1
|
Tian Y, Wu X, Luo S, Xiong D, Liu R, Hu L, Yuan Y, Shi G, Yao J, Huang Z, Fu F, Yang X, Tang Z, Zhang J, Hu K. A multi-omic single-cell landscape of cellular diversification in the developing human cerebral cortex. Comput Struct Biotechnol J 2024; 23:2173-2189. [PMID: 38827229 PMCID: PMC11141146 DOI: 10.1016/j.csbj.2024.05.019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2024] [Revised: 05/09/2024] [Accepted: 05/13/2024] [Indexed: 06/04/2024] Open
Abstract
The vast neuronal diversity in the human neocortex is vital for high-order brain functions, necessitating elucidation of the regulatory mechanisms underlying such unparalleled diversity. However, recent studies have yet to comprehensively reveal the diversity of neurons and the molecular logic of neocortical origin in humans at single-cell resolution through profiling transcriptomic or epigenomic landscapes, owing to the application of unimodal data alone to depict exceedingly heterogeneous populations of neurons. In this study, we generated a comprehensive compendium of the developing human neocortex by simultaneously profiling gene expression and open chromatin from the same cell. We computationally reconstructed the differentiation trajectories of excitatory projection neurons of cortical origin and inferred the regulatory logic governing lineage bifurcation decisions for neuronal diversification. We demonstrated that neuronal diversity arises from progenitor cell lineage specificity and postmitotic differentiation at distinct stages. Our data paves the way for understanding the primarily coordinated regulatory logic for neuronal diversification in the neocortex.
Collapse
Affiliation(s)
- Yuhan Tian
- Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510275, China
| | - Xia Wu
- Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510275, China
| | - Songhao Luo
- School of Mathematics, Sun Yat-sen University, Guangzhou 510275, China
| | - Dan Xiong
- Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510275, China
| | - Rong Liu
- Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510275, China
| | - Lanqi Hu
- Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510275, China
| | - Yuchen Yuan
- Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510275, China
| | - Guowei Shi
- Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510275, China
| | - Junjie Yao
- Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510275, China
| | - Zhiwei Huang
- School of Mathematics, Sun Yat-sen University, Guangzhou 510275, China
| | - Fang Fu
- Guangzhou Women and Children’s Medical Center, Guangzhou Medical University, Guangzhou 511436, China
| | - Xin Yang
- Guangzhou Women and Children’s Medical Center, Guangzhou Medical University, Guangzhou 511436, China
| | - Zhonghui Tang
- Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510275, China
| | - Jiajun Zhang
- School of Mathematics, Sun Yat-sen University, Guangzhou 510275, China
| | - Kunhua Hu
- Guangdong Provincial Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510275, China
- Public Platform Laboratory, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou 510630, China
| |
Collapse
|
2
|
Wood JA, Chaparala S, Bantang C, Chattopadhyay A, Wesesky MA, Kinchington PR, Nimgaonkar VL, Bloom DC, D'Aiuto L. RNA-Seq time-course analysis of neural precursor cell transcriptome in response to herpes simplex Virus-1 infection. J Neurovirol 2024; 30:131-145. [PMID: 38478163 PMCID: PMC11371869 DOI: 10.1007/s13365-024-01198-8] [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: 11/27/2023] [Revised: 02/12/2024] [Accepted: 02/20/2024] [Indexed: 09/04/2024]
Abstract
The neurogenic niches within the central nervous system serve as essential reservoirs for neural precursor cells (NPCs), playing a crucial role in neurogenesis. However, these NPCs are particularly vulnerable to infection by the herpes simplex virus 1 (HSV-1). In the present study, we investigated the changes in the transcriptome of NPCs in response to HSV-1 infection using bulk RNA-Seq, compared to those of uninfected samples, at different time points post infection and in the presence or absence of antivirals. The results showed that NPCs upon HSV-1 infection undergo a significant dysregulation of genes playing a crucial role in aspects of neurogenesis, including genes affecting NPC proliferation, migration, and differentiation. Our analysis revealed that the CREB signaling, which plays a crucial role in the regulation of neurogenesis and memory consolidation, was the most consistantly downregulated pathway, even in the presence of antivirals. Additionally, cholesterol biosynthesis was significantly downregulated in HSV-1-infected NPCs. The findings from this study, for the first time, offer insights into the intricate molecular mechanisms that underlie the neurogenesis impairment associated with HSV-1 infection.
Collapse
Affiliation(s)
- Joel A Wood
- Western Psychiatric Institute and Clinic, Department of Psychiatry, University of Pittsburgh School of Medicine, 3811 O'Hara Street, 15213, Pittsburgh, PA, USA
| | - Srilakshmi Chaparala
- Molecular Biology Information Service, Health Sciences Library System / Falk Library, University of Pittsburgh, M722 Alan Magee Scaife Hall / 3550 Terrace Street, 15261, Pittsburgh, PA, USA
| | - Cecilia Bantang
- Western Psychiatric Institute and Clinic, Department of Psychiatry, University of Pittsburgh School of Medicine, 3811 O'Hara Street, 15213, Pittsburgh, PA, USA
| | - Ansuman Chattopadhyay
- Molecular Biology Information Service, Health Sciences Library System / Falk Library, University of Pittsburgh, M722 Alan Magee Scaife Hall / 3550 Terrace Street, 15261, Pittsburgh, PA, USA
| | - Maribeth A Wesesky
- Western Psychiatric Institute and Clinic, Department of Psychiatry, University of Pittsburgh School of Medicine, 3811 O'Hara Street, 15213, Pittsburgh, PA, USA
| | - Paul R Kinchington
- Department of Ophthalmology, University of Pittsburgh, Suite 820, Eye & Ear Building, 203 Lothrop Street, 15213, Pittsburgh, PA, USA
| | - Vishwajit L Nimgaonkar
- Western Psychiatric Institute and Clinic, Department of Psychiatry, University of Pittsburgh School of Medicine, 3811 O'Hara Street, 15213, Pittsburgh, PA, USA
- VA Pittsburgh Healthcare system at U.S. Department of Veterans Affairs, Pittsburgh, PA, USA
| | - David C Bloom
- Academic Research Building, Department of Molecular Genetics and Microbiology, University of Florida, 1200 Newell Drive, R2-231, 32610, Gainesville, FL, USA
| | - Leonardo D'Aiuto
- Western Psychiatric Institute and Clinic, Department of Psychiatry, University of Pittsburgh School of Medicine, 3811 O'Hara Street, 15213, Pittsburgh, PA, USA.
| |
Collapse
|
3
|
Zaki ZMM, Ali SA, Ghazali MM, Jam FA. Genetic Modifications of Developmental Dyslexia and Its Representation Using In Vivo, In Vitro Model. Glob Med Genet 2024; 11:76-85. [PMID: 38414980 PMCID: PMC10898997 DOI: 10.1055/s-0044-1781456] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/29/2024] Open
Abstract
Dyslexia is a genetic and heritable disorder that has yet to discover the treatment of it, especially at the molecular and drug intervention levels. This review provides an overview of the current findings on the environmental and genetic factors involved in developmental dyslexia. The latest techniques used in diagnosing the disease and macromolecular factors findings may contribute to a higher degree of development in detangling the proper management and treatment for dyslexic individuals. Furthermore, this review tried to put together all the models used in the current dyslexia research for references in future studies that include animal models as well as in vitro models and how the previous research has provided consistent data across many years and regions. Thus, we suggest furthering the studies using an organoid model based on the existing gene polymorphism, pathways, and neuronal function input.
Collapse
Affiliation(s)
- Zakiyyah M M Zaki
- Department of Neurosciences, School of Medical Sciences, Universiti Sains Malaysia, Kelantan, Malaysia
| | - Siti A Ali
- Department of Electronic Engineering, Faculty of Engineering and Green Technology, Universiti Tunku Abdul Rahman, Kampar, Perak, Malaysia
- Centre for Healthcare Science and Technology, Universiti Tunku Abdul Rahman, Kampar, Perak, Malaysia
| | - Mazira M Ghazali
- Department of Neurosciences, School of Medical Sciences, Universiti Sains Malaysia, Kelantan, Malaysia
- Faculty of Medicine, Universiti Sultan Zainal Abidin, Terengganu, Malaysia
| | - Faidruz A Jam
- Department of Biochemistry, Faculty of Medicine, Manipal University College Malaysia, Melaka, Malaysia
| |
Collapse
|
4
|
Leung CS, Rosenzweig SJ, Yoon B, Marinelli NA, Hollingsworth EW, Maguire AM, Cowen MH, Schmidt M, Imitola J, Gamsiz Uzun ED, Lizarraga SB. Dysregulation of the chromatin environment leads to differential alternative splicing as a mechanism of disease in a human model of autism spectrum disorder. Hum Mol Genet 2023; 32:1634-1646. [PMID: 36621967 PMCID: PMC10162432 DOI: 10.1093/hmg/ddad002] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2022] [Revised: 12/24/2022] [Accepted: 01/03/2023] [Indexed: 01/10/2023] Open
Abstract
Autism spectrum disorder (ASD) affects 1 in 44 children. Chromatin regulatory proteins are overrepresented among genes that contain high risk variants in ASD. Disruption of the chromatin environment leads to widespread dysregulation of gene expression, which is traditionally thought of as a mechanism of disease pathogenesis associated with ASD. Alternatively, alterations in chromatin dynamics could also lead to dysregulation of alternative splicing, which is understudied as a mechanism of ASD pathogenesis. The anticonvulsant valproic acid (VPA) is a well-known environmental risk factor for ASD that acts as a class I histone deacetylase inhibitor. However, the precise molecular mechanisms underlying defects in human neuronal development associated with exposure to VPA are understudied. To dissect how VPA exposure and subsequent chromatin hyperacetylation influence molecular signatures involved in ASD pathogenesis, we conducted RNA sequencing (RNA-seq) in human cortical neurons that were treated with VPA. We observed that differentially expressed genes (DEGs) were enriched for mRNA splicing, mRNA processing, histone modification and metabolism related gene sets. Furthermore, we observed widespread increases in the number and the type of alternative splicing events. Analysis of differential transcript usage (DTU) showed that exposure to VPA induces extensive alterations in transcript isoform usage across neurodevelopmentally important genes. Finally, we find that DEGs and genes that display DTU overlap with known ASD-risk genes. Altogether, these findings suggest that, in addition to differential gene expression, changes in alternative splicing correlated with alterations in the chromatin environment could act as an additional mechanism of disease in ASD.
