1
|
Brooks ER, Moorman AR, Bhattacharya B, Prudhomme I, Land M, Alcorn HL, Sharma R, Pe’er D, Zallen JA. A single-cell atlas of spatial and temporal gene expression in the mouse cranial neural plate. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.08.25.609458. [PMID: 39229123 PMCID: PMC11370589 DOI: 10.1101/2024.08.25.609458] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/05/2024]
Abstract
The formation of the mammalian brain requires regionalization and morphogenesis of the cranial neural plate, which transforms from an epithelial sheet into a closed tube that provides the structural foundation for neural patterning and circuit formation. Sonic hedgehog (SHH) signaling is important for cranial neural plate patterning and closure, but the transcriptional changes that give rise to the spatially regulated cell fates and behaviors that build the cranial neural tube have not been systematically analyzed. Here we used single-cell RNA sequencing to generate an atlas of gene expression at six consecutive stages of cranial neural tube closure in the mouse embryo. Ordering transcriptional profiles relative to the major axes of gene expression predicted spatially regulated expression of 870 genes along the anterior-posterior and mediolateral axes of the cranial neural plate and reproduced known expression patterns with over 85% accuracy. Single-cell RNA sequencing of embryos with activated SHH signaling revealed distinct SHH-regulated transcriptional programs in the developing forebrain, midbrain, and hindbrain, suggesting a complex interplay between anterior-posterior and mediolateral patterning systems. These results define a spatiotemporally resolved map of gene expression during cranial neural tube closure and provide a resource for investigating the transcriptional events that drive early mammalian brain development.
Collapse
Affiliation(s)
- Eric R. Brooks
- HHMI and Developmental Biology Program, Sloan Kettering Institute
- North Carolina State University
| | - Andrew R. Moorman
- HHMI and Computational and Systems Biology Program, Sloan Kettering Institute
| | | | - Ian Prudhomme
- HHMI and Developmental Biology Program, Sloan Kettering Institute
| | - Max Land
- HHMI and Computational and Systems Biology Program, Sloan Kettering Institute
| | | | - Roshan Sharma
- HHMI and Computational and Systems Biology Program, Sloan Kettering Institute
| | - Dana Pe’er
- HHMI and Computational and Systems Biology Program, Sloan Kettering Institute
| | | |
Collapse
|
2
|
Hudson DT, Bromell JS, Day RC, McInnes T, Ward JM, Beck CW. Gene expression analysis of the Xenopus laevis early limb bud proximodistal axis. Dev Dyn 2022; 251:1880-1896. [PMID: 35809036 PMCID: PMC9796579 DOI: 10.1002/dvdy.517] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2022] [Revised: 06/30/2022] [Accepted: 07/06/2022] [Indexed: 01/01/2023] Open
Abstract
BACKGROUND Limb buds develop as bilateral outgrowths of the lateral plate mesoderm and are patterned along three axes. Current models of proximal to distal patterning of early amniote limb buds suggest that two signals, a distal organizing signal from the apical epithelial ridge (AER, Fgfs) and an opposing proximal (retinoic acid [RA]) act early on pattern this axis. RESULTS Transcriptional analysis of stage 51 Xenopus laevis hindlimb buds sectioned along the proximal-distal axis showed that the distal region is distinct from the rest of the limb. Expression of capn8.3, a novel calpain, was located in cells immediately flanking the AER. The Wnt antagonist Dkk1 was AER-specific in Xenopus limbs. Two transcription factors, sall1 and zic5, were expressed in distal mesenchyme. Zic5 has no described association with limb development. We also describe expression of two proximal genes, gata5 and tnn, not previously associated with limb development. Differentially expressed genes were associated with Fgf, Wnt, and RA signaling as well as differential cell adhesion and proliferation. CONCLUSIONS We identify new candidate genes for early proximodistal limb patterning. Our analysis of RA-regulated genes supports a role for transient RA gradients in early limb bud in proximal-to-distal patterning in this anamniote model organism.
Collapse
Affiliation(s)
- Daniel T. Hudson
- Department of ZoologyUniversity of OtagoDunedinNew Zealand,Oritain GlobalDunedinNew Zealand
| | - Jessica S. Bromell
- Department of ZoologyUniversity of OtagoDunedinNew Zealand,Dairy Goat Co‐operativeHamiltonNew Zealand
| | - Robert C. Day
- Department of BiochemistryUniversity of OtagoDunedinNew Zealand
| | - Tyler McInnes
- Department of ZoologyUniversity of OtagoDunedinNew Zealand
| | - Joanna M. Ward
- Department of ZoologyUniversity of OtagoDunedinNew Zealand
| | | |
Collapse
|
3
|
Luo S, Dong X, Guo S, Wang Q, Dai X, Jiang Y, Zhu W, Zhou W, Song W. Regulation of the Human IL-10RB Gene Expression by Sp8 and Sp9. J Alzheimers Dis 2022; 88:1469-1485. [PMID: 35811529 DOI: 10.3233/jad-220321] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
BACKGROUND Interleukin-10 (IL-10) is a classic anti-inflammatory cytokine that exerts its effects via the receptor complexes IL-10RA and IL-10RB. Loss of IL-10RB results in many diseases. Moreover, IL-10RB is closely associated with neuronal survival and synaptic formation. However, the regulation of IL-10RB gene expression remains elusive. OBJECTIVE To investigate whether the expression of IL-10RB gene is increased in brain of Alzheimer's disease (AD) and its transcriptional regulation. METHODS We examined the gene expression of AD patient brain from public database and detected the protein expression of AD model mouse brain by western blot. We constructed a variety of reporter gene plasmids with different lengths or mutation sites, tested the promoter activity and defined the functional region of the promoter with the luciferase reporter assay. The protein-DNA binding between transcription factors and the promoter was analyzed using chromatin immunoprecipitation (ChIP) and electrophoretic mobility shift assay (EMSA). RESULTS We found that the IL-10RB is elevated in the brain of AD patient and AD model mice. The minimal promoter of the IL-10RB gene is located in the -90 to +51 bp region (relative to the transcriptional start site) and is sufficient for high-level expression of the IL-10RB gene. Transcription factors Sp8 and Sp9 bind to the IL-10RB promoter in vitro. The overexpression or knockdown of Sp8 and Sp9 affected the IL-10RB promoter activity and its gene expression. CONCLUSION Our study functionally characterized the promoter of the IL-10RB gene and demonstrated that Sp8 and Sp9 regulated its expression.
Collapse
Affiliation(s)
- Shuyue Luo
- Chongqing Key Laboratory of Translational Medical Research in Cognitive Development and Learning and Memory Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, National Clinical Research Center for Child Health and Disorders, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Children's Hospital of Chongqing Medical University, Chongqing, China
| | - Xiangjun Dong
- Chongqing Key Laboratory of Translational Medical Research in Cognitive Development and Learning and Memory Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, National Clinical Research Center for Child Health and Disorders, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Children's Hospital of Chongqing Medical University, Chongqing, China
| | - Shipeng Guo
- Chongqing Key Laboratory of Translational Medical Research in Cognitive Development and Learning and Memory Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, National Clinical Research Center for Child Health and Disorders, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Children's Hospital of Chongqing Medical University, Chongqing, China
| | - Qunxian Wang
- Chongqing Key Laboratory of Translational Medical Research in Cognitive Development and Learning and Memory Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, National Clinical Research Center for Child Health and Disorders, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Children's Hospital of Chongqing Medical University, Chongqing, China
| | - Xi Dai
- Chongqing Key Laboratory of Translational Medical Research in Cognitive Development and Learning and Memory Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, National Clinical Research Center for Child Health and Disorders, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Children's Hospital of Chongqing Medical University, Chongqing, China
| | - Yanshuang Jiang
- Chongqing Key Laboratory of Translational Medical Research in Cognitive Development and Learning and Memory Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, National Clinical Research Center for Child Health and Disorders, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Children's Hospital of Chongqing Medical University, Chongqing, China
| | - Weiyi Zhu
- Chongqing Key Laboratory of Translational Medical Research in Cognitive Development and Learning and Memory Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, National Clinical Research Center for Child Health and Disorders, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Children's Hospital of Chongqing Medical University, Chongqing, China
| | - Weihui Zhou
- Chongqing Key Laboratory of Translational Medical Research in Cognitive Development and Learning and Memory Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, National Clinical Research Center for Child Health and Disorders, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Children's Hospital of Chongqing Medical University, Chongqing, China
| | - Weihong Song
- Chongqing Key Laboratory of Translational Medical Research in Cognitive Development and Learning and Memory Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, National Clinical Research Center for Child Health and Disorders, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Children's Hospital of Chongqing Medical University, Chongqing, China.,Institute of Aging, Key Laboratory of Alzheimer's Disease of Zhejiang Province, Zhejiang Provincial Clinical Research Center for Mental Disorders, School of Mental Health and Kangning Hospital, The Second Affiliated Hospital and Yuying Children's Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, China.,Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), Wenzhou, Zhejiang, China
| |
Collapse
|
4
|
Choudhury SR, Dutta S, Bhaduri U, Rao MRS. LncRNA Hmrhl regulates expression of cancer related genes in chronic myelogenous leukemia through chromatin association. NAR Cancer 2021; 3:zcab042. [PMID: 34734184 PMCID: PMC8559160 DOI: 10.1093/narcan/zcab042] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2021] [Revised: 10/11/2021] [Accepted: 10/19/2021] [Indexed: 12/13/2022] Open
Abstract
Long non-coding RNA has emerged as a key regulator of myriad gene functions. One such lncRNA mrhl, reported by our group, was found to have important role in spermatogenesis and embryonic development in mouse. Recently, its human homolog, Hmrhl was shown to have differential expression in several type of cancers. In the present study, we further characterize molecular features of Hmrhl and gain insight into its functional role in leukemia by gene silencing and transcriptome-based studies. Results indicate its high expression in CML patient samples as well as in K562 cell line. Silencing experiments suggest role of Hmrhl in cell proliferation, migration & invasion. RNA-seq and ChiRP-seq data analysis further revealed its association with important biological processes, including perturbed expression of crucial TFs and cancer-related genes. Among them ZIC1, PDGRFβ and TP53 were identified as regulatory targets, with high possibility of triplex formation by Hmrhl at their promoter site. Further, overexpression of PDGRFβ in Hmrhl silenced cells resulted in rescue effect of cancer associated cellular phenotypes. In addition, we also found TAL-1 to be a potential regulator of Hmrhl expression in K562 cells. Thus, we hypothesize that Hmrhl lncRNA may play a significant role in the pathobiology of CML.
Collapse
Affiliation(s)
- Subhendu Roy Choudhury
- Molecular Biology and Genetics Unit, Jawaharlal Nehru Centre for Advance Scientific Research, Bangalore, India
| | - Sangeeta Dutta
- Molecular Biology and Genetics Unit, Jawaharlal Nehru Centre for Advance Scientific Research, Bangalore, India
| | - Utsa Bhaduri
- Molecular Biology and Genetics Unit, Jawaharlal Nehru Centre for Advance Scientific Research, Bangalore, India
| | | |
Collapse
|
5
|
Shih Y, Wang K, Kumano G, Nishida H. Expression and Functional Analyses of Ectodermal Transcription Factors FoxJ-r, SoxF, and SP8/9 in Early Embryos of the Ascidian Halocynthia roretzi. Zoolog Sci 2021; 38:26-35. [PMID: 33639715 DOI: 10.2108/zs200128] [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: 08/11/2020] [Accepted: 09/06/2020] [Indexed: 11/17/2022]
Abstract
The spatiotemporal expression of zygotic genes is regulated by transcription factors, which mediate cell fate decision and morphogenesis. Investigation of the expression patterns and their transcriptional regulatory relationships is crucial to understand embryonic development. Staged RNA-seq of the ascidian Halocynthia roretzi has previously shown that nine genes encoding transcription factors are transiently expressed at the blastula stage, which is the stage at which cell fates are specified and differentiation starts. Six of these transcription factors have already been found to play important roles during early development. However, the functions of the other transcription factors (FoxJ-r, SoxF, and SP8/9) remain unknown. The study of the spatial and temporal expression patterns showed that all three genes were expressed in the animal hemisphere as early as the 16-cell stage. This is likely due to transcription factor genes that are expressed in the vegetal hemisphere, which have been extensively and comprehensively analyzed in previous studies of ascidians. Functional analyses using FoxJ-r morphants showed that they resulted in the disruption of laterality and the absence of epidermal mono-cilia, suggesting FoxJ-r functions in cilia formation and, consequently, in the generation of left-right asymmetry, as observed in vertebrates. SoxF knockdown resulted in incomplete epiboly by the ectoderm during gastrulation, while SP8/9 knockdown showed no phenotype until the tailbud stage in the present study, although it was expressed during blastula stages. Our results indicate that transcription factor genes expressed at the cleavage stages play roles in diverse functions, and are not limited to cell fate specification.
Collapse
Affiliation(s)
- Yu Shih
- Department of Biological Sciences, Graduate School of Science, Osaka University, Toyonaka, Osaka 560-0043, Japan,
| | - Kai Wang
- Department of Biological Sciences, Graduate School of Science, Osaka University, Toyonaka, Osaka 560-0043, Japan
| | - Gaku Kumano
- Research Center for Marine Biology, Graduate School of Life Sciences, Tohoku University, Aomori 039-3501, Japan
| | - Hiroki Nishida
- Department of Biological Sciences, Graduate School of Science, Osaka University, Toyonaka, Osaka 560-0043, Japan
| |
Collapse
|
6
|
Powell DL, Payne C, Banerjee SM, Keegan M, Bashkirova E, Cui R, Andolfatto P, Rosenthal GG, Schumer M. The Genetic Architecture of Variation in the Sexually Selected Sword Ornament and Its Evolution in Hybrid Populations. Curr Biol 2021; 31:923-935.e11. [PMID: 33513352 DOI: 10.1016/j.cub.2020.12.049] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2020] [Revised: 10/27/2020] [Accepted: 12/25/2020] [Indexed: 10/22/2022]
Abstract
Biologists since Darwin have been fascinated by the evolution of sexually selected ornaments, particularly those that reduce viability. Uncovering the genetic architecture of these traits is key to understanding how they evolve and are maintained. Here, we investigate the genetic architecture and evolutionary loss of a sexually selected ornament, the "sword" fin extension that characterizes many species of swordtail fish (Xiphophorus). Using sworded and swordless sister species of Xiphophorus, we generated a mapping population and show that the sword ornament is polygenic-with ancestry across the genome explaining substantial variation in the trait. After accounting for the impacts of genome-wide ancestry, we identify one major-effect quantitative trait locus (QTL) that explains ~5% of the overall variation in the trait. Using a series of approaches, we narrow this large QTL interval to several likely candidate genes, including genes involved in fin regeneration and growth. Furthermore, we find evidence of selection on ancestry at one of these candidates in four natural hybrid populations, consistent with selection against the sword in these populations.
