1
|
Abedini SS, Akhavantabasi S, Liang Y, Heng JIT, Alizadehsani R, Dehzangi I, Bauer DC, Alinejad-Rokny H. A critical review of the impact of candidate copy number variants on autism spectrum disorder. MUTATION RESEARCH. REVIEWS IN MUTATION RESEARCH 2024; 794:108509. [PMID: 38977176 DOI: 10.1016/j.mrrev.2024.108509] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/04/2023] [Revised: 04/14/2024] [Accepted: 07/02/2024] [Indexed: 07/10/2024]
Abstract
Autism spectrum disorder (ASD) is a complex neurodevelopmental disorder (NDD) influenced by genetic, epigenetic, and environmental factors. Recent advancements in genomic analysis have shed light on numerous genes associated with ASD, highlighting the significant role of both common and rare genetic mutations, as well as copy number variations (CNVs), single nucleotide polymorphisms (SNPs) and unique de novo variants. These genetic variations disrupt neurodevelopmental pathways, contributing to the disorder's complexity. Notably, CNVs are present in 10 %-20 % of individuals with autism, with 3 %-7 % detectable through cytogenetic methods. While the role of submicroscopic CNVs in ASD has been recently studied, their association with genomic loci and genes has not been thoroughly explored. In this review, we focus on 47 CNV regions linked to ASD, encompassing 1632 genes, including protein-coding genes and long non-coding RNAs (lncRNAs), of which 659 show significant brain expression. Using a list of ASD-associated genes from SFARI, we detect 17 regions harboring at least one known ASD-related protein-coding gene. Of the remaining 30 regions, we identify 24 regions containing at least one protein-coding gene with brain-enriched expression and a nervous system phenotype in mouse mutants, and one lncRNA with both brain-enriched expression and upregulation in iPSC to neuron differentiation. This review not only expands our understanding of the genetic diversity associated with ASD but also underscores the potential of lncRNAs in contributing to its etiology. Additionally, the discovered CNVs will be a valuable resource for future diagnostic, therapeutic, and research endeavors aimed at prioritizing genetic variations in ASD.
Collapse
Affiliation(s)
- Seyedeh Sedigheh Abedini
- UNSW BioMedical Machine Learning Lab (BML), The Graduate School of Biomedical Engineering, UNSW Sydney, Sydney, NSW 2052, Australia; School of Biotechnology & Biomolecular Sciences, UNSW Sydney, Sydney, NSW 2052, Australia
| | - Shiva Akhavantabasi
- Department of Molecular Biology and Genetics, Yeni Yuzyil University, Istanbul, Turkey; Ghiaseddin Jamshid Kashani University, Andisheh University Town, Danesh Blvd, 3441356611, Abyek, Qazvin, Iran
| | - Yuheng Liang
- UNSW BioMedical Machine Learning Lab (BML), The Graduate School of Biomedical Engineering, UNSW Sydney, Sydney, NSW 2052, Australia
| | - Julian Ik-Tsen Heng
- Curtin Health Innovation Research Institute, Curtin University, Bentley 6845, Australia
| | - Roohallah Alizadehsani
- Institute for Intelligent Systems Research and Innovation (IISRI), Deakin University, Victoria, Australia
| | - Iman Dehzangi
- Center for Computational and Integrative Biology, Rutgers University, Camden, NJ 08102, USA; Department of Computer Science, Rutgers University, Camden, NJ 08102, USA
| | - Denis C Bauer
- Transformational Bioinformatics, Commonwealth Scientific and Industrial Research Organisation (CSIRO), Sydney, Australia; Applied BioSciences, Faculty of Science and Engineering, Macquarie University, Macquarie Park, Australia
| | - Hamid Alinejad-Rokny
- UNSW BioMedical Machine Learning Lab (BML), The Graduate School of Biomedical Engineering, UNSW Sydney, Sydney, NSW 2052, Australia; Tyree Institute of Health Engineering (IHealthE), UNSW Sydney, Sydney, NSW 2052, Australia.
| |
Collapse
|
2
|
Jiang J, Wang D, Jiang Y, Yang X, Sun R, Chang J, Zhu W, Yao P, Song K, Chang S, Wang H, Zhou L, Zhang XS, Li H, Li N. The gut metabolite indole-3-propionic acid activates ERK1 to restore social function and hippocampal inhibitory synaptic transmission in a 16p11.2 microdeletion mouse model. MICROBIOME 2024; 12:66. [PMID: 38549163 PMCID: PMC10976717 DOI: 10.1186/s40168-024-01755-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 04/04/2023] [Accepted: 01/04/2024] [Indexed: 04/02/2024]
Abstract
BACKGROUND Microdeletion of the human chromosomal region 16p11.2 (16p11.2+ / - ) is a prevalent genetic factor associated with autism spectrum disorder (ASD) and other neurodevelopmental disorders. However its pathogenic mechanism remains unclear, and effective treatments for 16p11.2+ / - syndrome are lacking. Emerging evidence suggests that the gut microbiota and its metabolites are inextricably linked to host behavior through the gut-brain axis and are therefore implicated in ASD development. Despite this, the functional roles of microbial metabolites in the context of 16p11.2+ / - are yet to be elucidated. This study aims to investigate the therapeutic potential of indole-3-propionic acid (IPA), a gut microbiota metabolite, in addressing behavioral and neural deficits associated with 16p11.2+ / - , as well as the underlying molecular mechanisms. RESULTS Mice with the 16p11.2+ / - showed dysbiosis of the gut microbiota and a significant decrease in IPA levels in feces and blood circulation. Further, these mice exhibited significant social and cognitive memory impairments, along with hyperactivation of hippocampal dentate gyrus neurons and reduced inhibitory synaptic transmission in this region. However, oral administration of IPA effectively mitigated the histological and electrophysiological alterations, thereby ameliorating the social and cognitive deficits of the mice. Remarkably, IPA treatment significantly increased the phosphorylation level of ERK1, a protein encoded by the Mapk3 gene in the 16p11.2 region, without affecting the transcription and translation of the Mapk3 gene. CONCLUSIONS Our study reveals that 16p11.2+ / - leads to a decline in gut metabolite IPA levels; however, IPA supplementation notably reverses the behavioral and neural phenotypes of 16p11.2+ / - mice. These findings provide new insights into the critical role of gut microbial metabolites in ASD pathogenesis and present a promising treatment strategy for social and cognitive memory deficit disorders, such as 16p11.2 microdeletion syndrome. Video Abstract.
Collapse
Affiliation(s)
- Jian Jiang
- Tomas Lindahl Nobel Laureate Laboratory, The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen, China
| | - Dilong Wang
- Tomas Lindahl Nobel Laureate Laboratory, The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen, China
- Department of Pediatrics, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Youheng Jiang
- Tomas Lindahl Nobel Laureate Laboratory, The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen, China
| | - Xiuyan Yang
- Tomas Lindahl Nobel Laureate Laboratory, The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen, China
| | - Runfeng Sun
- Institute of Molecular Physiology, Shenzhen Bay Laboratory, Shenzhen, China
| | - Jinlong Chang
- Tomas Lindahl Nobel Laureate Laboratory, The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen, China
| | - Wenhui Zhu
- Tomas Lindahl Nobel Laureate Laboratory, The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen, China
| | - Peijia Yao
- Tomas Lindahl Nobel Laureate Laboratory, The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen, China
| | - Kun Song
- Brain Research Centre, Department of Biology, School of Life Sciences, Southern University of Science and Technology, Shenzhen, China
| | - Shuwen Chang
- The Brain Cognition and Brain Disease Institute (BCBDI), Shenzhen-Hong Kong Institute of Brain Science Shenzhen Fundamental Research Institutions, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Hong Wang
- The Brain Cognition and Brain Disease Institute (BCBDI), Shenzhen-Hong Kong Institute of Brain Science Shenzhen Fundamental Research Institutions, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Lei Zhou
- Institute of Molecular Physiology, Shenzhen Bay Laboratory, Shenzhen, China
| | - Xue-Song Zhang
- Center for Advanced Biotechnology and Medicine, Rutgers University, Piscataway, NJ, USA.
| | - Huiliang Li
- Wolfson Institute for Biomedical Research, Division of Medicine, Faculty of Medical Sciences, University College London, London, UK.
| | - Ningning Li
- Tomas Lindahl Nobel Laureate Laboratory, The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen, China.
- China-UK Institute for Frontier Science, Shenzhen, China.
- Department of Anesthesiology, The Afliated Hospital of Youjiang Medical University for Nationalities, Baise, China.
| |
Collapse
|
3
|
Ioannidis V, Pandey R, Bauer HF, Schön M, Bockmann J, Boeckers TM, Lutz AK. Disrupted extracellular matrix and cell cycle genes in autism-associated Shank3 deficiency are targeted by lithium. Mol Psychiatry 2024; 29:704-717. [PMID: 38123724 PMCID: PMC11153165 DOI: 10.1038/s41380-023-02362-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Revised: 11/20/2023] [Accepted: 12/01/2023] [Indexed: 12/23/2023]
Abstract
The Shank3 gene encodes the major postsynaptic scaffolding protein SHANK3. Its mutation causes a syndromic form of autism spectrum disorder (ASD): Phelan-McDermid Syndrome (PMDS). It is characterized by global developmental delay, intellectual disorders (ID), ASD behavior, affective symptoms, as well as extra-cerebral symptoms. Although Shank3 deficiency causes a variety of molecular alterations, they do not suffice to explain all clinical aspects of this heterogenic syndrome. Since global gene expression alterations in Shank3 deficiency remain inadequately studied, we explored the transcriptome in vitro in primary hippocampal cells from Shank3∆11(-/-) mice, under control and lithium (Li) treatment conditions, and confirmed the findings in vivo. The Shank3∆11(-/-) genotype affected the overall transcriptome. Remarkably, extracellular matrix (ECM) and cell cycle transcriptional programs were disrupted. Accordingly, in the hippocampi of adolescent Shank3∆11(-/-) mice we found proteins of the collagen family and core cell cycle proteins downregulated. In vitro Li treatment of Shank3∆11(-/-) cells had a rescue-like effect on the ECM and cell cycle gene sets. Reversed ECM gene sets were part of a network, regulated by common transcription factors (TF) such as cAMP responsive element binding protein 1 (CREB1) and β-Catenin (CTNNB1), which are known downstream effectors of synaptic activity and targets of Li. These TFs were less abundant and/or hypo-phosphorylated in hippocampi of Shank3∆11(-/-) mice and could be rescued with Li in vitro and in vivo. Our investigations suggest the ECM compartment and cell cycle genes as new players in the pathophysiology of Shank3 deficiency, and imply involvement of transcriptional regulators, which can be modulated by Li. This work supports Li as potential drug in the management of PMDS symptoms, where a Phase III study is ongoing.
Collapse
Affiliation(s)
- Valentin Ioannidis
- Institute for Anatomy and Cell Biology, Ulm University, 89081, Ulm, Germany
| | - Rakshita Pandey
- Institute for Anatomy and Cell Biology, Ulm University, 89081, Ulm, Germany
- International Graduate School in Molecular Medicine Ulm, Ulm University, Ulm, Germany
| | - Helen Friedericke Bauer
- Institute for Anatomy and Cell Biology, Ulm University, 89081, Ulm, Germany
- International Graduate School in Molecular Medicine Ulm, Ulm University, Ulm, Germany
| | - Michael Schön
- Institute for Anatomy and Cell Biology, Ulm University, 89081, Ulm, Germany
| | - Jürgen Bockmann
- Institute for Anatomy and Cell Biology, Ulm University, 89081, Ulm, Germany
| | - Tobias M Boeckers
- Institute for Anatomy and Cell Biology, Ulm University, 89081, Ulm, Germany
- German Center for Neurodegenerative Diseases (DZNE), Ulm site, 89081, Ulm, Germany
| | - Anne-Kathrin Lutz
- Institute for Anatomy and Cell Biology, Ulm University, 89081, Ulm, Germany.
| |
Collapse
|
4
|
László K, Vörös D, Correia P, Fazekas CL, Török B, Plangár I, Zelena D. Vasopressin as Possible Treatment Option in Autism Spectrum Disorder. Biomedicines 2023; 11:2603. [PMID: 37892977 PMCID: PMC10603886 DOI: 10.3390/biomedicines11102603] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2023] [Revised: 09/13/2023] [Accepted: 09/21/2023] [Indexed: 10/29/2023] Open
Abstract
Autism spectrum disorder (ASD) is rather common, presenting with prevalent early problems in social communication and accompanied by repetitive behavior. As vasopressin was implicated not only in salt-water homeostasis and stress-axis regulation, but also in social behavior, its role in the development of ASD might be suggested. In this review, we summarized a wide range of problems associated with ASD to which vasopressin might contribute, from social skills to communication, motor function problems, autonomous nervous system alterations as well as sleep disturbances, and altered sensory information processing. Beside functional connections between vasopressin and ASD, we draw attention to the anatomical background, highlighting several brain areas, including the paraventricular nucleus of the hypothalamus, medial preoptic area, lateral septum, bed nucleus of stria terminalis, amygdala, hippocampus, olfactory bulb and even the cerebellum, either producing vasopressin or containing vasopressinergic receptors (presumably V1a). Sex differences in the vasopressinergic system might underline the male prevalence of ASD. Moreover, vasopressin might contribute to the effectiveness of available off-label therapies as well as serve as a possible target for intervention. In this sense, vasopressin, but paradoxically also V1a receptor antagonist, were found to be effective in some clinical trials. We concluded that although vasopressin might be an effective candidate for ASD treatment, we might assume that only a subgroup (e.g., with stress-axis disturbances), a certain sex (most probably males) and a certain brain area (targeting by means of virus vectors) would benefit from this therapy.
Collapse
Affiliation(s)
- Kristóf László
- Institute of Physiology, Medical School, University of Pécs, 7624 Pecs, Hungary; (K.L.); (D.V.); (P.C.); (C.L.F.); (B.T.); (I.P.)
- Center of Neuroscience, University of Pécs, 7624 Pecs, Hungary
- Szentágothai Research Center, University of Pécs, 7624 Pecs, Hungary
| | - Dávid Vörös
- Institute of Physiology, Medical School, University of Pécs, 7624 Pecs, Hungary; (K.L.); (D.V.); (P.C.); (C.L.F.); (B.T.); (I.P.)
- Center of Neuroscience, University of Pécs, 7624 Pecs, Hungary
- Szentágothai Research Center, University of Pécs, 7624 Pecs, Hungary
| | - Pedro Correia
- Institute of Physiology, Medical School, University of Pécs, 7624 Pecs, Hungary; (K.L.); (D.V.); (P.C.); (C.L.F.); (B.T.); (I.P.)
