1
|
Cui Y, Rolova T, Fagerholm SC. The role of integrins in brain health and neurodegenerative diseases. Eur J Cell Biol 2024; 103:151441. [PMID: 39002282 DOI: 10.1016/j.ejcb.2024.151441] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2024] [Revised: 06/28/2024] [Accepted: 07/02/2024] [Indexed: 07/15/2024] Open
Abstract
Integrins are heterodimeric membrane proteins expressed on the surface of most cells. They mediate adhesion and signaling processes relevant for a wealth of physiological processes, including nervous system development and function. Interestingly, integrins are also recognized therapeutic targets for inflammatory diseases, such as multiple sclerosis. Here, we discuss the role of integrins in brain development and function, as well as in neurodegenerative diseases affecting the brain (Alzheimer's disease, multiple sclerosis, stroke). Furthermore, we discuss therapeutic targeting of these adhesion receptors in inflammatory diseases of the brain.
Collapse
Affiliation(s)
- Yunhao Cui
- Molecular and Integrative Biosciences Research Programme, Faculty of Biological and Environmental Sciences, University of Helsinki, Helsinki 00790, Finland
| | - Taisia Rolova
- Neuroscience Center, HiLIFE, University of Helsinki, Helsinki 00290, Finland
| | - Susanna C Fagerholm
- Molecular and Integrative Biosciences Research Programme, Faculty of Biological and Environmental Sciences, University of Helsinki, Helsinki 00790, Finland.
| |
Collapse
|
2
|
Nautiyal H, Jaiswar A, Jha PK, Dwivedi S. Exploring key genes and pathways associated with sex differences in autism spectrum disorder: integrated bioinformatic analysis. Mamm Genome 2024; 35:280-295. [PMID: 38594551 DOI: 10.1007/s00335-024-10036-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2023] [Accepted: 02/20/2024] [Indexed: 04/11/2024]
Abstract
Autism spectrum disorder (ASD) is a heterogenous neurodevelopmental disorder marked by functional abnormalities in brain that causes social and linguistic difficulties. The incidence of ASD is more prevalent in males compared to females, but the underlying mechanism, as well as molecular indications for identifying sex-specific differences in ASD symptoms remain unknown. Thus, impacting the development of personalized strategy towards pharmacotherapy of ASD. The current study employs an integrated bioinformatic approach to investigate the genes and pathways uniquely associated with sex specific differences in autistic individuals. Based on microarray dataset (GSE6575) extracted from the gene expression omnibus, the dysregulated genes between the autistic and the neurotypical individuals for both sexes were identified. Gene set enrichment analysis was performed to ascertain biological activities linked to the dysregulated genes. Protein-protein interaction network analysis was carried out to identify hub genes. The identified hub genes were examined to determine their functions and involvement in the associated pathways using Enrichr. Additionally, hub genes were validated from autism-associated databases and the potential small molecules targeting the hub genes were identified. The present study utilized whole blood transcriptomic gene expression analysis data and identified 2211 and 958 differentially expressed unique genes in males and females respectively. The functional enrichment analysis revealed that male hub genes were functionally associated with RNA polymerase II mediated transcriptional regulation whereas female hub genes were involved in intracellular signal transduction and cell migration. The top male hub genes exhibited functional enrichment in tyrosine kinase signalling pathway. The pathway enrichment analysis of male hub genes indicates the enrichment of papillomavirus infection. Female hub genes were enriched in androgen receptor signalling pathway and functionally enriched in focal adhesion specific excision repair. Identified drug like candidates targeting these genes may serve as a potential sex specific therapeutics. Wortmannin for males, 5-Fluorouracil for females had the highest scores. Targeted and sex-specific pharmacotherapies may be created for the management of ASD. The current investigation identifies sex-specific molecular signatures derived from whole blood which may serve as a potential peripheral sex-specific biomarkers for ASD. The study also uncovers the possible pharmacological interventions against the selected genes/pathway, providing support in development of therapeutic strategies to mitigate ASD. However, experimental proofs on biological systems are warranted.
Collapse
Affiliation(s)
- Himani Nautiyal
- Department of Pharmaceutical Sciences, School of Health Sciences and Technology, UPES, Dehradun, 248001, India
| | - Akanksha Jaiswar
- Laboratory of Human Disease Multiomics, Mossakowski Medical Research Institute Polish Academy of Sciences, Warsaw, Poland
| | - Prabhash Kumar Jha
- Center for Excellence in Vascular Biology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Shubham Dwivedi
- Department of Pharmaceutical Sciences, School of Health Sciences and Technology, UPES, Dehradun, 248001, India.
| |
Collapse
|
3
|
Esposito D, Cruciani G, Zaccaro L, Di Carlo E, Spitoni GF, Manti F, Carducci C, Fiori E, Leuzzi V, Pascucci T. A Systematic Review on Autism and Hyperserotonemia: State-of-the-Art, Limitations, and Future Directions. Brain Sci 2024; 14:481. [PMID: 38790459 PMCID: PMC11119126 DOI: 10.3390/brainsci14050481] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2024] [Revised: 04/29/2024] [Accepted: 05/02/2024] [Indexed: 05/26/2024] Open
Abstract
Hyperserotonemia is one of the most studied endophenotypes in autism spectrum disorder (ASD), but there are still no unequivocal results about its causes or biological and behavioral outcomes. This systematic review summarizes the studies investigating the relationship between blood serotonin (5-HT) levels and ASD, comparing diagnostic tools, analytical methods, and clinical outcomes. A literature search on peripheral 5-HT levels and ASD was conducted. In total, 1104 publications were screened, of which 113 entered the present systematic review. Of these, 59 articles reported hyperserotonemia in subjects with ASD, and 26 presented correlations between 5-HT levels and ASD-core clinical outcomes. The 5-HT levels are increased in about half, and correlations between hyperserotonemia and clinical outcomes are detected in a quarter of the studies. The present research highlights a large amount of heterogeneity in this field, ranging from the characterization of ASD and control groups to diagnostic and clinical assessments, from blood sampling procedures to analytical methods, allowing us to delineate critical topics for future studies.
Collapse
Affiliation(s)
- Dario Esposito
- Department of Human Neuroscience, Unit of Child Neurology and Psychiatry, Sapienza University of Rome, Via dei Sabelli 108, 00185 Rome, Italy; (D.E.); (F.M.)
| | - Gianluca Cruciani
- Department of Dynamic and Clinical Psychology, and Health Studies, Sapienza University of Rome, Via degli Apuli 1, 00185 Rome, Italy; (G.C.); (G.F.S.)
| | - Laura Zaccaro
- Department of Psychology, Sapienza University, Via dei Marsi 78, 00185 Rome, Italy; (L.Z.); (T.P.)
| | - Emanuele Di Carlo
- Department of Experimental Medicine, Sapienza University, Viale del Policlinico 155, 00161 Rome, Italy; (E.D.C.); (C.C.)
| | - Grazia Fernanda Spitoni
- Department of Dynamic and Clinical Psychology, and Health Studies, Sapienza University of Rome, Via degli Apuli 1, 00185 Rome, Italy; (G.C.); (G.F.S.)
- Cognitive and Motor Rehabilitation and Neuroimaging Unit, IRCCS Fondazione Santa Lucia, Via Ardeatina 306-354, 00179 Rome, Italy
| | - Filippo Manti
- Department of Human Neuroscience, Unit of Child Neurology and Psychiatry, Sapienza University of Rome, Via dei Sabelli 108, 00185 Rome, Italy; (D.E.); (F.M.)
| | - Claudia Carducci
- Department of Experimental Medicine, Sapienza University, Viale del Policlinico 155, 00161 Rome, Italy; (E.D.C.); (C.C.)
| | - Elena Fiori
- Rome Technopole Foundation, P.le Aldo Moro, 5, 00185 Rome, Italy;
| | - Vincenzo Leuzzi
- Department of Human Neuroscience, Unit of Child Neurology and Psychiatry, Sapienza University of Rome, Via dei Sabelli 108, 00185 Rome, Italy; (D.E.); (F.M.)
| | - Tiziana Pascucci
- Department of Psychology, Sapienza University, Via dei Marsi 78, 00185 Rome, Italy; (L.Z.); (T.P.)
- Centro “Daniel Bovet”, Sapienza University, P.le Aldo Moro 5, 00185 Rome, Italy
- Fondazione Santa Lucia Istituto di Ricovero e Cura a Carattere Scientifico, Via Ardeatina 306, 00179 Rome, Italy
| |
Collapse
|
4
|
Ivanova M, Belaya I, Kucháriková N, de Sousa Maciel I, Saveleva L, Alatalo A, Juvonen I, Thind N, Andrès C, Lampinen R, Chew S, Kanninen KM. Upregulation of Integrin beta-3 in astrocytes upon Alzheimer's disease progression in the 5xFAD mouse model. Neurobiol Dis 2024; 191:106410. [PMID: 38220131 DOI: 10.1016/j.nbd.2024.106410] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2023] [Revised: 01/11/2024] [Accepted: 01/11/2024] [Indexed: 01/16/2024] Open
Abstract
Integrins are receptors that have been linked to various brain disorders, including Alzheimer's disease (AD), the most prevalent neurodegenerative disorder. While Integrin beta-3 (ITGB3) is known to participate in multiple cellular processes such as adhesion, migration, and signaling, its specific role in AD remains poorly understood, particularly in astrocytes, the main glial cell type in the brain. In this study, we investigated alterations in ITGB3 gene and protein expression during aging in different brain regions of the 5xFAD mouse model of AD and assessed the interplay between ITGB3 and astrocytes. Primary cultures from adult mouse brains were used to gain further insight into the connection between ITGB3 and amyloid beta (Aβ) in astrocytes. In vivo studies showed a correlation between ITGB3 and the astrocytic marker GFAP in the 5xFAD brains, indicating its association with reactive astrocytes. In vitro studies revealed increased gene expression of ITGB3 upon Aβ treatment. Our findings underscore the potential significance of ITGB3 in astrocyte reactivity in the context of Alzheimer's disease.
Collapse
Affiliation(s)
- Mariia Ivanova
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
| | - Irina Belaya
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
| | - Nina Kucháriková
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
| | - Izaque de Sousa Maciel
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
| | - Liudmila Saveleva
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
| | - Arto Alatalo
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
| | - Ilona Juvonen
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
| | - Navjot Thind
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
| | - Clarisse Andrès
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
| | - Riikka Lampinen
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
| | - Sweelin Chew
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
| | - Katja M Kanninen
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland.
| |
Collapse
|
5
|
He B, Wang Y, Li H, Huang Y. The role of integrin beta in schizophrenia: a preliminary exploration. CNS Spectr 2023; 28:561-570. [PMID: 36274632 DOI: 10.1017/s1092852922001080] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
Integrins are transmembrane heterodimeric (αβ) receptors that transduce mechanical signals between the extracellular milieu and the cell in a bidirectional manner. Extensive research has shown that the integrin beta (β) family is widely expressed in the brain and that they control various aspects of brain development and function. Schizophrenia is a relatively common neurological disorder of unknown etiology and has been found to be closely related to neurodevelopment and neurochemicals in neuropathological studies of schizophrenia. Here, we review literature from recent years that shows that schizophrenia involves multiple signaling pathways related to neuronal migration, axon guidance, cell adhesion, and actin cytoskeleton dynamics, and that dysregulation of these processes affects the normal function of neurons and synapses. In fact, alterations in integrin β structure, expression and signaling for neural circuits, cortex, and synapses are likely to be associated with schizophrenia. We explored several aspects of the possible association between integrin β and schizophrenia in an attempt to demonstrate the role of integrin β in schizophrenia, which may help to provide new insights into the study of the pathogenesis and treatment of schizophrenia.
Collapse
Affiliation(s)
- Binshan He
- Department of Blood Transfusion, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Yuhan Wang
- Department of Blood Transfusion, Ya'an People's Hospital, Ya'an, China
| | - Huang Li
- Department of Clinical Medicine, Southwest Medical University, Luzhou, China
| | - Yuanshuai Huang
- Department of Blood Transfusion, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| |
Collapse
|
6
|
Jaudon F, Thalhammer A, Zentilin L, Cingolani LA. CRISPR-mediated activation of autism gene Itgb3 restores cortical network excitability via mGluR5 signaling. MOLECULAR THERAPY - NUCLEIC ACIDS 2022; 29:462-480. [PMID: 36035754 PMCID: PMC9382421 DOI: 10.1016/j.omtn.2022.07.013] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/28/2021] [Accepted: 07/15/2022] [Indexed: 01/12/2023]
Abstract
Many mutations in autism spectrum disorder (ASD) affect a single allele, indicating a key role for gene dosage in ASD susceptibility. Recently, haplo-insufficiency of ITGB3, the gene encoding the extracellular matrix receptor β3 integrin, was associated with ASD. Accordingly, Itgb3 knockout (KO) mice exhibit autism-like phenotypes. The pathophysiological mechanisms of Itgb3 remain, however, unknown, and the potential of targeting this gene for developing ASD therapies uninvestigated. By combining molecular, biochemical, imaging, and pharmacological analyses, we establish that Itgb3 haplo-insufficiency impairs cortical network excitability by promoting extra-synaptic over synaptic signaling of the metabotropic glutamate receptor mGluR5, which is similarly dysregulated in fragile X syndrome, the most frequent monogenic form of ASD. To assess the therapeutic potential of regulating Itgb3 gene dosage, we implemented CRISPR activation and compared its efficacy with that of a pharmacological rescue strategy for fragile X syndrome. Correction of neuronal Itgb3 haplo-insufficiency by CRISPR activation rebalanced network excitability as effectively as blockade of mGluR5 with the selective antagonist MPEP. Our findings reveal an unexpected functional interaction between two ASD genes, thereby validating the pathogenicity of ITGB3 haplo-insufficiency. Further, they pave the way for exploiting CRISPR activation as gene therapy for normalizing gene dosage and network excitability in ASD.
