMID: 31960908 PMCID: PMC7104674 DOI: 10.1093/hmg/ddz308] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2019] [Revised: 12/05/2019] [Accepted: 12/11/2019] [Indexed: 12/26/2022] Open
Abstract
Saliva, as a biofluid, is inexpensive and non-invasive to obtain, and provides a vital tool to investigate oral health and its interaction with systemic health conditions. There is growing interest in salivary biomarkers for systemic diseases, notably cardiovascular disease. Whereas hundreds of genetic loci have been shown to be involved in the regulation of blood metabolites, leading to significant insights into the pathogenesis of complex human diseases, little is known about the impact of host genetics on salivary metabolites. Here we report the first genome-wide association study exploring 476 salivary metabolites in 1419 subjects from the TwinsUK cohort (discovery phase), followed by replication in the Study of Health in Pomerania (SHIP-2) cohort. A total of 14 distinct locus-metabolite associations were identified in the discovery phase, most of which were replicated in SHIP-2. While only a limited number of the loci that are known to regulate blood metabolites were also associated with salivary metabolites in our study, we identified several novel saliva-specific locus-metabolite associations, including associations for the AGMAT (with the metabolites 4-guanidinobutanoate and beta-guanidinopropanoate), ATP13A5 (with the metabolite creatinine) and DPYS (with the metabolites 3-ureidopropionate and 3-ureidoisobutyrate) loci. Our study suggests that there may be regulatory pathways of particular relevance to the salivary metabolome. In addition, some of our findings may have clinical significance, such as the utility of the pyrimidine (uracil) degradation metabolites in predicting 5-fluorouracil toxicity and the role of the agmatine pathway metabolites as biomarkers of oral health.
Collapse
Affiliation(s)
- Abhishek Nag
- Department of Twin Research and Genetic Epidemiology, King’s College London, London SE1 7EH, UK
- Wellcome Centre for Human Genetics, University of Oxford, Oxford OX3 7BN, UK
| | - Yuko Kurushima
- Department of Twin Research and Genetic Epidemiology, King’s College London, London SE1 7EH, UK
| | - Ruth C E Bowyer
- Department of Twin Research and Genetic Epidemiology, King’s College London, London SE1 7EH, UK
| | - Philippa M Wells
- Department of Twin Research and Genetic Epidemiology, King’s College London, London SE1 7EH, UK
| | - Stefan Weiss
- Interfaculty Institute of Genetics and Functional Genomics, University Medicine Greifswald & University of Greifswald, 17489 Greifswald, Germany
| | - Maik Pietzner
- Institute of Clinical Chemistry and Laboratory Medicine, University Medicine Greifswald, 17489 Greifswald, Germany
| | - Thomas Kocher
- Department of Restorative Dentistry, Periodontology, Endodontology, and Preventive and Pediatric Dentistry, University Medicine Greifswald, 17489 Greifswald, Germany
| | - Johannes Raffler
- Institute of Bioinformatics and Systems Biology, Helmholtz Zentrum München, German Research Center for Environmental Health, D-85764 Neuherberg, Germany
| | - Uwe Völker
- Interfaculty Institute of Genetics and Functional Genomics, University Medicine Greifswald & University of Greifswald, 17489 Greifswald, Germany
| | - Massimo Mangino
- Department of Twin Research and Genetic Epidemiology, King’s College London, London SE1 7EH, UK
| | - Timothy D Spector
- Department of Twin Research and Genetic Epidemiology, King’s College London, London SE1 7EH, UK
| | - Michael V Milburn
- Discovery and Translational Sciences, Metabolon, Inc., Morrisville, NC 27560, USA
| | - Gabi Kastenmüller
- Institute of Bioinformatics and Systems Biology, Helmholtz Zentrum München, German Research Center for Environmental Health, D-85764 Neuherberg, Germany
| | - Robert P Mohney
- Discovery and Translational Sciences, Metabolon, Inc., Morrisville, NC 27560, USA
| | - Karsten Suhre
- Department of Physiology and Biophysics, Weill Cornell Medicine-Qatar, Doha 24144, Qatar
| | - Cristina Menni
- Department of Twin Research and Genetic Epidemiology, King’s College London, London SE1 7EH, UK
| | - Claire J Steves
- Department of Twin Research and Genetic Epidemiology, King’s College London, London SE1 7EH, UK
| |
Collapse
|
18
|
Duarte-Silva E, Filho AJMC, Barichello T, Quevedo J, Macedo D, Peixoto C. Phosphodiesterase-5 inhibitors: Shedding new light on the darkness of depression? J Affect Disord 2020; 264:138-149. [PMID: 32056743 DOI: 10.1016/j.jad.2019.11.114] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/24/2019] [Revised: 11/22/2019] [Accepted: 11/26/2019] [Indexed: 12/13/2022]
Abstract
BACKGROUND Phosphodiesterase-5 inhibitors (PDE5Is) are used to treat erectile dysfunction (ED). Recently, the antidepressant-like effect of PDE5Is was demonstrated in animal models of depression. In clinical settings, PDE5Is were studied only for ED associated depression. Hence, there are no studies evaluating the effects of PDE5Is for the treatment of major depressive disorder (MDD) without ED. In this review article, we aimed to discuss the use of PDE5Is in the context of MDD, highlighting the roles of PDE genes in the development of MDD, the potential mechanisms by which PDE5Is can be beneficial for MDD and the potentials and limitations of PDE5Is repurposing to treat MDD. METHODS We used PubMed (MEDLINE) database to collect the studies cited in this review. Papers written in English language regardless the year of publication were selected. RESULTS A few preclinical studies support the antidepressant-like activity of PDE5Is. Clinical studies in men with ED and depression suggest that PDE5Is improve depressive symptoms. No clinical studies were conducted in subjects suffering from depression without ED. Antidepressant effect of PDE5Is may be explained by multiple mechanisms including inhibition of brain inflammation and modulation of neuroplasticity. LIMITATIONS The low number of preclinical and absence of clinical studies to support the antidepressant effect of PDE5Is. CONCLUSIONS No clinical trial was conducted to date evaluating PDE5Is in depressed patients without ED. PDE5Is' anti-inflammatory and neuroplasticity mechanisms may justify the potential antidepressant effect of these drugs. Despite this, clinical trials evaluating their efficacy in depressed patients need to be conducted.
Collapse
Affiliation(s)
- Eduardo Duarte-Silva
- Laboratory of Ultrastructure, Aggeu Magalhães Institute (IAM), Oswaldo Cruz Foundation (FIOCRUZ-PE), Recife, PE, Brazil; Graduate Program in Biosciences and Biotechnology for Health (PPGBBS), Aggeu Magalhães Institute (IAM), Recife, PE, Brazil.
| | - Adriano José Maia Chaves Filho
- Neuropsychopharmacology Laboratory, Drug Research and Development Center, Faculty of Medicine, Universidade Federal do Ceará, Fortaleza, CE, Brazil
| | - Tatiana Barichello
- Department of Psychiatry and Behavioral Sciences, McGovern Medical School, The University of Texas Health Science Center at Houston, 1941 East Road, Houston, TX 77054, United States; Laboratory of Neurosciences, Graduate Program in Health Sciences, Health Sciences Unit, University of Southern Santa Catarina-UNESC, Criciúma, SC, Brazil; Center of Excellence on Mood Disorders, Department of Psychiatry and Behavioral Sciences, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, United States.
| | - João Quevedo
- Department of Psychiatry and Behavioral Sciences, McGovern Medical School, The University of Texas Health Science Center at Houston, 1941 East Road, Houston, TX 77054, United States; Laboratory of Neurosciences, Graduate Program in Health Sciences, Health Sciences Unit, University of Southern Santa Catarina-UNESC, Criciúma, SC, Brazil; Center of Excellence on Mood Disorders, Department of Psychiatry and Behavioral Sciences, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, United States.
| | - Danielle Macedo
- Neuropsychopharmacology Laboratory, Drug Research and Development Center, Faculty of Medicine, Universidade Federal do Ceará, Fortaleza, CE, Brazil; Department of Physiology and Pharmacology, Faculty of Medicine, Universidade Federal do Ceará, Fortaleza, CE, Brazil; National Institute for Translational Medicine (INCT-TM, CNPq), Ribeirão Preto, Brazil
| | - Christina Peixoto
- Laboratory of Ultrastructure, Aggeu Magalhães Institute (IAM), Oswaldo Cruz Foundation (FIOCRUZ-PE), Recife, PE, Brazil; National Institute of Science and Technology on Neuroimmunomodulation (INCT-NIM), Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro, Brazil.
| |
Collapse
|
19
|
Multi-omic biomarker identification and validation for diagnosing warzone-related post-traumatic stress disorder. Mol Psychiatry 2020; 25:3337-3349. [PMID: 31501510 PMCID: PMC7714692 DOI: 10.1038/s41380-019-0496-z] [Citation(s) in RCA: 59] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/18/2018] [Revised: 05/15/2019] [Accepted: 06/24/2019] [Indexed: 01/01/2023]
Abstract
Post-traumatic stress disorder (PTSD) impacts many veterans and active duty soldiers, but diagnosis can be problematic due to biases in self-disclosure of symptoms, stigma within military populations, and limitations identifying those at risk. Prior studies suggest that PTSD may be a systemic illness, affecting not just the brain, but the entire body. Therefore, disease signals likely span multiple biological domains, including genes, proteins, cells, tissues, and organism-level physiological changes. Identification of these signals could aid in diagnostics, treatment decision-making, and risk evaluation. In the search for PTSD diagnostic biomarkers, we ascertained over one million molecular, cellular, physiological, and clinical features from three cohorts of male veterans. In a discovery cohort of 83 warzone-related PTSD cases and 82 warzone-exposed controls, we identified a set of 343 candidate biomarkers. These candidate biomarkers were selected from an integrated approach using (1) data-driven methods, including Support Vector Machine with Recursive Feature Elimination and other standard or published methodologies, and (2) hypothesis-driven approaches, using previous genetic studies for polygenic risk, or other PTSD-related literature. After reassessment of ~30% of these participants, we refined this set of markers from 343 to 28, based on their performance and ability to track changes in phenotype over time. The final diagnostic panel of 28 features was validated in an independent cohort (26 cases, 26 controls) with good performance (AUC = 0.80, 81% accuracy, 85% sensitivity, and 77% specificity). The identification and validation of this diverse diagnostic panel represents a powerful and novel approach to improve accuracy and reduce bias in diagnosing combat-related PTSD.
Collapse
|
20
|
Younis RM, Taylor RM, Beardsley PM, McClay JL. The ANKS1B gene and its associated phenotypes: focus on CNS drug response. Pharmacogenomics 2019; 20:669-684. [PMID: 31250731 PMCID: PMC6912848 DOI: 10.2217/pgs-2019-0015] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2019] [Accepted: 04/26/2019] [Indexed: 12/21/2022] Open
Abstract
The ANKS1B gene was a top finding in genome-wide association studies (GWAS) of antipsychotic drug response. Subsequent GWAS findings for ANKS1B include cognitive ability, educational attainment, body mass index, response to corticosteroids and drug dependence. We review current human association evidence for ANKS1B, in addition to functional studies that include two published mouse knockouts. The several GWAS findings in humans indicate that phenotypically relevant variation is segregating at the ANKS1B locus. ANKS1B shows strong plausibility for involvement in CNS drug response because it encodes a postsynaptic effector protein that mediates long-term changes to neuronal biology. Forthcoming data from large biobanks should further delineate the role of ANKS1B in CNS drug response.
Collapse
Affiliation(s)
- Rabha M Younis
- Department of Pharmacotherapy & Outcomes Science, Virginia Commonwealth University School of Pharmacy, Richmond, VA 23298, USA
| | - Rachel M Taylor
- Center for Military Psychiatry & Neuroscience, Walter Reed Army Institute of Research, Silver Spring, MA 20910, USA
| | - Patrick M Beardsley
- Department of Pharmacology & Toxicology, Virginia Commonwealth University School of Medicine, Richmond, VA 23298, USA
- Center for Biomarker Research & Personalized Medicine, Virginia Commonwealth University, Richmond, VA 23298, USA
| | - Joseph L McClay
- Department of Pharmacotherapy & Outcomes Science, Virginia Commonwealth University School of Pharmacy, Richmond, VA 23298, USA
| |
Collapse
|
21
|
Liu Y, Fu X, Tang Z, Li C, Xu Y, Zhang F, Zhou D, Zhu C. Altered expression of the CSMD1 gene in the peripheral blood of schizophrenia patients. BMC Psychiatry 2019; 19:113. [PMID: 30987620 PMCID: PMC6466712 DOI: 10.1186/s12888-019-2089-4] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/30/2018] [Accepted: 03/27/2019] [Indexed: 01/08/2023] Open
Abstract
BACKGROUND Schizophrenia (SCZ) is a heritable, refractory, and devastating psychiatric disorder. Previous studies have shown that the variants of CUB and sushi multiple domains 1 (CSMD1) demonstrate significant genome-wide association with SCZ. However, few studies have been conducted on the effect of antipsychotics on the expression levels of CSMD1. This study explored whether a change occurs in the expression of the CSMD1 gene before and after antipsychotic treatment in SCZ patients. METHODS The study population comprised Han Chinese patients from eastern China, including 32 SCZ patients and 48 healthy controls. The expression of CSMD1 before and after treatment in the SCZ group and between the two groups was analyzed using real-time quantitative polymerase chain reaction (RT-qPCR). RESULTS The expression levels of the CSMD1 gene in the peripheral blood mononuclear cells (PBMCs) of SCZ patients were lower than those in the healthy controls. The expression levels of the CSMD1 gene in the PBMCs of the SCZ patients after antipsychotic treatment were higher than those in the baseline SCZ patients (all P < 0.05). CONCLUSIONS Our results showed that the expression levels of CSMD1 are correlated with the development and treatment of SCZ, providing further evidence for the involvement of CSMD1 in SCZ.
