Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
1.
J Comp Neurol ; 531(16): 1715-1750, 2023 11.
Article in English | MEDLINE | ID: mdl-37695031

ABSTRACT

The globus pallidus (GP) of primates is divided conventionally into distinct internal and external parts. The literature repeats since 1930 the opinion that the homolog of the primate internal pallidum in rodents is the hypothalamic entopeduncular nucleus (embedded within fiber tracts of the cerebral peduncle). To test this idea, we explored its historic fundaments, checked the development and genoarchitecture of mouse entopeduncular and pallidal neurons, and examined relevant comparative connectivity data. We found that the extratelencephalic mouse entopeduncular structure consists of four different components arrayed along a dorsoventral sequence in the alar hypothalamus. The ventral entopeduncular nucleus (EPV), with GABAergic neurons expressing Dlx5&6 and Nkx2-1, lies within the hypothalamic peduncular subparaventricular area. Three other formations-the dorsal entopeduncular nucleus (EPD), the prereticular entopeduncular nucleus (EPPRt ), and the preeminential entopeduncular nucleus (EPPEm )-lie within the overlying paraventricular area, under the subpallium. EPD contains glutamatergic neurons expressing Tbr1, Otp, and Pax6. The EPPRt has GABAergic cells expressing Isl1 and Meis2, whereas the EPPEm population expresses Foxg1 and may be glutamatergic. Genoarchitectonic observations on relevant areas of the mouse pallidal/diagonal subpallium suggest that the GP of rodents is constituted as in primates by two adjacent but molecularly and hodologically differentiable telencephalic portions (both expressing Foxg1). These and other reported data oppose the notion that the rodent extratelencephalic entopeduncular nucleus is homologous to the primate internal pallidum. We suggest instead that all mammals, including rodents, have dual subpallial GP components, whereas primates probably also have a comparable set of hypothalamic entopeduncular nuclei. Remarkably, there is close similarity in some gene expression properties of the telencephalic internal GP and the hypothalamic EPV. This apparently underlies their notable functional analogy, sharing GABAergic neurons and thalamopetal connectivity.


Subject(s)
Globus Pallidus , Rodentia , Animals , Mice , Entopeduncular Nucleus , Hypothalamus , Primates , GABAergic Neurons , Transcription Factors/genetics , Nerve Tissue Proteins , Forkhead Transcription Factors
2.
Mol Neurobiol ; 60(2): 687-731, 2023 Feb.
Article in English | MEDLINE | ID: mdl-36357614

ABSTRACT

The lateral hypothalamus (LH) has a heterogeneous cytoarchitectonic organization that has not been elucidated in detail. In this work, we analyzed within the framework of the prosomeric model the differential expression pattern of 59 molecular markers along the ventrodorsal dimension of the medial forebrain bundle in the mouse, considering basal and alar plate subregions of the LH. We found five basal (LH1-LH5) and four alar (LH6-LH9) molecularly distinct sectors of the LH with neuronal cell groups that correlate in topography with previously postulated alar and basal hypothalamic progenitor domains. Most peptidergic populations were restricted to one of these LH sectors though some may have dispersed into a neighboring sector. For instance, histaminergic Hdc-positive neurons were mostly contained within the basal LH3, Nts (neurotensin)- and Tac2 (tachykinin 2)-expressing cells lie strictly within LH4, Hcrt (hypocretin/orexin)-positive and Pmch (pro-melanin-concentrating hormone)-positive neurons appeared within separate LH5 subdivisions, Pnoc (prepronociceptin)-expressing cells were mainly restricted to LH6, and Sst (somatostatin)-positive cells were identified within the LH7 sector. The alar LH9 sector, a component of the Foxg1-positive telencephalo-opto-hypothalamic border region, selectively contained Satb2-expressing cells. Published studies of rodent LH subdivisions have not described the observed pattern. Our genoarchitectonic map should aid in systematic approaches to elucidate LH connectivity and function.


Subject(s)
Hypothalamic Area, Lateral , Neuropeptides , Mice , Animals , Hypothalamic Area, Lateral/metabolism , Orexins/metabolism , Neuropeptides/metabolism , Hypothalamus/metabolism , Neurons/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Nerve Tissue Proteins/metabolism , Forkhead Transcription Factors/metabolism
3.
Commun Biol ; 4(1): 95, 2021 01 21.
Article in English | MEDLINE | ID: mdl-33479483

