1
|
Saumweber E, Mzoughi S, Khadra A, Werberger A, Schumann S, Guccione E, Schmeisser MJ, Kühl SJ. Prdm15 acts upstream of Wnt4 signaling in anterior neural development of Xenopus laevis. Front Cell Dev Biol 2024; 12:1316048. [PMID: 38444828 PMCID: PMC10912572 DOI: 10.3389/fcell.2024.1316048] [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: 10/10/2023] [Accepted: 02/02/2024] [Indexed: 03/07/2024] Open
Abstract
Mutations in PRDM15 lead to a syndromic form of holoprosencephaly (HPE) known as the Galloway-Mowat syndrome (GAMOS). While a connection between PRDM15, a zinc finger transcription factor, and WNT/PCP signaling has been established, there is a critical need to delve deeper into their contributions to early development and GAMOS pathogenesis. We used the South African clawed frog Xenopus laevis as the vertebrate model organism and observed that prdm15 was enriched in the tissues and organs affected in GAMOS. Furthermore, we generated a morpholino oligonucleotide-mediated prdm15 knockdown model showing that the depletion of Prdm15 leads to abnormal eye, head, and brain development, effectively recapitulating the anterior neural features in GAMOS. An analysis of the underlying molecular basis revealed a reduced expression of key genes associated with eye, head, and brain development. Notably, this reduction could be rescued by the introduction of wnt4 RNA, particularly during the induction of the respective tissues. Mechanistically, our data demonstrate that Prdm15 acts upstream of both canonical and non-canonical Wnt4 signaling during anterior neural development. Our findings describe severe ocular and anterior neural abnormalities upon Prdm15 depletion and elucidate the role of Prdm15 in canonical and non-canonical Wnt4 signaling.
Collapse
Affiliation(s)
- Ernestine Saumweber
- Institute of Biochemistry and Molecular Biology, Ulm University, Ulm, Germany
| | - Slim Mzoughi
- Center of OncoGenomics and Innovative Therapeutics (COGIT), Department of Oncological Sciences, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New-York, NY, United States
| | - Arin Khadra
- Institute of Biochemistry and Molecular Biology, Ulm University, Ulm, Germany
| | - Anja Werberger
- Institute of Biochemistry and Molecular Biology, Ulm University, Ulm, Germany
| | - Sven Schumann
- Institute of Anatomy, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
| | - Ernesto Guccione
- Center of OncoGenomics and Innovative Therapeutics (COGIT), Department of Oncological Sciences, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New-York, NY, United States
| | - Michael J. Schmeisser
- Institute of Anatomy, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
- Focus Program Translational Neurosciences, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
| | - Susanne J. Kühl
- Institute of Biochemistry and Molecular Biology, Ulm University, Ulm, Germany
| |
Collapse
|
2
|
Belrose JL, Prasad A, Sammons MA, Gibbs KM, Szaro BG. Comparative gene expression profiling between optic nerve and spinal cord injury in Xenopus laevis reveals a core set of genes inherent in successful regeneration of vertebrate central nervous system axons. BMC Genomics 2020; 21:540. [PMID: 32758133 PMCID: PMC7430912 DOI: 10.1186/s12864-020-06954-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2019] [Accepted: 07/27/2020] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND The South African claw-toed frog, Xenopus laevis, is uniquely suited for studying differences between regenerative and non-regenerative responses to CNS injury within the same organism, because some CNS neurons (e.g., retinal ganglion cells after optic nerve crush (ONC)) regenerate axons throughout life, whereas others (e.g., hindbrain neurons after spinal cord injury (SCI)) lose this capacity as tadpoles metamorphose into frogs. Tissues from these CNS regions (frog ONC eye, tadpole SCI hindbrain, frog SCI hindbrain) were used in a three-way RNA-seq study of axotomized CNS axons to identify potential core gene expression programs for successful CNS axon regeneration. RESULTS Despite tissue-specific changes in expression dominating the injury responses of each tissue, injury-induced changes in gene expression were nonetheless shared between the two axon-regenerative CNS regions that were not shared with the non-regenerative region. These included similar temporal patterns of gene expression and over 300 injury-responsive genes. Many of these genes and their associated cellular functions had previously been associated with injury responses of multiple tissues, both neural and non-neural, from different species, thereby demonstrating deep phylogenetically conserved commonalities between successful CNS axon regeneration and tissue regeneration in general. Further analyses implicated the KEGG adipocytokine signaling pathway, which links leptin with metabolic and gene regulatory pathways, and a novel gene regulatory network with genes regulating chromatin accessibility at its core, as important hubs in the larger network of injury response genes involved in successful CNS axon regeneration. CONCLUSIONS This study identifies deep, phylogenetically conserved commonalities between CNS axon regeneration and other examples of successful tissue regeneration and provides new targets for studying the molecular underpinnings of successful CNS axon regeneration, as well as a guide for distinguishing pro-regenerative injury-induced changes in gene expression from detrimental ones in mammals.
Collapse
Affiliation(s)
- Jamie L Belrose
- Department of Biological Sciences, University at Albany, State University of New York, 1400 Washington Avenue, Albany, NY, 12222, USA
- Center for Neuroscience Research, University at Albany, State University of New York, 1400 Washington Avenue, Albany, NY, 12222, USA
| | - Aparna Prasad
- Department of Biological Sciences, University at Albany, State University of New York, 1400 Washington Avenue, Albany, NY, 12222, USA
- Center for Neuroscience Research, University at Albany, State University of New York, 1400 Washington Avenue, Albany, NY, 12222, USA
| | - Morgan A Sammons
- Department of Biological Sciences, University at Albany, State University of New York, 1400 Washington Avenue, Albany, NY, 12222, USA
| | - Kurt M Gibbs
- Department of Biology and Chemistry, Morehead State University, Morehead, KY, 40351, USA
| | - Ben G Szaro
- Department of Biological Sciences, University at Albany, State University of New York, 1400 Washington Avenue, Albany, NY, 12222, USA.
- Center for Neuroscience Research, University at Albany, State University of New York, 1400 Washington Avenue, Albany, NY, 12222, USA.
| |
Collapse
|
3
|
Zhang S, Shuai L, Wang D, Huang T, Yang S, Miao M, Liu F, Xu J. Pim-1 Protects Retinal Ganglion Cells by Enhancing Their Regenerative Ability Following Optic Nerve Crush. Exp Neurobiol 2020; 29:249-272. [PMID: 32624507 PMCID: PMC7344373 DOI: 10.5607/en20019] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2020] [Revised: 06/22/2020] [Accepted: 06/22/2020] [Indexed: 11/19/2022] Open
Abstract
Provirus integration site Moloney murine leukemia virus (Pim-1) is a proto-oncogene reported to be associated with cell proliferation, differentiation and survival. This study was to explore the neuroprotective role of Pim-1 in a rat model subjected to optic nerve crush (ONC), and discuss its related molecules in improving the intrinsic regeneration ability of retinal ganglion cells (RGCs). Immunofluorescence staining showed that AAV2- Pim-1 infected 71% RGCs and some amacrine cells in the retina. Real-time PCR and Western blotting showed that retina infection with AAV2- Pim-1 up-regulated the Pim-1 mRNA and protein expressions compared with AAV2-GFP group. Hematoxylin-Eosin (HE) staining, γ-synuclein immunohistochemistry, Cholera toxin B (CTB) tracing and TUNEL showed that RGCs transduction with AAV2-Pim-1 prior to ONC promoted the survival of damaged RGCs and decreased cell apoptosis. RITC anterograde labeling showed that Pim-1 overexpression increased axon regeneration and promoted the recovery of visual function by pupillary light reflex and flash visual evoked potential. Western blotting showed that Pim- 1 overexpression up-regulated the expression of Stat3, p-Stat3, Akt1, p-Akt1, Akt2 and p-Akt2, as well as βIII-tubulin, GAP-43 and 4E-BP1, and downregulated the expression of SOCS1 and SOCS3, Cleaved caspase 3, Bad and Bax. These results demonstrate that Pim-1 exerted a neuroprotective effect by promoting nerve regeneration and functional recovery of RGCs. In addition, it enhanced the intrinsic regeneration capacity of RGCs after ONC by activating Stat3, Akt1 and Akt2 pathways, and inhibiting the mitochondrial apoptosis pathways. These findings suggest that Pim-1 may prove to be a potential therapeutic target for the clinical treatment of optic nerve injury.
Collapse
Affiliation(s)
- Shoumei Zhang
- Department of Anatomy, Second Military Medical University, Shanghai 200433, China.,Translational Medical Center for Stem Cell Therapy, Shanghai East Hospital, Tongji University School of Medicine, Shanghai 200120, China
| | - Li Shuai
- Department of Health Administration, Second Military Medical University, Shanghai 200433, China
| | - Dong Wang
- Department of Anatomy, Second Military Medical University, Shanghai 200433, China
| | - Tingting Huang
- Department of Anatomy, Second Military Medical University, Shanghai 200433, China
| | - Shengsheng Yang
- Department of Biochemistry and Molecular Biology, Second Military Medical University, Shanghai 200433, China
| | - Mingyong Miao
- Department of Biochemistry and Molecular Biology, Second Military Medical University, Shanghai 200433, China
| | - Fang Liu
- Department of Anatomy, Second Military Medical University, Shanghai 200433, China
| | - Jiajun Xu
- Department of Anatomy, Second Military Medical University, Shanghai 200433, China
| |
Collapse
|
4
|
Hausrat TJ, Radwitz J, Lombino FL, Breiden P, Kneussel M. Alpha- and beta-tubulin isotypes are differentially expressed during brain development. Dev Neurobiol 2020; 81:333-350. [PMID: 32293117 DOI: 10.1002/dneu.22745] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2019] [Revised: 01/21/2020] [Accepted: 04/08/2020] [Indexed: 12/14/2022]
Abstract
Alpha- and beta-tubulin dimers polymerize into protofilaments that associate laterally to constitute a hollow tube, the microtubule. A dynamic network of interlinking filaments forms the microtubule cytoskeleton, which maintains the structure of cells and is key to various cellular processes including cell division, cell migration, and intracellular transport. Individual microtubules have an identity that depends on the differential integration of specific alpha- and beta-tubulin isotypes and is further specified by a variety of posttranslational modifications (PTMs). It is barely understood to which extent neighboring microtubules differ in their tubulin composition or whether specific tubulin isotypes cluster along the polymer. Furthermore, our knowledge about the spatio-temporal expression patterns of tubulin isotypes is limited, not at least due to the lack of antibodies or antibody cross-reactivities. Here, we asked which alpha- and beta-tubulin mRNAs and proteins are expressed in developing hippocampal neuron cultures and ex vivo brain tissue lysates. Using heterologous expression of GFP-tubulin fusion proteins, we systematically tested antibody-specificities against various tubulin isotypes. Our data provide quantitative information about tubulin expression levels in the mouse brain and classify tubulin isotypes during pre- and postnatal development.