Collapse
Affiliation(s)
- Fanny Jaudon
- Center for Synaptic Neuroscience and Technology (NSYN), Fondazione Istituto Italiano di Tecnologia (IIT), 16132 Genoa, Italy
- Department of Life Sciences, University of Trieste, 34127 Trieste, Italy
| | - Agnes Thalhammer
- Department of Life Sciences, University of Trieste, 34127 Trieste, Italy
- IRCCS Ospedale Policlinico San Martino, 16132 Genoa, Italy
| | - Lorena Zentilin
- AAV Vector Unit, International Centre for Genetic Engineering and Biotechnology (ICGEB), 34149 Trieste, Italy
| | - Lorenzo A. Cingolani
- Center for Synaptic Neuroscience and Technology (NSYN), Fondazione Istituto Italiano di Tecnologia (IIT), 16132 Genoa, Italy
- Department of Life Sciences, University of Trieste, 34127 Trieste, Italy
- Corresponding author Lorenzo A. Cingolani, Department of Life Sciences, University of Trieste, 34127 Trieste, Italy.
| |
Collapse
|
7
|
Iyshwarya B, Vajagathali M, Ramakrishnan V. Investigation of Genetic Polymorphism in Autism Spectrum Disorder: a Pathogenesis of the Neurodevelopmental Disorder. ADVANCES IN NEURODEVELOPMENTAL DISORDERS 2022; 6:136-146. [DOI: 10.1007/s41252-022-00251-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 03/12/2022] [Indexed: 12/07/2023]
|
8
|
Lopuch AJ, Swinehart BD, Widener EL, Holley ZL, Bland KM, Handwerk CJ, Brett CA, Cook HN, Kalinowski AR, Rodriguez HV, Song MI, Vidal GS. Integrin β3 in forebrain Emx1-expressing cells regulates repetitive self-grooming and sociability in mice. BMC Neurosci 2022; 23:12. [PMID: 35247972 PMCID: PMC8897866 DOI: 10.1186/s12868-022-00691-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2021] [Accepted: 01/28/2022] [Indexed: 12/02/2022] Open
Abstract
BACKGROUND Autism spectrum disorder (ASD) is characterized by repetitive behaviors, deficits in communication, and overall impaired social interaction. Of all the integrin subunit mutations, mutations in integrin β3 (Itgb3) may be the most closely associated with ASD. Integrin β3 is required for normal structural plasticity of dendrites and synapses specifically in excitatory cortical and hippocampal circuitry. However, the behavioral consequences of Itgb3 function in the forebrain have not been assessed. We tested the hypothesis that behaviors that are typically abnormal in ASD-such as self-grooming and sociability behaviors-are disrupted with conditional Itgb3 loss of function in forebrain circuitry in male and female mice. METHODS We generated male and female conditional knockouts (cKO) and conditional heterozygotes (cHET) of Itgb3 in excitatory neurons and glia that were derived from Emx1-expressing forebrain cells during development. We used several different assays to determine whether male and female cKO and cHET mice have repetitive self-grooming behaviors, anxiety-like behaviors, abnormal locomotion, compulsive-like behaviors, or abnormal social behaviors, when compared to male and female wildtype (WT) mice. RESULTS Our findings indicate that only self-grooming and sociability are altered in cKO, but not cHET or WT mice, suggesting that Itgb3 is specifically required in forebrain Emx1-expressing cells for normal repetitive self-grooming and social behaviors. Furthermore, in cKO (but not cHET or WT), we observed an interaction effect for sex and self-grooming environment and an interaction effect for sex and sociability test chamber. LIMITATIONS While this study demonstrated a role for forebrain Itgb3 in specific repetitive and social behaviors, it was unable to determine whether forebrain Itgb3 is required for a preference for social novelty, whether cHET are haploinsufficient with respect to repetitive self-grooming and social behaviors, or the nature of the interaction effect for sex and environment/chamber in affected behaviors of cKO. CONCLUSIONS Together, these findings strengthen the idea that Itgb3 has a specific role in shaping forebrain circuitry that is relevant to endophenotypes of autism spectrum disorder.
Collapse
Affiliation(s)
- Andrew J Lopuch
- Department of Biology, James Madison University, 951 Carrier Drive, Harrisonburg, VA, 22807, USA
| | - Brian D Swinehart
- Department of Biology, James Madison University, 951 Carrier Drive, Harrisonburg, VA, 22807, USA
| | - Eden L Widener
- Department of Biology, James Madison University, 951 Carrier Drive, Harrisonburg, VA, 22807, USA
| | - Z Logan Holley
- Department of Biology, James Madison University, 951 Carrier Drive, Harrisonburg, VA, 22807, USA
| | - Katherine M Bland
- Department of Biology, James Madison University, 951 Carrier Drive, Harrisonburg, VA, 22807, USA
| | - Christopher J Handwerk
- Department of Biology, James Madison University, 951 Carrier Drive, Harrisonburg, VA, 22807, USA
| | - Cooper A Brett
- Department of Biology, James Madison University, 951 Carrier Drive, Harrisonburg, VA, 22807, USA
| | - Hollyn N Cook
- Department of Biology, James Madison University, 951 Carrier Drive, Harrisonburg, VA, 22807, USA
| | - Anna R Kalinowski
- Department of Biology, James Madison University, 951 Carrier Drive, Harrisonburg, VA, 22807, USA
| | - Hilda V Rodriguez
- Department of Biology, James Madison University, 951 Carrier Drive, Harrisonburg, VA, 22807, USA
| | - M Irene Song
- Department of Biology, James Madison University, 951 Carrier Drive, Harrisonburg, VA, 22807, USA
| | - George S Vidal
- Department of Biology, James Madison University, 951 Carrier Drive, Harrisonburg, VA, 22807, USA.
| |
Collapse
|
9
|
Swinehart BD, Bland KM, Holley ZL, Lopuch AJ, Casey ZO, Handwerk CJ, Vidal GS. Integrin β3 organizes dendritic complexity of cerebral cortical pyramidal neurons along a tangential gradient. Mol Brain 2020; 13:168. [PMID: 33317577 PMCID: PMC7734815 DOI: 10.1186/s13041-020-00707-0] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2020] [Accepted: 12/01/2020] [Indexed: 11/10/2022] Open
Abstract
Dysfunctional dendritic arborization is a key feature of many developmental neurological disorders. Across various human brain regions, basal dendritic complexity is known to increase along a caudal-to-rostral gradient. We recently discovered that basal dendritic complexity of layer II/III cortical pyramidal neurons in the mouse increases along a caudomedial-to-rostrolateral gradient spanning multiple regions, but at the time, no molecules were known to regulate that exquisite pattern. Integrin subunits have been implicated in dendritic development, and the subunit with the strongest associations with autism spectrum disorder and intellectual disability is integrin β3 (Itgb3). In mice, global knockout of Itgb3 leads to autistic-like neuroanatomy and behavior. Here, we tested the hypothesis that Itgb3 is required for increasing dendritic complexity along the recently discovered tangential gradient among layer II/III cortical pyramidal neurons. We targeted a subset of layer II/III cortical pyramidal neurons for Itgb3 loss-of-function via Cre-loxP-mediated excision of Itgb3. We tracked the rostrocaudal and mediolateral position of the targeted neurons and reconstructed their dendritic arbors. In contrast to controls, the basal dendritic complexity of Itgb3 mutant neurons was not related to their cortical position. Basal dendritic complexity of mutant and control neurons differed because of overall changes in branch number across multiple branch orders (primary, secondary, etc.), rather than any changes in the average length at those branch orders. Furthermore, dendritic spine density was related to cortical position in control but not mutant neurons. Thus, the autism susceptibility gene Itgb3 is required for establishing a tangential pattern of basal dendritic complexity among layer II/III cortical pyramidal neurons, suggesting an early role for this molecule in the developing brain.
Collapse
Affiliation(s)
- Brian D Swinehart
- Department of Biology, James Madison University, 951 Carrier Drive, Harrisonburg, VA, 22801, USA
| | - Katherine M Bland
- Department of Biology, James Madison University, 951 Carrier Drive, Harrisonburg, VA, 22801, USA
| | - Z Logan Holley
- Department of Biology, James Madison University, 951 Carrier Drive, Harrisonburg, VA, 22801, USA
| | - Andrew J Lopuch
- Department of Biology, James Madison University, 951 Carrier Drive, Harrisonburg, VA, 22801, USA
| | - Zachary O Casey
- Department of Biology, James Madison University, 951 Carrier Drive, Harrisonburg, VA, 22801, USA
| | - Christopher J Handwerk
- Department of Biology, James Madison University, 951 Carrier Drive, Harrisonburg, VA, 22801, USA
| | - George S Vidal
- Department of Biology, James Madison University, 951 Carrier Drive, Harrisonburg, VA, 22801, USA.
| |
Collapse
|
10
|
Javier-Torrent M, Zimmer-Bensch G, Nguyen L. Mechanical Forces Orchestrate Brain Development. Trends Neurosci 2020; 44:110-121. [PMID: 33203515 DOI: 10.1016/j.tins.2020.10.012] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2020] [Revised: 10/05/2020] [Accepted: 10/26/2020] [Indexed: 02/06/2023]
Abstract
During brain development, progenitors generate successive waves of neurons that populate distinct cerebral regions, where they settle and differentiate within layers or nuclei. While migrating and differentiating, neurons are subjected to mechanical forces arising from the extracellular matrix, and their interaction with neighboring cells. Changes in brain biomechanical properties, during its formation or aging, are converted in neural cells by mechanotransduction into intracellular signals that control key neurobiological processes. Here, we summarize recent findings that support the contribution of mechanobiology to neurodevelopment, with focus on the cerebral cortex. Also discussed are the existing toolbox and emerging technologies made available to assess and manipulate the physical properties of neurons and their environment.
Collapse
Affiliation(s)
- Míriam Javier-Torrent
- GIGA Stem Cells, GIGA-Neurosciences, University of Liège, CHU Sart Tilman, Liège 4000, Belgium
| | | | - Laurent Nguyen
- GIGA Stem Cells, GIGA-Neurosciences, University of Liège, CHU Sart Tilman, Liège 4000, Belgium.
| |
Collapse
|
11
|
Chakraborty N, Hammamieh R, Gautam A, Miller SA, Condlin ML, Jett M, Scrimgeour AG. TBI weight-drop model with variable impact heights differentially perturbs hippocampus-cerebellum specific transcriptomic profile. Exp Neurol 2020; 335:113516. [PMID: 33172833 DOI: 10.1016/j.expneurol.2020.113516] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2020] [Revised: 09/28/2020] [Accepted: 10/21/2020] [Indexed: 01/14/2023]
Abstract
The degree of brain injury is the governing factor for the magnitude of the patient's psycho- and physiological deficits post-injury, and the associated long-term consequences. The present scaling method used to segregate the patients among mild, moderate and severe phases of traumatic brain injury (TBI) has major limitations; however, a more continuous stratification of TBI is still elusive. With the anticipation that differentiating molecular markers could be the backbone of a robust method to triage TBI, we used a modified closed-head injury (CHI) Marmarou model with two impact heights (IH). By definition, IH directly correlates with the impact force causing TBI. In our modified CHI model, the rat skull was fitted with a helmet to permit a diffuse axonal injury. With the frontal cortex as the focal point of injury, the adjacent brain regions (hippocampus, HC and cerebellum, CB) were susceptible to diffuse secondary shock injury. At 8 days post injury (po.i.), rats impacted by 120 cm IH (IH120) took a longer time to find an escape route in the Barnes maze as compared to those impacted by 100 cm IH (IH100). Using a time-resolved interrogation of the transcriptomic landscape of HC and CB tissues, we mined those genes that altered their regulations in correlation with the variable IHs. At 14 days po.i., when all rats demonstrated nearly normal visuomotor performance, the bio-functional analysis suggested an advanced healing mechanism in the HC of IH100 group. In contrast, the HC of IH120 group displayed a delayed healing with evidence of active cell death networks. Combining whole genome rat microarrays with behavioral analysis provided the insight of neuroprotective signals that could be the foundation of the next generation triage for TBI patients.
Collapse
Affiliation(s)
- Nabarun Chakraborty
- Geneva Foundation, Medical Readiness Systems Biology, Walter Reed Army Institute of Research, Silver Spring, MD 20910, United States of America; Medical Readiness Systems Biology, Walter Reed Army Institute of Research, Silver Spring, MD 20910, United States of America.
| | - Rasha Hammamieh
- Medical Readiness Systems Biology, Walter Reed Army Institute of Research, Silver Spring, MD 20910, United States of America
| | - Aarti Gautam
- Medical Readiness Systems Biology, Walter Reed Army Institute of Research, Silver Spring, MD 20910, United States of America
| | - Stacy-Ann Miller
- Medical Readiness Systems Biology, Walter Reed Army Institute of Research, Silver Spring, MD 20910, United States of America; ORISE, Medical Readiness Systems Biology, Walter Reed Army Institute of Research, Silver Spring, MD 20910, United States of America
| | - Michelle L Condlin
- Military Nutrition Division, US Army Research Institute of Environmental Medicine, 10 General Greene Ave, Bldg 42, Natick, MA 01760, United States of America
| | - Marti Jett
- Medical Readiness Systems Biology, Walter Reed Army Institute of Research, Silver Spring, MD 20910, United States of America
| | - Angus G Scrimgeour
- Military Nutrition Division, US Army Research Institute of Environmental Medicine, 10 General Greene Ave, Bldg 42, Natick, MA 01760, United States of America
| |
Collapse
|
12
|
Jaudon F, Thalhammer A, Cingolani LA. Integrin adhesion in brain assembly: From molecular structure to neuropsychiatric disorders. Eur J Neurosci 2020; 53:3831-3850. [PMID: 32531845 DOI: 10.1111/ejn.14859] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2020] [Revised: 05/21/2020] [Accepted: 06/02/2020] [Indexed: 02/07/2023]
Abstract
Integrins are extracellular matrix receptors that mediate biochemical and mechanical bi-directional signals between the extracellular and intracellular environment of a cell thanks to allosteric conformational changes. In the brain, they are found in both neurons and glial cells, where they play essential roles in several aspects of brain development and function, such as cell migration, axon guidance, synaptogenesis, synaptic plasticity and neuro-inflammation. Although there are many successful examples of how regulating integrin adhesion and signaling can be used for therapeutic purposes, for example for halting tumor progression, this is not the case for the brain, where the growing evidence of the importance of integrins for brain pathophysiology has not translated yet into medical applications. Here, we review recent literature showing how alterations in integrin structure, expression and signaling may be involved in the etiology of autism spectrum disorder, epilepsy, schizophrenia, addiction, depression and Alzheimer's disease. We focus on common mechanisms and recurrent signaling pathways, trying to bridge studies on the genetics and molecular structure of integrins with those on synaptic physiology and brain pathology. Further, we discuss integrin-targeting strategies and their potential benefits for therapeutic purposes in neuropsychiatric disorders.