Collapse
Affiliation(s)
- Daniel L Powell
- Department of Biology, Stanford University, 327 Campus Drive, Stanford, CA 94305, USA; Centro de Investigaciones Científicas de las Huastecas "Aguazarca," A.C., 16 de Septiembre, 392 Barrio Aguazarca, Calnali, Hidalgo 43240, México; Department of Biology, Texas A&M University, 3258 TAMU, College Station, TX 77843, USA.
| | - Cheyenne Payne
- Department of Biology, Stanford University, 327 Campus Drive, Stanford, CA 94305, USA; Centro de Investigaciones Científicas de las Huastecas "Aguazarca," A.C., 16 de Septiembre, 392 Barrio Aguazarca, Calnali, Hidalgo 43240, México
| | - Shreya M Banerjee
- Department of Biology, Stanford University, 327 Campus Drive, Stanford, CA 94305, USA; Centro de Investigaciones Científicas de las Huastecas "Aguazarca," A.C., 16 de Septiembre, 392 Barrio Aguazarca, Calnali, Hidalgo 43240, México
| | - Mackenzie Keegan
- Department of Biology, Northeastern University, 360 Huntington Avenue, Boston, MA 02115, USA
| | - Elizaveta Bashkirova
- Department of Biochemistry and Molecular Biophysics, Columbia University, 701 West 168th Street, New York, NY 10032, USA; Integrated Program in Cellular, Molecular and Biomedical Studies, Columbia University Irving Medical Center, 622 West 168th Street, New York, NY 10032, USA
| | - Rongfeng Cui
- Centro de Investigaciones Científicas de las Huastecas "Aguazarca," A.C., 16 de Septiembre, 392 Barrio Aguazarca, Calnali, Hidalgo 43240, México; Department of Biology, Texas A&M University, 3258 TAMU, College Station, TX 77843, USA; Max Planck Institute for the Biology of Aging, Postfach 41 06 23, 50931 Cologne, Germany; School of Ecology, Sun Yat-sen University, 135 Xingang West Road, Binjiang Road, Haizhu District, Guangdong Province, China
| | - Peter Andolfatto
- Department of Biological Sciences, Columbia University, 1212 Amsterdam Avenue, New York, NY 10027, USA
| | - Gil G Rosenthal
- Centro de Investigaciones Científicas de las Huastecas "Aguazarca," A.C., 16 de Septiembre, 392 Barrio Aguazarca, Calnali, Hidalgo 43240, México; Department of Biology, Texas A&M University, 3258 TAMU, College Station, TX 77843, USA
| | - Molly Schumer
- Department of Biology, Stanford University, 327 Campus Drive, Stanford, CA 94305, USA; Centro de Investigaciones Científicas de las Huastecas "Aguazarca," A.C., 16 de Septiembre, 392 Barrio Aguazarca, Calnali, Hidalgo 43240, México; Howard Hughes Medical Institute, 327 Campus Drive, Stanford, CA 94305, USA.
| |
Collapse
|
7
|
McCulloch KJ, Koenig KM. Krüppel-like factor/specificity protein evolution in the Spiralia and the implications for cephalopod visual system novelties. Proc Biol Sci 2020; 287:20202055. [PMID: 33081641 PMCID: PMC7661307 DOI: 10.1098/rspb.2020.2055] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
The cephalopod visual system is an exquisite example of convergence in biological complexity. However, we have little understanding of the genetic and molecular mechanisms underpinning its elaboration. The generation of new genetic material is considered a significant contributor to the evolution of biological novelty. We sought to understand if this mechanism may be contributing to cephalopod-specific visual system novelties. Specifically, we identified duplications in the Krüppel-like factor/specificity protein (KLF/SP) sub-family of C2H2 zinc-finger transcription factors in the squid Doryteuthis pealeii. We cloned and analysed gene expression of the KLF/SP family, including two paralogs of the DpSP6-9 gene. These duplicates showed overlapping expression domains but one paralog showed unique expression in the developing squid lens, suggesting a neofunctionalization of DpSP6-9a. To better understand this neofunctionalization, we performed a thorough phylogenetic analysis of SP6-9 orthologues in the Spiralia. We find multiple duplications and losses of the SP6-9 gene throughout spiralian lineages and at least one cephalopod-specific duplication. This work supports the hypothesis that gene duplication and neofunctionalization contribute to novel traits like the cephalopod image-forming eye and to the diversity found within Spiralia.
Collapse
Affiliation(s)
- Kyle J McCulloch
- Department of Organismic and Evolutionary Biology, Harvard University Cambridge, MA 02138, USA.,John Harvard Distinguished Science Fellows, Harvard University, Cambridge, MA 02138, USA
| | - Kristen M Koenig
- Department of Organismic and Evolutionary Biology, Harvard University Cambridge, MA 02138, USA.,John Harvard Distinguished Science Fellows, Harvard University, Cambridge, MA 02138, USA
| |
Collapse
|
8
|
Blom-Dahl D, Córdoba S, Gabilondo H, Carr-Baena P, Díaz-Benjumea FJ, Estella C. In vivo analysis of the evolutionary conserved BTD-box domain of Sp1 and Btd during Drosophila development. Dev Biol 2020; 466:77-89. [PMID: 32738261 DOI: 10.1016/j.ydbio.2020.07.011] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2020] [Revised: 07/20/2020] [Accepted: 07/21/2020] [Indexed: 10/23/2022]
Abstract
The Sp family of transcription factors plays important functions during development and disease. An evolutionary conserved role for some Sp family members is the control of limb development. The family is characterized by the presence of three C2H2-type zinc fingers and an adjacent 10 aa region with an unknown function called the Buttonhead (BTD) box. The presence of this BTD-box in all Sp family members identified from arthropods to vertebrates, suggests that it plays an essential role during development. However, despite its conservation, the in vivo function of the BTD-box has never been studied. In this work, we have generated specific BTD-box deletion alleles for the Drosophila Sp family members Sp1 and buttonhead (btd) using gene editing tools and analyzed its role during development. Unexpectedly, btd and Sp1 mutant alleles that lack the BTD-box are viable and have almost normal appendages. However, in a sensitized background the requirement of this domain to fully regulate some of Sp1 and Btd target genes is revealed. Furthermore, we have also identified a novel Sp1 role promoting leg vs antenna identity through the repression of spineless (ss) expression in the leg, a function that also depends on the Sp1 BTD-box.
Collapse
Affiliation(s)
- David Blom-Dahl
- Centro de Biología Molecular "Severo Ochoa", CSIC-UAM, Nicolás Cabrera 1, 28049, Madrid, Spain
| | - Sergio Córdoba
- Centro de Biología Molecular "Severo Ochoa", CSIC-UAM, Nicolás Cabrera 1, 28049, Madrid, Spain
| | - Hugo Gabilondo
- Centro de Biología Molecular "Severo Ochoa", CSIC-UAM, Nicolás Cabrera 1, 28049, Madrid, Spain
| | - Pablo Carr-Baena
- Centro de Biología Molecular "Severo Ochoa", CSIC-UAM, Nicolás Cabrera 1, 28049, Madrid, Spain
| | | | - Carlos Estella
- Centro de Biología Molecular "Severo Ochoa", CSIC-UAM, Nicolás Cabrera 1, 28049, Madrid, Spain.
| |
Collapse
|
9
|
Guo T, Liu G, Du H, Wen Y, Wei S, Li Z, Tao G, Shang Z, Song X, Zhang Z, Xu Z, You Y, Chen B, Rubenstein JL, Yang Z. Dlx1/2 are Central and Essential Components in the Transcriptional Code for Generating Olfactory Bulb Interneurons. Cereb Cortex 2019; 29:4831-4849. [PMID: 30796806 PMCID: PMC6917526 DOI: 10.1093/cercor/bhz018] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2018] [Revised: 01/03/2019] [Accepted: 01/26/2019] [Indexed: 12/22/2022] Open
Abstract
Generation of olfactory bulb (OB) interneurons requires neural stem/progenitor cell specification, proliferation, differentiation, and young interneuron migration and maturation. Here, we show that the homeobox transcription factors Dlx1/2 are central and essential components in the transcriptional code for generating OB interneurons. In Dlx1/2 constitutive null mutants, the differentiation of GSX2+ and ASCL1+ neural stem/progenitor cells in the dorsal lateral ganglionic eminence is blocked, resulting in a failure of OB interneuron generation. In Dlx1/2 conditional mutants (hGFAP-Cre; Dlx1/2F/- mice), GSX2+ and ASCL1+ neural stem/progenitor cells in the postnatal subventricular zone also fail to differentiate into OB interneurons. In contrast, overexpression of Dlx1&2 in embryonic mouse cortex led to ectopic production of OB-like interneurons that expressed Gad1, Sp8, Sp9, Arx, Pbx3, Etv1, Tshz1, and Prokr2. Pax6 mutants generate cortical ectopia with OB-like interneurons, but do not do so in compound Pax6; Dlx1/2 mutants. We propose that DLX1/2 promote OB interneuron development mainly through activating the expression of Sp8/9, which further promote Tshz1 and Prokr2 expression. Based on this study, in combination with earlier ones, we propose a transcriptional network for the process of OB interneuron development.
Collapse
Affiliation(s)
- Teng Guo
- State Key Laboratory of Medical Neurobiology, Institutes of Brain Science, MOE Frontier Research Center for Brain Science, Department of Neurology, Zhongshan Hospital, Fudan University, Shanghai 200032, PR China
| | - Guoping Liu
- State Key Laboratory of Medical Neurobiology, Institutes of Brain Science, MOE Frontier Research Center for Brain Science, Department of Neurology, Zhongshan Hospital, Fudan University, Shanghai 200032, PR China
| | - Heng Du
- State Key Laboratory of Medical Neurobiology, Institutes of Brain Science, MOE Frontier Research Center for Brain Science, Department of Neurology, Zhongshan Hospital, Fudan University, Shanghai 200032, PR China
| | - Yan Wen
- State Key Laboratory of Medical Neurobiology, Institutes of Brain Science, MOE Frontier Research Center for Brain Science, Department of Neurology, Zhongshan Hospital, Fudan University, Shanghai 200032, PR China
| | - Song Wei
- State Key Laboratory of Medical Neurobiology, Institutes of Brain Science, MOE Frontier Research Center for Brain Science, Department of Neurology, Zhongshan Hospital, Fudan University, Shanghai 200032, PR China
| | - Zhenmeiyu Li
- State Key Laboratory of Medical Neurobiology, Institutes of Brain Science, MOE Frontier Research Center for Brain Science, Department of Neurology, Zhongshan Hospital, Fudan University, Shanghai 200032, PR China
| | - Guangxu Tao
- State Key Laboratory of Medical Neurobiology, Institutes of Brain Science, MOE Frontier Research Center for Brain Science, Department of Neurology, Zhongshan Hospital, Fudan University, Shanghai 200032, PR China
| | - Zicong Shang
- State Key Laboratory of Medical Neurobiology, Institutes of Brain Science, MOE Frontier Research Center for Brain Science, Department of Neurology, Zhongshan Hospital, Fudan University, Shanghai 200032, PR China
| | - Xiaolei Song
- State Key Laboratory of Medical Neurobiology, Institutes of Brain Science, MOE Frontier Research Center for Brain Science, Department of Neurology, Zhongshan Hospital, Fudan University, Shanghai 200032, PR China
| | - Zhuangzhi Zhang
- State Key Laboratory of Medical Neurobiology, Institutes of Brain Science, MOE Frontier Research Center for Brain Science, Department of Neurology, Zhongshan Hospital, Fudan University, Shanghai 200032, PR China
| | - Zhejun Xu
- State Key Laboratory of Medical Neurobiology, Institutes of Brain Science, MOE Frontier Research Center for Brain Science, Department of Neurology, Zhongshan Hospital, Fudan University, Shanghai 200032, PR China
| | - Yan You
- State Key Laboratory of Medical Neurobiology, Institutes of Brain Science, MOE Frontier Research Center for Brain Science, Department of Neurology, Zhongshan Hospital, Fudan University, Shanghai 200032, PR China
| | - Bin Chen
- Department of Molecular, Cell and Developmental Biology, University of California, Santa Cruz, CA 95064, USA
| | - John L Rubenstein
- Department of Psychiatry, Nina Ireland Laboratory of Developmental Neurobiology, UCSF Weill Institute for Neurosciences, University of California, San Francisco, CA 94158, USA
| | - Zhengang Yang
- State Key Laboratory of Medical Neurobiology, Institutes of Brain Science, MOE Frontier Research Center for Brain Science, Department of Neurology, Zhongshan Hospital, Fudan University, Shanghai 200032, PR China
| |
Collapse
|
10
|
Li J, Wang C, Zhang Z, Wen Y, An L, Liang Q, Xu Z, Wei S, Li W, Guo T, Liu G, Tao G, You Y, Du H, Fu Z, He M, Chen B, Campbell K, Alvarez-Buylla A, Rubenstein JL, Yang Z. Transcription Factors Sp8 and Sp9 Coordinately Regulate Olfactory Bulb Interneuron Development. Cereb Cortex 2019; 28:3278-3294. [PMID: 28981617 DOI: 10.1093/cercor/bhx199] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2017] [Accepted: 07/13/2017] [Indexed: 01/01/2023] Open
Abstract
Neural stem cells in the postnatal telencephalic ventricular-subventricular zone (V-SVZ) generate new interneurons, which migrate tangentially through the rostral migratory stream (RMS) into the olfactory bulb (OB). The Sp8 and Sp9 transcription factors are expressed in neuroblasts, as well as in the immature and mature interneurons in the V-SVZ-RMS-OB system. Here we show that Sp8 and Sp9 coordinately regulate OB interneuron development: although Sp9 null mutants show no major OB interneuron defect, conditional deletion of both Sp8 and Sp9 resulted in a much more severe reduction of OB interneuron number than that observed in the Sp8 conditional mutant mice, due to defects in neuronal differentiation, tangential and radial migration, and increased cell death in the V-SVZ-RMS-OB system. RNA-Seq and RNA in situ hybridization reveal that, in Sp8/Sp9 double mutant mice, but not in Sp8 or Sp9 single mutant mice, newly born neuroblasts in the V-SVZ-RMS-OB system fail to express Prokr2 and Tshz1 expression, genes with known roles in promoting OB interneuron differentiation and migration, and that are involved in human Kallmann syndrome.