Collapse
Affiliation(s)
- Calvin S Leung
- Department of Molecular Biology, Cell Biology and Biochemistry, Brown University, Providence, RI 02912, USA
- Center for Translational Neuroscience, Carney Institute for Brain Science and Brown Institute for Translational Science (BITS), Brown University, Providence, RI 02912, USA
| | - Shoshana J Rosenzweig
- Center for Computational Molecular Biology, Brown University, Providence, RI 02906, USA
- Department of Pathology and Laboratory Medicine, Warren Alpert Medical School of Brown University, Providence, RI 02912, USA
- Department of Pathology and Laboratory Medicine, Rhode Island Hospital and Lifespan Academic Medical Center, Providence, RI 02903, USA
| | - Brian Yoon
- Department of Biological Sciences, University of South Carolina, Columbia, SC 29208, USA
| | - Nicholas A Marinelli
- Department of Biological Sciences, University of South Carolina, Columbia, SC 29208, USA
| | - Ethan W Hollingsworth
- UCONN Health Comprehensive Multiple Sclerosis Center, Department of Neurology, University of Connecticut School of Medicine, Farmington, CT 06030, USA
- Division of Multiple Sclerosis and Translational Neuroimmunology, Department of Neurology, University of Connecticut School of Medicine, Farmington, CT 06030, USA
| | - Abbie M Maguire
- Department of Molecular Biology, Cell Biology and Biochemistry, Brown University, Providence, RI 02912, USA
- Center for Translational Neuroscience, Carney Institute for Brain Science and Brown Institute for Translational Science (BITS), Brown University, Providence, RI 02912, USA
| | - Mara H Cowen
- Department of Biological Sciences, University of South Carolina, Columbia, SC 29208, USA
| | - Michael Schmidt
- Department of Molecular Biology, Cell Biology and Biochemistry, Brown University, Providence, RI 02912, USA
- Center for Translational Neuroscience, Carney Institute for Brain Science and Brown Institute for Translational Science (BITS), Brown University, Providence, RI 02912, USA
| | - Jaime Imitola
- UCONN Health Comprehensive Multiple Sclerosis Center, Department of Neurology, University of Connecticut School of Medicine, Farmington, CT 06030, USA
- Division of Multiple Sclerosis and Translational Neuroimmunology, Department of Neurology, University of Connecticut School of Medicine, Farmington, CT 06030, USA
| | - Ece D Gamsiz Uzun
- Center for Computational Molecular Biology, Brown University, Providence, RI 02906, USA
- Department of Pathology and Laboratory Medicine, Warren Alpert Medical School of Brown University, Providence, RI 02912, USA
- Department of Pathology and Laboratory Medicine, Rhode Island Hospital and Lifespan Academic Medical Center, Providence, RI 02903, USA
| | - Sofia B Lizarraga
- Department of Molecular Biology, Cell Biology and Biochemistry, Brown University, Providence, RI 02912, USA
- Center for Translational Neuroscience, Carney Institute for Brain Science and Brown Institute for Translational Science (BITS), Brown University, Providence, RI 02912, USA
| |
Collapse
|
5
|
Liu T, Yang Z, Liu S, Wei J. Parkinson's Disease as a Risk Factor for Prostate Adenocarcinoma: A Molecular Point of View. Gerontology 2023; 69:986-1001. [PMID: 36921580 DOI: 10.1159/000530088] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2022] [Accepted: 02/23/2023] [Indexed: 03/17/2023] Open
Abstract
INTRODUCTION Cancer and neurodegeneration are two major leading causes of morbidity and death worldwide. Neurodegeneration results in excessive neuronal cell death, and cancer emerges from increased proliferation and resistance to cell death. Although most epidemiological studies support an inverse association between the risk for the development of neurodegenerative diseases and cancer, increasing evidence points to a positive correlation between specific types of cancer, like prostate adenocarcinoma (PRAD), and neurodegenerative diseases, like Parkinson's disease (PD). METHODS PD and PRAD differential genes were screened through the GEO database, and the differential genes were analyzed using David, String, GEPIA, Kaplan-Meier plotter, TIMER2.0, proteinatlas, cBioPortal, and CTD databases to elucidate the biological function and molecular mechanism of PD and PRAD-related genes. RESULTS Studies have shown that the hub gene and differentially expressed genes (DEGs) in PD were differentially expressed in PRAD, including CDC20, HSPA4L, ROBO1, DMKN, IFI27L2, LUZP2, PTN, PTGDS. In PRAD, the high expression of HSPA4L, ROBO1, DMKN, IFI27L2, PTN, and PTGDS genes was associated with longer survival, while the patients with low expression of CDC20 and LUZP2 genes had longer survival. The mRNA of CDC20 and LUZP2 were highly expressed, while the mRNAs of HSPA4L, ROBO1, DMKN, IFI27L2, and PTGDS were low expressed. Gene methylation did not affect the survival of patients. The high expression of miR-142, miR-186, miR-30a, miR-497, miR-590, miR-28, and miR-576 in microRNA (miRNA) might potentially be used as biomarkers for the progression of PD and PRAD and for the early diagnosis of PD and PRAD in the populations. The genes in this study were highly associated with B cells, CD8+ T cells, CD4+ T cells, macrophages, neutrophils, and dendritic cells. Somatic mutation mainly focused on missense mutation. Therapeutic drugs included acetaminophen and valproic acid (VPA). CONCLUSION Bioinformatics was used to identify potential targets and novel molecular mechanisms that may serve as clinical markers for the diagnosis and treatment of PD and PRAD.
Collapse
Affiliation(s)
- Tingting Liu
- Institute for Brain Sciences Research, School of Life Sciences, Henan University, Kaifeng, China
| | - Zhengjia Yang
- Institute for Brain Sciences Research, School of Life Sciences, Henan University, Kaifeng, China
| | - Shufen Liu
- Institute for Brain Sciences Research, School of Life Sciences, Henan University, Kaifeng, China
| | - Jianshe Wei
- Institute for Brain Sciences Research, School of Life Sciences, Henan University, Kaifeng, China
| |
Collapse
|
6
|
Price KM, Wigg KG, Eising E, Feng Y, Blokland K, Wilkinson M, Kerr EN, Guger SL, Fisher SE, Lovett MW, Strug LJ, Barr CL. Hypothesis-driven genome-wide association studies provide novel insights into genetics of reading disabilities. Transl Psychiatry 2022; 12:495. [PMID: 36446759 PMCID: PMC9709072 DOI: 10.1038/s41398-022-02250-z] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/10/2022] [Revised: 10/24/2022] [Accepted: 11/03/2022] [Indexed: 11/30/2022] Open
Abstract
Reading Disability (RD) is often characterized by difficulties in the phonology of the language. While the molecular mechanisms underlying it are largely undetermined, loci are being revealed by genome-wide association studies (GWAS). In a previous GWAS for word reading (Price, 2020), we observed that top single-nucleotide polymorphisms (SNPs) were located near to or in genes involved in neuronal migration/axon guidance (NM/AG) or loci implicated in autism spectrum disorder (ASD). A prominent theory of RD etiology posits that it involves disturbed neuronal migration, while potential links between RD-ASD have not been extensively investigated. To improve power to identify associated loci, we up-weighted variants involved in NM/AG or ASD, separately, and performed a new Hypothesis-Driven (HD)-GWAS. The approach was applied to a Toronto RD sample and a meta-analysis of the GenLang Consortium. For the Toronto sample (n = 624), no SNPs reached significance; however, by gene-set analysis, the joint contribution of ASD-related genes passed the threshold (p~1.45 × 10-2, threshold = 2.5 × 10-2). For the GenLang Cohort (n = 26,558), SNPs in DOCK7 and CDH4 showed significant association for the NM/AG hypothesis (sFDR q = 1.02 × 10-2). To make the GenLang dataset more similar to Toronto, we repeated the analysis restricting to samples selected for reading/language deficits (n = 4152). In this GenLang selected subset, we found significant association for a locus intergenic between BTG3-C21orf91 for both hypotheses (sFDR q < 9.00 × 10-4). This study contributes candidate loci to the genetics of word reading. Data also suggest that, although different variants may be involved, alleles implicated in ASD risk may be found in the same genes as those implicated in word reading. This finding is limited to the Toronto sample suggesting that ascertainment influences genetic associations.
Collapse
Affiliation(s)
- Kaitlyn M Price
- Division of Experimental and Translational Neuroscience, Krembil Research Institute, University Health Network, Toronto, Ontario, Canada
- Program in Neuroscience and Mental Health, Hospital for Sick Children, Toronto, Ontario, Canada
- Department of Physiology, University of Toronto, Toronto, Ontario, Canada
| | - Karen G Wigg
- Division of Experimental and Translational Neuroscience, Krembil Research Institute, University Health Network, Toronto, Ontario, Canada
| | - Else Eising
- Language and Genetics Department, Max Planck Institute for Psycholinguistics, Nijmegen, The Netherlands
| | - Yu Feng
- Division of Experimental and Translational Neuroscience, Krembil Research Institute, University Health Network, Toronto, Ontario, Canada
| | - Kirsten Blokland
- Program in Neuroscience and Mental Health, Hospital for Sick Children, Toronto, Ontario, Canada
| | - Margaret Wilkinson
- Program in Neuroscience and Mental Health, Hospital for Sick Children, Toronto, Ontario, Canada
| | - Elizabeth N Kerr
- Department of Psychology, Hospital for Sick Children, Toronto, Ontario, Canada
- Department of Pediatrics, University of Toronto, Toronto, Ontario, Canada
| | - Sharon L Guger
- Department of Psychology, Hospital for Sick Children, Toronto, Ontario, Canada
| | - Simon E Fisher
- Language and Genetics Department, Max Planck Institute for Psycholinguistics, Nijmegen, The Netherlands
- Donders Institute for Brain, Cognition and Behaviour, Radboud University, Nijmegen, The Netherlands
| | - Maureen W Lovett
- Program in Neuroscience and Mental Health, Hospital for Sick Children, Toronto, Ontario, Canada
- Department of Pediatrics, University of Toronto, Toronto, Ontario, Canada
| | - Lisa J Strug
- Genetics and Genome Biology, Hospital for Sick Children, Toronto, Ontario, Canada
- Departments of Statistical Sciences and Computer Science, Faculty of Arts and Science and Division of Biostatistics, Dalla Lana School of Public Health, University of Toronto, Toronto, Ontario, Canada
| | - Cathy L Barr
- Division of Experimental and Translational Neuroscience, Krembil Research Institute, University Health Network, Toronto, Ontario, Canada.
- Program in Neuroscience and Mental Health, Hospital for Sick Children, Toronto, Ontario, Canada.
- Department of Physiology, University of Toronto, Toronto, Ontario, Canada.
| |
Collapse
|
7
|
Kim SH, Kim TJ, Shin D, Hur KJ, Hong SH, Lee JY, Ha US. ROBO1 protein expression is independently associated with biochemical recurrence in prostate cancer patients who underwent radical prostatectomy in Asian patients. Gland Surg 2021; 10:2956-2965. [PMID: 34804883 DOI: 10.21037/gs-21-406] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2021] [Accepted: 09/03/2021] [Indexed: 11/06/2022]
Abstract
Background The purpose of this study is to investigate the correlation between ROBO1 expression and prostate cancer aggressiveness. Methods ROBO1 expression was evaluated in normal prostate epithelial cells (PrEC) and different prostate cancer cell lines by Western blot analysis. The migration and invasion of native and ROBO1 knockdown cells were evaluated using migration chambers and a Matrigel-coated membrane, respectively. Samples from 145 patients who underwent radical prostatectomy between June 2000 and June 2008, were retrieved from the paraffin files for tissue microarray (TMA) with immunohistochemical analysis. Biochemical recurrence (BCR)-free survival curves were estimated using the Kaplan-Meier and Cox regression methods in two groups of patients classified according to the degree of ROBO1 expression (low or high expression). Results ROBO1 is highly expressed in the prostate cancer cell lines. All ROBO1 knockdown cells (PC3, 22Rv1 and DU 145) showed markedly decreased migration and invasiveness compared to native cells. In 145 patients with radical prostatectomy, the Kaplan-Meier curves and log-rank test for BCR-free survival stratified by ROBO1 expression in organ-confined (pT2) or not (pT3), showed significant differences in 10-year survival between the ROBO1 high and low expression groups (87.2% versus 52.6% in pT2; P=0.047, 51.0% versus 36.9% in pT3; P=0.033). The multivariable-adjusted model showed a markedly increased hazard ratio (HR) in patients with high ROBO1 expression compared to the patients with low ROBO1expression in every model. Conclusions ROBO1 may play an important role in the migration and invasion of prostate cancer cells, and was independently associated with BCR.