Collapse
Affiliation(s)
- Fanny Jaudon
- Center for Synaptic Neuroscience and Technology, Istituto Italiano di Tecnologia (IIT), Genoa, Italy.,IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| | - Agnes Thalhammer
- Center for Synaptic Neuroscience and Technology, Istituto Italiano di Tecnologia (IIT), Genoa, Italy.,IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| | - Lorenzo A Cingolani
- Center for Synaptic Neuroscience and Technology, Istituto Italiano di Tecnologia (IIT), Genoa, Italy.,Department of Life Sciences, University of Trieste, Trieste, Italy
| |
Collapse
|
13
|
Lew CH, Groeniger KM, Hanson KL, Cuevas D, Greiner DMZ, Hrvoj-Mihic B, Bellugi U, Schumann CM, Semendeferi K. Serotonergic innervation of the amygdala is increased in autism spectrum disorder and decreased in Williams syndrome. Mol Autism 2020; 11:12. [PMID: 32024554 PMCID: PMC7003328 DOI: 10.1186/s13229-019-0302-4] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2019] [Accepted: 12/04/2019] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Williams syndrome (WS) and autism spectrum disorder (ASD) are neurodevelopmental disorders that demonstrate overlapping genetic associations, dichotomous sociobehavioral phenotypes, and dichotomous pathological differences in neuronal distribution in key social brain areas, including the prefrontal cortex and the amygdala. The serotonergic system is critical to many processes underlying neurodevelopment and is additionally an important neuromodulator associated with behavioral variation. The amygdala is heavily innervated by serotonergic projections, suggesting that the serotonergic system is a significant mediator of neuronal activity. Disruptions to the serotonergic system, and atypical structure and function of the amygdala, are implicated in both WS and ASD. METHODS We quantified the serotonergic axon density in the four major subdivisions of the amygdala in the postmortem brains of individuals diagnosed with ASD and WS and neurotypical (NT) brains. RESULTS We found opposing directions of change in serotonergic innervation in the two disorders, with ASD displaying an increase in serotonergic axons compared to NT and WS displaying a decrease. Significant differences (p < 0.05) were observed between WS and ASD data sets across multiple amygdala nuclei. LIMITATIONS This study is limited by the availability of human postmortem tissue. Small sample size is an unavoidable limitation of most postmortem human brain research and particularly postmortem research in rare disorders. CONCLUSIONS Differential alterations to serotonergic innervation of the amygdala may contribute to differences in sociobehavioral phenotype in WS and ASD. These findings will inform future work identifying targets for future therapeutics in these and other disorders characterized by atypical social behavior.
Collapse
Affiliation(s)
- C H Lew
- Department of Anthropology, University of California, San Diego, USA
| | - K M Groeniger
- Department of Anthropology, University of California, San Diego, USA
| | - K L Hanson
- Department of Anthropology, University of California, San Diego, USA
| | - D Cuevas
- Department of Biological Sciences, University of California, San Diego, USA
| | - D M Z Greiner
- Department of Biological Sciences, University of California, San Diego, USA
| | - B Hrvoj-Mihic
- Department of Anthropology, University of California, San Diego, USA
| | - U Bellugi
- Salk Institute for Biological Sciences, San Diego, USA
| | - C M Schumann
- Department of Psychiatry and Behavioral Sciences, University of California, Davis School of Medicine, the MIND Institute, Sacramento, USA
| | - K Semendeferi
- Department of Anthropology, University of California, San Diego, USA.
| |
Collapse
|
14
|
Gabriele S, Canali M, Lintas C, Sacco R, Tirindelli MC, Ricciardello A, Persico AM. Evidence that ITGB3 promoter variants increase serotonin blood levels by regulating platelet serotonin transporter trafficking. Hum Mol Genet 2019; 28:1153-1161. [PMID: 30535103 DOI: 10.1093/hmg/ddy421] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2018] [Revised: 11/29/2018] [Accepted: 11/30/2018] [Indexed: 12/30/2022] Open
Abstract
Elevated serotonin (5-HT) blood levels, the first biomarker identified in autism research, has been consistently found in 20-30% of patients with Autism Spectrum Disorder (ASD). Hyperserotonemia is mainly due to greater 5-HT uptake into platelets, mediated by the 5-HT transporter (SERT) located at the platelet plasma membrane. The protein complex involved in platelet SERT trafficking and externalization includes integrin β3, the beta subunit of the platelet membrane adhesive GP IIb/IIIa. Integrin β3 is encoded by the ITGB3 gene, previously identified as a quantitative trait locus (QTL) for 5-HT blood levels in ASD at single nucleotide polymorphism (SNP) rs2317385. The present study aims to identify the functional ITGB3 gene variants contributing to hyperserotonemia. ITGB3 gene sequencing in 20 individuals selected on the basis of rs2317385 genotypes defined four haplotypes encompassing six SNPs located in the ITGB3 gene promoter region, all in linkage disequilibrium with rs2317385. Luciferase assays in two hematopoietic cell lines, K-562 and HEL 92.1.7, demonstrate that ITGB3 gene promoter activity is enhanced by the presence of the C allele at rs55827077 specifically during differentiation into megakaryocytes (P < 0.01), with modulatory effects by flanking SNPs. This same allele is strongly associated with (a) higher 5-HT blood levels in 176 autistic individuals (P < 0.001), (b) greater platelet integrin β3 protein expression (P < 0.05) and (c) enhanced SERT trafficking from the cytosol toward the platelet plasma membrane (P = 4.05 × 10-11). Our results support rs55827077 as the functional ITGB3 gene promoter variant contributing to elevated 5-HT blood levels in ASD and define a mechanistic chain of events linking ITGB3 to hyperserotonemia.
Collapse
Affiliation(s)
- Stefano Gabriele
- Center for Neurodevelopmental Disorders & Laboratory of Molecular Psychiatry and Neurogenetics, Department of Medicine, University Campus Bio-Medico, Rome, Italy
| | - Marco Canali
- Center for Neurodevelopmental Disorders & Laboratory of Molecular Psychiatry and Neurogenetics, Department of Medicine, University Campus Bio-Medico, Rome, Italy
| | - Carla Lintas
- Center for Neurodevelopmental Disorders & Laboratory of Molecular Psychiatry and Neurogenetics, Department of Medicine, University Campus Bio-Medico, Rome, Italy
| | - Roberto Sacco
- Center for Neurodevelopmental Disorders & Laboratory of Molecular Psychiatry and Neurogenetics, Department of Medicine, University Campus Bio-Medico, Rome, Italy
| | | | - Arianna Ricciardello
- Interdepartmental Program "Autism 0-90", "Gaetano Martino" University Hospital, University of Messina, Messina, Italy
| | - Antonio M Persico
- Interdepartmental Program "Autism 0-90", "Gaetano Martino" University Hospital, University of Messina, Messina, Italy.,Mafalda Luce Center for Pervasive Developmental Disorders, Milan, Italy
| |
Collapse
|
15
|
Ellegood J, Yee Y, Kerr TM, Muller CL, Blakely RD, Henkelman RM, Veenstra-VanderWeele J, Lerch JP. Analysis of neuroanatomical differences in mice with genetically modified serotonin transporters assessed by structural magnetic resonance imaging. Mol Autism 2018; 9:24. [PMID: 29651330 PMCID: PMC5894125 DOI: 10.1186/s13229-018-0210-z] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2017] [Accepted: 03/21/2018] [Indexed: 02/03/2023] Open
Abstract
Background The serotonin (5-HT) system has long been implicated in autism spectrum disorder (ASD) as indicated by elevated whole blood and platelet 5-HT, altered platelet and brain receptor and transporter binding, and genetic linkage and association findings. Based upon work in genetically modified mice, 5-HT is known to influence several aspects of brain development, but systematic neuroimaging studies have not previously been reported. In particular, the 5-HT transporter (serotonin transporter, SERT; 5-HTT) gene, Slc6a4, has been extensively studied. Methods Using a 7-T MRI and deformation-based morphometry, we assessed neuroanatomical differences in an Slc6a4 knockout mouse on a C57BL/6 genetic background, along with an Slc6a4 Ala56 knockin mouse on two different genetic backgrounds (129S and C57BL/6). Results Individually (same sex, same background, same genotype), the only differences found were in the female Slc6a4 knockout mouse; all the others had no significant differences. However, an analysis of variance across the whole study sample revealed a significant effect of Slc6a4 on the amygdala, thalamus, dorsal raphe nucleus, and lateral and frontal cortices. Conclusions This work shows that an increase or decrease in SERT function has a significant effect on the neuroanatomy in 5-HT relevant regions, particularly the raphe nuclei. Notably, the Slc6a4 Ala56 knockin alone appears to have an insignificant, but suggestive, effect compared to the KO, which is consistent with Slc6a4 function. Despite the small number of 5-HT neurons and their localization to the brainstem, it is clear that 5-HT plays an important role in neuroanatomical organization. Electronic supplementary material The online version of this article (10.1186/s13229-018-0210-z) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Jacob Ellegood
- 1Mouse Imaging Centre (MICe), Hospital for Sick Children, 25 Orde Street, Toronto, Ontario M5T 3H7 Canada
| | - Yohan Yee
- 1Mouse Imaging Centre (MICe), Hospital for Sick Children, 25 Orde Street, Toronto, Ontario M5T 3H7 Canada.,4Department of Medical Biophysics, University of Toronto, Toronto, ON M5S Canada
| | - Travis M Kerr
- 3Department of Psychiatry, Vanderbilt University, Nashville, TN 37235 USA
| | | | - Randy D Blakely
- 2Department of Pharmacology, Vanderbilt University, Nashville, TN 37235 USA.,3Department of Psychiatry, Vanderbilt University, Nashville, TN 37235 USA.,5Department of Biomedical Science and Brain Institute, Florida Atlantic University, Jupiter, FL 33431 USA
| | - R Mark Henkelman
- 1Mouse Imaging Centre (MICe), Hospital for Sick Children, 25 Orde Street, Toronto, Ontario M5T 3H7 Canada.,4Department of Medical Biophysics, University of Toronto, Toronto, ON M5S Canada
| | - Jeremy Veenstra-VanderWeele
- 2Department of Pharmacology, Vanderbilt University, Nashville, TN 37235 USA.,6Department of Psychiatry, Columbia University, New York, NY 10027 USA
| | - Jason P Lerch
- 1Mouse Imaging Centre (MICe), Hospital for Sick Children, 25 Orde Street, Toronto, Ontario M5T 3H7 Canada.,4Department of Medical Biophysics, University of Toronto, Toronto, ON M5S Canada
| |
Collapse
|
16
|
Zhang N, Ma D, Wang L, Zhu X, Pan Q, Zhao Y, Zhu W, Zhou J, Wang L, Chai Z, Ao J, Sun H, Tang Z. Insufficient Radiofrequency Ablation Treated Hepatocellular Carcinoma Cells Promote Metastasis by Up-Regulation ITGB3. J Cancer 2017; 8:3742-3754. [PMID: 29151962 PMCID: PMC5688928 DOI: 10.7150/jca.20816] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2017] [Accepted: 08/16/2017] [Indexed: 12/27/2022] Open
Abstract
Radiofrequency ablation (RFA) is one of the standards of care for early stage hepatocellular carcinoma (HCC). However, rapid progression of residual tumor after RFA has been confirmed. The aim of this study was to investigate the underlying mechanism of this phenomenon. Human HCC cell lines HCCLM3 and HepG2 were employed to establish insufficient RFA models in vivo and in vitro, respectively. The effects of insufficient RFA on metastatic potential of residual tumors were evaluated. The molecular changes after insufficient RFA were evaluated by PCR array, western blot, immunofluorescence, and immunohistochemistry. Results showed that insufficient RFA significantly promoted lung and intrahepatic residual tumor cells in vivo, and heat intervention promoted migration and invasion of hepatoma cells in vitro. PCR array revealed that the expression of integrin β3 (ITGB3) and MMP2 were up-regulated in the residual tumors of HCCLM3 xenograft model. The up-regulation of ITGB3 was confirmed by qRT-PCR, Western blot and immunohistochemistry. Knockdown ITGB3 expression in HCCLM3 cells by shRNA significantly lowered the pro-metastatic effects of insufficient RFA. Mechanism studies indicated that ITGB3 mediated the expression of MMP2 by activing FAK/PI3K/AKT signaling pathway. The up-regulation of ITGB3 contributed to enhanced metastatic potential of residual cancer in HCCLM3 model after insufficient RFA. Targeting ITGB3 expression may further improve the clinical effects of RFA.
Collapse
Affiliation(s)
- Ning Zhang
- Department of Liver Surgery, Fudan University Shanghai Cancer Center, Cancer Hospital, Shanghai, China
| | - Dening Ma
- Department of Colorectal Cancer Surgery, Zhejiang Cancer Hospital, Hangzhou, Zhejiang, China
| | - Lu Wang
- Department of Liver Surgery, Fudan University Shanghai Cancer Center, Cancer Hospital, Shanghai, China
| | - Xiaodong Zhu
- Liver Cancer Institute, Zhongshan Hospital, Fudan University; Key Laboratory of Carcinogenesis and Cancer Invasion (Fudan University), Ministry of Education, Shanghai, China
| | - Qi Pan
- Department of Liver Surgery, Fudan University Shanghai Cancer Center, Cancer Hospital, Shanghai, China
| | - Yiming Zhao
- Department of Liver Surgery, Fudan University Shanghai Cancer Center, Cancer Hospital, Shanghai, China
| | - Weiping Zhu
- Department of Liver Surgery, Fudan University Shanghai Cancer Center, Cancer Hospital, Shanghai, China
| | - Jiamin Zhou
- Department of Liver Surgery, Fudan University Shanghai Cancer Center, Cancer Hospital, Shanghai, China
| | - Longrong Wang
- Department of Liver Surgery, Fudan University Shanghai Cancer Center, Cancer Hospital, Shanghai, China
| | - Zongtao Chai
- Department of Hepatic Surgery VI, Eastern Hepatobiliary Surgery Hospital, Second Military Medical University, Shanghai, China
| | - Jianyang Ao
- Department of Hepatobiliary Surgery, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Huichuan Sun
- Liver Cancer Institute, Zhongshan Hospital, Fudan University; Key Laboratory of Carcinogenesis and Cancer Invasion (Fudan University), Ministry of Education, Shanghai, China
| | - Zhaoyou Tang
- Liver Cancer Institute, Zhongshan Hospital, Fudan University; Key Laboratory of Carcinogenesis and Cancer Invasion (Fudan University), Ministry of Education, Shanghai, China
| |
Collapse
|
17
|
The Gain-of-Function Integrin β3 Pro33 Variant Alters the Serotonin System in the Mouse Brain. J Neurosci 2017; 37:11271-11284. [PMID: 29038237 DOI: 10.1523/jneurosci.1482-17.2017] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2017] [Revised: 08/21/2017] [Accepted: 08/22/2017] [Indexed: 12/26/2022] Open
Abstract
Engagement of integrins by the extracellular matrix initiates signaling cascades that drive a variety of cellular functions, including neuronal migration and axonal pathfinding in the brain. Multiple lines of evidence link the ITGB3 gene encoding the integrin β3 subunit with the serotonin (5-HT) system, likely via its modulation of the 5-HT transporter (SERT). The ITGB3 coding polymorphism Leu33Pro (rs5918, PlA2) produces hyperactive αvβ3 receptors that influence whole-blood 5-HT levels and may influence the risk for autism spectrum disorder (ASD). Using a phenome-wide scan of psychiatric diagnoses, we found significant, male-specific associations between the Pro33 allele and attention-deficit hyperactivity disorder and ASDs. Here, we used knock-in (KI) mice expressing an Itgb3 variant that phenocopies the human Pro33 variant to elucidate the consequences of constitutively enhanced αvβ3 signaling to the 5-HT system in the brain. KI mice displayed deficits in multiple behaviors, including anxiety, repetitive, and social behaviors. Anatomical studies revealed a significant decrease in 5-HT synapses in the midbrain, accompanied by decreases in SERT activity and reduced localization of SERTs to integrin adhesion complexes in synapses of KI mice. Inhibition of focal adhesion kinase (FAK) rescued SERT function in synapses of KI mice, demonstrating that constitutive active FAK signaling downstream of the Pro32Pro33 integrin αvβ3 suppresses SERT activity. Our studies identify a complex regulation of 5-HT homeostasis and behaviors by integrin αvβ3, revealing an important role for integrins in modulating risk for neuropsychiatric disorders.SIGNIFICANCE STATEMENT The integrin β3 Leu33Pro coding polymorphism has been associated with autism spectrum disorders (ASDs) within a subgroup of patients with elevated blood 5-HT levels, linking integrin β3, 5-HT, and ASD risk. We capitalized on these interactions to demonstrate that the Pro33 coding variation in the murine integrin β3 recapitulates the sex-dependent neurochemical and behavioral attributes of ASD. Using state-of-the-art techniques, we show that presynaptic 5-HT function is altered in these mice, and that the localization of 5-HT transporters to specific compartments within the synapse, disrupted by the integrin β3 Pro33 mutation, is critical for appropriate reuptake of 5-HT. Our studies provide fundamental insight into the genetic network regulating 5-HT neurotransmission in the CNS that is also associated with ASD risk.
