Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 48
Filter
1.
J Neurosci ; 39(11): 2025-2040, 2019 03 13.
Article in English | MEDLINE | ID: mdl-30647151

ABSTRACT

Microglia play important roles in shaping the developing CNS, and at early stages they have been proposed to regulate progenitor proliferation, differentiation, and neuronal survival. However, these studies reveal contradictory outcomes, highlighting the complexity of these cell-cell interactions. Here, we investigate microglia function during embryonic mouse retina development, where only microglia, progenitors, and neurons are present. In both sexes, we determine that microglia primarily interact with retinal neurons and find that depletion of microglia via conditional KO of the Csf1 receptor results in increased density of retinal ganglion cells (RGCs). Pharmacological inhibition of microglia also results in an increase in RGCs, with no effect on retinal progenitor proliferation, RGC genesis, or apoptosis. We show that microglia in the embryonic retina are enriched for phagocytic markers and observe engulfment of nonapoptotic Brn3-labeled RGCs. We investigate the molecular pathways that can mediate cell engulfment by microglia and find selective downregulation of complement pathway components with microglia inhibition, and further show that C1q protein marks a subset of RGCs in the embryonic retina. KO of complement receptor 3 (CR3; Itgam), which is only expressed by microglia, results in increased RGC density, similar to what we observed after depletion or inhibition of microglia. Thus, our data suggest that microglia regulate neuron elimination in the embryonic mouse retina by complement-mediated phagocytosis of non-apoptotic newborn RGCs.SIGNIFICANCE STATEMENT Microglia are emerging as active and important participants in regulating neuron number in development, during adult neurogenesis, and following stem cell therapies. However, their role in these contexts and the mechanisms involved are not fully defined. Using a well-characterized in vivo system, we provide evidence that microglia regulate neuronal elimination by complement-mediated engulfment of nonapoptotic neurons. This work provides a significant advancement of the field by defining in vivo molecular mechanisms for microglia-mediated cell elimination. Our data add to a growing body of evidence that microglia are essential for proper nervous system development. In addition, we elucidate microglia function in the developing retina, which may shed light on microglia involvement in the context of retinal injury and disease.


Subject(s)
Complement System Proteins/physiology , Microglia/physiology , Phagocytosis/physiology , Retina/growth & development , Retinal Ganglion Cells/physiology , Animals , Cell Count , Female , Macrophage Colony-Stimulating Factor/genetics , Male , Mice, Knockout
2.
Dev Dyn ; 248(1): 98-117, 2019 01.
Article in English | MEDLINE | ID: mdl-30444278

ABSTRACT

Microglia are engineers of the central nervous system (CNS) both in health and disease. In addition to the canonical immunological roles of clearing damaging entities and limiting the spread of toxicity and death, microglia remodel the CNS throughout life. While they have been extensively studied in disease and injury, due to their highly variable functions, their precise role in these contexts still remains uncertain. Over the past decade, we have greatly expanded our understanding of microglial function, including their essential homeostatic roles during development. Here, we review these developmental roles, identify parallels in disease, and speculate whether developmental mechanisms re-emerge in disease and injury. Developmental Dynamics 248:98-117, 2019. © 2018 Wiley Periodicals, Inc.


Subject(s)
Central Nervous System Diseases/pathology , Central Nervous System/growth & development , Homeostasis , Microglia/physiology , Animals , Central Nervous System Diseases/etiology , Humans
3.
Dev Biol ; 437(1): 27-40, 2018 05 01.
Article in English | MEDLINE | ID: mdl-29518376