Collapse
Affiliation(s)
- Sang Hoon Kim
- Department of Urology, Eunpyeong St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Tae-Jung Kim
- Department of Hospital Pathology, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Dongho Shin
- Department of Urology, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Kyung Jae Hur
- Department of Urology, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Sung-Hoo Hong
- Department of Urology, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Ji Youl Lee
- Department of Urology, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - U-Syn Ha
- Department of Urology, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea.,Cancer Research Institute, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| |
Collapse
|
8
|
Franchini LF. Genetic Mechanisms Underlying Cortical Evolution in Mammals. Front Cell Dev Biol 2021; 9:591017. [PMID: 33659245 PMCID: PMC7917222 DOI: 10.3389/fcell.2021.591017] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2020] [Accepted: 01/08/2021] [Indexed: 12/13/2022] Open
Abstract
The remarkable sensory, motor, and cognitive abilities of mammals mainly depend on the neocortex. Thus, the emergence of the six-layered neocortex in reptilian ancestors of mammals constitutes a fundamental evolutionary landmark. The mammalian cortex is a columnar epithelium of densely packed cells organized in layers where neurons are generated mainly in the subventricular zone in successive waves throughout development. Newborn cells move away from their site of neurogenesis through radial or tangential migration to reach their specific destination closer to the pial surface of the same or different cortical area. Interestingly, the genetic programs underlying neocortical development diversified in different mammalian lineages. In this work, I will review several recent studies that characterized how distinct transcriptional programs relate to the development and functional organization of the neocortex across diverse mammalian lineages. In some primates such as the anthropoids, the neocortex became extremely large, especially in humans where it comprises around 80% of the brain. It has been hypothesized that the massive expansion of the cortical surface and elaboration of its connections in the human lineage, has enabled our unique cognitive capacities including abstract thinking, long-term planning, verbal language and elaborated tool making capabilities. I will also analyze the lineage-specific genetic changes that could have led to the modification of key neurodevelopmental events, including regulation of cell number, neuronal migration, and differentiation into specific phenotypes, in order to shed light on the evolutionary mechanisms underlying the diversity of mammalian brains including the human brain.
Collapse
Affiliation(s)
- Lucía Florencia Franchini
- Instituto de Investigaciones en Ingeniería Genética y Biología Molecular (INGEBI), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
| |
Collapse
|
9
|
Lin HY, Wang X, Tseng TS, Kao YH, Fang Z, Molina PE, Cheng CH, Berglund AE, Eeles RA, Muir KR, Pashayan N, Haiman CA, Brenner H, Consortium TP, Park JY. Alcohol Intake and Alcohol-SNP Interactions Associated with Prostate Cancer Aggressiveness. J Clin Med 2021; 10:553. [PMID: 33540941 PMCID: PMC7867322 DOI: 10.3390/jcm10030553] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2020] [Revised: 01/25/2021] [Accepted: 01/28/2021] [Indexed: 12/24/2022] Open
Abstract
Excessive alcohol intake is a well-known modifiable risk factor for many cancers. It is still unclear whether genetic variants or single nucleotide polymorphisms (SNPs) can modify alcohol intake's impact on prostate cancer (PCa) aggressiveness. The objective is to test the alcohol-SNP interactions of the 7501 SNPs in the four pathways (angiogenesis, mitochondria, miRNA, and androgen metabolism-related pathways) associated with PCa aggressiveness. We evaluated the impacts of three excessive alcohol intake behaviors in 3306 PCa patients with European ancestry from the PCa Consortium. We tested the alcohol-SNP interactions using logistic models with the discovery-validation study design. All three excessive alcohol intake behaviors were not significantly associated with PCa aggressiveness. However, the interactions of excessive alcohol intake and three SNPs (rs13107662 [CAMK2D, p = 6.2 × 10-6], rs9907521 [PRKCA, p = 7.1 × 10-5], and rs11925452 [ROBO1, p = 8.2 × 10-4]) were significantly associated with PCa aggressiveness. These alcohol-SNP interactions revealed contrasting effects of excessive alcohol intake on PCa aggressiveness according to the genotypes in the identified SNPs. We identified PCa patients with the rs13107662 (CAMK2D) AA genotype, the rs11925452 (ROBO1) AA genotype, and the rs9907521 (PRKCA) AG genotype were more vulnerable to excessive alcohol intake for developing aggressive PCa. Our findings support that the impact of excessive alcohol intake on PCa aggressiveness was varied by the selected genetic profiles.
Collapse
Affiliation(s)
- Hui-Yi Lin
- Biostatistics Program, School of Public Health, Louisiana State University Health Sciences Center, New Orleans, LA 70112, USA
| | - Xinnan Wang
- Biostatistics Program, School of Public Health, Louisiana State University Health Sciences Center, New Orleans, LA 70112, USA
| | - Tung-Sung Tseng
- Behavioral and Community Health Sciences Program, School of Public Health, Louisiana State University Health Sciences Center, New Orleans, LA 70112, USA
| | - Yu-Hsiang Kao
- Behavioral and Community Health Sciences Program, School of Public Health, Louisiana State University Health Sciences Center, New Orleans, LA 70112, USA
| | - Zhide Fang
- Biostatistics Program, School of Public Health, Louisiana State University Health Sciences Center, New Orleans, LA 70112, USA
| | - Patricia E Molina
- Department of Physiology, School of Medicine, Louisiana State University Health Sciences Center, New Orleans, LA 70112, USA
- Comprehensive Alcohol Research Center, School of Medicine, Louisiana State University Health Sciences Center, New Orleans, LA 70112, USA
| | - Chia-Ho Cheng
- Biostatistics and Bioinformatics Shared Resource, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL 33612, USA
| | - Anders E Berglund
- Department of Biostatistics and Bioinformatics, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL 33612, USA
| | - Rosalind A Eeles
- The Institute of Cancer Research, and The Royal Marsden NHS Foundation Trust, London, SM2 5NG, UK
| | - Kenneth R Muir
- Division of Population Health, Health Services Research, and Primary Care, University of Manchester, Oxford Road, Manchester, M139PT, UK
| | - Nora Pashayan
- Department of Applied Health Research, University College London, WC1E 7HB, London, UK
| | - Christopher A Haiman
- Center for Genetic Epidemiology, Department of Preventive Medicine, Keck School of Medicine, University of Southern California/Norris Comprehensive Cancer Center, Los Angeles, CA 90015, USA
| | - Hermann Brenner
- Division of Clinical Epidemiology and Aging Research, German Cancer Research Center (DKFZ), D-69120 Heidelberg, Germany
- Division of Preventive Oncology, German Cancer Research Center (DKFZ) and National Center for Tumor Diseases (NCT), D-69120 Heidelberg, Germany
- German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), D-69120 Heidelberg, Germany
| | - The Practical Consortium
- The Prostate Cancer Association Group to Investigate Cancer Associated Alterations in the Genome Consortium (PRACTICAL, http://practical.icr.ac.uk/), London SM2 5NG, UK. Additional members from The PRACTICAL Consortium were provided in the Supplement
| | - Jong Y Park
- Department of Cancer Epidemiology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL 33612, USA
| |
Collapse
|
10
|
Gonda Y, Namba T, Hanashima C. Beyond Axon Guidance: Roles of Slit-Robo Signaling in Neocortical Formation. Front Cell Dev Biol 2020; 8:607415. [PMID: 33425915 PMCID: PMC7785817 DOI: 10.3389/fcell.2020.607415] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2020] [Accepted: 12/07/2020] [Indexed: 12/11/2022] Open
Abstract
The formation of the neocortex relies on intracellular and extracellular signaling molecules that are involved in the sequential steps of corticogenesis, ranging from the proliferation and differentiation of neural progenitor cells to the migration and dendrite formation of neocortical neurons. Abnormalities in these steps lead to disruption of the cortical structure and circuit, and underly various neurodevelopmental diseases, including dyslexia and autism spectrum disorder (ASD). In this review, we focus on the axon guidance signaling Slit-Robo, and address the multifaceted roles of Slit-Robo signaling in neocortical development. Recent studies have clarified the roles of Slit-Robo signaling not only in axon guidance but also in progenitor cell proliferation and migration, and the maturation of neocortical neurons. We further discuss the etiology of neurodevelopmental diseases, which are caused by defects in Slit-Robo signaling during neocortical formation.
Collapse
Affiliation(s)
- Yuko Gonda
- Department of Histology and Neuroanatomy, Tokyo Medical University, Tokyo, Japan
| | - Takashi Namba
- Max Planck Institute of Molecular Cell Biology and Genetics, Dresden, Germany
- Neuroscience Center, HiLIFE – Helsinki Institute of Life Science, University of Helsinki, Helsinki, Finland
| | - Carina Hanashima
- Faculty of Education and Integrated Arts and Sciences, Waseda University, Tokyo, Japan
- Graduate School of Advanced Science and Engineering, Waseda University, Tokyo, Japan
| |
Collapse
|
11
|
Mascheretti S, Riva V, Feng B, Trezzi V, Andreola C, Giorda R, Villa M, Dionne G, Gori S, Marino C, Facoetti A. The Mediation Role of Dynamic Multisensory Processing Using Molecular Genetic Data in Dyslexia. Brain Sci 2020; 10:brainsci10120993. [PMID: 33339203 PMCID: PMC7765588 DOI: 10.3390/brainsci10120993] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2020] [Revised: 12/04/2020] [Accepted: 12/11/2020] [Indexed: 12/21/2022] Open
Abstract
Although substantial heritability has been reported and candidate genes have been identified, we are far from understanding the etiopathogenetic pathways underlying developmental dyslexia (DD). Reading-related endophenotypes (EPs) have been established. Until now it was unknown whether they mediated the pathway from gene to reading (dis)ability. Thus, in a sample of 223 siblings from nuclear families with DD and 79 unrelated typical readers, we tested four EPs (i.e., rapid auditory processing, rapid automatized naming, multisensory nonspatial attention and visual motion processing) and 20 markers spanning five DD-candidate genes (i.e., DYX1C1, DCDC2, KIAA0319, ROBO1 and GRIN2B) using a multiple-predictor/multiple-mediator framework. Our results show that rapid auditory and visual motion processing are mediators in the pathway from ROBO1-rs9853895 to reading. Specifically, the T/T genotype group predicts impairments in rapid auditory and visual motion processing which, in turn, predict poorer reading skills. Our results suggest that ROBO1 is related to reading via multisensory temporal processing. These findings support the use of EPs as an effective approach to disentangling the complex pathways between candidate genes and behavior.
Collapse
Affiliation(s)
- Sara Mascheretti
- Child Psychopathology Unit, Scientific Institute, IRCCS E. Medea, 23842 Bosisio Parini, Italy; (S.M.); (V.R.); (V.T.); (C.A.)
| | - Valentina Riva
- Child Psychopathology Unit, Scientific Institute, IRCCS E. Medea, 23842 Bosisio Parini, Italy; (S.M.); (V.R.); (V.T.); (C.A.)
| | - Bei Feng
- École de Psychologie, Laval University, Québec, QC G1V 0A6, Canada; (B.F.); (G.D.)
| | - Vittoria Trezzi
- Child Psychopathology Unit, Scientific Institute, IRCCS E. Medea, 23842 Bosisio Parini, Italy; (S.M.); (V.R.); (V.T.); (C.A.)
| | - Chiara Andreola
- Child Psychopathology Unit, Scientific Institute, IRCCS E. Medea, 23842 Bosisio Parini, Italy; (S.M.); (V.R.); (V.T.); (C.A.)