Collapse
|
18
|
Reilly J, Gallagher L, Chen JL, Leader G, Shen S. Bio-collections in autism research. Mol Autism 2017; 8:34. [PMID: 28702161 PMCID: PMC5504648 DOI: 10.1186/s13229-017-0154-8] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2017] [Accepted: 06/23/2017] [Indexed: 01/06/2023] Open
Abstract
Autism spectrum disorder (ASD) is a group of complex neurodevelopmental disorders with diverse clinical manifestations and symptoms. In the last 10 years, there have been significant advances in understanding the genetic basis for ASD, critically supported through the establishment of ASD bio-collections and application in research. Here, we summarise a selection of major ASD bio-collections and their associated findings. Collectively, these include mapping ASD candidate genes, assessing the nature and frequency of gene mutations and their association with ASD clinical subgroups, insights into related molecular pathways such as the synapses, chromatin remodelling, transcription and ASD-related brain regions. We also briefly review emerging studies on the use of induced pluripotent stem cells (iPSCs) to potentially model ASD in culture. These provide deeper insight into ASD progression during development and could generate human cell models for drug screening. Finally, we provide perspectives concerning the utilities of ASD bio-collections and limitations, and highlight considerations in setting up a new bio-collection for ASD research.
Collapse
Affiliation(s)
- Jamie Reilly
- Regenerative Medicine Institute, School of Medicine, BioMedical Sciences Building, National University of Ireland (NUI), Galway, Ireland
| | - Louise Gallagher
- Trinity Translational Medicine Institute and Department of Psychiatry, Trinity Centre for Health Sciences, St. James Hospital Street, Dublin 8, Ireland
| | - June L. Chen
- Department of Special Education, Faculty of Education, East China Normal University, Shanghai, 200062 China
| | - Geraldine Leader
- Irish Centre for Autism and Neurodevelopmental Research (ICAN), Department of Psychology, National University of Ireland Galway, University Road, Galway, Ireland
| | - Sanbing Shen
- Regenerative Medicine Institute, School of Medicine, BioMedical Sciences Building, National University of Ireland (NUI), Galway, Ireland
| |
Collapse
|
19
|
Mariath LM, Silva AMD, Kowalski TW, Gattino GS, Araujo GAD, Figueiredo FG, Tagliani-Ribeiro A, Roman T, Vianna FSL, Schuler-Faccini L, Schuch JB. Music genetics research: Association with musicality of a polymorphism in the AVPR1A gene. Genet Mol Biol 2017; 40:421-429. [PMID: 28534928 PMCID: PMC5488451 DOI: 10.1590/1678-4685-gmb-2016-0021] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2016] [Accepted: 12/15/2016] [Indexed: 01/01/2023] Open
Abstract
Musicality is defined as a natural tendency, sensibility, knowledge, or talent to
create, perceive, and play music. Musical abilities involve a great range of social
and cognitive behaviors, which are influenced by both environmental and genetic
factors. Although a number of studies have yielded insights into music genetics
research, genes and biological pathways related to these traits are not fully
understood. Our hypothesis in the current study is that genes associated with
different behaviors could also influence the musical phenotype. Our aim was to
investigate whether polymorphisms in six genes (AVPR1A, SLC6A4, ITGB3, COMT, DRD2 and
DRD4) related to social and cognitive traits are associated with musicality in a
sample of children. Musicality was assessed through an individualized music therapy
assessment profile (IMTAP) which has been validated in Brazil to measure musical
ability. We show here that the RS1 microsatellite of the AVPR1A gene is nominally
associated with musicality, corroborating previous results linking AVPR1A with
musical activity. This study is one of the first to investigate musicality in a
comprehensive way, and it contributes to better understand the genetic basis
underlying musical ability.
Collapse
Affiliation(s)
- Luiza Monteavaro Mariath
- Programa de Pós-Graduação em Genética e Biologia Molecular, Departamento de Genética, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Alexandre Mauat da Silva
- Programa de Pós-Graduação em Genética e Biologia Molecular, Departamento de Genética, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Thayne Woycinck Kowalski
- Programa de Pós-Graduação em Genética e Biologia Molecular, Departamento de Genética, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Gustavo Schulz Gattino
- Programa de Pos-Graduação em Saúde da Criança e Adolescente, Faculdade de Medicina, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Gustavo Andrade de Araujo
- Programa de Pos-Graduação em Saúde da Criança e Adolescente, Faculdade de Medicina, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Felipe Grahl Figueiredo
- Programa de Pos-Graduação em Saúde da Criança e Adolescente, Faculdade de Medicina, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Alice Tagliani-Ribeiro
- Programa de Pós-Graduação em Genética e Biologia Molecular, Departamento de Genética, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Tatiana Roman
- Programa de Pós-Graduação em Genética e Biologia Molecular, Departamento de Genética, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Fernanda Sales Luiz Vianna
- Programa de Pós-Graduação em Genética e Biologia Molecular, Departamento de Genética, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Lavínia Schuler-Faccini
- Programa de Pós-Graduação em Genética e Biologia Molecular, Departamento de Genética, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Jaqueline Bohrer Schuch
- Programa de Pós-Graduação em Genética e Biologia Molecular, Departamento de Genética, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
| |
Collapse
|
20
|
Mitra I, Lavillaureix A, Yeh E, Traglia M, Tsang K, Bearden CE, Rauen KA, Weiss LA. Reverse Pathway Genetic Approach Identifies Epistasis in Autism Spectrum Disorders. PLoS Genet 2017; 13:e1006516. [PMID: 28076348 PMCID: PMC5226683 DOI: 10.1371/journal.pgen.1006516] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2016] [Accepted: 12/01/2016] [Indexed: 02/08/2023] Open
Abstract
Although gene-gene interaction, or epistasis, plays a large role in complex traits in model organisms, genome-wide by genome-wide searches for two-way interaction have limited power in human studies. We thus used knowledge of a biological pathway in order to identify a contribution of epistasis to autism spectrum disorders (ASDs) in humans, a reverse-pathway genetic approach. Based on previous observation of increased ASD symptoms in Mendelian disorders of the Ras/MAPK pathway (RASopathies), we showed that common SNPs in RASopathy genes show enrichment for association signal in GWAS (P = 0.02). We then screened genome-wide for interactors with RASopathy gene SNPs and showed strong enrichment in ASD-affected individuals (P < 2.2 x 10-16), with a number of pairwise interactions meeting genome-wide criteria for significance. Finally, we utilized quantitative measures of ASD symptoms in RASopathy-affected individuals to perform modifier mapping via GWAS. One top region overlapped between these independent approaches, and we showed dysregulation of a gene in this region, GPR141, in a RASopathy neural cell line. We thus used orthogonal approaches to provide strong evidence for a contribution of epistasis to ASDs, confirm a role for the Ras/MAPK pathway in idiopathic ASDs, and to identify a convergent candidate gene that may interact with the Ras/MAPK pathway.
Collapse
Affiliation(s)
- Ileena Mitra
- Department of Psychiatry, University of California San Francisco, San Francisco, California, United States of America
- Institute for Human Genetics, University of California San Francisco, San Francisco, California, United States of America
| | - Alinoë Lavillaureix
- Department of Psychiatry, University of California San Francisco, San Francisco, California, United States of America
- Institute for Human Genetics, University of California San Francisco, San Francisco, California, United States of America
- Université Paris Descartes, Sorbonne Paris Cité, Faculty of Medicine, Paris, France
| | - Erika Yeh
- Department of Psychiatry, University of California San Francisco, San Francisco, California, United States of America
- Institute for Human Genetics, University of California San Francisco, San Francisco, California, United States of America
| | - Michela Traglia
- Department of Psychiatry, University of California San Francisco, San Francisco, California, United States of America
- Institute for Human Genetics, University of California San Francisco, San Francisco, California, United States of America
| | - Kathryn Tsang
- Department of Psychiatry, University of California San Francisco, San Francisco, California, United States of America
- Institute for Human Genetics, University of California San Francisco, San Francisco, California, United States of America
| | - Carrie E. Bearden
- Department of Psychiatry and Biobehavioral Sciences, Semel Institute for Neuroscience and Human Behavior, University of California Los Angeles, Los Angeles, California, United States of America
- Department of Psychology, University of California Los Angeles, Los Angeles, California, United States of America
| | - Katherine A. Rauen
- Institute for Human Genetics, University of California San Francisco, San Francisco, California, United States of America
- Department of Pediatrics, School of Medicine, University of California San Francisco, San Francisco, California, United States of America
| | - Lauren A. Weiss
- Department of Psychiatry, University of California San Francisco, San Francisco, California, United States of America
- Institute for Human Genetics, University of California San Francisco, San Francisco, California, United States of America
| |
Collapse
|
21
|
Abstract
Integrins are a large family of extracellular matrix (ECM) receptors. In the developing and adult brain, many integrins are present at high levels at synapses. The tetrapartite structure of synapses - which comprises presynaptic and postsynaptic neurons, the ECM and glial processes - places synaptic integrins in an excellent position to sense dynamic changes in the synaptic environment and use this information to coordinate further changes in synapse structure and function that will shape neural circuit properties. Recent developments in our understanding of the cellular and physiological roles of integrins, which range from control of neural process outgrowth and synapse formation to regulation of synaptic plasticity and memory, enable us to attempt a synthesis of synaptic integrin function.
Collapse
|
22
|
Baig DN, Yanagawa T, Tabuchi K. Distortion of the normal function of synaptic cell adhesion molecules by genetic variants as a risk for autism spectrum disorders. Brain Res Bull 2016; 129:82-90. [PMID: 27743928 DOI: 10.1016/j.brainresbull.2016.10.006] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2016] [Revised: 10/08/2016] [Accepted: 10/10/2016] [Indexed: 12/15/2022]
Abstract
Synaptic cell adhesion molecules (SCAMs) are a functional category of cell adhesion molecules that connect pre- and postsynapses by the protein-protein interaction via their extracellular cell adhesion domains. Countless numbers of common genetic variants and rare mutations in SCAMs have been identified in the patients with autism spectrum disorders (ASDs). Among these, NRXN and NLGN family proteins cooperatively function at synaptic terminals both of which genes are strongly implicated as risk genes for ASDs. Knock-in mice carrying a single rare point mutation of NLGN3 (NLGN3 R451C) discovered in the patients with ASDs display a deficit in social interaction and an enhancement of spatial learning and memory ability reminiscent of the clinical phenotype of ASDs. NLGN4 knockout (KO) and NRXN2α KO mice also show a deficit in sociability as well as some specific neuropsychiatric behaviors. In this review, we selected NRXNs/NLGNs, CNTNAP2/CNTNAP4, CNTN4, ITGB3, and KIRREL3 as strong ASD risk genes based on SFARI score and summarize the protein structures, functions at synapses, representative discoveries in human genetic studies, and phenotypes of the mutant model mice in light of the pathophysiology of ASDs.
Collapse
Affiliation(s)
- Deeba Noreen Baig
- Department of Biological Sciences, Forman Christian College, Zahoor Elahi Rd, Lahore, 54600, Pakistan
| | - Toru Yanagawa
- Department of Oral and Maxillofacial Surgery, Faculty of Medicine, University of Tsukuba, Ibaraki, 305-8575, Japan
| | - Katsuhiko Tabuchi
- Department of Molecular and Cellular Physiology, Shinshu University School of Medicine, 3-1-1 Asahi, Matsumoto, 390-8621, Japan; Institute for Biomedical Sciences, Interdisciplinary Cluster for Cutting Edge Research, Shinshu University, Matsumoto, 390-8621, Japan; PRESTO, JST, Saitama, 332-0012, Japan.
| |
Collapse
|
23
|
Drago A, Crisafulli C, Sidoti A, Calabrò M, Serretti A. The microtubule-associated molecular pathways may be genetically disrupted in patients with Bipolar Disorder. Insights from the molecular cascades. J Affect Disord 2016; 190:429-438. [PMID: 26551401 DOI: 10.1016/j.jad.2015.10.016] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/24/2015] [Revised: 09/24/2015] [Accepted: 10/10/2015] [Indexed: 01/15/2023]
Abstract
Bipolar Disorder is a severe disease characterized by pathological mood swings from major depressive episodes to manic ones and vice versa. The biological underpinnings of Bipolar Disorder have yet to be defined. As a consequence, pharmacological treatments are suboptimal. In the present paper we test the hypothesis that the molecular pathways involved with the direct targets of lithium, hold significantly more genetic variations associated with BD. A molecular pathway approach finds its rationale in the polygenic nature of the disease. The pathways were tested in a sample of ∼ 7,000 patients and controls. Data are available from the public NIMH database. The definition of the pathways was conducted according to the National Cancer Institute (http://pid.nci.nih.gov/). As a result, 3 out of the 18 tested pathways related to lithium action resisted the permutation analysis and were found to be associated with BD. These pathways were related to Reelin, Integrins and Aurora. A pool of genes selected from the ones linked with the above pathways was further investigated in order to identify the fine molecular mechanics shared by our significant pathways and also their link with lithium mechanism of action. The data obtained point out to a possible involvement of microtubule-related mechanics.
