Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 34
Filter
Add more filters










Publication year range
1.
Proc Natl Acad Sci U S A ; 120(50): e2311019120, 2023 Dec 12.
Article in English | MEDLINE | ID: mdl-38064506

ABSTRACT

The prevalence of obesity is increasing in older adults and contributes to age-related decline. Caloric restriction (CR) alleviates obesity phenotypes and delays the onset of age-related changes. However, how late in life organisms benefit from switching from a high-(H) to a low-calorie (L) diet is unclear. We transferred male flies from a H to a L (HL) diet or vice versa (LH) at different times during life. Both shifts immediately change fly rate of aging even when applied late in life. HL shift rapidly reduces fly mortality rate to briefly lower rate than in flies on a constant L diet, and extends lifespan. Transcriptomic analysis uncovers that flies aged on H diet have acquired increased stress response, which may have temporal advantage over flies aged on L diet and leads to rapid decrease in mortality rate after HL switch. Conversely, a LH shift increases mortality rate, which is temporarily higher than in flies aged on a H diet, and shortens lifespan. Unexpectedly, more abundant transcriptomic changes accompanied LH shift, including increase in ribosome biogenesis, stress response and growth. These changes reflect protection from sudden release of ROS, energy storage, and use of energy to growth, which all likely contribute to higher mortality rate. As the beneficial effects of CR on physiology and lifespan are conserved across many organisms, our study provides framework to study underlying mechanisms of CR interventions that counteract the detrimental effects of H diets and reduce rate of aging even when initiated later in life.


Subject(s)
Energy Intake , Longevity , Animals , Male , Longevity/physiology , Aging/physiology , Caloric Restriction , Drosophila melanogaster/physiology , Obesity
2.
Sci Adv ; 8(13): eabl9156, 2022 04.
Article in English | MEDLINE | ID: mdl-35363520

ABSTRACT

Recent studies using single-cell RNA-sequencing have revealed cellular heterogeneity in the developing mammalian cerebellum, yet the regulatory logic underlying this cellular diversity remains to be elucidated. Using integrated single-cell RNA and ATAC analyses, we resolved developmental trajectories of cerebellar progenitors and identified putative trans- and cis-elements that control cell state transition. We reverse engineered gene regulatory networks (GRNs) of each cerebellar cell type. Through in silico simulations and in vivo experiments, we validated the efficacy of GRN analyses and uncovered the molecular control of a posterior transitory zone (PTZ), a distinct progenitor zone residing immediately anterior to the morphologically defined rhombic lip (RL). We showed that perturbing cell fate specification in the PTZ and RL causes posterior cerebellar vermis hypoplasia, the most common cerebellar birth defect in humans. Our study provides a foundation for comprehensive studies of developmental programs of the mammalian cerebellum.


Subject(s)
Nervous System Malformations , Transcriptome , Animals , Cell Differentiation/genetics , Cerebellum/metabolism , Epigenesis, Genetic , Mammals/genetics , Mice , Nervous System Malformations/genetics , Nervous System Malformations/metabolism
3.
Front Genet ; 12: 681206, 2021.
Article in English | MEDLINE | ID: mdl-34512717

ABSTRACT

Single-cell transcriptional and epigenomics profiles have been applied in a variety of tissues and diseases for discovering new cell types, differentiation trajectories, and gene regulatory networks. Many methods such as Monocle 2/3, URD, and STREAM have been developed for tree-based trajectory building. Here, we propose a fast and flexible trajectory learning method, LISA2, for single-cell data analysis. This new method has two distinctive features: (1) LISA2 utilizes specified leaves and root to reduce the complexity for building the developmental trajectory, especially for some special cases such as rare cell populations and adjacent terminal cell states; and (2) LISA2 is applicable for both transcriptomics and epigenomics data. LISA2 visualizes complex trajectories using 3D Landmark ISOmetric feature MAPping (L-ISOMAP). We apply LISA2 to simulation and real datasets in cerebellum, diencephalon, and hematopoietic stem cells including both single-cell transcriptomics data and single-cell assay for transposase-accessible chromatin data. LISA2 is efficient in estimating single-cell trajectory and expression trends for different kinds of molecular state of cells.

