1
|
Millard N, Chen JH, Palshikar MG, Pelka K, Spurrell M, Price C, He J, Hacohen N, Raychaudhuri S, Korsunsky I. Batch correcting single-cell spatial transcriptomics count data with Crescendo improves visualization and detection of spatial gene patterns. Genome Biol 2025; 26:36. [PMID: 40001084 PMCID: PMC11863647 DOI: 10.1186/s13059-025-03479-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2024] [Accepted: 01/21/2025] [Indexed: 02/27/2025] Open
Abstract
Spatial transcriptomics facilitates gene expression analysis of cells in their spatial anatomical context. Batch effects hinder visualization of gene spatial patterns across samples. We present the Crescendo algorithm to correct for batch effects at the gene expression level and enable accurate visualization of gene expression patterns across multiple samples. We show Crescendo's utility and scalability across three datasets ranging from 170,000 to 7 million single cells across spatial and single-cell RNA sequencing technologies. By correcting for batch effects, Crescendo enhances spatial transcriptomics analyses to detect gene colocalization and ligand-receptor interactions and enables cross-technology information transfer.
Collapse
Affiliation(s)
- Nghia Millard
- Division of Rheumatology, Inflammation and Immunity, Brigham and Women's Hospital, Boston, MA, USA
- Division of Genetics, Brigham and Women's Hospital, Boston, MA, USA
- Center for Data Sciences, Brigham and Women's Hospital, Boston, MA, USA
- Department of Biomedical Informatics, Harvard Medical School, Boston, MA, USA
- Department of Medicine, Harvard Medical School, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Jonathan H Chen
- Harvard Medical School, Boston, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Massachusetts General Hospital (MGH) Cancer Center, Harvard Medical School, Boston, MA, USA
- Department of Pathology, MGH, Boston, MA, USA
| | - Mukta G Palshikar
- Division of Genetics, Brigham and Women's Hospital, Boston, MA, USA
- Center for Data Sciences, Brigham and Women's Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Karin Pelka
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Massachusetts General Hospital (MGH) Cancer Center, Harvard Medical School, Boston, MA, USA
- UCSF Institute of Genomic Immunology, Gladstone Institutes, San Francisco, CA, USA
| | - Maxwell Spurrell
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Massachusetts General Hospital (MGH) Cancer Center, Harvard Medical School, Boston, MA, USA
- Department of Pathology, MGH, Boston, MA, USA
| | | | | | - Nir Hacohen
- Department of Immunology, Harvard Medical School, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Massachusetts General Hospital (MGH) Cancer Center, Harvard Medical School, Boston, MA, USA
| | - Soumya Raychaudhuri
- Division of Rheumatology, Inflammation and Immunity, Brigham and Women's Hospital, Boston, MA, USA.
- Division of Genetics, Brigham and Women's Hospital, Boston, MA, USA.
- Center for Data Sciences, Brigham and Women's Hospital, Boston, MA, USA.
- Department of Biomedical Informatics, Harvard Medical School, Boston, MA, USA.
- Department of Medicine, Harvard Medical School, Boston, MA, USA.
- Harvard Medical School, Boston, MA, USA.
- Broad Institute of MIT and Harvard, Cambridge, MA, USA.
| | - Ilya Korsunsky
- Division of Genetics, Brigham and Women's Hospital, Boston, MA, USA.
- Center for Data Sciences, Brigham and Women's Hospital, Boston, MA, USA.
- Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
2
|
Zhou W, Munoz JR, Henry Ho HY, Hanamura K, Dalva MB. Specific neuroblast-derived signals control both cell migration and fate in the rostral migratory stream. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.02.18.638163. [PMID: 40027825 PMCID: PMC11870606 DOI: 10.1101/2025.02.18.638163] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 03/05/2025]
Abstract
Functional neuronal circuits require neuroblasts migrate to appropriate locations and then differentiate into neuronal subtypes. However, it remains unknown how neuroblasts in the subventricular zone (SVZ) are guided through the rostral migratory stream (RMS) to the olfactory bulb (OB). Here we define EphB2 as a neuroblast-derived cue that controls migration along the RMS and helps to determine cell fate. Within the RMS, EphB2 is expressed selectively in, kinase-active in, and required for the migration of neuroblasts. As neuroblasts enter the OB and differentiate, EphB kinase activity is down-regulated, and in the granule cell layer (GCL), EphB2 expression is down-regulated. Blocking EphB kinase activity or knocking down EphB2 results in defects in migration and premature cellular differentiation in the RMS. Unexpectedly, premature loss of EphB2 expression causes neuroblasts to stop migrating and differentiate into astrocyte-like cells. Thus, EphB2 kinase activity and expression are linked to migration and specification of neuroblast fate.
Collapse
Affiliation(s)
- Wei Zhou
- Department of Neuroscience and the Jefferson Synaptic Biology Center, Thomas Jefferson University, 900 Walnut Street, Philadelphia, PA 19107, USA
| | - James R. Munoz
- Present address: Department of Psychology and Neuroscience, Nova Southeastern University, 3301 College Avenue, Fort Lauderdale, FL 33314, USA
| | - Hsin-Yi Henry Ho
- Department of Cell Biology and Human Anatomy, UC Davis School of Medicine, One Shields Avenue, Davis, CA 95616
| | - Kenji Hanamura
- Department of Neuroscience and the Jefferson Synaptic Biology Center, Thomas Jefferson University, 900 Walnut Street, Philadelphia, PA 19107, USA
- Present address: Faculty of Medical Technology, Department of Radiological Technology, Niigata University of Health and Welfare, 1398 Shimami-cho, Kita-ku, Niigata-City, 950-3198, Japan
| | - Matthew B. Dalva
- Department of Neuroscience and the Jefferson Synaptic Biology Center, Thomas Jefferson University, 900 Walnut Street, Philadelphia, PA 19107, USA
- Present Address: Tulane Brain Institute, Tulane University, 201 Flower Hall, 6823 St. Charles Avenue, New Orleans, LA 70118
| |
Collapse
|
3
|
Wozna‐Wysocka M, Jazurek‐Ciesiolka M, Przybyl L, Wronka D, Misiorek JO, Suszynska‐Zajczyk J, Figura G, Ciesiolka A, Sobieszczanska P, Zeller A, Niemira M, Switonski PM, Fiszer A. Insights into RNA-mediated pathology in new mouse models of Huntington's disease. FASEB J 2024; 38:e70182. [PMID: 39604147 PMCID: PMC11602643 DOI: 10.1096/fj.202401465r] [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: 06/28/2024] [Revised: 10/09/2024] [Accepted: 11/04/2024] [Indexed: 11/29/2024]
Abstract
Huntington's disease (HD) is a neurodegenerative polyglutamine (polyQ) disease resulting from the expansion of CAG repeats located in the ORF of the huntingtin gene (HTT). The extent to which mutant mRNA-driven disruptions contribute to HD pathogenesis, particularly in comparison to the dominant mechanisms related to the gain-of-function effects of the mutant polyQ protein, is still debatable. To evaluate this contribution in vivo, we generated two mouse models through a knock-in strategy at the Rosa26 locus. These models expressed distinct variants of human mutant HTT cDNA fragment: a translated variant (HD/100Q model, serving as a reference) and a nontranslated variant (HD/100CAG model). The cohorts of animals were subjected to a broad spectrum of molecular, behavioral, and cognitive analysis for 21 months. Behavioral testing revealed alterations in both models, with the HD/100Q model exhibiting late disease phenotype. The rotarod, static rod, and open-field tests showed some motor deficits in HD/100CAG and HD/100Q model mice during the light phase, while ActiMot indicated hyperkinesis during the dark phase. Both models also exhibited certain gene deregulations in the striatum that are related to disrupted pathways and phenotype alterations observed in HD. In conclusion, we provide in vivo evidence for a minor contributory role of mutant RNA in HD pathogenesis. The separated effects resulting from the presence of mutant RNA in the HD/100CAG model led to less severe but, to some extent, similar types of impairments as in the HD/100Q model. Increased anxiety was one of the most substantial effects caused by mutant HTT RNA.
Collapse
Affiliation(s)
| | | | - Lukasz Przybyl
- Institute of Bioorganic ChemistryPolish Academy of SciencesPoznanPoland
| | - Dorota Wronka
- Institute of Bioorganic ChemistryPolish Academy of SciencesPoznanPoland
| | | | | | - Grzegorz Figura
- Institute of Bioorganic ChemistryPolish Academy of SciencesPoznanPoland
- Present address:
Department of Bioenergetics, Institute of Molecular Biology and BiotechnologyAdam Mickiewicz UniversityPoznanPoland
| | - Adam Ciesiolka
- Institute of Bioorganic ChemistryPolish Academy of SciencesPoznanPoland
- Present address:
Department of Gene Expression, Institute of Molecular Biology and BiotechnologyAdam Mickiewicz UniversityPoznanPoland
| | | | - Anna Zeller
- Genomics and Epigenomics Laboratory, Clinical Research CentreMedical University of BialystokBialystokPoland
| | - Magdalena Niemira
- Genomics and Epigenomics Laboratory, Clinical Research CentreMedical University of BialystokBialystokPoland
| | | | - Agnieszka Fiszer
- Institute of Bioorganic ChemistryPolish Academy of SciencesPoznanPoland
| |
Collapse
|
4
|
Madrid A, Koueik J, Papale LA, Chebel R, Renteria I, Cannon E, Hogan KJ, Alisch RS, Iskandar BJ. Folate-mediated transgenerational inheritance of sperm DNA methylation patterns correlate with spinal axon regeneration. Epigenetics 2024; 19:2380930. [PMID: 39066680 PMCID: PMC11285217 DOI: 10.1080/15592294.2024.2380930] [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: 02/10/2024] [Revised: 06/01/2024] [Accepted: 07/11/2024] [Indexed: 07/30/2024] Open
Abstract
In mammals, the molecular mechanisms underlying transgenerational inheritance of phenotypic traits in serial generations of progeny after ancestral environmental exposures, without variation in DNA sequence, remain elusive. We've recently described transmission of a beneficial trait in rats and mice, in which F0 supplementation of methyl donors, including folic acid, generates enhanced axon regeneration after sharp spinal cord injury in untreated F1 to F3 progeny linked to differential DNA methylation levels in spinal cord tissue. To test whether the transgenerational effect of folic acid is transmitted via the germline, we performed whole-genome methylation sequencing on sperm DNA from F0 mice treated with either folic acid or vehicle control, and their F1, F2, and F3 untreated progeny. Transgenerational differentially methylated regions (DMRs) are observed in each consecutive generation and distinguish folic acid from untreated lineages, predominate outside of CpG islands and in regions of the genome that regulate gene expression, including promoters, and overlap at both the differentially methylated position (DMP) and gene levels. These findings indicate that molecular changes between generations are caused by ancestral folate supplementation. In addition, 29,719 DMPs exhibit serial increases or decreases in DNA methylation levels in successive generations of untreated offspring, correlating with a serial increase in the phenotype across generations, consistent with a 'wash-in' effect. Sibship-specific DMPs annotate to genes that participate in axon- and synapse-related pathways.
Collapse
Affiliation(s)
- Andy Madrid
- Department of Neurological Surgery, University of Wisconsin School of Medicine and Public Health, Madison, USA
| | - Joyce Koueik
- Department of Neurological Surgery, University of Wisconsin School of Medicine and Public Health, Madison, USA
| | - Ligia A. Papale
- Department of Neurological Surgery, University of Wisconsin School of Medicine and Public Health, Madison, USA
| | - Roy Chebel
- Department of Neurological Surgery, University of Wisconsin School of Medicine and Public Health, Madison, USA
| | - Isabelle Renteria
- Department of Neurological Surgery, University of Wisconsin School of Medicine and Public Health, Madison, USA
| | - Emily Cannon
- Department of Neurological Surgery, University of Wisconsin School of Medicine and Public Health, Madison, USA
| | - Kirk J. Hogan
- Department of Anesthesiology, University of Wisconsin School of Medicine and Public Health, Madison, USA
| | - Reid S. Alisch
- Department of Neurological Surgery, University of Wisconsin School of Medicine and Public Health, Madison, USA
| | - Bermans J. Iskandar
- Department of Neurological Surgery, University of Wisconsin School of Medicine and Public Health, Madison, USA
| |
Collapse
|
5
|
Mack CM, Tsui-Bowen A, Smith AR, Jensen KF, Kodavanti PRS, Moser VC, Mundy WR, Shafer TJ, Herr DW. Identification of neural-relevant toxcast high-throughput assay intended gene targets: Applicability to neurotoxicity and neurotoxicant putative molecular initiating events. Neurotoxicology 2024; 103:256-265. [PMID: 38977203 DOI: 10.1016/j.neuro.2024.07.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2024] [Revised: 06/29/2024] [Accepted: 07/01/2024] [Indexed: 07/10/2024]
Abstract
The US EPA's Toxicity Forecaster (ToxCast) is a suite of high-throughput in vitro assays to screen environmental toxicants and predict potential toxicity of uncharacterized chemicals. This work examines the relevance of ToxCast assay intended gene targets to putative molecular initiating events (MIEs) of neurotoxicants. This effort is needed as there is growing interest in the regulatory and scientific communities about developing new approach methodologies (NAMs) to screen large numbers of chemicals for neurotoxicity and developmental neurotoxicity. Assay gene function (GeneCards, NCBI-PUBMED) was used to categorize gene target neural relevance (1 = neural, 2 = neural development, 3 = general cellular process, 3 A = cellular process critical during neural development, 4 = unlikely significance). Of 481 unique gene targets, 80 = category 1 (16.6 %); 16 = category 2 (3.3 %); 303 = category 3 (63.0 %); 97 = category 3 A (20.2 %); 82 = category 4 (17.0 %). A representative list of neurotoxicants (548) was researched (ex. PUBMED, PubChem) for neurotoxicity associated MIEs/Key Events (KEs). MIEs were identified for 375 compounds, whereas only KEs for 173. ToxCast gene targets associated with MIEs were primarily neurotransmitter (ex. dopaminergic, GABA)receptors and ion channels (calcium, sodium, potassium). Conversely, numerous MIEs associated with neurotoxicity were absent. Oxidative stress (OS) mechanisms were 79.1 % of KEs. In summary, 40 % of ToxCast assay gene targets are relevant to neurotoxicity mechanisms. Additional receptor and ion channel subtypes and increased OS pathway coverage are identified for potential future assay inclusion to provide more complete coverage of neural and developmental neural targets in assessing neurotoxicity.
Collapse
Affiliation(s)
- Cina M Mack
- Center for Public Health and Environmental Assessment, US Environmental Protection Agency, Research Triangle Park, NC 27711, USA.
| | | | - Alicia R Smith
- Oak Ridge Institute for Science Education, Oak Ridge, TN 37830, USA.
| | - Karl F Jensen
- US Environmental Protection Agency, Research Triangle Park, NC 27711, USA
| | - Prasada Rao S Kodavanti
- Center for Public Health and Environmental Assessment, US Environmental Protection Agency, Research Triangle Park, NC 27711, USA.
| | - Virginia C Moser
- US Environmental Protection Agency, Research Triangle Park, NC 27711, USA.
| | - William R Mundy
- US Environmental Protection Agency, Research Triangle Park, NC 27711, USA.
| | - Timothy J Shafer
- Center for Computational Toxicology and Exposure, US Environmental Protection Agency, Research Triangle Park, NC 27711, USA.
| | - David W Herr
- Center for Public Health and Environmental Assessment, US Environmental Protection Agency, Research Triangle Park, NC 27711, USA.
| |
Collapse
|
6
|
Dalton GD, Siecinski SK, Nikolova VD, Cofer GP, Hornburg KJ, Qi Y, Johnson GA, Jiang YH, Moy SS, Gregory SG. Transcriptome analysis identifies an ASD-Like phenotype in oligodendrocytes and microglia from C58/J amygdala that is dependent on sex and sociability. BEHAVIORAL AND BRAIN FUNCTIONS : BBF 2024; 20:14. [PMID: 38898502 PMCID: PMC11188533 DOI: 10.1186/s12993-024-00240-3] [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: 02/02/2024] [Accepted: 05/27/2024] [Indexed: 06/21/2024]
Abstract
BACKGROUND Autism Spectrum Disorder (ASD) is a group of neurodevelopmental disorders with higher incidence in males and is characterized by atypical verbal/nonverbal communication, restricted interests that can be accompanied by repetitive behavior, and disturbances in social behavior. This study investigated brain mechanisms that contribute to sociability deficits and sex differences in an ASD animal model. METHODS Sociability was measured in C58/J and C57BL/6J mice using the 3-chamber social choice test. Bulk RNA-Seq and snRNA-Seq identified transcriptional changes in C58/J and C57BL/6J amygdala within which DMRseq was used to measure differentially methylated regions in amygdala. RESULTS C58/J mice displayed divergent social strata in the 3-chamber test. Transcriptional and pathway signatures revealed immune-related biological processes differ between C58/J and C57BL/6J amygdala. Hypermethylated and hypomethylated genes were identified in C58/J versus C57BL/6J amygdala. snRNA-Seq data in C58/J amygdala identified differential transcriptional signatures within oligodendrocytes and microglia characterized by increased ASD risk gene expression and predicted impaired myelination that was dependent on sex and sociability. RNA velocity, gene regulatory network, and cell communication analysis showed diminished oligodendrocyte/microglia differentiation. Findings were verified using Bulk RNA-Seq and demonstrated oxytocin's beneficial effects on myelin gene expression. LIMITATIONS Our findings are significant. However, limitations can be noted. The cellular mechanisms linking reduced oligodendrocyte differentiation and reduced myelination to an ASD phenotype in C58/J mice need further investigation. Additional snRNA-Seq and spatial studies would determine if effects in oligodendrocytes/microglia are unique to amygdala or if this occurs in other brain regions. Oxytocin's effects need further examination to understand its' potential as an ASD therapeutic. CONCLUSIONS Our work demonstrates the C58/J mouse model's utility in evaluating the influence of sex and sociability on the transcriptome in concomitant brain regions involved in ASD. Our single-nucleus transcriptome analysis elucidates potential pathological roles of oligodendrocytes and microglia in ASD. This investigation provides details regarding regulatory features disrupted in these cell types, including transcriptional gene dysregulation, aberrant cell differentiation, altered gene regulatory networks, and changes to key pathways that promote microglia/oligodendrocyte differentiation. Our studies provide insight into interactions between genetic risk and epigenetic processes associated with divergent affiliative behavior and lack of positive sociability.
