Role of Gsk3ß in Behavioral Abnormalities Induced by Serotonin

Total Page:16

File Type:pdf, Size:1020Kb

Role of Gsk3ß in Behavioral Abnormalities Induced by Serotonin Role of GSK3␤ in behavioral abnormalities induced by serotonin deficiency Jean-Martin Beaulieu*†, Xiaodong Zhang*‡, Ramona M. Rodriguiz§, Tatyana D. Sotnikova*, Michael J. Cools§, William C. Wetsel*§¶, Raul R. Gainetdinov*, and Marc G. Caron*¶ʈ** Departments of *Cell Biology, ʈMedicine, ¶Neurobiology, and §Psychiatry and Behavioral Sciences, Mouse Behavioral and Neuroendocrine Analyses Core Facility, Duke University Medical Center, Durham, NC 27710 Communicated by L. L. Iversen, University of Oxford, Oxford, United Kingdom, December 6, 2007 (received for review November 6, 2007) Dysregulation of brain serotonin (5-HT) neurotransmission is pharmacological stimulation of 5-HT1 receptors enhances phos- thought to underlie mental conditions as diverse as depression, phorylation of the Ser-9 residue, thus leading to inhibition of the anxiety disorders, bipolar disorder, autism, and schizophrenia. kinase (7). In contrast, 5-HT2 receptor signaling reduces GSK3␤ Despite treatment of these conditions with serotonergic drugs, the phosphorylation thus leading to kinase activation (7). Hence, a molecular mechanisms by which 5-HT is involved in the regulation general 5-HT deficiency may exert differential effects on GSK3␤ of aberrant emotional behaviors are poorly understood. Here, we activity in brain regions expressing diverse sets of 5-HT generated knockin mice expressing a mutant form of the brain receptors. 5-HT synthesis enzyme, tryptophan hydroxylase 2 (Tph2). This Several lines of evidence indicate that the rate-limiting en- mutant is equivalent to a rare human variant (R441H) identified in zyme for 5-HT synthesis in adult brain is tryptophan hydroxylase few individuals with unipolar major depression. Expression of 2 (Tph2) (15, 16). This enzyme catalyzes the conversion of the mutant Tph2 in mice results in markedly reduced (Ϸ80%) brain amino acid tryptophan to 5-hydroxytryptophan (5-HTP) that is 5-HT production and leads to behavioral abnormalities in tests subsequently decarboxylated to 5-HT by L-aromatic amino acid assessing 5-HT-mediated emotional states. This reduction in brain decarboxylase. Recent investigations of naturally occurring ge- 5-HT levels is accompanied by activation of glycogen synthase netic polymorphisms in humans and mice have identified func- kinase 3␤ (GSK3␤), a signaling molecule modulated by many tional mutations in Tph2 that lead to pronounced reduction in psychiatric therapeutic agents. Importantly, inactivation of GSK3␤ enzyme activity (15, 17–19). A rare R441H TPH2 variant has NEUROSCIENCE in Tph2 knockin mice, using pharmacological or genetic ap- been identified in individuals from a small cohort of elderly proaches, alleviates the aberrant behaviors produced by 5-HT patients with major unipolar depression. This mutation causes a deficiency. These findings establish a critical role of Tph2 in the reduction of Ϸ80% of enzyme activity when expressed in maintenance of brain serotonin homeostasis and identify GSK3␤ transfected PC12 cells (18). To investigate more directly the signaling as an important pathway through which brain 5-HT effects of this mutation on in vivo 5-HT synthesis, cellular deficiency induces abnormal behaviors. Targeting GSK3␤ and re- signaling, and behavior, we generated mice where the R441H lated signaling events may afford therapeutic advantages for the TPH2 mutation was engineered at the equivalent R439H amino management of certain 5-HT-related psychiatric conditions. acid residue of the mouse Tph2. Here we show that this mutation recapitulates in the mouse the neurochemical changes predicted GSK-3 ͉ mood disorders ͉ serotonin ͉ Tph2 ͉ functional polymorphism from cellular studies of the human mutant Tph2 and reveals an important role of the GSK3 signaling pathway in the behavioral erotonin (5-HT) is involved in multiple aspects of normal abnormalities