Collapse
Affiliation(s)
- Yansong Liu
- Department of Clinical Psychology, Suzhou Guangji Hospital, The Affiliated Guangji Hospital of Soochow University, Suzhou, 215137, Jiangsu, China
| | - Xiaoqian Fu
- Department of Clinical Psychology, Suzhou Guangji Hospital, The Affiliated Guangji Hospital of Soochow University, Suzhou, 215137, Jiangsu, China
| | - Zhen Tang
- Department of Clinical Psychology, Suzhou Guangji Hospital, The Affiliated Guangji Hospital of Soochow University, Suzhou, 215137, Jiangsu, China
| | - Cui Li
- Department of Psychology, Xinghua People's Hospital, Xinghua, 225700, Jiangsu, China
| | - Yong Xu
- Department of Psychiatry, First Clinical Medical College/First Hospital of Shanxi Medical University, Taiyuan, China
| | - Fuquan Zhang
- Department of Clinical Psychology, Wuxi Mental Health Center, Nanjing Medical University, 156 Qianrong Road, Wuxi, Jiangsu Province, 214151, China
| | - Deyi Zhou
- Department of Clinical Psychology, Wuxi Mental Health Center, Nanjing Medical University, 156 Qianrong Road, Wuxi, Jiangsu Province, 214151, China.
| | - Chunming Zhu
- Department of Clinical Psychology, Wuxi Mental Health Center, Nanjing Medical University, 156 Qianrong Road, Wuxi, Jiangsu Province, 214151, China.
| |
Collapse
|
22
|
Loohuis LM, Albersen M, de Jong S, Wu T, Luykx JJ, Jans JJM, Verhoeven-Duif NM, Ophoff RA. The Alkaline Phosphatase (ALPL) Locus Is Associated with B6 Vitamer Levels in CSF and Plasma. Genes (Basel) 2018; 10:genes10010008. [PMID: 30583557 PMCID: PMC6357176 DOI: 10.3390/genes10010008] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2018] [Revised: 12/13/2018] [Accepted: 12/14/2018] [Indexed: 01/27/2023] Open
Abstract
The active form of vitamin B6, pyridoxal phosphate (PLP), is essential for human metabolism. The brain is dependent on vitamin B6 for its neurotransmitter balance. To obtain insight into the genetic determinants of vitamin B6 homeostasis, we conducted a genome-wide association study (GWAS) of the B6 vitamers pyridoxal (PL), PLP and the degradation product of vitamin B6, pyridoxic acid (PA). We collected a unique sample set of cerebrospinal fluid (CSF) and plasma from the same healthy human subjects of Dutch ancestry (n = 493) and included concentrations and ratios in and between these body fluids in our analysis. Based on a multivariate joint analysis of all B6 vitamers and their ratios, we identified a genome-wide significant association at a locus on chromosome 1 containing the ALPL (alkaline phosphatase) gene (minimal p = 7.89 × 10−10, rs1106357, minor allele frequency (MAF) = 0.46), previously associated with vitamin B6 levels in blood. Subjects homozygous for the minor allele showed a 1.4-times-higher ratio between PLP and PL in plasma, and even a 1.6-times-higher ratio between PLP and PL in CSF than subjects homozygous for the major allele. In addition, we observed a suggestive association with the CSF:plasma ratio of PLP on chromosome 15 (minimal p = 7.93 × 10−7, and MAF = 0.06 for rs28789220). Even though this finding is not reaching genome-wide significance, it highlights the potential of our experimental setup for studying transport and metabolism across the blood–CSF barrier. This GWAS of B6 vitamers identifies alkaline phosphatase as a key regulator in human vitamin B6 metabolism in CSF as well as plasma. Furthermore, our results demonstrate the potential of genetic studies of metabolites in plasma and CSF to elucidate biological aspects underlying metabolite generation, transport and degradation.
Collapse
Affiliation(s)
- Loes M Loohuis
- Center for Neurobehavioral Genetics, Semel Institute for Neuroscience and Human Behavior, University of California Los Angeles (UCLA), Los Angeles, CA 90095, USA.
| | - Monique Albersen
- Section Metabolic Diagnostics, Department of Genetics, University Medical Center (UMC), 3584 EA, Utrecht, The Netherlands.
| | - Simone de Jong
- Center for Neurobehavioral Genetics, Semel Institute for Neuroscience and Human Behavior, University of California Los Angeles (UCLA), Los Angeles, CA 90095, USA.
| | - Timothy Wu
- Center for Neurobehavioral Genetics, Semel Institute for Neuroscience and Human Behavior, University of California Los Angeles (UCLA), Los Angeles, CA 90095, USA.
| | - Jurjen J Luykx
- Department of Psychiatry, Rudolf Magnus Institute of Neuroscience, University Medical Center (UMC), 3584 CG, Utrecht, The Netherlands.
- Department of Translational Neuroscience, Human Neurogenetics Unit, Brain Center Rudolf Magnus, University Medical Center Utrecht (UMC), 3584 CG, Utrecht, The Netherlands.
| | - Judith J M Jans
- Section Metabolic Diagnostics, Department of Genetics, University Medical Center (UMC), 3584 EA, Utrecht, The Netherlands.
| | - Nanda M Verhoeven-Duif
- Section Metabolic Diagnostics, Department of Genetics, University Medical Center (UMC), 3584 EA, Utrecht, The Netherlands.
| | - Roel A Ophoff
- Center for Neurobehavioral Genetics, Semel Institute for Neuroscience and Human Behavior, University of California Los Angeles (UCLA), Los Angeles, CA 90095, USA.
| |
Collapse
|
23
|
Yoshimura R, Kishi T, Atake K, Katsuki A, Iwata N. Serum Brain-Derived Neurotrophic Factor, and Plasma Catecholamine Metabolites in People with Major Depression: Preliminary Cross-Sectional Study. Front Psychiatry 2018; 9:52. [PMID: 29541037 PMCID: PMC5835900 DOI: 10.3389/fpsyt.2018.00052] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/20/2023] Open
Abstract
BACKGROUND There are complicated interactions between catecholaminergic neurons and brain-derived neurotrophic factor (BDNF) in the brain. However, no reports have addressed the relationship among 3-methoxy-4-hydroxyphenylglycol (MHPG), homovanillic acid (HVA), and BDNF in the blood. OBJECTIVE This paper sought to investigate correlations between serum BDNF and plasma levels of MHPG and HVA in people with major depression (MD). MATERIALS AND METHODS A total of 148 patients (male/female 65/83, age 49.5 ± 12.1 years old) who satisfied criteria for MD based on the Diagnostic and Statistical Manual of Mental Disorders IV were enrolled in the present study. Plasma levels of MHPG and HVA were analyzed using high-performance liquid chromatography, and serum BDNF was measured using ELISA. RESULTS No interactions were observed between plasma HVA levels (mean ± SD = 4.5 ± 1.5 ng/mL) and age, sex, HAMD scores, or serum BDNF levels (mean ± SD = 9.8 ± 2.9 ng/mL). No correlations were not also observed between plasma MHPG levels (mean ± SD = 5.9 ± 2.1 ng/mL) and age, sex, the HAMD17 scores (mean ± SD = 22.2 ± 2.9 ng/mL), or serum BDNF levels. Serum BDNF levels were negatively associated with HAMD17 scores. CONCLUSION The results suggest that there are no significant correlations between catecholamine metabolites and BDNF in the blood for MDD patients.
Collapse
Affiliation(s)
- Reiji Yoshimura
- Department of Psychiatry, School of Medicine, University of Occupational and Environmental Health, Kitakyushu, Japan
| | - Taro Kishi
- Department of Psychiatry, School of Medicine, Fujita Health University, Toyoake, Japan
| | - Kiyokazu Atake
- Department of Psychiatry, School of Medicine, University of Occupational and Environmental Health, Kitakyushu, Japan
| | - Asuka Katsuki
- Department of Psychiatry, School of Medicine, University of Occupational and Environmental Health, Kitakyushu, Japan
| | - Nakao Iwata
- Department of Psychiatry, School of Medicine, Fujita Health University, Toyoake, Japan
| |
Collapse
|
24
|
Polimanti R, Zhao H, Farrer LA, Kranzler HR, Gelernter J. Ancestry-specific and sex-specific risk alleles identified in a genome-wide gene-by-alcohol dependence interaction study of risky sexual behaviors. Am J Med Genet B Neuropsychiatr Genet 2017; 174:846-853. [PMID: 28990359 PMCID: PMC5861711 DOI: 10.1002/ajmg.b.32604] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/26/2017] [Revised: 09/07/2017] [Accepted: 09/18/2017] [Indexed: 01/26/2023]
Abstract
We previously mapped loci for the genome-wide association studies (GWAS) and genome-wide gene-by-alcohol dependence interaction (GW-GxAD) analyses of risky sexual behaviors (RSB). This study extends those findings by analyzing the ancestry- and sex-specific AD-stratified effects on RSB. We examined the concordance of findings for the AD-stratified GWAS and the GW-GxAD analysis of RSB, with concordance defined as genome-wide significance in one analysis and at least nominal significance in the second analysis. A total of 2,173 African-American (AA) and 1,751 European-American (EA) subjects were investigated. Information regarding RSB (lifetime experiences of unprotected sex and multiple sexual partners) and DSM-IV diagnosis of lifetime AD were derived from the Semi-Structured Assessment for Drug Dependence and Alcoholism (SSADDA). In our ancestry- and sex-specific analyses, we identified four independent genome-wide significant (GWS) loci (p < 5*10-8 ) and one suggestive locus (p < 6*10-8 ). In men, we observed a GWS signal in FAM162A (rs2002594, p = 4.96*10-8 ). In women, there was a suggestive locus in PLGRKT (rs3824435, p = 5.52*10-8 ). In AAs, there was a GWS signal in GRK5 (rs1316543, p = 1.25*10-9 ). In AA men, we observed an intergenic GWS signal (rs12898370, p = 4.49*10-8 ) near LINGO1. In EA men, there was a GWS signal in CCSER1 (rs62313897; p = 7.93*10-10 ). The loci identified in this GWAS implicate molecular mechanisms related to psychiatric illness and personality features, suggesting that the interplay between AD and RSB is mediated by alleles associated with behavioral traits.
Collapse
Affiliation(s)
- Renato Polimanti
- Department of Psychiatry, Yale University School of Medicine, West Haven, CT, USA
- VA CT Healthcare Center, West Haven, CT, USA
| | - Hongyu Zhao
- Department of Biostatistics, Yale University School of Public Health, New Haven, CT, USA
- Department of Genetics, Yale University School of Medicine, New Haven, CT, USA
| | - Lindsay A. Farrer
- Departments of Medicine (Biomedical Genetics), Neurology, Ophthalmology, Biostatistics, and Epidemiology, Boston University Schools of Medicine and Public Health, Boston, MA, USA
| | - Henry R. Kranzler
- Department of Psychiatry, University of Pennsylvania School of Medicine and VISN 4 MIRECC, Philadelphia VAMC, Philadelphia, PA, USA
| | - Joel Gelernter
- Department of Psychiatry, Yale University School of Medicine, West Haven, CT, USA
- VA CT Healthcare Center, West Haven, CT, USA
- Department of Genetics, Yale University School of Medicine, New Haven, CT, USA
- Department of Neurobiology, Yale University School of Medicine, New Haven, CT, USA
| |
Collapse
|
25
|
Fitzgerald JC, Zimprich A, Carvajal Berrio DA, Schindler KM, Maurer B, Schulte C, Bus C, Hauser AK, Kübler M, Lewin R, Bobbili DR, Schwarz LM, Vartholomaiou E, Brockmann K, Wüst R, Madlung J, Nordheim A, Riess O, Martins LM, Glaab E, May P, Schenke-Layland K, Picard D, Sharma M, Gasser T, Krüger R. Metformin reverses TRAP1 mutation-associated alterations in mitochondrial function in Parkinson's disease. Brain 2017; 140:2444-2459. [PMID: 29050400 DOI: 10.1093/brain/awx202] [Citation(s) in RCA: 79] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2017] [Accepted: 07/04/2017] [Indexed: 12/11/2022] Open
Abstract
The mitochondrial proteins TRAP1 and HTRA2 have previously been shown to be phosphorylated in the presence of the Parkinson's disease kinase PINK1 but the downstream signalling is unknown. HTRA2 and PINK1 loss of function causes parkinsonism in humans and animals. Here, we identified TRAP1 as an interactor of HTRA2 using an unbiased mass spectrometry approach. In our human cell models, TRAP1 overexpression is protective, rescuing HTRA2 and PINK1-associated mitochondrial dysfunction and suggesting that TRAP1 acts downstream of HTRA2 and PINK1. HTRA2 regulates TRAP1 protein levels, but TRAP1 is not a direct target of HTRA2 protease activity. Following genetic screening of Parkinson's disease patients and healthy controls, we also report the first TRAP1 mutation leading to complete loss of functional protein in a patient with late onset Parkinson's disease. Analysis of fibroblasts derived from the patient reveal that oxygen consumption, ATP output and reactive oxygen species are increased compared to healthy individuals. This is coupled with an increased pool of free NADH, increased mitochondrial biogenesis, triggering of the mitochondrial unfolded protein response, loss of mitochondrial membrane potential and sensitivity to mitochondrial removal and apoptosis. These data highlight the role of TRAP1 in the regulation of energy metabolism and mitochondrial quality control. Interestingly, the diabetes drug metformin reverses mutation-associated alterations on energy metabolism, mitochondrial biogenesis and restores mitochondrial membrane potential. In summary, our data show that TRAP1 acts downstream of PINK1 and HTRA2 for mitochondrial fine tuning, whereas TRAP1 loss of function leads to reduced control of energy metabolism, ultimately impacting mitochondrial membrane potential. These findings offer new insight into mitochondrial pathologies in Parkinson's disease and provide new prospects for targeted therapies.