Collapse
Affiliation(s)
- George D Dalton
- Duke Molecular Physiology Institute, Duke University School of Medicine, Durham, NC, 27701, USA
| | - Stephen K Siecinski
- Duke Molecular Physiology Institute, Duke University School of Medicine, Durham, NC, 27701, USA
| | - Viktoriya D Nikolova
- Department of Psychiatry, The University of North Carolina at Chapel Hill, Chapel Hill, NC, 27516, USA
| | - Gary P Cofer
- Center for In Vivo Microscopy, Duke University, Durham, NC, 27710, USA
| | | | - Yi Qi
- Center for In Vivo Microscopy, Duke University, Durham, NC, 27710, USA
| | - G Allan Johnson
- Center for In Vivo Microscopy, Duke University, Durham, NC, 27710, USA
| | - Yong-Hui Jiang
- Department of Genetics, Neuroscience, and Pediatrics, Yale University School of Medicine, New Haven, CT, 06520, USA
| | - Sheryl S Moy
- Department of Psychiatry, The University of North Carolina at Chapel Hill, Chapel Hill, NC, 27516, USA
| | - Simon G Gregory
- Duke Molecular Physiology Institute, Duke University School of Medicine, Durham, NC, 27701, USA.
- Department of Neurology, Duke University School of Medicine, Durham, NC, 27710, USA.
- Department of Neurology, Molecular Genetics and Microbiology Duke Molecular Physiology Institute, 300 N. Duke Street, DUMC 104775, Durham, NC, 27701, USA.
| |
Collapse
|
7
|
Yao N, Ma Q, Yi W, Zhu Y, Liu Y, Gao X, Zhang Q, Luo W. Artesunate attenuates the tumorigenesis of choroidal melanoma via inhibiting EFNA3 through Stat3/Akt signaling pathway. J Cancer Res Clin Oncol 2024; 150:202. [PMID: 38630320 PMCID: PMC11024049 DOI: 10.1007/s00432-024-05711-8] [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: 02/04/2024] [Accepted: 03/18/2024] [Indexed: 04/19/2024]
Abstract
PURPOSE Choroidal melanoma (CM), a kind of malignant tumor, is the main type of Uveal melanoma and one half of CM patients develop metastases. As a member of Eph/ephrin pathway that plays vital role in tumors, EphrinA3 (EFNA3) has been proved to promote tumorigenesis in many tumors. But the effect of EFNA3 in CM has not been studied yet. Through inhibiting angiogenesis, inducing apoptosis and autophagy and so on, Artesunate (ART) plays a key anti-tumor role in many tumors, including CM. However, the exact mechanisms of anti-tumor in CM remain unclear. METHODS The UALCAN and TIMER v2.0 database analyzed the role of EFNA3 in CM patients. Quantitative real time polymerase chain reaction (qPCR) and Western blot were used to detect the expression of EFNA3 in CM. The growth ability of CM was tested by clonogenic assay and Cell counting kit-8 assay, and the migration ability using Transwell assay. RESULTS Our results found EFNA3 boosted CM cells' growth and migration through activating Stat3/Akt signaling pathway, while ART inhibited the tumor promoting effect of CM via downregulating EFNA3. In xenograft tumor model, EFNA3 knockdown and ART significantly inhibited tumor growth. CONCLUSION EFNA3 could be a valuable prognostic factor in CM.
Collapse
Affiliation(s)
- Ningning Yao
- Department of Ophthalmology of The Affiliated Hospital of Qingdao University, Qingdao, 266003, China
| | - Qingyue Ma
- Department of Ophthalmology of The Affiliated Hospital of Qingdao University, Qingdao, 266003, China
| | - Wendan Yi
- Department of Ophthalmology of The Affiliated Hospital of Qingdao University, Qingdao, 266003, China
| | - Yuanzhang Zhu
- Department of Ophthalmology of The Affiliated Hospital of Qingdao University, Qingdao, 266003, China
| | - Yichong Liu
- Department of Ophthalmology of The Affiliated Hospital of Qingdao University, Qingdao, 266003, China
| | - Xiaodi Gao
- Department of Ophthalmology of The Affiliated Hospital of Qingdao University, Qingdao, 266003, China
| | - Qian Zhang
- Department of Ophthalmology of The Affiliated Hospital of Qingdao University, Qingdao, 266003, China
| | - Wenjuan Luo
- Department of Ophthalmology of The Affiliated Hospital of Qingdao University, Qingdao, 266003, China.
| |
Collapse
|
8
|
Primak A, Bozov K, Rubina K, Dzhauari S, Neyfeld E, Illarionova M, Semina E, Sheleg D, Tkachuk V, Karagyaur M. Morphogenetic theory of mental and cognitive disorders: the role of neurotrophic and guidance molecules. Front Mol Neurosci 2024; 17:1361764. [PMID: 38646100 PMCID: PMC11027769 DOI: 10.3389/fnmol.2024.1361764] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2023] [Accepted: 03/04/2024] [Indexed: 04/23/2024] Open
Abstract
Mental illness and cognitive disorders represent a serious problem for the modern society. Many studies indicate that mental disorders are polygenic and that impaired brain development may lay the ground for their manifestation. Neural tissue development is a complex and multistage process that involves a large number of distant and contact molecules. In this review, we have considered the key steps of brain morphogenesis, and the major molecule families involved in these process. The review provides many indications of the important contribution of the brain development process and correct functioning of certain genes to human mental health. To our knowledge, this comprehensive review is one of the first in this field. We suppose that this review may be useful to novice researchers and clinicians wishing to navigate the field.
Collapse
Affiliation(s)
- Alexandra Primak
- Faculty of Medicine, Lomonosov Moscow State University, Moscow, Russia
| | - Kirill Bozov
- Faculty of Medicine, Lomonosov Moscow State University, Moscow, Russia
| | - Kseniya Rubina
- Faculty of Medicine, Lomonosov Moscow State University, Moscow, Russia
| | - Stalik Dzhauari
- Faculty of Medicine, Lomonosov Moscow State University, Moscow, Russia
| | - Elena Neyfeld
- Faculty of Medicine, Lomonosov Moscow State University, Moscow, Russia
- Federal State Budgetary Educational Institution of the Higher Education “A.I. Yevdokimov Moscow State University of Medicine and Dentistry” of the Ministry of Healthcare of the Russian Federation, Moscow, Russia
| | - Maria Illarionova
- Faculty of Medicine, Lomonosov Moscow State University, Moscow, Russia
| | - Ekaterina Semina
- Faculty of Medicine, Lomonosov Moscow State University, Moscow, Russia
| | - Dmitriy Sheleg
- Faculty of Medicine, Lomonosov Moscow State University, Moscow, Russia
- Federal State Budgetary Educational Institution of the Higher Education “A.I. Yevdokimov Moscow State University of Medicine and Dentistry” of the Ministry of Healthcare of the Russian Federation, Moscow, Russia
| | - Vsevolod Tkachuk
- Faculty of Medicine, Lomonosov Moscow State University, Moscow, Russia
- Institute for Regenerative Medicine, Medical Research and Education Center, Lomonosov Moscow State University, Moscow, Russia
| | - Maxim Karagyaur
- Faculty of Medicine, Lomonosov Moscow State University, Moscow, Russia
- Institute for Regenerative Medicine, Medical Research and Education Center, Lomonosov Moscow State University, Moscow, Russia
| |
Collapse
|
9
|
Chatzikalil E, Stergiou IE, Papadakos SP, Konstantinidis I, Theocharis S. The Clinical Relevance of the EPH/Ephrin Signaling Pathway in Pediatric Solid and Hematologic Malignancies. Int J Mol Sci 2024; 25:3834. [PMID: 38612645 PMCID: PMC11011407 DOI: 10.3390/ijms25073834] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2024] [Revised: 03/26/2024] [Accepted: 03/26/2024] [Indexed: 04/14/2024] Open
Abstract
Pediatric neoplasms represent a complex group of malignancies that pose unique challenges in terms of diagnosis, treatment, and understanding of the underlying molecular pathogenetic mechanisms. Erythropoietin-producing hepatocellular receptors (EPHs), the largest family of receptor tyrosine kinases and their membrane-tethered ligands, ephrins, orchestrate short-distance cell-cell signaling and are intricately involved in cell-pattern morphogenesis and various developmental processes. Unraveling the role of the EPH/ephrin signaling pathway in the pathophysiology of pediatric neoplasms and its clinical implications can contribute to deciphering the intricate landscape of these malignancies. The bidirectional nature of the EPH/ephrin axis is underscored by emerging evidence revealing its capacity to drive tumorigenesis, fostering cell-cell communication within the tumor microenvironment. In the context of carcinogenesis, the EPH/ephrin signaling pathway prompts a reevaluation of treatment strategies, particularly in pediatric oncology, where the modest progress in survival rates and enduring treatment toxicity necessitate novel approaches. Molecularly targeted agents have emerged as promising alternatives, prompting a shift in focus. Through a nuanced understanding of the pathway's intricacies, we aim to lay the groundwork for personalized diagnostic and therapeutic strategies, ultimately contributing to improved outcomes for young patients grappling with neoplastic challenges.
Collapse
Affiliation(s)
- Elena Chatzikalil
- Division of Pediatric Hematology-Oncology, First Department of Pediatrics, School of Medicine, National and Kapodistrian University of Athens, 11527 Athens, Greece;
| | - Ioanna E. Stergiou
- Department of Pathophysiology, School of Medicine, National and Kapodistrian University of Athens, 11527 Athens, Greece;
| | - Stavros P. Papadakos
- First Department of Pathology, School of Medicine, National and Kapodistrian University of Athens, 11527 Athens, Greece;
| | | | - Stamatios Theocharis
- First Department of Pathology, School of Medicine, National and Kapodistrian University of Athens, 11527 Athens, Greece;
| |
Collapse
|
10
|
Shiadeh SMJ, Goretta F, Svedin P, Jansson T, Mallard C, Ardalan M. Long-term impact of maternal obesity on the gliovascular unit and ephrin signaling in the hippocampus of adult offspring. J Neuroinflammation 2024; 21:39. [PMID: 38308309 PMCID: PMC10837922 DOI: 10.1186/s12974-024-03030-w] [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: 11/10/2023] [Accepted: 01/24/2024] [Indexed: 02/04/2024] Open
Abstract
BACKGROUND Children born to obese mothers are at increased risk of developing mood disorders and cognitive impairment. Experimental studies have reported structural changes in the brain such as the gliovascular unit as well as activation of neuroinflammatory cells as a part of neuroinflammation processing in aged offspring of obese mothers. However, the molecular mechanisms linking maternal obesity to poor neurodevelopmental outcomes are not well established. The ephrin system plays a major role in a variety of cellular processes including cell-cell interaction, synaptic plasticity, and long-term potentiation. Therefore, in this study we determined the impact of maternal obesity in pregnancy on cortical, hippocampal development, vasculature and ephrin-A3/EphA4-signaling, in the adult offspring in mice. METHODS Maternal obesity was induced in mice by a high fat/high sugar Western type of diet (HF/HS). We collected brain tissue (prefrontal cortex and hippocampus) from 6-month-old offspring of obese and lean (control) dams. Hippocampal volume, cortical thickness, myelination of white matter, density of astrocytes and microglia in relation to their activity were analyzed using 3-D stereological quantification. mRNA expression of ephrin-A3, EphA4 and synaptic markers were measured by qPCR in the brain tissue. Moreover, expression of gap junction protein connexin-43, lipocalin-2, and vascular CD31/Aquaporin 4 were determined in the hippocampus by immunohistochemistry. RESULTS Volume of hippocampus and cortical thickness were significantly smaller, and myelination impaired, while mRNA levels of hippocampal EphA4 and post-synaptic density (PSD) 95 were significantly lower in the hippocampus in the offspring of obese dams as compared to offspring of controls. Further analysis of the hippocampal gliovascular unit indicated higher coverage of capillaries by astrocytic end-feet, expression of connexin-43 and lipocalin-2 in endothelial cells in the offspring of obese dams. In addition, offspring of obese dams demonstrated activation of microglia together with higher density of cells, while astrocyte cell density was lower. CONCLUSION Maternal obesity affects brain size, impairs myelination, disrupts the hippocampal gliovascular unit and decreases the mRNA expression of EphA4 and PSD-95 in the hippocampus of adult offspring. These results indicate that the vasculature-glia cross-talk may be an important mediator of altered synaptic plasticity, which could be a link between maternal obesity and neurodevelopmental/neuropsychiatric disorders in the offspring.
Collapse
Affiliation(s)
- Seyedeh Marziyeh Jabbari Shiadeh
- Department of Physiology, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Department of Clinical Medicine, Translational Neuropsychiatry Unit, Aarhus University, Aarhus, Denmark
| | - Fanny Goretta
- Department of Physiology, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Pernilla Svedin
- Department of Physiology, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Thomas Jansson
- Division of Reproductive Sciences, Department of OB/GYN, University of Colorado, Anschutz Medical Campus, Aurora, CO, USA
| | - Carina Mallard
- Department of Physiology, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Maryam Ardalan
- Department of Physiology, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden.
- Department of Clinical Medicine, Translational Neuropsychiatry Unit, Aarhus University, Aarhus, Denmark.
| |
Collapse
|
11
|
Gavrish M, Kustova A, Celis Suescún JC, Bessa P, Mitina N, Tarabykin V. Molecular mechanisms of corpus callosum development: a four-step journey. Front Neuroanat 2024; 17:1276325. [PMID: 38298831 PMCID: PMC10827913 DOI: 10.3389/fnana.2023.1276325] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2023] [Accepted: 12/18/2023] [Indexed: 02/02/2024] Open
Abstract
The Corpus Callosum (CC) is a bundle of axons connecting the cerebral hemispheres. It is the most recent structure to have appeared during evolution of placental mammals. Its development is controlled by a very complex interplay of many molecules. In humans it contains almost 80% of all commissural axons in the brain. The formation of the CC can be divided into four main stages, each controlled by numerous intracellular and extracellular molecular factors. First, a newborn neuron has to specify an axon, leave proliferative compartments, the Ventricular Zone (VZ) and Subventricular Zone (SVZ), migrate through the Intermediate Zone (IZ), and then settle at the Cortical Plate (CP). During the second stage, callosal axons navigate toward the midline within a compact bundle. Next stage is the midline crossing into contralateral hemisphere. The last step is targeting a defined area and synapse formation. This review provides an insight into these four phases of callosal axons development, as well as a description of the main molecular players involved.