Collapse
Affiliation(s)
- Antonio Drago
- Department of Biomedical and Neuromotor Sciences - DIBINEM - University of Bologna, Bologna, Italy
| | - Concetta Crisafulli
- Department of Biomedical Science and Morphological and Functional Images, University of Messina, Via Consolare Valeria, 98125 Messina, Italy.
| | - Antonina Sidoti
- Department of Biomedical Science and Morphological and Functional Images, University of Messina, Via Consolare Valeria, 98125 Messina, Italy
| | - Marco Calabrò
- Department of Biomedical Science and Morphological and Functional Images, University of Messina, Via Consolare Valeria, 98125 Messina, Italy
| | - Alessandro Serretti
- Department of Biomedical and Neuromotor Sciences - DIBINEM - University of Bologna, Bologna, Italy
| |
Collapse
|
24
|
Mosconi MW, Wang Z, Schmitt LM, Tsai P, Sweeney JA. The role of cerebellar circuitry alterations in the pathophysiology of autism spectrum disorders. Front Neurosci 2015; 9:296. [PMID: 26388713 PMCID: PMC4555040 DOI: 10.3389/fnins.2015.00296] [Citation(s) in RCA: 66] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2015] [Accepted: 08/06/2015] [Indexed: 01/23/2023] Open
Abstract
The cerebellum has been repeatedly implicated in gene expression, rodent model and post-mortem studies of autism spectrum disorder (ASD). How cellular and molecular anomalies of the cerebellum relate to clinical manifestations of ASD remains unclear. Separate circuits of the cerebellum control different sensorimotor behaviors, such as maintaining balance, walking, making eye movements, reaching, and grasping. Each of these behaviors has been found to be impaired in ASD, suggesting that multiple distinct circuits of the cerebellum may be involved in the pathogenesis of patients' sensorimotor impairments. We will review evidence that the development of these circuits is disrupted in individuals with ASD and that their study may help elucidate the pathophysiology of sensorimotor deficits and core symptoms of the disorder. Preclinical studies of monogenetic conditions associated with ASD also have identified selective defects of the cerebellum and documented behavioral rescues when the cerebellum is targeted. Based on these findings, we propose that cerebellar circuits may prove to be promising targets for therapeutic development aimed at rescuing sensorimotor and other clinical symptoms of different forms of ASD.
Collapse
Affiliation(s)
- Matthew W Mosconi
- Clinical Child Psychology Program and Schiefelbusch Institute for Life Span Studies, University of Kansas Lawrence, KS, USA ; Center for Autism and Developmental Disabilities, University of Texas Southwestern Dallas, TX, USA ; Department of Psychiatry, University of Texas Southwestern Dallas, TX, USA ; Department of Pediatrics, University of Texas Southwestern Dallas, TX, USA
| | - Zheng Wang
- Center for Autism and Developmental Disabilities, University of Texas Southwestern Dallas, TX, USA ; Department of Psychiatry, University of Texas Southwestern Dallas, TX, USA
| | - Lauren M Schmitt
- Center for Autism and Developmental Disabilities, University of Texas Southwestern Dallas, TX, USA ; Department of Psychiatry, University of Texas Southwestern Dallas, TX, USA
| | - Peter Tsai
- Center for Autism and Developmental Disabilities, University of Texas Southwestern Dallas, TX, USA ; Department of Psychiatry, University of Texas Southwestern Dallas, TX, USA ; Department of Pediatrics, University of Texas Southwestern Dallas, TX, USA ; Department of Neurology and Neurotherapeutics, University of Texas Southwestern Dallas, TX, USA ; Department of Neuroscience, University of Texas Southwestern Dallas, TX, USA
| | - John A Sweeney
- Center for Autism and Developmental Disabilities, University of Texas Southwestern Dallas, TX, USA ; Department of Psychiatry, University of Texas Southwestern Dallas, TX, USA ; Department of Pediatrics, University of Texas Southwestern Dallas, TX, USA
| |
Collapse
|
25
|
Mazalouskas M, Jessen T, Varney S, Sutcliffe JS, Veenstra-VanderWeele J, Cook EH, Carneiro AMD. Integrin β3 Haploinsufficiency Modulates Serotonin Transport and Antidepressant-Sensitive Behavior in Mice. Neuropsychopharmacology 2015; 40:2015-24. [PMID: 25684064 PMCID: PMC4839525 DOI: 10.1038/npp.2015.51] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/20/2014] [Revised: 02/06/2015] [Accepted: 02/08/2015] [Indexed: 12/13/2022]
Abstract
Converging lines of evidence have identified genetic interactions between the serotonin transporter (SERT) gene and ITGB3, which encodes the β3 subunit that forms the αIIbβ3 and αvβ3 integrin receptor complexes. Here we examine the consequences of haploinsufficiency in the mouse integrin β3 subunit gene (Itgb3) on SERT function and selective 5-hydroxytryptamine (5-HT) reuptake inhibitor (SSRI) effectiveness in vivo. Biochemical fractionation studies and immunofluorescent staining of murine brain slices reveal that αvβ3 receptors and SERTs are enriched in presynaptic membranes from several brain regions and that αvβ3 colocalizes with a subpopulation of SERT-containing synapses in raphe nuclei. Notably, we establish that loss of a single allele of Itgb3 in murine neurons is sufficient to decrease 5-HT uptake by SERT in midbrain synaptosomes. Pharmacological assays to elucidate the αvβ3-mediated mechanism of reduced SERT function indicate that decreased integrin β3 subunit expression scales down the population size of active SERT molecules and, as a consequence, lowers the effective dose of SSRIs. These data are consistent with the existence of a subpopulation of SERTs that are tightly modulated by integrin αvβ3 and significantly contribute to global SERT function at 5-HT synapses in the midbrain. Importantly, our screen of a normal human population for single nucleotide polymorphisms in human ITGB3 identified a variant associated with reductions in integrin β3 expression levels that parallel our mouse findings. Thus, polymorphisms in human ITGB3 may contribute to the differential responsiveness of select patients to SSRIs.
Collapse
Affiliation(s)
- Matthew Mazalouskas
- Department of Pharmacology, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Tammy Jessen
- Department of Pharmacology, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Seth Varney
- Department of Pharmacology, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - James S Sutcliffe
- Department of Psychiatry, Vanderbilt University School of Medicine, Nashville, TN, USA,Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, Nashville, TN, USA
| | | | - Edwin H Cook
- Department of Psychiatry, University of Illinois at Chicago, Chicago, IL, USA
| | - Ana M D Carneiro
- Department of Pharmacology, Vanderbilt University School of Medicine, Nashville, TN, USA,Department of Pharmacology, Vanderbilt University School of Medicine, 461 Preston Research Building, 23rd Avenue South at Pierce, Nashville, TN 37232, USA, Tel: +1 615 875 5635, Fax: 615-343-1084, E-mail:
| |
Collapse
|
26
|
Namjou B, Marsolo K, Caroll RJ, Denny JC, Ritchie MD, Verma SS, Lingren T, Porollo A, Cobb BL, Perry C, Kottyan LC, Rothenberg ME, Thompson SD, Holm IA, Kohane IS, Harley JB. Phenome-wide association study (PheWAS) in EMR-linked pediatric cohorts, genetically links PLCL1 to speech language development and IL5-IL13 to Eosinophilic Esophagitis. Front Genet 2014; 5:401. [PMID: 25477900 PMCID: PMC4235428 DOI: 10.3389/fgene.2014.00401] [Citation(s) in RCA: 64] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2014] [Accepted: 10/31/2014] [Indexed: 02/06/2023] Open
Abstract
Objective: We report the first pediatric specific Phenome-Wide Association Study (PheWAS) using electronic medical records (EMRs). Given the early success of PheWAS in adult populations, we investigated the feasibility of this approach in pediatric cohorts in which associations between a previously known genetic variant and a wide range of clinical or physiological traits were evaluated. Although computationally intensive, this approach has potential to reveal disease mechanistic relationships between a variant and a network of phenotypes. Method: Data on 5049 samples of European ancestry were obtained from the EMRs of two large academic centers in five different genotyped cohorts. Recently, these samples have undergone whole genome imputation. After standard quality controls, removing missing data and outliers based on principal components analyses (PCA), 4268 samples were used for the PheWAS study. We scanned for associations between 2476 single-nucleotide polymorphisms (SNP) with available genotyping data from previously published GWAS studies and 539 EMR-derived phenotypes. The false discovery rate was calculated and, for any new PheWAS findings, a permutation approach (with up to 1,000,000 trials) was implemented. Results: This PheWAS found a variety of common variants (MAF > 10%) with prior GWAS associations in our pediatric cohorts including Juvenile Rheumatoid Arthritis (JRA), Asthma, Autism and Pervasive Developmental Disorder (PDD) and Type 1 Diabetes with a false discovery rate < 0.05 and power of study above 80%. In addition, several new PheWAS findings were identified including a cluster of association near the NDFIP1 gene for mental retardation (best SNP rs10057309, p = 4.33 × 10−7, OR = 1.70, 95%CI = 1.38 − 2.09); association near PLCL1 gene for developmental delays and speech disorder [best SNP rs1595825, p = 1.13 × 10−8, OR = 0.65(0.57 − 0.76)]; a cluster of associations in the IL5-IL13 region with Eosinophilic Esophagitis (EoE) [best at rs12653750, p = 3.03 × 10−9, OR = 1.73 95%CI = (1.44 − 2.07)], previously implicated in asthma, allergy, and eosinophilia; and association of variants in GCKR and JAZF1 with allergic rhinitis in our pediatric cohorts [best SNP rs780093, p = 2.18 × 10−5, OR = 1.39, 95%CI = (1.19 − 1.61)], previously demonstrated in metabolic disease and diabetes in adults. Conclusion: The PheWAS approach with re-mapping ICD-9 structured codes for our European-origin pediatric cohorts, as with the previous adult studies, finds many previously reported associations as well as presents the discovery of associations with potentially important clinical implications.
Collapse
Affiliation(s)
- Bahram Namjou
- Center for Autoimmune Genomics and Etiology, Cincinnati Children's Hospital Medical Center Cincinnati, OH, USA ; College of Medicine, University of Cincinnati Cincinnati, OH, USA
| | - Keith Marsolo
- College of Medicine, University of Cincinnati Cincinnati, OH, USA ; Division of Biomedical Informatics, Cincinnati Children's Hospital Medical Center Cincinnati, OH, USA
| | - Robert J Caroll
- Department of Biomedical Informatics, Vanderbilt University School of Medicine Nashville, TN, USA
| | - Joshua C Denny
- Department of Biomedical Informatics, Vanderbilt University School of Medicine Nashville, TN, USA ; Department of Medicine, Vanderbilt University School of Medicine Nashville, TN, USA
| | - Marylyn D Ritchie
- Center for Systems Genomics, The Pennsylvania State University Philadelphia, PA, USA
| | - Shefali S Verma
- Center for Systems Genomics, The Pennsylvania State University Philadelphia, PA, USA
| | - Todd Lingren
- College of Medicine, University of Cincinnati Cincinnati, OH, USA ; Division of Biomedical Informatics, Cincinnati Children's Hospital Medical Center Cincinnati, OH, USA
| | - Aleksey Porollo
- Center for Autoimmune Genomics and Etiology, Cincinnati Children's Hospital Medical Center Cincinnati, OH, USA ; College of Medicine, University of Cincinnati Cincinnati, OH, USA ; Division of Biomedical Informatics, Cincinnati Children's Hospital Medical Center Cincinnati, OH, USA
| | - Beth L Cobb
- Center for Autoimmune Genomics and Etiology, Cincinnati Children's Hospital Medical Center Cincinnati, OH, USA
| | - Cassandra Perry
- Division of Genetics and Genomics, Boston Children's Hospital Boston, MA, USA
| | - Leah C Kottyan
- Center for Autoimmune Genomics and Etiology, Cincinnati Children's Hospital Medical Center Cincinnati, OH, USA ; College of Medicine, University of Cincinnati Cincinnati, OH, USA ; Division of Allergy and Immunology, Department of Pediatrics, Cincinnati Children's Hospital Medical Center Cincinnati, OH, USA
| | - Marc E Rothenberg
- Division of Allergy and Immunology, Department of Pediatrics, Cincinnati Children's Hospital Medical Center Cincinnati, OH, USA
| | - Susan D Thompson
- Center for Autoimmune Genomics and Etiology, Cincinnati Children's Hospital Medical Center Cincinnati, OH, USA ; College of Medicine, University of Cincinnati Cincinnati, OH, USA
| | - Ingrid A Holm
- Division of Genetics and Genomics, Department of Pediatrics, The Manton Center for Orphan Disease Research, Harvard Medical School, Boston Children's Hospital Boston, MA, USA
| | - Isaac S Kohane
- Children's Hospital Informatics Program, Center for Biomedical Informatics, Harvard Medical School Boston, MA, USA
| | - John B Harley
- Center for Autoimmune Genomics and Etiology, Cincinnati Children's Hospital Medical Center Cincinnati, OH, USA ; College of Medicine, University of Cincinnati Cincinnati, OH, USA ; U.S. Department of Veterans Affairs Medical Center Cincinnati, OH, USA
| |
Collapse
|
27
|
Schuch JB, Muller D, Endres RG, Bosa CA, Longo D, Schuler-Faccini L, Ranzan J, Becker MM, dos Santos Riesgo R, Roman T. The role of β3 integrin gene variants in Autism Spectrum Disorders--diagnosis and symptomatology. Gene 2014; 553:24-30. [PMID: 25280596 DOI: 10.1016/j.gene.2014.09.058] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2014] [Revised: 09/12/2014] [Accepted: 09/29/2014] [Indexed: 11/15/2022]
Abstract
Autism Spectrum Disorders (ASDs) represent a group of very complex early-onset neurodevelopmental diseases. In this study, we analyzed 5 SNPs (rs2317385, rs5918, rs15908, rs12603582, rs3809865) at the β3 integrin locus (ITGB3), which has been suggested as a possible susceptibility gene, both as single markers and as part of haplotypes in 209 ASD children and their biological parents. We tested for association with the following: a) DSM-IV ASD diagnosis; b) clinical symptoms common in ASD patients (repetitive behaviors, echolalia, seizures and epilepsy, mood instability, aggression, psychomotor agitation, sleep disorders); and c) dimensional scores obtained with the Autism Screening Questionnaire and the Childhood Autism Rating Scale. These hypotheses were investigated using family-based tests, logistic regression models and analysis of covariance. The family-based tests showed an association with the H5 haplotype (composed by GTCGA alleles, the order of SNPs as above), which was transmitted less often than expected by chance (P=0.006; Pcorr=0.036). The analyses of the clinical symptoms showed a trend for an association with rs12603582 (P=0.008; Pcorr=0.064) and positive results for the haplotype composed of rs15908 and rs12603582 (Pglcorr=0.048; Pindcorr=0.015), both in symptoms of echolalia. Other nominal associations with different variants were found and involved epilepsy/seizures, aggression symptoms and higher ASQ scores. Although our positive results are not definitive, they suggest small effect associations of the ITGB3 gene with both ASD diagnosis and symptoms of echolalia. Other studies are nonetheless needed to fully understand the involvement of this locus on the etiology of ASDs and its different clinical aspects.
