1
|
Currey L, Harris L, Piper M. Single cell transcriptomics of the cerebral cortex of mice lacking the PRC2 gene eed. BMC Res Notes 2024; 17:382. [PMID: 39719612 DOI: 10.1186/s13104-024-07008-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2024] [Accepted: 11/18/2024] [Indexed: 12/26/2024] Open
Abstract
OBJECTIVE The Polycomb Repressive Complex 2 (PRC2) regulates neural stem cell behaviour during development of the cerebral cortex, yet how the loss of PRC2 developmentally influences cell identity in the mature brain is poorly defined. Using a mouse model in which the PRC2 gene Embryonic ectoderm development (Eed) was conditionally deleted from the developing mouse dorsal telencephalon, we performed single nuclei RNA sequencing (snRNA-seq) on the cortical plate of an adult heterozygote Eed knockout mouse and an adult homozygote Eed knockout mouse compared to a littermate control. This work was part of a larger effort to understand consequences of mutations to PRC2 within the mature brain. RESULTS Here we provide snRNA-seq data from the cortical plate of an adult heterozygous conditional Eed knockout, an adult homozygous conditional Eed knockout and an adult control mouse. This data provides insight on how loss of PRC2 function during development affects cell identity in the mature cortex.
Collapse
Affiliation(s)
- Laura Currey
- School of Biomedical Sciences, The University of Queensland, Brisbane, QLD, 4072, Australia.
| | - Lachlan Harris
- School of Biomedical Sciences, The University of Queensland, Brisbane, QLD, 4072, Australia
- QIMR Berghofer Medical Research Institute, Brisbane, QLD, 4006, Australia
| | - Michael Piper
- School of Biomedical Sciences, The University of Queensland, Brisbane, QLD, 4072, Australia.
- Queensland Brain Institute, The University of Queensland, Brisbane, QLD, 4072, Australia.
| |
Collapse
|
2
|
Zanin JP, Pandya MA, Espinoza D, Friedman WJ, Shiflett MW. Excess cerebellar granule neurons induced by the absence of p75NTR during development elicit social behavior deficits in mice. Front Mol Neurosci 2023; 16:1147597. [PMID: 37305555 PMCID: PMC10249730 DOI: 10.3389/fnmol.2023.1147597] [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: 01/18/2023] [Accepted: 04/24/2023] [Indexed: 06/13/2023] Open
Abstract
Introduction Recently, the cerebellum has been implicated with non-motor functions, including cognitive and emotional behavior. Anatomical and functional studies demonstrate bidirectional cerebellar connections with brain regions involved in social cognition. Cerebellar developmental abnormalities and injury are often associated with several psychiatric and mental disorders including autism spectrum disorders and anxiety. The cerebellar granule neurons (CGN) are essential for cerebellar function since they provide sensorimotor, proprioceptive, and contextual information to Purkinje cells to modify behavior in different contexts. Therefore, alterations to the CGN population are likely to compromise cerebellar processing and function. Previously we demonstrated that the p75 neurotrophin receptor (p75NTR) was fundamental for the development of the CGN. In the absence of p75NTR, we observed increased proliferation of the granule cell precursors (GCPs), followed by increased GCP migration toward the internal granule layer. The excess granule cells were incorporated into the cerebellar network, inducing alterations in cerebellar circuit processing. Methods In the present study, we used two conditional mouse lines to specifically delete the expression of p75NTR in CGN. In both mouse lines, deletion of the target gene was under the control of the transcription factor Atoh-1 promotor, however, one of the lines was also tamoxifen-inducible. Results We observed a loss of p75NTR expression from the GCPs in all cerebellar lobes. Compared to control animals, both mouse lines exhibited a reduced preference for social interactions when presented with a choice to interact with a mouse or an object. Open-field locomotor behavior and operant reward learning were unaffected in both lines. Lack of preference for social novelty and increased anxiety-related behavior was present in mice with constitutive p75NTR deletion; however, these effects were not present in the tamoxifen-inducible mice with p75NTR deletion that more specifically targeted the GCPs. Discussion Our findings demonstrate that alterations to CGN development by loss of p75NTR alter social behavior, and contribute to the increasing evidence that the cerebellum plays a role in non-motor-related behaviors, including social behavior.
Collapse
Affiliation(s)
- Juan Pablo Zanin
- Department of Biological Sciences, Rutgers, The State University of New Jersey, Newark, NJ, United States
| | - Mansi A. Pandya
- Department of Biological Sciences, Rutgers, The State University of New Jersey, Newark, NJ, United States
| | - Diego Espinoza
- Department of Psychology, Rutgers, The State University of New Jersey, Newark, NJ, United States
| | - Wilma J. Friedman
- Department of Biological Sciences, Rutgers, The State University of New Jersey, Newark, NJ, United States
| | - Michael W. Shiflett
- Department of Psychology, Rutgers, The State University of New Jersey, Newark, NJ, United States
| |
Collapse
|
3
|
Kittock CM, Pilaz LJ. Advances in in utero electroporation. Dev Neurobiol 2023; 83:73-90. [PMID: 36861639 DOI: 10.1002/dneu.22910] [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: 12/14/2022] [Revised: 02/02/2023] [Accepted: 02/21/2023] [Indexed: 03/03/2023]
Abstract
In utero electroporation (IUE) is a technique developed in the early 2000s to transfect the neurons and neural progenitors of embryonic brains, thus enabling continued development in utero and subsequent analyses of neural development. Early IUE experiments focused on ectopic expression of plasmid DNA to analyze parameters such as neuron morphology and migration. Recent advances made in other fields, such as CRISPR/CAS9 genome editing, have been incorporated into IUE techniques as they were developed. Here, we provide a general review of the mechanics and techniques involved in IUE and explore the breadth of approaches that can be used in conjunction with IUE to study cortical development in a rodent model, with a focus on the novel advances in IUE techniques. We also highlight a few cases that exemplify the potential of IUE to study a broad range of questions in neural development.
