Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 32
Filter
Add more filters










Publication year range
1.
J Comp Neurol ; 527(9): 1461-1477, 2019 05 15.
Article in English | MEDLINE | ID: mdl-30689213

ABSTRACT

In the mouse, two telencephalic signaling centers orchestrate embryonic patterning of the cerebral cortex. From the rostral patterning center in the telencephalon, the Fibroblast Growth Factor, FGF8, disperses as a morphogen to establish the rostral to caudal axis of the neocortical area map. FGF8 coordinates with Wnt3a from the cortical hem to regulate graded expression of transcription factors that position neocortical areas, and control hippocampal development. Whether similar signaling centers pattern the much larger cortices of carnivore and primate species, however, is unclear. The limited dispersion range of FGF8 and Wnt3a is inconsistent with patterning larger cortical primordia. Yet the implication that different mechanisms organize cortex in different mammals flies in the face of the tenet that developmental patterning mechanisms are conserved across vertebrate species. In the present study, both signaling centers were identified in the ferret telencephalon, as were expression gradients of the patterning transcription factor genes regulated by FGF8 and Wnt3a. Notably, at the stage corresponding to the peak period of FGF8 signaling in the mouse neocortical primordium (NP), the NP was the same size in ferret and mouse, which would allow morphogen patterning of the ferret NP. Subsequently, the size of ferret neocortex shot past that of the mouse. Images from online databases further suggest that NP growth in humans, too, is slowed in early cortical development. We propose that if early growth in larger brains is held back, mechanisms that pattern the neocortical area map in the mouse could be conserved across mammalian species.


Subject(s)
Ferrets/embryology , Lissencephaly/embryology , Neocortex/embryology , Animals , Female , Fibroblast Growth Factor 8/biosynthesis , Fibroblast Growth Factor 8/genetics , Fibroblast Growth Factor 8/physiology , Gene Expression Regulation, Developmental , Gestational Age , Humans , In Situ Hybridization , Lissencephaly/pathology , Male , Mice , Models, Animal , Models, Neurological , Neocortex/pathology , Organ Size , Organogenesis , Signal Transduction/physiology , Somites/ultrastructure , Species Specificity , Transcription Factors/biosynthesis , Transcription Factors/genetics , Wnt3A Protein/biosynthesis , Wnt3A Protein/genetics , Wnt3A Protein/physiology
2.
J Neurosci ; 38(42): 9105-9121, 2018 10 17.
Article in English | MEDLINE | ID: mdl-30143575

ABSTRACT

Specification of dorsoventral regional identity in progenitors of the developing telencephalon is a first pivotal step in the development of the cerebral cortex and basal ganglia. Previously, we demonstrated that the two zinc finger doublesex and mab-3 related (Dmrt) genes, Dmrt5 (Dmrta2) and Dmrt3, which are coexpressed in high caudomedial to low rostrolateral gradients in the cerebral cortical primordium, are separately needed for normal formation of the cortical hem, hippocampus, and caudomedial neocortex. We have now addressed the role of Dmrt3 and Dmrt5 in controlling dorsoventral division of the telencephalon in mice of either sex by comparing the phenotypes of single knock-out (KO) with double KO embryos and by misexpressing Dmrt5 in the ventral telencephalon. We find that DMRT3 and DMRT5 act as critical regulators of progenitor cell dorsoventral identity by repressing ventralizing regulators. Early ventral fate transcriptional regulators expressed in the dorsal lateral ganglionic eminence, such as Gsx2, are upregulated in the dorsal telencephalon of Dmrt3;Dmrt5 double KO embryos and downregulated when ventral telencephalic progenitors express ectopic Dmrt5 Conditional overexpression of Dmrt5 throughout the telencephalon produces gene expression and structural defects that are highly consistent with reduced GSX2 activity. Further, Emx2;Dmrt5 double KO embryos show a phenotype similar to Dmrt3;Dmrt5 double KO embryos, and both DMRT3, DMRT5 and the homeobox transcription factor EMX2 bind to a ventral telencephalon-specific enhancer in the Gsx2 locus. Together, our findings uncover cooperative functions of DMRT3, DMRT5, and EMX2 in dividing dorsal from ventral in the telencephalon.SIGNIFICANCE STATEMENT We identified the DMRT3 and DMRT5 zinc finger transcription factors as novel regulators of dorsoventral patterning in the telencephalon. Our data indicate that they have overlapping functions and compensate for one another. The double, but not the single, knock-out produces a dorsal telencephalon that is ventralized, and olfactory bulb tissue takes over most remaining cortex. Conversely, overexpressing Dmrt5 throughout the telencephalon causes expanded expression of dorsal gene determinants and smaller olfactory bulbs. Furthermore, we show that the homeobox transcription factor EMX2 that is coexpressed with DMRT3 and DMRT5 in cortical progenitors cooperates with them to maintain dorsoventral patterning in the telencephalon. Our study suggests that DMRT3/5 function with EMX2 in positioning the pallial-subpallial boundary by antagonizing the ventral homeobox transcription factor GSX2.