ABSTRACT

GABAergic neurons of the hypothalamus regulate many innate behaviors, but little is known about the mechanisms that control their development. We previously identified hypothalamic neurons that express the LIM homeodomain transcription factor Lhx6, a master regulator of cortical interneuron development, as sleep-promoting. In contrast to telencephalic interneurons, hypothalamic Lhx6 neurons do not undergo long-distance tangential migration and do not express cortical interneuronal markers such as Pvalb. Here, we show that Lhx6 is necessary for the survival of hypothalamic neurons. Dlx1/2, Nkx2-2, and Nkx2-1 are each required for specification of spatially distinct subsets of hypothalamic Lhx6 neurons, and that Nkx2-2+/Lhx6+ neurons of the zona incerta are responsive to sleep pressure. We further identify multiple neuropeptides that are enriched in spatially segregated subsets of hypothalamic Lhx6 neurons, and that are distinct from those seen in cortical neurons. These findings identify common and divergent molecular mechanisms by which Lhx6 controls the development of GABAergic neurons in the hypothalamus.


Subject(s)
Cell Differentiation , GABAergic Neurons/physiology , Gene Regulatory Networks , Hypothalamus/cytology , LIM-Homeodomain Proteins/metabolism , Nerve Tissue Proteins/metabolism , Transcription Factors/metabolism , Animals , Cell Survival , Homeobox Protein Nkx-2.2 , Homeodomain Proteins/metabolism , Hypothalamus/metabolism , Mice , Nuclear Proteins , Sleep/physiology
4.
J Comp Neurol ; 529(2): 367-420, 2021 02.
Article in English | MEDLINE | ID: mdl-32420617

ABSTRACT

We present here a thorough and complete analysis of mouse P0-P140 prethalamic histogenetic subdivisions and corresponding nuclear derivatives, in the context of local tract landmarks. The study used as fundamental material brains from a transgenic mouse line that expresses LacZ under the control of an intragenic enhancer of Dlx5 and Dlx6 (Dlx5/6-LacZ). Subtle shadings of LacZ signal, jointly with pan-DLX immunoreaction, and several other ancillary protein or RNA markers, including Calb2 and Nkx2.2 ISH (for the prethalamic eminence, and derivatives of the rostral zona limitans shell domain, respectively) were mapped across the prethalamus. The resulting model of the prethalamic region postulates tetrapartite rostrocaudal and dorsoventral subdivisions, as well as a tripartite radial stratification, each cell population showing a characteristic molecular profile. Some novel nuclei are proposed, and some instances of potential tangential cell migration were noted.


Subject(s)
Chromosome Mapping/methods , Homeodomain Proteins/genetics , Lac Operon/genetics , Thalamus/embryology , Animals , Animals, Newborn , Female , Gene Expression , Homeodomain Proteins/biosynthesis , Mice , Mice, Inbred C57BL , Mice, Transgenic , Pregnancy , Thalamus/growth & development , Thalamus/metabolism , Zebrafish
5.
Cell Rep ; 31(2): 107495, 2020 04 14.
Article in English | MEDLINE | ID: mdl-32294447

ABSTRACT

Tbr1 is a high-confidence autism spectrum disorder (ASD) gene encoding a transcription factor with distinct pre- and postnatal functions. Postnatally, Tbr1 conditional knockout (CKO) mutants and constitutive heterozygotes have immature dendritic spines and reduced synaptic density. Tbr1 regulates expression of several genes that underlie synaptic defects, including a kinesin (Kif1a) and a WNT-signaling ligand (Wnt7b). Furthermore, Tbr1 mutant corticothalamic neurons have reduced thalamic axonal arborization. LiCl and a GSK3ß inhibitor, two WNT-signaling agonists, robustly rescue the dendritic spines and the synaptic and axonal defects, suggesting that this could have relevance for therapeutic approaches in some forms of ASD.


Subject(s)
Dendritic Spines/metabolism , T-Box Domain Proteins/metabolism , Wnt Signaling Pathway/physiology , Animals , Autism Spectrum Disorder/genetics , DNA-Binding Proteins/metabolism , Dendritic Spines/physiology , Female , HEK293 Cells , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Neurogenesis/physiology , Neurons/metabolism , Neurons/physiology , Synapses/metabolism , T-Box Domain Proteins/genetics , T-Box Domain Proteins/physiology , Thalamus/metabolism , Wnt Signaling Pathway/genetics
6.
Brain Struct Funct ; 221(6): 3095-109, 2016 07.
Article in English | MEDLINE | ID: mdl-26311466