- Laboratoire de Psychologie du Développement et de l’Éducation de l’Enfant (LaPsyDÉ), Universitè de Paris, 75005 Paris, France
| | - Roberto Giorda
- Molecular Biology Laboratory, Scientific Institute, IRCCS E. Medea, 23842 Bosisio Parini, Italy; (R.G.); (M.V.)
| | - Marco Villa
- Molecular Biology Laboratory, Scientific Institute, IRCCS E. Medea, 23842 Bosisio Parini, Italy; (R.G.); (M.V.)
| | - Ginette Dionne
- École de Psychologie, Laval University, Québec, QC G1V 0A6, Canada; (B.F.); (G.D.)
| | - Simone Gori
- Department of Human and Social Sciences, University of Bergamo, 24100 Bergamo, Italy;
| | - Cecilia Marino
- Child Psychopathology Unit, Scientific Institute, IRCCS E. Medea, 23842 Bosisio Parini, Italy; (S.M.); (V.R.); (V.T.); (C.A.)
- Department of Psychiatry, University of Toronto, Toronto, ON M5T 1R8, Canada
- The Division of Child and Youth Psychiatry, Centre for Addiction and Mental Health (CAMH), Toronto, ON M6J 1H4, Canada
- Correspondence: (C.M.); (A.F.)
| | - Andrea Facoetti
- Developmental Cognitive Neuroscience Lab, Department of General Psychology, University of Padua, 35131 Padua, Italy
- Correspondence: (C.M.); (A.F.)
| |
Collapse
|
12
|
Liu JYW, Reeves C, van der Pijl R, Thom M. Glial regenerative cell types in the superficial cortex in cortical dysplasia subtypes. Epilepsy Res 2020; 169:106529. [PMID: 33370704 PMCID: PMC7829594 DOI: 10.1016/j.eplepsyres.2020.106529] [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: 08/07/2020] [Revised: 11/23/2020] [Accepted: 12/04/2020] [Indexed: 10/29/2022]
Abstract
PURPOSE Focal Cortical Dysplasias (FCD) are localized malformative brain lesions in epilepsy. FCD3a associated with hippocampal sclerosis, affects the superficial cortex and is presumed to have an 'acquired' rather than developmental origin. Precursor cells may arise outside neurogenic zones including cortical layer I. Our aim was to characterise subsets of glial progenitor cells in the superficial cortical layers, known to be involved in gliosis and gliogenesis and that could distinguish FCD3a from other subtypes. METHODS Using immunohistochemistry we quantified the density of glial progenitor subsets in superficial cortex layers using markers against PAX6, GFAP, Olig2 and PDGFRβ and proliferation marker MCM2 in ten FCD3a cases compared to 18 other FCD types and 11 non-FCD controls. KEY FINDINGS Glial progenitor cells types were present in the cortical layer I and II in all FCD groups. GFAP cells frequently expressed PAX6 and significantly higher GFAP/PAX6 than GFAP/MCM2 cell densities were identified in the FCD3a group (p < 0.05). Olig2 cell densities were significantly higher in FCD3b than FCD3a (p = 0.002) and significantly higher GFAP/MCM2 compared to PDGFRβ/MCM2 cell densities were identified in both FCD3b and FCD2 groups. There was no correlation between cell densities and the age of patients at surgery and between cortical regions. SIGNIFICANCE Immature and proliferative glial populations across FCD variants reflect reactive cell types and differences may provide insight into underlying pathomechanisms. Higher PAX6 expression in astroglial cells in FCD3a may indicate a switch to astrocytic maturation and enhanced superficial gliosis. Higher Olig2 and GFAP/MCM2 densities in FCD3b may reflect margins of the tumour infiltration zone rather than true cortical dysplasia.
Collapse
Affiliation(s)
- Joan Y W Liu
- Department of Clinical and Experimental Epilepsy, UCL Institute of Neurology and National Hospital for Neurology and Neurosurgery, Queen Square, London, WC1N 3BG, UK; School of Life Sciences, University of Westminster, London, W1W 6UW, UK
| | - Cheryl Reeves
- Department of Clinical and Experimental Epilepsy, UCL Institute of Neurology and National Hospital for Neurology and Neurosurgery, Queen Square, London, WC1N 3BG, UK
| | - Rianne van der Pijl
- Department of Clinical and Experimental Epilepsy, UCL Institute of Neurology and National Hospital for Neurology and Neurosurgery, Queen Square, London, WC1N 3BG, UK
| | - Maria Thom
- Department of Clinical and Experimental Epilepsy, UCL Institute of Neurology and National Hospital for Neurology and Neurosurgery, Queen Square, London, WC1N 3BG, UK; Division of Neuropathology, National Hospital for Neurology and Neurosurgery, Queen Square, London, WC1N 3BG, UK.
| |
Collapse
|
13
|
Reelin-Nrp1 Interaction Regulates Neocortical Dendrite Development in a Context-Specific Manner. J Neurosci 2020; 40:8248-8261. [PMID: 33009002 DOI: 10.1523/jneurosci.1907-20.2020] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2020] [Revised: 09/14/2020] [Accepted: 09/23/2020] [Indexed: 11/21/2022] Open
Abstract
Reelin plays versatile roles in neocortical development. The C-terminal region (CTR) of Reelin is required for the correct formation of the superficial structure of the neocortex; however, the mechanisms by which this position-specific effect occurs remain largely unknown. In this study, we demonstrate that Reelin with an intact CTR binds to neuropilin-1 (Nrp1), a transmembrane protein. Both male and female mice were used. Nrp1 is localized with very-low-density lipoprotein receptor (VLDLR), a canonical Reelin receptor, in the superficial layers of the developing neocortex. It forms a complex with VLDLR, and this interaction is modulated by the alternative splicing of VLDLR. Reelin with an intact CTR binds more strongly to the VLDLR/Nrp1 complex than to VLDLR alone. Knockdown of Nrp1 in neurons leads to the accumulation of Dab1 protein. Since the degradation of Dab1 is induced by Reelin signaling, it is suggested that Nrp1 augments Reelin signaling. The interaction between Reelin and Nrp1 is required for normal dendritic development in superficial-layer neurons. All of these characteristics of Reelin are abrogated by proteolytic processing of the six C-terminal amino acid residues of Reelin (0.17% of the whole protein). Therefore, Nrp1 is a coreceptor molecule for Reelin and, together with the proteolytic processing of Reelin, can account for context-specific Reelin function in brain development.SIGNIFICANCE STATEMENT Reelin often exhibits a context-dependent function during brain development; however, its underlying mechanism is not well understood. We found that neuropilin-1 (Nrp1) specifically binds to the CTR of Reelin and acts as a coreceptor for very-low-density lipoprotein receptor (VLDLR). The Nrp1/VLDLR complex is localized in the superficial layers of the neocortex, and its interaction with Reelin is essential for proper dendritic development in superficial-layer neurons. This study provides the first mechanistic evidence of the context-specific function of Reelin (>3400 residues) regulated by the C-terminal residues and Nrp1, a component of the canonical Reelin receptor complex.
Collapse
|
14
|
Skeide MA, Wehrmann K, Emami Z, Kirsten H, Hartmann AM, Rujescu D. Neurobiological origins of individual differences in mathematical ability. PLoS Biol 2020; 18:e3000871. [PMID: 33090992 PMCID: PMC7580992 DOI: 10.1371/journal.pbio.3000871] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2020] [Accepted: 09/18/2020] [Indexed: 01/23/2023] Open
Abstract
Mathematical ability is heritable and related to several genes expressing proteins in the brain. It is unknown, however, which intermediate neural phenotypes could explain how these genes relate to mathematical ability. Here, we examined genetic effects on cerebral cortical volume of 3-6-year-old children without mathematical training to predict mathematical ability in school at 7-9 years of age. To this end, we followed an exploration sample (n = 101) and an independent replication sample (n = 77). We found that ROBO1, a gene known to regulate prenatal growth of cerebral cortical layers, is associated with the volume of the right parietal cortex, a key region for quantity representation. Individual volume differences in this region predicted up to a fifth of the behavioral variance in mathematical ability. Our findings indicate that a fundamental genetic component of the quantity processing system is rooted in the early development of the parietal cortex.
Collapse
Affiliation(s)
- Michael A. Skeide
- Department of Neuropsychology, Max Planck Institute for Human Cognitive and Brain Sciences, Leipzig, Germany
| | - Katharina Wehrmann
- Department of Neuropsychology, Max Planck Institute for Human Cognitive and Brain Sciences, Leipzig, Germany
- Institute of Psychology, Humboldt University of Berlin, Berlin, Germany
- Department of Psychiatry, University of Bern, Bern, Switzerland
| | - Zahra Emami
- Department of Neuropsychology, Max Planck Institute for Human Cognitive and Brain Sciences, Leipzig, Germany
- The Hospital for Sick Children, Toronto, Canada
| | - Holger Kirsten
- Institute for Medical Informatics, Statistics and Epidemiology, University of Leipzig, Leipzig, Germany
| | - Annette M. Hartmann
- Department of Psychiatry, Psychotherapy and Psychosomatics, Martin-Luther-University Halle-Wittenberg, Halle (Saale), Germany
| | - Dan Rujescu
- Department of Psychiatry, Psychotherapy and Psychosomatics, Martin-Luther-University Halle-Wittenberg, Halle (Saale), Germany
| | | |
Collapse
|
15
|
Ferrari MG, Ganaie AA, Shabenah A, Mansini AP, Wang L, Murugan P, Davicioni E, Wang J, Deng Y, Hoeppner LH, Warlick CA, Konety BR, Saleem M. Identifying and treating ROBO1 -ve /DOCK1 +ve prostate cancer: An aggressive cancer subtype prevalent in African American patients. Prostate 2020; 80:1045-1057. [PMID: 32687658 PMCID: PMC7556361 DOI: 10.1002/pros.24018] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/17/2020] [Accepted: 05/15/2020] [Indexed: 12/19/2022]
Abstract
BACKGROUND There is a need to develop novel therapies which could be beneficial to patients with prostate cancer (CaP) including those who are predisposed to poor outcome, such as African-Americans. This study investigates the role of ROBO1-pathway in predicting outcome and race-based disparity in patients with CaP. METHODS AND RESULTS Aided by RNA sequencing-based DECIPHER-testing and immunohistochemical (IHC) analysis of tumors we show that ROBO1 is lost during the progressive stages of CaP, a prevalent feature in African-Americans. We show that the loss of ROBO1 predicts high-risk of recurrence, metastasis and poor outcome of androgen-deprivation therapy in radical prostatectomy-treated patients. These data identified an aggressive ROBO1deficient /DOCK1+ve sub-class of CaP. Combined genetic and IHC data showed that ROBO1 loss is accompanied by DOCK1/Rac1 elevation in grade-III/IV primary-tumors and Mets. We observed that the hypermethylation of ROBO1-promoter contributes to loss of expression that is highly prevalent in African-Americans. Because of limitations in restoring ROBO1 function, we asked if targeting the DOCK1 could be an ideal strategy to inhibit progression or treat ROBO1deficient metastatic-CaP. We tested the pharmacological efficacy of CPYPP, a selective inhibitor of DOCK1 under in vitro and in vivo conditions. Using ROBO1-ve and ROBO1+ve CaP models, we determined the median effective concentration of CPYPP for growth. DOCK1-inhibitor treatment significantly decreased the (a) Rac1-GTP/β-catenin activity, (b) transmigration of ROBO1deficient cells across endothelial lining, and (c) metastatic spread of ROBO1deficient cells through the vasculature of transgenicfl Zebrafish model. CONCLUSION We suggest that ROBO1 status forms as predictive biomarker of outcome in high-risk populations such as African-Americans and DOCK1-targeting therapy has a clinical potential for treating metastatic-CaP.