Subject(s)
Homeodomain Proteins/physiology , Neural Stem Cells/physiology , Neurons/physiology , Telencephalon/embryology , Transcription Factors/physiology , Animals , Female , Gene Expression Profiling , Gene Expression Regulation, Developmental , Homeodomain Proteins/genetics , Male , Mice, Inbred C57BL , Mice, Knockout , Neural Stem Cells/metabolism , Neurons/metabolism , Telencephalon/metabolism , Transcription Factors/genetics
3.
Cereb Cortex ; 28(2): 493-509, 2018 02 01.
Article in English | MEDLINE | ID: mdl-28031177

ABSTRACT

Mice that are constitutively null for the zinc finger doublesex and mab-3 related (Dmrt) gene, Dmrt5/Dmrta2, show a variety of patterning abnormalities in the cerebral cortex, including the loss of the cortical hem, a powerful cortical signaling center. In conditional Dmrt5 gain of function and loss of function mouse models, we generated bidirectional changes in the neocortical area map without affecting the hem. Analysis indicated that DMRT5, independent of the hem, directs the rostral-to-caudal pattern of the neocortical area map. Thus, DMRT5 joins a small number of transcription factors shown to control directly area size and position in the neocortex. Dmrt5 deletion after hem formation also reduced hippocampal size and shifted the position of the neocortical/paleocortical boundary. Dmrt3, like Dmrt5, is expressed in a gradient across the cortical primordium. Mice lacking Dmrt3 show cortical patterning defects akin to but milder than those in Dmrt5 mutants, perhaps in part because Dmrt5 expression increases in the absence of Dmrt3. DMRT5 upregulates Dmrt3 expression and negatively regulates its own expression, which may stabilize the level of DMRT5. Together, our findings indicate that finely tuned levels of DMRT5, together with DMRT3, regulate patterning of the cerebral cortex.


Subject(s)
Embryonic Development/physiology , Hippocampus/metabolism , Neocortex/metabolism , Transcription Factors/biosynthesis , Animals , Hippocampus/embryology , Hippocampus/growth & development , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Neocortex/embryology , Neocortex/growth & development , Neurogenesis/physiology
4.
Cereb Cortex ; 27(5): 2841-2856, 2017 05 01.
Article in English | MEDLINE | ID: mdl-27178193

ABSTRACT

A unique population of cells, called "lot cells," circumscribes the path of the lateral olfactory tract (LOT) in the rodent brain and acts to restrict its position at the lateral margin of the telencephalon. Lot cells were believed to originate in the dorsal pallium (DP). We show that Lhx2 null mice that lack a DP show a significant increase in the number of mGluR1/lot cells in the piriform cortex, indicating a non-DP origin of these cells. Since lot cells present common developmental features with Cajal-Retzius (CR) cells, we analyzed Wnt3a- and Dbx1-reporter mouse lines and found that mGluR1/lot cells are not generated in the cortical hem, ventral pallium, or septum, the best characterized sources of CR cells. Finally, we identified a novel origin for the lot cells by combining in utero electroporation assays and histochemical characterization. We show that mGluR1/lot cells are specifically generated in the lateral thalamic eminence and that they express mitral cell markers, although a minority of them express ΔNp73 instead. We conclude that most mGluR1/lot cells are prospective mitral cells migrating to the accessory olfactory bulb (OB), whereas mGluR1+, ΔNp73+ cells are CR cells that migrate through the LOT to the piriform cortex and the OB.