Collapse
Affiliation(s)
- Torben J Hausrat
- Department of Molecular Neurogenetics, ZMNH, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Jennifer Radwitz
- Department of Molecular Neurogenetics, ZMNH, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Franco L Lombino
- Department of Molecular Neurogenetics, ZMNH, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Petra Breiden
- Department of Molecular Neurogenetics, ZMNH, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Matthias Kneussel
- Department of Molecular Neurogenetics, ZMNH, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| |
Collapse
|
5
|
Warchol ME, Massoodnia R, Pujol R, Cox BC, Stone JS. Development of hair cell phenotype and calyx nerve terminals in the neonatal mouse utricle. J Comp Neurol 2019; 527:1913-1928. [PMID: 30724338 DOI: 10.1002/cne.24658] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2018] [Revised: 01/26/2019] [Accepted: 02/01/2019] [Indexed: 01/03/2023]
Abstract
The vestibular organs of reptiles, birds, and mammals possess Type I and Type II sensory hair cells, which have distinct morphologies, physiology, and innervation. Little is known about how vestibular hair cells adopt a Type I or Type II identity or acquire proper innervation. One distinguishing marker is the transcription factor Sox2, which is expressed in all developing hair cells but persists only in Type II hair cells in maturity. We examined Sox2 expression and formation of afferent nerve terminals in mouse utricles between postnatal days 0 (P0) and P17. Between P3 and P14, many hair cells lost Sox2 immunoreactivity and the density of calyceal afferent nerve terminals (specific to Type I hair cells) increased in all regions of the utricle. At early time points, many calyces enclosed Sox2-labeled hair cells, while some Sox2-negative hair cells within the striola had not yet developed a calyx. These observations indicate that calyx maturation is not temporally correlated with loss of Sox2 expression in Type I hair cells. To determine which type(s) of hair cells are formed postnatally, we fate-mapped neonatal supporting cells by injecting Plp-CreER T2 :Rosa26 tdTomato mice with tamoxifen at P2 and P3. At P9, tdTomato-positive hair cells were immature and not classifiable by type. At P30, tdTomato-positive hair cells increased 1.8-fold compared to P9, and 91% of tdTomato-labeled hair cells were Type II. Our findings show that most neonatally-derived hair cells become Type II, and many Type I hair cells (formed before P2) downregulate Sox2 and acquire calyces between P0 and P14.
Collapse
Affiliation(s)
- Mark E Warchol
- Department of Otolaryngology, Washington University, St Louis, Missouri
| | - Roxanna Massoodnia
- Department of Otolaryngology-Head and Neck Surgery and the Virginia Merrill Bloedel Hearing Research Center, University of Washington, Seattle, Washington
| | - Remy Pujol
- Department of Otolaryngology-Head and Neck Surgery and the Virginia Merrill Bloedel Hearing Research Center, University of Washington, Seattle, Washington.,INSERM Unit 1051, Institute of Neuroscience, University of Montpellier, Montpellier, France
| | - Brandon C Cox
- Departments of Pharmacology and Surgery, Division of Otolaryngology, Southern Illinois University School of Medicine, Springfield, Illinois
| | - Jennifer S Stone
- Department of Otolaryngology-Head and Neck Surgery and the Virginia Merrill Bloedel Hearing Research Center, University of Washington, Seattle, Washington
| |
Collapse
|
6
|
Priscilla R, Szaro BG. Comparisons of SOCS mRNA and protein levels in Xenopus provide insights into optic nerve regenerative success. Brain Res 2019; 1704:150-160. [PMID: 30315759 DOI: 10.1016/j.brainres.2018.10.012] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2018] [Revised: 08/23/2018] [Accepted: 10/09/2018] [Indexed: 01/21/2023]
Abstract
In vertebrates from fishes to mammals, optic nerve injury induces increased expression ofSuppressor of Cytokine Signaling 3(SOCS3) mRNA, a modulator of cytokine signaling that is known to inhibit CNS axon regeneration. Unlike amniotes, however, anamniotes successfully regenerate optic axons, despite this increase. To address this seeming paradox, we examined the SOCS3 response to optic nerve injury in the frog,Xenopus laevis, at both the mRNA and protein levels. Far from being only transiently induced, SOCS3 mRNA expression increased throughout regeneration in retinal ganglion cells, but immunostaining and Western blots indicated that this increase was reflected at the protein level in regenerating optic axons but not in ganglion cell bodies. Polysome profiling provided additional evidence that SOCS3 protein levels were regulated post-translationally by demonstrating that the mRNA was efficiently translated in the injured eye. In tumor cells, another member of theSOCS gene family,SOCS2, is known to mediate SOCS3 degradation by targeting it for proteasomal degradation. Unlike the SOCS2 response in mammalian optic nerve injury, SOCS2 expression increased inXenopusretinal ganglion cells after injury, at both the mRNA and protein levels; it was, however, largely absent from both uninjured and regenerating optic axons. We propose a similar degradation mechanism may be spatially restricted inXenopusto keep SOCS3 protein levels sufficiently in check within ganglion cell bodies, where SOCS3 would otherwise inhibit transcription of genes needed for regeneration, but allow them to rise within the axons, where SOCS3 has pro-regenerative effects.
Collapse
Affiliation(s)
- Rupa Priscilla
- Department of Biological Sciences and the Center for Neuroscience Research, University at Albany, State University of New York, 1400 Washington Avenue, Albany, NY 12222, USA
| | - Ben G Szaro
- Department of Biological Sciences and the Center for Neuroscience Research, University at Albany, State University of New York, 1400 Washington Avenue, Albany, NY 12222, USA.
| |
Collapse
|
7
|
Edwards-Faret G, Cebrián-Silla A, Méndez-Olivos EE, González-Pinto K, García-Verdugo JM, Larraín J. Cellular composition and organization of the spinal cord central canal during metamorphosis of the frog Xenopus laevis. J Comp Neurol 2018; 526:1712-1732. [PMID: 29603210 DOI: 10.1002/cne.24441] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2017] [Revised: 03/12/2018] [Accepted: 03/21/2018] [Indexed: 01/12/2023]
Abstract
Studying the cellular composition and morphological changes of cells lining the central canal during Xenopus laevis metamorphosis could contribute to understand postnatal development and spinal cord regeneration. Here we report the analysis of central canal cells at different stages during metamorphosis using immunofluorescence for protein markers expression, transmission and scanning electron microscopy and cell proliferation assays. The central canal was regionalized according to expression of glial markers, ultrastructure, and proliferation in dorsal, lateral, and ventral domains with differences between larvae and froglets. In regenerative larvae, all cell types were uniciliated, have a radial morphology, and elongated nuclei with lax chromatin, resembling radial glial cells. Important differences in cells of nonregenerative froglets were observed, although uniciliated cells were found, the most abundant cells had multicilia and revealed extensive changes in the maturation and differentiation state. The majority of dividing cells in larvae corresponded to uniciliated cells at dorsal and lateral domains in a cervical-lumbar gradient, correlating with undifferentiated features. Neurons contacting the lumen of the central canal were detected in both stages and revealed extensive changes in the maturation and differentiation state. However, in froglets a very low proportion of cells incorporate 5-ethynyl-2'-deoxyuridine (EdU), associated with the differentiated profile and with the increase of multiciliated cells. Our work showed progressive changes in the cell types lining the central canal of Xenopus laevis spinal cord which are correlated with the regenerative capacities.
Collapse
Affiliation(s)
- Gabriela Edwards-Faret
- Center for Aging and Regeneration, Faculty of Biological Sciences, P. Universidad Católica de Chile, Avenida Libertador Bernardo O'Higgins 340, Santiago, Chile
| | - Arantxa Cebrián-Silla
- Laboratorio de Neurobiologia Comparada, Instituto Cavanilles, Universidad de Valencia, Valencia 46980, CIBERNED, Valencia, Spain
| | - Emilio E Méndez-Olivos
- Center for Aging and Regeneration, Faculty of Biological Sciences, P. Universidad Católica de Chile, Avenida Libertador Bernardo O'Higgins 340, Santiago, Chile
| | - Karina González-Pinto
- Center for Aging and Regeneration, Faculty of Biological Sciences, P. Universidad Católica de Chile, Avenida Libertador Bernardo O'Higgins 340, Santiago, Chile.,Universidad Arturo Prat del Estado de Chile, Iquique, Chile
| | - José Manuel García-Verdugo
- Laboratorio de Neurobiologia Comparada, Instituto Cavanilles, Universidad de Valencia, Valencia 46980, CIBERNED, Valencia, Spain
| | - Juan Larraín
- Center for Aging and Regeneration, Faculty of Biological Sciences, P. Universidad Católica de Chile, Avenida Libertador Bernardo O'Higgins 340, Santiago, Chile
| |
Collapse
|
8
|
Fragile X Mental Retardation Protein Is Required to Maintain Visual Conditioning-Induced Behavioral Plasticity by Limiting Local Protein Synthesis. J Neurosci 2017; 36:7325-39. [PMID: 27383604 DOI: 10.1523/jneurosci.4282-15.2016] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2015] [Accepted: 05/28/2016] [Indexed: 12/30/2022] Open
Abstract
UNLABELLED Fragile X mental retardation protein (FMRP) is thought to regulate neuronal plasticity by limiting dendritic protein synthesis, but direct demonstration of a requirement for FMRP control of local protein synthesis during behavioral plasticity is lacking. Here we tested whether FMRP knockdown in Xenopus optic tectum affects local protein synthesis in vivo and whether FMRP knockdown affects protein synthesis-dependent visual avoidance behavioral plasticity. We tagged newly synthesized proteins by incorporation of the noncanonical amino acid azidohomoalanine and visualized them with fluorescent noncanonical amino acid tagging (FUNCAT). Visual conditioning and FMRP knockdown produce similar increases in FUNCAT in tectal neuropil. Induction of visual conditioning-dependent behavioral plasticity occurs normally in FMRP knockdown animals, but plasticity degrades over 24 h. These results indicate that FMRP affects visual conditioning-induced local protein synthesis and is required to maintain the visual conditioning-induced behavioral plasticity. SIGNIFICANCE STATEMENT Fragile X syndrome (FXS) is the most common form of inherited intellectual disability. Exaggerated dendritic protein synthesis resulting from loss of fragile X mental retardation protein (FMRP) is thought to underlie cognitive deficits in FXS, but no direct evidence has demonstrated that FMRP-regulated dendritic protein synthesis affects behavioral plasticity in intact animals. Xenopus tadpoles exhibit a visual avoidance behavior that improves with visual conditioning in a protein synthesis-dependent manner. We showed that FMRP knockdown and visual conditioning dramatically increase protein synthesis in neuronal processes. Furthermore, induction of visual conditioning-dependent behavioral plasticity occurs normally after FMRP knockdown, but performance rapidly deteriorated in the absence of FMRP. These studies show that FMRP negatively regulates local protein synthesis and is required to maintain visual conditioning-induced behavioral plasticity in vivo.
Collapse
|
9
|
Motahari Z, Martinez-De Luna RI, Viczian AS, Zuber ME. Tbx3 represses bmp4 expression and, with Pax6, is required and sufficient for retina formation. Development 2016; 143:3560-3572. [PMID: 27578778 DOI: 10.1242/dev.130955] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2015] [Accepted: 08/05/2016] [Indexed: 12/30/2022]
Abstract
Vertebrate eye formation begins in the anterior neural plate in the eye field. Seven eye field transcription factors (EFTFs) are expressed in eye field cells and when expressed together are sufficient to generate retina from pluripotent cells. The EFTF Tbx3 can regulate the expression of some EFTFs; however, its role in retina formation is unknown. Here, we show that Tbx3 represses bmp4 transcription and is required in the eye field for both neural induction and normal eye formation in Xenopus laevis Although sufficient for neural induction, Tbx3-expressing pluripotent cells only form retina in the context of the eye field. Unlike Tbx3, the neural inducer Noggin can generate retina both within and outside the eye field. We found that the neural and retina-inducing activity of Noggin requires Tbx3. Noggin, but not Tbx3, induces Pax6 and coexpression of Tbx3 and Pax6 is sufficient to determine pluripotent cells to a retinal lineage. Our results suggest that Tbx3 represses bmp4 expression and maintains eye field neural progenitors in a multipotent state; then, in combination with Pax6, Tbx3 causes eye field cells to form retina.