ABSTRACT

Neural basic helix-loop helix (bHLH) transcription factors promote progenitor cell differentiation by activation of downstream target genes that coordinate neuronal differentiation. Here we characterize a neural bHLH target gene in Xenopus laevis, vexin (vxn; previously sbt1), that is homologous to human c8orf46 and is conserved across vertebrate species. C8orf46 has been implicated in cancer progression, but its function is unknown. Vxn is transiently expressed in differentiating progenitors in the developing central nervous system (CNS), and is required for neurogenesis in the neural plate and retina. Its function is conserved, since overexpression of either Xenopus or mouse vxn expands primary neurogenesis and promotes early retinal cell differentiation in cooperation with neural bHLH factors. Vxn protein is localized to the cell membrane and the nucleus, but functions in the nucleus to promote neural differentiation. Vxn inhibits cell proliferation, and works with the cyclin-dependent kinase inhibitor p27Xic1 (cdkn1b) to enhance neurogenesis and increase levels of the proneural protein Neurog2. We propose that vxn provides a key link between neural bHLH activity and execution of the neurogenic program.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/genetics , Neurogenesis/genetics , Xenopus Proteins/genetics , Animals , Blotting, Western , Cell Differentiation/genetics , Cyclin-Dependent Kinase Inhibitor p27/metabolism , Gene Expression Regulation, Developmental , In Situ Hybridization , Mice , Nerve Tissue Proteins/metabolism , Neural Plate/embryology , Neural Plate/metabolism , Retina/embryology , Retina/metabolism , Xenopus laevis
4.
Mol Ther ; 26(10): 2379-2396, 2018 10 03.
Article in English | MEDLINE | ID: mdl-30217731

ABSTRACT

Dysregulation of the complement system is implicated in neurodegeneration, including human and animal glaucoma. Optic nerve and retinal damage in glaucoma is preceded by local complement upregulation and activation, but whether targeting this early innate immune response could have therapeutic benefit remains undefined. Because complement signals through three pathways that intersect at complement C3 activation, here we targeted this step to restore complement balance in the glaucomatous retina and to determine its contribution to degeneration onset and/or progression. To achieve this, we combined adeno-associated virus retinal gene therapy with the targeted C3 inhibitor CR2-Crry. We show that intravitreal injection of AAV2.CR2-Crry produced sustained Crry overexpression in the retina and reduced deposition of the activation product complement C3d on retinal ganglion cells and the inner retina of DBA/2J mice. This resulted in neuroprotection of retinal ganglion cell axons and somata despite continued intraocular pressure elevation, suggesting a direct restriction of neurodegeneration onset and progression and significant delay to terminal disease stages. Our study uncovers a damaging effect of complement C3 or downstream complement activation in glaucoma, and it establishes AAV2.CR2-Crry as a viable therapeutic strategy to target pathogenic C3-mediated complement activation in the glaucomatous retina.


Subject(s)
Complement C3/genetics , Glaucoma/therapy , Nerve Degeneration/therapy , Recombinant Fusion Proteins/genetics , Animals , Complement C3/antagonists & inhibitors , Dependovirus/genetics , Disease Models, Animal , Disease Progression , Gene Expression Regulation/drug effects , Genetic Therapy , Glaucoma/genetics , Glaucoma/pathology , Humans , Intraocular Pressure/drug effects , Intravitreal Injections , Mice , Nerve Degeneration/genetics , Nerve Degeneration/pathology , Recombinant Fusion Proteins/administration & dosage , Retina/drug effects , Retina/pathology , Retinal Ganglion Cells/drug effects , Retinal Ganglion Cells/pathology
5.
Dev Biol ; 403(2): 128-38, 2015 Jul 15.
Article in English | MEDLINE | ID: mdl-25989023

ABSTRACT

Epigenetic regulation, including histone modification, is a critical component of gene regulation, although precisely how this contributes to the development of complex tissues such as the neural retina is still being explored. We show that during retinal development in mouse, there are dynamic patterns of expression of the polycomb repressive complex 2 (PRC2) catalytic subunit EZH2 in retinal progenitors and some differentiated cells, as well as dynamic changes in the histone modification H3K27me3. Using conditional knockout of Ezh2 using either Pax6-αCre or Six3-Cre, we find selective reduction in postnatal retinal progenitor proliferation, disruption of retinal lamination, and enhanced differentiation of several late born cell types in the early postnatal retina, including photoreceptors and Müller glia, which are ultimately increased in number and become reactive. RNA-seq identifies many non-retinal genes upregulated with loss of Ezh2, including multiple Hox genes and the cell cycle regulator Cdkn2a, which are established targets of EZH2-mediated repression. ChIP analysis confirms loss of the H3K27me3 modification at these loci. Similar gene upregulation is observed in retinal explants treated with an EZH2 chemical inhibitor. There is considerable overlap with EZH2-regulated genes reported in non-neural tissues, suggesting that EZH2 can regulate similar genes in multiple lineages. Our findings reveal a conserved role for EZH2 in constraining the expression of potent developmental regulators to maintain lineage integrity and retinal progenitor proliferation, as well as regulating the timing of late differentiation.


