1
|
Bogart AH, Brooks ER. Canonical Wnt pathway modulation is required to correctly execute multiple independent cellular dynamic programs during cranial neural tube closure. Dev Biol 2025; 523:115-131. [PMID: 40280384 DOI: 10.1016/j.ydbio.2025.04.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2024] [Revised: 04/21/2025] [Accepted: 04/22/2025] [Indexed: 04/29/2025]
Abstract
Defects in cranial neural tube closure are among the most common and deleterious human structural birth defects. Correct cranial closure requires the coordination of multiple cell dynamic programs including cell proliferation and cell shape change. Mutations that impact Wnt signaling, including loss of the pathway co-receptor LRP6, lead to defects in cranial neural tube closure, but the cellular dynamics under control of the Wnt pathway during this critical morphogenetic process remain unclear. Here, we use mice mutant for LRP6 to examine the consequences of conditional and global reduction in Wnt signaling and mutants with conditional inactivation of APC to examine the consequences of pathway hyperactivation. Strikingly, we find that regulated Wnt signaling is required for two independent events during cranial neural tube closure. First, global reduction of Wnt leads to a surprising hyperplasia of the cranial neural folds driven by excessive cell proliferation at early pre-elevation stages, with the increased tissue volume creating a mechanical blockade to efficient closure despite normal apical constriction and cell polarization at later stages. Conversely, conditional hyperactivation of the pathway at later elevation stages prevents correct actin organization, blocking apical constriction and neural fold elevation without impacting tissue scaling. Together these data reveal that Wnt signaling levels must be modulated to restrict proliferation at early stages and promote apical constriction at later elevation stages to drive efficient closure of the cranial neural tube.
Collapse
Affiliation(s)
- Amber Huffine Bogart
- Department of Molecular Biomedical Sciences, College of Veterinary Medicine, North Carolina State University, United States
| | - Eric R Brooks
- Department of Molecular Biomedical Sciences, College of Veterinary Medicine, North Carolina State University, United States.
| |
Collapse
|
2
|
Göksu AY. A review article on the development of dopaminergic neurons and establishment of dopaminergic neuron-based in vitro models by using immortal cell lines or stem cells to study and treat Parkinson's disease. Int J Dev Neurosci 2024; 84:817-842. [PMID: 39379284 DOI: 10.1002/jdn.10383] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2023] [Revised: 09/09/2024] [Accepted: 09/12/2024] [Indexed: 10/10/2024] Open
Abstract
The primary pathological hallmark of Parkinson's disease (PD) is the degeneration of dopaminergic (DA) neurons in the substantia nigra pars compacta, a critical midbrain region. In vitro models based on DA neurons provide a powerful platform for investigating the cellular and molecular mechanisms of PD and testing novel therapeutic strategies. A deep understanding of DA neuron development, including the signalling pathways and transcription factors involved, is essential for advancing PD research. This article first explores the differentiation and maturation processes of DA neurons in the midbrain, detailing the relevant signalling pathways. It then compares various in vitro models, including primary cells, immortalized cell lines, and stem cell-based models, focusing on the advantages and limitations of each. Special attention is given to the role of immortalized and stem cell models in PD research. This review aims to guide researchers in selecting the most appropriate model for their specific research goals. Ethical considerations and clinical implications of using stem cells in PD research are also discussed.
Collapse
Affiliation(s)
- Azize Yasemin Göksu
- Department of Histology and Embryology, Department of Gene and Cell Therapy, Akdeniz University, School of Medicine, Antalya, Turkey
| |
Collapse
|
3
|
Matsuda M, Rozman J, Ostvar S, Kasza KE, Sokol SY. Mechanical control of neural plate folding by apical domain alteration. Nat Commun 2023; 14:8475. [PMID: 38123550 PMCID: PMC10733383 DOI: 10.1038/s41467-023-43973-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2023] [Accepted: 11/23/2023] [Indexed: 12/23/2023] Open
Abstract
Vertebrate neural tube closure is associated with complex changes in cell shape and behavior, however, the relative contribution of these processes to tissue folding is not well understood. At the onset of Xenopus neural tube folding, we observed alternation of apically constricted and apically expanded cells. This apical domain heterogeneity was accompanied by biased cell orientation along the anteroposterior axis, especially at neural plate hinges, and required planar cell polarity signaling. Vertex models suggested that dispersed isotropically constricting cells can cause the elongation of adjacent cells. Consistently, in ectoderm, cell-autonomous apical constriction was accompanied by neighbor expansion. Thus, a subset of isotropically constricting cells may initiate neural plate bending, whereas a 'tug-of-war' contest between the force-generating and responding cells reduces its shrinking along the body axis. This mechanism is an alternative to anisotropic shrinking of cell junctions that are perpendicular to the body axis. We propose that apical domain changes reflect planar polarity-dependent mechanical forces operating during neural folding.
Collapse
Affiliation(s)
- Miho Matsuda
- Department of Cell, Developmental and Regenerative Biology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Jan Rozman
- Rudolf Peierls Centre for Theoretical Physics, University of Oxford, Oxford, UK
| | - Sassan Ostvar
- Department of Mechanical Engineering, Columbia University, New York, NY, USA
| | - Karen E Kasza
- Department of Mechanical Engineering, Columbia University, New York, NY, USA
| | - Sergei Y Sokol
- Department of Cell, Developmental and Regenerative Biology, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
| |
Collapse
|
4
|
Azbazdar Y, Pera EM, De Robertis EM. Head organizer: Cerberus and IGF cooperate in brain induction in Xenopus embryos. Cells Dev 2023:203897. [PMID: 38109998 DOI: 10.1016/j.cdev.2023.203897] [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: 11/21/2023] [Accepted: 12/09/2023] [Indexed: 12/20/2023]
Abstract
Neural induction by cell-cell signaling was discovered a century ago by the organizer transplantations of Spemann and Mangold in amphibians. Spemann later found that early dorsal blastopore lips induced heads and late organizers trunk-tail structures. Identifying region-specific organizer signals has been a driving force in the progress of animal biology. Head induction in the absence of trunk is designated archencephalic differentiation. Two specific head inducers, Cerberus and Insulin-like growth factors (IGFs), that induce archencephalic brain but not trunk-tail structures have been described previously. However, whether these two signals interact with each other had not been studied to date and was the purpose of the present investigation. It was found that Cerberus, a multivalent growth factor antagonist that inhibits Nodal, BMP and Wnt signals, strongly cooperated with IGF2, a growth factor that provides a positive signal through tyrosine kinase IGF receptors that activate MAPK and other pathways. The ectopic archencephalic structures induced by the combination of Cerberus and IGF2 are of higher frequency and larger than either one alone. They contain brain, a cyclopic eye and multiple olfactory placodes, without trace of trunk structures such as notochord or somites. A dominant-negative secreted IGF receptor 1 blocked Cerberus activity, indicating that endogenous IGF signals are required for ectopic brain formation. In a sensitized embryonic system, in which embryos were depleted of β-catenin, IGF2 did not by itself induce neural tissue while in combination with Cerberus it greatly enhanced formation of circular brain structures expressing the anterior markers Otx2 and Rx2a, but not spinal cord or notochord markers. The main conclusion of this work is that IGF provides a positive signal initially uniformly expressed throughout the embryo that potentiates the effect of an organizer-specific negative signal mediated by Cerberus. The results are discussed in the context of the history of neural induction.
Collapse
Affiliation(s)
- Yagmur Azbazdar
- Department of Biological Chemistry, David Geffen School of Medicine, University of California, Los Angeles, 90095-1662, USA
| | - Edgar M Pera
- Vertebrate Developmental Biology Laboratory, Department of Laboratory Medicine, Lund Stem Cell Center, University of Lund, 22184 Lund, Sweden
| | - Edward M De Robertis
- Department of Biological Chemistry, David Geffen School of Medicine, University of California, Los Angeles, 90095-1662, USA.
| |
Collapse
|
5
|
Asmar AJ, Abrams SR, Hsin J, Collins JC, Yazejian RM, Wu Y, Cho J, Doyle AD, Cinthala S, Simon M, van Jaarsveld RH, Beck DB, Kerosuo L, Werner A. A ubiquitin-based effector-to-inhibitor switch coordinates early brain, craniofacial, and skin development. Nat Commun 2023; 14:4499. [PMID: 37495603 PMCID: PMC10371987 DOI: 10.1038/s41467-023-40223-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2022] [Accepted: 07/18/2023] [Indexed: 07/28/2023] Open
Abstract
The molecular mechanisms that coordinate patterning of the embryonic ectoderm into spatially distinct lineages to form the nervous system, epidermis, and neural crest-derived craniofacial structures are unclear. Here, biochemical disease-variant profiling reveals a posttranslational pathway that drives early ectodermal differentiation in the vertebrate head. The anteriorly expressed ubiquitin ligase CRL3-KLHL4 restricts signaling of the ubiquitous cytoskeletal regulator CDC42. This regulation relies on the CDC42-activating complex GIT1-βPIX, which CRL3-KLHL4 exploits as a substrate-specific co-adaptor to recognize and monoubiquitylate PAK1. Surprisingly, we find that ubiquitylation converts the canonical CDC42 effector PAK1 into a CDC42 inhibitor. Loss of CRL3-KLHL4 or a disease-associated KLHL4 variant reduce PAK1 ubiquitylation causing overactivation of CDC42 signaling and defective ectodermal patterning and neurulation. Thus, tissue-specific restriction of CDC42 signaling by a ubiquitin-based effector-to-inhibitor is essential for early face, brain, and skin formation, revealing how cell-fate and morphometric changes are coordinated to ensure faithful organ development.
Collapse
Affiliation(s)
- Anthony J Asmar
- Stem Cell Biochemistry Unit, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Shaun R Abrams
- Stem Cell Biochemistry Unit, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD, 20892, USA
- Neural Crest Development & Disease Unit, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Jenny Hsin
- Neural Crest Development & Disease Unit, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Jason C Collins
- Stem Cell Biochemistry Unit, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Rita M Yazejian
- Neural Crest Development & Disease Unit, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Youmei Wu
- Stem Cell Biochemistry Unit, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Jean Cho
- Stem Cell Biochemistry Unit, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Andrew D Doyle
- NIDCR Imaging Core, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Samhitha Cinthala
- Stem Cell Biochemistry Unit, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Marleen Simon
- Department of Genetics, University Medical Center Utrecht, Utrecht, the Netherlands
| | | | - David B Beck
- Division of Rheumatology, Department of Medicine, New York University Grossman School of Medicine, New York, NY, USA
- Center for Human Genetics and Genomics, New York University Grossman School of Medicine, New York, NY, USA
| | - Laura Kerosuo
- Neural Crest Development & Disease Unit, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD, 20892, USA.
| | - Achim Werner
- Stem Cell Biochemistry Unit, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD, 20892, USA.
| |
Collapse
|
6
|
Cao Y. Neural induction drives body axis formation during embryogenesis, but a neural induction-like process drives tumorigenesis in postnatal animals. Front Cell Dev Biol 2023; 11:1092667. [PMID: 37228646 PMCID: PMC10203556 DOI: 10.3389/fcell.2023.1092667] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2022] [Accepted: 04/17/2023] [Indexed: 05/27/2023] Open
Abstract
Characterization of cancer cells and neural stem cells indicates that tumorigenicity and pluripotency are coupled cell properties determined by neural stemness, and tumorigenesis represents a process of progressive loss of original cell identity and gain of neural stemness. This reminds of a most fundamental process required for the development of the nervous system and body axis during embryogenesis, i.e., embryonic neural induction. Neural induction is that, in response to extracellular signals that are secreted by the Spemann-Mangold organizer in amphibians or the node in mammals and inhibit epidermal fate in ectoderm, the ectodermal cells lose their epidermal fate and assume the neural default fate and consequently, turn into neuroectodermal cells. They further differentiate into the nervous system and also some non-neural cells via interaction with adjacent tissues. Failure in neural induction leads to failure of embryogenesis, and ectopic neural induction due to ectopic organizer or node activity or activation of embryonic neural genes causes a formation of secondary body axis or a conjoined twin. During tumorigenesis, cells progressively lose their original cell identity and gain of neural stemness, and consequently, gain of tumorigenicity and pluripotency, due to various intra-/extracellular insults in cells of a postnatal animal. Tumorigenic cells can be induced to differentiation into normal cells and integrate into normal embryonic development within an embryo. However, they form tumors and cannot integrate into animal tissues/organs in a postnatal animal because of lack of embryonic inducing signals. Combination of studies of developmental and cancer biology indicates that neural induction drives embryogenesis in gastrulating embryos but a similar process drives tumorigenesis in a postnatal animal. Tumorigenicity is by nature the manifestation of aberrant occurrence of pluripotent state in a postnatal animal. Pluripotency and tumorigenicity are both but different manifestations of neural stemness in pre- and postnatal stages of animal life, respectively. Based on these findings, I discuss about some confusion in cancer research, propose to distinguish the causality and associations and discriminate causal and supporting factors involved in tumorigenesis, and suggest revisiting the focus of cancer research.
Collapse
Affiliation(s)
- Ying Cao
- Shenzhen Research Institute of Nanjing University, Shenzhen, China
- MOE Key Laboratory of Model Animals for Disease Study, Model Animal Research Center of Medical School, Nanjing University, Nanjing, China
- Jiangsu Key Laboratory of Molecular Medicine of Medical School, Nanjing University, Nanjing, China
| |
Collapse
|
7
|
Matsuda M, Rozman J, Ostvar S, Kasza KE, Sokol SY. Mechanical control of neural plate folding by apical domain alteration. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.02.10.528047. [PMID: 36798359 PMCID: PMC9934705 DOI: 10.1101/2023.02.10.528047] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/13/2023]
Abstract
Vertebrate neural tube closure is associated with complex changes in cell shape and behavior, however, the relative contribution of these processes to tissue folding is not well understood. In this study, we evaluated morphology of the superficial cell layer in the Xenopus neural plate. At the stages corresponding to the onset of tissue folding, we observed the alternation of cells with apically constricting and apically expanding apical domains. The cells had a biased orientation along the anteroposterior (AP) axis. This apical domain heterogeneity required planar cell polarity (PCP) signaling and was especially pronounced at neural plate hinges. Vertex model simulations suggested that spatially dispersed isotropically constricting cells cause the elongation of their non-constricting counterparts along the AP axis. Consistent with this hypothesis, cell-autonomous induction of apical constriction in Xenopus ectoderm cells was accompanied by the expansion of adjacent non-constricting cells. Our observations indicate that a subset of isotropically constricting cells can initiate neural plate bending, whereas a 'tug-of-war' contest between the force-generating and responding cells reduces its shrinking along the AP axis. This mechanism is an alternative to anisotropic shrinking of cell junctions that are perpendicular to the body axis. We propose that neural folding relies on PCP-dependent transduction of mechanical signals between neuroepithelial cells.