Collapse
Affiliation(s)
- Zahra Motahari
- The Center for Vision Research, Department of Ophthalmology, Upstate Medical University, Syracuse, NY 13210, USA Department of Biochemistry and Molecular Biology, Upstate Medical University, Syracuse, NY 13210, USA
| | - Reyna I Martinez-De Luna
- The Center for Vision Research, Department of Ophthalmology, Upstate Medical University, Syracuse, NY 13210, USA
| | - Andrea S Viczian
- The Center for Vision Research, Department of Ophthalmology, Upstate Medical University, Syracuse, NY 13210, USA Department of Biochemistry and Molecular Biology, Upstate Medical University, Syracuse, NY 13210, USA Department of Neuroscience and Physiology, Upstate Medical University, Syracuse, NY 13210, USA Department of Cell and Developmental Biology, Upstate Medical University, Syracuse, NY 13210, USA
| | - Michael E Zuber
- The Center for Vision Research, Department of Ophthalmology, Upstate Medical University, Syracuse, NY 13210, USA Department of Biochemistry and Molecular Biology, Upstate Medical University, Syracuse, NY 13210, USA Department of Neuroscience and Physiology, Upstate Medical University, Syracuse, NY 13210, USA
| |
Collapse
|
10
|
Zhang S, Li J, Lea R, Amaya E. Assessing Primary Neurogenesis in Xenopus Embryos Using Immunostaining. J Vis Exp 2016:e53949. [PMID: 27166855 PMCID: PMC4941913 DOI: 10.3791/53949] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023] Open
Abstract
Primary neurogenesis is a dynamic and complex process during embryonic development that sets up the initial layout of the central nervous system. During this process, a portion of neural stem cells undergo differentiation and give rise to the first populations of differentiated primary neurons within the nascent central nervous system. Several vertebrate model organisms have been used to explore the mechanisms of neural cell fate specification, patterning, and differentiation. Among these is the African clawed frog, Xenopus, which provides a powerful system for investigating the molecular and cellular mechanisms responsible for primary neurogenesis due to its rapid and accessible development and ease of embryological and molecular manipulations. Here, we present a convenient and rapid method to observe the different populations of neuronal cells within Xenopus central nervous system. Using antibody staining and immunofluorescence on sections of Xenopus embryos, we are able to observe the locations of neural stem cells and differentiated primary neurons during primary neurogenesis.
Collapse
Affiliation(s)
- Siwei Zhang
- The Healing Foundation Centre, Faculty of Life Sciences, University of Manchester; Department of Cell and Molecular Biology, Feinberg School of Medicine, Northwestern University
| | - Jingjing Li
- The Healing Foundation Centre, Faculty of Life Sciences, University of Manchester; Department of Craniofacial Development and Stem Cell Biology, Dental Institute, King's College London
| | - Robert Lea
- The Healing Foundation Centre, Faculty of Life Sciences, University of Manchester
| | - Enrique Amaya
- The Healing Foundation Centre, Faculty of Life Sciences, University of Manchester;
| |
Collapse
|
11
|
Hutchins EJ, Belrose JL, Szaro BG. Phosphorylation of heterogeneous nuclear ribonucleoprotein K at an extracellular signal-regulated kinase phosphorylation site promotes neurofilament-medium protein expression and axon outgrowth in Xenopus. Neurosci Lett 2015; 607:59-65. [DOI: 10.1016/j.neulet.2015.09.027] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2015] [Revised: 08/27/2015] [Accepted: 09/22/2015] [Indexed: 12/18/2022]
|
12
|
Ware M, Dupé V, Schubert FR. Evolutionary Conservation of the Early Axon Scaffold in the Vertebrate Brain. Dev Dyn 2015; 244:1202-14. [PMID: 26228689 DOI: 10.1002/dvdy.24312] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2015] [Revised: 07/20/2015] [Accepted: 07/20/2015] [Indexed: 11/11/2022] Open
Abstract
The early axon scaffold is the first axonal structure to appear in the rostral brain of vertebrates, paving the way for later, more complex connections. Several early axon scaffold components are conserved between all vertebrates; most notably two main ventral longitudinal tracts, the tract of the postoptic commissure and the medial longitudinal fascicle. While the overall structure is remarkably similar, differences both in the organization and the development of the early tracts are apparent. This review will bring together extensive data from the last 25 years in different vertebrates and for the first time, the timing and anatomy of these early tracts have been directly compared. Representatives of major vertebrate clades, including cat shark, Xenopus, chick, and mouse embryos, will be compared using immunohistochemistry staining based on previous results. There is still confusion over the nomenclature and homology of these tracts which this review will aim to address. The discussion here is relevant both for understanding the evolution of the early axon scaffold and for future studies into the molecular regulation of its formation.
Collapse
Affiliation(s)
- Michelle Ware
- Institute of Biomedical and Biomolecular Sciences, University of Portsmouth, Portsmouth, United Kingdom.,Institut de Génétique et Développement, CNRS UMR6290, Université de Rennes1, IFR140, GFAS, Faculté de Médecine, Rennes, France
| | - Valérie Dupé
- Institut de Génétique et Développement, CNRS UMR6290, Université de Rennes1, IFR140, GFAS, Faculté de Médecine, Rennes, France
| | - Frank R Schubert
- Institute of Biomedical and Biomolecular Sciences, University of Portsmouth, Portsmouth, United Kingdom
| |
Collapse
|
13
|
Liu Y, Wang C, Destin G, Szaro BG. Microtubule-associated protein tau promotes neuronal class II β-tubulin microtubule formation and axon elongation in embryonic Xenopus laevis. Eur J Neurosci 2015; 41:1263-75. [PMID: 25656701 DOI: 10.1111/ejn.12848] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2014] [Revised: 12/13/2014] [Accepted: 01/07/2015] [Indexed: 01/06/2023]
Abstract
Compared with its roles in neurodegeneration, much less is known about microtubule-associated protein tau's normal functions in vivo, especially during development. The external development and ease of manipulating gene expression of Xenopus laevis embryos make them especially useful for studying gene function during early development. To study tau's functions in axon outgrowth, we characterized the most prominent tau isoforms of Xenopus embryos and manipulated their expression. None of these four isoforms were strictly analogous to those commonly studied in mammals, as all constitutively contained exon 10, which is preferentially removed from mammalian fetal tau isoforms, as well as exon 8, which in mammals is rare. Nonetheless, like mammalian tau, Xenopus tau exhibited alternative splicing of exon 4a, which in mammals distinguishes 'big' tau of peripheral neurons, and exon 6. Strongly suppressing tau expression with antisense morpholino oligonucleotides only modestly compromised peripheral nerve outgrowth of intact tadpoles, but severely disrupted neuronal microtubules containing class II β-tubulins while leaving other microtubules largely unperturbed. Thus, the relatively mild dependence of axon development on tau likely resulted from having only a single class of microtubules disrupted by its loss. Also, consistent with its greater expression in long peripheral axons, boosting expression of 'big' tau increased neurite outgrowth significantly and enhanced tubulin acetylation more so than did the smaller isoform. These data demonstrate the utility of Xenopus as a tool to gain new insights into tau's functions in vivo.
Collapse
Affiliation(s)
- Yuanyuan Liu
- Department of Biological Sciences and the Center for Neuroscience Research, University at Albany, State University of New York, 1400 Washington Avenue, Albany, NY, 12222, USA
| | - Chen Wang
- Department of Biological Sciences and the Center for Neuroscience Research, University at Albany, State University of New York, 1400 Washington Avenue, Albany, NY, 12222, USA
| | - Giovanny Destin
- Department of Biological Sciences and the Center for Neuroscience Research, University at Albany, State University of New York, 1400 Washington Avenue, Albany, NY, 12222, USA
| | - Ben G Szaro
- Department of Biological Sciences and the Center for Neuroscience Research, University at Albany, State University of New York, 1400 Washington Avenue, Albany, NY, 12222, USA
| |
Collapse
|
14
|
Wang C, Szaro BG. A method for using direct injection of plasmid DNA to study cis-regulatory element activity in F0 Xenopus embryos and tadpoles. Dev Biol 2014; 398:11-23. [PMID: 25448690 DOI: 10.1016/j.ydbio.2014.11.010] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2014] [Revised: 10/31/2014] [Accepted: 11/11/2014] [Indexed: 11/17/2022]
Abstract
The ability to express exogenous reporter genes in intact, externally developing embryos, such as Xenopus, is a powerful tool for characterizing the activity of cis-regulatory gene elements during development. Although methods exist for generating transgenic Xenopus lines, more simplified methods for use with F0 animals would significantly speed the characterization of these elements. We discovered that injecting 2-cell stage embryos with a plasmid bearing a ϕC31 integrase-targeted attB element and two dual β-globin HS4 insulators flanking a reporter transgene in opposite orientations relative to each other yielded persistent expression with sufficiently high penetrance for characterizing the activity of the promoter without having to coinject integrase RNA. Expression began appropriately during development and persisted into swimming tadpole stages without perturbing the expression of the cognate endogenous gene. Coinjected plasmids having the same elements but expressing different reporter proteins were reliably coexpressed within the same cells, providing a useful control for variations in injections between animals. To overcome the high propensity of these plasmids to undergo recombination, we developed a method for generating them using conventional cloning methods and DH5α cells for propagation. We conclude that this method offers a convenient and reliable way to evaluate the activity of cis-regulatory gene elements in the intact F0 embryo.
Collapse
Affiliation(s)
- Chen Wang
- Department of Biological Sciences and the Center for Neuroscience Research, University at Albany, State University of New York, 1400 Washington Avenue, Albany, NY 12222, USA.
| | - Ben G Szaro
- Department of Biological Sciences and the Center for Neuroscience Research, University at Albany, State University of New York, 1400 Washington Avenue, Albany, NY 12222, USA.
| |
Collapse
|
15
|
Sustained neurochemical plasticity in central terminals of mouse DRG neurons following colitis. Cell Tissue Res 2014; 356:309-17. [DOI: 10.1007/s00441-014-1832-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2013] [Accepted: 01/30/2014] [Indexed: 12/16/2022]
|
16
|
Taha MF, Javeri A, Kheirkhah O, Majidizadeh T, Khalatbary AR. Neural differentiation of mouse embryonic and mesenchymal stem cells in a simple medium containing synthetic serum replacement. J Biotechnol 2014; 172:1-10. [DOI: 10.1016/j.jbiotec.2013.11.028] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2013] [Revised: 11/17/2013] [Accepted: 11/29/2013] [Indexed: 01/23/2023]
|
17
|
c-Jun N-terminal kinase phosphorylation of heterogeneous nuclear ribonucleoprotein K regulates vertebrate axon outgrowth via a posttranscriptional mechanism. J Neurosci 2013; 33:14666-80. [PMID: 24027268 DOI: 10.1523/jneurosci.4821-12.2013] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
c-Jun N-terminal kinase (JNK) mediates cell signaling essential for axon outgrowth, but the associated substrates and underlying mechanisms are poorly understood. We identified in Xenopus laevis embryos a novel posttranscriptional mechanism whereby JNK regulates axonogenesis by phosphorylating a specific site on heterogeneous nuclear ribonucleoprotein K (hnRNP K). Both JNK inhibition and hnRNP K knockdown inhibited axon outgrowth and translation of hnRNP K-regulated cytoskeletal RNAs (tau and neurofilament medium), effects that were alleviated by expressing phosphomimetic, but not phosphodeficient, forms of hnRNP K. Immunohistochemical and biochemical analyses indicated that JNK phosphorylation of hnRNP K occurred within the cytoplasm and was necessary for the translational initiation of hnRNP K-targeted RNAs but not for hnRNP K intracellular localization or RNA binding. Thus, in addition to its known roles in transcription and cytoskeletal organization, JNK acts posttranscriptionally through hnRNP K to regulate translation of proteins crucial for axonogenesis.