Collapse
Affiliation(s)
- Claire M Kittock
- Pediatrics and Rare Diseases Group, Sanford Research, Sioux Falls, South Dakota, USA
- Sanford School of Medicine, University of South Dakota, Sioux Falls, South Dakota, USA
| | - Louis-Jan Pilaz
- Pediatrics and Rare Diseases Group, Sanford Research, Sioux Falls, South Dakota, USA
- Sanford School of Medicine, University of South Dakota, Sioux Falls, South Dakota, USA
| |
Collapse
|
4
|
Leung RF, George AM, Roussel EM, Faux MC, Wigle JT, Eisenstat DD. Genetic Regulation of Vertebrate Forebrain Development by Homeobox Genes. Front Neurosci 2022; 16:843794. [PMID: 35546872 PMCID: PMC9081933 DOI: 10.3389/fnins.2022.843794] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2021] [Accepted: 03/14/2022] [Indexed: 01/19/2023] Open
Abstract
Forebrain development in vertebrates is regulated by transcription factors encoded by homeobox, bHLH and forkhead gene families throughout the progressive and overlapping stages of neural induction and patterning, regional specification and generation of neurons and glia from central nervous system (CNS) progenitor cells. Moreover, cell fate decisions, differentiation and migration of these committed CNS progenitors are controlled by the gene regulatory networks that are regulated by various homeodomain-containing transcription factors, including but not limited to those of the Pax (paired), Nkx, Otx (orthodenticle), Gsx/Gsh (genetic screened), and Dlx (distal-less) homeobox gene families. This comprehensive review outlines the integral role of key homeobox transcription factors and their target genes on forebrain development, focused primarily on the telencephalon. Furthermore, links of these transcription factors to human diseases, such as neurodevelopmental disorders and brain tumors are provided.
Collapse
Affiliation(s)
- Ryan F. Leung
- Murdoch Children’s Research Institute, The Royal Children’s Hospital Melbourne, Parkville, VIC, Australia
- Department of Paediatrics, University of Melbourne, Parkville, VIC, Australia
| | - Ankita M. George
- Murdoch Children’s Research Institute, The Royal Children’s Hospital Melbourne, Parkville, VIC, Australia
| | - Enola M. Roussel
- Murdoch Children’s Research Institute, The Royal Children’s Hospital Melbourne, Parkville, VIC, Australia
| | - Maree C. Faux
- Murdoch Children’s Research Institute, The Royal Children’s Hospital Melbourne, Parkville, VIC, Australia
- Department of Paediatrics, University of Melbourne, Parkville, VIC, Australia
- Department of Surgery, Royal Melbourne Hospital, The University of Melbourne, Parkville, VIC, Australia
| | - Jeffrey T. Wigle
- Department of Biochemistry and Medical Genetics, Max Rady College of Medicine, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, MB, Canada
- Institute of Cardiovascular Sciences, St. Boniface Hospital Albrechtsen Research Centre, Winnipeg, MB, Canada
| | - David D. Eisenstat
- Murdoch Children’s Research Institute, The Royal Children’s Hospital Melbourne, Parkville, VIC, Australia
- Department of Paediatrics, University of Melbourne, Parkville, VIC, Australia
- Department of Medical Genetics, University of Alberta, Edmonton, AB, Canada
- Department of Pediatrics, University of Alberta, Edmonton, AB, Canada
| |
Collapse
|
5
|
Martín-Fernández F, Bermejo-Santos A, Bragg-Gonzalo L, Briz CG, Serrano-Saiz E, Nieto M. Role of Nrp1 in controlling cortical inter-hemispheric circuits. eLife 2022; 11:69776. [PMID: 35230240 PMCID: PMC8887897 DOI: 10.7554/elife.69776] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2021] [Accepted: 02/03/2022] [Indexed: 01/09/2023] Open
Abstract
Axons of the corpus callosum (CC) mediate the interhemispheric communication required for complex perception in mammals. In the somatosensory (SS) cortex, the CC exchanges inputs processed by the primary (S1) and secondary (S2) areas, which receive tactile and pain stimuli. During early postnatal life, a multistep process involving axonal navigation, growth, and refinement, leads to precise CC connectivity. This process is often affected in neurodevelopmental disorders such as autism and epilepsy. We herein show that in mice, expression of the axonal signaling receptor Neuropilin 1 (Nrp1) in SS layer (L) 2/3 is temporary and follows patterns that determine CC connectivity. At postnatal day 4, Nrp1 expression is absent in the SS cortex while abundant in the motor area, creating a sharp border. During the following 3 weeks, Nrp1 is transiently upregulated in subpopulations of SS L2/3 neurons, earlier and more abundantly in S2 than in S1. In vivo knock-down and overexpression experiments demonstrate that transient expression of Nrp1 does not affect the initial development of callosal projections in S1 but is required for subsequent S2 innervation. Moreover, knocking-down Nrp1 reduces the number of S2L2/3 callosal neurons due to excessive postnatal refinement. Thus, an exquisite temporal and spatial regulation of Nrp1 expression determines SS interhemispheric maps.
Collapse
Affiliation(s)
- Fernando Martín-Fernández
- Department of Cellular and Molecular Biology, Centro Nacional de Biotecnología, Consejo Superior de Investigaciones Científicas (CNB-CSIC), Campus de Cantoblanco, Darwin, Madrid, Spain
| | - Ana Bermejo-Santos
- Centro de Biología Molecular Severo Ochoa (CSIC/UAM), Campus de Cantoblanco, Nicolás Cabrera, Madrid, Spain
| | - Lorena Bragg-Gonzalo
- Department of Cellular and Molecular Biology, Centro Nacional de Biotecnología, Consejo Superior de Investigaciones Científicas (CNB-CSIC), Campus de Cantoblanco, Darwin, Madrid, Spain
| | - Carlos G Briz
- Department of Cellular and Molecular Biology, Centro Nacional de Biotecnología, Consejo Superior de Investigaciones Científicas (CNB-CSIC), Campus de Cantoblanco, Darwin, Madrid, Spain
| | - Esther Serrano-Saiz
- Centro de Biología Molecular Severo Ochoa (CSIC/UAM), Campus de Cantoblanco, Nicolás Cabrera, Madrid, Spain
| | - Marta Nieto
- Department of Cellular and Molecular Biology, Centro Nacional de Biotecnología, Consejo Superior de Investigaciones Científicas (CNB-CSIC), Campus de Cantoblanco, Darwin, Madrid, Spain
| |
Collapse
|
6
|
Ran S, Zuo Z, Li C, Yin X, Qu W, Tang Q, Wang Y, Shi Y, Li H. Atrophic Corpus Callosum Associated with Altered Functional Asymmetry in Major Depressive Disorder. Neuropsychiatr Dis Treat 2020; 16:1473-1482. [PMID: 32606700 PMCID: PMC7293967 DOI: 10.2147/ndt.s245078] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/22/2020] [Accepted: 05/12/2020] [Indexed: 11/23/2022] Open
Abstract
PURPOSE Impairments in intra- and inter-hemispheric information transfer circuits have been reported in patients with major depressive disorder (MDD). However, the specific anomalous connection (intra- and/or inter-hemispheric) and hemisphere (left and/or right) in which this connection plays a more dominant role in the pathogenic mechanism underlying MDD are still poorly understood. PATIENTS AND METHODS Structural magnetic resonance imaging and resting-state functional magnetic resonance imaging were performed in 33 patients with MDD and 33 healthy controls. The intra- and inter-hemispheric functional connectivity (FC) strength in the default mode network areas and volume of the callosal subregions were computed using independent samples t-tests. The partial correlations between the volumes and FCs were also computed. RESULTS The patients with MDD had smaller volumes in the genu of the corpus callosum than the controls. The intrahemispheric FCs of the bilateral posterior cingulate gyrus, left precuneus, left medial superior frontal gyrus, left medial orbitofrontal gyrus, left angular gyrus and left middle temporal gyrus, and interhemispheric FCs of the bilateral posterior cingulate gyrus in the patients with MDD were lower than those in the controls. Moreover, the intrahemispheric FCs of the precuneus and interhemispheric FCs of middle frontal gyrus, orbital middle frontal gyrus, and anterior cingulate gyrus in the patients with MDD showed right-lateralized asymmetry, which were opposite from the asymmetry patterns observed in the controls. The functional asymmetry of the anterior cingulate gyrus was correlated with the volume of the genu of the corpus callosum and disease duration. CONCLUSION These findings provide robust evidence that intra- and inter-hemispheric disconnections are involved in MDD, and that functional disruptions in the left hemisphere may be more relevant to the pathophysiology of MDD. Furthermore, imbalanced interhemispheric exchanges may contribute to the anatomical deficits in the corpus callosum in patients with MDD.
