1
|
Chitre AS, Hebda-Bauer EK, Blandino P, Bimschleger H, Nguyen KM, Maras P, Li F, Ozel AB, Pan Y, Polesskaya O, Cheng R, Flagel SB, Watson SJ, Li J, Akil H, Palmer AA. Genome-wide association study in a rat model of temperament identifies multiple loci for exploratory locomotion and anxiety-like traits. Front Genet 2023; 13:1003074. [PMID: 36712851 PMCID: PMC9873817 DOI: 10.3389/fgene.2022.1003074] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2022] [Accepted: 10/20/2022] [Indexed: 01/12/2023] Open
Abstract
Common genetic factors likely contribute to multiple psychiatric diseases including mood and substance use disorders. Certain stable, heritable traits reflecting temperament, termed externalizing or internalizing, play a large role in modulating vulnerability to these disorders. To model these heritable tendencies, we selectively bred rats for high and low exploration in a novel environment [bred High Responders (bHR) vs. Low Responders (bLR)]. To identify genes underlying the response to selection, we phenotyped and genotyped 538 rats from an F2 cross between bHR and bLR. Several behavioral traits show high heritability, including the selection trait: exploratory locomotion (EL) in a novel environment. There were significant phenotypic and genetic correlations between tests that capture facets of EL and anxiety. There were also correlations with Pavlovian conditioned approach (PavCA) behavior despite the lower heritability of that trait. Ten significant and conditionally independent loci for six behavioral traits were identified. Five of the six traits reflect different facets of EL that were captured by three behavioral tests. Distance traveled measures from the open field and the elevated plus maze map onto different loci, thus may represent different aspects of novelty-induced locomotor activity. The sixth behavioral trait, number of fecal boli, is the only anxiety-related trait mapping to a significant locus on chromosome 18 within which the Pik3c3 gene is located. There were no significant loci for PavCA. We identified a missense variant in the Plekhf1 gene on the chromosome 1:95 Mb QTL and Fancf and Gas2 as potential candidate genes that may drive the chromosome 1:107 Mb QTL for EL traits. The identification of a locomotor activity-related QTL on chromosome 7 encompassing the Pkhd1l1 and Trhr genes is consistent with our previous finding of these genes being differentially expressed in the hippocampus of bHR vs. bLR rats. The strong heritability coupled with identification of several loci associated with exploratory locomotion and emotionality provide compelling support for this selectively bred rat model in discovering relatively large effect causal variants tied to elements of internalizing and externalizing behaviors inherent to psychiatric and substance use disorders.
Collapse
Affiliation(s)
- Apurva S. Chitre
- Department of Psychiatry, University of California San Diego, La Jolla, CA, United States
| | - Elaine K. Hebda-Bauer
- Michigan Neuroscience Institute, University of Michigan, Ann Arbor, MI, United States
| | - Peter Blandino
- Michigan Neuroscience Institute, University of Michigan, Ann Arbor, MI, United States
| | - Hannah Bimschleger
- Department of Psychiatry, University of California San Diego, La Jolla, CA, United States
| | - Khai-Minh Nguyen
- Department of Psychiatry, University of California San Diego, La Jolla, CA, United States
| | - Pamela Maras
- Michigan Neuroscience Institute, University of Michigan, Ann Arbor, MI, United States
| | - Fei Li
- Michigan Neuroscience Institute, University of Michigan, Ann Arbor, MI, United States
| | - A. Bilge Ozel
- Department of Human Genetics, University of Michigan, Ann Arbor, MI, United States
| | - Yanchao Pan
- Department of Human Genetics, University of Michigan, Ann Arbor, MI, United States
| | - Oksana Polesskaya
- Department of Psychiatry, University of California San Diego, La Jolla, CA, United States
| | - Riyan Cheng
- Department of Psychiatry, University of California San Diego, La Jolla, CA, United States
| | - Shelly B. Flagel
- Michigan Neuroscience Institute, University of Michigan, Ann Arbor, MI, United States
| | - Stanley J. Watson
- Michigan Neuroscience Institute, University of Michigan, Ann Arbor, MI, United States
| | - Jun Li
- Department of Human Genetics, University of Michigan, Ann Arbor, MI, United States
| | - Huda Akil
- Michigan Neuroscience Institute, University of Michigan, Ann Arbor, MI, United States
| | - Abraham A. Palmer
- Department of Psychiatry, University of California San Diego, La Jolla, CA, United States,Institute for Genomic Medicine, University of California San Diego, La Jolla, CA, United States,*Correspondence: Abraham A. Palmer,
| |
Collapse
|
2
|
Grosjean I, Roméo B, Domdom MA, Belaid A, D’Andréa G, Guillot N, Gherardi RK, Gal J, Milano G, Marquette CH, Hung RJ, Landi MT, Han Y, Brest P, Von Bergen M, Klionsky DJ, Amos CI, Hofman P, Mograbi B. Autophagopathies: from autophagy gene polymorphisms to precision medicine for human diseases. Autophagy 2022; 18:2519-2536. [PMID: 35383530 PMCID: PMC9629091 DOI: 10.1080/15548627.2022.2039994] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2022] [Revised: 01/20/2022] [Accepted: 02/06/2022] [Indexed: 12/15/2022] Open
Abstract
At a time when complex diseases affect globally 280 million people and claim 14 million lives every year, there is an urgent need to rapidly increase our knowledge into their underlying etiologies. Though critical in identifying the people at risk, the causal environmental factors (microbiome and/or pollutants) and the affected pathophysiological mechanisms are not well understood. Herein, we consider the variations of autophagy-related (ATG) genes at the heart of mechanisms of increased susceptibility to environmental stress. A comprehensive autophagy genomic resource is presented with 263 single nucleotide polymorphisms (SNPs) for 69 autophagy-related genes associated with 117 autoimmune, inflammatory, infectious, cardiovascular, neurological, respiratory, and endocrine diseases. We thus propose the term 'autophagopathies' to group together a class of complex human diseases the etiology of which lies in a genetic defect of the autophagy machinery, whether directly related or not to an abnormal flux in autophagy, LC3-associated phagocytosis, or any associated trafficking. The future of precision medicine for common diseases will lie in our ability to exploit these ATG SNP x environment relationships to develop new polygenetic risk scores, new management guidelines, and optimal therapies for afflicted patients.Abbreviations: ATG, autophagy-related; ALS-FTD, amyotrophic lateral sclerosis-frontotemporal dementia; ccRCC, clear cell renal cell carcinoma; CD, Crohn disease; COPD, chronic obstructive pulmonary disease; eQTL, expression quantitative trait loci; HCC, hepatocellular carcinoma; HNSCC, head and neck squamous cell carcinoma; GTEx, genotype-tissue expression; GWAS, genome-wide association studies; LAP, LC3-associated phagocytosis; LC3-II, phosphatidylethanolamine conjugated form of LC3; LD, linkage disequilibrium; LUAD, lung adenocarcinoma; MAF, minor allele frequency; MAP1LC3/LC3: microtubule associated protein 1 light chain 3; NSCLC, non-small cell lung cancer; OS, overall survival; PtdIns3K CIII, class III phosphatidylinositol 3 kinase; PtdIns3P, phosphatidylinositol-3-phosphate; SLE, systemic lupus erythematosus; SNPs, single-nucleotide polymorphisms; mQTL, methylation quantitative trait loci; ULK, unc-51 like autophagy activating kinase; UTRs, untranslated regions; WHO, World Health Organization.
Collapse
Affiliation(s)
- Iris Grosjean
- University Côte d’Azur, CNRS, INSERM, IRCAN, FHU-OncoAge, Centre Antoine Lacassagne, France
| | - Barnabé Roméo
- University Côte d’Azur, CNRS, INSERM, IRCAN, FHU-OncoAge, Centre Antoine Lacassagne, France
| | - Marie-Angela Domdom
- University Côte d’Azur, CNRS, INSERM, IRCAN, FHU-OncoAge, Centre Antoine Lacassagne, France
| | - Amine Belaid
- Université Côte d’Azur (UCA), INSERM U1065, C3M, Team 5, F-06204, France
| | - Grégoire D’Andréa
- University Côte d’Azur, CNRS, INSERM, IRCAN, FHU-OncoAge, Centre Antoine Lacassagne, France
- ENT and Head and Neck surgery department, Institut Universitaire de la Face et du Cou, CHU de Nice, University Hospital, Côte d’Azur University, Nice, France
| | - Nicolas Guillot
- University Côte d’Azur, CNRS, INSERM, IRCAN, FHU-OncoAge, Centre Antoine Lacassagne, France
| | - Romain K Gherardi
- INSERM U955 Team Relais, Faculty of Health, Paris Est University, France
| | - Jocelyn Gal
- University Côte d’Azur, Centre Antoine Lacassagne, Epidemiology and Biostatistics Department, Nice, France
| | - Gérard Milano
- Université Côte d’Azur, Centre Antoine Lacassagne, UPR7497, Nice, France
| | - Charles Hugo Marquette
- University Côte d’Azur, CNRS, INSERM, IRCAN, FHU-OncoAge, Centre Antoine Lacassagne, France
- University Côte d’Azur, FHU-OncoAge, Department of Pulmonary Medicine and Oncology, CHU de Nice, Nice, France
| | - Rayjean J. Hung
- Lunenfeld-Tanenbaum Research Institute, Sinai Health System, Toronto, Ontario, Canada; Division of Epidemiology, Dalla Lana School of Public Health, University of Toronto, Toronto, Ontario, Canada
| | - Maria Teresa Landi
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Younghun Han
- Institute for Clinical and Translational Research, Baylor College of Medicine, Houston, TX, USA
| | - Patrick Brest
- University Côte d’Azur, CNRS, INSERM, IRCAN, FHU-OncoAge, Centre Antoine Lacassagne, France
| | - Martin Von Bergen
- Helmholtz Centre for Environmental Research GmbH - UFZ, Dep. of Molecular Systems Biology; University of Leipzig, Faculty of Life Sciences, Institute of Biochemistry, Leipzig, Germany
| | - Daniel J. Klionsky
- University of Michigan, Life Sciences Institute, Ann Arbor, MI, 48109, USA
| | - Christopher I. Amos
- Institute for Clinical and Translational Research, Baylor College of Medicine, Houston, TX, USA
| | - Paul Hofman
- University Côte d’Azur, CNRS, INSERM, IRCAN, FHU-OncoAge, Centre Antoine Lacassagne, France
- University Côte d’Azur, FHU-OncoAge, CHU de Nice, Laboratory of Clinical and Experimental Pathology (LPCE) Biobank(BB-0033-00025), Nice, France
| | - Baharia Mograbi
- University Côte d’Azur, CNRS, INSERM, IRCAN, FHU-OncoAge, Centre Antoine Lacassagne, France
| |
Collapse
|
3
|
Phosphoinositide 3-Kinases as Potential Targets for Thrombosis Prevention. Int J Mol Sci 2022; 23:ijms23094840. [PMID: 35563228 PMCID: PMC9105564 DOI: 10.3390/ijms23094840] [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: 03/29/2022] [Revised: 04/23/2022] [Accepted: 04/25/2022] [Indexed: 02/01/2023] Open
Abstract
As integral parts of pathological arterial thrombi, platelets are the targets of pharmacological regimens designed to treat and prevent thrombosis. A detailed understanding of platelet biology and function is thus key to design treatments that prevent thrombotic cardiovascular disease without significant disruption of the haemostatic balance. Phosphoinositide 3-kinases (PI3Ks) are a group of lipid kinases critical to various aspects of platelet biology. There are eight PI3K isoforms, grouped into three classes. Our understanding of PI3K biology has recently progressed with the targeting of specific isoforms emerging as an attractive therapeutic strategy in various human diseases, including for thrombosis. This review will focus on the role of PI3K subtypes in platelet function and subsequent thrombus formation. Understanding the mechanisms by which platelet function is regulated by the various PI3Ks edges us closer toward targeting specific PI3K isoforms for anti-thrombotic therapy.
Collapse
|
4
|
Hashimoto D, Fujimoto K, Morioka S, Ayabe S, Kataoka T, Fukumura R, Ueda Y, Kajimoto M, Hyuga T, Suzuki K, Hara I, Asamura S, Wakana S, Yoshiki A, Gondo Y, Tamura M, Sasaki T, Yamada G. Establishment of mouse line showing inducible priapism-like phenotypes. Reprod Med Biol 2022; 21:e12472. [PMID: 35765371 PMCID: PMC9207557 DOI: 10.1002/rmb2.12472] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2022] [Revised: 05/19/2022] [Accepted: 05/25/2022] [Indexed: 11/11/2022] Open
Abstract
Purpose Penile research is expected to reveal new targets for treatment and prevention of the complex mechanisms of its disorder including erectile dysfunction (ED). Thus, analyses of the molecular processes of penile ED and continuous erection as priapism are essential issues of reproductive medicine. Methods By performing mouse N-ethyl-N-nitrosourea mutagenesis and exome sequencing, we established a novel mouse line displaying protruded genitalia phenotype (PGP; priapism-like phenotype) and identified a novel Pitpna gene mutation for PGP. Extensive histological analyses on the Pitpna mutant and intracavernous pressure measurement (ICP) and liquid chromatography-electrospray ionization tandem mass spectrometry (LC-ESI/MS)/MS analyses were performed. Results We evaluated the role of phospholipids during erection for the first time and showed the mutants of inducible phenotypes of priapism. Moreover, quantitative analysis using LC-ESI/MS/MS revealed that the level of phosphatidylinositol (PI) was significantly lower in the mutant penile samples. These results imply that PI may contribute to penile erection by PITPα. Conclusions Our findings suggest that the current mutant is a mouse model for priapism and abnormalities in PI signaling pathways through PITPα may lead to priapism providing an attractive novel therapeutic target in its treatment.