Collapse
|
18
|
McKeown CR, Sharma P, Sharipov HE, Shen W, Cline HT. Neurogenesis is required for behavioral recovery after injury in the visual system of Xenopus laevis. J Comp Neurol 2013; 521:2262-78. [PMID: 23238877 DOI: 10.1002/cne.23283] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2012] [Revised: 11/30/2012] [Accepted: 12/11/2012] [Indexed: 12/22/2022]
Abstract
Nonmammalian vertebrates have a remarkable capacity to regenerate brain tissue in response to central nervous system (CNS) injury. Nevertheless, it is not clear whether animals recover lost function after injury or whether injury-induced cell proliferation mediates recovery. We address these questions using the visual system and visually-guided behavior in Xenopus laevis tadpoles. We established a reproducible means to produce a unilateral focal injury to optic tectal neurons without damaging retinotectal axons. We then assayed a tectally-mediated visual avoidance behavior to evaluate behavioral impairment and recovery. Focal ablation of part of the optic tectum prevents the visual avoidance response to moving stimuli. Animals recover the behavior over the week following injury. Injury induces a burst of proliferation of tectal progenitor cells based on phospho-histone H3 immunolabeling and experiments showing that Musashi-immunoreactive tectal progenitors incorporate the thymidine analog chlorodeoxyuridine after injury. Pulse chase experiments indicate that the newly-generated cells differentiate into N-β-tubulin-immunoreactive neurons. Furthermore, in vivo time-lapse imaging shows that Sox2-expressing neural progenitors divide in response to injury and generate neurons with elaborate dendritic arbors. These experiments indicate that new neurons are generated in response to injury. To test if neurogenesis is necessary for recovery from injury, we blocked cell proliferation in vivo and found that recovery of the visual avoidance behavior is inhibited by drugs that block cell proliferation. Moreover, behavioral recovery is facilitated by changes in visual experience that increase tectal progenitor cell proliferation. Our data indicate that neurogenesis in the optic tectum is critical for recovery of visually-guided behavior after injury.
Collapse
Affiliation(s)
- Caroline R McKeown
- Department of Molecular and Cellular Neuroscience, Dorris Neuroscience Center, Scripps Research Institute, La Jolla, California 92037, USA
| | | | | | | | | |
Collapse
|
19
|
Neurulation and neurite extension require the zinc transporter ZIP12 (slc39a12). Proc Natl Acad Sci U S A 2013; 110:9903-8. [PMID: 23716681 DOI: 10.1073/pnas.1222142110] [Citation(s) in RCA: 97] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Zn(2+) is required for many aspects of neuronal structure and function. However, the regulation of Zn(2+) in the nervous system remains poorly understood. Systematic analysis of tissue-profiling microarray data showed that the zinc transporter ZIP12 (slc39a12) is highly expressed in the human brain. In the work reported here, we confirmed that ZIP12 is a Zn(2+) uptake transporter with a conserved pattern of high expression in the mouse and Xenopus nervous system. Mouse neurons and Neuro-2a cells produce fewer and shorter neurites after ZIP12 knockdown without affecting cell viability. Zn(2+) chelation or loading in cells to alter Zn(2+) availability respectively mimicked or reduced the effects of ZIP12 knockdown on neurite outgrowth. ZIP12 knockdown reduces cAMP response element-binding protein activation and phosphorylation at serine 133, which is a critical pathway for neuronal differentiation. Constitutive cAMP response element-binding protein activation restores impairments in neurite outgrowth caused by Zn(2+) chelation or ZIP12 knockdown. ZIP12 knockdown also reduces tubulin polymerization and increases sensitivity to nocodazole following neurite outgrowth. We find that ZIP12 is expressed during neurulation and early nervous system development in Xenopus tropicalis, where ZIP12 antisense morpholino knockdown impairs neural tube closure and arrests development during neurulation with concomitant reduction in tubulin polymerization in the neural plate. This study identifies a Zn(2+) transporter that is specifically required for nervous system development and provides tangible links between Zn(2+), neurulation, and neuronal differentiation.
Collapse
|
20
|
Fini JB, Le Mével S, Palmier K, Darras VM, Punzon I, Richardson SJ, Clerget-Froidevaux MS, Demeneix BA. Thyroid hormone signaling in the Xenopus laevis embryo is functional and susceptible to endocrine disruption. Endocrinology 2012; 153:5068-81. [PMID: 22968643 DOI: 10.1210/en.2012-1463] [Citation(s) in RCA: 60] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Thyroid hormone (TH) is essential for vertebrate brain development. Most research on TH and neuronal development focuses on late development, mainly the perinatal period in mammals. However, in human infants neuromotor development correlates best with maternal TH levels in the first trimester of pregnancy, suggesting that TH signaling could affect early brain development. Studying TH signaling in early embryogenesis in mammals is experimentally challenging. In contrast, free-living embryos, such as Xenopus laevis, permit physiological experimentation independent of maternal factors. We detailed key elements of TH signaling: ligands, receptors (TR), and deiodinases during early X. laevis development, before embryonic thyroid gland formation. Dynamic profiles for all components were found. Between developmental stages 37 and 41 (~48 h after hatching, coincident with a phase of continuing neurogenesis) significant increases in T(3) levels as well as in mRNA encoding deiodinases and TR occurred. Exposure of embryos at this developmental stage for 24 h to either a TH antagonist, NH-3, or to tetrabromobisphenol A, a flame retardant and known TH disruptor, differentially modulated the expression of a number of TH target genes implicated in neural stem cell function or neural differentiation. Moreover, 24-h exposure to either NH-3 or tetrabromobisphenol A diminished cell proliferation in the brain. Thus, these data show first, that TH signaling exerts regulatory roles in early X. laevis neurogenesis and second, that this period represents a potential window for endocrine disruption.
Collapse
Affiliation(s)
- J B Fini
- Unité Mixte de Recherche Centre National de la Recherche Scientifique 7221, Evolution des Régulations Endocriniennes CNRS UMR 7221/Muséum National d'Histoire Naturelle Département Régulations, Développement et Diversité Moléculaire, 75231 Paris, France
| | | | | | | | | | | | | | | |
Collapse
|
21
|
Heterogeneous nuclear ribonucleoprotein K, an RNA-binding protein, is required for optic axon regeneration in Xenopus laevis. J Neurosci 2012; 32:3563-74. [PMID: 22399778 DOI: 10.1523/jneurosci.5197-11.2012] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Axotomized optic axons of Xenopus laevis, in contrast to those of mammals, retain their ability to regenerate throughout life. To better understand the molecular basis for this successful regeneration, we focused on the role of an RNA-binding protein, heterogeneous nuclear ribonucleoprotein (hnRNP) K, because it is required for axonogenesis during development and because several of its RNA targets are under strong post-transcriptional control during regeneration. At 11 d after optic nerve crush, hnRNP K underwent significant translocation into the nucleus of retinal ganglion cells (RGCs), indicating that the protein became activated during regeneration. To suppress its expression, we intravitreously injected an antisense Vivo-Morpholino oligonucleotide targeting hnRNP K. In uninjured eyes, it efficiently knocked down hnRNP K expression in only the RGCs, without inducing either an axotomy response or axon degeneration. After optic nerve crush, staining for multiple markers of regenerating axons showed no regrowth of axons beyond the lesion site with hnRNP K knockdown. RGCs nonetheless responded to the injury by increasing expression of multiple growth-associated RNAs and experienced no additional neurodegeneration above that normally seen with optic nerve injury. At the molecular level, hnRNP K knockdown during regeneration inhibited protein, but not mRNA, expression of several known hnRNP K RNA targets (NF-M, GAP-43) by compromising their efficient nuclear transport and disrupting their loading onto polysomes for translation. Our study therefore provides evidence of a novel post-transcriptional regulatory pathway orchestrated by hnRNP K that is essential for successful CNS axon regeneration.
Collapse
|
22
|
Lepore G, Gadau S, Mura A, Zedda M, Farina V. Aromatase immunoreactivity in fetal ovine neuronal cell cultures exposed to oxidative injury. Eur J Histochem 2012; 53:e28. [PMID: 22073360 PMCID: PMC3167340 DOI: 10.4081/ejh.2009.e28] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/05/2009] [Indexed: 12/03/2022] Open
Abstract
A lot of evidence testifies that aromatase is expressed in the central nervous system where it has been detected not only in hypothalamic and limbic regions but also in the cerebral cortex and spinal cord. In physiological conditions, aromatase is expressed exclusively by neurons, where it has been mainly found in cell bodies, processes and synaptic terminals. Moreover, primary cultured cortical astrocytes from female rats are more resistant to oxidant cell death than those from males, suggesting a protective role of estradiol. The aim of this study was to evaluate changes in aromatase expression in response to 3-nitro-L-tyrosine, a marker of oxidative stress, in primary neuronal cell cultures from brains of 60-day old sheep fetuses. Cells were identified as neurons by using class III β-tubulin, a marker of neuronal cells. Two morphological types were consistently recognizable: i) bipolar cells with an oval cell body; ii) multipolar cells whose processes formed a wide net with those of adjacent cells. In situ hybridization technique performed on 60-day old fetal neurons revealed that in baseline conditions aromatase gene expression occurs. Importantly, cells exposed to 360 µM 3-nitro-L-tyrosine were fewer and showed more globular shape and shorter cytoplasmic processes in comparison to control cells. The immunocytochemical study with anti-aromatase antibody revealed that cells exposed to 360 µM 3-nitro-L-tyrosine were significantly more immunoreactive than control cells. Thus, it can be postulated that the oxidant effects of the amino acid analogue 3-nitro-L-tyrosine could be counterbalanced by an increase in aromatase expression that in turn can lead to the formation of neuroprotective estradiol via aromatization of testosterone.
Collapse
Affiliation(s)
- G Lepore
- Department of Animal Biology, University of Sassari, Italy.
| | | | | | | | | |
Collapse
|
23
|
Rogers CD, Ferzli GS, Casey ES. The response of early neural genes to FGF signaling or inhibition of BMP indicate the absence of a conserved neural induction module. BMC DEVELOPMENTAL BIOLOGY 2011; 11:74. [PMID: 22172147 PMCID: PMC3271986 DOI: 10.1186/1471-213x-11-74] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/28/2011] [Accepted: 12/15/2011] [Indexed: 01/06/2023]
Abstract
BACKGROUND The molecular mechanism that initiates the formation of the vertebrate central nervous system has long been debated. Studies in Xenopus and mouse demonstrate that inhibition of BMP signaling is sufficient to induce neural tissue in explants or ES cells respectively, whereas studies in chick argue that instructive FGF signaling is also required for the expression of neural genes. Although additional signals may be involved in neural induction and patterning, here we focus on the roles of BMP inhibition and FGF8a. RESULTS To address the question of necessity and sufficiency of BMP inhibition and FGF signaling, we compared the temporal expression of the five earliest genes expressed in the neuroectoderm and determined their requirements for induction at the onset of neural plate formation in Xenopus. Our results demonstrate that the onset and peak of expression of the genes vary and that they have different regulatory requirements and are therefore unlikely to share a conserved neural induction regulatory module. Even though all require inhibition of BMP for expression, some also require FGF signaling; expression of the early-onset pan-neural genes sox2 and foxd5α requires FGF signaling while other early genes, sox3, geminin and zicr1 are induced by BMP inhibition alone. CONCLUSIONS We demonstrate that BMP inhibition and FGF signaling induce neural genes independently of each other. Together our data indicate that although the spatiotemporal expression patterns of early neural genes are similar, the mechanisms involved in their expression are distinct and there are different signaling requirements for the expression of each gene.
