1
|
Ng B, Tasaki S, Greathouse KM, Walker CK, Zhang A, Covitz S, Cieslak M, Weber AJ, Adamson AB, Andrade JP, Poovey EH, Curtis KA, Muhammad HM, Seidlitz J, Satterthwaite T, Bennett DA, Seyfried NT, Vogel J, Gaiteri C, Herskowitz JH. Integration across biophysical scales identifies molecular and cellular correlates of person-to-person variability in human brain connectivity. Nat Neurosci 2024; 27:2240-2252. [PMID: 39482360 PMCID: PMC11537986 DOI: 10.1038/s41593-024-01788-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2023] [Accepted: 09/16/2024] [Indexed: 11/03/2024]
Abstract
Brain connectivity arises from interactions across biophysical scales, ranging from molecular to cellular to anatomical to network level. To date, there has been little progress toward integrated analysis across these scales. To bridge this gap, from a unique cohort of 98 individuals, we collected antemortem neuroimaging and genetic data, as well as postmortem dendritic spine morphometric, proteomic and gene expression data from the superior frontal and inferior temporal gyri. Through the integration of the molecular and dendritic spine morphology data, we identified hundreds of proteins that explain interindividual differences in functional connectivity and structural covariation. These proteins are enriched for synaptic structures and functions, energy metabolism and RNA processing. By integrating data at the genetic, molecular, subcellular and tissue levels, we link specific biochemical changes at synapses to connectivity between brain regions. These results demonstrate the feasibility of integrating data from vastly different biophysical scales to provide a more comprehensive understanding of brain connectivity.
Collapse
Affiliation(s)
- Bernard Ng
- Rush Alzheimer's Disease Center, Rush University Medical Center, Chicago, IL, USA
- Department of Psychiatry, SUNY Upstate Medical University, Syracuse, NY, USA
| | - Shinya Tasaki
- Rush Alzheimer's Disease Center, Rush University Medical Center, Chicago, IL, USA
| | - Kelsey M Greathouse
- Department of Neurology, Center for Neurodegeneration and Experimental Therapeutics, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Courtney K Walker
- Department of Neurology, Center for Neurodegeneration and Experimental Therapeutics, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Ada Zhang
- Department of Psychiatry, SUNY Upstate Medical University, Syracuse, NY, USA
| | - Sydney Covitz
- Penn/CHOP Lifespan Brain Institute, University of Pennsylvania, Philadelphia, PA, USA
- Department of Psychiatry, University of Pennsylvania, Philadelphia, PA, USA
- Penn Lifespan Informatics and Neuroimaging Center, University of Pennsylvania, Philadelphia, PA, USA
| | - Matt Cieslak
- Penn/CHOP Lifespan Brain Institute, University of Pennsylvania, Philadelphia, PA, USA
- Department of Psychiatry, University of Pennsylvania, Philadelphia, PA, USA
- Penn Lifespan Informatics and Neuroimaging Center, University of Pennsylvania, Philadelphia, PA, USA
| | - Audrey J Weber
- Department of Neurology, Center for Neurodegeneration and Experimental Therapeutics, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Ashley B Adamson
- Department of Neurology, Center for Neurodegeneration and Experimental Therapeutics, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Julia P Andrade
- Department of Neurology, Center for Neurodegeneration and Experimental Therapeutics, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Emily H Poovey
- Department of Neurology, Center for Neurodegeneration and Experimental Therapeutics, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Kendall A Curtis
- Department of Neurology, Center for Neurodegeneration and Experimental Therapeutics, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Hamad M Muhammad
- Department of Neurology, Center for Neurodegeneration and Experimental Therapeutics, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Jakob Seidlitz
- Penn/CHOP Lifespan Brain Institute, University of Pennsylvania, Philadelphia, PA, USA
- Department of Psychiatry, University of Pennsylvania, Philadelphia, PA, USA
- Penn Lifespan Informatics and Neuroimaging Center, University of Pennsylvania, Philadelphia, PA, USA
- Department of Child and Adolescent Psychiatry and Behavioral Science, The Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Ted Satterthwaite
- Penn/CHOP Lifespan Brain Institute, University of Pennsylvania, Philadelphia, PA, USA
- Department of Psychiatry, University of Pennsylvania, Philadelphia, PA, USA
- Penn Lifespan Informatics and Neuroimaging Center, University of Pennsylvania, Philadelphia, PA, USA
| | - David A Bennett
- Rush Alzheimer's Disease Center, Rush University Medical Center, Chicago, IL, USA
| | - Nicholas T Seyfried
- Department of Biochemistry, Emory University School of Medicine, Atlanta, GA, USA
| | - Jacob Vogel
- Penn/CHOP Lifespan Brain Institute, University of Pennsylvania, Philadelphia, PA, USA
- Department of Psychiatry, University of Pennsylvania, Philadelphia, PA, USA
- Penn Lifespan Informatics and Neuroimaging Center, University of Pennsylvania, Philadelphia, PA, USA
- Department of Clinical Science, Malmö, SciLifeLab, Lund University, Lund, Sweden
| | - Chris Gaiteri
- Rush Alzheimer's Disease Center, Rush University Medical Center, Chicago, IL, USA.
- Department of Psychiatry, SUNY Upstate Medical University, Syracuse, NY, USA.
| | - Jeremy H Herskowitz
- Department of Neurology, Center for Neurodegeneration and Experimental Therapeutics, University of Alabama at Birmingham, Birmingham, AL, USA.
| |
Collapse
|
2
|
Salluzzo M, Vianello C, Flotta F, Rimondini R, Carboni L. MicroRNAs Associated with IgLON Cell Adhesion Molecule Expression. Curr Issues Mol Biol 2024; 46:7702-7718. [PMID: 39057097 PMCID: PMC11276434 DOI: 10.3390/cimb46070456] [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: 06/28/2024] [Revised: 07/15/2024] [Accepted: 07/16/2024] [Indexed: 07/28/2024] Open
Abstract
The IgLON family of cell adhesion molecules consists of five members (LSAMP, OPCML, neurotrimin, NEGR1, and IgLON5) discovered as supporters of neuronal development, axon growth and guidance, and synapse formation and maintenance. Tumour suppression properties have recently been emerging based on antiproliferative effects through the modulation of oncogenic pathways. Available evidence endorses a role for non-coding RNAs or microRNAs as relevant controllers of IgLON molecule expression that can impact their critical physiological and pathological roles. Current findings support a function for long non-coding RNAs and microRNAs in the modulation of LSAMP expression in cell senescence, cancer biogenesis, addiction, and pulmonary hypertension. For OPCML, data point to a role for several microRNAs in the control of tumorigenesis. MicroRNAs were detected in neurotrimin-mediated functions in cancer biogenesis and in Schwann cell responses to peripheral nerve injury. For NEGR1, studies have mainly investigated microRNA involvement in neuronal responses to ischaemic injury, although data also exist about tumorigenesis and endothelial cell dysfunction. For IgLON5, information is only available about microRNA involved in myocardial infarction. In conclusion, despite much information being still missing and further research needed, the emerging picture favours a model in which non-coding RNAs exert a crucial role in modulating IgLON expression, ultimately affecting their important physiological functions.
Collapse
Affiliation(s)
- Marco Salluzzo
- Department of Pharmacy and Biotechnology, Alma Mater Studiorum University of Bologna, 40126 Bologna, Italy;
| | - Clara Vianello
- Department for Life Quality Studies, Alma Mater Studiorum University of Bologna, 47921 Rimini, Italy;
| | - Francesca Flotta
- Department of Medical and Surgical Sciences, Alma Mater Studiorum University of Bologna, 40126 Bologna, Italy; (F.F.); (R.R.)
| | - Roberto Rimondini
- Department of Medical and Surgical Sciences, Alma Mater Studiorum University of Bologna, 40126 Bologna, Italy; (F.F.); (R.R.)
| | - Lucia Carboni
- Department of Pharmacy and Biotechnology, Alma Mater Studiorum University of Bologna, 40126 Bologna, Italy;
| |
Collapse
|
3
|
Perry AS, Farber-Eger E, Gonzales T, Tanaka T, Robbins JM, Murthy VL, Stolze LK, Zhao S, Huang S, Colangelo LA, Deng S, Hou L, Lloyd-Jones DM, Walker KA, Ferrucci L, Watts EL, Barber JL, Rao P, Mi MY, Gabriel KP, Hornikel B, Sidney S, Houstis N, Lewis GD, Liu GY, Thyagarajan B, Khan SS, Choi B, Washko G, Kalhan R, Wareham N, Bouchard C, Sarzynski MA, Gerszten RE, Brage S, Wells QS, Nayor M, Shah RV. Proteomic analysis of cardiorespiratory fitness for prediction of mortality and multisystem disease risks. Nat Med 2024; 30:1711-1721. [PMID: 38834850 PMCID: PMC11186767 DOI: 10.1038/s41591-024-03039-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2023] [Accepted: 04/30/2024] [Indexed: 06/06/2024]
Abstract
Despite the wide effects of cardiorespiratory fitness (CRF) on metabolic, cardiovascular, pulmonary and neurological health, challenges in the feasibility and reproducibility of CRF measurements have impeded its use for clinical decision-making. Here we link proteomic profiles to CRF in 14,145 individuals across four international cohorts with diverse CRF ascertainment methods to establish, validate and characterize a proteomic CRF score. In a cohort of around 22,000 individuals in the UK Biobank, a proteomic CRF score was associated with a reduced risk of all-cause mortality (unadjusted hazard ratio 0.50 (95% confidence interval 0.48-0.52) per 1 s.d. increase). The proteomic CRF score was also associated with multisystem disease risk and provided risk reclassification and discrimination beyond clinical risk factors, as well as modulating high polygenic risk of certain diseases. Finally, we observed dynamicity of the proteomic CRF score in individuals who undertook a 20-week exercise training program and an association of the score with the degree of the effect of training on CRF, suggesting potential use of the score for personalization of exercise recommendations. These results indicate that population-based proteomics provides biologically relevant molecular readouts of CRF that are additive to genetic risk, potentially modifiable and clinically translatable.
Collapse
Affiliation(s)
- Andrew S Perry
- Vanderbilt Translational and Clinical Cardiovascular Research Center, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Eric Farber-Eger
- Vanderbilt Translational and Clinical Cardiovascular Research Center, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Tomas Gonzales
- MRC Epidemiology Unit, University of Cambridge, Cambridge, UK
| | - Toshiko Tanaka
- Longtidudinal Studies Section, Translational Gerontology Branch, National Institute on Aging, NIH, Baltimore, MD, USA
| | - Jeremy M Robbins
- Cardiovascular Institute, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | | | - Lindsey K Stolze
- Department of Biostatistics, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Shilin Zhao
- Department of Biostatistics, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Shi Huang
- Department of Biostatistics, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Laura A Colangelo
- Department of Preventive Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Shuliang Deng
- Cardiovascular Institute, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Lifang Hou
- Department of Preventive Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Donald M Lloyd-Jones
- Department of Preventive Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Keenan A Walker
- Multimodal Imaging of Neurodegenerative Disease (MIND) Unit, National Institute on Aging, NIH, Baltimore, MD, USA
| | - Luigi Ferrucci
- Longtidudinal Studies Section, Translational Gerontology Branch, National Institute on Aging, NIH, Baltimore, MD, USA
| | - Eleanor L Watts
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Rockville, MD, USA
| | - Jacob L Barber
- Cardiovascular Institute, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Prashant Rao
- Cardiovascular Institute, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Michael Y Mi
- Cardiovascular Institute, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Kelley Pettee Gabriel
- Department of Epidemiology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Bjoern Hornikel
- Department of Epidemiology, University of Alabama at Birmingham, Birmingham, AL, USA
| | | | - Nicholas Houstis
- Cardiology Division, Massachusetts General Hospital, Boston, MA, USA
| | - Gregory D Lewis
- Cardiology Division, Massachusetts General Hospital, Boston, MA, USA
| | - Gabrielle Y Liu
- Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Medicine, University of California Davis, Sacramento, CA, USA
| | - Bharat Thyagarajan
- Department of Laboratory Medicine and Pathology, University of Minnesota, Minnesota, MN, USA
| | - Sadiya S Khan
- Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Bina Choi
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Brigham and Women's Hospital, Boston, MA, USA
| | - George Washko
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Brigham and Women's Hospital, Boston, MA, USA
| | - Ravi Kalhan
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Nick Wareham
- MRC Epidemiology Unit, University of Cambridge, Cambridge, UK
| | - Claude Bouchard
- Human Genomic Laboratory, Pennington Biomedical Research Center, Baton Rouge, LA, USA
| | - Mark A Sarzynski
- Department of Exercise Science, University of South Carolina Columbia, Columbia, SC, USA
| | - Robert E Gerszten
- Cardiovascular Institute, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Soren Brage
- MRC Epidemiology Unit, University of Cambridge, Cambridge, UK
| | - Quinn S Wells
- Vanderbilt Translational and Clinical Cardiovascular Research Center, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Matthew Nayor
- Sections of Cardiovascular Medicine and Preventive Medicine and Epidemiology, Department of Medicine, Boston University School of Medicine, Boston, MA, USA
| | - Ravi V Shah
- Vanderbilt Translational and Clinical Cardiovascular Research Center, Vanderbilt University School of Medicine, Nashville, TN, USA.
| |
Collapse
|
4
|
Chen PB, Chen R, LaPierre N, Chen Z, Mefford J, Marcus E, Heffel MG, Soto DC, Ernst J, Luo C, Flint J. Complementation testing identifies genes mediating effects at quantitative trait loci underlying fear-related behavior. CELL GENOMICS 2024; 4:100545. [PMID: 38697120 PMCID: PMC11099346 DOI: 10.1016/j.xgen.2024.100545] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/03/2024] [Revised: 02/23/2024] [Accepted: 04/04/2024] [Indexed: 05/04/2024]
Abstract
Knowing the genes involved in quantitative traits provides an entry point to understanding the biological bases of behavior, but there are very few examples where the pathway from genetic locus to behavioral change is known. To explore the role of specific genes in fear behavior, we mapped three fear-related traits, tested fourteen genes at six quantitative trait loci (QTLs) by quantitative complementation, and identified six genes. Four genes, Lamp, Ptprd, Nptx2, and Sh3gl, have known roles in synapse function; the fifth, Psip1, was not previously implicated in behavior; and the sixth is a long non-coding RNA, 4933413L06Rik, of unknown function. Variation in transcriptome and epigenetic modalities occurred preferentially in excitatory neurons, suggesting that genetic variation is more permissible in excitatory than inhibitory neuronal circuits. Our results relieve a bottleneck in using genetic mapping of QTLs to uncover biology underlying behavior and prompt a reconsideration of expected relationships between genetic and functional variation.