4.
J Cell Physiol ; 234(11): 19933-19941, 2019 11.
Article in English | MEDLINE | ID: mdl-30972783

ABSTRACT

Existing methodologies to produce human neural stem cells and neurons from embryonic stem cells frequently involve multistep processes and the use of complex and expensive media components, cytokines or small molecules. Here, we report a simple technique to generate human neuroepithelial progenitors and neurons by periodic mechanical dissection and adherent-cell depletion on regular cell-culture grade plastic surfaces. This neural induction technique does not employ growth factors, small molecules or peptide inhibitors, apart from those present in serum-free supplements. Suggestive of their central nervous system origin, we found that neural progenitors formed by this technique expressed radial glia markers, and, when differentiated, expressed TUBB3, RBFOX3 (NeuN) and serotonin, but not markers for peripheral neurons. With these data, we postulate that incorporation of periodic mechanical stimuli and plastic surface-mediated cell selection could improve and streamline existing human neuron production protocols.


Subject(s)
Cell Culture Techniques/methods , Neural Stem Cells/cytology , Neurons/cytology , Animals , Cell Adhesion , Cell Differentiation , Cell Line , Human Embryonic Stem Cells/cytology , Humans , Mice
5.
Development ; 146(12)2019 04 01.
Article in English | MEDLINE | ID: mdl-30872278

ABSTRACT

The embryonic diencephalon forms integration centers and relay stations in the forebrain. Anecdotal expression studies suggest that the diencephalon contains multiple developmental compartments and subdivisions. Here, we utilized single cell RNA sequencing to profile transcriptomes of dissociated cells from the diencephalon of E12.5 mouse embryos. We identified the divergence of different progenitors, intermediate progenitors, and emerging neurons. By mapping the identified cell groups to their spatial origins, we characterized the molecular features of cell types and cell states arising from various diencephalic domains. Furthermore, we reconstructed the developmental trajectory of distinct cell lineages, and thereby identified the genetic cascades and gene regulatory networks underlying the progression of the cell cycle, neurogenesis and cellular diversification. The analysis provides new insights into the molecular mechanisms underlying the amplification of intermediate progenitor cells in the thalamus. The single cell-resolved trajectories not only confirm a close relationship between the rostral thalamus and prethalamus, but also uncover an unexpected close relationship between the caudal thalamus, epithalamus and rostral pretectum. Our data provide a useful resource for systematic studies of cell heterogeneity and differentiation kinetics within the diencephalon.


Subject(s)
Epithalamus/embryology , Gene Expression Regulation, Developmental , Neurons/cytology , Pretectal Region/embryology , Single-Cell Analysis/methods , Thalamus/embryology , Animals , Body Patterning , Cell Differentiation , Cell Lineage , Gene Expression Profiling , Gene Regulatory Networks , Homeodomain Proteins/genetics , Mice , Neurogenesis , Sequence Analysis, RNA/methods , Stem Cells , Tissue Array Analysis
6.
Sci Adv ; 4(10): eaat2681, 2018 10.
Article in English | MEDLINE | ID: mdl-30338292

ABSTRACT

While deregulation of mitochondrial metabolism and cytosolic glycolysis has been well recognized in tumor cells, the role of coordinated mitochondrial oxidation and cytosolic fermentation of pyruvate, a key metabolite derived from glucose, in physiological processes is not well understood. Here, we report that knockout of PTPMT1, a mitochondrial phosphoinositide phosphatase, completely blocked postnatal cerebellar development. Proliferation of granule cell progenitors, the most actively replicating cells in the developing cerebellum, was only moderately decreased, and proliferation of Purkinje cell progenitors did not seem to be affected in knockout mice. In contrast, generation of functional Bergmann glia from multipotent precursor cells (radial glia), which is essential for cerebellar corticogenesis, was totally disrupted. Moreover, despite a low turnover rate, neural stem cells were impaired in self-renewal in knockout mice. Mechanistically, loss of PTPMT1 decreased mitochondrial aerobic metabolism by limiting utilization of pyruvate, which resulted in bioenergetic stress in neural precursor/stem cells but not in progenitor or mature cells, leading to cell cycle arrest through activation of the AMPK-p19/p21 pathway. This study suggests that mitochondrial oxidation of the carbohydrate fuel is required for postnatal cerebellar development, and identifies a bioenergetic stress-induced cell cycle checkpoint in neural precursor/stem cells.