Collapse
Affiliation(s)
- Daiki Hashimoto
- Department of Developmental GeneticsInstitute of Advanced Medicine, Wakayama Medical UniversityWakayamaJapan
- Department of Plastic and Reconstructive SurgeryWakayama Medical UniversityWakayamaJapan
| | - Kota Fujimoto
- Department of Developmental GeneticsInstitute of Advanced Medicine, Wakayama Medical UniversityWakayamaJapan
- Department of Plastic and Reconstructive SurgeryWakayama Medical UniversityWakayamaJapan
| | - Shin Morioka
- Department of Biochemical Pathophysiology/Lipid BiologyMedical Research InstituteTokyo Medical and Dental University (TMDU)TokyoJapan
| | - Shinya Ayabe
- Experimental Animal DivisionRIKEN BioResource Research CenterIbarakiJapan
| | - Tomoya Kataoka
- Department of Clinical PharmaceuticsGraduate School of Medical SciencesNagoya City UniversityNagoyaJapan
| | - Ryutaro Fukumura
- Clinical Laboratories Department sSRL & Shizuoka Cancer Center Collaborative Laboratories, IncShizuoka PrefJapan
| | - Yuko Ueda
- Department of Developmental GeneticsInstitute of Advanced Medicine, Wakayama Medical UniversityWakayamaJapan
- Department of UrologyWakayama Medical UniversityWakayamaJapan
| | - Mizuki Kajimoto
- Department of Developmental GeneticsInstitute of Advanced Medicine, Wakayama Medical UniversityWakayamaJapan
- Department of Plastic and Reconstructive SurgeryWakayama Medical UniversityWakayamaJapan
| | - Taiju Hyuga
- Department of Pediatric UrologyChildren's Medical Center TochigiJichi Medical UniversityTochigiJapan
| | - Kentaro Suzuki
- Department of Developmental GeneticsInstitute of Advanced Medicine, Wakayama Medical UniversityWakayamaJapan
- Department of Plastic and Reconstructive SurgeryWakayama Medical UniversityWakayamaJapan
| | - Isao Hara
- Department of UrologyWakayama Medical UniversityWakayamaJapan
| | - Shinichi Asamura
- Department of Plastic and Reconstructive SurgeryWakayama Medical UniversityWakayamaJapan
| | - Shigeharu Wakana
- Department of Animal ExperimentationFoundation for Biomedical Research and Innovation at KobeCreative Lab for Innovation in Kobe 5F 6‐3‐7KobeHyogoJapan
| | - Atsushi Yoshiki
- Experimental Animal DivisionRIKEN BioResource Research CenterIbarakiJapan
| | - Yoichi Gondo
- Department of Molecular Life SciencesDivision of Basic Medical Science and Molecular MedicineTokai University School of MedicineIsehara‐shiKanagawaJapan
| | - Masaru Tamura
- Technology and Development Team for Mouse Phenotype AnalysisRIKEN BioResource Research CenterTsukubaIbarakiJapan
| | - Takehiko Sasaki
- Department of Biochemical Pathophysiology/Lipid BiologyMedical Research InstituteTokyo Medical and Dental University (TMDU)TokyoJapan
| | - Gen Yamada
- Department of Developmental GeneticsInstitute of Advanced Medicine, Wakayama Medical UniversityWakayamaJapan
- Department of Plastic and Reconstructive SurgeryWakayama Medical UniversityWakayamaJapan
| |
Collapse
|
5
|
Class III PI3K Biology. Curr Top Microbiol Immunol 2022; 436:69-93. [DOI: 10.1007/978-3-031-06566-8_3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
|
6
|
Murray AJ, Rogers JC, Katshu MZUH, Liddle PF, Upthegrove R. Oxidative Stress and the Pathophysiology and Symptom Profile of Schizophrenia Spectrum Disorders. Front Psychiatry 2021; 12:703452. [PMID: 34366935 PMCID: PMC8339376 DOI: 10.3389/fpsyt.2021.703452] [Citation(s) in RCA: 59] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Accepted: 06/28/2021] [Indexed: 12/12/2022] Open
Abstract
Schizophrenia is associated with increased levels of oxidative stress, as reflected by an increase in the concentrations of damaging reactive species and a reduction in anti-oxidant defences to combat them. Evidence has suggested that whilst not the likely primary cause of schizophrenia, increased oxidative stress may contribute to declining course and poor outcomes associated with schizophrenia. Here we discuss how oxidative stress may be implicated in the aetiology of schizophrenia and examine how current understanding relates associations with symptoms, potentially via lipid peroxidation induced neuronal damage. We argue that oxidative stress may be a good target for future pharmacotherapy in schizophrenia and suggest a multi-step model of illness progression with oxidative stress involved at each stage.
Collapse
Affiliation(s)
- Alex J. Murray
- Institute for Mental Health, University of Birmingham, Birmingham, United Kingdom
| | - Jack C. Rogers
- Institute for Mental Health, University of Birmingham, Birmingham, United Kingdom
| | - Mohammad Zia Ul Haq Katshu
- Institute of Mental Health, Division of Mental Health and Neurosciences University of Nottingham, Nottingham, United Kingdom
- Nottinghamshire Healthcare National Health Service Foundation Trust, Nottingham, United Kingdom
| | - Peter F. Liddle
- Institute of Mental Health, Division of Mental Health and Neurosciences University of Nottingham, Nottingham, United Kingdom
| | - Rachel Upthegrove
- Institute for Mental Health, University of Birmingham, Birmingham, United Kingdom
- Early Intervention Service, Birmingham Women's and Children's National Health Service Foundation Trust, Centre for Human Brain Health, University of Birmingham, Birmingham, United Kingdom
| |
Collapse
|
7
|
Integrative analysis of genome-wide association studies identifies novel loci associated with neuropsychiatric disorders. Transl Psychiatry 2021; 11:69. [PMID: 33479212 PMCID: PMC7820351 DOI: 10.1038/s41398-020-01195-5] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/21/2020] [Revised: 12/10/2020] [Accepted: 12/11/2020] [Indexed: 01/30/2023] Open
Abstract
Neuropsychiatric disorders, such as autism spectrum disorder (ASD), attention deficit hyperactivity disorder (ADHD), schizophrenia (SCZ), bipolar disorder (BIP), and major depressive disorder (MDD) share common clinical presentations, suggesting etiologic overlap. A substantial proportion of SNP-based heritability for neuropsychiatric disorders is attributable to genetic components, and genome-wide association studies (GWASs) focusing on individual diseases have identified multiple genetic loci shared between these diseases. Here, we aimed at identifying novel genetic loci associated with individual neuropsychiatric diseases and genetic loci shared by neuropsychiatric diseases. We performed multi-trait joint analyses and meta-analysis across five neuropsychiatric disorders based on their summary statistics from the Psychiatric Genomics Consortium (PGC), and further carried out a replication study of ADHD among 2726 cases and 16299 controls in an independent pediatric cohort. In the multi-trait joint analyses, we found five novel genome-wide significant loci for ADHD, one novel locus for BIP, and ten novel loci for MDD. We further achieved modest replication in our independent pediatric dataset. We conducted fine-mapping and functional annotation through an integrative multi-omics approach and identified causal variants and potential target genes at each novel locus. Gene expression profile and gene-set enrichment analysis further suggested early developmental stage expression pattern and postsynaptic membrane compartment enrichment of candidate genes at the genome-wide significant loci of these neuropsychiatric disorders. Therefore, through a multi-omics approach, we identified novel genetic loci associated with the five neuropsychiatric disorders which may help to better understand the underlying molecular mechanism of neuropsychiatric diseases.
Collapse
|
8
|
A reference catalog of DNA palindromes in the human genome and their variations in 1000 Genomes. Hum Genome Var 2020; 7:40. [PMID: 33298903 PMCID: PMC7680136 DOI: 10.1038/s41439-020-00127-5] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2020] [Revised: 05/24/2020] [Accepted: 06/08/2020] [Indexed: 02/07/2023] Open
Abstract
A palindrome in DNA is like a palindrome in language, but when read backwards, it is a complement of the forward sequence; effectively, the two halves of a sequence complement each other from its midpoint like in a double strand of DNA. Palindromes are distributed throughout the human genome and play significant roles in gene expression and regulation. Palindromic mutations are linked to many human diseases, such as neuronal disorders, mental retardation, and various cancers. In this work, we computed and analyzed the palindromic sequences in the human genome and studied their conservation in personal genomes using 1000 Genomes data. We found that ~30% of the palindromes exhibit variation, some of which are caused by rare variants. The analysis of disease/trait-associated single-nucleotide polymorphisms in palindromic regions showed that disease-associated risk variants are 14 times more likely to be present in palindromic regions than in other regions. The catalog of palindromes in the reference genome and 1000 Genomes is being made available here with details on their variations in each individual genome to serve as a resource for future and retrospective whole-genome studies identifying statistically significant palindrome variations associated with diseases or traits and their roles in disease mechanisms.
Collapse
|
9
|
Tamargo-Gómez I, Fernández ÁF, Mariño G. Pathogenic Single Nucleotide Polymorphisms on Autophagy-Related Genes. Int J Mol Sci 2020; 21:ijms21218196. [PMID: 33147747 PMCID: PMC7672651 DOI: 10.3390/ijms21218196] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2020] [Revised: 10/28/2020] [Accepted: 10/30/2020] [Indexed: 02/06/2023] Open
Abstract
In recent years, the study of single nucleotide polymorphisms (SNPs) has gained increasing importance in biomedical research, as they can either be at the molecular origin of a determined disorder or directly affect the efficiency of a given treatment. In this regard, sequence variations in genes involved in pro-survival cellular pathways are commonly associated with pathologies, as the alteration of these routes compromises cellular homeostasis. This is the case of autophagy, an evolutionarily conserved pathway that counteracts extracellular and intracellular stressors by mediating the turnover of cytosolic components through lysosomal degradation. Accordingly, autophagy dysregulation has been extensively described in a wide range of human pathologies, including cancer, neurodegeneration, or inflammatory alterations. Thus, it is not surprising that pathogenic gene variants in genes encoding crucial effectors of the autophagosome/lysosome axis are increasingly being identified. In this review, we present a comprehensive list of clinically relevant SNPs in autophagy-related genes, highlighting the scope and relevance of autophagy alterations in human disease.
Collapse
Affiliation(s)
- Isaac Tamargo-Gómez
- Instituto de Investigación Sanitaria del Principado de Asturias, 33011 Oviedo, Spain;
- Departamento de Biología Funcional, Universidad de Oviedo, 33011 Oviedo, Spain
| | - Álvaro F. Fernández
- Instituto de Investigación Sanitaria del Principado de Asturias, 33011 Oviedo, Spain;
- Departamento de Biología Funcional, Universidad de Oviedo, 33011 Oviedo, Spain
- Correspondence: (Á.F.F.); (G.M.); Tel.: +34-985652416 (G.M.)
| | - Guillermo Mariño
- Instituto de Investigación Sanitaria del Principado de Asturias, 33011 Oviedo, Spain;
- Departamento de Biología Funcional, Universidad de Oviedo, 33011 Oviedo, Spain
- Correspondence: (Á.F.F.); (G.M.); Tel.: +34-985652416 (G.M.)
| |
Collapse
|
10
|
Bellio M, Caux M, Vauclard A, Chicanne G, Gratacap MP, Terrisse AD, Severin S, Payrastre B. Phosphatidylinositol 3 monophosphate metabolizing enzymes in blood platelet production and in thrombosis. Adv Biol Regul 2019; 75:100664. [PMID: 31604685 DOI: 10.1016/j.jbior.2019.100664] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2019] [Revised: 09/19/2019] [Accepted: 09/30/2019] [Indexed: 02/09/2023]
Abstract
Blood platelets, produced by the fragmentation of megakaryocytes, play a key role in hemostasis and thrombosis. Being implicated in atherothrombosis and other thromboembolic disorders, they represent a major therapeutic target for antithrombotic drug development. Several recent studies have highlighted an important role for the lipid phosphatidylinositol 3 monophosphate (PtdIns3P) in megakaryocytes and platelets. PtdIns3P, present in small amounts in mammalian cells, is involved in the control of endocytic trafficking and autophagy. Its metabolism is finely regulated by specific kinases and phosphatases. Class II (α, β and γ) and III (Vps34) phosphoinositide-3-kinases (PI3Ks), INPP4 and Fig4 are involved in the production of PtdIns3P whereas PIKFyve, myotubularins (MTMs) and type II PIPK metabolize PtdIns3P. By regulating the turnover of different pools of PtdIns3P, class II (PI3KC2α) and class III (Vps34) PI3Ks have been recently involved in the regulation of platelet production and functions. These pools of PtdIns3P appear to modulate membrane organization and intracellular trafficking. Moreover, PIKFyve and INPP4 have been recently implicated in arterial thrombosis. In this review, we will discuss the role of PtdIns3P metabolizing enzymes in platelet production and function. Potential new anti-thrombotic therapeutic perspectives based on inhibitors targeting specifically PtdIns3P metabolizing enzymes will also be commented.
Collapse
Affiliation(s)
- Marie Bellio
- Inserm U1048 and Université Paul Sabatier, Institut des Maladies Métaboliques et Cardiovasculaires, Toulouse, France
| | - Manuella Caux
- Inserm U1048 and Université Paul Sabatier, Institut des Maladies Métaboliques et Cardiovasculaires, Toulouse, France
| | - Alicia Vauclard
- Inserm U1048 and Université Paul Sabatier, Institut des Maladies Métaboliques et Cardiovasculaires, Toulouse, France
| | - Gaëtan Chicanne
- Inserm U1048 and Université Paul Sabatier, Institut des Maladies Métaboliques et Cardiovasculaires, Toulouse, France
| | - Marie-Pierre Gratacap
- Inserm U1048 and Université Paul Sabatier, Institut des Maladies Métaboliques et Cardiovasculaires, Toulouse, France
| | - Anne-Dominique Terrisse
- Inserm U1048 and Université Paul Sabatier, Institut des Maladies Métaboliques et Cardiovasculaires, Toulouse, France
| | - Sonia Severin
- Inserm U1048 and Université Paul Sabatier, Institut des Maladies Métaboliques et Cardiovasculaires, Toulouse, France
| | - Bernard Payrastre
- Inserm U1048 and Université Paul Sabatier, Institut des Maladies Métaboliques et Cardiovasculaires, Toulouse, France; Laboratoire d'Hématologie, Hopital Universitaire de Toulouse, Toulouse, France.
| |
Collapse
|
11
|
Crespi BJ. Comparative psychopharmacology of autism and psychotic-affective disorders suggests new targets for treatment. Evol Med Public Health 2019; 2019:149-168. [PMID: 31548888 PMCID: PMC6748779 DOI: 10.1093/emph/eoz022] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2018] [Accepted: 08/07/2019] [Indexed: 12/13/2022] Open
Abstract
The first treatments showing effectiveness for some psychiatric disorders, such as lithium for bipolar disorder and chlorpromazine for schizophrenia, were discovered by accident. Currently, psychiatric drug design is seen as a scientific enterprise, limited though it remains by the complexity of brain development and function. Relatively few novel and effective drugs have, however, been developed for many years. The purpose of this article is to demonstrate how evolutionary biology can provide a useful framework for psychiatric drug development. The framework is based on a diametrical nature of autism, compared with psychotic-affective disorders (mainly schizophrenia, bipolar disorder and depression). This paradigm follows from two inferences: (i) risks and phenotypes of human psychiatric disorders derive from phenotypes that have evolved along the human lineage and (ii) biological variation is bidirectional (e.g. higher vs lower, faster vs slower, etc.), such that dysregulation of psychological traits varies in two opposite ways. In this context, the author review the evidence salient to the hypothesis that autism and psychotic-affective disorders represent diametrical disorders in terms of current, proposed and potential psychopharmacological treatments. Studies of brain-derived neurotrophic factor, the PI3K pathway, the NMDA receptor, kynurenic acid metabolism, agmatine metabolism, levels of the endocannabinoid anandamide, antidepressants, anticonvulsants, antipsychotics, and other treatments, demonstrate evidence of diametric effects in autism spectrum disorders and phenotypes compared with psychotic-affective disorders and phenotypes. These findings yield insights into treatment mechanisms and the development of new pharmacological therapies, as well as providing an explanation for the longstanding puzzle of antagonism between epilepsy and psychosis. Lay Summary: Consideration of autism and schizophrenia as caused by opposite alterations to brain development and function leads to novel suggestions for pharmacological treatments.