Subject(s)
Gene Expression Regulation, Developmental/physiology , Receptors, Metabotropic Glutamate/metabolism , Stem Cells/physiology , Thalamus/cytology , Thalamus/metabolism , Animals , Cell Movement , Cells, Cultured , Embryo, Mammalian , Female , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , LIM-Homeodomain Proteins/genetics , LIM-Homeodomain Proteins/metabolism , Mice , Mice, Inbred C57BL , Mice, Inbred ICR , Mice, Transgenic , Nerve Tissue Proteins/metabolism , Neurogenesis/physiology , Pregnancy , Receptors, Metabotropic Glutamate/genetics , T-Box Domain Proteins/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism , Tumor Protein p73/genetics , Tumor Protein p73/metabolism
5.
Neuron ; 92(2): 435-448, 2016 Oct 19.
Article in English | MEDLINE | ID: mdl-27693257

ABSTRACT

The neocortex undergoes extensive developmental growth, but how its architecture adapts to expansion remains largely unknown. Here, we investigated how early born Cajal-Retzius (CR) neurons, which regulate the assembly of cortical circuits, maintain a dense superficial distribution in the growing neocortex. We found that CR cell density is sustained by an activity-dependent importation of olfactory CR cells, which migrate into the neocortex after they have acted as axonal guidepost cells in the olfactory system. Furthermore, using mouse genetics, we showed that CR cell density severely affects the architecture of layer 1, a key site of input integration for neocortical networks, leading to an excitation/inhibition ratio imbalance. Our study reveals that neurons reenter migration several days after their initial positioning, thereby performing sequential developmental roles in olfactory cortex and neocortex. This atypical process is essential to regulate CR cell density during growth, which in turn ensures the correct wiring of neocortical circuitry. VIDEO ABSTRACT.


Subject(s)
Cell Count , Neocortex/embryology , Neurons/physiology , Olfactory Bulb/embryology , Olfactory Cortex/embryology , Animals , Axons , Cell Movement , Interneurons/physiology , Mice , Olfactory Bulb/cytology
6.
Proc Natl Acad Sci U S A ; 111(29): E2996-3004, 2014 Jul 22.
Article in English | MEDLINE | ID: mdl-25002511

ABSTRACT

Celsr3 and Fzd3, members of "core planar cell polarity" (PCP) genes, were shown previously to control forebrain axon guidance and wiring by acting in axons and/or guidepost cells. Here, we show that Celsr2 acts redundantly with Celsr3, and that their combined mutation mimics that of Fzd3. The phenotypes generated upon inactivation of Fzd3 in different forebrain compartments are similar to those in conditional Celsr2-3 mutants, indicating that Fzd3 and Celsr2-3 act in the same population of cells. Inactivation of Celsr2-3 or Fzd3 in thalamus does not affect forebrain wiring, and joint inactivation in cortex and thalamus adds little to cortical inactivation alone in terms of thalamocortical projections. On the other hand, joint inactivation perturbs strongly the formation of the barrel field, which is unaffected upon single cortical or thalamic inactivation, indicating a role for interactions between thalamic axons and cortical neurons in cortical arealization. Unexpectedly, forebrain wiring is normal in mice defective in Vangl1 and Vangl2, showing that, contrary to epithelial PCP, axon guidance can be Vangl independent in some contexts. Our results suggest that Celsr2-3 and Fzd3 regulate axonal navigation in the forebrain by using mechanisms different from classical epithelial PCP, and require interacting partners other than Vangl1-2 that remain to be identified.


Subject(s)
Cadherins/metabolism , Carrier Proteins/metabolism , Frizzled Receptors/metabolism , Membrane Proteins/metabolism , Nerve Net/metabolism , Nerve Tissue Proteins/metabolism , Prosencephalon/metabolism , Receptors, Cell Surface/metabolism , Animals , Axons/metabolism , Cerebral Cortex/metabolism , Gene Silencing , Integrases/metabolism , Mice , Mutation/genetics , Phenotype , Thalamus/metabolism
7.
Development ; 141(14): 2855-65, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24948604

ABSTRACT

The cortical hem, a source of Wingless-related (WNT) and bone morphogenetic protein (BMP) signaling in the dorsomedial telencephalon, is the embryonic organizer for the hippocampus. Whether the hem is a major regulator of cortical patterning outside the hippocampus has not been investigated. We examined regional organization across the entire cerebral cortex in mice genetically engineered to lack the hem. Indicating that the hem regulates dorsoventral patterning in the cortical hemisphere, the neocortex, particularly dorsomedial neocortex, was reduced in size in late-stage hem-ablated embryos, whereas cortex ventrolateral to the neocortex expanded dorsally. Unexpectedly, hem ablation also perturbed regional patterning along the rostrocaudal axis of neocortex. Rostral neocortical domains identified by characteristic gene expression were expanded, and caudal domains diminished. A similar shift occurs when fibroblast growth factor (FGF) 8 is increased at the rostral telencephalic organizer, yet the FGF8 source was unchanged in hem-ablated brains. Rather we found that hem WNT or BMP signals, or both, have opposite effects to those of FGF8 in regulating transcription factors that control the size and position of neocortical areas. When the hem is ablated a necessary balance is perturbed, and cerebral cortex is rostralized. Our findings reveal a much broader role for the hem in cortical development than previously recognized, and emphasize that two major signaling centers interact antagonistically to pattern cerebral cortex.