Subject(s)
Carbohydrate Metabolism/physiology , Cerebellum/growth & development , Cerebellum/metabolism , Mitochondria/metabolism , Neural Stem Cells/physiology , Animals , Animals, Newborn , Cell Cycle Checkpoints/genetics , Cerebellum/cytology , Female , Glycolysis , Male , Mice, Knockout , Neural Stem Cells/cytology , PTEN Phosphohydrolase/genetics , PTEN Phosphohydrolase/metabolism , Pyruvic Acid/metabolism
7.
Cerebellum ; 17(1): 42-48, 2018 02.
Article in English | MEDLINE | ID: mdl-29218544

ABSTRACT

Evolution of complex behaviors in higher vertebrates and primates require the development of sophisticated neuronal circuitry and the expansion of brain surface area to accommodate the vast number of neuronal and glial populations. To achieve these goals, the neocortex in primates and the cerebellum in amniotes have developed specialized types of basal progenitors to aid the folding of their cortices. In the cerebellum, Bergmann glia constitute such a basal progenitor population, having a distinctive morphology and playing a critical role in cerebellar corticogenesis. Here, we review recent studies on the induction of Bergmann glia and their crucial role in mediating folding of the cerebellar cortex. These studies uncover a key function of FGF-ERK-ETV signaling cascade in the transformation of Bergmann glia from radial glia in the ventricular zone. Remarkably, in the neocortex, the same signaling axis operates to facilitate the transformation of ventricular radial glia into basal radial glia, a Bergmann glia-like basal progenitor population, which have been implicated in the establishment of neocortical gyri. These new findings draw a striking similarity in the function and ontogeny of the two basal progenitor populations born in distinct brain compartments.


Subject(s)
Cerebellum/cytology , Cerebellum/physiology , Extracellular Signal-Regulated MAP Kinases/metabolism , Neuroglia/physiology , Signal Transduction/physiology , Animals , Humans , Protein Tyrosine Phosphatase, Non-Receptor Type 11/genetics , Protein Tyrosine Phosphatase, Non-Receptor Type 11/metabolism
8.
Development ; 143(20): 3661-3673, 2016 10 15.
Article in English | MEDLINE | ID: mdl-27578777

ABSTRACT

The embryonic tectum displays an anteroposterior gradient in development and produces the superior colliculus and inferior colliculus. Studies suggest that partition of the tectum is controlled by different strengths and durations of FGF signals originated from the so-called isthmic organizer at the mid/hindbrain junction; however, the underlying mechanism is unclear. We show that deleting Ptpn11, which links FGF with the ERK pathway, prevents inferior colliculus formation by depleting a previously uncharacterized stem cell zone. The stem-zone loss is attributed to shortening of S phase and acceleration of cell cycle exit and neurogenesis. Expression of a constitutively active Mek1 (Mek1DD), the known ERK activator, restores the tectal stem zone and the inferior colliculus without Ptpn11. By contrast, Mek1DD expression fails to rescue the tectal stem zone and the inferior colliculus in the absence of Fgf8 and the isthmic organizer, indicating that FGF and Mek1DD initiate qualitatively and/or quantitatively distinctive signaling. Together, our data show that the formation of the inferior colliculus relies on the provision of new cells from the tectal stem zone. Furthermore, distinctive ERK signaling mediates Fgf8 in the control of cell survival, tissue polarity and cytogenetic gradient during the development of the tectum.


Subject(s)
Extracellular Signal-Regulated MAP Kinases/metabolism , Fibroblast Growth Factors/metabolism , Inferior Colliculi/cytology , Inferior Colliculi/metabolism , Stem Cells/cytology , Stem Cells/metabolism , Animals , Apoptosis/genetics , Apoptosis/physiology , Axons/metabolism , Body Patterning/genetics , Body Patterning/physiology , Cell Cycle/genetics , Cell Cycle/physiology , Cell Death/genetics , Cell Death/physiology , Extracellular Signal-Regulated MAP Kinases/genetics , Fibroblast Growth Factor 8/genetics , Fibroblast Growth Factor 8/metabolism , Fibroblast Growth Factors/genetics , Gene Expression Regulation, Developmental/genetics , Gene Expression Regulation, Developmental/physiology , MAP Kinase Kinase 1/genetics , MAP Kinase Kinase 1/metabolism , Mesencephalon/cytology , Mesencephalon/metabolism , Mice, Knockout , Neurogenesis/genetics , Neurogenesis/physiology , Protein Tyrosine Phosphatase, Non-Receptor Type 11/genetics , Protein Tyrosine Phosphatase, Non-Receptor Type 11/metabolism
9.
Dev Biol ; 407(1): 26-39, 2015 Nov 01.
Article in English | MEDLINE | ID: mdl-26297811