Collapse
Affiliation(s)
- Maria Gavrish
- Laboratory of Genetics of Brain Development, Research Institute of Neurosciences, Lobachevsky State University of Nizhny Novgorod, Nizhny Novgorod, Russia
| | - Angelina Kustova
- Laboratory of Genetics of Brain Development, Research Institute of Neurosciences, Lobachevsky State University of Nizhny Novgorod, Nizhny Novgorod, Russia
| | - Juan C. Celis Suescún
- Laboratory of Genetics of Brain Development, Research Institute of Neurosciences, Lobachevsky State University of Nizhny Novgorod, Nizhny Novgorod, Russia
| | - Paraskevi Bessa
- Charité Hospital, Institute of Cell Biology and Neurobiology, Berlin, Germany
| | - Natalia Mitina
- Laboratory of Genetics of Brain Development, Research Institute of Neurosciences, Lobachevsky State University of Nizhny Novgorod, Nizhny Novgorod, Russia
| | - Victor Tarabykin
- Charité Hospital, Institute of Cell Biology and Neurobiology, Berlin, Germany
| |
Collapse
|
12
|
Dalton GD, Siecinski SK, Nikolova VD, Cofer GP, Hornburg K, Qi Y, Johnson GA, Jiang YH, Moy SS, Gregory SG. Transcriptome Analysis Identifies An ASD-Like Phenotype In Oligodendrocytes And Microglia From C58/J Amygdala That Is Dependent On Sex and Sociability. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.01.15.575733. [PMID: 38293238 PMCID: PMC10827122 DOI: 10.1101/2024.01.15.575733] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/01/2024]
Abstract
Background Autism Spectrum Disorder (ASD) is a group of neurodevelopmental disorders with higher incidence in males and is characterized by atypical verbal/nonverbal communication, restricted interests that can be accompanied by repetitive behavior, and disturbances in social behavior. This study investigated brain mechanisms that contribute to sociability deficits and sex differences in an ASD animal model. Methods Sociability was measured in C58/J and C57BL/6J mice using the 3-chamber social choice test. Bulk RNA-Seq and snRNA-Seq identified transcriptional changes in C58/J and C57BL/6J amygdala within which DMRseq was used to measure differentially methylated regions in amygdala. Results C58/J mice displayed divergent social strata in the 3-chamber test. Transcriptional and pathway signatures revealed immune-related biological processes differ between C58/J and C57BL/6J amygdala. Hypermethylated and hypomethylated genes were identified in C58/J versus C57BL/6J amygdala. snRNA-Seq data in C58/J amygdala identified differential transcriptional signatures within oligodendrocytes and microglia characterized by increased ASD risk gene expression and predicted impaired myelination that was dependent on sex and sociability. RNA velocity, gene regulatory network, and cell communication analysis showed diminished oligodendrocyte/microglia differentiation. Findings were verified using bulk RNA-Seq and demonstrated oxytocin's beneficial effects on myelin gene expression. Limitations Our findings are significant. However, limitations can be noted. The cellular mechanisms linking reduced oligodendrocyte differentiation and reduced myelination to an ASD phenotype in C58/J mice need further investigation. Additional snRNA-Seq and spatial studies would determine if effects in oligodendrocytes/microglia are unique to amygdala or if this occurs in other brain regions. Oxytocin's effects need further examination to understand its potential as an ASD therapeutic. Conclusions Our work demonstrates the C58/J mouse model's utility in evaluating the influence of sex and sociability on the transcriptome in concomitant brain regions involved in ASD. Our single-nucleus transcriptome analysis elucidates potential pathological roles of oligodendrocytes and microglia in ASD. This investigation provides details regarding regulatory features disrupted in these cell types, including transcriptional gene dysregulation, aberrant cell differentiation, altered gene regulatory networks, and changes to key pathways that promote microglia/oligodendrocyte differentiation. Our studies provide insight into interactions between genetic risk and epigenetic processes associated with divergent affiliative behavior and lack of positive sociability.
Collapse
|
13
|
Calaf GM, Roy D, Jara L, Aguayo F, Crispin LA. Gene Signature Associated with Nervous System in an Experimental Radiation- and Estrogen-Induced Breast Cancer Model. Biomedicines 2023; 11:3111. [PMID: 38137332 PMCID: PMC10740914 DOI: 10.3390/biomedicines11123111] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2023] [Revised: 10/17/2023] [Accepted: 11/16/2023] [Indexed: 12/24/2023] Open
Abstract
Breast cancer is frequently the most diagnosed female cancer in the world. The experimental studies on cancer seldom focus on the relationship between the central nervous system and cancer. Despite extensive research into the treatment of breast cancer, chemotherapy resistance is an important issue limiting the efficacy of treatment. Novel biomarkers to predict prognosis or sensitivity to chemotherapy are urgently needed. This study examined nervous-system-related genes. The profiling of differentially expressed genes indicated that high-LET radiation, such as that emitted by radon progeny, in the presence of estrogen, induced a cascade of events indicative of tumorigenicity in human breast epithelial cells. Bioinformatic tools allowed us to analyze the genes involved in breast cancer and associated with the nervous system. The results indicated that the gene expression of the Ephrin A1 gene (EFNA1), the roundabout guidance receptor 1 (ROBO1), and the kallikrein-related peptidase 6 (KLK6) was greater in T2 and A5 than in the A3 cell line; the LIM domain kinase 2 gene (LIMK2) was greater in T2 than A3 and A5; the kallikrein-related peptidase 7 (KLK7), the neuroligin 4 X-linked gene (NLGN4X), and myelin basic protein (MBP) were greater than A3 only in T2; and the neural precursor cell expressed, developmentally down-regulated 9 gene (NEDD9) was greater in A5 than in the A3 and E cell lines. Concerning the correlation, it was found a positive correlation between ESR1 and EFNA1 in BRCA-LumA patients; with ROBO1 in BRCA-Basal patients, but this correlation was negative with the kallikrein-related peptidase 6 (KLK6) in BRCA-LumA and -LumB, as well as with LIMK2 and ROBO1 in all BRCA. It was also positive with neuroligin 4 X-linked (NLGN4X) in BRCA-Her2 and BRCA-LumB, and with MBP in BRCA-LumA and -LumB, but negative with KLK7 in all BRCA and BRCA-LumA and NEDD9 in BRCA-Her2. The differential gene expression levels between the tumor and adjacent tissue indicated that the ROBO1, KLK6, LIMK2, KLK7, NLGN4X, MBP, and NEDD9 gene expression levels were higher in normal tissues than in tumors; however, EFNA1 was higher in the tumor than the normal ones. EFNA1, LIMK2, ROBO1, KLK6, KLK7, and MBP gene expression had a negative ER status, whereas NEDD9 and NLGN4X were not significant concerning ER status. In conclusion, important markers have been analyzed concerning genes related to the nervous system, opening up a new avenue of studies in breast cancer therapy.
Collapse
Affiliation(s)
- Gloria M. Calaf
- Instituto de Alta Investigación, Universidad de Tarapacá, Arica 1000000, Chile;
| | - Debasish Roy
- Department of Natural Sciences, Hostos College of the City University of New York, Bronx, NY 10451, USA;
| | - Lilian Jara
- Laboratorio de Genética Humana, Programa de Genética Humana, Instituto de Ciencias Biomédicas (ICBM), Facultad de Medicina, Universidad de Chile, Santiago 8380000, Chile;
| | - Francisco Aguayo
- Laboratorio de Oncovirología, Programa de Virología, Instituto de Ciencias Biomédicas (ICBM), Facultad de Medicina, Universidad de Chile, Santiago 8380000, Chile;
| | - Leodan A. Crispin
- Instituto de Alta Investigación, Universidad de Tarapacá, Arica 1000000, Chile;
| |
Collapse
|
14
|
Yan K, Bormuth I, Bormuth O, Tutukova S, Renner A, Bessa P, Schaub T, Rosário M, Tarabykin V. TrkB-dependent EphrinA reverse signaling regulates callosal axon fasciculate growth downstream of Neurod2/6. Cereb Cortex 2023; 33:1752-1767. [PMID: 35462405 DOI: 10.1093/cercor/bhac170] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2017] [Revised: 04/04/2022] [Accepted: 04/05/2022] [Indexed: 11/14/2022] Open
Abstract
Abnormal development of corpus callosum is relatively common and causes a broad spectrum of cognitive impairments in humans. We use acallosal Neurod2/6-deficient mice to study callosal axon guidance within the ipsilateral cerebral cortex. Initial callosal tracts form but fail to traverse the ipsilateral cingulum and are not attracted towards the midline in the absence of Neurod2/6. We show that the restoration of Ephrin-A4 (EfnA4) expression in the embryonic neocortex of Neurod2/6-deficient embryos is sufficient to partially rescue targeted callosal axon growth towards the midline. EfnA4 cannot directly mediate reverse signaling within outgrowing axons, but it forms co-receptor complexes with TrkB (Ntrk2). The ability of EfnA4 to rescue the guided growth of a subset of callosal axons in Neurod2/6-deficient mice is abolished by the co-expression of dominant negative TrkBK571N (kinase-dead) or TrkBY515F (SHC-binding deficient) variants, but not by TrkBY816F (PLCγ1-binding deficient). Additionally, EphA4 is repulsive to EfnA4-positive medially projecting axons in organotypic brain slice culture. Collectively, we suggest that EfnA4-mediated reverse signaling acts via TrkB-SHC and is required for ipsilateral callosal axon growth accuracy towards the midline downstream of Neurod family factors.
Collapse
Affiliation(s)
- Kuo Yan
- Institute of Cell Biology and Neurobiology, Charité - Universitätsmedizin Berlin, D-10117, Berlin, Germany
| | - Ingo Bormuth
- Institute of Cell Biology and Neurobiology, Charité - Universitätsmedizin Berlin, D-10117, Berlin, Germany
| | - Olga Bormuth
- Institute of Cell Biology and Neurobiology, Charité - Universitätsmedizin Berlin, D-10117, Berlin, Germany.,Institute of Neuroscience, Lobachevsky State University of Nizhny Novgorod, 603950, Nizhny Novgorod Oblast, Russia
| | - Svetlana Tutukova
- Institute of Neuroscience, Lobachevsky State University of Nizhny Novgorod, 603950, Nizhny Novgorod Oblast, Russia.,Research Institute of Medical Genetics, Tomsk National Research Medical Center of the Russian Academy of Sciences, 634009, Tomsk, Russia
| | - Ana Renner
- Institute of Cell Biology and Neurobiology, Charité - Universitätsmedizin Berlin, D-10117, Berlin, Germany
| | - Paraskevi Bessa
- Institute of Cell Biology and Neurobiology, Charité - Universitätsmedizin Berlin, D-10117, Berlin, Germany
| | - Theres Schaub
- Institute of Cell Biology and Neurobiology, Charité - Universitätsmedizin Berlin, D-10117, Berlin, Germany
| | - Marta Rosário
- Institute of Cell Biology and Neurobiology, Charité - Universitätsmedizin Berlin, D-10117, Berlin, Germany
| | - Victor Tarabykin
- Institute of Cell Biology and Neurobiology, Charité - Universitätsmedizin Berlin, D-10117, Berlin, Germany.,Institute of Neuroscience, Lobachevsky State University of Nizhny Novgorod, 603950, Nizhny Novgorod Oblast, Russia.,Research Institute of Medical Genetics, Tomsk National Research Medical Center of the Russian Academy of Sciences, 634009, Tomsk, Russia
| |
Collapse
|
15
|
Systems level analysis of sex-dependent gene expression changes in Parkinson's disease. NPJ Parkinsons Dis 2023; 9:8. [PMID: 36681675 PMCID: PMC9867746 DOI: 10.1038/s41531-023-00446-8] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2022] [Accepted: 01/03/2023] [Indexed: 01/22/2023] Open
Abstract
Parkinson's disease (PD) is a heterogeneous disorder, and among the factors which influence the symptom profile, biological sex has been reported to play a significant role. While males have a higher age-adjusted disease incidence and are more frequently affected by muscle rigidity, females present more often with disabling tremors. The molecular mechanisms involved in these differences are still largely unknown, and an improved understanding of the relevant factors may open new avenues for pharmacological disease modification. To help address this challenge, we conducted a meta-analysis of disease-associated molecular sex differences in brain transcriptomics data from case/control studies. Both sex-specific (alteration in only one sex) and sex-dimorphic changes (changes in both sexes, but with opposite direction) were identified. Using further systems level pathway and network analyses, coordinated sex-related alterations were studied. These analyses revealed significant disease-associated sex differences in mitochondrial pathways and highlight specific regulatory factors whose activity changes can explain downstream network alterations, propagated through gene regulatory cascades. Single-cell expression data analyses confirmed the main pathway-level changes observed in bulk transcriptomics data. Overall, our analyses revealed significant sex disparities in PD-associated transcriptomic changes, resulting in coordinated modulations of molecular processes. Among the regulatory factors involved, NR4A2 has already been reported to harbor rare mutations in familial PD and its pharmacological activation confers neuroprotective effects in toxin-induced models of Parkinsonism. Our observations suggest that NR4A2 may warrant further research as a potential adjuvant therapeutic target to address a subset of pathological molecular features of PD that display sex-associated profiles.
Collapse
|
16
|
Schwend T. Wiring the ocular surface: A focus on the comparative anatomy and molecular regulation of sensory innervation of the cornea. Differentiation 2023:S0301-4681(23)00010-5. [PMID: 36997455 DOI: 10.1016/j.diff.2023.01.002] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Accepted: 01/23/2023] [Indexed: 01/29/2023]
Abstract
The cornea is richly innervated with sensory nerves that function to detect and clear harmful debris from the surface of the eye, promote growth and survival of the corneal epithelium and hasten wound healing following ocular disease or trauma. Given their importance to eye health, the neuroanatomy of the cornea has for many years been a source of intense investigation. Resultantly, complete nerve architecture maps exist for adult human and many animal models and these maps reveal few major differences across species. Interestingly, recent work has revealed considerable variation across species in how sensory nerves are acquired during developmental innervation of the cornea. Highlighting such species-distinct key differences, but also similarities, this review provides a full, comparative anatomy analysis of sensory innervation of the cornea for all species studied to date. Further, this article comprehensively describes the molecules that have been shown to guide and direct nerves toward, into and through developing corneal tissue as the final architectural pattern of the cornea's neuroanatomy is established. Such knowledge is useful for researchers and clinicians seeking to better understand the anatomical and molecular basis of corneal nerve pathologies and to hasten neuro-regeneration following infection, trauma or surgery that damage the ocular surface and its corneal nerves.
Collapse
|
17
|
Krasewicz J, Yu WM. Eph and ephrin signaling in the development of the central auditory system. Dev Dyn 2023; 252:10-26. [PMID: 35705527 PMCID: PMC9751234 DOI: 10.1002/dvdy.506] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2022] [Revised: 06/10/2022] [Accepted: 06/12/2022] [Indexed: 01/17/2023] Open
Abstract
Acoustic communication relies crucially on accurate interpretation of information about the intensity, frequency, timing, and location of diverse sound stimuli in the environment. To meet this demand, neurons along different levels of the auditory system form precisely organized neural circuits. The assembly of these precise circuits requires tight regulation and coordination of multiple developmental processes. Several groups of axon guidance molecules have proven critical in controlling these processes. Among them, the family of Eph receptors and their ephrin ligands emerge as one group of key players. They mediate diverse functions at multiple levels of the auditory pathway, including axon guidance and targeting, topographic map formation, as well as cell migration and tissue pattern formation. Here, we review our current knowledge of how Eph and ephrin molecules regulate different processes in the development and maturation of central auditory circuits.
Collapse
Affiliation(s)
| | - Wei-Ming Yu
- Correspondence: Wei-Ming Yu, Department of Biology, Loyola University of Chicago, 1032 W Sheridan Rd, LSB 226, Chicago, IL 60660, , Tel: +1-773-508-3325, Fax: +1-773-508-3646
| |
Collapse
|
18
|
Chromosome-level genome and population genomics reveal evolutionary characteristics and conservation status of Chinese indigenous geese. Commun Biol 2022; 5:1191. [DOI: 10.1038/s42003-022-04125-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2021] [Accepted: 10/18/2022] [Indexed: 11/09/2022] Open
Abstract
AbstractGeese are herbivorous birds that play an essential role in the agricultural economy. We construct the chromosome-level genome of a Chinese indigenous goose (the Xingguo gray goose, XGG; Anser cygnoides) and analyze the adaptation of fat storage capacity in the goose liver during the evolution of Anatidae. Genomic resequencing of 994 geese is used to investigate the genetic relationships of geese, which supports the dual origin of geese (Anser cygnoides and Anser anser). Chinese indigenous geese show higher genetic diversity than European geese, and a scientific conservation program can be established to preserve genetic variation for each breed. We also find that a 14-bp insertion in endothelin receptor B subtype 2 (EDNRB2) that determines the white plumage of Chinese domestic geese is a natural mutation, and the linkaged alleles rapidly increase in frequency as a result of genetic hitchhiking, leading to the formation of completely different haplotypes of white geese under strong artificial selection. These genomic resources and our findings will facilitate marker-assisted breeding of geese and provide a foundation for further research on geese genetics and evolution.
Collapse
|
19
|
Wolff DW, Deng Z, Bianchi-Smiraglia A, Foley CE, Han Z, Wang X, Shen S, Rosenberg MM, Moparthy S, Yun DH, Chen J, Baker BK, Roll MV, Magiera AJ, Li J, Hurley E, Feltri ML, Cox AO, Lee J, Furdui CM, Liu L, Bshara W, LaConte LE, Kandel ES, Pasquale EB, Qu J, Hedstrom L, Nikiforov MA. Phosphorylation of guanosine monophosphate reductase triggers a GTP-dependent switch from pro- to anti-oncogenic function of EPHA4. Cell Chem Biol 2022; 29:970-984.e6. [PMID: 35148834 PMCID: PMC9620470 DOI: 10.1016/j.chembiol.2022.01.007] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2021] [Revised: 11/19/2021] [Accepted: 01/11/2022] [Indexed: 12/11/2022]
Abstract
Signal transduction pathways post-translationally regulating nucleotide metabolism remain largely unknown. Guanosine monophosphate reductase (GMPR) is a nucleotide metabolism enzyme that decreases GTP pools by converting GMP to IMP. We observed that phosphorylation of GMPR at Tyr267 is critical for its activity and found that this phosphorylation by ephrin receptor tyrosine kinase EPHA4 decreases GTP pools in cell protrusions and levels of GTP-bound RAC1. EPHs possess oncogenic and tumor-suppressor activities, although the mechanisms underlying switches between these two modes are poorly understood. We demonstrated that GMPR plays a key role in EPHA4-mediated RAC1 suppression. This supersedes GMPR-independent activation of RAC1 by EPHA4, resulting in a negative overall effect on melanoma cell invasion and tumorigenicity. Accordingly, EPHA4 levels increase during melanoma progression and inversely correlate with GMPR levels in individual melanoma tumors. Therefore, phosphorylation of GMPR at Tyr267 is a metabolic signal transduction switch controlling GTP biosynthesis and transformed phenotypes.