Subject(s)
Body Patterning/genetics , Neocortex/embryology , Neocortex/metabolism , Organizers, Embryonic/embryology , Organizers, Embryonic/metabolism , Animals , Bone Morphogenetic Proteins/metabolism , Cell Proliferation , Female , Fibroblast Growth Factor 8/metabolism , Gene Deletion , Gene Expression Regulation, Developmental , Hippocampus/cytology , Hippocampus/metabolism , Mice , Neocortex/cytology , Organ Size , Organizers, Embryonic/cytology , Phenotype , Signal Transduction/genetics , Stem Cells/cytology , Stem Cells/metabolism , Wnt Proteins/metabolism
8.
Neuron ; 77(3): 472-84, 2013 Feb 06.
Article in English | MEDLINE | ID: mdl-23395374

ABSTRACT

Major outputs of the neocortex are conveyed by corticothalamic axons (CTAs), which form reciprocal connections with thalamocortical axons, and corticosubcerebral axons (CSAs) headed to more caudal parts of the nervous system. Previous findings establish that transcriptional programs define cortical neuron identity and suggest that CTAs and thalamic axons may guide each other, but the mechanisms governing CTA versus CSA pathfinding remain elusive. Here, we show that thalamocortical axons are required to guide pioneer CTAs away from a default CSA-like trajectory. This process relies on a hold in the progression of cortical axons, or waiting period, during which thalamic projections navigate toward cortical axons. At the molecular level, Sema3E/PlexinD1 signaling in pioneer cortical neurons mediates a "waiting signal" required to orchestrate the mandatory meeting with reciprocal thalamic axons. Our study reveals that temporal control of axonal progression contributes to spatial pathfinding of cortical projections and opens perspectives on brain wiring.


Subject(s)
Cerebral Cortex/physiology , Neural Pathways/physiology , Thalamus/physiology , Age Factors , Animals , Axons/physiology , Body Patterning/genetics , Calbindin 2 , Cerebral Cortex/cytology , Contactin 2/metabolism , Cytoskeletal Proteins , DNA-Binding Proteins/metabolism , Embryo, Mammalian , Gene Expression Regulation, Developmental/genetics , Glycoproteins/genetics , Homeodomain Proteins/genetics , Intracellular Signaling Peptides and Proteins , Leukocyte L1 Antigen Complex/metabolism , Luminescent Proteins/genetics , Luminescent Proteins/metabolism , Membrane Glycoproteins/genetics , Membrane Proteins/genetics , Mice , Mice, Inbred C57BL , Mice, Transgenic , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Nuclear Proteins/metabolism , POU Domain Factors/genetics , Repressor Proteins/metabolism , S100 Calcium Binding Protein G/metabolism , Semaphorins , T-Box Domain Proteins , Thalamus/cytology , Thyroid Nuclear Factor 1 , Transcription Factors/genetics , Transcription Factors/metabolism , Tumor Suppressor Proteins/metabolism , Wnt3A Protein/genetics , tau Proteins/genetics
9.
J Neurosci ; 32(21): 7191-201, 2012 May 23.
Article in English | MEDLINE | ID: mdl-22623663

ABSTRACT

The concept of an "organizer" is basic to embryology. An organizer is a portion of the embryo producing signals that lead to the creation of a patterned mature structure from an embryonic primordium. Fibroblast growth factor 8 (FGF8) is a morphogen that disperses from a rostromedial source in the neocortical primordium (NP), forms a rostral-to-caudal (R/C) gradient, and regulates embryonic and neonatal R/C patterns of gene expression in neocortex. Whether FGF8 also has organizer activity that generates the postnatal neocortical area map is uncertain. To test this possibility, new sources of FGF8 were introduced into the mouse NP with in utero microelectroporation at embryonic day 10.5, close to the estimated peak of area patterning. Results differed depending on the position of ectopic FGF8. Ectopic FGF8 in the caudalmost NP could duplicate somatosensory cortex (S1) and primary visual cortex (V1). FGF8 delivered to the midlateral NP generated a sulcus separating rostral and caudal portions of the NP, in effect creating duplicate NPs. In the caudal NP, ectopic FGF8 induced a second, inclusive area map, containing frontal cortex, S1, V1, and primary auditory areas. Moreover, duplicate S1 showed plasticity to sensory deprivation, and duplicate V1 responded to visual stimuli. Our findings implicate FGF8 as an organizer signal, and its source in the rostromedial telencephalon as an organizer of the neocortical area map.