ABSTRACT

The thalamus and habenula, two important nodes of the forebrain circuitry, are derived from a single developmental compartment, called prosomere 2, in the diencephalon. Habenular and thalamic neurons display distinct molecular identity, neurochemistry, and connectivity. Furthermore, their progenitors exhibit distinctive neurogenic patterns with a marked delay in the onset of neurogenesis in the thalamus. However, the progenitors in prosomere 2 express many common developmental regulators and the mechanism underlying the specification and differentiation of these two populations of neurons remains unknown. Gbx2, coding for a homeodomain transcription factor, is initially expressed in thalamic neuronal precursors that have just exited the cell cycle, and its expression is maintained in many mature thalamic neurons in adults. Deletion of Gbx2 severely disrupts histogenesis of the thalamus and abolishes thalamocortical projections in mice. Here, by using genome-wide transcriptional profiling, we show that Gbx2 promotes thalamic but inhibits habenular molecular characters. Remarkably, although Gbx2 is expressed in postmitotic neuronal precursors, deletion of Gbx2 changes gene expression and cell proliferation in dividing progenitors in the developing thalamus. These defects are partially rescued by the mosaic presence of wild-type cells, demonstrating a cell non-autonomous role of Gbx2 in regulating the development of thalamic progenitors. Our results suggest that Gbx2 is essential for the acquisition of the thalamic neuronal identity by repressing habenular identity through a feedback signaling from postmitotic neurons to progenitors.


Subject(s)
Habenula/embryology , Homeodomain Proteins/physiology , Neurons/cytology , Thalamus/embryology , Animals , Homeodomain Proteins/genetics , Mice , Transcription Factors/genetics , Transcriptome
10.
J Exp Med ; 212(8): 1323-36, 2015 Jul 27.
Article in English | MEDLINE | ID: mdl-26195727

ABSTRACT

Generation of T lymphocytes in the thymus is guided by signal transduction from the T cell receptor (TCR), but the underlying mechanism is incompletely understood. Here we have identified a Golgi-associated factor, TRAF3-interacting protein 3 (TRAF3IP3), as a crucial mediator of thymocyte development. TRAF3IP3 deficiency in mice attenuates the generation of mature thymocytes caused by impaired thymocyte-positive selection. TRAF3IP3 mediates TCR-stimulated activation of the mitogen-activated protein kinase (MAPK) extracellular signal-regulated kinase (ERK) and its upstream kinase mitogen/extracellular signal-regulated kinase (MEK). Interestingly, TRAF3IP3 exerts this signaling function through recruiting MEK to the Golgi and, thereby, facilitating the interaction of MEK with its activator BRAF. Transgenic expression of a constitutively active MEK rescues the T cell development block in Traf3ip3 knockout mice. These findings establish TRAF3IP3 as a novel regulator of T cell development and suggest a Golgi-specific ERK signaling mechanism that regulates thymocyte development.


Subject(s)
Carrier Proteins/metabolism , Cell Differentiation/immunology , Golgi Apparatus/metabolism , Membrane Proteins/metabolism , Microtubule-Associated Proteins/metabolism , Signal Transduction/immunology , T-Lymphocytes/immunology , Thymus Gland/cytology , Adoptive Transfer , Animals , Flow Cytometry , HEK293 Cells , Humans , Immunoblotting , Immunoprecipitation , Mice , Mice, Inbred C57BL , Mice, Transgenic , Thymus Gland/immunology
11.
Dev Biol ; 399(1): 177-187, 2015 Mar 01.
Article in English | MEDLINE | ID: mdl-25592224

ABSTRACT

Wnt1 and Wnt3a secreted from the dorsal neural tube were previously shown to regulate a gene expression program in the dorsal otic vesicle that is necessary for vestibular morphogenesis (Riccomagno et al., 2005. Genes Dev. 19, 1612-1623). Unexpectedly, Wnt1(-/-); Wnt3a(-/-) embryos also displayed a pronounced defect in the outgrowth of the ventrally derived cochlear duct. To determine how Wnt signaling in the dorsal otocyst contributes to cochlear development we performed a series of genetic fate mapping experiments using two independent Wnt responsive driver strains (TopCreER and Gbx2(CreER)) that when crossed to inducible responder lines (Rosa(lacZ) or Rosa(zsGreen)) permanently labeled dorsomedial otic progenitors and their derivatives. Tamoxifen time course experiments revealed that most vestibular structures showed some degree of labeling when recombination was induced between E7.75 and E12.5, consistent with continuous Wnt signaling activity in this tissue. Remarkably, a population of Wnt responsive cells in the dorsal otocyst was also found to contribute to the sensory epithelium of the cochlear duct, including auditory hair and support cells. Similar results were observed with both TopCreER and Gbx2(CreER) strains. The ventral displacement of Wnt responsive cells followed a spatiotemporal sequence that initiated in the anterior otic cup at, or immediately prior to, the 17-somite stage (E9) and then spread progressively to the posterior pole of the otic vesicle by the 25-somite stage (E9.5). These lineage-tracing experiments identify the earliest known origin of auditory sensory progenitors within a population of Wnt responsive cells in the dorsomedial otic cup.