Collapse
Affiliation(s)
- Patrick B Chen
- Department of Psychiatry and Biobehavioral Sciences, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA
| | - Rachel Chen
- Department of Psychiatry and Biobehavioral Sciences, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA
| | - Nathan LaPierre
- Department of Computer Science, Samueli School of Engineering, University of California, Los Angeles, Los Angeles, CA, USA; Department of Human Genetics, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA
| | - Zeyuan Chen
- Department of Computer Science, Samueli School of Engineering, University of California, Los Angeles, Los Angeles, CA, USA
| | - Joel Mefford
- Department of Human Genetics, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA
| | - Emilie Marcus
- Department of Biological Chemistry, University of California, Los Angeles, Los Angeles, CA, USA
| | - Matthew G Heffel
- Department of Human Genetics, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA
| | - Daniela C Soto
- Department of Psychiatry and Biobehavioral Sciences, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA
| | - Jason Ernst
- Department of Computer Science, Samueli School of Engineering, University of California, Los Angeles, Los Angeles, CA, USA; Department of Biological Chemistry, University of California, Los Angeles, Los Angeles, CA, USA
| | - Chongyuan Luo
- Department of Human Genetics, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA
| | - Jonathan Flint
- Department of Psychiatry and Biobehavioral Sciences, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA.
| |
Collapse
|
5
|
Chen PB, Chen R, LaPierre N, Chen Z, Mefford J, Marcus E, Heffel MG, Soto DC, Ernst J, Luo C, Flint J. Complementation testing identifies causal genes at quantitative trait loci underlying fear related behavior. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.01.03.574060. [PMID: 38260483 PMCID: PMC10802323 DOI: 10.1101/2024.01.03.574060] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/24/2024]
Abstract
Knowing the genes involved in quantitative traits provides a critical entry point to understanding the biological bases of behavior, but there are very few examples where the pathway from genetic locus to behavioral change is known. Here we address a key step towards that goal by deploying a test that directly queries whether a gene mediates the effect of a quantitative trait locus (QTL). To explore the role of specific genes in fear behavior, we mapped three fear-related traits, tested fourteen genes at six QTLs, and identified six genes. Four genes, Lsamp, Ptprd, Nptx2 and Sh3gl, have known roles in synapse function; the fifth gene, Psip1, is a transcriptional co-activator not previously implicated in behavior; the sixth is a long non-coding RNA 4933413L06Rik with no known function. Single nucleus transcriptomic and epigenetic analyses implicated excitatory neurons as likely mediating the genetic effects. Surprisingly, variation in transcriptome and epigenetic modalities between inbred strains occurred preferentially in excitatory neurons, suggesting that genetic variation is more permissible in excitatory than inhibitory neuronal circuits. Our results open a bottleneck in using genetic mapping of QTLs to find novel biology underlying behavior and prompt a reconsideration of expected relationships between genetic and functional variation.
Collapse
|
6
|
Salluzzo M, Vianello C, Abdullatef S, Rimondini R, Piccoli G, Carboni L. The Role of IgLON Cell Adhesion Molecules in Neurodegenerative Diseases. Genes (Basel) 2023; 14:1886. [PMID: 37895235 PMCID: PMC10606101 DOI: 10.3390/genes14101886] [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: 09/01/2023] [Revised: 09/26/2023] [Accepted: 09/27/2023] [Indexed: 10/29/2023] Open
Abstract
In the brain, cell adhesion molecules (CAMs) are critical for neurite outgrowth, axonal fasciculation, neuronal survival and migration, and synapse formation and maintenance. Among CAMs, the IgLON family comprises five members: Opioid Binding Protein/Cell Adhesion Molecule Like (OPCML or OBCAM), Limbic System Associated Membrane Protein (LSAMP), neurotrimin (NTM), Neuronal Growth Regulator 1 (NEGR1), and IgLON5. IgLONs exhibit three N-terminal C2 immunoglobulin domains; several glycosylation sites; and a glycosylphosphatidylinositol anchoring to the membrane. Interactions as homo- or heterodimers in cis and in trans, as well as binding to other molecules, appear critical for their functions. Shedding by metalloproteases generates soluble factors interacting with cellular receptors and activating signal transduction. The aim of this review was to analyse the available data implicating a role for IgLONs in neuropsychiatric disorders. Starting from the identification of a pathological role for antibodies against IgLON5 in an autoimmune neurodegenerative disease with a poorly understood mechanism of action, accumulating evidence links IgLONs to neuropsychiatric disorders, albeit with still undefined mechanisms which will require future thorough investigations.
Collapse
Affiliation(s)
- Marco Salluzzo
- Department of Pharmacy and Biotechnology, Alma Mater Studiorum University of Bologna, 40126 Bologna, Italy;
| | - Clara Vianello
- Department of Medical and Surgical Sciences, Alma Mater Studiorum University of Bologna, 40126 Bologna, Italy; (C.V.); (R.R.)
| | - Sandra Abdullatef
- Department of Cellular, Computational and Integrative Biology, University of Trento, 38123 Trento, Italy; (S.A.); (G.P.)
| | - Roberto Rimondini
- Department of Medical and Surgical Sciences, Alma Mater Studiorum University of Bologna, 40126 Bologna, Italy; (C.V.); (R.R.)
| | - Giovanni Piccoli
- Department of Cellular, Computational and Integrative Biology, University of Trento, 38123 Trento, Italy; (S.A.); (G.P.)
| | - Lucia Carboni
- Department of Pharmacy and Biotechnology, Alma Mater Studiorum University of Bologna, 40126 Bologna, Italy;
| |
Collapse
|
7
|
Yamashita A, Shichino Y, Fujii K, Koshidaka Y, Adachi M, Sasagawa E, Mito M, Nakagawa S, Iwasaki S, Takao K, Shiina N. ILF3 prion-like domain regulates gene expression and fear memory under chronic stress. iScience 2023; 26:106229. [PMID: 36876121 PMCID: PMC9982275 DOI: 10.1016/j.isci.2023.106229] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2022] [Revised: 01/11/2023] [Accepted: 02/14/2023] [Indexed: 02/22/2023] Open
Abstract
The prion-like domain (PrLD) is a class of intrinsically disordered regions. Although its propensity to form condensates has been studied in the context of neurodegenerative diseases, the physiological role of PrLD remains unclear. Here, we investigated the role of PrLD in the RNA-binding protein NFAR2, generated by a splicing variant of the Ilf3 gene. Removal of the PrLD in mice did not impair the function of NFAR2 required for survival, but did affect the responses to chronic water immersion and restraint stress (WIRS). The PrLD was required for WIRS-sensitive nuclear localization of NFAR2 and WIRS-induced changes in mRNA expression and translation in the amygdala, a fear-related brain region. Consistently, the PrLD conferred resistance to WIRS in fear-associated memory formation. Our study provides insights into the PrLD-dependent role of NFAR2 for chronic stress adaptation in the brain.
Collapse
Affiliation(s)
- Akira Yamashita
- Laboratory of Neuronal Cell Biology, National Institute for Basic Biology, Okazaki, Aichi 444-8585, Japan
- Department of Basic Biology, The Graduate University for Advanced Studies, SOKENDAI, Okazaki, Aichi 444-8585, Japan
| | - Yuichi Shichino
- RNA Systems Biochemistry Laboratory, RIKEN Cluster for Pioneering Research, Wako, Saitama 351-0198, Japan
| | - Kazuki Fujii
- Department of Behavioral Physiology, Faculty of Medicine, University of Toyama, Toyama 930-0194, Japan
- Life Science Research Center, University of Toyama, Toyama 930-0194, Japan
- Research Center for Idling Brain Science, University of Toyama, Toyama 930-0194, Japan
| | - Yumie Koshidaka
- Life Science Research Center, University of Toyama, Toyama 930-0194, Japan
| | - Mayumi Adachi
- Life Science Research Center, University of Toyama, Toyama 930-0194, Japan
| | - Eri Sasagawa
- Department of Behavioral Physiology, Graduate School of Innovative Life Science, University of Toyama, Toyama 930-0194, Japan
| | - Mari Mito
- RNA Systems Biochemistry Laboratory, RIKEN Cluster for Pioneering Research, Wako, Saitama 351-0198, Japan
| | - Shinichi Nakagawa
- RNA Biology Laboratory, Faculty of Pharmaceutical Sciences, Hokkaido University, Sapporo Hokkaido 060-0812, Japan
| | - Shintaro Iwasaki
- RNA Systems Biochemistry Laboratory, RIKEN Cluster for Pioneering Research, Wako, Saitama 351-0198, Japan
- Department of Computational Biology and Medical Sciences, Graduate School of Frontier Sciences, The University of Tokyo, Kashiwa, Chiba 277-8561, Japan
| | - Keizo Takao
- Department of Behavioral Physiology, Faculty of Medicine, University of Toyama, Toyama 930-0194, Japan
- Life Science Research Center, University of Toyama, Toyama 930-0194, Japan
- Research Center for Idling Brain Science, University of Toyama, Toyama 930-0194, Japan
- Department of Behavioral Physiology, Graduate School of Innovative Life Science, University of Toyama, Toyama 930-0194, Japan
| | - Nobuyuki Shiina
- Laboratory of Neuronal Cell Biology, National Institute for Basic Biology, Okazaki, Aichi 444-8585, Japan
- Department of Basic Biology, The Graduate University for Advanced Studies, SOKENDAI, Okazaki, Aichi 444-8585, Japan
- Exploratory Research Center on Life and Living Systems (ExCELLS), National Institutes of Natural Sciences, Okazaki, Aichi 444-8585, Japan
- Corresponding author
| |
Collapse
|
8
|
Machado PC, Brito LF, Martins R, Pinto LFB, Silva MR, Pedrosa VB. Genome-Wide Association Analysis Reveals Novel Loci Related with Visual Score Traits in Nellore Cattle Raised in Pasture-Based Systems. Animals (Basel) 2022; 12:ani12243526. [PMID: 36552446 PMCID: PMC9774243 DOI: 10.3390/ani12243526] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2022] [Revised: 12/06/2022] [Accepted: 12/10/2022] [Indexed: 12/15/2022] Open
Abstract
Body conformation traits assessed based on visual scores are widely used in Zebu cattle breeding programs. The aim of this study was to identify genomic regions and biological pathways associated with body conformation (CONF), finishing precocity (PREC), and muscling (MUSC) in Nellore cattle. The measurements based on visual scores were collected in 20,807 animals raised in pasture-based systems in Brazil. In addition, 2775 animals were genotyped using a 35 K SNP chip, which contained 31,737 single nucleotide polymorphisms after quality control. Single-step GWAS was performed using the BLUPF90 software while candidate genes were identified based on the Ensembl Genes 69. PANTHER and REVIGO platforms were used to identify key biological pathways and STRING to create gene networks. Novel candidate genes were revealed associated with CONF, including ALDH9A1, RXRG, RAB2A, and CYP7A1, involved in lipid metabolism. The genes associated with PREC were ELOVL5, PID1, DNER, TRIP12, and PLCB4, which are related to the synthesis of long-chain fatty acids, lipid metabolism, and muscle differentiation. For MUSC, the most important genes associated with muscle development were SEMA6A, TIAM2, UNC5A, and UIMC1. The polymorphisms identified in this study can be incorporated in commercial genotyping panels to improve the accuracy of genomic evaluations for visual scores in beef cattle.
Collapse
Affiliation(s)
- Pamela C. Machado
- Department of Animal Sciences, State University of Ponta Grossa, Ponta Grossa 84030-900, PR, Brazil
| | - Luiz F. Brito
- Department of Animal Sciences, Purdue University, West Lafayette, IN 47907, USA
| | - Rafaela Martins
- Department of Animal Sciences, State University of Ponta Grossa, Ponta Grossa 84030-900, PR, Brazil
| | - Luis Fernando B. Pinto
- Department of Animal Science, Federal University of Bahia, Av. Adhemar de Barros 500, Ondina, Salvador 40170-110, BA, Brazil
| | - Marcio R. Silva
- Melhore Animal and Katayama Agropecuaria Lda, Guararapes 16700-000, SP, Brazil
| | - Victor B. Pedrosa
- Department of Animal Sciences, State University of Ponta Grossa, Ponta Grossa 84030-900, PR, Brazil
- Department of Animal Sciences, Purdue University, West Lafayette, IN 47907, USA
- Correspondence:
| |
Collapse
|
9
|
Feleke M, Feng W, Rothzerg E, Song D, Wei Q, Kõks S, Wood D, Liu Y, Xu J. Single-cell RNA-seq identification of four differentially expressed survival-related genes by a TARGET: Osteosarcoma database analysis. Exp Biol Med (Maywood) 2022; 247:921-930. [PMID: 35285281 PMCID: PMC9189571 DOI: 10.1177/15353702221080131] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2021] [Accepted: 01/23/2022] [Indexed: 09/05/2023] Open
Abstract
Osteosarcoma (OS) differentially expressed genes (DEGs) have been predicted using the data portal of the Therapeutically Applicable Research to Generate Effective Treatments (TARGET). In this study, we sought to identify cell types that specially express key DEGs (MUC1, COL13A1, JAG2, and KAZALD1) in each of the nine identified cell populations derived from tissues of OS tumors with single-cell RNA-sequencing data. Gene expression levels were pairwise compared between cell clusters and a p value < 0.05 was considered differentially expressed. It was revealed that MUC1 is expressed at high levels in osteoblastic OS cells followed by carcinoma-associated fibroblasts (CAFs) and plasmocytes, respectively. COL13A1 is highly expressed in osteoblastic OS cells, CAFs, and endothelial cells (ECs), respectively. The KAZALD1 gene is expressed in CAFs and osteoblastic OS cells at high levels, but at very low levels in plasmocytes, osteoclasts, NK/T, myeloid cells 1, myeloid cells 2, ECs, and B cells. JAG2 is expressed at significantly high levels in ECs and osteoblastic OS cells, and at relatively lower levels in all other cell types. Interestingly, LSAMP, as an established gene in the development of OS shows high expression in osteoblastic OS cells and CAFs but low in other cells such as osteoclasts. Our findings here highlight the heterogeneity of OS cells and cell-type-dependent DEGs which have potential as therapeutic targets in OS.
Collapse
Affiliation(s)
- Mesalie Feleke
- School of Biomedical Sciences, The University of Western Australia, Perth, WA 6009, Australia
| | - Wenyu Feng
- Department of Orthopaedics, First Affiliated Hospital of Guangxi Medical University, Nanning 530021, China
| | - Emel Rothzerg
- School of Biomedical Sciences, The University of Western Australia, Perth, WA 6009, Australia
| | - Dezhi Song
- School of Biomedical Sciences, The University of Western Australia, Perth, WA 6009, Australia
- Research Centre for Regenerative Medicine, Guangxi Medical University, Nanning 530021, China
| | - Qingjun Wei
- Department of Orthopaedics, First Affiliated Hospital of Guangxi Medical University, Nanning 530021, China
| | - Sulev Kõks
- Perron Institute for Neurological and Translational Science, Queen Elizabeth II Medical Centre, Nedlands, WA 6009, Australia
- Centre for Molecular Medicine and Innovative Therapeutics, Murdoch University, Murdoch, WA 6150, Australia
| | - David Wood
- Medical School, The University of Western Australia, Perth, WA 6009, Australia
| | - Yun Liu
- School of Biomedical Sciences, The University of Western Australia, Perth, WA 6009, Australia
- Department of Orthopaedics, First Affiliated Hospital of Guangxi Medical University, Nanning 530021, China
| | - Jiake Xu
- School of Biomedical Sciences, The University of Western Australia, Perth, WA 6009, Australia
| |
Collapse
|
10
|
Bregin A, Kaare M, Jagomäe T, Karis K, Singh K, Laugus K, Innos J, Leidmaa E, Heinla I, Visnapuu T, Oja EM, Kõiv K, Lilleväli K, Harro J, Philips MA, Vasar E. Expression and impact of Lsamp neural adhesion molecule in the serotonergic neurotransmission system. Pharmacol Biochem Behav 2020; 198:173017. [PMID: 32828972 DOI: 10.1016/j.pbb.2020.173017] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/19/2020] [Revised: 08/15/2020] [Accepted: 08/17/2020] [Indexed: 12/22/2022]
Abstract
Limbic system associated membrane protein (Lsamp) is a neural adhesion protein which has been recently found to be differentially expressed between serotonergic neuron subtypes. We have previously shown elevated serotonin (5-HT) turnover rate in Lsamp-deficient mice. The purpose of the current study was to elucidate the role of Lsamp in serotonergic neurotransmission. Chronic (18 days) administration of serotonin reuptake inhibitor (SSRI) escitalopram (10 mg/kg) significantly increased general activity in wild-type mice in the open field and protected exploration in Lsamp-/- mice in the elevated-plus maze. An important psychopathology-related endophenotype, elevated 5-HT turnover in the brain of Lsamp-deficient mice, was reproduced in the saline group. Escitalopram restored the elevated 5-HT turnover of Lsamp-deficient mice to a level comparable with their wild-type littermates, suggesting that high 5-HT turnover in mutants is mediated by the increased activity of serotonin transporter (SERT protein encoded by Slc6a4 gene). The baseline level of Slc6a4 transcript was not changed in Lsamp-deficient mice, however, our immunohistochemical analysis showed partial co-expression of Lsamp with both SERT and Tph2 proteins in raphe. Overactivity of SERT in Lsamp-/- mice is further supported by significant elevation of Maoa transcript and increase of DOPAC, another Mao A product, specifically in the raphe. Again, elevation of DOPAC was reduced to the level of wild-type by chronic SSRI treatment. The activity of Lsamp gene promoters varied in 5-HT producing nuclei: both Lsamp 1a and 1b promoters were active in the dorsal raphe; most of the expression in the median raphe was from 1b promoter, whereas Lsamp 1a promoter was almost exclusively active in the caudal subgroup of raphe nuclei. We suggest that Lsamp may have an impact on the integrity of serotonergic synapses, which is possibly the neurochemical basis of the anxiety- and sociability-related phenotype in Lsamp-deficient mice.
