Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 20
Filter
Add more filters










Publication year range
1.
Cancer Rep (Hoboken) ; 7(4): e2048, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38599791

ABSTRACT

BACKGROUND: Medulloblastoma (MB) is a rare primitive neuroectodermal tumors originating from the cerebellum. MB is the most common malignant primary brain tumor of childhood. MB originates from neural precursor cells in distinctive regions of the rhombic lip, and their maturation occurs in the cerebellum or the brain stem during embryonal development. Also, apoptosis is a programmed cell death associated with numerous physiological as well as pathological regulations. RECENT FINDINGS: Irradiation (IR)-induce apoptosis triggers cell death, with or without intervening mitosis within a few hours of IR and these share different morphologic alteration such as, loss of normal nuclear structure as well as degradation of DNA. Moreover, MB is strikingly sensitive to DNA-damaging therapies and the role of apoptosis a key treatment modality. Furthermore, in MB, the apoptotic pathways are made up of several triggers, modulators, as well as effectors. Notably, IR-induced apoptotic mechanisms in MB therapy are very complex and they either induce radiosensitivity or inhibit radioresistance leading to potential effective treatment strategies for MB. CONCLUSION: This review explicitly explores the pivotal roles of IR-induced apoptosis in the pathogenesis and therapy of MB.


Subject(s)
Cerebellar Neoplasms , Embryonic Structures , Medulloblastoma , Metencephalon/embryology , Neural Stem Cells , Humans , Medulloblastoma/radiotherapy , Medulloblastoma/genetics , Neural Stem Cells/metabolism , Neural Stem Cells/pathology , Apoptosis , Cerebellar Neoplasms/radiotherapy , Cerebellar Neoplasms/pathology , DNA
2.
Dev Neurobiol ; 84(2): 74-92, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38509451

ABSTRACT

The organization of neurons into distinct layers, known as lamination, is a common feature of the nervous system. This process, which arises from the direct coupling of neurogenesis and neuronal migration, plays a crucial role in the development of the cerebellum, a structure exhibiting a distinct folding cytoarchitecture with cells arranged in discrete layers. Disruptions to neuronal migration can lead to various neurodevelopmental disorders, highlighting the significance of understanding the molecular regulation of lamination. We report a role Mllt11/Af1q/Tcf7c (myeloid/lymphoid or mixed-lineage leukemia; translocated to chromosome 11/All1 fused gene from chromosome 1q, also known as Mllt11 transcriptional cofactor 7; henceforth referred to Mllt11) in the migration of cerebellar granule cells (GCs). We now show that Mllt11 plays a role in both the tangential and radial migration of GCs. Loss of Mllt11 led to an accumulation of GC precursors in the rhombic lip region and a reduction in the number of GCs successfully populating developing folia. Consequently, this results in smaller folia and an overall reduction in cerebellar size. Furthermore, analysis of the anchoring centers reveals disruptions in the perinatal folia cytoarchitecture, including alterations in the Bergmann glia fiber orientation and reduced infolding of the Purkinje cell plate. Lastly, we demonstrate that Mllt11 interacts with non-muscle myosin IIB (NMIIB) and Mllt11 loss-reduced NMIIB expression. We propose that the dysregulation of NMIIB underlies altered GC migratory behavior. Taken together, the findings reported herein demonstrate a role for Mllt11 in regulating neuronal migration within the developing cerebellum, which is necessary for its proper neuroanatomical organization.


Subject(s)
Cerebellum , Embryonic Structures , Metencephalon/embryology , Neurons , Pregnancy , Female , Humans , Neurons/metabolism , Neuroglia/metabolism , Cell Movement/physiology
3.
Nature ; 609(7929): 1012-1020, 2022 09.
Article in English | MEDLINE | ID: mdl-36131015

ABSTRACT

Medulloblastoma, a malignant childhood cerebellar tumour, segregates molecularly into biologically distinct subgroups, suggesting that a personalized approach to therapy would be beneficial1. Mouse modelling and cross-species genomics have provided increasing evidence of discrete, subgroup-specific developmental origins2. However, the anatomical and cellular complexity of developing human tissues3-particularly within the rhombic lip germinal zone, which produces all glutamatergic neuronal lineages before internalization into the cerebellar nodulus-makes it difficult to validate previous inferences that were derived from studies in mice. Here we use multi-omics to resolve the origins of medulloblastoma subgroups in the developing human cerebellum. Molecular signatures encoded within a human rhombic-lip-derived lineage trajectory aligned with photoreceptor and unipolar brush cell expression profiles that are maintained in group 3 and group 4 medulloblastoma, suggesting a convergent basis. A systematic diagnostic-imaging review of a prospective institutional cohort localized the putative anatomical origins of group 3 and group 4 tumours to the nodulus. Our results connect the molecular and phenotypic features of clinically challenging medulloblastoma subgroups to their unified beginnings in the rhombic lip in the early stages of human development.