Collapse
Affiliation(s)
- Jiwen Li
- Department of Translational Neuroscience, Shanghai Pudong Hospital, State Key Laboratory of Medical Neurobiology, Institutes of Brain Science, and Collaborative Innovation Center for Brain Science, Fudan University, Shanghai, China
| | - Chunyang Wang
- Department of Translational Neuroscience, Shanghai Pudong Hospital, State Key Laboratory of Medical Neurobiology, Institutes of Brain Science, and Collaborative Innovation Center for Brain Science, Fudan University, Shanghai, China
| | - Zhuangzhi Zhang
- Department of Translational Neuroscience, Shanghai Pudong Hospital, State Key Laboratory of Medical Neurobiology, Institutes of Brain Science, and Collaborative Innovation Center for Brain Science, Fudan University, Shanghai, China
| | - Yan Wen
- Department of Translational Neuroscience, Shanghai Pudong Hospital, State Key Laboratory of Medical Neurobiology, Institutes of Brain Science, and Collaborative Innovation Center for Brain Science, Fudan University, Shanghai, China
| | - Lei An
- Department of Translational Neuroscience, Shanghai Pudong Hospital, State Key Laboratory of Medical Neurobiology, Institutes of Brain Science, and Collaborative Innovation Center for Brain Science, Fudan University, Shanghai, China
| | - Qifei Liang
- Department of Translational Neuroscience, Shanghai Pudong Hospital, State Key Laboratory of Medical Neurobiology, Institutes of Brain Science, and Collaborative Innovation Center for Brain Science, Fudan University, Shanghai, China
| | - Zhejun Xu
- Department of Translational Neuroscience, Shanghai Pudong Hospital, State Key Laboratory of Medical Neurobiology, Institutes of Brain Science, and Collaborative Innovation Center for Brain Science, Fudan University, Shanghai, China
| | - Song Wei
- Department of Translational Neuroscience, Shanghai Pudong Hospital, State Key Laboratory of Medical Neurobiology, Institutes of Brain Science, and Collaborative Innovation Center for Brain Science, Fudan University, Shanghai, China
| | - Weiwei Li
- Department of Translational Neuroscience, Shanghai Pudong Hospital, State Key Laboratory of Medical Neurobiology, Institutes of Brain Science, and Collaborative Innovation Center for Brain Science, Fudan University, Shanghai, China
| | - Teng Guo
- Department of Translational Neuroscience, Shanghai Pudong Hospital, State Key Laboratory of Medical Neurobiology, Institutes of Brain Science, and Collaborative Innovation Center for Brain Science, Fudan University, Shanghai, China
| | - Guoping Liu
- Department of Translational Neuroscience, Shanghai Pudong Hospital, State Key Laboratory of Medical Neurobiology, Institutes of Brain Science, and Collaborative Innovation Center for Brain Science, Fudan University, Shanghai, China
| | - Guangxu Tao
- Department of Translational Neuroscience, Shanghai Pudong Hospital, State Key Laboratory of Medical Neurobiology, Institutes of Brain Science, and Collaborative Innovation Center for Brain Science, Fudan University, Shanghai, China
| | - Yan You
- Department of Translational Neuroscience, Shanghai Pudong Hospital, State Key Laboratory of Medical Neurobiology, Institutes of Brain Science, and Collaborative Innovation Center for Brain Science, Fudan University, Shanghai, China
| | - Heng Du
- Department of Translational Neuroscience, Shanghai Pudong Hospital, State Key Laboratory of Medical Neurobiology, Institutes of Brain Science, and Collaborative Innovation Center for Brain Science, Fudan University, Shanghai, China
| | - Zhuoning Fu
- Department of Translational Neuroscience, Shanghai Pudong Hospital, State Key Laboratory of Medical Neurobiology, Institutes of Brain Science, and Collaborative Innovation Center for Brain Science, Fudan University, Shanghai, China
| | - Miao He
- Department of Translational Neuroscience, Shanghai Pudong Hospital, State Key Laboratory of Medical Neurobiology, Institutes of Brain Science, and Collaborative Innovation Center for Brain Science, Fudan University, Shanghai, China
| | - Bin Chen
- Department of Molecular, Cell and Developmental Biology, University of California, Santa Cruz, CA, USA
| | - Kenneth Campbell
- Division of Developmental Biology, Cincinnati Children's Hospital Medical Center, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Arturo Alvarez-Buylla
- Department of Neurological Surgery, The Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California, San Francisco, CA, USA
| | - John L Rubenstein
- Department of Psychiatry, Nina Ireland Laboratory of Developmental Neurobiology, UCSF Weill Institute for Neurosciences, University of California, San Francisco, CA, USA
| | - Zhengang Yang
- Department of Translational Neuroscience, Shanghai Pudong Hospital, State Key Laboratory of Medical Neurobiology, Institutes of Brain Science, and Collaborative Innovation Center for Brain Science, Fudan University, Shanghai, China
| |
Collapse
|
11
|
Richieri-Costa A, Zechi-Ceide RM, Candido-Souza RM, Monteiro RAC, Tonello C, de Freitas ML, Kokitsu-Nakata NM, Vendramini-Pittoli S, Mazzeu JF, Overes M, Ali-Amin R, van Slegtenhorst M, Hoefsloot LH, Jehee FS. Holoprosencephaly, orofacial cleft, and frontonaso-orbital encephaloceles: Genetic evaluation of a possible new syndrome. Am J Med Genet A 2019; 179:2170-2177. [PMID: 31353810 DOI: 10.1002/ajmg.a.61305] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2018] [Revised: 06/18/2019] [Accepted: 07/09/2019] [Indexed: 11/06/2022]
Abstract
Here we report on a Brazilian child who presented semilobar holoprosencephaly, frontonasal encephaloceles and bilateral cleft lip and palate. Malformations also included agenesis of the corpus callosum, abnormal cortical gyres, dilation of the aqueduct, bilateral endolymphatic sac, bilateral cystic cocci-vestibular malformation, and a cribriform defect. The 3D TC craniofacial images showed abnormal frontonasal transition region, with a bone bifurcation, and partial agenesis of nasal bone. The trunk and upper and lower limbs were normal. To our knowledge, this rare association of holoprocensephaly with frontonaso-orbital encephaloceles without limb anomalies has never been reported before. Karyotype was normal. SNP-array showed no copy-number alterations but revealed 25% of regions of homozygosity (ROH) with normal copy number, indicating a high coefficient of inbreeding, which significantly increases the risk for an autosomal recessive disorder. Whole exome sequencing analysis did not reveal any pathogenic or likely pathogenic variants. We discuss the possible influence of two variants of uncertain significance found within the patient's ROHs. First, a missense p.(Gly394Ser) in PCSK9, a gene involved in the regulation of plasma low-density lipoprotein cholesterol. Second, an inframe duplication p.(Ala75_Ala81dup) in SP8, a zinc-finger transcription factor that regulates signaling centers during craniofacial development. Further studies and/or the identification of other patients with a similar phenotype will help elucidate the genetic etiology of this complex case.
Collapse
Affiliation(s)
- Antonio Richieri-Costa
- Department of Clinical Genetics, Hospital for Rehabilitation of Craniofacial Anomalies (HRCA), University of São Paulo, Bauru, São Paulo, Brazil
| | - Roseli M Zechi-Ceide
- Department of Clinical Genetics, Hospital for Rehabilitation of Craniofacial Anomalies (HRCA), University of São Paulo, Bauru, São Paulo, Brazil
| | - Rosana M Candido-Souza
- Department of Clinical Genetics, Hospital for Rehabilitation of Craniofacial Anomalies (HRCA), University of São Paulo, Bauru, São Paulo, Brazil
| | - Rejane A C Monteiro
- Institute of Education and Research, Santa Casa Belo Horizonte, Belo Horizonte, Minas Gerais, Brazil
| | - Cristiano Tonello
- Craniofacial Team Hospital for Rehabilitation of Craniofacial Anomalies (HRCA), University of São Paulo, Bauru, São Paulo, Brazil
| | - Mariana L de Freitas
- Institute of Education and Research, Santa Casa Belo Horizonte, Belo Horizonte, Minas Gerais, Brazil
| | - Nancy M Kokitsu-Nakata
- Department of Clinical Genetics, Hospital for Rehabilitation of Craniofacial Anomalies (HRCA), University of São Paulo, Bauru, São Paulo, Brazil
| | - Siulan Vendramini-Pittoli
- Department of Clinical Genetics, Hospital for Rehabilitation of Craniofacial Anomalies (HRCA), University of São Paulo, Bauru, São Paulo, Brazil
| | - Juliana F Mazzeu
- Faculty of Medicine, University of Brasília, Brasília, Distrito Federal, Brazil
| | - Madelief Overes
- Department of Clinical Genetics, Erasmus MC, Rotterdam, CN, The Netherlands
| | - Roza Ali-Amin
- Department of Clinical Genetics, Erasmus MC, Rotterdam, CN, The Netherlands
| | | | - Lies H Hoefsloot
- Department of Clinical Genetics, Erasmus MC, Rotterdam, CN, The Netherlands
| | - Fernanda S Jehee
- Institute of Education and Research, Santa Casa Belo Horizonte, Belo Horizonte, Minas Gerais, Brazil.,Department of Clinical Genetics, Erasmus MC, Rotterdam, CN, The Netherlands
| |
Collapse
|
12
|
Wei S, Du H, Li Z, Tao G, Xu Z, Song X, Shang Z, Su Z, Chen H, Wen Y, Liu G, You Y, Zhang Z, Yang Z. Transcription factors
Sp8
and
Sp9
regulate the development of caudal ganglionic eminence‐derived cortical interneurons. J Comp Neurol 2019; 527:2860-2874. [DOI: 10.1002/cne.24712] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2019] [Revised: 04/16/2019] [Accepted: 05/03/2019] [Indexed: 12/13/2022]
Affiliation(s)
- Song Wei
- State Key Laboratory of Medical Neurobiology, Institutes of Brain Science, MOE Frontier Research Center for Brain Science, Department of Neurology, Zhongshan HospitalFudan University Shanghai China
| | - Heng Du
- State Key Laboratory of Medical Neurobiology, Institutes of Brain Science, MOE Frontier Research Center for Brain Science, Department of Neurology, Zhongshan HospitalFudan University Shanghai China
| | - Zhenmeiyu Li
- State Key Laboratory of Medical Neurobiology, Institutes of Brain Science, MOE Frontier Research Center for Brain Science, Department of Neurology, Zhongshan HospitalFudan University Shanghai China
| | - Guangxu Tao
- State Key Laboratory of Medical Neurobiology, Institutes of Brain Science, MOE Frontier Research Center for Brain Science, Department of Neurology, Zhongshan HospitalFudan University Shanghai China
| | - Zhejun Xu
- State Key Laboratory of Medical Neurobiology, Institutes of Brain Science, MOE Frontier Research Center for Brain Science, Department of Neurology, Zhongshan HospitalFudan University Shanghai China
| | - Xiaolei Song
- State Key Laboratory of Medical Neurobiology, Institutes of Brain Science, MOE Frontier Research Center for Brain Science, Department of Neurology, Zhongshan HospitalFudan University Shanghai China
| | - Zicong Shang
- State Key Laboratory of Medical Neurobiology, Institutes of Brain Science, MOE Frontier Research Center for Brain Science, Department of Neurology, Zhongshan HospitalFudan University Shanghai China
| | - Zihao Su
- State Key Laboratory of Medical Neurobiology, Institutes of Brain Science, MOE Frontier Research Center for Brain Science, Department of Neurology, Zhongshan HospitalFudan University Shanghai China
| | - Haotian Chen
- State Key Laboratory of Medical Neurobiology, Institutes of Brain Science, MOE Frontier Research Center for Brain Science, Department of Neurology, Zhongshan HospitalFudan University Shanghai China
| | - Yan Wen
- State Key Laboratory of Medical Neurobiology, Institutes of Brain Science, MOE Frontier Research Center for Brain Science, Department of Neurology, Zhongshan HospitalFudan University Shanghai China
| | - Guoping Liu
- State Key Laboratory of Medical Neurobiology, Institutes of Brain Science, MOE Frontier Research Center for Brain Science, Department of Neurology, Zhongshan HospitalFudan University Shanghai China
| | - Yan You
- State Key Laboratory of Medical Neurobiology, Institutes of Brain Science, MOE Frontier Research Center for Brain Science, Department of Neurology, Zhongshan HospitalFudan University Shanghai China
| | - Zhuangzhi Zhang
- State Key Laboratory of Medical Neurobiology, Institutes of Brain Science, MOE Frontier Research Center for Brain Science, Department of Neurology, Zhongshan HospitalFudan University Shanghai China
| | - Zhengang Yang
- State Key Laboratory of Medical Neurobiology, Institutes of Brain Science, MOE Frontier Research Center for Brain Science, Department of Neurology, Zhongshan HospitalFudan University Shanghai China
| |
Collapse
|
13
|
Pregnancy Promotes Maternal Hippocampal Neurogenesis in Guinea Pigs. Neural Plast 2019; 2019:5765284. [PMID: 31097956 PMCID: PMC6487096 DOI: 10.1155/2019/5765284] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2018] [Revised: 02/08/2019] [Accepted: 02/21/2019] [Indexed: 11/26/2022] Open
Abstract
Adult neurogenesis in the hippocampal dentate gyrus (DG) modulates cognition and behavior in mammals, while motherhood is associated with cognitive and behavioral changes essential for the care of the young. In mice and rats, hippocampal neurogenesis is reported to be reduced or unchanged during pregnancy, with few data available from other species. In guinea pigs, pregnancy lasts ~9 weeks; we set to explore if hippocampal neurogenesis is altered in these animals, relative to gestational stages. Time-pregnant primigravidas (3-5 months old) and age-matched nonpregnant females were examined, with neurogenic potential evaluated via immunolabeling of Ki67, Sp8, doublecortin (DCX), and neuron-specific nuclear antigen (NeuN) combined with bromodeoxyuridine (BrdU) birth-dating. Relative to control, subgranular Ki67, Sp8, and DCX-immunoreactive (+) cells tended to increase from early gestation to postpartum and peaked at the late gestational stage. In BrdU pulse-chasing experiments in nonpregnant females surviving for different time points (2-120 days), BrdU+ cells in the DG colocalized with DCX partially from 2 to 42 days (most frequently at 14-30 days) and with NeuN increasingly from 14 to 120 days. In pregnant females that received BrdU at early, middle, and late gestational stages and survived for 42 days, the density of BrdU+ cells in the DG was mostly high in the late gestational group. The rates of BrdU/DCX and BrdU/NeuN colocalization were similar among these groups and comparable to those among the corresponding control group. Together, the findings suggest that pregnancy promotes maternal hippocampal neurogenesis in guinea pigs, at least among primigravidas.
Collapse
|
14
|
Tao G, Li Z, Wen Y, Song X, Wei S, Du H, Yang Z, Xu Z, You Y. Transcription Factors Sp8 and Sp9 Regulate Medial Ganglionic Eminence-Derived Cortical Interneuron Migration. Front Mol Neurosci 2019; 12:75. [PMID: 31001083 PMCID: PMC6454190 DOI: 10.3389/fnmol.2019.00075] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2019] [Accepted: 03/11/2019] [Indexed: 12/11/2022] Open
Abstract
Cortical interneurons are derived from the subpallium and reach the developing cortex through long tangential migration. Mature cortical interneurons are characterized by remarkable morphological, molecular, and functional diversity. The calcium-binding protein parvalbumin (PV) and neuropeptide somatostatin (SST) identify most medial ganglionic eminence (MGE)-derived cortical interneurons. Previously, we demonstrated that Sp9 plays a curial transcriptional role in regulating MGE-derived cortical interneuron development. Here, we show that SP8 protein is weekly expressed in the MGE mantle zone of wild type mice but upregulated in Sp9 null mutants. PV+ cortical interneurons were severely lost in Sp8/Sp9 double conditional knockouts due to defects in tangential migration compared with Sp9 single mutants, suggesting that Sp8/9 coordinately regulate PV+ cortical interneuron development. We provide evidence that Sp8/Sp9 activity is required for normal MGE-derived cortical interneuron migration, at least in part, through regulating the expression of EphA3, Ppp2r2c, and Rasgef1b.