ABSTRACT

The establishment of the brain structural complexity requires a precisely orchestrated interplay between extrinsic and intrinsic signals modulating cellular mechanisms to guide neuronal differentiation. However, little is known about the nature of these signals in the diencephalon, a complex brain region that processes and relays sensory and motor information to and from the cerebral cortex and subcortical structures. Morphogenetic signals from brain organizers regulate histogenetic processes such as cellular proliferation, migration, and differentiation. Sonic hedgehog (Shh) in the key signal of the ZLI, identified as the diencephalic organizer. Fgf15, the mouse gene orthologous of human, chick, and zebrafish Fgf19, is induced by Shh signal and expressed in the diencephalic alar plate progenitors during histogenetic developmental stages. This work investigates the role of Fgf15 signal in diencephalic development. In the absence of Fgf15, the complementary expression pattern of proneural genes: Ascl1 and Nng2, is disrupted and the GABAergic thalamic cells do not differentiate; in addition dorsal thalamic progenitors failed to exit from the mitotic cycle and to differentiate into neurons. Therefore, our findings indicate that Fgf15 is the Shh downstream signal to control thalamic regionalization, neurogenesis, and neuronal differentiation by regulating the expression and mutual segregation of neurogenic and proneural regulatory genes.


Subject(s)
Fibroblast Growth Factors/metabolism , Gene Expression Regulation, Developmental , Neural Stem Cells/metabolism , Neural Stem Cells/physiology , Thalamus/embryology , Thalamus/metabolism , Animals , Basic Helix-Loop-Helix Transcription Factors/metabolism , Cell Differentiation , Cell Proliferation , Fibroblast Growth Factor 8/metabolism , GABAergic Neurons/metabolism , GABAergic Neurons/physiology , Hedgehog Proteins/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Neurogenesis , Neurons/metabolism , Neurons/physiology , Receptor, Fibroblast Growth Factor, Type 3/metabolism , Wnt1 Protein/metabolism
7.
Proc Natl Acad Sci U S A ; 109(34): 13829-34, 2012 Aug 21.
Article in English | MEDLINE | ID: mdl-22753490

ABSTRACT

Inhibitory interneurons regulate the responses of cortical circuits. In auditory cortical areas, inhibition from these neurons narrows spectral tuning and shapes response dynamics. Acute disruptions of inhibition expand spectral receptive fields. However, the effects of long-term perturbations of inhibitory circuitry on auditory cortical responses are unknown. We ablated ~30% of dendrite-targeting cortical inhibitory interneurons after the critical period by studying mice with a conditional deletion of Dlx1. Following the loss of interneurons, baseline firing rates rose and tone-evoked responses became less sparse in auditory cortex. However, contrary to acute blockades of inhibition, the sizes of spectral receptive fields were reduced, demonstrating both higher thresholds and narrower bandwidths. Furthermore, long-latency responses at the edge of the receptive field were absent. On the basis of changes in response dynamics, the mechanism for the reduction in receptive field size appears to be a compensatory loss of cortico-cortically (CC) driven responses. Our findings suggest chronic conditions that feature changes in inhibitory circuitry are not likely to be well modeled by acute network manipulations, and compensation may be a critical component of chronic neuronal conditions.


Subject(s)
Acoustic Stimulation , Auditory Cortex/physiology , Homeodomain Proteins/genetics , Interneurons/physiology , Neural Inhibition/physiology , Neurons/physiology , Transcription Factors/genetics , Action Potentials/physiology , Animals , Dendrites/metabolism , Electroencephalography/methods , Female , Male , Mice , Mice, Knockout , Models, Genetic , Neurons/drug effects , Phenotype , Time Factors
8.
J Neurosci ; 31(2): 549-64, 2011 Jan 12.
Article in English | MEDLINE | ID: mdl-21228164

ABSTRACT

The molecular mechanisms regulating fate divergence of closely related, but distinct, layer 6 corticothalamic and layer 5 subcerebral projection neurons are largely unknown. We present evidence for central transcriptional mechanisms that regulate fate specification of corticothalamic (layer 6) and subcerebral (layer 5) projection neurons. We found that TBR1 promotes the identity of corticothalamic neurons and represses subcerebral fates through reducing expression of Fezf2 and CTIP2. These conclusions are based on the following: (1) In Tbr1(-/-) mice, the number of cells expressing layer 6 markers was reduced, and the number of cells expressing layer 5 markers was increased. Early-born (birthdated on E11.5) neurons ectopically expressed subcerebral neuronal markers, and extended their axons into subcerebral targets. (2) Ectopic Tbr1 expression in layer 5 neurons prevented them from extending axons into the brainstem and the spinal cord. (3) Chromatin immunoprecipitation analysis using TBR1 antibodies showed that TBR1 bound to a conserved region in the Fezf2 gene. (4) Analysis of Fezf2 mutants and Tbr1(-/-); Fezf2(-/-) compound mutants provided evidence that Fezf2 blocks corticothalamic fate in layer 5 by reducing Tbr1 expression in subcerebral neurons. All neocortical regions appear to use this core transcriptional program to specify corticothalamic (layer 6) and subcerebral (layer 5) projection neurons.