- Center of Neuroscience, University of Pécs, 7624 Pecs, Hungary
- Szentágothai Research Center, University of Pécs, 7624 Pecs, Hungary
- Hungarian Research Network, Institute of Experimental Medicine, 1083 Budapest, Hungary
| | - Csilla Lea Fazekas
- Institute of Physiology, Medical School, University of Pécs, 7624 Pecs, Hungary; (K.L.); (D.V.); (P.C.); (C.L.F.); (B.T.); (I.P.)
- Center of Neuroscience, University of Pécs, 7624 Pecs, Hungary
- Szentágothai Research Center, University of Pécs, 7624 Pecs, Hungary
- Hungarian Research Network, Institute of Experimental Medicine, 1083 Budapest, Hungary
| | - Bibiána Török
- Institute of Physiology, Medical School, University of Pécs, 7624 Pecs, Hungary; (K.L.); (D.V.); (P.C.); (C.L.F.); (B.T.); (I.P.)
- Center of Neuroscience, University of Pécs, 7624 Pecs, Hungary
- Szentágothai Research Center, University of Pécs, 7624 Pecs, Hungary
- Hungarian Research Network, Institute of Experimental Medicine, 1083 Budapest, Hungary
| | - Imola Plangár
- Institute of Physiology, Medical School, University of Pécs, 7624 Pecs, Hungary; (K.L.); (D.V.); (P.C.); (C.L.F.); (B.T.); (I.P.)
- Center of Neuroscience, University of Pécs, 7624 Pecs, Hungary
- Szentágothai Research Center, University of Pécs, 7624 Pecs, Hungary
| | - Dóra Zelena
- Institute of Physiology, Medical School, University of Pécs, 7624 Pecs, Hungary; (K.L.); (D.V.); (P.C.); (C.L.F.); (B.T.); (I.P.)
- Center of Neuroscience, University of Pécs, 7624 Pecs, Hungary
- Szentágothai Research Center, University of Pécs, 7624 Pecs, Hungary
- Hungarian Research Network, Institute of Experimental Medicine, 1083 Budapest, Hungary
| |
Collapse
|
5
|
Subasi Turgut F, Karadag M, Taysi S, Hangül Z, Gokcen C. NRF2, KEAP1 and GSK-3 levels in autism spectrum disorder: a case control study. INTERNATIONAL JOURNAL OF DEVELOPMENTAL DISABILITIES 2023; 70:1441-1451. [PMID: 39713511 PMCID: PMC11660297 DOI: 10.1080/20473869.2023.2185959] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/01/2022] [Revised: 02/27/2023] [Accepted: 02/27/2023] [Indexed: 12/24/2024]
Abstract
Recent studies show that oxidative stress has an important role in the etiology of autism. In our study, Nrf2, which is the main regulator of cellular antioxidant response, and Keap1 and Gsk-3β, which are the main proteins that regulate this pathway, were compared between children with autism and healthy controls. To the best of our knowledge, our study is the first in which Nrf2, Keap1 and Gsk-3β levels were evaluated together in children with ASD. In our study, Nrf2 level was found to be lower and Keap1 level higher in children with autism. Although GSK-3β is increased in many psychiatric and neurodegenerative diseases, it was found to be low in the autistic group in accordance with the literature. In conclusion, considering the versatile modulation of the Nrf2 pathway, this article does not provide any mechanistic insight into the pathway, but it suggests that Nrf2, Keap1 and Gsk-3β, which have central roles in oxidative stress, may play a role in the pathophysiology of autism.
Collapse
Affiliation(s)
| | | | | | - Zehra Hangül
- Gaziantep Universitesi Tip Fakultesi, Gaziantep, Turkey
| | - Cem Gokcen
- Gaziantep Universitesi Tip Fakultesi, Gaziantep, Turkey
| |
Collapse
|
6
|
Zawadzka A, Cieślik M, Adamczyk A. The Role of Maternal Immune Activation in the Pathogenesis of Autism: A Review of the Evidence, Proposed Mechanisms and Implications for Treatment. Int J Mol Sci 2021; 22:ijms222111516. [PMID: 34768946 PMCID: PMC8584025 DOI: 10.3390/ijms222111516] [Citation(s) in RCA: 75] [Impact Index Per Article: 18.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2021] [Revised: 10/21/2021] [Accepted: 10/22/2021] [Indexed: 12/15/2022] Open
Abstract
Autism spectrum disorder (ASD) is a neurodevelopmental disease that is characterized by a deficit in social interactions and communication, as well as repetitive and restrictive behaviors. Increasing lines of evidence suggest an important role for immune dysregulation and/or inflammation in the development of ASD. Recently, a relationship between inflammation, oxidative stress, and mitochondrial dysfunction has been reported in the brain tissue of individuals with ASD. Some recent studies have also reported oxidative stress and mitochondrial abnormalities in animal models of maternal immune activation (MIA). This review is focused on the hypothesis that MIA induces microglial activation, oxidative stress, and mitochondrial dysfunction, a deleterious trio in the brain that can lead to neuroinflammation and neurodevelopmental pathologies in offspring. Infection during pregnancy activates the mother’s immune system to release proinflammatory cytokines, such as IL-6, TNF-α, and others. Furthermore, these cytokines can directly cross the placenta and enter the fetal circulation, or activate resident immune cells, resulting in an increased production of proinflammatory cytokines, including IL-6. Proinflammatory cytokines that cross the blood–brain barrier (BBB) may initiate a neuroinflammation cascade, starting with the activation of the microglia. Inflammatory processes induce oxidative stress and mitochondrial dysfunction that, in turn, may exacerbate oxidative stress in a self-perpetuating vicious cycle that can lead to downstream abnormalities in brain development and behavior.
Collapse
Affiliation(s)
| | - Magdalena Cieślik
- Correspondence: (M.C.); (A.A.); Tel.: +48-22-6086420 (M.C.); +48-22-6086572 (A.A.)
| | - Agata Adamczyk
- Correspondence: (M.C.); (A.A.); Tel.: +48-22-6086420 (M.C.); +48-22-6086572 (A.A.)
| |
Collapse
|
7
|
Characterizing Sleep Problems in 16p11.2 Deletion and Duplication. J Autism Dev Disord 2021; 53:1462-1475. [PMID: 34633643 DOI: 10.1007/s10803-021-05311-2] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/24/2021] [Indexed: 10/20/2022]
Abstract
Studies of 16p11.2 copy number variants (CNVs) provide an avenue to identify mechanisms of impairment and develop targeted treatments for individuals with neurodevelopmental disorders. 16p11.2 deletion and duplication phenotypes are currently being ascertained; however, sleep disturbances are minimally described. In this study, we examine sleep disturbance in a well-characterized national sample of 16p11.2 CNVs, the Simons Foundation Autism Research Initiative (SFARI) database of youth and adults (n = 692). Factor analyses and multilevel models of derived sleep questionnaires for youth (n = 345) and adults (n = 347) indicate that 16p11.2 carriers show elevated sleep disturbance relative to community controls. Non-carrier family members also show elevated sleep disturbance. However, sleep duration does not differ between carriers and controls. Further studies of sleep in 16p11.2 are needed.
Collapse
|
8
|
Tiwari A, Rahi S, Mehan S. Elucidation of Abnormal Extracellular Regulated Kinase (ERK) Signaling and Associations with Syndromic and Non-syndromic Autism. Curr Drug Targets 2021; 22:1071-1086. [PMID: 33081671 DOI: 10.2174/1389450121666201020155010] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2020] [Revised: 09/21/2020] [Accepted: 09/26/2020] [Indexed: 11/22/2022]
Abstract
Autism is a highly inherited and extremely complex disorder in which results from various cases indicate chromosome anomalies, unusual single-gene mutations, and multiplicative effects of particular gene variants, characterized primarily by impaired speech and social interaction and restricted behavior. The precise etiology of Autism Spectrum Disorder (ASD) is currently unclear. The extracellular signal-regulated kinase (ERK) signaling mechanism affects neurogenesis and neuronal plasticity during the development of the central nervous mechanism. In this regard, the pathway of ERK has recently gained significant interest in the pathogenesis of ASD. The mutation occurs in a few ERK components. Besides, the ERK pathway dysfunction lies in the upstream of modified translation and contributes to synapse pathology in syndromic types of autism. In this review, we highlight the ERK pathway as a target for neurodevelopmental disorder autism. In addition, we summarize the regulation of the ERK pathway with ERK inhibitors in neurological disorders. In conclusion, a better understanding of the ERK signaling pathway provides a range of therapeutic options for autism spectrum disorder.
Collapse
Affiliation(s)
- Aarti Tiwari
- Neuropharmacology Division, Department of Pharmacology, ISF College of Pharmacy, Moga, Punjab, India
| | - Saloni Rahi
- Neuropharmacology Division, Department of Pharmacology, ISF College of Pharmacy, Moga, Punjab, India
| | - Sidharth Mehan
- Neuropharmacology Division, Department of Pharmacology, ISF College of Pharmacy, Moga, Punjab, India
| |
Collapse
|
9
|
Boolean Networks: A Primer. SYSTEMS MEDICINE 2021. [DOI: 10.1016/b978-0-12-801238-3.11518-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
|
10
|
Farr CV, El-Kasaby A, Freissmuth M, Sucic S. The Creatine Transporter Unfolded: A Knotty Premise in the Cerebral Creatine Deficiency Syndrome. Front Synaptic Neurosci 2020; 12:588954. [PMID: 33192443 PMCID: PMC7644880 DOI: 10.3389/fnsyn.2020.588954] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2020] [Accepted: 09/18/2020] [Indexed: 12/16/2022] Open
Abstract
Creatine provides cells with high-energy phosphates for the rapid reconstitution of hydrolyzed adenosine triphosphate. The eponymous creatine transporter (CRT1/SLC6A8) belongs to a family of solute carrier 6 (SLC6) proteins. The key role of CRT1 is to translocate creatine across tissue barriers and into target cells, such as neurons and myocytes. Individuals harboring mutations in the coding sequence of the human CRT1 gene develop creatine transporter deficiency (CTD), one of the pivotal underlying causes of cerebral creatine deficiency syndrome. CTD encompasses an array of clinical manifestations, including severe intellectual disability, epilepsy, autism, development delay, and motor dysfunction. CTD is characterized by the absence of cerebral creatine, which implies an indispensable role for CRT1 in supplying the brain cells with creatine. CTD-associated variants dramatically reduce or abolish creatine transport activity by CRT1. Many of these are point mutations that are known to trigger folding defects, leading to the retention of encoded CRT1 proteins in the endoplasmic reticulum and precluding their delivery to the cell surface. Misfolding of several related SLC6 transporters also gives rise to detrimental pathologic conditions in people; e.g., mutations in the dopamine transporter induce infantile parkinsonism/dystonia, while mutations in the GABA transporter 1 cause treatment-resistant epilepsy. In some cases, folding defects are amenable to rescue by small molecules, known as pharmacological and chemical chaperones, which restore the cell surface expression and transport activity of the previously non-functional proteins. Insights from the recent molecular, animal and human case studies of CTD add toward our understanding of this complex disorder and reveal the wide-ranging effects elicited upon CRT1 dysfunction. This grants novel therapeutic prospects for the treatment of patients afflicted with CTD, e.g., modifying the creatine molecule to facilitate CRT1-independent entry into brain cells, or correcting folding-deficient and loss-of-function CTD variants using pharmacochaperones and/or allosteric modulators. The latter justifies a search for additional compounds with a capacity to correct mutation-specific defects.
Collapse
Affiliation(s)
| | | | | | - Sonja Sucic
- Institute of Pharmacology, Center of Physiology and Pharmacology, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
11
|
Lynch JF, Ferri SL, Angelakos C, Schoch H, Nickl-Jockschat T, Gonzalez A, O'Brien WT, Abel T. Comprehensive Behavioral Phenotyping of a 16p11.2 Del Mouse Model for Neurodevelopmental Disorders. Autism Res 2020; 13:1670-1684. [PMID: 32857907 DOI: 10.1002/aur.2357] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2020] [Revised: 05/07/2020] [Accepted: 05/18/2020] [Indexed: 01/24/2023]
Abstract
The microdeletion of copy number variant 16p11.2 is one of the most common genetic mutations associated with neurodevelopmental disorders, such as Autism Spectrum Disorders (ASDs). Here, we describe our comprehensive behavioral phenotyping of the 16p11.2 deletion line developed by Alea Mills on a C57BL/6J and 129S1/SvImJ F1 background (Delm ). Male and female Delm mice were tested in developmental milestones as preweanlings (PND2-PND12), and were tested in open field activity, elevated zero maze, rotarod, novel object recognition, fear conditioning, social approach, and other measures during post-weaning (PND21), adolescence (PND42), and adulthood (>PND70). Developmentally, Delm mice show distinct weight reduction that persists into adulthood. Delm males also have reduced grasp reflexes and limb strength during development, but no other reflexive deficits whereas Delm females show limb strength deficits and decreased sensitivity to heat. In a modified version of a rotarod task that measures balance and coordinated motor activity, Delm males, but not females, show improved performance at high speeds. Delm males and females also show age-specific reductions in anxiety-like behavior compared with WTs, but neither sex show deficits in a social preference task. When assessing learning and memory, Delm males and females show age-specific impairments in a novel object or spatial object recognition, but no deficits in contextual fear memory. This work extends the understanding of the behavioral phenotypes seen with 16p11.2 deletion by emphasizing age and sex-specific deficits; important variables to consider when studying mouse models for neurodevelopmental disorders. LAY SUMMARY: Autism spectrum disorder is a common neurodevelopmental disorder that causes repetitive behavior and impairments in social interaction and communication. Here, we assess the effects of one of the most common genetic alterations in ASDs, a deletion of one copy of 29 genes, using a mouse model. These animals show differences in behavior between males and females and across ages compared with control animals, including changes in development, cognition, and motor coordination. Autism Res 2020, 13: 1670-1684. © 2020 International Society for Autism Research and Wiley Periodicals LLC.