Collapse
Affiliation(s)
- David W. Wolff
- Department of Biomedical Engineering, Pratt School of Engineering, Duke University, Durham, NC 27708, USA,Department of Cancer Biology, Wake Forest School of Medicine, Winston Salem, NC 27157, USA
| | - Zhiyong Deng
- Department of Cancer Biology, Wake Forest School of Medicine, Winston Salem, NC 27157, USA
| | - Anna Bianchi-Smiraglia
- Department of Cell Stress Biology, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14203, USA
| | - Colleen E. Foley
- Department of Cell Stress Biology, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14203, USA
| | - Zhannan Han
- Department of Biomedical Engineering, Pratt School of Engineering, Duke University, Durham, NC 27708, USA,Department of Cancer Biology, Wake Forest School of Medicine, Winston Salem, NC 27157, USA
| | - Xingyou Wang
- Department of Chemistry, Brandeis University, Waltham, MA 02453, USA
| | - Shichen Shen
- Department of Pharmaceutical Sciences, University at Buffalo, Buffalo, NY 14214, USA
| | | | - Sudha Moparthy
- Department of Cancer Biology, Wake Forest School of Medicine, Winston Salem, NC 27157, USA
| | - Dong Hyun Yun
- Department of Cancer Biology, Wake Forest School of Medicine, Winston Salem, NC 27157, USA
| | - Jialin Chen
- Department of Biomedical Engineering, Pratt School of Engineering, Duke University, Durham, NC 27708, USA,Department of Cancer Biology, Wake Forest School of Medicine, Winston Salem, NC 27157, USA
| | - Brian K. Baker
- Department of Cancer Biology, Wake Forest School of Medicine, Winston Salem, NC 27157, USA
| | - Matthew V. Roll
- Department of Biomedical Engineering, Pratt School of Engineering, Duke University, Durham, NC 27708, USA,Department of Cancer Biology, Wake Forest School of Medicine, Winston Salem, NC 27157, USA
| | - Andrew J. Magiera
- Department of Cancer Biology, Wake Forest School of Medicine, Winston Salem, NC 27157, USA
| | - Jun Li
- Department of Pharmaceutical Sciences, University at Buffalo, Buffalo, NY 14214, USA
| | - Edward Hurley
- Department of Biochemistry and Neurology, Hunter James Kelly Research Institute, University at Buffalo, Buffalo NY, USA
| | - Maria Laura Feltri
- Department of Biochemistry and Neurology, Hunter James Kelly Research Institute, University at Buffalo, Buffalo NY, USA
| | - Anderson O. Cox
- Department of Internal Medicine, Section of Molecular Medicine, Wake Forest School of Medicine, Winston-Salem NC, USA
| | - Jingyun Lee
- Department of Internal Medicine, Section of Molecular Medicine, Wake Forest School of Medicine, Winston-Salem NC, USA
| | - Cristina M. Furdui
- Department of Internal Medicine, Section of Molecular Medicine, Wake Forest School of Medicine, Winston-Salem NC, USA
| | - Liang Liu
- Department of Cancer Biology, Wake Forest School of Medicine, Winston Salem, NC 27157, USA
| | - Wiam Bshara
- Department of Pathology, Roswell Park Comprehensive Cancer Center, Buffalo NY 14203, USA
| | - Leslie E.W. LaConte
- Fralin Biomedical Research Institute at Virginia Tech Carilion School of Medicine, Roanoke, VA 24016, USA
| | - Eugene S. Kandel
- Department of Cell Stress Biology, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14203, USA
| | - Elena B. Pasquale
- Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA 92037, USA
| | - Jun Qu
- Department of Chemistry, Brandeis University, Waltham, MA 02453, USA
| | - Lizbeth Hedstrom
- Department of Chemistry, Brandeis University, Waltham, MA 02453, USA,Department of Biology, Brandeis University, Waltham, MA 02453, USA
| | - Mikhail A. Nikiforov
- Department of Biomedical Engineering, Pratt School of Engineering, Duke University, Durham, NC 27708, USA,Department of Cancer Biology, Wake Forest School of Medicine, Winston Salem, NC 27157, USA,Department of Pathology, Duke University School of Medicine, Durham, NC 27710, USA,Corresponding author and lead contact: Mikhail A. Nikiforov,
| |
Collapse
|
20
|
Wu J, Zhu ZY, Fan ZW, Chen Y, Yang RY, Li Y. Downregulation of EphB2 by RNA interference attenuates glial/fibrotic scar formation and promotes axon growth. Neural Regen Res 2022; 17:362-369. [PMID: 34269211 PMCID: PMC8463997 DOI: 10.4103/1673-5374.317988] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/02/2022] Open
Abstract
The rapid formation of a glial/fibrotic scar is one of the main factors hampering axon growth after spinal cord injury. The bidirectional EphB2/ephrin-B2 signaling of the fibroblast-astrocyte contact-dependent interaction is a trigger for glial/fibrotic scar formation. In the present study, a new in vitro model was produced by coculture of fibroblasts and astrocytes wounded by scratching to mimic glial/fibrotic scar-like structures using an improved slide system. After treatment with RNAi to downregulate EphB2, changes in glial/fibrotic scar formation and the growth of VSC4.1 motoneuron axons were examined. Following RNAi treatment, fibroblasts and astrocytes dispersed without forming a glial/fibrotic scar-like structure. Furthermore, the expression levels of neurocan, NG2 and collagen I in the coculture were reduced, and the growth of VSC4.1 motoneuron axons was enhanced. These findings suggest that suppression of EphB2 expression by RNAi attenuates the formation of a glial/fibrotic scar and promotes axon growth. This study was approved by the Laboratory Animal Ethics Committee of Jiangsu Province, China (approval No. 2019-0506-002) on May 6, 2019.
Collapse
Affiliation(s)
- Jian Wu
- Department of Histology and Embryology, Medical College, Nantong University, Nantong, Jiangsu Province, China
| | - Zhen-Yu Zhu
- Department of Histology and Embryology, Medical College, Nantong University, Nantong, Jiangsu Province, China
| | - Zhi-Wei Fan
- Department of Histology and Embryology, Medical College, Nantong University, Nantong, Jiangsu Province, China
| | - Ying Chen
- Department of Histology and Embryology, Medical College, Nantong University, Nantong, Jiangsu Province, China
| | - Ri-Yun Yang
- Department of Histology and Embryology, Medical College, Nantong University, Nantong, Jiangsu Province, China
| | - Yi Li
- Department of Histology and Embryology, Medical College, Nantong University, Nantong, Jiangsu Province, China
| |
Collapse
|
21
|
Hoshino N, Altarshan Y, Alzein A, Fernando AM, Nguyen HT, Majewski EF, Chen VCF, William Rochlin M, Yu WM. Ephrin-A3 is required for tonotopic map precision and auditory functions in the mouse auditory brainstem. J Comp Neurol 2021; 529:3633-3654. [PMID: 34235739 PMCID: PMC8490280 DOI: 10.1002/cne.25213] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2021] [Revised: 06/22/2021] [Accepted: 07/02/2021] [Indexed: 01/09/2023]
Abstract
Tonotopy is a prominent feature of the vertebrate auditory system and forms the basis for sound discrimination, but the molecular mechanism that underlies its formation remains largely elusive. Ephrin/Eph signaling is known to play important roles in axon guidance during topographic mapping in other sensory systems, so we investigated its possible role in the establishment of tonotopy in the mouse cochlear nucleus. We found that ephrin-A3 molecules are differentially expressed along the tonotopic axis in the cochlear nucleus during innervation. Ephrin-A3 forward signaling is sufficient to repel auditory nerve fibers in a developmental stage-dependent manner. In mice lacking ephrin-A3, the tonotopic map is degraded and isofrequency bands of neuronal activation upon pure tone exposure become imprecise in the anteroventral cochlear nucleus. Ephrin-A3 mutant mice also exhibit a delayed second wave in auditory brainstem responses upon sound stimuli and impaired detection of sound frequency changes. Our findings establish an essential role for ephrin-A3 in forming precise tonotopy in the auditory brainstem to ensure accurate sound discrimination.
Collapse
Affiliation(s)
- Natalia Hoshino
- Department of Biology, Loyola University of Chicago, Chicago, Illinois
| | - Yazan Altarshan
- Department of Biology, Loyola University of Chicago, Chicago, Illinois
| | - Ahmad Alzein
- Department of Biology, Loyola University of Chicago, Chicago, Illinois
| | - Amali M. Fernando
- Department of Biology, Loyola University of Chicago, Chicago, Illinois
| | - Hieu T. Nguyen
- Department of Biology, Loyola University of Chicago, Chicago, Illinois
| | - Emma F. Majewski
- Department of Biology, Loyola University of Chicago, Chicago, Illinois
| | | | | | - Wei-Ming Yu
- Department of Biology, Loyola University of Chicago, Chicago, Illinois
| |
Collapse
|
22
|
Lévy J, Schell B, Nasser H, Rachid M, Ruaud L, Couque N, Callier P, Faivre L, Marle N, Engwerda A, van Ravenswaaij-Arts CMA, Plutino M, Karmous-Benailly H, Benech C, Redon S, Boute O, Boudry Labis E, Rama M, Kuentz P, Assoumani J, Maldergem LV, Dupont C, Verloes A, Tabet AC. EPHA7 haploinsufficiency is associated with a neurodevelopmental disorder. Clin Genet 2021; 100:396-404. [PMID: 34176129 DOI: 10.1111/cge.14017] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2020] [Revised: 06/21/2021] [Accepted: 06/23/2021] [Indexed: 11/26/2022]
Abstract
Ephrin receptor and their ligands, the ephrins, are widely expressed in the developing brain. They are implicated in several developmental processes that are crucial for brain development. Deletions in genes encoding for members of the Eph/ephrin receptor family were reported in several neurodevelopmental disorders. The ephrin receptor A7 gene (EPHA7) encodes a member of ephrin receptor subfamily of the protein-tyrosine kinase family. EPHA7 plays a role in corticogenesis processes, determines brain size and shape, and is involved in development of the central nervous system. One patient only was reported so far with a de novo deletion encompassing EPHA7 in 6q16.1. We report 12 additional patients from nine unrelated pedigrees with similar deletions. The deletions were inherited in nine out of 12 patients, suggesting variable expressivity and incomplete penetrance. Four patients had tiny deletions involving only EPHA7, suggesting a critical role of EPHA7 in a neurodevelopmental disability phenotype. We provide further evidence for EPHA7 deletion as a risk factor for neurodevelopmental disorder and delineate its clinical phenotype.
Collapse
Affiliation(s)
- Jonathan Lévy
- Genetics Department, APHP, Robert-Debré University Hospital, Paris, France
| | - Bérénice Schell
- Genetics Department, APHP, Robert-Debré University Hospital, Paris, France
| | - Hala Nasser
- Genetics Department, APHP, Robert-Debré University Hospital, Paris, France
| | - Myriam Rachid
- Genetics Department, APHP, Robert-Debré University Hospital, Paris, France
| | - Lyse Ruaud
- Genetics Department, APHP, Robert-Debré University Hospital, Paris, France.,Université de Paris Medical School, Paris, France.,INSERM UMR1141, Paris University, APHP, Robert-Debré Hospital, Paris, France
| | - Nathalie Couque
- Genetics Department, APHP, Robert-Debré University Hospital, Paris, France
| | - Patrick Callier
- Centre de Génétique et Centre de référence "Anomalies du Développement et Syndromes Malformatifs", Hôpital d'Enfants, Centre Hospitalier Universitaire de Dijon, Dijon, France.,Laboratoire de Génétique Chromosomique et Moléculaire, Plateau Technique de Biologie, Centre Hospitalier Universitaire de Dijon, Dijon, France.,UMR-Inserm 1231 GAD Team, Génétique des Anomalies du développement, Université de Bourgogne Franche-Comté, Dijon, France
| | - Laurence Faivre
- Centre de Génétique et Centre de référence "Anomalies du Développement et Syndromes Malformatifs", Hôpital d'Enfants, Centre Hospitalier Universitaire de Dijon, Dijon, France.,UMR-Inserm 1231 GAD Team, Génétique des Anomalies du développement, Université de Bourgogne Franche-Comté, Dijon, France
| | - Nathalie Marle
- Laboratoire de Génétique Chromosomique et Moléculaire, Plateau Technique de Biologie, Centre Hospitalier Universitaire de Dijon, Dijon, France.,UMR-Inserm 1231 GAD Team, Génétique des Anomalies du développement, Université de Bourgogne Franche-Comté, Dijon, France
| | - Aafke Engwerda
- Department of Genetics, University of Groningen, University Medical Centre Groningen, Groningen, The Netherlands
| | | | - Morgane Plutino
- Service de Génétique Médicale, Centre Hospitalier Universitaire de Nice, Nice, France
| | | | | | - Sylvia Redon
- Laboratoire de Génétique Moléculaire et Histocompatibilité, Service de Génétique Médicale, CHRU, Brest, France
| | - Odile Boute
- CHU Lille, Clinique de Génétique "Guy Fontaine", Lille, France
| | | | - Mélanie Rama
- CHU Lille, Institut de Génétique Médicale, Lille, France
| | - Paul Kuentz
- UMR-Inserm 1231 GAD Team, Génétique des Anomalies du développement, Université de Bourgogne Franche-Comté, Dijon, France.,Génétique Biologique, PCBio, Centre Hospitalier Universitaire de Besançon, Besançon, France
| | | | - Lionel Van Maldergem
- Clinical Investigation Center 1431, INSERM, Besançon, France.,Center of Human Genetics, University of Franche-Comté, Besançon, France
| | - Céline Dupont
- Genetics Department, APHP, Robert-Debré University Hospital, Paris, France
| | - Alain Verloes
- Genetics Department, APHP, Robert-Debré University Hospital, Paris, France.,Université de Paris Medical School, Paris, France.,INSERM UMR1141, Paris University, APHP, Robert-Debré Hospital, Paris, France
| | - Anne-Claude Tabet
- Genetics Department, APHP, Robert-Debré University Hospital, Paris, France.,Neuroscience Department, Human Genetics and Cognitive Function Unit, Pasteur Institute, Paris, France
| |
Collapse
|
23
|
Raj V, Jagadish C, Gautam V. Understanding, engineering, and modulating the growth of neural networks: An interdisciplinary approach. BIOPHYSICS REVIEWS 2021; 2:021303. [PMID: 38505122 PMCID: PMC10903502 DOI: 10.1063/5.0043014] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/05/2021] [Accepted: 05/25/2021] [Indexed: 03/21/2024]
Abstract
A deeper understanding of the brain and its function remains one of the most significant scientific challenges. It not only is required to find cures for a plethora of brain-related diseases and injuries but also opens up possibilities for achieving technological wonders, such as brain-machine interface and highly energy-efficient computing devices. Central to the brain's function is its basic functioning unit (i.e., the neuron). There has been a tremendous effort to understand the underlying mechanisms of neuronal growth on both biochemical and biophysical levels. In the past decade, this increased understanding has led to the possibility of controlling and modulating neuronal growth in vitro through external chemical and physical methods. We provide a detailed overview of the most fundamental aspects of neuronal growth and discuss how researchers are using interdisciplinary ideas to engineer neuronal networks in vitro. We first discuss the biochemical and biophysical mechanisms of neuronal growth as we stress the fact that the biochemical or biophysical processes during neuronal growth are not independent of each other but, rather, are complementary. Next, we discuss how utilizing these fundamental mechanisms can enable control over neuronal growth for advanced neuroengineering and biomedical applications. At the end of this review, we discuss some of the open questions and our perspectives on the challenges and possibilities related to controlling and engineering the growth of neuronal networks, specifically in relation to the materials, substrates, model systems, modulation techniques, data science, and artificial intelligence.
Collapse
Affiliation(s)
- Vidur Raj
- Department of Electronic Materials Engineering, Research School of Physics, The Australian National University, Canberra, Australian Capital Territory 2601, Australia
| | | | - Vini Gautam
- Department of Biomedical Engineering, Faculty of Engineering and Information Technology, The University of Melbourne, Melbourne, Victoria 3010, Australia
| |
Collapse
|
24
|
Kaczmarek R, Zimmer K, Gajdzis P, Gajdzis M. The Role of Eph Receptors and Ephrins in Corneal Physiology and Diseases. Int J Mol Sci 2021; 22:ijms22094567. [PMID: 33925443 PMCID: PMC8123804 DOI: 10.3390/ijms22094567] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2021] [Revised: 04/21/2021] [Accepted: 04/26/2021] [Indexed: 11/29/2022] Open
Abstract
The cornea, while appearing to be simple tissue, is actually an extremely complex structure. In order for it to retain its biomechanical and optical properties, perfect organization of its cells is essential. Proper regeneration is especially important after injuries and in the course of various diseases. Eph receptors and ephrin are mainly responsible for the proper organization of tissues as well as cell migration and communication. In this review, we present the current state of knowledge on the role of Eph and ephrins in corneal physiology and diseases, in particular, we focused on the functions of the epithelium and endothelium. Since the role of Eph and ephrins in the angiogenesis process has been well established, we also analyzed their influence on conditions with corneal neovascularization.