Collapse
Affiliation(s)
- Julia C Fitzgerald
- Department of Neurodegenerative Diseases, Center of Neurology and Hertie-Institute for Clinical Brain Research, University of Tübingen and German Centre for Neurodegenerative Diseases, Tübingen, Germany
| | | | - Daniel A Carvajal Berrio
- Department of Women's Health, Research Institute for Women's Health, University of Tübingen, Tübingen, Germany
| | - Kevin M Schindler
- Department of Neurodegenerative Diseases, Center of Neurology and Hertie-Institute for Clinical Brain Research, University of Tübingen and German Centre for Neurodegenerative Diseases, Tübingen, Germany.,University of Tübingen, Interfaculty Institute of Biochemistry, Tübingen, Germany
| | - Brigitte Maurer
- Department of Neurodegenerative Diseases, Center of Neurology and Hertie-Institute for Clinical Brain Research, University of Tübingen and German Centre for Neurodegenerative Diseases, Tübingen, Germany
| | - Claudia Schulte
- Department of Neurodegenerative Diseases, Center of Neurology and Hertie-Institute for Clinical Brain Research, University of Tübingen and German Centre for Neurodegenerative Diseases, Tübingen, Germany
| | - Christine Bus
- Department of Neurodegenerative Diseases, Center of Neurology and Hertie-Institute for Clinical Brain Research, University of Tübingen and German Centre for Neurodegenerative Diseases, Tübingen, Germany
| | - Anne-Kathrin Hauser
- Department of Neurodegenerative Diseases, Center of Neurology and Hertie-Institute for Clinical Brain Research, University of Tübingen and German Centre for Neurodegenerative Diseases, Tübingen, Germany
| | - Manuela Kübler
- Department of Neurodegenerative Diseases, Center of Neurology and Hertie-Institute for Clinical Brain Research, University of Tübingen and German Centre for Neurodegenerative Diseases, Tübingen, Germany
| | - Rahel Lewin
- Department of Neurodegenerative Diseases, Center of Neurology and Hertie-Institute for Clinical Brain Research, University of Tübingen and German Centre for Neurodegenerative Diseases, Tübingen, Germany
| | - Dheeraj Reddy Bobbili
- Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, Esch-sur-Alzette, Luxembourg
| | - Lisa M Schwarz
- Department of Neurodegenerative Diseases, Center of Neurology and Hertie-Institute for Clinical Brain Research, University of Tübingen and German Centre for Neurodegenerative Diseases, Tübingen, Germany.,Graduate Training Centre of Neuroscience, International Max Planck Research School, Tübingen, Germany
| | | | - Kathrin Brockmann
- Department of Neurodegenerative Diseases, Center of Neurology and Hertie-Institute for Clinical Brain Research, University of Tübingen and German Centre for Neurodegenerative Diseases, Tübingen, Germany
| | - Richard Wüst
- Department of Neurodegenerative Diseases, Center of Neurology and Hertie-Institute for Clinical Brain Research, University of Tübingen and German Centre for Neurodegenerative Diseases, Tübingen, Germany.,Department of Psychiatry and Psychotherapie, University Hospital Tübingen, Germany
| | - Johannes Madlung
- University of Tübingen, Interfaculty Institute for Cell Biology, Proteome Center Tübingen, Tübingen, Germany
| | - Alfred Nordheim
- University of Tübingen, Interfaculty Institute of Cell Biology, Unit of Molecular Biology, Tübingen, Germany
| | - Olaf Riess
- University of Tübingen, Institute of Medical Genetics and Applied Genomics, Tübingen, Germany
| | | | - Enrico Glaab
- Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, Esch-sur-Alzette, Luxembourg
| | - Patrick May
- Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, Esch-sur-Alzette, Luxembourg
| | - Katja Schenke-Layland
- Department of Women's Health, Research Institute for Women's Health, University of Tübingen, Tübingen, Germany.,Department of Cell and Tissue Engineering, Fraunhofer Institute for Interfacial Engineering and Biotechnology IGB Stuttgart, Germany.,Department of Medicine/ Cardiology, CVRL, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | - Didier Picard
- University of Geneva, Department of Cell Biology, Geneva, Switzerland
| | - Manu Sharma
- Centre for Genetic Epidemiology, Institute for Clinical Epidemiology and Applied Biometry, University of Tübingen, Germany
| | - Thomas Gasser
- Department of Neurodegenerative Diseases, Center of Neurology and Hertie-Institute for Clinical Brain Research, University of Tübingen and German Centre for Neurodegenerative Diseases, Tübingen, Germany
| | - Rejko Krüger
- Department of Neurodegenerative Diseases, Center of Neurology and Hertie-Institute for Clinical Brain Research, University of Tübingen and German Centre for Neurodegenerative Diseases, Tübingen, Germany.,Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, Esch-sur-Alzette, Luxembourg.,Parkinson Research Clinic, Centre Hospitalier de Luxembourg (CHL), Luxembourg
| |
Collapse
|
26
|
Paparelli A, Iwata K, Wakuda T, Iyegbe C, Murray RM, Takei N. Perinatal Asphyxia in Rat Alters Expression of Novel Schizophrenia Risk Genes. Front Mol Neurosci 2017; 10:341. [PMID: 29163023 PMCID: PMC5663725 DOI: 10.3389/fnmol.2017.00341] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2017] [Accepted: 10/06/2017] [Indexed: 11/17/2022] Open
Abstract
Epidemiological studies suggest that obstetric complications, particularly those related to hypoxia during labor and delivery, are a risk factor for development of schizophrenia. The impact of perinatal asphyxia on postnatal life has been studied in a rodent model of global hypoxia, which is accompanied by cesarean section birth. This asphyxia model shows several behavioral, pharmacological, neurochemical, and neuroanatomical abnormalities in adulthood that have relevance to schizophrenia. Further, it is suggested that schizophrenia has a strong genetic component, and indeed novel candidate genes were recently identified by a genome-wide association study. Here, we examined alteration in the novel schizophrenia risk genes, CNNM2, CSMD1, and MMP16 in the brains of rats undergoing cesarean section with or without global hypoxia. The brain regions studied were the prefrontal cortex, striatum, and hippocampus, which are all relevant to schizophrenia. Risk gene expression was measured at three time periods: neonatal, adolescence, and adulthood. We also performed an in vitro analysis to determine involvement of these genes in CNS maturation during differentiation of human neuronal and glial cell lines. Cnnm2 expression was altered in the brains of asphyxia model rats. However, Csmd1 and Mmp16 showed altered expression by exposure to cesarean section only. These findings suggest that altered expression of these risk genes via asphyxia and cesarean section may be associated, albeit through distinct pathways, with the pathobiology of schizophrenia.
Collapse
Affiliation(s)
- Alessandra Paparelli
- Department of Psychosis Studies, Institute of Psychiatry, King's College London, London, United Kingdom
| | - Keiko Iwata
- Research Center for Child Mental Development, University of Fukui, Fukui, Japan.,Department of Biology, University of Padova, Padova, Italy
| | - Tomoyasu Wakuda
- Department of Psychiatry, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | - Conrad Iyegbe
- Department of Psychosis Studies, Institute of Psychiatry, King's College London, London, United Kingdom
| | - Robin M Murray
- Department of Psychosis Studies, Institute of Psychiatry, King's College London, London, United Kingdom
| | - Nori Takei
- Department of Psychosis Studies, Institute of Psychiatry, King's College London, London, United Kingdom.,Division of Neuropsychological Development and Health Sciences, United Graduate School of Child Development, Osaka University, Kanazawa University, Hamamatsu University School of Medicine, Chiba University and University of Fukui, Hamamatsu, Japan.,Research Center for Child Mental Development, Hamamatsu University School of Medicine, Hamamatsu, Japan
| |
Collapse
|
27
|
Wang FL, Chassin L. Negative Urgency Mediates the Relation between Genetically-Influenced Serotonin Functioning and Alcohol Problems. Clin Psychol Sci 2017; 6:106-122. [PMID: 29354329 DOI: 10.1177/2167702617733817] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
Serotonin (5-HT) functioning is associated with alcohol problems. However, the mechanisms underlying this association remain unclear. The current study tested whether five separate dimensions of impulsivity (UPPS-P) mediated the relation between a polygenic score indexing 5-HT functioning and alcohol problems and whether any of these paths were moderated by age. Results showed that a 5-HT polygenic score predicted alcohol problems indirectly through negative urgency, but not any other facet of impulsivity. The 5-HT polygenic score also directly predicted alcohol problems. No age moderation was found. Findings suggest that negative urgency might be one important mechanism underlying the relation between genetically-influenced 5-HT functioning and alcohol problems. However, genetically-influenced 5-HT functioning likely influences alcohol problems through additional mechanisms. More broadly, results suggest that the previously observed transdiagnostic nature of 5-HT functioning on diverse types of psychopathology might be, in part, explained by its effect on negative urgency.
Collapse
Affiliation(s)
- Frances L Wang
- Department of Psychology, Arizona State University.,Department of Psychiatry, University of Pittsburgh Medical Center
| | | |
Collapse
|
28
|
Serotonin functioning and adolescents' alcohol use: A genetically informed study examining mechanisms of risk. Dev Psychopathol 2017; 30:213-233. [PMID: 28534453 DOI: 10.1017/s095457941700058x] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
The current study used data from two longitudinal samples to test whether self-regulation, depressive symptoms, and aggression/antisociality were mediators in the relation between a polygenic score indexing serotonin (5-HT) functioning and alcohol use in adolescence. The results from an independent genome-wide association study of 5-hydroxyindoleacetic acid in the cerebrospinal fluid were used to create 5-HT polygenic risk scores. Adolescents and/or parents reported on adolescents' self-regulation (Time 1), depressive symptoms (Time 2), aggression/antisociality (Time 2), and alcohol use (Time 3). The results showed that 5-HT polygenic risk did not predict self-regulation. However, adolescents with higher levels of 5-HT polygenic risk showed greater depression and aggression/antisociality. Adolescents' aggression/antisociality mediated the relation between 5-HT polygenic risk and later alcohol use. Deficits in self-regulation also predicted depression and aggression/antisociality, and indirectly predicted alcohol use through aggression/antisociality. Pathways to alcohol use were especially salient for males from families with low parental education in one of the two samples. The results provide insights into the longitudinal mechanisms underlying the relation between 5-HT functioning and alcohol use (i.e., earlier aggression/antisociality). There was no evidence that genetically based variation in 5-HT functioning predisposed individuals to deficits in self-regulation. Genetically based variation in 5-HT functioning and self-regulation might be separate, transdiagnostic risk factors for several types of psychopathology.
Collapse
|
29
|
Manchia M, Carpiniello B, Valtorta F, Comai S. Serotonin Dysfunction, Aggressive Behavior, and Mental Illness: Exploring the Link Using a Dimensional Approach. ACS Chem Neurosci 2017; 8:961-972. [PMID: 28378993 DOI: 10.1021/acschemneuro.6b00427] [Citation(s) in RCA: 49] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022] Open
Abstract
Aggressive individuals have higher rates of mental illness compared to non-aggressive individuals. Multiple factors, including psychosocial, genetic, and neurobiological determinants modulate the liability to both aggressive behavior and mental illness. Concerning the latter factors, multiple lines of evidence have shown a dysfunction in the serotonin (5-HT) system occurring in aggressive and in mentally ill individuals. In particular, reduced 5-HT activity has been associated with depression as well as with aggressive behavior, especially with impulsive aggression. Consistently, psychopharmacological interventions aimed at boosting the 5-HT system (e.g., with selective serotonin reuptake inhibitors) have demonstrated therapeutic efficacy in a high percentage of patients with either or both pathological conditions. Current knowledge does not yet allow to clearly disentangle whether 5-HT dysfunction, most often a 5-HT deficiency, is the cause or the consequence of the aggressive/violent behavior, of the underlying mental disease/s, or the expression of the comorbidity. Future studies are thus needed to clarify the association between changes in 5-HT levels, altered activity of 5-HT receptors and their intracellular signaling cascades, and modifications of 5-HT genes, and in particular the neurobiological link between the altered 5-HT machinery and aggressive behavior in the context or in the absence of mental illness. In this Review, we employ a dimensional approach to discuss the trivariate relationship among the 5-HT system, aggressive behavior, and mental illness, focusing our attention on 5-HT levels, 5-HT receptors, metabolic enzymes, and their genes. Emphasis is given to controversial findings, still unanswered questions, and future perspectives.
