1
|
Koyama E, Kant T, Takata A, Kennedy JL, Zai CC. Genetics of child aggression, a systematic review. Transl Psychiatry 2024; 14:252. [PMID: 38862490 PMCID: PMC11167064 DOI: 10.1038/s41398-024-02870-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/17/2023] [Revised: 03/07/2024] [Accepted: 03/11/2024] [Indexed: 06/13/2024] Open
Abstract
Excessive and persistent aggressiveness is the most common behavioral problem that leads to psychiatric referrals among children. While half of the variance in childhood aggression is attributed to genetic factors, the biological mechanism and the interplay between genes and environment that results in aggression remains elusive. The purpose of this systematic review is to provide an overview of studies examining the genetics of childhood aggression irrespective of psychiatric diagnosis. PubMed, PsycINFO, and MEDLINE databases were searched using predefined search terms for aggression, genes and the specific age group. From the 652 initially yielded studies, eighty-seven studies were systematically extracted for full-text review and for further quality assessment analyses. Findings show that (i) investigation of candidate genes, especially of MAOA (17 studies), DRD4 (13 studies), and COMT (12 studies) continue to dominate the field, although studies using other research designs and methods including genome-wide association and epigenetic studies are increasing, (ii) the published articles tend to be moderate in sizes, with variable methods of assessing aggressive behavior and inconsistent categorizations of tandem repeat variants, resulting in inconclusive findings of genetic main effects, gene-gene, and gene-environment interactions, (iii) the majority of studies are conducted on European, male-only or male-female mixed, participants. To our knowledge, this is the first study to systematically review the effects of genes on youth aggression. To understand the genetic underpinnings of childhood aggression, more research is required with larger, more diverse sample sets, consistent and reliable assessments and standardized definition of the aggression phenotypes. The search for the biological mechanisms underlying child aggression will also benefit from more varied research methods, including epigenetic studies, transcriptomic studies, gene system and genome-wide studies, longitudinal studies that track changes in risk/ameliorating factors and aggression-related outcomes, and studies examining causal mechanisms.
Collapse
Affiliation(s)
- Emiko Koyama
- Tanenbaum Centre for Pharmacogenetics, Molecular Brain Science, Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, ON, Canada
- Laboratory for Molecular Pathology of Psychiatric Disorders, RIKEN Center for Brain Science, Wako, Japan
| | - Tuana Kant
- Tanenbaum Centre for Pharmacogenetics, Molecular Brain Science, Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, ON, Canada
| | - Atsushi Takata
- Laboratory for Molecular Pathology of Psychiatric Disorders, RIKEN Center for Brain Science, Wako, Japan
| | - James L Kennedy
- Tanenbaum Centre for Pharmacogenetics, Molecular Brain Science, Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, ON, Canada
- Department of Psychiatry, University of Toronto, Toronto, ON, Canada
- Institute of Medical Science, University of Toronto, Toronto, ON, Canada
| | - Clement C Zai
- Tanenbaum Centre for Pharmacogenetics, Molecular Brain Science, Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, ON, Canada.
- Department of Psychiatry, University of Toronto, Toronto, ON, Canada.
- Institute of Medical Science, University of Toronto, Toronto, ON, Canada.
- Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada.
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA.
| |
Collapse
|
2
|
Potenzieri A, Uccella S, Preiti D, Pisoni M, Rosati S, Lavarello C, Bartolucci M, Debellis D, Catalano F, Petretto A, Nobili L, Fellin T, Tucci V, Ramenghi LA, Savardi A, Cancedda L. Early IGF-1 receptor inhibition in mice mimics preterm human brain disorders and reveals a therapeutic target. SCIENCE ADVANCES 2024; 10:eadk8123. [PMID: 38427732 PMCID: PMC10906931 DOI: 10.1126/sciadv.adk8123] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/13/2023] [Accepted: 01/29/2024] [Indexed: 03/03/2024]
Abstract
Besides recent advances in neonatal care, preterm newborns still develop sex-biased behavioral alterations. Preterms fail to receive placental insulin-like growth factor-1 (IGF-1), a major fetal growth hormone in utero, and low IGF-1 serum levels correlate with preterm poor neurodevelopmental outcomes. Here, we mimicked IGF-1 deficiency of preterm newborns in mice by perinatal administration of an IGF-1 receptor antagonist. This resulted in sex-biased brain microstructural, functional, and behavioral alterations, resembling those of ex-preterm children, which we characterized performing parallel mouse/human behavioral tests. Pharmacological enhancement of GABAergic tonic inhibition by the U.S. Food and Drug Administration-approved drug ganaxolone rescued functional/behavioral alterations in mice. Establishing an unprecedented mouse model of prematurity, our work dissects the mechanisms at the core of abnormal behaviors and identifies a readily translatable therapeutic strategy for preterm brain disorders.
Collapse
Affiliation(s)
- Alberto Potenzieri
- Brain Development and Disease Laboratory, Istituto Italiano di Tecnologia, via Morego, 30, 16163 Genoa, Italy
- Università degli Studi di Genova, via Balbi, 5, 16126 Genoa, Italy
| | - Sara Uccella
- Department of Neurosciences, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health (DINOGMI), University of Genoa, 16132 Genoa, Italy
- Child Neuropsychiatry Unit, IRCCS Istituto Giannina Gaslini, Genoa, Italy
- Patologia Neonatale, IRCCS Istituto Giannina Gaslini, Genoa, Italy
| | - Deborah Preiti
- Child Neuropsychiatry Unit, IRCCS Istituto Giannina Gaslini, Genoa, Italy
- Patologia Neonatale, IRCCS Istituto Giannina Gaslini, Genoa, Italy
| | - Matteo Pisoni
- Optical Approaches to Brain Function Laboratory, Istituto Italiano di Tecnologia, via Morego, 30, 16163 Genoa, Italy
| | - Silvia Rosati
- Brain Development and Disease Laboratory, Istituto Italiano di Tecnologia, via Morego, 30, 16163 Genoa, Italy
| | - Chiara Lavarello
- Core Facilities - Clinical Proteomics and Metabolomics, IRCCS Istituto Giannina Gaslini, via Gerolamo Gaslini 5, 16147 Genoa, Italy
| | - Martina Bartolucci
- Core Facilities - Clinical Proteomics and Metabolomics, IRCCS Istituto Giannina Gaslini, via Gerolamo Gaslini 5, 16147 Genoa, Italy
| | - Doriana Debellis
- Electron Microscopy Facility, Istituto Italiano di Tecnologia, via Morego, 30, 16163 Genoa, Italy
| | - Federico Catalano
- Electron Microscopy Facility, Istituto Italiano di Tecnologia, via Morego, 30, 16163 Genoa, Italy
| | - Andrea Petretto
- Core Facilities - Clinical Proteomics and Metabolomics, IRCCS Istituto Giannina Gaslini, via Gerolamo Gaslini 5, 16147 Genoa, Italy
| | - Lino Nobili
- Department of Neurosciences, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health (DINOGMI), University of Genoa, 16132 Genoa, Italy
- Child Neuropsychiatry Unit, IRCCS Istituto Giannina Gaslini, Genoa, Italy
| | - Tommaso Fellin
- Optical Approaches to Brain Function Laboratory, Istituto Italiano di Tecnologia, via Morego, 30, 16163 Genoa, Italy
| | - Valter Tucci
- Genetics and Epigenetics of Behavior (GEB) Laboratory, Istituto Italiano di Tecnologia, via Morego, 30, 16163 Genoa, Italy
| | - Luca A. Ramenghi
- Department of Neurosciences, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health (DINOGMI), University of Genoa, 16132 Genoa, Italy
- Patologia Neonatale, IRCCS Istituto Giannina Gaslini, Genoa, Italy
| | - Annalisa Savardi
- Brain Development and Disease Laboratory, Istituto Italiano di Tecnologia, via Morego, 30, 16163 Genoa, Italy
| | - Laura Cancedda
- Brain Development and Disease Laboratory, Istituto Italiano di Tecnologia, via Morego, 30, 16163 Genoa, Italy
| |
Collapse
|
3
|
Utami KH, Yusof NABM, Garcia-Miralles M, Skotte NH, Nama S, Sampath P, Langley SR, Pouladi MA. Dysregulated COMT Expression in Fragile X Syndrome. Neuromolecular Med 2023; 25:644-649. [PMID: 37684514 DOI: 10.1007/s12017-023-08754-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2023] [Accepted: 08/20/2023] [Indexed: 09/10/2023]
Abstract
Transcriptional and proteomics analyses in human fragile X syndrome (FXS) neurons identified markedly reduced expression of COMT, a key enzyme involved in the metabolism of catecholamines, including dopamine, epinephrine and norepinephrine. FXS is the most common genetic cause of intellectual disability and autism spectrum disorders. COMT encodes for catechol-o-methyltransferase and its association with neuropsychiatric disorders and cognitive function has been extensively studied. We observed a significantly reduced level of COMT in in FXS human neural progenitors and neurons, as well as hippocampal neurons from Fmr1 null mice. We show that deficits in COMT were associated with an altered response in an assay of dopaminergic activity in Fmr1 null mice. These findings demonstrate that loss of FMRP downregulates COMT expression and affects dopamine signaling in FXS, and supports the notion that targeting catecholamine metabolism may be useful in regulating certain neuropsychiatric aspects of FXS.
Collapse
Affiliation(s)
- Kagistia Hana Utami
- Translational Laboratory in Genetic Medicine, Agency for Science, Technology and Research, Singapore (A*STAR), 8A Biomedical Grove, Immunos, Level 5, Singapore, 138648, Singapore
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Singapore
| | - Nur Amirah Binte Muhammed Yusof
- Translational Laboratory in Genetic Medicine, Agency for Science, Technology and Research, Singapore (A*STAR), 8A Biomedical Grove, Immunos, Level 5, Singapore, 138648, Singapore
| | - Marta Garcia-Miralles
- Translational Laboratory in Genetic Medicine, Agency for Science, Technology and Research, Singapore (A*STAR), 8A Biomedical Grove, Immunos, Level 5, Singapore, 138648, Singapore
- Department of Molecular Embryology, Medical Faculty, Institute of Anatomy and Cell Biology, Albert-Ludwigs-University Freiburg, 79104, Freiburg, Germany
| | - Niels Henning Skotte
- Proteomics Program, Novo Nordisk Foundation Center for Protein Research, Faculty of Health Sciences, University of Copenhagen, 2200, Copenhagen, Denmark
- Department of Drug Design and Pharmacology, University of Copenhagen, Copenhagen, Denmark
| | - Srikanth Nama
- Institute of Medical Biology, Agency for Science, Technology and Research, Singapore (A*STAR), 8A Biomedical Grove, Immunos, Level 5, Singapore, 138648, Singapore
| | - Prabha Sampath
- Agency for Science, Technology and Research, Genome Institute of Singapore, 60 Biopolis Street, Genome, Singapore, 138672, Singapore
- Program in Cancer and Stem Cell Biology, Duke-NUS Medical School, 8 College Road, Singapore, 169857, Singapore
| | - Sarah R Langley
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Singapore
- School of Biosciences, Cardiff University, Cardiff, CF10 3AX, UK
| | - Mahmoud A Pouladi
- Translational Laboratory in Genetic Medicine, Agency for Science, Technology and Research, Singapore (A*STAR), 8A Biomedical Grove, Immunos, Level 5, Singapore, 138648, Singapore.
- Department of Medical Genetics, Centre for Molecular Medicine & Therapeutics, Djavad Mowafaghian Centre for Brain Health, British Columbia Children's Hospital Research Institute, University of British Columbia, 950 West 28th Avenue, Vancouver, BC, V5Z 4H4, Canada.
| |
Collapse
|
4
|
Managò F, Scheggia D, Pontillo M, Mereu M, Mastrogiacomo R, Udayan G, Valentini P, Tata MC, Weinberger DR, Weickert CS, Pompa PP, De Luca MA, Vicari S, Papaleo F. Dopaminergic signalling and behavioural alterations by Comt-Dtnbp1 genetic interaction and their clinical relevance. Br J Pharmacol 2023; 180:2514-2531. [PMID: 37218669 DOI: 10.1111/bph.16147] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2022] [Revised: 04/26/2023] [Accepted: 05/02/2023] [Indexed: 05/24/2023] Open
Abstract
BACKGROUND AND PURPOSE Cognitive and motor functions are modulated by dopaminergic signalling, which is shaped by several genetic factors. The biological effects of single genetic variants might differ depending on epistatic interactions that can be functionally multi-directional and non-linear. EXPERIMENTAL APPROACH We performed behavioural and neurochemical assessments in genetically modified mice and behavioural assessments and genetic screening in human patients with 22q11.2 deletion syndrome (22q11.2DS). KEY RESULTS Here, we confirm a genetic interaction between the Comt (catechol-O-methyltransferase, human orthologue: COMT) and Dtnbp1 (dystrobrevin binding protein 1, alias dysbindin, human orthologue: DTNBP1) genes that modulate cortical and striatal dopaminergic signalling in a manner not predictable by the effects of each single gene. In mice, Comt-by-Dtnbp1 concomitant reduction leads to a hypoactive mesocortical and a hyperactive mesostriatal dopamine pathway, associated with specific cognitive abnormalities. Like mice, in subjects with the 22q11.2DS (characterized by COMT hemideletion and dopamine alterations), COMT-by-DTNBP1 concomitant reduction was associated with analogous cognitive disturbances. We then developed an easy and inexpensive colourimetric kit for the genetic screening of common COMT and DTNBP1 functional genetic variants for clinical application. CONCLUSIONS AND IMPLICATIONS These findings illustrate an epistatic interaction of two dopamine-related genes and their functional effects, supporting the need to address genetic interaction mechanisms at the base of complex behavioural traits.
Collapse
Affiliation(s)
- Francesca Managò
- Genetics of Cognition Laboratory, Neuroscience Area, Istituto Italiano di Tecnologia, Genoa, Italy
| | - Diego Scheggia
- Genetics of Cognition Laboratory, Neuroscience Area, Istituto Italiano di Tecnologia, Genoa, Italy
- Department of Pharmacological and Biomolecular Sciences, University of Milan, Milan, Italy
| | - Maria Pontillo
- Department of Neuroscience, Bambino Gesù Children's Hospital, Rome, Italy
| | - Maddalena Mereu
- Genetics of Cognition Laboratory, Neuroscience Area, Istituto Italiano di Tecnologia, Genoa, Italy
| | - Rosa Mastrogiacomo
- Genetics of Cognition Laboratory, Neuroscience Area, Istituto Italiano di Tecnologia, Genoa, Italy
| | - Gayatri Udayan
- Nanobiointeractions and Nanodiagnostics, Istituto Italiano di Tecnologia, Genoa, Italy
- Department of Engineering for Innovation, University of Salento, Lecce, Italy
| | - Paola Valentini
- Nanobiointeractions and Nanodiagnostics, Istituto Italiano di Tecnologia, Genoa, Italy
| | | | - Daniel R Weinberger
- Lieber Institute for Brain Development, Johns Hopkins University Medical Campus, Baltimore, Maryland, USA
| | - Cynthia S Weickert
- Schizophrenia Research Laboratory, Neuroscience Research Australia, Sydney, Australia
| | - Pier Paolo Pompa
- Nanobiointeractions and Nanodiagnostics, Istituto Italiano di Tecnologia, Genoa, Italy
| | - Maria A De Luca
- Department of Biomedical Sciences, University of Cagliari, Cagliari, Italy
| | - Stefano Vicari
- Department of Neuroscience, Bambino Gesù Children's Hospital, Rome, Italy
| | - Francesco Papaleo
- Genetics of Cognition Laboratory, Neuroscience Area, Istituto Italiano di Tecnologia, Genoa, Italy
- Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| |
Collapse
|
5
|
Wyns A, Hendrix J, Lahousse A, De Bruyne E, Nijs J, Godderis L, Polli A. The Biology of Stress Intolerance in Patients with Chronic Pain—State of the Art and Future Directions. J Clin Med 2023; 12:jcm12062245. [PMID: 36983246 PMCID: PMC10057496 DOI: 10.3390/jcm12062245] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2023] [Revised: 03/07/2023] [Accepted: 03/10/2023] [Indexed: 03/16/2023] Open
Abstract
Stress has been consistently linked to negative impacts on physical and mental health. More specifically, patients with chronic pain experience stress intolerance, which is an exacerbation or occurrence of symptoms in response to any type of stress. The pathophysiological mechanisms underlying this phenomenon remain unsolved. In this state-of-the-art paper, we summarised the role of the autonomic nervous system (ANS) and hypothalamus-pituitary-adrenal (HPA) axis, the two major stress response systems in stress intolerance. We provided insights into such mechanisms based on evidence from clinical studies in both patients with chronic pain, showing dysregulated stress systems, and healthy controls supported by preclinical studies, highlighting the link between these systems and symptoms of stress intolerance. Furthermore, we explored the possible regulating role for (epi)genetic mechanisms influencing the ANS and HPA axis. The link between stress and chronic pain has become an important area of research as it has the potential to inform the development of interventions to improve the quality of life for individuals living with chronic pain. As stress has become a prevalent concern in modern society, understanding the connection between stress, HPA axis, ANS, and chronic health conditions such as chronic pain is crucial to improve public health and well-being.