Collapse
Affiliation(s)
- Aleksandr Bregin
- Department of Physiology, Institute of Biomedicine and Translational Medicine, University of Tartu, 19 Ravila Street, 50411 Tartu, Estonia; Centre of Excellence in Genomics and Translational Medicine, University of Tartu, Tartu, Estonia
| | - Maria Kaare
- Department of Physiology, Institute of Biomedicine and Translational Medicine, University of Tartu, 19 Ravila Street, 50411 Tartu, Estonia; Centre of Excellence in Genomics and Translational Medicine, University of Tartu, Tartu, Estonia
| | - Toomas Jagomäe
- Department of Physiology, Institute of Biomedicine and Translational Medicine, University of Tartu, 19 Ravila Street, 50411 Tartu, Estonia; Centre of Excellence in Genomics and Translational Medicine, University of Tartu, Tartu, Estonia
| | - Karina Karis
- Department of Physiology, Institute of Biomedicine and Translational Medicine, University of Tartu, 19 Ravila Street, 50411 Tartu, Estonia; Centre of Excellence in Genomics and Translational Medicine, University of Tartu, Tartu, Estonia
| | - Katyayani Singh
- Department of Physiology, Institute of Biomedicine and Translational Medicine, University of Tartu, 19 Ravila Street, 50411 Tartu, Estonia; Centre of Excellence in Genomics and Translational Medicine, University of Tartu, Tartu, Estonia
| | - Karita Laugus
- Division of Neuropsychopharmacology, Department of Psychology, Estonian Centre of Behavioural and Health Sciences, University of Tartu, Estonia
| | - Jürgen Innos
- Department of Physiology, Institute of Biomedicine and Translational Medicine, University of Tartu, 19 Ravila Street, 50411 Tartu, Estonia; Centre of Excellence in Genomics and Translational Medicine, University of Tartu, Tartu, Estonia
| | - Este Leidmaa
- Institute of Molecular Psychiatry, Medical Faculty, University of Bonn, Bonn, Germany
| | - Indrek Heinla
- Department of Psychology, UiT The Arctic University of Norway, Tromsø, Norway
| | - Tanel Visnapuu
- Department of Physiology, Institute of Biomedicine and Translational Medicine, University of Tartu, 19 Ravila Street, 50411 Tartu, Estonia; Centre of Excellence in Genomics and Translational Medicine, University of Tartu, Tartu, Estonia
| | - Eva-Maria Oja
- Department of Physiology, Institute of Biomedicine and Translational Medicine, University of Tartu, 19 Ravila Street, 50411 Tartu, Estonia; Centre of Excellence in Genomics and Translational Medicine, University of Tartu, Tartu, Estonia
| | - Kadri Kõiv
- Division of Neuropsychopharmacology, Department of Psychology, Estonian Centre of Behavioural and Health Sciences, University of Tartu, Estonia
| | - Kersti Lilleväli
- Department of Physiology, Institute of Biomedicine and Translational Medicine, University of Tartu, 19 Ravila Street, 50411 Tartu, Estonia; Centre of Excellence in Genomics and Translational Medicine, University of Tartu, Tartu, Estonia
| | - Jaanus Harro
- Division of Neuropsychopharmacology, Department of Psychology, Estonian Centre of Behavioural and Health Sciences, University of Tartu, Estonia
| | - Mari-Anne Philips
- Department of Physiology, Institute of Biomedicine and Translational Medicine, University of Tartu, 19 Ravila Street, 50411 Tartu, Estonia; Centre of Excellence in Genomics and Translational Medicine, University of Tartu, Tartu, Estonia.
| | - Eero Vasar
- Department of Physiology, Institute of Biomedicine and Translational Medicine, University of Tartu, 19 Ravila Street, 50411 Tartu, Estonia; Centre of Excellence in Genomics and Translational Medicine, University of Tartu, Tartu, Estonia
| |
Collapse
|
11
|
Forebrain Transcriptional Response to Transient Changes in Circulating Androgens in a Cichlid Fish. G3-GENES GENOMES GENETICS 2020; 10:1971-1982. [PMID: 32276961 PMCID: PMC7263668 DOI: 10.1534/g3.119.400947] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
It has been hypothesized that androgens respond to the social interactions as a way to adjust the behavior of individuals to the challenges of the social environment in an adaptive manner. Therefore, it is expected that transient changes in circulating androgen levels within physiological scope should impact the state of the brain network that regulates social behavior, which should translate into adaptive behavioral changes. Here, we examined the effect that a transient peak in androgen circulating levels, which mimics socially driven changes in androgen levels, has on the forebrain state, which harbors most nuclei of the social decision-making network. For this purpose, we successfully induced transient changes in circulating androgen levels in an African cichlid fish (Mozambique tilapia, Oreochromis mossambicus) commonly used as a model in behavioral neuroendocrinology by injecting 11-ketotestosterone or testosterone, and compared the forebrain transcriptome of these individuals to control fish injected with vehicle. Forebrain samples were collected 30 min and 60 min after injection and analyzed using RNAseq. Our results showed that a transient peak in 11-ketotestosterone drives more accentuated changes in forebrain transcriptome than testosterone, and that transcriptomic impact was greater at the 30 min than at the 60 min post-androgen administration. Several genes involved in the regulation of translation, steroid metabolism, ion channel membrane receptors, and genes involved in epigenetic mechanisms were differentially expressed after 11-ketotestosterone or testosterone injection. In summary, this study identified specific candidate genes that may regulate socially driven changes in behavioral flexibility mediated by androgens.
Collapse
|
12
|
Noh K, Park JC, Han JS, Lee SJ. From Bound Cells Comes a Sound Mind: The Role of Neuronal Growth Regulator 1 in Psychiatric Disorders. Exp Neurobiol 2020; 29:1-10. [PMID: 32122104 PMCID: PMC7075657 DOI: 10.5607/en.2020.29.1.1] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2020] [Revised: 02/23/2020] [Accepted: 02/25/2020] [Indexed: 12/21/2022] Open
Abstract
Cell-to-cell adhesion is important for maintenance of brain structure and function. Abnormal neuronal cell adhesion and loss of its connectivity are considered a main cause of psychiatric disorders such as major depressive disorder (MDD). Various cell adhesion molecules (CAMs) are involved in neuronal cell adhesions and thereby affect brain functions such as learning and memory, cognitive functions, and psychiatric functions. Compared with other CAMs, neuronal growth regulator 1 (Negr1) has a distinct functioning mechanism in terms of its cross-talk with cytokine receptor signaling. Negr1 is a member of the immunoglobulin LON (IgLON) family of proteins and is involved in neuronal outgrowth, dendritic arborization, and synapse formation. In humans, Negr1 is a risk gene for obesity based on a genome-wide association study. More recently, accumulating evidence supports that it also plays a critical role in psychiatric disorders. In this review, we discuss the recent findings on the role of Negr1 in MDD, focusing on its regulatory mechanism. We also provide evidence of putative involvement of Negr1 in other psychiatric disorders based on the novel behavioral phenotypes of Negr1 knockout mice.
Collapse
Affiliation(s)
- Kyungchul Noh
- Department of Physiology and Neuroscience, Dental Research Institute, Seoul National University School of Dentistry, Seoul 08826, Korea
| | - Jung-Cheol Park
- Department of Biological Science, Konkuk University, Seoul 05029, Korea
| | - Jung-Soo Han
- Department of Biological Science, Konkuk University, Seoul 05029, Korea
| | - Sung Joong Lee
- Department of Physiology and Neuroscience, Dental Research Institute, Seoul National University School of Dentistry, Seoul 08826, Korea
| |
Collapse
|
13
|
Nissen MS, Blaabjerg M. Anti-IgLON5 Disease: A Case With 11-Year Clinical Course and Review of the Literature. Front Neurol 2019; 10:1056. [PMID: 31632341 PMCID: PMC6783555 DOI: 10.3389/fneur.2019.01056] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2019] [Accepted: 09/18/2019] [Indexed: 11/13/2022] Open
Abstract
Background: Anti-IgLON5 disease is a novel disorder with a complex interplay between inflammation and neurodegeneration. Patients develop antibodies against IgLON5 but also deposition of neuronal tau protein. Symptoms often have an insidious onset, slow progression and mimic other neurological disorders. Here we report a case with severely prolonged 11-year disease course and provide a review of current reported cases with focus on presentation, work-up, treatment, and outcome. Method: All reported cases of anti-IgLON5 disease were evaluated. Cases reported twice (in case series and as single case reports), were carefully excluded. Results: Most patients display a characteristic sleep disorder with severe insomnia, non rapid eye movement (NREM) parasomnia, with finalistic movements and sleep disordered breathing (stridor and obstructive sleep apnea). Other symptoms are bulbar involvement, gait instability, movement disorders, oculomotor abnormalities, dysautonomia, and peripheral symptoms. Antibodies are present in both serum and CSF and there is a strong correlation with human leukocyte antigen (HLA) DRB1*10:01 and HLA-DQB1*05:01. Neuropathological examination reveals neurodegeneration with neuronal tau deposits in regions that correlate with the clinical presentation (e.g., predominantly hypothalamus and tegmentum of the brain stem). Majority of cases respond partially to immunotherapy. Cases, who received no treatment or treatment with IV corticosteroids alone, had a higher mortality than cases treated with more potent immunotherapy. Conclusion: The clinical spectrum of Anti-IgLON5 disease continues to expand. Further studies are needed to elucidate the pathophysiology, therapeutic strategies and outcome in this novel disorder. Aggressive immunotherapy seems to increase survival.
Collapse
Affiliation(s)
- Mette Scheller Nissen
- Department of Neurology, Odense University Hospital, Odense, Denmark.,Department of Clinical Research, University of Southern Denmark, Odense, Denmark.,BRIDGE, Brain Research - Inter-Disciplinary Guided Excellence, University of Southern Denmark, Odense, Denmark
| | - Morten Blaabjerg
- Department of Neurology, Odense University Hospital, Odense, Denmark.,Department of Clinical Research, University of Southern Denmark, Odense, Denmark.,BRIDGE, Brain Research - Inter-Disciplinary Guided Excellence, University of Southern Denmark, Odense, Denmark
| |
Collapse
|
14
|
Kubick N, Brösamle D, Mickael ME. Molecular Evolution and Functional Divergence of the IgLON Family. Evol Bioinform Online 2018; 14:1176934318775081. [PMID: 29844654 PMCID: PMC5967153 DOI: 10.1177/1176934318775081] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2018] [Accepted: 04/09/2018] [Indexed: 11/15/2022] Open
Abstract
IgLON family is a subgroup of cell adhesion molecules which is known to have diverse roles in neuronal development. IgLONs are characterized by possessing 3 Ig-like C2 domains, which play a part in mediating various cellular interactions. Recently, IgLONs have been shown to be expressed at the blood-brain barrier (BBB). However, our understanding of the genetic divergence patterns and evolutionary rates of these proteins in relation to their functions, in general, and at the BBB, in particular, remains inadequate. In this study, 12 species were explored to shed more light on the phylogenetic origins, structure, functional specificity, and divergence of this family. A total of 40 IgLON genes were identified from vertebrates and invertebrates. The absence of IgLON family genes in Hydra vulgaris and Nematostella vectensis but not in Drosophila melanogaster suggests that this family appeared during the time of divergence of Arthropoda 455 Mya. In general, IgLON genes have been subject to strong positive selection in vertebrates. Our study, based on IgLONs’ structural similarity, suggests that they may play a role in the evolutionary changes in the brain anatomy towards complexity including regulating neural growth and BBB permeability. IgLONs’ functions seem to be performed through complex interactions on the level of motifs as well as single residues. We identified several IgLON motifs that could be influencing cellular migration and proliferation as well as BBB integrity through interactions with SH3 or integrin. Our motif analysis also revealed that NEGR1 might be involved in MAPK pathway as a form of a signal transmitting receptor through its motif (KKVRVVVNF). We found several residues that were both positively selected and with highly functional specificity. We also located functional divergent residues that could act as drug targets to regulate BBB permeability. Furthermore, we identified several putative metalloproteinase cleavage sites that support the ectodomain shedding hypothesis of the IgLONs. In conclusion, our results present a bridge between IgLONs’ molecular evolution and their functions.
Collapse
Affiliation(s)
- Norwin Kubick
- Institute of Biochemistry, Molecular Cell Biology, University Clinic Hamburg-Eppendorf, Hamburg, Germany
| | - Desiree Brösamle
- Institute of Medical Systems Biology, Center for Molecular Neurobiology Hamburg, University Clinic Hamburg-Eppendorf, Hamburg, Germany
| | - Michel-Edwar Mickael
- Institute of Medical Systems Biology, Center for Molecular Neurobiology Hamburg, University Clinic Hamburg-Eppendorf, Hamburg, Germany
| |
Collapse
|
15
|
Singh K, Lilleväli K, Gilbert SF, Bregin A, Narvik J, Jayaram M, Rahi M, Innos J, Kaasik A, Vasar E, Philips MA. The combined impact of IgLON family proteins Lsamp and Neurotrimin on developing neurons and behavioral profiles in mouse. Brain Res Bull 2018; 140:5-18. [PMID: 29605488 DOI: 10.1016/j.brainresbull.2018.03.013] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2017] [Revised: 02/26/2018] [Accepted: 03/23/2018] [Indexed: 12/13/2022]
Abstract
Cell surface neural adhesion proteins are critical components in the complex orchestration of cell proliferation, apoptosis, and neuritogenesis essential for proper brain construction and behavior. We focused on the impact of two plasticity-associated IgLON family neural adhesion molecules, Neurotrimin (Ntm) and Limbic system associated membrane protein (Lsamp), on mouse behavior and its underlying neural development. Phenotyping neurons derived from the hippocampi of Lsamp-/-, Ntm-/- and Lsamp-/-Ntm-/- mice was performed in parallel with behavioral testing. While the anatomy of mutant brains revealed no gross changes, the Ntm-/- hippocampal neurons exhibited premature sprouting of neurites and manifested accelerated neurite elongation and branching. We propose that Ntm exerts an inhibitory impact on neurite outgrowth, whereas Lsamp appears to be an enhancer of the said process as premature neuritogenesis in Ntm-/- neurons is apparent only in the presence of Lsamp. We also show interplay between Lsamp and Ntm in regulating tissue homeostasis: the impact of Ntm on cellular proliferation was dependent on Lsamp, and Lsamp appeared to be a positive regulator of apoptosis in the presence of Ntm. Behavioral phenotyping indicated test-specific interactions between Lsamp and Ntm. The phenotypes of single mutant lines, such as reduced swimming speed in Morris water maze and increased activity in the elevated plus maze, were magnified in Lsamp-/-Ntm-/- mice. Altogether, evidence both from behavioral experiments and cultured hippocampal cells show combined and differential interactions between Ntm and Lsamp in the formation of hippocampal circuits and behavioral profiles. We demonstrate that mutual interactions between IgLON molecules regulate the initiation of neurite sprouting at very early ages, and even cell-autonomously, independent of their regulation of cell-cell adhesion.