Subject(s)
Cochlea/metabolism , Ear, Inner/metabolism , Epithelium/metabolism , Wnt Signaling Pathway/genetics , Animals , Cell Lineage/genetics , Cell Movement/genetics , Cell Proliferation/genetics , Cochlea/cytology , Cochlea/embryology , Ear, Inner/cytology , Ear, Inner/embryology , Embryo, Mammalian/drug effects , Embryo, Mammalian/embryology , Embryo, Mammalian/metabolism , Epithelium/embryology , Estrogen Antagonists/pharmacology , Female , Gene Expression Regulation, Developmental , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Immunohistochemistry , In Situ Hybridization , Male , Mice, Transgenic , Microscopy, Confocal , Morphogenesis/drug effects , Morphogenesis/genetics , Tamoxifen/pharmacology , Time Factors
12.
J Neurosci ; 34(3): 922-31, 2014 Jan 15.
Article in English | MEDLINE | ID: mdl-24431450

ABSTRACT

Folding of the cortex and the persistence of radial glia (RG)-like cells called Bergmann glia (BG) are hallmarks of the mammalian cerebellum. Similar to basal RG in the embryonic neocortex, BG maintain only basal processes and continuously express neural stem cell markers. Past studies had focused on the function of BG in granule cell migration and how granule cell progenitors (GCP) regulate cerebellar foliation. The molecular control of BG generation and its role in cerebellar foliation are less understood. Here, we have analyzed the function of the protein tyrosine phosphatase Shp2 in mice by deleting its gene Ptpn11 in the entire cerebellum or selectively in the GCP lineage. Deleting Ptpn11 in the entire cerebellum by En1-cre blocks transformation of RG into BG but preserves other major cerebellar cell types. In the absence of BG, inward invagination of GCP persists but is uncoupled from the folding of the Purkinje cell layer and the basement membrane, leading to disorganized lamination and an absence of cerebellar folia. In contrast, removing Ptpn11 in the GCP lineage by Atoh1-cre has no effect on cerebellar development, indicating that Shp2 is not cell autonomously required in GCP. Furthermore, we demonstrate that Ptpn11 interacts with Fgf8 and is essential for ERK activation in RG and nascent BG. Finally, expressing constitutively active MEK1 rescues BG formation and cerebellar foliation in Shp2-deficient cerebella. Our results demonstrate an essential role of Shp2 in BG specification via fibroblast growth factor/extracellular signal-regulated protein kinase signaling, and reveal a crucial function of BG in organizing cerebellar foliation.


Subject(s)
Cerebellum/cytology , Cerebellum/physiology , MAP Kinase Signaling System/physiology , Neuroglia/physiology , Protein Tyrosine Phosphatase, Non-Receptor Type 11/physiology , Animals , Ependymoglial Cells/physiology , Female , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Protein Tyrosine Phosphatase, Non-Receptor Type 11/deficiency
13.
Water Sci Technol ; 68(11): 2382-90, 2013.
Article in English | MEDLINE | ID: mdl-24334886

ABSTRACT

Evaluations of benefits of implementing low impact development (LID) stormwater management techniques can extend up to a watershed scale. This presents a challenge for representing them in watershed models, since they are typically orders of magnitude smaller in size. This paper presents an approach that is focused on trying to evaluate the benefits of implementing LIDs on a lot level. The methodology uses the concept of urban hydrological response Unit and results in developing and applying performance curves that are a function of lot properties to estimate the potential benefit of large-scale LID implementation. Lot properties are determined using a municipal geographic information system database and processed to determine groups of lots with similar properties. A representative lot from each group is modeled over a typical rainfall year using USEPA Stormwater Management Model to develop performance functions that relate the lot properties and the change in annual runoff volume and corresponding phosphorus loading with different LIDs implemented. The results of applying performance functions on all urban areas provide the potential locations, benefit and cost of implementation of all LID techniques, guiding future decisions for LID implementation by watershed area municipalities.