Collapse
Affiliation(s)
- Arne Wyns
- Pain in Motion Research Group (PAIN), Department of Physiotherapy, Human Physiology and Anatomy, Faculty of Physical Education and Physiotherapy, Vrije Universiteit Brussel, 1090 Brussels, Belgium; (A.W.); (A.L.); (J.N.); (A.P.)
| | - Jolien Hendrix
- Pain in Motion Research Group (PAIN), Department of Physiotherapy, Human Physiology and Anatomy, Faculty of Physical Education and Physiotherapy, Vrije Universiteit Brussel, 1090 Brussels, Belgium; (A.W.); (A.L.); (J.N.); (A.P.)
- Department of Public Health and Primary Care, Centre for Environment & Health, KU Leuven, Kapucijnenvoer 35, 3000 Leuven, Belgium;
- Flanders Research Foundation-FWO, 1090 Brussels, Belgium
- Correspondence:
| | - Astrid Lahousse
- Pain in Motion Research Group (PAIN), Department of Physiotherapy, Human Physiology and Anatomy, Faculty of Physical Education and Physiotherapy, Vrije Universiteit Brussel, 1090 Brussels, Belgium; (A.W.); (A.L.); (J.N.); (A.P.)
- Flanders Research Foundation-FWO, 1090 Brussels, Belgium
- Chronic Pain Rehabilitation, Department of Physical Medicine and Physiotherapy, University Hospital, 1090 Brussels, Belgium
- Rehabilitation Research (RERE) Research Group, Department of Physiotherapy, Human Physiology and Anatomy, Faculty of Physical Education & Physiotherapy (KIMA), Vrije Universiteit Brussel, 1090 Brussels, Belgium
| | - Elke De Bruyne
- Department of Hematology and Immunology-Myeloma Center Brussels, Vrije Universiteit Brussel, 1090 Brussels, Belgium;
| | - Jo Nijs
- Pain in Motion Research Group (PAIN), Department of Physiotherapy, Human Physiology and Anatomy, Faculty of Physical Education and Physiotherapy, Vrije Universiteit Brussel, 1090 Brussels, Belgium; (A.W.); (A.L.); (J.N.); (A.P.)
- Chronic Pain Rehabilitation, Department of Physical Medicine and Physiotherapy, University Hospital, 1090 Brussels, Belgium
- Unit of Physiotherapy, Department of Health and Rehabilitation, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, 405 30 Gothenburg, Sweden
| | - Lode Godderis
- Department of Public Health and Primary Care, Centre for Environment & Health, KU Leuven, Kapucijnenvoer 35, 3000 Leuven, Belgium;
- External Service for Prevention and Protection at Work, IDEWE, 3001 Heverlee, Belgium
| | - Andrea Polli
- Pain in Motion Research Group (PAIN), Department of Physiotherapy, Human Physiology and Anatomy, Faculty of Physical Education and Physiotherapy, Vrije Universiteit Brussel, 1090 Brussels, Belgium; (A.W.); (A.L.); (J.N.); (A.P.)
- Department of Public Health and Primary Care, Centre for Environment & Health, KU Leuven, Kapucijnenvoer 35, 3000 Leuven, Belgium;
- Flanders Research Foundation-FWO, 1090 Brussels, Belgium
| |
Collapse
|
6
|
Fiksinski AM, Hoftman GD, Vorstman JAS, Bearden CE. A genetics-first approach to understanding autism and schizophrenia spectrum disorders: the 22q11.2 deletion syndrome. Mol Psychiatry 2023; 28:341-353. [PMID: 36192458 PMCID: PMC9812786 DOI: 10.1038/s41380-022-01783-5] [Citation(s) in RCA: 27] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/09/2021] [Revised: 08/31/2022] [Accepted: 09/05/2022] [Indexed: 02/03/2023]
Abstract
Recently, increasing numbers of rare pathogenic genetic variants have been identified that are associated with variably elevated risks of a range of neurodevelopmental outcomes, notably including Autism Spectrum Disorders (ASD), Schizophrenia Spectrum Disorders (SSD), and Intellectual Disability (ID). This review is organized along three main questions: First, how can we unify the exclusively descriptive basis of our current psychiatric diagnostic classification system with the recognition of an identifiable, highly penetrant genetic risk factor in an increasing proportion of patients with ASD or SSD? Second, what can be learned from studies of individuals with ASD or SSD who share a common genetic basis? And third, what accounts for the observed variable penetrance and pleiotropy of neuropsychiatric phenotypes in individuals with the same pathogenic variant? In this review, we focus on findings of clinical and preclinical studies of the 22q11.2 deletion syndrome (22q11DS). This particular variant is not only one of the most common among the increasing list of known rare pathogenic variants, but also one that benefits from a relatively long research history. Consequently, 22q11DS is an appealing model as it allows us to: (1) elucidate specific genotype-phenotype associations, (2) prospectively study behaviorally defined classifications, such as ASD or SSD, in the context of a known, well-characterized genetic basis, and (3) elucidate mechanisms underpinning variable penetrance and pleiotropy, phenomena with far-reaching ramifications for research and clinical practice. We discuss how findings from animal and in vitro studies relate to observations in human studies and can help elucidate factors, including genetic, environmental, and stochastic, that impact the expression of neuropsychiatric phenotypes in 22q11DS, and how this may inform mechanisms underlying neurodevelopmental expression in the general population. We conclude with research priorities for the field, which may pave the way for novel therapeutics.
Collapse
Affiliation(s)
- Ania M Fiksinski
- Department of Psychology and Department of Pediatrics, Wilhelmina Children's Hospital, University Medical Center Utrecht, Utrecht, The Netherlands
- Department of Psychiatry and Neuropsychology, Division of Mental Health, MHeNS, Maastricht University, Maastricht, The Netherlands
| | - Gil D Hoftman
- Department of Psychiatry and Biobehavioral Sciences, Semel Institute for Neuroscience and Human Behavior, University of California, Los Angeles, CA, USA
| | - Jacob A S Vorstman
- Program in Genetics and Genome Biology, Research Institute, and Department of Psychiatry, The Hospital for Sick Children, Toronto, ON, Canada
- Department of Psychiatry, University of Toronto, Toronto, ON, Canada
| | - Carrie E Bearden
- Department of Psychiatry and Biobehavioral Sciences, Semel Institute for Neuroscience and Human Behavior, University of California, Los Angeles, CA, USA.
- Department of Psychology, University of California, Los Angeles, CA, USA.
| |
Collapse
|
7
|
Kaki S, DeRosa H, Timmerman B, Brummelte S, Hunter RG, Kentner AC. Developmental Manipulation-Induced Changes in Cognitive Functioning. Curr Top Behav Neurosci 2023; 63:241-289. [PMID: 36029460 PMCID: PMC9971379 DOI: 10.1007/7854_2022_389] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/15/2022]
Abstract
Schizophrenia is a complex neurodevelopmental disorder with as-yet no identified cause. The use of animals has been critical to teasing apart the potential individual and intersecting roles of genetic and environmental risk factors in the development of schizophrenia. One way to recreate in animals the cognitive impairments seen in people with schizophrenia is to disrupt the prenatal or neonatal environment of laboratory rodent offspring. This approach can result in congruent perturbations in brain physiology, learning, memory, attention, and sensorimotor domains. Experimental designs utilizing such animal models have led to a greatly improved understanding of the biological mechanisms that could underlie the etiology and symptomology of schizophrenia, although there is still more to be discovered. The implementation of the Research and Domain Criterion (RDoC) has been critical in taking a more comprehensive approach to determining neural mechanisms underlying abnormal behavior in people with schizophrenia through its transdiagnostic approach toward targeting mechanisms rather than focusing on symptoms. Here, we describe several neurodevelopmental animal models of schizophrenia using an RDoC perspective approach. The implementation of animal models, combined with an RDoC framework, will bolster schizophrenia research leading to more targeted and likely effective therapeutic interventions resulting in better patient outcomes.
Collapse
Affiliation(s)
- Sahith Kaki
- School of Arts and Sciences, Health Psychology Program, Massachusetts College of Pharmacy and Health Sciences, Boston, MA, USA
| | - Holly DeRosa
- School of Arts and Sciences, Health Psychology Program, Massachusetts College of Pharmacy and Health Sciences, Boston, MA, USA
- University of Massachusetts Boston, Boston, MA, USA
| | - Brian Timmerman
- Department of Psychology, Wayne State University, Detroit, MI, USA
| | - Susanne Brummelte
- Department of Psychology, Wayne State University, Detroit, MI, USA
- Translational Neuroscience Program, Wayne State University, Detroit, MI, USA
| | | | - Amanda C Kentner
- School of Arts and Sciences, Health Psychology Program, Massachusetts College of Pharmacy and Health Sciences, Boston, MA, USA.
| |
Collapse
|
8
|
Gregorio I, Mereu M, Contarini G, Bello L, Semplicini C, Burgio F, Russo L, Sut S, Dall'Acqua S, Braghetta P, Semenza C, Pegoraro E, Papaleo F, Bonaldo P, Cescon M. Collagen VI deficiency causes behavioral abnormalities and cortical dopaminergic dysfunction. Dis Model Mech 2022; 15:276265. [PMID: 35946603 PMCID: PMC9548377 DOI: 10.1242/dmm.049481] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2022] [Accepted: 07/29/2022] [Indexed: 11/20/2022] Open
Abstract
Mutations of genes coding for Collagen VI (COL6) cause muscle diseases, including Ullrich congenital muscular dystrophy (UCMD) and Bethlem myopathy (BM). Although more recently COL6 genetic variants were linked to brain pathologies, the impact of COL6 deficiency in brain function is still largely unknown. Here, a thorough behavioral characterization of COL6 null (Col6a1-/-) mice unexpectedly revealed that COL6 deficiency leads to a significant impairment in sensorimotor gating and memory/attention functions. In keeping with these behavioral abnormalities, Col6a1-/- mice displayed alterations in dopaminergic signalling, primarily in the prefrontal cortex (PFC). In vitro co-culture of SH-SY5Y neural cells with primary meningeal fibroblasts from wild-type and Col6a1-/- mice confirmed a direct link between COL6 ablation and defective dopaminergic activity, through a mechanism involving the inability of meningeal cells to sustain dopaminergic differentiation. Finally, patients affected by COL6-related myopathies were evaluated with an ad hoc neuropsychological protocol, revealing distinctive defects in attentional control abilities. Altogether, these findings point at a novel role for COL6 in the proper maintenance of dopamine circuitry function and its related neurobehavioral features in both mice and humans.
Collapse
Affiliation(s)
- Ilaria Gregorio
- Department of Molecular Medicine, University of Padova, 35131 Padova, Italy
| | - Maddalena Mereu
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, 35131, Italy.,Genetics of Cognition Laboratory, Neuroscience Area, Istituto Italiano di Tecnologia, 16163, Genova, Italy
| | - Gabriella Contarini
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, 35131, Italy.,Genetics of Cognition Laboratory, Neuroscience Area, Istituto Italiano di Tecnologia, 16163, Genova, Italy
| | - Luca Bello
- ERN Neuromuscular Center, Department of Neurosciences, University of Padova, 35129 Padova, Italy
| | - Claudio Semplicini
- ERN Neuromuscular Center, Department of Neurosciences, University of Padova, 35129 Padova, Italy
| | | | - Loris Russo
- Department of Molecular Medicine, University of Padova, 35131 Padova, Italy
| | - Stefania Sut
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, 35131, Italy
| | - Stefano Dall'Acqua
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, 35131, Italy
| | - Paola Braghetta
- Department of Molecular Medicine, University of Padova, 35131 Padova, Italy
| | - Carlo Semenza
- ERN Neuromuscular Center, Department of Neurosciences, University of Padova, 35129 Padova, Italy.,IRCCS San Camillo Hospital, 30126 Venice, Italy
| | - Elena Pegoraro
- ERN Neuromuscular Center, Department of Neurosciences, University of Padova, 35129 Padova, Italy
| | - Francesco Papaleo
- Genetics of Cognition Laboratory, Neuroscience Area, Istituto Italiano di Tecnologia, 16163, Genova, Italy
| | - Paolo Bonaldo
- Department of Molecular Medicine, University of Padova, 35131 Padova, Italy
| | - Matilde Cescon
- Department of Molecular Medicine, University of Padova, 35131 Padova, Italy
| |
Collapse
|
9
|
Martens MAG, Dalton N, Scaife J, Harmer CJ, Harrison PJ, Tunbridge EM. Catechol-O-methyltransferase activity does not influence emotional processing in men. J Psychopharmacol 2022; 36:768-775. [PMID: 35443830 PMCID: PMC9150146 DOI: 10.1177/02698811221089032] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
BACKGROUND Catechol-O-methyltransferase (COMT) regulates cortical dopaminergic transmission and prefrontal-dependent cognitive function. However, its role in other cognitive processes, including emotional processing, is relatively unexplored. We therefore investigated the separate and interactive influences of COMT inhibition and Val158Met (rs4680) genotype on performance on an emotional test battery. METHODS We recruited 74 healthy men homozygous for the functional COMT Val158Met polymorphism. Volunteers were administered either a single 200 mg dose of the brain-penetrant COMT inhibitor tolcapone or placebo in a double-blind, randomised manner. Emotional processing was assessed using the emotional test battery, and mood was rated using visual analogue scales and the Profile of Mood States (POMS) questionnaire across the test day. RESULTS There were no main or interactive effects of Val158Met genotype or tolcapone on any of the emotional processing measures or mood ratings. CONCLUSIONS Our findings suggest that, at least in healthy adult men, COMT has little or no effect on emotional processing or mood. These findings contrast with several neuroimaging studies that suggest that COMT modulates neural activity during emotional processing. Thus, further studies are required to understand how COMT impacts on the relationship between behavioural output and neural activity during emotional processing. Nevertheless, our data suggest that novel COMT inhibitors under development for treating cognitive dysfunction are unlikely to have acute off target effects on emotional behaviours.