Subject(s)
Cell Lineage , Cerebellar Neoplasms , Medulloblastoma , Metencephalon , Animals , Cerebellar Neoplasms/classification , Cerebellar Neoplasms/embryology , Cerebellar Neoplasms/pathology , Cerebellum/embryology , Humans , Medulloblastoma/classification , Medulloblastoma/embryology , Medulloblastoma/pathology , Metencephalon/embryology , Mice , Neurons/pathology , Prospective Studies
4.
Nature ; 609(7929): 1021-1028, 2022 09.
Article in English | MEDLINE | ID: mdl-36131014

ABSTRACT

Medulloblastoma (MB) comprises a group of heterogeneous paediatric embryonal neoplasms of the hindbrain with strong links to early development of the hindbrain1-4. Mutations that activate Sonic hedgehog signalling lead to Sonic hedgehog MB in the upper rhombic lip (RL) granule cell lineage5-8. By contrast, mutations that activate WNT signalling lead to WNT MB in the lower RL9,10. However, little is known about the more commonly occurring group 4 (G4) MB, which is thought to arise in the unipolar brush cell lineage3,4. Here we demonstrate that somatic mutations that cause G4 MB converge on the core binding factor alpha (CBFA) complex and mutually exclusive alterations that affect CBFA2T2, CBFA2T3, PRDM6, UTX and OTX2. CBFA2T2 is expressed early in the progenitor cells of the cerebellar RL subventricular zone in Homo sapiens, and G4 MB transcriptionally resembles these progenitors but are stalled in developmental time. Knockdown of OTX2 in model systems relieves this differentiation blockade, which allows MB cells to spontaneously proceed along normal developmental differentiation trajectories. The specific nature of the split human RL, which is destined to generate most of the neurons in the human brain, and its high level of susceptible EOMES+KI67+ unipolar brush cell progenitor cells probably predisposes our species to the development of G4 MB.


Subject(s)
Cell Differentiation , Cerebellar Neoplasms , Medulloblastoma , Metencephalon , Cell Differentiation/genetics , Cell Lineage , Cerebellar Neoplasms/classification , Cerebellar Neoplasms/genetics , Cerebellar Neoplasms/pathology , Cerebellum/embryology , Cerebellum/pathology , Core Binding Factor alpha Subunits/genetics , Hedgehog Proteins/metabolism , Histone Demethylases , Humans , Ki-67 Antigen/metabolism , Medulloblastoma/classification , Medulloblastoma/genetics , Medulloblastoma/pathology , Metencephalon/embryology , Metencephalon/pathology , Muscle Proteins , Mutation , Otx Transcription Factors/deficiency , Otx Transcription Factors/genetics , Repressor Proteins , T-Box Domain Proteins/metabolism , Transcription Factors
5.
Science ; 339(6116): 204-7, 2013 Jan 11.
Article in English | MEDLINE | ID: mdl-23307742

ABSTRACT

We investigated the role of histone methyltransferase Ezh2 in tangential migration of mouse precerebellar pontine nuclei, the main relay between neocortex and cerebellum. By counteracting the sonic hedgehog pathway, Ezh2 represses Netrin1 in dorsal hindbrain, which allows normal pontine neuron migration. In Ezh2 mutants, ectopic Netrin1 derepression results in abnormal migration and supernumerary nuclei integrating in brain circuitry. Moreover, intrinsic topographic organization of pontine nuclei according to rostrocaudal progenitor origin is maintained throughout migration and correlates with patterned cortical input. Ezh2 maintains spatially restricted Hox expression, which, in turn, regulates differential expression of the repulsive receptor Unc5b in migrating neurons; together, they generate subsets with distinct responsiveness to environmental Netrin1. Thus, Ezh2-dependent epigenetic regulation of intrinsic and extrinsic transcriptional programs controls topographic neuronal guidance and connectivity in the cortico-ponto-cerebellar pathway.


Subject(s)
Cerebellum/embryology , Neural Pathways/embryology , Neurons/physiology , Polycomb Repressive Complex 2/metabolism , Pons/embryology , Animals , Cell Movement , Cerebellum/cytology , Cerebellum/metabolism , Cerebral Cortex/embryology , Cerebral Cortex/physiology , Enhancer of Zeste Homolog 2 Protein , Epigenesis, Genetic , Gene Expression Regulation, Developmental , Genes, Homeobox , Homeodomain Proteins/metabolism , Metencephalon/embryology , Mice , Mice, Transgenic , Nerve Growth Factors/genetics , Nerve Growth Factors/metabolism , Netrin Receptors , Netrin-1 , Neural Pathways/physiology , Polycomb Repressive Complex 2/genetics , Pons/cytology , Pons/metabolism , Receptors, Cell Surface/genetics , Receptors, Cell Surface/metabolism , Transcription, Genetic , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/metabolism
6.
Dev Growth Differ ; 54(4): 463-73, 2012 May.
Article in English | MEDLINE | ID: mdl-22404534

ABSTRACT

Fibroblast growth factor 8 (FGF8) functions as a local organizing signal for the tectum and cerebellum. FGF8 activates Ras-ERK signaling pathway to induce cerebellar development. We paid attention to the difference in the expression pattern of the molecules that are induced by FGF8 in the mid-hind brain region during normal development and after FGF8 misexpression; some are expressed in the area corresponding to the ERK activation domain but the others are expressed corresponding to the Fgf8 expression domain. Since some of the FGF family members are localized in the nucleus, we wondered if FGF8 could localize in the nuclei and function in the nucleus. We first show that in cultured NIH3T3 cells transfected FGF8b could localize in the nucleus. Transfected FGF8b could also localize in the nucleus of the cells in the chick neural tube. In mouse embryonic neural tube, we detected endogenous FGF8 in the nuclei. Implantation of an FGF8b-soaked bead showed that exogenous FGF8b could be translocated to the nuclei in the isthmus. Furthermore, signal-peptide-deletion mutant of FGF8b mainly localized in the nuclei, and induced Sprouty2 without activating ERK in the mesencephalon. Signal-peptide-deletion mutant of FGF8b could not induce Pax2 expression. Taken together, we concluded that FGF8b could be translocated to the nuclei, and that the nuclear FGF8 could function as transcriptional regulator to induce Sprouty2 in the isthmus.