associated with reduced brain 5-HT levels. brain functions ranging from the regulation of mood to the S Results control of appetite and social interactions (1–3). Several studies have suggested a contribution of abnormal 5-HT transmission in Generation of R439H Tph2 Knockin Mice. R439H Tph2 knockin mice various human psychiatric conditions and drugs acting on 5-HT were generated by using a homologous recombination strategy neurotransmission are commonly used for the management of that resulted in the insertion of the mutation in exon 11 and of major depression, anxiety disorder, obsessive-compulsive disor- a residual loxP site in the ninth intron of the gene (Fig. 1A). der, autism, and schizophrenia (1–3). Although drugs that Homozygous (HO) and heterozygous (HET) mutant mice were influence the 5-HT system can affect histone acetylation, mod- first identified using a PCR genotyping protocol detecting the ulate production of brain-derived neurotrophic factor, increase presence of the LoxP insertion in intron 9 (Fig. 1B). Subse- neural progenitor cell proliferation, and inhibit glycogen syn- quently, the presence of the mutated tph2 alleles in both HO and thase kinase 3␤ (GSK3␤) in certain brain areas (4, 5), the HET knockin mice was confirmed (Fig. 1C) by ARMS-PCR (15, mechanisms underlying the regulation of behavior by 5-HT are 20). Mice carrying the R439H mutation were viable, developed still obscure. without overt phenotype, and reproduced normally. RT-PCR There are indications that pharmacologic manipulations of confirmed the integrity of the mutated R439H Tph2 transcript 5-HT levels by different classes of drugs can affect distinct neuronal signaling mechanisms (6–9). For instance, multiple Author contributions: J.-M.B. and X.Z. contributed equally to this work; J.-M.B., X.Z., R.M.R., classes of 5-HT drugs, including selective 5-HT reuptake inhib- W.C.W., R.R.G., and M.G.C. designed research; J.-M.B., X.Z., R.M.R., T.D.S., M.J.C., and R.R.G. itors (SSRIs), tricyclic antidepressants, monoamine oxidase in- performed research; J.-M.B., X.Z., R.M.R., T.D.S., and R.R.G. analyzed data; and J.-M.B., X.Z., hibitors, and atypical antipsychotics (5, 7, 9), inhibit brain W.C.W., R.R.G., and M.G.C. wrote the paper. GSK3␤ signaling. GSK3␤ is also inhibited in vivo by lithium (6, The authors declare no conflict of interest. 10–12), which is often used in combination with antidepressants †Present address: CRULRG, Research Institute, Universite´Laval, Que´bec City, Canada. for the management of certain mood disorders (13). However, ‡Present address: Duke–National University of Singapore Graduate Medical School, whether these changes in GSK3␤ are incidental or contribute to Singapore. the regulation of 5-HT-related abnormal behaviors is unex- **To whom correspondence should be addressed. E-mail: [email protected]. plored (5). GSK3␤ is a constitutively active kinase that is This article contains supporting information online at www.pnas.org/cgi/content/full/ inhibited following the phosphorylation of the Ser-9 residue 0711496105/DC1. located in its amino-terminal domain (14). In mouse brain, © 2008 by The National Academy of Sciences of the USA www.pnas.org͞cgi͞doi͞10.1073͞pnas.0711496105 PNAS ͉ January 29, 2008 ͉ vol. 105 ͉ no. 4 ͉ 1333–1338 Downloaded by guest on September 25, 2021 Fig. 2. Neurochemical measurements of 5-HT synthesis rates and contents in R439H Tph2 knockin mice. (A) 5-HTP synthesis rates in striatum [ANOVA, FGenotype(2,21) ϭ 5.412, P Ͻ 0.012], frontal cortex [ANOVA, FGenotype(2,21) ϭ 11.791, P Ͻ 0.003], and hippocampus [ANOVA, FGenotype(2,21) ϭ 17.426, P Ͻ 0.001] from WT, HET and HO Tph2 mice. (B) 5-HT tissue contents in striatum [ANOVA, FGenotype(2,21) ϭ 19.236, P Ͻ 0.001], frontal cortex [ANOVA, FGenotype(2,21) ϭ 17.675, P Ͻ 0.001], and hippocampus [ANOVA, FGenotype(2,21) ϭ 15.319, P Ͻ 0.001]. (C) 5-HIAA tissue contents in striatum [ANOVA, FGenotype(2,21) ϭ 9.678, P Ͻ 0.001], frontal cortex [ANOVA, FGenotype(2,21) ϭ 6.238, P Ͻ 0.007], and hippocampus [ANOVA, FGenotype(2,21) ϭ8.112, PϽ0.002] of WT, HET and HO Tph2 mice (nϭ4–11 mice per group). Data are means Ϯ SEM. Bonferroni corrected pair-wise compar- isons: *, P Յ 0.05; ***, P Յ 0.005 from the WT control. hydroxybenzylhydrazine, provides a direct assessment of Tph2 activity in vivo (15). Synthesis rates in the striatum, frontal cortex (FC), and hippocampus were reduced by Ϸ40% and Ϸ80% in Fig. 1. Generation of mice expressing the R439H allele of Tph2.