Collapse
Affiliation(s)
- Marieke AG Martens
- Department of Psychiatry, University of Oxford, Oxford, UK,Oxford Health NHS Foundation Trust, Oxford, UK,Marieke AG Martens, Department of Psychiatry, University of Oxford, Warneford Hospital, Oxford OX3 7JX, UK.
| | - Nina Dalton
- Department of Psychiatry, University of Oxford, Oxford, UK
| | - Jessica Scaife
- Department of Psychiatry, University of Oxford, Oxford, UK,Oxford Health NHS Foundation Trust, Oxford, UK
| | - Catherine J Harmer
- Department of Psychiatry, University of Oxford, Oxford, UK,Oxford Health NHS Foundation Trust, Oxford, UK
| | - Paul J Harrison
- Department of Psychiatry, University of Oxford, Oxford, UK,Oxford Health NHS Foundation Trust, Oxford, UK
| | - Elizabeth M Tunbridge
- Department of Psychiatry, University of Oxford, Oxford, UK,Oxford Health NHS Foundation Trust, Oxford, UK
| |
Collapse
|
10
|
Porceddu PF, Ciampoli M, Romeo E, Garrone B, Durando L, Milanese C, Di Giorgio FP, Reggiani A. The novel potent GSK3 inhibitor AF3581 reverts fragile X syndrome phenotype. Hum Mol Genet 2021; 31:839-849. [PMID: 34596681 DOI: 10.1093/hmg/ddab251] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2021] [Revised: 08/24/2021] [Accepted: 08/25/2021] [Indexed: 11/14/2022] Open
Abstract
Glycogen synthase kinase 3 (GSK3) is a kinase mediating phosphorylation on serine and threonine amino acid residues of several target molecules. The enzyme is involved in the regulation of many cellular processes and aberrant activity of GSK3 has been linked to several disease conditions such as Fragile X Syndrome (FXS). Recent evidences demonstrating an increased activity of GSK3 in murine models of FXS, suggest that dysregulation/hyperactivation of the GSK3 path should contribute to FXS development. A likely possibility could be that in FXS there is a functional impairment of the upstream inhibitory input over GSK3 thus making overactive the kinase. Since GSK3 signaling is a central regulatory node for critical neurodevelopmental pathways, understanding the contribution of GSK3 dysregulation to FXS, may provide novel targets for therapeutic interventions for this disease. In this study we used AF3581, a potent GSK3 inhibitor that we recently discovered, in an in vivo FXS mouse model to elucidate the crucial role of GSK3 in specific behavioral patterns (locomotor activity, sensorimotor gating and social behavior) associated with this disease. All the behavioral alterations manifested by Fmr1 knockout mice were reverted after a chronic treatment with our GSK3 inhibitor, confirming the importance of this pathway as a therapeutic target.
Collapse
Affiliation(s)
- Pier Francesca Porceddu
- D3 Validation Research Line, Istituto Italiano di Tecnologia, Via Morego 30, 16163 Genova, Italy
| | - Mariasole Ciampoli
- D3 Validation Research Line, Istituto Italiano di Tecnologia, Via Morego 30, 16163 Genova, Italy
| | - Elisa Romeo
- D3 Validation Research Line, Istituto Italiano di Tecnologia, Via Morego 30, 16163 Genova, Italy
| | | | - Lucia Durando
- Angelini Pharma S.p.A., Viale Amelia 70, 00181 Rome, Italy
| | | | | | - Angelo Reggiani
- D3 Validation Research Line, Istituto Italiano di Tecnologia, Via Morego 30, 16163 Genova, Italy
| |
Collapse
|
11
|
Leso V, Fontana L, Finiello F, De Cicco L, Luigia Ercolano M, Iavicoli I. Noise induced epigenetic effects: A systematic review. Noise Health 2021; 22:77-89. [PMID: 33402608 PMCID: PMC8000140 DOI: 10.4103/nah.nah_17_20] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Background: Noise-induced hearing loss (NIHL) is one of the leading causes of acquired sensorineural hearing loss. However, molecular mechanisms responsible for its pathogenesis remain to be elucidated. Epigenetic changes, i.e. DNA methylation, histone and microRNA expression modifications may function as a link between noise exposure and hearing loss. Therefore, the aim of the present review was to assess whether epigenetic alterations may serve as biomarkers of noise exposure or early effect. Materials and Methods: A systematic review of studies available in Pubmed, Scopus, and ISI Web of Science databases was performed. Results: Noise exposure was able to induce alterations in DNA methylation levels in workers and animal models, resulting in expression changes of genes related to hearing loss and also to extra-auditory effects. Differently expressed microRNAs were determined in NIHL workers compared to noise-exposed subjects with normal hearing, supporting their possible role as biomarkers of effect. Acoustic trauma affected histon acethylation and methylation levels in animals, suggesting their influence in the pathogenesis of acute noise-induced damage and their role as targets for potential therapeutic treatments. Conclusions: Although preliminary data suggest a relationship between noise and epigenetic effects, the limited number of studies, their different methodologies and the lack of adequate characterization of acoustic insults prevent definite conclusions. In this context, further research aimed to define the epigenetic impact of workplace noise exposure and the role of such alterations in predicting hearing loss may be important for the adoption of correct risk assessment and management strategies in occupational settings.
Collapse
Affiliation(s)
- Veruscka Leso
- Section of Occupational Medicine, Department of Public Health, University of Naples Federico II, Naples, Italy
| | - Luca Fontana
- Section of Occupational Medicine, Department of Public Health, University of Naples Federico II, Naples, Italy
| | - Ferdinando Finiello
- Section of Occupational Medicine, Department of Public Health, University of Naples Federico II, Naples, Italy
| | - Luigi De Cicco
- Section of Occupational Medicine, Department of Public Health, University of Naples Federico II, Naples, Italy
| | - Maria Luigia Ercolano
- Section of Occupational Medicine, Department of Public Health, University of Naples Federico II, Naples, Italy
| | - Ivo Iavicoli
- Section of Occupational Medicine, Department of Public Health, University of Naples Federico II, Naples, Italy
| |
Collapse
|
12
|
Ciampoli M, Scheggia D, Papaleo F. Automatic Intra-/Extra-Dimensional Attentional Set-Shifting Task in Adolescent Mice. Front Behav Neurosci 2021; 15:704684. [PMID: 34349628 PMCID: PMC8326460 DOI: 10.3389/fnbeh.2021.704684] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2021] [Accepted: 06/16/2021] [Indexed: 11/13/2022] Open
Abstract
Adolescence is a developmental period crucial for the maturation of higher-order cognitive functions. Indeed, adolescence deficits in executive functions are strong predictors of increased vulnerability to several mental disabilities later in life. Here, we tested adolescent mice in a fully-automated attentional set-shifting task equivalent to the humans' Wisconsin Card Sorting Test (WCST) and the Cambridge Neuropsychological Test Automated Battery Intra-/Extra-Dimensional set-shift task (ID/ED). Compared to an adult, adolescent mice required more time to complete the task (≈16 days), and a higher percentage failed to finish the entire task. Nevertheless, adolescent mice completing this demanding task showed an increased effort in solving the extradimensional shift stage (EDS) compared to previous stages. Moreover, we found that this paradigm can be used to detect early cognitive dysfunctions in adolescent genetically modified mice. Thus, this automatic paradigm provides a further tool to assess attentional control in adolescent mice, and the development of dysfunctional executive functions from adolescence to adulthood.
Collapse
Affiliation(s)
- Mariasole Ciampoli
- Genetics of Cognition Laboratory, Neuroscience Area, Istituto Italiano di Tecnologia, Genoa, Italy
| | - Diego Scheggia
- Genetics of Cognition Laboratory, Neuroscience Area, Istituto Italiano di Tecnologia, Genoa, Italy.,Department of Pharmacological and Biomolecular Sciences, University of Milan, Milan, Italy
| | - Francesco Papaleo
- Genetics of Cognition Laboratory, Neuroscience Area, Istituto Italiano di Tecnologia, Genoa, Italy.,Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| |
Collapse
|
13
|
Wang J, Xu H, Wang D, Wei G, Zhou H, Wang L, Zhou Y, Zhang X. The interactive effect of genetic polymorphisms of IL-10 and COMT on cognitive function in schizophrenia. J Psychiatr Res 2021; 136:501-507. [PMID: 33127070 DOI: 10.1016/j.jpsychires.2020.10.021] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/01/2020] [Revised: 09/24/2020] [Accepted: 10/16/2020] [Indexed: 12/13/2022]
Abstract
Dopaminergic and inflammatory systems have been proven to play an important role in the cognitive deficits of schizophrenia. Although increasing evidence indicates two systems have strong interaction, the relevant research on this interaction is still limited. Catechol-o-methyltransferase (COMT) and Interleukin-10 (IL-10) play critical functions in dopaminergic and inflammatory systems respectively, and their genetic polymorphisms are both associated with cognitive function. However, the interactive effect of their genetic polymorphisms has not been investigated. In this study, COMT Val158Met (rs4680) and IL-10 -592A/C (rs1800872) polymorphisms were measured in patients with chronic schizophrenia (n = 244) and healthy controls (n = 396), and their cognitive functions were assessed using the "Repeatable Battery for the Assessment of Neuropsychological Status" (RBANS). We found that IL-10 alone had no effect on cognitive function, while COMT affected language ability and interacted with the schizophrenia (case vs control) or sex in multiple RBANS indexes. Additionally, we found there was a significant interactive effect between IL-10 and COMT polymorphisms on multiple cognitive indexes of RBANS. In detail, the analysis showed that the IL-10 polymorphism had opposite effects on cognitive function in different COMT genotype carriers; meanwhile, the polymorphism of COMT only had a significant effect on cognitive function in IL-10 C carriers. And this interaction was more significant in schizophrenia than in controls. Our study discovered for the first time, there is an interactive effect between IL-10 and COMT genetic polymorphisms on cognitive function, which is valuable for further investigations and drug administrations associated with both systems.
Collapse
Affiliation(s)
- Jiesi Wang
- CAS Key Laboratory of Mental Health, Institute of Psychology, Chinese Academy of Sciences, Beijing, China; Department of Psychology, University of Chinese Academy of Sciences, Beijing, China
| | - Hang Xu
- CAS Key Laboratory of Mental Health, Institute of Psychology, Chinese Academy of Sciences, Beijing, China; Department of Psychology, University of Chinese Academy of Sciences, Beijing, China
| | - Dongmei Wang
- CAS Key Laboratory of Mental Health, Institute of Psychology, Chinese Academy of Sciences, Beijing, China; Department of Psychology, University of Chinese Academy of Sciences, Beijing, China
| | - Gaoxia Wei
- CAS Key Laboratory of Mental Health, Institute of Psychology, Chinese Academy of Sciences, Beijing, China; Department of Psychology, University of Chinese Academy of Sciences, Beijing, China
| | - Huixia Zhou
- CAS Key Laboratory of Mental Health, Institute of Psychology, Chinese Academy of Sciences, Beijing, China; Department of Psychology, University of Chinese Academy of Sciences, Beijing, China
| | - Li Wang
- CAS Key Laboratory of Mental Health, Institute of Psychology, Chinese Academy of Sciences, Beijing, China; Department of Psychology, University of Chinese Academy of Sciences, Beijing, China
| | - Yongjie Zhou
- Research Center for Psychological and Health Sciences, China University of Geosciences, Wuhan, China; Affiliated Wuhan Mental Health Center, Tongji Medical College of Huazhong University of Science & Technology, Wuhan, China
| | - Xiangyang Zhang
- CAS Key Laboratory of Mental Health, Institute of Psychology, Chinese Academy of Sciences, Beijing, China; Department of Psychology, University of Chinese Academy of Sciences, Beijing, China.
| |
Collapse
|
14
|
The epistatic interaction between the dopamine D3 receptor and dysbindin-1 modulates higher-order cognitive functions in mice and humans. Mol Psychiatry 2021; 26:1272-1285. [PMID: 31492942 DOI: 10.1038/s41380-019-0511-4] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/18/2018] [Revised: 07/15/2019] [Accepted: 07/26/2019] [Indexed: 11/08/2022]
Abstract
The dopamine D2 and D3 receptors are implicated in schizophrenia and its pharmacological treatments. These receptors undergo intracellular trafficking processes that are modulated by dysbindin-1 (Dys). Indeed, Dys variants alter cognitive responses to antipsychotic drugs through D2-mediated mechanisms. However, the mechanism by which Dys might selectively interfere with the D3 receptor subtype is unknown. Here, we revealed an interaction between functional genetic variants altering Dys and D3. Specifically, both in patients with schizophrenia and in genetically modified mice, concomitant reduction in D3 and Dys functionality was associated with improved executive and working memory abilities. This D3/Dys interaction produced a D2/D3 imbalance favoring increased D2 signaling in the prefrontal cortex (PFC) but not in the striatum. No epistatic effects on the clinical positive and negative syndrome scale (PANSS) scores were evident, while only marginal effects on sensorimotor gating, locomotor functions, and social behavior were observed in mice. This genetic interaction between D3 and Dys suggests the D2/D3 imbalance in the PFC as a target for patient stratification and procognitive treatments in schizophrenia.
Collapse
|
15
|
Sohn R, Rösch G, Junker M, Meurer A, Zaucke F, Jenei-Lanzl Z. Adrenergic signalling in osteoarthritis. Cell Signal 2021; 82:109948. [PMID: 33571663 DOI: 10.1016/j.cellsig.2021.109948] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2020] [Revised: 02/03/2021] [Accepted: 02/04/2021] [Indexed: 12/14/2022]
Abstract
Adrenoceptors (ARs) mediate the effects of the sympathetic neurotransmitters norepinephrine (NE) and epinephrine (E) in the human body and play a central role in physiologic and pathologic processes. Therefore, ARs have long been recognized as targets for therapeutic agents, especially in the field of cardiovascular medicine. During the past decades, the contribution of the sympathetic nervous system (SNS) and particularly of its major peripheral catecholamine NE to the pathogenesis of osteoarthritis (OA) attracted growing interest. OA is the most common degenerative joint disorder worldwide and a disease of the whole joint. It is characterized by progressive degradation of articular cartilage, synovial inflammation, osteophyte formation, and subchondral bone sclerosis mostly resulting in chronic pain. The subchondral bone marrow, the periosteum, the synovium, the vascular meniscus and numerous tendons and ligaments are innervated by tyrosine hydroxylase-positive (TH+) sympathetic nerve fibers that release NE into the synovial fluid and cells of all abovementioned joint tissues express at least one out of nine AR subtypes. During the past decades, several in vitro studies explored the AR-mediated effects of NE on different cell types in the joint. So far, only a few studies used animal OA models to investigate the contribution of distinct AR subtypes to OA pathogenesis in vivo. This narrative review shortly summarizes the current background knowledge about ARs and their signalling pathways at first. In the second part, we focus on recent findings in the field of NE-induced AR-mediated signalling in different joint tissues during OA pathogenesis and at the end, we will delineate the potential of targeting the adrenergic signalling for OA prevention or treatment. We used the PubMed bibliographic database to search for keywords such as 'joint' or 'cartilage' or 'synovium' or 'bone' and 'osteoarthritis' and/or 'trauma' and 'sympathetic nerve fibers' and/or 'norepinephrine' and 'adrenergic receptors / adrenoceptors' as well as 'adrenergic therapy'.
Collapse
Affiliation(s)
- Rebecca Sohn
- Dr. Rolf M. Schwiete Research Unit for Osteoarthritis, Department of Orthopedics (Friedrichsheim), University Hospital Frankfurt, Goethe University, Frankfurt / Main, Germany
| | - Gundula Rösch
- Dr. Rolf M. Schwiete Research Unit for Osteoarthritis, Department of Orthopedics (Friedrichsheim), University Hospital Frankfurt, Goethe University, Frankfurt / Main, Germany
| | - Marius Junker
- Dr. Rolf M. Schwiete Research Unit for Osteoarthritis, Department of Orthopedics (Friedrichsheim), University Hospital Frankfurt, Goethe University, Frankfurt / Main, Germany
| | - Andrea Meurer
- Dr. Rolf M. Schwiete Research Unit for Osteoarthritis, Department of Orthopedics (Friedrichsheim), University Hospital Frankfurt, Goethe University, Frankfurt / Main, Germany
| | - Frank Zaucke
- Dr. Rolf M. Schwiete Research Unit for Osteoarthritis, Department of Orthopedics (Friedrichsheim), University Hospital Frankfurt, Goethe University, Frankfurt / Main, Germany
| | - Zsuzsa Jenei-Lanzl
- Dr. Rolf M. Schwiete Research Unit for Osteoarthritis, Department of Orthopedics (Friedrichsheim), University Hospital Frankfurt, Goethe University, Frankfurt / Main, Germany.
| |
Collapse
|
16
|
Low catechol-O-methyltransferase and stress potentiate functional pain and depressive behavior, especially in female mice. Pain 2021; 161:446-458. [PMID: 31972854 DOI: 10.1097/j.pain.0000000000001734] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Low levels of catechol-O-methyltransferase (COMT), an enzyme that metabolizes catecholamines, and stress, which potentiates catecholamine release from sympathetic nerves, are fundamental to chronic functional pain syndromes and comorbid depression, which predominantly affect females. Here, we sought to examine the independent and joint contributions of low COMT and stress to chronic functional pain and depression at the behavioral and molecular level. Male and female C57BL/6 mice received sustained systemic delivery of the COMT inhibitor OR486 over 14 days and underwent a swim stress paradigm on days 8 to 10. Pain and depressive-like behavior were measured over 14 days, and brain-derived neurotrophic factor (BDNF; a factor involved in nociception and depression) and glucocorticoid receptor (GR; a stress-related receptor) expression were measured on day 14. We found that stress potentiates the effect of low COMT on functional pain and low COMT potentiates the effect of stress on depressive-like behavior. The joint effects of low COMT and stress on functional pain and depressive-like behavior were significantly greater in females vs males. Consistent with behavioral data, we found that stress potentiates COMT-dependent increases in spinal BDNF and low COMT potentiates stress-dependent decreases in hippocampal BDNF in females, but not males. Although low COMT increases spinal GR and stress increases hippocampal GR expression, these increases are not potentiated in the OR486 + stress group and are not sex-specific. These results suggest that genetic and environmental factors that enhance catecholamine bioavailability cause abnormalities in BDNF signaling and increase risk of comorbid functional pain and depression, especially among females.