Collapse
Affiliation(s)
- Mirko Manchia
- Section of Psychiatry,
Department of Medical Sciences and Public Health, University of Cagliari, 09121 Cagliari, Italy
- Department of Pharmacology, Dalhousie University, Halifax, Nova Scotia B3H 4R2, Canada
| | - Bernardo Carpiniello
- Section of Psychiatry,
Department of Medical Sciences and Public Health, University of Cagliari, 09121 Cagliari, Italy
| | - Flavia Valtorta
- San Raffaele Scientific Institute and Vita Salute University, Via Olgettina 58, 20132 Milano, Italy
| | - Stefano Comai
- San Raffaele Scientific Institute and Vita Salute University, Via Olgettina 58, 20132 Milano, Italy
| |
Collapse
|
30
|
Perkovic MN, Erjavec GN, Strac DS, Uzun S, Kozumplik O, Pivac N. Theranostic Biomarkers for Schizophrenia. Int J Mol Sci 2017; 18:E733. [PMID: 28358316 PMCID: PMC5412319 DOI: 10.3390/ijms18040733] [Citation(s) in RCA: 70] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2017] [Revised: 03/23/2017] [Accepted: 03/27/2017] [Indexed: 12/14/2022] Open
Abstract
Schizophrenia is a highly heritable, chronic, severe, disabling neurodevelopmental brain disorder with a heterogeneous genetic and neurobiological background, which is still poorly understood. To allow better diagnostic procedures and therapeutic strategies in schizophrenia patients, use of easy accessible biomarkers is suggested. The most frequently used biomarkers in schizophrenia are those associated with the neuroimmune and neuroendocrine system, metabolism, different neurotransmitter systems and neurotrophic factors. However, there are still no validated and reliable biomarkers in clinical use for schizophrenia. This review will address potential biomarkers in schizophrenia. It will discuss biomarkers in schizophrenia and propose the use of specific blood-based panels that will include a set of markers associated with immune processes, metabolic disorders, and neuroendocrine/neurotrophin/neurotransmitter alterations. The combination of different markers, or complex multi-marker panels, might help in the discrimination of patients with different underlying pathologies and in the better classification of the more homogenous groups. Therefore, the development of the diagnostic, prognostic and theranostic biomarkers is an urgent and an unmet need in psychiatry, with the aim of improving diagnosis, therapy monitoring, prediction of treatment outcome and focus on the personal medicine approach in order to improve the quality of life in patients with schizophrenia and decrease health costs worldwide.
Collapse
Affiliation(s)
| | | | - Dubravka Svob Strac
- Rudjer Boskovic Institute, Division of Molecular Medicine, 10000 Zagreb, Croatia.
| | - Suzana Uzun
- Clinic for Psychiatry Vrapce, 10090 Zagreb, Croatia.
| | | | - Nela Pivac
- Rudjer Boskovic Institute, Division of Molecular Medicine, 10000 Zagreb, Croatia.
| |
Collapse
|
31
|
Bocharova AV, Stepanov VA, Marusin AV, Kharkov VN, Vagaitseva KV, Fedorenko OY, Bokhan NA, Semke AV, Ivanova SA. Association study of genetic markers of schizophrenia and its cognitive endophenotypes. RUSS J GENET+ 2017. [DOI: 10.1134/s1022795417010033] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
|
32
|
Enga RM, Rice AC, Weller P, Subler MA, Lee D, Hall CP, Windle JJ, Beardsley PM, van den Oord EJ, McClay JL. Initial characterization of behavior and ketamine response in a mouse knockout of the post-synaptic effector gene Anks1b. Neurosci Lett 2017; 641:26-32. [PMID: 28115237 DOI: 10.1016/j.neulet.2017.01.044] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2016] [Revised: 01/13/2017] [Accepted: 01/17/2017] [Indexed: 12/19/2022]
Abstract
The human ANKS1B gene encodes an activity-dependent effector of post-synaptic signaling. It was recently associated with neuropsychiatric phenotypes in genome-wide studies. While the biological function of ANKS1B has been partly elucidated, its role in behavior is poorly understood. Here, we breed and characterize a full knockout (KO) for murine Anks1b. We found that the homozygous KO genotype was partially lethal, showing significant deviation from expected segregation ratios at weaning. Behaviorally, KOs exhibited no difference in baseline acoustic startle response, but showed deficits in prepulse inhibition (PPI). KOs also exhibited locomotor hyperactivity and increased stereotypy at baseline. Administration of ketamine, a non-competitive NMDA-receptor antagonist, greatly exacerbated locomotor activity in the KOs at lower doses, but genotype groups were almost indistinguishable as dose increased. Stereotypy showed a complex response to ketamine in the KOs, with elevated stereotypy at lower doses and markedly less at high doses, compared to wild type. Our study is the first to probe the behavioral phenotypes associated with ablation of Anks1b. Deficits in PPI, locomotor hyperactivity, elevated stereotypy and altered response to NMDA receptor antagonism are murine behavioral outcomes with translational relevance for psychiatric disorders. These findings are also consistent with the role of Anks1b as an effector of glutamatergic signaling. As an intermediary between post-synaptic receptor stimulation and long-term changes to neuronal protein expression, further investigation of Anks1b is warranted.
Collapse
Affiliation(s)
- Rachel M Enga
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, Richmond, VA, USA
| | - Ann C Rice
- Department of Neurology, Virginia Commonwealth University, Richmond, VA, USA
| | - Pamela Weller
- Department of Human and Molecular Genetics, Virginia Commonwealth University, Richmond, VA, USA
| | - Mark A Subler
- Department of Human and Molecular Genetics, Virginia Commonwealth University, Richmond, VA, USA
| | - Daiyoon Lee
- Department of Pharmacotherapy and Outcomes Science, Virginia Commonwealth University, Richmond, VA, USA
| | - Chelsea P Hall
- Department of Pharmacotherapy and Outcomes Science, Virginia Commonwealth University, Richmond, VA, USA
| | - Jolene J Windle
- Department of Human and Molecular Genetics, Virginia Commonwealth University, Richmond, VA, USA
| | - Patrick M Beardsley
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, Richmond, VA, USA; Center for Biomarker Research and Precision Medicine, Virginia Commonwealth University, Richmond, VA, USA
| | - Edwin J van den Oord
- Center for Biomarker Research and Precision Medicine, Virginia Commonwealth University, Richmond, VA, USA
| | - Joseph L McClay
- Department of Pharmacotherapy and Outcomes Science, Virginia Commonwealth University, Richmond, VA, USA.
| |
Collapse
|
33
|
Luykx JJ, Olde Loohuis LM, Neeleman M, Strengman E, Bakker SC, Lentjes E, Borgdorff P, van Dongen EPA, Bruins P, Kahn RS, Horvath S, de Jong S, Ophoff RA. Peripheral blood gene expression profiles linked to monoamine metabolite levels in cerebrospinal fluid. Transl Psychiatry 2016; 6:e983. [PMID: 27959337 PMCID: PMC5290339 DOI: 10.1038/tp.2016.245] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/07/2016] [Accepted: 10/15/2016] [Indexed: 01/07/2023] Open
Abstract
The blood-brain barrier separates circulating blood from the central nervous system (CNS). The scope of this barrier is not fully understood which limits our ability to relate biological measurements from peripheral to central phenotypes. For example, it is unknown to what extent gene expression levels in peripheral blood are reflective of CNS metabolism. In this study, we examine links between central monoamine metabolite levels and whole-blood gene expression to better understand the connection between peripheral systems and the CNS. To that end, we correlated the prime monoamine metabolites in cerebrospinal fluid (CSF) with whole-genome gene expression microarray data from blood (N=240 human subjects). We additionally applied gene-enrichment analysis and weighted gene co-expression network analyses (WGCNA) to identify modules of co-expressed genes in blood that may be involved with monoamine metabolite levels in CSF. Transcript levels of two genes were significantly associated with CSF serotonin metabolite levels after Bonferroni correction for multiple testing: THAP7 (P=2.8 × 10-8, β=0.08) and DDX6 (P=2.9 × 10-7, β=0.07). Differentially expressed genes were significantly enriched for genes expressed in the brain tissue (P=6.0 × 10-52). WGCNA revealed significant correlations between serotonin metabolism and hub genes with known functions in serotonin metabolism, for example, HTR2A and COMT. We conclude that gene expression levels in whole blood are associated with monoamine metabolite levels in the human CSF. Our results, including the strong enrichment of brain-expressed genes, illustrate that gene expression profiles in peripheral blood can be relevant for quantitative metabolic phenotypes in the CNS.
Collapse
Affiliation(s)
- J J Luykx
- Department of Psychiatry, Brain Center Rudolf Magnus, University Medical Center Utrecht, Utrecht, The Netherlands,Department of Translational Neuroscience Human Neurogenetics Unit, Brain Center Rudolf Magnus, University Medical Center Utrecht, Utrecht, The Netherlands,Department of Psychiatry, ZNA Hospitals, Antwerp, Belgium
| | - L M Olde Loohuis
- Center for Neurobehavioral Genetics, Semel Institute for Neuroscience and Human Behavior, University of California, Los Angeles, Los Angeles, CA, USA
| | - M Neeleman
- Department of Psychiatry, Brain Center Rudolf Magnus, University Medical Center Utrecht, Utrecht, The Netherlands
| | - E Strengman
- Department of Pathology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - S C Bakker
- Department of Psychiatry, Brain Center Rudolf Magnus, University Medical Center Utrecht, Utrecht, The Netherlands
| | - E Lentjes
- Department of Clinical Chemistry and Hematology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - P Borgdorff
- Department of Anesthesiology, Intensive Care and Pain Management, Diakonessenhuis Hospital, Utrecht, The Netherlands
| | - E P A van Dongen
- Department of Anesthesiology, Intensive Care and Pain Management, University Medical Center Utrecht, Utrecht, The Netherlands
| | - P Bruins
- Department of Anesthesiology, Intensive Care and Pain Management, University Medical Center Utrecht, Utrecht, The Netherlands
| | - R S Kahn
- Department of Psychiatry, Brain Center Rudolf Magnus, University Medical Center Utrecht, Utrecht, The Netherlands
| | - S Horvath
- Department of Human Genetics, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA,Department of Biostatistics, Fielding School of Public Health, University of California, Los Angeles, Los Angeles, CA, USA
| | - S de Jong
- Center for Neurobehavioral Genetics, Semel Institute for Neuroscience and Human Behavior, University of California, Los Angeles, Los Angeles, CA, USA
| | - R A Ophoff
- Department of Psychiatry, Brain Center Rudolf Magnus, University Medical Center Utrecht, Utrecht, The Netherlands,Center for Neurobehavioral Genetics, Semel Institute for Neuroscience and Human Behavior, University of California, Los Angeles, Los Angeles, CA, USA,Department of Human Genetics, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA,Department of Human Genetics, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095 USA. E-mail:
| |
Collapse
|
34
|
Liu Y, Cheng Z, Wang J, Jin C, Yuan J, Wang G, Zhang F, Zhao X. No association between the rs10503253 polymorphism in the CSMD1 gene and schizophrenia in a Han Chinese population. BMC Psychiatry 2016; 16:206. [PMID: 27377754 PMCID: PMC4932686 DOI: 10.1186/s12888-016-0923-5] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/29/2015] [Accepted: 06/15/2016] [Indexed: 11/16/2022] Open
Abstract
BACKGROUND Schizophrenia (SCZ) is a complex, heritable, and devastating psychiatric disorder. Recent genome-wide association studies have identified a single-nucleotide polymorphism (SNP; rs10503253) in the CUB and SUSHI multiple domains 1 (CSMD1) gene as a risk factor for SCZ. In this study, we investigated whether the rs10503253 in CSMD1 contributes to the risk of SCZ in a Han Chinese population. METHODS We conducted a case-control study in a population from eastern China, involving 1378 SCZ patients and 1091 unrelated healthy controls, using the ligase detection reaction-polymerase chain reaction method to genotype the rs10503253 polymorphism in the CSMD1 gene. RESULTS No significant association was found between the SCZ patients and controls for any allele or genotype frequency of the SNP rs10503253 (all P > 0.05). CONCLUSIONS Our findings do not support an association between CSMD1 rs10503253 and SCZ in a Han Chinese population.