Collapse
Affiliation(s)
- Bernard J Crespi
- Department of Biological Sciences, Simon Fraser University, 8888 University Drive, Burnaby, BC V5A 1S6, Canada
| |
Collapse
|
12
|
Zhang Z, Jia Y, Almeida P, Mank JE, van Tuinen M, Wang Q, Jiang Z, Chen Y, Zhan K, Hou S, Zhou Z, Li H, Yang F, He Y, Ning Z, Yang N, Qu L. Whole-genome resequencing reveals signatures of selection and timing of duck domestication. Gigascience 2018; 7:4965113. [PMID: 29635409 PMCID: PMC6007426 DOI: 10.1093/gigascience/giy027] [Citation(s) in RCA: 68] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2017] [Accepted: 03/18/2018] [Indexed: 12/28/2022] Open
Abstract
Background The genetic basis of animal domestication remains poorly understood, and systems with
substantial phenotypic differences between wild and domestic populations are useful for
elucidating the genetic basis of adaptation to new environments as well as the genetic
basis of rapid phenotypic change. Here, we sequenced the whole genome of 78 individual
ducks, from two wild and seven domesticated populations, with an average sequencing
depth of 6.42X per individual. Results Our population and demographic analyses indicate a complex history of domestication,
with early selection for separate meat and egg lineages. Genomic comparison of wild to
domesticated populations suggests that genes that affect brain and neuronal development
have undergone strong positive selection during domestication. Our FST
analysis also indicates that the duck white plumage is the result of selection at the
melanogenesis-associated transcription factor locus. Conclusions Our results advance the understanding of animal domestication and selection for complex
phenotypic traits.
Collapse
Affiliation(s)
- Zebin Zhang
- State Key Laboratory of Animal Nutrition, Department of Animal Genetics and Breeding, National Engineering Laboratory for Animal Breeding, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Yaxiong Jia
- Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing, China
| | - Pedro Almeida
- Department of Genetics, Evolution and Environment, University College London, London, UK
| | - Judith E Mank
- Department of Genetics, Evolution and Environment, University College London, London, UK.,Department of Organismal Biology, Evolutionary Biology Centre, Uppsala University, Uppsala, Sweden
| | - Marcel van Tuinen
- Centre of Evolutionary and Ecological Studies, Marine Evolution and Conservation Group, University of Groningen, Groningen, The Netherlands
| | - Qiong Wang
- State Key Laboratory of Animal Nutrition, Department of Animal Genetics and Breeding, National Engineering Laboratory for Animal Breeding, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Zhihua Jiang
- Department of Animal Sciences, Center for Reproductive Biology, Veterinary and Biomedical Research Building, Washington State University, Pullman, United States
| | - Yu Chen
- Beijing Municipal General Station of Animal Science, Beijing, China
| | - Kai Zhan
- Institute of Animal Husbandry and Veterinary Medicine, Anhui Academy of Agricultural Sciences, Hefei, China
| | - Shuisheng Hou
- Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing, China
| | - Zhengkui Zhou
- Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing, China
| | - Huifang Li
- Poultry Institute, Chinese Academy of Agriculture Science, Yangzhou, China
| | | | - Yong He
- Cherry Valley farms (xianghe) Co., Ltd, Langfang, China
| | - Zhonghua Ning
- State Key Laboratory of Animal Nutrition, Department of Animal Genetics and Breeding, National Engineering Laboratory for Animal Breeding, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Ning Yang
- State Key Laboratory of Animal Nutrition, Department of Animal Genetics and Breeding, National Engineering Laboratory for Animal Breeding, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Lujiang Qu
- State Key Laboratory of Animal Nutrition, Department of Animal Genetics and Breeding, National Engineering Laboratory for Animal Breeding, College of Animal Science and Technology, China Agricultural University, Beijing, China
| |
Collapse
|
13
|
Gstrein T, Edwards A, Přistoupilová A, Leca I, Breuss M, Pilat-Carotta S, Hansen AH, Tripathy R, Traunbauer AK, Hochstoeger T, Rosoklija G, Repic M, Landler L, Stránecký V, Dürnberger G, Keane TM, Zuber J, Adams DJ, Flint J, Honzik T, Gut M, Beltran S, Mechtler K, Sherr E, Kmoch S, Gut I, Keays DA. Mutations in Vps15 perturb neuronal migration in mice and are associated with neurodevelopmental disease in humans. Nat Neurosci 2018; 21:207-217. [PMID: 29311744 PMCID: PMC5897053 DOI: 10.1038/s41593-017-0053-5] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2016] [Accepted: 11/22/2017] [Indexed: 01/31/2023]
Abstract
The formation of the vertebrate brain requires the generation, migration, differentiation and survival of neurons. Genetic mutations that perturb these critical cellular events can result in malformations of the telencephalon, providing a molecular window into brain development. Here we report the identification of an N-ethyl-N-nitrosourea-induced mouse mutant characterized by a fractured hippocampal pyramidal cell layer, attributable to defects in neuronal migration. We show that this is caused by a hypomorphic mutation in Vps15 that perturbs endosomal-lysosomal trafficking and autophagy, resulting in an upregulation of Nischarin, which inhibits Pak1 signaling. The complete ablation of Vps15 results in the accumulation of autophagic substrates, the induction of apoptosis and severe cortical atrophy. Finally, we report that mutations in VPS15 are associated with cortical atrophy and epilepsy in humans. These data highlight the importance of the Vps15-Vps34 complex and the Nischarin-Pak1 signaling hub in the development of the telencephalon.
Collapse
Affiliation(s)
- Thomas Gstrein
- Institute of Molecular Pathology (IMP), Vienna Biocentre (VBC), Vienna, Austria
| | - Andrew Edwards
- Wellcome Trust Center for Human Genetics (WTCHG), Oxford, UK
| | - Anna Přistoupilová
- Institute of Inherited Metabolic Disorders, Charles University, Prague, Czech Republic
- CNAG-CRG, Centre for Genomic Regulation (CRG), Barcelona Institute of Science and Technology (BIST), Barcelona, Spain
- Universitat Pompeu Fabra (UPF), Barcelona, Spain
| | - Ines Leca
- Institute of Molecular Pathology (IMP), Vienna Biocentre (VBC), Vienna, Austria
| | - Martin Breuss
- Institute of Molecular Pathology (IMP), Vienna Biocentre (VBC), Vienna, Austria
| | | | - Andi H Hansen
- Institute of Molecular Pathology (IMP), Vienna Biocentre (VBC), Vienna, Austria
| | - Ratna Tripathy
- Institute of Molecular Pathology (IMP), Vienna Biocentre (VBC), Vienna, Austria
| | - Anna K Traunbauer
- Institute of Molecular Pathology (IMP), Vienna Biocentre (VBC), Vienna, Austria
| | - Tobias Hochstoeger
- Institute of Molecular Pathology (IMP), Vienna Biocentre (VBC), Vienna, Austria
| | - Gavril Rosoklija
- Institute of Molecular Pathology (IMP), Vienna Biocentre (VBC), Vienna, Austria
| | - Marco Repic
- Institute for Molecular Biotechnology (IMBA), Vienna, Austria
| | - Lukas Landler
- Institute of Molecular Pathology (IMP), Vienna Biocentre (VBC), Vienna, Austria
| | - Viktor Stránecký
- Institute of Inherited Metabolic Disorders, Charles University, Prague, Czech Republic
| | - Gerhard Dürnberger
- Institute of Molecular Pathology (IMP), Vienna Biocentre (VBC), Vienna, Austria
| | - Thomas M Keane
- Wellcome Trust Sanger Institute, Hinxton, Cambridgeshire, UK
| | - Johannes Zuber
- Institute of Molecular Pathology (IMP), Vienna Biocentre (VBC), Vienna, Austria
| | - David J Adams
- Wellcome Trust Sanger Institute, Hinxton, Cambridgeshire, UK
| | - Jonathan Flint
- Wellcome Trust Center for Human Genetics (WTCHG), Oxford, UK
| | - Tomas Honzik
- Department of Pediatrics and Adolescent Medicine, First Faculty of Medicine, Charles University, Prague, Czech Republic
| | - Marta Gut
- CNAG-CRG, Centre for Genomic Regulation (CRG), Barcelona Institute of Science and Technology (BIST), Barcelona, Spain
- Universitat Pompeu Fabra (UPF), Barcelona, Spain
| | - Sergi Beltran
- CNAG-CRG, Centre for Genomic Regulation (CRG), Barcelona Institute of Science and Technology (BIST), Barcelona, Spain
- Universitat Pompeu Fabra (UPF), Barcelona, Spain
| | - Karl Mechtler
- Institute of Molecular Pathology (IMP), Vienna Biocentre (VBC), Vienna, Austria
| | - Elliott Sherr
- Institute of Human Genetics, University of California, San Francisco, San Francisco, CA, USA
| | - Stanislav Kmoch
- Institute of Inherited Metabolic Disorders, Charles University, Prague, Czech Republic
| | - Ivo Gut
- CNAG-CRG, Centre for Genomic Regulation (CRG), Barcelona Institute of Science and Technology (BIST), Barcelona, Spain
- Universitat Pompeu Fabra (UPF), Barcelona, Spain
| | - David A Keays
- Institute of Molecular Pathology (IMP), Vienna Biocentre (VBC), Vienna, Austria.
| |
Collapse
|
14
|
Epigenetic and genetic variants in the HTR1B gene and clinical improvement in children and adolescents treated with fluoxetine. Prog Neuropsychopharmacol Biol Psychiatry 2017; 75:28-34. [PMID: 28025020 DOI: 10.1016/j.pnpbp.2016.12.003] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/09/2016] [Revised: 12/09/2016] [Accepted: 12/19/2016] [Indexed: 12/15/2022]
Abstract
The serotonin 1B receptor (5-HT1B) is important to both the pathogenesis of major depressive disorder and the antidepressant effects of selective serotonin reuptake inhibitors. Although fluoxetine has been shown to be effective and safe in children and adolescents, not all patients experience a proper clinical response, which has led to further study into the main factors involved in this inter-individual variability. Our aim was to study the effect of epigenetic and genetic factors that could affect 5-hydroxytryptamine receptor 1B (HTR1B) gene expression, and thereby response to fluoxetine. A total of 83 children and adolescents were clinically assessed 12weeks after of initiating an antidepressant treatment with fluoxetine for the first time. We evaluated the influence of single nucleotide polymorphisms (SNPs) specifically located in transcription factor binding sites (TFBSs) on their clinical improvement. A combined genetic analysis considering the significant SNPs together with the functional variant rs130058 previously associated in our population was also performed. Moreover, we assessed, for the first time in the literature, whether methylation levels of the HTR1B promoter region could be associated with the pharmacological response. Two, rs9361233 and rs9361235, were significantly associated with clinical improvement after treatment with fluoxetine. The heterozygous genotype combination analysis showed a negative correlation with clinical improvement. The lowest improvement was experienced by patients who were heterozygous for all three SNPs. Moreover, a negative correlation was found between clinical improvement and the average methylation level of the HTR1B promoter. These results give new evidence for the role of epigenetic and genetic factors which could modulate HTR1B expression in the pharmacological response to antidepressants.
Collapse
|
15
|
Matsumoto J, Nakanishi H, Kunii Y, Sugiura Y, Yuki D, Wada A, Hino M, Niwa SI, Kondo T, Waki M, Hayasaka T, Masaki N, Akatsu H, Hashizume Y, Yamamoto S, Sato S, Sasaki T, Setou M, Yabe H. Decreased 16:0/20:4-phosphatidylinositol level in the post-mortem prefrontal cortex of elderly patients with schizophrenia. Sci Rep 2017; 7:45050. [PMID: 28332626 PMCID: PMC5362900 DOI: 10.1038/srep45050] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2016] [Accepted: 02/17/2017] [Indexed: 01/17/2023] Open
Abstract
The etiology of schizophrenia includes phospholipid abnormalities. Phospholipids are bioactive substances essential for brain function. To analyze differences in the quantity and types of phospholipids present in the brain tissue of patients with schizophrenia, we performed a global analysis of phospholipids in multiple brain samples using liquid chromatography electrospray ionization mass/mass spectrometry (LC-ESI/MS/MS) and imaging mass spectrometry (IMS). We found significantly decreased 16:0/20:4-phosphatidylinositol (PI) levels in the prefrontal cortex (PFC) in the brains from patients with schizophrenia in the LC-ESI/MS/MS, and that the 16:0/20:4-PI in grey matter was most prominently diminished according to the IMS experiments. Previous reports investigating PI pathology of schizophrenia did not identify differences in the sn-1 and sn-2 fatty acyl chains. This study is the first to clear the fatty acid composition of PI in brains from patients with schizophrenia. Alteration in the characteristic fatty acid composition of PI may also affect neuronal function, and could play a role in the etiology of schizophrenia. Although further studies are necessary to understand the role of reduced 16:0/20:4-PI levels within the prefrontal cortex in the etiology of schizophrenia, our results provide insight into the development of a novel therapy for the clinical treatment of schizophrenia.