(A) Diagram HET and HO R439H Tph2 knockin mice, respectively (Fig. 2A). of the Tph2 locus and the targeting vector. The floxed PGK-NEO/HSV-TK Due to decreased synthesis, tissue contents of 5-HT were also cassette, flanked by the PGK-DT cassette and targeting arms, is illustrated. The substantially reduced in these same brain regions of HO R439H G1449A coding mutation was engineered in exon 11 of the mouse Tph2 gene. mice (Fig. 2B). However, in HET mice, a significant reduction After homologous recombination, ES cell clones carrying the mutated exon 11 in 5-HT contents was only apparent in the FC, suggesting that were selected using ARMS-PCR and transfected with a CRE recombinase homeostatic regulation of 5-HT levels may differ among various expression vector to remove the PGK-NEO/HSV-TK cassette resulting in a Tph2 mutant allele carrying a mutated exon 11 and a residual LoxP site in intron 9. brain regions. Finally, no differences between WT and HET The WT and mutant (Mut) alleles are shown; the location of PCR primers is R439H mice were evident in tissue levels of the 5-HT degrada- indicated, red arrows: LoxP primers, green arrows: ARMS-PCR external prim- tion product 5-hydroxyindoleacetic acid (5-HIAA), whereas the ers, blue or purple arrows: ARMS-PCR allele-specific primers. (B and C) PCR metabolite was reduced to almost undetectable levels in all brain genotyping of R439H Tph2 knockin mice.
Recommended publications
  • Developmental Plasticity and Circuit Mechanisms of Dopamine-Modulated Aggression Darshini Mahadevia Submitted in Partial Fulfill
    Developmental plasticity and circuit mechanisms of dopamine-modulated aggression Darshini Mahadevia Submitted in partial fulfillment of the requirements for the degree of Doctor of Philosophy under the Executive Committee of the Graduate School of Arts and Sciences COLUMBIA UNIVERSITY 2018 © 2018 Darshini Mahadevia All rights reserved ABSTRACT Developmental plasticity and circuit mechanisms of dopamine-modulated aggression Darshini Mahadevia Aggression and violence pose a significant public health concern to society. Aggression is a highly conserved behavior that shares common biological correlates across species. While aggression developed as an evolutionary adaptation to competition, its untimely and uncontrolled expression is maladaptive and presents itself in a number of neuropsychiatric disorders. A mechanistic hypothesis for pathological aggression links aberrant behavior with heightened dopamine function. However, while dopamine hyper-activity is a neural correlate of aggression, the developmental aspects and circuit level contributions of dopaminergic signaling have not been elucidated. In this dissertation, I aim to address these questions regarding the specifics of dopamine function in a murine model of aggressive behavior. In chapter I, I provide a review of the literature that describes the current state of research on aggression. I describe the background elements that lay the foundation for experimental questions and original data presented in later chapters. I introduce, in detail, published studies that describe the clinical manifestation and epidemiological spread, the dominant categories, the anatomy and physiology, and the pharmacology of aggression, with a particular emphasis on the dopaminergic system. Finally, I describe instances of genetic and environmental risk factors impacting aggression, concluding with studies revealing an important role for interactions among genetics, environmental factors, and age in the development of aggression.