Collapse
Affiliation(s)
- Shuhua Ran
- Department of Radiology, Southwest Hospital, Army Medical University, Chongqing, People’s Republic of China
| | - Zhiwei Zuo
- Department of Radiology, General Hospital of Western Theater Command, Chengdu, People’s Republic of China
| | - Chang Li
- Department of Radiology, Daping Hospital, Army Medical University, Chongqing, People’s Republic of China
| | - Xuntao Yin
- Department of Medical Imaging, Guizhou Provincial People’s Hospital, Guizhou, People’s Republic of China
| | - Wei Qu
- Department of Psychology, Southwest Hospital, Army Medical University, Chongqing, People’s Republic of China
| | - Qianying Tang
- Department of Psychology, Southwest Hospital, Army Medical University, Chongqing, People’s Republic of China
| | - Yao Wang
- Department of Radiology, Southwest Hospital, Army Medical University, Chongqing, People’s Republic of China
| | - Yanshu Shi
- Department of Radiology, Southwest Hospital, Army Medical University, Chongqing, People’s Republic of China
| | - Haitao Li
- Department of Radiology, Southwest Hospital, Army Medical University, Chongqing, People’s Republic of China
| |
Collapse
|
7
|
Bottom RT, Krubitzer LA, Huffman KJ. Early postnatal gene expression in the developing neocortex of prairie voles (Microtus ochrogaster) is related to parental rearing style. J Comp Neurol 2020; 528:3008-3022. [PMID: 31930725 DOI: 10.1002/cne.24856] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2019] [Revised: 01/08/2020] [Accepted: 01/09/2020] [Indexed: 11/10/2022]
Abstract
The earliest and most prevalent sensory experience includes tactile, thermal, and olfactory stimulation delivered to the young via contact with the mother, and in some mammals, the father. Prairie voles (Microtus ochrogaster), like humans, are biparental and serve as a model for understanding the impact of parent/offspring interactions on the developing brain. Prairie voles also exhibit natural variation in the level of tactile stimulation delivered by the parents to the offspring, and this has been well documented and quantified. Previous studies revealed that adult prairie vole offspring who received either high (HC) or low (LC) tactile contact from their parents have differences in the size of cortical fields and the connections of somatosensory cortex. In the current investigation, we examined gene expression, intraneocortical connectivity, and cortical thickness in newborn voles to appreciate when differences in HC and LC offspring begin to emerge. We observed differences in developmentally regulated genes, as well as variation in prelimbic and anterior cingulate cortical thickness at postnatal Day 1 (P1) in HC and LC voles. Results from this study suggest that parenting styles, such as those involving high or low physical contact, impact the developing neocortex via very early sensory experience as well as differences in epigenetic modifications that may emerge in HC and LC voles.
Collapse
Affiliation(s)
- Riley T Bottom
- Interdepartmental Neuroscience Program, University of California, Riverside, Riverside, California
| | - Leah A Krubitzer
- Center for Neuroscience, University of California, Davis, Davis, California.,Department of Psychology, University of California, Davis, Davis, California
| | - Kelly J Huffman
- Interdepartmental Neuroscience Program, University of California, Riverside, Riverside, California.,Department of Psychology, University of California, Riverside, Riverside, California
| |
Collapse
|
8
|
Xu X, Liu T, Wang Y, Fu J, Yang Q, Wu J, Zhou H. miRNA-mRNA Associated With Survival in Endometrial Cancer. Front Genet 2019; 10:743. [PMID: 31481972 PMCID: PMC6710979 DOI: 10.3389/fgene.2019.00743] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2018] [Accepted: 07/16/2019] [Indexed: 12/29/2022] Open
Abstract
Although various factors may contribute to its initiation and progression, the etiology and prognostic factors of endometrial carcinoma (EC) remains not fully understood. We sought to understand the role of changes in transcriptome during the progress of EC by exploring public datasets. The aberrant expression characteristics of EC based on RNA-Seq and miRNA-seq data from The Cancer Genome Atlas (TCGA) were analyzed. Kaplan-Meier analysis was performed to assess the relationship between differently expressed genes (DEGs) and patient survival. As a result, 320 out of 4,613 differently expressed mRNAs (DE mRNAs) and 68 out of 531 differently expressed miRNAs (DE miRNAs) with a significantly poorer survival were determined. We predicted eight paired DE miRNAs and DE mRNAs through TargetScan. Patients with three out of the eight paired low rate of miRNA/mRNA (miR-497/EMX1, miR-23c/DMBX1, and miR-670/KCNS1) expression had a significantly poorer survival. Furthermore, the simultaneous presence of these selected low miRNA/mRNA pairs occurred in most patients and resulted in a significantly poorer survival rate. Luciferase reporter assay identified that EMX1 was a direct target of miR-497. Both low expression of miR-497 and overexpression of EMX1 were significantly associated with more advanced clinicopathologic characteristics (stage, tumor status, grade, and histology) besides survival (all P values < 0.05). Multivariate analysis also demonstrated that miR-497 remained an independent prognostic variable for overall survival. In summary, we identified that a series of DE mRNAs and miRNAs, including eight paired DE miRNAs and mRNAs, were associated with survival in EC. Clinical evaluation of downregulated miR-497 and paired upregulated EMX1 confirmed the value of our prediction analysis. The simultaneous presence of low rate of these selected low miRNA/mRNA pairs (miR-497/EMX1, miR-23c/DMBX1, and miR-670/KCNS1) might have a better prediction value. Therefore, further studies are required to validate these findings.