Collapse
Affiliation(s)
- Radoslaw Kaczmarek
- Department of Ophthalmology, Wroclaw Medical University, 50-556 Wroclaw, Poland; (R.K.); (K.Z.)
| | - Katarzyna Zimmer
- Department of Ophthalmology, Wroclaw Medical University, 50-556 Wroclaw, Poland; (R.K.); (K.Z.)
| | - Pawel Gajdzis
- Department of Pathomorphology and Oncological Cytology, Wroclaw Medical University, 50-556 Wroclaw, Poland;
| | - Malgorzata Gajdzis
- Department of Ophthalmology, Wroclaw Medical University, 50-556 Wroclaw, Poland; (R.K.); (K.Z.)
- Correspondence: ; Tel.: +48-71-736-43-00
| |
Collapse
|
25
|
Golan M, Krivitsky A, Mausner-Fainberg K, Benhamou M, Vigiser I, Regev K, Kolb H, Karni A. Increased Expression of Ephrins on Immune Cells of Patients with Relapsing Remitting Multiple Sclerosis Affects Oligodendrocyte Differentiation. Int J Mol Sci 2021; 22:ijms22042182. [PMID: 33671716 PMCID: PMC7927032 DOI: 10.3390/ijms22042182] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2021] [Revised: 02/17/2021] [Accepted: 02/19/2021] [Indexed: 12/02/2022] Open
Abstract
The effect of the inflammatory response on regenerative processes in the brain is complex. This complexity is even greater when the cause of the tissue damage is an autoimmune response. Multiple sclerosis (MS) is an immune-mediated disease in which demyelination foci are formed in the central nervous system. The degree of repair through oligodendrocyte regeneration and remyelination is insufficient. Ephrins are membrane-bound ligands activating tyrosine kinase signaling proteins that are known to have an inhibitory effect on oligodendrocyte regeneration. In this study, we examined the expression of ephrins on immune cells of 43 patients with relapsing-remitting (RR) MS compared to 27 matched healthy controls (HC). We found an increased expression of ephrin-A2, -A3 and -B3, especially on T cell subpopulations. We also showed overexpression of ephrins on immune cells of patients with RR-MS that increases the forward signaling pathway and that expression of ephrins on immune cells has an inhibitory effect on the differentiation of oligodendrocyte precursor cells (OPCs) in vitro. Our study findings support the concept that the immune activity of T cells in patients with RR-MS has an inhibitory effect on the differentiation capacity of OPCs through the expression and forward signaling of ephrins.
Collapse
Affiliation(s)
- Maya Golan
- The Neuroimmunology and Multiple Sclerosis Unit, Neurology Institute, Tel Aviv Sourasky Medical Center, Tel Aviv 64239, Israel; (M.G.); (A.K.); (K.M.-F.); (M.B.); (I.V.); (K.R.); (H.K.)
| | - Avivit Krivitsky
- The Neuroimmunology and Multiple Sclerosis Unit, Neurology Institute, Tel Aviv Sourasky Medical Center, Tel Aviv 64239, Israel; (M.G.); (A.K.); (K.M.-F.); (M.B.); (I.V.); (K.R.); (H.K.)
- Sackler School of Medicine, Tel Aviv University, Tel Aviv 69978, Israel
| | - Karin Mausner-Fainberg
- The Neuroimmunology and Multiple Sclerosis Unit, Neurology Institute, Tel Aviv Sourasky Medical Center, Tel Aviv 64239, Israel; (M.G.); (A.K.); (K.M.-F.); (M.B.); (I.V.); (K.R.); (H.K.)
| | - Moshe Benhamou
- The Neuroimmunology and Multiple Sclerosis Unit, Neurology Institute, Tel Aviv Sourasky Medical Center, Tel Aviv 64239, Israel; (M.G.); (A.K.); (K.M.-F.); (M.B.); (I.V.); (K.R.); (H.K.)
- Sackler School of Medicine, Tel Aviv University, Tel Aviv 69978, Israel
| | - Ifat Vigiser
- The Neuroimmunology and Multiple Sclerosis Unit, Neurology Institute, Tel Aviv Sourasky Medical Center, Tel Aviv 64239, Israel; (M.G.); (A.K.); (K.M.-F.); (M.B.); (I.V.); (K.R.); (H.K.)
| | - Keren Regev
- The Neuroimmunology and Multiple Sclerosis Unit, Neurology Institute, Tel Aviv Sourasky Medical Center, Tel Aviv 64239, Israel; (M.G.); (A.K.); (K.M.-F.); (M.B.); (I.V.); (K.R.); (H.K.)
| | - Hadar Kolb
- The Neuroimmunology and Multiple Sclerosis Unit, Neurology Institute, Tel Aviv Sourasky Medical Center, Tel Aviv 64239, Israel; (M.G.); (A.K.); (K.M.-F.); (M.B.); (I.V.); (K.R.); (H.K.)
| | - Arnon Karni
- The Neuroimmunology and Multiple Sclerosis Unit, Neurology Institute, Tel Aviv Sourasky Medical Center, Tel Aviv 64239, Israel; (M.G.); (A.K.); (K.M.-F.); (M.B.); (I.V.); (K.R.); (H.K.)
- Sackler School of Medicine, Tel Aviv University, Tel Aviv 69978, Israel
- Sagol School of Neuroscience, Tel Aviv University, Tel Aviv 69978, Israel
- Correspondence:
| |
Collapse
|
26
|
Arthur A, Gronthos S. Eph-Ephrin Signaling Mediates Cross-Talk Within the Bone Microenvironment. Front Cell Dev Biol 2021; 9:598612. [PMID: 33634116 PMCID: PMC7902060 DOI: 10.3389/fcell.2021.598612] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2020] [Accepted: 01/15/2021] [Indexed: 12/18/2022] Open
Abstract
Skeletal integrity is maintained through the tightly regulated bone remodeling process that occurs continuously throughout postnatal life to replace old bone and to repair skeletal damage. This is maintained primarily through complex interactions between bone resorbing osteoclasts and bone forming osteoblasts. Other elements within the bone microenvironment, including stromal, osteogenic, hematopoietic, endothelial and neural cells, also contribute to maintaining skeletal integrity. Disruption of the dynamic interactions between these diverse cellular systems can lead to poor bone health and an increased susceptibility to skeletal diseases including osteopenia, osteoporosis, osteoarthritis, osteomalacia, and major fractures. Recent reports have implicated a direct role for the Eph tyrosine kinase receptors and their ephrin ligands during bone development, homeostasis and skeletal repair. These membrane-bound molecules mediate contact-dependent signaling through both the Eph receptors, termed forward signaling, and through the ephrin ligands, referred to as reverse signaling. This review will focus on Eph/ ephrin cross-talk as mediators of hematopoietic and stromal cell communication, and how these interactions contribute to blood/ bone marrow function and skeletal integrity during normal steady state or pathological conditions.
Collapse
Affiliation(s)
- Agnieszka Arthur
- Mesenchymal Stem Cell Laboratory, Faculty of Health and Medical Sciences, Adelaide Medical School, University of Adelaide, Adelaide, SA, Australia.,Precision Medicine Theme, South Australian Health and Medical Research Institute, Adelaide, SA, Australia
| | - Stan Gronthos
- Mesenchymal Stem Cell Laboratory, Faculty of Health and Medical Sciences, Adelaide Medical School, University of Adelaide, Adelaide, SA, Australia.,Precision Medicine Theme, South Australian Health and Medical Research Institute, Adelaide, SA, Australia
| |
Collapse
|
27
|
Pensold D, Gehrmann J, Pitschelatow G, Walberg A, Braunsteffer K, Reichard J, Ravaei A, Linde J, Lampert A, Costa IG, Zimmer-Bensch G. The Expression of the Cancer-Associated lncRNA Snhg15 Is Modulated by EphrinA5-Induced Signaling. Int J Mol Sci 2021; 22:1332. [PMID: 33572758 PMCID: PMC7866228 DOI: 10.3390/ijms22031332] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2020] [Revised: 01/21/2021] [Accepted: 01/26/2021] [Indexed: 12/16/2022] Open
Abstract
The Eph receptor tyrosine kinases and their respective ephrin-ligands are an important family of membrane receptors, being involved in developmental processes such as proliferation, migration, and in the formation of brain cancer such as glioma. Intracellular signaling pathways, which are activated by Eph receptor signaling, are well characterized. In contrast, it is unknown so far whether ephrins modulate the expression of lncRNAs, which would enable the transduction of environmental stimuli into our genome through a great gene regulatory spectrum. Applying a combination of functional in vitro assays, RNA sequencing, and qPCR analysis, we found that the proliferation and migration promoting stimulation of mouse cerebellar granule cells (CB) with ephrinA5 diminishes the expression of the cancer-related lncRNA Snhg15. In a human medulloblastoma cell line (DAOY) ephrinA5 stimulation similarly reduced SNHG15 expression. Computational analysis identified triple-helix-mediated DNA-binding sites of Snhg15 in promoters of genes found up-regulated upon ephrinA5 stimulation and known to be involved in tumorigenic processes. Our findings propose a crucial role of Snhg15 downstream of ephrinA5-induced signaling in regulating gene transcription in the nucleus. These findings could be potentially relevant for the regulation of tumorigenic processes in the context of glioma.
Collapse
Affiliation(s)
- Daniel Pensold
- Division of Functional Epigenetics, Institute of Zoology (Biology 2), RWTH Aachen University, Worringerweg 3, 52074 Aachen, Germany; (D.P.); (G.P.); (A.W.); (K.B.); (J.R.); (J.L.)
| | - Julia Gehrmann
- RWTH Aachen Medical Faculty, Institute for Computational Genomics, 52074 Aachen, Germany; (J.G.); (I.G.C.)
| | - Georg Pitschelatow
- Division of Functional Epigenetics, Institute of Zoology (Biology 2), RWTH Aachen University, Worringerweg 3, 52074 Aachen, Germany; (D.P.); (G.P.); (A.W.); (K.B.); (J.R.); (J.L.)
| | - Asa Walberg
- Division of Functional Epigenetics, Institute of Zoology (Biology 2), RWTH Aachen University, Worringerweg 3, 52074 Aachen, Germany; (D.P.); (G.P.); (A.W.); (K.B.); (J.R.); (J.L.)
| | - Kai Braunsteffer
- Division of Functional Epigenetics, Institute of Zoology (Biology 2), RWTH Aachen University, Worringerweg 3, 52074 Aachen, Germany; (D.P.); (G.P.); (A.W.); (K.B.); (J.R.); (J.L.)
| | - Julia Reichard
- Division of Functional Epigenetics, Institute of Zoology (Biology 2), RWTH Aachen University, Worringerweg 3, 52074 Aachen, Germany; (D.P.); (G.P.); (A.W.); (K.B.); (J.R.); (J.L.)
- Research Training Group 2416 Multi Senses—Multi Scales, RWTH Aachen University, 52074 Aachen, Germany;
| | - Amin Ravaei
- Department of Neurosciences and Rehabilitation, Section of Medical Biochemistry, Molecular Biology and Genetics, University of Ferrara, 44100 Ferrara, Italy;
| | - Jenice Linde
- Division of Functional Epigenetics, Institute of Zoology (Biology 2), RWTH Aachen University, Worringerweg 3, 52074 Aachen, Germany; (D.P.); (G.P.); (A.W.); (K.B.); (J.R.); (J.L.)
- Research Training Group 2416 Multi Senses—Multi Scales, RWTH Aachen University, 52074 Aachen, Germany;
| | - Angelika Lampert
- Research Training Group 2416 Multi Senses—Multi Scales, RWTH Aachen University, 52074 Aachen, Germany;
- RWTH Aachen Medical Faculty, Institute of Physiology, 52074 Aachen, Germany
| | - Ivan G. Costa
- RWTH Aachen Medical Faculty, Institute for Computational Genomics, 52074 Aachen, Germany; (J.G.); (I.G.C.)
| | - Geraldine Zimmer-Bensch
- Division of Functional Epigenetics, Institute of Zoology (Biology 2), RWTH Aachen University, Worringerweg 3, 52074 Aachen, Germany; (D.P.); (G.P.); (A.W.); (K.B.); (J.R.); (J.L.)
- Research Training Group 2416 Multi Senses—Multi Scales, RWTH Aachen University, 52074 Aachen, Germany;
| |
Collapse
|
28
|
Thulasi Raman SN, Latreille E, Gao J, Zhang W, Wu J, Russell MS, Walrond L, Cyr T, Lavoie JR, Safronetz D, Cao J, Sauve S, Farnsworth A, Chen W, Shi PY, Wang Y, Wang L, Rosu-Myles M, Li X. Dysregulation of Ephrin receptor and PPAR signaling pathways in neural progenitor cells infected by Zika virus. Emerg Microbes Infect 2020; 9:2046-2060. [PMID: 32873194 PMCID: PMC7534353 DOI: 10.1080/22221751.2020.1818631] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2020] [Accepted: 08/28/2020] [Indexed: 12/13/2022]
Abstract
Zika virus (ZIKV) infection is a serious public threat with cases reported in about 70 countries and territories. One of the most serious consequences of ZIKV infection is congenital microcephaly in babies. Congenital microcephaly has been suggested to result from infection of neural progenitor cells (NPCs) in the developing fetal brain. However, the molecular and cellular mechanisms underlying microcephaly development remains to be fully elucidated. In this study, we employed quantitative proteomics to determine protein expression profile that occur during viral replication in NPCs. Bioinformatics analysis of the protein expression changes resulted in the identification of a wide range of cell signaling pathways. Specifically, pathways involved in neurogenesis and embryonic development were markedly altered, along with those associated with cell cycle, apoptosis, lipid metabolism and oxidative stress. Notably, the differential regulation of Ephrin Receptor and PPAR signaling pathways, as revealed by quantitative proteomics and validated by qPCR array, underscores the need to explore these pathways in disease development. Collectively, these results indicate that ZIKV-induced pathogenesis involves complex virus-host reactions; the findings reported here could help shed light on the mechanisms underlying ZIKV-induced microcephaly and ZIKV replication in NPCs.
Collapse
Affiliation(s)
- Sathya N. Thulasi Raman
- Centre for Biologics Evaluation, Biologics and Radiopharmaceutical Drugs Directorate, HPFB, Health Canada and WHO Collaborating Centre for Standardization and Evaluation of Biologicals, Ottawa, ON, Canada
| | - Elyse Latreille
- Centre for Biologics Evaluation, Biologics and Radiopharmaceutical Drugs Directorate, HPFB, Health Canada and WHO Collaborating Centre for Standardization and Evaluation of Biologicals, Ottawa, ON, Canada
| | - Jun Gao
- Centre for Biologics Evaluation, Biologics and Radiopharmaceutical Drugs Directorate, HPFB, Health Canada and WHO Collaborating Centre for Standardization and Evaluation of Biologicals, Ottawa, ON, Canada
| | - Wanyue Zhang
- Centre for Biologics Evaluation, Biologics and Radiopharmaceutical Drugs Directorate, HPFB, Health Canada and WHO Collaborating Centre for Standardization and Evaluation of Biologicals, Ottawa, ON, Canada
- Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada
| | - Jianguo Wu
- Centre for Biologics Evaluation, Biologics and Radiopharmaceutical Drugs Directorate, HPFB, Health Canada and WHO Collaborating Centre for Standardization and Evaluation of Biologicals, Ottawa, ON, Canada
| | - Marsha S. Russell
- Centre for Biologics Evaluation, Biologics and Radiopharmaceutical Drugs Directorate, HPFB, Health Canada and WHO Collaborating Centre for Standardization and Evaluation of Biologicals, Ottawa, ON, Canada
| | - Lisa Walrond
- Centre for Biologics Evaluation, Biologics and Radiopharmaceutical Drugs Directorate, HPFB, Health Canada and WHO Collaborating Centre for Standardization and Evaluation of Biologicals, Ottawa, ON, Canada
| | - Terry Cyr
- Centre for Biologics Evaluation, Biologics and Radiopharmaceutical Drugs Directorate, HPFB, Health Canada and WHO Collaborating Centre for Standardization and Evaluation of Biologicals, Ottawa, ON, Canada
| | - Jessie R. Lavoie
- Centre for Biologics Evaluation, Biologics and Radiopharmaceutical Drugs Directorate, HPFB, Health Canada and WHO Collaborating Centre for Standardization and Evaluation of Biologicals, Ottawa, ON, Canada
| | - David Safronetz
- National Microbiology Laboratory, Public Health Agency of Canada, Winnipeg, MB, Canada
| | - Jingxin Cao
- National Microbiology Laboratory, Public Health Agency of Canada, Winnipeg, MB, Canada
| | - Simon Sauve
- Centre for Biologics Evaluation, Biologics and Radiopharmaceutical Drugs Directorate, HPFB, Health Canada and WHO Collaborating Centre for Standardization and Evaluation of Biologicals, Ottawa, ON, Canada
| | - Aaron Farnsworth
- Centre for Biologics Evaluation, Biologics and Radiopharmaceutical Drugs Directorate, HPFB, Health Canada and WHO Collaborating Centre for Standardization and Evaluation of Biologicals, Ottawa, ON, Canada
| | - Wangxue Chen
- National Research Council of Canada, Human Health Therapeutics, Ottawa, ON, Canada
| | - Pei-Yong Shi
- Department of Biochemistry & Molecular Biology, University of Texas Medical Branch, Galveston, TX, USA
| | - Youchun Wang
- National Institute for Food and Drug Control and WHO Collaborating Center for Standardization and Evaluation of Biologicals, Beijing, People’s Republic of China
| | - Lisheng Wang
- Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada
| | - Michael Rosu-Myles
- Centre for Biologics Evaluation, Biologics and Radiopharmaceutical Drugs Directorate, HPFB, Health Canada and WHO Collaborating Centre for Standardization and Evaluation of Biologicals, Ottawa, ON, Canada
- Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada
| | - Xuguang Li
- Centre for Biologics Evaluation, Biologics and Radiopharmaceutical Drugs Directorate, HPFB, Health Canada and WHO Collaborating Centre for Standardization and Evaluation of Biologicals, Ottawa, ON, Canada
- Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada
| |
Collapse
|
29
|
Ankamreddy H, Bok J, Groves AK. Uncovering the secreted signals and transcription factors regulating the development of mammalian middle ear ossicles. Dev Dyn 2020; 249:1410-1424. [PMID: 33058336 DOI: 10.1002/dvdy.260] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2020] [Revised: 10/11/2020] [Accepted: 10/11/2020] [Indexed: 12/22/2022] Open
Abstract
The mammalian middle ear comprises a chain of ossicles, the malleus, incus, and stapes that act as an impedance matching device during the transmission of sound from the tympanic membrane to the inner ear. These ossicles are derived from cranial neural crest cells that undergo endochondral ossification and subsequently differentiate into their final functional forms. Defects that occur during middle ear development can result in conductive hearing loss. In this review, we summarize studies describing the crucial roles played by signaling molecules such as sonic hedgehog, bone morphogenetic proteins, fibroblast growth factors, notch ligands, and chemokines during the differentiation of neural crest into the middle ear ossicles. In addition to these cell-extrinsic signals, we also discuss studies on the function of transcription factor genes such as Foxi3, Tbx1, Bapx1, Pou3f4, and Gsc in regulating the development and morphology of the middle ear ossicles.