Collapse
Affiliation(s)
- Jaqueline Bohrer Schuch
- Department of Genetics, Biosciences Institute, Federal University of Rio Grande do Sul, Avenida Bento Gonçalves, 9500, 91501-970 Porto Alegre, Brazil.
| | - Diana Muller
- Department of Genetics, Biosciences Institute, Federal University of Rio Grande do Sul, Avenida Bento Gonçalves, 9500, 91501-970 Porto Alegre, Brazil
| | - Renata Giuliani Endres
- Department of Psychology, Federal University of Rio Grande do Sul, Rua Ramiro Barcelos 2600, 90035-003 Porto Alegre, Brazil
| | - Cleonice Alves Bosa
- Department of Psychology, Federal University of Rio Grande do Sul, Rua Ramiro Barcelos 2600, 90035-003 Porto Alegre, Brazil.
| | - Dânae Longo
- Department of Genetics, Biosciences Institute, Federal University of Rio Grande do Sul, Avenida Bento Gonçalves, 9500, 91501-970 Porto Alegre, Brazil
| | - Lavinia Schuler-Faccini
- Department of Genetics, Biosciences Institute, Federal University of Rio Grande do Sul, Avenida Bento Gonçalves, 9500, 91501-970 Porto Alegre, Brazil
| | - Josiane Ranzan
- Child Neurology Unit, Clinics Hospital of Porto Alegre, Federal University of Rio Grande do Sul, Rua Ramiro Barcelos, 2350, 90035-903 Porto Alegre, Brazil
| | - Michele Michelin Becker
- Child Neurology Unit, Clinics Hospital of Porto Alegre, Federal University of Rio Grande do Sul, Rua Ramiro Barcelos, 2350, 90035-903 Porto Alegre, Brazil
| | - Rudimar dos Santos Riesgo
- Child Neurology Unit, Clinics Hospital of Porto Alegre, Federal University of Rio Grande do Sul, Rua Ramiro Barcelos, 2350, 90035-903 Porto Alegre, Brazil.
| | - Tatiana Roman
- Department of Genetics, Biosciences Institute, Federal University of Rio Grande do Sul, Avenida Bento Gonçalves, 9500, 91501-970 Porto Alegre, Brazil.
| |
Collapse
|
28
|
Gabriele S, Sacco R, Persico AM. Blood serotonin levels in autism spectrum disorder: a systematic review and meta-analysis. Eur Neuropsychopharmacol 2014; 24:919-29. [PMID: 24613076 DOI: 10.1016/j.euroneuro.2014.02.004] [Citation(s) in RCA: 226] [Impact Index Per Article: 20.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/29/2013] [Revised: 01/09/2014] [Accepted: 02/12/2014] [Indexed: 12/14/2022]
Abstract
Elevated blood serotonin (5-HT) levels were the first biomarker identified in autism research. Many studies have contrasted blood 5-HT levels in autistic patients and controls, but different measurement protocols, technologies, and biomaterials have been used through the years. We performed a systematic review and meta-analysis to provide an overall estimate of effect size and between-study heterogeneity, while verifying whether and to what extent different methodological approaches influence the strength of this association. Our literature search strategy identified 551 papers, from which 22 studies providing patient and control blood 5-HT values were selected for meta-analysis. Significantly higher 5-HT levels in autistic patients compared to controls were recorded both in whole blood (WB) [O.R.=4.6; (3.1-5.2); P=1.0×10(-12]), and in platelet-rich plasma (PRP) [O.R.=2.6 (1.8-3.9); P=2.7×10(-7)]. Predictably, studies measuring 5-HT levels in platelet-poor plasma (PPP) yielded no significant group difference [O.R.=0.54 (0.2-2-0); P=0.36]. Altogether, elevated 5-HT blood levels were recorded in 28.3% in WB and 22.5% in PRP samples of autistic individuals, as reported in 15 and 4 studies, respectively. Studies employing HPLC vs fluorometric assays yield similar cumulative effect sizes, but the former display much lower variability. In summary, despite some limitations mainly due to small study sample sizes, our results significantly reinforce the reliability of elevated 5-HT blood levels as a biomarker in ASD, providing practical indications potentially useful for its inclusion in multi-marker diagnostic panels for clinical use.
Collapse
Affiliation(s)
- Stefano Gabriele
- Unit of Child and Adolescent NeuroPsychiatry, Laboratory of Molecular Psychiatry and Neurogenetics, University "Campus Bio-Medico", Via Alvaro del Portillo 21, I-00128 Rome, Italy; Department of Experimental Neurosciences, I.R.C.C.S. "Fondazione Santa Lucia", Rome, Italy
| | - Roberto Sacco
- Unit of Child and Adolescent NeuroPsychiatry, Laboratory of Molecular Psychiatry and Neurogenetics, University "Campus Bio-Medico", Via Alvaro del Portillo 21, I-00128 Rome, Italy; Department of Experimental Neurosciences, I.R.C.C.S. "Fondazione Santa Lucia", Rome, Italy
| | - Antonio M Persico
- Unit of Child and Adolescent NeuroPsychiatry, Laboratory of Molecular Psychiatry and Neurogenetics, University "Campus Bio-Medico", Via Alvaro del Portillo 21, I-00128 Rome, Italy; Department of Experimental Neurosciences, I.R.C.C.S. "Fondazione Santa Lucia", Rome, Italy; Mafalda Luce Center for Pervasive Developmental Disorders, Milan, Italy.
| |
Collapse
|
29
|
Petrinovic MM, Künnecke B. Neuroimaging endophenotypes in animal models of autism spectrum disorders: lost or found in translation? Psychopharmacology (Berl) 2014; 231:1167-89. [PMID: 23852013 DOI: 10.1007/s00213-013-3200-z] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/28/2013] [Accepted: 06/26/2013] [Indexed: 11/26/2022]
Abstract
RATIONALE Autism spectrum disorder(s) (ASDs) is a neurodevelopmental disorder characterized by stereotyped behaviours and impairments in communication and social interactions. This heterogeneity has been a major obstacle in uncovering the aetiology and biomarkers of ASDs. Rodent models with genetic modifications or environmental insults have been created to study particular endophenotypes and bridge the gap between genetics and behavioural phenotypes. Translational neuroimaging modalities with their ability to screen the brain noninvasively and yield structural, biochemical and functional information provide a unique platform for discovery and evaluation of such endophenotypes in preclinical and clinical research. OBJECTIVES We reviewed literature on translational neuroimaging in rodent models of ASDs. The most prominent models will be described and the respective neuroimaging endophenotypes will be discussed with reference to human data. A perspective on future directions of translational neuroimaging in animal models of ASDs will be given. RESULTS AND CONCLUSIONS To date, we experience a proliferation of rodent models which recapitulate specific liabilities identified in ASDs patients. Translational neuroimaging in these models is emerging but is skewed towards magnetic resonance imaging (MRI) modalities. Volumetric and structural assessments of the brain are dominating and a host of endophenotypes have been reported that allude to findings in ASDs patients but with only few to converge among the models. Caveats of current studies are the diverging biological conditions related to genetic background and age of the animals. It is anticipated that longitudinal and functional assessments will gain much importance and will help elucidating mechanistic relationship between behavioural and structural endophenotypes.
Collapse
Affiliation(s)
- Marija M Petrinovic
- F. Hoffmann-La Roche AG, pRED, Pharma Research and Early Development, DTA Neuroscience, Building 68, Room 327A, Grenzacherstrasse 124, 4070, Basel, Switzerland
| | | |
Collapse
|
30
|
Carayol J, Schellenberg GD, Dombroski B, Amiet C, Génin B, Fontaine K, Rousseau F, Vazart C, Cohen D, Frazier TW, Hardan AY, Dawson G, Rio Frio T. A scoring strategy combining statistics and functional genomics supports a possible role for common polygenic variation in autism. Front Genet 2014; 5:33. [PMID: 24600472 PMCID: PMC3927086 DOI: 10.3389/fgene.2014.00033] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2013] [Accepted: 01/29/2014] [Indexed: 12/23/2022] Open
Abstract
Autism spectrum disorders (ASD) are highly heritable complex neurodevelopmental disorders with a 4:1 male: female ratio. Common genetic variation could explain 40-60% of the variance in liability to autism. Because of their small effect, genome-wide association studies (GWASs) have only identified a small number of individual single-nucleotide polymorphisms (SNPs). To increase the power of GWASs in complex disorders, methods like convergent functional genomics (CFG) have emerged to extract true association signals from noise and to identify and prioritize genes from SNPs using a scoring strategy combining statistics and functional genomics. We adapted and applied this approach to analyze data from a GWAS performed on families with multiple children affected with autism from Autism Speaks Autism Genetic Resource Exchange (AGRE). We identified a set of 133 candidate markers that were localized in or close to genes with functional relevance in ASD from a discovery population (545 multiplex families); a gender specific genetic score (GS) based on these common variants explained 1% (P = 0.01 in males) and 5% (P = 8.7 × 10(-7) in females) of genetic variance in an independent sample of multiplex families. Overall, our work demonstrates that prioritization of GWAS data based on functional genomics identified common variants associated with autism and provided additional support for a common polygenic background in autism.
Collapse
Affiliation(s)
| | - Gerard D. Schellenberg
- Department of Pathology and Laboratory Medicine, University of PennsylvaniaPhiladelphia, PA, USA
| | - Beth Dombroski
- Department of Pathology and Laboratory Medicine, University of PennsylvaniaPhiladelphia, PA, USA
| | | | | | | | | | | | - David Cohen
- Groupe Hospitalier Pitié-Salpêtrière, Department of Child and Adolescent Psychiatry, AP-HP, Université Pierre et Marie CurieParis, France
| | - Thomas W. Frazier
- Center for Pediatric Behavioral Health and Center for Autism, Cleveland ClinicCleveland, OH, USA
| | - Antonio Y. Hardan
- Department of Psychiatry and Behavioral Sciences, Stanford UniversityStanford, CA, USA
| | - Geraldine Dawson
- Department of Psychiatry and Behavioral Sciences, Duke University Medical CenterDurham, NC, USA
| | | |
Collapse
|
31
|
Kerrisk ME, Cingolani LA, Koleske AJ. ECM receptors in neuronal structure, synaptic plasticity, and behavior. PROGRESS IN BRAIN RESEARCH 2014; 214:101-31. [PMID: 25410355 DOI: 10.1016/b978-0-444-63486-3.00005-0] [Citation(s) in RCA: 69] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
During central nervous system development, extracellular matrix (ECM) receptors and their ligands play key roles as guidance molecules, informing neurons where and when to send axonal and dendritic projections, establish connections, and form synapses between pre- and postsynaptic cells. Once stable synapses are formed, many ECM receptors transition in function to control the maintenance of stable connections between neurons and regulate synaptic plasticity. These receptors bind to and are activated by ECM ligands. In turn, ECM receptor activation modulates downstream signaling cascades that control cytoskeletal dynamics and synaptic activity to regulate neuronal structure and function and thereby impact animal behavior. The activities of cell adhesion receptors that mediate interactions between pre- and postsynaptic partners are also strongly influenced by ECM composition. This chapter highlights a number of ECM receptors, their roles in the control of synapse structure and function, and the impact of these receptors on synaptic plasticity and animal behavior.
Collapse
Affiliation(s)
- Meghan E Kerrisk
- Department of Molecular Biophysics and Biochemistry, Yale University, New Haven, CT, USA
| | - Lorenzo A Cingolani
- Department of Neuroscience and Brain Technologies, Istituto Italiano di Tecnologia, Genoa, Italy
| | - Anthony J Koleske
- Department of Molecular Biophysics and Biochemistry, Yale University, New Haven, CT, USA; Department of Neurobiology, Yale University, New Haven, CT, USA; Interdepartmental Neuroscience Program, Yale University, New Haven, CT, USA; Program in Cellular Neuroscience, Neurodegeneration, and Repair, Yale University, New Haven, CT, USA.
| |
Collapse
|
32
|
Yilmaz U, Zeybek U, Kahraman OT, Kafadar AM, Toptas B, Yamak N, Celik F, Yaylim I. Investigation of ICAM-1 and β3 integrin gene variations in patients with brain tumors. Asian Pac J Cancer Prev 2013; 14:5929-34. [PMID: 24289603 DOI: 10.7314/apjcp.2013.14.10.5929] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Primary brain tumors constitute a small percent of all malignant cancers, but their etiology remains poorly understood. β3 integrin (ITGB3) has been recognized to play influential roles in angiogenesis, tumor growth and metastasis. Intercellular adhesion molecule-1 (ICAM-1) is a surface glycoprotein important for tumor invasion and angiogenesis. The aim of this study was to investigate whether specific genetic polymorphisms of ICAM-1 and ITGB3 could be associated with brain cancer development and progression in a Turkish population. Our study is the first to our knowledge to investigate the relationship between brain tumor risk and ICAM-1 and β3 integrin gene polymorphisms. MATERIALS AND METHODS The study covered 92 patients with primary brain tumors and 92 age-matched healthy control subjects. Evaluation of β3 integrin (Leu33Pro (rs5918)) and ICAM-1 (R241G (rs1799969) and K469E (rs5498)) gene polymorphisms was performed by polymerase chain reaction-restriction fragment length polymorphism (PCR-RFLP). RESULTS According to results of our research, the A allele of the ICAM-1 R241G gene polymorphism appeared to be a risk factor for primary brain tumors (p<0.001). Similarly, the frequency of the A mutant allele of ICAM-1 R241G was statistically significant in patients with brain tumors classified as glioma (p<0.001). When allele and genotype distributions of ICAM- 1 K469E, ICAM-1 R241G and β3 integrin Leu33Pro gene polymorphisms were evaluated with age, sex, and smoking, there were no statistically significant differences. Haplotype analysis revealed that the frequencies of GAC (rs1799969-rs5498-rs5918) and GAT (rs1799969-rs5498-rs5918) haplotypes were significantly lower in patients as compared with controls (p=0.001; p=0.036 respectively). CONCLUSIONS This study provides the first evidence that ICAM-1 R241G SNP significantly contributes to the risk of primary brain tumors in a Turkish population. In addition, our results suggest that ICAM-1 R241G in combination ICAM-1 K469E may have protective effects against the development of brain cancer.
Collapse
Affiliation(s)
- Umit Yilmaz
- Department of Molecular Medicine, The Institute of Experimental Medicine, Istanbul University, Istanbul, Turkey E-mail :
| | | | | | | | | | | | | | | |
Collapse
|
33
|
Steadman PE, Ellegood J, Szulc KU, Turnbull DH, Joyner AL, Henkelman RM, Lerch JP. Genetic effects on cerebellar structure across mouse models of autism using a magnetic resonance imaging atlas. Autism Res 2013; 7:124-37. [PMID: 24151012 DOI: 10.1002/aur.1344] [Citation(s) in RCA: 123] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2013] [Accepted: 09/17/2013] [Indexed: 12/19/2022]
Abstract
Magnetic resonance imaging (MRI) of autism populations is confounded by the inherent heterogeneity in the individuals' genetics and environment, two factors difficult to control for. Imaging genetic animal models that recapitulate a mutation associated with autism quantify the impact of genetics on brain morphology and mitigate the confounding factors in human studies. Here, we used MRI to image three genetic mouse models with single mutations implicated in autism: Neuroligin-3 R451C knock-in, Methyl-CpG binding protein-2 (MECP2) 308-truncation and integrin β3 homozygous knockout. This study identified the morphological differences specific to the cerebellum, a structure repeatedly linked to autism in human neuroimaging and postmortem studies. To accomplish a comparative analysis, a segmented cerebellum template was created and used to segment each study image. This template delineated 39 different cerebellar structures. For Neuroligin-3 R451C male mutants, the gray (effect size (ES) = 1.94, FDR q = 0.03) and white (ES = 1.84, q = 0.037) matter of crus II lobule and the gray matter of the paraflocculus (ES = 1.45, q = 0.045) were larger in volume. The MECP2 mutant mice had cerebellar volume changes that increased in scope depending on the genotype: hemizygous males to homozygous females. The integrin β3 mutant mouse had a drastically smaller cerebellum than controls with 28 out of 39 cerebellar structures smaller. These imaging results are discussed in relation to repetitive behaviors, sociability, and learning in the context of autism. This work further illuminates the cerebellum's role in autism.