Collapse
Affiliation(s)
- Marina G. Ferrari
- Department of Urology, School of Medicine, Masonic Cancer Center, University of Minnesota
| | - Arsheed A. Ganaie
- Department of Urology, School of Medicine, Masonic Cancer Center, University of Minnesota
| | - Ashraf Shabenah
- Institute for Health Informatics, Masonic Cancer Center, University of Minnesota
| | - Adrian P. Mansini
- Department of Urology, School of Medicine, Masonic Cancer Center, University of Minnesota
| | - Li Wang
- Hormel Institute, University of Minnesota, Austin, MN
| | - Paari Murugan
- Department of Laboratory Medicine and Pathology, University of Minnesota
| | | | - Jinhua Wang
- Institute for Health Informatics, Masonic Cancer Center, University of Minnesota
| | - Yibin Deng
- Department of Urology, School of Medicine, Masonic Cancer Center, University of Minnesota
| | | | - Christopher A. Warlick
- Department of Urology, School of Medicine, Masonic Cancer Center, University of Minnesota
| | - Badrinath R. Konety
- Department of Urology, School of Medicine, Masonic Cancer Center, University of Minnesota
| | - Mohammad Saleem
- Department of Urology, School of Medicine, Masonic Cancer Center, University of Minnesota
| |
Collapse
|
16
|
Sasaki T, Komatsu Y, Yamamori T. Expression patterns of SLIT/ROBO mRNAs reveal a characteristic feature in the entorhinal-hippocampal area of macaque monkeys. BMC Res Notes 2020; 13:262. [PMID: 32460877 PMCID: PMC7251749 DOI: 10.1186/s13104-020-05100-7] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [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: 05/19/2020] [Indexed: 01/06/2023] Open
Abstract
OBJECTIVE SLITs are secreted glycoproteins that bind to Roundabouts (ROBOs) which are a family member of transmembrane receptors. SLIT signaling has well-conserved roles in mediating axon repulsion in a developing nervous system. We previously reported that SLIT1 mRNA is enriched in middle layers of the prefrontal cortex of macaque monkeys in a developmentally regulated manner. Other SLIT (SLIT2 and SLIT3) mRNAs showed preferential expressions in the prefrontal cortex with a distinct laminar pattern. To obtain further clues to the role of SLIT signaling in the organization of the primate brain, we performed ISH analysis of SLIT and ROBO mRNAs using adult macaque brain tissues. RESULTS In this study, we examined the expression patterns of SLITs and ROBOs (ROBO1 and ROBO2) in other brain regions, and found intense and characteristic expression patterns of these genes in the entorhinal-hippocampal area. In situ hybridization analysis revealed that SLIT1 and SLIT2 mRNAs showed marked complementary distribution in the entorhinal cortex. SLIT and ROBO mRNAs were widely expressed in the hippocampus with modest regional preference. These findings suggest that each SLIT gene has a specialized role that is particularly important for prefrontal as well as hippocampal connectivity in the primate cortex.
Collapse
Affiliation(s)
- Tetsuya Sasaki
- Division of Brain Biology, National Institute for Basic Biology, 38 Nishigonaka Myodaiji, Okazaki, 444-8585, Japan.
- Department of Anatomy and Neuroscience, Faculty of Medicine, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki, 305-8577, Japan.
- Ph.D Program of Neurosciences, Graduate School of Comprehensive Human Sciences, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki, 305-8577, Japan.
| | - Yusuke Komatsu
- Division of Brain Biology, National Institute for Basic Biology, 38 Nishigonaka Myodaiji, Okazaki, 444-8585, Japan
- Laboratory of Veterinary Hygiene, Graduate School of Veterinary Medicine, Hokkaido University, Sapporo, 060-0816, Japan
| | - Tetsuo Yamamori
- Division of Brain Biology, National Institute for Basic Biology, 38 Nishigonaka Myodaiji, Okazaki, 444-8585, Japan.
- Laboratory for Molecular Analysis of Higher Brain Function, RIKEN Center for Brain Science, Wako, Saitama, 351-0198, Japan.
| |
Collapse
|
17
|
Price KM, Wigg KG, Feng Y, Blokland K, Wilkinson M, He G, Kerr EN, Carter TC, Guger SL, Lovett MW, Strug LJ, Barr CL. Genome-wide association study of word reading: Overlap with risk genes for neurodevelopmental disorders. GENES BRAIN AND BEHAVIOR 2020; 19:e12648. [PMID: 32108986 DOI: 10.1111/gbb.12648] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/22/2019] [Revised: 01/28/2020] [Accepted: 02/24/2020] [Indexed: 12/12/2022]
Abstract
Reading disabilities (RD) are the most common neurocognitive disorder, affecting 5% to 17% of children in North America. These children often have comorbid neurodevelopmental/psychiatric disorders, such as attention deficit/hyperactivity disorder (ADHD). The genetics of RD and their overlap with other disorders is incompletely understood. To contribute to this, we performed a genome-wide association study (GWAS) for word reading. Then, using summary statistics from neurodevelopmental/psychiatric disorders, we computed polygenic risk scores (PRS) and used them to predict reading ability in our samples. This enabled us to test the shared aetiology between RD and other disorders. The GWAS consisted of 5.3 million single nucleotide polymorphisms (SNPs) and two samples; a family-based sample recruited for reading difficulties in Toronto (n = 624) and a population-based sample recruited in Philadelphia [Philadelphia Neurodevelopmental Cohort (PNC)] (n = 4430). The Toronto sample SNP-based analysis identified suggestive SNPs (P ~ 5 × 10-7 ) in the ARHGAP23 gene, which is implicated in neuronal migration/axon pathfinding. The PNC gene-based analysis identified significant associations (P < 2.72 × 10-6 ) for LINC00935 and CCNT1, located in the region of the KANSL2/CCNT1/LINC00935/SNORA2B/SNORA34/MIR4701/ADCY6 genes on chromosome 12q, with near significant SNP-based analysis. PRS identified significant overlap between word reading and intelligence (R2 = 0.18, P = 7.25 × 10-181 ), word reading and educational attainment (R2 = 0.07, P = 4.91 × 10-48 ) and word reading and ADHD (R2 = 0.02, P = 8.70 × 10-6 ; threshold for significance = 7.14 × 10-3 ). Overlap was also found between RD and autism spectrum disorder (ASD) as top-ranked genes were previously implicated in autism by rare and copy number variant analyses. These findings support shared risk between word reading, cognitive measures, educational outcomes and neurodevelopmental disorders, including ASD.
Collapse
Affiliation(s)
- Kaitlyn M Price
- Genetics and Development Division, Krembil Research Institute, University Health Network, Toronto, Ontario, Canada.,Program in Neuroscience and Mental Health, Hospital for Sick Children, Toronto, Ontario, Canada.,Department of Physiology, University of Toronto, Toronto, Ontario, Canada
| | - Karen G Wigg
- Genetics and Development Division, Krembil Research Institute, University Health Network, Toronto, Ontario, Canada
| | - Yu Feng
- Genetics and Development Division, Krembil Research Institute, University Health Network, Toronto, Ontario, Canada
| | - Kirsten Blokland
- Program in Neuroscience and Mental Health, Hospital for Sick Children, Toronto, Ontario, Canada
| | - Margaret Wilkinson
- Program in Neuroscience and Mental Health, Hospital for Sick Children, Toronto, Ontario, Canada
| | - Gengming He
- Genetics and Genome Biology, Hospital for Sick Children, Toronto, Ontario, Canada
| | - Elizabeth N Kerr
- Department of Psychology, Hospital for Sick Children, Toronto, Ontario, Canada.,Department of Pediatrics, University of Toronto, Toronto, Ontario, Canada
| | - Tasha-Cate Carter
- Program in Neuroscience and Mental Health, Hospital for Sick Children, Toronto, Ontario, Canada.,Holland Bloorview Rehabilitation Hospital, Toronto, Ontario, Canada
| | - Sharon L Guger
- Department of Psychology, Hospital for Sick Children, Toronto, Ontario, Canada
| | - Maureen W Lovett
- Program in Neuroscience and Mental Health, Hospital for Sick Children, Toronto, Ontario, Canada.,Department of Pediatrics, University of Toronto, Toronto, Ontario, Canada
| | - Lisa J Strug
- Genetics and Genome Biology, Hospital for Sick Children, Toronto, Ontario, Canada.,Dalla Lana School of Public Health, University of Toronto, Toronto, Ontario, Canada
| | - Cathy L Barr
- Genetics and Development Division, Krembil Research Institute, University Health Network, Toronto, Ontario, Canada.,Program in Neuroscience and Mental Health, Hospital for Sick Children, Toronto, Ontario, Canada.,Department of Physiology, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
18
|
Kaneyama T, Shirasaki R. Post-crossing segment of dI1 commissural axons forms collateral branches to motor neurons in the developing spinal cord. J Comp Neurol 2019; 526:1943-1961. [PMID: 29752714 DOI: 10.1002/cne.24464] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2017] [Revised: 03/30/2018] [Accepted: 05/03/2018] [Indexed: 11/09/2022]
Abstract
The dI1 commissural axons in the developing spinal cord, upon crossing the midline through the floor plate, make a sharp turn to grow rostrally. These post-crossing axons initially just extend adjacent to the floor plate without entering nearby motor columns. However, it remains poorly characterized how these post-crossing dI1 axons behave subsequently to this process. In the present study, to address this issue, we examined in detail the behavior of post-crossing dI1 axons in mice, using the Atoh1 enhancer-based conditional expression system that enables selective and sparse labeling of individual dI1 axons, together with Hb9 and ChAT immunohistochemistry for precise identification of spinal motor neurons (MNs). We found unexpectedly that the post-crossing segment of dI1 axons later gave off collateral branches that extended laterally to invade motor columns. Interestingly, these collateral branches emerged at around the time when their primary growth cones initiated invasion into motor columns. In addition, although the length of the laterally growing collateral branches increased with age, the majority of them remained within motor columns. Strikingly, these collateral branches further gave rise to multiple secondary branches in the region of MNs that innervate muscles close to the body axis. Moreover, these axonal branches formed presynaptic terminals on MNs. These observations demonstrate that dI1 commissural neurons develop axonal projection to spinal MNs via collateral branches arising later from the post-crossing segment of these axons. Our findings thus reveal a previously unrecognized projection of dI1 commissural axons that may contribute directly to generating proper motor output.