Subject(s)
DNA-Binding Proteins/physiology , Neocortex/cytology , Nerve Tissue Proteins/physiology , Neurons/cytology , Animals , Axons/physiology , Brain Stem/physiology , Chromatin Immunoprecipitation , DNA-Binding Proteins/biosynthesis , DNA-Binding Proteins/genetics , Immunohistochemistry , Mice , Mice, Knockout , Mutation , Neocortex/embryology , Neocortex/metabolism , Nerve Tissue Proteins/biosynthesis , Nerve Tissue Proteins/genetics , Neurogenesis , Neurons/metabolism , Protein Binding , Repressor Proteins/biosynthesis , Spinal Cord/physiology , T-Box Domain Proteins , Thalamus/cytology , Thalamus/embryology , Thalamus/metabolism , Transcription, Genetic , Tumor Suppressor Proteins/biosynthesis
9.
Brain Res Bull ; 75(2-4): 231-5, 2008 Mar 18.
Article in English | MEDLINE | ID: mdl-18331876

ABSTRACT

The paraventricular nucleus complex (Pa) is a component of central neural circuitry that regulates several homeostatic variables. The paraventricular nucleus is composed of magnocellular neurons that project to the posterior pituitary and parvicellular neurons that project to numerous sites in the central nervous system. According to the revised prosomeric model, the paraventricular nucleus is located caudal to the eye stalk along the rostrocaudal dimension of the dorsal hypothalamic alar plate. Caudally, the paraventricular nucleus abuts the prethalamus (prosomere 3), and the entire complex is flanked ventrally and dorsally by Dlx5-expressing domains of the alar plate. The homeodomain transcription factor Orthopedia (Otp) is expressed in several separate hypothalamic sites: the paraventricular nucleus, perimammillary region and arcuate nucleus. In this study, we compared Otp expression in the hypothalamus of mouse (Mus musculus), chick (Gallus gallus), frog (Rana perezi) and axolotol (Ambystoma mexicanum), using immunohistochemical and in situ hybridization techniques. In all cases, Otp-positive cells in the paraventricular nucleus were excluded from Dlx5-expressing adjacent domains. Other positive neuronal populations were observed in the arcuate nucleus and oblique perimammillary band. Expression in the medial amygdala appears to be continuous with the Otp-expressing paraventricular nucleus complex. This area is relatively unevaginated in the amphibian brains, barely evaginated in the chick, and fully evaginated in the mouse. These data led us to conclude that the expression pattern of Otp is topologically highly conserved in tetrapods and is plesiomorphic among chordates.


Subject(s)
Homeodomain Proteins/metabolism , Hypothalamus/cytology , Nerve Tissue Proteins/metabolism , Neurons/metabolism , Ambystoma , Animals , Anura , Chick Embryo , Embryo, Nonmammalian , Homeodomain Proteins/genetics , Mice , Nerve Tissue Proteins/genetics
10.
J Neurosci ; 27(17): 4786-98, 2007 Apr 25.
Article in English | MEDLINE | ID: mdl-17460091

ABSTRACT

ARX loss-of-function mutations cause X-linked lissencephaly with ambiguous genitalia (XLAG), a severe neurological condition that results in profound brain malformations, including microcephaly, absence of corpus callosum, and impairment of the basal ganglia. Despite such dramatic defects, their nature and origin remain largely unknown. Here, we used Arx mutant mice as a model to characterize the cellular and molecular mechanisms underlying the basal ganglia alterations. In these animals, the early differentiation of this tissue appeared normal, whereas subsequent differentiation was impaired, leading to the periventricular accumulation of immature neurons in both the lateral ganglionic eminence and medial ganglionic eminence (MGE). Both tangential migration toward the cortex and striatum and radial migration to the globus pallidus and striatum were greatly reduced in the mutants, causing a periventricular accumulation of NPY+ or calretinin+ neurons in the MGE. Arx mutant neurons retained their differentiation potential in vitro but exhibited deficits in morphology and migration ability. These findings imply that cell-autonomous defects in migration underlie the neuronal localization defects. Furthermore, Arx mutants lacked a large fraction of cholinergic neurons and displayed a strong impairment of thalamocortical projections, in which major axon fiber tracts failed to traverse the basal ganglia. Altogether, these results highlight the critical functions of Arx in promoting neural migration and regulating basal ganglia differentiation in mice, consistent with the phenotype of XLAG patients.