Collapse
|
17
|
Durairaja A, Fendt M. Orexin deficiency modulates cognitive flexibility in a sex-dependent manner. GENES BRAIN AND BEHAVIOR 2020; 20:e12707. [PMID: 33070452 DOI: 10.1111/gbb.12707] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/28/2020] [Revised: 10/01/2020] [Accepted: 10/14/2020] [Indexed: 12/18/2022]
Abstract
Cognitive flexibility is an important executive function and refers to the ability to adapt behaviors in response to changes in the environment. Of note, many brain disorders are associated with impairments in cognitive flexibility. Several classical neurotransmitter systems including dopamine, acetylcholine and noradrenaline are shown to be important for cognitive flexibility, however, there is not much known about the role of neuropeptides. The neuropeptide orexin, which is brain-widely released by neurons in the lateral hypothalamus, is a major player in maintaining sleep/wake cycle, feeding behavior, arousal, and motivational behavior. Recent studies showed a role of orexin in attention, cognition and stress-induced attenuation of cognitive flexibility by disrupting orexin signaling locally or systemically. However, it is not known so far whether brain-wide reduction or loss of orexin affects cognitive flexibility. We investigated this question by testing male and female orexin-deficient mice in the attentional set shifting task (ASST), an established paradigm of cognitive flexibility. We found that orexin deficiency impaired the intra-dimensional shift phase of the ASST selectively in female homozygous orexin-deficient mice and improved the first reversal learning phase selectively in male homozygous orexin-deficient mice. We also found that these orexin-mediated sex-based modulations of cognitive flexibility were not correlated with trait anxiety, narcoleptic episodes, and reward consumption. Our findings highlight a sexually dimorphic role of orexin in regulating cognitive flexibility and the need for further investigations of sex-specific functions of the orexin circuitry.
Collapse
Affiliation(s)
- Archana Durairaja
- Institute for Pharmacology and Toxicology, Otto-von-Guericke University, Magdeburg, Germany
| | - Markus Fendt
- Institute for Pharmacology and Toxicology, Otto-von-Guericke University, Magdeburg, Germany.,Center of Behavioral Brain Sciences, Otto-von-Guericke University, Magdeburg, Germany
| |
Collapse
|
18
|
Leggio GM, Torrisi SA, Papaleo F. The Discrete Paired-trial Variable-delay T-maze Task to Assess Working Memory in Mice. Bio Protoc 2020; 10:e3664. [PMID: 33659334 DOI: 10.21769/bioprotoc.3664] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2020] [Revised: 04/23/2020] [Accepted: 04/27/2020] [Indexed: 11/02/2022] Open
Abstract
Working memory abnormalities involving the prefrontal cortex (PFC) dramatically contribute to poor functional outcomes in patients with schizophrenia and still represent an unmet therapeutic need. Studies in rodents might provide essential tools to understand the mechanisms underlying PFC-dependent working memory dysfunctions, as well as precious tools for genetic and pharmacological testing. However, proper tests assessing working memory and sensitive to PFC-dependent functions must be used. In this regard, the discrete paired-trial variable-delay T-maze task, equivalent to delayed non-match to sample tasks used in humans, has proved to be an effective paradigm to test PFC-dependent working memory dysfunctions with high predictive validity in human studies.
Collapse
Affiliation(s)
- Gian Marco Leggio
- Department of Biomedical and Biotechnological Sciences, University of Catania, Catania, Italy
| | | | - Francesco Papaleo
- Genetics of Cognition laboratory, Neuroscience area, Istituto Italiano di Tecnologia, Genova, Italy
| |
Collapse
|
19
|
Yang JH, Presby RE, Cayer S, Rotolo RA, Perrino PA, Fitch RH, Correa M, Chesler EJ, Salamone JD. Effort-related decision making in humanized COMT mice: Effects of Val 158Met polymorphisms and possible implications for negative symptoms in humans. Pharmacol Biochem Behav 2020; 196:172975. [PMID: 32593787 DOI: 10.1016/j.pbb.2020.172975] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/20/2020] [Revised: 06/05/2020] [Accepted: 06/21/2020] [Indexed: 01/05/2023]
Abstract
Catechol-o-methyltransferase (COMT) is an enzyme that metabolizes catecholamines, and is crucial for clearance of dopamine (DA) in prefrontal cortex. Val158Met polymorphism, which causes a valine (Val) to methionine (Met) substitution at codon 158, is reported to be associated with human psychopathologies in some studies. The Val/Val variant of the enzyme results in higher dopamine metabolism, which results in reduced dopamine transmission. Thus, it is important to investigate the relation between Val158Met polymorphisms using rodent models of psychiatric symptoms, including negative symptoms such as motivational dysfunction. In the present study, humanized COMT transgenic mice with two genotype groups (Val/Val (Val) and Met/Met (Met) homozygotes) and wild-type (WT) mice from the S129 background were tested using a touchscreen effort-based choice paradigm. Mice were trained to choose between delivery of a preferred liquid diet that reinforced panel pressing on various fixed ratio (FR) schedules (high-effort alternative), vs. intake of pellets concurrently available in the chamber (low-effort alternative). Panel pressing requirements were controlled by varying the FR levels (FR1, 2, 4, 8, 16) in ascending and descending sequences across weeks of testing. All mice were able to acquire the initial touchscreen operant training, and there was an inverse relationship between the number of reinforcers delivered by panel pressing and pellet intake across different FR levels. There was a significant group x FR level interaction in the ascending limb, with panel presses in the Val group being significantly lower than the WT group in FR1-8, and lower than Met in FR4. These findings indicate that the humanized Val allele in mice modulates FR/pellet-choice performance, as marked by lower levels of panel pressing in the Val group when the ratio requirement was moderately high. These studies may contribute to the understanding of the role of COMT polymorphisms in negative symptoms such as motivational dysfunctions in schizophrenic patients.
Collapse
Affiliation(s)
- Jen-Hau Yang
- Behavioral Neuroscience Division, Department of Psychology, University of Connecticut, Storrs, CT, USA; Present address: Dept. of Psychiatry, Yale University, New Haven, CT, USA
| | - Rose E Presby
- Behavioral Neuroscience Division, Department of Psychology, University of Connecticut, Storrs, CT, USA
| | - Suzanne Cayer
- Behavioral Neuroscience Division, Department of Psychology, University of Connecticut, Storrs, CT, USA
| | - Renee A Rotolo
- Behavioral Neuroscience Division, Department of Psychology, University of Connecticut, Storrs, CT, USA
| | - Peter A Perrino
- Behavioral Neuroscience Division, Department of Psychology, University of Connecticut, Storrs, CT, USA
| | - R Holly Fitch
- Behavioral Neuroscience Division, Department of Psychology, University of Connecticut, Storrs, CT, USA
| | - Merce Correa
- Behavioral Neuroscience Division, Department of Psychology, University of Connecticut, Storrs, CT, USA; Area de Psicobiologia, Universitat Jaume I, Castelló, Spain
| | | | - John D Salamone
- Behavioral Neuroscience Division, Department of Psychology, University of Connecticut, Storrs, CT, USA.
| |
Collapse
|
20
|
Fan P, Qi X, Sweet RA, Wang L. Network Systems Pharmacology-Based Mechanism Study on the Beneficial Effects of Vitamin D against Psychosis in Alzheimer's Disease. Sci Rep 2020; 10:6136. [PMID: 32273551 PMCID: PMC7145835 DOI: 10.1038/s41598-020-63021-8] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2019] [Accepted: 03/21/2020] [Indexed: 11/08/2022] Open
Abstract
Alzheimer's disease (AD) is a chronic neurodegenerative disease with significant financial costs and negative impacts on quality of life. Psychotic symptoms, i.e., the presence of delusions and/or hallucinations, is a frequent complication of AD. About 50% of AD patients will develop psychotic symptoms (AD with Psychosis, or AD + P) and these patients will experience an even more rapid cognitive decline than AD patients without psychosis (AD-P). In a previous analysis on medication records of 776 AD patients, we had shown that use of Vitamin D was associated with delayed time to psychosis in AD patients and Vitamin D was used more by AD-P than AD + P patients. To explore the potential molecular mechanism behind our findings, we applied systems pharmacology approaches to investigate the crosstalk between AD and psychosis. Specifically, we built protein-protein interaction (PPI) networks with proteins encoded by AD- and psychosis-related genes and Vitamin D-perturbed genes. Using network analysis we identified several high-impact genes, including NOTCH4, COMT, CACNA1C and DRD3 which are related to calcium homeostasis. The new findings highlight the key role of calcium-related signaling pathways in AD + P development and may provide a new direction and facilitate hypothesis generation for future drug development.
Collapse
Affiliation(s)
- Peihao Fan
- Department of Pharmaceutical Sciences, Computational Chemical Genomics Screening Center, University of Pittsburgh School of Pharmacy, Pittsburgh, USA
| | - Xiguang Qi
- Department of Pharmaceutical Sciences, Computational Chemical Genomics Screening Center, University of Pittsburgh School of Pharmacy, Pittsburgh, USA
| | - Robert A Sweet
- Department of Neurology, University of Pittsburgh School of Medicine, Pittsburgh, USA.
- Department of Psychiatry, University of Pittsburgh School of Medicine, Pittsburgh, USA.
| | - Lirong Wang
- Department of Pharmaceutical Sciences, Computational Chemical Genomics Screening Center, University of Pittsburgh School of Pharmacy, Pittsburgh, USA.
| |
Collapse
|
21
|
Morè L, Lauterborn JC, Papaleo F, Brambilla R. Enhancing cognition through pharmacological and environmental interventions: Examples from preclinical models of neurodevelopmental disorders. Neurosci Biobehav Rev 2020; 110:28-45. [PMID: 30981451 DOI: 10.1016/j.neubiorev.2019.02.003] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2018] [Revised: 02/04/2019] [Accepted: 02/05/2019] [Indexed: 12/29/2022]
Abstract
In this review we discuss the role of environmental and pharmacological treatments to enhance cognition with special regards to neurodevelopmental related disorders and aging. How the environment influences brain structure and function, and the interactions between rearing conditions and gene expression, are fundamental questions that are still poorly understood. We propose a model that can explain some of the discrepancies in findings for effects of environmental enrichment on outcome measures. Evidence of a direct causal correlation of nootropics and treatments that enhanced cognition also will be presented, and possible molecular mechanisms that include neurotrophin signaling and downstream pathways underlying these processes are discussed. Finally we review recent findings achieved with a wide set of behavioral and cognitive tasks that have translational validity to humans, and should be useful for future work on devising appropriate therapies. As will be discussed, the collective findings suggest that a combinational therapeutic approach of environmental enrichment and nootropics could be particularly successful for improving learning and memory in both developmental disorders and normal aging.
Collapse
Affiliation(s)
- Lorenzo Morè
- School of Pharmacy and Biomedical Sciences, University of Central Lancashire, PR1 2XT, Preston, UK.
| | - Julie C Lauterborn
- Department of Anatomy & Neurobiology, School of Medicine, University of California, Irvine, CA, 92617, USA.
| | - Francesco Papaleo
- Genetics of Cognition Laboratory, Istituto Italiano di Tecnologia, Via Morego, 30, 16163, Genova, Italy.
| | - Riccardo Brambilla
- Neuroscience and Mental Health Research Institute (NMHRI), Division of Neuroscience, School of Biosciences, Cardiff University, CF24 4HQ, Cardiff, UK.
| |
Collapse
|
22
|
Maul S, Giegling I, Fabbri C, Corponi F, Serretti A, Rujescu D. Genetics of resilience: Implications from genome-wide association studies and candidate genes of the stress response system in posttraumatic stress disorder and depression. Am J Med Genet B Neuropsychiatr Genet 2020; 183:77-94. [PMID: 31583809 DOI: 10.1002/ajmg.b.32763] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/09/2019] [Revised: 07/22/2019] [Accepted: 09/03/2019] [Indexed: 12/28/2022]
Abstract
Resilience is the ability to cope with critical situations through the use of personal and socially mediated resources. Since a lack of resilience increases the risk of developing stress-related psychiatric disorders such as posttraumatic stress disorder (PTSD) and major depressive disorder (MDD), a better understanding of the biological background is of great value to provide better prevention and treatment options. Resilience is undeniably influenced by genetic factors, but very little is known about the exact underlying mechanisms. A recently published genome-wide association study (GWAS) on resilience has identified three new susceptibility loci, DCLK2, KLHL36, and SLC15A5. Further interesting results can be found in association analyses of gene variants of the stress response system, which is closely related to resilience, and PTSD and MDD. Several promising genes, such as the COMT (catechol-O-methyltransferase) gene, the serotonin transporter gene (SLC6A4), and neuropeptide Y (NPY) suggest gene × environment interaction between genetic variants, childhood adversity, and the occurrence of PTSD and MDD, indicating an impact of these genes on resilience. GWAS on PTSD and MDD provide another approach to identifying new disease-associated loci and, although the functional significance for disease development for most of these risk genes is still unknown, they are potential candidates due to the overlap of stress-related psychiatric disorders and resilience. In the future, it will be important for genetic studies to focus more on resilience than on pathological phenotypes, to develop reasonable concepts for measuring resilience, and to establish international cooperations to generate sufficiently large samples.
Collapse
Affiliation(s)
- Stephan Maul
- Department of Psychiatry, Psychotherapy, and Psychosomatics, Martin-Luther-University Halle-Wittenberg, Halle, Germany
| | - Ina Giegling
- Department of Psychiatry, Psychotherapy, and Psychosomatics, Martin-Luther-University Halle-Wittenberg, Halle, Germany
| | - Chiara Fabbri
- Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy
| | - Filippo Corponi
- Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy
| | - Alessandro Serretti
- Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy
| | - Dan Rujescu
- Department of Psychiatry, Psychotherapy, and Psychosomatics, Martin-Luther-University Halle-Wittenberg, Halle, Germany
| |
Collapse
|
23
|
Cognition- and circuit-based dysfunction in a mouse model of 22q11.2 microdeletion syndrome: effects of stress. Transl Psychiatry 2020; 10:41. [PMID: 32066701 PMCID: PMC7026063 DOI: 10.1038/s41398-020-0687-z] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/10/2019] [Revised: 11/19/2019] [Accepted: 11/27/2019] [Indexed: 12/24/2022] Open
Abstract
Genetic microdeletion at the 22q11 locus is associated with very high risk for schizophrenia. The 22q11.2 microdeletion (Df(h22q11)/+) mouse model shows cognitive deficits observed in this disorder, some of which can be linked to dysfunction of the prefrontal cortex (PFC). We used behavioral (n = 10 per genotype), electrophysiological (n = 7 per genotype per group), and neuroanatomical (n = 5 per genotype) techniques to investigate schizophrenia-related pathology of Df(h22q11)/+ mice, which showed a significant decrease in the total number of parvalbumin positive interneurons in the medial PFC. The Df(h22q11)/+ mice when tested on PFC-dependent behavioral tasks, including gambling tasks, perform significantly worse than control animals while exhibiting normal behavior on hippocampus-dependent tasks. They also show a significant decrease in hippocampus-medial Prefrontal cortex (H-PFC) synaptic plasticity (long-term potentiation, LTP). Acute platform stress almost abolished H-PFC LTP in both wild-type and Df(h22q11)/+ mice. H-PFC LTP was restored to prestress levels by clozapine (3 mg/kg i.p.) in stressed Df(h22q11)/+ mice, but the restoration of stress-induced LTP, while significant, was similar between wild-type and Df(h22q11)/+ mice. A medial PFC dysfunction may underlie the negative and cognitive symptoms in human 22q11 deletion carriers, and these results are relevant to the current debate on the utility of clozapine in such subjects.