Collapse
Affiliation(s)
- Junya Matsumoto
- Department of Neuropsychiatry, School of Medicine, Fukushima Medical University, 1 Hikarigaoka, Fukushima, Fukushima 960-1295, Japan
| | - Hiroki Nakanishi
- Research Center for Biosignal, Akita University, 1-1-1 Hondo, Akita, Akita 010-8543, Japan
- Akita Lipid Technologies, LLC.,1-2, Nukazuka, Yanagida, Akita, 010-0825, Japan
| | - Yasuto Kunii
- Department of Neuropsychiatry, School of Medicine, Fukushima Medical University, 1 Hikarigaoka, Fukushima, Fukushima 960-1295, Japan
- Department of Psychiatry, Aizu Medical Center, Fukushima Medical University, 21-2 Maeda, Yazawa Kawahigashimachi, Aizuwakamatsu, Fukushima 969-3492, Japan
| | - Yuki Sugiura
- Department of Cellular and Molecular Anatomy, Hamamatsu University School of Medicine, 1-20-1, Handayama, Higashi-ku, Hamamatsu, Shizuoka 431-3192, Japan
| | - Dai Yuki
- Department of Cellular and Molecular Anatomy, Hamamatsu University School of Medicine, 1-20-1, Handayama, Higashi-ku, Hamamatsu, Shizuoka 431-3192, Japan
| | - Akira Wada
- Department of Neuropsychiatry, School of Medicine, Fukushima Medical University, 1 Hikarigaoka, Fukushima, Fukushima 960-1295, Japan
- Department of Neuropsychiatry, The University of Tokyo Hospital, 7-3-1, Hongo, Bunkyo-ku, Tokyo 113-8655, Japan
| | - Mizuki Hino
- Department of Neuropsychiatry, School of Medicine, Fukushima Medical University, 1 Hikarigaoka, Fukushima, Fukushima 960-1295, Japan
| | - Shin-Ichi Niwa
- Department of Psychiatry, Aizu Medical Center, Fukushima Medical University, 21-2 Maeda, Yazawa Kawahigashimachi, Aizuwakamatsu, Fukushima 969-3492, Japan
| | - Takeshi Kondo
- Department of Cellular and Molecular Anatomy, Hamamatsu University School of Medicine, 1-20-1, Handayama, Higashi-ku, Hamamatsu, Shizuoka 431-3192, Japan
| | - Michihiko Waki
- Department of Cellular and Molecular Anatomy, Hamamatsu University School of Medicine, 1-20-1, Handayama, Higashi-ku, Hamamatsu, Shizuoka 431-3192, Japan
| | - Takahiro Hayasaka
- Department of Cellular and Molecular Anatomy, Hamamatsu University School of Medicine, 1-20-1, Handayama, Higashi-ku, Hamamatsu, Shizuoka 431-3192, Japan
| | - Noritaka Masaki
- Department of Cellular and Molecular Anatomy, Hamamatsu University School of Medicine, 1-20-1, Handayama, Higashi-ku, Hamamatsu, Shizuoka 431-3192, Japan
| | - Hiroyasu Akatsu
- Choju Medical Institute, Fukushimura Hospital, 19-14 Yamanaka, Noyori-cho, Toyohashi, Aichi 441-8124, Japan
- Department of Community-based Medical Education/Department of Community-based Medicine, Nagoya City University Graduate School of Medical Science, 1, Kawasumi, Mizuho-cho, Mizuho-ku, Nagoya, Aichi 467-8601, Japan
| | - Yoshio Hashizume
- Choju Medical Institute, Fukushimura Hospital, 19-14 Yamanaka, Noyori-cho, Toyohashi, Aichi 441-8124, Japan
| | - Sakon Yamamoto
- Choju Medical Institute, Fukushimura Hospital, 19-14 Yamanaka, Noyori-cho, Toyohashi, Aichi 441-8124, Japan
| | - Shinji Sato
- Choju Medical Institute, Fukushimura Hospital, 19-14 Yamanaka, Noyori-cho, Toyohashi, Aichi 441-8124, Japan
- Quests Research Institute, Otsuka Pharmaceutical Co. Ltd., 463-10 Kagasuno, Kawauchi-cho, Tokushima, Tokushima 771-0192, Japan
| | - Takehiko Sasaki
- Research Center for Biosignal, Akita University, 1-1-1 Hondo, Akita, Akita 010-8543, Japan
- Akita Lipid Technologies, LLC.,1-2, Nukazuka, Yanagida, Akita, 010-0825, Japan
- Department of Medical Biology Graduate School of Medicine, Akita University, 1-1-1 Hondo, Akita, Tokushima 010-8543, Japan
| | - Mitsutoshi Setou
- Department of Cellular and Molecular Anatomy, Hamamatsu University School of Medicine, 1-20-1, Handayama, Higashi-ku, Hamamatsu, Shizuoka 431-3192, Japan
- International Mass Imaging Center, Hamamatsu University School of Medicine, 1-20-1 Handayama, Higashi-ku, Hamamatsu, Shizuoka 431-3192, Japan
- Preeminent Medical Photonics Education & Research Center, Hamamatsu University School of Medicine, 1-20-1 Handayama, Higashi-ku, Hamamatsu, Shizuoka 431-3192, Japan
- Department of Anatomy, The university of Hong Kong, 6/F, William MW Mong Block 21 Sassoon Road, Pokfulam, Hong Kong SAR, China
- Division of Neural Systematics, National Institute for Physiological Sciences, 38 Nishigonaka Myodaiji, Okazaki, Aichi, 444-8585, Japan
- Riken Center for Molecular Imaging Science, 6-7-3 Minatojima-minamimachi, Chuo-ku, Kobe, Hyogo 650-0047, Japan
| | - Hirooki Yabe
- Department of Neuropsychiatry, School of Medicine, Fukushima Medical University, 1 Hikarigaoka, Fukushima, Fukushima 960-1295, Japan
| |
Collapse
|
16
|
Khanzada NS, Butler MG, Manzardo AM. GeneAnalytics Pathway Analysis and Genetic Overlap among Autism Spectrum Disorder, Bipolar Disorder and Schizophrenia. Int J Mol Sci 2017; 18:ijms18030527. [PMID: 28264500 PMCID: PMC5372543 DOI: 10.3390/ijms18030527] [Citation(s) in RCA: 58] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2017] [Revised: 02/15/2017] [Accepted: 02/23/2017] [Indexed: 12/18/2022] Open
Abstract
Bipolar disorder (BPD) and schizophrenia (SCH) show similar neuropsychiatric behavioral disturbances, including impaired social interaction and communication, seen in autism spectrum disorder (ASD) with multiple overlapping genetic and environmental influences implicated in risk and course of illness. GeneAnalytics software was used for pathway analysis and genetic profiling to characterize common susceptibility genes obtained from published lists for ASD (792 genes), BPD (290 genes) and SCH (560 genes). Rank scores were derived from the number and nature of overlapping genes, gene-disease association, tissue specificity and gene functions subdivided into categories (e.g., diseases, tissues or functional pathways). Twenty-three genes were common to all three disorders and mapped to nine biological Superpathways including Circadian entrainment (10 genes, score = 37.0), Amphetamine addiction (five genes, score = 24.2), and Sudden infant death syndrome (six genes, score = 24.1). Brain tissues included the medulla oblongata (11 genes, score = 2.1), thalamus (10 genes, score = 2.0) and hypothalamus (nine genes, score = 2.0) with six common genes (BDNF, DRD2, CHRNA7, HTR2A, SLC6A3, and TPH2). Overlapping genes impacted dopamine and serotonin homeostasis and signal transduction pathways, impacting mood, behavior and physical activity level. Converging effects on pathways governing circadian rhythms support a core etiological relationship between neuropsychiatric illnesses and sleep disruption with hypoxia and central brain stem dysfunction.
Collapse
Affiliation(s)
- Naveen S Khanzada
- Department of Psychiatry and Behavioral Sciences, University of Kansas Medical Center, Kansas City, KS 66160, USA.
| | - Merlin G Butler
- Department of Psychiatry and Behavioral Sciences, University of Kansas Medical Center, Kansas City, KS 66160, USA.
- Department of Pediatrics, University of Kansas Medical Center, Kansas City, KS 66160, USA.
| | - Ann M Manzardo
- Department of Psychiatry and Behavioral Sciences, University of Kansas Medical Center, Kansas City, KS 66160, USA.
| |
Collapse
|
17
|
Papadopoulos T, Rhee HJ, Subramanian D, Paraskevopoulou F, Mueller R, Schultz C, Brose N, Rhee JS, Betz H. Endosomal Phosphatidylinositol 3-Phosphate Promotes Gephyrin Clustering and GABAergic Neurotransmission at Inhibitory Postsynapses. J Biol Chem 2016; 292:1160-1177. [PMID: 27941024 DOI: 10.1074/jbc.m116.771592] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2016] [Indexed: 11/06/2022] Open
Abstract
The formation of neuronal synapses and the dynamic regulation of their efficacy depend on the proper assembly of the postsynaptic neurotransmitter receptor apparatus. Receptor recruitment to inhibitory GABAergic postsynapses requires the scaffold protein gephyrin and the guanine nucleotide exchange factor collybistin (Cb). In vitro, the pleckstrin homology domain of Cb binds phosphoinositides, specifically phosphatidylinositol 3-phosphate (PI3P). However, whether PI3P is required for inhibitory postsynapse formation is currently unknown. Here, we investigated the role of PI3P at developing GABAergic postsynapses by using a membrane-permeant PI3P derivative, time-lapse confocal imaging, electrophysiology, as well as knockdown and overexpression of PI3P-metabolizing enzymes. Our results provide the first in cellula evidence that PI3P located at early/sorting endosomes regulates the postsynaptic clustering of gephyrin and GABAA receptors and the strength of inhibitory, but not excitatory, postsynapses in cultured hippocampal neurons. In human embryonic kidney 293 cells, stimulation of gephyrin cluster formation by PI3P depends on Cb. We therefore conclude that the endosomal pool of PI3P, generated by the class III phosphatidylinositol 3-kinase, is important for the Cb-mediated recruitment of gephyrin and GABAA receptors to developing inhibitory postsynapses and thus the formation of postsynaptic membrane specializations.
Collapse
Affiliation(s)
- Theofilos Papadopoulos
- From the Department of Molecular Biology, Center of Biochemistry and Molecular Cell Biology, Universitätsmedizin Göttingen, Humboldtallee 23, 37073 Göttingen, Germany,
| | - Hong Jun Rhee
- the Department of Molecular Neurobiology, Max Planck Institute of Experimental Medicine, Hermann-Rein-Strasse 3, 37075 Göttingen, Germany
| | - Devaraj Subramanian
- the European Molecular Biology Laboratory (EMBL), Cell Biology and Biophysics Unit, Meyerhofstrasse 1, 69117 Heidelberg, Germany
| | - Foteini Paraskevopoulou
- From the Department of Molecular Biology, Center of Biochemistry and Molecular Cell Biology, Universitätsmedizin Göttingen, Humboldtallee 23, 37073 Göttingen, Germany
| | - Rainer Mueller
- the European Molecular Biology Laboratory (EMBL), Cell Biology and Biophysics Unit, Meyerhofstrasse 1, 69117 Heidelberg, Germany
| | - Carsten Schultz
- the European Molecular Biology Laboratory (EMBL), Cell Biology and Biophysics Unit, Meyerhofstrasse 1, 69117 Heidelberg, Germany.,the Department of Physiology and Pharmacology, Oregon Health and Science University, Portland, Oregon 97239-3098
| | - Nils Brose
- the Department of Molecular Neurobiology, Max Planck Institute of Experimental Medicine, Hermann-Rein-Strasse 3, 37075 Göttingen, Germany
| | - Jeong-Seop Rhee
- the Department of Molecular Neurobiology, Max Planck Institute of Experimental Medicine, Hermann-Rein-Strasse 3, 37075 Göttingen, Germany
| | - Heinrich Betz
- the Department of Neurochemistry, Max Planck Institute for Brain Research, Deutschordenstrasse 46, 60528 Frankfurt am Main, Germany, and.,the Max Planck Institute of Medical Research, Jahnstrasse 29, 69120 Heidelberg, Germany
| |
Collapse
|
18
|
Jensen KP. A Review of Genome-Wide Association Studies of Stimulant and Opioid Use Disorders. MOLECULAR NEUROPSYCHIATRY 2016; 2:37-45. [PMID: 27606319 DOI: 10.1159/000444755] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/07/2015] [Accepted: 02/16/2016] [Indexed: 12/27/2022]
Abstract
Substance use disorders (SUD) are a major contributor to disability and disease burden worldwide. Risk for developing SUDs is influenced by variation in the genome. Identifying the genetic variants that influence SUD risk may help us to understand the biological mechanisms for the disorders and improve treatments. Genome-wide association studies (GWAS) have been successful in identifying many regions of the genome associated with common human disorders. Here, findings from recent GWAS of SUDs that involve illicit substances will be reviewed. Several GWAS have been reported, including studies on opioid and stimulant use disorder (cocaine and methamphetamine). Several of these GWAS report associations that are biologically interesting and statistically robust. Replication of the associations in independent samples and functional studies to understand the basis for the statistical associations will be important next steps.
Collapse
Affiliation(s)
- Kevin P Jensen
- Department of Psychiatry, Yale University School of Medicine, New Haven, Conn., and VA Connecticut Healthcare System, West Haven, Conn., USA
| |
Collapse
|
19
|
Enriquez-Barreto L, Morales M. The PI3K signaling pathway as a pharmacological target in Autism related disorders and Schizophrenia. MOLECULAR AND CELLULAR THERAPIES 2016; 4:2. [PMID: 26877878 PMCID: PMC4751644 DOI: 10.1186/s40591-016-0047-9] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/13/2015] [Accepted: 01/25/2016] [Indexed: 01/01/2023]
Abstract
This review is focused in PI3K’s involvement in two widespread mental disorders: Autism and Schizophrenia. A large body of evidence points to synaptic dysfunction as a cause of these diseases, either during the initial phases of brain synaptic circuit’s development or later modulating synaptic function and plasticity. Autism related disorders and Schizophrenia are complex genetic conditions in which the identification of gene markers has proved difficult, although the existence of single-gene mutations with a high prevalence in both diseases offers insight into the role of the PI3K signaling pathway. In the brain, components of the PI3K pathway regulate synaptic formation and plasticity; thus, disruption of this pathway leads to synapse dysfunction and pathological behaviors. Here, we recapitulate recent evidences that demonstrate the imbalance of several PI3K elements as leading causes of Autism and Schizophrenia, together with the plausible new pharmacological paths targeting this signaling pathway.