Collapse
Affiliation(s)
| | - Jinwoong Bok
- Department of Anatomy, Yonsei University College of Medicine, Seoul, South Korea.,Department of Otorhinolaryngology, Yonsei University College of Medicine, Seoul, South Korea
| | - Andrew K Groves
- Department of Neuroscience, Baylor College of Medicine, Houston, Texas, USA.,Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas, USA
| |
Collapse
|
30
|
Berlandi J, Chaouch A, De Jay N, Tegeder I, Thiel K, Shirinian M, Kleinman CL, Jeibmann A, Lasko P, Jabado N, Hasselblatt M. Identification of genes functionally involved in the detrimental effects of mutant histone H3.3-K27M in Drosophila melanogaster. Neuro Oncol 2020; 21:628-639. [PMID: 30715493 DOI: 10.1093/neuonc/noz021] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Recurrent specific mutations in evolutionarily conserved histone 3 (H3) variants drive pediatric high-grade gliomas (HGGs), but little is known about their downstream effects. The aim of this study was to identify genes involved in the detrimental effects of mutant H3.3-K27M, the main genetic driver in lethal midline HGG, in a transgenic Drosophila model. METHODS Mutant and wild-type histone H3.3-expressing flies were generated using a φC31-based integration system. Genetic modifier screens were performed by crossing H3.3-K27M expressing driver strains and 194 fly lines expressing short hairpin RNA targeting genes selected based on their potential role in the detrimental effects of mutant H3. Expression of the human orthologues of genes with functional relevance in the fly model was validated in H3-K27M mutant HGG. RESULTS Ubiquitous and midline glia-specific expression of H3.3-K27M but not wild-type H3.3 caused pupal lethality, morphological alterations, and decreased H3K27me3. Knockdown of 17 candidate genes shifted the lethal phenotype to later stages of development. These included histone modifying and chromatin remodeling genes as well as genes regulating cell differentiation and proliferation. Notably, several of these genes were overexpressed in mutant H3-K27M mutated HGG. CONCLUSIONS Rapid screening, identification, and validation of relevant targets in "oncohistone" mediated pathogenesis have proven a challenge and a barrier to providing novel therapies. Our results provide further evidence on the role of chromatin modifiers in the genesis of H3.3-K27M. Notably, they validate Drosophila as a model system for rapid identification of relevant genes functionally involved in the detrimental effects of H3.3-K27M mutagenesis.
Collapse
Affiliation(s)
- Johannes Berlandi
- Institute of Neuropathology, University Hospital Münster, Münster, Germany
| | - Amel Chaouch
- Department of Biology, McGill University, Montreal, Quebec, Canada
| | - Nicolas De Jay
- Department of Human Genetics, McGill University, Montreal, Quebec, Canada.,The Lady Davis Institute, Jewish General Hospital, Montreal, Quebec, Canada
| | - Isabel Tegeder
- Institute of Neuropathology, University Hospital Münster, Münster, Germany
| | - Katharina Thiel
- Institute of Neuropathology, University Hospital Münster, Münster, Germany
| | - Margret Shirinian
- Department of Experimental Pathology, Immunology, and Microbiology Faculty of Medicine, American University of Beirut, Beirut, Lebanon
| | - Claudia L Kleinman
- Department of Human Genetics, McGill University, Montreal, Quebec, Canada.,The Lady Davis Institute, Jewish General Hospital, Montreal, Quebec, Canada
| | - Astrid Jeibmann
- Institute of Neuropathology, University Hospital Münster, Münster, Germany
| | - Paul Lasko
- Department of Biology, McGill University, Montreal, Quebec, Canada
| | - Nada Jabado
- Department of Paediatrics, McGill University and the McGill University Health Center Research Institute, Montreal, Quebec, Canada
| | - Martin Hasselblatt
- Institute of Neuropathology, University Hospital Münster, Münster, Germany
| |
Collapse
|
31
|
Anderson D, Neri JICF, Souza CRM, Valverde JG, De Araújo JMG, Nascimento MDSB, Branco RCC, Arrais NMR, Lassmann T, Blackwell JM, Jeronimo SMB. Zika Virus Changes Methylation of Genes Involved in Immune Response and Neural Development in Brazilian Babies Born With Congenital Microcephaly. J Infect Dis 2020; 223:435-440. [PMID: 32614431 DOI: 10.1093/infdis/jiaa383] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2020] [Accepted: 06/25/2020] [Indexed: 11/14/2022] Open
Abstract
The recent increase in babies born with brain and eye malformations in Brazil is associated with Zika virus (ZIKV) infection in utero. ZIKV alters host DNA methylation in vitro. Using genome-wide DNA methylation profiling we compared 18 babies born with congenital ZIKV microcephaly with 20 controls. We found ZIKV-associated alteration of host methylation patterns, notably at RABGAP1L which is important in brain development, at viral host immunity genes MX1 and ISG15, and in an epigenetic module containing the causal microcephaly gene MCPH1. Our data support the hypothesis that clinical signs of congenital ZIKV are associated with changes in DNA methylation.
Collapse
Affiliation(s)
- Denise Anderson
- Telethon Kids Institute, University of Western Australia, Perth Children's Hospital, Perth, Western Australia, Australia
| | - João I C F Neri
- Institute of Tropical Medicine of Rio Grande do Norte, Department of Biochemistry, Universidade Federal do Rio Grande do Norte, Natal, Rio de Grande do Norte, Brazil
| | - Cássio R M Souza
- Institute of Tropical Medicine of Rio Grande do Norte, Department of Biochemistry, Universidade Federal do Rio Grande do Norte, Natal, Rio de Grande do Norte, Brazil
| | - Joanna G Valverde
- Institute of Tropical Medicine of Rio Grande do Norte, Department of Biochemistry, Universidade Federal do Rio Grande do Norte, Natal, Rio de Grande do Norte, Brazil
| | - Josélio M G De Araújo
- Institute of Tropical Medicine of Rio Grande do Norte, Department of Biochemistry, Universidade Federal do Rio Grande do Norte, Natal, Rio de Grande do Norte, Brazil
| | | | - Rebeca C C Branco
- Department of Pathology, Federal University of Maranhão, São Luis, Maranhão, Brazil
| | - Nivia M R Arrais
- Department of Pediatrics, Federal University of Rio Grande do Norte, Empresa Brasileira de Servicos Hospitalares, Natal, Rio de Grande do Norte, Brazil
| | - Timo Lassmann
- Telethon Kids Institute, University of Western Australia, Perth Children's Hospital, Perth, Western Australia, Australia
| | - Jenefer M Blackwell
- Telethon Kids Institute, University of Western Australia, Perth Children's Hospital, Perth, Western Australia, Australia
| | - Selma M B Jeronimo
- Institute of Tropical Medicine of Rio Grande do Norte, Department of Biochemistry, Universidade Federal do Rio Grande do Norte, Natal, Rio de Grande do Norte, Brazil.,National Institute of Science and Technology of Tropical Diseases, Natal, Rio de Grande do Norte, Brazil
| |
Collapse
|
32
|
Chen Q, Song H, Liu C, Xu J, Wei C, Wang W, Han F. The Interaction of EphA4 With PDGFRβ Regulates Proliferation and Neuronal Differentiation of Neural Progenitor Cells in vitro and Promotes Neurogenesis in vivo. Front Aging Neurosci 2020; 12:7. [PMID: 32116646 PMCID: PMC7026009 DOI: 10.3389/fnagi.2020.00007] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2019] [Accepted: 01/13/2020] [Indexed: 01/07/2023] Open
Abstract
Neural progenitor cells (NPCs) have great potentials in cell replacement therapy for neurodegenerative diseases, such as Alzheimer’s disease (AD), by promoting neurogenesis associated with hippocampal memory improvement. Ephrin receptors and angiogenic growth factor receptors have a marked impact on the proliferation and differentiation of NPCs. Although ephrin receptor A4 (EphA4) was shown to directly interact with platelet-derived growth factor receptor β (PDGFRβ), the functional effects of this interaction on neurogenesis in cultured NPCs and adult hippocampus have not yet been studied. Immunoprecipitation demonstrated that EphA4 directly interacted with PDGFRβ in NPCs under ligand stimulation. Ephrin-A1 and PDGF-platelet-derived growth factor BB (BB) significantly increased proliferation and neuronal differentiation of NPCs, which was further augmented by combined treatment of Ephrin-A1 and PDGF-BB. We also found that ligand-dependent proliferation and neuronal differentiation were inhibited by the dominant-negative EphA4 mutant or a PDGFR inhibitor. Most importantly, injection of ephrin-A1 and/or PDGF-BB promoted hippocampal NPC proliferation in the APP/PS1 mouse model of AD, indicating that direct interaction of EphA4 with PDGFRβ plays a functional role on neurogenesis in vivo. Finally, studies in NPCs showed that the EphA4/PDGFRβ/FGFR1/FRS2α complex formed by ligand stimulation is involved in neurogenesis via ERK signaling. The present findings provided a novel insight into the functional role of direct interaction of EphA4 and PDGFRβ in neurogenesis, implicating its potential use for treating neurodegenerative diseases.
Collapse
Affiliation(s)
- Qingfa Chen
- The Institute for Tissue Engineering and Regenerative Medicine, The Liaocheng University/Liaocheng People's Hospital, Liaocheng, China
| | - Hao Song
- The Institute for Tissue Engineering and Regenerative Medicine, The Liaocheng University/Liaocheng People's Hospital, Liaocheng, China
| | - Chuanguo Liu
- The Translational Research Laboratory of Stem Cells and Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Jun Xu
- Department of Neurology, Qilu Hospital at Qingdao, Shandong University, Qingdao, China
| | - Chuanfei Wei
- The Institute for Tissue Engineering and Regenerative Medicine, The Liaocheng University/Liaocheng People's Hospital, Liaocheng, China
| | - Wei Wang
- The Institute for Tissue Engineering and Regenerative Medicine, The Liaocheng University/Liaocheng People's Hospital, Liaocheng, China
| | - Fabin Han
- The Institute for Tissue Engineering and Regenerative Medicine, The Liaocheng University/Liaocheng People's Hospital, Liaocheng, China.,The Translational Research Laboratory of Stem Cells and Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| |
Collapse
|
33
|
Teng S, Palmieri A, Maita I, Zheng C, Das G, Park J, Zhou R, Alder J, Thakker-Varia S. Inhibition of EphA/Ephrin-A signaling using genetic and pharmacologic approaches improves recovery following traumatic brain injury in mice. Brain Inj 2019; 33:1385-1401. [DOI: 10.1080/02699052.2019.1641622] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Affiliation(s)
- Shavonne Teng
- Department of Neuroscience and Cell Biology, Rutgers Robert Wood Johnson Medical School, Piscataway, New Jersey, USA
| | - Alicia Palmieri
- Department of Neuroscience and Cell Biology, Rutgers Robert Wood Johnson Medical School, Piscataway, New Jersey, USA
| | - Isabella Maita
- Department of Neuroscience and Cell Biology, Rutgers Robert Wood Johnson Medical School, Piscataway, New Jersey, USA
| | - Cynthia Zheng
- Department of Neuroscience and Cell Biology, Rutgers Robert Wood Johnson Medical School, Piscataway, New Jersey, USA
| | - Gitanjali Das
- Department of Chemical Biology, Ernest Mario School of Pharmacy, Rutgers University, Piscataway, New Jersey, USA
| | - Juyeon Park
- Department of Neuroscience and Cell Biology, Rutgers Robert Wood Johnson Medical School, Piscataway, New Jersey, USA
| | - Renping Zhou
- Department of Chemical Biology, Ernest Mario School of Pharmacy, Rutgers University, Piscataway, New Jersey, USA
| | - Janet Alder
- Department of Neuroscience and Cell Biology, Rutgers Robert Wood Johnson Medical School, Piscataway, New Jersey, USA
| | - Smita Thakker-Varia
- Department of Neuroscience and Cell Biology, Rutgers Robert Wood Johnson Medical School, Piscataway, New Jersey, USA
| |
Collapse
|
34
|
Fritzsch B, Elliott KL, Pavlinkova G. Primary sensory map formations reflect unique needs and molecular cues specific to each sensory system. F1000Res 2019; 8:F1000 Faculty Rev-345. [PMID: 30984379 PMCID: PMC6439788 DOI: 10.12688/f1000research.17717.1] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 03/21/2019] [Indexed: 12/21/2022] Open
Abstract
Interaction with the world around us requires extracting meaningful signals to guide behavior. Each of the six mammalian senses (olfaction, vision, somatosensation, hearing, balance, and taste) has a unique primary map that extracts sense-specific information. Sensory systems in the periphery and their target neurons in the central nervous system develop independently and must develop specific connections for proper sensory processing. In addition, the regulation of sensory map formation is independent of and prior to central target neuronal development in several maps. This review provides an overview of the current level of understanding of primary map formation of the six mammalian senses. Cell cycle exit, combined with incompletely understood molecules and their regulation, provides chemoaffinity-mediated primary maps that are further refined by activity. The interplay between cell cycle exit, molecular guidance, and activity-mediated refinement is the basis of dominance stripes after redundant organ transplantations in the visual and balance system. A more advanced level of understanding of primary map formation could benefit ongoing restoration attempts of impaired senses by guiding proper functional connection formations of restored sensory organs with their central nervous system targets.
Collapse
Affiliation(s)
- Bernd Fritzsch
- Department of Biology, University of Iowa, Iowa City, USA
| | | | - Gabriela Pavlinkova
- Institute of Biotechnology of the Czech Academy of Sciences, Vestec, Czech Republic
| |
Collapse
|
35
|
Franco M, Carmena A. Eph signaling controls mitotic spindle orientation and cell proliferation in neuroepithelial cells. J Cell Biol 2019; 218:1200-1217. [PMID: 30808706 PMCID: PMC6446852 DOI: 10.1083/jcb.201807157] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2018] [Revised: 12/16/2018] [Accepted: 01/28/2019] [Indexed: 12/17/2022] Open
Abstract
In this study, Franco and Carmena uncover a function for Eph signaling as a novel extrinsic mechanism controlling mitotic spindle alignment in Drosophila neuroepithelial cells through aPKC activity–dependent myosin II regulation. Additionally, Eph loss leads to a Rho signaling–dependent activation of the PI3K–Akt1 pathway, enhancing cell proliferation within this neuroepithelium. Mitotic spindle orientation must be tightly regulated during development and adult tissue homeostasis. It determines cell-fate specification and tissue architecture during asymmetric and symmetric cell division, respectively. Here, we uncover a novel role for Ephrin–Eph intercellular signaling in controlling mitotic spindle alignment in Drosophila optic lobe neuroepithelial cells through aPKC activity–dependent myosin II regulation. We show that conserved core components of the mitotic spindle orientation machinery, including Discs Large1, Mud/NuMA, and Canoe/Afadin, mislocalize in dividing Eph mutant neuroepithelial cells and produce spindle alignment defects in these cells when they are down-regulated. In addition, the loss of Eph leads to a Rho signaling–dependent activation of the PI3K–Akt1 pathway, enhancing cell proliferation within this neuroepithelium. Hence, Eph signaling is a novel extrinsic mechanism that regulates both spindle orientation and cell proliferation in the Drosophila optic lobe neuroepithelium. Similar mechanisms could operate in other Drosophila and vertebrate epithelia.