Collapse
|
8
|
Takebayashi-Suzuki K, Uchida M, Suzuki A. Zbtb21 is required for the anterior-posterior patterning of neural tissue in the early Xenopus embryo. Biochem Biophys Res Commun 2022; 630:190-197. [PMID: 36166855 DOI: 10.1016/j.bbrc.2022.09.048] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2022] [Accepted: 09/12/2022] [Indexed: 12/01/2022]
Abstract
The vertebrate body is organized along the dorsal-ventral (DV) and anterior-posterior (AP) axes by the BMP and Wnt pathways, respectively. We previously reported that Xenopus Zbtb14 promotes dorsalization (neural induction) of ectoderm by inhibiting BMP signaling and also posteriorizes neural tissue by activating Wnt signaling, thereby coordinating the patterning of the DV and AP axes during early development. Although it has been reported that human ZBTB21 binds to ZBTB14 and is involved in gene expression in cultured mammalian cells, the function of Zbtb21 in early embryonic development remains unknown. Here, we show that Xenopus Zbtb21 plays an essential role in AP axis formation in the early Xenopus embryo. zbtb21 and zbtb14 are co-expressed in the dorsal region of embryos during gastrulation. Simultaneous overexpression of Zbtb21 and Zbtb14 in ectodermal explants enhances the neural-inducing activity of Zbtb14. Moreover, knockdown experiments showed that Zbtb21 is required for the formation of posterior neural tissue and the suppression of anterior neural development. Collectively, these results suggest that in cooperation with Zbtb14, Zbtb21 has a crucial function in AP patterning during early Xenopus embryogenesis.
Collapse
Affiliation(s)
- Kimiko Takebayashi-Suzuki
- Amphibian Research Center, Graduate School of Integrated Sciences for Life, Hiroshima University, 1-3-1 Kagamiyama, Higashi-Hiroshima, Hiroshima, 739-8526, Japan.
| | - Misa Uchida
- Amphibian Research Center, Graduate School of Integrated Sciences for Life, Hiroshima University, 1-3-1 Kagamiyama, Higashi-Hiroshima, Hiroshima, 739-8526, Japan
| | - Atsushi Suzuki
- Amphibian Research Center, Graduate School of Integrated Sciences for Life, Hiroshima University, 1-3-1 Kagamiyama, Higashi-Hiroshima, Hiroshima, 739-8526, Japan.
| |
Collapse
|
9
|
Hongo I, Okamoto H. FGF/MAPK/Ets signaling in Xenopus ectoderm contributes to neural induction and patterning in an autonomous and paracrine manner, respectively. Cells Dev 2022; 170:203769. [DOI: 10.1016/j.cdev.2022.203769] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2021] [Revised: 01/16/2022] [Accepted: 02/15/2022] [Indexed: 10/19/2022]
|
10
|
Umair Z, Kumar V, Goutam RS, Kumar S, Lee U, Kim J. Goosecoid Controls Neuroectoderm Specification via Dual Circuits of Direct Repression and Indirect Stimulation in Xenopus Embryos. Mol Cells 2021; 44:723-735. [PMID: 34711690 PMCID: PMC8560583 DOI: 10.14348/molcells.2021.0055] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2021] [Revised: 08/03/2021] [Accepted: 08/16/2021] [Indexed: 11/29/2022] Open
Abstract
Spemann organizer is a center of dorsal mesoderm and itself retains the mesoderm character, but it has a stimulatory role for neighboring ectoderm cells in becoming neuroectoderm in gastrula embryos. Goosecoid (Gsc) overexpression in ventral region promotes secondary axis formation including neural tissues, but the role of gsc in neural specification could be indirect. We examined the neural inhibitory and stimulatory roles of gsc in the same cell and neighboring cells contexts. In the animal cap explant system, Gsc overexpression inhibited expression of neural specific genes including foxd4l1.1, zic3, ncam, and neurod. Genome-wide chromatin immunoprecipitation sequencing (ChIP-seq) and promoter analysis of early neural genes of foxd4l1.1 and zic3 were performed to show that the neural inhibitory mode of gsc was direct. Site-directed mutagenesis and serially deleted construct studies of foxd4l1.1 promoter revealed that Gsc directly binds within the foxd4l1.1 promoter to repress its expression. Conjugation assay of animal cap explants was also performed to demonstrate an indirect neural stimulatory role for gsc. The genes for secretory molecules, Chordin and Noggin, were up-regulated in gsc injected cells with the neural fate only achieved in gsc uninjected neighboring cells. These experiments suggested that gsc regulates neuroectoderm formation negatively when expressed in the same cell and positively in neighboring cells via soluble factors. One is a direct suppressive circuit of neural genes in gsc expressing mesoderm cells and the other is an indirect stimulatory circuit for neurogenesis in neighboring ectoderm cells via secreted BMP antagonizers.
Collapse
Affiliation(s)
- Zobia Umair
- Department of Biochemistry, Institute of Cell Differentiation and Aging, College of Medicine, Hallym University, Chuncheon 24252, Korea
- Department of Molecular Medicine, School of Medicine, Gachon University, Incheon 21999, Korea
| | - Vijay Kumar
- Department of Biochemistry, Institute of Cell Differentiation and Aging, College of Medicine, Hallym University, Chuncheon 24252, Korea
| | - Ravi Shankar Goutam
- Department of Biochemistry, Institute of Cell Differentiation and Aging, College of Medicine, Hallym University, Chuncheon 24252, Korea
| | - Shiv Kumar
- Department of Biochemistry, Institute of Cell Differentiation and Aging, College of Medicine, Hallym University, Chuncheon 24252, Korea
| | - Unjoo Lee
- Department of Electrical Engineering, Hallym University, Chuncheon 24252, Korea
| | - Jaebong Kim
- Department of Biochemistry, Institute of Cell Differentiation and Aging, College of Medicine, Hallym University, Chuncheon 24252, Korea
| |
Collapse
|
11
|
Neural is Fundamental: Neural Stemness as the Ground State of Cell Tumorigenicity and Differentiation Potential. Stem Cell Rev Rep 2021; 18:37-55. [PMID: 34714532 DOI: 10.1007/s12015-021-10275-y] [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] [Accepted: 10/01/2021] [Indexed: 01/07/2023]
Abstract
Tumorigenic cells are similar to neural stem cells or embryonic neural cells in regulatory networks, tumorigenicity and pluripotent differentiation potential. By integrating the evidence from developmental biology, tumor biology and evolution, I will make a detailed discussion on the observations and propose that neural stemness underlies two coupled cell properties, tumorigenicity and pluripotent differentiation potential. Neural stemness property of tumorigenic cells can hopefully integrate different observations/concepts underlying tumorigenesis. Neural stem cells and tumorigenic cells share regulatory networks; both exhibit neural stemness, tumorigenicity and pluripotent differentiation potential; both depend on expression or activation of ancestral genes; both rely primarily on aerobic glycolytic metabolism; both can differentiate into various cells/tissues that are derived from three germ layers, leading to tumor formation resembling severely disorganized or more degenerated process of embryonic tissue differentiation; both are enriched in long genes with more splice variants that provide more plastic scaffolds for cell differentiation, etc. Neural regulatory networks, which include higher levels of basic machineries of cell physiological functions and developmental programs, work concertedly to define a basic state with fast cell cycle and proliferation. This is predestined by the evolutionary advantage of neural state, the ground or initial state for multicellularity with adaptation to an ancient environment. Tumorigenesis might represent a process of restoration of neural ground state, thereby restoring a state with fast proliferation and pluripotent differentiation potential in somatic cells. Tumorigenesis and pluripotent differentiation potential might be better understood from understanding neural stemness, and cancer therapy should benefit more from targeting neural stemness.
Collapse
|
12
|
Abstract
It has previously been reported that in ex vivo planar explants prepared from Xenopus laevis embryos, the intracellular pH (pHi) increases in cells of the dorsal ectoderm from stage 10.5 to 11.5 (i.e. 11-12.5 hpf). It was proposed that such increases (potentially due to H+ being extruded, sequestered, or buffered in some manner), play a role in regulating neural induction. Here, we used an extracellular ion-selective electrode to non-invasively measure H+ fluxes at eight locations around the equatorial circumference of intact X. laevis embryos between stages 9-12 (˜7-13.25 hpf). We showed that at stages 9-11, there was a small H+ efflux recorded from all the measuring positions. At stage 12 there was a small, but significant, increase in the efflux of H+ from most locations, but the efflux from the dorsal side of the embryo was significantly greater than from the other positions. Embryos were also treated from stages 9-12 with bafilomycin A1, to block the activity of the ATP-driven H+ pump. By stage 22 (24 hpf), these embryos displayed retarded development, arresting before the end of gastrulation and therefore did not display the usual anterior and neural structures, which were observed in the solvent-control embryos. In addition, expression of the early neural gene, Zic3, was absent in treated embryos compared with the solvent controls. Together, our new in vivo data corroborated and extended the earlier explant-derived report describing changes in pHi that were suggested to play a role during neural induction in X. laevis embryos.
Collapse
|
13
|
Mancini P, Ossipova O, Sokol SY. The dorsal blastopore lip is a source of signals inducing planar cell polarity in the Xenopus neural plate. Biol Open 2021; 10:bio058761. [PMID: 34259326 PMCID: PMC8325942 DOI: 10.1242/bio.058761] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2021] [Accepted: 06/04/2021] [Indexed: 12/03/2022] Open
Abstract
Coordinated polarization of cells in the tissue plane, known as planar cell polarity (PCP), is associated with a signaling pathway critical for the control of morphogenetic processes. Although the segregation of PCP components to opposite cell borders is believed to play a critical role in this pathway, whether PCP derives from egg polarity or preexistent long-range gradient, or forms in response to a localized cue, remains a challenging question. Here we investigate the Xenopus neural plate, a tissue that has been previously shown to exhibit PCP. By imaging Vangl2 and Prickle3, we show that PCP is progressively acquired in the neural plate and requires a signal from the posterior region of the embryo. Tissue transplantations indicated that PCP is triggered in the neural plate by a planar cue from the dorsal blastopore lip. The PCP cue did not depend on the orientation of the graft and was distinct from neural inducers. These observations suggest that neuroectodermal PCP is not instructed by a preexisting molecular gradient but induced by a signal from the dorsal blastopore lip.
Collapse
Affiliation(s)
| | | | - Sergei Y. Sokol
- Department of Cell, Developmental and Regenerative Biology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| |
Collapse
|
14
|
Becerra-Solano LE, Mateos-Sánchez L, López-Muñoz E. Microcephaly, an etiopathogenic vision. Pediatr Neonatol 2021; 62:354-360. [PMID: 34112604 DOI: 10.1016/j.pedneo.2021.05.008] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/04/2020] [Revised: 03/30/2021] [Accepted: 05/17/2021] [Indexed: 11/18/2022] Open
Abstract
Microcephaly is defined by an occipital-frontal head circumference (OFD) 2 standard deviations (SD) smaller than the average expected for age, gender and population. Its incidence has been reported between 1.3 and 150 cases per 100,000 births. Currently, new clinical characteristics, causes and pathophysiological mechanisms related to microcephaly continue to be identified. Its etiology is varied and heterogeneous, with genetic and non-genetic factors that produce alterations in differentiation, proliferation, migration, repair of damage to deoxyribonucleic acid and neuronal apoptosis. It requires a multidisciplinary diagnostic approach that includes a medical history, detailed prenatal and postnatal clinical evaluation, cerebral magnetic resonance imaging, neuropsychological evaluation, and in some cases complementary tests such as metabolic screening, tests to rule out infectious processes and genetic testing. There is no specific treatment or intervention to increase cerebral growth; however, timely intervention strategies and programs can be established to improve motor and neurocognitive development, as well as to provide genetic counseling. The objective of this work is to review the available information and reinforce the proposal to carry out an etiopathogenic approach for microcephaly diagnosis and management.