Subject(s)
Cell Nucleus/metabolism , Fibroblast Growth Factor 8/metabolism , Gene Expression Regulation, Developmental , Membrane Proteins/metabolism , Active Transport, Cell Nucleus , Adaptor Proteins, Signal Transducing , Animals , Cell Nucleus/genetics , Chick Embryo , Electroporation , Embryo, Mammalian/cytology , Embryo, Mammalian/embryology , Embryonic Development , Female , Fibroblast Growth Factor 8/genetics , Immunohistochemistry , Intracellular Signaling Peptides and Proteins , MAP Kinase Signaling System , Membrane Proteins/genetics , Mesencephalon/cytology , Mesencephalon/embryology , Metencephalon/cytology , Metencephalon/embryology , Mice , Mice, Inbred ICR , Microscopy, Confocal , NIH 3T3 Cells , Neural Tube/cytology , Neural Tube/embryology , PAX2 Transcription Factor/genetics , PAX2 Transcription Factor/metabolism , Protein Isoforms/genetics , Protein Isoforms/metabolism , Protein Serine-Threonine Kinases , Protein Sorting Signals , Protein Structure, Tertiary , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Sequence Deletion , Transfection
7.
Dev Biol ; 347(2): 392-403, 2010 Nov 15.
Article in English | MEDLINE | ID: mdl-20816794

ABSTRACT

Otx2 is expressed in each step and site of head development. To dissect each Otx2 function we have identified a series of Otx2 enhancers. The Otx2 expression in the anterior neuroectoderm is regulated by the AN enhancer and the subsequent expression in forebrain and midbrain later than E8.5 by FM1 and FM2 enhancers; the Otx1 expression takes place at E8.0. In telencephalon later than E9.5 Otx1 continues to be expressed in the entire pallium, while the Otx2 expression is confined to the most medial pallium. To determine the Otx functions in forebrain and midbrain development we have generated mouse mutants that lack both FM1 and FM2 enhancers (DKO: Otx2(ΔFM1ΔFM2/ΔFM1ΔFM2)) and examined the TKO (Otx1(-/-)Otx2(ΔFM1ΔFM2/ΔFM1ΔFM2)) phenotype. The mutants develop normally until E8.0, but subsequently by E9.5 the diencephalon, including thalamic eminence and prethalamus, and the mesencephalon are caudalized into metencephalon consisting of isthmus and rhombomere 1; the caudalization does not extend to rhombomere 2 and more caudal rhombomeres. In rostral forebrain, neopallium, ganglionic eminences and hypothalamus in front of prethalamus develop; we propose that they become insensitive to the caudalization with the switch from the Otx2 expression under the AN enhancer to that under FM1 and FM2 enhancers. In contrast, the medial pallium requires Otx1 and Otx2 for its development later than E9.5, and the Otx2 expression in diencepalon and mesencephalon later than E9.5 is also directed by an enhancer other than FM1 and FM2 enhancers.


Subject(s)
Brain/embryology , Brain/metabolism , Otx Transcription Factors/metabolism , Animals , Base Sequence , Body Patterning , DNA Primers/genetics , Diencephalon/embryology , Diencephalon/metabolism , Enhancer Elements, Genetic , Female , Gene Expression Regulation, Developmental , Mesencephalon/embryology , Mesencephalon/metabolism , Metencephalon/embryology , Metencephalon/metabolism , Mice , Mice, Knockout , Mice, Mutant Strains , Mice, Transgenic , Otx Transcription Factors/deficiency , Otx Transcription Factors/genetics , Pregnancy
8.
Dev Dyn ; 235(4): 928-33, 2006 Apr.
Article in English | MEDLINE | ID: mdl-16496282

ABSTRACT

Detailed reconstruction of the spatiotemporal history of embryonic cells is key to understanding tissue formation processes but is often complicated by the large number of cells involved, particularly so in vertebrates. Through a combination of high-resolution time-lapse lineage tracing and antibody staining, we have analyzed the movement of mesencephalic and metencephalic cell populations in the early zebrafish embryo. To facilitate the analysis of our cell tracking data, we have created TracePilot, a software tool that allows interactive manipulation and visualization of tracking data. We demonstrate its utility by showing novel visualizations of cell movement in the developing zebrafish brain. TracePilot (http://www.mpi-cbg.de/tracepilot) is Java-based, available free of charge, and has a program structure that allows the incorporation of additional analysis tools.