Collapse
Affiliation(s)
- Xiaofeng Xu
- Department of Gynecology, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, China
| | - Tao Liu
- Department of Gynecology, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, China
- Medical College, Nanjing University, Nanjing, China
| | - Yijin Wang
- Department of Gynecology, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, China
- Medical College, Southeast University, Nanjing, China
| | - Jian Fu
- Department of Gynecology, Suqian People’s Hospital of Nanjing Drum Tower Hospital Group, Suqian, China
| | - Qian Yang
- Department of Gynecology and Obstetrics, The Pukou Hospital of Nanjing, The Fourth Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Jun Wu
- Department of Gynecology, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, China
| | - Huaijun Zhou
- Department of Gynecology, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, China
| |
Collapse
|
9
|
Hossain MM, Tsuzuki T, Sakakibara K, Imaizumi F, Ikegaya A, Inagaki M, Takahashi I, Ito T, Takamatsu H, Kumanogoh A, Negishi T, Yukawa K. PlexinA1 is crucial for the midline crossing of callosal axons during corpus callosum development in BALB/cAJ mice. PLoS One 2019; 14:e0221440. [PMID: 31430342 PMCID: PMC6701775 DOI: 10.1371/journal.pone.0221440] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2019] [Accepted: 08/06/2019] [Indexed: 12/04/2022] Open
Abstract
The corpus callosum (CC) is the biggest commissure that links cerebral hemispheres. Guidepost structures develop in the cortical midline during CC development and express axon guidance molecules that instruct neurons regarding the proper direction of axonal elongation toward and across the cortical midline. Neuropilin-1 (Npn1), a high affinity receptor for class 3 semaphorins (Sema3s) localized on cingulate pioneering axons, plays a crucial role in axon guidance to the midline through interactions with Sema3s. However, it remains unclear which type of Plexin is a component of Sema3 holoreceptors with Npn1 during the guidance of cingulate pioneering axons. To address the role of PlexinA1 in CC development, we examined with immunohistochemistry the localization of PlexinA1, Npn1, and Sema3s using embryonic brains from wild-type (WT) and PlexinA1-deficient (PlexinA1 knock-out (KO)) mice with a BALB/cAJ background. The immunohistochemistry confirmed the expression of PlexinA1 in callosal axons derived from the cingulate and neocortex of the WT mice on embryonic day 17.5 (E17.5) but not in the PlexinA1 KO mice. To examine the role of PlexinA1 in the navigation of callosal axons, the extension of callosal axons toward and across the midline was traced in brains of WT and PlexinA1 KO mice at E17.5. As a result, callosal axons in the PlexinA1 KO brains had a significantly lower incidence of midline crossing at E17.5 compared with the WT brains. To further examine the role of PlexinA1 in CC development, the CC phenotype was examined in PlexinA1 KO mice at postnatal day 0.5 (P0.5). Most of the PlexinA1 KO mice at P0.5 showed agenesis of the CC. These results indicate the crucial involvement of PlexinA1 in the midline crossing of callosal axons during CC development in BALB/cAJ mice.
Collapse
Affiliation(s)
| | - Takamasa Tsuzuki
- Department of Physiology, Faculty of Pharmacy, Meijo University, Nagoya, Japan
| | - Kazuki Sakakibara
- Department of Physiology, Faculty of Pharmacy, Meijo University, Nagoya, Japan
| | - Fumitaka Imaizumi
- Department of Physiology, Faculty of Pharmacy, Meijo University, Nagoya, Japan
| | - Akihiro Ikegaya
- Department of Physiology, Faculty of Pharmacy, Meijo University, Nagoya, Japan
| | - Mami Inagaki
- Department of Physiology, Faculty of Pharmacy, Meijo University, Nagoya, Japan
| | - Ikuko Takahashi
- Radioisotope Center, Faculty of Pharmacy, Meijo University, Nagoya, Japan
| | - Takuji Ito
- Department of Physiology, Faculty of Pharmacy, Meijo University, Nagoya, Japan
| | - Hyota Takamatsu
- Department of Immunopathology, Immunology Frontier Research Center, Osaka University, Suita, Japan
| | - Atsushi Kumanogoh
- Department of Immunopathology, Immunology Frontier Research Center, Osaka University, Suita, Japan
| | - Takayuki Negishi
- Department of Physiology, Faculty of Pharmacy, Meijo University, Nagoya, Japan
| | - Kazunori Yukawa
- Department of Physiology, Faculty of Pharmacy, Meijo University, Nagoya, Japan
- * E-mail:
| |
Collapse
|
10
|
Morales EE, Handa N, Drummond BE, Chambers JM, Marra AN, Addiego A, Wingert RA. Homeogene emx1 is required for nephron distal segment development in zebrafish. Sci Rep 2018; 8:18038. [PMID: 30575756 PMCID: PMC6303317 DOI: 10.1038/s41598-018-36061-4] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2018] [Accepted: 10/19/2018] [Indexed: 02/08/2023] Open
Abstract
Vertebrate kidneys contain nephron functional units where specialized epithelial cell types are organized into segments with discrete physiological roles. Many gaps remain in our understanding of how segment regions develop. Here, we report that the transcription factor empty spiracles homeobox gene 1 (emx1) is a novel nephron segment regulator during embryonic kidney development in zebrafish. emx1 loss of function altered the domains of distal segments without changes in cell turnover or traits like size and morphology, indicating that emx1 directs distal segment fates during nephrogenesis. In exploring how emx1 influences nephron patterning, we found that retinoic acid (RA), a morphogen that induces proximal and represses distal segments, negatively regulates emx1 expression. Next, through a series of genetic studies, we found that emx1 acts downstream of a cascade involving mecom and tbx2b, which encode essential distal segment transcription factors. Finally, we determined that emx1 regulates the expression domains of irx3b and irx1a to control distal segmentation, and sim1a to control corpuscle of Stannius formation. Taken together, our work reveals for the first time that emx1 is a key component of the pronephros segmentation network, which has implications for understanding the genetic regulatory cascades that orchestrate vertebrate nephron patterning.