Collapse
Affiliation(s)
- Yansong Liu
- />Department of Psychosomatic Medicine, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, 200092 China
| | - Zaohuo Cheng
- />Wuxi Mental Health Center, Wuxi, 214151 Jiangsu Province China
| | - Jun Wang
- />Wuxi Mental Health Center, Wuxi, 214151 Jiangsu Province China
| | - Chunhui Jin
- />Wuxi Mental Health Center, Wuxi, 214151 Jiangsu Province China
| | - Jianmin Yuan
- />Wuxi Mental Health Center, Wuxi, 214151 Jiangsu Province China
| | - Guoqiang Wang
- />Wuxi Mental Health Center, Wuxi, 214151 Jiangsu Province China
| | - Fuquan Zhang
- />Wuxi Mental Health Center, Wuxi, 214151 Jiangsu Province China
| | - Xudong Zhao
- />Department of Psychosomatic Medicine, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, 200092 China
| |
Collapse
|
35
|
Genome-wide association study of NMDA receptor coagonists in human cerebrospinal fluid and plasma. Mol Psychiatry 2015; 20:1557-64. [PMID: 25666758 DOI: 10.1038/mp.2014.190] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/18/2014] [Revised: 11/05/2014] [Accepted: 12/08/2014] [Indexed: 01/13/2023]
Abstract
The N-methyl-D-aspartate receptor (NMDAR) coagonists glycine, D-serine and L-proline play crucial roles in NMDAR-dependent neurotransmission and are associated with a range of neuropsychiatric disorders. We conducted the first genome-wide association study of concentrations of these coagonists and their enantiomers in plasma and cerebrospinal fluid (CSF) of human subjects from the general population (N=414). Genetic variants at chromosome 22q11.2, located in and near PRODH (proline dehydrogenase), were associated with L-proline in plasma (β=0.29; P=6.38 × 10(-10)). The missense variant rs17279437 in the proline transporter SLC6A20 was associated with L-proline in CSF (β=0.28; P=9.68 × 10(-9)). Suggestive evidence of association was found for the D-serine plasma-CSF ratio at the D-amino-acid oxidase (DAO) gene (β=-0.28; P=9.08 × 10(-8)), whereas a variant in SRR (that encodes serine racemase and is associated with schizophrenia) constituted the most strongly associated locus for the L-serine to D-serine ratio in CSF. All these genes are highly expressed in rodent meninges and choroid plexus, anatomical regions relevant to CSF physiology. The enzymes and transporters they encode may be targeted to further construe the nature of NMDAR coagonist involvement in NMDAR gating. Furthermore, the highlighted genetic variants may be followed up in clinical populations, for example, schizophrenia and 22q11 deletion syndrome. Overall, this targeted metabolomics approach furthers the understanding of NMDAR coagonist concentration variability and sets the stage for non-targeted CSF metabolomics projects.
Collapse
|
36
|
Burkhardt R, Kirsten H, Beutner F, Holdt LM, Gross A, Teren A, Tönjes A, Becker S, Krohn K, Kovacs P, Stumvoll M, Teupser D, Thiery J, Ceglarek U, Scholz M. Integration of Genome-Wide SNP Data and Gene-Expression Profiles Reveals Six Novel Loci and Regulatory Mechanisms for Amino Acids and Acylcarnitines in Whole Blood. PLoS Genet 2015; 11:e1005510. [PMID: 26401656 PMCID: PMC4581711 DOI: 10.1371/journal.pgen.1005510] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2015] [Accepted: 08/17/2015] [Indexed: 01/23/2023] Open
Abstract
Profiling amino acids and acylcarnitines in whole blood spots is a powerful tool in the laboratory diagnosis of several inborn errors of metabolism. Emerging data suggests that altered blood levels of amino acids and acylcarnitines are also associated with common metabolic diseases in adults. Thus, the identification of common genetic determinants for blood metabolites might shed light on pathways contributing to human physiology and common diseases. We applied a targeted mass-spectrometry-based method to analyze whole blood concentrations of 96 amino acids, acylcarnitines and pathway associated metabolite ratios in a Central European cohort of 2,107 adults and performed genome-wide association (GWA) to identify genetic modifiers of metabolite concentrations. We discovered and replicated six novel loci associated with blood levels of total acylcarnitine, arginine (both on chromosome 6; rs12210538, rs17657775), propionylcarnitine (chromosome 10; rs12779637), 2-hydroxyisovalerylcarnitine (chromosome 21; rs1571700), stearoylcarnitine (chromosome 1; rs3811444), and aspartic acid traits (chromosome 8; rs750472). Based on an integrative analysis of expression quantitative trait loci in blood mononuclear cells and correlations between gene expressions and metabolite levels, we provide evidence for putative causative genes: SLC22A16 for total acylcarnitines, ARG1 for arginine, HLCS for 2-hydroxyisovalerylcarnitine, JAM3 for stearoylcarnitine via a trans-effect at chromosome 1, and PPP1R16A for aspartic acid traits. Further, we report replication and provide additional functional evidence for ten loci that have previously been published for metabolites measured in plasma, serum or urine. In conclusion, our integrative analysis of SNP, gene-expression and metabolite data points to novel genetic factors that may be involved in the regulation of human metabolism. At several loci, we provide evidence for metabolite regulation via gene-expression and observed overlaps with GWAS loci for common diseases. These results form a strong rationale for subsequent functional and disease-related studies.
Collapse
Affiliation(s)
- Ralph Burkhardt
- LIFE Leipzig Research Center for Civilization Diseases, University of Leipzig, Leipzig Germany
- Institute of Laboratory Medicine, Clinical Chemistry and Molecular Diagnostics, University Hospital Leipzig, Leipzig, Germany
| | - Holger Kirsten
- LIFE Leipzig Research Center for Civilization Diseases, University of Leipzig, Leipzig Germany
- Institute for Medical Informatics, Statistics and Epidemiology, University of Leipzig, Leipzig, Germany
- Department for Cell Therapy, Fraunhofer Institute for Cell Therapy and Immunology, Leipzig, Germany
| | - Frank Beutner
- LIFE Leipzig Research Center for Civilization Diseases, University of Leipzig, Leipzig Germany
- Heart Center Leipzig, Leipzig, Germany
| | - Lesca M. Holdt
- LIFE Leipzig Research Center for Civilization Diseases, University of Leipzig, Leipzig Germany
- Institute for Laboratory Medicine, Ludwig-Maximilians University Munich, Munich, Germany
| | - Arnd Gross
- LIFE Leipzig Research Center for Civilization Diseases, University of Leipzig, Leipzig Germany
- Institute for Medical Informatics, Statistics and Epidemiology, University of Leipzig, Leipzig, Germany
| | - Andrej Teren
- LIFE Leipzig Research Center for Civilization Diseases, University of Leipzig, Leipzig Germany
- Heart Center Leipzig, Leipzig, Germany
| | - Anke Tönjes
- Medical Department, Clinic for Endocrinology and Nephrology, University of Leipzig, Leipzig, Germany
| | - Susen Becker
- LIFE Leipzig Research Center for Civilization Diseases, University of Leipzig, Leipzig Germany
- Institute of Laboratory Medicine, Clinical Chemistry and Molecular Diagnostics, University Hospital Leipzig, Leipzig, Germany
| | - Knut Krohn
- LIFE Leipzig Research Center for Civilization Diseases, University of Leipzig, Leipzig Germany
- Interdisciplinary Centre for Clinical Research, University of Leipzig, Leipzig, Germany
| | - Peter Kovacs
- Integrated Research and Treatment Center Adiposity Diseases, University of Leipzig, Leipzig Germany
| | - Michael Stumvoll
- Medical Department, Clinic for Endocrinology and Nephrology, University of Leipzig, Leipzig, Germany
- Integrated Research and Treatment Center Adiposity Diseases, University of Leipzig, Leipzig Germany
| | - Daniel Teupser
- LIFE Leipzig Research Center for Civilization Diseases, University of Leipzig, Leipzig Germany
- Institute for Laboratory Medicine, Ludwig-Maximilians University Munich, Munich, Germany
| | - Joachim Thiery
- LIFE Leipzig Research Center for Civilization Diseases, University of Leipzig, Leipzig Germany
- Institute of Laboratory Medicine, Clinical Chemistry and Molecular Diagnostics, University Hospital Leipzig, Leipzig, Germany
| | - Uta Ceglarek
- LIFE Leipzig Research Center for Civilization Diseases, University of Leipzig, Leipzig Germany
- Institute of Laboratory Medicine, Clinical Chemistry and Molecular Diagnostics, University Hospital Leipzig, Leipzig, Germany
| | - Markus Scholz
- LIFE Leipzig Research Center for Civilization Diseases, University of Leipzig, Leipzig Germany
- Institute for Medical Informatics, Statistics and Epidemiology, University of Leipzig, Leipzig, Germany
- * E-mail:
| |
Collapse
|
37
|
Fond G, d'Albis MA, Jamain S, Tamouza R, Arango C, Fleischhacker WW, Glenthøj B, Leweke M, Lewis S, McGuire P, Meyer-Lindenberg A, Sommer IE, Winter-van Rossum I, Kapur S, Kahn RS, Rujescu D, Leboyer M. The promise of biological markers for treatment response in first-episode psychosis: a systematic review. Schizophr Bull 2015; 41:559-73. [PMID: 25759473 PMCID: PMC4393702 DOI: 10.1093/schbul/sbv002] [Citation(s) in RCA: 75] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Successful treatment of first-episode psychosis is one of the major factors that impacts long-term prognosis. Currently, there are no satisfactory biological markers (biomarkers) to predict which patients with a first-episode psychosis will respond to which treatment. In addition, a non-negligible rate of patients does not respond to any treatment or may develop side effects that affect adherence to the treatments as well as negatively impact physical health. Thus, there clearly is a pressing need for defining biomarkers that may be helpful to predict response to treatment and sensitivity to side effects in first-episode psychosis. The present systematic review provides (1) trials that assessed biological markers associated with antipsychotic response or side effects in first-episode psychosis and (2) potential biomarkers associated with biological disturbances that may guide the choice of conventional treatments or the prescription of innovative treatments. Trials including first-episode psychoses are few in number. Most of the available data focused on pharmacogenetics markers with so far only preliminary results. To date, these studies yielded-beside markers for metabolism of antipsychotics-no or only a few biomarkers for response or side effects, none of which have been implemented in daily clinical practice. Other biomarkers exploring immunoinflammatory, oxidative, and hormonal disturbances emerged as biomarkers of first-episode psychoses in the last decades, and some of them have been associated with treatment response. In addition to pharmacogenetics, further efforts should focus on the association of emergent biomarkers with conventional treatments or with innovative therapies efficacy, where some preliminary data suggest promising results.
Collapse
Affiliation(s)
| | | | | | - Ryad Tamouza
- Jean Dausset Laboratory & INSERM, UMRS 940, Hôpital Saint Louis, Paris, France
| | - Celso Arango
- Child and Adolescent Psychiatry Department, Hospital General Universitario Gregorio Marañón, IiSGM, School of Medicine, Universidad Complutense, CIBERSAM, Madrid, Spain
| | | | - Birte Glenthøj
- Center for Neuropsychiatric Schizophrenia Research & Center for Clinical Intervention and Neuropsychiatric Schizophrenia Research, Psychiatric Hospital Center Glostrup, University of Copenhagen, Faculty of Health and Medical Sciences, Denmark
| | - Markus Leweke
- Central Institute of Mental Health, University of Heidelberg/Medical Faculty Mannheim, Mannheim, Germany
| | - Shôn Lewis
- Institute of Brain, Behaviour and Mental Health, University of Manchester, Manchester, UK
| | - Phillip McGuire
- Department of Psychosis Studies, Institute of Psychiatry, King's College London, London, UK
| | - Andreas Meyer-Lindenberg
- Central Institute of Mental Health, University of Heidelberg/Medical Faculty Mannheim, Mannheim, Germany
| | - Iris E Sommer
- Department of Psychiatry, Brain Center Rudolf Magnus, UMC Utrecht, Utrecht, The Netherlands
| | - Inge Winter-van Rossum
- Department of Psychiatry, Brain Center Rudolf Magnus, UMC Utrecht, Utrecht, The Netherlands
| | - Shitij Kapur
- Institute of Psychiatry, King's College London, London, UK
| | - René S Kahn
- Department of Psychiatry, Brain Center Rudolf Magnus, UMC Utrecht, Utrecht, The Netherlands
| | - Dan Rujescu
- Department of Psychiatry, University of Halle, Halle, Germany
| | | |
Collapse
|
38
|
Re-annotation of presumed noncoding disease/trait-associated genetic variants by integrative analyses. Sci Rep 2015; 5:9453. [PMID: 25819875 PMCID: PMC4377585 DOI: 10.1038/srep09453] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2014] [Accepted: 03/02/2015] [Indexed: 11/08/2022] Open
Abstract
Using RefSeq annotations, most disease/trait-associated genetic variants identified by genome-wide association studies (GWAS) appear to be located within intronic or intergenic regions, which makes it difficult to interpret their functions. We reassessed GWAS-Associated single-nucleotide polymorphisms (herein termed as GASs) for their potential functionalities using integrative approaches. 8834 of 9184 RefSeq “noncoding” GASs were reassessed to have potential regulatory functionalities. As examples, 3 variants (rs3130320, rs3806932 and rs6890853) were shown to have regulatory properties in HepG2, A549 and 293T cells. Except rs3130320 as a known expression quantitative trait loci (eQTL), rs3806932 and rs6890853 were not reported as eQTLs in previous reports. 1999 of 9184 “noncoding” GASs were re-annotated to the promoters or intragenic regions using Ensembl, UCSC and AceView gene annotations but they were not annotated into corresponding regions in RefSeq database. Moreover, these GAS-harboring genes were broadly expressed across different tissues and a portion of them was expressed in a tissue-specific manner, suggesting that they could be functional. Collectively, our study demonstrates the benefits of using integrative analyses to interpret genetic variants and may help to predict or explain disease susceptibility more accurately and comprehensively.