Collapse
Affiliation(s)
- Luis Eduardo Becerra-Solano
- Medical Research Unit in Reproductive Medicine, Unidad Médica de Alta Especialidad, Hospital de Gineco Obstetricia No. 4, "Luis Castelazo Ayala", Instituto Mexicano del Seguro Social, Río Magdalena 289, Level 6, Laboratory K, Colonia Tizapan San Ángel, Alcaldía Álvaro Obregón, C.P. 01090, Mexico City, Mexico
| | - Leovigildo Mateos-Sánchez
- Neonatal Intensive Care Unit, Unidad Médica de Alta Especialidad, Hospital de Gineco Obstetricia No. 4, "Luis Castelazo Ayala", Instituto Mexicano del Seguro Social, Río Magdalena 289, Mezzanine, Colonia Tizapan San Ángel, Alcaldía Álvaro Obregón, C.P. 01090, Mexico City, Mexico
| | - Eunice López-Muñoz
- Medical Research Unit in Reproductive Medicine, Unidad Médica de Alta Especialidad, Hospital de Gineco Obstetricia No. 4, "Luis Castelazo Ayala", Instituto Mexicano del Seguro Social, Río Magdalena 289, Level 6, Laboratory K, Colonia Tizapan San Ángel, Alcaldía Álvaro Obregón, C.P. 01090, Mexico City, Mexico.
| |
Collapse
|
15
|
Birt IA, Hagenauer MH, Clinton SM, Aydin C, Blandino P, Stead JD, Hilde KL, Meng F, Thompson RC, Khalil H, Stefanov A, Maras P, Zhou Z, Hebda-Bauer EK, Goldman D, Watson SJ, Akil H. Genetic Liability for Internalizing Versus Externalizing Behavior Manifests in the Developing and Adult Hippocampus: Insight From a Meta-analysis of Transcriptional Profiling Studies in a Selectively Bred Rat Model. Biol Psychiatry 2021; 89:339-355. [PMID: 32762937 PMCID: PMC7704921 DOI: 10.1016/j.biopsych.2020.05.024] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/27/2019] [Revised: 04/29/2020] [Accepted: 05/19/2020] [Indexed: 01/05/2023]
Abstract
BACKGROUND For more than 16 years, we have selectively bred rats for either high or low levels of exploratory activity within a novel environment. These bred high-responder (bHR) and bred low-responder (bLR) rats model temperamental extremes, exhibiting large differences in internalizing and externalizing behaviors relevant to mood and substance use disorders. METHODS We characterized persistent differences in gene expression related to bHR/bLR phenotype across development and adulthood in the hippocampus, a region critical for emotional regulation, by meta-analyzing 8 transcriptional profiling datasets (microarray and RNA sequencing) spanning 43 generations of selective breeding (postnatal day 7: n = 22; postnatal day 14: n = 49; postnatal day 21: n = 21; adult: n = 46; all male). We cross-referenced expression differences with exome sequencing within our colony to pinpoint candidates likely to mediate the effect of selective breeding on behavioral phenotype. The results were compared with hippocampal profiling from other bred rat models. RESULTS Genetic and transcriptional profiling results converged to implicate multiple candidate genes, including two previously associated with metabolism and mood: Trhr and Ucp2. Results also highlighted bHR/bLR functional differences in the hippocampus, including a network essential for neurodevelopmental programming, proliferation, and differentiation, centering on Bmp4 and Mki67. Finally, we observed differential expression related to microglial activation, which is important for synaptic pruning, including 2 genes within implicated chromosomal regions: C1qa and Mfge8. CONCLUSIONS These candidate genes and functional pathways may direct bHR/bLR rats along divergent developmental trajectories and promote a widely different reactivity to the environment.
Collapse
Affiliation(s)
- Isabelle A. Birt
- Molecular and Behavioral Neuroscience Institute, University of
Michigan, Ann Arbor, Michigan
| | - Megan H. Hagenauer
- Molecular and Behavioral Neuroscience Institute, University of
Michigan, Ann Arbor, Michigan
| | | | - Cigdem Aydin
- Molecular and Behavioral Neuroscience Institute, University of
Michigan, Ann Arbor, Michigan
| | - Peter Blandino
- Molecular and Behavioral Neuroscience Institute, University of
Michigan, Ann Arbor, Michigan
| | - John D.H. Stead
- Department of Neuroscience, Carleton University, Ottawa, Ontario,
Canada
| | - Kathryn L. Hilde
- Molecular and Behavioral Neuroscience Institute, University of
Michigan, Ann Arbor, Michigan
| | - Fan Meng
- Molecular and Behavioral Neuroscience Institute, University of
Michigan, Ann Arbor, Michigan
| | - Robert C. Thompson
- Molecular and Behavioral Neuroscience Institute, University of
Michigan, Ann Arbor, Michigan
| | - Huzefa Khalil
- Molecular and Behavioral Neuroscience Institute, University of
Michigan, Ann Arbor, Michigan
| | - Alex Stefanov
- Molecular and Behavioral Neuroscience Institute, University of
Michigan, Ann Arbor, Michigan
| | - Pamela Maras
- Molecular and Behavioral Neuroscience Institute, University of
Michigan, Ann Arbor, Michigan
| | - Zhifeng Zhou
- National Institute on Alcohol Abuse and Alcoholism, National
Institutes of Health, Bethesda, Maryland
| | - Elaine K. Hebda-Bauer
- Molecular and Behavioral Neuroscience Institute, University of
Michigan, Ann Arbor, Michigan
| | - David Goldman
- National Institute on Alcohol Abuse and Alcoholism, National
Institutes of Health, Bethesda, Maryland
| | - Stanley J. Watson
- Molecular and Behavioral Neuroscience Institute, University of
Michigan, Ann Arbor, Michigan
| | - Huda Akil
- Molecular and Behavioral Neuroscience Institute, University of
Michigan, Ann Arbor, Michigan
| |
Collapse
|
16
|
Kumar S, Umair Z, Kumar V, Kumar S, Lee U, Kim J. Foxd4l1.1 negatively regulates transcription of neural repressor ventx1.1 during neuroectoderm formation in Xenopus embryos. Sci Rep 2020; 10:16780. [PMID: 33033315 PMCID: PMC7545198 DOI: 10.1038/s41598-020-73662-4] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2020] [Accepted: 09/16/2020] [Indexed: 11/09/2022] Open
Abstract
Neuroectoderm formation is the first step in development of a proper nervous system for vertebrates. The developmental decision to form a non-neural ectoderm versus a neural one involves the regulation of BMP signaling, first reported many decades ago. However, the precise regulatory mechanism by which this is accomplished has not been fully elucidated, particularly for transcriptional regulation of certain key transcription factors. BMP4 inhibition is a required step in eliciting neuroectoderm from ectoderm and Foxd4l1.1 is one of the earliest neural genes highly expressed in the neuroectoderm and conserved across vertebrates, including humans. In this work, we focused on how Foxd4l1.1 downregulates the neural repressive pathway. Foxd4l1.1 inhibited BMP4/Smad1 signaling and triggered neuroectoderm formation in animal cap explants of Xenopus embryos. Foxd4l1.1 directly bound within the promoter of endogenous neural repressor ventx1.1 and inhibited ventx1.1 transcription. Foxd4l1.1 also physically interacted with Xbra in the nucleus and inhibited Xbra-induced ventx1.1 transcription. In addition, Foxd4l1.1 also reduced nuclear localization of Smad1 to inhibit Smad1-mediated ventx1.1 transcription. Foxd4l1.1 reduced the direct binding of Xbra and Smad1 on ventx1.1 promoter regions to block Xbra/Smad1-induced synergistic activation of ventx1.1 transcription. Collectively, Foxd4l1.1 negatively regulates transcription of a neural repressor ventx1.1 by multiple mechanisms in its exclusively occupied territory of neuroectoderm, and thus leading to primary neurogenesis. In conjunction with the results of our previous findings that ventx1.1 directly represses foxd4l1.1, the reciprocal repression of ventx1.1 and foxd4l1.1 is significant in at least in part specifying the mechanism for the non-neural versus neural ectoderm fate determination in Xenopus embryos.
Collapse
Affiliation(s)
- Shiv Kumar
- Department of Biochemistry, Institute of Cell Differentiation and Aging, College of Medicine, Hallym University, Gangwon-Do, Chuncheon, 24252, Republic of Korea
| | - Zobia Umair
- Department of Biochemistry, Institute of Cell Differentiation and Aging, College of Medicine, Hallym University, Gangwon-Do, Chuncheon, 24252, Republic of Korea
| | - Vijay Kumar
- Department of Biochemistry, Institute of Cell Differentiation and Aging, College of Medicine, Hallym University, Gangwon-Do, Chuncheon, 24252, Republic of Korea
| | - Santosh Kumar
- Department of Biochemistry, Institute of Cell Differentiation and Aging, College of Medicine, Hallym University, Gangwon-Do, Chuncheon, 24252, Republic of Korea
| | - Unjoo Lee
- Department of Electrical Engineering, Hallym University, Gangwon-Do, Chuncheon, 24252, Republic of Korea.
| | - Jaebong Kim
- Department of Biochemistry, Institute of Cell Differentiation and Aging, College of Medicine, Hallym University, Gangwon-Do, Chuncheon, 24252, Republic of Korea.
| |
Collapse
|
17
|
Rao C, Malaguti M, Mason JO, Lowell S. The transcription factor E2A drives neural differentiation in pluripotent cells. Development 2020; 147:dev184093. [PMID: 32487737 PMCID: PMC7328008 DOI: 10.1242/dev.184093] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2019] [Accepted: 05/26/2020] [Indexed: 12/21/2022]
Abstract
The intrinsic mechanisms that link extracellular signalling to the onset of neural differentiation are not well understood. In pluripotent mouse cells, BMP blocks entry into the neural lineage via transcriptional upregulation of inhibitor of differentiation (Id) factors. We have previously identified the major binding partner of Id proteins in pluripotent cells as the basic helix-loop-helix (bHLH) transcription factor (TF) E2A. Id1 can prevent E2A from forming heterodimers with bHLH TFs or from forming homodimers. Here, we show that overexpression of a forced E2A homodimer is sufficient to drive robust neural commitment in pluripotent cells, even under non-permissive conditions. Conversely, we find that E2A null cells display a defect in their neural differentiation capacity. E2A acts as an upstream activator of neural lineage genes, including Sox1 and Foxd4, and as a repressor of Nodal signalling. Our results suggest a crucial role for E2A in establishing neural lineage commitment in pluripotent cells.
Collapse
Affiliation(s)
- Chandrika Rao
- MRC Centre for Regenerative Medicine, Institute for Stem Cell Research, School of Biological Sciences, University of Edinburgh, 5 Little France Drive, Edinburgh EH16 4UU, UK
| | - Mattias Malaguti
- MRC Centre for Regenerative Medicine, Institute for Stem Cell Research, School of Biological Sciences, University of Edinburgh, 5 Little France Drive, Edinburgh EH16 4UU, UK
| | - John O Mason
- Centre for Discovery Brain Sciences, University of Edinburgh, 15 George Square, Edinburgh EH8 9XD, UK
- Simons Initiative for the Developing Brain, Hugh Robson Building, George Square, Edinburgh EH8 9XD, UK
| | - Sally Lowell
- MRC Centre for Regenerative Medicine, Institute for Stem Cell Research, School of Biological Sciences, University of Edinburgh, 5 Little France Drive, Edinburgh EH16 4UU, UK
| |
Collapse
|
18
|
Leibovich A, Edri T, Klein SL, Moody SA, Fainsod A. Natural size variation among embryos leads to the corresponding scaling in gene expression. Dev Biol 2020; 462:165-179. [PMID: 32259520 PMCID: PMC8073595 DOI: 10.1016/j.ydbio.2020.03.014] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2019] [Revised: 02/27/2020] [Accepted: 03/23/2020] [Indexed: 11/17/2022]
Abstract
Xenopus laevis frogs from laboratory stocks normally lay eggs exhibiting extensive size variability. We find that these initial size differences subsequently affect the size of the embryos prior to the onset of growth, and the size of tadpoles during the growth period. Even though these tadpoles differ in size, their tissues, organs, and structures always seem to be properly proportioned, i.e. they display static allometry. Initial axial patterning events in Xenopus occur in a spherical embryo, allowing easy documentation of their size-dependent features. We examined the size distribution of early Xenopus laevis embryos and measured diameters that differed by about 38% with a median of about 1.43 mm. This range of embryo sizes corresponds to about a 1.9-fold difference in surface area and a 2.6-fold difference in volume. We examined the relationship between embryo size and gene expression and observed a significant correlation between diameter and RNA content during gastrula stages. In addition, we investigated the expression levels of genes that pattern the mesoderm, induce the nervous system and mediate the progression of ectodermal cells to neural precursors in large and small embryos. We found that most of these factors were expressed at levels that scaled with the different embryo sizes and total embryo RNA content. In agreement with the changes in transcript levels, the expression domains in larger embryos increased proportionally with the increase in surface area, maintaining their relative expression domain size in relation to the total size of the embryo. Thus, our study identified a mechanism for adapting gene expression domains to embryo size by adjusting the transcript levels of the genes regulating mesoderm induction and patterning. In the neural plate, besides the scaling of the expression domains, we observed similar cell sizes and cell densities in small and large embryos suggesting that additional cell divisions took place in large embryos to compensate for the increased size. Our results show in detail the size variability among Xenopus laevis embryos and the transcriptional adaptation to scale gene expression with size. The observations further support the involvement of BMP/ADMP signaling in the scaling process.
Collapse
Affiliation(s)
- Avi Leibovich
- Department of Developmental Biology and Cancer Research, Institute for Medical Research Israel-Canada, Faculty of Medicine, The Hebrew University of Jerusalem, Israel
| | - Tamir Edri
- Department of Developmental Biology and Cancer Research, Institute for Medical Research Israel-Canada, Faculty of Medicine, The Hebrew University of Jerusalem, Israel
| | - Steven L Klein
- Department of Anatomy and Cell Biology, George Washington University School of Medicine and Health Sciences, USA
| | - Sally A Moody
- Department of Anatomy and Cell Biology, George Washington University School of Medicine and Health Sciences, USA
| | - Abraham Fainsod
- Department of Developmental Biology and Cancer Research, Institute for Medical Research Israel-Canada, Faculty of Medicine, The Hebrew University of Jerusalem, Israel.
| |
Collapse
|
19
|
Inomata C, Yuikawa T, Nakayama-Sadakiyo Y, Kobayashi K, Ikeda M, Chiba M, Konishi C, Ishioka A, Tsuda S, Yamasu K. Involvement of an Oct4-related PouV gene, pou5f3/pou2, in neurogenesis in the early neural plate of zebrafish embryos. Dev Biol 2020; 457:30-42. [DOI: 10.1016/j.ydbio.2019.09.002] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2019] [Revised: 09/09/2019] [Accepted: 09/10/2019] [Indexed: 01/03/2023]
|
20
|
Kameneva P, Adameyko I. Recent advances in our understanding of central and peripheral nervous system progenitors. Curr Opin Cell Biol 2019; 61:24-30. [PMID: 31369951 DOI: 10.1016/j.ceb.2019.07.003] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2019] [Revised: 06/25/2019] [Accepted: 07/02/2019] [Indexed: 10/26/2022]
Abstract
Several decades of intense research provided us with a grand framework describing the emergence of neurons in central (CNS) and peripheral (PNS) nervous systems. However, the specifics of molecular events and lineage control leading to a plethora of neuronal subtypes stayed largely unclear. Today, the advances in single cell omics, sample clearing and 3D-microscopy techniques, brain organoids, and synaptic connectivity tracing enabled systematic and unbiased understanding of neuronal diversity, development, circuitry and cell identity control. Novel technological advancements stimulated the transition from conceptual scheme of neuronal differentiation into precise maps of molecular events leading to the diversity of specific neuronal subtypes in relation to their locations and microenvironment. These high-resolution data opened a set of new questions including how transcriptional and epigenetics states control the proportionality of neuronal subpopulations or what are the evolutionary mechanisms of origin of different neuronal subtypes. In this review, we outline the most recent advancements in our understanding of how the neuronal diversity is generated in CNS and PNS and briefly address the challenges and questions arising in the field of neurogenesis.
