Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
1.
Development ; 148(10)2021 05 15.
Article in English | MEDLINE | ID: mdl-34032267

ABSTRACT

The choroid plexus (ChP) produces cerebrospinal fluid and forms an essential brain barrier. ChP tissues form in each brain ventricle, each one adopting a distinct shape, but remarkably little is known about the mechanisms underlying ChP development. Here, we show that epithelial WNT5A is crucial for determining fourth ventricle (4V) ChP morphogenesis and size in mouse. Systemic Wnt5a knockout, or forced Wnt5a overexpression beginning at embryonic day 10.5, profoundly reduced ChP size and development. However, Wnt5a expression was enriched in Foxj1-positive epithelial cells of 4V ChP plexus, and its conditional deletion in these cells affected the branched, villous morphology of the 4V ChP. We found that WNT5A was enriched in epithelial cells localized to the distal tips of 4V ChP villi, where WNT5A acted locally to activate non-canonical WNT signaling via ROR1 and ROR2 receptors. During 4V ChP development, MEIS1 bound to the proximal Wnt5a promoter, and gain- and loss-of-function approaches demonstrated that MEIS1 regulated Wnt5a expression. Collectively, our findings demonstrate a dual function of WNT5A in ChP development and identify MEIS transcription factors as upstream regulators of Wnt5a in the 4V ChP epithelium.


Subject(s)
Choroid Plexus/embryology , Epithelium/metabolism , Fourth Ventricle/embryology , Myeloid Ecotropic Viral Integration Site 1 Protein/metabolism , Wnt-5a Protein/metabolism , Animals , Brain/embryology , CRISPR-Cas Systems/genetics , Cell Line , Epithelial Cells/metabolism , Female , HEK293 Cells , Humans , Mice , Mice, Knockout , Promoter Regions, Genetic/genetics , Receptor Tyrosine Kinase-like Orphan Receptors/metabolism , Signal Transduction/physiology , Wnt-5a Protein/genetics
3.
Am J Pathol ; 189(12): 2440-2449, 2019 12.
Article in English | MEDLINE | ID: mdl-31541646

ABSTRACT

Cells of the developing central nervous system are particularly susceptible to formation of double-stranded DNA breaks (DSBs) arising from physiological and/or environmental insults. Therefore, efficient repair of DSBs is especially vital for maintaining cellular health and proper functioning in the developing brain. Here, increased expression of DSB initiating and nonhomologous end joining repair machinery in newborn neurons in the developing brains of both mouse and human are demonstrated. In parallel, the first characterization is provided of the brain phenotype in the Lig4R278H/R278H (Lig4R/R) mouse model of DNA Ligase 4 (LIG4) syndrome, in which a hypomorphic Lig4 mutation, originally identified in patients, impedes nonhomologous end joining. It is shown that Lig4R/R mice develop nonprogressive microcephaly, resulting primarily from apoptotic death of newborn neurons that is both spatially and temporally specific during peak cortical neurogenesis. This apoptosis leads to a reduction in neurons throughout the postnatal cerebral cortex, but with a more prominent impact on those of the lower cortical layers. Together, these findings begin to uncover the pathogenesis of microcephaly in LIG4 syndrome and open avenues to more focused investigations on the critical roles of DSB formation and repair in vulnerable neuronal populations of the brain.


Subject(s)
Apoptosis , Cerebral Cortex/pathology , Craniofacial Abnormalities/complications , DNA Ligase ATP/metabolism , Disease Models, Animal , Growth Disorders/complications , Immunologic Deficiency Syndromes/complications , Microcephaly/etiology , Neurons/pathology , Animals , Cerebral Cortex/metabolism , DNA Breaks, Double-Stranded , DNA Ligase ATP/genetics , Female , Gene Knock-In Techniques , Male , Mice , Microcephaly/pathology , Mutation , Neurons/metabolism , Spatio-Temporal Analysis
4.
Nat Rev Neurosci ; 16(8): 445-57, 2015 Aug.
Article in English | MEDLINE | ID: mdl-26174708

ABSTRACT

The choroid plexus (ChP) is the principal source of cerebrospinal fluid (CSF), which has accepted roles as a fluid cushion and a sink for nervous system waste in vertebrates. Various animal models have provided insights into how the ChP-CSF system develops and matures. In addition, recent studies have uncovered new, active roles for this dynamic system in the regulation of neural stem cells, critical periods and the overall health of the nervous system. Together, these findings have brought about a paradigm shift in our understanding of brain development and health, and have stimulated new initiatives for the treatment of neurological disease.