Subject(s)
Cell Movement , Mesencephalon/cytology , Mesencephalon/embryology , Metencephalon/cytology , Metencephalon/embryology , Zebrafish/embryology , Animals , Cell Lineage , Computer Graphics , Data Interpretation, Statistical , Embryo, Nonmammalian , Mesencephalon/physiology , Metencephalon/physiology , Microscopy, Video , Software , Time Factors
9.
Development ; 132(14): 3163-73, 2005 Jul.
Article in English | MEDLINE | ID: mdl-15944182

ABSTRACT

The mesencephalic and metencephalic region (MMR) of the vertebrate central nervous system develops in response to signals produced by the isthmic organizer (IsO). We have previously reported that the LIM homeobox transcription factor Lmx1b is expressed within the chick IsO, where it is sufficient to maintain expression of the secreted factor wnt1. In this paper, we show that zebrafish express two Lmx1b orthologs, lmx1b.1 and lmx1b.2, in the rostral IsO, and demonstrate that these genes are necessary for key aspects of MMR development. Simultaneous knockdown of Lmx1b.1 and Lmx1b.2 using morpholino antisense oligos results in a loss of wnt1, wnt3a, wnt10b, pax8 and fgf8 expression at the IsO, leading ultimately to programmed cell death and the loss of the isthmic constriction and cerebellum. Single morpholino knockdown of either Lmx1b.1 or Lmx1b.2 has no discernible effect on MMR development. Maintenance of lmx1b.1 and lmx1b.2 expression at the isthmus requires the function of no isthmus/pax2.1, as well as Fgf signaling. Transient misexpression of Lmx1b.1 or Lmx1b.2 during early MMR development induces ectopic wnt1 and fgf8 expression in the MMR, as well as throughout much of the embryo. We propose that Lmx1b.1- and Lmx1b.2-mediated regulation of wnt1, wnt3a, wnt10b, pax8 and fgf8 maintains cell survival in the isthmocerebellar region.


Subject(s)
Homeodomain Proteins/physiology , Mesencephalon/embryology , Metencephalon/embryology , Transcription Factors/physiology , Zebrafish/embryology , Amino Acid Sequence , Animals , DNA-Binding Proteins/metabolism , Fibroblast Growth Factor 8 , Fibroblast Growth Factors/genetics , Fibroblast Growth Factors/metabolism , Gene Expression Regulation, Developmental/physiology , Homeodomain Proteins/genetics , Intercellular Signaling Peptides and Proteins/biosynthesis , Intercellular Signaling Peptides and Proteins/genetics , LIM-Homeodomain Proteins , Molecular Sequence Data , PAX2 Transcription Factor , Protein Isoforms/genetics , Protein Isoforms/physiology , Transcription Factors/genetics , Transcription Factors/metabolism , Wnt Proteins , Wnt1 Protein , Zebrafish/genetics , Zebrafish Proteins
10.
Int J Dev Biol ; 49(2-3): 231-5, 2005.
Article in English | MEDLINE | ID: mdl-15906236

ABSTRACT

The brain vesicles that are formed at an early stage of neural development are the fundamentals of the brain plan. Heterotopic transplantation revealed that the diencephalon could change its fate when juxtaposed to the isthmus (mes-metencephalic boundary), which indicated that the isthmus functions as an organizer for the mesencephalon and metencephalon. Fgf8 is identified as an isthmus organizing signal. Misexpression of Fgf8a and Fgf8b indicated that a strong Fgf8 signal organizes cerebellar development. The transcription factors define the fate of the region. Overlapping expression of Otx2, En1 and Pax2 may define the mesencephalic region and additional expression of Pax3/7 may instruct the mesencephalic region to differentiate into the tectum. The di-mesencephalic boundary is determined by repressive interaction between Pax6 and En1/Pax2 and the mes-metencephalic boundary is defined by repressive interaction between Otx2 and Gbx2. Fgf8 is induced at the border of the Otx2 and Gbx2 expression domain, overlapping with Gbx2 expression.


Subject(s)
Brain/embryology , Mesencephalon/embryology , Metencephalon/embryology , Organizers, Embryonic/cytology , Animals , Body Patterning , Gene Expression Regulation, Developmental , Gene Silencing , Gene Transfer Techniques , Organizers, Embryonic/physiology
11.
Development ; 131(14): 3319-31, 2004 Jul.
Article in English | MEDLINE | ID: mdl-15201224

ABSTRACT

Otx2 expression in the forebrain and midbrain was found to be regulated by two distinct enhancers (FM and FM2) located at 75 kb 5' upstream and 115 kb 3' downstream. The activities of these two enhancers were absent in anterior neuroectoderm earlier than E8.0; however, at E9.5 their regions of activity spanned the entire mesencephalon and diencephalon with their caudal limits at the boundary with the metencephalon or isthmus. In telencephalon, activities were found only in the dorsomedial aspect. Potential binding sites of OTX and TCF were essential to FM activity, and TCF sites were also essential to FM2 activity. The FM2 enhancer appears to be unique to rodent; however, the FM enhancer region is deeply conserved in gnathostomes. Studies of mutants lacking FM or FM2 enhancer demonstrated that these enhancers indeed regulate Otx2 expression in forebrain and midbrain. Development of mesencephalic and diencephalic regions was differentially regulated in a dose-dependent manner by the cooperation between Otx1 and Otx2 under FM and FM2 enhancers: the more caudal the structure the higher the OTX dose requirement. At E10.5 Otx1-/-Otx2DeltaFM/DeltaFM mutants, in which Otx2 expression under the FM2 enhancer remained, exhibited almost complete loss of the entire diencephalon and mesencephalon; the telencephalon did, however, develop.