Collapse
Affiliation(s)
- Guangxu Tao
- State Key Laboratory of Medical Neurobiology, MOE Frontier Research Center for Brain Science, Department of Neurology, Institutes of Brain Science, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Zhenmeiyu Li
- State Key Laboratory of Medical Neurobiology, MOE Frontier Research Center for Brain Science, Department of Neurology, Institutes of Brain Science, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Yan Wen
- State Key Laboratory of Medical Neurobiology, MOE Frontier Research Center for Brain Science, Department of Neurology, Institutes of Brain Science, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Xiaolei Song
- State Key Laboratory of Medical Neurobiology, MOE Frontier Research Center for Brain Science, Department of Neurology, Institutes of Brain Science, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Song Wei
- State Key Laboratory of Medical Neurobiology, MOE Frontier Research Center for Brain Science, Department of Neurology, Institutes of Brain Science, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Heng Du
- State Key Laboratory of Medical Neurobiology, MOE Frontier Research Center for Brain Science, Department of Neurology, Institutes of Brain Science, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Zhengang Yang
- State Key Laboratory of Medical Neurobiology, MOE Frontier Research Center for Brain Science, Department of Neurology, Institutes of Brain Science, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Zhejun Xu
- State Key Laboratory of Medical Neurobiology, MOE Frontier Research Center for Brain Science, Department of Neurology, Institutes of Brain Science, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Yan You
- State Key Laboratory of Medical Neurobiology, MOE Frontier Research Center for Brain Science, Department of Neurology, Institutes of Brain Science, Zhongshan Hospital, Fudan University, Shanghai, China
| |
Collapse
|
15
|
Linz DM, Hu Y, Moczek AP. The origins of novelty from within the confines of homology: the developmental evolution of the digging tibia of dung beetles. Proc Biol Sci 2019; 286:20182427. [PMID: 30963933 PMCID: PMC6408602 DOI: 10.1098/rspb.2018.2427] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2018] [Accepted: 01/23/2019] [Indexed: 11/12/2022] Open
Abstract
Understanding the origin of novel complex traits is among the most fundamental goals in evolutionary biology. The most widely used definition of novelty in evolution assumes the absence of homology, yet where homology ends and novelty begins is increasingly difficult to parse as evo devo continuously revises our understanding of what constitutes homology. Here, we executed a case study to explore the earliest stages of innovation by examining the tibial teeth of tunnelling dung beetles. Tibial teeth are a morphologically modest innovation, composed of relatively simple body wall projections and contained fully within the fore tibia, a leg segment whose own homology status is unambiguous. We first demonstrate that tibial teeth aid in multiple digging behaviours. We then show that the developmental evolution of tibial teeth was dominated by the redeployment of locally pre-existing gene networks. At the same time, we find that even at this very early stage of innovation, at least two genes that ancestrally function in embryonic patterning and thus entirely outside the spatial and temporal context of leg formation, have already become recruited to help shape the formation of tibial teeth. Our results suggest a testable model for how developmental evolution scaffolds innovation.
Collapse
|
16
|
Kantaputra PN, Carlson BM. Genetic regulatory pathways of split-hand/foot malformation. Clin Genet 2018; 95:132-139. [PMID: 30101460 DOI: 10.1111/cge.13434] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2018] [Accepted: 08/07/2018] [Indexed: 12/13/2022]
Abstract
Split-hand/foot malformation (SHFM) is caused by mutations in TP63, DLX5, DLX6, FGF8, FGFR1, WNT10B, and BHLHA9. The clinical features of SHFM caused by mutations of these genes are not distinguishable. This implies that in normal situations these SHFM-associated genes share an underlying regulatory pathway that is involved in the development of the central parts of the hands and feet. The mutations in SHFM-related genes lead to dysregulation of Fgf8 in the central portion of the apical ectodermal ridge (AER) and subsequently lead to misexpression of a number of downstream target genes, failure of stratification of the AER, and thus SHFM. Syndactyly of the remaining digits is most likely the effects of dysregulation of Fgf-Bmp-Msx signaling on apoptotic cell death. Loss of digit identity in SHFM is hypothesized to be the effects of misexpression of HOX genes, abnormal SHH gradient, or the loss of balance between GLI3A and GLI3R. Disruption of canonical and non-canonical Wnt signaling is involved in the pathogenesis of SHFM. Whatever the causative genes of SHFM are, the mutations seem to lead to dysregulation of Fgf8 in AER cells of the central parts of the hands and feet and disruption of Wnt-Bmp-Fgf signaling pathways in AER.
Collapse
Affiliation(s)
- Piranit N Kantaputra
- Center of Excellence in Medical Genetics Research, Chiang Mai University, Chiang Mai, Thailand.,Division of Pediatric Dentistry, Department of Orthodontics and Pediatric Dentistry, Faculty of Dentistry, Chiang Mai University, Chiang Mai, Thailand.,Dentaland Clinic, Chiang Mai, Thailand
| | - Bruce M Carlson
- Department of Anatomy and Cell Biology, University of Michigan, Ann Arbor, Michigan
| |
Collapse
|
17
|
Xu Z, Liang Q, Song X, Zhang Z, Lindtner S, Li Z, Wen Y, Liu G, Guo T, Qi D, Wang M, Wang C, Li H, You Y, Wang X, Chen B, Feng H, Rubenstein JL, Yang Z. SP8 and SP9 coordinately promote D2-type medium spiny neuron production by activating Six3 expression. Development 2018; 145:dev165456. [PMID: 29967281 PMCID: PMC6078334 DOI: 10.1242/dev.165456] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2018] [Accepted: 06/18/2018] [Indexed: 12/21/2022]
Abstract
Dopamine receptor DRD1-expressing medium spiny neurons (D1 MSNs) and dopamine receptor DRD2-expressing medium spiny neurons (D2 MSNs) are the principal projection neurons in the striatum, which is divided into dorsal striatum (caudate nucleus and putamen) and ventral striatum (nucleus accumbens and olfactory tubercle). Progenitors of these neurons arise in the lateral ganglionic eminence (LGE). Using conditional deletion, we show that mice lacking the transcription factor genes Sp8 and Sp9 lose virtually all D2 MSNs as a result of reduced neurogenesis in the LGE, whereas D1 MSNs are largely unaffected. SP8 and SP9 together drive expression of the transcription factor Six3 in a spatially restricted domain of the LGE subventricular zone. Conditional deletion of Six3 also prevents the formation of most D2 MSNs, phenocopying the Sp8/9 mutants. Finally, ChIP-Seq reveals that SP9 directly binds to the promoter and a putative enhancer of Six3 Thus, this study defines components of a transcription pathway in a regionally restricted LGE progenitor domain that selectively drives the generation of D2 MSNs.
Collapse
Affiliation(s)
- Zhejun Xu
- State Key Laboratory of Medical Neurobiology, Institutes of Brain Science, Department of Neurology, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Qifei Liang
- State Key Laboratory of Medical Neurobiology, Institutes of Brain Science, Department of Neurology, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Xiaolei Song
- State Key Laboratory of Medical Neurobiology, Institutes of Brain Science, Department of Neurology, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Zhuangzhi Zhang
- State Key Laboratory of Medical Neurobiology, Institutes of Brain Science, Department of Neurology, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Susan Lindtner
- Department of Psychiatry, Nina Ireland Laboratory of Developmental Neurobiology, UCSF Weill Institute for Neurosciences, University of California, San Francisco, CA 94158, USA
| | - Zhenmeiyu Li
- State Key Laboratory of Medical Neurobiology, Institutes of Brain Science, Department of Neurology, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Yan Wen
- State Key Laboratory of Medical Neurobiology, Institutes of Brain Science, Department of Neurology, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Guoping Liu
- State Key Laboratory of Medical Neurobiology, Institutes of Brain Science, Department of Neurology, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Teng Guo
- State Key Laboratory of Medical Neurobiology, Institutes of Brain Science, Department of Neurology, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Dashi Qi
- State Key Laboratory of Medical Neurobiology, Institutes of Brain Science, Department of Neurology, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Min Wang
- State Key Laboratory of Medical Neurobiology, Institutes of Brain Science, Department of Neurology, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Chunyang Wang
- State Key Laboratory of Medical Neurobiology, Institutes of Brain Science, Department of Neurology, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Hao Li
- State Key Laboratory of Medical Neurobiology, Institutes of Brain Science, Department of Neurology, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Yan You
- State Key Laboratory of Medical Neurobiology, Institutes of Brain Science, Department of Neurology, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Xin Wang
- State Key Laboratory of Medical Neurobiology, Institutes of Brain Science, Department of Neurology, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Bin Chen
- Department of Molecular, Cell and Developmental Biology, University of California, Santa Cruz, CA 95064, USA
| | - Hua Feng
- CAS Key Laboratory of Computational Biology, CAS-MPG Partner Institute for Computational Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - John L Rubenstein
- Department of Psychiatry, Nina Ireland Laboratory of Developmental Neurobiology, UCSF Weill Institute for Neurosciences, University of California, San Francisco, CA 94158, USA
| | - Zhengang Yang
- State Key Laboratory of Medical Neurobiology, Institutes of Brain Science, Department of Neurology, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| |
Collapse
|
18
|
Ruiz-Losada M, Blom-Dahl D, Córdoba S, Estella C. Specification and Patterning of Drosophila Appendages. J Dev Biol 2018; 6:jdb6030017. [PMID: 30011921 PMCID: PMC6162442 DOI: 10.3390/jdb6030017] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2018] [Revised: 07/10/2018] [Accepted: 07/12/2018] [Indexed: 02/06/2023] Open
Abstract
Appendages are external projections of the body that serve the animal for locomotion, feeding, or environment exploration. The appendages of the fruit fly Drosophilamelanogaster are derived from the imaginal discs, epithelial sac-like structures specified in the embryo that grow and pattern during larva development. In the last decades, genetic and developmental studies in the fruit fly have provided extensive knowledge regarding the mechanisms that direct the formation of the appendages. Importantly, many of the signaling pathways and patterning genes identified and characterized in Drosophila have similar functions during vertebrate appendage development. In this review, we will summarize the genetic and molecular mechanisms that lead to the specification of appendage primordia in the embryo and their posterior patterning during imaginal disc development. The identification of the regulatory logic underlying appendage specification in Drosophila suggests that the evolutionary origin of the insect wing is, in part, related to the development of ventral appendages.
Collapse
Affiliation(s)
- Mireya Ruiz-Losada
- Departamento de Biología Molecular and Centro de Biología Molecular Severo Ochoa, Universidad Autónoma de Madrid (UAM/CSIC), Nicolás Cabrera 1, 28049 Madrid, Spain.
| | - David Blom-Dahl
- Departamento de Biología Molecular and Centro de Biología Molecular Severo Ochoa, Universidad Autónoma de Madrid (UAM/CSIC), Nicolás Cabrera 1, 28049 Madrid, Spain.
| | - Sergio Córdoba
- Departamento de Biología Molecular and Centro de Biología Molecular Severo Ochoa, Universidad Autónoma de Madrid (UAM/CSIC), Nicolás Cabrera 1, 28049 Madrid, Spain.
| | - Carlos Estella
- Departamento de Biología Molecular and Centro de Biología Molecular Severo Ochoa, Universidad Autónoma de Madrid (UAM/CSIC), Nicolás Cabrera 1, 28049 Madrid, Spain.
| |
Collapse
|
19
|
Cooption of an appendage-patterning gene cassette in the head segmentation of arachnids. Proc Natl Acad Sci U S A 2018; 115:E3491-E3500. [PMID: 29581309 DOI: 10.1073/pnas.1720193115] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
The jointed appendages of arthropods have facilitated the spectacular diversity and success of this phylum. Key to the regulation of appendage outgrowth is the Krüppel-like factor (KLF)/specificity protein (Sp) family of zinc finger transcription factors. In the fruit fly, Drosophila melanogaster, the Sp6-9 homolog is activated by Wnt-1/wingless (wg) and establishes ventral appendage (leg) fate. Subsequently, Sp6-9 maintains expression of the axial patterning gene Distal-less (Dll), which promotes limb outgrowth. Intriguingly, in spiders, Dll has been reported to have a derived role as a segmentation gap gene, but the evolutionary origin and regulation of this function are not understood because functional investigations of the appendage-patterning regulatory network are restricted to insects. We tested the evolutionary conservation of the ancestral appendage-patterning network of arthropods with a functional approach in the spider. RNAi-mediated knockdown of the spider Sp6-9 ortholog resulted in diminution or loss of Dll expression and truncation of appendages, as well as loss of the two body segments specified by the early Dll function. In reciprocal experiments, Dll is shown not to be required for Sp6-9 expression. Knockdown of arrow (Wnt-1 coreceptor) disrupted segmentation and appendage development but did not affect the early Sp6-9 expression domain. Ectopic appendages generated in the spider "abdomen" by knockdown of the Hox gene Antennapedia-1 (Antp-1) expressed Sp6-9 comparably to wild-type walking legs. Our results support (i) the evolutionary conservation of an appendage-patterning regulatory network that includes canonical Wnt signaling, Sp6-9, and Dll and (ii) the cooption of the Sp6-9/Dll regulatory cassette in arachnid head segmentation.
Collapse
|
20
|
Boughner JC, van Eede MC, Spring S, Yu LX, Rostampour N, Henkelman RM. P63 expression plays a role in developmental rate, embryo size, and local morphogenesis. Dev Dyn 2018; 247:779-787. [DOI: 10.1002/dvdy.24622] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2017] [Revised: 01/23/2018] [Accepted: 01/30/2018] [Indexed: 12/16/2022] Open
Affiliation(s)
- Julia C. Boughner
- Department of Anatomy & Cell Biology, College of Medicine; University of Saskatchewan; Saskatoon Saskatchewan Canada
| | | | - Shoshana Spring
- Mouse Imaging Centre; Hospital for Sick Children; Toronto Ontario Canada
| | - Lisa X. Yu
- Mouse Imaging Centre; Hospital for Sick Children; Toronto Ontario Canada
| | - Nasim Rostampour
- Department of Anatomy & Cell Biology, College of Medicine; University of Saskatchewan; Saskatoon Saskatchewan Canada
| | - R. Mark Henkelman
- Mouse Imaging Centre; Hospital for Sick Children; Toronto Ontario Canada
- Department of Medical Biophysics; University of Toronto; Toronto Ontario Canada
| |
Collapse
|
21
|
Nguyen M, Singhal P, Piet JW, Shefelbine SJ, Maden M, Voss SR, Monaghan JR. Retinoic acid receptor regulation of epimorphic and homeostatic regeneration in the axolotl. Development 2017; 144:601-611. [PMID: 28087637 DOI: 10.1242/dev.139873] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2016] [Accepted: 12/30/2016] [Indexed: 12/16/2023]
Abstract
Salamanders are capable of regenerating amputated limbs by generating a mass of lineage-restricted cells called a blastema. Blastemas only generate structures distal to their origin unless treated with retinoic acid (RA), which results in proximodistal (PD) limb duplications. Little is known about the transcriptional network that regulates PD duplication. In this study, we target specific retinoic acid receptors (RARs) to either PD duplicate (RA treatment or RARγ agonist) or truncate (RARβ antagonist) regenerating limbs. RARE-EGFP reporter axolotls showed divergent reporter activity in limbs undergoing PD duplication versus truncation, suggesting differences in patterning and skeletal regeneration. Transcriptomics identified expression patterns that explain PD duplication, including upregulation of proximal homeobox gene expression and silencing of distal-associated genes, whereas limb truncation was associated with disrupted skeletal differentiation. RARβ antagonism in uninjured limbs induced a loss of skeletal integrity leading to long bone regression and loss of skeletal turnover. Overall, mechanisms were identified that regulate the multifaceted roles of RARs in the salamander limb including regulation of skeletal patterning during epimorphic regeneration, skeletal tissue differentiation during regeneration, and homeostatic regeneration of intact limbs.