Collapse
Affiliation(s)
- Katyayani Singh
- Department of Physiology, Institute of Biomedicine and Translational Medicine, University of Tartu, 19 Ravila Street, 50411, Tartu, Estonia; Centre of Excellence in Genomics and Translational Medicine, University of Tartu, 19 Ravila Street, 50411, Tartu, Estonia
| | - Kersti Lilleväli
- Department of Physiology, Institute of Biomedicine and Translational Medicine, University of Tartu, 19 Ravila Street, 50411, Tartu, Estonia; Centre of Excellence in Genomics and Translational Medicine, University of Tartu, 19 Ravila Street, 50411, Tartu, Estonia
| | - Scott F Gilbert
- Department of Biology, Swarthmore College, Swarthmore, PA, USA
| | - Aleksandr Bregin
- Department of Physiology, Institute of Biomedicine and Translational Medicine, University of Tartu, 19 Ravila Street, 50411, Tartu, Estonia; Centre of Excellence in Genomics and Translational Medicine, University of Tartu, 19 Ravila Street, 50411, Tartu, Estonia
| | - Jane Narvik
- Department of Physiology, Institute of Biomedicine and Translational Medicine, University of Tartu, 19 Ravila Street, 50411, Tartu, Estonia; Centre of Excellence in Genomics and Translational Medicine, University of Tartu, 19 Ravila Street, 50411, Tartu, Estonia
| | - Mohan Jayaram
- Department of Physiology, Institute of Biomedicine and Translational Medicine, University of Tartu, 19 Ravila Street, 50411, Tartu, Estonia; Centre of Excellence in Genomics and Translational Medicine, University of Tartu, 19 Ravila Street, 50411, Tartu, Estonia
| | - Märt Rahi
- Institute of Agricultural and Environmental Sciences, Estonian University of Life Sciences, Fr.R. Kreutzwaldi 5, 51014, Tartu, Estonia
| | - Jürgen Innos
- Department of Physiology, Institute of Biomedicine and Translational Medicine, University of Tartu, 19 Ravila Street, 50411, Tartu, Estonia; Centre of Excellence in Genomics and Translational Medicine, University of Tartu, 19 Ravila Street, 50411, Tartu, Estonia
| | - Allen Kaasik
- Department of Pharmacology, Institute of Biomedicine and Translational Medicine, University of Tartu, 19 Ravila Street, 50411, Tartu, Estonia
| | - Eero Vasar
- Department of Physiology, Institute of Biomedicine and Translational Medicine, University of Tartu, 19 Ravila Street, 50411, Tartu, Estonia; Centre of Excellence in Genomics and Translational Medicine, University of Tartu, 19 Ravila Street, 50411, Tartu, Estonia
| | - Mari-Anne Philips
- Department of Physiology, Institute of Biomedicine and Translational Medicine, University of Tartu, 19 Ravila Street, 50411, Tartu, Estonia; Centre of Excellence in Genomics and Translational Medicine, University of Tartu, 19 Ravila Street, 50411, Tartu, Estonia.
| |
Collapse
|
16
|
Singh K, Loreth D, Pöttker B, Hefti K, Innos J, Schwald K, Hengstler H, Menzel L, Sommer CJ, Radyushkin K, Kretz O, Philips MA, Haas CA, Frauenknecht K, Lilleväli K, Heimrich B, Vasar E, Schäfer MKE. Neuronal Growth and Behavioral Alterations in Mice Deficient for the Psychiatric Disease-Associated Negr1 Gene. Front Mol Neurosci 2018; 11:30. [PMID: 29479305 PMCID: PMC5811522 DOI: 10.3389/fnmol.2018.00030] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2017] [Accepted: 01/23/2018] [Indexed: 12/11/2022] Open
Abstract
Neuronal growth regulator 1 (NEGR1), a member of the immunoglobulin superfamily cell adhesion molecule subgroup IgLON, has been implicated in neuronal growth and connectivity. In addition, genetic variants in or near the NEGR1 locus have been associated with obesity and more recently with learning difficulties, intellectual disability and psychiatric disorders. However, experimental evidence is lacking to support a possible link between NEGR1, neuronal growth and behavioral abnormalities. Initial expression analysis of NEGR1 mRNA in C57Bl/6 wildtype (WT) mice by in situ hybridization demonstrated marked expression in the entorhinal cortex (EC) and dentate granule cells. In co-cultures of cortical neurons and NSC-34 cells overexpressing NEGR1, neurite growth of cortical neurons was enhanced and distal axons occupied an increased area of cells overexpressing NEGR1. Conversely, in organotypic slice co-cultures, Negr1-knockout (KO) hippocampus was less permissive for axons grown from EC of β-actin-enhanced green fluorescent protein (EGFP) mice compared to WT hippocampus. Neuroanatomical analysis revealed abnormalities of EC axons in the hippocampal dentate gyrus (DG) of Negr1-KO mice including increased numbers of axonal projections to the hilus. Neurotransmitter receptor ligand binding densities, a proxy of functional neurotransmitter receptor abundance, did not show differences in the DG of Negr1-KO mice but altered ligand binding densities to NMDA receptor and muscarinic acetylcholine receptors M1 and M2 were found in CA1 and CA3. Activity behavior, anxiety-like behavior and sensorimotor gating were not different between genotypes. However, Negr1-KO mice exhibited impaired social behavior compared to WT littermates. Moreover, Negr1-KO mice showed reversal learning deficits in the Morris water maze and increased susceptibility to pentylenetetrazol (PTZ)-induced seizures. Thus, our results from neuronal growth assays, neuroanatomical analyses and behavioral assessments provide first evidence that deficiency of the psychiatric disease-associated Negr1 gene may affect neuronal growth and behavior. These findings might be relevant to further evaluate the role of NEGR1 in cognitive and psychiatric disorders.
Collapse
Affiliation(s)
- Katyayani Singh
- Department of Physiology, Institute of Biomedicine and Translational Medicine, University of Tartu, Tartu, Estonia.,Centre of Excellence in Genomics and Translational Medicine, University of Tartu, Tartu, Estonia
| | - Desirée Loreth
- Department of Neuroanatomy, Institute of Anatomy and Cell Biology, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Bruno Pöttker
- Department of Anesthesiology, University Medical Center, Johannes Gutenberg-University Mainz, Mainz, Germany
| | - Kyra Hefti
- Institute of Neuropathology, University Medical Center, Johannes Gutenberg-University of Mainz, Mainz, Germany
| | - Jürgen Innos
- Department of Physiology, Institute of Biomedicine and Translational Medicine, University of Tartu, Tartu, Estonia.,Centre of Excellence in Genomics and Translational Medicine, University of Tartu, Tartu, Estonia
| | - Kathrin Schwald
- Department of Neuroanatomy, Institute of Anatomy and Cell Biology, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Heidi Hengstler
- Department of Neuroanatomy, Institute of Anatomy and Cell Biology, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Lutz Menzel
- Department of Anesthesiology, University Medical Center, Johannes Gutenberg-University Mainz, Mainz, Germany
| | - Clemens J Sommer
- Institute of Neuropathology, University Medical Center, Johannes Gutenberg-University of Mainz, Mainz, Germany.,Focus Program Translational Neurosciences, Johannes Gutenberg-University of Mainz, Mainz, Germany
| | - Konstantin Radyushkin
- Focus Program Translational Neurosciences, Johannes Gutenberg-University of Mainz, Mainz, Germany.,Mouse Behavioral Unit, Johannes Gutenberg-University of Mainz, Mainz, Germany
| | - Oliver Kretz
- Department of Neuroanatomy, Institute of Anatomy and Cell Biology, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Mari-Anne Philips
- Department of Physiology, Institute of Biomedicine and Translational Medicine, University of Tartu, Tartu, Estonia.,Centre of Excellence in Genomics and Translational Medicine, University of Tartu, Tartu, Estonia
| | - Carola A Haas
- Experimental Epilepsy Research, Department of Neurosurgery, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Katrin Frauenknecht
- Institute of Neuropathology, University Medical Center, Johannes Gutenberg-University of Mainz, Mainz, Germany.,Institute of Neuropathology, University Hospital Zurich, Zurich, Switzerland
| | - Kersti Lilleväli
- Department of Physiology, Institute of Biomedicine and Translational Medicine, University of Tartu, Tartu, Estonia.,Centre of Excellence in Genomics and Translational Medicine, University of Tartu, Tartu, Estonia
| | - Bernd Heimrich
- Department of Neuroanatomy, Institute of Anatomy and Cell Biology, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Eero Vasar
- Department of Physiology, Institute of Biomedicine and Translational Medicine, University of Tartu, Tartu, Estonia.,Centre of Excellence in Genomics and Translational Medicine, University of Tartu, Tartu, Estonia
| | - Michael K E Schäfer
- Department of Anesthesiology, University Medical Center, Johannes Gutenberg-University Mainz, Mainz, Germany.,Focus Program Translational Neurosciences, Johannes Gutenberg-University of Mainz, Mainz, Germany
| |
Collapse
|
17
|
Velásquez E, Nogueira FCS, Velásquez I, Schmitt A, Falkai P, Domont GB, Martins-de-Souza D. Synaptosomal Proteome of the Orbitofrontal Cortex from Schizophrenia Patients Using Quantitative Label-Free and iTRAQ-Based Shotgun Proteomics. J Proteome Res 2017; 16:4481-4494. [PMID: 28949146 DOI: 10.1021/acs.jproteome.7b00422] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Schizophrenia is a chronic and incurable neuropsychiatric disorder that affects about one percent of the world population. The proteomic characterization of the synaptosome fraction of the orbitofrontal cortex is useful for providing valuable information about the molecular mechanisms of synaptic functions in these patients. Quantitative analyses of synaptic proteins were made with eight paranoid schizophrenia patients and a pool of eight healthy controls free of mental diseases. Label-free and iTRAQ labeling identified a total of 2018 protein groups. Statistical analyses revealed 12 and 55 significantly dysregulated proteins by iTRAQ and label-free, respectively. Quantitative proteome analyses showed an imbalance in the calcium signaling pathway and proteins such as reticulon-1 and cytochrome c, related to endoplasmic reticulum stress and programmed cell death. Also, it was found that there is a significant increase in limbic-system-associated membrane protein and α-calcium/calmodulin-dependent protein kinase II, associated with the regulation of human behavior. Our data contribute to a better understanding about apoptosis as a possible pathophysiological mechanism of this disease as well as neural systems supporting social behavior in schizophrenia. This study also is a joint effort of the Chr 15 C-HPP team and the Human Brain Proteome Project of B/D-HPP. All MS proteomics data are deposited in the ProteomeXchange Repository under PXD006798.
Collapse
Affiliation(s)
- Erika Velásquez
- Proteomics Unit, Department of Biochemistry, Institute of Chemistry, Federal University of Rio de Janeiro , Rio de Janeiro, 21941-909 Rio de Janeiro, Brazil
| | - Fabio C S Nogueira
- Proteomics Unit, Department of Biochemistry, Institute of Chemistry, Federal University of Rio de Janeiro , Rio de Janeiro, 21941-909 Rio de Janeiro, Brazil.,Laboratory of Proteomics, LADETEC, Institute of Chemistry, Federal University of Rio de Janeiro , Rio de Janeiro, 21941-598 Rio de Janeiro, Brazil
| | | | - Andrea Schmitt
- Department of Psychiatry and Psychotherapy, Ludwig Maximilian University of Munich (LMU) , 80336 Munich, Germany
| | - Peter Falkai
- Department of Psychiatry and Psychotherapy, Ludwig Maximilian University of Munich (LMU) , 80336 Munich, Germany
| | - Gilberto B Domont
- Proteomics Unit, Department of Biochemistry, Institute of Chemistry, Federal University of Rio de Janeiro , Rio de Janeiro, 21941-909 Rio de Janeiro, Brazil
| | - Daniel Martins-de-Souza
- Laboratory of Neuroproteomics, Department of Biochemistry, Institute of Biology, University of Campinas (UNICAMP) , Campinas, 13083-862 São Paulo, Brazil.,UNICAMP's Neurobiology Center , Campinas, 13083-888 São Paulo, Brazil.,Instituto Nacional de Biomarcadores em Neuropsiquiatria (INBION), Conselho Nacional de Desenvolvimento Cientifico e Tecnologico , São Paulo, 01060-970 São Paulo, Brazil
| |
Collapse
|
18
|
Curran AM, Fogarty Draper C, Scott-Boyer MP, Valsesia A, Roche HM, Ryan MF, Gibney MJ, Kutmon M, Evelo CT, Coort SL, Astrup A, Saris WH, Brennan L, Kaput J. Sexual Dimorphism, Age, and Fat Mass Are Key Phenotypic Drivers of Proteomic Signatures. J Proteome Res 2017; 16:4122-4133. [DOI: 10.1021/acs.jproteome.7b00501] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Affiliation(s)
- Aoife M. Curran
- Institute
of Food and Health, Food for Health Ireland, University College Dublin, Dublin 4, Republic of Ireland
| | - Colleen Fogarty Draper
- Nestlé Institute of Health Sciences, 1015 Lausanne, Switzerland
- Leiden
Academic Centre for Drug Research, Analytical BioSciences, Leiden University, 2311 EZ Leiden, The Netherlands
| | - Marie-Pier Scott-Boyer
- The Microsoft Research − University of Trento Centre for Computational and Systems Biology (COSBI), 38068 Rovereto, Italy
| | - Armand Valsesia
- Nestlé Institute of Health Sciences, 1015 Lausanne, Switzerland
| | - Helen M. Roche
- Institute
of Food and Health, Food for Health Ireland, University College Dublin, Dublin 4, Republic of Ireland
- Nutrigenomics
Research Group, UCD Conway Institute of Biomolecular and Biomedical
Research and UCD Institute of Food and Health, School of Public Health,
Physiotherapy and Sports Science, University College Dublin, Belfield, Dublin 4 Republic of Ireland
| | - Miriam F. Ryan
- Institute
of Food and Health, Food for Health Ireland, University College Dublin, Dublin 4, Republic of Ireland
| | - Michael J. Gibney
- Institute
of Food and Health, Food for Health Ireland, University College Dublin, Dublin 4, Republic of Ireland
| | - Martina Kutmon
- Department
of Bioinformatics − BiGCaT, School of Nutrition and Translational
Research in Metabolism and Maastricht
Centre for Systems Biology (McCSBio), Maastricht University, 6211 LK Maastricht, The Netherlands
| | - Chris T. Evelo
- Department
of Bioinformatics − BiGCaT, School of Nutrition and Translational
Research in Metabolism and Maastricht
Centre for Systems Biology (McCSBio), Maastricht University, 6211 LK Maastricht, The Netherlands
| | - Susan L. Coort
- Department
of Bioinformatics − BiGCaT, School of Nutrition and Translational
Research in Metabolism and Maastricht
Centre for Systems Biology (McCSBio), Maastricht University, 6211 LK Maastricht, The Netherlands
| | - Arne Astrup
- Department
of Nutrition, Exercise and Sports, Faculty of Science, University of Copenhagen, 1165 Copenhagen, Denmark
| | - Wim H. Saris
- Department
of Human Biology, School of Nutrition and Translational Research in
Metabolism, Maastricht University Medical Centre, 6211 LK Maastricht, The Netherlands
| | - Lorraine Brennan
- Institute
of Food and Health, Food for Health Ireland, University College Dublin, Dublin 4, Republic of Ireland
| | - Jim Kaput
- Nestlé Institute of Health Sciences, 1015 Lausanne, Switzerland
| |
Collapse
|
19
|
Ectodomain shedding of Limbic System-Associated Membrane Protein (LSAMP) by ADAM Metallopeptidases promotes neurite outgrowth in DRG neurons. Sci Rep 2017; 7:7961. [PMID: 28801670 PMCID: PMC5554145 DOI: 10.1038/s41598-017-08315-0] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2017] [Accepted: 07/07/2017] [Indexed: 11/25/2022] Open
Abstract
IgLONs are members of the immunoglobulin superfamily of cell adhesion proteins implicated in the process of neuronal outgrowth, cell adhesion and subdomain target recognition. IgLONs form homophilic and heterophilic complexes on the cell surface that repress or promote growth depending on the neuronal population, the developmental stage and surface repertoire of IgLON family members. In the present study, we identified a metalloproteinase-dependent mechanism necessary to promote growth in embryonic dorsal root ganglion cells (DRGs). Treatment of embryonic DRG neurons with pan-metalloproteinase inhibitors, tissue inhibitor of metalloproteinase-3, or an inhibitor of ADAM Metallopeptidase Domain 10 (ADAM10) reduces outgrowth from DRG neurons indicating that metalloproteinase activity is important for outgrowth. The IgLON family members Neurotrimin (NTM) and Limbic System-Associated Membrane Protein (LSAMP) were identified as ADAM10 substrates that are shed from the cell surface of DRG neurons. Overexpression of LSAMP and NTM suppresses outgrowth from DRG neurons. Furthermore, LSAMP loss of function decreases the outgrowth sensitivity to an ADAM10 inhibitor. Together our findings support a role for ADAM-dependent shedding of cell surface LSAMP in promoting outgrowth from DRG neurons.