Subject(s)
Cerebrospinal Fluid/physiology , Choroid Plexus/growth & development , Choroid Plexus/physiology , Animals , Humans
5.
Brain ; 140(5): 1267-1279, 2017 05 01.
Article in English | MEDLINE | ID: mdl-28335020

ABSTRACT

Progressive encephalopathy with oedema, hypsarrhythmia, and optic atrophy (PEHO) syndrome is an early childhood onset, severe autosomal recessive encephalopathy characterized by extreme cerebellar atrophy due to almost total granule neuron loss. By combining homozygosity mapping in Finnish families with Sanger sequencing of positional candidate genes and with exome sequencing a homozygous missense substitution of leucine for serine at codon 31 in ZNHIT3 was identified as the primary cause of PEHO syndrome. ZNHIT3 encodes a nuclear zinc finger protein previously implicated in transcriptional regulation and in small nucleolar ribonucleoprotein particle assembly and thus possibly to pre-ribosomal RNA processing. The identified mutation affects a highly conserved amino acid residue in the zinc finger domain of ZNHIT3. Both knockdown and genome editing of znhit3 in zebrafish embryos recapitulate the patients' cerebellar defects, microcephaly and oedema. These phenotypes are rescued by wild-type, but not mutant human ZNHIT3 mRNA, suggesting that the patient missense substitution causes disease through a loss-of-function mechanism. Transfection of cell lines with ZNHIT3 expression vectors showed that the PEHO syndrome mutant protein is unstable. Immunohistochemical analysis of mouse cerebellar tissue demonstrated ZNHIT3 to be expressed in proliferating granule cell precursors, in proliferating and post-mitotic granule cells, and in Purkinje cells. Knockdown of Znhit3 in cultured mouse granule neurons and ex vivo cerebellar slices indicate that ZNHIT3 is indispensable for granule neuron survival and migration, consistent with the zebrafish findings and patient neuropathology. These results suggest that loss-of-function of a nuclear regulator protein underlies PEHO syndrome and imply that establishment of its spatiotemporal interaction targets will be the basis for developing therapeutic approaches and for improved understanding of cerebellar development.


Subject(s)
Brain Edema/genetics , Brain Edema/pathology , Cerebellum/pathology , Neurodegenerative Diseases/genetics , Neurodegenerative Diseases/pathology , Neurons/pathology , Nuclear Proteins/genetics , Nuclear Proteins/physiology , Optic Atrophy/genetics , Optic Atrophy/pathology , Spasms, Infantile/genetics , Spasms, Infantile/pathology , Animals , COP9 Signalosome Complex , Cell Movement/genetics , Cell Movement/physiology , Cell Survival/genetics , Cell Survival/physiology , Cerebellum/metabolism , Edema/complications , Edema/genetics , Exome/genetics , Gene Editing , Gene Knockdown Techniques , Humans , Mice , Microcephaly/complications , Microcephaly/genetics , Mutation, Missense/genetics , Mutation, Missense/physiology , Neurons/metabolism , Nuclear Proteins/biosynthesis , Sequence Analysis, DNA , Transcription Factors/biosynthesis , Zebrafish
6.
J Neurosci ; 35(12): 4903-16, 2015 Mar 25.
Article in English | MEDLINE | ID: mdl-25810521

ABSTRACT

A sheet of choroid plexus epithelial cells extends into each cerebral ventricle and secretes signaling factors into the CSF. To evaluate whether differences in the CSF proteome across ventricles arise, in part, from regional differences in choroid plexus gene expression, we defined the transcriptome of lateral ventricle (telencephalic) versus fourth ventricle (hindbrain) choroid plexus. We find that positional identities of mouse, macaque, and human choroid plexi derive from gene expression domains that parallel their axial tissues of origin. We then show that molecular heterogeneity between telencephalic and hindbrain choroid plexi contributes to region-specific, age-dependent protein secretion in vitro. Transcriptome analysis of FACS-purified choroid plexus epithelial cells also predicts their cell-type-specific secretome. Spatial domains with distinct protein expression profiles were observed within each choroid plexus. We propose that regional differences between choroid plexi contribute to dynamic signaling gradients across the mammalian cerebroventricular system.