Collapse
Affiliation(s)
- Lilian Enriquez-Barreto
- Institut de Neurociències, Departament de Bioquímica i Biologia Molecular, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Miguel Morales
- Institut de Neurociències, Departament de Bioquímica i Biologia Molecular, Universitat Autònoma de Barcelona, Barcelona, Spain
| |
Collapse
|
20
|
Kordi-Tamandani DM, Mir A. Relationship between phosphoinositide-3-kinase genetic polymorphism and schizophrenia. Nord J Psychiatry 2016; 70:272-5. [PMID: 26643470 DOI: 10.3109/08039488.2015.1092171] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
UNLABELLED Schizophrenia, with incidence of 1% worldwide, is a common mental disorder. Phosphoinositide-3-kinases (PI3Ks) are a family of enzymes involved in cellular functions such as cell growth, proliferation, differentiation, motility, intracellular trafficking, and survival. These enzymes play an important role in the PI3K/AKT signalling pathway. The PIK3CA gene encodes the alpha catalytic subunit of the PI3K enzyme. The present study analysed the role of three SNPs of the PIK3CA gene (rs6443624 (A/C), rs7640662(C/G) and rs7621329(C/T)) in the development of schizophrenia. METHODS In this case-controlled study, DNA was extracted from blood samples from 108 patients with schizophrenia and 108 healthy patients as controls. Genotypic analyses of PIK3CA SNPs rs6443624 (A/C), rs7640662(C/G) and rs7621329(C/T) were made using the tetra primer ARMS-PCR technique. RESULTS The outcome shows significant difference between CT and the combined genotype (CT + TT) of rs7621329 and the risk of schizophrenia (OR = 6.4, 95% CI = 3.023-14.23, p < 0.0001). Outcome showed no significant difference for were for analyses of the rs6443624 and rs7640662 genotypes. CONCLUSIONS These results indicate an association between PIK3CA gene polymorphism on the rs7621329(C/T) site and the risk of schizophrenia. Further study of the genetic population using a larger sample size is necessary in order to validate these present findings.
Collapse
Affiliation(s)
| | - Atefeh Mir
- a Department of Biology , University of Sistan and Baluchestan , Zahedan , Iran
| |
Collapse
|
21
|
Stahelin RV, Scott JL, Frick CT. Cellular and molecular interactions of phosphoinositides and peripheral proteins. Chem Phys Lipids 2014; 182:3-18. [PMID: 24556335 DOI: 10.1016/j.chemphyslip.2014.02.002] [Citation(s) in RCA: 87] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2014] [Revised: 02/05/2014] [Accepted: 02/05/2014] [Indexed: 12/23/2022]
Abstract
Anionic lipids act as signals for the recruitment of proteins containing cationic clusters to biological membranes. A family of anionic lipids known as the phosphoinositides (PIPs) are low in abundance, yet play a critical role in recruitment of peripheral proteins to the membrane interface. PIPs are mono-, bis-, or trisphosphorylated derivatives of phosphatidylinositol (PI) yielding seven species with different structure and anionic charge. The differential spatial distribution and temporal appearance of PIPs is key to their role in communicating information to target proteins. Selective recognition of PIPs came into play with the discovery that the substrate of protein kinase C termed pleckstrin possessed the first PIP binding region termed the pleckstrin homology (PH) domain. Since the discovery of the PH domain, more than ten PIP binding domains have been identified including PH, ENTH, FYVE, PX, and C2 domains. Representative examples of each of these domains have been thoroughly characterized to understand how they coordinate PIP headgroups in membranes, translocate to specific membrane docking sites in the cell, and function to regulate the activity of their full-length proteins. In addition, a number of novel mechanisms of PIP-mediated membrane association have emerged, such as coincidence detection-specificity for two distinct lipid headgroups. Other PIP-binding domains may also harbor selectivity for a membrane physical property such as charge or membrane curvature. This review summarizes the current understanding of the cellular distribution of PIPs and their molecular interaction with peripheral proteins.
Collapse
Affiliation(s)
- Robert V Stahelin
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine-South Bend, South Bend, IN 46617, United States; Department of Chemistry and Biochemistry, University of Notre Dame, Notre Dame, IN 46556, United States; Mike and Josie Harper Cancer Research Institute, University of Notre Dame, Notre Dame, IN 46556, United States.
| | - Jordan L Scott
- Department of Chemistry and Biochemistry, University of Notre Dame, Notre Dame, IN 46556, United States; Mike and Josie Harper Cancer Research Institute, University of Notre Dame, Notre Dame, IN 46556, United States
| | - Cary T Frick
- Department of Chemistry and Biochemistry, University of Notre Dame, Notre Dame, IN 46556, United States
| |
Collapse
|
22
|
Kim YK, Na KS, Hwang JA, Yoon HK, Lee HJ, Hahn SW, Lee BH, Jung HY. High insulin-like growth factor-1 in patients with bipolar I disorder: a trait marker? J Affect Disord 2013; 151:738-743. [PMID: 24012102 DOI: 10.1016/j.jad.2013.07.041] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/02/2013] [Revised: 07/28/2013] [Accepted: 07/30/2013] [Indexed: 01/21/2023]
Abstract
OBJECTIVES Neurotrophic factors exert substantial effects on the central nervous system. The present study investigates the roles of insulin-like growth factor-1 (IGF-1), β-nerve growth factor (β-NGF), and brain-derived neurotrophic factor (BDNF) in bipolar disorder. METHODS Baseline levels of culture-stimulated IGF-1, β-NGF, and BDNF were compared in 116 patients with bipolar I disorder and 123 healthy controls. Neurotrophic factors were also compared in patients before and after 6 weeks of pharmacotherapy. A multivariate logistic regression analysis was used to investigate the influence of the neurotrophic factors analyzed in quartile form, in relation to confounding variables, such as age, sex, and body mass index. RESULTS IGF-1 was significantly higher in patients (mean=514.57, SD=259.78) than in healthy controls (mean=316.82, SD=270.00, p<0.0001) at baseline. Furthermore, higher levels of IGF-1 substantially increased the risk for bipolar I disorder. IGF-1 level was not significantly changed at 6-weeks (mean=506.41, SD=313.66). No changes in BDNF or β-NGF-1 levels were found following the 6-week treatment period. IGF-1 and β-NGF were negatively correlated in healthy controls, but not in patients. Severity of manic symptoms was not associated with any of the neurotrophic factors. LIMITATIONS We did not measure cortisol, growth hormone, or IGF-1 receptors. This study is cross-sectional in design. CONCLUSIONS Elevated IGF-1 levels may be a trait marker for bipolar disorder. Further studies are needed to thoroughly investigate the role of IGF-1 in relation to other neuroendocrine factors and biological markers for bipolar disorder.
Collapse
Affiliation(s)
- Yong-Ku Kim
- Department of Psychiatry, College of Medicine, Korea University, Seoul, Republic of Korea
| | - Kyoung-Sae Na
- Department of Psychiatry, Gachon University Gil Medical Center, 1198, Guwol-dong, Namdong-Gu, Incheon, Republic of Korea.
| | - Jung-A Hwang
- Department of Psychiatry, College of Medicine, Korea University, Seoul, Republic of Korea
| | - Ho-Kyoung Yoon
- Department of Psychiatry, College of Medicine, Korea University, Seoul, Republic of Korea
| | - Heon-Jeong Lee
- Department of Psychiatry, College of Medicine, Korea University, Seoul, Republic of Korea
| | - Sang-Woo Hahn
- Department of Psychiatry, College of Medicine, Soonchunhyang University, Seoul, Republic of Korea
| | - Bun-Hee Lee
- Department of Psychiatry, Gangnam Eulji Hospital, Eulji University, Seoul, Republic of Korea
| | - Han-Yong Jung
- Department of Psychiatry, College of Medicine, Soonchunhyang University, Seoul, Republic of Korea
| |
Collapse
|
23
|
No effect of adjunctive, repeated dose intranasal insulin treatment on body metabolism in patients with schizophrenia. Schizophr Res 2013; 146:40-5. [PMID: 23434504 PMCID: PMC3622829 DOI: 10.1016/j.schres.2013.01.034] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/13/2012] [Revised: 01/22/2013] [Accepted: 01/28/2013] [Indexed: 11/22/2022]
Abstract
OBJECTIVE This study examined the effect of adjunctive intranasal insulin therapy on body metabolism in patients with schizophrenia. METHOD Each subject had a DSM-IV diagnosis of schizophrenia or schizoaffective disorder and had been on stable dose of antipsychotic agent for at least one month. In an 8-week randomized, double-blind, placebo-controlled study, subjects received either intranasal insulin (40 IU 4 times per day) or placebo. The whole body dual-energy X-ray absorptiometry (DXA) was used to assess body composition. Lipid particles were assessed using nuclear magnetic resonance (NMR) spectroscopy. All assessments were conducted at baseline, and repeated at week 8. RESULTS A total number of 39 subjects completed the study (18 in the insulin group, 21 in the placebo group). There were no significant differences between the two groups in week 8 changes for body weight, body mass index, waist circumference, as well as various measures of lipid particles (p's>0.100). The DXA assessment showed no significant differences between the two groups in week 8 changes for fat mass, lean mass or total mass (p's>0.100). CONCLUSION In the present study, adjunctive therapy of intranasal insulin did not seem to improve body metabolism in patients with schizophrenia. The implications for future studies were discussed.
Collapse
|
24
|
No effect of adjunctive, repeated-dose intranasal insulin treatment on psychopathology and cognition in patients with schizophrenia. J Clin Psychopharmacol 2013; 33:226-30. [PMID: 23422397 PMCID: PMC5366038 DOI: 10.1097/jcp.0b013e31828701d0] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
OBJECTIVE This study examined the effect of adjunctive intranasal insulin therapy on psychopathology and cognition in patients with schizophrenia. METHODS Each subject had a Diagnostic and Statistical Manual of Mental Disorders, Fourth Edition, diagnosis of schizophrenia or schizoaffective disorder and been on stable antipsychotics for at least 1 month. In an 8-week randomized, double-blind, placebo-controlled study, subjects received either intranasal insulin (40 IU 4 times per day) or placebo. Psychopathology was assessed using the Positive and Negative Syndrome Scale and the Scale for Assessment of Negative Symptoms. A neuropsychological battery was used to assess cognitive performance. The assessment for psychopathology and cognition was conducted at baseline, week 4, and week 8. RESULTS A total of 45 subjects were enrolled in the study (21 in the insulin group and 24 in the placebo group). The mixed model analysis showed that there were no significant differences between the 2 groups at week 8 on various psychopathology and cognitive measures (P > 0.1). CONCLUSIONS Adjunctive therapy with intranasal insulin did not seem to be beneficial in improving schizophrenia symptoms or cognition in the present study. The implications for future studies were discussed.
Collapse
|
25
|
Bechtel W, Helmstädter M, Balica J, Hartleben B, Kiefer B, Hrnjic F, Schell C, Kretz O, Liu S, Geist F, Kerjaschki D, Walz G, Huber TB. Vps34 deficiency reveals the importance of endocytosis for podocyte homeostasis. J Am Soc Nephrol 2013; 24:727-43. [PMID: 23492732 DOI: 10.1681/asn.2012070700] [Citation(s) in RCA: 113] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
The molecular mechanisms that maintain podocytes and consequently, the integrity of the glomerular filtration barrier are incompletely understood. Here, we show that the class III phosphoinositide 3-kinase vacuolar protein sorting 34 (Vps34) plays a central role in modulating endocytic pathways, maintaining podocyte homeostasis. In mice, podocyte-specific conditional knockout of Vps34 led to early proteinuria, glomerular scarring, and death within 3-9 weeks of age. Vps34-deficient podocytes exhibited substantial vacuolization and foot process effacement. Although the formation of autophagosomes and autophagic flux were impaired, comparisons between podocyte-specific Vps34-deficient mice, autophagy-deficient mice, and doubly deficient mice suggested that defective autophagy was not primarily responsible for the severe phenotype caused by the loss of Vps34. In fact, Rab5-positive endosomal compartments, endocytosis, and fluid-phase uptake were severely disrupted in Vps34-deficient podocytes. Vps34 deficiency in nephrocytes, the podocyte-like cells of Drosophila melanogaster, resulted in a block between Rab5- and Rab7-positive endosomal compartments. In summary, these data identify Vps34 as a major regulator of endolysosomal pathways in podocytes and underline the fundamental roles of endocytosis and fluid-phase uptake for the maintenance of the glomerular filtration barrier.
Collapse
Affiliation(s)
- Wibke Bechtel
- Renal Division, University Hospital Freiburg, Breisacher Str. 66, 79106 Freiburg, Germany
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
26
|
Menke A, Domschke K, Czamara D, Klengel T, Hennings J, Lucae S, Baune BT, Arolt V, Müller-Myhsok B, Holsboer F, Binder EB. Genome-wide association study of antidepressant treatment-emergent suicidal ideation. Neuropsychopharmacology 2012; 37:797-807. [PMID: 22030708 PMCID: PMC3260972 DOI: 10.1038/npp.2011.257] [Citation(s) in RCA: 56] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/28/2011] [Revised: 09/09/2011] [Accepted: 09/26/2011] [Indexed: 11/08/2022]
Abstract
Emergence of suicidal ideation (TESI) during treatment with antidepressants in major depression led to a black box warning. We performed a genome-wide association study to identify genetic markers, which increase the risk for this serious side effect. TESI was evaluated in depressed in-patients (N=397) and defined by an emergence of suicidal thoughts during hospitalization without suicidal thoughts at admission using the suicide item (3) of the Hamilton Depression Rating Scale. Genotype distribution of 405.383 single-nucleotide polymorphisms (SNPs) in patients with TESI (N=32/8.1%) was compared to patients without increase in suicidal ideation (N=329/82.9%) and to a subgroup never reported suicidal ideation (N=79/19.9%). Top results were analyzed in an independent sample (N=501). None variant reached genome-wide significance, the best associated SNP was rs1630535 (p-value=1.3 × 10(-7)). The top 79 SNPs could be analyzed in an independent sample, and 14 variants showed nominal significant association with the same risk allele in the replication sample. A discriminant analysis classifying patients using these 79 SNPs revealed a 91% probability to classify TESI vs non-TESI cases correctly in the replication sample. Although our data need to be interpreted carefully owing to the small numbers in both cohorts, they suggest that a combination of genetic markers might indeed be used to identify patients at risk for TESI.