Subject(s)
Fibroblast Growth Factor 8/physiology , Neocortex/growth & development , Animals , Brain Mapping/methods , Electroporation/methods , Female , Fibroblast Growth Factor 8/administration & dosage , Male , Mice , Mice, Inbred C57BL , Mice, Inbred ICR , Morphogenesis/physiology , Neocortex/blood supply , Neocortex/physiology , Neuronal Plasticity/physiology , Photic Stimulation/methods , Pregnancy , Sensory Deprivation/physiology , Visual Perception/physiology
10.
Nature ; 483(7389): 289-94, 2012 Mar 14.
Article in English | MEDLINE | ID: mdl-22422262

ABSTRACT

Neuroectodermal signalling centres induce and pattern many novel vertebrate brain structures but are absent, or divergent, in invertebrate chordates. This has led to the idea that signalling-centre genetic programs were first assembled in stem vertebrates and potentially drove morphological innovations of the brain. However, this scenario presumes that extant cephalochordates accurately represent ancestral chordate characters, which has not been tested using close chordate outgroups. Here we report that genetic programs homologous to three vertebrate signalling centres-the anterior neural ridge, zona limitans intrathalamica and isthmic organizer-are present in the hemichordate Saccoglossus kowalevskii. Fgf8/17/18 (a single gene homologous to vertebrate Fgf8, Fgf17 and Fgf18), sfrp1/5, hh and wnt1 are expressed in vertebrate-like arrangements in hemichordate ectoderm, and homologous genetic mechanisms regulate ectodermal patterning in both animals. We propose that these genetic programs were components of an unexpectedly complex, ancient genetic regulatory scaffold for deuterostome body patterning that degenerated in amphioxus and ascidians, but was retained to pattern divergent structures in hemichordates and vertebrates.


Subject(s)
Biological Evolution , Brain/anatomy & histology , Brain/embryology , Chordata/anatomy & histology , Chordata/embryology , Signal Transduction , Vertebrates/anatomy & histology , Vertebrates/embryology , Animals , Brain/physiology , Chordata/genetics , Chordata/physiology , Fibroblast Growth Factors/metabolism , Hedgehog Proteins/metabolism , Mice , Vertebrates/genetics , Vertebrates/physiology , Wnt Signaling Pathway
11.
Dev Biol ; 359(2): 242-50, 2011 Nov 15.
Article in English | MEDLINE | ID: mdl-21925158

ABSTRACT

In human holoprosencephaly (HPE), the forebrain does not separate fully into two hemispheres. Further, the border between the telencephalon and diencephalon, the telencephalic/diencephalic junction (TDJ), is often indistinct, and the ventricular system can be blocked at the third ventricle, creating a forebrain 'holosphere'. Mice deficient in Sonic Hedgehog (Shh) have previously been described to show HPE and associated cyclopia. Here we report that the third ventricle is blocked in Shh null mutants, similar to human HPE, and that characteristic telencephalic and diencephalic signaling centers, the cortical hem and zona limitans intrathalamica (ZLI), are merged, obliterating the TDJ. The resulting forebrain holosphere comprises Foxg1-positive telencephalic- and Foxg1-negative diencephalic territories. Loss of one functional copy of Gli3 in Shh nulls rescues ventricular collapse and substantially restores the TDJ. Characteristic regional gene expression patterns are rescued on the telencephalic side of the TDJ but not in the diencephalon. Further analysis of compound Shh;Gli3 mutants revealed an unexpected type of signaling center deregulation. In Shh;Gli3 mutants, adjacent rings of Fgf8 and Wnt3a expression are induced in the diencephalon at the ZLI, reminiscent of the Fgf8/Wnt1-expressing isthmic organizer. Neither Shh nor Gli3 single mutants show this forebrain double ring of Fgf/Wnt expression; thus both Shh and Gli3 are independently required to suppress it. Adjacent tissue is not respecified to a midbrain/hindbrain fate, but shows overgrowth, consistent with ectopic mitogen expression. Our observations indicate that the separation of the telencephalon and diencephalon depends on interactions between Shh and Gli3, and, moreover, demonstrate that both Shh and Gli3 suppress a potential Fgf/Wnt signaling source in the forebrain. That optional signaling centers are actively repressed in normal development is a striking new insight into the processes of vertebrate brain development.