Subject(s)
Basal Ganglia/abnormalities , Basal Ganglia/pathology , Genitalia/abnormalities , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Interneurons/pathology , Transcription Factors/genetics , Transcription Factors/metabolism , Animals , Animals, Newborn , Cell Differentiation , Cell Movement , Cells, Cultured , Cerebral Cortex/abnormalities , Cerebral Cortex/pathology , Doublecortin Protein , Female , Globus Pallidus/abnormalities , Globus Pallidus/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Mutant Strains , Organ Culture Techniques , Pregnancy , Septal Nuclei/abnormalities , Septal Nuclei/pathology , Substantia Nigra/abnormalities , Substantia Nigra/pathology , Thalamus/abnormalities , Thalamus/pathology , X Chromosome
11.
Science ; 310(5749): 805-10, 2005 Nov 04.
Article in English | MEDLINE | ID: mdl-16272112

ABSTRACT

The cerebral cortex of the human brain is a sheet of about 10 billion neurons divided into discrete subdivisions or areas that process particular aspects of sensation, movement, and cognition. Recent evidence has begun to transform our understanding of how cortical areas form, make specific connections with other brain regions, develop unique processing networks, and adapt to changes in inputs.


Subject(s)
Body Patterning , Cerebral Cortex/growth & development , Cerebral Cortex/physiology , Neuronal Plasticity , Animals , Axons/physiology , Brain Mapping , Cerebral Cortex/anatomy & histology , Cerebral Cortex/metabolism , Dominance, Ocular , Gene Expression Regulation, Developmental , Humans , Models, Neurological , Morphogenesis , Nerve Net/physiology , Neural Pathways/growth & development , Neural Pathways/physiology , Thalamus/anatomy & histology , Thalamus/growth & development , Thalamus/physiology
12.
J Neurosci ; 25(33): 7586-600, 2005 Aug 17.
Article in English | MEDLINE | ID: mdl-16107646

ABSTRACT

The vertebrate telencephalon is composed of many architectonically and functionally distinct areas and structures, with billions of neurons that are precisely connected. This complexity is fine-tuned during development by numerous genes. To identify genes involved in the regulation of telencephalic development, a specific subset of differentially expressed genes was characterized. Here, we describe a set of cDNAs encoded by genes preferentially expressed during development of the mouse telencephalon that was identified through a functional genomics approach. Of 832 distinct transcripts found, 223 (27%) are known genes. Of the remaining, 228 (27%) correspond to expressed sequence tags of unknown function, 58 (7%) are homologs or orthologs of known genes, and 323 (39%) correspond to novel rare transcripts, including 48 (14%) new putative noncoding RNAs. As an example of this latter group of novel precursor transcripts of micro-RNAs, telencephalic embryonic subtractive sequence (TESS) 24.E3 was functionally characterized, and one of its targets was identified: the zinc finger transcription factor ZFP9. The TESS transcriptome has been annotated, mapped for chromosome loci, and arrayed for its gene expression profiles during neural development and differentiation (in Neuro2a and neural stem cells). Within this collection, 188 genes were also characterized on embryonic and postnatal tissue by in situ hybridization, demonstrating that most are specifically expressed in the embryonic CNS. The full information has been organized into a searchable database linked to other genomic resources, allowing easy access to those who are interested in the dissection of the molecular basis of telencephalic development.


Subject(s)
DNA, Complementary/genetics , Gene Expression Regulation, Developmental/genetics , Telencephalon/embryology , Telencephalon/physiology , Animals , Base Sequence , Cell Line, Tumor , Cells, Cultured , DNA, Complementary/biosynthesis , Gene Expression Profiling/methods , Mice , MicroRNAs/biosynthesis , MicroRNAs/genetics , Molecular Sequence Data
13.
J Biol Chem ; 280(19): 19156-65, 2005 May 13.
Article in English | MEDLINE | ID: mdl-15743757

ABSTRACT

Gonadotropin-releasing hormone (GnRH) is the central regulator of the hypothalamic-pituitary-gonadal axis, controlling sexual maturation and fertility in diverse species from fish to humans. GnRH gene expression is limited to a discrete population of neurons that migrate through the nasal region into the hypothalamus during embryonic development. The GnRH regulatory region contains four conserved homeodomain binding sites (ATTA) that are essential for basal promoter activity and cell-specific expression of the GnRH gene. MSX and DLX are members of the Antennapedia class of non-Hox homeodomain transcription factors that regulate gene expression and influence development of the craniofacial structures and anterior forebrain. Here, we report that expression patterns of the Msx and Dlx families of homeodomain transcription factors largely coincide with the migratory route of GnRH neurons and co-express with GnRH in neurons during embryonic development. In addition, MSX and DLX family members bind directly to the ATTA consensus sequences and regulate transcriptional activity of the GnRH promoter. Finally, mice lacking MSX1 or DLX1 and 2 show altered numbers of GnRH-expressing cells in regions where these factors likely function. These findings strongly support a role for MSX and DLX in contributing to spatiotemporal regulation of GnRH transcription during development.