Collapse
Affiliation(s)
- Elvin E Morales
- Department of Biological Sciences, Center for Stem Cells and Regenerative Medicine, Center for Zebrafish Research, University of Notre Dame, Notre Dame, IN, 46556, USA
| | - Nicole Handa
- Department of Biological Sciences, Center for Stem Cells and Regenerative Medicine, Center for Zebrafish Research, University of Notre Dame, Notre Dame, IN, 46556, USA
| | - Bridgette E Drummond
- Department of Biological Sciences, Center for Stem Cells and Regenerative Medicine, Center for Zebrafish Research, University of Notre Dame, Notre Dame, IN, 46556, USA
| | - Joseph M Chambers
- Department of Biological Sciences, Center for Stem Cells and Regenerative Medicine, Center for Zebrafish Research, University of Notre Dame, Notre Dame, IN, 46556, USA
| | - Amanda N Marra
- Department of Biological Sciences, Center for Stem Cells and Regenerative Medicine, Center for Zebrafish Research, University of Notre Dame, Notre Dame, IN, 46556, USA
| | - Amanda Addiego
- Department of Biological Sciences, Center for Stem Cells and Regenerative Medicine, Center for Zebrafish Research, University of Notre Dame, Notre Dame, IN, 46556, USA
| | - Rebecca A Wingert
- Department of Biological Sciences, Center for Stem Cells and Regenerative Medicine, Center for Zebrafish Research, University of Notre Dame, Notre Dame, IN, 46556, USA.
| |
Collapse
|
11
|
Transcriptional control of long-range cortical projections. Curr Opin Neurobiol 2018; 53:57-65. [DOI: 10.1016/j.conb.2018.05.005] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2018] [Accepted: 05/23/2018] [Indexed: 12/11/2022]
|
12
|
Schanze I, Bunt J, Lim JWC, Schanze D, Dean RJ, Alders M, Blanchet P, Attié-Bitach T, Berland S, Boogert S, Boppudi S, Bridges CJ, Cho MT, Dobyns WB, Donnai D, Douglas J, Earl DL, Edwards TJ, Faivre L, Fregeau B, Genevieve D, Gérard M, Gatinois V, Holder-Espinasse M, Huth SF, Izumi K, Kerr B, Lacaze E, Lakeman P, Mahida S, Mirzaa GM, Morgan SM, Nowak C, Peeters H, Petit F, Pilz DT, Puechberty J, Reinstein E, Rivière JB, Santani AB, Schneider A, Sherr EH, Smith-Hicks C, Wieland I, Zackai E, Zhao X, Gronostajski RM, Zenker M, Richards LJ. NFIB Haploinsufficiency Is Associated with Intellectual Disability and Macrocephaly. Am J Hum Genet 2018; 103:752-768. [PMID: 30388402 PMCID: PMC6218805 DOI: 10.1016/j.ajhg.2018.10.006] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2018] [Accepted: 10/03/2018] [Indexed: 12/19/2022] Open
Abstract
The nuclear factor I (NFI) family of transcription factors play an important role in normal development of multiple organs. Three NFI family members are highly expressed in the brain, and deletions or sequence variants in two of these, NFIA and NFIX, have been associated with intellectual disability (ID) and brain malformations. NFIB, however, has not previously been implicated in human disease. Here, we present a cohort of 18 individuals with mild ID and behavioral issues who are haploinsufficient for NFIB. Ten individuals harbored overlapping microdeletions of the chromosomal 9p23-p22.2 region, ranging in size from 225 kb to 4.3 Mb. Five additional subjects had point sequence variations creating a premature termination codon, and three subjects harbored single-nucleotide variations resulting in an inactive protein as determined using an in vitro reporter assay. All individuals presented with additional variable neurodevelopmental phenotypes, including muscular hypotonia, motor and speech delay, attention deficit disorder, autism spectrum disorder, and behavioral abnormalities. While structural brain anomalies, including dysgenesis of corpus callosum, were variable, individuals most frequently presented with macrocephaly. To determine whether macrocephaly could be a functional consequence of NFIB disruption, we analyzed a cortex-specific Nfib conditional knockout mouse model, which is postnatally viable. Utilizing magnetic resonance imaging and histology, we demonstrate that Nfib conditional knockout mice have enlargement of the cerebral cortex but preservation of overall brain structure and interhemispheric connectivity. Based on our findings, we propose that haploinsufficiency of NFIB causes ID with macrocephaly.
Collapse
Affiliation(s)
- Ina Schanze
- Institute of Human Genetics, University Hospital Magdeburg, Otto-von-Guericke University, Magdeburg 39120, Germany
| | - Jens Bunt
- Queensland Brain Institute, The University of Queensland, Brisbane, QLD 4072, Australia.
| | - Jonathan W C Lim
- Queensland Brain Institute, The University of Queensland, Brisbane, QLD 4072, Australia
| | - Denny Schanze
- Institute of Human Genetics, University Hospital Magdeburg, Otto-von-Guericke University, Magdeburg 39120, Germany
| | - Ryan J Dean
- Queensland Brain Institute, The University of Queensland, Brisbane, QLD 4072, Australia
| | - Marielle Alders
- Department of Clinical Genetics, Academic Medical Center, University of Amsterdam, Amsterdam 1105 AZ, the Netherlands
| | - Patricia Blanchet
- INSERM U1183, Département de Génétique Médicale, Maladies Rares et Médecine Personnalisée, Génétique clinique, CHU Montpellier, Université Montpellier, Centre de référence anomalies du développement SORO, Montpellier 34295, France
| | - Tania Attié-Bitach
- INSERM U1163, Laboratory of Embryology and Genetics of Congenital Malformations, Paris Descartes University, Sorbonne Paris Cité and Imagine Institute, Paris 75015, France
| | - Siren Berland
- Department of Medical Genetics, Haukeland University Hospital, Bergen 5021, Norway
| | - Steven Boogert
- Institute of Human Genetics, University Hospital Magdeburg, Otto-von-Guericke University, Magdeburg 39120, Germany
| | - Sangamitra Boppudi
- Institute of Human Genetics, University Hospital Magdeburg, Otto-von-Guericke University, Magdeburg 39120, Germany
| | - Caitlin J Bridges
- Institute of Human Genetics, University Hospital Magdeburg, Otto-von-Guericke University, Magdeburg 39120, Germany