Subject(s)
Homeodomain Proteins , Mesencephalon/embryology , Nerve Tissue Proteins/biosynthesis , Nerve Tissue Proteins/genetics , Prosencephalon/embryology , Trans-Activators/biosynthesis , Trans-Activators/genetics , Alleles , Animals , Base Sequence , Binding Sites , Conserved Sequence , Diencephalon/embryology , Ectoderm/metabolism , Enhancer Elements, Genetic , Gene Expression Regulation, Developmental , Humans , In Situ Hybridization , Metencephalon/embryology , Mice , Mice, Transgenic , Models, Genetic , Molecular Sequence Data , Mutation , Otx Transcription Factors , RNA/metabolism , Sequence Homology, Nucleic Acid , Signal Transduction , Time Factors
12.
Development ; 129(22): 5269-77, 2002 Nov.
Article in English | MEDLINE | ID: mdl-12399317

ABSTRACT

The isthmus is the organizing center for the tectum and cerebellum. Fgf8 and Wnt1 are secreted molecules expressed around the isthmus. The function of Fgf8 has been well analyzed, and now accepted as the most important organizing signal. Involvement of Wnt1 in the isthmic organizing activity was suggested by analysis of Wnt1 knockout mice. But its role in isthmic organizing activity is still obscure. Recently, it has been shown that Lmx1b is expressed in the isthmic region and that it may occupy higher hierarchical position in the gene expression cascade in the isthmus. We have carried out misexpression experiment of Lmx1b and Wnt1, and considered their role in the isthmic organizing activity. Lmx1b or Wnt1 misexpression caused expansion of the tectum and cerebellum. Fgf8 was repressed in a cells that misexpress Lmx1b, but Fgf8 expression was induced around Lmx1b-misexpressing cells. As Lmx1b induced Wnt1 and Wnt1 induced Fgf8 expression in turn, Wnt1 may be involved in non cell-autonomous induction of Fgf8 expression by Lmx1b. Wnt1 could not induce Lmx1b expression so that Lmx1b may be put at the higher hierarchical position than Wnt1 in gene expression cascade in the isthmus. We have examined the relationship among isthmus related genes, and discuss the mechanism of the formation and maintenance of isthmic organizing activity.


Subject(s)
Gene Expression Regulation, Developmental , Homeodomain Proteins/metabolism , Mesencephalon/embryology , Metencephalon/embryology , Proto-Oncogene Proteins/metabolism , Zebrafish Proteins , Animals , Cerebellum/embryology , Chick Embryo , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Fibroblast Growth Factor 8 , Fibroblast Growth Factors/genetics , Fibroblast Growth Factors/metabolism , Genes, Dominant , Homeodomain Proteins/genetics , LIM-Homeodomain Proteins , Mesencephalon/metabolism , Metencephalon/metabolism , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Otx Transcription Factors , Proto-Oncogene Proteins/genetics , Repressor Proteins/genetics , Repressor Proteins/metabolism , Trans-Activators/genetics , Trans-Activators/metabolism , Transcription Factors , Wnt Proteins , Wnt1 Protein
13.
Development ; 128(24): 4979-91, 2001 Dec.
Article in English | MEDLINE | ID: mdl-11748135

ABSTRACT

Otx2 and Gbx2 are among the earliest genes expressed in the neuroectoderm, dividing it into anterior and posterior domains with a common border that marks the mid-hindbrain junction. Otx2 is required for development of the forebrain and midbrain, and Gbx2 for the anterior hindbrain. Furthermore, opposing interactions between Otx2 and Gbx2 play an important role in positioning the mid-hindbrain boundary, where an organizer forms that regulates midbrain and cerebellum development. We show that the expression domains of Otx2 and Gbx2 are initially established independently of each other at the early headfold stage, and then their expression rapidly becomes interdependent by the late headfold stage. As we demonstrate that the repression of Otx2 by retinoic acid is dependent on an induction of Gbx2 in the anterior brain, molecules other than retinoic acid must regulate the initial expression of Otx2 in vivo. In contrast to previous suggestions that an interaction between Otx2- and Gbx2-expressing cells may be essential for induction of mid-hindbrain organizer factors such as Fgf8, we find that Fgf8 and other essential mid-hindbrain genes are induced in a correct temporal manner in mouse embryos deficient for both Otx2 and Gbx2. However, expression of these genes is abnormally co-localized in a broad anterior region of the neuroectoderm. Finally, we find that by removing Otx2 function, development of rhombomere 3 is rescued in Gbx2(-/-) embryos, showing that Gbx2 plays a permissive, not instructive, role in rhombomere 3 development. Our results provide new insights into induction and maintenance of the mid-hindbrain genetic cascade by showing that a mid-hindbrain competence region is initially established independent of the division of the neuroectoderm into an anterior Otx2-positive domain and posterior Gbx2-positive domain. Furthermore, Otx2 and Gbx2 are required to suppress hindbrain and midbrain development, respectively, and thus allow establishment of the normal spatial domains of Fgf8 and other genes.