Collapse
Affiliation(s)
- Maribel Franco
- Developmental Neurobiology Department, Instituto de Neurociencias, Consejo Superior de Investigaciones Científicas/Universidad Miguel Hernández, Alicante, Spain
| | - Ana Carmena
- Developmental Neurobiology Department, Instituto de Neurociencias, Consejo Superior de Investigaciones Científicas/Universidad Miguel Hernández, Alicante, Spain
| |
Collapse
|
36
|
Suo G, Shen F, Sun B, Song H, Xu M, Wu Y. Abnormal expression of ephrin-A5 affects brain development of congenital hypothyroidism rats. Neuroreport 2019; 29:877-882. [PMID: 29762250 DOI: 10.1097/wnr.0000000000001047] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
EphA5 and its ligand ephrin-A5 interaction can trigger synaptogenesis during early hippocampus development. We have previously reported that abnormal EphA5 expression can result in synaptogenesis disorder in congenital hypothyroidism (CH) rats. To better understand its precise molecular mechanism, we further analyzed the characteristics of ephrin-A5 expression in the hippocampus of CH rats. Our study revealed that ephrin-A5 expression was downregulated by thyroid hormone deficiency in the developing hippocampus and hippocampal neurons in rats. Thyroxine treatment for hypothyroid hippocampus and triiodothyronine treatment for hypothyroid hippocampal neurons significantly improved ephrin-A5 expression but could not restore its expression to control levels. Hypothyroid hippocampal neurons in-vitro showed synaptogenesis disorder characterized by a reduction in the number and length of neurites. Furthermore, the synaptogenesis-associated molecular expressions of NMDAR-1 (NR1), PSD95 and CaMKII were all downregulated correspondingly. These results suggest that ephrin-A5 expression may be decreased in CH, and abnormal activation of ephrin-A5/EphA5 signaling affects synaptogenesis during brain development. Such findings provide an important basis for exploring the pathogenesis of CH genetically.
Collapse
|
37
|
Regulation of axonal EphA4 forward signaling is involved in the effect of EphA3 on chicken retinal ganglion cell axon growth during retinotectal mapping. Exp Eye Res 2019; 178:46-60. [DOI: 10.1016/j.exer.2018.09.007] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2018] [Revised: 09/06/2018] [Accepted: 09/16/2018] [Indexed: 12/22/2022]
|
38
|
Glycans and glycosaminoglycans in neurobiology: key regulators of neuronal cell function and fate. Biochem J 2018; 475:2511-2545. [PMID: 30115748 DOI: 10.1042/bcj20180283] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2018] [Revised: 07/14/2018] [Accepted: 07/18/2018] [Indexed: 12/16/2022]
Abstract
The aim of the present study was to examine the roles of l-fucose and the glycosaminoglycans (GAGs) keratan sulfate (KS) and chondroitin sulfate/dermatan sulfate (CS/DS) with selected functional molecules in neural tissues. Cell surface glycans and GAGs have evolved over millions of years to become cellular mediators which regulate fundamental aspects of cellular survival. The glycocalyx, which surrounds all cells, actuates responses to growth factors, cytokines and morphogens at the cellular boundary, silencing or activating downstream signaling pathways and gene expression. In this review, we have focused on interactions mediated by l-fucose, KS and CS/DS in the central and peripheral nervous systems. Fucose makes critical contributions in the area of molecular recognition and information transfer in the blood group substances, cytotoxic immunoglobulins, cell fate-mediated Notch-1 interactions, regulation of selectin-mediated neutrophil extravasation in innate immunity and CD-34-mediated new blood vessel development, and the targeting of neuroprogenitor cells to damaged neural tissue. Fucosylated glycoproteins regulate delivery of synaptic neurotransmitters and neural function. Neural KS proteoglycans (PGs) were examined in terms of cellular regulation and their interactive properties with neuroregulatory molecules. The paradoxical properties of CS/DS isomers decorating matrix and transmembrane PGs and the positive and negative regulatory cues they provide to neurons are also discussed.
Collapse
|
39
|
Zhen L, Shao T, Luria V, Li G, Li Z, Xu Y, Zhao X. EphB2 Deficiency Induces Depression-Like Behaviors and Memory Impairment: Involvement of NMDA 2B Receptor Dependent Signaling. Front Pharmacol 2018; 9:862. [PMID: 30131699 PMCID: PMC6090375 DOI: 10.3389/fphar.2018.00862] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2018] [Accepted: 07/17/2018] [Indexed: 01/06/2023] Open
Abstract
Receptor tyrosine kinase EphB2 mediates development of the neurogenic niche of excitatory neurons, suggesting the possibility that its inactivation plays a role in neuropsychiatric disorders including depression and memory impairment. While N-methyl-D-aspartate (NMDA) receptor is involved in regulating memory formation and neurogenesis in adult animal, it remains unclear how NMDA receptor subtypes mediate depression and cognitive deficits caused by EphB2 loss. The present study shows that EphB2 inactivation results in depression-like behaviors, memory impairment and defects of adult hippocampal neurogenesis. Compared to wild-type littermates, EphB2 KO mice exhibited depression-like behavior and deficits in spatial memory and cognition in forced swimming, tail suspension, Morris water maze, object recognition test and object location test. These behavioral abnormalities were accompanied by substantial decreases in the number of BrdU+ progenitor neurons, phosphorylation of cAMP-response element binding protein (pCREB) and brain derived neurotrophic factor (BDNF), and increased NMDA receptor 2B (NR2B) expression. These molecular, cellular and behavioral alterations induced by EphB2 inactivation were reversed by NR2B antagonist Ro25-6981, suggesting that EphB2 functions to prevent the progression of depression-like behavior and memory impairment by downregulating NR2B. Our findings highlight that NR2B is responsible for EphB2-dependent behavioral and morphological changes. EphB2 may thus be as an important candidate target for treating psychiatric and cognitive disorders.
Collapse
Affiliation(s)
- Linlin Zhen
- Department of Breast and Thyroid Surgery, The Affiliated Huai'an No. 1 People's Hospital of Nanjing Medical University, Huai'an, China
| | - Tuo Shao
- Brain Institute, School of Pharmacy, Wenzhou Medical University, Wenzhou, China
| | - Victor Luria
- Department of Systems Biology, Harvard University Medical School, Boston, MA, United States
| | - Gaowen Li
- Department of Pharmacology, Ningbo College of Health Sciences, Ningbo, China
| | - Zhi Li
- Department of Breast and Thyroid Surgery, The Affiliated Huai'an No. 1 People's Hospital of Nanjing Medical University, Huai'an, China.,School of Pharmacy and Pharmaceutical Sciences, University at Buffalo, State University of New York, Buffalo, NY, United States
| | - Ying Xu
- School of Pharmacy and Pharmaceutical Sciences, University at Buffalo, State University of New York, Buffalo, NY, United States
| | - Xin Zhao
- Department of Pharmacology, School of Medical Science, Ningbo University, Ningbo, China
| |
Collapse
|
40
|
Guo Q, Zhu H, Wang H, Zhang P, Wang S, Sun Z, Li S, Xue C, Gu X, Cui S. Transcriptomic Landscapes of Immune Response and Axonal Regeneration by Integrative Analysis of Molecular Pathways and Interactive Networks Post-sciatic Nerve Transection. Front Neurosci 2018; 12:457. [PMID: 30038556 PMCID: PMC6046400 DOI: 10.3389/fnins.2018.00457] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2017] [Accepted: 06/15/2018] [Indexed: 12/31/2022] Open
Abstract
Potential interaction between immune response and axonal regeneration has recently attracted much attention in peripheral nervous system (PNS). Previously, global mRNA expression changes in proximal nerve segments were profiled and merely focused on the differentially change of the key biological processes. To further uncover molecular mechanisms of peripheral nerve regeneration, here we focused on the interaction between immune response and axonal regeneration that associated with specific molecular pathways and interactive networks following sciatic nerve transection. To offer an outline of the specific molecular pathways elaborating axonal regeneration and immune response, and to figure out the molecular interaction between immune response and axonal regeneration post-sciatic nerve transection, we carried out comprehensive approaches, including gene expression profiling plus multi-level bioinformatics analysis and then further experimental validation. Alcam, Nrp1, Nrp2, Rac1, Creb1, and Runx3 were firstly considered as the key or hub genes of the protein-protein interaction (PPI) network in rat models of sciatic nerve transection, which are highly correlated with immune response and axonal regeneration. Our work provide a new way to figure out molecular mechanism of peripheral nerve regeneration and valuable resources to figure out the molecular courses which outline neural injury-induced micro-environmental variation to discover novel therapeutic targets for axonal regeneration.
Collapse
Affiliation(s)
- Qi Guo
- Department of Hand Surgery, China-Japan Union Hospital, Jilin University, Changchun, China.,Key Laboratory of Neuroregeneration, Ministry of Education and Jiangsu Province, Co-innovation Center of Neuroregeneration, Nantong University, Nantong, China
| | - Hui Zhu
- Key Laboratory of Neuroregeneration, Ministry of Education and Jiangsu Province, Co-innovation Center of Neuroregeneration, Nantong University, Nantong, China.,Jiangsu Clinical Medicine Center of Tissue Engineering and Nerve Injury Repair, Research Center of Clinical Medicine, Affiliated Hospital of Nantong University, Nantong, China
| | - Hongkui Wang
- Key Laboratory of Neuroregeneration, Ministry of Education and Jiangsu Province, Co-innovation Center of Neuroregeneration, Nantong University, Nantong, China
| | - Ping Zhang
- Key Laboratory of Neuroregeneration, Ministry of Education and Jiangsu Province, Co-innovation Center of Neuroregeneration, Nantong University, Nantong, China
| | - Shengran Wang
- Key Laboratory of Neuroregeneration, Ministry of Education and Jiangsu Province, Co-innovation Center of Neuroregeneration, Nantong University, Nantong, China
| | - Zhichao Sun
- Key Laboratory of Neuroregeneration, Ministry of Education and Jiangsu Province, Co-innovation Center of Neuroregeneration, Nantong University, Nantong, China
| | - Shiying Li
- Key Laboratory of Neuroregeneration, Ministry of Education and Jiangsu Province, Co-innovation Center of Neuroregeneration, Nantong University, Nantong, China
| | - Chengbin Xue
- Key Laboratory of Neuroregeneration, Ministry of Education and Jiangsu Province, Co-innovation Center of Neuroregeneration, Nantong University, Nantong, China.,Jiangsu Clinical Medicine Center of Tissue Engineering and Nerve Injury Repair, Research Center of Clinical Medicine, Affiliated Hospital of Nantong University, Nantong, China
| | - Xiaosong Gu
- Key Laboratory of Neuroregeneration, Ministry of Education and Jiangsu Province, Co-innovation Center of Neuroregeneration, Nantong University, Nantong, China.,Jiangsu Clinical Medicine Center of Tissue Engineering and Nerve Injury Repair, Research Center of Clinical Medicine, Affiliated Hospital of Nantong University, Nantong, China
| | - Shusen Cui
- Department of Hand Surgery, China-Japan Union Hospital, Jilin University, Changchun, China
| |
Collapse
|
41
|
|
42
|
Chen Z. Common cues wire the spinal cord: Axon guidance molecules in spinal neuron migration. Semin Cell Dev Biol 2018; 85:71-77. [PMID: 29274387 DOI: 10.1016/j.semcdb.2017.12.012] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2017] [Revised: 12/12/2017] [Accepted: 12/14/2017] [Indexed: 01/28/2023]
Abstract
Topographic arrangement of neuronal cell bodies and axonal tracts are crucial for proper wiring of the nervous system. This involves often-coordinated neuronal migration and axon guidance during development. Most neurons migrate from their birthplace to specific topographic coordinates as they adopt the final cell fates and extend axons. The axons follow temporospatial specific guidance cues to reach the appropriate targets. When neuronal or axonal migration or their coordination is disrupted, severe consequences including neurodevelopmental disorders and neurological diseases, can arise. Neuronal and axonal migration shares some molecular mechanisms, as genes originally identified as axon guidance molecules have been increasingly shown to direct both navigation processes. This review focuses on axon guidance pathways that are shown to also direct neuronal migration in the vertebrate spinal cord.
Collapse
Affiliation(s)
- Zhe Chen
- Department of MCD Biology, University of Colorado Boulder, Boulder, CO 80309, USA.
| |
Collapse
|
43
|
Daugherty DJ, Marquez A, Calcutt NA, Schubert D. A novel curcumin derivative for the treatment of diabetic neuropathy. Neuropharmacology 2018; 129:26-35. [PMID: 29122628 PMCID: PMC5841546 DOI: 10.1016/j.neuropharm.2017.11.007] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2017] [Revised: 11/03/2017] [Accepted: 11/04/2017] [Indexed: 12/16/2022]
Abstract
Neuropathy is a common complication of long-term diabetes. Proposed mechanisms of neuronal damage caused by diabetes that are downstream of hyperglycemia and/or loss of insulin signaling include ischemic hypoxia, inflammation and loss of neurotrophic support. The curcumin derivative J147 is a potent neurogenic and neuroprotective drug candidate initially developed for the treatment of neurodegenerative conditions associated with aging that impacts many pathways implicated in the pathogenesis of diabetic neuropathy. Here, we demonstrate efficacy of J147 in ameliorating multiple indices of neuropathy in the streptozotocin-induced mouse model of type 1 diabetes. Diabetes was determined by blood glucose, HbA1c, and insulin levels and efficacy of J147 by behavioral, physiologic, biochemical, proteomic, and transcriptomic assays. Biological efficacy of systemic J147 treatment was confirmed by its capacity to decrease TNFα pathway activation and several other markers of neuroinflammation in the CNS. Chronic oral treatment with J147 protected the sciatic nerve from progressive diabetes-induced slowing of large myelinated fiber conduction velocity while single doses of J147 rapidly and transiently reversed established touch-evoked allodynia. Conduction slowing and allodynia are clinically relevant markers of early diabetic neuropathy and neuropathic pain, respectively. RNA expression profiling suggests that one of the pathways by which J147 imparts its protection against diabetic induced neuropathy may be through activation of the AMP kinase pathway. The diverse biological and therapeutic effects of J147 suggest it as an alternative to the polypharmaceutical approaches required to treat the multiple pathogenic mechanisms that contribute to diabetic neuropathy.
Collapse
Affiliation(s)
| | | | | | - David Schubert
- The Salk Institute for Biological Studies, La Jolla, CA, USA
| |
Collapse
|
44
|
Single-cell transcriptomics of the developing lateral geniculate nucleus reveals insights into circuit assembly and refinement. Proc Natl Acad Sci U S A 2018; 115:E1051-E1060. [PMID: 29343640 PMCID: PMC5798372 DOI: 10.1073/pnas.1717871115] [Citation(s) in RCA: 57] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Neurons and nonneuronal cells in the developing brain dynamically regulate gene expression as neural connectivity is established. However, the specific gene programs activated in distinct cell populations during the assembly and refinement of many intact neuronal circuits have not been thoroughly characterized. In this study, we take advantage of recent advances in transcriptomic profiling techniques to characterize gene expression in the postnatal developing lateral geniculate nucleus (LGN) at single-cell resolution. Our data reveal that genes involved in brain development are dynamically regulated in all major cell types of the LGN, suggesting that the establishment of neural connectivity depends upon functional collaboration between multiple neuronal and nonneuronal cell types in this brain region. Coordinated changes in gene expression underlie the early patterning and cell-type specification of the central nervous system. However, much less is known about how such changes contribute to later stages of circuit assembly and refinement. In this study, we employ single-cell RNA sequencing to develop a detailed, whole-transcriptome resource of gene expression across four time points in the developing dorsal lateral geniculate nucleus (LGN), a visual structure in the brain that undergoes a well-characterized program of postnatal circuit development. This approach identifies markers defining the major LGN cell types, including excitatory relay neurons, oligodendrocytes, astrocytes, microglia, and endothelial cells. Most cell types exhibit significant transcriptional changes across development, dynamically expressing genes involved in distinct processes including retinotopic mapping, synaptogenesis, myelination, and synaptic refinement. Our data suggest that genes associated with synapse and circuit development are expressed in a larger proportion of nonneuronal cell types than previously appreciated. Furthermore, we used this single-cell expression atlas to identify the Prkcd-Cre mouse line as a tool for selective manipulation of relay neurons during a late stage of sensory-driven synaptic refinement. This transcriptomic resource provides a cellular map of gene expression across several cell types of the LGN, and offers insight into the molecular mechanisms of circuit development in the postnatal brain.