| | | | - William B Dobyns
- Department of Pediatrics (Genetics), University of Washington and Center for Integrative Brain Research, Seattle Children's Research Institute, Seattle, WA 98101, USA
| | - Dian Donnai
- Manchester Centre for Genomic Medicine, Manchester Academic Health Science Centre, Central Manchester University Hospitals NHS Foundation Trust; Division of Evolution and Genomic Sciences School of Biological Sciences, and University of Manchester, Manchester M13 9WL, UK
| | - Jessica Douglas
- Boston Children's Hospital - The Feingold Center, Waltham, MA 02115, USA
| | - Dawn L Earl
- Division of Genetic Medicine, Seattle Children's Hospital, Seattle, WA 98105, USA
| | - Timothy J Edwards
- Queensland Brain Institute, The University of Queensland, Brisbane, QLD 4072, Australia; The Faculty of Medicine Brisbane, The University of Queensland, Brisbane, QLD 4072, Australia
| | - Laurence Faivre
- UMR1231, Génétique des Anomalies du Développement, Université de Bourgogne, Dijon 21079, France; Centre de Génétique et Centre de Référence Anomalies du Développement et Syndromes Malformatifs de l'Interrégion Est et FHU TRANSLAD, Centre Hospitalier Universitaire Dijon, Dijon 21079, France
| | - Brieana Fregeau
- Department of Neurology, University of California, San Francisco, San Francisco, CA 94158, USA
| | - David Genevieve
- INSERM U1183, Département de Génétique Médicale, Maladies Rares et Médecine Personnalisée, Génétique clinique, CHU Montpellier, Université Montpellier, Centre de référence anomalies du développement SORO, Montpellier 34295, France
| | - Marion Gérard
- Service de Génétique, CHU de Caen - Hôpital Clémenceau, Caen Cedex 14000, France
| | - Vincent Gatinois
- INSERM U1183, Département de Génétique Médicale, Maladies Rares et Médecine Personnalisée, Génétique clinique, CHU Montpellier, Université Montpellier, Centre de référence anomalies du développement SORO, Montpellier 34295, France
| | - Muriel Holder-Espinasse
- Service de Génétique Clinique, Hôpital Jeanne de Flandre, CHU Lille, Lille 59000, France; Department of Clinical Genetics, Guy's Hospital, London SE1 9RT, UK
| | - Samuel F Huth
- Queensland Brain Institute, The University of Queensland, Brisbane, QLD 4072, Australia
| | - Kosuke Izumi
- Department of Pediatrics, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA; Division of Genetics, Department of Pediatrics, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Bronwyn Kerr
- Manchester Centre for Genomic Medicine, Manchester Academic Health Science Centre, Central Manchester University Hospitals NHS Foundation Trust; Division of Evolution and Genomic Sciences School of Biological Sciences, and University of Manchester, Manchester M13 9WL, UK
| | - Elodie Lacaze
- Department of genetics, Le Havre Hospital, 76600 Le Havre, France
| | - Phillis Lakeman
- Department of Clinical Genetics, Academic Medical Center, University of Amsterdam, Amsterdam 1105 AZ, the Netherlands
| | - Sonal Mahida
- Department of Neurogenetics, Kennedy Krieger Institute, Baltimore, MD 21205, USA
| | - Ghayda M Mirzaa
- Department of Pediatrics (Genetics), University of Washington and Center for Integrative Brain Research, Seattle Children's Research Institute, Seattle, WA 98101, USA
| | - Sian M Morgan
- All Wales Genetics Laboratory, Institute of Medical Genetics, University Hospital of Wales, Cardiff CF14 4XW, UK
| | - Catherine Nowak
- Boston Children's Hospital - The Feingold Center, Waltham, MA 02115, USA
| | - Hilde Peeters
- Center for Human Genetics, University Hospital Leuven, KU Leuven, Leuven 3000, Belgium
| | - Florence Petit
- Service de Génétique Clinique, Hôpital Jeanne de Flandre, CHU Lille, Lille 59000, France
| | - Daniela T Pilz
- West of Scotland Genetics Service, Queen Elizabeth University Hospital, Glasgow G51 4TF, UK
| | - Jacques Puechberty
- INSERM U1183, Département de Génétique Médicale, Maladies Rares et Médecine Personnalisée, Génétique clinique, CHU Montpellier, Université Montpellier, Centre de référence anomalies du développement SORO, Montpellier 34295, France
| | - Eyal Reinstein
- Medical Genetics Institute, Meir Medical Center, Kfar-Saba 4428164, Israel; Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 6997801, Israel
| | - Jean-Baptiste Rivière
- UMR1231, Génétique des Anomalies du Développement, Université de Bourgogne, Dijon 21079, France; Centre de Génétique et Centre de Référence Anomalies du Développement et Syndromes Malformatifs de l'Interrégion Est et FHU TRANSLAD, Centre Hospitalier Universitaire Dijon, Dijon 21079, France; Child Health and Human Development Program, Research Institute of the McGill University Health Centre, Montreal, QC H4A 3J1, Canada
| | - Avni B Santani
- Division of Genomic Diagnostics, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA; Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Anouck Schneider
- INSERM U1183, Département de Génétique Médicale, Maladies Rares et Médecine Personnalisée, Génétique clinique, CHU Montpellier, Université Montpellier, Centre de référence anomalies du développement SORO, Montpellier 34295, France
| | - Elliott H Sherr
- Department of Neurology, University of California, San Francisco, San Francisco, CA 94158, USA
| | | | - Ilse Wieland
- Institute of Human Genetics, University Hospital Magdeburg, Otto-von-Guericke University, Magdeburg 39120, Germany
| | - Elaine Zackai
- Department of Pediatrics, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Xiaonan Zhao
- Division of Genomic Diagnostics, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Richard M Gronostajski
- Department of Biochemistry, Program in Genetics, Genomics and Bioinformatics, Center of Excellence in Bioinformatics and Life Sciences, State University of New York at Buffalo, Buffalo, NY 14203, USA
| | - Martin Zenker
- Institute of Human Genetics, University Hospital Magdeburg, Otto-von-Guericke University, Magdeburg 39120, Germany.