Collapse
|
24
|
Ernst G, Akuma D, Au V, Buchler IP, Byers S, Carr GV, Defays S, de León P, Demaude T, DePasquale M, Durieu V, Huang Y, Jigorel E, Kimos M, Kolobova A, Montel F, Moureau F, Poslusney M, Swinnen D, Vandergeten MC, Van houtvin N, Wei H, White N, Wood M, Barrow JC. Synthesis and Evaluation of Bicyclic Hydroxypyridones as Inhibitors of Catechol O-Methyltransferase. ACS Med Chem Lett 2019; 10:1573-1578. [PMID: 32038769 PMCID: PMC7003998 DOI: 10.1021/acsmedchemlett.9b00345] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2019] [Accepted: 10/22/2019] [Indexed: 11/30/2022] Open
Abstract
![]()
A series
of bicyclic pyridones were identified as potent inhibitors
of catechol O-methyltransferase (COMT). Substituted
benzyl groups attached to the basic nitrogen of the core scaffold
gave the most potent inhibitors within this series. Rat pharmacokinetic
studies showed medium to high levels of clearance for this series,
but with high free fraction due to remarkably low levels of protein
and tissue binding. In rat biomarker studies, levels of unbound drug
exposure are seen in the brain, which exceed their respective IC50s, leading to changes in the levels of dopamine metabolites
in a manner consistent with COMT inhibition.
Collapse
Affiliation(s)
- Glen Ernst
- Lieber Institute for Brain Development, 855 North Wolfe Street, Baltimore, Maryland 21205, United States
| | - Daniel Akuma
- Lieber Institute for Brain Development, 855 North Wolfe Street, Baltimore, Maryland 21205, United States
| | - Vinh Au
- Lieber Institute for Brain Development, 855 North Wolfe Street, Baltimore, Maryland 21205, United States
| | - Ingrid P. Buchler
- Lieber Institute for Brain Development, 855 North Wolfe Street, Baltimore, Maryland 21205, United States
| | - Spencer Byers
- Lieber Institute for Brain Development, 855 North Wolfe Street, Baltimore, Maryland 21205, United States
| | - Gregory V. Carr
- Lieber Institute for Brain Development, 855 North Wolfe Street, Baltimore, Maryland 21205, United States
- Department of Pharmacology, Johns Hopkins University School of Medicine, 855 North Wolfe Street, Baltimore, Maryland 21287, United States
| | - Sabine Defays
- UCB Biopharma, SPRL, B-1420 Braine-L’Alleud, Belgium
| | - Pablo de León
- Lieber Institute for Brain Development, 855 North Wolfe Street, Baltimore, Maryland 21205, United States
| | | | - Michael DePasquale
- Lieber Institute for Brain Development, 855 North Wolfe Street, Baltimore, Maryland 21205, United States
| | | | - Yifang Huang
- Lieber Institute for Brain Development, 855 North Wolfe Street, Baltimore, Maryland 21205, United States
| | | | - Martha Kimos
- Lieber Institute for Brain Development, 855 North Wolfe Street, Baltimore, Maryland 21205, United States
| | - Anna Kolobova
- Lieber Institute for Brain Development, 855 North Wolfe Street, Baltimore, Maryland 21205, United States
| | | | | | - Michael Poslusney
- Lieber Institute for Brain Development, 855 North Wolfe Street, Baltimore, Maryland 21205, United States
| | | | | | | | - Huijun Wei
- Lieber Institute for Brain Development, 855 North Wolfe Street, Baltimore, Maryland 21205, United States
- Department of Pharmacology, Johns Hopkins University School of Medicine, 855 North Wolfe Street, Baltimore, Maryland 21287, United States
| | - Noelle White
- Lieber Institute for Brain Development, 855 North Wolfe Street, Baltimore, Maryland 21205, United States
| | - Martyn Wood
- UCB Biopharma, SPRL, B-1420 Braine-L’Alleud, Belgium
| | - James C. Barrow
- Lieber Institute for Brain Development, 855 North Wolfe Street, Baltimore, Maryland 21205, United States
- Department of Pharmacology, Johns Hopkins University School of Medicine, 855 North Wolfe Street, Baltimore, Maryland 21287, United States
| |
Collapse
|
25
|
Hiroi N, Yamauchi T. Modeling and Predicting Developmental Trajectories of Neuropsychiatric Dimensions Associated With Copy Number Variations. Int J Neuropsychopharmacol 2019; 22:488-500. [PMID: 31135887 PMCID: PMC6672556 DOI: 10.1093/ijnp/pyz026] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/15/2019] [Revised: 05/20/2019] [Accepted: 05/24/2019] [Indexed: 01/23/2023] Open
Abstract
Copy number variants, such as duplications and hemizygous deletions at chromosomal loci of up to a few million base pairs, are highly associated with psychiatric disorders. Hemizygous deletions at human chromosome 22q11.2 were found to be associated with elevated instances of schizophrenia and autism spectrum disorder in 1992 and 2002, respectively. Following these discoveries, many mouse models have been developed and tested to analyze the effects of gene dose alterations in small chromosomal segments and single genes of 22q11.2. Despite several limitations to modeling mental illness in mice, mouse models have identified several genes on 22q11.2-Tbx1, Dgcr8, Comt, Sept5, and Prodh-that contribute to dimensions of autism spectrum disorder and schizophrenia, including working memory, social communication and interaction, and sensorimotor gating. Mouse studies have identified that heterozygous deletion of Tbx1 results in defective social communication during the neonatal period and social interaction deficits during adolescence/adulthood. Overexpression of Tbx1 or Comt in adult neural progenitor cells in the hippocampus delays the developmental maturation of working memory capacity. Collectively, mouse models of variants of these 4 genes have revealed several potential neuronal mechanisms underlying various aspects of psychiatric disorders, including adult neurogenesis, microRNA processing, catecholamine metabolism, and synaptic transmission. The validity of the mouse data would be ultimately tested when therapies or drugs based on such potential mechanisms are applied to humans.
Collapse
Affiliation(s)
- Noboru Hiroi
- Department of Psychiatry and Behavioral Sciences, Albert Einstein College of Medicine, Bronx, New York
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, Bronx, New York
- Department of Genetics, Albert Einstein College of Medicine, Bronx, New York
| | - Takahira Yamauchi
- Department of Psychiatry and Behavioral Sciences, Albert Einstein College of Medicine, Bronx, New York
| |
Collapse
|
26
|
Hall KT, Loscalzo J, Kaptchuk TJ. Systems pharmacogenomics - gene, disease, drug and placebo interactions: a case study in COMT. Pharmacogenomics 2019; 20:529-551. [PMID: 31124409 PMCID: PMC6563236 DOI: 10.2217/pgs-2019-0001] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2019] [Accepted: 03/11/2019] [Indexed: 02/07/2023] Open
Abstract
Disease, drugs and the placebos used as comparators are inextricably linked in the methodology of the double-blind, randomized controlled trial. Nonetheless, pharmacogenomics, the study of how individuals respond to drugs based on genetic substrate, focuses primarily on the link between genes and drugs, while the link between genes and disease is often overlooked and the link between genes and placebos is largely ignored. Herein, we use the example of the enzyme catechol-O-methyltransferase to examine the hypothesis that genes can function as pharmacogenomic hubs across system-wide regulatory processes that, if perturbed in andomized controlled trials, can have primary and combinatorial effects on drug and placebo responses.
Collapse
Affiliation(s)
- Kathryn T Hall
- Department of Medicine, Brigham & Women’s Hospital, Boston, MA 02115, USA
- Division of Preventive Medicine, Brigham & Women’s Hospital, Boston, MA 02215, USA
- Harvard Medical School, Boston, MA 02115, USA
| | - Joseph Loscalzo
- Department of Medicine, Brigham & Women’s Hospital, Boston, MA 02115, USA
- Harvard Medical School, Boston, MA 02115, USA
| | - Ted J Kaptchuk
- Harvard Medical School, Boston, MA 02115, USA
- Program in Placebo Studies, Beth Israel Deaconess Medical Center, Boston, MA 02215, USA
| |
Collapse
|
27
|
Klaus K, Pennington K. Dopamine and Working Memory: Genetic Variation, Stress and Implications for Mental Health. Curr Top Behav Neurosci 2019; 41:369-391. [PMID: 31502081 DOI: 10.1007/7854_2019_113] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
At the molecular level, the neurotransmitter dopamine (DA) is a key regulatory component of executive function in the prefrontal cortex (PFC) and dysfunction in dopaminergic (DAergic) circuitry has been shown to result in impaired working memory (WM). Research has identified multiple common genetic variants suggested to impact on the DA system functionally and also behaviourally to alter WM task performance. In addition, environmental stressors impact on DAergic tone, and this may be one mechanism by which stressors confer vulnerability to the development of neuropsychiatric conditions. This chapter aims to evaluate the impact of key DAergic gene variants suggested to impact on both synaptic DA levels (COMT, DAT1, DBH, MAOA) and DA receptor function (ANKK1, DRD2, DRD4) in terms of their influence on visuospatial WM. The role of stressors and interaction with the genetic background is discussed in addition to discussion around some of the implications for precision psychiatry. This and future work in this area aim to disentangle the neural mechanisms underlying susceptibility to stress and their impact and relationship with cognitive processes known to influence mental health vulnerability.
Collapse
Affiliation(s)
- Kristel Klaus
- MRC Brain and Cognition Unit, University of Cambridge, Cambridge, UK
| | | |
Collapse
|
28
|
Kim S, Zhang X, O'Buckley SC, Cooter M, Park JJ, Nackley AG. Acupuncture Resolves Persistent Pain and Neuroinflammation in a Mouse Model of Chronic Overlapping Pain Conditions. THE JOURNAL OF PAIN 2018; 19:1384.e1-1384.e14. [PMID: 29981376 PMCID: PMC6289709 DOI: 10.1016/j.jpain.2018.05.013] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/04/2017] [Revised: 05/24/2018] [Accepted: 05/28/2018] [Indexed: 02/07/2023]
Abstract
Patients with chronic overlapping pain conditions have decreased levels of catechol-O-methyltransferase (COMT), an enzyme that metabolizes catecholamines. Consistent with clinical syndromes, we previously demonstrated that COMT inhibition in rodents produces persistent pain and heightened immune responses. Here, we sought to determine the efficacy of manual acupuncture in resolving persistent pain and neuroinflammation in the classic inbred C57BL/6 strain and the rapid-wound healing MRL/MpJ strain. Mice received subcutaneous osmotic minipumps to deliver the COMT inhibitor OR486 or vehicle for 13 days. On day 7 after pump implantation, acupuncture was performed at the Zusanli (ST36) point or a non-acupoint for 6 consecutive days. Behavioral responses to mechanical stimuli were measured throughout the experiment. Immunohistochemical analysis of spinal phosphorylated p38 mitogen-activated protein kinase, a marker of inflammation, and glial fibrillary acidic protein, a marker of astrogliosis, was performed on day 13. Results demonstrated that ST36, but not sham, acupuncture resolved mechanical hypersensitivity and reduced OR486-dependent increases in phosphorylated p38 and glial fibrillary acidic protein in both strains. The magnitude of the analgesic response was greater in MRL/MpJ mice. These findings indicate acupuncture as an effective treatment for persistent pain linked to abnormalities in catecholamine signaling and, furthermore, that analgesic efficacy may be influenced by genetic differences. PERSPECTIVE: Chronic overlapping pain conditions remain ineffectively managed by conventional pharmacotherapies. Here, we demonstrate that acupuncture alleviates persistent pain and neuroinflammation linked to heightened catecholaminergic tone. Mice with superior healing capacity exhibit greater analgesic efficacy. Findings indicate acupuncture as an effective treatment for chronic overlapping pain conditions and provide insight into treatment response variability.
Collapse
Affiliation(s)
- Seungtae Kim
- Center for Translational Pain Medicine, Department of Anesthesiology, Duke University School of Medicine, Durham, North Carolina; Division of Meridian and Structural Medicine, School of Korean Medicine, Pusan National University, Yangsan, Korea
| | - Xin Zhang
- Center for Translational Pain Medicine, Department of Anesthesiology, Duke University School of Medicine, Durham, North Carolina; Pain Management Center, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| | - Sandra C O'Buckley
- Center for Translational Pain Medicine, Department of Anesthesiology, Duke University School of Medicine, Durham, North Carolina
| | - Mary Cooter
- Department of Anesthesiology, Duke University Medical Center, Durham, North Carolina
| | - Jongbae J Park
- Center for Translational Pain Medicine, Department of Anesthesiology, Duke University School of Medicine, Durham, North Carolina
| | - Andrea G Nackley
- Center for Translational Pain Medicine, Department of Anesthesiology, Duke University School of Medicine, Durham, North Carolina.
| |
Collapse
|
29
|
Buchler I, Akuma D, Au V, Carr G, de León P, DePasquale M, Ernst G, Huang Y, Kimos M, Kolobova A, Poslusney M, Wei H, Swinnen D, Montel F, Moureau F, Jigorel E, Schulze MSED, Wood M, Barrow JC. Optimization of 8-Hydroxyquinolines as Inhibitors of Catechol O-Methyltransferase. J Med Chem 2018; 61:9647-9665. [PMID: 30272964 DOI: 10.1021/acs.jmedchem.8b01126] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
A series of 8-hydroxy quinolines were identified as potent inhibitors of catechol O-methyltransferase (COMT) with selectivity for the membrane-bound form of the enzyme. Small substituents at the 7-position of the quinoline were found to increase metabolic stability without sacrificing potency. Compounds with good pharmacokinetics and brain penetration were identified and demonstrated in vivo modulation of dopamine metabolites in the brain. An X-ray cocrystal structure of compound 21 in the S-COMT active site shows chelation of the active site magnesium similar to catechol-based inhibitors. These compounds should prove useful for treatment of many neurological and psychiatric conditions associated with compromised cortical dopamine signaling.