Collapse
Affiliation(s)
- Polina Kameneva
- Department of Physiology and Pharmacology, Karolinska Institutet, 17177 Stockholm, Sweden
| | - Igor Adameyko
- Department of Physiology and Pharmacology, Karolinska Institutet, 17177 Stockholm, Sweden; Department of Molecular Neurosciences, Center for Brain Research, Medical University Vienna, 1090 Vienna, Austria.
| |
Collapse
|
21
|
Polevoy H, Gutkovich YE, Michaelov A, Volovik Y, Elkouby YM, Frank D. New roles for Wnt and BMP signaling in neural anteroposterior patterning. EMBO Rep 2019; 20:embr.201845842. [PMID: 30936121 DOI: 10.15252/embr.201845842] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2018] [Revised: 03/04/2019] [Accepted: 03/07/2019] [Indexed: 01/19/2023] Open
Abstract
During amphibian development, neural patterning occurs via a two-step process. Spemann's organizer secretes BMP antagonists that induce anterior neural tissue. A subsequent caudalizing step re-specifies anterior fated cells to posterior fates such as hindbrain and spinal cord. The neural patterning paradigm suggests that a canonical Wnt-signaling gradient acts along the anteroposterior axis to pattern the nervous system. Wnt activity is highest in the posterior, inducing spinal cord, at intermediate levels in the trunk, inducing hindbrain, and is lowest in anterior fated forebrain, while BMP-antagonist levels are constant along the axis. Our results in Xenopus laevis challenge this paradigm. We find that inhibition of canonical Wnt signaling or its downstream transcription factors eliminates hindbrain, but not spinal cord fates, an observation not compatible with a simple high-to-low Wnt gradient specifying all fates along the neural anteroposterior axis. Additionally, we find that BMP activity promotes posterior spinal cord cell fate formation in an FGF-dependent manner, while inhibiting hindbrain fates. These results suggest a need to re-evaluate the paradigms of neural anteroposterior pattern formation during vertebrate development.
Collapse
Affiliation(s)
- Hanna Polevoy
- Department of Biochemistry, Faculty of Medicine, The Rappaport Family Institute for Research in the Medical Sciences, Technion - Israel Institute of Technology, Haifa, Israel
| | - Yoni E Gutkovich
- Department of Biochemistry, Faculty of Medicine, The Rappaport Family Institute for Research in the Medical Sciences, Technion - Israel Institute of Technology, Haifa, Israel
| | - Ariel Michaelov
- Department of Biochemistry, Faculty of Medicine, The Rappaport Family Institute for Research in the Medical Sciences, Technion - Israel Institute of Technology, Haifa, Israel
| | - Yael Volovik
- Department of Biochemistry, Faculty of Medicine, The Rappaport Family Institute for Research in the Medical Sciences, Technion - Israel Institute of Technology, Haifa, Israel
| | - Yaniv M Elkouby
- Department of Biochemistry, Faculty of Medicine, The Rappaport Family Institute for Research in the Medical Sciences, Technion - Israel Institute of Technology, Haifa, Israel
| | - Dale Frank
- Department of Biochemistry, Faculty of Medicine, The Rappaport Family Institute for Research in the Medical Sciences, Technion - Israel Institute of Technology, Haifa, Israel
| |
Collapse
|
22
|
Durston AJ. Two Tier Hox Collinearity Mediates Vertebrate Axial Patterning. Front Cell Dev Biol 2018; 6:102. [PMID: 30234110 PMCID: PMC6131192 DOI: 10.3389/fcell.2018.00102] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2018] [Accepted: 08/10/2018] [Indexed: 12/04/2022] Open
Abstract
A two tier mechanism mediates Hox collinearity. Besides the familiar collinear chromatin modification within each Hox cluster (nanocollinearity), there is also a macrocollinearity tier. Individual Hox clusters and individual cells are coordinated and synchronized to generate multiscale (macro and nano) collinearity in the early vertebrate embryo. Macro-collinearity is mediated by three non-cell autonomous Hox–Hox interactions. These mediate temporal collinearity in early NOM (non-organizer mesoderm), time space translation where temporal collinearity is translated to spatial collinearity along the early embryo’s main body axis and neural transformation, where Hox expression is copied monospecifically from NOM mesoderm to overlying neurectoderm in the late gastrula. Unlike nanocollinearity, which is Hox cluster restricted, axial macrocollinearity extends into the head and EAD domains, thus covering the whole embryonic anterior-posterior (A-P) axis. EAD: extreme anterior domain, the only A-P axial domain anterior to the head. The whole time space translation mechanism interacts with A-P signaling pathways via “decision points,” separating different domains on the axis.
Collapse
Affiliation(s)
- Antony J Durston
- Faculty of Science, Institute of Biology Leiden, Leiden University, Leiden, Netherlands
| |
Collapse
|
23
|
Verrier L, Davidson L, Gierliński M, Dady A, Storey KG. Neural differentiation, selection and transcriptomic profiling of human neuromesodermal progenitor-like cells in vitro. Development 2018; 145:dev166215. [PMID: 29899136 PMCID: PMC6124542 DOI: 10.1242/dev.166215] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2018] [Accepted: 05/30/2018] [Indexed: 01/06/2023]
Abstract
Robust protocols for directed differentiation of human pluripotent cells are required to determine whether mechanisms operating in model organisms are relevant to our own development. Recent work in vertebrate embryos has identified neuromesodermal progenitors as a bipotent cell population that contributes to paraxial mesoderm and spinal cord. However, precise protocols for in vitro differentiation of human spinal cord progenitors are lacking. Informed by signalling in amniote embryos, we show here that transient dual-SMAD inhibition, together with retinoic acid (dSMADi-RA), provides rapid and reproducible induction of human spinal cord progenitors from neuromesodermal progenitor-like cells. Using CRISPR-Cas9 to engineer human embryonic stem cells with a GFP-reporter for neuromesodermal progenitor-associated gene Nkx1.2 we facilitate selection of this cell population. RNA-sequencing was then used to identify human and conserved neuromesodermal progenitor transcriptional signatures, to validate this differentiation protocol and to reveal new pathways/processes in human neural differentiation. This optimised protocol, novel reporter line and transcriptomic data are useful resources with which to dissect molecular mechanisms regulating human spinal cord generation and allow the scaling-up of distinct cell populations for global analyses, including proteomic, biochemical and chromatin interrogation.
Collapse
Affiliation(s)
- Laure Verrier
- Division of Cell and Developmental Biology, School of Life Sciences, University of Dundee, Dow Street, Dundee DD1 5EH, UK
| | - Lindsay Davidson
- Human Pluripotent Cell Facility, Division of Cell and Developmental Biology, School of Life Sciences, University of Dundee, Dow Street, Dundee DD1 5EH, UK
| | - Marek Gierliński
- Data analysis group, Division of Cell and Developmental Biology, School of Life Sciences, University of Dundee, Dow Street, Dundee DD1 5EH, UK
| | - Alwyn Dady
- Division of Cell and Developmental Biology, School of Life Sciences, University of Dundee, Dow Street, Dundee DD1 5EH, UK
| | - Kate G Storey
- Division of Cell and Developmental Biology, School of Life Sciences, University of Dundee, Dow Street, Dundee DD1 5EH, UK
| |
Collapse
|
24
|
Pla P, Monsoro-Burq AH. The neural border: Induction, specification and maturation of the territory that generates neural crest cells. Dev Biol 2018; 444 Suppl 1:S36-S46. [PMID: 29852131 DOI: 10.1016/j.ydbio.2018.05.018] [Citation(s) in RCA: 68] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2018] [Revised: 05/23/2018] [Accepted: 05/23/2018] [Indexed: 11/17/2022]
Abstract
The neural crest is induced at the edge between the neural plate and the nonneural ectoderm, in an area called the neural (plate) border, during gastrulation and neurulation. In recent years, many studies have explored how this domain is patterned, and how the neural crest is induced within this territory, that also participates to the prospective dorsal neural tube, the dorsalmost nonneural ectoderm, as well as placode derivatives in the anterior area. This review highlights the tissue interactions, the cell-cell signaling and the molecular mechanisms involved in this dynamic spatiotemporal patterning, resulting in the induction of the premigratory neural crest. Collectively, these studies allow building a complex neural border and early neural crest gene regulatory network, mostly composed by transcriptional regulations but also, more recently, including novel signaling interactions.
Collapse
Affiliation(s)
- Patrick Pla
- Univ. Paris Sud, Université Paris Saclay, CNRS UMR 3347, INSERM U1021, Centre Universitaire, 15, rue Georges Clémenceau, F-91405 Orsay, France; Institut Curie Research Division, PSL Research University, CNRS UMR 3347, INSERM U1021, F-91405 Orsay, France
| | - Anne H Monsoro-Burq
- Univ. Paris Sud, Université Paris Saclay, CNRS UMR 3347, INSERM U1021, Centre Universitaire, 15, rue Georges Clémenceau, F-91405 Orsay, France; Institut Curie Research Division, PSL Research University, CNRS UMR 3347, INSERM U1021, F-91405 Orsay, France; Institut Universitaire de France, F-75005, Paris.
| |
Collapse
|
25
|
Plouhinec JL, Medina-Ruiz S, Borday C, Bernard E, Vert JP, Eisen MB, Harland RM, Monsoro-Burq AH. A molecular atlas of the developing ectoderm defines neural, neural crest, placode, and nonneural progenitor identity in vertebrates. PLoS Biol 2017; 15:e2004045. [PMID: 29049289 PMCID: PMC5663519 DOI: 10.1371/journal.pbio.2004045] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2016] [Revised: 10/31/2017] [Accepted: 09/29/2017] [Indexed: 12/18/2022] Open
Abstract
During vertebrate neurulation, the embryonic ectoderm is patterned into lineage progenitors for neural plate, neural crest, placodes and epidermis. Here, we use Xenopus laevis embryos to analyze the spatial and temporal transcriptome of distinct ectodermal domains in the course of neurulation, during the establishment of cell lineages. In order to define the transcriptome of small groups of cells from a single germ layer and to retain spatial information, dorsal and ventral ectoderm was subdivided along the anterior-posterior and medial-lateral axes by microdissections. Principal component analysis on the transcriptomes of these ectoderm fragments primarily identifies embryonic axes and temporal dynamics. This provides a genetic code to define positional information of any ectoderm sample along the anterior-posterior and dorsal-ventral axes directly from its transcriptome. In parallel, we use nonnegative matrix factorization to predict enhanced gene expression maps onto early and mid-neurula embryos, and specific signatures for each ectoderm area. The clustering of spatial and temporal datasets allowed detection of multiple biologically relevant groups (e.g., Wnt signaling, neural crest development, sensory placode specification, ciliogenesis, germ layer specification). We provide an interactive network interface, EctoMap, for exploring synexpression relationships among genes expressed in the neurula, and suggest several strategies to use this comprehensive dataset to address questions in developmental biology as well as stem cell or cancer research. Vertebrate embryo germ layers become progressively regionalized by evolutionarily conserved molecular processes. Catching the early steps of this dynamic spatial cell diversification at the scale of the transcriptome was challenging, even with the advent of efficient RNA sequencing. We have microdissected complementary and defined areas of a single germ layer, the developing ectoderm, and explored how the transcriptome changes over time and space in the ectoderm during the differentiation of frog epidermis, neural plate, and neural crest. We have created EctoMap, a searchable interface using these regional transcriptomes, to predict the expression of the 31 thousand genes expressed in neurulae and their networks of co-expression, predictive of functional relationships. Through several examples, we illustrate how these data provide insights in development, cancer, evolution and stem cell biology.
Collapse
Affiliation(s)
- Jean-Louis Plouhinec
- Université Paris Sud, Université Paris Saclay, CNRS UMR 3347, INSERM U1021, Orsay, France
- Institut Curie Research Division, PSL Research University, CNRS UMR 3347, INSERM U1021, Orsay, France
- MINES ParisTech, PSL Research University, CBIO - Centre for Computational Biology, Paris, France
| | - Sofía Medina-Ruiz
- Department of Molecular and Cell Biology, Division of Genetics, Genomics and Development Biology, University of California, Berkeley, Berkeley, California, United States of America
| | - Caroline Borday
- Université Paris Sud, Université Paris Saclay, CNRS UMR 3347, INSERM U1021, Orsay, France
- Institut Curie Research Division, PSL Research University, CNRS UMR 3347, INSERM U1021, Orsay, France
| | - Elsa Bernard
- MINES ParisTech, PSL Research University, CBIO - Centre for Computational Biology, Paris, France
- Institut Curie, INSERM U900, Paris, France
- INSERM U900, Paris, France
| | - Jean-Philippe Vert
- MINES ParisTech, PSL Research University, CBIO - Centre for Computational Biology, Paris, France
- Institut Curie, INSERM U900, Paris, France
- INSERM U900, Paris, France
| | - Michael B. Eisen
- Department of Molecular and Cell Biology, Division of Genetics, Genomics and Development Biology, University of California, Berkeley, Berkeley, California, United States of America
- Howard Hughes Medical Institute, University of California, Berkeley, Berkeley, California, United States of America
| | - Richard M. Harland
- Department of Molecular and Cell Biology, Division of Genetics, Genomics and Development Biology, University of California, Berkeley, Berkeley, California, United States of America
| | - Anne H. Monsoro-Burq
- Université Paris Sud, Université Paris Saclay, CNRS UMR 3347, INSERM U1021, Orsay, France
- Institut Curie Research Division, PSL Research University, CNRS UMR 3347, INSERM U1021, Orsay, France
- Institut Universitaire de France, Paris, France
- * E-mail:
| |
Collapse
|
26
|
Maruyama-Koide Y, Mikawa S, Sasaki T, Sato K. Bone morphogenetic protein-4 and bone morphogenetic protein receptors expressions in the adult rat eye. Eur J Histochem 2017; 61:2797. [PMID: 29046048 PMCID: PMC5592339 DOI: 10.4081/ejh.2017.2797] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2017] [Revised: 06/22/2017] [Accepted: 06/23/2017] [Indexed: 11/22/2022] Open
Abstract
We investigated the expressions of bone morphogenetic protein-4 (BMP4) and its receptors, bone morphogenetic protein receptor IA (BMPRIA), bone morphogenetic protein receptor IB (BMPRIB) and bone morphogenetic protein receptor II (BMPRII) in the adult rat eye. Interesting differences in expression profile were observed between BMPRIA and BMPRIB in the retina. BMPRIA-like immunoreactivity (IR) was very intensely seen in the photoreceptor layer, while BMPRIB-IR was mainly observed in the other layers. In the cornea, BMP4, BMPRIA, BMPRIB and BMPRII-IRs were abundantly seen in the cell body of basal cells in the corneal epithelium, and endothelium. In the lens, BMP4, BMPRIA, BMPRIB and BMPRIIIRs were observed in epithelial cells, lens cortical fiber cells, however they were not seen in the capsule and the central region of the lens. In the iris and ciliary body, strong BMP4 and BMPRIB-IRs were observed in nonpigmented epithelium. These results suggest that different kinds of BMP signaling should be needed in different areas in the adult eye to keep the shapes, differentiation levels, and functions of various cells.