Collapse
|
20
|
Son AI, Fu X, Suto F, Liu JS, Hashimoto-Torii K, Torii M. Proteome dynamics during postnatal mouse corpus callosum development. Sci Rep 2017; 7:45359. [PMID: 28349996 PMCID: PMC5368975 DOI: 10.1038/srep45359] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2016] [Accepted: 02/27/2017] [Indexed: 02/08/2023] Open
Abstract
Formation of cortical connections requires the precise coordination of numerous discrete phases. This is particularly significant with regard to the corpus callosum, whose development undergoes several dynamic stages including the crossing of axon projections, elimination of exuberant projections, and myelination of established tracts. To comprehensively characterize the molecular events in this dynamic process, we set to determine the distinct temporal expression of proteins regulating the formation of the corpus callosum and their respective developmental functions. Mass spectrometry-based proteomic profiling was performed on early postnatal mouse corpus callosi, for which limited evidence has been obtained previously, using stable isotope of labeled amino acids in mammals (SILAM). The analyzed corpus callosi had distinct proteomic profiles depending on age, indicating rapid progression of specific molecular events during this period. The proteomic profiles were then segregated into five separate clusters, each with distinct trajectories relevant to their intended developmental functions. Our analysis both confirms many previously-identified proteins in aspects of corpus callosum development, and identifies new candidates in understudied areas of development including callosal axon refinement. We present a valuable resource for identifying new proteins integral to corpus callosum development that will provide new insights into the development and diseases afflicting this structure.
Collapse
Affiliation(s)
- Alexander I Son
- Center for Neuroscience Research, Children's Research Institute, Children's National Medical Center, Washington, DC 20010, USA
| | - Xiaoqin Fu
- Center for Neuroscience Research, Children's Research Institute, Children's National Medical Center, Washington, DC 20010, USA
| | - Fumikazu Suto
- Department of Ultrastructural Research, National Institute of Neuroscience, National Center of Neurology and Psychiatry, Tokyo 187-8502, Japan
| | - Judy S Liu
- Center for Neuroscience Research, Children's Research Institute, Children's National Medical Center, Washington, DC 20010, USA.,Department of Pediatrics, Pharmacology and Physiology, School of Medicine and Health Sciences, The George Washington University, Washington, DC 20052, USA
| | - Kazue Hashimoto-Torii
- Center for Neuroscience Research, Children's Research Institute, Children's National Medical Center, Washington, DC 20010, USA.,Department of Pediatrics, Pharmacology and Physiology, School of Medicine and Health Sciences, The George Washington University, Washington, DC 20052, USA.,Department of Neurobiology and Kavli Institute for Neuroscience, School of Medicine, Yale University, New Haven, CT 06510, USA
| | - Masaaki Torii
- Center for Neuroscience Research, Children's Research Institute, Children's National Medical Center, Washington, DC 20010, USA.,Department of Pediatrics, Pharmacology and Physiology, School of Medicine and Health Sciences, The George Washington University, Washington, DC 20052, USA.,Department of Neurobiology and Kavli Institute for Neuroscience, School of Medicine, Yale University, New Haven, CT 06510, USA
| |
Collapse
|
21
|
Tien WS, Chen JH, Wu KP. SheddomeDB: the ectodomain shedding database for membrane-bound shed markers. BMC Bioinformatics 2017; 18:42. [PMID: 28361715 PMCID: PMC5374707 DOI: 10.1186/s12859-017-1465-7] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND A number of membrane-anchored proteins are known to be released from cell surface via ectodomain shedding. The cleavage and release of membrane proteins has been shown to modulate various cellular processes and disease pathologies. Numerous studies revealed that cell membrane molecules of diverse functional groups are subjected to proteolytic cleavage, and the released soluble form of proteins may modulate various signaling processes. Therefore, in addition to the secreted protein markers that undergo secretion through the secretory pathway, the shed membrane proteins may comprise an additional resource of noninvasive and accessible biomarkers. In this context, identifying the membrane-bound proteins that will be shed has become important in the discovery of clinically noninvasive biomarkers. Nevertheless, a data repository for biological and clinical researchers to review the shedding information, which is experimentally validated, for membrane-bound protein shed markers is still lacking. RESULTS In this study, the database SheddomeDB was developed to integrate publicly available data of the shed membrane proteins. A comprehensive literature survey was performed to collect the membrane proteins that were verified to be cleaved or released in the supernatant by immunological-based validation experiments. From 436 studies on shedding, 401 validated shed membrane proteins were included, among which 199 shed membrane proteins have not been annotated or validated yet by existing cleavage databases. SheddomeDB attempted to provide a comprehensive shedding report, including the regulation of shedding machinery and the related function or diseases involved in the shedding events. In addition, our published tool ShedP was embedded into SheddomeDB to support researchers for predicting the shedding event on unknown or unrecorded membrane proteins. CONCLUSIONS To the best of our knowledge, SheddomeDB is the first database for the identification of experimentally validated shed membrane proteins and currently may provide the most number of membrane proteins for reviewing the shedding information. The database included membrane-bound shed markers associated with numerous cellular processes and diseases, and some of these markers are potential novel markers because they are not annotated or validated yet in other databases. SheddomeDB may provide a useful resource for discovering membrane-bound shed markers. The interactive web of SheddomeDB is publicly available at http://bal.ym.edu.tw/SheddomeDB/ .
Collapse
Affiliation(s)
- Wei-Sheng Tien
- Institute of Biomedical Informatics, National Yang Ming University, Taipei, 112, Taiwan.,Bioinformatics Program, Taiwan International Graduate Program, Academia Sinica, Taipei, 115, Taiwan
| | - Jun-Hong Chen
- Department of Computer Science, National Taipei University of Education, Taipei, 106, Taiwan
| | - Kun-Pin Wu
- Institute of Biomedical Informatics, National Yang Ming University, Taipei, 112, Taiwan.
| |
Collapse
|
22
|
Vanaveski T, Singh K, Narvik J, Eskla KL, Visnapuu T, Heinla I, Jayaram M, Innos J, Lilleväli K, Philips MA, Vasar E. Promoter-Specific Expression and Genomic Structure of IgLON Family Genes in Mouse. Front Neurosci 2017; 11:38. [PMID: 28210208 PMCID: PMC5288359 DOI: 10.3389/fnins.2017.00038] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2016] [Accepted: 01/19/2017] [Indexed: 01/20/2023] Open
Abstract
IgLON family is composed of five genes: Lsamp, Ntm, Opcml, Negr1, and Iglon5; encoding for five highly homologous neural adhesion proteins that regulate neurite outgrowth and synapse formation. In the current study we performed in silico analysis revealing that Ntm and Opcml display similar genomic structure as previously reported for Lsamp, characterized by two alternative promotors 1a and 1b. Negr1 and Iglon5 transcripts have uniform 5′ region, suggesting single promoter. Iglon5, the recently characterized family member, shares high level of conservation and structural qualities characteristic to IgLON family such as N-terminal signal peptide, three Ig domains, and GPI anchor binding site. By using custom 5′-isoform-specific TaqMan gene-expression assay, we demonstrated heterogeneous expression of IgLON transcripts in different areas of mouse brain and several-fold lower expression in selected tissues outside central nervous system. As an example, the expression of IgLON transcripts in urogenital and reproductive system is in line with repeated reports of urogenital tumors accompanied by mutations in IgLON genes. Considering the high levels of intra-family homology shared by IgLONs, we investigated potential compensatory effects at the level of IgLON isoforms in the brains of mice deficient of one or two family members. We found that the lack of IgLONs is not compensated by a systematic quantitative increase of the other family members. On the contrary, the expression of Ntm 1a transcript and NEGR1 protein was significantly reduced in the frontal cortex of Lsamp-deficient mice suggesting that the expression patterns within IgLON family are balanced coherently. The actions of individual IgLONs, however, can be antagonistic as demonstrated by differential expression of Syp in deletion mutants of IgLONs. In conclusion, we show that the genomic twin-promoter structure has impact on both anatomical distribution and intra-family interactions of IgLON family members. Remarkable variety in the activity levels of 1a and 1b promoters both in the brain and in other tissues, suggests complex functional regulation of IgLONs by alternative signal peptides driven by 1a and 1b promoters.
Collapse
Affiliation(s)
- Taavi Vanaveski
- Department of Physiology, Institute of Biomedicine and Translational Medicine, University of Tartu Tartu, Estonia
| | - Katyayani Singh
- Department of Physiology, Institute of Biomedicine and Translational Medicine, University of Tartu Tartu, Estonia
| | - Jane Narvik
- Department of Physiology, Institute of Biomedicine and Translational Medicine, University of Tartu Tartu, Estonia
| | - Kattri-Liis Eskla
- Department of Physiology, Institute of Biomedicine and Translational Medicine, University of Tartu Tartu, Estonia
| | - Tanel Visnapuu
- Department of Physiology, Institute of Biomedicine and Translational Medicine, University of TartuTartu, Estonia; Division of Pharmacology and Pharmacotherapy, Faculty of Pharmacy, University of HelsinkiHelsinki, Finland
| | - Indrek Heinla
- Department of Physiology, Institute of Biomedicine and Translational Medicine, University of Tartu Tartu, Estonia
| | - Mohan Jayaram
- Department of Physiology, Institute of Biomedicine and Translational Medicine, University of Tartu Tartu, Estonia
| | - Jürgen Innos
- Department of Physiology, Institute of Biomedicine and Translational Medicine, University of Tartu Tartu, Estonia
| | - Kersti Lilleväli
- Department of Physiology, Institute of Biomedicine and Translational Medicine, University of Tartu Tartu, Estonia
| | - Mari-Anne Philips
- Department of Physiology, Institute of Biomedicine and Translational Medicine, University of Tartu Tartu, Estonia
| | - Eero Vasar
- Department of Physiology, Institute of Biomedicine and Translational Medicine, University of Tartu Tartu, Estonia
| |
Collapse
|
23
|
Cell type- and brain region-resolved mouse brain proteome. Nat Neurosci 2015; 18:1819-31. [PMID: 26523646 DOI: 10.1038/nn.4160] [Citation(s) in RCA: 579] [Impact Index Per Article: 57.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2015] [Accepted: 10/06/2015] [Indexed: 12/13/2022]
Abstract
Brain transcriptome and connectome maps are being generated, but an equivalent effort on the proteome is currently lacking. We performed high-resolution mass spectrometry-based proteomics for in-depth analysis of the mouse brain and its major brain regions and cell types. Comparisons of the 12,934 identified proteins in oligodendrocytes, astrocytes, microglia and cortical neurons with deep sequencing data of the transcriptome indicated deep coverage of the proteome. Cell type-specific proteins defined as tenfold more abundant than average expression represented about a tenth of the proteome, with an overrepresentation of cell surface proteins. To demonstrate the utility of our resource, we focused on this class of proteins and identified Lsamp, an adhesion molecule of the IgLON family, as a negative regulator of myelination. Our findings provide a framework for a system-level understanding of cell-type diversity in the CNS and serves as a rich resource for analyses of brain development and function.
Collapse
|
24
|
Barøy T, Kresse SH, Skårn M, Stabell M, Castro R, Lauvrak S, Llombart-Bosch A, Myklebost O, Meza-Zepeda LA. Reexpression of LSAMP inhibits tumor growth in a preclinical osteosarcoma model. Mol Cancer 2014; 13:93. [PMID: 24885297 PMCID: PMC4029956 DOI: 10.1186/1476-4598-13-93] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2013] [Accepted: 03/11/2014] [Indexed: 11/16/2022] Open
Abstract
BACKGROUND Osteosarcomas are the most common primary malignant tumors of bone, showing complex chromosomal rearrangements with multiple gains and losses. A frequent deletion within the chromosomal region 3q13.31 has been identified by us and others, and is mainly reported to be present in osteosarcomas. The purpose of the study was to further characterize the frequency and the extent of the deletion in an extended panel of osteosarcoma samples, and the expression level of the affected genes within the region. We have identified LSAMP as the target gene for the deletion, and have studied the functional implications of LSAMP-reexpression. METHODS LSAMP copy number, expression level and protein level were investigated by quantitative PCR and western blotting in an osteosarcoma panel. The expression of LSAMP was restored in an osteosarcoma cell line, and differences in proliferation rate, tumor formation, gene expression, migration rate, differentiation capabilities, cell cycle distribution and apoptosis were investigated by metabolic dyes, tumor formation in vivo, gene expression profiling, time-lapse photography, differentiation techniques and flow cytometry, respectively. RESULTS We found reduced copy number of LSAMP in 45/76 osteosarcoma samples, reduced expression level in 25/42 samples and protein expression in 9/42 samples. By restoring the expression of LSAMP in a cell line with a homozygous deletion of the gene, the proliferation rate in vitro was significantly reduced and tumor growth in vivo was significantly delayed. In response to reexpression of LSAMP, mRNA expression profiling revealed consistent upregulation of the genes hairy and enhancer of split 1 (HES1), cancer/testis antigen 2 (CTAG2) and kruppel-like factor 10 (KLF10). CONCLUSIONS The high frequency and the specificity of the deletion indicate that it is important for the development of osteosarcomas. The deletion targets the tumor suppressor LSAMP, and based on the functional evidence, the tumor suppressor function of LSAMP is most likely exerted by reducing the proliferation rate of the tumor cells, possibly by indirectly upregulating one or more of the genes HES1, CTAG2 or KLF10. To our knowledge, this study describes novel functions of LSAMP, a first step to understanding the functional role of this specific deletion in osteosarcomas.