Subject(s)
Cerebrospinal Fluid/metabolism , Choroid Plexus/metabolism , Fourth Ventricle/metabolism , Lateral Ventricles/metabolism , Transcriptome , Aging/metabolism , Animals , Epithelial Cells/metabolism , Female , Humans , Macaca mulatta , Male , Mice
7.
Nat Cell Biol ; 18(4): 418-30, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26999738

ABSTRACT

Aberrant Notch signalling has been linked to many cancers including choroid plexus (CP) tumours, a group of rare and predominantly paediatric brain neoplasms. We developed animal models of CP tumours, by inducing sustained expression of Notch1, that recapitulate properties of human CP tumours with aberrant NOTCH signalling. Whole-transcriptome and functional analyses showed that tumour cell proliferation is associated with Sonic Hedgehog (Shh) in the tumour microenvironment. Unlike CP epithelial cells, which have multiple primary cilia, tumour cells possess a solitary primary cilium as a result of Notch-mediated suppression of multiciliate differentiation. A Shh-driven signalling cascade in the primary cilium occurs in tumour cells but not in epithelial cells. Lineage studies show that CP tumours arise from monociliated progenitors in the roof plate characterized by elevated Notch signalling. Abnormal SHH signalling and distinct ciliogenesis are detected in human CP tumours, suggesting the SHH pathway and cilia differentiation as potential therapeutic avenues.


Subject(s)
Cell Proliferation/genetics , Choroid Plexus Neoplasms/genetics , Hedgehog Proteins/genetics , Receptor, Notch1/genetics , Animals , Blotting, Western , Choroid Plexus/metabolism , Choroid Plexus/pathology , Choroid Plexus/ultrastructure , Choroid Plexus Neoplasms/metabolism , Choroid Plexus Neoplasms/pathology , Cilia/metabolism , Cilia/ultrastructure , Female , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Hedgehog Proteins/metabolism , Humans , Male , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Transgenic , Microscopy, Confocal , Microscopy, Electron, Transmission , Oligonucleotide Array Sequence Analysis , Receptor, Notch1/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction/genetics , Tumor Cells, Cultured , Tumor Microenvironment/genetics
8.
Dev Cell ; 35(6): 789-802, 2015 Dec 21.
Article in English | MEDLINE | ID: mdl-26702835

ABSTRACT

After neural tube closure, amniotic fluid (AF) captured inside the neural tube forms the nascent cerebrospinal fluid (CSF). Neuroepithelial stem cells contact CSF-filled ventricles, proliferate, and differentiate to form the mammalian brain, while neurogenic placodes, which generate cranial sensory neurons, remain in contact with the AF. Using in vivo ultrasound imaging, we quantified the expansion of the embryonic ventricular-CSF space from its inception. We developed tools to obtain pure AF and nascent CSF, before and after neural tube closure, and to define how the AF and CSF proteomes diverge during mouse development. Using embryonic neural explants, we demonstrate that age-matched fluids promote Sox2-positive neurogenic identity in developing forebrain and olfactory epithelia. Nascent CSF also stimulates SOX2-positive self-renewal of forebrain progenitor cells, some of which is attributable to LIFR signaling. Our Resource should facilitate the investigation of fluid-tissue interactions during this highly vulnerable stage of early brain development.


Subject(s)
Amniotic Fluid/metabolism , Cell Differentiation/physiology , Cerebrospinal Fluid/metabolism , Neural Tube/metabolism , Neurons/cytology , Proteome/metabolism , Animals , Cells, Cultured , Female , Mice , Neuroepithelial Cells/metabolism , Pregnancy , Signal Transduction/physiology , Stem Cells/cytology
SELECTION OF CITATIONS
SEARCH DETAIL