Collapse
|
27
|
Fang Y, Liu Z, Chen Z, Xu X, Xiao M, Yu Y, Zhang Y, Zhang X, Du Y, Jiang C, Zhao Y, Wang Y, Fan B, Terheyden-Keighley D, Liu Y, Shi L, Hui Y, Zhang X, Zhang B, Feng H, Ma L, Zhang Q, Jin G, Yang Y, Xiang B, Liu L, Zhang X. Smad5 acts as an intracellular pH messenger and maintains bioenergetic homeostasis. Cell Res 2017; 27:1083-1099. [PMID: 28675158 PMCID: PMC5587853 DOI: 10.1038/cr.2017.85] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2017] [Revised: 02/22/2017] [Accepted: 05/23/2017] [Indexed: 12/16/2022] Open
Abstract
Both environmental cues and intracellular bioenergetic states profoundly affect intracellular pH (pHi). How a cell responds to pHi changes to maintain bioenergetic homeostasis remains elusive. Here we show that Smad5, a well-characterized downstream component of bone morphogenetic protein (BMP) signaling responds to pHi changes. Cold, basic or hypertonic conditions increase pHi, which in turn dissociates protons from the charged amino acid clusters within the MH1 domain of Smad5, prompting its relocation from the nucleus to the cytoplasm. On the other hand, heat, acidic or hypotonic conditions decrease pHi, blocking the nuclear export of Smad5, and thus causing its nuclear accumulation. Active nucleocytoplasmic shuttling of Smad5 induced by environmental changes and pHi fluctuation is independent of BMP signaling, carboxyl terminus phosphorylation and Smad4. In addition, ablation of Smad5 causes chronic and irreversible dysregulation of cellular bioenergetic homeostasis and disrupted normal neural developmental processes as identified in a differentiation model of human pluripotent stem cells. Importantly, these metabolic and developmental deficits in Smad5-deficient cells could be rescued only by cytoplasmic Smad5. Cytoplasmic Smad5 physically interacts with hexokinase 1 and accelerates glycolysis. Together, our findings indicate that Smad5 acts as a pHi messenger and maintains the bioenergetic homeostasis of cells by regulating cytoplasmic metabolic machinery.
Collapse
Affiliation(s)
- Yujiang Fang
- Shanghai Tenth People's Hospital, and Neuroregeneration Key Laboratory of Shanghai Universities, Tongji University School of Medicine, Shanghai 200092, China
| | - Zhongliang Liu
- Shanghai Tenth People's Hospital, and Neuroregeneration Key Laboratory of Shanghai Universities, Tongji University School of Medicine, Shanghai 200092, China
| | - Zhenyu Chen
- Shanghai Tenth People's Hospital, and Neuroregeneration Key Laboratory of Shanghai Universities, Tongji University School of Medicine, Shanghai 200092, China
| | - Xiangjie Xu
- Shanghai Tenth People's Hospital, and Neuroregeneration Key Laboratory of Shanghai Universities, Tongji University School of Medicine, Shanghai 200092, China
| | - Mengtao Xiao
- China Novartis Institutes for BioMedical Research, Shanghai 201203, China
| | - Yanyan Yu
- China Novartis Institutes for BioMedical Research, Shanghai 201203, China
| | - Yuanyuan Zhang
- Shanghai Tenth People's Hospital, and Neuroregeneration Key Laboratory of Shanghai Universities, Tongji University School of Medicine, Shanghai 200092, China
| | - Xiaobai Zhang
- The School of Life Sciences and Technology, Tongji University, Shanghai 200092, China
| | - Yanhua Du
- The School of Life Sciences and Technology, Tongji University, Shanghai 200092, China
| | - Cizhong Jiang
- The School of Life Sciences and Technology, Tongji University, Shanghai 200092, China
| | - Yuzheng Zhao
- Synthetic Biology and Biotechnology Laboratory, State Key Laboratory of Bioreactor Engineering, Shanghai Collaborative Innovation Center for Biomanufacturing Technology, East China University of Science and Technology, Shanghai 200237, China
| | - Yiran Wang
- Shanghai Tenth People's Hospital, and Neuroregeneration Key Laboratory of Shanghai Universities, Tongji University School of Medicine, Shanghai 200092, China
| | - Beibei Fan
- Shanghai Tenth People's Hospital, and Neuroregeneration Key Laboratory of Shanghai Universities, Tongji University School of Medicine, Shanghai 200092, China
| | - Daniel Terheyden-Keighley
- Shanghai Tenth People's Hospital, and Neuroregeneration Key Laboratory of Shanghai Universities, Tongji University School of Medicine, Shanghai 200092, China
| | - Yang Liu
- Shanghai Tenth People's Hospital, and Neuroregeneration Key Laboratory of Shanghai Universities, Tongji University School of Medicine, Shanghai 200092, China
| | - Lei Shi
- Shanghai Tenth People's Hospital, and Neuroregeneration Key Laboratory of Shanghai Universities, Tongji University School of Medicine, Shanghai 200092, China
| | - Yi Hui
- Department of Anatomy and Neurobiology, the Jiangsu Key Laboratory of Neuroregeneration, Nantong University, Nantong, Jiangsu 226001, China
| | - Xin Zhang
- Department of Anatomy and Neurobiology, the Jiangsu Key Laboratory of Neuroregeneration, Nantong University, Nantong, Jiangsu 226001, China
| | - Bowen Zhang
- Shanghai Tenth People's Hospital, and Neuroregeneration Key Laboratory of Shanghai Universities, Tongji University School of Medicine, Shanghai 200092, China
| | - Hexi Feng
- Shanghai Tenth People's Hospital, and Neuroregeneration Key Laboratory of Shanghai Universities, Tongji University School of Medicine, Shanghai 200092, China
| | - Lin Ma
- Shanghai Tenth People's Hospital, and Neuroregeneration Key Laboratory of Shanghai Universities, Tongji University School of Medicine, Shanghai 200092, China
| | - Quanbin Zhang
- Shanghai Tenth People's Hospital, and Neuroregeneration Key Laboratory of Shanghai Universities, Tongji University School of Medicine, Shanghai 200092, China
| | - Guohua Jin
- Department of Anatomy and Neurobiology, the Jiangsu Key Laboratory of Neuroregeneration, Nantong University, Nantong, Jiangsu 226001, China
| | - Yi Yang
- Synthetic Biology and Biotechnology Laboratory, State Key Laboratory of Bioreactor Engineering, Shanghai Collaborative Innovation Center for Biomanufacturing Technology, East China University of Science and Technology, Shanghai 200237, China
| | - Bin Xiang
- China Novartis Institutes for BioMedical Research, Shanghai 201203, China
| | - Ling Liu
- Shanghai Tenth People's Hospital, and Neuroregeneration Key Laboratory of Shanghai Universities, Tongji University School of Medicine, Shanghai 200092, China
- Tongji University Advanced Institute of Translational Medicine, Shanghai 200092, China
| | - Xiaoqing Zhang
- Shanghai Tenth People's Hospital, and Neuroregeneration Key Laboratory of Shanghai Universities, Tongji University School of Medicine, Shanghai 200092, China
- Tongji University Advanced Institute of Translational Medicine, Shanghai 200092, China
- The Collaborative Innovation Center for Brain Science, Tongji University, Shanghai 200092, China
| |
Collapse
|
28
|
Brafman D, Willert K. Wnt/β-catenin signaling during early vertebrate neural development. Dev Neurobiol 2017; 77:1239-1259. [PMID: 28799266 DOI: 10.1002/dneu.22517] [Citation(s) in RCA: 63] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2017] [Revised: 07/24/2017] [Accepted: 08/09/2017] [Indexed: 12/29/2022]
Abstract
The vertebrate central nervous system (CNS) is comprised of vast number of distinct cell types arranged in a highly organized manner. This high degree of complexity is achieved by cellular communication, including direct cell-cell contact, cell-matrix interactions, and cell-growth factor signaling. Among the several developmental signals controlling the development of the CNS, Wnt proteins have emerged as particularly critical and, hence, have captivated the attention of many researchers. With Wnts' evolutionarily conserved function as primordial symmetry breaking signals, these proteins and their downstream effects are responsible for simultaneously establishing cellular diversity and tissue organization. With their expansive repertoire of secreted agonists and antagonists, cell surface receptors, signaling cascades and downstream biological effects, Wnts are ideally suited to control the complex processes underlying vertebrate neural development. In this review, we will describe the mechanisms by which Wnts exert their potent effects on cells and tissues and highlight the many roles of Wnt signaling during neural development, starting from the initial induction of the neural plate, the subsequent patterning along the embryonic axes, to the intricately organized structure of the CNS. © 2017 Wiley Periodicals, Inc. Develop Neurobiol 77: 1239-1259, 2017.
Collapse
Affiliation(s)
- David Brafman
- School of Biological and Health Systems Engineering, Arizona State University, Tempe, Arizona, 85287
| | - Karl Willert
- Department of Cellular & Molecular Medicine, University of California San Diego, La Jolla, CA, 92093-0695
| |
Collapse
|
29
|
Jagadeesan M, Khetan V, Mallipatna A. Genetic perspective of retinoblastoma: From present to future. Indian J Ophthalmol 2017; 64:332-6. [PMID: 27380971 PMCID: PMC4966369 DOI: 10.4103/0301-4738.185585] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
Retinoblastoma (RB) is the most common malignant intraocular tumor in children. In the last decade, basic research has led to a better understanding of events after two hits in RB susceptibility gene (RB1), molecular mechanism of tumor growth, the cell of origin of RB, etc. This would pave way to identify biomarkers and molecular targeted therapy for better treatment option in the future. Furthermore, improvement in molecular techniques has led to enhanced diagnostic methods for early diagnosis, genetic counseling, and prevention of the disease. This review will help to understand the essence of basic research work conducted in recent times and its implication in the management of RB in the future.
Collapse
Affiliation(s)
- Madhavan Jagadeesan
- Department of Genetics and Molecular Biology, Vasan Medical Research Trust, Chennai, Tamil Nadu; Dual Helix Genetic Diagnostics Private Limited, Chennai, Tamil Nadu, India
| | - Vikas Khetan
- Sri Bhagavan Mahavir Vitreo-Retinal Services, Medical Research Foundation, Sankara Nethralaya, Chennai, Tamil Nadu, India
| | | |
Collapse
|
30
|
Waqas M, Sun S, Xuan C, Fang Q, Zhang X, Islam IU, Qi J, Zhang S, Gao X, Tang M, Shi H, Li H, Chai R. Bone morphogenetic protein 4 promotes the survival and preserves the structure of flow-sorted Bhlhb5+ cochlear spiral ganglion neurons in vitro. Sci Rep 2017; 7:3506. [PMID: 28615657 PMCID: PMC5471210 DOI: 10.1038/s41598-017-03810-w] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2017] [Accepted: 05/04/2017] [Indexed: 01/22/2023] Open
Abstract
SGNs are the primary auditory neurons, and damage or loss of SGNs leads to sensorineural hearing loss. BMP4 is a growth factor that belongs to the TGF-β superfamily and has been shown to play a key role during development, but little is known about its effect on postnatal cochlear SGNs in mice. In this study, we used the P3 Bhlhb5-cre/tdTomato transgenic mouse model and FACS to isolate a pure population of Bhlhb5+ SGNs. We found that BMP4 significantly promoted SGN survival after 7 days of culture. We observed fewer apoptotic cells and decreased expression of pro-apoptotic marker genes after BMP4 treatment. We also found that BMP4 promoted monopolar neurite outgrowth of isolated SGNs, and high concentrations of BMP4 preserved the number and the length of neurites in the explant culture of the modiolus harboring the SGNs. We showed that high concentration of BMP4 enhanced neurite growth as determined by the higher average number of filopodia and the larger area of the growth cone. Finally, we found that high concentrations of BMP4 significantly elevated the synapse density of SGNs in explant culture. Thus, our findings suggest that BMP4 has the potential to promote the survival and preserve the structure of SGNs.