Collapse
Affiliation(s)
- Crystal D Rogers
- Department of Biology, Georgetown University, Washington DC, USA
| | - George S Ferzli
- Department of Biology, Georgetown University, Washington DC, USA
| | - Elena S Casey
- Department of Biology, Georgetown University, Washington DC, USA
| |
Collapse
|
24
|
Szaro BG, Strong MJ. Regulation of Cytoskeletal Composition in Neurons: Transcriptional and Post-transcriptional Control in Development, Regeneration, and Disease. ADVANCES IN NEUROBIOLOGY 2011. [DOI: 10.1007/978-1-4419-6787-9_24] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/12/2023]
|
25
|
Sharma P, Cline HT. Visual activity regulates neural progenitor cells in developing xenopus CNS through musashi1. Neuron 2010; 68:442-55. [PMID: 21040846 DOI: 10.1016/j.neuron.2010.09.028] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/19/2010] [Indexed: 11/30/2022]
Abstract
Regulation of progenitor cell fate determines the numbers of neurons in the developing brain. While proliferation of neural progenitors predominates during early central nervous system (CNS) development, progenitor cell fate shifts toward differentiation as CNS circuits develop, suggesting that signals from developing circuits may regulate proliferation and differentiation. We tested whether activity regulates neurogenesis in vivo in the developing visual system of Xenopus tadpoles. Both cell proliferation and the number of musashi1-immunoreactive progenitors in the optic tectum decrease as visual system connections become stronger. Visual deprivation for 2 days increased proliferation of musashi1-immunoreactive radial glial progenitors, while visual experience increased neuronal differentiation. Morpholino-mediated knockdown and overexpression of musashi1 indicate that musashi1 is necessary and sufficient for neural progenitor proliferation in the CNS. These data demonstrate a mechanism by which increased brain activity in developing circuits decreases cell proliferation and increases neuronal differentiation through the downregulation of musashi1 in response to circuit activity.
Collapse
Affiliation(s)
- Pranav Sharma
- The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, CA 92037, USA
| | | |
Collapse
|
26
|
Abstract
Tubulin antibodies are among the most extensively used immunological reagents in basic and applied cell and molecular biology. In this chapter, we provide a brief overview of the practices and reagents developed in our laboratory during the past 25 years for characterizing anti-beta-tubulin antibodies.
Collapse
Affiliation(s)
- Anthony J Spano
- Department of Biology, University of Virginia, Charlottesville, Virginia 22904, USA
| | | |
Collapse
|
27
|
Brundage CM, Taylor BE. Timing and duration of developmental nicotine exposure contribute to attenuation of the tadpole hypercapnic neuroventilatory response. Dev Neurobiol 2009; 69:451-61. [PMID: 19360722 DOI: 10.1002/dneu.20720] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
The ability for air-breathing vertebrates to adjust ventilation in response to increased CO(2) (hypercapnia) is fundamental to maintaining pH homeostasis. Developmental nicotine exposure has been shown to impair tadpole neuroventilatory responses to hypercapnia following 8-12 weeks of exposure. It is not clear, however, to what extent the timing of exposure during development and/or the duration over which the exposure takes place contribute to this impairment. Here, tadpoles were exposed to 30 microg/L of nicotine for 3- or 10-week durations, either early or late in tadpole development. Correlates of tadpole lung neuroventilation were monitored during normocapnic (1.5% CO(2)) and hypercapnic (5% CO(2)) conditions of isolated brainstems. Preparations derived from early metamorphic tadpoles failed to increase lung neuroventilation in response to hypercapnia whether they had been exposed to nicotine for 3 or 10 weeks. Preparations derived from late metamorphic tadpoles failed to respond to hypercapnia after being exposed to nicotine for 10 weeks. These results suggest that both the developmental timing and duration of exposure are important when considering nicotine's effect on the hypercapnic neuroventilatory response.
Collapse
Affiliation(s)
- Cord M Brundage
- Institute of Arctic Biology, Department of Biology and Wildlife, University of Alaska Fairbanks, Fairbanks, AK, USA
| | | |
Collapse
|
28
|
Generation of stable Xenopus laevis transgenic lines expressing a transgene controlled by weak promoters. Transgenic Res 2009; 18:815-27. [DOI: 10.1007/s11248-009-9273-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2009] [Accepted: 04/16/2009] [Indexed: 10/20/2022]
|
29
|
Liu Y, Gervasi C, Szaro BG. A crucial role for hnRNP K in axon development in Xenopus laevis. Development 2008; 135:3125-35. [PMID: 18725517 DOI: 10.1242/dev.022236] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
We report that hnRNP K, an RNA-binding protein implicated in multiple aspects of post-transcriptional gene control, is essential for axon outgrowth in Xenopus. Its intracellular localization was found to be consistent with one of its known roles as an mRNA shuttling protein. In early embryos, it was primarily nuclear, whereas later it occupied both the nucleus and cytoplasm to varying degrees in different neuronal subtypes. Antisense hnRNP K morpholino oligonucleotides (MOs) microinjected into blastomeres suppressed hnRNP K expression from neural plate stages through to at least stage 40. Differentiating neural cells in these embryos expressed several markers for terminally differentiated neurons but failed to make axons. Rescue experiments and the use of two separate hnRNP K MOs were carried out to confirm that these effects were specifically caused by knockdown of hnRNP K expression. For insights into the involvement of hnRNP K in neuronal post-transcriptional gene control at the molecular level, we compared effects on expression of the medium neurofilament protein (NF-M), the RNA for which binds hnRNP K, with that of peripherin, another intermediate filament protein, the RNA for which does not bind hnRNP K. hnRNP K knockdown compromised NF-M mRNA nucleocytoplasmic export and translation, but had no effect on peripherin. Because eliminating NF-M from Xenopus axons attenuates, but does not abolish, their outgrowth, hnRNP K must target additional RNAs needed for axon development. Our study supports the idea that translation of at least a subset of RNAs involved in axon development is controlled by post-transcriptional regulatory modules that have hnRNP K as an essential element.
Collapse
Affiliation(s)
- Yuanyuan Liu
- The Department of Biological Sciences and the Center for Neuroscience Research, University at Albany, State University of New York, Albany, NY 12222, USA
| | | | | |
Collapse
|
30
|
Verdier-Pinard P, Pasquier E, Xiao H, Burd B, Villard C, Lafitte D, Miller LM, Angeletti RH, Horwitz SB, Braguer D. Tubulin proteomics: towards breaking the code. Anal Biochem 2008; 384:197-206. [PMID: 18840397 DOI: 10.1016/j.ab.2008.09.020] [Citation(s) in RCA: 46] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2008] [Revised: 09/12/2008] [Accepted: 09/15/2008] [Indexed: 01/02/2023]
Affiliation(s)
- Pascal Verdier-Pinard
- INSERM UMR 911 CRO2, Aix-Marseille Université, Faculté de Pharmacie, 27 bd Jean Moulin, 13285 Marseille cedex 05, France.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Falk J, Drinjakovic J, Leung KM, Dwivedy A, Regan AG, Piper M, Holt CE. Electroporation of cDNA/Morpholinos to targeted areas of embryonic CNS in Xenopus. BMC DEVELOPMENTAL BIOLOGY 2007; 7:107. [PMID: 17900342 PMCID: PMC2147031 DOI: 10.1186/1471-213x-7-107] [Citation(s) in RCA: 64] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/08/2007] [Accepted: 09/27/2007] [Indexed: 02/07/2023]
Abstract
Background Blastomere injection of mRNA or antisense oligonucleotides has proven effective in analyzing early gene function in Xenopus. However, functional analysis of genes involved in neuronal differentiation and axon pathfinding by this method is often hampered by earlier function of these genes during development. Therefore, fine spatio-temporal control of over-expression or knock-down approaches is required to specifically address the role of a given gene in these processes. Results We describe here an electroporation procedure that can be used with high efficiency and low toxicity for targeting DNA and antisense morpholino oligonucleotides (MOs) into spatially restricted regions of the Xenopus CNS at a critical time-window of development (22–50 hour post-fertilization) when axonal tracts are first forming. The approach relies on the design of "electroporation chambers" that enable reproducible positioning of fixed-spaced electrodes coupled with accurate DNA/MO injection. Simple adjustments can be made to the electroporation chamber to suit the shape of different aged embryos and to alter the size and location of the targeted region. This procedure can be used to electroporate separate regions of the CNS in the same embryo allowing separate manipulation of growing axons and their intermediate and final targets in the brain. Conclusion Our study demonstrates that electroporation can be used as a versatile tool to investigate molecular pathways involved in axon extension during Xenopus embryogenesis. Electroporation enables gain or loss of function studies to be performed with easy monitoring of electroporated cells. Double-targeted transfection provides a unique opportunity to monitor axon-target interaction in vivo. Finally, electroporated embryos represent a valuable source of MO-loaded or DNA transfected cells for in vitro analysis. The technique has broad applications as it can be tailored easily to other developing organ systems and to other organisms by making simple adjustments to the electroporation chamber.
Collapse
Affiliation(s)
- Julien Falk
- Department of Physiology, Development and Neuroscience, University of Cambridge, Downing Street, Cambridge CB2 3DY, UK
| | - Jovana Drinjakovic
- Department of Physiology, Development and Neuroscience, University of Cambridge, Downing Street, Cambridge CB2 3DY, UK
| | - Kin Mei Leung
- Department of Physiology, Development and Neuroscience, University of Cambridge, Downing Street, Cambridge CB2 3DY, UK
| | - Asha Dwivedy
- Department of Physiology, Development and Neuroscience, University of Cambridge, Downing Street, Cambridge CB2 3DY, UK
| | - Aoife G Regan
- Department of Physiology, Development and Neuroscience, University of Cambridge, Downing Street, Cambridge CB2 3DY, UK
| | - Michael Piper
- Department of Physiology, Development and Neuroscience, University of Cambridge, Downing Street, Cambridge CB2 3DY, UK
- The Queensland Brain Institute, The University of Queensland, St Lucia, QLD, 4072, Australia
| | - Christine E Holt
- Department of Physiology, Development and Neuroscience, University of Cambridge, Downing Street, Cambridge CB2 3DY, UK
| |
Collapse
|
32
|
Huang T, He D, Kleiner G, Kuluz J. Neuron-like differentiation of adipose-derived stem cells from infant piglets in vitro. J Spinal Cord Med 2007; 30 Suppl 1:S35-40. [PMID: 17874685 PMCID: PMC2031970 DOI: 10.1080/10790268.2007.11753967] [Citation(s) in RCA: 83] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/21/2022] Open
Abstract
BACKGROUND/OBJECTIVE Adipose-derived stem cells (ADSCs) are mesenchymal stem cells (MSCs) that can be extracted from adipose tissue and obtained by a less invasive method and in larger quantities compared with bone marrow-derived MSCs. The objective of this study was to harvest ADSCs from piglets and to explore their neuronal differentiation potential. METHODS Adipose tissue from piglet facial or abdominal fat was digested with collagenase type XI, followed by filter and centrifugation; the isolated adipose stromal cells were cultured in dishes. MSC markers were measured by flow cytometry; 2 to 5 passage cells were used for in vitro differentiation. Adipogenic, chondrogenic, osteogenic, and neuronal differentiation was induced by incubation of the ADSCs with different induction media. RESULTS ADSCs were easily expanded to beyond 15 passages, maintaining the undifferentiated state and exhibiting MSC characteristics and markers CD29, CD44, and CD90. ADSCs differentiated into other mesodermal cells including adipocytes, chondrocytes, and osteocytes. These cells were induced to differentiate into neuron-like cells as evidenced by neuronal morphology and the presence of neuronal markers including microtubule-associated protein 2, neuronal nuclear antigen, and beta-tubulin III. CONCLUSIONS ADSCs can be readily obtained from a small amount fat tissue and expanded in culture. Adipose tissue may be an alternative source of stem cell therapy for nervous system injury.