    [Show full text]
  • JAD 5478.Pdf
    JAD-05478; No of Pages 9 Journal of Affective Disorders xxx (2012) xxx–xxx Contents lists available at SciVerse ScienceDirect Journal of Affective Disorders journal homepage: www.elsevier.com/locate/jad Research report Association of TPH1, TPH2, and 5HTTLPR with PTSD and depressive symptoms Armen K. Goenjian a,b,e,⁎, Julia N. Bailey c,d, David P. Walling b, Alan M. Steinberg a, Devon Schmidt b, Uma Dandekar d, Ernest P. Noble e a UCLA/Duke University National Center for Child Traumatic Stress, Department of Psychiatry and Biobehavioral Sciences, University of California, Los Angeles (UCLA), United States b Collaborative Neuroscience Network, Garden Grove, CA 92845, United States c Department of Epidemiology, UCLA School of Public Health, Los Angeles, CA, United States d Epilepsy Genetics/Genomics Laboratories, VA GLAHS, Los Angeles, CA, United States e Alcohol Research Center, Department of Psychiatry and Biobehavioral Sciences, University of California, Los Angeles, United States article info abstract Article history: Objective: To examine the potential contribution of the serotonin hydroxylase (TPH1 and Received 29 January 2012 TPH2) genes, and the serotonin transporter promoter polymorphism (5HTTLPR) to the unique Accepted 5 February 2012 and pleiotropic risk of PTSD symptoms and depressive symptoms. Available online xxxx Methods: Participants included 200 adults exposed to the 1988 Spitak earthquake from 12 multigenerational families (3 to 5 generations). Severity of trauma exposure, PTSD, and de- Keywords: pressive symptoms were assessed using standard psychometric instruments. Pedigree-based Genetics variance component analysis was used to assess the association between select genes and PTSD the phenotypes. Depression Results: After adjusting for age, sex, exposure and environmental variables, there was a signif- Tryptophan hydroxylase icant association of PTSD symptoms with the ‘t’ allele of TPH1 SNP rs2108977 (pb0.004), Serotonin transporter explaining 3% of the phenotypic variance.
    [Show full text]
  • A Computational Approach for Defining a Signature of Β-Cell Golgi Stress in Diabetes Mellitus
    Page 1 of 781 Diabetes A Computational Approach for Defining a Signature of β-Cell Golgi Stress in Diabetes Mellitus Robert N. Bone1,6,7, Olufunmilola Oyebamiji2, Sayali Talware2, Sharmila Selvaraj2, Preethi Krishnan3,6, Farooq Syed1,6,7, Huanmei Wu2, Carmella Evans-Molina 1,3,4,5,6,7,8* Departments of 1Pediatrics, 3Medicine, 4Anatomy, Cell Biology & Physiology, 5Biochemistry & Molecular Biology, the 6Center for Diabetes & Metabolic Diseases, and the 7Herman B. Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN 46202; 2Department of BioHealth Informatics, Indiana University-Purdue University Indianapolis, Indianapolis, IN, 46202; 8Roudebush VA Medical Center, Indianapolis, IN 46202. *Corresponding Author(s): Carmella Evans-Molina, MD, PhD ([email protected]) Indiana University School of Medicine, 635 Barnhill Drive, MS 2031A, Indianapolis, IN 46202, Telephone: (317) 274-4145, Fax (317) 274-4107 Running Title: Golgi Stress Response in Diabetes Word Count: 4358 Number of Figures: 6 Keywords: Golgi apparatus stress, Islets, β cell, Type 1 diabetes, Type 2 diabetes 1 Diabetes Publish Ahead of Print, published online August 20, 2020 Diabetes Page 2 of 781 ABSTRACT The Golgi apparatus (GA) is an important site of insulin processing and granule maturation, but whether GA organelle dysfunction and GA stress are present in the diabetic β-cell has not been tested. We utilized an informatics-based approach to develop a transcriptional signature of β-cell GA stress using existing RNA sequencing and microarray datasets generated using human islets from donors with diabetes and islets where type 1(T1D) and type 2 diabetes (T2D) had been modeled ex vivo. To narrow our results to GA-specific genes, we applied a filter set of 1,030 genes accepted as GA associated.