Collapse
Affiliation(s)
- Matthew Nguyen
- Department of Biology, Northeastern University, Boston, MA 02115, USA
| | - Pankhuri Singhal
- Department of Biology, Northeastern University, Boston, MA 02115, USA
| | - Judith W Piet
- Mechanical and Industrial Engineering, Northeastern University, Boston, MA 02115, USA
| | - Sandra J Shefelbine
- Mechanical and Industrial Engineering, Northeastern University, Boston, MA 02115, USA
| | - Malcolm Maden
- Department of Biology and UF Genetics Institute, University of Florida, Gainesville, FL 32611, USA
| | - S Randal Voss
- Department of Biology, University of Kentucky, Lexington, KY 40506, USA
- Spinal Cord and Brain Injury Research Center, University of Kentucky, College of Medicine, Lexington, KY 40506, USA
| | - James R Monaghan
- Department of Biology, Northeastern University, Boston, MA 02115, USA
| |
Collapse
|
22
|
Masuda T, Ishitani T. Context-dependent regulation of the β-catenin transcriptional complex supports diverse functions of Wnt/β-catenin signaling. J Biochem 2016; 161:9-17. [PMID: 28013224 DOI: 10.1093/jb/mvw072] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2016] [Accepted: 09/14/2016] [Indexed: 12/15/2022] Open
Abstract
Wnt/β-catenin signaling is activated repeatedly during an animal's lifespan, and it controls gene expression through its essential nuclear effector, β-catenin, to regulate embryogenesis, organogenesis, and adult homeostasis. Although the β-catenin transcriptional complex has the ability to induce the expression of many genes to exert its diverse roles, it chooses and transactivates a specific gene set from among its numerous target genes depending on the context. For example, the β-catenin transcriptional complex stimulates the expression of cell cycle-related genes and consequent cell proliferation in neural progenitor cells, while it promotes the expression of neural differentiation-related genes in differentiating neurons. Recent studies using animal and cell culture models have gradually improved our understanding of the molecular basis underlying such context-dependent actions of the β-catenin transcriptional complex. Here, we describe eight mechanisms that support β-catenin-mediated context-dependent gene regulation, and their spatio-temporal regulation during vertebrate development. In addition, we discuss their contribution to the diverse functions of Wnt/β-catenin signaling.
Collapse
Affiliation(s)
- Takamasa Masuda
- Division of Cell Regulation Systems, Medical Institute of Bioregulation, Kyushu University, 3-1-1 Maidashi, Higashi-Ku, Fukuoka 812-8582, Japan
| | - Tohru Ishitani
- Division of Cell Regulation Systems, Medical Institute of Bioregulation, Kyushu University, 3-1-1 Maidashi, Higashi-Ku, Fukuoka 812-8582, Japan
| |
Collapse
|
23
|
Tbx5 Buffers Inherent Left/Right Asymmetry Ensuring Symmetric Forelimb Formation. PLoS Genet 2016; 12:e1006521. [PMID: 27992425 PMCID: PMC5215935 DOI: 10.1371/journal.pgen.1006521] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2016] [Revised: 01/05/2017] [Accepted: 12/02/2016] [Indexed: 11/19/2022] Open
Abstract
The forelimbs and hindlimbs of vertebrates are bilaterally symmetric. The mechanisms that ensure symmetric limb formation are unknown but they can be disrupted in disease. In Holt-Oram Syndrome (HOS), caused by mutations in TBX5, affected individuals have left-biased upper/forelimb defects. We demonstrate a role for the transcription factor Tbx5 in ensuring the symmetric formation of the left and right forelimb. In our mouse model, bilateral hypomorphic levels of Tbx5 produces asymmetric forelimb defects that are consistently more severe in the left limb than the right, phenocopying the left-biased limb defects seen in HOS patients. In Tbx hypomorphic mutants maintained on an INV mutant background, with situs inversus, the laterality of defects is reversed. Our data demonstrate an early, inherent asymmetry in the left and right limb-forming regions and that threshold levels of Tbx5 are required to overcome this asymmetry to ensure symmetric forelimb formation. Externally, the human form appears bilaterally symmetric. For example, each of our pairs of arms and legs are the same length. This external symmetry masks many asymmetries found in internal organs. In most people the heart is found on the left side of the chest. The stomach, liver and spleen are also positioned asymmetrically. The authors of this study demonstrate, using a mouse model, that bilateral symmetry of the arms is not a default, passive state but that mechanisms are in place that ensure symmetrical formation of the left and right limbs. Bilateral symmetry of the arms is achieved by the action of a gene Tbx5 that masks the effects of signals that acted earlier during embryogenesis, many days before limb formation, and imposed asymmetries on the forming internal organs. Maintaining bilateral symmetry of the arms is important for them to carry out their normal functions but this process can go wrong. Holt-Oram syndrome patients have upper limb defects, including shortened arms. Consistently the defects are more severe in their left arm than right. This birth defect is caused by disruption of the TBX5 gene. By linking the action of Tbx5 to symmetrical limb formation, the authors provide an explanation for why Holt-Oram syndrome patients have more severe defects in the left arms than right.
Collapse
|
24
|
Zheng X, Xiang L, Liang J, Xie L, Zhang R. Pf-Sp8/9, a novel member of the specificity protein family in Pinctada fucata, potentially participates in biomineralization. J Struct Biol 2016; 196:119-126. [DOI: 10.1016/j.jsb.2016.05.002] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2015] [Revised: 05/09/2016] [Accepted: 05/10/2016] [Indexed: 02/07/2023]
|
25
|
Li Z, Zhang T, Lin Z, Hou C, Zhang J, Men Y, Li H, Gao J. Lgl1 Is Required for Olfaction and Development of Olfactory Bulb in Mice. PLoS One 2016; 11:e0162126. [PMID: 27603780 PMCID: PMC5014313 DOI: 10.1371/journal.pone.0162126] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2016] [Accepted: 08/17/2016] [Indexed: 11/27/2022] Open
Abstract
Lethal giant larvae 1 (Lgl1) was initially identified as a tumor suppressor in Drosophila and functioned as a key regulator of epithelial polarity and asymmetric cell division. In this study, we generated Lgl1 conditional knockout mice mediated by Pax2-Cre, which is expressed in olfactory bulb (OB). Next, we examined the effects of Lgl1 loss in the OB. First, we determined the expression patterns of Lgl1 in the neurogenic regions of the embryonic dorsal region of the LGE (dLGE) and postnatal OB. Furthermore, the Lgl1 conditional mutants exhibited abnormal morphological characteristics of the OB. Our behavioral analysis exhibited greatly impaired olfaction in Lgl1 mutant mice. To elucidate the possible mechanisms of impaired olfaction in Lgl1 mutant mice, we investigated the development of the OB. Interestingly, reduced thickness of the MCL and decreased density of mitral cells (MCs) were observed in Lgl1 mutant mice. Additionally, we observed a dramatic loss in SP8+ interneurons (e.g. calretinin and GABAergic/non-dopaminergic interneurons) in the GL of the OB. Our results demonstrate that Lgl1 is required for the development of the OB and the deletion of Lgl1 results in impaired olfaction in mice.
Collapse
Affiliation(s)
- Zhenzu Li
- Institute of Developmental Biology, School of Life Science, Shandong University, Jinan, 250100, Shandong, China
| | - Tingting Zhang
- Institute of Developmental Biology, School of Life Science, Shandong University, Jinan, 250100, Shandong, China
| | - Zhuchun Lin
- Jinan First People's Hospital, Jinan, 250011, Shandong, China
| | - Congzhe Hou
- The Second Hospital of Shandong University, Jinan, 250000, Shandong, China
| | - Jian Zhang
- Institute of Developmental Biology, School of Life Science, Shandong University, Jinan, 250100, Shandong, China
| | - Yuqin Men
- Institute of Developmental Biology, School of Life Science, Shandong University, Jinan, 250100, Shandong, China
| | - Huashun Li
- SARITEX Center for Stem Cell, Engineering Translational Medicine, Shanghai East Hospital, Advanced Institute of Translational Medicine, Tongji University School of Medicine, Shanghai, 200123, China.,Center for Stem Cell&Nano-Medicine, Shanghai Advanced Research Institute, Chinese Academy of Sciences, Shanghai, 200123, China.,Shenzhen Key Laboratory for Molecular Biology of Neural Development, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518000, Guangdong, China
| | - Jiangang Gao
- Institute of Developmental Biology, School of Life Science, Shandong University, Jinan, 250100, Shandong, China
| |
Collapse
|
26
|
Time-Course Gene Expression Profiling Reveals a Novel Role of Non-Canonical WNT Signaling During Neural Induction. Sci Rep 2016; 6:32600. [PMID: 27600186 PMCID: PMC5013468 DOI: 10.1038/srep32600] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2016] [Accepted: 08/10/2016] [Indexed: 12/22/2022] Open
Abstract
The process of neuroepithelial differentiation from human pluripotent stem cells (PSCs) resembles in vivo neuroectoderm induction in the temporal course, morphogenesis, and biochemical changes. This in vitro model is therefore well-suited to reveal previously unknown molecular mechanisms underlying neural induction in humans. By transcriptome analysis of cells along PSC differentiation to early neuroepithelia at day 6 and definitive neuroepithelia at day 10, we found downregulation of genes that are associated with TGF-β and canonical WNT/β-CATENIN signaling, confirming the roles of classical signaling in human neural induction. Interestingly, WNT/Ca2+ signaling was upregulated. Pharmacological inhibition of the downstream effector of WNT/Ca2+ pathway, Ca2+/calmodulin-dependent protein kinase II (CaMKII), led to an inhibition of the neural marker PAX6 and upregulation of epidermal marker K18, suggesting that Ca2+/CaMKII signaling promotes neural induction by preventing the alternative epidermal fate. In addition, our analyses revealed known and novel expression patterns of genes that are involved in DNA methylation, histone modification, as well as epithelial-mesenchymal transition, highlighting potential roles of those genes and signaling pathways during neural differentiation.
Collapse
|
27
|
Córdoba S, Requena D, Jory A, Saiz A, Estella C. The evolutionary conserved transcription factor Sp1 controls appendage growth through Notch signaling. Development 2016; 143:3623-3631. [DOI: 10.1242/dev.138735] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2016] [Accepted: 08/12/2016] [Indexed: 12/25/2022]
Abstract
The appendages of arthropods and vertebrates are not homologous structures, although the underlying genetic mechanisms that pattern them are highly conserved. Members of the Sp family of transcription factors are expressed in the developing limbs and their function is required for limb growth in both insects and chordates. Despite the fundamental and conserved role that these transcription factors play during appendage development, their target genes and the mechanisms in which they participate to control limb growth are mostly unknown. We analyzed here the individual contributions of two Drosophila Sp members, buttonhead (btd) and Sp1, during leg development. We show that Sp1 plays a more prominent role controlling leg growth than btd. We identified a regulatory function of Sp1 in Notch signaling, and performed a genome wide transcriptome analysis to identify other potential Sp1 target genes contributing to leg growth. Our data suggest a mechanism by which the Sp factors control appendage growth through the Notch signaling.
Collapse
Affiliation(s)
- Sergio Córdoba
- Departamento de Biología Molecular and Centro de Biología Molecular Severo Ochoa, Universidad Autónoma de Madrid (UAM), Nicolás Cabrera 1, 28049 Madrid, Spain
| | - David Requena
- Departamento de Biología Molecular and Centro de Biología Molecular Severo Ochoa, Universidad Autónoma de Madrid (UAM), Nicolás Cabrera 1, 28049 Madrid, Spain
| | - Aurelie Jory
- Department of Biochemistry and Molecular Biophysics, Columbia University, New York, New York, United States of America
| | - Almudena Saiz
- Departamento de Biología Molecular and Centro de Biología Molecular Severo Ochoa, Universidad Autónoma de Madrid (UAM), Nicolás Cabrera 1, 28049 Madrid, Spain
| | - Carlos Estella
- Departamento de Biología Molecular and Centro de Biología Molecular Severo Ochoa, Universidad Autónoma de Madrid (UAM), Nicolás Cabrera 1, 28049 Madrid, Spain
| |
Collapse
|
28
|
Zhang XM, Cai Y, Wang F, Wu J, Mo L, Zhang F, Patrylo PR, Pan A, Ma C, Fu J, Yan XX. Sp8 expression in putative neural progenitor cells in guinea pig and human cerebrum. Dev Neurobiol 2015; 76:939-55. [PMID: 26585436 DOI: 10.1002/dneu.22367] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2015] [Revised: 10/05/2015] [Accepted: 11/12/2015] [Indexed: 12/19/2022]
Abstract
Neural stem/progenitor cells have been characterized at neurogenic sites in adult mammalian brain with various molecular markers. Here it has been demonstrated that Sp8, a transcription factor typically expressed among mature GABAergic interneurons, also labels putative neural precursors in adult guinea pig and human cerebrum. In guinea pigs, Sp8 immunoreactive (Sp8+) cells were localized largely in the superficial layers of the cortex including layer I, as well as the subventricular zone (SVZ) and subgranular zone (SGZ). Sp8+ cells at the SGZ showed little colocalization with mature and immature neuronal markers, but co-expressed neural stem cell markers including Sox2. Some layer I Sp8+ cells also co-expressed Sox2. The amount of Sp8+ cells in the dentate gyrus was maintained 2 weeks after X-ray irradiation, while that of doublecortin (DCX+) cells was greatly reduced. Mild ischemic insult caused a transient increase of Sp8+ cells in the SGZ and layer I, with the subgranular Sp8+ cells exhibited an increased colabeling for the mitotic marker Ki67 and pulse-chased bromodeoxyuridine (BrdU). Sp8+ cells in the dentate gyrus showed an age-related decline in guinea pigs, in parallel with the loss of DCX+ cells in the same region. In adult humans, Sp8+ cells exhibited comparable morphological features as seen in guinea pigs, with those at the SGZ and some in cortical layer I co-expressed Sox2. Together, these results suggested that Sp8 may label putative neural progenitors in guinea pig and human cerebrum, with the labeled cells in the SGZ appeared largely not mitotically active under normal conditions. © 2015 Wiley Periodicals, Inc. Develop Neurobiol 76: 939-955, 2016.