Collapse
Affiliation(s)
- Joseph F Lynch
- Department of Psychology, Franklin and Marshall College, Lancaster, Pennsylvania, USA
| | - Sarah L Ferri
- Department of Neuroscience and Pharmacology, Iowa Neuroscience Institute, University of Iowa, Iowa City, Iowa, USA
| | - Christopher Angelakos
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, California, USA
| | - Hannah Schoch
- Eison S. Floyd College of Medicine, Washington State University Spokane, Spokane, Washington, USA
| | - Thomas Nickl-Jockschat
- Department of Psychiatry, Iowa Neuroscience Institute, University of Iowa, Iowa City, Iowa, USA
| | - Arnold Gonzalez
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | | | - Ted Abel
- Department of Neuroscience and Pharmacology, Iowa Neuroscience Institute, University of Iowa, Iowa City, Iowa, USA
| |
Collapse
|
12
|
Szczurkowska J, Pischedda F, Pinto B, Managò F, Haas CA, Summa M, Bertorelli R, Papaleo F, Schäfer MK, Piccoli G, Cancedda L. NEGR1 and FGFR2 cooperatively regulate cortical development and core behaviours related to autism disorders in mice. Brain 2019; 141:2772-2794. [PMID: 30059965 PMCID: PMC6113639 DOI: 10.1093/brain/awy190] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2017] [Accepted: 06/04/2018] [Indexed: 12/15/2022] Open
Abstract
Autism spectrum disorders are neurodevelopmental conditions with diverse aetiologies, all characterized by common core symptoms such as impaired social skills and communication, as well as repetitive behaviour. Cell adhesion molecules, receptor tyrosine kinases and associated downstream signalling have been strongly implicated in both neurodevelopment and autism spectrum disorders. We found that downregulation of the cell adhesion molecule NEGR1 or the receptor tyrosine kinase fibroblast growth factor receptor 2 (FGFR2) similarly affects neuronal migration and spine density during mouse cortical development in vivo and results in impaired core behaviours related to autism spectrum disorders. Mechanistically, NEGR1 physically interacts with FGFR2 and modulates FGFR2-dependent extracellular signal-regulated kinase (ERK) and protein kinase B (AKT) signalling by decreasing FGFR2 degradation from the plasma membrane. Accordingly, FGFR2 overexpression rescues all defects due to Negr1 knockdown in vivo. Negr1 knockout mice present phenotypes similar to Negr1-downregulated animals. These data indicate that NEGR1 and FGFR2 cooperatively regulate cortical development and suggest a role for defective NEGR1-FGFR2 complex and convergent downstream ERK and AKT signalling in autism spectrum disorders.
Collapse
Affiliation(s)
- Joanna Szczurkowska
- Local Micro-environment and Brain Development Laboratory, Italian Institute of Technology, Genoa, Italy.,Università degli Studi di Genova, Via Balbi, 5, Genoa, Italy
| | - Francesca Pischedda
- Laboratory of Biology of Synapse. Center for Integrative Biology (CIBIO), University of Trento, Trento, Italy
| | - Bruno Pinto
- Local Micro-environment and Brain Development Laboratory, Italian Institute of Technology, Genoa, Italy.,Bio@SNS, Scuola Normale Superiore, Pisa, Italy
| | - Francesca Managò
- Genetics of Cognition Laboratory, Italian Institute of Technology, Genoa, Italy
| | - Carola A Haas
- Experimental Epilepsy Research, Department of Neurosurgery, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Maria Summa
- Department of Drug Discovery and Development, Italian Institute of Technology, Genoa, Italy
| | - Rosalia Bertorelli
- Department of Drug Discovery and Development, Italian Institute of Technology, Genoa, Italy
| | - Francesco Papaleo
- Genetics of Cognition Laboratory, Italian Institute of Technology, Genoa, Italy
| | - Michael K Schäfer
- Department of Anesthesiology and Focus Program Translational Neurosciences, University Medical Center of the Johannes Gutenberg-University Mainz, Germany
| | - Giovanni Piccoli
- Laboratory of Biology of Synapse. Center for Integrative Biology (CIBIO), University of Trento, Trento, Italy.,Dulbecco Telethon Institute, Varese Street 16b - 00185 Rome, Italy
| | - Laura Cancedda
- Local Micro-environment and Brain Development Laboratory, Italian Institute of Technology, Genoa, Italy.,Dulbecco Telethon Institute, Varese Street 16b - 00185 Rome, Italy
| |
Collapse
|
13
|
Komenoi S, Suzuki Y, Asami M, Murakami C, Hoshino F, Chiba S, Takahashi D, Kado S, Sakane F. Microarray analysis of gene expression in the diacylglycerol kinase η knockout mouse brain. Biochem Biophys Rep 2019; 19:100660. [PMID: 31297456 PMCID: PMC6597918 DOI: 10.1016/j.bbrep.2019.100660] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2019] [Revised: 06/17/2019] [Accepted: 06/19/2019] [Indexed: 12/18/2022] Open
Abstract
We have revealed that diacylglycerol kinase η (DGKη)-knockout (KO) mice display bipolar disorder (BPD) remedy-sensitive mania-like behaviors. However, the molecular mechanisms causing the mania-like abnormal behaviors remain unclear. In the present study, microarray analysis was performed to determine global changes in gene expression in the DGKη-KO mouse brain. We found that the DGKη-KO brain had 43 differentially expressed genes and the following five affected biological pathways: "neuroactive ligand-receptor interaction", "transcription by RNA polymerase II", "cytosolic calcium ion concentration", "Jak-STAT signaling pathway" and "ERK1/2 cascade". Interestingly, mRNA levels of prolactin and growth hormone, which are augmented in BPD patients and model animals, were most strongly increased. Notably, all five biological pathways include at least one gene among prolactin, growth hormone, forkhead box P3, glucagon-like peptide 1 receptor and interleukin 1β, which were previously implicated in BPD. Consistent with the microarray data, phosphorylated ERK1/2 levels were decreased in the DGKη-KO brain. Microarray analysis showed that the expression levels of several glycerolipid metabolism-related genes were also changed. Liquid chromatography-mass spectrometry revealed that several polyunsaturated fatty acid (PUFA)-containing phosphatidic acid (PA) molecular species were significantly decreased as a result of DGKη deficiency, suggesting that the decrease affects PUFA metabolism. Intriguingly, the PUFA-containing lysoPA species were markedly decreased in DGKη-KO mouse blood. Taken together, our study provides not only key broad knowledge to gain novel insights into the underlying mechanisms for the mania-like behaviors but also information for developing BPD diagnostics.
Collapse
Key Words
- BPD, bipolar disorder
- Bipolar disorder
- DAVID, Database for AnnotationVisualization and Integrated Discovery
- DG, diacylglycerol
- DGK, diacylglycerol kinase
- Diacylglycerol kinase
- ERK, extracellular signal-regulated kinase
- Fpr2, N-formyl peptide receptor 2
- GO:BP, Gene Ontology: Biological Process
- GWAS, genome-wide association study
- Gh, growth hormone
- Glp1r, glucagon-like peptide 1 receptor
- Growth hormone
- Il1b, interleukin 1β
- KEGG, Kyoto Encyclopedia of Genes and Genomes
- KO, knockout
- LC-MS, liquid chromatography-mass spectrometry
- LPA, lysophosphatidic acid
- Lysophosphatidic acid
- MEK, mitogen-activated protein kinase/ERK kinase
- PA, phosphatidic acid
- PI, phosphatidylinositol
- PUFA, polyunsaturated fatty acid
- Phosphatidic acid
- Prl, prolactin
- Prolactin
- SERT, serotonin transporter
- WT, wild type
Collapse
Affiliation(s)
- Suguru Komenoi
- Department of Chemistry, Graduate School of Science, 1-33 Yayoi-cho, Inage-ku, Chiba, 263-8522, Japan
| | - Yuji Suzuki
- Department of Chemistry, Graduate School of Science, 1-33 Yayoi-cho, Inage-ku, Chiba, 263-8522, Japan
| | - Maho Asami
- Department of Chemistry, Graduate School of Science, 1-33 Yayoi-cho, Inage-ku, Chiba, 263-8522, Japan
| | - Chiaki Murakami
- Department of Chemistry, Graduate School of Science, 1-33 Yayoi-cho, Inage-ku, Chiba, 263-8522, Japan
| | - Fumi Hoshino
- Department of Chemistry, Graduate School of Science, 1-33 Yayoi-cho, Inage-ku, Chiba, 263-8522, Japan
| | - Sohei Chiba
- Department of Chemistry, Graduate School of Science, 1-33 Yayoi-cho, Inage-ku, Chiba, 263-8522, Japan
| | - Daisuke Takahashi
- Department of Chemistry, Graduate School of Science, 1-33 Yayoi-cho, Inage-ku, Chiba, 263-8522, Japan
| | - Sayaka Kado
- Center for Analytical Instrumentation, Chiba University, 1-33 Yayoi-cho, Inage-ku, Chiba, 263-8522, Japan
| | - Fumio Sakane
- Department of Chemistry, Graduate School of Science, 1-33 Yayoi-cho, Inage-ku, Chiba, 263-8522, Japan
- Corresponding author. Department of Chemistry, Graduate School of Science, Chiba University, 1-33 Yayoi-cho, Inage-ku, Chiba, 263-8522, Japan.
| |
Collapse
|
14
|
Giusti L, Molinaro A, Alessandrì MG, Boldrini C, Ciregia F, Lacerenza S, Ronci M, Urbani A, Cioni G, Mazzoni MR, Pizzorusso T, Lucacchini A, Baroncelli L. Brain mitochondrial proteome alteration driven by creatine deficiency suggests novel therapeutic venues for creatine deficiency syndromes. Neuroscience 2019; 409:276-289. [PMID: 31029731 DOI: 10.1016/j.neuroscience.2019.03.030] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2019] [Revised: 03/09/2019] [Accepted: 03/12/2019] [Indexed: 01/15/2023]
Abstract
Creatine (Cr) is a small metabolite with a central role in energy metabolism and mitochondrial function. Creatine deficiency syndromes are inborn errors of Cr metabolism causing Cr depletion in all body tissues and particularly in the nervous system. Patient symptoms involve intellectual disability, language and behavioral disturbances, seizures and movement disorders suggesting that brain cells are particularly sensitive to Cr depletion. Cr deficiency was found to affect metabolic activity and structural abnormalities of mitochondrial organelles; however a detailed analysis of molecular mechanisms linking Cr deficit, energy metabolism alterations and brain dysfunction is still missing. Using a proteomic approach we evaluated the proteome changes of the brain mitochondrial fraction induced by the deletion of the Cr transporter (CrT) in developing mutant mice. We found a marked alteration of the mitochondrial proteomic landscape in the brain of CrT deficient mice, with the overexpression of many proteins involved in energy metabolism and response to oxidative stress. Moreover, our data suggest possible abnormalities of dendritic spines, synaptic function and plasticity, network excitability and neuroinflammatory response. Intriguingly, the alterations occurred in coincidence with the developmental onset of neurological symptoms. Thus, cerebral mitochondrial alterations could represent an early response to Cr deficiency that could be targeted for therapeutic intervention.
Collapse
Affiliation(s)
- Laura Giusti
- Department of Clinical and Experimental Medicine, University of Pisa, I-56126, Pisa, Italy; School of Pharmacy, University of Camerino, I-62032 Camerino, Italy
| | - Angelo Molinaro
- Department of Neuroscience, Psychology, Drug Research and Child Health NEUROFARBA, University of Florence, I-50135, Florence, Italy; Institute of Neuroscience, National Research Council (CNR), I-56124, Pisa, Italy
| | - Maria Grazia Alessandrì
- Department of Developmental Neuroscience, IRCCS Stella Maris Foundation, I-56128 Pisa, Italy
| | - Claudia Boldrini
- Department of Pharmacy, University of Pisa, I-56126, Pisa, Italy
| | - Federica Ciregia
- Department of Pharmacy, University of Pisa, I-56126, Pisa, Italy; Department of Rheumatology, GIGA Research, Centre Hospitalier Universitaire (CHU) de Liège, B-4000, Liège, Belgium
| | - Serena Lacerenza
- Department of Pharmacy, University of Pisa, I-56126, Pisa, Italy
| | - Maurizio Ronci
- Department of Medical, Oral and Biotechnological Sciences, University G. d'Annunzio of Chieti-Pescara, I-66100, Chieti, Italy
| | - Andrea Urbani
- Institute of Biochemistry and Clinical Chemistry, Catholic university of the sacred heart, I-00168, Rome, Italy
| | - Giovanni Cioni
- Department of Clinical and Experimental Medicine, University of Pisa, I-56126, Pisa, Italy; Department of Developmental Neuroscience, IRCCS Stella Maris Foundation, I-56128 Pisa, Italy
| | | | - Tommaso Pizzorusso
- Department of Neuroscience, Psychology, Drug Research and Child Health NEUROFARBA, University of Florence, I-50135, Florence, Italy; Institute of Neuroscience, National Research Council (CNR), I-56124, Pisa, Italy
| | - Antonio Lucacchini
- Department of Clinical and Experimental Medicine, University of Pisa, I-56126, Pisa, Italy
| | - Laura Baroncelli
- Institute of Neuroscience, National Research Council (CNR), I-56124, Pisa, Italy; Department of Developmental Neuroscience, IRCCS Stella Maris Foundation, I-56128 Pisa, Italy.
| |
Collapse
|
15
|
Pérez-Sen R, Queipo MJ, Gil-Redondo JC, Ortega F, Gómez-Villafuertes R, Miras-Portugal MT, Delicado EG. Dual-Specificity Phosphatase Regulation in Neurons and Glial Cells. Int J Mol Sci 2019; 20:ijms20081999. [PMID: 31018603 PMCID: PMC6514851 DOI: 10.3390/ijms20081999] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2019] [Revised: 04/19/2019] [Accepted: 04/19/2019] [Indexed: 01/03/2023] Open
Abstract
Dual-specificity protein phosphatases comprise a protein phosphatase subfamily with selectivity towards mitogen-activated protein (MAP) kinases, also named MKPs, or mitogen-activated protein kinase (MAPK) phosphatases. As powerful regulators of the intensity and duration of MAPK signaling, a relevant role is envisioned for dual-specificity protein phosphatases (DUSPs) in the regulation of biological processes in the nervous system, such as differentiation, synaptic plasticity, and survival. Important neural mediators include nerve growth factor (NGF) and brain-derived neurotrophic factor (BDNF) that contribute to DUSP transcriptional induction and post-translational mechanisms of DUSP protein stabilization to maintain neuronal survival and differentiation. Potent DUSP gene inducers also include cannabinoids, which preserve DUSP activity in inflammatory conditions. Additionally, nucleotides activating P2X7 and P2Y13 nucleotide receptors behave as novel players in the regulation of DUSP function. They increase cell survival in stressful conditions, regulating DUSP protein turnover and inducing DUSP gene expression. In general terms, in the context of neural cells exposed to damaging conditions, the recovery of DUSP activity is neuroprotective and counteracts pro-apoptotic over-activation of p38 and JNK. In addition, remarkable changes in DUSP function take place during the onset of neuropathologies. The restoration of proper DUSP levels and recovery of MAPK homeostasis underlie the therapeutic effect, indicating that DUSPs can be relevant targets for brain diseases.