Subject(s)
Diencephalon/metabolism , Hedgehog Proteins/genetics , Kruppel-Like Transcription Factors/genetics , Nerve Tissue Proteins/genetics , Prosencephalon/metabolism , Telencephalon/metabolism , Animals , Diencephalon/embryology , Embryo, Mammalian/embryology , Embryo, Mammalian/metabolism , Female , Fibroblast Growth Factor 8/genetics , Gene Expression Regulation, Developmental , Holoprosencephaly/embryology , Holoprosencephaly/genetics , Humans , In Situ Hybridization , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Prosencephalon/embryology , Signal Transduction/genetics , Telencephalon/embryology , Third Ventricle/embryology , Third Ventricle/metabolism , Time Factors , Wnt Proteins/genetics , Wnt1 Protein/genetics , Zinc Finger Protein Gli3
12.
Genes Dev ; 25(17): 1759-62, 2011 Sep 01.
Article in English | MEDLINE | ID: mdl-21896652

ABSTRACT

In canonical Wnt signaling, ß-catenin translocates to the cell nucleus, interacting with Tcf/Lef factors to activate transcription of Wnt target genes. In this issue of Genes & Development, Vacik and colleagues (pp. 1783-1795) report that a highly conserved sequence in intron 5 of Tcf7l2 conceals an internal promoter region that, when activated by Vax2, drives transcription of truncated Tcf7l2 mRNAs. The encoded Tcf7l2 protein binds to DNA, but not ß-catenin, and therefore acts as a dominant-negative Wnt antagonist.


Subject(s)
Gene Expression Regulation, Developmental , Signal Transduction , Wnt Proteins/metabolism , Xenopus laevis/embryology , Animals , Humans
13.
Neuron ; 69(6): 1046-60, 2011 Mar 24.
Article in English | MEDLINE | ID: mdl-21435552

ABSTRACT

The primary cilium is a cellular organelle that is almost ubiquitous in eukaryotes, yet its functions in vertebrates have been slow to emerge. The last fifteen years have been marked by accelerating insight into the biology of primary cilia, arising from the synergy of three major lines of research. These research programs describe a specialized mode of protein trafficking in cilia, reveal that genetic disruptions of primary cilia cause complex human disease syndromes, and establish that Sonic hedgehog (Shh) signal transduction requires the primary cilium. New lines of research have branched off to investigate the role of primary cilia in neuronal signaling, adult neurogenesis, and brain tumor formation. We review a fast expanding literature to determine what we now know about the primary cilium in the developing and adult CNS and what new directions should lead to further clarity.


Subject(s)
Central Nervous System/physiology , Cilia/physiology , Animals , Hedgehog Proteins/metabolism , Humans , Neurogenesis/physiology , Signal Transduction/physiology
14.
Cereb Cortex ; 21(4): 748-55, 2011 Apr.
Article in English | MEDLINE | ID: mdl-20713502

ABSTRACT

Cerebral cortical γ-aminobutyric acid (GABA)ergic interneurons originate from the basal forebrain and migrate into the cortex in 2 phases. First, interneurons cross the boundary between the developing striatum and the cortex to migrate tangentially through the cortical primordium. Second, interneurons migrate radially to their correct neocortical layer position. A previous study demonstrated that mice in which the cortical hem was genetically ablated displayed a massive reduction of Cajal-Retzius (C-R) cells in the neocortical marginal zone (MZ), thereby losing C-R cell-generated reelin in the MZ. Surprisingly, pyramidal cell migration and subsequent layering were almost normal. In contrast, we find that the timing of migration of cortical GABAergic interneurons is abnormal in hem-ablated mice. Migrating interneurons both advance precociously along their tangential path and switch prematurely from tangential to radial migration to invade the cortical plate (CP). We propose that the cortical hem is responsible for establishing cues that control the timing of interneuron migration. In particular, we suggest that loss of a repellant signal from the medial neocortex, which is greatly decreased in size in hem-ablated mice, allows the early advance of interneurons and that reduction of another secreted molecule from C-R cells, the chemokine SDF-1/CXCL12, permits early radial migration into the CP.