Subject(s)
Cell Differentiation , Polycomb Repressive Complex 2/metabolism , Retina/cytology , Retina/metabolism , Animals , Cell Proliferation , Chromatin Assembly and Disassembly , Enhancer of Zeste Homolog 2 Protein , Gene Expression Regulation , Mice , Stem Cells/cytology , Stem Cells/metabolism , Transcription, Genetic
6.
Development ; 140(14): 2867-78, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23739135

ABSTRACT

The histone methyltransferase complex PRC2 controls key steps in developmental transitions and cell fate choices; however, its roles in vertebrate eye development remain unknown. Here, we report that in Xenopus, PRC2 regulates the progression of retinal progenitors from proliferation to differentiation. We show that the PRC2 core components are enriched in retinal progenitors and downregulated in differentiated cells. Knockdown of the PRC2 core component Ezh2 leads to reduced retinal progenitor proliferation, in part due to upregulation of the Cdk inhibitor p15(Ink4b). In addition, although PRC2 knockdown does not alter eye patterning, retinal progenitor gene expression or expression of the neural competence factor Sox2, it does cause suppression of proneural bHLH gene expression, indicating that PRC2 is crucial for the initiation of neural differentiation in the retina. Consistent with this, knocking down or blocking PRC2 function constrains the generation of most retinal neural cell types and promotes a Müller glial cell fate decision. We also show that Wnt/ß-catenin signaling acting through the receptor Frizzled 5, but independent of Sox2, regulates expression of key PRC2 subunits in the developing retina. This is consistent with a role for this pathway in coordinating proliferation and the transition to neurogenesis in the Xenopus retina. Our data establish PRC2 as a regulator of proliferation and differentiation during eye development.


Subject(s)
Polycomb Repressive Complex 2/metabolism , Retina/embryology , Wnt Signaling Pathway , Xenopus Proteins/metabolism , Xenopus laevis/embryology , Animals , Cell Differentiation , Cell Proliferation , Enhancer of Zeste Homolog 2 Protein , Frizzled Receptors/metabolism , Gene Knockdown Techniques , Histones/metabolism , Methylation , Polycomb Repressive Complex 2/genetics , Repressor Proteins/metabolism , Retina/cytology , Retina/metabolism , Xenopus Proteins/genetics
7.
Exp Eye Res ; 150: 34-43, 2016 09.
Article in English | MEDLINE | ID: mdl-26851485

ABSTRACT

Within the white matter, axonal loss by neurodegeneration is coupled to glial cell changes in gene expression, structure and function commonly termed gliosis. Recently, we described the highly variable expansion of gliosis alebosco@neuro.utah.edu in degenerative optic nerves from the DBA/2J mouse model of chronic, age-related glaucoma. Here, to estimate and compare the levels of axonal loss with the expansion of glial coverage and axonal degeneration in DBA/2J nerves, we combined semiautomatic axon counts with threshold-based segmentation of total glial/scar areas and degenerative axonal profiles in plastic cross-sections. In nerves ranging from mild to severe degeneration, we found that the progression of axonal dropout is coupled to an increase of gliotic area. We detected a strong correlation between axon loss and the aggregate coverage by glial cells and scar, whereas axon loss did not correlate with the small fraction of degenerating profiles. Nerves with low to medium levels of axon loss displayed moderate glial reactivity, consisting of hypertrophic astrocytes, activated microglia and normal distribution of oligodendrocytes, with minimal reorganization of the tissue architecture. In contrast, nerves with extensive axonal loss showed prevalent rearrangement of the nerve, with loss of axon fascicle territories and enlarged or almost continuous gliotic and scar domains, containing reactive astrocytes, oligodendrocytes and activated microglia. These findings support the value of optic nerve gliotic expansion as a quantitative estimate of optic neuropathy that correlates with axon loss, applicable to grade the severity of optic nerve damage in mouse chronic glaucoma.