Collapse
Affiliation(s)
- Raquel Pérez-Sen
- Departamento de Bioquímica y Biología Molecular, Facultad de Veterinaria, Instituto Universitario de Investigación en Neuroquímica (IUIN), Instituto de Investigación Sanitaria del Hospital Clínico San Carlos (IdiSSC), Universidad Complutense Madrid, 28040 Madrid, Spain.
| | - María José Queipo
- Departamento de Bioquímica y Biología Molecular, Facultad de Veterinaria, Instituto Universitario de Investigación en Neuroquímica (IUIN), Instituto de Investigación Sanitaria del Hospital Clínico San Carlos (IdiSSC), Universidad Complutense Madrid, 28040 Madrid, Spain.
| | - Juan Carlos Gil-Redondo
- Departamento de Bioquímica y Biología Molecular, Facultad de Veterinaria, Instituto Universitario de Investigación en Neuroquímica (IUIN), Instituto de Investigación Sanitaria del Hospital Clínico San Carlos (IdiSSC), Universidad Complutense Madrid, 28040 Madrid, Spain.
| | - Felipe Ortega
- Departamento de Bioquímica y Biología Molecular, Facultad de Veterinaria, Instituto Universitario de Investigación en Neuroquímica (IUIN), Instituto de Investigación Sanitaria del Hospital Clínico San Carlos (IdiSSC), Universidad Complutense Madrid, 28040 Madrid, Spain.
| | - Rosa Gómez-Villafuertes
- Departamento de Bioquímica y Biología Molecular, Facultad de Veterinaria, Instituto Universitario de Investigación en Neuroquímica (IUIN), Instituto de Investigación Sanitaria del Hospital Clínico San Carlos (IdiSSC), Universidad Complutense Madrid, 28040 Madrid, Spain.
| | - María Teresa Miras-Portugal
- Departamento de Bioquímica y Biología Molecular, Facultad de Veterinaria, Instituto Universitario de Investigación en Neuroquímica (IUIN), Instituto de Investigación Sanitaria del Hospital Clínico San Carlos (IdiSSC), Universidad Complutense Madrid, 28040 Madrid, Spain.
| | - Esmerilda G Delicado
- Departamento de Bioquímica y Biología Molecular, Facultad de Veterinaria, Instituto Universitario de Investigación en Neuroquímica (IUIN), Instituto de Investigación Sanitaria del Hospital Clínico San Carlos (IdiSSC), Universidad Complutense Madrid, 28040 Madrid, Spain.
| |
Collapse
|
16
|
Adamo AM, Liu X, Mathieu P, Nuttall JR, Supasai S, Oteiza PI. Early Developmental Marginal Zinc Deficiency Affects Neurogenesis Decreasing Neuronal Number and Altering Neuronal Specification in the Adult Rat Brain. Front Cell Neurosci 2019; 13:62. [PMID: 30890920 PMCID: PMC6414196 DOI: 10.3389/fncel.2019.00062] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2018] [Accepted: 02/08/2019] [Indexed: 12/03/2022] Open
Abstract
During pregnancy, a decreased availability of zinc to the fetus can disrupt the development of the central nervous system leading to defects ranging from severe malformations to subtle neurological and cognitive effects. We previously found that marginal zinc deficiency down-regulates the extracellular signal-regulated kinase 1/2 (ERK1/2) signaling pathway and affects neural progenitor cell (NPC) proliferation. This study investigated if marginal zinc deficiency during gestation in rats could disrupt fetal neurogenesis and affect the number and specification of neurons in the adult offspring brain cortex. Rats were fed a marginal zinc deficient or adequate diet throughout gestation and until postnatal day (P) 2, and subsequently the zinc adequate diet until P56. Neurogenesis was evaluated in the offspring at embryonic day (E)14, E19, P2, and P56 measuring parameters of NPC proliferation and differentiation by Western blot and/or immunofluorescence. At E14 and E19, major signals (i.e., ERK1/2, Sox2, and Pax6) that stimulate NPC proliferation and self-renewal were markedly downregulated in the marginal zinc deficient fetal brain. These alterations were associated to a lower number of Ki67 positive cells in the ventricular (VZs) and subventricular zones (SVZs). Following the progression of NPCs into intermediate progenitor cells (IPCs) and into neurons, Pax6, Tbr2 and Tbr1 were affected in the corresponding areas of the brain at E19 and P2. The above signaling alterations led to a lower density of neurons and a selective decrease of glutamatergic neurons in the young adult brain cortex exposed to maternal marginal zinc deficiency from E14 to P2. Current results supports the concept that marginal zinc deficiency during fetal development can disrupt neurogenesis and alter cortical structure potentially leading to irreversible neurobehavioral impairments later in life.
Collapse
Affiliation(s)
- Ana M Adamo
- Department of Biological Chemistry and IQUIFIB (UBA-CONICET), Facultad de Farmacia y Bioquimica, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Xiuzhen Liu
- Department of Nutrition and Department of Environmental Toxicology, University of California, Davis, Davis, CA, United States
| | - Patricia Mathieu
- Department of Biological Chemistry and IQUIFIB (UBA-CONICET), Facultad de Farmacia y Bioquimica, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Johnathan R Nuttall
- Department of Nutrition and Department of Environmental Toxicology, University of California, Davis, Davis, CA, United States
| | - Suangsuda Supasai
- Department of Nutrition and Department of Environmental Toxicology, University of California, Davis, Davis, CA, United States.,Department of Molecular Tropical Medicine and Genetics, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
| | - Patricia I Oteiza
- Department of Nutrition and Department of Environmental Toxicology, University of California, Davis, Davis, CA, United States
| |
Collapse
|
17
|
Rosina E, Battan B, Siracusano M, Di Criscio L, Hollis F, Pacini L, Curatolo P, Bagni C. Disruption of mTOR and MAPK pathways correlates with severity in idiopathic autism. Transl Psychiatry 2019; 9:50. [PMID: 30705255 PMCID: PMC6355879 DOI: 10.1038/s41398-018-0335-z] [Citation(s) in RCA: 75] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/01/2018] [Revised: 11/04/2018] [Accepted: 11/13/2018] [Indexed: 12/15/2022] Open
Abstract
The molecular signature underlying autism spectrum disorder remains largely unknown. This study identifies differential expression of mTOR and MAPK pathways in patients affected by mild and severe idiopathic autism. A total of 55 subjects were enrolled, of which 22 were typically developing individuals and 33 were patients aged between 3 and 11 years, with autism spectrum disorder. A detailed history, including physical examination, developmental evaluation, mental health history and autism diagnostic observation schedule were performed for each patient. Components of the mTOR and MAPK signalling pathways were analysed from peripheral blood at the protein level. Patients were then stratified according to their clinical phenotypes, and the molecular profiling was analysed in relation to the degree of autism severity. In this cohort of patients, we identified increased activity of mTOR and the MAPK pathways, key regulators of synaptogenesis and protein synthesis. Specifically, rpS6, p-eIF4E, TSC1 and p-MNK1 expression discriminated patients according to their clinical diagnosis, suggesting that components of protein synthesis signalling pathways might constitute a molecular signature of clinical severity in autism spectrum disorder.
Collapse
Affiliation(s)
- Eleonora Rosina
- Department of Biomedicine and Prevention, University of Rome Tor Vergata, Rome, Italy
- Department of Fundamental Neurosciences, University of Lausanne, Lausanne, Switzerland
| | - Barbara Battan
- Department of Systems Medicine, Division of Child Neurology and Psychiatry, University Hospital of Tor Vergata, Rome, Italy
| | - Martina Siracusano
- Department of Biomedicine and Prevention, University of Rome Tor Vergata, Rome, Italy
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, L'Aquila, Italy
| | - Lorena Di Criscio
- Department of Systems Medicine, Division of Child Neurology and Psychiatry, University Hospital of Tor Vergata, Rome, Italy
| | - Fiona Hollis
- Department of Fundamental Neurosciences, University of Lausanne, Lausanne, Switzerland
| | - Laura Pacini
- Department of Biomedicine and Prevention, University of Rome Tor Vergata, Rome, Italy
| | - Paolo Curatolo
- Department of Systems Medicine, Division of Child Neurology and Psychiatry, University Hospital of Tor Vergata, Rome, Italy
| | - Claudia Bagni
- Department of Biomedicine and Prevention, University of Rome Tor Vergata, Rome, Italy.
- Department of Fundamental Neurosciences, University of Lausanne, Lausanne, Switzerland.
| |
Collapse
|
18
|
Xiao X, Zhang C, Grigoroiu-Serbanescu M, Wang L, Li L, Zhou D, Yuan TF, Wang C, Chang H, Wu Y, Li Y, Wu DD, Yao YG, Li M. The cAMP responsive element-binding (CREB)-1 gene increases risk of major psychiatric disorders. Mol Psychiatry 2018; 23:1957-1967. [PMID: 29158582 DOI: 10.1038/mp.2017.243] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/04/2017] [Revised: 08/25/2017] [Accepted: 09/14/2017] [Indexed: 12/11/2022]
Abstract
Bipolar disorder (BPD), schizophrenia (SCZ) and unipolar major depressive disorder (MDD) are primary psychiatric disorders sharing substantial genetic risk factors. We previously reported that two single-nucleotide polymorphisms (SNPs) rs2709370 and rs6785 in the cAMP responsive element-binding (CREB)-1 gene (CREB1) were associated with the risk of BPD and abnormal hippocampal function in populations of European ancestry. In the present study, we further expanded our analyses of rs2709370 and rs6785 in multiple BPD, SCZ and MDD data sets, including the published Psychiatric Genomics Consortium (PGC) genome-wide association study, the samples used in our previous CREB1 study, and six additional cohorts (three new BPD samples, two new SCZ samples and one new MDD sample). Although the associations of both CREB1 SNPs with each illness were not replicated in the new cohorts (BPD analysis in 871 cases and 1089 controls (rs2709370, P=0.0611; rs6785, P=0.0544); SCZ analysis in 1273 cases and 1072 controls (rs2709370, P=0.230; rs6785, P=0.661); and MDD analysis in 129 cases and 100 controls (rs2709370, P=0.114; rs6785, P=0.188)), an overall meta-analysis of all included samples suggested that both SNPs were significantly associated with increased risk of BPD (11 105 cases and 51 331 controls; rs2709370, P=2.33 × 10-4; rs6785, P=6.33 × 10-5), SCZ (34 913 cases and 44 528 controls; rs2709370, P=3.96 × 10-5; rs6785, P=2.44 × 10-5) and MDD (9369 cases and 9619 controls; rs2709370, P=0.0144; rs6785, P=0.0314), with the same direction of allelic effects across diagnostic categories. We then examined the impact of diagnostic status on CREB1 mRNA expression using data obtained from independent brain tissue samples, and observed that the mRNA expression of CREB1 was significantly downregulated in psychiatric patients compared with healthy controls. The protein-protein interaction analyses showed that the protein encoded by CREB1 directly interacted with several risk genes of psychiatric disorders identified by GWAS. In conclusion, the current study suggests that CREB1 might be a common risk gene for major psychiatric disorders, and further investigations are necessary.
Collapse
Affiliation(s)
- X Xiao
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences and Yunnan Province, Kunming Institute of Zoology, Kunming, China
| | - C Zhang
- Schizophrenia Program, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| | - M Grigoroiu-Serbanescu
- Biometric Psychiatric Genetics Research Unit, Alexandru Obregia Clinical Psychiatric Hospital, Bucharest, Romania.
| | - L Wang
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences and Yunnan Province, Kunming Institute of Zoology, Kunming, China
| | - L Li
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences and Yunnan Province, Kunming Institute of Zoology, Kunming, China
| | - D Zhou
- Ningbo Kangning Hospital, Ningbo, China
| | - T-F Yuan
- Shanghai Key Laboratory of Psychotic Disorders, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - C Wang
- Department of Pharmacology, and Provincial Key Laboratory of Pathophysiology in Ningbo University School of Medicine, Ningbo, China
| | - H Chang
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences and Yunnan Province, Kunming Institute of Zoology, Kunming, China
| | - Y Wu
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences and Yunnan Province, Kunming Institute of Zoology, Kunming, China
| | - Y Li
- Laboratory for Conservation and Utilization of Bio-Resource, Yunnan University, Kunming, China
| | - D-D Wu
- State Key Laboratory of Genetic Resources and Evolution, Chinese Academy of Sciences, Kunming, China
| | - Y-G Yao
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences and Yunnan Province, Kunming Institute of Zoology, Kunming, China.,CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, China
| | - M Li
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences and Yunnan Province, Kunming Institute of Zoology, Kunming, China. .,CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, China.
| |
Collapse
|
19
|
Pharmacological Inhibition of ERK Signaling Rescues Pathophysiology and Behavioral Phenotype Associated with 16p11.2 Chromosomal Deletion in Mice. J Neurosci 2018; 38:6640-6652. [PMID: 29934348 DOI: 10.1523/jneurosci.0515-17.2018] [Citation(s) in RCA: 61] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2017] [Revised: 06/05/2018] [Accepted: 06/06/2018] [Indexed: 12/17/2022] Open
Abstract
The human 16p11.2 microdeletion is one of the most common gene copy number variations linked to autism, but the pathophysiology associated with this chromosomal abnormality is largely unknown. The 593 kb deletion contains the ERK1 gene and other genes that converge onto the ERK/MAP kinase pathway. Perturbations in ERK signaling are linked to a group of related neurodevelopmental disorders hallmarked by intellectual disability, including autism. We report that mice harboring the 16p11.2 deletion exhibit a paradoxical elevation of ERK activity, cortical cytoarchitecture abnormalities and behavioral deficits. Importantly, we show that treatment with a novel ERK pathway inhibitor during a critical period of brain development rescues the molecular, anatomical and behavioral deficits in the 16p11.2 deletion mice. The ERK inhibitor treatment administered to adult mice ameliorates a subset of these behavioral deficits. Our findings provide evidence for potential targeted therapeutic intervention in 16p11.2 deletion carriers.SIGNIFICANCE STATEMENT The ERK/MAPK pathway is genetically linked to autism spectrum disorders and other syndromes typified by intellectual disability. We provide direct evidence connecting the ERK/MAP kinases to the developmental abnormalities in neurogenesis and cortical cytoarchitecture associated with the 16p11.2 chromosomal deletion. Most importantly, we demonstrate that treatment with a novel ERK-specific inhibitor during development rescues aberrant cortical cytoarchitecture and restores normal levels of cell-cycle regulators during cortical neurogenesis. These treatments partially reverse the behavioral deficits observed in the 16p11.2del mouse model, including hyperactivity, memory as well as olfaction, and maternal behavior. We also report a rescue of a subset of these deficits upon treatment of adult 16p11.2del mice. These data provide a strong rationale for therapeutic approaches to this disorder.