Subject(s)
Drainage, Sanitary , Models, Theoretical , Cities
14.
Dev Biol ; 379(2): 208-20, 2013 Jul 15.
Article in English | MEDLINE | ID: mdl-23643939

ABSTRACT

Sensory and endoneurocrine tissues as diverse as the lens, the olfactory epithelium, the inner ear, the cranial sensory ganglia, and the anterior pituitary arise from a common pool of progenitors in the preplacodal ectoderm (PPE). Around late gastrulation, the PPE forms at the border surrounding the anterior neural plate, and expresses a unique set of evolutionarily conserved transcription regulators including Six1, Eya 1 and Eya2. Here, we describe the first report to generate and characterize the SIX1(+) PPE cells from human embryonic stem (ES) cells by adherent differentiation. Before forming PPE cells, differentiating cultures first expressed the non-neural ectoderm specific transcriptional factors TFAP2A, GATA2, GATA3, DLX3, and DLX5, which are crucial in establishing the PPE competence. We demonstrated that bone morphogenetic protein (BMP) activity plays a transient but essential role in inducing expression of these PPE competence factors and eventually the PPE cells. Interestingly, we found that attenuating BMP signaling after establishing the competence state induces anterior placode precursors. By manipulating BMP and hedgehog signaling pathways, we further differentiate these precursors into restricted lineages including the lens placode and the oral ectoderm (pituitary precursor) cells. Finally, we also show that sensory neurons can be generated from human PPE cells, demonstrating the multipotency of the human ES-derived PPE cells.


Subject(s)
Bone Morphogenetic Protein 4/metabolism , Cell Differentiation/physiology , Ectoderm/embryology , Embryonic Stem Cells/physiology , Signal Transduction/physiology , Amides , Analysis of Variance , Animals , Bone Morphogenetic Protein 4/pharmacology , Carrier Proteins , Cattle , Cell Differentiation/drug effects , Cornea/cytology , DNA Primers/genetics , Ectoderm/cytology , Embryonic Stem Cells/cytology , Homeodomain Proteins/metabolism , Humans , Immunohistochemistry , Pyrazoles , Pyridines , Pyrimidines , Reverse Transcriptase Polymerase Chain Reaction , Sensory Receptor Cells/cytology , Signal Transduction/genetics , Tretinoin
15.
Development ; 139(24): 4633-43, 2012 Dec.
Article in English | MEDLINE | ID: mdl-23136391

ABSTRACT

Combinatorial expression of transcription factors forms transcriptional codes to confer neuronal identities and connectivity. However, how these intrinsic factors orchestrate the spatiotemporal expression of guidance molecules to dictate the responsiveness of axons to guidance cues is less understood. Thalamocortical axons (TCAs) represent the major input to the neocortex and modulate cognitive functions, consciousness and alertness. TCAs travel a long distance and make multiple target choices en route to the cortex. The homeodomain transcription factor Gbx2 is essential for TCA development, as loss of Gbx2 abolishes TCAs in mice. Using a novel TCA-specific reporter, we have discovered that thalamic axons are mostly misrouted to the ventral midbrain and dorsal midline of the diencephalon in Gbx2-deficient mice. Furthermore, conditionally deleting Gbx2 at different embryonic stages has revealed a sustained role of Gbx2 in regulating TCA navigation and targeting. Using explant culture and mosaic analyses, we demonstrate that Gbx2 controls the intrinsic responsiveness of TCAs to guidance cues. The guidance defects of Gbx2-deficient TCAs are associated with abnormal expression of guidance receptors Robo1 and Robo2. Finally, we demonstrate that Gbx2 controls Robo expression by regulating LIM-domain transcription factors through three different mechanisms: Gbx2 and Lhx2 compete for binding to the Lmo3 promoter and exert opposing effects on its transcription; repressing Lmo3 by Gbx2 is essential for Lhx2 activity to induce Robo2; and Gbx2 represses Lhx9 transcription, which in turn induces Robo1. Our findings illustrate the transcriptional control of differential expression of Robo1 and Robo2, which may play an important role in establishing the topography of TCAs.


Subject(s)
Axons/physiology , Homeodomain Proteins/physiology , LIM-Homeodomain Proteins/genetics , Nerve Tissue Proteins/genetics , Thalamus/embryology , Animals , Axons/metabolism , Cells, Cultured , Cerebral Cortex/embryology , Cerebral Cortex/metabolism , Cerebral Cortex/physiology , Embryo, Mammalian , Female , Gene Expression Regulation, Developmental , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , LIM-Homeodomain Proteins/metabolism , Mice , Mice, Transgenic , Nerve Tissue Proteins/metabolism , Neurogenesis/genetics , Neurogenesis/physiology , Pregnancy , Receptors, Immunologic/genetics , Receptors, Immunologic/metabolism , Thalamus/metabolism , Thalamus/physiology , Transcription Factors/genetics , Transcription Factors/metabolism , Roundabout Proteins
16.
PLoS One ; 7(10): e47111, 2012.
Article in English | MEDLINE | ID: mdl-23056596