Collapse
Affiliation(s)
- Ingrid Buchler
- Lieber Institute for Brain Development , 855 North Wolfe Street , Baltimore , Maryland 21205 , United States
| | - Daniel Akuma
- Lieber Institute for Brain Development , 855 North Wolfe Street , Baltimore , Maryland 21205 , United States
| | - Vinh Au
- Lieber Institute for Brain Development , 855 North Wolfe Street , Baltimore , Maryland 21205 , United States
| | - Gregory Carr
- Lieber Institute for Brain Development , 855 North Wolfe Street , Baltimore , Maryland 21205 , United States.,Department of Pharmacology , Johns Hopkins University School of Medicine , 855 North Wolfe Street , Baltimore , Maryland 21287 , United States
| | - Pablo de León
- Lieber Institute for Brain Development , 855 North Wolfe Street , Baltimore , Maryland 21205 , United States
| | - Michael DePasquale
- Lieber Institute for Brain Development , 855 North Wolfe Street , Baltimore , Maryland 21205 , United States
| | - Glen Ernst
- Lieber Institute for Brain Development , 855 North Wolfe Street , Baltimore , Maryland 21205 , United States
| | - Yifang Huang
- Lieber Institute for Brain Development , 855 North Wolfe Street , Baltimore , Maryland 21205 , United States
| | - Martha Kimos
- Lieber Institute for Brain Development , 855 North Wolfe Street , Baltimore , Maryland 21205 , United States
| | - Anna Kolobova
- Lieber Institute for Brain Development , 855 North Wolfe Street , Baltimore , Maryland 21205 , United States
| | - Michael Poslusney
- Lieber Institute for Brain Development , 855 North Wolfe Street , Baltimore , Maryland 21205 , United States
| | - Huijun Wei
- Lieber Institute for Brain Development , 855 North Wolfe Street , Baltimore , Maryland 21205 , United States.,Department of Pharmacology , Johns Hopkins University School of Medicine , 855 North Wolfe Street , Baltimore , Maryland 21287 , United States
| | | | | | | | | | | | - Martyn Wood
- UCB Biopharma SPRL , B-1420 Braine-l'Alleud , Belgium
| | - James C Barrow
- Lieber Institute for Brain Development , 855 North Wolfe Street , Baltimore , Maryland 21205 , United States.,Department of Pharmacology , Johns Hopkins University School of Medicine , 855 North Wolfe Street , Baltimore , Maryland 21287 , United States
| |
Collapse
|
30
|
Knisely MR, Conley YP, Kober KM, Smoot B, Paul SM, Levine JD, Miaskowski C. Associations Between Catecholaminergic and Serotonergic Genes and Persistent Breast Pain Phenotypes After Breast Cancer Surgery. THE JOURNAL OF PAIN 2018; 19:1130-1146. [DOI: 10.1016/j.jpain.2018.04.007] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/13/2017] [Revised: 03/04/2018] [Accepted: 04/12/2018] [Indexed: 12/20/2022]
|
31
|
Nikiforuk A. Assessment of cognitive functions in animal models of schizophrenia. Pharmacol Rep 2018; 70:639-649. [DOI: 10.1016/j.pharep.2018.01.009] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2017] [Revised: 01/19/2018] [Accepted: 01/31/2018] [Indexed: 12/16/2022]
|
32
|
Trusel M, Baldrighi M, Marotta R, Gatto F, Pesce M, Frasconi M, Catelani T, Papaleo F, Pompa PP, Tonini R, Giordani S. Internalization of Carbon Nano-onions by Hippocampal Cells Preserves Neuronal Circuit Function and Recognition Memory. ACS APPLIED MATERIALS & INTERFACES 2018; 10:16952-16963. [PMID: 29669213 DOI: 10.1021/acsami.7b17827] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/08/2023]
Abstract
One area where nanomedicine may offer superior performances and efficacy compared to current strategies is in the diagnosis and treatment of central nervous system (CNS) diseases. However, the application of nanomaterials in such complex arenas is still in its infancy and an optimal vector for the therapy of CNS diseases has not been identified. Graphitic carbon nano-onions (CNOs) represent a class of carbon nanomaterials that shows promising potential for biomedical purposes. To probe the possible applications of graphitic CNOs as a platform for therapeutic and diagnostic interventions on CNS diseases, fluorescently labeled CNOs were stereotaxically injected in vivo in mice hippocampus. Their diffusion within brain tissues and their cellular localization were analyzed ex vivo by confocal microscopy, electron microscopy, and correlative light-electron microscopy techniques. The subsequent fluorescent staining of hippocampal cells populations indicates they efficiently internalize the nanomaterial. Furthermore, the inflammatory potential of the CNOs injection was found comparable to sterile vehicle infusion, and it did not result in manifest neurophysiological and behavioral alterations of hippocampal-mediated functions. These results clearly demonstrate that CNOs can interface effectively with several cell types, which encourages further their development as possible brain disease-targeted diagnostics or therapeutics nanocarriers.
Collapse
Affiliation(s)
- Massimo Trusel
- Neuroscience and Brain Technology , Istituto Italiano di Tecnologia , via Morego 30 , Genova , Italy
| | - Michele Baldrighi
- Nano Carbon Materials , Istituto Italiano di Tecnologia , via Morego 30 , Genova , Italy
| | - Roberto Marotta
- Electron Microscopy Laboratory , Istituto Italiano di Tecnologia , via Morego 30 , Genova , Italy
| | - Francesca Gatto
- Nanobiointeractions & Nanodiagnostics , Istituto Italiano di Tecnologia , via Morego 30 , Genova , Italy
- Department of Engineering for Innovation , University of Salento , Via per Monteroni , Lecce , Italy
| | - Mattia Pesce
- Neuroscience and Brain Technology , Istituto Italiano di Tecnologia , via Morego 30 , Genova , Italy
| | - Marco Frasconi
- Nano Carbon Materials , Istituto Italiano di Tecnologia , via Morego 30 , Genova , Italy
| | - Tiziano Catelani
- Electron Microscopy Laboratory , Istituto Italiano di Tecnologia , via Morego 30 , Genova , Italy
| | - Francesco Papaleo
- Neuroscience and Brain Technology , Istituto Italiano di Tecnologia , via Morego 30 , Genova , Italy
| | - Pier Paolo Pompa
- Nanobiointeractions & Nanodiagnostics , Istituto Italiano di Tecnologia , via Morego 30 , Genova , Italy
| | - Raffaella Tonini
- Neuroscience and Brain Technology , Istituto Italiano di Tecnologia , via Morego 30 , Genova , Italy
| | - Silvia Giordani
- Nano Carbon Materials , Istituto Italiano di Tecnologia , via Morego 30 , Genova , Italy
- Department of Chemistry , University of Turin , Via Giuria 7 , Turin , Italy
| |
Collapse
|
33
|
Hiroi N. Critical reappraisal of mechanistic links of copy number variants to dimensional constructs of neuropsychiatric disorders in mouse models. Psychiatry Clin Neurosci 2018; 72:301-321. [PMID: 29369447 PMCID: PMC5935536 DOI: 10.1111/pcn.12641] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/14/2017] [Revised: 12/27/2017] [Accepted: 01/19/2018] [Indexed: 12/17/2022]
Abstract
Copy number variants are deletions and duplications of a few thousand to million base pairs and are associated with extraordinarily high levels of autism spectrum disorder, schizophrenia, intellectual disability, or attention-deficit hyperactivity disorder. The unprecedented levels of robust and reproducible penetrance of copy number variants make them one of the most promising and reliable entry points to delve into the mechanistic bases of many mental disorders. However, the precise mechanistic bases of these associations still remain elusive in humans due to the many genes encoded in each copy number variant and the diverse associated phenotypic features. Genetically engineered mice have provided a technical means to ascertain precise genetic mechanisms of association between copy number variants and dimensional aspects of mental illnesses. Molecular, cellular, and neuronal phenotypes can be detected as potential mechanistic substrates for various behavioral constructs of mental illnesses. However, mouse models come with many technical pitfalls. Genetic background is not well controlled in many mouse models, leading to rather obvious interpretative issues. Dose alterations of many copy number variants and single genes within copy number variants result in some molecular, cellular, and neuronal phenotypes without a behavioral phenotype or with a behavioral phenotype opposite to what is seen in humans. In this review, I discuss technical and interpretative pitfalls of mouse models of copy number variants and highlight well-controlled studies to suggest potential neuronal mechanisms of dimensional aspects of mental illnesses. Mouse models of copy number variants represent toeholds to achieve a better understanding of the mechanistic bases of dimensions of neuropsychiatric disorders and thus for development of mechanism-based therapeutic options in humans.
Collapse
Affiliation(s)
- Noboru Hiroi
- Department of Psychiatry and Behavioral Sciences, Albert Einstein College of Medicine, New York, USA.,Department of Neuroscience, Albert Einstein College of Medicine, New York, USA.,Department of Genetics, Albert Einstein College of Medicine, New York, USA
| |
Collapse
|
34
|
Crum AJ, Akinola M, Turnwald BP, Kaptchuk TJ, Hall KT. Catechol-O-Methyltransferase moderates effect of stress mindset on affect and cognition. PLoS One 2018; 13:e0195883. [PMID: 29677196 PMCID: PMC5909917 DOI: 10.1371/journal.pone.0195883] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2017] [Accepted: 03/30/2018] [Indexed: 12/12/2022] Open
Abstract
There is evidence that altering stress mindset-the belief that stress is enhancing vs. debilitating-can change cognitive, affective and physiological responses to stress. However individual differences in responsiveness to stress mindset manipulations have not been explored. Given the previously established role of catecholamines in both placebo effects and stress, we hypothesized that genetic variation in catechol-O-methyltransferase (COMT), an enzyme that metabolizes catecholamines, would moderate responses to an intervention intended to alter participants' mindsets about stress. Participants (N = 107) were exposed to a stress mindset manipulation (videos highlighting either the enhancing or debilitating effects of stress) prior to engaging in a Trier Social Stress task and subsequent cognitive tasks. The associations of the COMT rs4680 polymorphism with the effect of stress mindset video manipulations on cognitive and affective responses were examined. Genetic variation at rs4680 modified the effects of stress mindset on affective and cognitive responses to stress. Individuals homozygous for rs4680 low-activity allele (met/met) were responsive to the stress-is-enhancing mindset manipulation as indicated by greater increases in positive affect, improved cognitive functioning, and happiness bias in response to stress. Conversely, individuals homozygous for the high-activity allele (val/val) were not as responsive to the stress mindset manipulation. These results suggest that responses to stress mindset intervention may vary with COMT genotype. These findings contribute to the understanding of gene by environment interactions for mindset interventions and stress reactivity and therefore warrant further investigations.
Collapse
Affiliation(s)
- Alia J. Crum
- Department of Psychology, Stanford University, Stanford, CA, United States of America
| | - Modupe Akinola
- Columbia Business School, Columbia University, New York, NY, United States of America
| | - Bradley P. Turnwald
- Department of Psychology, Stanford University, Stanford, CA, United States of America
| | - Ted J. Kaptchuk
- Division of General Medicine and Primary Care, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, United States of America
- Program in Placebo Studies, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, United States of America
| | - Kathryn T. Hall
- Program in Placebo Studies, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, United States of America
- Division of Preventive Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, United States of America
| |
Collapse
|
35
|
Scheggia D, Zamberletti E, Realini N, Mereu M, Contarini G, Ferretti V, Managò F, Margiani G, Brunoro R, Rubino T, De Luca MA, Piomelli D, Parolaro D, Papaleo F. Remote memories are enhanced by COMT activity through dysregulation of the endocannabinoid system in the prefrontal cortex. Mol Psychiatry 2018. [PMID: 28630452 DOI: 10.1038/mp.2017.126] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The prefrontal cortex (PFC) is a crucial hub for the flexible modulation of recent memories (executive functions) as well as for the stable organization of remote memories. Dopamine in the PFC is implicated in both these processes and genetic variants affecting its neurotransmission might control the unique balance between cognitive stability and flexibility present in each individual. Functional genetic variants in the catechol-O-methyltransferase (COMT) gene result in a different catabolism of dopamine in the PFC. However, despite the established role played by COMT genetic variation in executive functions, its impact on remote memory formation and recall is still poorly explored. Here we report that transgenic mice overexpressing the human COMT-Val gene (COMT-Val-tg) present exaggerated remote memories (>50 days) while having unaltered recent memories (<24 h). COMT selectively and reversibly modulated the recall of remote memories as silencing COMT Val overexpression starting from 30 days after the initial aversive conditioning normalized remote memories. COMT genetic overactivity produced a selective overdrive of the endocannabinoid system within the PFC, but not in the striatum and hippocampus, which was associated with enhanced remote memories. Indeed, acute pharmacological blockade of CB1 receptors was sufficient to rescue the altered remote memory recall in COMT-Val-tg mice and increased PFC dopamine levels. These results demonstrate that COMT genetic variations modulate the retrieval of remote memories through the dysregulation of the endocannabinoid system in the PFC.
Collapse
Affiliation(s)
- D Scheggia
- Department of Neuroscience and Brain Technologies, Istituto Italiano di Tecnologia, Genoa, Italy
| | - E Zamberletti
- Department of Biotechnology and Life Sciences, and Neuroscience Center, University of Insubria, Busto Arsizio, Italy
| | - N Realini
- Drug Discovery and Development, Istituto Italiano di Tecnologia, Genoa, Italy
| | - M Mereu
- Department of Neuroscience and Brain Technologies, Istituto Italiano di Tecnologia, Genoa, Italy.,Dipartimento di Scienze del Farmaco, Universita' degli Studi di Padova, Largo Meneghetti, Padova, Italy
| | - G Contarini
- Dipartimento di Scienze del Farmaco, Universita' degli Studi di Padova, Largo Meneghetti, Padova, Italy
| | - V Ferretti
- Department of Neuroscience and Brain Technologies, Istituto Italiano di Tecnologia, Genoa, Italy
| | - F Managò
- Department of Neuroscience and Brain Technologies, Istituto Italiano di Tecnologia, Genoa, Italy
| | - G Margiani
- Department of Biomedical Sciences, Università di Cagliari, Cagliari, Italy
| | - R Brunoro
- Department of Neuroscience and Brain Technologies, Istituto Italiano di Tecnologia, Genoa, Italy
| | - T Rubino
- Department of Biotechnology and Life Sciences, and Neuroscience Center, University of Insubria, Busto Arsizio, Italy
| | - M A De Luca
- Department of Biomedical Sciences, Università di Cagliari, Cagliari, Italy
| | - D Piomelli
- Drug Discovery and Development, Istituto Italiano di Tecnologia, Genoa, Italy.,Department of Anatomy and Neurobiology, Pharmacology and Biological Chemistry, University of California, Irvine, Irvine, CA, USA
| | - D Parolaro
- Department of Biotechnology and Life Sciences, and Neuroscience Center, University of Insubria, Busto Arsizio, Italy
| | - F Papaleo
- Department of Neuroscience and Brain Technologies, Istituto Italiano di Tecnologia, Genoa, Italy
| |
Collapse
|
36
|
Copy number elevation of 22q11.2 genes arrests the developmental maturation of working memory capacity and adult hippocampal neurogenesis. Mol Psychiatry 2018; 23:985-992. [PMID: 28827761 PMCID: PMC5823706 DOI: 10.1038/mp.2017.158] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/28/2016] [Revised: 06/07/2017] [Accepted: 06/12/2017] [Indexed: 01/08/2023]
Abstract
Working memory capacity, a critical component of executive function, expands developmentally from childhood through adulthood. Anomalies in this developmental process are seen in individuals with autism spectrum disorder (ASD), schizophrenia and intellectual disabilities (ID), implicating this atypical process in the trajectory of developmental neuropsychiatric disorders. However, the cellular and neuronal substrates underlying this process are not understood. Duplication and triplication of copy number variants of 22q11.2 are consistently and robustly associated with cognitive deficits of ASD and ID in humans, and overexpression of small 22q11.2 segments recapitulates dimensional aspects of developmental neuropsychiatric disorders in mice. We capitalized on these two lines of evidence to delve into the cellular substrates for this atypical development of working memory. Using a region- and cell-type-selective gene expression approach, we demonstrated that copy number elevations of catechol-O-methyl-transferase (COMT) or Tbx1, two genes encoded in the two small 22q11.2 segments, in adult neural stem/progenitor cells in the hippocampus prevents the developmental maturation of working memory capacity in mice. Moreover, copy number elevations of COMT or Tbx1 reduced the proliferation of adult neural stem/progenitor cells in a cell-autonomous manner in vitro and migration of their progenies in the hippocampus granular layer in vivo. Our data provide evidence for the novel hypothesis that copy number elevations of these 22q11.2 genes alter the developmental trajectory of working memory capacity via suboptimal adult neurogenesis in the hippocampus.