Collapse
|
45
|
Wan Y, Yang JS, Xu LC, Huang XJ, Wang W, Xie MJ. Roles of Eph/ephrin bidirectional signaling during injury and recovery of the central nervous system. Neural Regen Res 2018; 13:1313-1321. [PMID: 30106032 PMCID: PMC6108204 DOI: 10.4103/1673-5374.235217] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Multiple cellular components, including neuronal, glial and endothelial cells, are involved in the sophisticated pathological processes following central nervous system injury. The pathological process cannot reduce damage or improve functional recovery by merely targeting the molecular mechanisms of neuronal cell death after central nerve system injuries. Eph receptors and ephrin ligands have drawn wide attention since the discovery of their extensive distribution and unique bidirectional signaling between astrocytes and neurons. The roles of Eph/ephrin bidirectional signaling in the developmental processes have been reported in previous research. Recent observations suggest that Eph/ephrin bidirectional signaling continues to be expressed in most regions and cell types in the adult central nervous system, playing diverse roles. The Eph/ephrin complex mediates neurogenesis and angiogenesis, promotes glial scar formation, regulates endocrine levels, inhibits myelin formation and aggravates inflammation and nerve pain caused by injury. The interaction between Eph and ephrin is also considered to be the key to angiogenesis. This review focuses on the roles of Eph/ephrin bidirectional signaling in the repair of central nervous system injuries.
Collapse
Affiliation(s)
- Yue Wan
- Department of Neurology, The Third People's Hospital of Hubei Province, Wuhan, Hubei Province, China
| | - Jin-Shan Yang
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province; Department of Neurology, First Affiliated Hospital, Fujian Medical University, Fuzhou, Fujian Province, China
| | - Li-Cai Xu
- Department of Neurological Rehabilitation Center, The Third People's Hospital of Hubei Province, Wuhan, Hubei Province, China
| | - Xiao-Jiang Huang
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China
| | - Wei Wang
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China
| | - Min-Jie Xie
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China
| |
Collapse
|
46
|
Gugliandolo A, Diomede F, Cardelli P, Bramanti A, Scionti D, Bramanti P, Trubiani O, Mazzon E. Transcriptomic analysis of gingival mesenchymal stem cells cultured on 3D bioprinted scaffold: A promising strategy for neuroregeneration. J Biomed Mater Res A 2017; 106:126-137. [PMID: 28879677 DOI: 10.1002/jbm.a.36213] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2017] [Revised: 08/02/2017] [Accepted: 08/22/2017] [Indexed: 01/01/2023]
Abstract
The combined approach of mesenchymal stem cells (MSCs) and scaffolds has been proposed as a potential therapeutic tool for the treatment of neurodegenerative diseases. Indeed, even if MSCs can promote neuronal regeneration, replacing lost neurons or secreting neurotrophic factors, many limitations still exist for their application in regenerative medicine, including the low survival and differentiation rate. The scaffolds, by mimicking the endogenous microenvironment, have shown to promote cell survival, proliferation, and differentiation. In this work, gingival mesenchymal stem cells (GMSCs), isolated from healthy donors, were expanded in vitro, by culturing them adherent in plastic dishes (CTR-GMSCs) or on a poly(lactic acid) scaffold (SC-GMSCs). In order to evaluate the survival and the neurogenic differentiation potential, we performed a comparative transcriptomic analysis between CTR-GMSCs and SC-GMSCs by next generation sequencing. We found that SC-GMSCs showed an increased expression of neurogenic and prosurvival genes. In particular, genes involved in neurotrophin signaling and PI3K/Akt pathways were upregulated. On the contrary, proapoptotic and negative regulator of neuronal growth genes were downregulated. Moreover, nestin and GAP-43 protein levels increased in SC-GMSCs, confirming the neurogenic commitment of these cells. In conclusion, the scaffold, providing a trophic support for MSCs, may promote GMSCs differentiation toward a neuronal phenotype and survival. © 2017 Wiley Periodicals, Inc. J Biomed Mater Res Part A: 106A: 126-137, 2018.
Collapse
Affiliation(s)
- Agnese Gugliandolo
- IRCCS Centro Neurolesi "Bonino-Pulejo," Via Provinciale Palermo, Contrada Casazza, 98124 Messina, Italy
| | - Francesca Diomede
- Stem Cells and Regenerative Medicine Laboratory, Department of Medical, Oral and Biotechnological Sciences, University "G. D'Annunzio," Chieti-Pescara, via dei Vestini, 31, 66100, Chieti, Italy
| | - Paolo Cardelli
- Stem Cells and Regenerative Medicine Laboratory, Department of Medical, Oral and Biotechnological Sciences, University "G. D'Annunzio," Chieti-Pescara, via dei Vestini, 31, 66100, Chieti, Italy
| | - Alessia Bramanti
- IRCCS Centro Neurolesi "Bonino-Pulejo," Via Provinciale Palermo, Contrada Casazza, 98124 Messina, Italy.,Institute of Applied Science and Intelligent Systems "ISASI Eduardo Caianiello,", National Research Council of Italy, Messina, Italy
| | - Domenico Scionti
- IRCCS Centro Neurolesi "Bonino-Pulejo," Via Provinciale Palermo, Contrada Casazza, 98124 Messina, Italy
| | - Placido Bramanti
- IRCCS Centro Neurolesi "Bonino-Pulejo," Via Provinciale Palermo, Contrada Casazza, 98124 Messina, Italy
| | - Oriana Trubiani
- Stem Cells and Regenerative Medicine Laboratory, Department of Medical, Oral and Biotechnological Sciences, University "G. D'Annunzio," Chieti-Pescara, via dei Vestini, 31, 66100, Chieti, Italy
| | - Emanuela Mazzon
- IRCCS Centro Neurolesi "Bonino-Pulejo," Via Provinciale Palermo, Contrada Casazza, 98124 Messina, Italy
| |
Collapse
|
47
|
Nagaraja S, Vitanza NA, Woo PJ, Taylor KR, Liu F, Zhang L, Li M, Meng W, Ponnuswami A, Sun W, Ma J, Hulleman E, Swigut T, Wysocka J, Tang Y, Monje M. Transcriptional Dependencies in Diffuse Intrinsic Pontine Glioma. Cancer Cell 2017; 31:635-652.e6. [PMID: 28434841 PMCID: PMC5462626 DOI: 10.1016/j.ccell.2017.03.011] [Citation(s) in RCA: 275] [Impact Index Per Article: 34.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/29/2016] [Revised: 12/27/2016] [Accepted: 03/22/2017] [Indexed: 12/12/2022]
Abstract
Diffuse intrinsic pontine glioma (DIPG) is a fatal pediatric cancer with limited therapeutic options. The majority of cases of DIPG exhibit a mutation in histone-3 (H3K27M) that results in oncogenic transcriptional aberrancies. We show here that DIPG is vulnerable to transcriptional disruption using bromodomain inhibition or CDK7 blockade. Targeting oncogenic transcription through either of these methods synergizes with HDAC inhibition, and DIPG cells resistant to HDAC inhibitor therapy retain sensitivity to CDK7 blockade. Identification of super-enhancers in DIPG provides insights toward the cell of origin, highlighting oligodendroglial lineage genes, and reveals unexpected mechanisms mediating tumor viability and invasion, including potassium channel function and EPH receptor signaling. The findings presented demonstrate transcriptional vulnerabilities and elucidate previously unknown mechanisms of DIPG pathobiology.
Collapse
Affiliation(s)
- Surya Nagaraja
- Department of Neurology, Stanford University, Palo Alto, CA 94305, USA
| | | | - Pamelyn J Woo
- Department of Neurology, Stanford University, Palo Alto, CA 94305, USA
| | - Kathryn R Taylor
- Department of Neurology, Stanford University, Palo Alto, CA 94305, USA
| | - Fang Liu
- Key Laboratory of Cell Differentiation and Apoptosis of National Ministry of Education, Department of Pathophysiology, Shanghai Jiao Tong University School of Medicine, 280 South Chongqing Road, Shanghai 200025, P.R. China
| | - Lei Zhang
- Key Laboratory of Cell Differentiation and Apoptosis of National Ministry of Education, Department of Pathophysiology, Shanghai Jiao Tong University School of Medicine, 280 South Chongqing Road, Shanghai 200025, P.R. China
| | - Meng Li
- Key Laboratory of Cell Differentiation and Apoptosis of National Ministry of Education, Department of Pathophysiology, Shanghai Jiao Tong University School of Medicine, 280 South Chongqing Road, Shanghai 200025, P.R. China
| | - Wei Meng
- Department of Pediatric Neurosurgery, Xin Hua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200092, P.R. China
| | - Anitha Ponnuswami
- Department of Neurology, Stanford University, Palo Alto, CA 94305, USA
| | - Wenchao Sun
- Department of Neurology, Stanford University, Palo Alto, CA 94305, USA
| | - Jie Ma
- Department of Pediatric Neurosurgery, Xin Hua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200092, P.R. China
| | - Esther Hulleman
- Department of Pediatric Oncology, VU University Medical Center, 1081 HV Amsterdam, the Netherlands
| | - Tomek Swigut
- Department of Chemical and Systems Biology, Stanford University, Palo Alto, CA 94305, USA
| | - Joanna Wysocka
- Department of Chemical and Systems Biology, Stanford University, Palo Alto, CA 94305, USA; Institute for Stem Cell Biology and Regenerative Medicine, Stanford University, Palo Alto, California 94305, USA; Department of Developmental Biology, Stanford University, Palo Alto, California 94305, USA; Howard Hughes Medical Institute, Stanford School of Medicine, Stanford University, Palo Alto, California 94305, USA
| | - Yujie Tang
- Department of Neurology, Stanford University, Palo Alto, CA 94305, USA; Key Laboratory of Cell Differentiation and Apoptosis of National Ministry of Education, Department of Pathophysiology, Shanghai Jiao Tong University School of Medicine, 280 South Chongqing Road, Shanghai 200025, P.R. China; Department of Pediatric Neurosurgery, Xin Hua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200092, P.R. China.
| | - Michelle Monje
- Department of Neurology, Stanford University, Palo Alto, CA 94305, USA; Institute for Stem Cell Biology and Regenerative Medicine, Stanford University, Palo Alto, California 94305, USA.
| |
Collapse
|
48
|
EphA4 Regulates Neuroblast and Astrocyte Organization in a Neurogenic Niche. J Neurosci 2017; 37:3331-3341. [PMID: 28258169 DOI: 10.1523/jneurosci.3738-16.2017] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2016] [Revised: 02/10/2017] [Accepted: 02/13/2017] [Indexed: 11/21/2022] Open
Abstract
Significant migration cues are required to guide and contain newly generated rodent subventricular zone (SVZ) neuroblasts as they transit along the lateral ventricles and then through the anterior forebrain to their ultimate site of differentiation in the olfactory bulbs (OBs). These cues enforce strict neuroblast spatial boundaries within the dense astroglial meshwork of the SVZ and rostral migratory stream (RMS), yet are permissive to large-scale neuroblast migration. Therefore, the molecular mechanisms that define these cues and control dynamic interactions between migratory neuroblasts and surrounding astrocytes are of particular interest. We found that deletion of EphA4 and specifically ablation of EphA4 kinase activity resulted in misaligned neuroblasts and disorganized astrocytes in the RMS/SVZ, linking EphA4 forward signaling to SVZ and RMS spatial organization, orientation, and regulation. In addition, within a 3 week period, there was a significant reduction in the number of neuroblasts that reached the OB and integrated into the periglomerular layer, revealing a crucial role for EphA4 in facilitating efficient neuroblast migration to the OB. Single-cell analysis revealed that EPHA4 and its EFN binding partners are expressed by subpopulations of neuroblasts and astrocytes within the SVZ/RMS/OB system resulting in a cell-specific mosaic, suggesting complex EphA4 signaling involving both homotypic and heterotypic cell-cell interactions. Together, our studies reveal a novel molecular mechanism involving EphA4 signaling that functions in stem cell niche organization and ultimately neuroblast migration in the anterior forebrain.SIGNIFICANCE STATEMENT The subventricular zone neurogenic stem cell niche generates highly migratory neuroblasts that transit the anterior forebrain along a defined pathway to the olfactory bulb. Postnatal and adult brain organization dictates strict adherence to a narrow migration corridor. Subventricular zone neuroblasts are aligned in tightly bundled chains within a meshwork of astrocytes; however, the cell-cell cues that organize this unique, cell-dense migration pathway are largely unknown. Our studies show that forward signaling through the EphA4 tyrosine kinase receptor, mediated by ephrins expressed by subpopulations of neuroblasts and astrocytes, is required for compact, directional organization of neuroblasts and astrocytes within the pathway and efficient transit of neuroblasts through the anterior forebrain to the olfactory bulb.
Collapse
|
49
|
EphrinA5 Signaling Is Required for the Distinctive Targeting of Raphe Serotonin Neurons in the Forebrain. eNeuro 2017; 4:eN-NWR-0327-16. [PMID: 28197551 PMCID: PMC5292598 DOI: 10.1523/eneuro.0327-16.2017] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2016] [Accepted: 01/06/2017] [Indexed: 12/13/2022] Open
Abstract
Serotonin (5-HT) neurotransmission in the brain relies on a widespread axon terminal network originating from the hindbrain raphe nuclei. These projections are topographically organized such that the dorsal (DR), and median raphe (MnR) nuclei have different brain targets. However, the guidance molecules involved in this selective targeting in development are unknown. Here, we show the implication of ephrinA5 signaling in this process. We find that the EphA5 gene is selectively expressed in a subset of 5-HT neurons during embryonic and postnatal development. Highest coexpression of EphA5 and the 5-HT marker Tph2 is found in the DR, with lower coexpression in the MnR, and hardly any colocalization of the caudal raphe in the medulla. Accordingly, ephrinA induced a dose-dependent collapse response of 5-HT growth cones cultured from rostral but not caudal raphe. Ectopic expression of ephrinA3, after in utero electroporation in the amygdala and piriform cortex, repelled 5-HT raphe fiber ingrowth. Conversely, misplaced DR 5-HT axons were found in ephrin A5 knockout mice in brain regions that are normally only targeted by MnR 5-HT axons. This causes an overall increase in the density of 5-HT innervation in the ventromedial hypothalamus, the suprachiasmatic nucleus, and the olfactory bulb. All these brain areas have high expression of ephrinAs at the time of 5-HT fiber ingrowth. Present results show for the first time the role of a guidance molecule for the region-specific targeting of raphe neurons. This has important implications to understand how functional parsing of central 5-HT neurons is established during development.
Collapse
|
50
|
Braud M, Magee DA, Park SDE, Sonstegard TS, Waters SM, MacHugh DE, Spillane C. Genome-Wide microRNA Binding Site Variation between Extinct Wild Aurochs and Modern Cattle Identifies Candidate microRNA-Regulated Domestication Genes. Front Genet 2017; 8:3. [PMID: 28197171 PMCID: PMC5281612 DOI: 10.3389/fgene.2017.00003] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2016] [Accepted: 01/09/2017] [Indexed: 12/21/2022] Open
Abstract
The domestication of cattle from the now-extinct wild aurochs (Bos primigenius) involved selection for physiological and behavioral traits, with underlying genetic factors that remain largely unknown. Non-coding microRNAs have emerged as key regulators of the spatio-temporal expression of target genes controlling mammalian growth and development, including in livestock species. During the domestication process, selection of mutational changes in miRNAs and/or miRNA binding sites could have provided a mechanism to generate some of the traits that differentiate domesticated cattle from wild aurochs. To investigate this, we analyzed the open reading frame DNA sequence of 19,994 orthologous protein-coding gene pairs from extant Bos taurus genomes and a single extinct B. primigenius genome. We identified miRNA binding site polymorphisms in the 3′ UTRs of 1,620 of these orthologous genes. These 1,620 genes with altered miRNA binding sites between the B. taurus and B. primigenius lineages represent candidate domestication genes. Using a novel Score Site ratio metric we have ranked these miRNA-regulated genes according to the extent of divergence between miRNA binding site presence, frequency and copy number between the orthologous genes from B. taurus and B. primigenius. This provides an unbiased approach to identify cattle genes that have undergone the most changes in miRNA binding (i.e., regulation) between the wild aurochs and modern-day cattle breeds. In addition, we demonstrate that these 1,620 candidate domestication genes are enriched for roles in pigmentation, fertility, neurobiology, metabolism, immunity and production traits (including milk quality and feed efficiency). Our findings suggest that directional selection of miRNA regulatory variants was important in the domestication and subsequent artificial selection that gave rise to modern taurine cattle.
Collapse
Affiliation(s)
- Martin Braud
- Genetics and Biotechnology Lab, Plant and AgriBiosciences Research Centre, School of Natural Sciences, National University of Ireland Galway, University Road Galway, Ireland
| | - David A Magee
- Animal Genomics Laboratory, UCD School of Agriculture and Food Science, University College Dublin Dublin, Ireland
| | - Stephen D E Park
- IdentiGEN Ltd, Unit 2, Trinity Enterprise Centre Dublin, Ireland
| | | | - Sinead M Waters
- Animal and Bioscience Research Department, Animal and Grassland Research and Innovation Centre, Teagasc Dunsany, Ireland
| | - David E MacHugh
- Animal Genomics Laboratory, UCD School of Agriculture and Food Science, University College DublinDublin, Ireland; UCD Conway Institute of Biomolecular and Biomedical Research, University College DublinDublin, Ireland
| | - Charles Spillane
- Genetics and Biotechnology Lab, Plant and AgriBiosciences Research Centre, School of Natural Sciences, National University of Ireland Galway, University Road Galway, Ireland
| |
Collapse
|