ABSTRACT

Unlike the laminar arrangement of neurons in the neocortex, thalamic neurons aggregate to form about dozens of nuclei, many of which make topographic connections with specific areas in the neocortex. The molecular mechanisms underlying the formation of thalamic nuclei remain largely unknown. Homeodomain transcription factor Gbx2 is specifically expressed in the developing thalamus. Deleting Gbx2 leads to severe disruption of the histogenesis of the thalamus in mice, demonstrating an essential role of Gbx2 in this brain structure. Using inducible genetic fate mapping, we have previously shown that the neuronal precursors for different sets of thalamic nuclei have distinctive onset and duration of Gbx2 expression, suggesting that the dynamic expression of Gbx2 plays an important role in the specification and differentiation of thalamic nuclei. Here, we showed that the Gbx2 lineage exclusively gives rise to neurons but not glia in the thalamus. We performed conditional deletion to examine the temporal requirements of Gbx2 in the developing thalamus in mice. Corresponding to the dynamic and differential expression of Gbx2 in various thalamic nucleus groups, deleting Gbx2 at different embryonic stages disrupts formation of distinct sets of thalamic nuclei. Interestingly, different thalamic nuclei have remarkably different requirements of Gbx2 for the survival of thalamic neurons. Furthermore, although Gbx2 expression persists in many thalamic nuclei until adulthood, only the initial expression of Gbx2 following neurogenesis is crucial for the differentiation of thalamic nuclei. Our results indicate that the dynamic expression of Gbx2 may act as an important determinant in coupling with other developmental programs to generate distinct thalamic nuclei.


Subject(s)
Homeodomain Proteins/metabolism , Neurons/cytology , Neurons/metabolism , Thalamic Nuclei/cytology , Thalamic Nuclei/metabolism , Animals , Cell Survival/genetics , Cell Survival/physiology , Cerebral Cortex/cytology , Cerebral Cortex/metabolism , Female , Fluorescent Antibody Technique , Gene Expression Regulation, Developmental/genetics , Gene Expression Regulation, Developmental/physiology , Homeodomain Proteins/genetics , In Situ Hybridization , Male , Mice
17.
Front Neurosci ; 6: 66, 2012.
Article in English | MEDLINE | ID: mdl-22593732

ABSTRACT

The diencephalon gives rise to structures that play an important role in connecting the anterior forebrain with the rest of the central nervous system. The thalamus is the major diencephalic derivative that functions as a relay station between the cortex and other lower order sensory systems. Almost two decades ago, neuromeric/prosomeric models were proposed describing the subdivision and potential segmentation of the diencephalon. Unlike the laminar structure of the cortex, the diencephalon is progressively divided into distinct functional compartments consisting principally of thalamus, epithalamus, pretectum, and hypothalamus. Neurons generated within these domains further aggregate to form clusters called nuclei, which form specific structural and functional units. We review the recent advances in understanding the genetic mechanisms that are involved in the patterning and compartment formation of the diencephalon.

18.
J Neurosci ; 32(13): 4372-85, 2012 Mar 28.
Article in English | MEDLINE | ID: mdl-22457488

ABSTRACT

The assembly of neural circuits is dependent upon the generation of specific neuronal subtypes, each subtype displaying unique properties that direct the formation of selective connections with appropriate target cells. Actions of transcription factors in neural progenitors and postmitotic cells are key regulators in this process. LIM-homeodomain transcription factors control crucial aspects of neuronal differentiation, including subtype identity and axon guidance. Nonetheless, their regulation during development is poorly understood and the identity of the downstream molecular effectors of their activity remains largely unknown. Here, we demonstrate that the Lhx2 transcription factor is dynamically regulated in distinct pools of thalamic neurons during the development of thalamocortical connectivity in mice. Indeed, overexpression of Lhx2 provokes defective thalamocortical axon guidance in vivo, while specific conditional deletion of Lhx2 in the thalamus produces topographic defects that alter projections from the medial geniculate nucleus and from the caudal ventrobasal nucleus in particular. Moreover, we demonstrate that Lhx2 influences axon guidance and the topographical sorting of axons by regulating the expression of Robo1 and Robo2 guidance receptors, which are essential for these axons to establish correct connections in the cerebral cortex. Finally, augmenting Robo1 function restores normal axon guidance in Lhx2-overexpressing neurons. By regulating axon guidance receptors, such as Robo1 and Robo2, Lhx2 differentially regulates the axon guidance program of distinct populations of thalamic neurons, thus enabling the establishment of specific neural connections.