Collapse
|
37
|
McCane AM, DeLory MJ, Timm MM, Janetsian-Fritz SS, Lapish CC, Czachowski CL. Differential COMT expression and behavioral effects of COMT inhibition in male and female Wistar and alcohol preferring rats. Alcohol 2018; 67:15-22. [PMID: 29310047 PMCID: PMC5818329 DOI: 10.1016/j.alcohol.2017.08.007] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2017] [Revised: 08/14/2017] [Accepted: 08/15/2017] [Indexed: 10/19/2022]
Abstract
Polymorphisms of the catechol-O-methyl transferase (COMT) gene have been associated with alcoholism, suggesting that alterations in the metabolism of catecholamines may be a critical component of the neuropathology of alcoholism. In the current experiments, the COMT inhibitor tolcapone was utilized in an operant behavioral model of reinforcer-seeking and drinking to determine if this compound was capable of remediating the excessive seeking and drinking phenotype of the alcohol-preferring P rat. Tolcapone was administered to male and female alcohol-reinforced P and Wistar rats. Additionally, tolcapone was administered to male sucrose-reinforced P and Wistar rats to determine if its effects also extended to a natural reinforcer. Animals were trained to make an operant response that resulted in 20 min uninterrupted access to the reinforcer solutions. Tolcapone had no effect in female rats on either seeking or consumption of ethanol. However, reductions of both reinforcer seeking and consumption were observed in male P rats, but only of seeking in Wistars. In separate experiments, using reinforcer naïve male and female animals, COMT expression was assessed via Western Blot analysis. Sex differences in COMT expression were also observed, where male P rats exhibited a marked reduction in protein expression relative to females in the PFC. Sex differences were not observed for Wistars or in the striatum and hippocampus. These data complement our previous findings in which tolcapone reduced cue-evoked responses in P rats and further suggest clinical utility of COMT inhibitors in the treatment of addiction disorders, specifically in male high drinkers.
Collapse
Affiliation(s)
- Aqilah M McCane
- Department of Psychology, Indiana University Purdue University Indianapolis, Indianapolis, IN 46202, USA.
| | - Michael J DeLory
- Department of Psychology, Indiana University Purdue University Indianapolis, Indianapolis, IN 46202, USA
| | - Maureen M Timm
- Department of Psychology, Indiana University Purdue University Indianapolis, Indianapolis, IN 46202, USA
| | - Sarine S Janetsian-Fritz
- Department of Psychology, Indiana University Purdue University Indianapolis, Indianapolis, IN 46202, USA
| | - Christopher C Lapish
- Department of Psychology, Indiana University Purdue University Indianapolis, Indianapolis, IN 46202, USA; Stark Neuroscience Research Institute, Indiana University School of Medicine, Indianapolis, IN 46202, USA; Institute for Mathematical Modeling and Computational Sciences, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Cristine L Czachowski
- Department of Psychology, Indiana University Purdue University Indianapolis, Indianapolis, IN 46202, USA; Stark Neuroscience Research Institute, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| |
Collapse
|
38
|
Tait DS, Bowman EM, Neuwirth LS, Brown VJ. Assessment of intradimensional/extradimensional attentional set-shifting in rats. Neurosci Biobehav Rev 2018; 89:72-84. [PMID: 29474818 DOI: 10.1016/j.neubiorev.2018.02.013] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2017] [Revised: 02/06/2018] [Accepted: 02/19/2018] [Indexed: 01/07/2023]
Abstract
The rat intradimensional/extradimensional (ID/ED) task, first described by Birrell and Brown 18 years ago, has become the predominant means by which attentional set-shifting is investigated in rodents: the use of rats in the task has been described in over 135 publications by researchers from nearly 90 universities and pharmaceutical companies. There is variation in the protocols used by different groups, including differences in apparatus, stimuli (both stimulus dimensions and exemplars within), and also the methodology. Nevertheless, most of these variations seem to be of little consequence: there is remarkable similarity in the profile of published data, with consistency of learning rates and in the size and reliability of the set-shifting and reversal 'costs'. However, we suspect that there may be inconsistent data that is unpublished or perhaps 'failed experiments' that may have been caused by unintended deviations from effective protocols. The purpose of this review is to describe our approach and the rationale behind certain aspects of the protocol, including common pitfalls that are encountered when establishing an effective local protocol.
Collapse
Affiliation(s)
- David S Tait
- School of Psychology and Neuroscience, University of St Andrews, St Mary's Quad, South Street, St Andrews, Fife, KY16 9JP, UK.
| | - Eric M Bowman
- School of Psychology and Neuroscience, University of St Andrews, St Mary's Quad, South Street, St Andrews, Fife, KY16 9JP, UK
| | - Lorenz S Neuwirth
- Department of Psychology, SUNY Old Westbury, Old Westbury, NY, 11568, USA; SUNY Neuroscience Research Institute, Old Westbury, NY, 11568, USA
| | - Verity J Brown
- School of Psychology and Neuroscience, University of St Andrews, St Mary's Quad, South Street, St Andrews, Fife, KY16 9JP, UK
| |
Collapse
|
39
|
Amato D, Vernon AC, Papaleo F. Dopamine, the antipsychotic molecule: A perspective on mechanisms underlying antipsychotic response variability. Neurosci Biobehav Rev 2018; 85:146-159. [DOI: 10.1016/j.neubiorev.2017.09.027] [Citation(s) in RCA: 49] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2017] [Revised: 09/20/2017] [Accepted: 09/26/2017] [Indexed: 12/12/2022]
|
40
|
Mony TJ, Hong M, Lee HJ. Empathy Study in Rodent Model of Autism Spectrum Disorders. Psychiatry Investig 2018; 15:104-110. [PMID: 29475229 PMCID: PMC5900397 DOI: 10.30773/pi.2017.06.20] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/01/2017] [Revised: 05/14/2017] [Accepted: 06/20/2017] [Indexed: 11/27/2022] Open
Abstract
There is a highly cognitive and social context to empathy behavior in human. In various social contexts, rodents also display remarkable affective sensitivity and exhibit primitive forms of empathy similar to human. Therefore, we aimed to elaborate the concept of empathy about various components of empathetic behavior in rodents with the similar contexts of a human. In this review, we highlighted the behavioral paradigm that already examined different aspects of rodent empathetic behavior in response to conspecific distress. Additionally, we summarized homologous brain parts of human and rodents to express the empathetic behavior. Integrating the findings with corresponding experiments in the human will provide a novel insight into therapeutic intervention or expanded experimental approaches for neuropsychiatric disorders like autism spectrum disorders associated with empathetic behavior.
Collapse
Affiliation(s)
- Tamanna Jahan Mony
- Department of Pharmacology, School of Medicine, Kangwon National University, Chuncheon, Republic of Korea
| | - Minha Hong
- Department of Psychiatry, School of Medicine, Seonam University, Goyang, Republic of Korea
| | - Hee Jae Lee
- Department of Pharmacology, School of Medicine, Kangwon National University, Chuncheon, Republic of Korea
| |
Collapse
|
41
|
Zhang H, Li J, Yang B, Ji T, Long Z, Xing Q, Shao D, Bai H, Sun J, Cao F. The divergent impact of catechol-O-methyltransferase (COMT) Val 158Met genetic polymorphisms on executive function in adolescents with discrete patterns of childhood adversity. Compr Psychiatry 2018; 81:33-41. [PMID: 29222971 DOI: 10.1016/j.comppsych.2017.11.004] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/29/2017] [Revised: 11/07/2017] [Accepted: 11/17/2017] [Indexed: 12/12/2022] Open
Abstract
PURPOSE Catechol-O-methyltransferase (COMT) Val158Met functional polymorphisms play a crucial role in the development of executive function (EF), but their effect may be moderated by environmental factors such as childhood adversity. The present study aimed at testing the divergent impact of the COMT Val158Met genotype on EF in non-clinical adolescents with discrete patterns of childhood adversity. METHODS A total of 341 participants completed the Childhood Trauma Questionnaire, the self-reported version of the Behavior Rating Inventory of Executive Function, and self-administered questionnaires on familial function. The participants' COMT Val158Met genotype was determined. Associations among the variables were explored using latent class analysis and general linear models. RESULTS We found that Val/Val homozygotes showed significantly worse performance on behavioral shift, relative to Met allele carriers (F=5.921, p=0.015, Partial η2=0.018). Moreover, three typical patterns of childhood adversity, namely, low childhood adversity (23.5%), childhood neglect (59.8%), and high childhood adversity (16.7%), were found. Both childhood neglect and high childhood adversity had a negative impact on each aspect of EF and on global EF performance. Importantly, these results provided evidence for significant interaction effects, as adolescents with the Val/Val genotype showed inferior behavioral shift performance than Met carriers (F=6.647, p=0.010, Partial η2=0.020) in the presence of high childhood adversity. Furthermore, there were no differences between the genotypes for childhood neglect and low childhood adversity. CONCLUSIONS Overall, this is the first study to show that an interaction between the COMT genotype and childhood adversity affects EF in non-clinical adolescents. These results suggest that the COMT genotype may operate as a susceptibility gene vulnerable to an adverse environment.
Collapse
Affiliation(s)
- Huihui Zhang
- School of Nursing, Shandong University, No. 44 Wenhua Xi Road, Jinan, Shandong 250012, PR China
| | - Jie Li
- School of Nursing, Shandong University, No. 44 Wenhua Xi Road, Jinan, Shandong 250012, PR China
| | - Bei Yang
- School of Nursing, Shandong University, No. 44 Wenhua Xi Road, Jinan, Shandong 250012, PR China
| | - Tao Ji
- Zaozhuang Vocational College, Zaozhuang, Shandong, PR China
| | - Zhouting Long
- Zaozhuang Vocational College, Zaozhuang, Shandong, PR China
| | - Qiquan Xing
- Zaozhuang Vocational College, Zaozhuang, Shandong, PR China
| | - Di Shao
- School of Nursing, Shandong University, No. 44 Wenhua Xi Road, Jinan, Shandong 250012, PR China
| | - Huayu Bai
- School of Nursing, Shandong University, No. 44 Wenhua Xi Road, Jinan, Shandong 250012, PR China
| | - Jiwei Sun
- School of Nursing, Shandong University, No. 44 Wenhua Xi Road, Jinan, Shandong 250012, PR China
| | - Fenglin Cao
- School of Nursing, Shandong University, No. 44 Wenhua Xi Road, Jinan, Shandong 250012, PR China.
| |
Collapse
|
42
|
Laviola G, Zoratto F, Ingiosi D, Carito V, Huzard D, Fiore M, Macrì S. Low empathy-like behaviour in male mice associates with impaired sociability, emotional memory, physiological stress reactivity and variations in neurobiological regulations. PLoS One 2017; 12:e0188907. [PMID: 29200428 PMCID: PMC5714342 DOI: 10.1371/journal.pone.0188907] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2017] [Accepted: 11/15/2017] [Indexed: 12/30/2022] Open
Abstract
Deficits in empathy have been proposed to constitute a hallmark of several psychiatric disturbances like conduct disorder, antisocial and narcissistic personality disorders. Limited sensitivity to punishment, shallow or deficient affect and reduced physiological reactivity to environmental stressors have been often reported to co-occur with limited empathy and contribute to the onset of antisocial phenotypes. Empathy in its simplest form (i.e. emotional contagion) is addressed in preclinical models through the evaluation of the social transmission of emotional states: mice exposed to a painful stimulus display a higher response if in the presence of a familiar individual experiencing a higher degree of discomfort, than in isolation. In the present study, we investigated whether a reduction of emotional contagion can be considered a predictor of reduced sociality, sensitivity to punishment and physiological stress reactivity. To this aim, we first evaluated emotional contagion in a group of Balb/cJ mice and then discretised their values in four quartiles. The upper (i.e. Emotional Contagion Prone, ECP) and the lower (i.e. Emotional Contagion Resistant, ECR) quartiles constituted the experimental groups. Our results indicate that mice in the lower quartile are characterized by reduced sociability, impaired memory of negative events and dampened hypothalamic-pituitary-adrenocortical reactivity to external stressors. Furthermore, in the absence of changes in oxytocin receptor density, we show that these mice exhibit elevated concentrations of oxytocin and vasopressin and reduced density of BDNF receptors in behaviourally-relevant brain areas. Thus, not only do present results translate to the preclinical investigation of psychiatric disturbances, but also they can contribute to the study of emotional contagion in terms of its adaptive significance.
Collapse
Affiliation(s)
- Giovanni Laviola
- Reference Centre for Behavioural Sciences and Mental Health, Istituto Superiore di Sanità (ISS), Rome, Italy
- * E-mail:
| | - Francesca Zoratto
- Reference Centre for Behavioural Sciences and Mental Health, Istituto Superiore di Sanità (ISS), Rome, Italy
| | - Danilo Ingiosi
- Reference Centre for Behavioural Sciences and Mental Health, Istituto Superiore di Sanità (ISS), Rome, Italy
| | - Valentina Carito
- Institute of Cell Biology and Neurobiology, National Research Council of Italy (CNR), Rome, Italy
| | - Damien Huzard
- Laboratory of Behavioural Genetics, Brain Mind Institute, Ecole Polytechnique Federale de Lausanne (EPFL), Lausanne, Switzerland
| | - Marco Fiore
- Institute of Cell Biology and Neurobiology, National Research Council of Italy (CNR), Rome, Italy
| | - Simone Macrì
- Reference Centre for Behavioural Sciences and Mental Health, Istituto Superiore di Sanità (ISS), Rome, Italy
| |
Collapse
|
43
|
Brain catechol-O-methyltransferase (COMT) inhibition by tolcapone counteracts recognition memory deficits in normal and chronic phencyclidine-treated rats and in COMT-Val transgenic mice. Behav Pharmacol 2017; 27:415-21. [PMID: 26919286 DOI: 10.1097/fbp.0000000000000208] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
The critical involvement of dopamine in cognitive processes has been well established, suggesting that therapies targeting dopamine metabolism may alleviate cognitive dysfunction. Catechol-O-methyl transferase (COMT) is a catecholamine-degrading enzyme, the substrates of which include dopamine, epinephrine, and norepinephrine. The present work illustrates the potential therapeutic efficacy of COMT inhibition in alleviating cognitive impairment. A brain-penetrant COMT inhibitor, tolcapone, was tested in normal and phencyclidine-treated rats and COMT-Val transgenic mice. In a novel object recognition procedure, tolcapone counteracted a 24-h-dependent forgetting of a familiar object as well as phencyclidine-induced recognition deficits in the rats at doses ranging from 7.5 to 30 mg/kg. In contrast, entacapone, a COMT inhibitor that does not readily cross the blood-brain barrier, failed to show efficacy at doses up to 30 mg/kg. Tolcapone at a dose of 30 mg/kg also improved novel object recognition performance in transgenic mice, which showed clear recognition deficits. Complementing earlier studies, our results indicate that central inhibition of COMT positively impacts recognition memory processes and might constitute an appealing treatment for cognitive dysfunction related to neuropsychiatric disorders.
Collapse
|
44
|
Carr GV, Maltese F, Sibley DR, Weinberger DR, Papaleo F. The Dopamine D5 Receptor Is Involved in Working Memory. Front Pharmacol 2017; 8:666. [PMID: 29056909 PMCID: PMC5635435 DOI: 10.3389/fphar.2017.00666] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2017] [Accepted: 09/06/2017] [Indexed: 12/18/2022] Open
Abstract
Pharmacological studies indicate that dopamine D1-like receptors (D1 and D5) are critically involved in cognitive function. However, the lack of pharmacological ligands selective for either the D1 or D5 receptors has made it difficult to determine the unique contributions of the D1-like family members. To circumvent these pharmacological limitations, we used D5 receptor homozygous (-/-) and heterozygous (+/-) knockout mice, to identify the specific role of this receptor in higher order cognitive functions. We identified a novel role for D5 receptors in the regulation of spatial working memory and temporal order memory function. The D5 mutant mice acquired a discrete paired-trial variable-delay T-maze task at normal rates. However, both [Formula: see text] and [Formula: see text] mice exhibited impaired performance compared to [Formula: see text] littermates when a higher burden on working memory faculties was imposed. In a temporal order object recognition task, [Formula: see text] exhibited significant memory deficits. No D5-dependent differences in locomotor functions and interest in exploring objects were evident. Molecular biomarkers of dopaminergic functions within the prefrontal cortex (PFC) revealed a selective gene-dose effect on Akt phosphorylation at Ser473 with increased levels in [Formula: see text] knockout mice. A trend toward reduced levels in CaMKKbeta brain-specific band (64 kDa) in [Formula: see text] compared to [Formula: see text] was also evident. These findings highlight a previously unidentified role for D5 receptors in working memory function and associated molecular signatures within the PFC.