Collapse
Affiliation(s)
- Tingting Huang
- University of Miami School of Medicine, Miami, Florida
- Please address correspondence to John Kuluz, MD, Pediatric ICU, 6006 Holtz Center—JMH East Tower R131, Miami, FL 33136; phone: 303.243.3399; fax: 303.243.4607; (e-mail: )
| | - Dansha He
- University of Miami School of Medicine, Miami, Florida
| | - Gary Kleiner
- University of Miami School of Medicine, Miami, Florida
| | - John Kuluz
- University of Miami School of Medicine, Miami, Florida
| |
Collapse
|
33
|
Smith A, Gervasi C, Szaro BG. Neurofilament content is correlated with branch length in developing collateral branches of Xenopus spinal cord neurons. Neurosci Lett 2006; 403:283-7. [PMID: 16725258 DOI: 10.1016/j.neulet.2006.04.055] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2006] [Revised: 04/26/2006] [Accepted: 04/29/2006] [Indexed: 11/29/2022]
Abstract
During development, axons form interstitial collateral branches, which are initially dynamic but gradually stabilize as the projection sharpens. The initial outgrowth of collaterals is characterized by transitions in growth dynamics that occur at different lengths. Below 10 microm, collateral branches start out as unstable, thin filopodia. Above 30 microm, the branches stabilize. Although the relationship between branch length and the presence of microfilaments and microtubules has been well characterized, relatively less is known about the development of the neurofilament cytoskeleton in collateral branches. In the main axon, successive stages of outgrowth are accompanied by changes in the polypeptide composition of neurofilaments (NFs), which shifts from being rich in Type III neuronal intermediate filament proteins (nIFs) to progressively favoring Type IV subunits. To characterize the NF composition of developing collateral branches, antibodies to peripherin (a Type III nIF) and NF-M (a Type IV nIF) were used to stain newly differentiating embryonic Xenopus laevis spinal cord neurons in culture. In contrast to what happens in the main axon, staining for both subunits coincided in collaterals. Branches shorter than 10 microm seldom had NFs, whereas all branches longer than 30 microm did. In branches that had NFs staining either extended all the way to branch tip or terminated approximately 10mum from it. These lengths correspond remarkably well with lengths associated with branch stabilization. Given that NFs are the most stable of the cytoskeletal polymers, we speculate that they may contribute to this stabilization.
Collapse
Affiliation(s)
- Andrew Smith
- Department of Biological Sciences and the Neuroscience Research Center, University at Albany, State University of New York, 1400 Washington Avenue, Albany, NY 12222, USA
| | | | | |
Collapse
|
34
|
Yoshino J, Tochinai S. Functional regeneration of the olfactory bulb requires reconnection to the olfactory nerve in Xenopus larvae. Dev Growth Differ 2006; 48:15-24. [PMID: 16466389 DOI: 10.1111/j.1440-169x.2006.00840.x] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Larvae of the South African clawed frog (Xenopus laevis) can regenerate the telencephalon, which consists of the olfactory bulb and the cerebrum, after it has been partially removed. Some authors have argued that the telencephalon, once removed, must be reconnected to the olfactory nerve in order to regenerate. However, considerable regeneration has been observed before reconnection. Therefore, we have conducted several experiments to learn whether or not reconnection is a prerequisite for regeneration. We found that the olfactory bulb did not regenerate without reconnection, while the cerebrum regenerated by itself. On the other hand, when the brain was reconnected by the olfactory nerve, both the cerebrum and the olfactory bulb regenerated. Morphological and histological investigation showed that the regenerated telencephalon was identical to the intact one in morphology, types and distributions of cells, and connections between neurons. Froglets with a regenerated telencephalon also recovered olfaction, the primary function of the frog telencephalon. These results suggest that the Xenopus larva requires reconnection of the regenerating brain to the olfactory nerve in order to regenerate the olfactory bulb, and thus the regenerated brain functions, in order to process olfactory information.
Collapse
Affiliation(s)
- Jun Yoshino
- Division of Biological Sciences, Graduate School of Science, Hokkaido University, N10W8, Sapporo 060-0810, Japan.
| | | |
Collapse
|
35
|
Dickinson AJG, Sive H. Development of the primary mouth in Xenopus laevis. Dev Biol 2006; 295:700-13. [PMID: 16678148 DOI: 10.1016/j.ydbio.2006.03.054] [Citation(s) in RCA: 46] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2006] [Revised: 03/30/2006] [Accepted: 03/31/2006] [Indexed: 11/19/2022]
Abstract
The initial opening between the gut and the outside of the deuterostome embryo breaks through at the extreme anterior. This region is unique in that ectoderm and endoderm are directly juxtaposed, without intervening mesoderm. This opening has been called the stomodeum, buccopharyngeal membrane or oral cavity at various stages of its formation, however, in order to clarify its function, we have termed this the "primary mouth". In vertebrates, the neural crest grows around the primary mouth to form the face and a "secondary mouth" forms. The primary mouth then becomes the pharyngeal opening. In order to establish a molecular understanding of primary mouth formation, we have begun to examine this process during Xenopus laevis development. An early step during this process occurs at tailbud and involves dissolution of the basement membrane between the ectoderm and endoderm. This is followed by ectodermal invagination to create the stomodeum. A subsequent step involves localized cell death in the ectoderm, which may lead to ectodermal thinning. Subsequently, ectoderm and endoderm apparently intercalate to generate one to two cell layers. The final step is perforation, where (after hatching) the primary mouth opens. Fate mapping has defined the ectodermal and endodermal regions that will form the primary mouth. Extirpations and transplants of these and adjacent regions indicate that, at tailbud, the oral ectoderm is not specifically required for primary mouth formation. In contrast, underlying endoderm and surrounding regions are crucial, presumably sources of necessary signals. This study indicates the complexity of primary mouth formation, and lays the groundwork for future molecular analyses of this important structure.
Collapse
|
36
|
Kay GW, Oswald MJ, Palmer DN. The development and characterisation of complex ovine neuron cultures from fresh and frozen foetal neurons. J Neurosci Methods 2006; 155:98-108. [PMID: 16487596 PMCID: PMC1525139 DOI: 10.1016/j.jneumeth.2006.01.008] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2005] [Revised: 11/21/2005] [Accepted: 01/06/2006] [Indexed: 10/25/2022]
Abstract
Cultures of ovine cerebral and cerebellar neurons from mid-term sheep foetal brains, 9-15 weeks old, have been established for the first time. These foetal brains are relatively mature, being at similar stages of development as peri and post-natal rodent brains. Cultures were routinely maintained for 3-4 weeks, and longer. Nearly all the cells from the younger foetuses adhered as neurons. The proportion of glial cells increased with age, as did the risk of cultures being overtaken by glial cells. Cultured neurons were bipolar, tripolar and multipolar, similar to the morphologies of neurons in vivo. Older foetuses also yield more complex neurons, notably giant cells. Other properties of the cultured neurons also mimic in vivo observations, including neurite beading, complexity in neurotransmitter class (GABAergic and glutamatergic) and calcium binding protein (calbindin and calretinin) content. Single cell divisions of neurons were observed in younger cultures by time-lapse photography and the occurrence of telophase nuclei. The advantage of the high yield of genetically identical cells obtained from a single sheep foetus, 150 million, was extended by cryopreservation of neurons after snap freezing, and later culture. These cultures showed the same characteristics as cultures from the freshly plated cells.
Collapse
Affiliation(s)
| | | | - David N. Palmer
- * Correspondence to: Dr D N Palmer, Agricultural and Life Sciences Division, PO Box 84, Lincoln University, Canterbury, New Zealand, Tel.: +64-3-325-2811, Fax : +64-3-325-3851, E-mail:
| |
Collapse
|
37
|
Kim J, Lauderdale JD. Analysis of Pax6 expression using a BAC transgene reveals the presence of a paired-less isoform of Pax6 in the eye and olfactory bulb. Dev Biol 2006; 292:486-505. [PMID: 16464444 DOI: 10.1016/j.ydbio.2005.12.041] [Citation(s) in RCA: 68] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2005] [Revised: 09/08/2005] [Accepted: 12/21/2005] [Indexed: 11/19/2022]
Abstract
Pax6, a member of the paired-family of transcription factors, exhibits restricted expression and essential functions in the developing eye, olfactory system, central nervous system, and pancreas. To understand Pax6 function, which critically depends on induction of proper expression levels during development, it is necessary to elucidate the molecular mechanisms governing Pax6 transcription. Although previous studies using classic transgenic approaches have provided a wealth of information about the distribution and types of regulatory elements involved in Pax6 regulation, genetic studies in both humans and mice indicate that these enhancers alone are not sufficient for fully regulated Pax6 expression. We report here our analysis of mice transgenic for a 160 kb mouse Pax6 BAC transgene, which was generated as a necessary first step towards testing the long-range control of Pax6 expression in vivo. We show that this BAC transgene replicates Pax6 expression in the eye. This is the first time that a reporter transgene has been expressed in a normal Pax6-like pattern in all of the tissues of the eye and defines an eye regulatory region within the Pax6 downstream regulatory region (DRR). Second, we show that this BAC transgene contains all of the cis regulatory elements required for normal Pax6 expression within the developing embryo, except for within the diencephalon and olfactory bulb. Third, we show that this transgene is subject to Pax6 autoregulation. Lastly, we identify, for the first time in mammals, an isoform of the Pax6 protein lacking the paired domain. This isoform is expressed in the developing olfactory bulb and eye. Over-expression of Pax6DeltaPD causes a microphthalmic phenotype in both Pax6(+/+) mice and Pax6(+/-) mice. These results demonstrate a role for Pax6DeltaPD in eye development, which appears to be different than that ascribed to either canonical Pax6 or Pax6(5a).
Collapse
Affiliation(s)
- Jiha Kim
- Department of Cellular Biology, The University of Georgia, Athens, GA 30602, USA
| | | |
Collapse
|
38
|
Huang S, Yan B, Sullivan SA, Moody SA. Noggin signaling fromXenopus animal blastomere lineages promotes a neural fate in neighboring vegetal blastomere lineages. Dev Dyn 2006; 236:171-83. [PMID: 17096409 DOI: 10.1002/dvdy.20944] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
In Xenopus, localized factors begin to regionalize embryonic fates prior to the inductive interactions that occur during gastrulation. We previously reported that an animal-to-vegetal signal that occurs prior to gastrulation promotes primary spinal neuron fate in vegetal equatorial (C-tier) blastomere lineages. Herein we demonstrate that maternal mRNA encoding noggin is enriched in animal tiers and at low concentrations in the C-tier, suggesting that the neural fates of C-tier blastomeres may be responsive to early signaling from their neighboring cells. In support of this hypothesis, experimental alteration of the levels of Noggin from animal equatorial (B-tier) or BMP4 from vegetal (D-tier) blastomeres significantly affects the numbers of primary spinal neurons derived from their neighboring C-tier blastomeres. These effects are duplicated in blastomere explants isolated at cleavage stages and cultured in the absence of gastrulation interactions. Co-culture with animal blastomeres enhanced the expression of zygotic neural markers in C-tier blastomere explants, whereas co-culture with vegetal blastomeres repressed them. The expression of these markers in C-tier explants was promoted when Noggin was transiently added to the culture during cleavage/morula stages, and repressed with the transient addition of BMP4. Reduction of Noggin translation in B-tier blastomeres by antisense morpholino oligonucleotides significantly reduced the efficacy of neural marker induction in C-tier explants. These experiments indicate that early anti-BMP signaling from the animal hemisphere recruits vegetal equatorial cells into the neural precursor pool prior to interactions that occur during gastrulation.