Collapse
|
20
|
Duda M, Zhang H, Li HD, Wall DP, Burmeister M, Guan Y. Brain-specific functional relationship networks inform autism spectrum disorder gene prediction. Transl Psychiatry 2018; 8:56. [PMID: 29507298 PMCID: PMC5838237 DOI: 10.1038/s41398-018-0098-6] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/11/2017] [Revised: 10/20/2017] [Accepted: 12/30/2017] [Indexed: 11/09/2022] Open
Abstract
Autism spectrum disorder (ASD) is a neuropsychiatric disorder with strong evidence of genetic contribution, and increased research efforts have resulted in an ever-growing list of ASD candidate genes. However, only a fraction of the hundreds of nominated ASD-related genes have identified de novo or transmitted loss of function (LOF) mutations that can be directly attributed to the disorder. For this reason, a means of prioritizing candidate genes for ASD would help filter out false-positive results and allow researchers to focus on genes that are more likely to be causative. Here we constructed a machine learning model by leveraging a brain-specific functional relationship network (FRN) of genes to produce a genome-wide ranking of ASD risk genes. We rigorously validated our gene ranking using results from two independent sequencing experiments, together representing over 5000 simplex and multiplex ASD families. Finally, through functional enrichment analysis on our highly prioritized candidate gene network, we identified a small number of pathways that are key in early neural development, providing further support for their potential role in ASD.
Collapse
Affiliation(s)
- Marlena Duda
- 0000000086837370grid.214458.eDepartment of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, MI USA
| | - Hongjiu Zhang
- 0000000086837370grid.214458.eDepartment of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, MI USA
| | - Hong-Dong Li
- 0000000086837370grid.214458.eDepartment of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, MI USA ,0000 0001 0379 7164grid.216417.7Center for Bioinformatics, School of Information Science and Engineering, Central South University, Changsha, China
| | - Dennis P. Wall
- 0000000419368956grid.168010.eDepartment of Pediatrics, Division of Systems Medicine, Stanford University, Stanford, CA USA ,0000000419368956grid.168010.eDepartment of Biomedical Data Science, Stanford University, Stanford, CA USA
| | - Margit Burmeister
- 0000000086837370grid.214458.eDepartment of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, MI USA ,0000000086837370grid.214458.eMolecular and Behavioral Neuroscience Institute, University of Michigan, Ann Arbor, MI USA ,0000000086837370grid.214458.eDepartment of Human Genetics, University of Michigan, Ann Arbor, MI USA ,0000000086837370grid.214458.eDepartment of Psychiatry, University of Michigan, Ann Arbor, MI USA
| | - Yuanfang Guan
- Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, MI, USA. .,Department of Internal Medicine, Usniversity of Michigan, Ann Arbor, MI, USA. .,Department of Electrical Engineering and Computer Science, University of Michigan, Ann Arbor, MI, USA.
| |
Collapse
|
21
|
Yoon SY, Kwon SG, Kim YH, Yeo JH, Ko HG, Roh DH, Kaang BK, Beitz AJ, Lee JH, Oh SB. A critical role of spinal Shank2 proteins in NMDA-induced pain hypersensitivity. Mol Pain 2017; 13:1744806916688902. [PMID: 28326932 PMCID: PMC5302174 DOI: 10.1177/1744806916688902] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Background Self-injurious behaviors (SIBs) are devastating traits in autism spectrum disorder (ASD). Although deficits in pain sensation might be one of the contributing factors underlying the development of SIBs, the mechanisms have yet to be addressed. Recently, the Shank2 synaptic protein has been considered to be a key component in ASD, and mutations of SHANK2 gene induce the dysfunction of N-methyl-D-aspartate (NMDA) receptors, suggesting a link between Shank2 and NMDA receptors in ASD. Given that spinal NMDA receptors play a pivotal role in pain hypersensitivity, we investigated the possible role of Shank2 in nociceptive hypersensitivity by examining changes in spontaneous pain following intrathecal NMDA injection in Shank2−/− (Shank2 knock-out, KO) mice. Results Intrathecal NMDA injection evoked spontaneous nociceptive behaviors. These NMDA-induced nociceptive responses were significantly reduced in Shank2 KO mice. We also observed a significant decrease of NMDA currents in the spinal dorsal horn of Shank2 KO mice. Subsequently, we examined whether mitogen-activated protein kinase or AKT signaling is involved in this reduced pain behavior in Shank2 KO mice because the NMDA receptor is closely related to these signaling molecules. Western blotting and immunohistochemistry revealed that spinally administered NMDA increased the expression of a phosphorylated form of extracellular signal-regulated kinase (p-ERK) which was significantly reduced in Shank2 KO mice. However, p38, JNK, or AKT were not changed by NMDA administration. The ERK inhibitor, PD98059, decreased NMDA-induced spontaneous pain behaviors in a dose-dependent manner in wild-type mice. Moreover, it was found that the NMDA-induced increase in p-ERK was primarily colocalized with Shank2 proteins in the spinal cord dorsal horn. Conclusion Shank2 protein is involved in spinal NMDA receptor-mediated pain, and mutations of Shank2 may suppress NMDA-ERK signaling in spinal pain transmission. This study provides new clues into the mechanisms underlying pain deficits associated with SIB and deserves further study in patients with ASD.
Collapse
Affiliation(s)
- Seo-Yeon Yoon
- 1 Department of Brain and Cognitive Sciences, College of Natural Sciences, Pain Cognitive Function Research Center, Dental Research Institute, Seoul National University, Seoul, Republic of Korea.,2 Department of Neurobiology and Physiology School of Dentistry, Seoul National University, Seoul, Republic of Korea
| | - Soon-Gu Kwon
- 3 Department of Oral Physiology and Research Center for Tooth and Periodontal Tissue Regeneration, School of Dentistry, Kyung Hee University, Seoul, Republic of Korea
| | - Yong Ho Kim
- 1 Department of Brain and Cognitive Sciences, College of Natural Sciences, Pain Cognitive Function Research Center, Dental Research Institute, Seoul National University, Seoul, Republic of Korea.,2 Department of Neurobiology and Physiology School of Dentistry, Seoul National University, Seoul, Republic of Korea.,4 Department of Physiology, College of Medicine, Gachon University, Incheon, Republic of Korea
| | - Ji-Hee Yeo
- 3 Department of Oral Physiology and Research Center for Tooth and Periodontal Tissue Regeneration, School of Dentistry, Kyung Hee University, Seoul, Republic of Korea
| | - Hyoung-Gon Ko
- 5 Department of Biological Sciences, College of Natural Sciences, Seoul National University, Seoul, Republic of Korea
| | - Dae-Hyun Roh
- 3 Department of Oral Physiology and Research Center for Tooth and Periodontal Tissue Regeneration, School of Dentistry, Kyung Hee University, Seoul, Republic of Korea
| | - Bong-Kiun Kaang
- 5 Department of Biological Sciences, College of Natural Sciences, Seoul National University, Seoul, Republic of Korea.,6 Center for Neuron and Disease, Frontier Institutes of Science and Technology, Xi'an Jiaotong University, Xi'an, China
| | - Alvin J Beitz
- 7 Department of Veterinary and Biomedical Sciences, College of Veterinary Medicine, University of Minnesota, St Paul, MN, USA
| | - Jang-Hern Lee
- 8 Department of Veterinary Physiology, BK21 PLUS Program for Creative Veterinary Science Research, Seoul National University, Seoul, Republic of Korea.,9 Research Institute for Veterinary Science, College of Veterinary Medicine, Seoul National University, Seoul, Republic of Korea
| | - Seog Bae Oh
- 1 Department of Brain and Cognitive Sciences, College of Natural Sciences, Pain Cognitive Function Research Center, Dental Research Institute, Seoul National University, Seoul, Republic of Korea.,2 Department of Neurobiology and Physiology School of Dentistry, Seoul National University, Seoul, Republic of Korea
| |
Collapse
|
22
|
Rao AR, Yourshaw M, Christensen B, Nelson SF, Kerner B. Rare deleterious mutations are associated with disease in bipolar disorder families. Mol Psychiatry 2017; 22:1009-1014. [PMID: 27725659 PMCID: PMC5388596 DOI: 10.1038/mp.2016.181] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/25/2015] [Revised: 06/19/2016] [Accepted: 08/01/2016] [Indexed: 12/18/2022]
Abstract
Bipolar disorder (BD) is a common, complex and heritable psychiatric disorder characterized by episodes of severe mood swings. The identification of rare, damaging genomic mutations in families with BD could inform about disease mechanisms and lead to new therapeutic interventions. To determine whether rare, damaging mutations shared identity-by-descent in families with BD could be associated with disease, exome sequencing was performed in multigenerational families of the NIMH BD Family Study followed by in silico functional prediction. Disease association and disease specificity was determined using 5090 exomes from the Sweden-Schizophrenia (SZ) Population-Based Case-Control Exome Sequencing study. We identified 14 rare and likely deleterious mutations in 14 genes that were shared identity-by-descent among affected family members. The variants were associated with BD (P<0.05 after Bonferroni's correction) and disease specificity was supported by the absence of the mutations in patients with SZ. In addition, we found rare, functional mutations in known causal genes for neuropsychiatric disorders including holoprosencephaly and epilepsy. Our results demonstrate that exome sequencing in multigenerational families with BD is effective in identifying rare genomic variants of potential clinical relevance and also disease modifiers related to coexisting medical conditions. Replication of our results and experimental validation are required before disease causation could be assumed.
Collapse
Affiliation(s)
- Aliz R Rao
- Department of Human Genetics, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Michael Yourshaw
- Department of Pediatrics, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | | | - Stanley F Nelson
- Department of Human Genetics, University of California, Los Angeles, Los Angeles, CA 90095, USA
- Department of Psychiatry and Biobehavioral Sciences at the David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
- Department of Pathology and Laboratory Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Berit Kerner
- Semel Institute for Neuroscience and Human Behavior, University of California, Los Angeles, Los Angeles, CA 90095, USA
| |
Collapse
|
23
|
Schaefer TL, Davenport MH, Grainger LM, Robinson CK, Earnheart AT, Stegman MS, Lang AL, Ashworth AA, Molinaro G, Huber KM, Erickson CA. Acamprosate in a mouse model of fragile X syndrome: modulation of spontaneous cortical activity, ERK1/2 activation, locomotor behavior, and anxiety. J Neurodev Disord 2017; 9:6. [PMID: 28616095 PMCID: PMC5467053 DOI: 10.1186/s11689-017-9184-y] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/12/2016] [Accepted: 01/13/2017] [Indexed: 01/08/2023] Open
Abstract
BACKGROUND Fragile X Syndrome (FXS) occurs as a result of a silenced fragile X mental retardation 1 gene (FMR1) and subsequent loss of fragile X mental retardation protein (FMRP) expression. Loss of FMRP alters excitatory/inhibitory signaling balance, leading to increased neuronal hyperexcitability and altered behavior. Acamprosate (the calcium salt of N-acetylhomotaurinate), a drug FDA-approved for relapse prevention in the treatment of alcohol dependence in adults, is a novel agent with multiple mechanisms that may be beneficial for people with FXS. There are questions regarding the neuroactive effects of acamprosate and the significance of the molecule's calcium moiety. Therefore, the electrophysiological, cellular, molecular, and behavioral effects of acamprosate were assessed in the Fmr1-/y (knock out; KO) mouse model of FXS controlling for the calcium salt in several experiments. METHODS Fmr1 KO mice and their wild-type (WT) littermates were utilized to assess acamprosate treatment on cortical UP state parameters, dendritic spine density, and seizure susceptibility. Brain extracellular-signal regulated kinase 1/2 (ERK1/2) activation was used to investigate this signaling molecule as a potential biomarker for treatment response. Additional adult mice were used to assess chronic acamprosate treatment and any potential effects of the calcium moiety using CaCl2 treatment on behavior and nuclear ERK1/2 activation. RESULTS Acamprosate attenuated prolonged cortical UP state duration, decreased elevated ERK1/2 activation in brain tissue, and reduced nuclear ERK1/2 activation in the dentate gyrus in KO mice. Acamprosate treatment modified behavior in anxiety and locomotor tests in Fmr1 KO mice in which control-treated KO mice were shown to deviate from control-treated WT mice. Mice treated with CaCl2 were not different from saline-treated mice in the adult behavior battery or nuclear ERK1/2 activation. CONCLUSIONS These data indicate that acamprosate, and not calcium, improves function reminiscent of reduced anxiety-like behavior and hyperactivity in Fmr1 KO mice and that acamprosate attenuates select electrophysiological and molecular dysregulation that may play a role in the pathophysiology of FXS. Differences between control-treated KO and WT mice were not evident in a recognition memory test or in examination of acoustic startle response/prepulse inhibition which impeded conclusions from being made about the treatment effects of acamprosate in these instances.