Collapse
Affiliation(s)
- Xue-Mei Zhang
- Department of Neurology, The Second Affiliated Hospital, Harbin Medical University, Harbin, Heilongjiang, China
| | - Yan Cai
- Department of Anatomy and Neurobiology, Central South University School of Basic Medicine, Changsha, Hunan, China
| | - Fang Wang
- Department of Neurology, The Second Affiliated Hospital, Harbin Medical University, Harbin, Heilongjiang, China
| | - Jun Wu
- Department of Neurology, The Second Affiliated Hospital, Harbin Medical University, Harbin, Heilongjiang, China
| | - Lin Mo
- Department of Neurology, The Second Affiliated Hospital, Harbin Medical University, Harbin, Heilongjiang, China
| | - Feng Zhang
- Department of Radiation Oncology, The Second Affiliated Hospital, Harbin Medical University, Harbin, Heilongjiang, China
| | - Peter R Patrylo
- Southern Illinois University School of Medicine, Center for Integrated Research in Cognitive and Neural Sciences, Carbondale, Illinois
| | - Aihua Pan
- Department of Anatomy and Neurobiology, Central South University School of Basic Medicine, Changsha, Hunan, China
| | - Chao Ma
- Department of Human Anatomy, Histology & Embryology, Institute of Basic Medical Sciences, Neuroscience Center, Chinese Academy of Medical Sciences, School of Basic Medicine, Peking Union Medical College, Beijing, China
| | - Jin Fu
- Department of Neurology, The Second Affiliated Hospital, Harbin Medical University, Harbin, Heilongjiang, China
| | - Xiao-Xin Yan
- Department of Anatomy and Neurobiology, Central South University School of Basic Medicine, Changsha, Hunan, China.,Key Laboratory of Hunan Province in Neurodegenerative Disorders, Changsha, Hunan, China
| |
Collapse
|
29
|
Identifying Regulators of Morphogenesis Common to Vertebrate Neural Tube Closure and Caenorhabditis elegans Gastrulation. Genetics 2015; 202:123-39. [PMID: 26434722 DOI: 10.1534/genetics.115.183137] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2015] [Accepted: 09/29/2015] [Indexed: 12/18/2022] Open
Abstract
Neural tube defects including spina bifida are common and severe congenital disorders. In mice, mutations in more than 200 genes can result in neural tube defects. We hypothesized that this large gene set might include genes whose homologs contribute to morphogenesis in diverse animals. To test this hypothesis, we screened a set of Caenorhabditis elegans homologs for roles in gastrulation, a topologically similar process to vertebrate neural tube closure. Both C. elegans gastrulation and vertebrate neural tube closure involve the internalization of surface cells, requiring tissue-specific gene regulation, actomyosin-driven apical constriction, and establishment and maintenance of adhesions between specific cells. Our screen identified several neural tube defect gene homologs that are required for gastrulation in C. elegans, including the transcription factor sptf-3. Disruption of sptf-3 in C. elegans reduced the expression of early endodermally expressed genes as well as genes expressed in other early cell lineages, establishing sptf-3 as a key contributor to multiple well-studied C. elegans cell fate specification pathways. We also identified members of the actin regulatory WAVE complex (wve-1, gex-2, gex-3, abi-1, and nuo-3a). Disruption of WAVE complex members reduced the narrowing of endodermal cells' apical surfaces. Although WAVE complex members are expressed broadly in C. elegans, we found that expression of a vertebrate WAVE complex member, nckap1, is enriched in the developing neural tube of Xenopus. We show that nckap1 contributes to neural tube closure in Xenopus. This work identifies in vivo roles for homologs of mammalian neural tube defect genes in two manipulable genetic model systems.
Collapse
|
30
|
Le Duc D, Renaud G, Krishnan A, Almén MS, Huynen L, Prohaska SJ, Ongyerth M, Bitarello BD, Schiöth HB, Hofreiter M, Stadler PF, Prüfer K, Lambert D, Kelso J, Schöneberg T. Kiwi genome provides insights into evolution of a nocturnal lifestyle. Genome Biol 2015; 16:147. [PMID: 26201466 PMCID: PMC4511969 DOI: 10.1186/s13059-015-0711-4] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2015] [Accepted: 07/01/2015] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Kiwi, comprising five species from the genus Apteryx, are endangered, ground-dwelling bird species endemic to New Zealand. They are the smallest and only nocturnal representatives of the ratites. The timing of kiwi adaptation to a nocturnal niche and the genomic innovations, which shaped sensory systems and morphology to allow this adaptation, are not yet fully understood. RESULTS We sequenced and assembled the brown kiwi genome to 150-fold coverage and annotated the genome using kiwi transcript data and non-redundant protein information from multiple bird species. We identified evolutionary sequence changes that underlie adaptation to nocturnality and estimated the onset time of these adaptations. Several opsin genes involved in color vision are inactivated in the kiwi. We date this inactivation to the Oligocene epoch, likely after the arrival of the ancestor of modern kiwi in New Zealand. Genome comparisons between kiwi and representatives of ratites, Galloanserae, and Neoaves, including nocturnal and song birds, show diversification of kiwi's odorant receptors repertoire, which may reflect an increased reliance on olfaction rather than sight during foraging. Further, there is an enrichment of genes influencing mitochondrial function and energy expenditure among genes that are rapidly evolving specifically on the kiwi branch, which may also be linked to its nocturnal lifestyle. CONCLUSIONS The genomic changes in kiwi vision and olfaction are consistent with changes that are hypothesized to occur during adaptation to nocturnal lifestyle in mammals. The kiwi genome provides a valuable genomic resource for future genome-wide comparative analyses to other extinct and extant diurnal ratites.
Collapse
Affiliation(s)
- Diana Le Duc
- Institute of Biochemistry, Medical Faculty, University of Leipzig, Johannisallee 30, Leipzig, 04103, Germany.
- Department of Evolutionary Genetics, Max Planck Institute for Evolutionary Anthropology, Leipzig, 04103, Germany.
| | - Gabriel Renaud
- Department of Evolutionary Genetics, Max Planck Institute for Evolutionary Anthropology, Leipzig, 04103, Germany.
| | - Arunkumar Krishnan
- Department of Neuroscience, Unit of Functional Pharmacology, Uppsala University, Box 593, Husargatan 3, Uppsala, 751 24, Sweden.
| | - Markus Sällman Almén
- Department of Neuroscience, Unit of Functional Pharmacology, Uppsala University, Box 593, Husargatan 3, Uppsala, 751 24, Sweden.
| | - Leon Huynen
- Griffith School of Environment and School of Biomolecular and Physical Sciences, Griffith University, Nathan, Queensland, 4111, Australia.
| | - Sonja J Prohaska
- Department of Computer Science, and Interdisciplinary Center for Bioinformatics, University of Leipzig, Leipzig, 04103, Germany.
| | - Matthias Ongyerth
- Department of Evolutionary Genetics, Max Planck Institute for Evolutionary Anthropology, Leipzig, 04103, Germany.
| | - Bárbara D Bitarello
- Department of Genetics and Evolutionary Biology, University of São Paulo, São Paulo, SP, 05508-090, Brazil.
| | - Helgi B Schiöth
- Department of Neuroscience, Unit of Functional Pharmacology, Uppsala University, Box 593, Husargatan 3, Uppsala, 751 24, Sweden.
| | - Michael Hofreiter
- Adaptive Evolutionary Genomics, Institute for Biochemistry and Biology, University Potsdam, Potsdam, 14469, Germany.
| | - Peter F Stadler
- Department of Computer Science, and Interdisciplinary Center for Bioinformatics, University of Leipzig, Leipzig, 04103, Germany.
| | - Kay Prüfer
- Department of Evolutionary Genetics, Max Planck Institute for Evolutionary Anthropology, Leipzig, 04103, Germany.
| | - David Lambert
- Griffith School of Environment and School of Biomolecular and Physical Sciences, Griffith University, Nathan, Queensland, 4111, Australia.
| | - Janet Kelso
- Department of Evolutionary Genetics, Max Planck Institute for Evolutionary Anthropology, Leipzig, 04103, Germany.
| | - Torsten Schöneberg
- Institute of Biochemistry, Medical Faculty, University of Leipzig, Johannisallee 30, Leipzig, 04103, Germany.
| |
Collapse
|
31
|
Raines AM, Magella B, Adam M, Potter SS. Key pathways regulated by HoxA9,10,11/HoxD9,10,11 during limb development. BMC DEVELOPMENTAL BIOLOGY 2015; 15:28. [PMID: 26186931 PMCID: PMC4506574 DOI: 10.1186/s12861-015-0078-5] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/27/2015] [Accepted: 07/07/2015] [Indexed: 11/17/2022]
Abstract
Background The 39 mammalian Hox genes show problematic patterns of functional overlap. In order to more fully define the developmental roles of Hox genes it is necessary to remove multiple combinations of paralogous and flanking genes. In addition, the downstream molecular pathways regulated by Hox genes during limb development remain incompletely delineated. Results In this report we examine limb development in mice with frameshift mutations in six Hox genes, Hoxa9,10,11 and Hoxd9,10,11. The mice were made with a novel recombineering method that allows the simultaneous targeting of frameshift mutations into multiple flanking genes. The Hoxa9,10,11−/−/Hoxd9,10,11−/− mutant mice show a reduced ulna and radius that is more severe than seen in Hoxa11−/−/Hoxd11−/− mice, indicating a minor role for the flanking Hox9,10 genes in zeugopod development, as well as their primary function in stylopod development. The mutant mice also show severe reduction of Shh expression in the zone of polarizing activity, and decreased Fgf8 expression in the apical ectodermal ridge, thereby better defining the roles of these specific Hox genes in the regulation of critical signaling centers during limb development. Importantly, we also used laser capture microdissection coupled with RNA-Seq to characterize the gene expression programs in wild type and mutant limbs. Resting, proliferative and hypertrophic compartments of E15.5 forelimb zeugopods were examined. The results provide an RNA-Seq characterization of the progression of gene expression patterns during normal endochondral bone formation. In addition of the Hox mutants showed strongly altered expression of Pknox2, Zfp467, Gdf5, Bmpr1b, Dkk3, Igf1, Hand2, Shox2, Runx3, Bmp7 and Lef1, all of which have been previously shown to play important roles in bone formation. Conclusions The recombineering based frameshift mutation of the six flanking and paralogous Hoxa9,10,11 and Hoxd9,10,11 genes provides a resource for the analysis of their overlapping functions. Analysis of the Hoxa9,10,11−/−/Hoxd9,10,11−/− mutant limbs confirms and extends the results of previous studies using mice with Hox mutations in single paralogous groups or with entire Hox cluster deletions. The RNA-Seq analysis of specific compartments of the normal and mutant limbs defines the multiple key perturbed pathways downstream of these Hox genes. Electronic supplementary material The online version of this article (doi:10.1186/s12861-015-0078-5) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Anna M Raines
- Division of Developmental Biology, Cincinnati Children's Medical Center, 3333 Burnet Ave., Cincinnati, OH, 45229, USA.
| | - Bliss Magella
- Division of Developmental Biology, Cincinnati Children's Medical Center, 3333 Burnet Ave., Cincinnati, OH, 45229, USA.
| | - Mike Adam
- Division of Developmental Biology, Cincinnati Children's Medical Center, 3333 Burnet Ave., Cincinnati, OH, 45229, USA.
| | - S Steven Potter
- Division of Developmental Biology, Cincinnati Children's Medical Center, 3333 Burnet Ave., Cincinnati, OH, 45229, USA.
| |
Collapse
|
32
|
VanderMeer JE, Smith RP, Jones SL, Ahituv N. Genome-wide identification of signaling center enhancers in the developing limb. Development 2014; 141:4194-8. [PMID: 25273087 PMCID: PMC4302890 DOI: 10.1242/dev.110965] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
The limb is widely used as a model developmental system and changes to gene expression patterns in its signaling centers, notably the zone of polarizing activity (ZPA) and the apical ectodermal ridge (AER), are known to cause limb malformations and evolutionary differences in limb morphology. Although several genes that define these limb signaling centers have been described, the identification of regulatory elements that are active within these centers has been limited. By dissecting mouse E11.5 limbs that fluorescently mark the ZPA or AER, followed by fluorescence-activated cell sorting and low-cell H3K27ac ChIP-seq, we identified thousands of specific signaling-center enhancers. Our ChIP-seq datasets show strong correlation with ZPA- and AER-expressed genes, previously characterized functional ZPA and AER enhancers and enrichment for relevant biological terms related to limb development and malformation for the neighboring genes. Using transgenic assays, we show that several of these sequences function as ZPA and AER enhancers. Our results identify novel ZPA and AER enhancers that could be important regulators of genes involved in the establishment of these specialized regions and the patterning of tetrapod limbs.
Collapse
Affiliation(s)
- Julia E VanderMeer
- Department of Bioengineering and Therapeutic Sciences, University of California San Francisco, San Francisco, CA 94158, USA Institute for Human Genetics, University of California San Francisco, San Francisco, CA 94158, USA
| | - Robin P Smith
- Department of Bioengineering and Therapeutic Sciences, University of California San Francisco, San Francisco, CA 94158, USA Institute for Human Genetics, University of California San Francisco, San Francisco, CA 94158, USA
| | - Stacy L Jones
- Department of Bioengineering and Therapeutic Sciences, University of California San Francisco, San Francisco, CA 94158, USA
| | - Nadav Ahituv
- Department of Bioengineering and Therapeutic Sciences, University of California San Francisco, San Francisco, CA 94158, USA Institute for Human Genetics, University of California San Francisco, San Francisco, CA 94158, USA
| |
Collapse
|
33
|
Sp6 and Sp8 transcription factors control AER formation and dorsal-ventral patterning in limb development. PLoS Genet 2014; 10:e1004468. [PMID: 25166858 PMCID: PMC4148220 DOI: 10.1371/journal.pgen.1004468] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2013] [Accepted: 05/14/2014] [Indexed: 12/27/2022] Open
Abstract
The formation and maintenance of the apical ectodermal ridge (AER) is critical for the outgrowth and patterning of the vertebrate limb. The induction of the AER is a complex process that relies on integrated interactions among the Fgf, Wnt, and Bmp signaling pathways that operate within the ectoderm and between the ectoderm and the mesoderm of the early limb bud. The transcription factors Sp6 and Sp8 are expressed in the limb ectoderm and AER during limb development. Sp6 mutant mice display a mild syndactyly phenotype while Sp8 mutants exhibit severe limb truncations. Both mutants show defects in AER maturation and in dorsal-ventral patterning. To gain further insights into the role Sp6 and Sp8 play in limb development, we have produced mice lacking both Sp6 and Sp8 activity in the limb ectoderm. Remarkably, the elimination or significant reduction in Sp6;Sp8 gene dosage leads to tetra-amelia; initial budding occurs, but neither Fgf8 nor En1 are activated. Mutants bearing a single functional allele of Sp8 (Sp6−/−;Sp8+/−) exhibit a split-hand/foot malformation phenotype with double dorsal digit tips probably due to an irregular and immature AER that is not maintained in the center of the bud and on the abnormal expansion of Wnt7a expression to the ventral ectoderm. Our data are compatible with Sp6 and Sp8 working together and in a dose-dependent manner as indispensable mediators of Wnt/βcatenin and Bmp signaling in the limb ectoderm. We suggest that the function of these factors links proximal-distal and dorsal-ventral patterning. In this report we examined the functional roles of Sp6 and Sp8 during limb development using compound loss-of-function mutants. Sp6 and Sp8, two members of the Sp gene family, are expressed in the limb bud ectoderm and function downstream of WNT/βcatenin signaling for Fgf8 induction. The analysis of the allelic series shows that the progressive reduction in the dose of Sp6 and Sp8 gene products leads to predictable morphology, from syndactyly, to split hand/foot malformation, oligodactyly, truncation and finally amelia, indicating that these two factors act in a complementary manner. The molecular characterization of the mutant limbs reveal that Sp6/Sp8 are required in a dose-dependent manner for Fgf8 and En1 induction, thereby placing them as an important link between the induction of the AER and the establishment of dorsal-ventral patterning during limb development.