Collapse
Affiliation(s)
- Muhammad Waqas
- Key Laboratory for Developmental Genes and Human Disease, Ministry of Education, Institute of Life Sciences, Southeast University, Nanjing, 210096, China.,Department of Otolaryngology Head and Neck Surgery, Zhongda Hospital, Southeast University, Nanjing, 210096, China.,Department of Biotechnology, Federal Urdu University of Arts, Science and Technology, Gulshan-e-Iqbal campus, Karachi, Pakistan
| | - Shan Sun
- Department of Otorhinolaryngology, Affiliated Eye and ENT Hospital, State Key Laboratory of Medical Neurobiology, Fudan University, Shanghai, 200031, China.,Key Laboratory of Hearing Medicine of NHFPC, Shanghai, 200031, China
| | - Chuanyin Xuan
- Key Laboratory for Developmental Genes and Human Disease, Ministry of Education, Institute of Life Sciences, Southeast University, Nanjing, 210096, China.,Department of Otolaryngology Head and Neck Surgery, Zhongda Hospital, Southeast University, Nanjing, 210096, China.,Co-innovation Center of Neuroregeneration, Nantong University, Nantong, 226001, China
| | - Qiaojun Fang
- Key Laboratory for Developmental Genes and Human Disease, Ministry of Education, Institute of Life Sciences, Southeast University, Nanjing, 210096, China.,Department of Otolaryngology Head and Neck Surgery, Zhongda Hospital, Southeast University, Nanjing, 210096, China.,Co-innovation Center of Neuroregeneration, Nantong University, Nantong, 226001, China
| | - Xiaoli Zhang
- Department of Otolaryngology, Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, 210008, China
| | - Irum-Us Islam
- Department of Biotechnology, Federal Urdu University of Arts, Science and Technology, Gulshan-e-Iqbal campus, Karachi, Pakistan
| | - Jieyu Qi
- Key Laboratory for Developmental Genes and Human Disease, Ministry of Education, Institute of Life Sciences, Southeast University, Nanjing, 210096, China.,Department of Otolaryngology Head and Neck Surgery, Zhongda Hospital, Southeast University, Nanjing, 210096, China.,Co-innovation Center of Neuroregeneration, Nantong University, Nantong, 226001, China
| | - Shasha Zhang
- Key Laboratory for Developmental Genes and Human Disease, Ministry of Education, Institute of Life Sciences, Southeast University, Nanjing, 210096, China.,Department of Otolaryngology Head and Neck Surgery, Zhongda Hospital, Southeast University, Nanjing, 210096, China.,Co-innovation Center of Neuroregeneration, Nantong University, Nantong, 226001, China
| | - Xia Gao
- Department of Otolaryngology, Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, 210008, China
| | - Mingliang Tang
- Key Laboratory for Developmental Genes and Human Disease, Ministry of Education, Institute of Life Sciences, Southeast University, Nanjing, 210096, China.,Department of Otolaryngology Head and Neck Surgery, Zhongda Hospital, Southeast University, Nanjing, 210096, China.,Co-innovation Center of Neuroregeneration, Nantong University, Nantong, 226001, China
| | - Haibo Shi
- Department of Otorhinolaryngology Head & Neck Surgery, The Sixth People's Hospital Affiliated to Shanghai Jiao Tong University, Shanghai, 200233, China.
| | - Huawei Li
- Department of Otorhinolaryngology, Affiliated Eye and ENT Hospital, State Key Laboratory of Medical Neurobiology, Fudan University, Shanghai, 200031, China. .,Key Laboratory of Hearing Medicine of NHFPC, Shanghai, 200031, China. .,Institutes of Biomedical Sciences, Fudan University, Shanghai, 200032, China. .,Shanghai Engineering Research Centre of Cochlear Implants, Shanghai, 200031, China. .,The Institutes of Brain Science and the Collaborative Innovation Center for Brain Science, Fudan University, Shanghai, 200032, China.
| | - Renjie Chai
- Key Laboratory for Developmental Genes and Human Disease, Ministry of Education, Institute of Life Sciences, Southeast University, Nanjing, 210096, China. .,Department of Otolaryngology Head and Neck Surgery, Zhongda Hospital, Southeast University, Nanjing, 210096, China. .,Co-innovation Center of Neuroregeneration, Nantong University, Nantong, 226001, China.
| |
Collapse
|
31
|
De Almeida I, Oliveira NMM, Randall RA, Hill CS, McCoy JM, Stern CD. Calreticulin is a secreted BMP antagonist, expressed in Hensen's node during neural induction. Dev Biol 2017; 421:161-170. [PMID: 27919666 PMCID: PMC5231319 DOI: 10.1016/j.ydbio.2016.12.001] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2016] [Revised: 11/29/2016] [Accepted: 12/01/2016] [Indexed: 11/27/2022]
Abstract
Hensen's node is the "organizer" of the avian and mammalian early embryo. It has many functions, including neural induction and patterning of the ectoderm and mesoderm. Some of the signals responsible for these activities are known but these do not explain the full complexity of organizer activity. Here we undertake a functional screen to discover new secreted factors expressed by the node at this time of development. Using a Signal Sequence Trap in yeast, we identify several candidates. Here we focus on Calreticulin. We show that in addition to its known functions in intracellular Calcium regulation and protein folding, Calreticulin is secreted, it can bind to BMP4 and act as a BMP antagonist in vivo and in vitro. Calreticulin is not sufficient to account for all organizer functions but may contribute to the complexity of its activity.
Collapse
Affiliation(s)
- Irene De Almeida
- Department of Cell & Developmental Biology, University College London, Gower Street, London WC1E 6BT, UK
| | - Nidia M M Oliveira
- Department of Cell & Developmental Biology, University College London, Gower Street, London WC1E 6BT, UK
| | | | | | | | - Claudio D Stern
- Department of Cell & Developmental Biology, University College London, Gower Street, London WC1E 6BT, UK
| |
Collapse
|
32
|
Cho GS, Park DS, Choi SC, Han JK. Tbx2 regulates anterior neural specification by repressing FGF signaling pathway. Dev Biol 2016; 421:183-193. [PMID: 27913219 DOI: 10.1016/j.ydbio.2016.11.020] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2016] [Revised: 11/28/2016] [Accepted: 11/28/2016] [Indexed: 10/20/2022]
Abstract
During early embryogenesis, FGF signals regulate the antero-posterior (AP) patterning of the neural plate by promoting posterior cell fates. In particular, BMP signal-mediated attenuation of FGF pathway plays a critical role in the determination of the anterior neural region. Here we show that Tbx2, a T-box transcriptional repressor regulates anterior neural specification by suppressing FGF8 signaling pathway in Xenopus embryo. Tbx2 is expressed in the anterior edge of the neural plate in early neurulae. Overexpression and knockdown of Tbx2 induce expansion and reduction in the expression of anterior neural markers, respectively. It also suppresses FGF8-induced ERK phosphorylation and neural caudalization. Tbx2, which is a target gene of BMP signal, down-regulates FGF8 signaling by inhibiting the expression of Flrt3, a positive regulator of this pathway. We found that Tbx2 binds directly to the T-box element located in the promoter region of Flrt3 gene, thereby interfering with the activity of the promoter. Consistently, Tbx2 augmentation of anterior neural formation is inhibited by co-expression of Flrt3. Furthermore, disruption of the anterior-most structures such as eyes in Tbx2-depleted embryos can be rescued by inhibition of Flrt3 function or FGF signaling. Taken together, our results suggest that Tbx2 mediates BMP signal to down-regulate FGF signaling pathway by repressing Flrt3 expression for anterior tissue formation.
Collapse
Affiliation(s)
- Gun-Sik Cho
- Division of Molecular and Life Sciences, Pohang University of Science and Technology, San 31, Hyoja-dong, Nam-gu, Pohang, Kyungbuk 790-784, Republic of Korea
| | - Dong-Seok Park
- Department of Biomedical Sciences, University of Ulsan College of Medicine, Pungnap-Dong, Songpa-Gu, Seoul 138-736, Republic of Korea
| | - Sun-Cheol Choi
- Department of Biomedical Sciences, University of Ulsan College of Medicine, Pungnap-Dong, Songpa-Gu, Seoul 138-736, Republic of Korea.
| | - Jin-Kwan Han
- Division of Molecular and Life Sciences, Pohang University of Science and Technology, San 31, Hyoja-dong, Nam-gu, Pohang, Kyungbuk 790-784, Republic of Korea.
| |
Collapse
|
33
|
Yoshida M, Kajikawa E, Kurokawa D, Noro M, Iwai T, Yonemura S, Kobayashi K, Kiyonari H, Aizawa S. Conserved and divergent expression patterns of markers of axial development in reptilian embryos: Chinese soft-shell turtle and Madagascar ground gecko. Dev Biol 2016; 415:122-142. [DOI: 10.1016/j.ydbio.2016.05.005] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2016] [Revised: 05/06/2016] [Accepted: 05/06/2016] [Indexed: 12/18/2022]
|
34
|
Luehders K, Sasai N, Davaapil H, Kurosawa-Yoshida M, Hiura H, Brah T, Ohnuma SI. The small leucine-rich repeat secreted protein Asporin induces eyes in Xenopus embryos through the IGF signalling pathway. Development 2016; 142:3351-61. [PMID: 26443635 DOI: 10.1242/dev.124438] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Small leucine-rich repeat proteoglycan (SLRP) family proteins play important roles in a number of biological events. Here, we demonstrate that the SLRP family member Asporin (ASPN) plays a crucial role in the early stages of eye development in Xenopus embryos. During embryogenesis, ASPN is broadly expressed in the neuroectoderm of the embryo. Overexpression of ASPN causes the induction of ectopic eyes. By contrast, blocking ASPN function with a morpholino oligonucleotide (ASPN-MO) inhibits eye formation, indicating that ASPN is an essential factor for eye development. Detailed molecular analyses revealed that ASPN interacts with insulin growth factor receptor (IGFR) and is essential for activating the IGF receptor-mediated intracellular signalling pathway. Moreover, ASPN perturbed the Wnt, BMP and Activin signalling pathways, suggesting that ASPN thereby creates a favourable environment in which the IGF signal can dominate. ASPN is thus a novel secreted molecule essential for eye induction through the coordination of multiple signalling pathways.
Collapse
Affiliation(s)
- Kristin Luehders
- Ocular Biology and Therapeutic unit (ORBIT), Institute of Ophthalmology, University College London, 11-43 Bath Street, London EC1V 9EL, UK
| | - Noriaki Sasai
- Ocular Biology and Therapeutic unit (ORBIT), Institute of Ophthalmology, University College London, 11-43 Bath Street, London EC1V 9EL, UK Developmental Biomedical Science, Graduate School of Biological Sciences, Nara Institute of Science and Technology (NAIST), 8916-5, Takayama-cho, Ikoma 630-0192, Japan
| | - Hongorzul Davaapil
- Ocular Biology and Therapeutic unit (ORBIT), Institute of Ophthalmology, University College London, 11-43 Bath Street, London EC1V 9EL, UK
| | - Maiko Kurosawa-Yoshida
- Department of Oncology, The Hutchison/MRC Research Centre, University of Cambridge, Hills Road, Cambridge CB2 2XZ, UK
| | - Hitoshi Hiura
- Ocular Biology and Therapeutic unit (ORBIT), Institute of Ophthalmology, University College London, 11-43 Bath Street, London EC1V 9EL, UK
| | - Tara Brah
- Ocular Biology and Therapeutic unit (ORBIT), Institute of Ophthalmology, University College London, 11-43 Bath Street, London EC1V 9EL, UK
| | - Shin-ichi Ohnuma
- Ocular Biology and Therapeutic unit (ORBIT), Institute of Ophthalmology, University College London, 11-43 Bath Street, London EC1V 9EL, UK Department of Oncology, The Hutchison/MRC Research Centre, University of Cambridge, Hills Road, Cambridge CB2 2XZ, UK
| |
Collapse
|
35
|
Durand BC. Stem cell-like Xenopus Embryonic Explants to Study Early Neural Developmental Features In Vitro and In Vivo. J Vis Exp 2016:e53474. [PMID: 26863402 DOI: 10.3791/53474] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
Understanding the genetic programs underlying neural development is an important goal of developmental and stem cell biology. In the amphibian blastula, cells from the roof of the blastocoel are pluripotent. These cells can be isolated, and programmed to generate various tissues through manipulation of genes expression or induction by morphogens. In this manuscript protocols are described for the use of Xenopus laevis blastocoel roof explants as an assay system to investigate key in vivo and in vitro features of early neural development. These protocols allow the investigation of fate acquisition, cell migration behaviors, and cell autonomous and non-autonomous properties. The blastocoel roof explants can be cultured in a serum-free defined medium and grafted into host embryos. This transplantation into an embryo allows the investigation of the long-term lineage commitment, the inductive properties, and the behavior of transplanted cells in vivo. These assays can be exploited to investigate molecular mechanisms, cellular processes and gene regulatory networks underlying neural development. In the context of regenerative medicine, these assays provide a means to generate neural-derived cell types in vitro that could be used in drug screening.
Collapse
Affiliation(s)
- Beatrice C Durand
- Institut Curie; UMR 3387, CNRS; PSL Research University; Université Paris-Sud;
| |
Collapse
|
36
|
Abstract
Anterior-posterior (A-P) patterning of the vertebrate main body axis regulated by timing. Anterior structures are specified early, posterior late. (1) Timing involves timed decision points as emphasised by the Wnt studies of Sokol and colleagues. It also involves complex timers, where large parts of the axis are patterned sequentially by a common upstream mechanism (articles by Durston et al., Mullins et al., Oates et al.,). (2) A gastrula BMP-anti BMP dependent time-space translation (TST) mechanism was demonstrated for the trunk section of the axis (Durston). (3) Thisses' studies emphasise the importance of BMP-anti BMP and the organiser inducing factor nodal for A-P patterning. (4) Meinhardt's interesting studies on the organiser and A-P patterning are reviewed in relation to TST. (5) Mullins' investigations show that anti-BMP dependent TST starts earlier (at the blastula stage) and extends further anteriorly (to the anterior head). Sive's studies imply it may extend further still to the "extreme anterior domain" (EAD). (6) The somitogenesis timer (clock) is presented. Stern's and Oates' findings are discussed. (7) Relations between somitogenesis and axial TST are discussed. (8) Relations of classical axial patterning pathways to TST decision points and somitogenesis are inventarised. In conclusion, all of these findings point to an integral BMP-anti BMP dependent A-P TST mechanism, running from cement gland in the EAD, Six3 and the anterior tip of the forebrain at blastula stages to Hox13 and the tip of the tail by the mid neurula stage. TST acts via sequential timed transitions between ventral (unstable, timed) and dorsal (stabilised) states. In the trunk-tail, the timer is thought to be Hox temporal collinearity and TST depends on Hox function. In the head, TST is under investigation. The somitogenesis clock is upstream of the TST timer, providing precision in the posterior part of the axis at least. Classical A-P signalling pathways: retinoids, FGFs and Wnts, change behaviour at functional decision points on the axis.