Subject(s)
Gene Expression Regulation, Developmental , Homeodomain Proteins/metabolism , Nerve Tissue Proteins/metabolism , Rhombencephalon/embryology , Trans-Activators/metabolism , Animals , Body Patterning , Fibroblast Growth Factor 8 , Fibroblast Growth Factors/biosynthesis , Head/abnormalities , Homeodomain Proteins/genetics , Mesencephalon/embryology , Metencephalon/embryology , Mice , Mice, Mutant Strains , Nerve Tissue Proteins/genetics , Organizers, Embryonic , Otx Transcription Factors , Prosencephalon/embryology , Somites , Tissue Distribution , Trans-Activators/genetics , Tretinoin/pharmacology
14.
Brain Res Mol Brain Res ; 93(2): 137-47, 2001 Sep 30.
Article in English | MEDLINE | ID: mdl-11589991

ABSTRACT

The floor plate (FP) is a transient structure of the embryonic central nervous system (CNS) which plays a key role in development driving cell differentiation and patterning in the ventral neural tube. The fact that antisera raised against subcommissural organ (SCO) secretion immunostain FP cells and react with high-molecular-mass proteins in FP extracts, prompted us to investigate the expression of a SCO-related polypeptide in FP cells. RNA from bovine FP was analyzed by means of reverse transcriptase polymerase chain reaction (RT-PCR), using primers derived from the 3' end of SCO-spondin which revealed products of 233, 237, 519 and 783 bp. Sequence analysis of the 233 bp PCR fragment confirmed the identity between this FP product and SCO-spondin. FP-translation of the SCO-spondin encoded polypeptide(s) was demonstrated by Western blot analysis and immunocytochemistry, using antisera raised against (i) the glycoproteins secreted by the bovine SCO, and (ii) a peptide derived from the open reading frame of the major SCO secretory protein, SCO-spondin, respectively. Additional evidence pointing to active transcription and translation of a SCO-spondin related gene was obtained in long term FP organ cultures. On the basis of partial sequence homologies of SCO-spondin with protein domains implicated in cell-cell contacts, cell-matrix interactions and neurite outgrowth it is possible to suggest that the SCO-spondin secreted by the FP is involved in CNS development.


Subject(s)
Cell Adhesion Molecules, Neuronal/biosynthesis , Central Nervous System/embryology , Fetal Proteins/biosynthesis , Gene Expression Regulation, Developmental , RNA, Messenger/biosynthesis , Subcommissural Organ/metabolism , Animals , Base Sequence , Blotting, Southern , Blotting, Western , Cattle , Cell Adhesion Molecules, Neuronal/genetics , Female , Fetal Proteins/genetics , Immune Sera , Metencephalon/embryology , Metencephalon/metabolism , Molecular Sequence Data , Molecular Weight , Organ Culture Techniques , Organ Specificity , Protein Biosynthesis , Protein Structure, Tertiary , Repetitive Sequences, Amino Acid , Reverse Transcriptase Polymerase Chain Reaction , Subcommissural Organ/embryology , Subcommissural Organ/growth & development
15.
Development ; 128(13): 2461-9, 2001 Jul.
Article in English | MEDLINE | ID: mdl-11493563

ABSTRACT

The mes/metencephalic boundary (isthmus) has an organizing activity for mesencephalon and metencephalon. The candidate signaling molecule is Fgf8 whose mRNA is localized in the region where the cerebellum differentiates. Responding to this signal, the cerebellum differentiates in the metencephalon and the tectum differentiates in the mesencephalon. Based on the assumption that strong Fgf8 signal induces the cerebellum and that the Fgf8b signal is stronger than that of Fgf8a, we carried out experiments to misexpress Fgf8b and Fgf8a in chick embryos. Fgf8a did not affect the expression pattern of Otx2, Gbx2 or Irx2. En2 expression was upregulated in the mesencephalon and in the diencephalon by Fgf8a. Consequently, Fgf8a misexpression resulted in the transformation of the presumptive diencephalon to the fate of the mesencephalon. In contrast, Fgf8b repressed Otx2 expression, but upregulated Gbx2 and Irx2 expression in the mesencephalon. As a result, Fgf8b completely changed the fate of the mesencephalic alar plate to cerebellum. Quantitative analysis showed that Fgf8b signal is 100 times stronger than Fgf8a signal. Co-transfection of Fgf8b with Otx2 indicates that Otx2 is a key molecule in mesencephalic generation. We have shown by RT-PCR that both Fgf8a and Fgf8b are expressed, Fgf8b expression prevailing in the isthmic region. The results all support our working hypothesis that the strong Fgf8 signal induces the neural tissue around the isthmus to differentiate into the cerebellum.