Collapse
Affiliation(s)
- Gregory V Carr
- Lieber Institute for Brain Development, Baltimore, MD, United States.,Department of Pharmacology and Molecular Sciences, Johns Hopkins School of Medicine, Baltimore, MD, United States.,Clinical Brain Disorders Branch, Genes, Cognition and Psychosis Program, National Institute of Mental Health, National Institutes of Health, Bethesda, MD, United States
| | - Federica Maltese
- Department of Neuroscience and Brain Technologies, Istituto Italiano di Tecnologia, Genova, Italy
| | - David R Sibley
- Molecular Neuropharmacology Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, United States
| | - Daniel R Weinberger
- Lieber Institute for Brain Development, Baltimore, MD, United States.,Clinical Brain Disorders Branch, Genes, Cognition and Psychosis Program, National Institute of Mental Health, National Institutes of Health, Bethesda, MD, United States.,Departments of Psychiatry and Behavioral Sciences, Neurology, and Neuroscience, The McKusick-Nathans Institute of Genetic Medicine, Johns Hopkins School of Medicine, Baltimore, MD, United States
| | - Francesco Papaleo
- Clinical Brain Disorders Branch, Genes, Cognition and Psychosis Program, National Institute of Mental Health, National Institutes of Health, Bethesda, MD, United States.,Department of Neuroscience and Brain Technologies, Istituto Italiano di Tecnologia, Genova, Italy
| |
Collapse
|
45
|
O'Tuathaigh CMP, Moran PM, Zhen XC, Waddington JL. Translating advances in the molecular basis of schizophrenia into novel cognitive treatment strategies. Br J Pharmacol 2017; 174:3173-3190. [PMID: 28667666 DOI: 10.1111/bph.13938] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2017] [Revised: 06/07/2017] [Accepted: 06/12/2017] [Indexed: 02/06/2023] Open
Abstract
The presence and severity of cognitive symptoms, including working memory, executive dysfunction and attentional impairment, contributes materially to functional impairment in schizophrenia. Cognitive symptoms have proved to be resistant to both first- and second-generation antipsychotic drugs. Efforts to develop a consensus set of cognitive domains that are both disrupted in schizophrenia and are amenable to cross-species validation (e.g. the National Institute of Mental Health Cognitive Neuroscience Treatment Research to Improve Cognition in Schizophrenia and Research Domain Criteria initiatives) are an important step towards standardization of outcome measures that can be used in preclinical testing of new drugs. While causative genetic mutations have not been identified, new technologies have identified novel genes as well as hitherto candidate genes previously implicated in the pathophysiology of schizophrenia and/or mechanisms of antipsychotic efficacy. This review comprises a selective summary of these developments, particularly phenotypic data arising from preclinical genetic models for cognitive dysfunction in schizophrenia, with the aim of indicating potential new directions for pro-cognitive therapeutics. Linked Articles This article is part of a themed section on Pharmacology of Cognition: a Panacea for Neuropsychiatric Disease? To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v174.19/issuetoc.
Collapse
Affiliation(s)
- Colm M P O'Tuathaigh
- School of Medicine, University College Cork, Brookfield Health Sciences Complex, Cork, Ireland
| | - Paula M Moran
- School of Psychology, University of Nottingham, Nottingham, UK
| | - Xuechu C Zhen
- Jiangsu Key Laboratory of Translational Research & Therapy for Neuropsychiatric Disorders and Department of Pharmacology, College of Pharmaceutical Sciences, Soochow University, Suzhou, China
| | - John L Waddington
- Jiangsu Key Laboratory of Translational Research & Therapy for Neuropsychiatric Disorders and Department of Pharmacology, College of Pharmaceutical Sciences, Soochow University, Suzhou, China.,Molecular and Cellular Therapeutics, Royal College of Surgeons in Ireland, Dublin 2, Ireland
| |
Collapse
|
46
|
Szlachta M, Pabian P, Kuśmider M, Solich J, Kolasa M, Żurawek D, Dziedzicka-Wasylewska M, Faron-Górecka A. Effect of clozapine on ketamine-induced deficits in attentional set shift task in mice. Psychopharmacology (Berl) 2017; 234:2103-2112. [PMID: 28405711 PMCID: PMC5486929 DOI: 10.1007/s00213-017-4613-x] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/16/2016] [Accepted: 03/24/2017] [Indexed: 01/26/2023]
Abstract
RATIONALE Clozapine (CLZ) is an effective treatment for schizophrenia, producing improvements in both negative symptoms and cognitive impairments. Cognitive impairments can be modelled in animals by ketamine (KET) and assessed using the attentional set-shift task (ASST). OBJECTIVE Our first aim was to determine whether CLZ improves cognitive function and reverses KET-induced cognitive impairments using the ASST. Our second aim was to assess dose dependency of these effects. RESULTS Our findings demonstrate that acute as well as sub-chronic administration of KET cause cognitive deficits observed as increase in number of trails and errors to reach the criterion in the EDS phase. CLZ 0.3 mg/kg reversed the effects of both acute and sub-chronic KET, with no effects on locomotor activity. However, clozapine's effect after sub-chronic administration of dose 0.3 mg/kg was not as explicit as in the case of acute treatment. Moreover, administration of 1 mg/kg CLZ to KET-treated mice induced or enhanced deficits in the extra-dimensional shift phase compared to 1 mg/kg CLZ administration to mice not receiving KET. Locomotor activity test showed sedation effects of CLZ 1 mg/kg after acute treatment; therefore, effect of CLZ 1 mg/kg on KET-induced cognitive deficits was not evaluated in the attentional set-shift task (ASST) test. CONCLUSIONS The present findings support dose-dependent effects of CLZ to reverse KET-induced cognitive deficits. The observed dose dependency may be mediated by activation of different receptors, including monomers and/or heterodimers.
Collapse
Affiliation(s)
- M Szlachta
- Department of Pharmacology, Institute of Pharmacology Polish Academy of Sciences, Smętna Street 12, 31-343, Kraków, Poland
| | - P Pabian
- Department of Pharmacology, Institute of Pharmacology Polish Academy of Sciences, Smętna Street 12, 31-343, Kraków, Poland
| | - M Kuśmider
- Department of Pharmacology, Institute of Pharmacology Polish Academy of Sciences, Smętna Street 12, 31-343, Kraków, Poland
| | - J Solich
- Department of Pharmacology, Institute of Pharmacology Polish Academy of Sciences, Smętna Street 12, 31-343, Kraków, Poland
| | - M Kolasa
- Department of Pharmacology, Institute of Pharmacology Polish Academy of Sciences, Smętna Street 12, 31-343, Kraków, Poland
| | - D Żurawek
- Department of Pharmacology, Institute of Pharmacology Polish Academy of Sciences, Smętna Street 12, 31-343, Kraków, Poland
| | - M Dziedzicka-Wasylewska
- Department of Pharmacology, Institute of Pharmacology Polish Academy of Sciences, Smętna Street 12, 31-343, Kraków, Poland
| | - A Faron-Górecka
- Department of Pharmacology, Institute of Pharmacology Polish Academy of Sciences, Smętna Street 12, 31-343, Kraków, Poland.
| |
Collapse
|
47
|
Behavioral, Neurophysiological, and Synaptic Impairment in a Transgenic Neuregulin1 (NRG1-IV) Murine Schizophrenia Model. J Neurosci 2017; 36:4859-75. [PMID: 27122041 DOI: 10.1523/jneurosci.4632-15.2016] [Citation(s) in RCA: 49] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2015] [Accepted: 03/22/2016] [Indexed: 11/21/2022] Open
Abstract
UNLABELLED Schizophrenia is a chronic, disabling neuropsychiatric disorder with complex genetic origins. The development of strategies for genome manipulation in rodents provides a platform for understanding the pathogenic role of genes and for testing novel therapeutic agents. Neuregulin 1 (NRG1), a critical developmental neurotrophin, is associated with schizophrenia. The NRG1 gene undergoes extensive alternative splicing and, to date, little is known about the neurobiology of a novel NRG1 isoform, NRG1-IV, which is increased in the brains of individuals with schizophrenia and associated with genetic risk variation. Here, we developed a transgenic mouse model (NRG1-IV/NSE-tTA) in which human NRG1-IV is selectively overexpressed in a neuronal specific manner. Using a combination of molecular, biochemical, electrophysiological, and behavioral analyses, we demonstrate that NRG1-IV/NSE-tTA mice exhibit abnormal behaviors relevant to schizophrenia, including impaired sensorimotor gating, discrimination memory, and social behaviors. These neurobehavioral phenotypes are accompanied by increases in cortical expression of the NRG1 receptor, ErbB4 and the downstream signaling target, PIK3-p110δ, along with disrupted dendritic development, synaptic pathology, and altered prefrontal cortical excitatory-inhibitory balance. Pharmacological inhibition of p110δ reversed sensorimotor gating and cognitive deficits. These data demonstrate a novel role for NRG1-IV in learning, memory, and neural circuit formation and a potential neurobiological mechanism for schizophrenia risk; show that deficits are pharmacologically reversible in adulthood; and further highlight p110δ as a target for antipsychotic drug development. SIGNIFICANCE STATEMENT Schizophrenia is a disabling psychiatric disorder with neurodevelopmental origins. Genes that increase risk for schizophrenia have been identified. Understanding how these genes affect brain development and function is necessary. This work is the first report of a newly generated humanized transgenic mouse model engineered to express human NRG1-IV, an isoform of the NRG1 (Neuregulin 1) gene that is increased in the brains of patients with schizophrenia in association with genetic risk. Using behavioral neuroscience, molecular biology, electrophysiology, and pharmacology, we identify a role for NRG1-IV in learning, memory, and cognition and determine that this relates to brain excitatory-inhibitory balance and changes in ErbB4/PI3K/AKT signaling. Moreover, the study further highlights the potential of targeting the PI3K pathway for the treatment of schizophrenia.
Collapse
|
48
|
PKBγ/AKT3 loss-of-function causes learning and memory deficits and deregulation of AKT/mTORC2 signaling: Relevance for schizophrenia. PLoS One 2017; 12:e0175993. [PMID: 28467426 PMCID: PMC5414975 DOI: 10.1371/journal.pone.0175993] [Citation(s) in RCA: 44] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2016] [Accepted: 04/04/2017] [Indexed: 12/17/2022] Open
Abstract
Psychiatric genetic studies have identified genome-wide significant loci for schizophrenia. The AKT3/1q44 locus is a principal risk region and gene-network analyses identify AKT3 polymorphisms as a constituent of several neurobiological pathways relevant to psychiatric risk; the neurobiological mechanisms remain unknown. AKT3 shows prenatal enrichment during human neocortical development and recurrent copy number variations involving the 1q43-44 locus are associated with cortical malformations and intellectual disability, implicating an essential role in early brain development. Here, we investigated the role of AKT3 as it relates to aspects of learning and memory and behavioral function, relevant to schizophrenia and cognitive disability, utilizing a novel murine model of Akt3 genetic deficiency. Akt3 heterozygous (Akt3-/+) or null mice (Akt3-/-) were assessed in a comprehensive test battery. Brain biochemical studies were conducted to assess the impact of Akt3 deficiency on cortical Akt/mTOR signaling. Akt3-/+ and Akt3-/- mice exhibited selective deficits of temporal order discrimination and spatial memory, tasks critically dependent on intact prefrontal-hippocampal circuitry, but showed normal prepulse inhibition, fear conditioned learning, memory for novel objects and social function. Akt3 loss-of-function, reduced brain size and dramatically impaired cortical Akt Ser473 activation in an allele-dose dependent manner. Such changes were observed in the absence of altered Akt1 or Akt2 protein expression. Concomitant reduction of the mTORC2 complex proteins, Rictor and Sin1 identifies a potential mechanism. Our findings provide novel insight into the neurodevelopmental role of Akt3, identify a non-redundant role for Akt3 in the development of prefrontal cortical-mediated cognitive function and show that Akt3 is potentially the dominant regulator of AKT/mTOR signaling in brain.
Collapse
|
49
|
Klaus K, Butler K, Durrant SJ, Ali M, Inglehearn CF, Hodgson TL, Gutierrez H, Pennington K. The effect of COMT Val158Met and DRD2 C957T polymorphisms on executive function and the impact of early life stress. Brain Behav 2017; 7:e00695. [PMID: 28523234 PMCID: PMC5434197 DOI: 10.1002/brb3.695] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/05/2016] [Revised: 02/21/2017] [Accepted: 02/22/2017] [Indexed: 12/15/2022] Open
Abstract
INTRODUCTION Previous research has indicated that variation in genes encoding catechol-O-methyltransferase (COMT) and dopamine receptor D2 (DRD2) may influence cognitive function and that this may confer vulnerability to the development of mental health disorders such as schizophrenia. However, increasing evidence suggests environmental factors such as early life stress may interact with genetic variants in affecting these cognitive outcomes. This study investigated the effect of COMT Val158Met and DRD2 C957T polymorphisms on executive function and the impact of early life stress in healthy adults. METHODS One hundred and twenty-two healthy adult males (mean age 35.2 years, range 21-63) were enrolled in the study. Cognitive function was assessed using Cambridge Neuropsychological Test Automated Battery and early life stress was assessed using the Childhood Traumatic Events Scale (Pennebaker & Susman, 1988). RESULTS DRD2 C957T was significantly associated with executive function, with CC homozygotes having significantly reduced performance in spatial working memory and spatial planning. A significant genotype-trauma interaction was found in Rapid Visual Information Processing test, a measure of sustained attention, with CC carriers who had experienced early life stress exhibiting impaired performance compared to the CC carriers without early life stressful experiences. There were no significant findings for COMT Val158Met. CONCLUSIONS This study supports previous findings that DRD2 C957T significantly affects performance on executive function related tasks in healthy individuals and shows for the first time that some of these effects may be mediated through the impact of childhood traumatic events. Future work should aim to clarify further the effect of stress on neuronal systems that are known to be vulnerable in mental health disorders and more specifically what the impact of this might be on cognitive function.
Collapse
Affiliation(s)
- Kristel Klaus
- School of Psychology University of Lincoln Lincoln UK
| | - Kevin Butler
- School of Psychology University of Lincoln Lincoln UK
| | | | - Manir Ali
- Section of Ophthalmology & Neuroscience Leeds Institute of Biomedical Sciences St James' Hospital University of Leeds Leeds UK
| | - Chris F Inglehearn
- Section of Ophthalmology & Neuroscience Leeds Institute of Biomedical Sciences St James' Hospital University of Leeds Leeds UK
| | | | | | | |
Collapse
|
50
|
Adolescence is the starting point of sex-dichotomous COMT genetic effects. Transl Psychiatry 2017; 7:e1141. [PMID: 28556830 PMCID: PMC5584523 DOI: 10.1038/tp.2017.109] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/12/2017] [Revised: 02/28/2017] [Accepted: 04/11/2017] [Indexed: 01/09/2023] Open
Abstract
The catechol-o-methyltransferase (COMT) genetic variations produce pleiotropic behavioral/neuroanatomical effects. Some of these effects may vary among sexes. However, the developmental trajectories of COMT-by-sex interactions are unclear. Here we found that extreme COMT reduction, in both humans (22q11.2 deletion syndrome COMT Met) and mice (COMT-/-), was associated to cortical thinning only after puberty and only in females. Molecular biomarkers, such as tyrosine hydroxylase, Akt and neuronal/cellular counting, confirmed that COMT-by-sex divergent effects started to appear at the cortical level during puberty. These biochemical differences were absent in infancy. Finally, developmental cognitive assessment in 22q11DS and COMT knockout mice established that COMT-by-sex-dichotomous effects in executive functions were already apparent in adolescence. These findings uncover that genetic variations severely reducing COMT result in detrimental cortical and cognitive development selectively in females after their sexual maturity. This highlights the importance of taking into account the combined effect of genetics, sex and developmental stage.
Collapse
|