Collapse
Affiliation(s)
- Takeshi Kaneyama
- Cellular and Molecular Neurobiology Laboratory, Graduate School of Frontier Biosciences, Osaka University, Suita, Osaka, 565-0871, Japan
| | - Ryuichi Shirasaki
- Cellular and Molecular Neurobiology Laboratory, Graduate School of Frontier Biosciences, Osaka University, Suita, Osaka, 565-0871, Japan
| |
Collapse
|
19
|
Abstract
Neuronal connectivity in the cortex is determined by the laminar positioning of neurons. An important determinant of laminar positioning is likely to be the control of leading process behavior during migration, maintaining their tips directed toward the pia. In this study, we provide evidence that pial bone morphogenetic protein (Bmp) signaling regulates cortical neuronal migration during cortical layer formation. Specific disruption of pial Bmp ligands impaired the positioning of early-born neurons in the deep layer; further, cell-autonomous inhibition of Smad4, a core nuclear factor mediating Bmp signaling, in the cortical radial glial cells or postmitotic cortical neurons also produced neuronal migration defects that blurred the cortical layers. We found that leading processes were abnormal and that this was accompanied by excess dephosphorylated cofilin-1, an actin-severing protein, in Smad4 mutant neurons. This suggested that regulation of cofilin-1 might transduce Bmp signaling in the migrating neurons. Ectopic expression of a phosphorylation-defective form of cofilin-1 in the late-born wild-type neurons led them to stall in the deep layer, similar to the Smad4 mutant neurons. Expression of a phosphomimetic variant of cofilin-1 in the Smad4 mutant neurons rescued the migration defects. This suggests that cofilin-1 activity underlies Bmp-mediated cortical neuronal migration. This study shows that cofilin-1 mediates pial Bmp signaling during the positioning of cortical neurons and the formation of cortical layers.
Collapse
Affiliation(s)
- Youngshik Choe
- Department of Neurology, University of California, San Francisco, San Francisco, CA, USA.,Korea Brain Research Institute, Dong-gu, Daegu, Korea
| | - Samuel J Pleasure
- Department of Neurology, University of California, San Francisco, San Francisco, CA, USA.,Department of Neuroscience, University of California, San Francisco, San Francisco, CA, USA.,Department of Developmental Stem Cell Biology, University of California, San Francisco, San Francisco, CA, USA.,Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research and University of California, San Francisco, San Francisco, CA, USA
| |
Collapse
|
20
|
Guidi LG, Velayos‐Baeza A, Martinez‐Garay I, Monaco AP, Paracchini S, Bishop DVM, Molnár Z. The neuronal migration hypothesis of dyslexia: A critical evaluation 30 years on. Eur J Neurosci 2018; 48:3212-3233. [PMID: 30218584 PMCID: PMC6282621 DOI: 10.1111/ejn.14149] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2018] [Revised: 08/06/2018] [Accepted: 08/13/2018] [Indexed: 12/29/2022]
Abstract
The capacity for language is one of the key features underlying the complexity of human cognition and its evolution. However, little is known about the neurobiological mechanisms that mediate normal or impaired linguistic ability. For developmental dyslexia, early postmortem studies conducted in the 1980s linked the disorder to subtle defects in the migration of neurons in the developing neocortex. These early studies were reinforced by human genetic analyses that identified dyslexia susceptibility genes and subsequent evidence of their involvement in neuronal migration. In this review, we examine recent experimental evidence that does not support the link between dyslexia and neuronal migration. We critically evaluate gene function studies conducted in rodent models and draw attention to the lack of robust evidence from histopathological and imaging studies in humans. Our review suggests that the neuronal migration hypothesis of dyslexia should be reconsidered, and the neurobiological basis of dyslexia should be approached with a fresh start.
Collapse
Affiliation(s)
- Luiz G. Guidi
- Department of Physiology, Anatomy, and GeneticsUniversity of OxfordOxfordUK
- Wellcome Centre for Human GeneticsUniversity of OxfordOxfordUK
| | - Antonio Velayos‐Baeza
- Department of Physiology, Anatomy, and GeneticsUniversity of OxfordOxfordUK
- Wellcome Centre for Human GeneticsUniversity of OxfordOxfordUK
| | - Isabel Martinez‐Garay
- Department of Physiology, Anatomy, and GeneticsUniversity of OxfordOxfordUK
- Division of NeuroscienceSchool of BiosciencesCardiff UniversityCardiffUK
| | | | | | | | - Zoltán Molnár
- Department of Physiology, Anatomy, and GeneticsUniversity of OxfordOxfordUK
| |
Collapse
|
21
|
Guidi LG, Mattley J, Martinez-Garay I, Monaco AP, Linden JF, Velayos-Baeza A, Molnár Z. Knockout Mice for Dyslexia Susceptibility Gene Homologs KIAA0319 and KIAA0319L have Unaffected Neuronal Migration but Display Abnormal Auditory Processing. Cereb Cortex 2017; 27:5831-5845. [PMID: 29045729 PMCID: PMC5939205 DOI: 10.1093/cercor/bhx269] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Developmental dyslexia is a neurodevelopmental disorder that affects reading ability caused by genetic and non-genetic factors. Amongst the susceptibility genes identified to date, KIAA0319 is a prime candidate. RNA-interference experiments in rats suggested its involvement in cortical migration but we could not confirm these findings in Kiaa0319-mutant mice. Given its homologous gene Kiaa0319L (AU040320) has also been proposed to play a role in neuronal migration, we interrogated whether absence of AU040320 alone or together with KIAA0319 affects migration in the developing brain. Analyses of AU040320 and double Kiaa0319;AU040320 knockouts (dKO) revealed no evidence for impaired cortical lamination, neuronal migration, neurogenesis or other anatomical abnormalities. However, dKO mice displayed an auditory deficit in a behavioral gap-in-noise detection task. In addition, recordings of click-evoked auditory brainstem responses revealed suprathreshold deficits in wave III amplitude in AU040320-KO mice, and more general deficits in dKOs. These findings suggest that absence of AU040320 disrupts firing and/or synchrony of activity in the auditory brainstem, while loss of both proteins might affect both peripheral and central auditory function. Overall, these results stand against the proposed role of KIAA0319 and AU040320 in neuronal migration and outline their relationship with deficits in the auditory system.
Collapse
Affiliation(s)
- Luiz G Guidi
- Department of Physiology, Anatomy, and Genetics, University of Oxford, Oxford OX1 3QX, UK
- Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford OX3 7BN, UK
| | - Jane Mattley
- Ear Institute, University College London, London WC1X 8EE, UK
| | - Isabel Martinez-Garay
- Department of Physiology, Anatomy, and Genetics, University of Oxford, Oxford OX1 3QX, UK
| | - Anthony P Monaco
- Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford OX3 7BN, UK
- Current address: Office of the President, Ballou Hall, Tufts University, Medford, MA 02155, USA
| | - Jennifer F Linden
- Ear Institute, University College London, London WC1X 8EE, UK
- Department of Neuroscience, Physiology & Pharmacology, University College London, London WC1E 6BT, UK
| | | | - Zoltán Molnár
- Department of Physiology, Anatomy, and Genetics, University of Oxford, Oxford OX1 3QX, UK
| |
Collapse
|
22
|
Oishi K, Nakajima K. Subtype Specification of Cerebral Cortical Neurons in Their Immature Stages. Neurochem Res 2017; 43:238-244. [PMID: 29185180 DOI: 10.1007/s11064-017-2441-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2017] [Revised: 11/21/2017] [Accepted: 11/23/2017] [Indexed: 10/18/2022]
Abstract
The diversification of neuronal subtypes during corticogenesis is fundamental to the establishment of the complex cortical structure. Although subtype specification has been assumed to occur in neural progenitor cells, increasing evidence has begun to reveal the plasticity of subtype determination in immature neurons. Here, we summarize recent findings regarding the regulation of subtype specification during later periods of neuronal differentiation, such as the post-mitotic and post-migratory stages. We also discuss thalamocortical axons as an extra-cortical cue that provides information on the subtype determination of immature cortical neurons.
Collapse
Affiliation(s)
- Koji Oishi
- Department of Anatomy, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo, 160-8582, Japan.
| | - Kazunori Nakajima
- Department of Anatomy, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo, 160-8582, Japan.
| |
Collapse
|
23
|
Fukuda T, Yanagi S. Psychiatric behaviors associated with cytoskeletal defects in radial neuronal migration. Cell Mol Life Sci 2017; 74:3533-3552. [PMID: 28516224 PMCID: PMC11107632 DOI: 10.1007/s00018-017-2539-4] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2017] [Revised: 04/21/2017] [Accepted: 05/11/2017] [Indexed: 12/17/2022]
Abstract
Normal development of the cerebral cortex is an important process for higher brain functions, such as language, and cognitive and social functions. Psychiatric disorders, such as schizophrenia and autism, are thought to develop owing to various dysfunctions occurring during the development of the cerebral cortex. Radial neuronal migration in the embryonic cerebral cortex is a complex process, which is achieved by strict control of cytoskeletal dynamics, and impairments in this process are suggested to cause various psychiatric disorders. Our recent findings indicate that radial neuronal migration as well as psychiatric behaviors is rescued by controlling microtubule stability during the embryonic stage. In this review, we outline the relationship between psychiatric disorders, such as schizophrenia and autism, and radial neuronal migration in the cerebral cortex by focusing on the cytoskeleton and centrosomes. New treatment strategies for psychiatric disorders will be discussed.
Collapse
Affiliation(s)
- Toshifumi Fukuda
- Laboratory of Molecular Biochemistry, School of Life Sciences, Tokyo University of Pharmacy and Life Sciences, Hachioji, Tokyo, 192-0392, Japan.
| | - Shigeru Yanagi
- Laboratory of Molecular Biochemistry, School of Life Sciences, Tokyo University of Pharmacy and Life Sciences, Hachioji, Tokyo, 192-0392, Japan.
| |
Collapse
|
24
|
Ng HX, Lee EP, Cavanagh BL, Britto JM, Tan SS. A method for isolating cortical interneurons sharing the same birthdays for gene expression studies. Exp Neurol 2017; 295:36-45. [PMID: 28511841 DOI: 10.1016/j.expneurol.2017.05.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2016] [Revised: 04/13/2017] [Accepted: 05/13/2017] [Indexed: 11/19/2022]
Abstract
The two neuronal populations in the cortex, pyramidal neurons and interneurons, can be separated based on neurotransmitter identity, however, within this segregation a large degree of diversity exists. Investigations into the molecular diversity of neurons are impeded by the inability to isolate cell populations born at different times for gene expression analysis. Developing interneurons may be distinguished by the expression of Glutamic Acid Decarboxylase-67 (GAD67). Neuronal birthdating using nucleoside analogs is an effective means of identifying coetaneous interneurons. Using these two features, neurotransmitter identity and birthdating, we have developed a method to isolate migrating interneurons using fluorescent-activated cell sorting (FACS) for RNA extraction and gene expression analysis. We utilized 5-ethynyl-2'-deoxyuridine (EdU) to birthdate interneuron cohorts and the GAD67 knock-in GFP transgenic mice to identify interneurons. In combination, we achieved simultaneous detection of GFP and EdU signals during FACS sorting of coetaneous interneurons with minimum loss of RNA integrity. RNA quality was deemed to be satisfactory by quantitative polymerase chain reaction (qPCR) for the interneuron-specific transcript Gad67.