Subject(s)
Cerebellum/embryology , Fibroblast Growth Factors/metabolism , Homeodomain Proteins , Signal Transduction , Superior Colliculi/embryology , Animals , Base Sequence , Cerebellum/metabolism , Chick Embryo , Diencephalon/embryology , Fibroblast Growth Factor 8 , Fibroblast Growth Factors/genetics , Fibroblast Growth Factors/physiology , Gene Expression , Mesencephalon/embryology , Metencephalon/embryology , Molecular Sequence Data , Nerve Tissue Proteins/genetics , Otx Transcription Factors , Superior Colliculi/metabolism , Trans-Activators/genetics
16.
Dev Biol ; 233(2): 513-25, 2001 May 15.
Article in English | MEDLINE | ID: mdl-11336511

ABSTRACT

In the chick metencephalon, oligodendrocyte precursors arise in distinct domains of the ventricular zone. During development, the earliest oligodendrocyte precursors appear in the metencephalic ventral ventricular zone adjacent to the midline, consistent with their location in the spinal cord. In contrast to spinal cord, however, distinct domains in the lateral and dorsal metencephalic ventricular zone subsequently generate oligodendrocyte precursors. All oligodendrogenic domains of the metencephalon appear in close apposition to regions that transiently express sonic hedgehog (Shh). Inhibition studies demonstrate a functional requirement for Shh signaling in the early appearance of metencephalic oligodendrocyte precursors, while in vitro studies suggest a dose-dependent increase in the number of oligodendrocyte precursors in response to Shh. In purified cultures of oligodendrocyte precursors, Shh promotes cell survival and proliferation, suggesting that Shh can act directly on these cells. These data suggest that Shh may be responsible for the localized appearance of oligodendrocyte precursors throughout the CNS, irrespective of the dorso-ventral neural axis.


Subject(s)
Metencephalon/embryology , Oligodendroglia/cytology , Proteins/physiology , Trans-Activators , Animals , Body Patterning/drug effects , Cell Division/drug effects , Cell Survival/drug effects , Chick Embryo , Culture Techniques , Hedgehog Proteins , Immunohistochemistry , Metencephalon/cytology , Metencephalon/metabolism , Oligodendroglia/drug effects , Oligodendroglia/metabolism , Proteins/pharmacology , Signal Transduction , Stem Cells/cytology , Stem Cells/drug effects , Stem Cells/metabolism
17.
Dev Biol ; 231(1): 77-86, 2001 Mar 01.
Article in English | MEDLINE | ID: mdl-11180953

ABSTRACT

The proliferation of oligodendrocyte lineage cells in the chick embryo central nervous system (CNS) was examined by double-immunolabeling with a lineage marker monoclonal antibody (mAb) O4 or mAb O1 and 5-bromo-3'-deoxyuridine (BrdU). In all regions examined, the first O4-positive (O4+) cells appeared in restricted regions of the ventricular zone (VZ), regarded as a site of oligodendrocyte origin. Within the O4+ focus, less than 20% of the O4+ cells incorporated BrdU. In contrast, O4+ cells in the parenchyma were mitotically active; for example, 40-50% of early O4+ cells were labeled with BrdU. Some of these were unipolar in shape, indicative of migratory precursor cells. The frequency of O4+/BrdU+ cell appearance decreased to less than 20% with further development. O1+ oligodendrocytes were largely mitotically inactive, with only approximately 5% of O1+ cells incorporating BrdU. These results clearly demonstrated that the VZ generates relatively few precursor cells and that these oligodendrocyte precursors actively generate their cohort in the parenchyma of the CNS.


Subject(s)
Metencephalon/embryology , Oligodendroglia/physiology , Spinal Cord/embryology , Stem Cells/physiology , Animals , Chick Embryo , Optic Nerve/embryology , Retina/embryology
18.
Trends Neurosci ; 24(1): 32-9, 2001 Jan.
Article in English | MEDLINE | ID: mdl-11163885

ABSTRACT

The optic tectum differentiates from the alar plate of the mesencephalon. Here, the molecular mechanisms for differentiation of the tectum are reviewed. Mis-expression of Pax2, Pax5 or En can change the fate of the presumptive diencephalon to become the tectum. En, Fgf8, Pax2 and Pax5, exist in a positive feedback loop for their expression so that mis-expression of any of these genes acts on the feedback loop resulting in induction of the optic tectum in the diencephalon. Otx2 and Gbx2 can repress the expression of each other and contribute to the formation of the posterior border of the tectum. Mis-expression of Otx2 in the metencephalon changed the fate of its alar plate to the tectum. The anterior border of the tectum might be determined as a result of repressive interaction of Pax6 with En1 and Pax2. Along the dorsoventral axis of the mesencephalon, Shh contributes to the ventralization of the tissue, that is, the area affected by Shh differentiates into the tegmentum. It is proposed that the brain vesicle that expresses Otx2, Pax2 and En1 might differentiate into the tectum.