Collapse
|
39
|
Abstract
This review considers pharmacogenetics of the so called 'second-generation' antipsychotics. Findings for polymorphisms replicating in more than one study are emphasized and compared and contrasted with larger-scale candidate gene studies and genome-wide association study analyses. Variants in three types of genes are discussed: pharmacokinetic genes associated with drug metabolism and disposition, pharmacodynamic genes encoding drug targets, and pharmacotypic genes impacting disease presentation and subtype. Among pharmacokinetic markers, CYP2D6 metabolizer phenotype has clear clinical significance, as it impacts dosing considerations for aripiprazole, iloperidone and risperidone, and variants of the ABCB1 gene hold promise as biomarkers for dosing for olanzapine and clozapine. Among pharmacodynamic variants, the TaqIA1 allele of the DRD2 gene, the DRD3 (Ser9Gly) polymorphism, and the HTR2C -759C/T polymorphism have emerged as potential biomarkers for response and/or side effects. However, large-scale candidate gene studies and genome-wide association studies indicate that pharmacotypic genes may ultimately prove to be the richest source of biomarkers for response and side effect profiles for second-generation antipsychotics.
Collapse
Affiliation(s)
- Mark D Brennan
- Department of Biochemistry & Molecular Biology, School of Medicine, University of Louisville, Louisville, KY 40292, USA.
| |
Collapse
|
40
|
Albersen M, Bosma M, Luykx JJ, Jans JJM, Bakker SC, Strengman E, Borgdorff PJ, Keijzers PJM, van Dongen EPA, Bruins P, de Sain-van der Velden MGM, Visser G, Knoers NVVAM, Ophoff RA, Verhoeven-Duif NM. Vitamin B-6 vitamers in human plasma and cerebrospinal fluid. Am J Clin Nutr 2014; 100:587-92. [PMID: 24808484 DOI: 10.3945/ajcn.113.082008] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
BACKGROUND Vitamin B-6 comprises a group of 6 interrelated vitamers and is essential for numerous physiologic processes, including brain functioning. Genetic disorders disrupting vitamin B-6 metabolism have severe clinical consequences. OBJECTIVE To adequately diagnose known and novel disorders in vitamin B-6 metabolism, a reference set is required containing information on all vitamin B-6 vitamers in plasma and cerebrospinal fluid (CSF). DESIGN Concentrations of vitamin B-6 vitamers in the plasma and CSF of 533 adult subjects were measured by ultra high-performance liquid chromatography-tandem mass spectrometry. RESULTS The relative vitamin B-6 vitamer composition of plasma [pyridoxal phosphate (PLP) > pyridoxic acid (PA) > pyridoxal] differed from that of CSF (pyridoxal > PLP > PA > pyridoxamine). Sex influenced vitamin B-6 vitamer concentrations in plasma and CSF and should therefore be taken into account when interpreting vitamin B-6 vitamer concentrations. The strict ratios and strong correlations between vitamin B-6 vitamers point to a tight regulation of vitamin B-6 vitamer concentrations in blood and CSF. Given the unique design of this study, with simultaneously withdrawn blood and CSF from a large number of subjects, reliable CSF:plasma ratios and correlations of vitamin B-6 vitamers could be established. CONCLUSIONS We provide an extensive reference set of vitamin B-6 vitamer concentrations in plasma and CSF. In addition to providing insight on the regulation of individual vitamers and their intercompartmental distribution, we anticipate that these data will prove to be a valuable reference set for the diagnosis and treatment of conditions associated with altered vitamin B-6 metabolism.
Collapse
Affiliation(s)
- Monique Albersen
- Department of Medical Genetics, University Medical Center Utrecht, Utrecht, Netherlands (MA, MB, JJMJ, ES, MGMdS-vdV, NVVAMK, and NMV-D); the Neurogenetics Unit (JJL) and the Department of Psychiatry (SCB and RAO), Brain Center Rudolf Magnus, University Medical Center Utrecht, Utrecht, Netherlands; the Department of Psychiatry, ZNA Hospitals, Antwerp, Belgium (JJL); the Department of Anesthesiology, Intensive Care and Pain Management, Diakonessenhuis Hospital, Utrecht, Netherlands (PJB); the Department of Anesthesiology, Central Military Hospital, Utrecht, Netherlands (PJMK); the Department of Anesthesiology, Intensive Care and Pain Management, St Antonius Hospital, Nieuwegein, Netherlands (EPAvD and PB); the Department of Pediatric Metabolic Diseases, Wilhelmina Children's Hospital, University Medical Center Utrecht, Utrecht, Netherlands (GV); and the Center for Neurobehavioral Genetics, Semel Institute for Neuroscience and Human Behavior, University of California Los Angeles, Los Angeles, CA (RAO)
| | - Marjolein Bosma
- Department of Medical Genetics, University Medical Center Utrecht, Utrecht, Netherlands (MA, MB, JJMJ, ES, MGMdS-vdV, NVVAMK, and NMV-D); the Neurogenetics Unit (JJL) and the Department of Psychiatry (SCB and RAO), Brain Center Rudolf Magnus, University Medical Center Utrecht, Utrecht, Netherlands; the Department of Psychiatry, ZNA Hospitals, Antwerp, Belgium (JJL); the Department of Anesthesiology, Intensive Care and Pain Management, Diakonessenhuis Hospital, Utrecht, Netherlands (PJB); the Department of Anesthesiology, Central Military Hospital, Utrecht, Netherlands (PJMK); the Department of Anesthesiology, Intensive Care and Pain Management, St Antonius Hospital, Nieuwegein, Netherlands (EPAvD and PB); the Department of Pediatric Metabolic Diseases, Wilhelmina Children's Hospital, University Medical Center Utrecht, Utrecht, Netherlands (GV); and the Center for Neurobehavioral Genetics, Semel Institute for Neuroscience and Human Behavior, University of California Los Angeles, Los Angeles, CA (RAO)
| | - Jurjen J Luykx
- Department of Medical Genetics, University Medical Center Utrecht, Utrecht, Netherlands (MA, MB, JJMJ, ES, MGMdS-vdV, NVVAMK, and NMV-D); the Neurogenetics Unit (JJL) and the Department of Psychiatry (SCB and RAO), Brain Center Rudolf Magnus, University Medical Center Utrecht, Utrecht, Netherlands; the Department of Psychiatry, ZNA Hospitals, Antwerp, Belgium (JJL); the Department of Anesthesiology, Intensive Care and Pain Management, Diakonessenhuis Hospital, Utrecht, Netherlands (PJB); the Department of Anesthesiology, Central Military Hospital, Utrecht, Netherlands (PJMK); the Department of Anesthesiology, Intensive Care and Pain Management, St Antonius Hospital, Nieuwegein, Netherlands (EPAvD and PB); the Department of Pediatric Metabolic Diseases, Wilhelmina Children's Hospital, University Medical Center Utrecht, Utrecht, Netherlands (GV); and the Center for Neurobehavioral Genetics, Semel Institute for Neuroscience and Human Behavior, University of California Los Angeles, Los Angeles, CA (RAO)
| | - Judith J M Jans
- Department of Medical Genetics, University Medical Center Utrecht, Utrecht, Netherlands (MA, MB, JJMJ, ES, MGMdS-vdV, NVVAMK, and NMV-D); the Neurogenetics Unit (JJL) and the Department of Psychiatry (SCB and RAO), Brain Center Rudolf Magnus, University Medical Center Utrecht, Utrecht, Netherlands; the Department of Psychiatry, ZNA Hospitals, Antwerp, Belgium (JJL); the Department of Anesthesiology, Intensive Care and Pain Management, Diakonessenhuis Hospital, Utrecht, Netherlands (PJB); the Department of Anesthesiology, Central Military Hospital, Utrecht, Netherlands (PJMK); the Department of Anesthesiology, Intensive Care and Pain Management, St Antonius Hospital, Nieuwegein, Netherlands (EPAvD and PB); the Department of Pediatric Metabolic Diseases, Wilhelmina Children's Hospital, University Medical Center Utrecht, Utrecht, Netherlands (GV); and the Center for Neurobehavioral Genetics, Semel Institute for Neuroscience and Human Behavior, University of California Los Angeles, Los Angeles, CA (RAO)
| | - Steven C Bakker
- From the Department of Medical Genetics, University Medical Center Utrecht, Utrecht, Netherlands (MA, MB, JJMJ, ES, MGMdS-vdV, NVVAMK, and NMV-D); the Neurogenetics Unit (JJL) and the Department of Psychiatry (SCB and RAO), Brain Center Rudolf Magnus, University Medical Center Utrecht, Utrecht, Netherlands; the Department of Psychiatry, ZNA Hospitals, Antwerp, Belgium (JJL); the Department of Anesthesiology, Intensive Care and Pain Management, Diakonessenhuis Hospital, Utrecht, Netherlands (PJB); the Department of Anesthesiology, Central Military Hospital, Utrecht, Netherlands (PJMK); the Department of Anesthesiology, Intensive Care and Pain Management, St Antonius Hospital, Nieuwegein, Netherlands (EPAvD and PB); the Department of Pediatric Metabolic Diseases, Wilhelmina Children's Hospital, University Medical Center Utrecht, Utrecht, Netherlands (GV); and the Center for Neurobehavioral Genetics, Semel Institute for Neuroscience and Human Behavior, University of California Los Angeles, Los Angeles, CA (RAO)
| | - Eric Strengman
- Department of Medical Genetics, University Medical Center Utrecht, Utrecht, Netherlands (MA, MB, JJMJ, ES, MGMdS-vdV, NVVAMK, and NMV-D); the Neurogenetics Unit (JJL) and the Department of Psychiatry (SCB and RAO), Brain Center Rudolf Magnus, University Medical Center Utrecht, Utrecht, Netherlands; the Department of Psychiatry, ZNA Hospitals, Antwerp, Belgium (JJL); the Department of Anesthesiology, Intensive Care and Pain Management, Diakonessenhuis Hospital, Utrecht, Netherlands (PJB); the Department of Anesthesiology, Central Military Hospital, Utrecht, Netherlands (PJMK); the Department of Anesthesiology, Intensive Care and Pain Management, St Antonius Hospital, Nieuwegein, Netherlands (EPAvD and PB); the Department of Pediatric Metabolic Diseases, Wilhelmina Children's Hospital, University Medical Center Utrecht, Utrecht, Netherlands (GV); and the Center for Neurobehavioral Genetics, Semel Institute for Neuroscience and Human Behavior, University of California Los Angeles, Los Angeles, CA (RAO)
| | - Paul J Borgdorff
- Department of Medical Genetics, University Medical Center Utrecht, Utrecht, Netherlands (MA, MB, JJMJ, ES, MGMdS-vdV, NVVAMK, and NMV-D); the Neurogenetics Unit (JJL) and the Department of Psychiatry (SCB and RAO), Brain Center Rudolf Magnus, University Medical Center Utrecht, Utrecht, Netherlands; the Department of Psychiatry, ZNA Hospitals, Antwerp, Belgium (JJL); the Department of Anesthesiology, Intensive Care and Pain Management, Diakonessenhuis Hospital, Utrecht, Netherlands (PJB); the Department of Anesthesiology, Central Military Hospital, Utrecht, Netherlands (PJMK); the Department of Anesthesiology, Intensive Care and Pain Management, St Antonius Hospital, Nieuwegein, Netherlands (EPAvD and PB); the Department of Pediatric Metabolic Diseases, Wilhelmina Children's Hospital, University Medical Center Utrecht, Utrecht, Netherlands (GV); and the Center for Neurobehavioral Genetics, Semel Institute for Neuroscience and Human Behavior, University of California Los Angeles, Los Angeles, CA (RAO)
| | - Peter J M Keijzers
- From the Department of Medical Genetics, University Medical Center Utrecht, Utrecht, Netherlands (MA, MB, JJMJ, ES, MGMdS-vdV, NVVAMK, and NMV-D); the Neurogenetics Unit (JJL) and the Department of Psychiatry (SCB and RAO), Brain Center Rudolf Magnus, University Medical Center Utrecht, Utrecht, Netherlands; the Department of Psychiatry, ZNA Hospitals, Antwerp, Belgium (JJL); the Department of Anesthesiology, Intensive Care and Pain Management, Diakonessenhuis Hospital, Utrecht, Netherlands (PJB); the Department of Anesthesiology, Central Military Hospital, Utrecht, Netherlands (PJMK); the Department of Anesthesiology, Intensive Care and Pain Management, St Antonius Hospital, Nieuwegein, Netherlands (EPAvD and PB); the Department of Pediatric Metabolic Diseases, Wilhelmina Children's Hospital, University Medical Center Utrecht, Utrecht, Netherlands (GV); and the Center for Neurobehavioral Genetics, Semel Institute for Neuroscience and Human Behavior, University of California Los Angeles, Los Angeles, CA (RAO)
| | - Eric P A van Dongen
- Department of Medical Genetics, University Medical Center Utrecht, Utrecht, Netherlands (MA, MB, JJMJ, ES, MGMdS-vdV, NVVAMK, and NMV-D); the Neurogenetics Unit (JJL) and the Department of Psychiatry (SCB and RAO), Brain Center Rudolf Magnus, University Medical Center Utrecht, Utrecht, Netherlands; the Department of Psychiatry, ZNA Hospitals, Antwerp, Belgium (JJL); the Department of Anesthesiology, Intensive Care and Pain Management, Diakonessenhuis Hospital, Utrecht, Netherlands (PJB); the Department of Anesthesiology, Central Military Hospital, Utrecht, Netherlands (PJMK); the Department of Anesthesiology, Intensive Care and Pain Management, St Antonius Hospital, Nieuwegein, Netherlands (EPAvD and PB); the Department of Pediatric Metabolic Diseases, Wilhelmina Children's Hospital, University Medical Center Utrecht, Utrecht, Netherlands (GV); and the Center for Neurobehavioral Genetics, Semel Institute for Neuroscience and Human Behavior, University of California Los Angeles, Los Angeles, CA (RAO)
| | - Peter Bruins
- Department of Medical Genetics, University Medical Center Utrecht, Utrecht, Netherlands (MA, MB, JJMJ, ES, MGMdS-vdV, NVVAMK, and NMV-D); the Neurogenetics Unit (JJL) and the Department of Psychiatry (SCB and RAO), Brain Center Rudolf Magnus, University Medical Center Utrecht, Utrecht, Netherlands; the Department of Psychiatry, ZNA Hospitals, Antwerp, Belgium (JJL); the Department of Anesthesiology, Intensive Care and Pain Management, Diakonessenhuis Hospital, Utrecht, Netherlands (PJB); the Department of Anesthesiology, Central Military Hospital, Utrecht, Netherlands (PJMK); the Department of Anesthesiology, Intensive Care and Pain Management, St Antonius Hospital, Nieuwegein, Netherlands (EPAvD and PB); the Department of Pediatric Metabolic Diseases, Wilhelmina Children's Hospital, University Medical Center Utrecht, Utrecht, Netherlands (GV); and the Center for Neurobehavioral Genetics, Semel Institute for Neuroscience and Human Behavior, University of California Los Angeles, Los Angeles, CA (RAO)