Collapse
Affiliation(s)
- Tori L Schaefer
- Division of Psychiatry, MLC 7004, Cincinnati Children's Research Foundation, 3333 Burnet Ave., Cincinnati, OH 45229-3039 USA
| | - Matthew H Davenport
- Division of Psychiatry, MLC 7004, Cincinnati Children's Research Foundation, 3333 Burnet Ave., Cincinnati, OH 45229-3039 USA
| | - Lindsay M Grainger
- Division of Psychiatry, MLC 7004, Cincinnati Children's Research Foundation, 3333 Burnet Ave., Cincinnati, OH 45229-3039 USA
| | - Chandler K Robinson
- Division of Psychiatry, MLC 7004, Cincinnati Children's Research Foundation, 3333 Burnet Ave., Cincinnati, OH 45229-3039 USA
| | - Anthony T Earnheart
- Division of Psychiatry, MLC 7004, Cincinnati Children's Research Foundation, 3333 Burnet Ave., Cincinnati, OH 45229-3039 USA
| | - Melinda S Stegman
- Division of Psychiatry, MLC 7004, Cincinnati Children's Research Foundation, 3333 Burnet Ave., Cincinnati, OH 45229-3039 USA.,Present address: Division of Nephrology and Hypertension, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229 USA
| | - Anna L Lang
- Division of Psychiatry, MLC 7004, Cincinnati Children's Research Foundation, 3333 Burnet Ave., Cincinnati, OH 45229-3039 USA.,Present address: Department of Pharmacology and Toxicology, University of Louisville, Louisville, KY 40202 USA
| | - Amy A Ashworth
- Division of Psychiatry, MLC 7004, Cincinnati Children's Research Foundation, 3333 Burnet Ave., Cincinnati, OH 45229-3039 USA.,Present address: BlackbookHR, Cincinnati, OH 45202 USA
| | - Gemma Molinaro
- Department of Neuroscience, University of Texas Southwestern Medical Center, Dallas, TX 75390 USA
| | - Kimberly M Huber
- Department of Neuroscience, University of Texas Southwestern Medical Center, Dallas, TX 75390 USA
| | - Craig A Erickson
- Division of Psychiatry, MLC 7004, Cincinnati Children's Research Foundation, 3333 Burnet Ave., Cincinnati, OH 45229-3039 USA
| |
Collapse
|
24
|
Muneer A. Wnt and GSK3 Signaling Pathways in Bipolar Disorder: Clinical and Therapeutic Implications. CLINICAL PSYCHOPHARMACOLOGY AND NEUROSCIENCE 2017; 15:100-114. [PMID: 28449557 PMCID: PMC5426498 DOI: 10.9758/cpn.2017.15.2.100] [Citation(s) in RCA: 65] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/29/2016] [Accepted: 11/20/2016] [Indexed: 01/19/2023]
Abstract
The neurobiology of bipolar disorder, a chronic and systemic ailment is not completely understood. The bipolar phenotype manifests in myriad ways, and psychopharmacological agents like lithium have long term beneficial effects. The enzyme glycogen synthase kinase 3 (GSK3) has come into focus, as lithium and several other mood stabilizing medications inhibit its activity. This kinase and its key upstream modulator, Wnt are dysregulated in mood disorders and there is a growing impetus to delineate the chief substrates involved in the development of these illnesses. In May 2016, a comprehensive literature search was undertaken which revealed that there is over activity of GSK3 in bipolar disorder with deleterious downstream effects like proinflammatory status, increased oxidative stress, and circadian dysregulation leading to declining neurotrophic support and enhanced apoptosis of neural elements. By developing specific GSK3 inhibitors the progressive worsening in bipolar disorder can be forestalled with improved prospects for the sufferers.
Collapse
Affiliation(s)
- Ather Muneer
- Department of Psychiatry, Islamic International Medical College, Riphah International University, Rawalpindi, Pakistan
| |
Collapse
|
25
|
Györffy BA, Gulyássy P, Gellén B, Völgyi K, Madarasi D, Kis V, Ozohanics O, Papp I, Kovács P, Lubec G, Dobolyi Á, Kardos J, Drahos L, Juhász G, Kékesi KA. Widespread alterations in the synaptic proteome of the adolescent cerebral cortex following prenatal immune activation in rats. Brain Behav Immun 2016; 56:289-309. [PMID: 27058163 DOI: 10.1016/j.bbi.2016.04.002] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/27/2016] [Revised: 03/23/2016] [Accepted: 04/04/2016] [Indexed: 01/08/2023] Open
Abstract
An increasing number of studies have revealed associations between pre- and perinatal immune activation and the development of schizophrenia and autism spectrum disorders (ASDs). Accordingly, neuroimmune crosstalk has a considerably large impact on brain development during early ontogenesis. While a plethora of heterogeneous abnormalities have already been described in established maternal immune activation (MIA) rodent and primate animal models, which highly correlate to those found in human diseases, the underlying molecular background remains obscure. In the current study, we describe the long-term effects of MIA on the neocortical pre- and postsynaptic proteome of adolescent rat offspring in detail. Molecular differences were revealed in sub-synaptic fractions, which were first thoroughly characterized using independent methods. The widespread proteomic examination of cortical samples from offspring exposed to maternal lipopolysaccharide administration at embryonic day 13.5 was conducted via combinations of different gel-based proteomic techniques and tandem mass spectrometry. Our experimentally validated proteomic data revealed more pre- than postsynaptic protein level changes in the offspring. The results propose the relevance of altered synaptic vesicle recycling, cytoskeletal structure and energy metabolism in the presynaptic region in addition to alterations in vesicle trafficking, the cytoskeleton and signal transduction in the postsynaptic compartment in MIA offspring. Differing levels of the prominent signaling regulator molecule calcium/calmodulin-dependent protein kinase II in the postsynapse was validated and identified specifically in the prefrontal cortex. Finally, several potential common molecular regulators of these altered proteins, which are already known to be implicated in schizophrenia and ASD, were identified and assessed. In summary, unexpectedly widespread changes in the synaptic molecular machinery in MIA rats were demonstrated which might underlie the pathological cortical functions that are characteristic of schizophrenia and ASD.
Collapse
Affiliation(s)
- Balázs A Györffy
- Laboratory of Proteomics, Institute of Biology, Eötvös Loránd University, Budapest H-1117, Hungary; MTA-ELTE NAP B Neuroimmunology Research Group, Department of Biochemistry, Institute of Biology, Eötvös Loránd University, Budapest H-1117, Hungary
| | - Péter Gulyássy
- MTA-TTK NAP B MS Neuroproteomics Group, Hungarian Academy of Sciences, Budapest H-1117, Hungary; Department of Pediatrics, Medical University of Vienna, Vienna A-1090, Austria
| | - Barbara Gellén
- Laboratory of Proteomics, Institute of Biology, Eötvös Loránd University, Budapest H-1117, Hungary; MTA-ELTE NAP B Laboratory of Molecular and Systems Neurobiology, Institute of Biology, Hungarian Academy of Sciences and Eötvös Loránd University, Budapest H-1117, Hungary
| | - Katalin Völgyi
- Laboratory of Proteomics, Institute of Biology, Eötvös Loránd University, Budapest H-1117, Hungary; MTA-ELTE NAP B Laboratory of Molecular and Systems Neurobiology, Institute of Biology, Hungarian Academy of Sciences and Eötvös Loránd University, Budapest H-1117, Hungary
| | - Dóra Madarasi
- Laboratory of Proteomics, Institute of Biology, Eötvös Loránd University, Budapest H-1117, Hungary
| | - Viktor Kis
- Department of Anatomy, Cell and Developmental Biology, Institute of Biology, Eötvös Loránd University, Budapest H-1117, Hungary
| | - Olivér Ozohanics
- MTA-TTK NAP B MS Neuroproteomics Group, Hungarian Academy of Sciences, Budapest H-1117, Hungary
| | | | | | - Gert Lubec
- Department of Pediatrics, Medical University of Vienna, Vienna A-1090, Austria
| | - Árpád Dobolyi
- MTA-ELTE NAP B Laboratory of Molecular and Systems Neurobiology, Institute of Biology, Hungarian Academy of Sciences and Eötvös Loránd University, Budapest H-1117, Hungary
| | - József Kardos
- MTA-ELTE NAP B Neuroimmunology Research Group, Department of Biochemistry, Institute of Biology, Eötvös Loránd University, Budapest H-1117, Hungary
| | - László Drahos
- MTA-TTK NAP B MS Neuroproteomics Group, Hungarian Academy of Sciences, Budapest H-1117, Hungary
| | - Gábor Juhász
- Laboratory of Proteomics, Institute of Biology, Eötvös Loránd University, Budapest H-1117, Hungary; MTA-TTK NAP B MS Neuroproteomics Group, Hungarian Academy of Sciences, Budapest H-1117, Hungary
| | - Katalin A Kékesi
- Laboratory of Proteomics, Institute of Biology, Eötvös Loránd University, Budapest H-1117, Hungary; Department of Physiology and Neurobiology, Institute of Biology, Eötvös Loránd University, Budapest H-1117, Hungary.
| |
Collapse
|
26
|
Wen Y, Alshikho MJ, Herbert MR. Pathway Network Analyses for Autism Reveal Multisystem Involvement, Major Overlaps with Other Diseases and Convergence upon MAPK and Calcium Signaling. PLoS One 2016; 11:e0153329. [PMID: 27055244 PMCID: PMC4824422 DOI: 10.1371/journal.pone.0153329] [Citation(s) in RCA: 88] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2015] [Accepted: 03/28/2016] [Indexed: 01/05/2023] Open
Abstract
We used established databases in standard ways to systematically characterize gene ontologies, pathways and functional linkages in the large set of genes now associated with autism spectrum disorders (ASDs). These conditions are particularly challenging—they lack clear pathognomonic biological markers, they involve great heterogeneity across multiple levels (genes, systemic biological and brain characteristics, and nuances of behavioral manifestations)—and yet everyone with this diagnosis meets the same defining behavioral criteria. Using the human gene list from Simons Foundation Autism Research Initiative (SFARI) we performed gene set enrichment analysis with the Kyoto Encyclopedia of Genes and Genomes (KEGG) Pathway Database, and then derived a pathway network from pathway-pathway functional interactions again in reference to KEGG. Through identifying the GO (Gene Ontology) groups in which SFARI genes were enriched, mapping the coherence between pathways and GO groups, and ranking the relative strengths of representation of pathway network components, we 1) identified 10 disease-associated and 30 function-associated pathways 2) revealed calcium signaling pathway and neuroactive ligand-receptor interaction as the most enriched, statistically significant pathways from the enrichment analysis, 3) showed calcium signaling pathways and MAPK signaling pathway to be interactive hubs with other pathways and also to be involved with pervasively present biological processes, 4) found convergent indications that the process “calcium-PRC (protein kinase C)-Ras-Raf-MAPK/ERK” is likely a major contributor to ASD pathophysiology, and 5) noted that perturbations associated with KEGG’s category of environmental information processing were common. These findings support the idea that ASD-associated genes may contribute not only to core features of ASD themselves but also to vulnerability to other chronic and systemic problems potentially including cancer, metabolic conditions and heart diseases. ASDs may thus arise, or emerge, from underlying vulnerabilities related to pleiotropic genes associated with pervasively important molecular mechanisms, vulnerability to environmental input and multiple systemic co-morbidities.
Collapse
Affiliation(s)
- Ya Wen
- TRANSCEND Research, Neurology Department, Massachusetts General Hospital, Charlestown, Massachusetts, United States of America
- Harvard Medical School, Harvard University, Boston, Massachusetts, United States of America
- Higher Synthesis Foundation, Cambridge, Massachusetts, United States of America
- * E-mail: (YW); (MRH)
| | - Mohamad J. Alshikho
- TRANSCEND Research, Neurology Department, Massachusetts General Hospital, Charlestown, Massachusetts, United States of America
- Harvard Medical School, Harvard University, Boston, Massachusetts, United States of America
| | - Martha R. Herbert
- TRANSCEND Research, Neurology Department, Massachusetts General Hospital, Charlestown, Massachusetts, United States of America
- Harvard Medical School, Harvard University, Boston, Massachusetts, United States of America
- Higher Synthesis Foundation, Cambridge, Massachusetts, United States of America
- * E-mail: (YW); (MRH)
| |
Collapse
|
27
|
Soueid J, Kourtian S, Makhoul NJ, Makoukji J, Haddad S, Ghanem SS, Kobeissy F, Boustany RM. RYR2, PTDSS1 and AREG genes are implicated in a Lebanese population-based study of copy number variation in autism. Sci Rep 2016; 6:19088. [PMID: 26742492 PMCID: PMC4705475 DOI: 10.1038/srep19088] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2015] [Accepted: 12/02/2015] [Indexed: 11/11/2022] Open
Abstract
Autism Spectrum Disorders (ASDs) are a group of neurodevelopmental disorders characterized by ritualistic-repetitive behaviors and impaired verbal and non-verbal communication. Objectives were to determine the contribution of genetic variation to ASDs in the Lebanese. Affymetrix Cytogenetics Whole-Genome 2.7 M and CytoScan(™) HD Arrays were used to detect CNVs in 41 Lebanese autistic children and 35 non-autistic, developmentally delayed and intellectually disabled patients. 33 normal participants were used as controls. 16 de novo CNVs and 57 inherited CNVs, including recognized pathogenic 16p11.2 duplications and 2p16.3 deletions were identified. A duplication at 1q43 classified as likely pathogenic encompasses RYR2 as a potential ASD candidate gene. This previously identified CNV has been classified as both pathogenic, and, of uncertain significance. A duplication of unknown significance at 10q11.22, proposed as a modulator for phenotypic disease expression in Rett syndrome, was also identified. The novel potential autism susceptibility genes PTDSS1 and AREG were uncovered and warrant further genetic and functional analyses. Previously described and novel genetic targets in ASD were identified in Lebanese families with autism. These findings may lead to improved diagnosis of ASDs and informed genetic counseling, and may also lead to untapped therapeutic targets applicable to Lebanese and non-Lebanese patients.
Collapse
MESH Headings
- Adolescent
- Amphiregulin/deficiency
- Amphiregulin/genetics
- Autism Spectrum Disorder/genetics
- Autism Spectrum Disorder/physiopathology
- Case-Control Studies
- Child
- Child, Preschool
- Chromosome Aberrations
- Chromosomes, Human, Pair 1
- Chromosomes, Human, Pair 10
- Chromosomes, Human, Pair 16
- Chromosomes, Human, Pair 2
- DNA Copy Number Variations
- Developmental Disabilities/genetics
- Developmental Disabilities/physiopathology
- Female
- Gene Deletion
- Genome, Human
- Humans
- Lebanon
- Male
- Nitrogenous Group Transferases/deficiency
- Nitrogenous Group Transferases/genetics
- Ryanodine Receptor Calcium Release Channel/deficiency
- Ryanodine Receptor Calcium Release Channel/genetics
Collapse
Affiliation(s)
- Jihane Soueid
- American University of Beirut Medical Center Special Kids Clinic, Neurogenetics Program and Division of Pediatric Neurology, Departments of Pediatrics and Adolescent Medicine, Lebanon
| | - Silva Kourtian
- American University of Beirut Medical Center Special Kids Clinic, Neurogenetics Program and Division of Pediatric Neurology, Departments of Pediatrics and Adolescent Medicine, Lebanon
- Department of Biological and Environmental Sciences, Faculty of Science, Beirut Arab University, Lebanon
| | - Nadine J. Makhoul
- American University of Beirut Medical Center Special Kids Clinic, Neurogenetics Program and Division of Pediatric Neurology, Departments of Pediatrics and Adolescent Medicine, Lebanon
| | - Joelle Makoukji
- American University of Beirut Medical Center Special Kids Clinic, Neurogenetics Program and Division of Pediatric Neurology, Departments of Pediatrics and Adolescent Medicine, Lebanon
| | - Sariah Haddad
- American University of Beirut Medical Center Special Kids Clinic, Neurogenetics Program and Division of Pediatric Neurology, Departments of Pediatrics and Adolescent Medicine, Lebanon
| | - Simona S. Ghanem
- American University of Beirut Medical Center Special Kids Clinic, Neurogenetics Program and Division of Pediatric Neurology, Departments of Pediatrics and Adolescent Medicine, Lebanon
| | - Firas Kobeissy
- Biochemistry and Molecular Genetics, American university of Beirut, Lebanon
| | - Rose-Mary Boustany
- American University of Beirut Medical Center Special Kids Clinic, Neurogenetics Program and Division of Pediatric Neurology, Departments of Pediatrics and Adolescent Medicine, Lebanon
- Biochemistry and Molecular Genetics, American university of Beirut, Lebanon
| |
Collapse
|
28
|
Ehrlich DE, Josselyn SA. Plasticity-related genes in brain development and amygdala-dependent learning. GENES BRAIN AND BEHAVIOR 2015; 15:125-43. [PMID: 26419764 DOI: 10.1111/gbb.12255] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/07/2015] [Revised: 09/12/2015] [Accepted: 09/14/2015] [Indexed: 12/31/2022]
Abstract
Learning about motivationally important stimuli involves plasticity in the amygdala, a temporal lobe structure. Amygdala-dependent learning involves a growing number of plasticity-related signaling pathways also implicated in brain development, suggesting that learning-related signaling in juveniles may simultaneously influence development. Here, we review the pleiotropic functions in nervous system development and amygdala-dependent learning of a signaling pathway that includes brain-derived neurotrophic factor (BDNF), extracellular signaling-related kinases (ERKs) and cyclic AMP-response element binding protein (CREB). Using these canonical, plasticity-related genes as an example, we discuss the intersection of learning-related and developmental plasticity in the immature amygdala, when aversive and appetitive learning may influence the developmental trajectory of amygdala function. We propose that learning-dependent activation of BDNF, ERK and CREB signaling in the immature amygdala exaggerates and accelerates neural development, promoting amygdala excitability and environmental sensitivity later in life.