Subject(s)
Gene Expression Regulation, Developmental/physiology , Superior Colliculi/embryology , Transcription Factors/metabolism , Animals , Antigens, CD , Antigens, Surface/metabolism , Chick Embryo , DNA-Binding Proteins/metabolism , Diencephalon/embryology , Diencephalon/metabolism , Feedback/physiology , Homeodomain Proteins/metabolism , Humans , Mesencephalon/embryology , Mesencephalon/metabolism , Metencephalon/embryology , Metencephalon/metabolism , Neural Tube Defects/metabolism , Nuclear Proteins/metabolism , PAX2 Transcription Factor , PAX5 Transcription Factor , Superior Colliculi/metabolism , Transfection/methods
19.
Mech Dev ; 99(1-2): 123-37, 2000 Dec.
Article in English | MEDLINE | ID: mdl-11091080

ABSTRACT

Three homeobox genes, one from Drosophila melanogaster (Drosophila Hmx gene) and two from mouse (murine Hmx2 and Hmx3) were isolated and the full-length cDNAs and corresponding genomic structures were characterized. The striking homeodomain similarity encoded by these three genes to previously identified genes in sea urchin, chick and human, as well as the recently cloned murine Hmx1 gene, and the low homology to other homeobox genes indicate that the Hmx genes comprise a novel gene family. The widespread existence of Hmx genes in the animal kingdom suggests that this gene family is of ancient origin. Drosophila Hmx was mapped to the 90B5 region of Chromosome 3 and at early embryonic stages is primarily expressed in distinct areas of the neuroectoderm and subsets of neuroblasts in the developing fly brain. Later its expression continues in rostral areas of the brain in a segmented pattern, suggesting a putative role in the development of the Drosophila central nervous system. During evolution, mouse Hmx2 and Hmx3 may have retained a primary function in central nervous system development as suggested by their expression in the postmitotic cells of the neural tube, as well as in the hypothalamus, the mesencephalon, metencephalon and discrete regions in the myelencephalon during embryogenesis. Hmx1 has diverged from other Hmx members by its expression in the dorsal root, sympathetic and vagal nerve (X) ganglia. Aside from their expression in the developing nervous system, all three Hmx genes display expression in sensory organ development, and in the adult uterus. Hmx2 and Hmx3 show identical expression in the otic vesicle, whereas Hmx1 is strongly expressed in the developing eye. Transgenic mouse lines were generated to examine the DNA regulatory elements controlling Hmx2 and Hmx3. Transgenic constructs spanning more than 31 kb of genomic DNA gave reproducible expression patterns in the developing central and peripheral nervous systems, eye, ear and other tissues, yet failed to fully recapitulate the endogenous expression pattern of either Hmx2 or Hmx3, suggesting both the presence and absence of certain critical enhancers in the transgenes, or the requirement of proximal enhancers to work synergistically.


Subject(s)
Brain/embryology , Drosophila Proteins , Drosophila/embryology , Embryo, Mammalian/metabolism , Evolution, Molecular , Genes, Homeobox/genetics , Homeodomain Proteins/biosynthesis , Homeodomain Proteins/genetics , Nerve Tissue Proteins/biosynthesis , Nerve Tissue Proteins/genetics , Nervous System/metabolism , Transcription Factors , Amino Acid Sequence , Animals , Base Sequence , Chromosome Mapping , DNA, Complementary/metabolism , Drosophila/genetics , Ectoderm/metabolism , Embryo, Nonmammalian/metabolism , Eye/embryology , Ganglia/embryology , Gene Library , Humans , Hypothalamus/embryology , In Situ Hybridization , Mesencephalon/embryology , Metencephalon/embryology , Mice , Mice, Transgenic , Models, Genetic , Molecular Sequence Data , Nervous System/embryology , Neural Crest/embryology , RNA/metabolism , Sequence Homology, Amino Acid , Time Factors , Transgenes
20.
Histochem Cell Biol ; 113(6): 415-26, 2000 Jun.
Article in English | MEDLINE | ID: mdl-10933218

ABSTRACT

The nature and the function of the compounds secreted by the floor plate (FP) of the metencephalon are little known. The FP cells of the hindbrain react with antibodies (AFRU) against the glycoproteins secreted by the subcommissural organ (SCO). One of the these proteins, RF-Gly I, is a 540-kDa core glycosylated protein. The aims of the present investigation were to identify by immunoblot the AFRU-immunoreactive compound secreted by the FP of chick embryos, to establish temporal and regional patterns of this secretory activity, and to obtain information about the fate of these compounds. It was established that the SCO and FP of chick embryos secrete two AFRU-immunoreactive compounds of 540 and 230 kDa. The two compounds secreted by the FP have been designated as FP-Gly I and FP-Gly II. The expression of these proteins was circumscribed to the metencephalic FP, and occurred from HH 29 to HH 36. Within the FP cells, FP-Gly I and FP-Gly II were confined to the supranuclear and apical regions, which under the electron microscope displayed numerous cisternae of the rough endoplasmic reticulum and granules. Aggregates of AFRU-immunoreactive material appeared on the free surface of the FP. The possibility that FP-Gly I and FP-Gly II are released into the ventricle to reach distant targets is discussed.


Subject(s)
Cell Adhesion Molecules, Neuronal , Metencephalon/embryology , Neurons/chemistry , Subcommissural Organ/embryology , Animals , Antibody Specificity , Blotting, Western , Cell Differentiation/physiology , Cerebral Ventricles/metabolism , Chick Embryo , Cytoplasm/chemistry , Cytoplasm/ultrastructure , Metencephalon/chemistry , Metencephalon/cytology , Microscopy, Electron , Microscopy, Electron, Scanning , Neurons/metabolism , Neurons/ultrastructure , Secretory Vesicles/chemistry , Secretory Vesicles/ultrastructure , Subcommissural Organ/chemistry , Subcommissural Organ/cytology
SELECTION OF CITATIONS
SEARCH DETAIL