| | - Monique G M de Sain-van der Velden
- Department of Medical Genetics, University Medical Center Utrecht, Utrecht, Netherlands (MA, MB, JJMJ, ES, MGMdS-vdV, NVVAMK, and NMV-D); the Neurogenetics Unit (JJL) and the Department of Psychiatry (SCB and RAO), Brain Center Rudolf Magnus, University Medical Center Utrecht, Utrecht, Netherlands; the Department of Psychiatry, ZNA Hospitals, Antwerp, Belgium (JJL); the Department of Anesthesiology, Intensive Care and Pain Management, Diakonessenhuis Hospital, Utrecht, Netherlands (PJB); the Department of Anesthesiology, Central Military Hospital, Utrecht, Netherlands (PJMK); the Department of Anesthesiology, Intensive Care and Pain Management, St Antonius Hospital, Nieuwegein, Netherlands (EPAvD and PB); the Department of Pediatric Metabolic Diseases, Wilhelmina Children's Hospital, University Medical Center Utrecht, Utrecht, Netherlands (GV); and the Center for Neurobehavioral Genetics, Semel Institute for Neuroscience and Human Behavior, University of California Los Angeles, Los Angeles, CA (RAO)
| | - Gepke Visser
- Department of Medical Genetics, University Medical Center Utrecht, Utrecht, Netherlands (MA, MB, JJMJ, ES, MGMdS-vdV, NVVAMK, and NMV-D); the Neurogenetics Unit (JJL) and the Department of Psychiatry (SCB and RAO), Brain Center Rudolf Magnus, University Medical Center Utrecht, Utrecht, Netherlands; the Department of Psychiatry, ZNA Hospitals, Antwerp, Belgium (JJL); the Department of Anesthesiology, Intensive Care and Pain Management, Diakonessenhuis Hospital, Utrecht, Netherlands (PJB); the Department of Anesthesiology, Central Military Hospital, Utrecht, Netherlands (PJMK); the Department of Anesthesiology, Intensive Care and Pain Management, St Antonius Hospital, Nieuwegein, Netherlands (EPAvD and PB); the Department of Pediatric Metabolic Diseases, Wilhelmina Children's Hospital, University Medical Center Utrecht, Utrecht, Netherlands (GV); and the Center for Neurobehavioral Genetics, Semel Institute for Neuroscience and Human Behavior, University of California Los Angeles, Los Angeles, CA (RAO)
| | - Nine V V A M Knoers
- Department of Medical Genetics, University Medical Center Utrecht, Utrecht, Netherlands (MA, MB, JJMJ, ES, MGMdS-vdV, NVVAMK, and NMV-D); the Neurogenetics Unit (JJL) and the Department of Psychiatry (SCB and RAO), Brain Center Rudolf Magnus, University Medical Center Utrecht, Utrecht, Netherlands; the Department of Psychiatry, ZNA Hospitals, Antwerp, Belgium (JJL); the Department of Anesthesiology, Intensive Care and Pain Management, Diakonessenhuis Hospital, Utrecht, Netherlands (PJB); the Department of Anesthesiology, Central Military Hospital, Utrecht, Netherlands (PJMK); the Department of Anesthesiology, Intensive Care and Pain Management, St Antonius Hospital, Nieuwegein, Netherlands (EPAvD and PB); the Department of Pediatric Metabolic Diseases, Wilhelmina Children's Hospital, University Medical Center Utrecht, Utrecht, Netherlands (GV); and the Center for Neurobehavioral Genetics, Semel Institute for Neuroscience and Human Behavior, University of California Los Angeles, Los Angeles, CA (RAO)
| | - Roel A Ophoff
- Department of Medical Genetics, University Medical Center Utrecht, Utrecht, Netherlands (MA, MB, JJMJ, ES, MGMdS-vdV, NVVAMK, and NMV-D); the Neurogenetics Unit (JJL) and the Department of Psychiatry (SCB and RAO), Brain Center Rudolf Magnus, University Medical Center Utrecht, Utrecht, Netherlands; the Department of Psychiatry, ZNA Hospitals, Antwerp, Belgium (JJL); the Department of Anesthesiology, Intensive Care and Pain Management, Diakonessenhuis Hospital, Utrecht, Netherlands (PJB); the Department of Anesthesiology, Central Military Hospital, Utrecht, Netherlands (PJMK); the Department of Anesthesiology, Intensive Care and Pain Management, St Antonius Hospital, Nieuwegein, Netherlands (EPAvD and PB); the Department of Pediatric Metabolic Diseases, Wilhelmina Children's Hospital, University Medical Center Utrecht, Utrecht, Netherlands (GV); and the Center for Neurobehavioral Genetics, Semel Institute for Neuroscience and Human Behavior, University of California Los Angeles, Los Angeles, CA (RAO)
| | - Nanda M Verhoeven-Duif
- Department of Medical Genetics, University Medical Center Utrecht, Utrecht, Netherlands (MA, MB, JJMJ, ES, MGMdS-vdV, NVVAMK, and NMV-D); the Neurogenetics Unit (JJL) and the Department of Psychiatry (SCB and RAO), Brain Center Rudolf Magnus, University Medical Center Utrecht, Utrecht, Netherlands; the Department of Psychiatry, ZNA Hospitals, Antwerp, Belgium (JJL); the Department of Anesthesiology, Intensive Care and Pain Management, Diakonessenhuis Hospital, Utrecht, Netherlands (PJB); the Department of Anesthesiology, Central Military Hospital, Utrecht, Netherlands (PJMK); the Department of Anesthesiology, Intensive Care and Pain Management, St Antonius Hospital, Nieuwegein, Netherlands (EPAvD and PB); the Department of Pediatric Metabolic Diseases, Wilhelmina Children's Hospital, University Medical Center Utrecht, Utrecht, Netherlands (GV); and the Center for Neurobehavioral Genetics, Semel Institute for Neuroscience and Human Behavior, University of California Los Angeles, Los Angeles, CA (RAO)
| |
Collapse
|
41
|
Plongthongkum N, van Eijk KR, de Jong S, Wang T, Sul JH, Boks MPM, Kahn RS, Fung HL, Ophoff RA, Zhang K. Characterization of genome-methylome interactions in 22 nuclear pedigrees. PLoS One 2014; 9:e99313. [PMID: 25019935 PMCID: PMC4096397 DOI: 10.1371/journal.pone.0099313] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2013] [Accepted: 05/13/2014] [Indexed: 12/19/2022] Open
Abstract
Genetic polymorphisms can shape the global landscape of DNA methylation, by either changing substrates for DNA methyltransferases or altering the DNA binding affinity of cis-regulatory proteins. The interactions between CpG methylation and genetic polymorphisms have been previously investigated by methylation quantitative trait loci (mQTL) and allele-specific methylation (ASM) analysis. However, it remains unclear whether these approaches can effectively and comprehensively identify all genetic variants that contribute to the inter-individual variation of DNA methylation levels. Here we used three independent approaches to systematically investigate the influence of genetic polymorphisms on variability in DNA methylation by characterizing the methylation state of 96 whole blood samples in 52 parent-child trios from 22 nuclear pedigrees. We performed targeted bisulfite sequencing with padlock probes to quantify the absolute DNA methylation levels at a set of 411,800 CpG sites in the human genome. With mid-parent offspring analysis (MPO), we identified 10,593 CpG sites that exhibited heritable methylation patterns, among which 70.1% were SNPs directly present in methylated CpG dinucleotides. We determined the mQTL analysis identified 49.9% of heritable CpG sites for which regulation occurred in a distal cis-regulatory manner, and that ASM analysis was only able to identify 5%. Finally, we identified hundreds of clusters in the human genome for which the degree of variation of CpG methylation, as opposed to whether or not CpG sites were methylated, was associated with genetic polymorphisms, supporting a recent hypothesis on the genetic influence of phenotypic plasticity. These results show that cis-regulatory SNPs identified by mQTL do not comprise the full extent of heritable CpG methylation, and that ASM appears overall unreliable. Overall, the extent of genome-methylome interactions is well beyond what is detectible with the commonly used mQTL and ASM approaches, and is likely to include effects on plasticity.
Collapse
Affiliation(s)
- Nongluk Plongthongkum
- Department of Bioengineering, University of California San Diego, La Jolla, California, United States of America
| | - Kristel R. van Eijk
- Department of Psychiatry, Rudolf Magnus Institute of Neuroscience, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Simone de Jong
- Center for Neurobehavioral Genetics, Semel Institute for Neuroscience and Behavior, David Geffen School of Medicine at the University of California Los Angeles, Los Angeles, California, United States of America
| | - Tina Wang
- Center for Neurobehavioral Genetics, Semel Institute for Neuroscience and Behavior, David Geffen School of Medicine at the University of California Los Angeles, Los Angeles, California, United States of America
| | - Jae Hoon Sul
- Department of Computer Science, University of California Los Angeles, Los Angeles, California, United States of America
| | - Marco P. M. Boks
- Department of Psychiatry, Rudolf Magnus Institute of Neuroscience, University Medical Center Utrecht, Utrecht, The Netherlands
| | - René S. Kahn
- Department of Psychiatry, Rudolf Magnus Institute of Neuroscience, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Ho-Lim Fung
- Department of Bioengineering, University of California San Diego, La Jolla, California, United States of America
| | - Roel A. Ophoff
- Center for Neurobehavioral Genetics, Semel Institute for Neuroscience and Behavior, David Geffen School of Medicine at the University of California Los Angeles, Los Angeles, California, United States of America
- Department of Psychiatry, Rudolf Magnus Institute of Neuroscience, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Kun Zhang
- Department of Bioengineering, University of California San Diego, La Jolla, California, United States of America
| |
Collapse
|
42
|
Yokoyama JS, Evans DS, Coppola G, Kramer JH, Tranah GJ, Yaffe K. Genetic modifiers of cognitive maintenance among older adults. Hum Brain Mapp 2014; 35:4556-65. [PMID: 24616004 PMCID: PMC4107001 DOI: 10.1002/hbm.22494] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2013] [Revised: 02/06/2014] [Accepted: 02/07/2014] [Indexed: 02/02/2023] Open
Abstract
Objective Identify genetic factors associated with cognitive maintenance in late life and assess their association with gray matter (GM) volume in brain networks affected in aging. Methods We conducted a genome‐wide association study of ∼2.4 M markers to identify modifiers of cognitive trajectories in Caucasian participants (N = 7,328) from two population‐based cohorts of non‐demented elderly. Standardized measures of global cognitive function (z‐scores) over 10 and 6 years were calculated among participants and mixed model regression was used to determine subject‐specific cognitive slopes. “Cognitive maintenance” was defined as a change in slope of ≥ 0 and was compared with all cognitive decliners (slope < 0). In an independent cohort of cognitively normal older Caucasians adults (N = 122), top association findings were then used to create genetic scores to assess whether carrying more cognitive maintenance alleles was associated with greater GM volume in specific brain networks using voxel‐based morphometry. Results The most significant association was on chromosome 11 (rs7109806, P = 7.8 × 10−8) near RIC3. RIC3 modulates activity of α7 nicotinic acetylcholine receptors, which have been implicated in synaptic plasticity and beta‐amyloid binding. In the neuroimaging cohort, carrying more cognitive maintenance alleles was associated with greater volume in the right executive control network (RECN; PFWE = 0.01). Conclusions These findings suggest that there may be genetic loci that promote healthy cognitive aging and that they may do so by conferring robustness to GM in the RECN. Future work is required to validate top candidate genes such as RIC3 for involvement in cognitive maintenance. Hum Brain Mapp 35:4556–4565, 2014. © 2014 The Authors. Human Brain Mapping Published by Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Jennifer S Yokoyama
- Department of Neurology, Memory and Aging Center, University of California, San Francisco, California
| | | | | | | | | | | |
Collapse
|
43
|
Onaivi ES, Schanz N, Lin ZC. Psychiatric disturbances regulate the innate immune system in CSF of conscious mice. Transl Psychiatry 2014; 4:e367. [PMID: 24594778 PMCID: PMC3966044 DOI: 10.1038/tp.2014.5] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/03/2013] [Revised: 11/26/2013] [Accepted: 12/07/2013] [Indexed: 12/18/2022] Open
Abstract
Environment may affect brain activity through cerebrospinal fluid (CSF) only if there are regulatory molecules or cascades in CSF that are sensitive to external stimuli. This study was designed to identify regulatory activity present in CSF, better elucidating environmental regulation of brain function. By using cannulation-based sequential CSF sampling coupled with mass spectrometry-based identification and quantification of proteins, we show that the naive mouse CSF harbors, among 22 other pathways, the innate immune system as a main pathway, which was downregulated and upregulated, respectively, by acute stressor (AS) and acute cocaine (AC) administrations. Among novel processes and molecular functions, AS also regulated schizophrenia-associated proteins. Furthermore, AC upregulated exosome-related proteins with a false discovery rate of 1.0 × 10(-)(16). These results suggest that psychiatric disturbances regulate the neuroimmune system and brain disorder-related proteins, presenting a sensitive approach to investigating extracellular mechanisms in conscious and various mouse models of psychiatric disorders.