Collapse
Affiliation(s)
- Sen Huang
- Department of Anatomy and Cell Biology, George Washington University, Washington, DC 20037, USA
| | | | | | | |
Collapse
|
39
|
Moon MS, Gomez TM. Adjacent pioneer commissural interneuron growth cones switch from contact avoidance to axon fasciculation after midline crossing. Dev Biol 2005; 288:474-86. [PMID: 16293241 DOI: 10.1016/j.ydbio.2005.09.049] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2005] [Revised: 09/21/2005] [Accepted: 09/30/2005] [Indexed: 11/23/2022]
Abstract
Commissural interneurons (CI) of the vertebrate spinal cord are guided ventrally toward the floor plate, but subsequently cross the midline and select a longitudinal fascicle at specific dorsal-ventral (D-V) positions. We examined at high resolution the detailed behaviors of individual pathfinding CI growth cones on the ipsilateral and contralateral sides of the spinal cord of living Xenopus embryos. We find that pre-crossing CI growth cones exhibit distinct pathfinding behaviors compared to post-crossing axons and that the behavioral switch occurs immediately upon crossing to the contralateral side. Groups of pioneer commissural axons typically extend simultaneously toward the ventral midline following discrete paths with separation between adjacent commissurals apparently maintained through contact inhibition. In contrast, shortly after crossing the midline, commissural axons turn longitudinally and begin to fasciculate with other crossed CIs. However, growth cones of crossed commissurals often select their final D-V longitudinal track through a series of rapid step-like dorsal adjustments that may be due to differential fasciculation with longitudinal axons. Together, our results suggest that guidance of commissural axons is controlled in part through interactions among CIs that switch rapidly from avoidance to fasciculation after midline crossing.
Collapse
Affiliation(s)
- Myung-Soon Moon
- Department of Anatomy, University of Wisconsin, 257 Bardeen Labs-SMI, 1300 University Ave., Madison, WI 53706, USA
| | | |
Collapse
|
40
|
Mas C, Guimiot-Maloum I, Guimiot F, Khelfaoui M, Nepote V, Bourgeois F, Boda B, Levacher B, Galat A, Moalic JM, Simonneau M. Molecular cloning and expression pattern of the Fkbp25 gene during cerebral cortical neurogenesis. Gene Expr Patterns 2005; 5:577-85. [PMID: 15908283 DOI: 10.1016/j.modgep.2005.03.006] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2004] [Revised: 03/22/2005] [Accepted: 03/22/2005] [Indexed: 11/18/2022]
Abstract
Neocortical neurons are generated predominantly from the cells that proliferate in the ventricular zone of the telencephalon. In order to understand the nature of these expanding cortical neuronal progenitor cells, we selected by differential display some transcripts that were enriched in the telencephalon as compared to the more caudal regions (diencephalon/mesencephalon). This systematic screening revealed one of the differentially expressed transcripts, namely the Fkbp25 mRNA that encodes a member of the FK506 binding proteins (FKBPs). Northern blot analysis showed that the expression of the single 1.4kb Fkbp25 transcript reached a maximum level on embryonic day 11.5 at the start of cortical neurogenesis in the mouse and was followed by a weak basal expression in the adult brain. In the embryo, Fkbp25 gene was strongly expressed in the telencephalon ventricular zone but also in areas active in myogenesis (walls of the ventricle and the atrium) and chondrogenesis (the cartilage of the rib and the hindlimb). An increase in the transcript levels of the Fkbp25 gene was also observed during the two successive proliferation waves of the cerebellum development. Immunostaining on primary cultures of embryonic day 10.5 telencephalon stem cells showed that the Fkbp25 protein was present in the cytoplasm and nuclei of cells cultured for 6h but exclusively in the nuclei of the Tuj-1 immunoreactive neurons obtained after 3 days of culture (The sequence data reported here have been submitted to GenBank under accession no. AF135595.).
Collapse
Affiliation(s)
- Christophe Mas
- Neurogénétique INSERM E9935, Hôpital Robert Debré, Paris, France
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Yamasaki Y, Kayashima T, Soejima H, Kinoshita A, Yoshiura KI, Matsumoto N, Ohta T, Urano T, Masuzaki H, Ishimaru T, Mukai T, Niikawa N, Kishino T. Neuron-specific relaxation of Igf2r imprinting is associated with neuron-specific histone modifications and lack of its antisense transcript Air. Hum Mol Genet 2005; 14:2511-20. [PMID: 16037066 DOI: 10.1093/hmg/ddi255] [Citation(s) in RCA: 60] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
The mouse insulin-like growth factor II receptor (Igf2r) gene and its antisense transcript Air are reciprocally imprinted in most tissues, but in the brain, Igf2r is biallelically expressed despite the imprinted Air expression. To investigate the molecular mechanisms of such brain-specific relaxation of Igf2r imprinting, we analyzed its expression and epigenetic modifications in neurons, glial cells and fibroblasts by the use of primary cortical cell cultures. In glial cells and fibroblasts, Igf2r was maternally expressed and Air was paternally expressed, whereas in the primary cultured neurons, Igf2r was biallelically expressed and Air was not expressed. In the differentially methylated region 2 (DMR2), which includes the Air promoter, allele-specific DNA methylation, differential H3 and H4 acetylation and H3K4 and K9 di-methylation were maintained in each cultured cell type. In DMR1, which includes the Igf2r promoter, maternal-allele-specific DNA hypomethylation, histones H3 and H4 acetylation and H3K4 di-methylation were apparent in glial cells and fibroblasts. However, in neurons, biallelic DNA hypomethylation and biallelic histones H3 and H4 acetylation and H3K4 di-methylation were detected. These data indicate that lack of reciprocal imprinting of Igf2r and Air in the brain results from neuron-specific relaxation of Igf2r imprinting associated with neuron-specific histone modifications in DMR1 and lack of Air expression. Our observation of biallelic Igf2r expression with no Air expression in neurons sheds light on the function of Air as a critical effector in Igf2r silencing and suggests that neuron-specific epigenetic modifications related to the lineage determination of neural stem cells play a critical role in controlling imprinting by antisense transcripts.
Collapse
Affiliation(s)
- Yoko Yamasaki
- Department of Human Genetics, Graduate School of Biomedical Sciences, Nagasaki 852-8523, Japan
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Oda E, Nakamura Y, Yamamoto M, Kojiro M. Immunohistochemical distribution of tubulin beta II in human normal and neoplastic tissues. Kurume Med J 2005; 52:117-25. [PMID: 16639982 DOI: 10.2739/kurumemedj.52.117] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/08/2023]
Abstract
Tubulin is the major constituent protein of microtubules. In mammals, there are seven beta-tubulins and six alpha-tubulins. Each beta-tubulin isotype has a unique tissue distribution. The purpose of this study was to describe the distribution of tubulin beta II in normal and neoplastic human tissues with immunohistochemical techniques. We obtained normal tissues from 33 cases (8 fetuses, 17 neonates, 3 children and 5 adults) and 121 samples of neoplastic tissue from surgical specimens or at autopsy. Immunohistochemical staining for tubulin beta II was performed using a monoclonal antibody, KNY379 developed in our laboratory. Tubulin beta II was detected in various normal tissues, particularly in fetal and neonatal tissues, such as the nervous system, pulmonary alveoli, bronchioles and bronchi, colon, pancreatic ducts and acini, renal convoluted tubuli, skin epidermis, body cavity mesothelial cells, smooth muscle and thymus. In the adult, broad expression was also observed; however, the immunoreactivity was weaker and the extent of its distribution decreased with age. In neoplastic tissues, tubulin beta II immunoreactivity was detected in various nervous system neoplasms and other neoplasms such as pancreatic solid cystic carcinoma, pleomorphic adenoma, Warthin's tumor, nephroblastoma, basal cell carcinoma and malignant mesothelioma. We conclude that our monoclonal antibody, KNY379, may be useful as a marker of nervous system neoplasm, pancreatic solid cystic carcinoma, pleomorphic adenoma, Warthin's tumor, nephroblastoma, basal cell carcinoma and malignant mesothelioma.
Collapse
Affiliation(s)
- Eriko Oda
- Department of Chemistry, Kurume University School of Medicine, Kurume 830-0011, Japan.
| | | | | | | |
Collapse
|
43
|
Pandur PD, Dirksen ML, Moore KB, Moody SA. Xenopus flotillin1, a novel gene highly expressed in the dorsal nervous system. Dev Dyn 2004; 231:881-7. [PMID: 15517583 DOI: 10.1002/dvdy.20191] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
The two paralogues of the Xenopus flotillin1 gene (flotillin1A and flotillin1B), which encodes a putative membrane-associated protein, were cloned from egg, cleavage, and tadpole cDNA libraries. Both mRNAs are present during oogenesis and cleavage stages. After the onset of zygotic transcription, flotillin1 transcripts are first expressed throughout the embryonic ectoderm and become enhanced in the presumptive neural ectoderm as the neural plate forms. As the neural tube forms and differentiates, flotillin1 transcripts become enriched in the dorsal half, with particularly high expression in dorsal primary neurons. At early tail bud stages, there is additional expression in the paraxial mesoderm. At late tail bud stages, flotillin1A is expressed in branchial arch mesenchyme, the overlying branchial ectoderm and in dorsal somitic mesoderm, whereas flotillin1B expression is more restricted in the dorsal neural tube and head sensory structures. This report is the first comprehensive developmental description in any animal of the expression pattern of this gene, whose protein product in several systems plays important roles in signal transduction events.
Collapse
Affiliation(s)
- Petra D Pandur
- Department of Anatomy and Cell Biology, The George Washington University, Washington, DC 20037, USA
| | | | | | | |
Collapse
|
44
|
Kuvbachieva A, Bestel AM, Tissir F, Maloum I, Guimiot F, Ramoz N, Bourgeois F, Moalic JM, Goffinet AM, Simonneau M. Identification of a novel brain-specific and reelin-regulated gene that encodes a protein colocalized with synapsin. Eur J Neurosci 2004; 20:603-10. [PMID: 15255972 DOI: 10.1111/j.1460-9568.2004.03473.x] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
We carried out a screening of genes that are differentially expressed in normal mice and reeler mutants and are characterized by abnormal neuronal migration and neurite deployment due to defective Reelin signalling. A novel gene, provisionally named C61, was overexpressed in Reelin-deficient embryonic mouse brain RNA. C61 encodes a 3.7 kb mRNA that is brain specific and developmentally regulated, with predominant expression in differentiating neurons. The predicted protein is 664 amino acids long, and contains LAG1 and Ezrin/Radixin/Moesin-Myosin-Filament motifs, suggesting that it may function as an intracellular adaptor. From E14.5 to birth, C61 was highly expressed in all neuronal differentiation fields, with the highest signal in the telencephalic cortical plate and mitral cells in the olfactory bulb. When expressed as a GFP fusion protein in transfected non-neuronal cells and primary neurons, this protein localizes, respectively, to the nuclear membrane or axonal outgrowths, indicating a function in axonal traffic or signalling.