Collapse
Affiliation(s)
- D E Ehrlich
- Department of Neuroscience and Physiology, Neuroscience Institute, NYU Langone Medical Center, New York, NY, USA.,Department of Otolaryngology, NYU Langone School of Medicine, New York, NY, USA
| | - S A Josselyn
- Program in Neurosciences & Mental Health, Hospital for Sick Children, Toronto, ON, Canada.,Department of Psychology, University of Toronto, Toronto, ON, Canada.,Institute of Medical Sciences, University of Toronto, Toronto, ON, Canada.,Department of Physiology, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
29
|
Kripke DF, Elliott JA, Welsh DK, Youngstedt SD. Photoperiodic and circadian bifurcation theories of depression and mania. F1000Res 2015; 4:107. [PMID: 26180634 PMCID: PMC4490783 DOI: 10.12688/f1000research.6444.1] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 05/05/2015] [Indexed: 12/26/2022] Open
Abstract
Seasonal effects on mood have been observed throughout much of human history. Seasonal changes in animals and plants are largely mediated through the changing photoperiod (i.e., the photophase or duration of daylight). We review that in mammals, daylight specifically regulates SCN (suprachiasmatic nucleus) circadian organization and its control of melatonin secretion. The timing of melatonin secretion interacts with gene transcription in the pituitary pars tuberalis to modulate production of TSH (thyrotropin), hypothalamic T3 (triiodothyronine), and tuberalin peptides which modulate pituitary production of regulatory gonadotropins and other hormones. Pituitary hormones largely mediate seasonal physiologic and behavioral variations. As a result of long winter nights or inadequate illumination, we propose that delayed morning offset of nocturnal melatonin secretion, suppressing pars tuberalis function, could be the main cause for winter depression and even cause depressions at other times of year. Irregularities of circadian sleep timing and thyroid homeostasis contribute to depression. Bright light and sleep restriction are antidepressant and conversely, sometimes trigger mania. We propose that internal desynchronization or bifurcation of SCN circadian rhythms may underlie rapid-cycling manic-depressive disorders and perhaps most mania. Much further research will be needed to add substance to these theories.
Collapse
Affiliation(s)
- Daniel F Kripke
- Department of Psychiatry and Center for Circadian Biology, University of California, San Diego, CA, 92093-0603, USA
| | - Jeffrey A Elliott
- Department of Psychiatry and Center for Circadian Biology, University of California, San Diego, CA, 92093-0603, USA
| | - David K Welsh
- Department of Psychiatry and Center for Circadian Biology, University of California, San Diego, CA, 92093-0603, USA
| | - Shawn D Youngstedt
- College of Nursing and Health Innovation, Arizona State University, Phoenix, AZ, 85004-4431, USA
| |
Collapse
|
30
|
The 16p11.2 deletion mouse model of autism exhibits altered cortical progenitor proliferation and brain cytoarchitecture linked to the ERK MAPK pathway. J Neurosci 2015; 35:3190-200. [PMID: 25698753 DOI: 10.1523/jneurosci.4864-13.2015] [Citation(s) in RCA: 129] [Impact Index Per Article: 12.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Autism spectrum disorders are complex, highly heritable neurodevelopmental disorders affecting ∼1 in 100 children. Copy number variations of human chromosomal region 16p11.2 are genetically linked to 1% of autism-related disorders. This interval contains the MAPK3 gene, which encodes the MAP kinase, ERK1. Mutations in upstream elements regulating the ERK pathway are genetically linked to autism and other disorders of cognition including the neuro-cardio-facial cutaneous syndromes and copy number variations. We report that a murine model of human 16p11.2 deletion exhibits a reduction in brain size and perturbations in cortical cytoarchitecture. We observed enhanced progenitor proliferation and premature cell cycle exit, which are a consequence of altered levels of downstream ERK effectors cyclin D1 and p27(Kip1) during mid-neurogenesis. The increased progenitor proliferation and cell cycle withdrawal resulted in premature depletion of progenitor pools, altering the number and frequency of neurons ultimately populating cortical lamina. Specifically, we found a reduced number of upper layer pyramidal neurons and an increase in layer VI corticothalamic projection neurons, reflecting the altered cortical progenitor proliferation dynamics in these mice. Importantly, we observed a paradoxical increase in ERK signaling in mid-neurogenesis in the 16p11.2del mice, which is coincident with the development of aberrant cortical cytoarchitecture. The 16p11.2del mice exhibit anxiety-like behaviors and impaired memory. Our findings provide evidence of ERK dysregulation, developmental abnormalities in neurogenesis, and behavioral impairment associated with the 16p11.2 chromosomal deletion.
Collapse
|
31
|
BDNF stimulation of protein synthesis in cortical neurons requires the MAP kinase-interacting kinase MNK1. J Neurosci 2015; 35:972-84. [PMID: 25609615 DOI: 10.1523/jneurosci.2641-14.2015] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Although the MAP kinase-interacting kinases (MNKs) have been known for >15 years, their roles in the regulation of protein synthesis have remained obscure. Here, we explore the involvement of the MNKs in brain-derived neurotrophic factor (BDNF)-stimulated protein synthesis in cortical neurons from mice. Using a combination of pharmacological and genetic approaches, we show that BDNF-induced upregulation of protein synthesis requires MEK/ERK signaling and the downstream kinase, MNK1, which phosphorylates eukaryotic initiation factor (eIF) 4E. Translation initiation is mediated by the interaction of eIF4E with the m(7)GTP cap of mRNA and with eIF4G. The latter interaction is inhibited by the interactions of eIF4E with partner proteins, such as CYFIP1, which acts as a translational repressor. We find that BDNF induces the release of CYFIP1 from eIF4E, and that this depends on MNK1. Finally, using a novel combination of BONCAT and SILAC, we identify a subset of proteins whose synthesis is upregulated by BDNF signaling via MNK1 in neurons. Interestingly, this subset of MNK1-sensitive proteins is enriched for functions involved in neurotransmission and synaptic plasticity. Additionally, we find significant overlap between our subset of proteins whose synthesis is regulated by MNK1 and those encoded by known FMRP-binding mRNAs. Together, our data implicate MNK1 as a key component of BDNF-mediated translational regulation in neurons.
Collapse
|
32
|
Berbel P, Navarro D, Román GC. An evo-devo approach to thyroid hormones in cerebral and cerebellar cortical development: etiological implications for autism. Front Endocrinol (Lausanne) 2014; 5:146. [PMID: 25250016 PMCID: PMC4158880 DOI: 10.3389/fendo.2014.00146] [Citation(s) in RCA: 63] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/12/2014] [Accepted: 08/25/2014] [Indexed: 12/11/2022] Open
Abstract
The morphological alterations of cortical lamination observed in mouse models of developmental hypothyroidism prompted the recognition that these experimental changes resembled the brain lesions of children with autism; this led to recent studies showing that maternal thyroid hormone deficiency increases fourfold the risk of autism spectrum disorders (ASD), offering for the first time the possibility of prevention of some forms of ASD. For ethical reasons, the role of thyroid hormones on brain development is currently studied using animal models, usually mice and rats. Although mammals have in common many basic developmental principles regulating brain development, as well as fundamental basic mechanisms that are controlled by similar metabolic pathway activated genes, there are also important differences. For instance, the rodent cerebral cortex is basically a primary cortex, whereas the primary sensory areas in humans account for a very small surface in the cerebral cortex when compared to the associative and frontal areas that are more extensive. Associative and frontal areas in humans are involved in many neurological disorders, including ASD, attention deficit-hyperactive disorder, and dyslexia, among others. Therefore, an evo-devo approach to neocortical evolution among species is fundamental to understand not only the role of thyroid hormones and environmental thyroid disruptors on evolution, development, and organization of the cerebral cortex in mammals but also their role in neurological diseases associated to thyroid dysfunction.
Collapse
Affiliation(s)
- Pere Berbel
- Departamento de Histología y Anatomía, Facultad de Medicina, Universidad Miguel Hernández, Alicante, Spain
| | - Daniela Navarro
- Departamento de Histología y Anatomía, Facultad de Medicina, Universidad Miguel Hernández, Alicante, Spain
| | - Gustavo C. Román
- Department of Neurology, Weill Cornell Medical College, Cornell University, New York, NY, USA
- Methodist Neurological Institute, Houston, TX, USA
| |
Collapse
|
33
|
Del Giudice M, Klimczuk AC, Traficonte DM, Maestripieri D. Autistic-like and schizotypal traits in a life history perspective: diametrical associations with impulsivity, sensation seeking, and sociosexual behavior. EVOL HUM BEHAV 2014. [DOI: 10.1016/j.evolhumbehav.2014.05.007] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
|
34
|
|
35
|
Long-term memory deficits are associated with elevated synaptic ERK1/2 activation and reversed by mGluR5 antagonism in an animal model of autism. Neuropsychopharmacology 2014; 39:1664-73. [PMID: 24448645 PMCID: PMC4023139 DOI: 10.1038/npp.2014.13] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/17/2013] [Revised: 12/31/2013] [Accepted: 01/07/2014] [Indexed: 12/21/2022]
Abstract
A significant proportion of patients with autism exhibit some degree of intellectual disability. The BTBR T(+) Itpr3(tf)/J mouse strain exhibits behaviors that align with the major diagnostic criteria of autism. To further evaluate the BTBR strain's cognitive impairments, we quantified hippocampus-dependent object location memory (OLM) and found that one-third of the BTBR mice exhibited robust memory, whereas the remainder did not. Fluorescence deconvolution tomography was used to test whether synaptic levels of activated extracellular signal-regulated kinase 1/2 (ERK1/2), a protein that contributes importantly to plasticity, correlate with OLM scores in individual mice. In hippocampal field CA1, the BTBRs had fewer post-synaptic densities associated with high levels of phosphorylated (p-) ERK1/2 as compared with C57BL/6 mice. Although counts of p-ERK1/2 immunoreactive synapses did not correlate with OLM performance, the intensity of synaptic p-ERK1/2 immunolabeling was negatively correlated with OLM scores across BTBRs. Metabotropic glutamate receptor (mGluR) 5 signaling activates ERK1/2. Therefore, we tested whether treatment with the mGluR5 antagonist MPEP normalizes synaptic and learning measures in BTBR mice: MPEP facilitated OLM and decreased synaptic p-ERK1/2 immunolabeling intensity without affecting numbers of p-ERK1/2+ synapses. In contrast, semi-chronic ampakine treatment, which facilitates memory in other models of cognitive impairment, had no effect on OLM in BTBRs. These results suggest that intellectual disabilities associated with different neurodevelopmental disorders on the autism spectrum require distinct therapeutic strategies based on underlying synaptic pathology.
Collapse
|
36
|
Ríos P, Nunes-Xavier CE, Tabernero L, Köhn M, Pulido R. Dual-specificity phosphatases as molecular targets for inhibition in human disease. Antioxid Redox Signal 2014; 20:2251-73. [PMID: 24206177 DOI: 10.1089/ars.2013.5709] [Citation(s) in RCA: 59] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
SIGNIFICANCE The dual-specificity phosphatases (DUSPs) constitute a heterogeneous group of cysteine-based protein tyrosine phosphatases, whose members exert a pivotal role in cell physiology by dephosphorylation of phosphoserine, phosphothreonine, and phosphotyrosine residues from proteins, as well as other non-proteinaceous substrates. RECENT ADVANCES A picture is emerging in which a selected group of DUSP enzymes display overexpression or hyperactivity that is associated with human disease, especially human cancer, making feasible targeted therapy approaches based on their inhibition. A panoply of molecular and functional studies on DUSPs have been performed in the previous years, and drug-discovery efforts are ongoing to develop specific and efficient DUSP enzyme inhibitors. This review summarizes the current status on inhibitory compounds targeting DUSPs that belong to the MAP kinase phosphatases-, small-sized atypical-, and phosphatases of regenerating liver subfamilies, whose inhibition could be beneficial for the prevention or mitigation of human disease. CRITICAL ISSUES Achieving specificity, potency, and bioavailability are the major challenges in the discovery of DUSP inhibitors for the clinics. Clinical validation of compounds or alternative inhibitory strategies of DUSP inhibition has yet to come. FUTURE DIRECTIONS Further work is required to understand the dual role of many DUSPs in human cancer, their function-structure properties, and to identify their physiologic substrates. This will help in the implementation of therapies based on DUSPs inhibition.