Subject(s)
Gene Expression Regulation, Developmental , Gonadotropin-Releasing Hormone/biosynthesis , Gonadotropin-Releasing Hormone/genetics , Homeodomain Proteins/chemistry , Transcription Factors/chemistry , Animals , Base Sequence , Binding Sites , Binding, Competitive , Cell Line , Cell Nucleus/metabolism , Embryo, Mammalian/metabolism , Hypothalamus/metabolism , Immunohistochemistry , In Situ Hybridization , MSX1 Transcription Factor , Mice , Mice, Inbred C57BL , Molecular Sequence Data , Mutation , NIH 3T3 Cells , Neurons/metabolism , Oligonucleotides, Antisense/pharmacology , Promoter Regions, Genetic , Prosencephalon/metabolism , Protein Binding , Protein Structure, Tertiary , Reverse Transcriptase Polymerase Chain Reaction , Time Factors , Transcription, Genetic , Transfection
14.
J Neurosci ; 24(41): 8917-23, 2004 Oct 13.
Article in English | MEDLINE | ID: mdl-15483110

ABSTRACT

The process of generating functionally distinct neocortical areas requires the formation of an intra-neocortical connectivity map. Here, we explore the early development of murine intra-neocortical projections and find that axons from rostral and caudal neurons remain, respectively, within large rostral and caudal domains of the neonatal neocortex. Despite evidence that thalamic input can regulate neocortical areal properties, we found that the neonatal intra-neocortical projection pattern was not perturbed when thalamic input was absent in Gbx2 mutants. On the contrary, in Fgf8 hypomorphic mutants, the rostral neocortex of which acquires more caudal molecular properties, caudally located neurons ectopically project axons into the rostral cortex. Therefore, neocortical patterning by Fgf8 also contributes to arealization through mediating early development of intra-neocortical connectivity.


Subject(s)
Fibroblast Growth Factors/physiology , Neocortex/embryology , Neocortex/metabolism , Neural Pathways/embryology , Neural Pathways/metabolism , Animals , Axons/physiology , Female , Fibroblast Growth Factor 8 , Fibroblast Growth Factors/metabolism , Histocytochemistry , Homeodomain Proteins/genetics , In Situ Hybridization , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Naphthalenes , Neocortex/cytology , Neural Pathways/cytology , Neurons/cytology , Neurons/metabolism , Organophosphonates , Thalamus/physiology
15.
Trends Neurosci ; 27(9): 533-9, 2004 Sep.
Article in English | MEDLINE | ID: mdl-15331235

ABSTRACT

Topography of axonal projections has been generally thought to arise from positional information located within the projecting and targeted structures, independent of events along the path or within the axonal bundle. Recent evidence suggests that in the projection from the dorsal thalamus to the neocortex, initial rostrocaudal targeting of axons is regulated at the level of an intermediate target, the subcortical telencephalon. In this system, thalamic axons are spatially positioned within the subcortical telencephalon, partly via interactions between EphAs and ephrin-As, and this positioning apparently determines the rostrocaudal level of the neocortex that the axons will initially target.


Subject(s)
Axons/physiology , Brain Mapping , Cerebral Cortex/anatomy & histology , Neural Pathways/anatomy & histology , Thalamus/anatomy & histology , Animals , Basic Helix-Loop-Helix Transcription Factors , Carrier Proteins/genetics , Carrier Proteins/physiology , Cerebral Cortex/embryology , Growth Cones , Homeodomain Proteins/physiology , Humans , Mice , Mice, Mutant Strains , Models, Neurological , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/physiology , Neural Pathways/embryology , Receptors, Eph Family/physiology , Thalamus/embryology , Transcription Factors/deficiency , Transcription Factors/physiology
16.
Trends Neurosci ; 26(9): 469-76, 2003 Sep.
Article in English | MEDLINE | ID: mdl-12948657

ABSTRACT

The prosomeric model attributes morphological meaning to gene expression patterns and other data in the forebrain. It divides this territory into the same transverse segments (prosomeres) and longitudinal zones in all vertebrates. The axis and longitudinal zones of this model are widely accepted but controversy subsists about the number of prosomeres and their nature as segments. We describe difficulties encountered in establishing continuity between prosomeric limits postulated in the hypothalamus and intra-telencephalic limits. Such difficulties throw doubt on the intersegmental nature of these limits. We sketch a simplified model, in which the secondary prosencephalon (telencephalon plus hypothalamus) is a complex protosegment not subdivided into prosomeres, which exhibits patterning singularities. By contrast, we continue to postulate that prosomeres p1-p3 (i.e. the pretectum, thalamus and prethalamus) are the caudal forebrain.