Subject(s)
Cell Movement/physiology , Cerebral Cortex/embryology , Interneurons/cytology , Neurogenesis/physiology , Animals , In Situ Hybridization, Fluorescence , Mice , Mice, Mutant Strains , Reelin Protein
15.
Development ; 137(20): 3439-48, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20843859

ABSTRACT

Gain- and loss-of-function experiments have demonstrated that a source of fibroblast growth factor (FGF) 8 regulates anterior to posterior (A/P) patterning in the neocortical area map. Whether FGF8 controls patterning as a classic diffusible morphogen has not been directly tested. We report evidence that FGF8 diffuses through the mouse neocortical primordium from a discrete source in the anterior telencephalon, forms a protein gradient across the entire A/P extent of the primordium, and acts directly at a distance from its source to determine area identity. FGF8 immunofluorescence revealed FGF8 protein distributed in an A/P gradient. Fate-mapping experiments showed that outside the most anterior telencephalon, neocortical progenitor cells did not express Fgf8, nor were they derived from Fgf8-expressing cells, suggesting that graded distribution of FGF8 results from protein diffusion from the anterior source. Supporting this conclusion, a dominant-negative high-affinity FGF8 receptor captured endogenous FGF8 at a distance from the FGF8 source. New FGF8 sources introduced by electroporation showed haloes of FGF8 immunofluorescence indicative of FGF8 diffusion, and surrounding cells reacted to a new source of FGF8 by upregulating different FGF8-responsive genes in concentric domains around the source. Reducing endogenous FGF8 with the dominant-negative receptor in the central neocortical primordium induced cells to adopt a more posterior area identity, demonstrating long-range area patterning by FGF8. These observations support FGF8 as a classic diffusible morphogen in neocortex, thereby guiding future studies of neocortical pattern formation.


Subject(s)
Body Patterning/physiology , Fibroblast Growth Factor 8/metabolism , Gene Expression Regulation, Developmental/physiology , Neocortex/embryology , Animals , Antibodies, Monoclonal , Electroporation , Fluorescent Antibody Technique , Immunohistochemistry , In Situ Hybridization , Mice , Microscopy, Confocal , Neocortex/metabolism , Receptors, Fibroblast Growth Factor/metabolism
16.
J Neurosci ; 30(18): 6291-301, 2010 May 05.
Article in English | MEDLINE | ID: mdl-20445055

ABSTRACT

The cortical hem is an embryonic signaling center that generates bone morphogenetic proteins (BMPs) and acts as an organizer for the hippocampus. The role of BMP signaling in hippocampal neurogenesis, however, has not been established. We therefore generated mice that were deficient in Bmpr1b constitutively, and deficient in Bmpr1a conditionally in the dorsal telencephalon. In double mutant male and female mice, the dentate gyrus (DG) was dramatically smaller than in control mice, reflecting decreased production of granule neurons at the peak period of DG neurogenesis. Additionally, the pool of cells that generates new DG neurons throughout life was reduced, commensurate with the smaller size of the DG. Effects of diminished BMP signaling on the cortical hem were at least partly responsible for these defects in DG development. Reduction of the DG and its major extrinsic output to CA3 raised the possibility that the DG was functionally compromised. We therefore looked for behavioral deficits in double mutants and found that the mice were less responsive to fear- or anxiety-provoking stimuli, whether the association of the stimulus with fear or anxiety was learned or innate. Given that no anatomical defects appeared in the double mutant telencephalon outside the DG, our observations support a growing literature that implicates the hippocampus in circuitry mediating fear and anxiety. Our results additionally indicate a requirement for BMP signaling in generating the dorsalmost neuronal lineage of the telencephalon, DG granule neurons, and in the development of the stem cell niche that makes neurons in the adult hippocampus.


Subject(s)
Bone Morphogenetic Protein 1 , Dentate Gyrus , Fear , Neurogenesis/genetics , Telencephalon/growth & development , Animals , Animals, Newborn , Behavior, Animal/physiology , Bone Morphogenetic Protein 1/genetics , Bone Morphogenetic Protein 1/metabolism , Bone Morphogenetic Protein 1/physiology , Dentate Gyrus/anatomy & histology , Dentate Gyrus/embryology , Dentate Gyrus/growth & development , Dentate Gyrus/metabolism , Gene Expression Regulation, Developmental , Maze Learning/physiology , Mice , Mice, Knockout , Mossy Fibers, Hippocampal/anatomy & histology , Mossy Fibers, Hippocampal/growth & development , Signal Transduction/genetics , Signal Transduction/physiology , Telencephalon/embryology , Wnt Proteins/genetics , Wnt Proteins/metabolism , Wnt3 Protein
17.
Development ; 135(13): 2203-6, 2008 Jul.
Article in English | MEDLINE | ID: mdl-18539920