Collapse
Affiliation(s)
- E S Onaivi
- Department of Biology, William Paterson University, Wayne, NJ, USA
| | - N Schanz
- Department of Biology, William Paterson University, Wayne, NJ, USA
| | - Z C Lin
- Department of Psychiatry, Harvard Medical School, Boston, MA, USA,NeuroDiscovery Center, Harvard Medical School, Boston, MA, USA,Laboratory of Psychiatric Neurogenomics, Division of Alcohol and Drug Abuse, McLean Hospital, Belmont, MA, USA,Laboratory of Psychiatric Neurogenomics, Division of Alcohol and Drug Abuse, McLean Hospital Mailstop 318, 115 Mill Street, Belmont 02478, MA, USA. E-mail:
| |
Collapse
|
44
|
De Jong S, Neeleman M, Luykx JJ, ten Berg MJ, Strengman E, Den Breeijen HH, Stijvers LC, Buizer-Voskamp JE, Bakker SC, Kahn RS, Horvath S, Van Solinge WW, Ophoff RA. Seasonal changes in gene expression represent cell-type composition in whole blood. Hum Mol Genet 2014; 23:2721-8. [PMID: 24399446 DOI: 10.1093/hmg/ddt665] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
Seasonal patterns in behavior and biological parameters are widespread. Here, we examined seasonal changes in whole blood gene expression profiles of 233 healthy subjects. Using weighted gene co-expression network analysis, we identified three co-expression modules showing circannual patterns. Enrichment analysis suggested that this signal stems primarily from red blood cells and blood platelets. Indeed, a large clinical database with 51 142 observations of blood cell counts over 3 years confirmed a corresponding seasonal pattern of counts of red blood cells, reticulocytes and platelets. We found no direct evidence that these changes are linked to genes known to be key players in regulating immune function or circadian rhythm. It is likely, however, that these seasonal changes in cell counts and gene expression profiles in whole blood represent biological and clinical relevant phenomena. Moreover, our findings highlight possible confounding factors relevant to the study of gene expression profiles in subjects collected at geographical locations with disparaging seasonality patterns.
Collapse
Affiliation(s)
- Simone De Jong
- Center for Neurobehavioral Genetics, Semel Institute for Neuroscience and Human Behavior, University of California, Los Angeles, Los Angeles, CA, USA
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Wolock S, Yates A, Petrill SA, Bohland JW, Blair C, Li N, Machiraju R, Huang K, Bartlett CW. Gene × smoking interactions on human brain gene expression: finding common mechanisms in adolescents and adults. J Child Psychol Psychiatry 2013; 54:1109-19. [PMID: 23909413 PMCID: PMC3809890 DOI: 10.1111/jcpp.12119] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 06/04/2013] [Indexed: 12/25/2022]
Abstract
BACKGROUND Numerous studies have examined gene × environment interactions (G × E) in cognitive and behavioral domains. However, these studies have been limited in that they have not been able to directly assess differential patterns of gene expression in the human brain. Here, we assessed G × E interactions using two publically available datasets to assess if DNA variation is associated with post-mortem brain gene expression changes based on smoking behavior, a biobehavioral construct that is part of a complex system of genetic and environmental influences. METHODS We conducted an expression quantitative trait locus (eQTL) study on two independent human brain gene expression datasets assessing G × E for selected psychiatric genes and smoking status. We employed linear regression to model the significance of the Gene × Smoking interaction term, followed by meta-analysis across datasets. RESULTS Overall, we observed that the effect of DNA variation on gene expression is moderated by smoking status. Expression of 16 genes was significantly associated with single nucleotide polymorphisms that demonstrated G × E effects. The strongest finding (p = 1.9 × 10⁻¹¹) was neurexin 3-alpha (NRXN3), a synaptic cell-cell adhesion molecule involved in maintenance of neural connections (such as the maintenance of smoking behavior). Other significant G × E associations include four glutamate genes. CONCLUSIONS This is one of the first studies to demonstrate G × E effects within the human brain. In particular, this study implicated NRXN3 in the maintenance of smoking. The effect of smoking on NRXN3 expression and downstream behavior is different based upon SNP genotype, indicating that DNA profiles based on SNPs could be useful in understanding the effects of smoking behaviors. These results suggest that better measurement of psychiatric conditions, and the environment in post-mortem brain studies may yield an important avenue for understanding the biological mechanisms of G × E interactions in psychiatry.
Collapse
Affiliation(s)
- Samuel Wolock
- Battelle Center for Mathematical Medicine, Nationwide Children’s Hospital, Columbus, OH, USA
| | - Andrew Yates
- Department of Biomedical Informatics, The Ohio State University College of Medicine, Columbus, OH, USA
| | | | - Jason W. Bohland
- Department of Health Sciences, Boston University, Boston, MA, USA
| | - Clancy Blair
- Department of Applied Psychology, New York University, New York, NY, USA
| | - Ning Li
- Battelle Center for Mathematical Medicine, Nationwide Children’s Hospital, Columbus, OH, USA
| | - Raghu Machiraju
- Department of Biomedical Informatics, The Ohio State University College of Medicine, Columbus, OH, USA
,Department of Computer Science and Engineering, The Ohio State University, Columbus, OH, USA
| | - Kun Huang
- Department of Biomedical Informatics, The Ohio State University College of Medicine, Columbus, OH, USA
,Department of Computer Science and Engineering, The Ohio State University, Columbus, OH, USA
,The CCC Biomedical Informatics Shared Resource, The Ohio State University Columbus, OH, USA
| | - Christopher W. Bartlett
- Battelle Center for Mathematical Medicine, Nationwide Children’s Hospital, Columbus, OH, USA
,Department of Pediatrics, The Ohio State University College of Medicine, Columbus, OH, USA
| |
Collapse
|
46
|
Luykx JJ, Bakker SC, van Boxmeer L, Vinkers CH, Smeenk HE, Visser WF, Verhoeven-Duif NM, Strengman E, Buizer-Voskamp JE, de Groene L, van Dongen EPA, Borgdorff P, Bruins P, de Koning TJ, Kahn RS, Ophoff RA. D-amino acid aberrations in cerebrospinal fluid and plasma of smokers. Neuropsychopharmacology 2013; 38:2019-26. [PMID: 23615666 PMCID: PMC3746686 DOI: 10.1038/npp.2013.103] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/06/2013] [Revised: 03/21/2013] [Accepted: 04/10/2013] [Indexed: 11/09/2022]
Abstract
The glutamatergic neurotransmission system and the N-methyl-D-aspartate receptor (NMDAR) have been implicated in smoking and alcohol consumption behavior. Preclinical studies have demonstrated that nicotine and ethanol influence NMDAR functionality, which may have a role in tendencies to consume these substances. Nonetheless, little is known about concentrations of NMDAR coagonists in the cerebrospinal fluid (CSF) and plasma of individuals who smoke or consume alcohol. Glycine and L- and D-stereoisomers of alanine, serine, and proline were therefore measured using ultra-high-performance liquid chromatography-tandem mass spectrometry in 403 healthy subjects. Nicotine and alcohol consumption were quantified using questionnaires. Possible differences in NMDAR coagonist concentrations in plasma and CSF were investigated using ANCOVA with age, body mass index, and storage duration as covariates. The significance threshold was Bonferroni corrected (α=0.00625). Compared with non-smokers, smokers displayed lower levels of D-proline in plasma (p=0.0027, Cohen's d=-0.41) and D-proline in CSF (p=0.0026, Cohen's d=-0.43). D-Serine in CSF was higher in smokers than in non-smokers (p=0.0052, Cohen's d=0.41). After subdividing participants based on smoking quantity, dose-dependent decreases were demonstrated in smokers for D-proline in plasma (F=5.65, p=0.0039) and D-proline in CSF (F=5.20, p=0.0060). No differences in NMDAR coagonist levels between alcohol consumption groups were detected. To our knowledge, this is the first report to implicate D-amino acids in smoking behavior of humans. Whether such concentration differences lie at the root of or result from smoking habits may be addressed in prospective studies.
Collapse
Affiliation(s)
- Jurjen J Luykx
- Department of Psychiatry, Rudolf Magnus Institute of Neuroscience, University Medical Center Utrecht (UMCU), Utrecht, the Netherlands,Department of Psychiatry, Ziekenhuis Netwerk Antwerpen (ZNA), Stuivenberg campus, Antwerp, Belgium
| | - Steven C Bakker
- Department of Psychiatry, Rudolf Magnus Institute of Neuroscience, University Medical Center Utrecht (UMCU), Utrecht, the Netherlands
| | - Loes van Boxmeer
- Department of Psychiatry, Rudolf Magnus Institute of Neuroscience, University Medical Center Utrecht (UMCU), Utrecht, the Netherlands
| | - Christiaan H Vinkers
- Department of Psychiatry, Rudolf Magnus Institute of Neuroscience, University Medical Center Utrecht (UMCU), Utrecht, the Netherlands,Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Rudolf Magnus Institute of Neuroscience, Utrecht University, Utrecht, the Netherlands
| | - Hanne E Smeenk
- Department of Psychiatry, Rudolf Magnus Institute of Neuroscience, University Medical Center Utrecht (UMCU), Utrecht, the Netherlands
| | - Wouter F Visser
- Department of Medical Genetics, UMCU, Utrecht, the Netherlands
| | | | - Eric Strengman
- Department of Medical Genetics, UMCU, Utrecht, the Netherlands
| | - Jacobine E Buizer-Voskamp
- Department of Psychiatry, Rudolf Magnus Institute of Neuroscience, University Medical Center Utrecht (UMCU), Utrecht, the Netherlands
| | - Lizzy de Groene
- Department of Anesthesiology, Intensive Care and Pain Management, St Antonius Hospital, Nieuwegein, the Netherlands
| | - Eric PA van Dongen
- Department of Anesthesiology, Intensive Care and Pain Management, St Antonius Hospital, Nieuwegein, the Netherlands
| | - Paul Borgdorff
- Department of Anesthesiology, Intensive Care and Pain Management, Diakonessenhuis Hospital, Utrecht, the Netherlands
| | - Peter Bruins
- Department of Anesthesiology, Intensive Care and Pain Management, St Antonius Hospital, Nieuwegein, the Netherlands
| | - Tom J de Koning
- Department of Genetics, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - René S Kahn
- Department of Psychiatry, Rudolf Magnus Institute of Neuroscience, University Medical Center Utrecht (UMCU), Utrecht, the Netherlands
| | - Roel A Ophoff
- Department of Psychiatry, Rudolf Magnus Institute of Neuroscience, University Medical Center Utrecht (UMCU), Utrecht, the Netherlands,Center for Neurobehavioral Genetics, Semel Institute for Neuroscience and Human Behavior, University of California Los Angeles, Los Angeles, CA, USA,Center for Neurobehavioral Genetics, Semel Institute for Neuroscience and Human Behavior, University of California–Los Angeles, Los Angeles, CA 90095, USA, Tel: +1 310 794 9602, Fax: +1 310 794 9613, E-mail:
| |
Collapse
|