Collapse
MESH Headings
- Antigens, Neoplasm/genetics
- Antigens, Neoplasm/metabolism
- Antigens, Surface/genetics
- Antigens, Surface/metabolism
- Basic Helix-Loop-Helix Transcription Factors/genetics
- Basic Helix-Loop-Helix Transcription Factors/metabolism
- Bone Neoplasms/genetics
- Bone Neoplasms/metabolism
- Bone Neoplasms/mortality
- Bone Neoplasms/pathology
- Cell Adhesion Molecules, Neuronal/genetics
- Cell Adhesion Molecules, Neuronal/metabolism
- Cell Line, Tumor
- Cell Proliferation/genetics
- Chromosome Mapping
- Chromosomes, Human, Pair 3
- Early Growth Response Transcription Factors/genetics
- Early Growth Response Transcription Factors/metabolism
- Female
- GPI-Linked Proteins/genetics
- GPI-Linked Proteins/metabolism
- Gene Deletion
- Gene Dosage
- Gene Expression Regulation, Neoplastic
- Genetic Complementation Test
- Homeodomain Proteins/genetics
- Homeodomain Proteins/metabolism
- Homozygote
- Humans
- Kruppel-Like Transcription Factors/genetics
- Kruppel-Like Transcription Factors/metabolism
- Male
- Mutation Rate
- Osteosarcoma/genetics
- Osteosarcoma/metabolism
- Osteosarcoma/mortality
- Osteosarcoma/pathology
- RNA, Messenger/genetics
- RNA, Messenger/metabolism
- Signal Transduction
- Survival Analysis
- Transcription Factor HES-1
Collapse
Affiliation(s)
- Tale Barøy
- Department of Tumor Biology, Institute for Cancer Research, Oslo University Hospital, The Norwegian Radium Hospital, Oslo, Norway
| | - Stine H Kresse
- Department of Tumor Biology, Institute for Cancer Research, Oslo University Hospital, The Norwegian Radium Hospital, Oslo, Norway
| | - Magne Skårn
- Department of Tumor Biology, Institute for Cancer Research, Oslo University Hospital, The Norwegian Radium Hospital, Oslo, Norway
| | - Marianne Stabell
- Department of Tumor Biology, Institute for Cancer Research, Oslo University Hospital, The Norwegian Radium Hospital, Oslo, Norway
| | - Russell Castro
- Department of Tumor Biology, Institute for Cancer Research, Oslo University Hospital, The Norwegian Radium Hospital, Oslo, Norway
| | - Silje Lauvrak
- Department of Tumor Biology, Institute for Cancer Research, Oslo University Hospital, The Norwegian Radium Hospital, Oslo, Norway
| | | | - Ola Myklebost
- Department of Tumor Biology, Institute for Cancer Research, Oslo University Hospital, The Norwegian Radium Hospital, Oslo, Norway
- Department of Molecular Biosciences, University of Oslo, Oslo, Norway
| | - Leonardo A Meza-Zepeda
- Department of Tumor Biology, Institute for Cancer Research, Oslo University Hospital, The Norwegian Radium Hospital, Oslo, Norway
- Genomics Core Facility, Oslo University Hospital, Oslo, Norway
| |
Collapse
|
25
|
Hiemer SE, Szymaniak AD, Varelas X. The transcriptional regulators TAZ and YAP direct transforming growth factor β-induced tumorigenic phenotypes in breast cancer cells. J Biol Chem 2014; 289:13461-74. [PMID: 24648515 DOI: 10.1074/jbc.m113.529115] [Citation(s) in RCA: 192] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Uncontrolled transforming growth factor-β (TGFβ) signaling promotes aggressive metastatic properties in late-stage breast cancers. However, how TGFβ-mediated cues are directed to induce tumorigenic events is poorly understood, particularly given that TGFβ has clear tumor suppressing activity in other contexts. Here, we demonstrate that the transcriptional regulators TAZ and YAP (TAZ/YAP), key effectors of the Hippo pathway, are necessary to promote and maintain TGFβ-induced tumorigenic phenotypes in breast cancer cells. Interactions between TAZ/YAP, TGFβ-activated SMAD2/3, and TEAD transcription factors reveal convergent roles for these factors in the nucleus. Genome-wide expression analyses indicate that TAZ/YAP, TEADs, and TGFβ-induced signals coordinate a specific pro-tumorigenic transcriptional program. Importantly, genes cooperatively regulated by TAZ/YAP, TEAD, and TGFβ, such as the novel targets NEGR1 and UCA1, are necessary for maintaining tumorigenic activity in metastatic breast cancer cells. Nuclear TAZ/YAP also cooperate with TGFβ signaling to promote phenotypic and transcriptional changes in nontumorigenic cells to overcome TGFβ-repressive effects. Our work thus identifies cross-talk between nuclear TAZ/YAP and TGFβ signaling in breast cancer cells, revealing novel insight into late-stage disease-driving mechanisms.
Collapse
Affiliation(s)
- Samantha E Hiemer
- From the Department of Biochemistry, Boston University School of Medicine, Boston, Massachusetts 02118
| | | | | |
Collapse
|
26
|
Innos J, Koido K, Philips MA, Vasar E. Limbic system associated membrane protein as a potential target for neuropsychiatric disorders. Front Pharmacol 2013; 4:32. [PMID: 23532449 PMCID: PMC3607788 DOI: 10.3389/fphar.2013.00032] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2012] [Accepted: 03/08/2013] [Indexed: 12/16/2022] Open
Abstract
The studies performed in laboratory animals and psychiatric patients suggest a possible role of limbic system-associated membrane protein (LAMP) in the mechanisms of psychiatric disorders. Stressful manipulations and genetic invalidation have revealed a role of the Lsamp gene in the regulation of anxiety in rodents. Besides that, Lsamp-deficient mice display reduced aggressiveness and impaired adaptation in novel and stressful environments. The behavioral effects of amphetamine were blunted in genetically modified mice. Recent pharmacological and biochemical studies point toward altered function of GABA-, 5-hydroxytryptamine-, and dopaminergic systems in Lsamp-deficient mice. Moreover, we found an association between the gene polymorphisms of LSAMP and major depressive disorder (MDD). Patients suffering from MDD had significantly increased ratio between risk and protective haplotypes of the LSAMP gene compared to healthy volunteers. However, the impact of these haplotypes for the function of LAMP is not clear and remains to be elucidated in future studies.
Collapse
Affiliation(s)
- Jürgen Innos
- Department of Physiology, University of Tartu Tartu, Estonia
| | | | | | | |
Collapse
|
27
|
Koido K, Traks T, Balõtšev R, Eller T, Must A, Koks S, Maron E, Tõru I, Shlik J, Vasar V, Vasar E. Associations between LSAMP gene polymorphisms and major depressive disorder and panic disorder. Transl Psychiatry 2012; 2:e152. [PMID: 22892717 PMCID: PMC3432189 DOI: 10.1038/tp.2012.74] [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] [Indexed: 01/19/2023] Open
Abstract
The purpose of this case-control genetic association study was to explore potential relationships between polymorphisms in the limbic system-associated membrane protein (LSAMP) gene and mood and anxiety disorders. A total of 21 single-nucleotide polymorphisms (SNPs) from the LSAMP gene were analyzed in 591 unrelated patients with the diagnoses of major depressive disorder (MDD) or panic disorder (PD) and in 384 healthy control subjects. The results showed a strong association between LSAMP SNPs and MDD, and a suggestive association between LSAMP SNPs and PD. This is the first evidence of a possible role of LSAMP gene in mood and anxiety disorders in humans.
Collapse
Affiliation(s)
- K Koido
- Department of Physiology, University of Tartu, Tartu, Estonia.
| | - T Traks
- Department of Physiology, University of Tartu, Tartu, Estonia,Centre of Excellence for Translational Medicine, University of Tartu, Tartu, Estonia
| | - R Balõtšev
- Department of Psychiatry, University of Tartu, Tartu, Estonia
| | - T Eller
- Department of Psychiatry, University of Tartu, Tartu, Estonia
| | - A Must
- Department of Physiology, University of Tartu, Tartu, Estonia,Centre of Excellence for Translational Medicine, University of Tartu, Tartu, Estonia
| | - S Koks
- Department of Physiology, University of Tartu, Tartu, Estonia,Centre of Excellence for Translational Medicine, University of Tartu, Tartu, Estonia
| | - E Maron
- Department of Psychiatry, University of Tartu, Tartu, Estonia,Department of Neuropsychopharmacology and Molecular Imaging, Imperial College London, London, UK
| | - I Tõru
- Department of Psychiatry, University of Tartu, Tartu, Estonia
| | - J Shlik
- Department of Psychiatry, University of Ottawa, Ottawa, ON, Canada
| | - V Vasar
- Department of Psychiatry, University of Tartu, Tartu, Estonia
| | - E Vasar
- Department of Physiology, University of Tartu, Tartu, Estonia,Centre of Excellence for Translational Medicine, University of Tartu, Tartu, Estonia
| |
Collapse
|
28
|
Bajor M, Michaluk P, Gulyassy P, Kekesi AK, Juhasz G, Kaczmarek L. Synaptic cell adhesion molecule-2 and collapsin response mediator protein-2 are novel members of the matrix metalloproteinase-9 degradome. J Neurochem 2012; 122:775-88. [DOI: 10.1111/j.1471-4159.2012.07829.x] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
|
29
|
Innos J, Philips MA, Raud S, Lilleväli K, Kõks S, Vasar E. Deletion of the Lsamp gene lowers sensitivity to stressful environmental manipulations in mice. Behav Brain Res 2011; 228:74-81. [PMID: 22155487 DOI: 10.1016/j.bbr.2011.11.033] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2011] [Revised: 11/20/2011] [Accepted: 11/24/2011] [Indexed: 11/29/2022]
Abstract
The Lsamp gene gives rise to limbic system-associated membrane protein (LAMP), which is expressed on the surface of somata and proximal dendrites of neurons. Lsamp-deficient mice have been shown to be slightly hyperactive in novel environments and less anxious, and they display alterations in swimming speed, fear reaction, fear conditioning and social behaviour. In human studies, links between the LSAMP gene and several psychiatric disorders have been found and LSAMP has been established as a tumour suppressor gene. To study the impact of environmental manipulations on the phenotype, we exposed male Lsamp-deficient mice to environmental enrichment (EE), a technique that has often been shown to abolish phenotypic deviations in knockout mice, and to social isolation, a stressful manipulation, after which all the mice were tested in a behavioural battery. EE abolished differences between the genotypes in body weight and anogenital sniffing, a behaviour related to aggressiveness, and amplified the anxiolytic-like phenotype of Lsamp-deficient mice both in the plus maze and motility box. Isolation abolished differences between the genotypes in body weight and anxiety and amplified the differences in swimming speed and anogenital sniffing. EE and isolation failed to modify the results as compared to standard housing in whisker trimming, locomotor activity, marble burying and corticosterone levels. In conclusion, Lsamp-deficient mice were less sensitive to isolation stress than their wild-type littermates. Lack of LAMP protein seemingly leads to a deterioration in the ability to adapt to novel stressful environments and stimuli.
Collapse
Affiliation(s)
- Jürgen Innos
- Department of Physiology, University of Tartu, 19 Ravila Street, 50411 Tartu, Estonia.
| | | | | | | | | | | |
Collapse
|
30
|
Innos J, Philips MA, Leidmaa E, Heinla I, Raud S, Reemann P, Plaas M, Nurk K, Kurrikoff K, Matto V, Visnapuu T, Mardi P, Kõks S, Vasar E. Lower anxiety and a decrease in agonistic behaviour in Lsamp-deficient mice. Behav Brain Res 2010; 217:21-31. [PMID: 20888367 DOI: 10.1016/j.bbr.2010.09.019] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2010] [Revised: 09/16/2010] [Accepted: 09/21/2010] [Indexed: 11/27/2022]
Abstract
In rodents, the Lsamp gene has been implicated in trait anxiety, fear reaction and fear conditioning. Human data link the LSAMP gene to several psychiatric disorders. In this study, we presented a general phenotypic characterization of Lsamp gene-deficient mouse line, created by deleting exon 1b. These mice displayed no gross sensory-motor deficiencies, no overt abnormalities and performed normally in memory and learning tests. However, they responded with increased activity to new environments. Moreover, they displayed reduced anxiety and notable deviations in social behaviour, such as lack of whisker trimming, reduced aggressiveness and reduced dominance. One possible explanation for the anxiolytic-like effect of the deletion of the Lsamp gene is a shift in balance in the Gabra1 and Gabra2 genes in the temporal lobe in favor of the Gabra2 transcript, encoding α2 subunit of GABA(A) receptors that mediate the stimulating effect of GABA agonists. The overall phenotype of Lsamp-deficient mice, characterized by decreased anxiety and several alterations in social behaviour, makes them a good model for studying the molecular mechanisms behind inadequate social behaviours observed in several psychiatric disorders.
Collapse
Affiliation(s)
- Jürgen Innos
- Department of Physiology, University of Tartu, Tartu, Estonia.
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Qiu S, Champagne DL, Peters M, Catania EH, Weeber EJ, Levitt P, Pimenta AF. Loss of limbic system-associated membrane protein leads to reduced hippocampal mineralocorticoid receptor expression, impaired synaptic plasticity, and spatial memory deficit. Biol Psychiatry 2010; 68:197-204. [PMID: 20385375 PMCID: PMC2900390 DOI: 10.1016/j.biopsych.2010.02.013] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/09/2009] [Revised: 02/10/2010] [Accepted: 02/12/2010] [Indexed: 11/25/2022]
Abstract
BACKGROUND The limbic system-associated membrane protein (LAMP) promotes development of neurons of limbic origin. We have previously shown that genetic deletion of LAMP results in heightened reactivity to novelty and reduced anxiety-like behaviors in mice. Here, we demonstrate a critical role of LAMP in hippocampal-dependent synaptic physiology and behavior. METHODS We tested spatial memory performance, hippocampal synaptic plasticity, and stress-related modalities in Lsamp(-/-) mice and their littermate control mice. RESULTS Lsamp(-/-) mice exhibit a pronounced deficit in spatial memory acquisition and poorly sustained CA1 long-term potentiation. We found reduced expression of mineralocorticoid receptor (MR) transcripts in the hippocampus and reduction in the corticosterone-induced, MR-mediated nongenomic modulatory effects on CA1 synaptic transmission. Importantly, the impaired long-term potentiation in Lsamp(-/-) mice can be rescued by stress-like levels of corticosterone in a MR-dependent manner. CONCLUSIONS Our study reveals a novel functional relationship between a cell adhesion molecule enriched in developing limbic circuits, glucocorticoid receptors, and cognitive functioning.
Collapse
Affiliation(s)
- Shenfeng Qiu
- Department of Pharmacology, Vanderbilt University, Nashville, TN 37232, USA, Vanderbilt Kennedy Center for Research on Human Development, Vanderbilt University, Nashville, TN 37232, USA
| | | | - Melinda Peters
- Molecular Pharmacology and Physiology, University of South Florida, Tampa, FL
| | - Elizabeth H. Catania
- Vanderbilt Kennedy Center for Research on Human Development, Vanderbilt University, Nashville, TN 37232, USA
| | - Edwin J. Weeber
- Molecular Pharmacology and Physiology, University of South Florida, Tampa, FL
| | - Pat Levitt
- Department of Pharmacology, Vanderbilt University, Nashville, TN 37232, USA, Vanderbilt Kennedy Center for Research on Human Development, Vanderbilt University, Nashville, TN 37232, USA
| | - Aurea F. Pimenta
- Department of Pharmacology, Vanderbilt University, Nashville, TN 37232, USA, Vanderbilt Kennedy Center for Research on Human Development, Vanderbilt University, Nashville, TN 37232, USA
| |
Collapse
|
32
|
Sugimoto C, Maekawa S, Miyata S. OBCAM, an immunoglobulin superfamily cell adhesion molecule, regulates morphology and proliferation of cerebral astrocytes. J Neurochem 2010; 112:818-28. [DOI: 10.1111/j.1471-4159.2009.06513.x] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
|
33
|
Expression profile of rat hippocampal neurons treated with the neuroprotective compound 2,4-dinitrophenol: up-regulation of cAMP signaling genes. Neurotox Res 2009; 18:112-23. [PMID: 19949915 DOI: 10.1007/s12640-009-9133-y] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2009] [Revised: 10/22/2009] [Accepted: 11/03/2009] [Indexed: 12/29/2022]
Abstract
2,4-Dinitrophenol (DNP) is classically known as a mitochondrial uncoupler and, at high concentrations, is toxic to a variety of cells. However, it has recently been shown that, at subtoxic concentrations, DNP protects neurons against a variety of insults and promotes neuronal differentiation and neuritogenesis. The molecular and cellular mechanisms underlying the beneficial neuroactive properties of DNP are still largely unknown. We have now used DNA microarray analysis to investigate changes in gene expression in rat hippocampal neurons in culture treated with low micromolar concentrations of DNP. Under conditions that did not affect neuronal viability, high-energy phosphate levels or mitochondrial oxygen consumption, DNP induced up-regulation of 275 genes and down-regulation of 231 genes. Significantly, several up-regulated genes were linked to intracellular cAMP signaling, known to be involved in neurite outgrowth, synaptic plasticity, and neuronal survival. Differential expression of specific genes was validated by quantitative RT-PCR using independent samples. Results shed light on molecular mechanisms underlying neuroprotection by DNP and point to possible targets for development of novel therapeutics for neurodegenerative disorders.