    [Show full text]
  • Effects of TPH2 Gene Variation and Childhood Trauma on the Clinical
    Movement disorders J Neurol Neurosurg Psychiatry: first published as 10.1136/jnnp-2019-322636 on 23 June 2020. Downloaded from ORIGINAL RESEARCH Effects of TPH2 gene variation and childhood trauma on the clinical and circuit- level phenotype of functional movement disorders Primavera A Spagnolo ,1 Gina Norato,2 Carine W Maurer,3 David Goldman,4 Colin Hodgkinson,4 Silvina Horovitz,1 Mark Hallett 1 ► Additional material is ABSTRact known about the contribution of genetic factors to published online only. To view, Background Functional movement disorders (FMDs), the pathophysiology of FMD. please visit the journal online (http:// dx. doi. org/ 10. 1136/ part of the wide spectrum of functional neurological Several studies have indicated that positive jnnp- 2019- 322636). disorders (conversion disorders), are common and often family history for FMD is associated with increased associated with a poor prognosis. Nevertheless, little morbidity risk among family members.2 3 However, 1 Human Motor Control is known about their neurobiological underpinnings, large- scale genetic epidemiology studies (eg, twin- Section, Medical Neurology particularly with regard to the contribution of genetic based, family- based, adoption- based and other Branch, National Institute on Nuerological Disorders and factors. Because FMD and stress-related disorders share population-based studies), which have provided a Stroke, National Institutes of a common core of biobehavioural manifestations, we necessary first step in establishing heritability and Health, Bethesda, MD, USA investigated whether variants in stress-related genes exploring genetic interactions in several neuropsy- 2 Office of Biostatistics, National also contributed, directly and interactively with childhood chiatric disorders, have not yet been carried out in Institute on Neurological Disorders and Stroke, National trauma, to the clinical and circuit-level phenotypes of patients with FMD.
    [Show full text]
  • Retrograde Inhibition by a Specific Subset of Interpeduncular Α5 Nicotinic Neurons Regulates Nicotine Preference
    Retrograde inhibition by a specific subset of interpeduncular α5 nicotinic neurons regulates nicotine preference Jessica L. Ablesa,b,c, Andreas Görlicha,1, Beatriz Antolin-Fontesa,2,CuidongWanga, Sylvia M. Lipforda, Michael H. Riada, Jing Rend,e,3,FeiHud,e,4,MinminLuod,e,PaulJ.Kennyc, Nathaniel Heintza,f,5, and Ines Ibañez-Tallona,5 aLaboratory of Molecular Biology, The Rockefeller University, New York, NY 10065; bDepartment of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY 10029; cDepartment of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY 10029; dNational Institute of Biological Sciences, Beijing 102206, China; eSchool of Life Sciences, Tsinghua University, Beijing 100084, China; and fHoward Hughes Medical Institute, The Rockefeller University, New York, NY 10065 Contributed by Nathaniel Heintz, October 23, 2017 (sent for review October 5, 2017; reviewed by Jean-Pierre Changeux and Lorna W. Role) Repeated exposure to drugs of abuse can produce adaptive changes nicotine withdrawal, and optical activation of IPN GABAergic cells that lead to the establishment of dependence. It has been shown that is sufficient to produce a withdrawal syndrome, while blockade of allelic variation in the α5 nicotinic acetylcholine receptor (nAChR) gene GABAergic cells in the IPN reduced symptoms of withdrawal (17). CHRNA5 is associated with higher risk of tobacco dependence. In the Taken together these studies highlight the critical role of α5in brain, α5-containing nAChRs are expressed at very high levels in the regulating behavioral responses to nicotine. Here we characterize two subpopulations of GABAergic interpeduncular nucleus (IPN). Here we identified two nonoverlapping Amigo1 Epyc α + α Amigo1 α Epyc neurons in the IPN that express α5: α5- and α5- neu- 5 cell populations ( 5- and 5- ) in mouse IPN that respond α Amigo1 α Epyc differentially to nicotine.