Collapse
Affiliation(s)
- Andreas Menke
- Max Planck Institute of Psychiatry, Munich, Germany.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
27
|
Abstract
A literature review was conducted on the genetic and developmental bases of autism in relation to genes and pathways associated with cancer risk. Convergent lines of evidence from four types of analysis: (1) recent theoretical studies on the causes of autism, (2) epidemiological studies, (3) genetic analyses linking autism with mutations in tumor suppressor genes and other cancer-associated genes and pathways, and (4) contrasts with schizophrenia, Parkinson's, and Alzheimer's disease indicate that autism may involve altered cancer risk. This evidence should motivate further epidemiological studies, and it provides useful insights into the nature of the genetic, epigenetic, and environmental factors underlying the etiologies of autism, other neurological conditions, and carcinogenesis.
Collapse
Affiliation(s)
- B Crespi
- Department of Biological Sciences, Simon Fraser University, Burnaby (B.C.), British Columbia, Canada.
| |
Collapse
|
28
|
Vounou M, Janousova E, Wolz R, Stein JL, Thompson PM, Rueckert D, Montana G. Sparse reduced-rank regression detects genetic associations with voxel-wise longitudinal phenotypes in Alzheimer's disease. Neuroimage 2011; 60:700-16. [PMID: 22209813 DOI: 10.1016/j.neuroimage.2011.12.029] [Citation(s) in RCA: 92] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2011] [Revised: 11/18/2011] [Accepted: 12/14/2011] [Indexed: 11/17/2022] Open
Abstract
Scanning the entire genome in search of variants related to imaging phenotypes holds great promise in elucidating the genetic etiology of neurodegenerative disorders. Here we discuss the application of a penalized multivariate model, sparse reduced-rank regression (sRRR), for the genome-wide detection of markers associated with voxel-wise longitudinal changes in the brain caused by Alzheimer's disease (AD). Using a sample from the Alzheimer's Disease Neuroimaging Initiative database, we performed three separate studies that each compared two groups of individuals to identify genes associated with disease development and progression. For each comparison we took a two-step approach: initially, using penalized linear discriminant analysis, we identified voxels that provide an imaging signature of the disease with high classification accuracy; then we used this multivariate biomarker as a phenotype in a genome-wide association study, carried out using sRRR. The genetic markers were ranked in order of importance of association to the phenotypes using a data re-sampling approach. Our findings confirmed the key role of the APOE and TOMM40 genes but also highlighted some novel potential associations with AD.
Collapse
Affiliation(s)
- Maria Vounou
- Statistics Section, Department of Mathematics, Imperial College London, UK
| | | | | | | | | | | | | |
Collapse
|
29
|
Carrard A, Salzmann A, Perroud N, Gafner J, Malafosse A, Karege F. Genetic association of the Phosphoinositide-3 kinase in schizophrenia and bipolar disorder and interaction with a BDNF gene polymorphism. Brain Behav 2011; 1:119-24. [PMID: 22399091 PMCID: PMC3236546 DOI: 10.1002/brb3.23] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/24/2011] [Revised: 08/08/2011] [Accepted: 09/10/2011] [Indexed: 01/14/2023] Open
Abstract
Phosphoinositide-3-kinase, class III (PIK3C3) is a member of the phosphoinosite-3-kinases family, involved in cell signaling, membrane trafficking, and neurodevelopment. Previous studies have indeed shown an association between PIK3C3 gene variants and both bipolar disorder (BD) and schizophrenia (SZ). Brain-derived neurotrophic factor (BDNF) is a neurodevelopmental factor, which can regulate the PI3K signaling pathway. Associations have been reported between BDNF gene polymorphisms and affective and psychotic disorders. The aim of the present study was to replicate an association between PIK3C3 and BDNF gene variants in SZ and BD and a putative epistasis between the two genes. Patients meeting the DSM-IV criteria of BD and SZ were included in this study (98 BD and 79 SZ) as well as 158 healthy controls. Blood DNA was extracted and genotyping was performed either by the polymerase chain reaction (PCR) technique followed by enzymatic digestion or by the high-resolution melt (HRM) method. Genotype and haplotype association was assessed with the UNPHASED statistical program.The results showed one nominal association with BD (P < 0.02) and two risk haplotypes in both SZ (P < 0.001) and BP (P < 0.0005), which survived multiple testing correction. A modest interaction between a BDNF variant and PI3KC3 polymorphism was observed (P < 0.04).These preliminary results confirm the genetic association of PI3K gene variants with both SZ and BD, and support the hypothesis that SZ and BD share a genetic background.
Collapse
Affiliation(s)
- Anthony Carrard
- Department of Psychiatry, University of Geneva, 2 ch Petit Bel‐Air, CH‐1225 Chêne‐Bourg, Geneva, Switzerland
| | - Annick Salzmann
- Department of Psychiatry, University of Geneva, 2 ch Petit Bel‐Air, CH‐1225 Chêne‐Bourg, Geneva, Switzerland
| | - Nader Perroud
- Department of Medical Genetics and Laboratory, Geneva University Hospitals, 2 ch Petit Bel‐Air, CH‐1225 Chêne‐Bourg, Geneva, Switzerland
| | - Jérémie Gafner
- Department of Psychiatry, University of Geneva, 2 ch Petit Bel‐Air, CH‐1225 Chêne‐Bourg, Geneva, Switzerland
| | - Alain Malafosse
- Department of Psychiatry, University of Geneva, 2 ch Petit Bel‐Air, CH‐1225 Chêne‐Bourg, Geneva, Switzerland
- Department of Medical Genetics and Laboratory, Geneva University Hospitals, 2 ch Petit Bel‐Air, CH‐1225 Chêne‐Bourg, Geneva, Switzerland
| | - Félicien Karege
- Department of Psychiatry, University of Geneva, 2 ch Petit Bel‐Air, CH‐1225 Chêne‐Bourg, Geneva, Switzerland
- Department of Medical Genetics and Laboratory, Geneva University Hospitals, 2 ch Petit Bel‐Air, CH‐1225 Chêne‐Bourg, Geneva, Switzerland
| |
Collapse
|
30
|
Heritability of genetic variants of resistin gene in patients with coronary artery disease: A family-based study. Clin Biochem 2011; 44:618-22. [DOI: 10.1016/j.clinbiochem.2011.02.013] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2010] [Revised: 01/29/2011] [Accepted: 02/23/2011] [Indexed: 12/20/2022]
|
31
|
Wang L, Budolfson K, Wang F. Pik3c3 deletion in pyramidal neurons results in loss of synapses, extensive gliosis and progressive neurodegeneration. Neuroscience 2011; 172:427-42. [PMID: 20955765 PMCID: PMC3010427 DOI: 10.1016/j.neuroscience.2010.10.035] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2010] [Revised: 10/04/2010] [Accepted: 10/11/2010] [Indexed: 11/18/2022]
Abstract
The lipid kinase PIK3C3 (also known as VPS34) regulates multiple aspects of endo-membrane trafficking processes. PIK3C3 is widely expressed by neurons in the CNS, and its catalytic product PI3P is enriched in dendritic spines. Here we generated a line of conditional mutant mouse in which Pik3c3 is specifically deleted in hippocampal and in small subsets of cortical pyramidal neurons using the CaMKII-Cre transgene. We found that Pik3c3-deficiency initially causes loss of dendritic spines accompanied with reactive gliosis, which is followed by progressive neuronal degeneration over a period of several months. Layers III and IV cortical neurons are more susceptible to Pik3c3-deletion than hippocampal neurons. Furthermore, in aged conditional Pik3c3 mutant animals, there are extensive gliosis and severe secondary loss of wild type neurons. Our analyses show that Pik3c3 is essential for CNS neuronal homeostasis and Pik3c3flox/flox; CaMKII-Cre mouse is a useful model for studying pathological changes in progressive forebrain neurodegeneration.
Collapse
Affiliation(s)
- Liangli Wang
- Department of Cell Biology, Duke University Medical Center, Box 3709, Durham, NC 27710
| | - Katie Budolfson
- Department of Cell Biology, Duke University Medical Center, Box 3709, Durham, NC 27710
| | - Fan Wang
- Department of Cell Biology, Duke University Medical Center, Box 3709, Durham, NC 27710
| |
Collapse
|
32
|
HIROSE K, TAKIZAWA T, FUKAWA K, ITO T, UEDA M, HAYASHI Y, TANAKA K. Association of an SNP marker in exon 24 of a class 3 phosphoinositide-3-kinase (PIK3C3) gene with production traits in Duroc pigs. Anim Sci J 2010; 82:46-51. [DOI: 10.1111/j.1740-0929.2010.00816.x] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
|
33
|
Chen X, Li X, Wang P, Liu Y, Zhang Z, Zhao G, Xu H, Zhu J, Qin X, Chen S, Hu L, Kong X. Novel association strategy with copy number variation for identifying new risk Loci of human diseases. PLoS One 2010; 5:e12185. [PMID: 20808825 PMCID: PMC2924882 DOI: 10.1371/journal.pone.0012185] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2010] [Accepted: 07/20/2010] [Indexed: 12/20/2022] Open
Abstract
Background Copy number variations (CNV) are important causal genetic variations for human disease; however, the lack of a statistical model has impeded the systematic testing of CNVs associated with disease in large-scale cohort. Methodology/Principal Findings Here, we developed a novel integrated strategy to test CNV-association in genome-wide case-control studies. We converted the single-nucleotide polymorphism (SNP) signal to copy number states using a well-trained hidden Markov model. We mapped the susceptible CNV-loci through SNP site-specific testing to cope with the physiological complexity of CNVs. We also ensured the credibility of the associated CNVs through further window-based CNV-pattern clustering. Genome-wide data with seven diseases were used to test our strategy and, in total, we identified 36 new susceptible loci that are associated with CNVs for the seven diseases: 5 with bipolar disorder, 4 with coronary artery disease, 1 with Crohn's disease, 7 with hypertension, 9 with rheumatoid arthritis, 7 with type 1 diabetes and 3 with type 2 diabetes. Fifteen of these identified loci were validated through genotype-association and physiological function from previous studies, which provide further confidence for our results. Notably, the genes associated with bipolar disorder converged in the phosphoinositide/calcium signaling, a well-known affected pathway in bipolar disorder, which further supports that CNVs have impact on bipolar disorder. Conclusions/Significance Our results demonstrated the effectiveness and robustness of our CNV-association analysis and provided an alternative avenue for discovering new associated loci of human diseases.
Collapse
Affiliation(s)
- Xianfeng Chen
- The Key Laboratory of Stem Cell Biology, Institute of Health Sciences, Shanghai Institutes for Biological Sciences (SIBS), Chinese Academy of Sciences (CAS) and Shanghai Jiao Tong University School of Medicine (SJTUSM), Shanghai, People's Republic of China
| | - Xinlei Li
- The Key Laboratory of Stem Cell Biology, Institute of Health Sciences, Shanghai Institutes for Biological Sciences (SIBS), Chinese Academy of Sciences (CAS) and Shanghai Jiao Tong University School of Medicine (SJTUSM), Shanghai, People's Republic of China
| | - Ping Wang
- The Key Laboratory of Stem Cell Biology, Institute of Health Sciences, Shanghai Institutes for Biological Sciences (SIBS), Chinese Academy of Sciences (CAS) and Shanghai Jiao Tong University School of Medicine (SJTUSM), Shanghai, People's Republic of China
| | - Yang Liu
- The Key Laboratory of Stem Cell Biology, Institute of Health Sciences, Shanghai Institutes for Biological Sciences (SIBS), Chinese Academy of Sciences (CAS) and Shanghai Jiao Tong University School of Medicine (SJTUSM), Shanghai, People's Republic of China
| | - Zhenguo Zhang
- The Key Laboratory of Stem Cell Biology, Institute of Health Sciences, Shanghai Institutes for Biological Sciences (SIBS), Chinese Academy of Sciences (CAS) and Shanghai Jiao Tong University School of Medicine (SJTUSM), Shanghai, People's Republic of China
| | - Guoping Zhao
- Chinese National Human Genome Center at Shanghai, Shanghai, People's Republic of China
| | - Haiming Xu
- Institute of Bioinformatics, College of Agriculture and Biotechnology, Zhejiang University, Hangzhou, People's Republic of China
| | - Jun Zhu
- Institute of Bioinformatics, College of Agriculture and Biotechnology, Zhejiang University, Hangzhou, People's Republic of China
| | - Xueying Qin
- State Key Laboratory of Computer Aided Design and Computer Graphics, Zhejiang University, Hangzhou, People's Republic of China
| | - Suchao Chen
- State Key Laboratory of Computer Aided Design and Computer Graphics, Zhejiang University, Hangzhou, People's Republic of China
| | - Landian Hu
- The Key Laboratory of Stem Cell Biology, Institute of Health Sciences, Shanghai Institutes for Biological Sciences (SIBS), Chinese Academy of Sciences (CAS) and Shanghai Jiao Tong University School of Medicine (SJTUSM), Shanghai, People's Republic of China
- State Key Laboratory of Medical Genomics, Ruijin Hospital, Shanghai Jiaotong University, Shanghai, People's Republic of China
| | - Xiangyin Kong
- The Key Laboratory of Stem Cell Biology, Institute of Health Sciences, Shanghai Institutes for Biological Sciences (SIBS), Chinese Academy of Sciences (CAS) and Shanghai Jiao Tong University School of Medicine (SJTUSM), Shanghai, People's Republic of China
- State Key Laboratory of Medical Genomics, Ruijin Hospital, Shanghai Jiaotong University, Shanghai, People's Republic of China
- * E-mail:
| |
Collapse
|
34
|
Promoter variant of PIK3C3 is associated with autoimmunity against Ro and Sm epitopes in African-American lupus patients. J Biomed Biotechnol 2010; 2010:826434. [PMID: 20671926 PMCID: PMC2910508 DOI: 10.1155/2010/826434] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2010] [Revised: 05/07/2010] [Accepted: 05/24/2010] [Indexed: 01/02/2023] Open
Abstract
The PIK3C3 locus was implicated in case-case genome-wide association study of systemic lupus erythematosus (SLE) which we had performed to detect genes associated with autoantibodies and serum interferon-alpha (IFN-α). Herein, we examine a PIK3C3 promoter variant (rs3813065/-442 C/T) in an independent multiancestral cohort of 478 SLE cases and 522 controls. rs3813065 C was strongly associated with the simultaneous presence of both anti-Ro and anti-Sm antibodies in African-American patients [OR = 2.24 (1.34–3.73), P = 2.0 × 10−3]. This autoantibody profile was associated with higher serum IFN-α (P = 7.6 × 10−6). In the HapMap Yoruba population, rs3813065 was associated with differential expression of ERAP2 (P = 2.0 × 10−5), which encodes an enzyme involved in MHC class I peptide processing. Thus, rs3813065 C is associated with a particular autoantibody profile and altered expression of an MHC peptide processing enzyme, suggesting that this variant modulates serologic autoimmunity in African-American SLE patients.