ABSTRACT

The RIKEN Center for Developmental Biology recently held its 2008 Symposium ;Turning Neurons into a Nervous System' in Kobe, Japan. The program, organized by Masatoshi Takeichi, Joshua Sanes, Hideki Enomoto and Raj Ladher, provided a rich sampling from current work in developmental neurobiology. Researchers from Japan, Europe and the USA gathered at this meeting to share insights into neural development and to admire the opening of the cherry blossom season.


Subject(s)
Nervous System/cytology , Nervous System/metabolism , Neurons/cytology , Neurons/physiology , Animals , Cell Differentiation , Cell Shape , Humans , Models, Biological , Nervous System/chemistry , Nervous System/growth & development , Retinal Ganglion Cells/cytology , Retinal Ganglion Cells/metabolism
19.
J Neurosci ; 27(43): 11595-603, 2007 Oct 24.
Article in English | MEDLINE | ID: mdl-17959802

ABSTRACT

Division of the telencephalic vesicle into hemispheres and specification of the cerebral cortex are key stages in forebrain development. We investigate the interplay in these processes of Sonic hedgehog (Shh), fibroblast growth factors (Fgfs), and the transcription factor Gli3, which in its repressor form (Gli3R) antagonizes Shh signaling and downregulates expression of several Fgf genes. Contrary to previous reports, Shh is not required for dorsal hemisphere separation. Mice lacking Shh develop a dorsal telencephalic midline, a cortical hem, and two cortical hemispheres. The hemispheres do not divide rostrally, probably because of reduced local Fgf gene expression, resulting from the loss of Shh inhibition of Gli3R. Removing one functional copy of Gli3 substantially rescues Fgf expression and rostral telencephalic morphology. In mice lacking Gli3 function, cortical development is arrested, and ventral gene expression invades the dorsal telencephalon. These defects are potentially explained by disinhibition of Shh activity. However, when both copies of Shh are removed from Gli3-null mice, dorsal telencephalic defects persist. One such defect is a large dorsal expansion of the expression of Fgf genes. Fgf15 expression, for example, expands from a discrete ventral domain throughout the dorsal telencephalon. We propose that Fgf signaling, known to ventralize the telencephalon in a Shh-independent manner, suppresses cortical fate in the absence of Gli3. Our findings point away from Shh involvement in dorsal telencephalic patterning and encourage additional exploration of Fgf signaling and Gli3 repression in corticogenesis.


Subject(s)
Hedgehog Proteins/physiology , Telencephalon/embryology , Telencephalon/metabolism , Animals , Gene Expression Regulation, Developmental/physiology , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Mutant Strains , Telencephalon/cytology
20.
J Comp Neurol ; 504(5): 550-69, 2007 Oct 10.
Article in English | MEDLINE | ID: mdl-17701978

ABSTRACT

The dorsal midline of the vertebrate neural tube is a source of signals that direct cell fate specification and proliferation. Using genetic fate mapping in the mouse and a previously generated Wnt3aCre line, we report here that genetically labeled cells of the Wnt3a lineage migrate widely from the dorsal midline into the dorsal half of the adult brain and spinal cord, contributing to diverse structures in the diencephalon, midbrain, and brainstem and extensively populating the rostral spinal cord. Conspicuously, many of these structures are linked in specific functional networks. Wnt3a lineage cells populate nuclei of the central auditory system from the medulla to thalamus, and the trigeminal sensory system from the cervical spinal cord to the midbrain. Our findings reveal the rich contributions of the Wnt3a lineage to a variety of brain structures and show that functionally integrated nuclei can share a molecular identity, provided by transient gene expression early in their development.


Subject(s)
Body Patterning/genetics , Central Nervous System/embryology , Central Nervous System/physiology , Wnt Proteins/genetics , Animals , Cell Differentiation/genetics , Cell Lineage/genetics , Cell Movement/genetics , Central Nervous System/cytology , Embryo, Mammalian , Gene Expression Regulation, Developmental/physiology , In Situ Hybridization , Mice , Mice, Transgenic , Wnt3 Protein , Wnt3A Protein , beta-Galactosidase/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...