| | - Linda J Richards
- Queensland Brain Institute, The University of Queensland, Brisbane, QLD 4072, Australia; School of Biomedical Sciences, The Faculty of Medicine Brisbane, The University of Queensland, Brisbane, QLD 4072, Australia
| |
Collapse
|
13
|
Frey S, Eichler A, Stonawski V, Kriebel J, Wahl S, Gallati S, Goecke TW, Fasching PA, Beckmann MW, Kratz O, Moll GH, Heinrich H, Kornhuber J, Golub Y. Prenatal Alcohol Exposure Is Associated With Adverse Cognitive Effects and Distinct Whole-Genome DNA Methylation Patterns in Primary School Children. Front Behav Neurosci 2018; 12:125. [PMID: 29997484 PMCID: PMC6028559 DOI: 10.3389/fnbeh.2018.00125] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2017] [Accepted: 06/06/2018] [Indexed: 12/22/2022] Open
Abstract
Prenatal alcohol exposure (PAE) is known to elicit a broad range of systemic effects, including neurophysiological alterations that result in adverse behavioral and cognitive outcomes. However, molecular pathways underlying these long-term intrauterine effects remain to be investigated. Here, we tested a hypothesis that PAE may lead to epigenetic alterations to the DNA resulting in attentional and cognitive alterations of the children. We report the results of the study that included 156 primary school children of the Franconian Cognition and Emotion Studies (FRANCES) cohort which were tested for an objective marker of PAE, ethyl glucuronide (EtG) in meconium at birth. Thirty-two newborns were found to be exposed to alcohol with EtG values above 30 ng/g (EtG+). Previously we described PAE being associated with lower IQ and smaller amplitude of the event-related potential component P3 in go trials (Go-P3), which indicates a reduced capacity of attentional resources. Whole-genome methylation analysis of the buccal cell DNA revealed 193 differentially methylated genes in children with positive meconium EtG, that were clustered into groups involved in epigenetic modifications, neurodegeneration, neurodevelopment, axon guidance and neuronal excitability. Furthermore, we detected mediation effects of the methylation changes in DPP10 and SLC16A9 genes on the EtG related cognitive and attention-related deficits. Our results suggest that system-wide epigenetic changes are involved in long-term effects of PAE. In particular, we show an epigenetic mediation of PAE effects on cognition and attention-related processes.
Collapse
Affiliation(s)
- Stefan Frey
- Department of Child and Adolescent Mental Health, University Hospital Erlangen, Friedrich-Alexander University Erlangen-Nürnberg, Erlangen, Germany
| | - Anna Eichler
- Department of Child and Adolescent Mental Health, University Hospital Erlangen, Friedrich-Alexander University Erlangen-Nürnberg, Erlangen, Germany
| | - Valeska Stonawski
- Department of Child and Adolescent Mental Health, University Hospital Erlangen, Friedrich-Alexander University Erlangen-Nürnberg, Erlangen, Germany
| | - Jennifer Kriebel
- Research Unit of Molecular Epidemiology, German Research Center for Environmental Health - Institute of Epidemiology II, Helmholtz Zentrum München, Munich, Germany
| | - Simone Wahl
- Research Unit of Molecular Epidemiology, German Research Center for Environmental Health - Institute of Epidemiology II, Helmholtz Zentrum München, Munich, Germany
| | - Sabina Gallati
- Division of Human Genetics, Department of Paediatrics, Inselspital University of Bern, Bern, Switzerland
| | - Tamme W Goecke
- Department of Obstetrics and Gynecology, University Hospital Erlangen, Friedrich-Alexander University Erlangen-Nürnberg, Erlangen, Germany.,Department of Obstetrics and Gynecology, RWTH Aachen University, Aachen, Germany
| | - Peter A Fasching
- Department of Obstetrics and Gynecology, RWTH Aachen University, Aachen, Germany
| | - Matthias W Beckmann
- Department of Obstetrics and Gynecology, RWTH Aachen University, Aachen, Germany
| | - Oliver Kratz
- Department of Child and Adolescent Mental Health, University Hospital Erlangen, Friedrich-Alexander University Erlangen-Nürnberg, Erlangen, Germany
| | - Gunther H Moll
- Department of Child and Adolescent Mental Health, University Hospital Erlangen, Friedrich-Alexander University Erlangen-Nürnberg, Erlangen, Germany
| | - Hartmut Heinrich
- Department of Child and Adolescent Mental Health, University Hospital Erlangen, Friedrich-Alexander University Erlangen-Nürnberg, Erlangen, Germany.,kbo-Heckscher-Klinikum, Munich, Germany
| | - Johannes Kornhuber
- Department of Psychiatry and Psychotherapy, University Hospital Erlangen, Friedrich-Alexander University Erlangen-Nürnberg, Erlangen, Germany
| | - Yulia Golub
- Department of Child and Adolescent Mental Health, University Hospital Erlangen, Friedrich-Alexander University Erlangen-Nürnberg, Erlangen, Germany.,Department of Child and Adolescent Psychiatry, Faculty of Medicine of the TU Dresden, Dresden, Germany
| |
Collapse
|
14
|
Bunt J, Osinski JM, Lim JW, Vidovic D, Ye Y, Zalucki O, O'Connor TR, Harris L, Gronostajski RM, Richards LJ, Piper M. Combined allelic dosage of Nfia and Nfib regulates cortical development. Brain Neurosci Adv 2017; 1:2398212817739433. [PMID: 32166136 PMCID: PMC7058261 DOI: 10.1177/2398212817739433] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2017] [Accepted: 10/01/2017] [Indexed: 12/02/2022] Open
Abstract
Background: Nuclear factor I family members nuclear factor I A and nuclear factor I B play important roles during cerebral cortical development. Nuclear factor I A and nuclear factor I B regulate similar biological processes, as their expression patterns, regulation of target genes and individual knockout phenotypes overlap. We hypothesised that the combined allelic loss of Nfia and Nfib would culminate in more severe defects in the cerebral cortex than loss of a single member. Methods: We combined immunofluorescence, co-immunoprecipitation, gene expression analysis and immunohistochemistry on knockout mouse models to investigate whether nuclear factor I A and nuclear factor I B function similarly and whether increasing allelic loss of Nfia and Nfib caused a more severe phenotype. Results: We determined that the biological functions of nuclear factor I A and nuclear factor I B overlap during early cortical development. These proteins are co-expressed and can form heterodimers in vivo. Differentially regulated genes that are shared between Nfia and Nfib knockout mice are highly enriched for nuclear factor I binding sites in their promoters and are associated with neurodevelopment. We found that compound heterozygous deletion of both genes resulted in a cortical phenotype similar to that of single homozygous Nfia or Nfib knockout embryos. This was characterised by retention of the interhemispheric fissure, dysgenesis of the corpus callosum and a malformed dentate gyrus. Double homozygous knockout of Nfia and Nfib resulted in a more severe phenotype, with increased ventricular enlargement and decreased numbers of differentiated glia and neurons. Conclusion: In the developing cerebral cortex, nuclear factor I A and nuclear factor I B share similar biological functions and function additively, as the combined allelic loss of these genes directly correlates with the severity of the developmental brain phenotype.