    [Show full text]
  • Understanding the Molecular Mechanisms of Aggression in BALB/C and TPH2-Deficient Mice
    Understanding the molecular mechanisms of aggression in BALB/c and TPH2-deficient mice Citation for published version (APA): Gorlova, A. (2020). Understanding the molecular mechanisms of aggression in BALB/c and TPH2- deficient mice. OneBook.ru. https://doi.org/10.26481/dis.20200305ag Document status and date: Published: 01/01/2020 DOI: 10.26481/dis.20200305ag Document Version: Publisher's PDF, also known as Version of record Please check the document version of this publication: • A submitted manuscript is the version of the article upon submission and before peer-review. There can be important differences between the submitted version and the official published version of record. People interested in the research are advised to contact the author for the final version of the publication, or visit the DOI to the publisher's website. • The final author version and the galley proof are versions of the publication after peer review. • The final published version features the final layout of the paper including the volume, issue and page numbers. Link to publication General rights Copyright and moral rights for the publications made accessible in the public portal are retained by the authors and/or other copyright owners and it is a condition of accessing publications that users recognise and abide by the legal requirements associated with these rights. • Users may download and print one copy of any publication from the public portal for the purpose of private study or research. • You may not further distribute the material or use it for any profit-making activity or commercial gain • You may freely distribute the URL identifying the publication in the public portal.
    [Show full text]
  • Electronic Supplementary Material (ESI) for Metallomics
    Electronic Supplementary Material (ESI) for Metallomics. This journal is © The Royal Society of Chemistry 2018 Uniprot Entry name Gene names Protein names Predicted Pattern Number of Iron role EC number Subcellular Membrane Involvement in disease Gene ontology (biological process) Id iron ions location associated 1 P46952 3HAO_HUMAN HAAO 3-hydroxyanthranilate 3,4- H47-E53-H91 1 Fe cation Catalytic 1.13.11.6 Cytoplasm No NAD biosynthetic process [GO:0009435]; neuron cellular homeostasis dioxygenase (EC 1.13.11.6) (3- [GO:0070050]; quinolinate biosynthetic process [GO:0019805]; response to hydroxyanthranilate oxygenase) cadmium ion [GO:0046686]; response to zinc ion [GO:0010043]; tryptophan (3-HAO) (3-hydroxyanthranilic catabolic process [GO:0006569] acid dioxygenase) (HAD) 2 O00767 ACOD_HUMAN SCD Acyl-CoA desaturase (EC H120-H125-H157-H161; 2 Fe cations Catalytic 1.14.19.1 Endoplasmic Yes long-chain fatty-acyl-CoA biosynthetic process [GO:0035338]; unsaturated fatty 1.14.19.1) (Delta(9)-desaturase) H160-H269-H298-H302 reticulum acid biosynthetic process [GO:0006636] (Delta-9 desaturase) (Fatty acid desaturase) (Stearoyl-CoA desaturase) (hSCD1) 3 Q6ZNF0 ACP7_HUMAN ACP7 PAPL PAPL1 Acid phosphatase type 7 (EC D141-D170-Y173-H335 1 Fe cation Catalytic 3.1.3.2 Extracellular No 3.1.3.2) (Purple acid space phosphatase long form) 4 Q96SZ5 AEDO_HUMAN ADO C10orf22 2-aminoethanethiol dioxygenase H112-H114-H193 1 Fe cation Catalytic 1.13.11.19 Unknown No oxidation-reduction process [GO:0055114]; sulfur amino acid catabolic process (EC 1.13.11.19) (Cysteamine
    [Show full text]
  • The Genetics of Bipolar Disorder
    Molecular Psychiatry (2008) 13, 742–771 & 2008 Nature Publishing Group All rights reserved 1359-4184/08 $30.00 www.nature.com/mp FEATURE REVIEW The genetics of bipolar disorder: genome ‘hot regions,’ genes, new potential candidates and future directions A Serretti and L Mandelli Institute of Psychiatry, University of Bologna, Bologna, Italy 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.