Collapse
Affiliation(s)
- Hui Xuan Ng
- Florey Institute of Neuroscience and Mental Health, Parkville, VIC, Australia; University of Melbourne, Parkville, VIC, Australia.
| | - Ean Phing Lee
- Florey Institute of Neuroscience and Mental Health, Parkville, VIC, Australia
| | | | - Joanne M Britto
- Florey Institute of Neuroscience and Mental Health, Parkville, VIC, Australia; University of Melbourne, Parkville, VIC, Australia
| | - Seong-Seng Tan
- Florey Institute of Neuroscience and Mental Health, Parkville, VIC, Australia; University of Melbourne, Parkville, VIC, Australia.
| |
Collapse
|
25
|
Darki F, Massinen S, Salmela E, Matsson H, Peyrard-Janvid M, Klingberg T, Kere J. Human ROBO1 regulates white matter structure in corpus callosum. Brain Struct Funct 2017; 222:707-716. [PMID: 27240594 PMCID: PMC5334444 DOI: 10.1007/s00429-016-1240-y] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2015] [Accepted: 05/20/2016] [Indexed: 12/17/2022]
Abstract
The axon guidance receptor, Robo1, controls the pathfinding of callosal axons in mice. To determine whether the orthologous ROBO1 gene is involved in callosal development also in humans, we studied polymorphisms in the ROBO1 gene and variation in the white matter structure in the corpus callosum using both structural magnetic resonance imaging and diffusion tensor magnetic resonance imaging. We found that five polymorphisms in the regulatory region of ROBO1 were associated with white matter density in the posterior part of the corpus callosum pathways. One of the polymorphisms, rs7631357, was also significantly associated with the probability of connections to the parietal cortical regions. Our results demonstrate that human ROBO1 may be involved in the regulation of the structure and connectivity of posterior part of corpus callosum.
Collapse
Affiliation(s)
- Fahimeh Darki
- Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Satu Massinen
- Research Programs Unit, Haartman Institute, University of Helsinki, Helsinki, Finland
- Folkhälsan Institute of Genetics, Helsinki, Finland
| | - Elina Salmela
- Research Programs Unit, Haartman Institute, University of Helsinki, Helsinki, Finland
- Folkhälsan Institute of Genetics, Helsinki, Finland
| | - Hans Matsson
- Department of Biosciences and Nutrition, Karolinska Institutet, Hälsovägen 7, 14183, Huddinge, Sweden
| | - Myriam Peyrard-Janvid
- Department of Biosciences and Nutrition, Karolinska Institutet, Hälsovägen 7, 14183, Huddinge, Sweden
| | - Torkel Klingberg
- Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Juha Kere
- Research Programs Unit, Haartman Institute, University of Helsinki, Helsinki, Finland.
- Folkhälsan Institute of Genetics, Helsinki, Finland.
- Department of Biosciences and Nutrition, Karolinska Institutet, Hälsovägen 7, 14183, Huddinge, Sweden.
- Science for Life Laboratory, Karolinska Institutet, Solna, Sweden.
| |
Collapse
|
26
|
Lin HY, Cheng CH, Chen DT, Chen YA, Park JY. Coexpression and expression quantitative trait loci analyses of the angiogenesis gene-gene interaction network in prostate cancer. Transl Cancer Res 2016; 5:S951-S963. [PMID: 28664150 DOI: 10.21037/tcr.2016.10.55] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
BACKGROUND Prostate cancer (PCa) shows a substantial clinical heterogeneity. The existing risk classification for PCa prognosis based on clinical factors is not sufficient. Although some biomarkers for PCa aggressiveness have been identified, their underlying functional mechanisms are still unclear. We previously reported a gene-gene interaction network associated with PCa aggressiveness based on single nucleotide polymorphism (SNP)-SNP interactions in the angiogenesis pathway. The goal of this study is to investigate potential functional evidence of the involvement of the genes in this gene-gene interaction network. METHODS A total of 11 angiogenesis genes were evaluated. The crosstalks among genes were examined through coexpression and expression quantitative trait loci (eQTL) analyses. The study population is 352 Caucasian PCa patients in the Cancer Genome Atlas (TCGA) study. The pairwise coexpressions among the genes of interest were evaluated using the Spearman coefficient. The eQTL analyses were tested using the Kruskal-Wallis test. RESULTS Among all within gene and 55 possible pairwise gene evaluations, 12 gene pairs and one gene (MMP16) showed strong coexpression or significant eQTL evidence. There are nine gene pairs with a strong correlation (Spearman correlation ≥0.6, P<1×10-13). The top coexpressed gene pairs are EGFR-SP1 (r=0.73), ITGB3-HSPG2 (r=0.71), ITGB3-CSF1 (r=0.70), MMP16-FBLN5 (r=0.68), ITGB3-MMP16 (r=0.65), ITGB3-ROBO1 (r=0.62), CSF1-HSPG2 (r=0.61), CSF1-FBLN5 (r=0.6), and CSF1-ROBO1 (r=0.60). One cis-eQTL in MMP16 and five trans-eQTLs (MMP16-ESR1, ESR1-ROBO1, CSF1-ROBO1, HSPG2-ROBO1, and FBLN5-CSF1) are significant with a false discovery rate q value less than 0.2. CONCLUSIONS These findings provide potential biological evidence for the gene-gene interactions in this angiogenesis network. These identified interactions between the angiogenesis genes not only provide information for PCa etiology mechanism but also may serve as integrated biomarkers for building a risk prediction model for PCa aggressiveness.
Collapse
Affiliation(s)
- Hui-Yi Lin
- Biostatistics Program, School of Public Health, Louisiana State University Health Sciences Center, New Orleans, LA 70112, USA
| | - Chia-Ho Cheng
- Department of Biostatistics and Bioinformatics, Moffitt Cancer Center & Research Institute, Tampa, FL 33612, USA
| | - Dung-Tsa Chen
- Department of Biostatistics and Bioinformatics, Moffitt Cancer Center & Research Institute, Tampa, FL 33612, USA
| | - Y Ann Chen
- Department of Biostatistics and Bioinformatics, Moffitt Cancer Center & Research Institute, Tampa, FL 33612, USA
| | - Jong Y Park
- Department of Cancer Epidemiology, Moffitt Cancer Center & Research Institute, Tampa, FL 33612, USA
| |
Collapse
|
27
|
Toma K, Hanashima C. Switching modes in corticogenesis: mechanisms of neuronal subtype transitions and integration in the cerebral cortex. Front Neurosci 2015; 9:274. [PMID: 26321900 PMCID: PMC4531338 DOI: 10.3389/fnins.2015.00274] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2015] [Accepted: 07/21/2015] [Indexed: 12/16/2022] Open
Abstract
Information processing in the cerebral cortex requires the activation of diverse neurons across layers and columns, which are established through the coordinated production of distinct neuronal subtypes and their placement along the three-dimensional axis. Over recent years, our knowledge of the regulatory mechanisms of the specification and integration of neuronal subtypes in the cerebral cortex has progressed rapidly. In this review, we address how the unique cytoarchitecture of the neocortex is established from a limited number of progenitors featuring neuronal identity transitions during development. We further illuminate the molecular mechanisms of the subtype-specific integration of these neurons into the cerebral cortex along the radial and tangential axis, and we discuss these key features to exemplify how neocortical circuit formation accomplishes economical connectivity while maintaining plasticity and evolvability to adapt to environmental changes.
Collapse
Affiliation(s)
- Kenichi Toma
- Laboratory for Neocortical Development, RIKEN Center for Developmental Biology Kobe, Japan
| | - Carina Hanashima
- Laboratory for Neocortical Development, RIKEN Center for Developmental Biology Kobe, Japan ; Department of Biology, Graduate School of Science, Kobe University Kobe, Japan
| |
Collapse
|
28
|
Bullmann T, Arendt T, Frey U, Hanashima C. A transportable, inexpensive electroporator for in utero electroporation. Dev Growth Differ 2015; 57:369-377. [PMID: 25988525 DOI: 10.1111/dgd.12216] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2015] [Revised: 03/31/2015] [Accepted: 04/08/2015] [Indexed: 01/14/2023]
Abstract
Electroporation is a useful technique to study gene function during development but its broad application is hampered due to the expensive equipment needed. We describe the construction of a transportable, simple and inexpensive electroporator delivering square pulses with varying length and amplitude. The device was successfully used for in utero electroporation in mouse with a performance comparable to that of commercial products.
Collapse
Affiliation(s)
- Torsten Bullmann
- Frey Initiative Research Unit, RIKEN Quantitative Biology Center, 2-2-3 Minatojima-minamimachi, Chuo-ku, Kobe, 650-0047, Japan.,Laboratory for Neocortical Development, RIKEN Center for Developmental Biology, 2-2-3 Minatojima-minamimachi, Chuo-ku, Kobe, 650-0047, Japan.,Department of Molecular and Cellular Mechanisms of Neurodegeneration, Paul Flechsig Institute of Brain Research, University of Leipzig, Liebigstraβe 19, 04103, Leipzig, Germany
| | - Thomas Arendt
- Department of Molecular and Cellular Mechanisms of Neurodegeneration, Paul Flechsig Institute of Brain Research, University of Leipzig, Liebigstraβe 19, 04103, Leipzig, Germany
| | - Urs Frey
- Frey Initiative Research Unit, RIKEN Quantitative Biology Center, 2-2-3 Minatojima-minamimachi, Chuo-ku, Kobe, 650-0047, Japan
| | - Carina Hanashima
- Laboratory for Neocortical Development, RIKEN Center for Developmental Biology, 2-2-3 Minatojima-minamimachi, Chuo-ku, Kobe, 650-0047, Japan
| |
Collapse
|
29
|
Parray A, Siddique HR, Kuriger JK, Mishra SK, Rhim JS, Nelson HH, Aburatani H, Konety BR, Koochekpour S, Saleem M. ROBO1, a tumor suppressor and critical molecular barrier for localized tumor cells to acquire invasive phenotype: study in African-American and Caucasian prostate cancer models. Int J Cancer 2014; 135:2493-506. [PMID: 24752651 PMCID: PMC4610361 DOI: 10.1002/ijc.28919] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2014] [Accepted: 04/07/2014] [Indexed: 12/20/2022]
Abstract
High-risk populations exhibit early transformation of localized prostate cancer (CaP) disease to metastasis which results in the mortality of such patients. The paucity of knowledge about the molecular mechanism involved in acquiring of metastatic behavior by primary tumor cells and non-availability of reliable phenotype-discriminating biomarkers are stumbling blocks in the management of CaP disease. Here, we determine the role and translational relevance of ROBO1 (an organogenesis-associated gene) in human CaP. Employing CaP-progression models and prostatic tissues of Caucasian and African-American patients, we show that ROBO1 expression is localized to cell-membrane and significantly lost in primary and metastatic tumors. While Caucasians exhibited similar ROBO1 levels in primary and metastatic phenotype, a significant difference was observed between tumor phenotypes in African-Americans. Epigenetic assays identified promoter methylation of ROBO1 specific to African-American metastatic CaP cells. Using African-American CaP models for further studies, we show that ROBO1 negatively regulates motility and invasiveness of primary CaP cells, and its loss causes these cells to acquire invasive trait. To understand the underlying mechanism, we employed ROBO1-expressing/ROBO1-C2C3-mutant constructs, immunoprecipitation, confocal-microscopy and luciferase-reporter techniques. We show that ROBO1 through its interaction with DOCK1 (at SH3-SH2-domain) controls the Rac-activation. However, loss of ROBO1 results in Rac1-activation which in turn causes E-Cadherin/β-catenin cytoskeleton destabilization and induction of cell migration. We suggest that ROBO1 is a predictive biomarker that has potential to discriminate among CaP types, and could be exploited as a molecular target to inhibit the progression of disease as well as treat metastasis in high-risk populations such as African-Americans.