Subject(s)
Gene Expression , Models, Neurological , Prosencephalon/metabolism , Animals , Humans , Hypothalamus/metabolism , Telencephalon/metabolism
17.
J Comp Neurol ; 457(4): 345-60, 2003 Mar 17.
Article in English | MEDLINE | ID: mdl-12561075

ABSTRACT

The homeobox transcription factors Emx1 and Emx2 are expressed in overlapping patterns that include cortical progenitors in the dorsal telencephalic neuroepithelium. We have addressed cooperation of Emx1 and Emx2 in cortical development by comparing phenotypes in Emx1; Emx2 double mutant mice with wild-type and Emx1 and Emx2 single mutants. Emx double mutant cortex is greatly reduced compared with wild types and Emx single mutants; the hippocampus and dentate gyrus are absent, and growth and lamination of the olfactory bulbs are defective. Cell proliferation and death are relatively normal early in cortical neurogenesis, suggesting that hypoplasia of the double mutant cortex is primarily due to earlier patterning defects. Expression of cortical markers persists in the reduced double mutant neocortex, but the laminar patterns exhibited are less sharp than normal, consistent with deficient cytoarchitecture, probably due in part to reduced numbers of preplate and Reelin-positive Cajal-Retzius neurons. Subplate neurons also exhibit abnormal differentiation in double mutants. Cortical efferent axons fail to exit the double mutant cortex, and TCAs pass through the striatum and approach the cortex but do not enter it. This TCA pathfinding defect appears to be non-cell autonomous and supports the hypothesis that cortical efferents are required scaffolds to guide TCAs into cortex. In double mutants, some TCAs fail to turn into ventral telencephalon and take an aberrant ventral trajectory; this pathfinding defect correlates with an Emx2 expression domain in ventral telencephalon. The more severe phenotypes in Emx double mutants suggest that Emx1 and Emx2 cooperate to regulate multiple features of cortical development.


Subject(s)
Afferent Pathways/pathology , Cerebral Cortex/pathology , Homeodomain Proteins , Neurons/pathology , Olfactory Bulb/pathology , Thalamus/pathology , Afferent Pathways/growth & development , Animals , Axons/pathology , Cell Death , Cell Differentiation , Cerebral Cortex/growth & development , Efferent Pathways/growth & development , Efferent Pathways/pathology , Embryonic and Fetal Development , Gene Expression Regulation, Developmental , Hippocampus/growth & development , Hippocampus/pathology , Homeodomain Proteins/genetics , Immunohistochemistry , In Situ Hybridization , Mice , Mice, Mutant Strains , Mutation , Olfactory Bulb/growth & development , Phenotype , Reelin Protein , Thalamus/growth & development , Transcription Factors/genetics
18.
Development ; 129(24): 5621-34, 2002 Dec.
Article in English | MEDLINE | ID: mdl-12421703

ABSTRACT

The prevailing model to explain the formation of topographic projections in the nervous system stipulates that this process is governed by information located within the projecting and targeted structures. In mammals, different thalamic nuclei establish highly ordered projections with specific neocortical domains and the mechanisms controlling the initial topography of these projections remain to be characterized. To address this issue, we examined Ebf1(-/-) embryos in which a subset of thalamic axons does not reach the neocortex. We show that the projections that do form between thalamic nuclei and neocortical domains have a shifted topography, in the absence of regionalization defects in the thalamus or neocortex. This shift is first detected inside the basal ganglia, a structure on the path of thalamic axons, and which develops abnormally in Ebf1(-/-) embryos. A similar shift in the topography of thalamocortical axons inside the basal ganglia and neocortex was observed in Dlx1/2(-/-) embryos, which also have an abnormal basal ganglia development. Furthermore, Dlx1 and Dlx2 are not expressed in the dorsal thalamus or in cortical projections neurons. Thus, our study shows that: (1) different thalamic nuclei do not establish projections independently of each other; (2) a shift in thalamocortical topography can occur in the absence of major regionalization defects in the dorsal thalamus and neocortex; and (3) the basal ganglia may contain decision points for thalamic axons' pathfinding and topographic organization. These observations suggest that the topography of thalamocortical projections is not strictly determined by cues located within the neocortex and may be regulated by the relative positioning of thalamic axons inside the basal ganglia.