Subject(s)
Glaucoma/pathology , Gliosis/complications , Neuroglia/pathology , Optic Nerve Diseases/pathology , Optic Nerve/pathology , Retinal Ganglion Cells/pathology , Animals , Astrocytes/pathology , Axons/pathology , Chronic Disease , Disease Models, Animal , Female , Glaucoma/complications , Gliosis/pathology , Male , Mice , Mice, Inbred DBA , Microscopy, Confocal , Optic Nerve Diseases/etiology , Photomicrography
8.
Dev Biol ; 367(2): 91-9, 2012 Jul 15.
Article in English | MEDLINE | ID: mdl-22565092

ABSTRACT

During organogenesis, tissues expand in size and eventually acquire consistent ratios of cells with dazzling diversity in morphology and function. During this process progenitor cells exit the cell cycle and execute differentiation programs through extensive genetic reprogramming that involves the silencing of proliferation genes and the activation of differentiation genes in a step-wise temporal manner. Recent years have witnessed expansion in our understanding of the epigenetic mechanisms that contribute to cellular differentiation and maturation during organ development, as this is a crucial step toward advancing regenerative therapy research for many intractable disorders. Among such epigenetic programs, the developmental roles of the polycomb repressive complex 2 (PRC2), a chromatin remodeling complex that mediates silencing of gene expression, have been under intensive examination. This review summarizes recent findings of how PRC2 functions to regulate the transition from proliferation to differentiation during organogenesis and discusses some aspects of the remaining questions associated with its regulation and mechanisms of action.


Subject(s)
Organogenesis/physiology , Repressor Proteins/physiology , Animals , Cell Differentiation , Cell Lineage , Chromatin Assembly and Disassembly , Epigenesis, Genetic , Gene Expression Regulation, Developmental , Humans , Muscle Development , Neurogenesis , Organogenesis/genetics , Polycomb-Group Proteins , Protein Processing, Post-Translational , Repressor Proteins/genetics
9.
Cell Rep ; 42(3): 112237, 2023 03 28.
Article in English | MEDLINE | ID: mdl-36924502

ABSTRACT

Transitions in competence underlie the ability of CNS progenitors to generate a diversity of neurons and glia. Retinal progenitor cells in mouse generate early-born cell types embryonically and late-born cell types largely postnatally. We find that the transition from early to late progenitor competence is regulated by Jarid2. Loss of Jarid2 results in extended production of early cell types and extended expression of early progenitor genes. Jarid2 can regulate histone modifications, and we find reduction of repressive mark H3K27me3 on a subset of early progenitor genes with loss of Jarid2, most notably Foxp1. We show that Foxp1 regulates the competence to generate early-born retinal cell types, promotes early and represses late progenitor gene expression, and is required for extending early retinal cell production after loss of Jarid2. We conclude that Jarid2 facilitates progression of retinal progenitor temporal identity by repressing Foxp1, which is a primary regulator of early temporal patterning.


Subject(s)
Polycomb Repressive Complex 2 , Retina , Mice , Animals , Cell Differentiation , Polycomb Repressive Complex 2/genetics , Polycomb Repressive Complex 2/metabolism , Retina/metabolism , Stem Cells/metabolism , Repressor Proteins/genetics , Repressor Proteins/metabolism , Forkhead Transcription Factors/genetics , Forkhead Transcription Factors/metabolism
10.
Dev Biol ; 358(1): 240-50, 2011 Oct 01.
Article in English | MEDLINE | ID: mdl-21839736

ABSTRACT

EBF proteins have diverse functions in the development of multiple lineages, including neurons, B cells and adipocytes. During Drosophila muscle development EBF proteins are expressed in muscle progenitors and are required for muscle cell differentiation, but there is no known function of EBF proteins in vertebrate muscle development. In this study, we examine the expression of ebf genes in Xenopus muscle tissue and show that EBF activity is necessary for aspects of Xenopus skeletal muscle development, including somite organization, migration of hypaxial muscle anlagen toward the ventral abdomen, and development of jaw muscle. From a microarray screen, we have identified multiple candidate targets of EBF activity with known roles in muscle development. The candidate targets we have verified are MYOD, MYF5, M-Cadherin and SEB-4. In vivo overexpression of the ebf2 and ebf3 genes leads to ectopic expression of these candidate targets, and knockdown of EBF activity causes downregulation of the endogenous expression of the candidate targets. Furthermore, we found that MYOD and MYF5 are likely to be direct targets. Finally we show that MYOD can upregulate the expression of ebf genes, indicating the presence of a positive feedback loop between EBF and MYOD that we find to be important for maintenance of MYOD expression in Xenopus. These results suggest that EBF activity is important for both stabilizing commitment and driving aspects of differentiation in Xenopus muscle cells.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/metabolism , Cell Differentiation/physiology , Gene Expression Regulation, Developmental/physiology , Muscle Development/physiology , Muscle, Skeletal/embryology , Xenopus Proteins/metabolism , Xenopus/embryology , Animals , Cadherins/metabolism , Feedback, Physiological/physiology , In Situ Hybridization , Microarray Analysis , MyoD Protein/metabolism , Myogenic Regulatory Factor 5/metabolism , RNA-Binding Proteins/metabolism , Reverse Transcriptase Polymerase Chain Reaction
11.
Development ; 136(19): 3289-99, 2009 Oct.
Article in English | MEDLINE | ID: mdl-19736324