Collapse
Affiliation(s)
- Pablo Ríos
- 1 Genome Biology Unit, European Molecular Biology Laboratory , Heidelberg, Germany
| | | | | | | | | |
Collapse
|
37
|
Thangavel M, Seelan RS, Lakshmanan J, Vadnal RE, Stagner JI, Parthasarathy LK, Casanova MF, El-Mallakh RS, Parthasarathy RN. Proteomic analysis of rat prefrontal cortex after chronic valproate treatment. J Neurosci Res 2014; 92:927-36. [DOI: 10.1002/jnr.23373] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2013] [Revised: 01/28/2014] [Accepted: 01/28/2014] [Indexed: 11/08/2022]
Affiliation(s)
- Muthusamy Thangavel
- Molecular Neuroscience and Bioinformatics Laboratories; Mental Health; Behavioral Science; and Research Services; Robley Rex Veterans Affairs Medical Center; Louisville Kentucky
- Department of Psychiatry and Behavioral Sciences; University of Louisville; Louisville Kentucky
| | - Ratnam S. Seelan
- Molecular Neuroscience and Bioinformatics Laboratories; Mental Health; Behavioral Science; and Research Services; Robley Rex Veterans Affairs Medical Center; Louisville Kentucky
- Department of Psychiatry and Behavioral Sciences; University of Louisville; Louisville Kentucky
- Department of Molecular; Cellular; and Craniofacial Biology; School of Dentistry, University of Louisville; Louisville Kentucky
| | - Jaganathan Lakshmanan
- Molecular Neuroscience and Bioinformatics Laboratories; Mental Health; Behavioral Science; and Research Services; Robley Rex Veterans Affairs Medical Center; Louisville Kentucky
- Price Institute of Surgical Research; Department of Surgery; School of Medicine, University of Louisville; Louisville Kentucky
| | - Robert E. Vadnal
- Eastern Colorado Health Care System; Department of Veterans Affairs; Pueblo Colorado
| | - John I. Stagner
- Molecular Neuroscience and Bioinformatics Laboratories; Mental Health; Behavioral Science; and Research Services; Robley Rex Veterans Affairs Medical Center; Louisville Kentucky
| | - Latha K. Parthasarathy
- Molecular Neuroscience and Bioinformatics Laboratories; Mental Health; Behavioral Science; and Research Services; Robley Rex Veterans Affairs Medical Center; Louisville Kentucky
- Department of Psychiatry and Behavioral Sciences; University of Louisville; Louisville Kentucky
| | - Manuel F. Casanova
- Department of Psychiatry and Behavioral Sciences; University of Louisville; Louisville Kentucky
| | - Rifaat Shody El-Mallakh
- Department of Psychiatry and Behavioral Sciences; University of Louisville; Louisville Kentucky
| | - Ranga N. Parthasarathy
- Molecular Neuroscience and Bioinformatics Laboratories; Mental Health; Behavioral Science; and Research Services; Robley Rex Veterans Affairs Medical Center; Louisville Kentucky
- Department of Psychiatry and Behavioral Sciences; University of Louisville; Louisville Kentucky
- Department of Biochemistry and Molecular Biology; University of Louisville; Louisville Kentucky
| |
Collapse
|
38
|
Yin A, Qiu Y, Jia B, Song T, Yu Y, Alberts I, Zhong M. The developmental pattern of the RAS/RAF/Erk1/2 pathway in the BTBR autism mouse model. Int J Dev Neurosci 2014; 39:2-8. [PMID: 24631207 DOI: 10.1016/j.ijdevneu.2014.01.006] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2013] [Revised: 01/22/2014] [Accepted: 01/22/2014] [Indexed: 12/01/2022] Open
Abstract
BTBR mice exhibit several autistic-like behaviors and are currently used as a model for understanding mechanisms that may be responsible for the pathogenesis of autism. Ras/Raf/ERK1/2 signaling has been suggested to play an important role in neural development, learning, memory, and cognition. Two studies reported that a deletion of a locus on chromosome 16 containing the mitogen-activated protein kinase 3 (MAPK3) gene, which encodes ERK1, is associated with autism. In the present study, Ras/Raf/ERK1/2 signaling was found to be up-regulated in BTBR mice relative to matched control B6 mice, to further suggest involvement in the pathogenesis of autism. To further characterize the developmental pattern of Ras/Raf/ERK1/2 signaling, varying stages during development were sampled to reveal an up-regulation in newborn and 2-week old BTBR mice relative to age-matched B6 mice. By the age of 3-week, Ras/Raf/ERK1/2 signaling in the brain of BTBR mice was unaltered relative to B6 mice, with this trend maintained in 6-week samples. These results suggest that the alteration of Ras/Raf/ERK signaling in the early developmental stages in mice could contribute to the noted autistic phenotype. Furthermore, these findings support the value of BTBR mice to serve as a human analog for autistic etiological research and aid in a better understanding of the developmental mechanisms of autism.
Collapse
Affiliation(s)
- Ailan Yin
- Department of Obstetrics & Gynecology, Nanfang Hospital, Guangzhou, China; Southern Medical University, Guangzhou, China
| | - Yuwen Qiu
- Department of Obstetrics & Gynecology, Nanfang Hospital, Guangzhou, China
| | - Bei Jia
- Department of Obstetrics & Gynecology, Nanfang Hospital, Guangzhou, China
| | - Tianrong Song
- Department of Obstetrics & Gynecology, Nanfang Hospital, Guangzhou, China
| | - Yanhong Yu
- Southern Medical University, Guangzhou, China
| | - Ian Alberts
- Department of Natural Sciences, LarGuardia CC, CUNY, NY, NY 11101, USA
| | - Mei Zhong
- Department of Obstetrics & Gynecology, Nanfang Hospital, Guangzhou, China.
| |
Collapse
|
39
|
Yoo J, Bakes J, Bradley C, Collingridge GL, Kaang BK. Shank mutant mice as an animal model of autism. Philos Trans R Soc Lond B Biol Sci 2014; 369:20130143. [PMID: 24298145 PMCID: PMC3843875 DOI: 10.1098/rstb.2013.0143] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
In this review, we focus on the role of the Shank family of proteins in autism. In recent years, autism research has been flourishing. With genetic, molecular, imaging and electrophysiological studies being supported by behavioural studies using animal models, there is real hope that we may soon understand the fundamental pathology of autism. There is also genuine potential to develop a molecular-level pharmacological treatment that may be able to deal with the most severe symptoms of autism, and clinical trials are already underway. The Shank family of proteins has been strongly implicated as a contributing factor in autism in certain individuals and sits at the core of the alleged autistic pathway. Here, we analyse studies that relate Shank to autism and discuss what light this sheds on the possible causes of autism.
Collapse
Affiliation(s)
- Juyoun Yoo
- Department of Biological Sciences, Seoul National University, Gwanangno 599, Gwanak-gu, Seoul 151-747, SouthKorea
| | - Joseph Bakes
- Department of Brain and Cognitive Sciences, College of Natural Sciences, Seoul National University, Gwanangno 599, Gwanak-gu, Seoul 151-747, SouthKorea
| | - Clarrisa Bradley
- Department of Brain and Cognitive Sciences, College of Natural Sciences, Seoul National University, Gwanangno 599, Gwanak-gu, Seoul 151-747, SouthKorea
| | - Graham L. Collingridge
- Department of Brain and Cognitive Sciences, College of Natural Sciences, Seoul National University, Gwanangno 599, Gwanak-gu, Seoul 151-747, SouthKorea
- Centre for Synaptic Plasticity, University of Bristol, Whitson Street, Bristol BS1 3NY, UK
| | - Bong-Kiun Kaang
- Department of Biological Sciences, Seoul National University, Gwanangno 599, Gwanak-gu, Seoul 151-747, SouthKorea
- Department of Brain and Cognitive Sciences, College of Natural Sciences, Seoul National University, Gwanangno 599, Gwanak-gu, Seoul 151-747, SouthKorea
| |
Collapse
|
40
|
de Lacy N, King BH. Revisiting the relationship between autism and schizophrenia: toward an integrated neurobiology. Annu Rev Clin Psychol 2013; 9:555-87. [PMID: 23537488 DOI: 10.1146/annurev-clinpsy-050212-185627] [Citation(s) in RCA: 72] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Schizophrenia and autism have been linked since their earliest descriptions. Both are disorders of cerebral specialization originating in the embryonic period. Genetic, molecular, and cytologic research highlights a variety of shared contributory mechanisms that may lead to patterns of abnormal connectivity arising from altered development and topology. Overt behavioral pathology likely emerges during or after neurosensitive periods in which resource demands overwhelm system resources and the individual's ability to compensate using interregional activation fails. We are at the threshold of being able to chart autism and schizophrenia from the inside out. In so doing, the door is opened to the consideration of new therapeutics that are developed based upon molecular, synaptic, and systems targets common to both disorders.
Collapse
Affiliation(s)
- Nina de Lacy
- University of Washington and Seattle Children's Hospital, Seattle, Washington 98195, USA
| | | |
Collapse
|
41
|
Eishingdrelo H, Kongsamut S. Minireview: Targeting GPCR Activated ERK Pathways for Drug Discovery. Curr Chem Genom Transl Med 2013; 7:9-15. [PMID: 24396730 PMCID: PMC3854659 DOI: 10.2174/2213988501307010009] [Citation(s) in RCA: 66] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2013] [Revised: 04/08/2013] [Accepted: 04/15/2013] [Indexed: 02/07/2023] Open
Abstract
It has become clear in recent years that multiple signal transduction pathways are employed upon GPCR
activation. One of the major cellular effectors activated by GPCRs is extracellular signal-regulated kinase (ERK). Both G-protein and β-arrestin mediated signaling pathways can lead to ERK activation. However, depending on activation
pathway, the subcellular destination of activated ERK1/2 may be different. G-protein -dependent ERK activation results in the translocation of active ERK to the nucleus, whereas ERK activated via an arrestin-dependent mechanism remains largely in the cytoplasm. The subcellular location of activated ERK1/2 determines the downstream signaling cascade. Many substrates of ERK1/2 are found in the nucleus: nuclear transcription factors that participate in gene transcription, cell proliferation and differentiation. ERK1/2 substrates are also found in cytosol and other cellular organelles: they may play roles in translation, mitosis, apoptosis and cross-talk with other signaling pathways. Therefore, determining specific subcellular locations of activated ERK1/2 mediated by GPCR ligands would be important in correlating signaling pathways with cellular physiological functions. While GPCR-stimulated selective ERK pathway activation has been studied in several receptor systems, exploitation of these different signaling cascades for therapeutics has not yet been seriously
pursued. Many old drug candidates were identified from screens based on G-protein signaling assays, and their activity on β-arrestin signaling pathways being mostly unknown, especially regarding their subcellular ERK pathways. With today’s knowledge of complicated GPCR signaling pathways, drug discovery can no longer rely on single-pathway approaches. Since ERK activation is an important signaling pathway and associated with many physiological functions, targeting the ERK pathway, especially specific subcellular activation pathways should provide new avenues for GPCR drug discovery.
Collapse
Affiliation(s)
- Haifeng Eishingdrelo
- BioInvenu Corporation, 50 Williams Parkway, East Hanover, New Jersey, 07936, USA
| | | |
Collapse
|
42
|
Frizzo ME. Putative role of glycogen as a peripheral biomarker of GSK3β activity. Med Hypotheses 2013; 81:376-8. [PMID: 23809426 DOI: 10.1016/j.mehy.2013.05.020] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2013] [Accepted: 05/19/2013] [Indexed: 11/29/2022]
Abstract
Glycogen synthase kinase 3-β (GSK3β) has a pivotal role in several intracellular signaling cascades that are involved in gene transcription, cytoskeletal reorganization, energy metabolism, cell cycle regulation, and apoptosis. This kinase has pleiotropic functions, and the importance of its activity has recently been shown in neurons and platelets. In addition to its regulatory function in several physiological events, changes in GSK3β activity have been associated with many psychiatric and neurodegenerative illnesses, such as Alzheimer's disease, schizophrenia and autism-spectrum disorders. Beside the reports of its involvement in several pathologies, it has become increasingly apparent that GSK3β might be a common therapeutic target for different classes of psychiatric drugs, and also that the GSK3β ratio may be a useful parameter to determine the biochemical changes that might occur during antidepressant treatment. Although GSK3β is commonly described as a key enzyme in a plethora of signaling cascades, originally it was identified as playing an important role in the regulation of glycogen synthesis, given its ability to inactivate glycogen synthase (GS) by phosphorylation. Acting as a constitutively active kinase, GSK3β phosphorylates GS, which results in a decrease of glycogen production. GSK3β phosphorylation increases glycogen synthesis and storage, while its dephosphorylation decreases glycogen synthesis. Inactivation of GSK3β leads to dephosphorylation of GS and increase in glycogen synthesis in the adipose tissue, muscle and liver. Glycogen levels are reduced by antidepressant treatment, and this effect seems to be related to an effect of drugs on GSK3β activity. Peripherally, glycogen is also abundantly found in platelets, where it is considered a major energy source, required for a variety of its functions, including the release reaction. Recently, analysis of platelets from patients with late-life major depression showed that active forms of GSK3β expression were upregulated by continuous treatment with sertraline. Here, we hypothesized that the quantification of glycogen in platelets might be used as a peripheral biomarker of GSK3β activity. Since it has been recently demonstrated that the modulation of GSK3β activity causes changes in glycogen stores, the glycogen levels in platelets could be used to assay the effects of drugs that have this kinase as a target, or diseases where its activity is affected. In conclusion, we hypothesized that the determination of glycogen peripherally may be useful to indicate a change in the activity of this enzyme, providing a faster and non-invasive approach to guide the therapeutic procedures for the patient.
Collapse
Affiliation(s)
- Marcos Emilio Frizzo
- Department of Morphological Sciences, Universidade Federal do Rio Grande do Sul, Rua Sarmento Leite, 500, CEP 90050-170 Porto Alegre, Brazil.
| |
Collapse
|
43
|
Hendriks WJAJ, Pulido R. Protein tyrosine phosphatase variants in human hereditary disorders and disease susceptibilities. Biochim Biophys Acta Mol Basis Dis 2013; 1832:1673-96. [PMID: 23707412 DOI: 10.1016/j.bbadis.2013.05.022] [Citation(s) in RCA: 86] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2013] [Revised: 05/14/2013] [Accepted: 05/16/2013] [Indexed: 12/18/2022]
Abstract
Reversible tyrosine phosphorylation of proteins is a key regulatory mechanism to steer normal development and physiological functioning of multicellular organisms. Phosphotyrosine dephosphorylation is exerted by members of the super-family of protein tyrosine phosphatase (PTP) enzymes and many play such essential roles that a wide variety of hereditary disorders and disease susceptibilities in man are caused by PTP alleles. More than two decades of PTP research has resulted in a collection of PTP genetic variants with corresponding consequences at the molecular, cellular and physiological level. Here we present a comprehensive overview of these PTP gene variants that have been linked to disease states in man. Although the findings have direct bearing for disease diagnostics and for research on disease etiology, more work is necessary to translate this into therapies that alleviate the burden of these hereditary disorders and disease susceptibilities in man.
Collapse
Affiliation(s)
- Wiljan J A J Hendriks
- Department of Cell Biology, Radboud University Nijmegen Medical Centre, Nijmegen, The Netherlands.
| | | |
Collapse
|