Collapse
Affiliation(s)
- Jens Bunt
- The Queensland Brain Institute, The University of Queensland, Brisbane, QLD, Australia
| | - Jason M Osinski
- Department of Biochemistry, Program in Genetics, Genomics and Bioinformatics, Center of Excellence in Bioinformatics and Life Sciences, State University of New York at Buffalo, Buffalo, New York, USA
| | - Jonathan Wc Lim
- The Queensland Brain Institute, The University of Queensland, Brisbane, QLD, Australia
| | - Diana Vidovic
- The School of Biomedical Sciences, Faculty of Medicine, The University of Queensland, Brisbane, QLD, Australia
| | - Yunan Ye
- The Queensland Brain Institute, The University of Queensland, Brisbane, QLD, Australia
| | - Oressia Zalucki
- The School of Biomedical Sciences, Faculty of Medicine, The University of Queensland, Brisbane, QLD, Australia
| | - Timothy R O'Connor
- School of Chemical and Molecular Biosciences and Institute for Molecular Bioscience, The University of Queensland, Brisbane, QLD, Australia
| | - Lachlan Harris
- The School of Biomedical Sciences, Faculty of Medicine, The University of Queensland, Brisbane, QLD, Australia
| | - Richard M Gronostajski
- Department of Biochemistry, Program in Genetics, Genomics and Bioinformatics, Center of Excellence in Bioinformatics and Life Sciences, State University of New York at Buffalo, Buffalo, New York, USA
| | - Linda J Richards
- The Queensland Brain Institute, The University of Queensland, Brisbane, QLD, Australia.,The School of Biomedical Sciences, Faculty of Medicine, The University of Queensland, Brisbane, QLD, Australia
| | - Michael Piper
- The Queensland Brain Institute, The University of Queensland, Brisbane, QLD, Australia.,The School of Biomedical Sciences, Faculty of Medicine, The University of Queensland, Brisbane, QLD, Australia
| |
Collapse
|
15
|
Kobeissy FH, Hansen K, Neumann M, Fu S, Jin K, Liu J. Deciphering the Role of Emx1 in Neurogenesis: A Neuroproteomics Approach. Front Mol Neurosci 2016; 9:98. [PMID: 27799894 PMCID: PMC5065984 DOI: 10.3389/fnmol.2016.00098] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2016] [Accepted: 09/26/2016] [Indexed: 12/18/2022] Open
Abstract
Emx1 has long been implicated in embryonic brain development. Previously we found that mice null of Emx1 gene had smaller dentate gyri and reduced neurogenesis, although the molecular mechanisms underlying this defect was not well understood. To decipher the role of Emx1 gene in neural regeneration and the timing of its involvement, we determine the frequency of neural stem cells (NSCs) in embryonic and adult forebrains of Emx1 wild type (WT) and knock out (KO) mice in the neurosphere assay. Emx1 gene deletion reduced the frequency and self-renewal capacity of NSCs of the embryonic brain but did not affect neuronal or glial differentiation. Emx1 KO NSCs also exhibited a reduced migratory capacity in response to serum or vascular endothelial growth factor (VEGF) in the Boyden chamber migration assay compared to their WT counterparts. A thorough comparison between NSC lysates from Emx1 WT and KO mice utilizing 2D-PAGE coupled with tandem mass spectrometry revealed 38 proteins differentially expressed between genotypes, including the F-actin depolymerization factor Cofilin. A global systems biology and cluster analysis identified several potential mechanisms and cellular pathways implicated in altered neurogenesis, all involving Cofilin1. Protein interaction network maps with functional enrichment analysis further indicated that the differentially expressed proteins participated in neural-specific functions including brain development, axonal guidance, synaptic transmission, neurogenesis, and hippocampal morphology, with VEGF as the upstream regulator intertwined with Cofilin1 and Emx1. Functional validation analysis indicated that apart from the overall reduced level of phosphorylated Cofilin1 (p-Cofilin1) in the Emx1 KO NSCs compared to WT NSCs as demonstrated in the western blot analysis, VEGF was able to induce more Cofilin1 phosphorylation and FLK expression only in the latter. Our results suggest that a defect in Cofilin1 phosphorylation induced by VEGF or other growth factors might contribute to the reduced neurogenesis in the Emx1 null mice during brain development.
Collapse
Affiliation(s)
- Firas H Kobeissy
- Department of Psychiatry, Center for Neuroproteomics and Biomarkers Research, University of Florida Gainesville, FL, USA
| | - Katharina Hansen
- Department of Neurological Surgery, University of California, San FranciscoSan Francisco, CA, USA; San Francisco VA Medical CenterSan Francisco, CA, USA
| | - Melanie Neumann
- Department of Neurological Surgery, University of California, San FranciscoSan Francisco, CA, USA; San Francisco VA Medical CenterSan Francisco, CA, USA
| | - Shuping Fu
- Department of Neurological Surgery, University of California, San FranciscoSan Francisco, CA, USA; San Francisco VA Medical CenterSan Francisco, CA, USA; Key Laboratory of Acupuncture and Medicine Research of Minister of Education, Nanjing University of Chinese MedicineNanjing, China
| | - Kulin Jin
- Pharmacology & Neuroscience, University of North Texas Health Science Center Fort Worth, TX, USA
| | - Jialing Liu
- Department of Neurological Surgery, University of California, San FranciscoSan Francisco, CA, USA; San Francisco VA Medical CenterSan Francisco, CA, USA
| |
Collapse
|
16
|
Stocker AM, O’Leary DDM. Emx1 Is Required for Neocortical Area Patterning. PLoS One 2016; 11:e0149900. [PMID: 26901526 PMCID: PMC4762677 DOI: 10.1371/journal.pone.0149900] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2015] [Accepted: 02/05/2016] [Indexed: 11/19/2022] Open
Abstract
Establishing appropriate area patterning in the neocortex is a critical developmental event, and transcription factors whose expression is graded across the developing neural axes have been implicated in this process. While previous reports suggested that the transcription factor Emx1 does not contribute to neocortical area patterning, those studies were performed at perinatal ages prior to the emergence of primary areas. We therefore examined two different Emx1 deletion mouse lines once primary areas possess mature features. Following the deletion of Emx1, the frontal and motor areas were expanded while the primary visual area was reduced, and overall the areas shifted posterio-medially. This patterning phenotype was consistent between the two Emx1 deletion strategies. The present study demonstrates that Emx1 is an area patterning transcription factor and is required for the specification of the primary visual area.
Collapse
Affiliation(s)
- Adam M. Stocker
- Molecular Neurobiology Laboratory, The Salk Institute, 10010 North Torrey Pines Road, La Jolla, California, 92037, United States of America
- * E-mail:
| | - Dennis D. M. O’Leary
- Molecular Neurobiology Laboratory, The Salk Institute, 10010 North Torrey Pines Road, La Jolla, California, 92037, United States of America
| |
Collapse
|