ABSTRACT

Progenitor cells in the central nervous system must leave the cell cycle to become neurons and glia, but the signals that coordinate this transition remain largely unknown. We previously found that Wnt signaling, acting through Sox2, promotes neural competence in the Xenopus retina by activating proneural gene expression. We now report that Wnt and Sox2 inhibit neural differentiation through Notch activation. Independently of Sox2, Wnt stimulates retinal progenitor proliferation and this, when combined with the block on differentiation, maintains retinal progenitor fates. Feedback inhibition by Sox2 on Wnt signaling and by the proneural transcription factors on Sox2 mean that each element of the core pathway activates the next element and inhibits the previous one, providing a directional network that ensures retinal cells make the transition from progenitors to neurons and glia.


Subject(s)
Retina/embryology , Retina/physiology , SOXB1 Transcription Factors/physiology , Wnt Proteins/physiology , Xenopus Proteins/physiology , Xenopus laevis/embryology , Xenopus laevis/physiology , beta Catenin/physiology , Animals , Animals, Genetically Modified , Cell Cycle , Cell Differentiation , Cell Proliferation , Embryonic Stem Cells/cytology , Embryonic Stem Cells/physiology , Gene Expression Regulation, Developmental , Models, Biological , Neurogenesis/genetics , Neurogenesis/physiology , Receptors, Notch/genetics , Receptors, Notch/physiology , SOXB1 Transcription Factors/genetics , Signal Transduction , Wnt Proteins/genetics , Xenopus Proteins/genetics , Xenopus laevis/genetics , beta Catenin/genetics
12.
Elife ; 112022 04 28.
Article in English | MEDLINE | ID: mdl-35481836

ABSTRACT

Microglia serve critical remodeling roles that shape the developing nervous system, responding to the changing neural environment with phagocytosis or soluble factor secretion. Recent single-cell sequencing (scRNAseq) studies have revealed the context-dependent diversity in microglial properties and gene expression, but the cues promoting this diversity are not well defined. Here, we ask how interactions with apoptotic neurons shape microglial state, including lysosomal and lipid metabolism gene expression and dependence on Colony-stimulating factor 1 receptor (CSF1R) for survival. Using early postnatal mouse retina, a CNS region undergoing significant developmental remodeling, we performed scRNAseq on microglia from mice that are wild-type, lack neuronal apoptosis (Bax KO), or are treated with CSF1R inhibitor (PLX3397). We find that interactions with apoptotic neurons drive multiple microglial remodeling states, subsets of which are resistant to CSF1R inhibition. We find that TAM receptor Mer and complement receptor 3 are required for clearance of apoptotic neurons, but that Mer does not drive expression of remodeling genes. We show TAM receptor Axl is negligible for phagocytosis or remodeling gene expression but is consequential for microglial survival in the absence of CSF1R signaling. Thus, interactions with apoptotic neurons shift microglia toward distinct remodeling states and through Axl, alter microglial dependence on survival pathway, CSF1R.


Subject(s)
Microglia , Receptors, Granulocyte-Macrophage Colony-Stimulating Factor , Animals , Apoptosis , Mice , Mice, Inbred C57BL , Microglia/metabolism , Phagocytosis , Receptor Protein-Tyrosine Kinases/metabolism , Receptors, Granulocyte-Macrophage Colony-Stimulating Factor/metabolism , Signal Transduction
13.
J Biol Chem ; 285(40): 30814-24, 2010 Oct 01.
Article in English | MEDLINE | ID: mdl-20547764