Subject(s)
DNA-Binding Proteins/genetics , Homeodomain Proteins/genetics , Thalamus/pathology , Trans-Activators/genetics , Animals , Axons/metabolism , DNA-Binding Proteins/physiology , Genotype , Heterozygote , Homeodomain Proteins/physiology , Homozygote , In Situ Hybridization , Mice , Mice, Inbred C57BL , Mice, Transgenic , Models, Biological , Mutation , Time Factors , Trans-Activators/physiology , Transcription Factors
19.
J Comp Neurol ; 447(1): 8-17, 2002 May 20.
Article in English | MEDLINE | ID: mdl-11967891

ABSTRACT

During development, cortical areas establish precise reciprocal projections with corresponding thalamic nuclei. Pioneer axons from the cortex and thalamus first meet in the intermediate zone of the subcortical telencephalon (subpallium). Their close interactions in the subpallium suggest that they may use each other for guidance. To test this hypothesis, the development of corticothalamic and thalamocortical connections was studied in mice with mutations of transcription factor genes expressed specifically in the cortex (Tbr1), the dorsal thalamus (Gbx2), or both (Pax6). In Tbr1 mutants, cortical pioneer axons entered the subpallium at the appropriate time, but most stopped growing without entering the diencephalon. Surprisingly, thalamic axons (which do not express Tbr1) deviated into the external capsule and amygdala regions, without entering the cortex. Conversely, in most Gbx2 mutants, thalamic axons were reduced in number and grew no farther than the subpallium. Cortical axons (which do not express Gbx2) grew into the subpallium but did not enter the diencephalon. In one Gbx2- /- case, sparse thalamocortical and corticothalamic projections both developed, but in no case did one projection reach its target and not the other. In Pax6 mutants, neither corticothalamic nor thalamocortical axons reached their targets. These results suggest that thalamocortical and corticothalamic projections may not form independently. After reaching the subpallium, each projection may require a molecularly intact reciprocal projection for further guidance. This type of mechanism ensures that thalamocortical and corticothalamic axons project reciprocally. However, the exact nature of the interaction between cortical and thalamic pioneer axons remains to be elucidated.


Subject(s)
Cerebral Cortex/embryology , DNA-Binding Proteins/deficiency , Growth Cones/metabolism , Homeodomain Proteins/metabolism , Mice, Knockout/embryology , Neural Pathways/embryology , Thalamus/embryology , Animals , Animals, Newborn , Carbocyanines , Cell Communication/genetics , Cell Differentiation/genetics , Cerebral Cortex/growth & development , Cerebral Cortex/metabolism , DNA-Binding Proteins/metabolism , Eye Proteins , Female , Fetus , Fluorescent Dyes , Growth Cones/ultrastructure , Homeodomain Proteins/genetics , Internal Capsule/embryology , Internal Capsule/growth & development , Internal Capsule/metabolism , Mice , Mice, Knockout/growth & development , Mice, Knockout/metabolism , Mutation/genetics , Nervous System Malformations/genetics , Nervous System Malformations/metabolism , Nervous System Malformations/pathology , Neural Pathways/growth & development , Neural Pathways/metabolism , PAX6 Transcription Factor , Paired Box Transcription Factors , Repressor Proteins , T-Box Domain Proteins , Thalamus/growth & development , Thalamus/metabolism
20.
Development ; 129(3): 761-73, 2002 Feb.
Article in English | MEDLINE | ID: mdl-11830575

ABSTRACT

We have investigated the mechanisms that control the guidance of corticofugal projections as they extend along different subdivisions of the forebrain. To this aim, we analyzed the development of cortical projections in mice that lack Nkx2-1, a homeobox gene whose expression is restricted to two domains within the forebrain: the basal telencephalon and the hypothalamus. Molecular respecification of the basal telencephalon and hypothalamus in Nkx2-1-deficient mice causes a severe defect in the guidance of layer 5 cortical projections and ascending fibers of the cerebral peduncle. These axon tracts take an abnormal path when coursing through both the basal telencephalon and hypothalamus. By contrast, loss of Nkx2-1 function does not impair guidance of corticothalamic or thalamocortical axons. In vitro experiments demonstrate that the basal telencephalon and the hypothalamus contain an activity that repels the growth of cortical axons, suggesting that loss of this activity is the cause of the defects observed in Nkx2-1 mutants. Furthermore, analysis of the expression of candidate molecules in the basal telencephalon and hypothalamus of Nkx2-1 mutants suggests that Slit2 contributes to this activity.


Subject(s)
Homeodomain Proteins/genetics , Hypothalamus/embryology , Neural Pathways/embryology , Telencephalon/embryology , Animals , Axons/physiology , Body Patterning , Cell Communication , Dopamine/metabolism , Hypothalamus/cytology , Intercellular Signaling Peptides and Proteins , Mice , Mice, Mutant Strains , Models, Neurological , Mutation , Nerve Tissue Proteins/metabolism , Prosencephalon/embryology , Pyramidal Tracts/cytology , Telencephalon/cytology
SELECTION OF CITATIONS
SEARCH DETAIL