Collapse
Affiliation(s)
- Patrick E Steadman
- Mouse Imaging Centre, Hospital for Sick Children, Toronto, Ontario, Canada; Department of Medical Biophysics, University of Toronto, Toronto, Ontario, Canada
| | | | | | | | | | | | | |
Collapse
|
34
|
Campbell NG, Zhu CB, Lindler KM, Yaspan BL, Kistner-Griffin E. Rare coding variants of the adenosine A3 receptor are increased in autism: on the trail of the serotonin transporter regulome. Mol Autism 2013; 4:28. [PMID: 23953133 PMCID: PMC3882891 DOI: 10.1186/2040-2392-4-28] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2012] [Accepted: 07/30/2013] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND Rare genetic variation is an important class of autism spectrum disorder (ASD) risk factors and can implicate biological networks for investigation. Altered serotonin (5-HT) signaling has been implicated in ASD, and we and others have discovered multiple, rare, ASD-associated variants in the 5-HT transporter (SERT) gene leading to elevated 5-HT re-uptake and perturbed regulation. We hypothesized that loci encoding SERT regulators harbor variants that impact SERT function and/or regulation and therefore could contribute to ASD risk. The adenosine A3 receptor (A3AR) regulates SERT via protein kinase G (PKG) and other signaling pathways leading to enhanced SERT surface expression and catalytic activity. METHODS To test our hypothesis, we asked whether rare variants in the A3AR gene (ADORA3) were increased in ASD cases vs. controls. Discovery sequencing in a case-control sample and subsequent analysis of comparison exome sequence data were conducted. We evaluated the functional impact of two variants from the discovery sample on A3AR signaling and SERT activity. RESULTS Sequencing discovery showed an increase of rare coding variants in cases vs. controls (P=0.013). While comparison exome sequence data did not show a significant enrichment (P=0.071), combined analysis strengthened evidence for association (P=0.0025). Two variants discovered in ASD cases (Leu90Val and Val171Ile) lie in or near the ligand-binding pocket, and Leu90Val was enriched individually in cases (P=0.040). In vitro analysis of cells expressing Val90-A3AR revealed elevated basal cGMP levels compared with the wildtype receptor. Additionally, a specific A3AR agonist increased cGMP levels across the full time course studied in Val90-A3AR cells, compared to wildtype receptor. In Val90-A3AR/SERT co-transfections, agonist stimulation elevated SERT activity over the wildtype receptor with delayed 5-HT uptake activity recovery. In contrast, Ile171-A3AR was unable to support agonist stimulation of SERT. Although both Val90 and Ile171 were present in greater numbers in these ASD cases, segregation analysis in families showed incomplete penetrance, consistent with other rare ASD risk alleles. CONCLUSIONS Our results validate the hypothesis that the SERT regulatory network harbors rare, functional variants that impact SERT activity and regulation in ASD, and encourages further investigation of this network for other variation that may impact ASD risk.
Collapse
Affiliation(s)
- Nicholas G Campbell
- Department of Molecular Physiology & Biophysics and Vanderbilt Brain Institute, Vanderbilt University School of Medicine, Nashville, TN 37232-8548, USA
| | - Chong-Bin Zhu
- Department of Pharmacology, Vanderbilt University School of Medicine, Nashville, TN 37232-8548, USA
| | - Kathryn M Lindler
- Department of Pharmacology, Vanderbilt University School of Medicine, Nashville, TN 37232-8548, USA
| | - Brian L Yaspan
- Department of Molecular Physiology & Biophysics and Vanderbilt Brain Institute, Vanderbilt University School of Medicine, Nashville, TN 37232-8548, USA
| | - Emily Kistner-Griffin
- Biostatistics and Epidemiology, Medical University of South Carolina, Charleston, SC 29425, USA
| |
Collapse
|
35
|
Singh AS, Chandra R, Guhathakurta S, Sinha S, Chatterjee A, Ahmed S, Ghosh S, Rajamma U. Genetic association and gene-gene interaction analyses suggest likely involvement of ITGB3 and TPH2 with autism spectrum disorder (ASD) in the Indian population. Prog Neuropsychopharmacol Biol Psychiatry 2013; 45:131-43. [PMID: 23628433 DOI: 10.1016/j.pnpbp.2013.04.015] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/14/2013] [Revised: 04/12/2013] [Accepted: 04/22/2013] [Indexed: 11/19/2022]
Abstract
BACKGROUND Serotoninergic dysfunction leads to neurodevelopmental abnormalities and behavioral impairments. Platelet hyperserotoninemia is reported as the best identified endophenotype for autism spectrum disorders. Therefore, in the present study we investigate the association of TPH2, the rate limiting enzyme in 5-HT biosynthesis and ITGB3, a serotonin quantitative trait locus with ASD in the Indian population. METHODS Population and family-based genetic association and gene-gene interaction analyses were performed to evaluate the role of ITGB3 and TPH2 markers in ASD etiology. RESULTS Association tests using ITGB3 markers revealed significant paternal overtransmission of T allele of rs5918 to male probands. Interestingly for TPH2, we observed significant overrepresentation of A-A (rs11179000-rs4290270), G-A (rs4570625-rs4290270), G-G-A (rs4570625-rs11179001-rs4290270) and A-G-A (rs11179000-rs11179001-rs4290270) haplotypes in the controls and maternal preferential transmission of A-A (rs11179001-rs7305115), T-A-A (rs4570625-rs11179001-rs7305115) and T-A-A (rs11179000-rs11179001-rs7305115) and nontransmission of G-G-A (rs4570625-rs11179001-rs7305115) haplotypes to the affected offspring. Moreover, interaction of ITGB3 marker, rs15908 with TPH2 markers was found to be significant and influenced by the sex of the probands. Predicted individual risk, which varied from very mild to moderate, supports combined effect of these markers in ASD. CONCLUSION Overall results of the present study indicate likely involvement of ITGB3 and TPH2 in the pathophysiology of ASD in the Indian population.
Collapse
Affiliation(s)
- Asem Surindro Singh
- Manovikas Biomedical Research & Diagnostic Centre, Manovikas Kendra, 482 Madudah, Plot I-24, Sector J, EM bypass, Kolkata, West Bengal, India.
| | | | | | | | | | | | | | | |
Collapse
|
36
|
Genetic Association Analysis of ITGB3 Polymorphisms with Age at Onset of Schizophrenia. J Mol Neurosci 2013; 51:446-53. [DOI: 10.1007/s12031-013-0059-8] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2013] [Accepted: 06/20/2013] [Indexed: 12/24/2022]
|
37
|
Flood ZC, Engel DLJ, Simon CC, Negherbon KR, Murphy LJ, Tamavimok W, Anderson GM, Janušonis S. Brain growth trajectories in mouse strains with central and peripheral serotonin differences: relevance to autism models. Neuroscience 2012; 210:286-95. [PMID: 22450231 DOI: 10.1016/j.neuroscience.2012.03.010] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2012] [Revised: 03/01/2012] [Accepted: 03/03/2012] [Indexed: 10/28/2022]
Abstract
The genetic heterogeneity of autism spectrum disorders (ASDs) suggests that their underlying neurobiology involves dysfunction at the neural network level. Understanding these neural networks will require a major collaborative effort and will depend on validated and widely accepted animal models. Many mouse models have been proposed in autism research, but the assessment of their validity often has been limited to measuring social interactions. However, two other well-replicated findings have been reported in ASDs: transient brain overgrowth in early postnatal life and elevated 5-HT (serotonin) levels in blood platelets (platelet hyperserotonemia). We examined two inbred mouse strains (C57BL/6 and BALB/c) with respect to these phenomena. The BALB/c strain is less social and exhibits some other autistic-like behaviors. In addition, it has a lower 5-HT synthesis rate in the central nervous system due to a single-nucleotide polymorphism in the tryptophan hydroxylase 2 (Tph2) gene. The postnatal growth of brain mass was analyzed with mixed-effects models that included litter effects. The volume of the hippocampal complex and the thickness of the somatosensory cortex were measured in 3D-brain reconstructions from serial sections. The postnatal whole-blood 5-HT levels were assessed with high-performance liquid chromatography. With respect to the BALB/c strain, the C57BL/6 strain showed transient brain overgrowth and persistent blood hyperserotonemia. The hippocampal volume was permanently enlarged in the C57BL/6 strain, with no change in the adult brain mass. These results indicate that, in mice, autistic-like shifts in the brain and periphery may be associated with less autistic-like behaviors. Importantly, they suggest that consistency among behavioral, anatomical, and physiological measures may expedite the validation of new and previously proposed mouse models of autism, and that the construct validity of models should be demonstrated when these measures are inconsistent.
Collapse
Affiliation(s)
- Z C Flood
- Department of Psychological and Brain Sciences, University of California, Santa Barbara, CA 93106, USA
| | | | | | | | | | | | | | | |
Collapse
|
38
|
Thanseem I, Anitha A, Nakamura K, Suda S, Iwata K, Matsuzaki H, Ohtsubo M, Ueki T, Katayama T, Iwata Y, Suzuki K, Minoshima S, Mori N. Elevated transcription factor specificity protein 1 in autistic brains alters the expression of autism candidate genes. Biol Psychiatry 2012; 71:410-8. [PMID: 22030357 DOI: 10.1016/j.biopsych.2011.09.020] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/08/2011] [Revised: 09/08/2011] [Accepted: 09/10/2011] [Indexed: 01/09/2023]
Abstract
BACKGROUND Profound changes in gene expression can result from abnormalities in the concentrations of sequence-specific transcription factors like specificity protein 1 (Sp1). Specificity protein 1 binding sites have been reported in the promoter regions of several genes implicated in autism. We hypothesize that dysfunction of Sp1 could affect the expression of multiple autism candidate genes, contributing to the heterogeneity of autism. METHODS We assessed any alterations in the expression of Sp1 and that of autism candidate genes in the postmortem brain (anterior cingulate gyrus [ACG], motor cortex, and thalamus) of autism patients (n = 8) compared with healthy control subjects (n = 13). Alterations in the expression of candidate genes upon Sp1/DNA binding inhibition with mithramycin and Sp1 silencing by RNAi were studied in SK-N-SH neuronal cells. RESULTS We observed elevated expression of Sp1 in ACG of autism patients (p = .010). We also observed altered expression of several autism candidate genes. GABRB3, RELN, and HTR2A showed reduced expression, whereas CD38, ITGB3, MAOA, MECP2, OXTR, and PTEN showed elevated expression in autism. In SK-N-SH cells, OXTR, PTEN, and RELN showed reduced expression upon Sp1/DNA binding inhibition and Sp1 silencing. The RNA integrity number was not available for any of the samples. CONCLUSIONS Transcription factor Sp1 is dysfunctional in the ACG of autistic brain. Consequently, the expression of potential autism candidate genes regulated by Sp1, especially OXTR and PTEN, could be affected. The diverse downstream pathways mediated by the Sp1-regulated genes, along with the environmental and intracellular signal-related regulation of Sp1, could explain the complex phenotypes associated with autism.
Collapse
Affiliation(s)
- Ismail Thanseem
- Department of Psychiatry and Neurology, Hamamatsu University School of Medicine, Japan
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Ellegood J, Henkelman RM, Lerch JP. Neuroanatomical Assessment of the Integrin β3 Mouse Model Related to Autism and the Serotonin System Using High Resolution MRI. Front Psychiatry 2012; 3:37. [PMID: 22557981 PMCID: PMC3337465 DOI: 10.3389/fpsyt.2012.00037] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/01/2012] [Accepted: 04/09/2012] [Indexed: 12/02/2022] Open
Abstract
The integrinβ3 (ITGβ3) gene has been associated with both autism and the serotonin system. The purpose of this study was to examine the volumetric differences in the brain of an ITGβ3 homozygous knockout mouse model compared with a corresponding wild-type mouse using high resolution magnetic resonance imaging and detailed statistical analyses. The most striking difference found was an 11% reduction in total brain volume. Moreover, 32 different regions were found to have significantly different relative volumes (percentage total brain volume) in the ITGβ3 mouse. A number of interesting differences relevant to autism were discovered including a smaller corpus callosum volume and bilateral decreases in the hippocampus, striatum, and cerebellum. Relative volume increases were also found in the frontal and parieto-temporal lobes as well as in the amygdala. Particularly intriguing were the changes in the lateral wings of the dorsal raphe nuclei since that nucleus is so integral to the development of many different brain regions and the serotonin system in general.
Collapse
Affiliation(s)
- Jacob Ellegood
- Mouse Imaging Centre, Hospital for Sick Children Toronto, ON, Canada
| | | | | |
Collapse
|
40
|
Genetics and Epigenetics of Autism Spectrum Disorders. RESEARCH AND PERSPECTIVES IN NEUROSCIENCES 2012. [DOI: 10.1007/978-3-642-27913-3_10] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
|
41
|
Carayol J, Schellenberg GD, Dombroski B, Genin E, Rousseau F, Dawson G. Autism risk assessment in siblings of affected children using sex-specific genetic scores. Mol Autism 2011; 2:17. [PMID: 22017886 PMCID: PMC3214848 DOI: 10.1186/2040-2392-2-17] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2011] [Accepted: 10/21/2011] [Indexed: 01/01/2023] Open
Abstract
Background The inheritance pattern in most cases of autism is complex. The risk of autism is increased in siblings of children with autism and previous studies have indicated that the level of risk can be further identified by the accumulation of multiple susceptibility single nucleotide polymorphisms (SNPs) allowing for the identification of a higher-risk subgroup among siblings. As a result of the sex difference in the prevalence of autism, we explored the potential for identifying sex-specific autism susceptibility SNPs in siblings of children with autism and the ability to develop a sex-specific risk assessment genetic scoring system. Methods SNPs were chosen from genes known to be associated with autism. These markers were evaluated using an exploratory sample of 480 families from the Autism Genetic Resource Exchange (AGRE) repository. A reproducibility index (RI) was proposed and calculated in all children with autism and in males and females separately. Differing genetic scoring models were then constructed to develop a sex-specific genetic score model designed to identify individuals with a higher risk of autism. The ability of the genetic scores to identify high-risk children was then evaluated and replicated in an independent sample of 351 affected and 90 unaffected siblings from families with at least 1 child with autism. Results We identified three risk SNPs that had a high RI in males, two SNPs with a high RI in females, and three SNPs with a high RI in both sexes. Using these results, genetic scoring models for males and females were developed which demonstrated a significant association with autism (P = 2.2 × 10-6 and 1.9 × 10-5, respectively). Conclusions Our results demonstrate that individual susceptibility associated SNPs for autism may have important differential sex effects. We also show that a sex-specific risk score based on the presence of multiple susceptibility associated SNPs allow for the identification of subgroups of siblings of children with autism who have a significantly higher risk of autism.