ABSTRACT

Zinc finger protein 423 encodes a 30 Zn-finger transcription factor involved in cerebellar and olfactory development. ZFP423 is a known interactor of SMAD1-SMAD4 and of Collier/Olf-1/EBF proteins, and acts as a modifier of retinoic acid-induced differentiation. In the present article, we show that ZFP423 interacts with the Notch1 intracellular domain in mammalian cell lines and in Xenopus neurula embryos, to activate the expression of the Notch1 target Hes5/ESR1. This effect is antagonized by EBF transcription factors, both in cultured cells and in Xenopus embryos, and amplified in vitro by BMP4, suggesting that ZFP423 acts to integrate BMP and Notch signaling, selectively promoting their convergence onto the Hes5 gene promoter.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/biosynthesis , Bone Morphogenetic Protein 4/metabolism , DNA-Binding Proteins/metabolism , Embryo, Mammalian/metabolism , Embryo, Nonmammalian/metabolism , Gene Expression Regulation, Developmental/physiology , Receptor, Notch1/metabolism , Repressor Proteins/biosynthesis , Signal Transduction/physiology , Transcription Factors/metabolism , Xenopus Proteins/biosynthesis , Xenopus Proteins/metabolism , Animals , Basic Helix-Loop-Helix Transcription Factors/genetics , Bone Morphogenetic Protein 4/genetics , COS Cells , Chlorocebus aethiops , DNA-Binding Proteins/genetics , Embryo, Mammalian/cytology , Embryo, Nonmammalian/cytology , Humans , Mice , Receptor, Notch1/genetics , Repressor Proteins/genetics , Transcription Factors/genetics , Up-Regulation/physiology , Xenopus Proteins/genetics , Xenopus laevis
14.
Dev Biol ; 326(2): 471-81, 2009 Feb 15.
Article in English | MEDLINE | ID: mdl-19059393

ABSTRACT

During central nervous system development the timing of progenitor differentiation must be precisely controlled to generate the proper number and complement of neuronal cell types. Proneural basic helix-loop-helix (bHLH) transcription factors play a central role in regulating neurogenesis, and thus the timing of their expression must be regulated to ensure that they act at the appropriate developmental time. In the developing retina, the expression of the bHLH factor Ath5 is controlled by multiple signals in early retinal progenitors, although less is known about how these signals are coordinated to ensure correct spatial and temporal pattern of gene expression. Here we identify a key distal Xath5 enhancer and show that this enhancer regulates the early phase of Xath5 expression, while the proximal enhancer we previously identified acts later. The distal enhancer responds to Pax6, a key patterning factor in the optic vesicle, while FGF signaling regulates Xath5 expression through sequences outside of this region. In addition, we have identified an inhibitory element adjacent to the conserved distal enhancer region that is required to prevent premature initiation of expression in the retina. This temporal regulation of Xath5 gene expression is comparable to proneural gene regulation in Drosophila, whereby separate enhancers regulate different temporal phases of expression.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors , Eye Proteins , Eye/embryology , Gene Expression Regulation, Developmental , Xenopus Proteins , Xenopus laevis , Animals , Animals, Genetically Modified , Base Sequence , Basic Helix-Loop-Helix Transcription Factors/genetics , Basic Helix-Loop-Helix Transcription Factors/metabolism , Enhancer Elements, Genetic , Eye/anatomy & histology , Eye/metabolism , Eye Proteins/genetics , Eye Proteins/metabolism , Fibroblast Growth Factors/metabolism , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Molecular Sequence Data , Morphogenesis/physiology , PAX6 Transcription Factor , Paired Box Transcription Factors/genetics , Paired Box Transcription Factors/metabolism , Repressor Proteins/genetics , Repressor Proteins/metabolism , Retina/cytology , Retina/embryology , Retina/metabolism , Signal Transduction/physiology , Transgenes , Xenopus Proteins/genetics , Xenopus Proteins/metabolism , Xenopus laevis/anatomy & histology , Xenopus laevis/embryology
15.
Dev Dyn ; 238(12): 3185-92, 2009 Dec.
Article in English | MEDLINE | ID: mdl-19877271

ABSTRACT

The Polycomb repressive complex 2 is a multimeric aggregate that mediates silencing of a broad range of genes, and is associated with important biological contexts such as stem cell maintenance and cancer progression. PRC2 mainly trimethylates lysine 27 of histone H3 and is composed of three essential core subunits: EZH2, EED, and SUZ12. The Xenopus orthologs of PRC2 subunits Ezh2 and Eed have been described but Suz12 remained unidentified. Here, we report the cloning of the Xenopus Suz12, and determine its spatiotemporal expression during development. Xsuz12 transcript is provided maternally and continues to be expressed throughout development, particularly in the anterior part of the developing central nervous system. Importantly, comparative analysis of the expression of the PRC2 subunits Xez, Xeed, and Xrbbp4 indicates that their expression largely coincides with Xsuz12 in the nervous system, suggesting that PRC2 may have unexplored functions in the development of the frog central nervous system.