    [Show full text]
  • Inducible Gene Manipulations in Serotonergic Neurons
    ORIGINAL RESEARCH ARTICLE published: 06 November 2009 MOLECULAR NEUROSCIENCE doi: 10.3389/neuro.02.024.2009 Inducible gene manipulations in serotonergic neurons Tillmann Weber1,2, Gerald Böhm1, Elke Hermann1, Günther Schütz 3, Kai Schönig1 and Dusan Bartsch1* 1 Department of Molecular Biology, Central Institute of Mental Health, Heidelberg University, Mannheim, Germany 2 Department of Addictive Behavior and Addiction Medicine, Central Institute of Mental Health, Heidelberg University, Mannheim, Germany 3 Division Molecular Biology of the Cell I, German Cancer Research Center, Heidelberg, Germany Edited by: An impairment of the serotonergic (5-HT) system has been implicated in the etiology of many William Wisden, Imperial College, UK neuropsychiatric disorders. Despite the considerable genetic evidence, the exact molecular Reviewed by: and pathophysiological mechanisms underlying this dysfunction remain largely unknown. To Gregg E. Homanics, University of Pittsburgh, USA; address the lack of instruments for the molecular dissection of gene function in serotonergic Bernhard Lüscher, Pennsylvania State neurons we have developed a new mouse transgenic tool that allows inducible Cre-mediated University, USA recombination of genes selectively in 5-HT neurons of all raphe nuclei. In this transgenic mouse *Correspondence: line, the tamoxifen-inducible CreERT2 recombinase is expressed under the regulatory control Dusan Bartsch, Department of of the mouse tryptophan hydroxylase 2 (Tph2) gene locus (177 kb). Tamoxifen treatment Molecular Biology, Central Institute of Mental Health, Heidelberg University, effi ciently induced recombination selectively in serotonergic neurons with minimal background J5, 68159 Mannheim, Germany. activity in vehicle-treated mice. These genetic manipulations can be initiated at any desired time e-mail: [email protected] during embryonic development, neonatal stage or adulthood.
    [Show full text]
  • Isoform-Specific Substrate Inhibition Mechanism of Human Tryptophan Hydroxylase
    View metadata,Downloaded citation and from similar orbit.dtu.dk papers on:at core.ac.uk Apr 01, 2019 brought to you by CORE provided by Online Research Database In Technology Isoform-Specific Substrate Inhibition Mechanism of Human Tryptophan Hydroxylase Tidemand, Kasper Damgaard; Peters, Günther H.J.; Harris, Pernille; Stensgaard, Eva; Christensen, Hans Erik Mølager Published in: Biochemistry Link to article, DOI: 10.1021/acs.biochem.7b00763 Publication date: 2017 Document Version Peer reviewed version Link back to DTU Orbit Citation (APA): Tidemand, K. D., Peters, G. H. J., Harris, P., Stensgaard, E., & Christensen, H. E. M. (2017). Isoform-Specific Substrate Inhibition Mechanism of Human Tryptophan Hydroxylase. Biochemistry, 56(46), 6155–6164. DOI: 10.1021/acs.biochem.7b00763 General rights Copyright and moral rights for the publications made accessible in the public portal are retained by the authors and/or other copyright owners and it is a condition of accessing publications that users recognise and abide by the legal requirements associated with these rights. Users may download and print one copy of any publication from the public portal for the purpose of private study or research. You may not further distribute the material or use it for any profit-making activity or commercial gain You may freely distribute the URL identifying the publication in the public portal If you believe that this document breaches copyright please contact us providing details, and we will remove access to the work immediately and investigate your claim. Isoform-Specific Substrate Inhibition Mechanism of Human Tryptophan Hydroxylase Kasper D. Tidemand, Günther H. Peters*, Pernille Harris, Eva Stensgaard, and Hans E.