Collapse
|
35
|
Karam CS, Ballon JS, Bivens NM, Freyberg Z, Girgis RR, Lizardi-Ortiz JE, Markx S, Lieberman JA, Javitch JA. Signaling pathways in schizophrenia: emerging targets and therapeutic strategies. Trends Pharmacol Sci 2010; 31:381-90. [PMID: 20579747 DOI: 10.1016/j.tips.2010.05.004] [Citation(s) in RCA: 127] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2010] [Revised: 05/17/2010] [Accepted: 05/20/2010] [Indexed: 02/07/2023]
Abstract
Dopamine D(2) receptor antagonism is a unifying property of all antipsychotic drugs in use for schizophrenia. While often effective at ameliorating psychosis, these drugs are largely ineffective at treating negative and cognitive symptoms. Increasing attention is being focused on the complex genetics of the illness and the signaling pathways implicated in its pathophysiology. We review targeted approaches for pharmacotherapy involving the glutamatergic, GABAergic and cholinergic pathways. We also describe several of the major genetic findings that identify signaling pathways representing potential targets for novel pharmacological intervention. These include genes in the 22q11 locus, DISC1, Neuregulin 1/ErbB4, and components of the Akt/GSK-3 pathway.
Collapse
Affiliation(s)
- Caline S Karam
- Department of Psychiatry, Columbia University College of Physicians and Surgeons, New York, NY 10032, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
36
|
Strong synaptic transmission impact by copy number variations in schizophrenia. Proc Natl Acad Sci U S A 2010; 107:10584-9. [PMID: 20489179 DOI: 10.1073/pnas.1000274107] [Citation(s) in RCA: 169] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Schizophrenia is a psychiatric disorder with onset in late adolescence and unclear etiology characterized by both positive and negative symptoms, as well as cognitive deficits. To identify copy number variations (CNVs) that increase the risk of schizophrenia, we performed a whole-genome CNV analysis on a cohort of 977 schizophrenia cases and 2,000 healthy adults of European ancestry who were genotyped with 1.7 million probes. Positive findings were evaluated in an independent cohort of 758 schizophrenia cases and 1,485 controls. The Gene Ontology synaptic transmission family of genes was notably enriched for CNVs in the cases (P = 1.5 x 10(-7)). Among these, CACNA1B and DOC2A, both calcium-signaling genes responsible for neuronal excitation, were deleted in 16 cases and duplicated in 10 cases, respectively. In addition, RET and RIT2, both ras-related genes important for neural crest development, were significantly affected by CNVs. RET deletion was exclusive to seven cases, and RIT2 deletions were overrepresented common variant CNVs in the schizophrenia cases. Our results suggest that novel variations involving the processes of synaptic transmission contribute to the genetic susceptibility of schizophrenia.
Collapse
|
37
|
Deletion of PIK3C3/Vps34 in sensory neurons causes rapid neurodegeneration by disrupting the endosomal but not the autophagic pathway. Proc Natl Acad Sci U S A 2010; 107:9424-9. [PMID: 20439739 DOI: 10.1073/pnas.0914725107] [Citation(s) in RCA: 192] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
The lipid kinase PIK3C3 (also called Vps34) regulates both the endosomal and autophagic pathways. However, the effect of inactivating PIK3C3 on neuronal endosomal versus autophagic processes in vivo has not been studied. We generated mice in which Pik3c3 was conditionally deleted in differentiated sensory neurons. Within a few days after Pik3c3 deletion, mutant large-diameter myelinated neurons accumulated numerous enlarged vacuoles and ubiquitin-positive aggregates and underwent rapid degeneration. By contrast, Pik3c3-deficient small-diameter unmyelinated neurons accumulated excessive numbers of lysosome-like organelles and degenerated more slowly. These differential degenerative phenotypes are unlikely caused by a disruption in the autophagy pathway, because inhibiting autophagy alone by conditional deletion of Atg7 results in a completely distinct phenotype in all sensory neurons (i.e., formation of very large intracellular inclusion bodies and slow degeneration over a period of several months). More surprisingly, a noncanonical PIK3C3-independent LC3-positive autophagosome formation pathway was activated in Pik3c3-deficient small-diameter neurons. Analyses of Pik3c3/Atg7 double mutant neurons revealed that this unconventional initiation pathway still depends on ATG7. Our studies represent in vivo characterization of PIK3C3 functions in mammals and provide insights into the complexity of neuronal endo-lysosomal and autophagic pathways.
Collapse
|
38
|
Crespi B, Stead P, Elliot M. Evolution in health and medicine Sackler colloquium: Comparative genomics of autism and schizophrenia. Proc Natl Acad Sci U S A 2010; 107 Suppl 1:1736-41. [PMID: 19955444 PMCID: PMC2868282 DOI: 10.1073/pnas.0906080106] [Citation(s) in RCA: 195] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
We used data from studies of copy-number variants (CNVs), single-gene associations, growth-signaling pathways, and intermediate phenotypes associated with brain growth to evaluate four alternative hypotheses for the genomic and developmental relationships between autism and schizophrenia: (i) autism subsumed in schizophrenia, (ii) independence, (iii) diametric, and (iv) partial overlap. Data from CNVs provides statistical support for the hypothesis that autism and schizophrenia are associated with reciprocal variants, such that at four loci, deletions predispose to one disorder, whereas duplications predispose to the other. Data from single-gene studies are inconsistent with a hypothesis based on independence, in that autism and schizophrenia share associated genes more often than expected by chance. However, differentiation between the partial overlap and diametric hypotheses using these data is precluded by limited overlap in the specific genetic markers analyzed in both autism and schizophrenia. Evidence from the effects of risk variants on growth-signaling pathways shows that autism-spectrum conditions tend to be associated with up-regulation of pathways due to loss of function mutations in negative regulators, whereas schizophrenia is associated with reduced pathway activation. Finally, data from studies of head and brain size phenotypes indicate that autism is commonly associated with developmentally-enhanced brain growth, whereas schizophrenia is characterized, on average, by reduced brain growth. These convergent lines of evidence appear most compatible with the hypothesis that autism and schizophrenia represent diametric conditions with regard to their genomic underpinnings, neurodevelopmental bases, and phenotypic manifestations as reflecting under-development versus dysregulated over-development of the human social brain.
Collapse
Affiliation(s)
- Bernard Crespi
- Department of Biological Sciences, Simon Fraser University, Burnaby, BC, Canada.
| | | | | |
Collapse
|
39
|
Jiang B, Kenna HA, Rasgon NL. Genetic overlap between polycystic ovary syndrome and bipolar disorder: The endophenotype hypothesis. Med Hypotheses 2009; 73:996-1004. [DOI: 10.1016/j.mehy.2008.12.056] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2008] [Revised: 12/03/2008] [Accepted: 12/07/2008] [Indexed: 12/13/2022]
|
40
|
Falasca M, Maffucci T. Rethinking phosphatidylinositol 3-monophosphate. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2009; 1793:1795-803. [PMID: 19852987 DOI: 10.1016/j.bbamcr.2009.10.003] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Received: 06/13/2009] [Revised: 10/06/2009] [Accepted: 10/13/2009] [Indexed: 11/27/2022]
Abstract
A generally accepted view considers phosphatidylinositol 3-monophosphate (PtdIns3P) as a lipid confined to the endosomal compartment where it regulates trafficking pathways and is produced constitutively and exclusively by class III phosphoinositide 3-kinase (PI3K). Recent evidence suggests that this phosphoinositide has a more complex role as a second messenger involved in different physiological and pathological events and that specific intracellular localization of kinases and/or phosphatases is critical for PtdIns3P synthesis and PtdIns3P-dependent intracellular functions. Here, we review the current knowledge of the regulation and function of PtdIns3P and discuss how the view of PtdIns3P changed in the last few years.
Collapse
Affiliation(s)
- Marco Falasca
- Queen Mary University of London, Barts and The London School of Medicine and Dentistry, Blizard Institute of Cell and Molecular Science, Centre for Diabetes, Inositide Signalling Group, 4 Newark Street, London E1 2AT, UK.
| | | |
Collapse
|
41
|
Sasaki T, Takasuga S, Sasaki J, Kofuji S, Eguchi S, Yamazaki M, Suzuki A. Mammalian phosphoinositide kinases and phosphatases. Prog Lipid Res 2009; 48:307-43. [PMID: 19580826 DOI: 10.1016/j.plipres.2009.06.001] [Citation(s) in RCA: 206] [Impact Index Per Article: 12.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Phosphoinositides are lipids that are present in the cytoplasmic leaflet of a cell's plasma and internal membranes and play pivotal roles in the regulation of a wide variety of cellular processes. Phosphoinositides are molecularly diverse due to variable phosphorylation of the hydroxyl groups of their inositol rings. The rapid and reversible configuration of the seven known phosphoinositide species is controlled by a battery of phosphoinositide kinases and phosphoinositide phosphatases, which are thus critical for phosphoinositide isomer-specific localization and functions. Significantly, a given phosphoinositide generated by different isozymes of these phosphoinositide kinases and phosphatases can have different biological effects. In mammals, close to 50 genes encode the phosphoinositide kinases and phosphoinositide phosphatases that regulate phosphoinositide metabolism and thus allow cells to respond rapidly and effectively to ever-changing environmental cues. Understanding the distinct and overlapping functions of these phosphoinositide-metabolizing enzymes is important for our knowledge of both normal human physiology and the growing list of human diseases whose etiologies involve these proteins. This review summarizes the structural and biological properties of all the known mammalian phosphoinositide kinases and phosphoinositide phosphatases, as well as their associations with human disorders.
Collapse
Affiliation(s)
- Takehiko Sasaki
- Department of Pathology and Immunology, Akita University, Graduate School of Medicine, Akita 010-8543, Japan.
| | | | | | | | | | | | | |
Collapse
|
42
|
Liu J, Kiehl KA, Pearlson G, Perrone-Bizzozero NI, Eichele T, Calhoun VD. Genetic determinants of target and novelty-related event-related potentials in the auditory oddball response. Neuroimage 2009; 46:809-16. [PMID: 19285141 PMCID: PMC2676714 DOI: 10.1016/j.neuroimage.2009.02.045] [Citation(s) in RCA: 52] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2008] [Revised: 12/24/2008] [Accepted: 02/23/2009] [Indexed: 10/21/2022] Open
Abstract
Processing of novel and target stimuli in the auditory target detection or 'oddball' task encompasses the chronometry of perception, attention and working memory and is reflected in scalp recorded event-related potentials (ERPs). A variety of ERP components related to target and novelty processing have been described and extensively studied, and linked to deficits of cognitive processing. However, little is known about associations of genotypes with ERP endophenotypes. Here we sought to elucidate the genetic underpinnings of auditory oddball ERP components using a novel data analysis technique. A parallel independent component analysis of the electrophysiology and single nucleotide polymorphism (SNP) data was used to extract relations between patterns of ERP components and SNP associations purely based on an analysis incorporating higher order statistics. The method allows for broader associations of genotypes with phenotypes than traditional hypothesis-driven univariate correlational analyses. We show that target detection and processing of novel stimuli are both associated with a shared cluster of genes linked to the adrenergic and dopaminergic pathways. These results provide evidence of genetic influences on normal patterns of ERP generation during auditory target detection and novelty processing at the SNP association level.
Collapse
Affiliation(s)
- Jingyu Liu
- The Mind Research Network, Albuquerque, NM 87131, USA.
| | | | | | | | | | | |
Collapse
|
43
|
McCrea HJ, De Camilli P. Mutations in phosphoinositide metabolizing enzymes and human disease. Physiology (Bethesda) 2009; 24:8-16. [PMID: 19196647 DOI: 10.1152/physiol.00035.2008] [Citation(s) in RCA: 134] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Phosphoinositides are implicated in the regulation of a wide variety of cellular functions. Their importance in cellular and organismal physiology is underscored by the growing number of human diseases linked to perturbation of kinases and phosphatases that catalyze interconversion from one phosphoinositide to another. Many such enzymes are attractive targets for therapeutic interventions. Here, we review diseases linked to inheritable or somatic mutations of these enzymes.
Collapse
Affiliation(s)
- Heather J McCrea
- Howard Hughes Medical Institute, Department of Cell Biology, Kavli Institute for Neuroscience, Program in Cellular Neuroscience, Neurodegeneration and Repair, Yale University School of Medicine, New Haven, Connecticut, USA
| | | |
Collapse
|
44
|
Kanahara N, Miyatake R, Sekine Y, Inada T, Ozaki N, Iwata N, Harano M, Komiyama T, Yamada M, Sora I, Ujike H, Iyo M, Hashimoto K. Association study between the PIK4CA gene and methamphetamine use disorder in a Japanese population. Am J Med Genet B Neuropsychiatr Genet 2009; 150B:233-8. [PMID: 18521859 DOI: 10.1002/ajmg.b.30797] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Accumulating evidence suggests that phosphatidylinositol (PI) pathways have been involved in the secretion of dopamine (DA) and the regulation of DA transporter, which is a target of methamphetamine (METH). A recent large-scale gene-association study in a Dutch population demonstrated that the PIK4CA gene was closely linked to schizophrenia [Jungerius et al. (2007); Mol Psychiatry]. Here, we conducted a case (N = 232)-control (N = 233) study of the PIK4CA gene on Japanese METH abusers, which can manifest severe psychosis similar to schizophrenia. The genotype and allelic distributions of all four single nucleotide polymorphisms (SNPs) did not differ significantly between the METH abusers and the controls. The comparisons based on the classification of the psychosis as transient or prolonged and on the presence or absence of spontaneous relapse revealed no significant distribution of the four SNPs compared to the controls. Furthermore, haplotype analyses showed almost the same frequencies between the METH abusers and the controls. The present study suggests that the PIK4CA gene does not play a significant role in the vulnerability to METH use disorder in the Japanese population.