Collapse
|
37
|
Gouti M, Metzis V, Briscoe J. The route to spinal cord cell types: a tale of signals and switches. Trends Genet 2015; 31:282-9. [PMID: 25823696 DOI: 10.1016/j.tig.2015.03.001] [Citation(s) in RCA: 72] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2015] [Revised: 02/28/2015] [Accepted: 03/02/2015] [Indexed: 01/20/2023]
Abstract
Understanding the mechanisms that control induction and elaboration of the vertebrate central nervous system (CNS) requires an analysis of the extrinsic signals and downstream transcriptional networks that assign cell fates in the correct space and time. We focus on the generation and patterning of the spinal cord. We summarize evidence that the origin of the spinal cord is distinct from the anterior regions of the CNS. We discuss how this affects the gene regulatory networks and cell state transitions that specify spinal cord cell subtypes, and we highlight how the timing of extracellular signals and dynamic control of transcriptional networks contribute to the correct spatiotemporal generation of different neural cell types.
Collapse
Affiliation(s)
- Mina Gouti
- The Francis Crick Institute, Mill Hill Laboratory, The Ridgeway, Mill Hill, London, NW7 1AA, UK
| | - Vicki Metzis
- The Francis Crick Institute, Mill Hill Laboratory, The Ridgeway, Mill Hill, London, NW7 1AA, UK
| | - James Briscoe
- The Francis Crick Institute, Mill Hill Laboratory, The Ridgeway, Mill Hill, London, NW7 1AA, UK.
| |
Collapse
|
38
|
Wong KA, Trembley M, Abd Wahab S, Viczian AS. Efficient retina formation requires suppression of both Activin and BMP signaling pathways in pluripotent cells. Biol Open 2015; 4:573-83. [PMID: 25750435 PMCID: PMC4400599 DOI: 10.1242/bio.20149977] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
Abstract
Retina formation requires the correct spatiotemporal patterning of key regulatory factors. While it is known that repression of several signaling pathways lead to specification of retinal fates, addition of only Noggin, a known BMP antagonist, can convert pluripotent Xenopus laevis animal cap cells to functional retinal cells. The aim of this study is to determine the intracellular molecular events that occur during this conversion. Surprisingly, blocking BMP signaling alone failed to mimic Noggin treatment. Overexpressing Noggin in pluripotent cells resulted in a concentration-dependent suppression of both Smad1 and Smad2 phosphorylation, which act downstream of BMP and Activin signaling, respectively. This caused a decrease in downstream targets: endothelial marker, xk81, and mesodermal marker, xbra. We treated pluripotent cells with dominant-negative receptors or the chemical inhibitors, dorsomorphin and SB431542, which each target either the BMP or Activin signaling pathway. We determined the effect of these treatments on retina formation using the Animal Cap Transplant (ACT) assay; in which treated pluripotent cells were transplanted into the eye field of host embryos. We found that inhibition of Activin signaling, in the presence of BMP signaling inhibition, promotes efficient retinal specification in Xenopus tissue, mimicking the affect of adding Noggin alone. In whole embryos, we found that the eye field marker, rax, expanded when adding both dominant-negative Smad1 and Smad2, as did treating the cells with both dorsomorphin and SB431542. Future studies could translate these findings to a mammalian culture assay, in order to more efficiently produce retinal cells in culture.
Collapse
Affiliation(s)
- Kimberly A Wong
- Department of Neuroscience and Physiology, SUNY Upstate Medical University, Syracuse, NY 13210, USA The Center for Vision Research, SUNY Eye Institute, Upstate Medical University, Syracuse, NY 13210, USA
| | - Michael Trembley
- Department of Pharmacology and Physiology, Aab Cardiovascular Research Institute, University of Rochester School of Medicine and Dentistry, Rochester, NY 14642, USA
| | - Syafiq Abd Wahab
- Department of Molecular Biology, Weill Cornell Graduate School of Medical Sciences, New York, NY 10021, USA
| | - Andrea S Viczian
- Department of Ophthalmology, SUNY Upstate Medical University, Syracuse, NY 13210, USA Department of Neuroscience and Physiology, SUNY Upstate Medical University, Syracuse, NY 13210, USA The Center for Vision Research, SUNY Eye Institute, Upstate Medical University, Syracuse, NY 13210, USA
| |
Collapse
|
39
|
Juraver-Geslin HA, Durand BC. Early development of the neural plate: new roles for apoptosis and for one of its main effectors caspase-3. Genesis 2015; 53:203-24. [PMID: 25619400 DOI: 10.1002/dvg.22844] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2014] [Accepted: 01/07/2015] [Indexed: 12/12/2022]
Abstract
Despite its tremendous complexity, the vertebrate nervous system emerges from a homogenous layer of neuroepithelial cells, the neural plate. Its formation relies on the time- and space-controlled progression of developmental programs. Apoptosis is a biological process that removes superfluous and potentially dangerous cells and is implemented through the activation of a molecular pathway conserved during evolution. Apoptosis and an unconventional function of one of its main effectors, caspase-3, contribute to the patterning and growth of the neuroepithelium. Little is known about the intrinsic and extrinsic cues controlling activities of the apoptotic machinery during development. The BarH-like (Barhl) proteins are homeodomain-containing transcription factors. The observations in Caenorhabditis elegans, Xenopus, and mice document that Barhl proteins act in cell survival and as cell type-specific regulators of a caspase-3 function that limits neural progenitor proliferation. In this review, we discuss the roles and regulatory modes of the apoptotic machinery in the development of the neural plate. We focus on the Barhl2, the Sonic Hedgehog, and the Wnt pathways and their activities in neural progenitor survival and proliferation.
Collapse
Affiliation(s)
- Hugo A Juraver-Geslin
- Department of Basic Science, Craniofacial Biology, College of Dentistry, New York University, New York, New York
| | | |
Collapse
|
40
|
Herhaus L, Al-Salihi MA, Dingwell KS, Cummins TD, Wasmus L, Vogt J, Ewan R, Bruce D, Macartney T, Weidlich S, Smith JC, Sapkota GP. USP15 targets ALK3/BMPR1A for deubiquitylation to enhance bone morphogenetic protein signalling. Open Biol 2015; 4:140065. [PMID: 24850914 PMCID: PMC4042855 DOI: 10.1098/rsob.140065] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023] Open
Abstract
Protein kinase ALK3/BMPR1A mediates bone morphogenetic protein (BMP) signalling through phosphorylation and activation of SMADs 1/5/8. SMAD6, a transcriptional target of BMP, negatively regulates the BMP pathway by recruiting E3 ubiquitin ligases and targeting ALK3 for ubiquitin-mediated degradation. Here, we identify a deubiquitylating enzyme USP15 as an interactor of SMAD6 and ALK3. We show that USP15 enhances BMP-induced phosphorylation of SMAD1 by interacting with and deubiquitylating ALK3. RNAi-mediated depletion of USP15 increases ALK3 K48-linked polyubiquitylation, and reduces both BMP-induced SMAD1 phosphorylation and transcription of BMP target genes. We also show that loss of USP15 expression from mouse myoblast cells inhibits BMP-induced osteoblast differentiation. Furthermore, USP15 modulates BMP-induced phosphorylation of SMAD1 and transcription during Xenopus embryogenesis.
Collapse
Affiliation(s)
- Lina Herhaus
- Medical Research Council Protein Phosphorylation and Ubiquitylation Unit, University of Dundee, Dow St., Dundee DD1 5EH, UK
| | - Mazin A Al-Salihi
- Medical Research Council Protein Phosphorylation and Ubiquitylation Unit, University of Dundee, Dow St., Dundee DD1 5EH, UK
| | - Kevin S Dingwell
- Division of Systems Biology, MRC National Institute for Medical Research, The Ridgeway, Mill Hill NW7 1AA, UK
| | - Timothy D Cummins
- Medical Research Council Protein Phosphorylation and Ubiquitylation Unit, University of Dundee, Dow St., Dundee DD1 5EH, UK
| | - Lize Wasmus
- Medical Research Council Protein Phosphorylation and Ubiquitylation Unit, University of Dundee, Dow St., Dundee DD1 5EH, UK
| | - Janis Vogt
- Medical Research Council Protein Phosphorylation and Ubiquitylation Unit, University of Dundee, Dow St., Dundee DD1 5EH, UK
| | - Richard Ewan
- Medical Research Council Protein Phosphorylation and Ubiquitylation Unit, University of Dundee, Dow St., Dundee DD1 5EH, UK
| | - David Bruce
- Medical Research Council Protein Phosphorylation and Ubiquitylation Unit, University of Dundee, Dow St., Dundee DD1 5EH, UK
| | - Thomas Macartney
- Medical Research Council Protein Phosphorylation and Ubiquitylation Unit, University of Dundee, Dow St., Dundee DD1 5EH, UK
| | - Simone Weidlich
- Medical Research Council Protein Phosphorylation and Ubiquitylation Unit, University of Dundee, Dow St., Dundee DD1 5EH, UK
| | - James C Smith
- Division of Systems Biology, MRC National Institute for Medical Research, The Ridgeway, Mill Hill NW7 1AA, UK
| | - Gopal P Sapkota
- Medical Research Council Protein Phosphorylation and Ubiquitylation Unit, University of Dundee, Dow St., Dundee DD1 5EH, UK
| |
Collapse
|
41
|
Watanabe H, Kuhn A, Fushiki M, Agata K, Özbek S, Fujisawa T, Holstein TW. Sequential actions of β-catenin and Bmp pattern the oral nerve net in Nematostella vectensis. Nat Commun 2014; 5:5536. [PMID: 25534229 PMCID: PMC4284808 DOI: 10.1038/ncomms6536] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2014] [Accepted: 10/10/2014] [Indexed: 12/16/2022] Open
Abstract
Animal evolution is closely linked to the emergence of the nervous system. At present it is unknown how the basic mechanisms of neural induction and formation of central nervous systems evolved. We addressed this question in Nematostella vectensis, a member of cnidarians, the ancient sister group of bilaterians. We found that β-catenin signalling is crucial for the early induction of the embryonic nervous system. β-Catenin activity at the blastopore induces specific neurogenic genes required for development of the oral nervous system. β-Catenin signalling induces also Bmp signalling, which, at later larval stages, becomes indispensible for the maintenance and asymmetric patterning of the oral nervous system along the primary and secondary (directive) axes. We hypothesize that the consecutive and functionally linked involvement of β-catenin and Bmp signalling in the formation of the cnidarian oral nervous system reflects an ancestral mechanism that evolved before the cnidarian/bilaterian split. The bilaterian central nervous system is thought to have evolved from a cnidarian-like ancestor, but the mechanisms of neural induction in cnidarians are largely unknown. Here the authors study the cnidarian Nematostella vectensis and show that β-catenin signalling is crucial for the early induction of its embryonic nervous system, suggesting evolutionary roots for this pathway.
Collapse
Affiliation(s)
- Hiroshi Watanabe
- Department of Molecular Evolution and Genomics, Centre for Organismal Studies (COS), Heidelberg University, Im Neuenheimer Feld 329, 69120 Heidelberg, Germany
| | - Anne Kuhn
- Department of Molecular Evolution and Genomics, Centre for Organismal Studies (COS), Heidelberg University, Im Neuenheimer Feld 329, 69120 Heidelberg, Germany
| | - Manami Fushiki
- 1] Department of Molecular Evolution and Genomics, Centre for Organismal Studies (COS), Heidelberg University, Im Neuenheimer Feld 329, 69120 Heidelberg, Germany [2] Department of Biophysics, Graduate School of Science, Kyoto University, Kitashirakawa-Oiwake, Sakyo-ku, Kyoto 606-8502, Japan
| | - Kiyokazu Agata
- Department of Biophysics, Graduate School of Science, Kyoto University, Kitashirakawa-Oiwake, Sakyo-ku, Kyoto 606-8502, Japan
| | - Suat Özbek
- Department of Molecular Evolution and Genomics, Centre for Organismal Studies (COS), Heidelberg University, Im Neuenheimer Feld 329, 69120 Heidelberg, Germany
| | - Toshitaka Fujisawa
- 1] Department of Molecular Evolution and Genomics, Centre for Organismal Studies (COS), Heidelberg University, Im Neuenheimer Feld 329, 69120 Heidelberg, Germany [2] Center for the Promotion of Integrated Sciences, The Graduate University for Advanced Studies, Higashiyama 5-1, Myodaiji, Okazaki 444-8585, Japan
| | - Thomas W Holstein
- Department of Molecular Evolution and Genomics, Centre for Organismal Studies (COS), Heidelberg University, Im Neuenheimer Feld 329, 69120 Heidelberg, Germany
| |
Collapse
|
42
|
An otx/nodal regulatory signature for posterior neural development in ascidians. PLoS Genet 2014; 10:e1004548. [PMID: 25121599 PMCID: PMC4133040 DOI: 10.1371/journal.pgen.1004548] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2014] [Accepted: 06/18/2014] [Indexed: 12/25/2022] Open
Abstract
In chordates, neural induction is the first step of a complex developmental process through which ectodermal cells acquire a neural identity. In ascidians, FGF-mediated neural induction occurs at the 32-cell stage in two blastomere pairs, precursors respectively of anterior and posterior neural tissue. We combined molecular embryology and cis-regulatory analysis to unveil in the ascidian Ciona intestinalis the remarkably simple proximal genetic network that controls posterior neural fate acquisition downstream of FGF. We report that the combined action of two direct FGF targets, the TGFβ factor Nodal, acting via Smad- and Fox-binding sites, and the transcription factor Otx suffices to trigger ascidian posterior neural tissue formation. Moreover, we found that this strategy is conserved in the distantly related ascidian Phallusia mammillata, in spite of extreme sequence divergence in the cis-regulatory sequences involved. Our results thus highlight that the modes of gene regulatory network evolution differ with the evolutionary scale considered. Within ascidians, developmental regulatory networks are remarkably robust to genome sequence divergence. Between ascidians and vertebrates, major fate determinants, such as Otx and Nodal, can be co-opted into different networks. Comparative developmental studies in ascidians with divergent genomes will thus uncover shared ascidian strategies, and contribute to a better understanding of the diversity of developmental strategies within chordates.