Collapse
|
34
|
Li X, Liu Z, Qiu M, Yang Z. Sp8 plays a supplementary role to Pax6 in establishing the pMN/p3 domain boundary in the spinal cord. Development 2014; 141:2875-84. [DOI: 10.1242/dev.105387] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Progenitor cells are segregated into multiple domains along the dorsoventral axis of the vertebrate neural tube, and each progenitor domain generates particular types of neurons. Selective cross-repressive interactions between pairs of class I and class II transcription factors play important roles in patterning neural progenitors into domains with clear boundaries. Here, we provide evidence that the zinc-finger protein Sp8 plays a supplementary role to Pax6 in establishing the pMN/p3 domain boundary through mutually repressive interactions with the class II protein Nkx2-2. The ventral limit of Sp8 expression is complementary to the dorsal limit of Nkx2-2 expression at the pMN/p3 boundary. Sp8 and Nkx2-2 exert cross-repressive interactions, and changing the expression of Sp8 and Nkx2-2 is coupled with pMN and p3 progenitor fate conversion. Sp8 exerts its neural patterning activities by acting as a transcriptional activator. The expression of a repressive form of Sp8 results in the selective inhibition of motor neuron generation and the ectopic induction of Nkx2-2 expression. Sp8 expression is positively regulated by, but not completely dependent on, Pax6. Furthermore, whereas loss of Pax6 function alone results in disruption of the pMN/p3 domain boundary only in the rostral levels of the spinal cord, loss of both Sp8 and Pax6 functions results in disruption of the pMN/p3 domain boundary along the whole rostrocaudal axis of the spinal cord. We conclude that Sp8 plays a supplementary role to Pax6 in specifying the pMN over p3 progenitor fate through cross-repressive interactions with Nkx2-2.
Collapse
Affiliation(s)
- Xiaosu Li
- Institutes of Brain Science and State Key Laboratory of Medical Neurobiology, Fudan University, Shanghai 200032, China
| | - Zhidong Liu
- Institutes of Brain Science and State Key Laboratory of Medical Neurobiology, Fudan University, Shanghai 200032, China
| | - Mengsheng Qiu
- Institute of Developmental and Regenerative Biology, Zhejiang Key Laboratory of Organ Development and Regeneration, Hangzhou Normal University, Hangzhou 310036, China
- Department of Anatomical Sciences and Neurobiology, School of Medicine, University of Louisville, Louisville, KY40392, USA
| | - Zhengang Yang
- Institutes of Brain Science and State Key Laboratory of Medical Neurobiology, Fudan University, Shanghai 200032, China
| |
Collapse
|
35
|
Safe S, Imanirad P, Sreevalsan S, Nair V, Jutooru I. Transcription factor Sp1, also known as specificity protein 1 as a therapeutic target. Expert Opin Ther Targets 2014; 18:759-69. [PMID: 24793594 DOI: 10.1517/14728222.2014.914173] [Citation(s) in RCA: 78] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
INTRODUCTION Specificity protein (Sp) transcription factors (TFs) are members of the Sp/Kruppel-like factor family, and Sp proteins play an important role in embryonic and early postnatal development. Sp1 has been the most extensively investigated member of this family, and expression of this protein decreases with age, whereas Sp1 and other family members (Sp3 and Sp4) are highly expressed in tumors and cancer cell lines. AREA COVERED The prognostic significance of Sp1 in cancer patients and the functional pro-oncogenic activities of Sp1, Sp3 and Sp4 in cancer cell lines are summarized. Several different approaches have been used to target downregulation of Sp TFs and Sp-regulated genes, and this includes identification of different structural classes of antineoplastic agents including NSAIDs, natural products and their synthetic analogs and several well-characterized drugs including arsenic trioxide, aspirin and metformin. The multiple pathways involved in drug-induced Sp downregulation are also discussed. EXPERT OPINION The recognition by the scientific and clinical community that experimental and clinically used antineoplastic agents downregulate Sp1, Sp3 and Sp4, and pro-oncogenic Sp-regulated genes will facilitate future clinical applications for individual drug and drug combination therapies that take advantage of their unusual effects.
Collapse
Affiliation(s)
- Stephen Safe
- Texas A&M University, Veterinary Physiology and Pharmacology , 4466 TAMU, College Station, TX 77843-4466 , USA
| | | | | | | | | |
Collapse
|
36
|
Abstract
A forward genetic screen of N-ethyl-N-nitrosourea mutagenized Xenopus tropicalis has identified an inner ear mutant named eclipse (ecl). Mutants developed enlarged otic vesicles and various defects of otoconia development; they also showed abnormal circular and inverted swimming patterns. Positional cloning identified specificity protein 8 (sp8), which was previously found to regulate limb and brain development. Two different loss-of-function approaches using transcription activator-like effector nucleases and morpholino oligonucleotides confirmed that the ecl mutant phenotype is caused by down-regulation of sp8. Depletion of sp8 resulted in otic dysmorphogenesis, such as uncompartmentalized and enlarged otic vesicles, epithelial dilation with abnormal sensory end organs. When overexpressed, sp8 was sufficient to induce ectopic otic vesicles possessing sensory hair cells, neurofilament innervation in a thickened sensory epithelium, and otoconia, all of which are found in the endogenous otic vesicle. We propose that sp8 is an important factor for initiation and elaboration of inner ear development.
Collapse
|
37
|
Dunty WC, Kennedy MWL, Chalamalasetty RB, Campbell K, Yamaguchi TP. Transcriptional profiling of Wnt3a mutants identifies Sp transcription factors as essential effectors of the Wnt/β-catenin pathway in neuromesodermal stem cells. PLoS One 2014; 9:e87018. [PMID: 24475213 PMCID: PMC3901714 DOI: 10.1371/journal.pone.0087018] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2013] [Accepted: 12/17/2013] [Indexed: 01/08/2023] Open
Abstract
Neuromesodermal (NM) stem cells reside in the primitive streak (PS) of gastrulating vertebrate embryos and generate precursors of the spinal cord and musculoskeletal system. Although Wnt3a/β-catenin signaling is crucial for NM stem cell maintenance and differentiation, few key transcriptional effectors have been identified. Through a concerted transcriptional profiling and genetic approach we have determined that two Zn2+-finger transcription factors, Sp5 and Sp8, are regulated by Wnt3a in the PS, and are essential for neural and musculoskeletal patterning. These results identify Sp5 and Sp8 as pivotal downstream effectors of Wnt3a, and suggest that they are essential for the self-renewal and differentiation of NM stem cells.
Collapse
Affiliation(s)
- William C. Dunty
- Cancer and Developmental Biology Laboratory, Center for Cancer Research, NCI-Frederick, NIH, Frederick, Maryland, United States of America
| | - Mark W. L. Kennedy
- Cancer and Developmental Biology Laboratory, Center for Cancer Research, NCI-Frederick, NIH, Frederick, Maryland, United States of America
| | - Ravindra B. Chalamalasetty
- Cancer and Developmental Biology Laboratory, Center for Cancer Research, NCI-Frederick, NIH, Frederick, Maryland, United States of America
| | - Kenneth Campbell
- Division of Developmental Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio, United States of America
| | - Terry P. Yamaguchi
- Cancer and Developmental Biology Laboratory, Center for Cancer Research, NCI-Frederick, NIH, Frederick, Maryland, United States of America
- * E-mail:
| |
Collapse
|
38
|
Lee HJ, Ryu JS, Choi NY, Park YS, Kim YI, Han DW, Ko K, Shin CY, Hwang HS, Kang KS, Ko K. Transgenerational effects of paternal alcohol exposure in mouse offspring. Anim Cells Syst (Seoul) 2013. [DOI: 10.1080/19768354.2013.865675] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022] Open
|
39
|
Kasberg AD, Brunskill EW, Steven Potter S. SP8 regulates signaling centers during craniofacial development. Dev Biol 2013; 381:312-23. [PMID: 23872235 DOI: 10.1016/j.ydbio.2013.07.007] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2013] [Revised: 07/10/2013] [Accepted: 07/11/2013] [Indexed: 01/23/2023]
Abstract
Much of the bone, cartilage and smooth muscle of the vertebrate face is derived from neural crest (NC) cells. During craniofacial development, the anterior neural ridge (ANR) and olfactory pit (OP) signaling centers are responsible for driving the outgrowth, survival, and differentiation of NC populated facial prominences, primarily via FGF. While much is known about the functional importance of signaling centers, relatively little is understood of how these signaling centers are made and maintained. In this report we describe a dramatic craniofacial malformation in mice mutant for the zinc finger transcription factor gene Sp8. At E14.5 they show facial prominences that are reduced in size and underdeveloped, giving an almost faceless phenotype. At later times they show severe midline defects, excencephaly, hyperterlorism, cleft palate, and a striking loss of many NC and paraxial mesoderm derived cranial bones. Sp8 expression was primarily restricted to the ANR and OP regions during craniofacial development. Analysis of an extensive series of conditional Sp8 mutants confirmed the critical role of Sp8 in signaling centers, and not directly in the NC and paraxial mesoderm cells. The NC cells of the Sp8 mutants showed increased levels of apoptosis and decreased cell proliferation, thereby explaining the reduced sizes of the facial prominences. Perturbed gene expression in the Sp8 mutants was examined by laser capture microdissection coupled with microarrays, as well as in situ hybridization and immunostaining. The most dramatic differences included striking reductions in Fgf8 and Fgf17 expression in the ANR and OP signaling centers. We were also able to achieve genetic and pharmaceutical partial rescue of the Sp8 mutant phenotype by reducing Sonic Hedgehog (SHH) signaling. These results show that Sp8 primarily functions to promote Fgf expression in the ANR and OP signaling centers that drive the survival, proliferation, and differentiation of the NC and paraxial mesoderm that make the face.
Collapse
Affiliation(s)
- Abigail D Kasberg
- Division of Developmental Biology, Cincinnati Children's Medical Center, Cincinnati, OH 45229, USA
| | | | | |
Collapse
|
40
|
Gurzu S, Gozar H, Derzsi Z, Szabo B, Jung I. Second case of a fetal sacrococcygeal neuroectodermal cyst in a male newborn. Pathology 2013; 45:188-91. [PMID: 23250044 DOI: 10.1097/pat.0b013e32835c7804] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
|
41
|
Lin C, Yin Y, Bell SM, Veith GM, Chen H, Huh SH, Ornitz DM, Ma L. Delineating a conserved genetic cassette promoting outgrowth of body appendages. PLoS Genet 2013; 9:e1003231. [PMID: 23358455 PMCID: PMC3554569 DOI: 10.1371/journal.pgen.1003231] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2012] [Accepted: 11/26/2012] [Indexed: 12/27/2022] Open
Abstract
The acquisition of the external genitalia allowed mammals to cope with terrestrial-specific reproductive needs for internal fertilization, and thus it represents one of the most fundamental steps in evolution towards a life on land. How genitalia evolved remains obscure, and the key to understanding this process may lie in the developmental genetics that underpins the early establishment of the genital primordium, the genital tubercle (GT). Development of the GT is similar to that of the limb, which requires precise regulation from a distal signaling epithelium. However, whether outgrowth of the GT and limbs is mediated by common instructive signals remains unknown. In this study, we used comprehensive genetic approaches to interrogate the signaling cascade involved in GT formation in comparison with limb formation. We demonstrate that the FGF ligand responsible for GT development is FGF8 expressed in the cloacal endoderm. We further showed that forced Fgf8 expression can rescue limb and GT reduction in embryos deficient in WNT signaling activity. Our studies show that the regulation of Fgf8 by the canonical WNT signaling pathway is mediated in part by the transcription factor SP8. Sp8 mutants elicit appendage defects mirroring WNT and FGF mutants, and abolishing Sp8 attenuates ectopic appendage development caused by a gain-of-function β-catenin mutation. These observations indicate that a conserved WNT-SP8-FGF8 genetic cassette is employed by both appendages for promoting outgrowth, and suggest a deep homology shared by the limb and external genitalia. Mammalian limbs and external genitalia are body appendages specialized for locomotion and internal fertilization, respectively. Despite their marked anatomical and functional differences, development of the limb and external genitalia appears to involve similar genetic controls, and some have suggested that regulatory mechanisms common to both might be evolutionarily linked. One essential aspect for appendage development is the establishment and maintenance of a separated proximodistal developmental axis apart from the main body axis, which is often instructed by a distal signaling epithelium. Herein, we adopted comprehensive mouse genetic approaches to investigate regulatory mechanisms underlying the distal signaling center in the limb and the GT, and uncovered a conserved genetic cassette that is utilized by both paired and unpaired appendages to establish a distal signaling center in the epithelium that mediates subsequent proximodistal outgrowth. Our results further suggested that the evolution of the external genital organ involved co-option of the same genetic program underpinning limb development.
Collapse
Affiliation(s)
- Congxing Lin
- Division of Dermatology, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri, United States of America
| | - Yan Yin
- Division of Dermatology, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri, United States of America
| | - Sheila M. Bell
- Perinatal Institute of Cincinnati Children's Hospital Medical Center, Division of Neonatology-and Pulmonary Biology, University of Cincinnati College of Medicine, Cincinnati, Ohio, United States of America
| | - G. Michael Veith
- Division of Dermatology, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri, United States of America
| | - Hong Chen
- Division of Dermatology, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri, United States of America
| | - Sung-Ho Huh
- Department of Developmental Biology, Washington University School of Medicine, St. Louis, Missouri, United States of America
| | - David M. Ornitz
- Department of Developmental Biology, Washington University School of Medicine, St. Louis, Missouri, United States of America
| | - Liang Ma
- Division of Dermatology, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri, United States of America
- Department of Developmental Biology, Washington University School of Medicine, St. Louis, Missouri, United States of America
- * E-mail:
| |
Collapse
|
42
|
Ing T, Tseng A, Sustar A, Schubiger G. Sp1 modifies leg-to-wing transdetermination in Drosophila. Dev Biol 2013; 373:290-9. [PMID: 23165292 DOI: 10.1016/j.ydbio.2012.11.008] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2012] [Revised: 10/20/2012] [Accepted: 11/09/2012] [Indexed: 10/27/2022]
Abstract
During Drosophila development, the transcription factor Sp1 is necessary for proper leg growth and also to repress wing development. Here we test the role of Sp1 during imaginal disc regeneration. Ubiquitous expression of wg induces a regeneration blastema in the dorsal aspect of the leg disc. Within this outgrowth, the wing selector gene vg is activated in some cells, changing their fate to wing identity in a process known as transdetermination. In this report we demonstrate that reducing the gene copy number of Sp1 significantly increases both the frequency and the area of transdetermination in regenerating leg discs. By examining the expression of known Sp1 target genes, we also show that the proximo-distal patterning gene dachshund is downregulated dorsally, leading to a break in its normal ring-shaped expression pattern. We further report that transdetermination, as evidenced by Vg expression, is only observed when there is a broken ring of Dachshund expression. Combined, these studies establish a role for Sp1 in leg-to-wing transdetermination.