Collapse
MESH Headings
- Black or African American/statistics & numerical data
- Blotting, Western
- Cadherins/genetics
- Cadherins/metabolism
- Cell Movement
- Cell Proliferation
- Cohort Studies
- Disease Progression
- Fluorescent Antibody Technique
- Gene Expression Regulation, Neoplastic
- Genes, Tumor Suppressor
- Humans
- Immunoenzyme Techniques
- Male
- Neoplasm Metastasis
- Neoplasm Staging
- Nerve Tissue Proteins/antagonists & inhibitors
- Nerve Tissue Proteins/genetics
- Nerve Tissue Proteins/metabolism
- Phenotype
- Promoter Regions, Genetic/genetics
- Prostatic Neoplasms/ethnology
- Prostatic Neoplasms/metabolism
- Prostatic Neoplasms/pathology
- RNA, Messenger/genetics
- RNA, Small Interfering/genetics
- Real-Time Polymerase Chain Reaction
- Receptors, Immunologic/antagonists & inhibitors
- Receptors, Immunologic/genetics
- Receptors, Immunologic/metabolism
- Reverse Transcriptase Polymerase Chain Reaction
- Tumor Cells, Cultured
- White People/statistics & numerical data
- Wound Healing
- beta Catenin/genetics
- beta Catenin/metabolism
- rac GTP-Binding Proteins/genetics
- rac GTP-Binding Proteins/metabolism
- rac1 GTP-Binding Protein/genetics
- rac1 GTP-Binding Protein/metabolism
- Roundabout Proteins
Collapse
Affiliation(s)
- Aijaz Parray
- Section of Molecular Chemoprevention and Therapeutics, The Hormel Institute, University of Minnesota, Austin, MN
| | - Hifzur R. Siddique
- Section of Molecular Chemoprevention and Therapeutics, The Hormel Institute, University of Minnesota, Austin, MN
| | - Jacquelyn K. Kuriger
- Division of Epidemiology and Community Health, University of Minnesota, Minneapolis, MN
| | - Shrawan K. Mishra
- Section of Molecular Chemoprevention and Therapeutics, The Hormel Institute, University of Minnesota, Austin, MN
| | - Johng S. Rhim
- Uniformed Services University of the Health Sciences, Bethesda, MD
| | - Heather H. Nelson
- Division of Epidemiology and Community Health, University of Minnesota, Minneapolis, MN
| | - Hiroyuki Aburatani
- Genome Science Division, Research Center for Advanced Science and Technology, University of Tokyo, Japan
| | - Badrinath R. Konety
- Department of Urology, Masonic Cancer Center, University of Minnesota, Minneapolis, MN
| | - Shahriar Koochekpour
- Center for Genetics and Pharmacology, Roswell Park Cancer Institute, Buffalo, NY
| | - Mohammad Saleem
- Section of Molecular Chemoprevention and Therapeutics, The Hormel Institute, University of Minnesota, Austin, MN
- Department of Urology, Masonic Cancer Center, University of Minnesota, Minneapolis, MN
- Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, MN
| |
Collapse
|
30
|
Blockus H, Chédotal A. The multifaceted roles of Slits and Robos in cortical circuits: from proliferation to axon guidance and neurological diseases. Curr Opin Neurobiol 2014; 27:82-8. [PMID: 24698714 DOI: 10.1016/j.conb.2014.03.003] [Citation(s) in RCA: 58] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2014] [Revised: 02/17/2014] [Accepted: 03/09/2014] [Indexed: 11/20/2022]
Abstract
Slit repulsion, mediated by Robo receptors, is known to play a major role in axon guidance in the nervous system. However, recent studies have revealed that in the mammalian cortex these molecules are highly versatile and that their function extends far beyond axon guidance. They act at all phases of development to control neurogenesis, neuronal migration, axon patterning, dendritic outgrowth and spinogenesis. The expression of Robo receptors in cortical and thalamocortical axons (TCAs) is tightly regulated by a combination of transcription factors (TFs), proteases and activity. These findings also suggest that Slit and Robos have influenced the evolution of cortical circuits. Last, novel genetic evidence associates various neurological disorders, such as autism, to abnormal Slit/Robo signaling.
Collapse
Affiliation(s)
- Heike Blockus
- INSERM UMR_S968, Institut de la Vision, F-75012 Paris, France; Sorbonne Universités, UPMC Univ Paris 06, UMR_S968, Institut de la vision, F-75012, France; CNRS, UMR7210, F-75012 Paris, France
| | - Alain Chédotal
- INSERM UMR_S968, Institut de la Vision, F-75012 Paris, France; Sorbonne Universités, UPMC Univ Paris 06, UMR_S968, Institut de la vision, F-75012, France; CNRS, UMR7210, F-75012 Paris, France.
| |
Collapse
|
31
|
Yeh ML, Gonda Y, Mommersteeg MTM, Barber M, Ypsilanti AR, Hanashima C, Parnavelas JG, Andrews WD. Robo1 modulates proliferation and neurogenesis in the developing neocortex. J Neurosci 2014; 34:5717-31. [PMID: 24741061 PMCID: PMC3988420 DOI: 10.1523/jneurosci.4256-13.2014] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2013] [Revised: 02/19/2014] [Accepted: 03/19/2014] [Indexed: 12/11/2022] Open
Abstract
The elaborate cytoarchitecture of the mammalian neocortex requires the timely production of its constituent pyramidal neurons and interneurons and their disposition in appropriate layers. Numerous chemotropic factors present in the forebrain throughout cortical development play important roles in the orchestration of these events. The Roundabout (Robo) family of receptors and their ligands, the Slit proteins, are expressed in the developing forebrain, and are known to play important roles in the generation and migration of cortical interneurons. However, few studies have investigated their function(s) in the development of pyramidal cells. Here, we observed expression of Robo1 and Slit genes (Slit1, Slit2) in cells lining the telencephalic ventricles, and found significant increases in progenitor cells (basal and apical) at embryonic day (E)12.5 and E14.5 in the developing cortex of Robo1(-/-), Slit1(-/-), and Slit1(-/-)/Slit2(-/-), but not in mice lacking the other Robo or Slit genes. Using layer-specific markers, we found that both early- and late-born pyramidal neuron populations were significantly increased in the cortices of Robo1(-/-) mice at the end of corticogenesis (E18.5). The excess number of cortical pyramidal neurons generated prenatally appears to die in early postnatal life. The observed increase in pyramidal neurons was due to prolonged proliferative activity of their progenitors and not due to changes in cell cycle events. This finding, confirmed by in utero electroporation with Robo1 short hairpin RNA (shRNA) or control constructs into progenitors along the ventricular zone as well as in dissociated cortical cell cultures, points to a novel role for Robo1 in regulating the proliferation and generation of pyramidal neurons.
Collapse
Affiliation(s)
- Mason L. Yeh
- Department of Cell and Developmental Biology, University College London, London, United Kingdom WC1E 6DE
| | - Yuko Gonda
- Laboratory for Neocortical Development, RIKEN Center for Developmental Biology, Kobe 650-0047, Japan
| | - Mathilda T. M. Mommersteeg
- Department of Cell and Developmental Biology, University College London, London, United Kingdom WC1E 6DE
| | - Melissa Barber
- Institut Jacques-Monod, Université Paris Diderot/CNRS, 75201 Paris, France, and
| | | | - Carina Hanashima
- Laboratory for Neocortical Development, RIKEN Center for Developmental Biology, Kobe 650-0047, Japan
| | - John G. Parnavelas
- Department of Cell and Developmental Biology, University College London, London, United Kingdom WC1E 6DE
| | - William D. Andrews
- Department of Cell and Developmental Biology, University College London, London, United Kingdom WC1E 6DE
| |
Collapse
|
32
|
Abstract
Roundabout receptors (Robo) and their Slit ligands were discovered in the 1990s and found to be key players in axon guidance. Slit was initially described s an extracellular matrix protein that was expressed by midline glia in Drosophila. A few years later, it was shown that, in vertebrates and invertebrates, Slits acted as chemorepellents for axons crossing the midline. Robo proteins were originally discovered in Drosophila in a mutant screen for genes involved in the regulation of midline crossing. This ligand-receptor pair has since been implicated in a variety of other neuronal and non-neuronal processes ranging from cell migration to angiogenesis, tumourigenesis and even organogenesis of tissues such as kidneys, lungs and breasts.
Collapse
|
33
|
Platt MP, Adler WT, Mehlhorn AJ, Johnson GC, Wright KA, Choi RT, Tsang WH, Poon MW, Yeung SY, Waye MMY, Galaburda AM, Rosen GD. Embryonic disruption of the candidate dyslexia susceptibility gene homolog Kiaa0319-like results in neuronal migration disorders. Neuroscience 2013; 248:585-93. [PMID: 23831424 DOI: 10.1016/j.neuroscience.2013.06.056] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2013] [Revised: 06/19/2013] [Accepted: 06/20/2013] [Indexed: 02/04/2023]
Abstract
Developmental dyslexia, the most common childhood learning disorder, is highly heritable, and recent studies have identified KIAA0319-Like (KIAA0319L) as a candidate dyslexia susceptibility gene at the 1p36-34 (DYX8) locus. In this experiment, we investigated the anatomical effects of knocking down this gene during rat corticogenesis. Cortical progenitor cells were transfected using in utero electroporation on embryonic day (E) 15.5 with plasmids encoding either: (1) Kiaa0319l small hairpin RNA (shRNA), (2) an expression construct for human KIAA0319L, (3) Kiaa0319l shRNA+KIAA0319L expression construct (rescue), or (4) controls (scrambled Kiaa0319l shRNA or empty expression vector). Mothers were injected with 5-bromo-2-deoxyuridine (BrdU) at either E13.5, E15.5, or E17.5. Disruption of Kiaa0319l function (by knockdown, overexpression, or rescue) resulted in the formation of large nodular periventricular heterotopia in approximately 25% of the rats, which can be seen as early as postnatal day 1. Only a small subset of heterotopic neurons had been transfected, indicating non-cell autonomous effects of the transfection. Most heterotopic neurons were generated in mid- to late-gestation, and laminar markers suggest that they were destined for upper cortical laminae. Finally, we found that transfected neurons in the cerebral cortex were located in their expected laminae. These results indicate that KIAA0319L is the fourth of four candidate dyslexia susceptibility genes that is involved in neuronal migration, which supports the association of abnormal neuronal migration with developmental dyslexia.
Collapse
Affiliation(s)
- M P Platt
- Department of Neurology, Beth Israel Deaconess Medical Center, Boston, MA, USA
| | - W T Adler
- Department of Neurology, Beth Israel Deaconess Medical Center, Boston, MA, USA
| | - A J Mehlhorn
- Department of Neurology, Beth Israel Deaconess Medical Center, Boston, MA, USA
| | - G C Johnson
- Department of Neurology, Beth Israel Deaconess Medical Center, Boston, MA, USA
| | - K A Wright
- Department of Neurology, Beth Israel Deaconess Medical Center, Boston, MA, USA
| | - R T Choi
- School of Biomedical Sciences & School of Life Sciences, The Chinese University of Hong Kong, Shatin, Hong Kong, China
| | - W H Tsang
- School of Biomedical Sciences & School of Life Sciences, The Chinese University of Hong Kong, Shatin, Hong Kong, China
| | - M W Poon
- School of Biomedical Sciences & School of Life Sciences, The Chinese University of Hong Kong, Shatin, Hong Kong, China
| | - S Y Yeung
- School of Biomedical Sciences & School of Life Sciences, The Chinese University of Hong Kong, Shatin, Hong Kong, China
| | - M M Y Waye
- School of Biomedical Sciences & School of Life Sciences, The Chinese University of Hong Kong, Shatin, Hong Kong, China
| | - A M Galaburda
- Department of Neurology, Beth Israel Deaconess Medical Center, Boston, MA, USA
| | - G D Rosen
- Department of Neurology, Beth Israel Deaconess Medical Center, Boston, MA, USA.
| |
Collapse
|