Collapse
|
43
|
Guo S, Mao X, He F, Liu H, Ming L. Activin A supplement in the hESCs culture enhances the endoderm differentiation efficiency. Cell Biol Int 2014; 38:849-56. [PMID: 24604611 DOI: 10.1002/cbin.10274] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2013] [Accepted: 02/21/2014] [Indexed: 01/25/2023]
Abstract
Activin A is a critical regulator in human embryonic stem cells (hESCs) maintenance and differentiation. Different concentrations of Activin A affect hESC maintenance and differentiation in different ways. A high concentration favors anterior primitive streak and gives rise to DE if the stimulation persists. hESCs were cultured with and without 10 ng/mL Activin A supplement respectively. The two groups of cells were differentiated into endoderm cells with 100 ng/mL Activin A and other reagents. Microarray-based DNA methylation was analyzed with the Infinium Human Methylation450 BeadChip on these two groups. There was a significant difference in endoderm differentiation efficiency (average efficiency: 71 vs. 58.5%, P < 0.05). hESCs cultured with Activin A supplement had an increased propensity to form definitive endoderm cells in response to Activin A and Wnt signal. Differentially Methylated Regions (DMRs) between these two groups were found. DMRs were related to the stem cell maintenance and gene regulation by peroxisome proliferators via PPARα, indicating that hESCs maintained with Activin A supplement had stronger "stemness."
Collapse
Affiliation(s)
- Shuren Guo
- Department of Clinical Laboratory, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450002, P.R. China
| | | | | | | | | |
Collapse
|
44
|
Vogt J, Dingwell KS, Herhaus L, Gourlay R, Macartney T, Campbell D, Smith JC, Sapkota GP. Protein associated with SMAD1 (PAWS1/FAM83G) is a substrate for type I bone morphogenetic protein receptors and modulates bone morphogenetic protein signalling. Open Biol 2014; 4:130210. [PMID: 24554596 PMCID: PMC3938053 DOI: 10.1098/rsob.130210] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Bone morphogenetic proteins (BMPs) control multiple cellular processes in embryos and adult tissues. BMPs signal through the activation of type I BMP receptor kinases, which then phosphorylate SMADs 1/5/8. In the canonical pathway, this triggers the association of these SMADs with SMAD4 and their translocation to the nucleus, where they regulate gene expression. BMPs can also signal independently of SMAD4, but this pathway is poorly understood. Here, we report the discovery and characterization of PAWS1/FAM83G as a novel SMAD1 interactor. PAWS1 forms a complex with SMAD1 in a SMAD4-independent manner, and BMP signalling induces the phosphorylation of PAWS1 through BMPR1A. The phosphorylation of PAWS1 in response to BMP is essential for activation of the SMAD4-independent BMP target genes NEDD9 and ASNS. Our findings identify PAWS1 as the first non-SMAD substrate for type I BMP receptor kinases and as a novel player in the BMP pathway. We also demonstrate that PAWS1 regulates the expression of several non-BMP target genes, suggesting roles for PAWS1 beyond the BMP pathway.
Collapse
Affiliation(s)
- Janis Vogt
- MRC Protein Phosphorylation and Ubiquitylation Unit, University of Dundee, Dow St., Dundee DD1 5EH, UK
| | | | | | | | | | | | | | | |
Collapse
|
45
|
Papanayotou C, De Almeida I, Liao P, Oliveira NMM, Lu SQ, Kougioumtzidou E, Zhu L, Shaw A, Sheng G, Streit A, Yu D, Wah Soong T, Stern CD. Calfacilitin is a calcium channel modulator essential for initiation of neural plate development. Nat Commun 2013; 4:1837. [PMID: 23673622 PMCID: PMC3674269 DOI: 10.1038/ncomms2864] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2012] [Accepted: 04/10/2013] [Indexed: 11/09/2022] Open
Abstract
Calcium fluxes have been implicated in the specification of the vertebrate embryonic nervous system for some time, but how these fluxes are regulated and how they relate to the rest of the neural induction cascade is unknown. Here we describe Calfacilitin, a transmembrane calcium channel facilitator that increases calcium flux by generating a larger window current and slowing inactivation of the L-type CaV1.2 channel. Calfacilitin binds to this channel and is co-expressed with it in the embryo. Regulation of intracellular calcium by Calfacilitin is required for expression of the neural plate specifiers Geminin and Sox2 and for neural plate formation. Loss-of-function of Calfacilitin can be rescued by ionomycin, which increases intracellular calcium. Our results elucidate the role of calcium fluxes in early neural development and uncover a new factor in the modulation of calcium signalling.
Collapse
Affiliation(s)
- Costis Papanayotou
- Department of Cell and Developmental Biology, University College London, Gower Street (Anatomy Building), London WC1E 6BT, UK.
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
46
|
Pera EM, Acosta H, Gouignard N, Climent M, Arregi I. Active signals, gradient formation and regional specificity in neural induction. Exp Cell Res 2013; 321:25-31. [PMID: 24315941 DOI: 10.1016/j.yexcr.2013.11.018] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2013] [Revised: 11/21/2013] [Accepted: 11/23/2013] [Indexed: 01/01/2023]
Abstract
The question of how the vertebrate embryo gives rise to a nervous system is of paramount interest in developmental biology. Neural induction constitutes the earliest step in this process and is tightly connected with development of the embryonic body axes. In the Xenopus embryo, perpendicular gradients of BMP and Wnt signals pattern the dorsoventral and anteroposterior body axes. Both pathways need to be inhibited to allow anterior neural induction to occur. FGF8 and IGF are active neural inducers that together with BMP and Wnt signals are integrated at the level of Smad 1/5/8 phosphorylation. Hedgehog (Hh) also contributes to anterior neural induction. Suppressor-of-fused plays an important role in intertwining the Hh and Wnt pathways. Distinct mechanisms are discussed that establish morphogen gradients and integrate retinoic acid and FGF signals during posterior development. These findings not only improve our understanding of regional specification in neural induction, but have profound implications for mammalian stem cell research and regenerative medicine.
Collapse
Affiliation(s)
- Edgar M Pera
- Lund Stem Cell Center, BMC, B12, Klinikgatan 26, Lund University, S-221 84 Lund, Sweden.
| | - Helena Acosta
- Lund Stem Cell Center, BMC, B12, Klinikgatan 26, Lund University, S-221 84 Lund, Sweden
| | - Nadège Gouignard
- Lund Stem Cell Center, BMC, B12, Klinikgatan 26, Lund University, S-221 84 Lund, Sweden
| | - Maria Climent
- Lund Stem Cell Center, BMC, B12, Klinikgatan 26, Lund University, S-221 84 Lund, Sweden
| | - Igor Arregi
- Lund Stem Cell Center, BMC, B12, Klinikgatan 26, Lund University, S-221 84 Lund, Sweden
| |
Collapse
|
47
|
Midbrain dopaminergic neurons: a review of the molecular circuitry that regulates their development. Dev Biol 2013; 379:123-38. [PMID: 23603197 DOI: 10.1016/j.ydbio.2013.04.014] [Citation(s) in RCA: 131] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2013] [Revised: 03/27/2013] [Accepted: 04/12/2013] [Indexed: 12/21/2022]
Abstract
Dopaminergic (DA) neurons of the ventral midbrain (VM) play vital roles in the regulation of voluntary movement, emotion and reward. They are divided into the A8, A9 and A10 subgroups. The development of the A9 group of DA neurons is an area of intense investigation to aid the generation of these neurons from stem cell sources for cell transplantation approaches to Parkinson's disease (PD). This review discusses the molecular processes that are involved in the identity, specification, maturation, target innervation and survival of VM DA neurons during development. The complex molecular interactions of a number of genetic pathways are outlined, as well as recent advances in the mechanisms that regulate subset identity within the VM DA neuronal pool. A thorough understanding of the cellular and molecular mechanisms involved in the development of VM DA neurons will greatly facilitate the use of cell replacement therapy for the treatment of PD.
Collapse
|
48
|
Shimizu M, Goto T, Sato A, Shibuya H. WNK4 is an essential effector of anterior formation in FGF signaling. Genes Cells 2013; 18:442-9. [PMID: 23517227 PMCID: PMC3712470 DOI: 10.1111/gtc.12048] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2013] [Accepted: 02/17/2013] [Indexed: 11/30/2022]
Abstract
With no lysine (K) (WNK) kinase family is conserved among many species and regulates SPAK/OSR1 and ion cotransporters. WNK is also involved in developmental and cellular processes, but the molecular mechanisms underlying its regulation in these processes remain unknown. In this study, we found that WNK4 is involved in fibroblast growth factor (FGF) signaling during Xenopus development. In Xenopus embryos, depletion of WNK4 by antisense morpholino oligonucleotides (MOs) results in a severe defect in anterior development and impaired expression of endogenous anterior markers. Defects in head formation or expression of anterior marker genes caused by suppression of endogenous WNK4 expression could be rescued by expression of wild-type WNK4, but not mutant WNK4 lacking its kinase activity. It is notable that morphants of Xenopus WNK4 inhibited the expression of anterior marker genes and the target genes induced by FGF signaling. Moreover, knockdown of Wnk4 significantly reduced the phosphorylation level of Osr1 induced by FGF. These results provide the first evidence that FGF signaling regulates WNK4 function required for anterior formation in Xenopus development.
Collapse
Affiliation(s)
- Masahiro Shimizu
- Department of Molecular Cell Biology, Medical Research Institute, Tokyo Medical and Dental University, 1-5-45 Yushima, Bunkyo-ku, Tokyo 113-8510, Japan
| | | | | | | |
Collapse
|
49
|
Kidwai FK, Liu H, Toh WS, Fu X, Jokhun DS, Movahednia MM, Li M, Zou Y, Squier CA, Phan TT, Cao T. Differentiation of human embryonic stem cells into clinically amenable keratinocytes in an autogenic environment. J Invest Dermatol 2012; 133:618-628. [PMID: 23235526 DOI: 10.1038/jid.2012.384] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
Human embryonic stem cells (hESCs)-derived keratinocytes hold great clinical and research potential. However, the current techniques are hampered by the use of xenogenic components that limits their clinical application. Here we demonstrated an efficient differentiation of H9 hESCs (H9-hESCs) into keratinocytes (H9-Kert) with the minimum use of animal-derived materials. For differentiation, we established two microenvironment systems originated from H9-hESCs (autogenic microenvironment). These autogenic microenvironment systems consist of an autogenic coculture system (ACC) and an autogenic feeder-free system (AFF). In addition, we showed a stage-specific effect of Activin in promoting keratinocyte differentiation from H9-hESCs while repressing the expression of early neural markers in the ACC system. Furthermore, we also explained the effect of Activin in construction of the AFF system made up of extracellular matrix similar to basement membrane extracted from H9-hESC-derived fibroblasts. H9-Kert differentiated in both systems expressed keratinocyte markers at mRNA and protein levels. H9-Kert were also able to undergo terminal differentiation in high Ca(2+) medium. These findings support the transition toward the establishment of an animal-free microenvironment for successful differentiation of hESCs into keratinocytes for potential clinical application.
Collapse
Affiliation(s)
- Fahad K Kidwai
- Oral Sciences Disciplines, Faculty of Dentistry, National University of Singapore, Singapore
| | - Hua Liu
- Oral Sciences Disciplines, Faculty of Dentistry, National University of Singapore, Singapore; Centre for Stem Cell and Tissue Engineering, School of Medicine, Zhejiang University, Hangzhou, Zhejiang Province, People's Republic of China
| | - Wei Seong Toh
- Oral Sciences Disciplines, Faculty of Dentistry, National University of Singapore, Singapore
| | - Xin Fu
- Oral Sciences Disciplines, Faculty of Dentistry, National University of Singapore, Singapore; Plastic Surgery Hospital (Institute), Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, People's Republic of China
| | - Doorgesh S Jokhun
- Department of Biological Sciences, Faculty of Science, National University of Singapore, Singapore
| | - Mohammad M Movahednia
- Oral Sciences Disciplines, Faculty of Dentistry, National University of Singapore, Singapore
| | - Mingming Li
- Oral Sciences Disciplines, Faculty of Dentistry, National University of Singapore, Singapore
| | - Yu Zou
- Oral Sciences Disciplines, Faculty of Dentistry, National University of Singapore, Singapore
| | - Christopher A Squier
- Department of Oral Pathology, Radiology and Medicine, and Dows, College of Dentistry, The University of Iowa, Iowa City, Iowa, USA
| | - Toan T Phan
- Department of Surgery, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Tong Cao
- Oral Sciences Disciplines, Faculty of Dentistry, National University of Singapore, Singapore.
| |
Collapse
|
50
|
Das S, Chang C. Regulation of early xenopus embryogenesis by Smad ubiquitination regulatory factor 2. Dev Dyn 2012; 241:1260-73. [PMID: 22674516 DOI: 10.1002/dvdy.23811] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/23/2012] [Indexed: 01/17/2023] Open
Abstract
BACKGROUND Smad ubiquitination regulatory factor (Smurf) 1 and 2 are E3 ubiquitin ligases originally identified as inhibitors of transforming growth factor beta signaling and are shown to modulate multiple cellular activities. The roles of Smurfs in vertebrate embryogenesis, however, are not completely understood. RESULTS Here we investigate the function of Smurf2 during early Xenopus development. We show that distinctly from Smurf1, overexpression of Smurf2 in presumptive mesoderm interfered with mesoderm induction and caused axial defects, whereas knockdown of Smurf2 with antisense morpholino oligonucleotides resulted in expansion of the mesoderm. These results imply that Smurf2 may modulate nodal-mediated mesodermal induction. Consistently, ventral expression of Smurf2 induced a partial secondary axis with head structures. In the ectoderm, Smurf2 resembled Smurf1 in controlling neural and epidermal marker expression and influencing head formation. Smurf1, but not Smurf2, additionally affected neural tube closure. Interestingly, both Smurfs could enhance as well as repress neural crest markers, implying that they modulate their targets dynamically during neural plate border specification. CONCLUSION Our data demonstrate that Smurf1 and Smurf2 have overlapping and distinct functionalities during early frog embryogenesis; collectively, they regulate ectodermal and mesodermal induction and patterning to ensure normal development of Xenopus embryos.
Collapse
Affiliation(s)
- Shaonli Das
- Department of Cell Biology, University of Alabama, Birmingham, Alabama, USA
| | | |
Collapse
|