Collapse
Affiliation(s)
- Nobuhisa Kanahara
- Department of Psychiatry, Chiba University Graduate School of Medicine, Japan
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Crespi B. Genomic imprinting in the development and evolution of psychotic spectrum conditions. Biol Rev Camb Philos Soc 2008; 83:441-493. [PMID: 18783362 DOI: 10.1111/j.1469-185x.2008.00050.x] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
I review and evaluate genetic and genomic evidence salient to the hypothesis that the development and evolution of psychotic spectrum conditions have been mediated in part by alterations of imprinted genes expressed in the brain. Evidence from the genetics and genomics of schizophrenia, bipolar disorder, major depression, Prader-Willi syndrome, Klinefelter syndrome, and other neurogenetic conditions support the hypothesis that the etiologies of psychotic spectrum conditions commonly involve genetic and epigenetic imbalances in the effects of imprinted genes, with a bias towards increased relative effects from imprinted genes with maternal expression or other genes favouring maternal interests. By contrast, autistic spectrum conditions, including Kanner autism, Asperger syndrome, Rett syndrome, Turner syndrome, Angelman syndrome, and Beckwith-Wiedemann syndrome, commonly engender increased relative effects from paternally expressed imprinted genes, or reduced effects from genes favouring maternal interests. Imprinted-gene effects on the etiologies of autistic and psychotic spectrum conditions parallel the diametric effects of imprinted genes in placental and foetal development, in that psychotic spectrum conditions tend to be associated with undergrowth and relatively-slow brain development, whereas some autistic spectrum conditions involve brain and body overgrowth, especially in foetal development and early childhood. An important role for imprinted genes in the etiologies of psychotic and autistic spectrum conditions is consistent with neurodevelopmental models of these disorders, and with predictions from the conflict theory of genomic imprinting.
Collapse
Affiliation(s)
- Bernard Crespi
- Department of Biosciences, Simon Fraser University, Burnaby BCV5A1S6, Canada.
| |
Collapse
|
46
|
Vamathevan JJ, Hasan S, Emes RD, Amrine-Madsen H, Rajagopalan D, Topp SD, Kumar V, Word M, Simmons MD, Foord SM, Sanseau P, Yang Z, Holbrook JD. The role of positive selection in determining the molecular cause of species differences in disease. BMC Evol Biol 2008; 8:273. [PMID: 18837980 PMCID: PMC2576240 DOI: 10.1186/1471-2148-8-273] [Citation(s) in RCA: 72] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2008] [Accepted: 10/06/2008] [Indexed: 11/20/2022] Open
Abstract
BACKGROUND Related species, such as humans and chimpanzees, often experience the same disease with varying degrees of pathology, as seen in the cases of Alzheimer's disease, or differing symptomatology as in AIDS. Furthermore, certain diseases such as schizophrenia, epithelial cancers and autoimmune disorders are far more frequent in humans than in other species for reasons not associated with lifestyle. Genes that have undergone positive selection during species evolution are indicative of functional adaptations that drive species differences. Thus we investigate whether biomedical disease differences between species can be attributed to positively selected genes. RESULTS We identified genes that putatively underwent positive selection during the evolution of humans and four mammals which are often used to model human diseases (mouse, rat, chimpanzee and dog). We show that genes predicted to have been subject to positive selection pressure during human evolution are implicated in diseases such as epithelial cancers, schizophrenia, autoimmune diseases and Alzheimer's disease, all of which differ in prevalence and symptomatology between humans and their mammalian relatives. In agreement with previous studies, the chimpanzee lineage was found to have more genes under positive selection than any of the other lineages. In addition, we found new evidence to support the hypothesis that genes that have undergone positive selection tend to interact with each other. This is the first such evidence to be detected widely among mammalian genes and may be important in identifying molecular pathways causative of species differences. CONCLUSION Our dataset of genes predicted to have been subject to positive selection in five species serves as an informative resource that can be consulted prior to selecting appropriate animal models during drug target validation. We conclude that studying the evolution of functional and biomedical disease differences between species is an important way to gain insight into their molecular causes and may provide a method to predict when animal models do not mirror human biology.
Collapse
Affiliation(s)
- Jessica J Vamathevan
- Department of Biology, University College London, Darwin Bldg, Gower Street, London WC1E 6BT, UK
| | - Samiul Hasan
- Computational Biology Division, Molecular Discovery Research, GlaxoSmithKline R&D Ltd., 1250 South Collegeville Road, Collegeville, PA 19426, USA
| | - Richard D Emes
- Institute for Science and Technology in Medicine, Keele University, Thornburrow Drive, Hartshill, Stoke-on-Trent, ST4 7QB, UK
| | - Heather Amrine-Madsen
- Computational Biology Division, Molecular Discovery Research, GlaxoSmithKline R&D Ltd., 1250 South Collegeville Road, Collegeville, PA 19426, USA
| | - Dilip Rajagopalan
- Computational Biology Division, Molecular Discovery Research, GlaxoSmithKline R&D Ltd., 1250 South Collegeville Road, Collegeville, PA 19426, USA
| | - Simon D Topp
- Computational Biology Division, Molecular Discovery Research, GlaxoSmithKline R&D Ltd., 1250 South Collegeville Road, Collegeville, PA 19426, USA
| | - Vinod Kumar
- Computational Biology Division, Molecular Discovery Research, GlaxoSmithKline R&D Ltd., 1250 South Collegeville Road, Collegeville, PA 19426, USA
| | - Michael Word
- Computational Biology Division, Molecular Discovery Research, GlaxoSmithKline R&D Ltd., 1250 South Collegeville Road, Collegeville, PA 19426, USA
| | - Mark D Simmons
- Molecular Discovery Research Information Technology, GlaxoSmithKline R&D Ltd., 1250 South Collegeville Road, Collegeville, PA 19426, USA
| | - Steven M Foord
- Computational Biology Division, Molecular Discovery Research, GlaxoSmithKline R&D Ltd., 1250 South Collegeville Road, Collegeville, PA 19426, USA
| | - Philippe Sanseau
- Computational Biology Division, Molecular Discovery Research, GlaxoSmithKline R&D Ltd., 1250 South Collegeville Road, Collegeville, PA 19426, USA
| | - Ziheng Yang
- Department of Biology, University College London, Darwin Bldg, Gower Street, London WC1E 6BT, UK
| | - Joanna D Holbrook
- Computational Biology Division, Molecular Discovery Research, GlaxoSmithKline R&D Ltd., 1250 South Collegeville Road, Collegeville, PA 19426, USA
| |
Collapse
|
47
|
Girgis RR, Javitch JA, Lieberman JA. Antipsychotic drug mechanisms: links between therapeutic effects, metabolic side effects and the insulin signaling pathway. Mol Psychiatry 2008; 13:918-29. [PMID: 18414407 PMCID: PMC3618283 DOI: 10.1038/mp.2008.40] [Citation(s) in RCA: 83] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
The exact therapeutic mechanism of action of antipsychotic drugs remains unclear. Recent evidence has shown that second-generation antipsychotic drugs (SGAs) are differentially associated with metabolic side effects compared to first-generation antipsychotic drugs (FGAs). Their proclivity to cause metabolic disturbances correlates, to some degree, with their comparative efficacy. This is particularly the case for clozapine and olanzapine. In addition, the insulin signaling pathway is vital for normal brain development and function. Abnormalities of this pathway have been found in persons with schizophrenia and antipsychotic drugs may ameliorate some of these alterations. This prompted us to hypothesize that the therapeutic antipsychotic and adverse metabolic effects of antipsychotic drugs might be related to a common pharmacologic mechanism. This article reviews insulin metabolism in the brain and related abnormalities associated with schizophrenia with the goals of gaining insight into antipsychotic drug effects and possibly also into the pathophysiology of schizophrenia. Finally, we speculate about one potential mechanism of action (that is, functional selectivity) that would be consistent with the data reviewed herein and make suggestions for the future investigation that is required before a therapeutic agent based on these data can be realized.
Collapse
Affiliation(s)
- RR Girgis
- Department of Psychiatry, College of Physicians and Surgeons, Columbia University, New York, NY, USA,New York State Psychiatric Institute, New York, NY, USA
| | - JA Javitch
- Department of Psychiatry, College of Physicians and Surgeons, Columbia University, New York, NY, USA,New York State Psychiatric Institute, New York, NY, USA
| | - JA Lieberman
- Department of Psychiatry, College of Physicians and Surgeons, Columbia University, New York, NY, USA,New York State Psychiatric Institute, New York, NY, USA
| |
Collapse
|
48
|
Lecompte O, Poch O, Laporte J. PtdIns5P regulation through evolution: roles in membrane trafficking? Trends Biochem Sci 2008; 33:453-60. [PMID: 18774718 DOI: 10.1016/j.tibs.2008.07.002] [Citation(s) in RCA: 64] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2008] [Revised: 07/01/2008] [Accepted: 07/02/2008] [Indexed: 01/27/2023]
Abstract
Phosphoinositides are lipid second messengers that are essential for many cellular processes, including signal transduction and cell compartmentalization. Among them, phosphatidylinositol 5-phosphate (PtdIns5P) is the least characterized, although several proteins involved in its regulation are implicated in human diseases. We studied the distribution of 32 PtdIns5P-metabolizing proteins in 39 eukaryotic genomes. Phylogenetic profiles identify four groups of co-evolving proteins, confirming known protein complexes and revealing new ones. The complexes comprise a phosphatase, a kinase and a regulator; this indicates that physical interactions between the three partners are necessary for the acute spatial regulation of PtdIns5P turnover. By examining PtdIns5P metabolism in this new perspective, we propose a role for PtdIns5P in membrane trafficking from late endosomal compartments to the plasma membrane.
Collapse
Affiliation(s)
- Odile Lecompte
- Department of Structural Biology and Genomics, rue Laurent Fries, Illkirch, F-67400 France
| | | | | |
Collapse
|
49
|
Serretti A, Mandelli L. The genetics of bipolar disorder: genome 'hot regions,' genes, new potential candidates and future directions. Mol Psychiatry 2008; 13:742-71. [PMID: 18332878 DOI: 10.1038/mp.2008.29] [Citation(s) in RCA: 131] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
Bipolar disorder (BP) is a complex disorder caused by a number of liability genes interacting with the environment. In recent years, a large number of linkage and association studies have been conducted producing an extremely large number of findings often not replicated or partially replicated. Further, results from linkage and association studies are not always easily comparable. Unfortunately, at present a comprehensive coverage of available evidence is still lacking. In the present paper, we summarized results obtained from both linkage and association studies in BP. Further, we indicated new potential interesting genes, located in genome 'hot regions' for BP and being expressed in the brain. We reviewed published studies on the subject till December 2007. We precisely localized regions where positive linkage has been found, by the NCBI Map viewer (http://www.ncbi.nlm.nih.gov/mapview/); further, we identified genes located in interesting areas and expressed in the brain, by the Entrez gene, Unigene databases (http://www.ncbi.nlm.nih.gov/entrez/) and Human Protein Reference Database (http://www.hprd.org); these genes could be of interest in future investigations. The review of association studies gave interesting results, as a number of genes seem to be definitively involved in BP, such as SLC6A4, TPH2, DRD4, SLC6A3, DAOA, DTNBP1, NRG1, DISC1 and BDNF. A number of promising genes, which received independent confirmations, and genes that have to be further investigated in BP, have been also systematically listed. In conclusion, the combination of linkage and association approaches provided a number of liability genes. Nevertheless, other approaches are required to disentangle conflicting findings, such as gene interaction analyses, interaction with psychosocial and environmental factors and, finally, endophenotype investigations.
Collapse
Affiliation(s)
- A Serretti
- Institute of Psychiatry, University of Bologna, Bologna, Italy.
| | | |
Collapse
|
50
|
Psychosis and autism as diametrical disorders of the social brain. Behav Brain Sci 2008; 31:241-61; discussion 261-320. [DOI: 10.1017/s0140525x08004214] [Citation(s) in RCA: 353] [Impact Index Per Article: 20.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
AbstractAutistic-spectrum conditions and psychotic-spectrum conditions (mainly schizophrenia, bipolar disorder, and major depression) represent two major suites of disorders of human cognition, affect, and behavior that involve altered development and function of the social brain. We describe evidence that a large set of phenotypic traits exhibit diametrically opposite phenotypes in autistic-spectrum versus psychotic-spectrum conditions, with a focus on schizophrenia. This suite of traits is inter-correlated, in that autism involves a general pattern of constrained overgrowth, whereas schizophrenia involves undergrowth. These disorders also exhibit diametric patterns for traits related to social brain development, including aspects of gaze, agency, social cognition, local versus global processing, language, and behavior. Social cognition is thus underdeveloped in autistic-spectrum conditions and hyper-developed on the psychotic spectrum.;>We propose and evaluate a novel hypothesis that may help to explain these diametric phenotypes: that the development of these two sets of conditions is mediated in part by alterations of genomic imprinting. Evidence regarding the genetic, physiological, neurological, and psychological underpinnings of psychotic-spectrum conditions supports the hypothesis that the etiologies of these conditions involve biases towards increased relative effects from imprinted genes with maternal expression, which engender a general pattern of undergrowth. By contrast, autistic-spectrum conditions appear to involve increased relative bias towards effects of paternally expressed genes, which mediate overgrowth. This hypothesis provides a simple yet comprehensive theory, grounded in evolutionary biology and genetics, for understanding the causes and phenotypes of autistic-spectrum and psychotic-spectrum conditions.
Collapse
|