Subject(s)
Axons/physiology , Cerebral Cortex/physiology , LIM-Homeodomain Proteins/physiology , Nerve Tissue Proteins/biosynthesis , Neurogenesis/physiology , Receptors, Immunologic/biosynthesis , Thalamus/physiology , Transcription Factors/physiology , Animals , Cerebral Cortex/growth & development , Cerebral Cortex/metabolism , Embryo, Mammalian , Gene Deletion , Gene Expression Regulation, Developmental , LIM-Homeodomain Proteins/metabolism , Mice , Mice, Inbred ICR , Mice, Knockout , Mice, Transgenic , Neural Pathways/growth & development , Neural Pathways/metabolism , Neural Pathways/physiology , Signal Transduction/physiology , Thalamus/growth & development , Thalamus/metabolism , Transcription Factors/metabolism , Roundabout Proteins
19.
J Cell Physiol ; 226(7): 1722-6, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21506104

ABSTRACT

Soluble growth factors play an important role in the coordination and integration of cell proliferation, differentiation, fate determination, and morphogenesis during development of multicellular organisms. Fibroblast growth factors (FGFs) are a large family of polypeptide growth factors that are present in organisms ranging from nematodes to humans. RNA alternative splicing of FGFs and their receptors further enhances the complexity of this ligand-receptor system. The mouse Fgf8 gene produces eight splice variants, which encode isoform proteins with different N-termini and distinct receptor-binding affinity and biological activity. In this article, we review the roles of Fgf8 in vertebrate development and summarize the recent findings on the in vivo function of different Fgf8 splice variants. We propose that multiple Fgf8 isoform proteins act in concert to regulate the overall function of Fgf8 and account for the diverse and essential role of Fgf8 during vertebrate development.


Subject(s)
Brain/metabolism , Fibroblast Growth Factor 8/metabolism , Signal Transduction , Animals , Brain/embryology , Brain/growth & development , Cell Differentiation , Cell Proliferation , Fibroblast Growth Factor 8/genetics , Fibroblast Growth Factors/metabolism , Humans , Mice , Morphogenesis , Protein Binding , Protein Isoforms , Receptors, Fibroblast Growth Factor/metabolism , Zebrafish Proteins/metabolism
20.
Development ; 138(4): 725-34, 2011 Feb.
Article in English | MEDLINE | ID: mdl-21266408

ABSTRACT

In vertebrates, the common expression border of two homeobox genes, Otx2 and Gbx2, demarcates the prospective midbrain-hindbrain border (MHB) in the neural plate at the end of gastrulation. The presence of a compartment boundary at the MHB has been demonstrated, but the mechanism and timing of its formation remain unclear. We show by genetic inducible fate mapping using a Gbx2(CreER) knock-in mouse line that descendants of Gbx2(+) cells as early as embryonic day (E) 7.5 do not cross the MHB. Without Gbx2, hindbrain-born cells abnormally populate the entire midbrain, demonstrating that Gbx2 is essential for specifying hindbrain fate. Gbx2(+) and Otx2(+) cells segregate from each other, suggesting that mutually exclusive expression of Otx2 and Gbx2 in midbrain and hindbrain progenitors is responsible for cell sorting in establishing the MHB. The MHB organizer gene Fgf8, which is expressed as a sharp transverse band immediately posterior to the lineage boundary at the MHB, is crucial in maintaining the lineage-restricted boundary after E7.5. Partial deletion of Fgf8 disrupts MHB lineage separation. Activation of FGF pathways has a cell-autonomous effect on cell sorting in midbrain progenitors. Therefore, Fgf8 from the MHB may signal the nearby mesencephalic cells to impart distinct cell surface characteristics or induce local cell-cell signaling, which consequently prevents cell movements across the MHB. Our findings reveal the distinct function of Gbx2 and Fgf8 in a stepwise process in the development of the compartment boundary at the MHB and that Fgf8, in addition to its organizer function, plays a crucial role in maintaining the lineage boundary at the MHB by restricting cell movement.


Subject(s)
Fibroblast Growth Factor 8/metabolism , Homeodomain Proteins/metabolism , Mesencephalon/embryology , Mesencephalon/metabolism , Rhombencephalon/embryology , Rhombencephalon/metabolism , Animals , Cell Lineage , Fibroblast Growth Factor 8/genetics , Gene Deletion , Gene Expression Regulation, Developmental , Homeodomain Proteins/genetics , Mesencephalon/cytology , Mice , Mice, Transgenic , Rhombencephalon/cytology , Signal Transduction
SELECTION OF CITATIONS
SEARCH DETAIL