Collapse
|
34
|
Kresse SH, Ohnstad HO, Paulsen EB, Bjerkehagen B, Szuhai K, Serra M, Schaefer KL, Myklebost O, Meza-Zepeda LA. LSAMP, a novel candidate tumor suppressor gene in human osteosarcomas, identified by array comparative genomic hybridization. Genes Chromosomes Cancer 2009; 48:679-93. [PMID: 19441093 DOI: 10.1002/gcc.20675] [Citation(s) in RCA: 71] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022] Open
Abstract
Osteosarcomas are the most common primary malignant tumor of bone, and almost all conventional osteosarcomas are high-grade tumors with complex karyotypes. We have examined DNA copy number changes in 36 osteosarcoma tumors and 20 cell lines using microarray-based comparative genomic hybridization. The most frequent minimal recurrent regions of gain identified in the tumor samples were in 1q21.2-q21.3 (78% of the samples), 1q21.3-q22 (78%), and 8q22.1 (72%). Minimal recurrent regions in 10q22.1-q22.2 (81%), 6q16.1 (67%), 13q14.2 (67%), and 13q21.1 (67%) were most frequently lost. A small region in 3q13.31 (2.1 Mb) containing the gene limbic system-associated membrane protein (LSAMP) was frequently deleted (56%). LSAMP has previously been reported to be a candidate tumor suppressor gene in other cancer types. The deletion was validated using fluorescence in situ hybridization, and the expression level and promoter methylation status of LSAMP were investigated using quantitative real-time reverse transcription PCR and methylation-specific PCR, respectively. LSAMP showed low expression compared to two normal bone samples in 6/15 tumors and 5/9 cell lines with deletion of 3q13.31, and also in 5/14 tumors and 3/11 cell lines with normal copy number or gain. Partial or full methylation of the investigated CpG island was identified in 3/30 tumors and 7/20 cell lines. Statistical analyses revealed that loss of 11p15.4-p15.3 and low expression of LSAMP (both P = 0.011) were significantly associated with poor survival. Our results show that LSAMP is a novel candidate tumor suppressor gene in osteosarcomas.
Collapse
Affiliation(s)
- Stine H Kresse
- Department of Tumor Biology, The Norwegian Radium Hospital, Rikshospitalet University Hospital, Oslo, Norway
| | | | | | | | | | | | | | | | | |
Collapse
|
35
|
Farina F, Botto L, Chinello C, Cunati D, Magni F, Masserini M, Palestini P. Characterization of prion protein-enriched domains, isolated from rat cerebellar granule cells in culture. J Neurochem 2009; 110:1038-48. [DOI: 10.1111/j.1471-4159.2009.06198.x] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
|
36
|
Lamprecht R, Dracheva S, Assoun S, LeDoux JE. Fear conditioning induces distinct patterns of gene expression in lateral amygdala. GENES BRAIN AND BEHAVIOR 2009; 8:735-43. [PMID: 19689454 DOI: 10.1111/j.1601-183x.2009.00515.x] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The lateral nucleus of the amygdala (LA) has been implicated in the formation of long-term associative memory (LTM) of stimuli associated with danger through fear conditioning. The current study aims to detect genes that are expressed in LA following associative fear conditioning. Using oligonucleotide microarrays, we monitored gene expression in rats subjected to paired training where a tone co-terminates with a footshock, or unpaired training where the tone and footshock are presented in a non-overlapping manner. The paired protocol consistently leads to auditory fear conditioning memory formation, whereas the unpaired protocol does not. When the paired group was compared with the unpaired group 5 h after training, the expression of genes coding for the limbic system-associated membrane protein (Lsamp), kinesin heavy chain member 2 (Kif2), N-ethylmaleimide-sensitive fusion protein (NSF) and Hippocalcin-like 4 protein (Hpcal4) was higher in the paired group. These genes encode proteins that regulate neuronal axonal morphology (Lsamp, Kif2), presynaptic vesicle cycling and release (Hpcal4 and NSF), and AMPA receptor maintenance in synapses (NSF). Quantitative real-time PCR (qPCR) showed that Kif2 and Lsamp are expressed hours following fear conditioning but minutes after unpaired training. Hpcal4 is induced by paired stimulation only 5 h after the training. These results show that fear conditioning induces a unique temporal activation of molecular pathways involved in regulating synaptic transmission and axonal morphology in LA, which is different from non-associative stimulation.
Collapse
Affiliation(s)
- R Lamprecht
- Department of Neurobiology and Ethology, University of Haifa, Haifa, Israel.
| | | | | | | |
Collapse
|
37
|
Takahata T, Komatsu Y, Watakabe A, Hashikawa T, Tochitani S, Yamamori T. Differential expression patterns of occ1-related genes in adult monkey visual cortex. ACTA ACUST UNITED AC 2008; 19:1937-51. [PMID: 19073625 PMCID: PMC2705702 DOI: 10.1093/cercor/bhn220] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
We have previously revealed that occ1 is preferentially expressed in the primary visual area (V1) of the monkey neocortex. In our attempt to identify more area-selective genes in the macaque neocortex, we found that testican-1, an occ1-related gene, and its family members also exhibit characteristic expression patterns along the visual pathway. The expression levels of testican-1 and testican-2 mRNAs as well as that of occ1 mRNA start of high in V1, progressively decrease along the ventral visual pathway, and end of low in the temporal areas. Complementary to them, the neuronal expression of SPARC mRNA is abundant in the association areas and scarce in V1. Whereas occ1, testican-1, and testican-2 mRNAs are preferentially distributed in thalamorecipient layers including “blobs,” SPARC mRNA expression avoids these layers. Neither SC1 nor testican-3 mRNA expression is selective to particular areas, but SC1 mRNA is abundantly observed in blobs. The expressions of occ1, testican-1, testican-2, and SC1 mRNA were downregulated after monocular tetrodotoxin injection. These results resonate with previous works on chemical and functional gradients along the primate occipitotemporal visual pathway and raise the possibility that these gradients and functional architecture may be related to the visual activity–dependent expression of these extracellular matrix glycoproteins.
Collapse
Affiliation(s)
- Toru Takahata
- Division of Brain Biology, National Institute for Basic Biology, Okazaki, Aichi 444-8585, Japan
| | | | | | | | | | | |
Collapse
|
38
|
Hirokawa J, Watakabe A, Ohsawa S, Yamamori T. Analysis of area-specific expression patterns of RORbeta, ER81 and Nurr1 mRNAs in rat neocortex by double in situ hybridization and cortical box method. PLoS One 2008; 3:e3266. [PMID: 18815614 PMCID: PMC2533703 DOI: 10.1371/journal.pone.0003266] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2008] [Accepted: 09/04/2008] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND The mammalian neocortex is subdivided into many areas, each of which exhibits distinctive lamina architecture. To investigate such area differences in detail, we chose three genes for comparative analyses, namely, RORbeta, ER81 and Nurr1, mRNAs of which have been reported to be mainly expressed in layers 4, 5 and 6, respectively. To analyze their qualitative and quantitative coexpression profiles in the rat neocortex, we used double in situ hybridization (ISH) histochemistry and cortical box method which we previously developed to integrate the data of different staining and individuals in a standard three-dimensional space. PRINCIPAL FINDINGS Our new approach resulted in three main observations. First, the three genes showed unique area distribution patterns that are mostly complementary to one another. The patterns revealed by cortical box method matched well with the cytoarchitectonic areas defined by Nissl staining. Second, at single cell level, RORbeta and ER81 mRNAs were coexpressed in a subpopulation of layer 5 neurons, whereas Nurr1 and ER81 mRNAs were not colocalized. Third, principal component analysis showed that the order of hierarchical processing in the cortex correlates well with the expression profiles of these three genes. Based on this analysis, the dysgranular zone (DZ) in the somatosensory area was considered to exhibit a profile of a higher order area, which is consistent with previous proposal. CONCLUSIONS/SIGNIFICANCE The tight relationship between the expression of the three layer specific genes and functional areas were revealed, demonstrating the usefulness of cortical box method in the study on the cerebral cortex. In particular, it allowed us to perform statistical evaluation and pattern matching, which would become important in interpreting the ever-increasing data of gene expression in the cortex.
Collapse
Affiliation(s)
- Junya Hirokawa
- Division of Brain Biology, National Institute for Basic Biology, Okazaki, Japan
- Department of Basic Biology, The Graduate University for Advanced Studies, Okazaki, Japan
| | - Akiya Watakabe
- Division of Brain Biology, National Institute for Basic Biology, Okazaki, Japan
- Department of Basic Biology, The Graduate University for Advanced Studies, Okazaki, Japan
| | - Sonoko Ohsawa
- Division of Brain Biology, National Institute for Basic Biology, Okazaki, Japan
| | - Tetsuo Yamamori
- Division of Brain Biology, National Institute for Basic Biology, Okazaki, Japan
- Department of Basic Biology, The Graduate University for Advanced Studies, Okazaki, Japan
| |
Collapse
|
39
|
Itoh S, Hachisuka A, Kawasaki N, Hashii N, Teshima R, Hayakawa T, Kawanishi T, Yamaguchi T. Glycosylation analysis of IgLON family proteins in rat brain by liquid chromatography and multiple-stage mass spectrometry. Biochemistry 2008; 47:10132-54. [PMID: 18729387 DOI: 10.1021/bi8009778] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
IgLON family proteins, including limbic-associated membrane protein (LAMP), opioid-binding cell adhesion molecule (OBCAM), neurotrimin, and Kilon, are immunoglobulin (Ig) superfamily cell adhesion molecules. These molecules are composed of three Ig domains and a glycosylphosphatidylinositol (GPI) anchor and contain six or seven potential N-glycosylation sites. Although their glycosylations are supposed to be associated with the development of the central nervous system like other Ig superfamily proteins, they are still unknown because of difficulty in isolating individual proteins with a high degree of homology in performing carbohydrate analysis. In this study, we conducted simultaneous site-specific glycosylation analysis of rat brain IgLON proteins by liquid chromatography and multiple-stage mass spectrometry (LC-MS ( n )). The rat brain GPI-linked proteins were enriched and separated by sodium dodecyl sulfate-polyacrylamide gel electrophoresis. The four proteins were extracted from the gel, and subjected to LC-MS ( n ) after proteinase digestions. A set of glycopeptide MS data, including the mass spectrum, the mass spectrum in the selected ion monitoring mode, and the product ion spectra, was selected from all data based on carbohydrate-related ions in the MS/MS spectrum. The peptide portion and the carbohydrate structure were identified on the basis of peptide-related ion and carbohydrate-related ions, and the accurate mass. The site-specific glycosylations of four proteins were elucidated as follows. N-Glycans near the N-terminal were disialic acid-conjugated complex- and hybrid-type oligosaccharides. The first Ig domains were occupied by Man-5-9. Diverse oligosaccharides, including Lewis a/x-modified glycans, a brain-specific glycan known as BA-2, and Man-5, were found to be attached to the third Ig domain. Three common structures of glycans were found in the GPI moiety of LAMP, OBCAM, and neurotrimin.
Collapse
Affiliation(s)
- Satsuki Itoh
- Division of Biological Chemistry and Biologicals, National Institute of Health Sciences, 1-18-1, Kamiyoga, Setagaya-ku, Tokyo 158-8501, Japan
| | | | | | | | | | | | | | | |
Collapse
|
40
|
Wang L, Hauser ER, Shah SH, Seo D, Sivashanmugam P, Exum ST, Gregory SG, Granger CB, Haines JL, Jones CJH, Crossman D, Haynes C, Kraus WE, Freedman NJ, Pericak-Vance MA, Goldschmidt-Clermont PJ, Vance JM. Polymorphisms of the tumor suppressor gene LSAMP are associated with left main coronary artery disease. Ann Hum Genet 2008; 72:443-53. [PMID: 18318786 DOI: 10.1111/j.1469-1809.2008.00433.x] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
Previous association mapping on chromosome 3q13-21 detected evidence for association at the limbic system-associated membrane protein (LSAMP) gene in individuals with late-onset coronary artery disease (CAD). LSAMP has never been implicated in the pathogenesis of CAD. We sought to thoroughly characterize the association and the gene. Non-redundant single nucleotide polymorphisms (SNPs) across the gene were examined in an initial dataset (168 cases with late-onset CAD, 149 controls). Stratification analysis on left main CAD (N = 102) revealed stronger association, which was further validated in a validation dataset (141 cases with left main CAD, 215 controls), a third control dataset (N = 255), and a family-based dataset (N = 2954). A haplotype residing in a novel alternative transcript of the LSAMP gene was significant in all independent case-control datasets (p = 0.0001 to 0.0205) and highly significant in the joint analysis (p = 0.00004). Lower expression of the novel alternative transcript was associated with the risk haplotype (p = 0.0002) and atherosclerosis burden in human aortas (p = 0.0001). Furthermore, silencing LSAMP expression in human aortic smooth muscle cells (SMCs) substantially augmented SMC proliferation (p<0.01). Therefore, the risk conferred by the LSAMP haplotype appears to be mediated by LSAMP down-regulation, which may promote SMC proliferation in the arterial wall and progression of atherosclerosis.
Collapse
Affiliation(s)
- L Wang
- Miami Institute of Human Genomics, University of Miami, Miami, FL 33101, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Hashimoto T, Yamada M, Maekawa S, Nakashima T, Miyata S. IgLON cell adhesion molecule Kilon is a crucial modulator for synapse number in hippocampal neurons. Brain Res 2008; 1224:1-11. [PMID: 18602091 DOI: 10.1016/j.brainres.2008.05.069] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2007] [Revised: 05/11/2008] [Accepted: 05/24/2008] [Indexed: 01/28/2023]
Abstract
Kilon is a member of the IgLON family belonging to the immunoglobulin superfamily of cell adhesion molecules. In the present study, we investigated temporal and spatial changes of Kilon expression and its modulatory functions for synapse number using hippocampal cultured neurons. Kilon was observed to localize chiefly at axons and presynaptic terminals at early culture stage, however, it was seen mainly at dendritic postsynaptic spine of mature neurons at late culture stages. Kilon was solubilized with detergent treatment at early culture stages, while it resisted to extraction of the detergent in mature neurons. The overexpression of Kilon gene using a plasmid vector decreased the number of dendritic synapses at early culture stages, whereas the overexpression increased the number of dendritic synapses at late culture. These results demonstrate the alteration of modulatory function of Kilon for the number of dendritic synapses concomitant with changes in its localization and detergent solubility during neuronal culture development.