Subject(s)
Embryonic Development/genetics , Repressor Proteins/genetics , Xenopus Proteins/genetics , Xenopus laevis/embryology , Xenopus laevis/genetics , Amino Acid Sequence , Animals , Cloning, Molecular , Embryo, Nonmammalian , Enhancer of Zeste Homolog 2 Protein , Gene Expression Profiling , Gene Expression Regulation, Developmental , Molecular Sequence Data , Nervous System/embryology , Nervous System/metabolism , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Phylogeny , Polycomb Repressive Complex 2 , Polycomb-Group Proteins , Protein Subunits/genetics , Protein Subunits/metabolism , Repressor Proteins/chemistry , Repressor Proteins/metabolism , Retinoblastoma-Binding Protein 4/genetics , Retinoblastoma-Binding Protein 4/metabolism , Sequence Homology, Amino Acid , Xenopus Proteins/metabolism , Xenopus laevis/metabolism
16.
Front Cell Dev Biol ; 8: 581136, 2020.
Article in English | MEDLINE | ID: mdl-33043015

ABSTRACT

Retinal development follows a conserved neurogenic program in vertebrates to orchestrate the generation of specific cell types from multipotent progenitors in sequential but overlapping waves. In this program, retinal ganglion cells (RGCs) are the first cell type generated. RGCs are the final output neurons of the retina and are essential for vision and circadian rhythm. Key molecular steps have been defined in multiple vertebrate species to regulate competence, specification, and terminal differentiation of this cell type. This involves neuronal-specific transcription factor networks, regulators of chromatin dynamics and miRNAs. In mammals, RGCs and their optic nerve axons undergo neurodegeneration and loss in glaucoma and other optic neuropathies, resulting in irreversible vision loss. The incapacity of RGCs and axons to regenerate reinforces the need for the design of efficient RGC replacement strategies. Here we describe the essential molecular pathways for the differentiation of RGCs in vertebrates, as well as experimental manipulations that extend the competence window for generation of this early cell type from late progenitors. We discuss recent advances in regeneration of retinal neurons in vivo in both mouse and zebrafish and discuss possible strategies and barriers to achieving RGC regeneration as a therapeutic approach for vision restoration in blinding diseases such as glaucoma.

17.
Genesis ; 47(3): 175-87, 2009 Mar.
Article in English | MEDLINE | ID: mdl-19208436

ABSTRACT

Activation of the bHLH factor Math5 (Atoh7) is an initiating event for mammalian retinal neurogenesis, as it is critically required for retinal ganglion cell formation. However, the cis-regulatory elements and trans-acting factors that control Math5 expression are largely unknown. Using a combination of transgenic mice and bioinformatics, we identified a phylogenetically conserved regulatory element that is required to activate Math5 transcription during early retinal neurogenesis. This element drives retinal expression in vivo, in a cross-species transgenic assay. Previously, Pax6 was shown to be necessary for the initiation of Math5 mRNA expression. We extend this finding by showing that the Math5 retinal enhancer also requires Pax6 for its activation, via Pax6 binding to a highly conserved binding site. In addition, our data reveal that other retinal factors are required for accurate regulation of Math5 by Pax6.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/biosynthesis , Eye Proteins/biosynthesis , Gene Expression Regulation, Developmental/physiology , Homeodomain Proteins/biosynthesis , Nerve Tissue Proteins/biosynthesis , Neurogenesis/physiology , Paired Box Transcription Factors/biosynthesis , Repressor Proteins/biosynthesis , Response Elements/physiology , Retinal Ganglion Cells/metabolism , Animals , Basic Helix-Loop-Helix Transcription Factors/genetics , Eye Proteins/genetics , Homeodomain Proteins/genetics , Mice , Mice, Mutant Strains , Mice, Transgenic , Nerve Tissue Proteins/genetics , PAX6 Transcription Factor , Paired Box Transcription Factors/genetics , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Repressor Proteins/genetics , Retinal Ganglion Cells/cytology , Transcription, Genetic/physiology , Xenopus
18.
Neuron ; 46(1): 23-36, 2005 Apr 07.
Article in English | MEDLINE | ID: mdl-15820691