Collapse
MESH Headings
- Amino Acid Motifs/physiology
- Amino Acid Sequence
- Animals
- Animals, Newborn
- Blotting, Northern/methods
- Brain/embryology
- Brain/growth & development
- Brain/metabolism
- Caenorhabditis elegans
- Cell Adhesion Molecules, Neuronal/deficiency
- Cell Adhesion Molecules, Neuronal/genetics
- Cell Adhesion Molecules, Neuronal/metabolism
- Cell Adhesion Molecules, Neuronal/physiology
- Cell Line
- Cloning, Molecular
- Drosophila
- Embryo, Mammalian
- Embryo, Nonmammalian
- Extracellular Matrix Proteins/deficiency
- Extracellular Matrix Proteins/genetics
- Extracellular Matrix Proteins/physiology
- Gene Expression Regulation, Developmental
- Green Fluorescent Proteins
- Humans
- Immunohistochemistry/methods
- In Situ Hybridization/methods
- Luminescent Proteins/metabolism
- Membrane Proteins
- Mice
- Mice, Inbred BALB C
- Mice, Neurologic Mutants
- Microfilament Proteins
- Microtubule-Associated Proteins/metabolism
- Nerve Tissue Proteins
- Neurofibromin 2/genetics
- Neurofibromin 2/metabolism
- Neurons/metabolism
- Organ Specificity
- RNA, Messenger/biosynthesis
- Reelin Protein
- Reverse Transcriptase Polymerase Chain Reaction/methods
- Serine Endopeptidases
- Synapsins/metabolism
- Transfection
- Tubulin/metabolism
- Zebrafish
Collapse
Affiliation(s)
- Anelia Kuvbachieva
- Unité de Neurobiologie, Facultés Universitaires ND de la Paix, Namur, Belgium
| | | | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Chen SS, Revoltella RP, Papini S, Michelini M, Fitzgerald W, Zimmerberg J, Margolis L. Multilineage differentiation of rhesus monkey embryonic stem cells in three-dimensional culture systems. Stem Cells 2004; 21:281-95. [PMID: 12743323 DOI: 10.1634/stemcells.21-3-281] [Citation(s) in RCA: 71] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
In the course of normal embryogenesis, embryonic stem (ES) cells differentiate along different lineages in the context of complex three-dimensional (3D) tissue structures. In order to study this phenomenon in vitro under controlled conditions, 3D culture systems are necessary. Here, we studied in vitro differentiation of rhesus monkey ES cells in 3D collagen matrixes (collagen gels and porous collagen sponges). Differentiation of ES cells in these 3D systems was different from that in monolayers. ES cells differentiated in collagen matrixes into neural, epithelial, and endothelial lineages. The abilities of ES cells to form various structures in two chemically similar but topologically different matrixes were different. In particular, in collagen gels ES cells formed gland-like circular structures, whereas in collagen sponges ES cells were scattered through the matrix or formed aggregates. Soluble factors produced by feeder cells or added to the culture medium facilitated ES cell differentiation into particular lineages. Coculture with fibroblasts in collagen gel facilitated ES cell differentiation into cells of a neural lineage expressing nestin, neural cell adhesion molecule, and class III beta-tubulin. In collagen sponges, keratinocytes facilitated ES cell differentiation into cells of an endothelial lineage expressing factor VIII. Exogenous granulocyte-macrophage colony-stimulating factor further enhanced endothelial differentiation. Thus, both soluble factors and the type of extracellular matrix seem to be critical in directing differentiation of ES cells and the formation of tissue-like structures. Three-dimensional culture systems are a valuable tool for studying the mechanisms of these phenomena.
Collapse
Affiliation(s)
- Silvia S Chen
- NASA/NIH Center for Three Dimensional Tissue Culture, Laboratory of Cellular and Molecular Biophysics, National Institute of Child Health and Human Development (NICHD), NIH, Bethesda, Maryland 20892, USA
| | | | | | | | | | | | | |
Collapse
|
46
|
Marsh-Armstrong N, Cai L, Brown DD. Thyroid hormone controls the development of connections between the spinal cord and limbs during Xenopus laevis metamorphosis. Proc Natl Acad Sci U S A 2004; 101:165-70. [PMID: 14691251 PMCID: PMC314156 DOI: 10.1073/pnas.2136755100] [Citation(s) in RCA: 48] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
During premetamorphic stages, Xenopus laevis tadpoles expressing either a dominant-negative thyroid hormone (TH) receptor or a type-III iodothyronine deiodinase transgene in the nervous system have reduced TH-induced proliferation in the spinal cord and produce fewer hindlimb-innervating motorneurons. During prometamorphic stages, innervation of the hindlimbs is reduced, and few functional neuromuscular connections are formed. By metamorphic climax, limb movement is impaired, ranging from uncoordinated leg swimming to complete quadriplegia. This phenotype is due to transgene action in the tadpole spinal cord. The requirement of TH for neurogenesis during premetamorphosis is the earliest TH-regulated process reported to date in the sequence of metamorphic changes in anurans. The muscle formed during limb growth was previously shown to be a direct target of TH control. Here, we show that the same is true of the development of spinal cord cells that innervate the limbs.
Collapse
Affiliation(s)
- Nicholas Marsh-Armstrong
- Department of Neuroscience, Johns Hopkins University School of Medicine, and Kennedy Krieger Institute, 707 North Broadway Street, Baltimore, MD 21205, USA.
| | | | | |
Collapse
|
47
|
Katsetos CD, Legido A, Perentes E, Mörk SJ. Class III beta-tubulin isotype: a key cytoskeletal protein at the crossroads of developmental neurobiology and tumor neuropathology. J Child Neurol 2003; 18:851-66; discussion 867. [PMID: 14736079 DOI: 10.1177/088307380301801205] [Citation(s) in RCA: 190] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
The expression of the cytoskeletal protein class III beta-tubulin isotype is reviewed in the context of human central nervous system development and neoplasia. Compared to systemic organs and tissues, class III beta-tubulin is abundant in the brain, where it is prominently expressed during fetal and postnatal development. As exemplified in cerebellar neurogenesis, the distribution of class III beta-tubulin is neuron associated, exhibiting different temporospatial gradients in the neuronal progeny of the external granule layer versus the neuroepithelial germinal matrix of the velum medullare. However, transient expression of this protein is also present in the telencephalic subventricular zones comprising putative neuronal and/or glial precursor cells. This temporospatially restricted, potentially non-neuronal expression of class III beta-tubulin may have implications in the accurate identification of presumptive neurons derived from transplanted embryonic stem cells. In the adult central nervous system, the distribution of class III beta-tubulin is almost exclusively neuron specific. Altered patterns of expression are noted in brain tumors. In "embryonal"-type neuronal/neuroblastic tumors of the central nervous system, such as the medulloblastomas, class III beta-tubulin expression is associated with neuronal differentiation and decreased cell proliferation. In contrast, the expression of class III beta-tubulin in gliomas is associated with an ascending grade of histologic malignancy and with correspondingly high proliferative indices. Thus, class III beta-tubulin expression in neuronal or neuroblastic tumors is differentiation dependent, whereas in glial tumors, it is aberrant and/or represents "dedifferentiation" associated with the acquisition of glial progenitor-like phenotype(s). From a diagnostic perspective, the detection of class III beta-tubulin immunostaining in neoplastic cells should not be construed as categorical evidence of divergent neuronal differentiation in tumors, which are otherwise phenotypically glial. Because class III beta-tubulin is present in neoplastic but not in normal differentiated glial cells, the elucidation of molecular mechanisms responsible for the altered expression of this isotype may provide critical insights into the dynamics of the microtubule cytoskeleton in the growth and progression of gliomas.
Collapse
Affiliation(s)
- Christos D Katsetos
- Department of Pediatrics, Drexel University College of Medicine, Philadelphia, PA, USA.
| | | | | | | |
Collapse
|
48
|
Katsetos CD, Herman MM, Mörk SJ. Class III beta-tubulin in human development and cancer. CELL MOTILITY AND THE CYTOSKELETON 2003; 55:77-96. [PMID: 12740870 DOI: 10.1002/cm.10116] [Citation(s) in RCA: 222] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
The differential cellular expression of class III beta-tubulin isotype (betaIII) is reviewed in the context of human embryological development and neoplasia. As compared to somatic organs and tissues, betaIII is abundant in the central and peripheral nervous systems (CNS and PNS) where it is prominently expressed during fetal and postnatal development. As exemplified in cerebellar and sympathoadrenal neurogenesis, the distribution of betaIII is neuron-associated, exhibiting distinct temporospatial gradients according to the regional neuroepithelia of origin. However, transient expression of this protein is also present in the subventricular zones of the CNS comprising putative neuronal- and/or glial precursor cells, as well as in Kulchitsky neuroendocrine cells of the fetal respiratory epithelium. This temporally restricted, potentially non-neuronal expression may have implications in the identification of presumptive neurons derived from embryonic stem cells. In adult tissues, the distribution of betaIII is almost exclusively neuron-specific. Altered patterns of expression are noted in cancer. In "embryonal"- and "adult-type" neuronal tumors of the CNS and PNS, betaIII is associated with neuronal differentiation and decreased cell proliferation. In contrast, the presence of betaIII in gliomas and lung cancer is associated with an ascending histological grade of malignancy. Thus, betaIII expression in neuronal tumors is differentiation-dependent, while in non-neuronal tumors it is aberrant and/or represents "dedifferentiation" associated with the acquisition of progenitor-like phenotypic properties. Increased expression in various epithelial cancer cell lines is associated with chemoresistance to taxanes. Because betaIII is present in subpopulations of neoplastic, but not in normal differentiated glial or somatic epithelial cells, the elucidation of mechanisms responsible for the altered expression of this isotype may provide insights into the role of the microtubule cytoskeleton in tumorigenesis and tumor progression.
Collapse
Affiliation(s)
- Christos D Katsetos
- Department of Pediatrics, Section of Neurology, Drexel University College of Medicine, Philadelphia, Pennsylvania, USA.
| | | | | |
Collapse
|
49
|
Economides KD, Zeltser L, Capecchi MR. Hoxb13 mutations cause overgrowth of caudal spinal cord and tail vertebrae. Dev Biol 2003; 256:317-30. [PMID: 12679105 DOI: 10.1016/s0012-1606(02)00137-9] [Citation(s) in RCA: 115] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
To address the expression and function of Hoxb13, the 5' most Hox gene in the HoxB cluster, we have generated mice with loss-of-function and beta-galactosidase reporter insertion alleles of this gene. Mice homozygous for Hoxb13 loss-of-function mutations show overgrowth in all major structures derived from the tail bud, including the developing secondary neural tube (SNT), the caudal spinal ganglia, and the caudal vertebrae. Using the beta-galactosidase reporter allele of Hoxb13, also a loss-of-function allele, we found that the expression patterns of Hoxb13 in the developing spinal cord and caudal mesoderm are closely associated with overgrowth phenotypes in the tails of homozygous mutant animals. These phenotypes can be explained by the observed increased cell proliferation and decreased levels of apoptosis within the tail of homozygous mutant mice. This analysis of Hoxb13 function suggests that this 5' Hox gene may act as an inhibitor of neuronal cell proliferation, an activator of apoptotic pathways in the SNT, and as a general repressor of growth in the caudal vertebrae.
Collapse
Affiliation(s)
- Kyriakos D Economides
- Howard Hughes Medical Center, Department of Human Genetics, University of Utah, Salt Lake City, UT 84112, USA
| | | | | |
Collapse
|
50
|
Nakamura Y, Yamamoto M, Oda E, Yamamoto A, Kanemura Y, Hara M, Suzuki A, Yamasaki M, Okano H. Expression of tubulin beta II in neural stem/progenitor cells and radial fibers during human fetal brain development. J Transl Med 2003; 83:479-89. [PMID: 12695551 DOI: 10.1097/01.lab.0000063930.75913.b3] [Citation(s) in RCA: 35] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
Recent studies revealed that the "radial glia" in fetal rodent brains are dividing neuronal precursor cells. However, in fetal primate brains, this issue remains unclear, with previous reports indicating that radial glia are a specialized form of astroglia. To investigate the relationship between radial fibers (RFs) and neural stem/progenitor cells in the fetal human brain, we generated polyclonal antibodies to human nestin protein and developed a new mAb, KNY-379, by screening for antibodies that immunostained RFs on paraffin-embedded human fetal brain specimens (12 gestational weeks). The immunostaining for KNY-379 antigen and nestin was seen over the RFs in brains at 8 gestational weeks. Furthermore, KNY-379 antigen and nestin were also detected in human neural stem/progenitor cells in neurosphere cultures. At 12 to 15 gestational weeks, the KNY-379 immunostaining of RFs remained in the periventricular zone and the deep part of the intermediate zone, but it also appeared in outgrowing axons in the cortical plate, in the superficial portion of the intermediate zone, and in apical dendrites in the molecular layer. In the later stages of fetal development (18-40 gestational weeks), this antigen remained in the outgrowing axons and dendrites, but was no longer associated with RFs. Expression cloning and immunoblot analysis demonstrated the antigen to be tubulin beta II, which would thus be a good marker for studying RFs and neural stem/progenitor cells in the early developing human brain.
Collapse
Affiliation(s)
- Yasuhiro Nakamura
- Department of Pathology, St Mary's Hospital, Kurume University School of Medicine, Kurume, Japan.
| | | | | | | | | | | | | | | | | |
Collapse
|