Collapse
|
42
|
Azmitia EC, Singh JS, Hou XP, Wegiel J. Dystrophic serotonin axons in postmortem brains from young autism patients. Anat Rec (Hoboken) 2011; 294:1653-62. [PMID: 21901837 PMCID: PMC4112519 DOI: 10.1002/ar.21243] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2010] [Accepted: 07/01/2010] [Indexed: 02/04/2023]
Abstract
Autism causes neuropathological changes in varied anatomical loci. A coherent neural mechanism to explain the spectrum of autistic symptomatology has not been proposed because most anatomical researchers focus on point-to-point functional neural systems (e.g., auditory and social networks) rather than considering global chemical neural systems. Serotonergic neurons have a global innervation pattern. Disorders Research Program, AS073234, Program Project (JW). Their cell bodies are found in the midbrain but they project their axons throughout the neural axis beginning in the fetal brain. This global system is implicated in autism by animal models and by biochemical, imaging, pharmacological, and genetics studies. However, no anatomical studies of the 5-HT innervation of autistic donors have been reported. Our review presents immunocytochemical evidence of an increase in 5-HT axons in postmortem brain tissue from autism donors aged 2.8-29 years relative to controls. This increase is observed in the principle ascending fiber bundles of the medial and lateral forebrain bundles, and in the innervation density of the amygdala and the piriform, superior temporal, and parahippocampal cortices. In autistic donors 8 years of age and up, several types of dystrophic 5-HT axons were seen in the termination fields. One class of these dystrophic axons, the thick heavily stained axons, was not seen in the brains of patients with neurodegenerative diseases. These findings provide morphological evidence for the involvement of serotonin neurons in the early etiology of autism, and suggest new therapies may be effective to blunt serotonin's trophic actions during early brain development in children.
Collapse
Affiliation(s)
- Efrain C Azmitia
- Department of Biology, New York University, New York, 10003, USA.
| | | | | | | |
Collapse
|
43
|
Holmes RS, Rout UK. Comparative studies of vertebrate Beta integrin genes and proteins: ancient genes in vertebrate evolution. Biomolecules 2011; 1:3-31. [PMID: 24970121 PMCID: PMC4030831 DOI: 10.3390/biom1010003] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2011] [Revised: 08/14/2011] [Accepted: 08/15/2011] [Indexed: 12/31/2022] Open
Abstract
Intregins are heterodimeric α- and β-subunit containing membrane receptor proteins which serve various cell adhesion roles in tissue repair, hemostasis, immune response, embryogenesis and metastasis. At least 18 α- (ITA or ITGA) and 8 β-integrin subunits (ITB or ITGB) are encoded on mammalian genomes. Comparative ITB amino acid sequences and protein structures and ITB gene locations were examined using data from several vertebrate genome projects. Vertebrate ITB genes usually contained 13-16 coding exons and encoded protein subunits with ~800 amino acids, whereas vertebrate ITB4 genes contained 36-39 coding exons and encoded larger proteins with ~1800 amino acids. The ITB sequences exhibited several conserved domains including signal peptide, extracellular β-integrin, β-tail domain and integrin β-cytoplasmic domains. Sequence alignments of the integrin β-cytoplasmic domains revealed highly conserved regions possibly for performing essential functions and its maintenance during vertebrate evolution. With the exception of the human ITB8 sequence, the other ITB sequences shared a predicted 19 residue α-helix for this region. Potential sites for regulating human ITB gene expression were identified which included CpG islands, transcription factor binding sites and microRNA binding sites within the 3'-UTR of human ITB genes. Phylogenetic analyses examined the relationships of vertebrate beta-integrin genes which were consistent with four major groups: 1: ITB1, ITB2, ITB7; 2: ITB3, ITB5, ITB6; 3: ITB4; and 4: ITB8 and a common evolutionary origin from an ancestral gene, prior to the appearance of fish during vertebrate evolution. The phylogenetic analyses revealed that ITB4 is the most likely primordial form of the vertebrate β integrin subunit encoding genes, that is the only β subunit expressed as a constituent of the sole integrin receptor 'α6β4' in the hemidesmosomes of unicellular organisms.
Collapse
Affiliation(s)
- Roger S Holmes
- School of Biomolecular and Physical Sciences, Griffith University, Nathan, 4111QLD, Australia.
| | - Ujjwal K Rout
- Department of Surgery, University of Mississippi Medical Center, Jackson, MS 38677, USA.
| |
Collapse
|
44
|
Gene expression studies in autism: moving from the genome to the transcriptome and beyond. Neurobiol Dis 2011; 45:69-75. [PMID: 21839838 DOI: 10.1016/j.nbd.2011.07.017] [Citation(s) in RCA: 52] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2011] [Accepted: 07/20/2011] [Indexed: 12/22/2022] Open
Abstract
Autism is a clinically and genetically heterogeneous neurodevelopmental disorder. Although multiple genes, risk alleles and copy number variants (CNVs) have been implicated in ASD, none of the currently established genetic causes of ASD accounts for more than 2% of the cases, and a genetic diagnosis is not yet possible for most autism patients. Thus, advancing our understanding of autism genetics requires the integration of genetic information with information on genome function, as provided by transcriptomic data. We review recent autism transcriptome studies, in the context of current knowledge of autism genetics, and discuss the utility of gene expression data in evaluating the functional relevance of genetic variants and identifying common molecular pathways dysregulated in autism.
Collapse
|
45
|
Carter MD, Shah CR, Muller CL, Crawley JN, Carneiro AMD, Veenstra-VanderWeele J. Absence of preference for social novelty and increased grooming in integrin β3 knockout mice: initial studies and future directions. Autism Res 2011; 4:57-67. [PMID: 21254450 PMCID: PMC3073711 DOI: 10.1002/aur.180] [Citation(s) in RCA: 88] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2010] [Accepted: 12/09/2010] [Indexed: 12/17/2022]
Abstract
Elevated whole blood serotonin 5-HT, or hyperserotonemia, is a common biomarker in autism spectrum disorder (ASD). The integrin β3 receptor subunit gene (ITGB3) is a quantitative trait locus for whole blood 5-HT levels. Recent work shows that integrin β3 interacts with the serotonin transporter (SERT) in both platelets and in the midbrain. Furthermore, multiple studies have now reported gene-gene interaction between the integrin β3 and SERT genes in association with ASD. Given the lack of previous data on the impact of integrin β3 on brain or behavioral phenotypes, we sought to compare mice with decreased or absent expression of the integrin β3 receptor subunit (Itgb3 +/- and -/-) with wildtype littermate controls in behavioral tasks relevant to ASD. These mice did not show deficits in activity level in the open field or anxiety-like behavior on the elevated plus maze, two potential confounds in the evaluation of mouse social behavior. In the three-chamber social test, mice lacking integrin β3 were shown to have normal sociability but did not show a preference for social novelty. Importantly, the absence of integrin β3 did not impair olfaction or the ability to recall familiar social odors. Additionally, mice lacking integrin β3 showed increased grooming behavior in novel environments. These preliminary studies reveal altered social and repetitive behavior in these mice, which suggests that the integrin β3 subunit may be involved in brain systems relevant to ASD. Further work is needed to fully characterize these behavioral changes and the underlying brain mechanisms.
Collapse
Affiliation(s)
- Michelle D. Carter
- Department of Psychiatry, Vanderbilt University, Nashville, TN, USA, 37232
| | - Charisma R. Shah
- Department of Psychiatry, Vanderbilt University, Nashville, TN, USA, 37232
| | | | - Jacqueline N. Crawley
- Laboratory of Behavioral Neuroscience, National Institute of Mental Health, NIH Building 35 Room 1C-903, Bethesda, MD 20892
| | - Ana M. D. Carneiro
- Department of Pharmacology, Vanderbilt University, Nashville, TN, USA, 37232
- Kennedy Center for Research on Human Development, 465 21 Ave S, 7158 MRB III, Vanderbilt University, Nashville, TN, USA, 37232
| | - Jeremy Veenstra-VanderWeele
- Department of Psychiatry, Vanderbilt University, Nashville, TN, USA, 37232
- Department of Pharmacology, Vanderbilt University, Nashville, TN, USA, 37232
- Department of Pediatrics, Vanderbilt University, Nashville, TN, USA, 37232
- Center for Molecular Neuroscience, Vanderbilt University, Nashville, TN, USA, 37232
- Kennedy Center for Research on Human Development, 465 21 Ave S, 7158 MRB III, Vanderbilt University, Nashville, TN, USA, 37232
| |
Collapse
|
46
|
Ye H, Liu J, Wu JY. Cell adhesion molecules and their involvement in autism spectrum disorder. Neurosignals 2011; 18:62-71. [PMID: 21212702 DOI: 10.1159/000322543] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2010] [Accepted: 11/08/2010] [Indexed: 12/18/2022] Open
Abstract
Autism spectrum disorder (ASD) is a group of neurodevelopmental disorders characterized by abnormalities in social interaction, language development and behavior. Recent genetic studies demonstrate that alterations in synaptic genes including those encoding cell adhesion molecules and their interaction partners play important roles in the pathogenesis of ASD. Systematic analyses of different cell adhesion molecule genes will help elucidate their normal functions and regulatory mechanisms in the establishment and maintenance of normal neural circuits and uncover genetic aberrations contributing to ASD.
Collapse
Affiliation(s)
- Haihong Ye
- The State Key Laboratory of Brain and Cognitive Science, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China
| | | | | |
Collapse
|
47
|
Cochrane LE, Tansey KE, Gill M, Gallagher L, Anney RJ. Lack of association between markers in the ITGA3, ITGAV, ITGA6 and ITGB3 and autism in an Irish sample. Autism Res 2010; 3:342-4. [DOI: 10.1002/aur.157] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2009] [Accepted: 07/19/2010] [Indexed: 01/20/2023]
|
48
|
Napolioni V, Lombardi F, Sacco R, Curatolo P, Manzi B, Alessandrelli R, Militerni R, Bravaccio C, Lenti C, Saccani M, Schneider C, Melmed R, Pascucci T, Puglisi-Allegra S, Reichelt KL, Rousseau F, Lewin P, Persico AM. Family-based association study of ITGB3 in autism spectrum disorder and its endophenotypes. Eur J Hum Genet 2010; 19:353-9. [PMID: 21102624 DOI: 10.1038/ejhg.2010.180] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
The integrin-β 3 gene (ITGB3), located on human chromosome 17q21.3, was previously identified as a quantitative trait locus (QTL) for 5-HT blood levels and has been implicated as a candidate gene for autism spectrum disorder (ASD). We performed a family-based association study in 281 simplex and 12 multiplex Caucasian families. ITGB3 haplotypes are significantly associated with autism (HBAT, global P=0.038). Haplotype H3 is largely over-transmitted to the affected offspring and doubles the risk of an ASD diagnosis (HBAT P=0.005; odds ratio (OR)=2.000), at the expense of haplotype H1, which is under-transmitted (HBAT P=0.018; OR=0.725). These two common haplotypes differ only at rs12603582 located in intron 11, which reaches a P-value of 0.072 in single-marker FBAT analyses. Interestingly, rs12603582 is strongly associated with pre-term delivery in our ASD patients (P=0.008). On the other hand, it is SNP rs2317385, located at the 5' end of the gene, that significantly affects 5-HT blood levels (Mann-Whitney U-test, P=0.001; multiple regression analysis, P=0.010). No gene-gene interaction between ITGB3 and SLC6A4 has been detected. In conclusion, we identify a significant association between a common ITGB3 haplotype and ASD. Distinct markers, located toward the 5' and 3' ends of the gene, seemingly modulate 5-HT blood levels and autism liability, respectively. Our results also raise interest into ITGB3 influences on feto-maternal immune interactions in autism.
Collapse
Affiliation(s)
- Valerio Napolioni
- Laboratory of Molecular Psychiatry and Neurogenetics, University Campus Bio-Medico, Rome, Italy
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Key role for gene dosage and synaptic homeostasis in autism spectrum disorders. Trends Genet 2010; 26:363-72. [PMID: 20609491 DOI: 10.1016/j.tig.2010.05.007] [Citation(s) in RCA: 236] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2009] [Revised: 05/26/2010] [Accepted: 05/26/2010] [Indexed: 12/19/2022]
Abstract
Autism spectrum disorders (ASD) are characterized by impairments in reciprocal social communication, and repetitive, stereotyped verbal and non-verbal behaviors. Genetic studies have provided a relatively large number of genes that constitute a comprehensive framework to better understand this complex and heterogeneous syndrome. Based on the most robust findings, three observations can be made. First, genetic contributions to ASD are highly heterogeneous and most probably involve a combination of alleles with low and high penetrance. Second, the majority of the mutations apparently affect a single allele, suggesting a key role for gene dosage in susceptibility to ASD. Finally, the broad expression and function of the causative genes suggest that alteration of synaptic homeostasis could be a common biological process associated with ASD. Understanding the mechanisms that regulate synaptic homeostasis should shed new light on the causes of ASD and could provide a means to modulate the severity of the symptoms.
Collapse
|
50
|
Silverman JL, Yang M, Lord C, Crawley JN. Behavioural phenotyping assays for mouse models of autism. Nat Rev Neurosci 2010; 11:490-502. [PMID: 20559336 PMCID: PMC3087436 DOI: 10.1038/nrn2851] [Citation(s) in RCA: 1141] [Impact Index Per Article: 76.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Autism is a heterogeneous neurodevelopmental disorder of unknown aetiology that affects 1 in 100-150 individuals. Diagnosis is based on three categories of behavioural criteria: abnormal social interactions, communication deficits and repetitive behaviours. Strong evidence for a genetic basis has prompted the development of mouse models with targeted mutations in candidate genes for autism. As the diagnostic criteria for autism are behavioural, phenotyping these mouse models requires behavioural assays with high relevance to each category of the diagnostic symptoms. Behavioural neuroscientists are generating a comprehensive set of assays for social interaction, communication and repetitive behaviours to test hypotheses about the causes of autism. Robust phenotypes in mouse models hold great promise as translational tools for discovering effective treatments for components of autism spectrum disorders.
Collapse
Affiliation(s)
- Jill L Silverman
- National Institute of Mental Health, Porter Neuroscience Research Center, Bethesda, MD 20892-3730, USA
| | | | | | | |
Collapse
|