Collapse
Affiliation(s)
- Takashi Hashimoto
- Department of Applied Biology, Kyoto Institute of Technology, Matsugasaki, Sakyo-ku, Kyoto 606-8585, Japan
| | | | | | | | | |
Collapse
|
42
|
Bekirov IH, Nagy V, Svoronos A, Huntley GW, Benson DL. Cadherin-8 and N-cadherin differentially regulate pre- and postsynaptic development of the hippocampal mossy fiber pathway. Hippocampus 2008; 18:349-63. [PMID: 18064706 DOI: 10.1002/hipo.20395] [Citation(s) in RCA: 55] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Cells sort into regions and groups in part by their selective surface expression of particular classic cadherins during development. In the nervous system, cadherin-based sorting can define axon tracts, restrict axonal and dendritic arbors to particular regions or layers, and may encode certain aspects of synapse specificity. The underlying model has been that afferents and their targets hold in common the expression of a particular cadherin, thereby providing a recognition code of homophilic cadherin binding. However, most neurons express multiple cadherins, and it is not clear whether multiple cadherins all act similarly in shaping neural circuitry. Here we asked how two such cadherins, cadherin-8 and N-cadherin, influence the guidance and differentiation of hippocampal mossy fibers. Using organotypic hippocampal cultures, we find that cadherin-8 regulates mossy fiber fasciculation and targeting, but has little effect on CA3 dendrites. In contrast, N-cadherin regulates mossy fiber fasciculation, but has little impact on axonal growth and targeting. However, N-cadherin is essential for CA3 dendrite arborization. Both cadherins are required for formation of proper numbers of presynaptic terminals. Mechanistically, such differential actions of these two cadherins could, in theory, reflect coupling to distinct intracellular binding partners. However, we find that both cadherins bind beta-catenin in dentate gyrus (DG). This suggests that cadherins may engage different intracellular signaling cascades downstream of beta-catenin, coopt different extracellular binding partners, or target distinct subcellular domains. Together our findings demonstrate that cadherin-8 and N-cadherin are critical for generating the mossy fiber pathway, but that each contributes differentially to afferent and target differentiation, thereby complementing one another in the assembly of a synaptic circuit.
Collapse
Affiliation(s)
- Iddil H Bekirov
- Fishberg Department of Neuroscience, Mount Sinai School of Medicine, New York, New York 10029, USA
| | | | | | | | | |
Collapse
|
43
|
Must A, Tasa G, Lang A, Vasar E, Kõks S, Maron E, Väli M. Association of limbic system-associated membrane protein (LSAMP) to male completed suicide. BMC MEDICAL GENETICS 2008; 9:34. [PMID: 18433483 PMCID: PMC2386445 DOI: 10.1186/1471-2350-9-34] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/25/2007] [Accepted: 04/23/2008] [Indexed: 11/21/2022]
Abstract
Background Neuroimaging studies have demonstrated volumetric abnormalities in limbic structures of suicide victims. The morphological changes might be caused by some inherited neurodevelopmental defect, such as failure to form proper axonal connections due to genetically determined dysfunction of neurite guidance molecules. Limbic system-associated membrane protein (LSAMP) is a neuronal adhesive molecule, preferentially expressed in developing limbic system neuronal dendrites and somata. Some evidence for the association between LSAMP gene and behavior has come from both animal as well as human studies but further investigation is required. In current study, polymorphic loci in human LSAMP gene were examined in order to reveal any associations between genetic variation in LSAMP and suicidal behaviour. Methods DNA was obtained from 288 male suicide victims and 327 healthy male volunteers. Thirty SNPs from LSAMP gene and adjacent region were selected by Tagger algorithm implemented in Haploview 3.32. Genotyping was performed using the SNPlex™ (Applied Biosystems) platform. Data was analyzed by Genemapper 3.7, Haploview 3.32 and SPSS 13.0. Results Chi square test revealed four allelic variants (rs2918215, rs2918213, rs9874470 and rs4821129) located in the intronic region of the gene to be associated with suicide, major alleles being overrepresented in suicide group. However, the associations did not survive multiple correction test. Defining the haplotype blocks using confidence interval algorithm implemented in Haploview 3.32, we failed to detect any associated haplotypes. Conclusion Despite a considerable amount of investigation on the nature of suicidal behaviour, its aetiology and pathogenesis remain unknown. This study examined the variability in LSAMP gene in relation to completed suicide. Our results indicate that LSAMP might play a role in pathoaetiology of suicidal behaviour but further studies are needed to understand its exact contribution.
Collapse
Affiliation(s)
- Anne Must
- Institute of Physiology, Tartu University, Ravila 19, Tartu 50411, Estonia.
| | | | | | | | | | | | | |
Collapse
|
44
|
Catania EH, Pimenta A, Levitt P. Genetic deletion of Lsamp causes exaggerated behavioral activation in novel environments. Behav Brain Res 2008; 188:380-90. [PMID: 18199495 PMCID: PMC2275759 DOI: 10.1016/j.bbr.2007.11.022] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2007] [Revised: 11/20/2007] [Accepted: 11/24/2007] [Indexed: 01/09/2023]
Abstract
The limbic system-associated membrane protein (LAMP) is a GPI-anchored cell adhesion molecule expressed heavily in limbic and limbic-associated regions of the developing and adult brain. Experimental studies show that LAMP promotes the growth of limbic neurons and guides the projections of limbic fibers. In order to examine the functional consequences of disrupting limbic circuit assembly, we generated a mouse line in which the Lsamp gene encoding LAMP was deleted. Basic neuroanatomical organization and sensory and motor development are normal in Lsamp(-/-) mice. The most profound change in behavior in both male and female Lsamp(-/-) mice is a heightened reactivity to novelty exhibited in several behavioral tests. Lsamp(-/-) mice display hyperactivity in a novel arena and both sexes habituate to the same activity levels as their wild type littermates, but at different rates. In the elevated plus maze, Lsamp(-/-) mice exhibit increased total arm entries, with a bias towards the open arms; they spend more time in the open arms and have a substantial increase in the amount of risk assessment in unprotected areas of the maze. In the y-maze, Lsamp(-/-) mice exhibit characteristic hyperactivity and a decreased level of spontaneous alternation during the period when their novelty-induced hyperactivity is at its peak. We hypothesize that Lsamp(-/-) mice may not simply exhibit a decrease in anxiety, but may have a heightened, and possibly maladaptive, response to novel environmental stressors. Genetic deletion of Lsamp may thus cause circumscribed changes in the fine connectivity of specific circuits that underlie these behaviors.
Collapse
|
45
|
Yamada M, Hashimoto T, Hayashi N, Higuchi M, Murakami A, Nakashima T, Maekawa S, Miyata S. Synaptic adhesion molecule OBCAM; synaptogenesis and dynamic internalization. Brain Res 2007; 1165:5-14. [PMID: 17658490 DOI: 10.1016/j.brainres.2007.04.062] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2007] [Revised: 03/13/2007] [Accepted: 04/22/2007] [Indexed: 11/23/2022]
Abstract
Opioid-binding cell adhesion molecule (OBCAM) is the member of the IgLON family, a subgroup of the immunoglobulin superfamily. In the present study, the functions and dynamics of OBCAM were investigated in hippocampal neurons in vitro. Western blotting revealed that OBCAM expression was low at early stages of culture but it was increased as culture development. Double labeling immunofluorescence microscopy showed that OBCAM immunoreactivity was localized mainly at postsynaptic spines labeled with phalloidin and anti-PSD-95. The inhibition of OBCAM function with the specific antibody resulted in a significant decrease in the number of synapses on dendrites compared with control mouse IgG. The suppression of OBCAM expression using the antisense oligodeoxynucleotide also impaired the formation of synapses compared with control universal ones. The overexpression of OBCAM mRNA using a plasmid vector augmented the formation of synapses. Moreover, the internalization of OBCAM was promoted with increased neuronal activity by 4-aminopyridine. This internalization was reduced with the treatment of filipin, a sterol agent, indicating that this process is a raft-dependent pathway. These results indicate that OBCAM is a synaptic cell adhesion molecule concerning synaptogenesis and its surface localization is dynamically regulated in response to neuronal activity.
Collapse
Affiliation(s)
- Mayumi Yamada
- Department of Applied Biology, Kyoto Institute of Technology, Matsugasaki, Sakyo-ku, Kyoto 606-8585, Japan
| | | | | | | | | | | | | | | |
Collapse
|
46
|
Nelovkov A, Areda T, Innos J, Kõks S, Vasar E. Rats displaying distinct exploratory activity also have different expression patterns of gamma-aminobutyric acid- and cholecystokinin-related genes in brain regions. Brain Res 2006; 1100:21-31. [PMID: 16769038 DOI: 10.1016/j.brainres.2006.05.007] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2005] [Revised: 04/24/2006] [Accepted: 05/03/2006] [Indexed: 11/20/2022]
Abstract
The aim of the present study was to evaluate the expression of genes in relation to the exploratory activity of rats. Limbic system-associated membrane protein (LsAMP) gene, gamma-aminobutyric acid-(GABA)- and cholecystokinin-(CCK)-related genes were analyzed in the frontal cortex, amygdala and periaqueductal gray (PAG) after exposure of rats to exploratory challenge. Two groups of animals displaying low and high exploratory activity in the elevated plus-maze were selected for gene expression studies from the population of 43 male Wistar rats. Eight rats were taken randomly from the same cages as animals exposed to the plus-maze, but they were not subjected to the exploratory test. This home-cage control group was also used for gene expression analysis in order to explore a possible impact of the plus-maze exposure. Rats with low and high exploratory activity displayed clearly distinct profiles in gene expression. Most prominent alterations were established in the amygdala where almost all GABA-related and CCK receptor genes were two- to five-fold up-regulated in low exploratory activity rats compared to high exploratory activity and home-cage control group. The expression of several GABA-related genes was also increased in the PAG of animals displaying low exploratory activity compared to the other groups. Moreover, we found reduced expression of GABA- and CCK-related genes in all three brain regions in animals with high exploratory activity compared to the home-cage control group. In addition to these findings, we established a significantly increased expression of the LsAMP gene in the amygdala and PAG of low exploratory activity animals compared with the other groups. In conclusion, low and high exploratory activity rats differed not only by their exploratory activity but also displayed opposite gene expression patterns. Low exploratory activity of rats correlated with the up-regulation of LsAMP and GABA-related genes in the amygdala and PAG and with the up-regulation of CCK receptors in the amygdala. High exploratory activity of rats was related to a significant down-regulation of CCK receptor genes in the amygdala and PAG. These rats also displayed the reduced expression of GABA-related genes in the frontal cortex and PAG.
Collapse
Affiliation(s)
- Aleksei Nelovkov
- Department of Physiology, University of Tartu, Biomedicum, 19 Ravila Street, 51014 Tartu, Estonia
| | | | | | | | | |
Collapse
|
47
|
Strigini M, Cantera R, Morin X, Bastiani MJ, Bate M, Karagogeos D. The IgLON protein Lachesin is required for the blood-brain barrier in Drosophila. Mol Cell Neurosci 2006; 32:91-101. [PMID: 16682215 DOI: 10.1016/j.mcn.2006.03.001] [Citation(s) in RCA: 34] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2006] [Revised: 03/05/2006] [Accepted: 03/09/2006] [Indexed: 10/24/2022] Open
Abstract
In the mammalian peripheral nervous system, nerve insulation depends on the integrity of paranodal junctions between axons and their ensheathing glia. Ultrastructurally, these junctions are similar to the septate junctions (SJ) of invertebrates. In Drosophila, SJ are found in epithelia and in the glia that form the blood-brain barrier (BBB). Drosophila NeurexinIV and Gliotactin, two components of SJ, play an important role in nerve ensheathment and insulation. Here, we report that Drosophila Lachesin (Lac), another SJ component, is also required for a functional BBB. In the developing nervous system, Lac is expressed in a dynamic pattern by surface glia and a subset of neurons. Ultrastructural analysis of Lac mutant embryos shows poorly developed SJ in surface glia and epithelia where Lac is expressed. Mutant embryos undergo a phase of hyperactivity, with unpatterned muscle contractions, and subsequently become paralyzed and fail to hatch. We propose that this phenotype reflects a failure in BBB function.
Collapse
Affiliation(s)
- Maura Strigini
- IMBB/FORTH, Vassilika Vouton, Iraklio, Crete GR-71110, Greece.
| | | | | | | | | | | |
Collapse
|
48
|
Ikegaya Y. [Potential roles for mossy fiber sprouting in temporal lobe epilepsy]. Nihon Yakurigaku Zasshi 2006; 127:355-61. [PMID: 16819240 DOI: 10.1254/fpj.127.355] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/10/2023]
|
49
|
Petryshen TL, Kirby A, Hammer RP, Purcell S, O'Leary SB, Singer JB, Hill AE, Nadeau JH, Daly MJ, Sklar P. Two quantitative trait loci for prepulse inhibition of startle identified on mouse chromosome 16 using chromosome substitution strains. Genetics 2005; 171:1895-904. [PMID: 15998716 PMCID: PMC1456091 DOI: 10.1534/genetics.105.045658] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2005] [Accepted: 06/28/2005] [Indexed: 11/18/2022] Open
Abstract
Prepulse inhibition (PPI) of acoustic startle is a genetically complex quantitative phenotype of considerable medical interest due to its impairment in psychiatric disorders such as schizophrenia. To identify quantitative trait loci (QTL) involved in mouse PPI, we studied mouse chromosome substitution strains (CSS) that each carry a homologous chromosome pair from the A/J inbred strain on a host C57BL/6J inbred strain background. We determined that the chromosome 16 substitution strain has elevated PPI compared to C57BL/6J (P = 1.6 x 10(-11)), indicating that chromosome 16 carries one or more PPI genes. QTL mapping using 87 F(2) intercross progeny identified two significant chromosome 16 loci with LODs of 3.9 and 4.7 (significance threshold LOD is 2.3). The QTL were each highly significant independently and do not appear to interact. Sequence variation between B6 and A/J was used to identify strong candidate genes in the QTL regions, some of which have known neuronal functions. In conclusion, we used mouse CSS to rapidly and efficiently identify two significant QTL for PPI on mouse chromosome 16. The regions contain a limited number of strong biological candidate genes that are potential risk genes for psychiatric disorders in which patients have PPI impairments.
Collapse
Affiliation(s)
- Tracey L Petryshen
- Psychiatric and Neurodevelopmental Genetics Unit, Center for Human Genetic Research, Broad Institute of Harvard, 185 Cambridge Street, Cambridge, MA 02139, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Schrimpf SP, Meskenaite V, Brunner E, Rutishauser D, Walther P, Eng J, Aebersold R, Sonderegger P. Proteomic analysis of synaptosomes using isotope-coded affinity tags and mass spectrometry. Proteomics 2005; 5:2531-41. [PMID: 15984043 DOI: 10.1002/pmic.200401198] [Citation(s) in RCA: 92] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Synaptosomes are isolated synapses produced by subcellular fractionation of brain tissue. They contain the complete presynaptic terminal, including mitochondria and synaptic vesicles, and portions of the postsynaptic side, including the postsynaptic membrane and the postsynaptic density (PSyD). A proteomic characterisation of synaptosomes isolated from mouse brain was performed employing the isotope-coded affinity tag (ICAT) method and tandem mass spectrometry (MS/MS). After isotopic labelling and tryptic digestion, peptides were fractionated by cation exchange chromatography and cysteine-containing peptides were isolated by affinity chromatography. The peptides were identified by microcapillary liquid chromatography-electrospray ionisation MS/MS (muLC-ESI MS/MS). In two experiments, peptides representing a total of 1131 database entries were identified. They are involved in different presynaptic and postsynaptic functions, including synaptic vesicle exocytosis for neurotransmitter release, vesicle endocytosis for synaptic vesicle recycling, as well as postsynaptic receptors and proteins constituting the PSyD. Moreover, a large number of soluble and membrane-bound molecules serving functions in synaptic signal transduction and metabolism were detected. The results provide an inventory of the synaptic proteome and confirm the suitability of the ICAT method for the assessment of synaptic structure, function and plasticity.
Collapse
Affiliation(s)
- Sabine P Schrimpf
- Department of Biochemistry, University of Zurich, Zurich, Switzerland
| | | | | | | | | | | | | | | |
Collapse
|