    [Show full text]
  • Genetic Association Studies of Bipolar Disorder
    Genetic Association Studies of Bipolar Disorder Dr Nicholas James Bass This thesis is submitted for the degree of Doctor of Medicine (MD) at University College London 2007 Molecular Psychiatry Laboratory Department of Mental Health Sciences Royal Free and University College London Medical School The Windeyer Institute of Medical Sciences 46 Cleveland Street London W 1T4JF 1 UMI Number: U591560 All rights reserved INFORMATION TO ALL USERS The quality of this reproduction is dependent upon the quality of the copy submitted. In the unlikely event that the author did not send a complete manuscript and there are missing pages, these will be noted. Also, if material had to be removed, a note will indicate the deletion. Dissertation Publishing UMI U591560 Published by ProQuest LLC 2013. Copyright in the Dissertation held by the Author. Microform Edition © ProQuest LLC. All rights reserved. This work is protected against unauthorized copying under Title 17, United States Code. ProQuest LLC 789 East Eisenhower Parkway P.O. Box 1346 Ann Arbor, Ml 48106-1346 Statement of Conjoint Work My work was very much part of a team effort. My specific contributions were: 1. UCL bipolar sample collection - recruitment, diagnostic interviewing, blood sampling and data management of 300 bipolar volunteers and over 100 control volunteers. 2. DNA preparation - extraction, quantification and plating out for PCR-based analysis. 3. Marker selection - from literature and using bioinformatic tools. 4. Microsatellite genotyping at BDNF, P2RX7 and COMT loci. 5. Microsatellite genotyping at DISCI locus in conjunction with Cindy Yang. 6. SNP genotyping at BDNF and COMT loci using Taqman, Amplifluor and RFLP assays.
    [Show full text]
  • Strand Breaks for P53 Exon 6 and 8 Among Different Time Course of Folate Depletion Or Repletion in the Rectosigmoid Mucosa
    SUPPLEMENTAL FIGURE COLON p53 EXONIC STRAND BREAKS DURING FOLATE DEPLETION-REPLETION INTERVENTION Supplemental Figure Legend Strand breaks for p53 exon 6 and 8 among different time course of folate depletion or repletion in the rectosigmoid mucosa. The input of DNA was controlled by GAPDH. The data is shown as ΔCt after normalized to GAPDH. The higher ΔCt the more strand breaks. The P value is shown in the figure. SUPPLEMENT S1 Genes that were significantly UPREGULATED after folate intervention (by unadjusted paired t-test), list is sorted by P value Gene Symbol Nucleotide P VALUE Description OLFM4 NM_006418 0.0000 Homo sapiens differentially expressed in hematopoietic lineages (GW112) mRNA. FMR1NB NM_152578 0.0000 Homo sapiens hypothetical protein FLJ25736 (FLJ25736) mRNA. IFI6 NM_002038 0.0001 Homo sapiens interferon alpha-inducible protein (clone IFI-6-16) (G1P3) transcript variant 1 mRNA. Homo sapiens UDP-N-acetyl-alpha-D-galactosamine:polypeptide N-acetylgalactosaminyltransferase 15 GALNTL5 NM_145292 0.0001 (GALNT15) mRNA. STIM2 NM_020860 0.0001 Homo sapiens stromal interaction molecule 2 (STIM2) mRNA. ZNF645 NM_152577 0.0002 Homo sapiens hypothetical protein FLJ25735 (FLJ25735) mRNA. ATP12A NM_001676 0.0002 Homo sapiens ATPase H+/K+ transporting nongastric alpha polypeptide (ATP12A) mRNA. U1SNRNPBP NM_007020 0.0003 Homo sapiens U1-snRNP binding protein homolog (U1SNRNPBP) transcript variant 1 mRNA. RNF125 NM_017831 0.0004 Homo sapiens ring finger protein 125 (RNF125) mRNA. FMNL1 NM_005892 0.0004 Homo sapiens formin-like (FMNL) mRNA. ISG15 NM_005101 0.0005 Homo sapiens interferon alpha-inducible protein (clone IFI-15K) (G1P2) mRNA. SLC6A14 NM_007231 0.0005 Homo sapiens solute carrier family 6 (neurotransmitter transporter) member 14 (SLC6A14) mRNA.
    [Show full text]