Collapse
Affiliation(s)
- Thomas Ing
- Department of Biology, University of Washington, Seattle, WA 98195, USA.
| | | | | | | |
Collapse
|
43
|
Borello U, Madhavan M, Vilinsky I, Faedo A, Pierani A, Rubenstein J, Campbell K. Sp8 and COUP-TF1 reciprocally regulate patterning and Fgf signaling in cortical progenitors. ACTA ACUST UNITED AC 2013; 24:1409-21. [PMID: 23307639 DOI: 10.1093/cercor/bhs412] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
To gain new insights into the transcriptional regulation of cortical development, we examined the role of the transcription factor Sp8, which is downstream of Fgf8 signaling and known to promote rostral cortical development. We have used a binary transgenic system to express Sp8 throughout the mouse telencephalon in a temporally restricted manner. Our results show that misexpression of Sp8 throughout the telencephalon, at early but not late embryonic stages, results in cortical hypoplasia, which is accompanied by increased cell death, reduced proliferation, and precocious neuronal differentiation. Misexpression of Sp8 at early developmental stages represses COUP-TF1 expression, a negative effector of Fgf signaling and a key promoter of posterior cortical identity, while ablation of Sp8 has the opposite effect. In addition, transgenic misexpression of COUP-TF1 resulted in downregulation of Sp8, indicating a reciprocal cross-regulation between these 2 transcription factors. Although Sp8 has been suggested to induce and/or maintain Fgf8 expression in the embryonic telencephalon, neither Fgf8 nor Fgf15 was upregulated using our gain-of-function approach. However, misexpression of Sp8 greatly increased the expression of Fgf target molecules, suggesting enhanced Fgf signaling. Thus, we propose that Sp8 promotes rostral and dorsomedial cortical development by repressing COUP-TF1 and promoting Fgf signaling in pallial progenitors.
Collapse
Affiliation(s)
- Ugo Borello
- Inserm U846, Stem Cell and Brain Research Institute, Bron 69500, France
| | | | | | | | | | | | | |
Collapse
|
44
|
Casanova JC, Badia-Careaga C, Uribe V, Sanz-Ezquerro JJ. Bambi and Sp8 expression mark digit tips and their absence shows that chick wing digits 2 and 3 are truncated. PLoS One 2012; 7:e52781. [PMID: 23285181 PMCID: PMC3532063 DOI: 10.1371/journal.pone.0052781] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2012] [Accepted: 11/20/2012] [Indexed: 11/19/2022] Open
Abstract
An often overlooked aspect of digit development is the special nature of the terminal phalanx, a specialized structure with characteristics distinct from other phalanges, for example the presence of ectodermal derivatives such as nails and claws. Here, we describe the unique ossification pattern of distal phalanges and characteristic gene expression in the digit tips of chick and duck embryos. Our results show that the distal phalanx of chick wing digit 1 is a genuine tip with a characteristic ossification pattern and expression of Bambi and Sp8; however, the terminal phalanx of digits 2* and 3 is not a genuine tip, and these are therefore truncated digits. Bambi and Sp8 expression in the chick wing provides a direct molecular assessment of digit identity changes after experimental manipulations of digit primordia. In contrast, digits 1 and 2 of the duck wing both possess true tips. Although chick wing-tip development was not rescued by application of Fgf8, this treatment induced the development of extra phalanges. Grafting experiments show that competence for tip formation, including nails, is latent in the interdigital tissue. Our results deepen understanding of the mechanisms of digit tip formation, highlighting its developmental autonomy and modular nature, with implications for digit reduction or loss during evolution. * Numbering of wing digits is 1, 2, 3 from anterior to posterior.
Collapse
Affiliation(s)
- Jesús C. Casanova
- Department of Cardiovascular Development and Repair, Centro Nacional de Investigaciones Cardiovasculares, CNIC, Madrid, Spain
- Department of Immunology and Oncology, Centro Nacional de Biotecnologia, CSIC, Madrid, Spain
| | - Claudio Badia-Careaga
- Department of Cardiovascular Development and Repair, Centro Nacional de Investigaciones Cardiovasculares, CNIC, Madrid, Spain
| | - Verónica Uribe
- Department of Cardiovascular Development and Repair, Centro Nacional de Investigaciones Cardiovasculares, CNIC, Madrid, Spain
| | - Juan José Sanz-Ezquerro
- Department of Cardiovascular Development and Repair, Centro Nacional de Investigaciones Cardiovasculares, CNIC, Madrid, Spain
- Department of Immunology and Oncology, Centro Nacional de Biotecnologia, CSIC, Madrid, Spain
- Department of Molecular and Cellular Biology, Centro Nacional de Biotecnologia, CSIC, Madrid, Spain
- * E-mail:
| |
Collapse
|
45
|
Rabinowitz AH, Vokes SA. Integration of the transcriptional networks regulating limb morphogenesis. Dev Biol 2012; 368:165-80. [PMID: 22683377 DOI: 10.1016/j.ydbio.2012.05.035] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2012] [Revised: 05/29/2012] [Accepted: 05/29/2012] [Indexed: 12/29/2022]
Abstract
The developing limb is one of the best described vertebrate systems for understanding how coordinated gene expression during embryogenesis leads to the structures present in the mature organism. This knowledge, derived from decades of research, is largely based upon gain- and loss-of-function experiments. These studies have provided limited information about how the key signaling pathways interact with each other and the downstream effectors of these pathways. We summarize our current understanding of known genetic interactions in the context of three temporally defined gene regulatory networks. These networks crystallize our current knowledge, depicting a dynamic process involving multiple feedback loops between the ectoderm and mesoderm. At the same time, they highlight the fact that many essential processes are still largely undescribed. Much of the dynamic transcriptional activity occurring during development is regulated by distal cis-regulatory elements. Modern genomic tools have provided new approaches for studying the function of cis-regulatory elements and we discuss the results of these studies in regard to understanding limb development. Ultimately, these genomic techniques will allow scientists to understand how multiple signaling pathways are integrated in space and time to drive gene expression and regulate the formation of the limb.
Collapse
Affiliation(s)
- Adam H Rabinowitz
- Section of Molecular Cell & Developmental Biology, Institute for Cellular and Molecular Biology, One University Station A4800, Austin, TX 78712, USA
| | | |
Collapse
|
46
|
Estella C, Voutev R, Mann RS. A dynamic network of morphogens and transcription factors patterns the fly leg. Curr Top Dev Biol 2012; 98:173-98. [PMID: 22305163 DOI: 10.1016/b978-0-12-386499-4.00007-0] [Citation(s) in RCA: 68] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Animal appendages require a proximodistal (PD) axis, which forms orthogonally from the two main body axes, anteroposterior and dorsoventral. In this review, we discuss recent advances that begin to provide insights into the molecular mechanisms controlling PD axis formation in the Drosophila leg. In this case, two morphogens, Wingless (Wg) and Decapentaplegic (Dpp), initiate a genetic cascade that, together with growth of the leg imaginal disc, establishes the PD axis. The analysis of cis-regulatory modules (CRMs) that control the expression of genes at different positions along the PD axis has been particularly valuable in dissecting this complex process. From these experiments, it appears that only one concentration of Wg and Dpp are required to initiate PD axis formation by inducing the expression of Distal-less (Dll), a homeodomain-encoding gene that is required for leg development. Once Dll is turned on, it activates the medially expressed gene dachshund (dac). Cross-regulation between Dll and dac, together with cell proliferation in the growing leg imaginal disc, results in the formation of a rudimentary PD axis. Wg and Dpp also initiate the expression of ligands for the EGFR pathway, which in turn induces the expression of a series of target genes that pattern the distal-most portion of the leg.
Collapse
Affiliation(s)
- Carlos Estella
- Department of Biochemistry and Molecular Biophysics, Columbia University, New York, New York, USA
| | | | | |
Collapse
|
47
|
Ma T, Zhang Q, Cai Y, You Y, Rubenstein JLR, Yang Z. A subpopulation of dorsal lateral/caudal ganglionic eminence-derived neocortical interneurons expresses the transcription factor Sp8. Cereb Cortex 2011; 22:2120-30. [PMID: 22021915 DOI: 10.1093/cercor/bhr296] [Citation(s) in RCA: 59] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Cortical GABAergic interneurons in rodents originate from subpallial progenitors and tangentially migrate to the cortex. While the majority of mouse neocortical interneurons are derived from the medial and caudal ganglionic eminence (MGE and CGE, respectively), it remains unknown whether the lateral ganglionic eminence (LGE) also contributes to a subpopulation of cortical interneurons. Here, we show that the transcription factor Sp8 is expressed in one-fifth of adult cortical interneurons, which appear to be derived from both the dorsal LGE and the dorsal CGE (dLGE and dCGE, respectively). Compared with the MGE-derived cortical interneurons, dLGE/dCGE-derived Sp8-expressing (Sp8+) ones are born at later embryonic stages with peak production occurring at embryonic day 15.5. They tangentially migrate mainly along the subventricular/intermediate zone (SVZ/IZ) route; some continue to express mitotic markers (Ki67 and PH3) in the neonatal cortical SVZ/IZ. Sp8+ interneurons continue to radially migrate from the SVZ/IZ into the cortical layers at early postnatal stages. In contrast to MGE-derived interneurons, dLGE/dCGE-derived Sp8+ interneurons follow an outside-in layering pattern, preferentially occupying superficial cortical layers.
Collapse
Affiliation(s)
- Tong Ma
- State Key Laboratory of Medical Neurobiology, Institutes of Brain Science, Fudan University, Shanghai, China
| | | | | | | | | | | |
Collapse
|
48
|
Krol AJ, Roellig D, Dequéant ML, Tassy O, Glynn E, Hattem G, Mushegian A, Oates AC, Pourquié O. Evolutionary plasticity of segmentation clock networks. Development 2011; 138:2783-92. [PMID: 21652651 DOI: 10.1242/dev.063834] [Citation(s) in RCA: 139] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
The vertebral column is a conserved anatomical structure that defines the vertebrate phylum. The periodic or segmental pattern of the vertebral column is established early in development when the vertebral precursors, the somites, are rhythmically produced from presomitic mesoderm (PSM). This rhythmic activity is controlled by a segmentation clock that is associated with the periodic transcription of cyclic genes in the PSM. Comparison of the mouse, chicken and zebrafish PSM oscillatory transcriptomes revealed networks of 40 to 100 cyclic genes mostly involved in Notch, Wnt and FGF signaling pathways. However, despite this conserved signaling oscillation, the identity of individual cyclic genes mostly differed between the three species, indicating a surprising evolutionary plasticity of the segmentation networks.
Collapse
Affiliation(s)
- Aurélie J Krol
- Stowers Institute for Medical Research, Kansas City, MO 64110, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Soncin F, Mohamet L, Ritson S, Hawkins K, Bobola N, Zeef L, Merry CLR, Ward CM. E-cadherin acts as a regulator of transcripts associated with a wide range of cellular processes in mouse embryonic stem cells. PLoS One 2011; 6:e21463. [PMID: 21779327 PMCID: PMC3136471 DOI: 10.1371/journal.pone.0021463] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2010] [Accepted: 06/01/2011] [Indexed: 11/18/2022] Open
Abstract
Background We have recently shown that expression of the cell adhesion molecule E-cadherin is required for LIF-dependent pluripotency of mouse embryonic stem (ES) cells. Methodology In this study, we have assessed global transcript expression in E-cadherin null (Ecad-/-) ES cells cultured in either the presence or absence of LIF and compared these to the parental cell line wtD3. Results We show that LIF has little effect on the transcript profile of Ecad-/- ES cells, with statistically significant transcript alterations observed only for Sp8 and Stat3. Comparison of Ecad-/- and wtD3 ES cells cultured in LIF demonstrated significant alterations in the transcript profile, with effects not only confined to cell adhesion and motility but also affecting, for example, primary metabolic processes, catabolism and genes associated with apoptosis. Ecad-/- ES cells share similar, although not identical, gene expression profiles to epiblast-derived pluripotent stem cells, suggesting that E-cadherin expression may inhibit inner cell mass to epiblast transition. We further show that Ecad-/- ES cells maintain a functional β-catenin pool that is able to induce β-catenin/TCF-mediated transactivation but, contrary to previous findings, do not display endogenous β-catenin/TCF-mediated transactivation. We conclude that loss of E-cadherin in mouse ES cells leads to significant transcript alterations independently of β-catenin/TCF transactivation.
Collapse
Affiliation(s)
- Francesca Soncin
- Core Technology Facility, Faculty of Medical and Human Sciences, The University of Manchester, Manchester, United Kingdom
- Materials Science Centre, The University of Manchester, Manchester, United Kingdom
| | - Lisa Mohamet
- Core Technology Facility, Faculty of Medical and Human Sciences, The University of Manchester, Manchester, United Kingdom
| | - Sarah Ritson
- Core Technology Facility, Faculty of Medical and Human Sciences, The University of Manchester, Manchester, United Kingdom
| | - Kate Hawkins
- Core Technology Facility, Faculty of Medical and Human Sciences, The University of Manchester, Manchester, United Kingdom
| | - Nicoletta Bobola
- Core Technology Facility, Faculty of Medical and Human Sciences, The University of Manchester, Manchester, United Kingdom
| | - Leo Zeef
- Microarray Facility, Faculty of Life Sciences, Michael Smith Building, The University of Manchester, Manchester, United Kingdom
| | | | - Christopher M. Ward
- Core Technology Facility, Faculty of Medical and Human Sciences, The University of Manchester, Manchester, United Kingdom
- * E-mail:
| |
Collapse
|
50
|
A symphony of regulations centered on p63 to control development of ectoderm-derived structures. J Biomed Biotechnol 2011; 2011:864904. [PMID: 21716671 PMCID: PMC3118300 DOI: 10.1155/2011/864904] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2010] [Revised: 01/25/2011] [Accepted: 03/16/2011] [Indexed: 12/27/2022] Open
Abstract
The p53-related transcription factor p63 is critically important for basic cellular functions during development of the ectoderm and derived structure and tissues, including skin, limb, palate, and hair. On the one side, p63 is required to sustain the proliferation of keratinocyte progenitors, while on the other side it is required for cell stratification, commitment to differentiate, cell adhesion, and epithelial-mesenchymal signaling. Molecules that are components or regulators of the p63 pathway(s) are rapidly being identified, and it comes with no surprise that alterations in the p63 pathway lead to congenital conditions in which the skin and other ectoderm-derived structures are affected. In this paper, we summarize the current knowledge of the molecular and cellular regulations centered on p63, derived from the comprehension of p63-linked human diseases and the corresponding animal models, as well as from cellular models and high-throughput molecular approaches. We point out common themes and features, that allow to speculate on the possible role of p63 downstream events and their potential exploitation in future attempts to correct the congenital defect in preclinical studies.
Collapse
|