ABSTRACT

Progenitors in the developing central nervous system acquire neural potential and proliferate to expand the pool of precursors competent to undergo neuronal differentiation. The formation and maintenance of neural-competent precursors are regulated by SoxB1 transcription factors, and evidence that their expression is regionally regulated suggests that specific signals regulate neural potential in subdomains of the developing nervous system. We show that the frizzled (Fz) transmembrane receptor Xfz5 selectively governs neural potential in the developing Xenopus retina by regulating the expression of Sox2. Blocking either Xfz5 or canonical Wnt signaling within the developing retina inhibits Sox2 expression, reduces cell proliferation, inhibits the onset of proneural gene expression, and biases individual progenitors toward a nonneural fate, without altering the expression of multiple progenitor markers. Blocking Sox2 function mimics these effects. Rescue experiments indicate that Sox2 is downstream of Xfz5. Thus, Fz signaling can regulate the neural potential of progenitors in the developing nervous system.


Subject(s)
Eye Proteins/metabolism , Neurons/cytology , Retina/embryology , Signal Transduction/physiology , Xenopus Proteins/metabolism , Animals , Animals, Genetically Modified , DNA-Binding Proteins/metabolism , Embryo, Nonmammalian , Frizzled Receptors , Gene Expression Regulation, Developmental , In Situ Hybridization , Intercellular Signaling Peptides and Proteins/metabolism , Nerve Tissue Proteins/metabolism , Neurons/metabolism , Retina/cytology , Stem Cells , Wnt Proteins , Xenopus
19.
J Neurosci ; 28(11): 2735-44, 2008 Mar 12.
Article in English | MEDLINE | ID: mdl-18337403

ABSTRACT

Glaucoma is characterized by retinal ganglion cell (RGC) pathology and a progressive loss of vision. Previous studies suggest RGC death is responsible for vision loss in glaucoma, yet evidence from other neurodegenerative diseases suggests axonal degeneration, in the absence of neuronal loss, can significantly affect neuronal function. To characterize RGC degeneration in the DBA/2 mouse model of glaucoma, we quantified RGCs in mice of various ages using neuronal-specific nuclear protein (NeuN) immunolabeling, retrograde labeling, and optic nerve axon counts. Surprisingly, the number of NeuN-labeled RGCs did not decline significantly until 18 months of age, at which time a significant decrease in RGC somal size was also observed. Axon dysfunction and degeneration occurred before loss of NeuN-positive RGCs, because significant declines in RGC number assayed by retrograde tracers and axon counts were observed at 13 months. To examine whether axonal dysfunction/degeneration affected gene expression in RGC axons or somas, NeuN and neurofilament-heavy (NF-H) immunolabeling was performed along with quantitative reverse transcription-PCR for RGC-specific genes in retinas of aged DBA/2 mice. Although these mice had similar numbers of NeuN-positive RGCs, the expression of neurofilament light, Brn-3b, and Sncg mRNA varied; this variation in RGC-specific gene expression was correlated with the appearance of NF-H immunoreactive RGC axons. Together, these data support a progression of RGC degeneration in this model of glaucoma, beginning with loss of retrograde label, where axon dysfunction and degeneration precede neuronal loss. This progression of degeneration suggests a need to examine the RGC axon as a locus of pathology in glaucoma.


Subject(s)
Disease Models, Animal , Glaucoma/pathology , Nerve Degeneration/pathology , Neurons/pathology , Retinal Ganglion Cells/pathology , Animals , Cell Count/methods , Cell Death/physiology , Disease Progression , Glaucoma/genetics , Mice , Mice, Inbred C57BL , Mice, Inbred DBA , Nerve Degeneration/genetics , Neurons/physiology , Retinal Ganglion Cells/physiology
20.
J Neurosci ; 28(2): 548-61, 2008 Jan 09.
Article in English | MEDLINE | ID: mdl-18184797

ABSTRACT

Little is known about molecular changes occurring within retinal ganglion cells (RGCs) before their death in glaucoma. Taking advantage of the fact that gamma-synuclein (Sncg) mRNA is expressed specifically and highly in adult mouse RGCs, we show in the DBA/2J mouse model of glaucoma that there is not only a loss of cells expressing this gene, but also a downregulation of gene expression of Sncg and many other genes within large numbers of RGCs. This downregulation of gene expression within RGCs occurs together with reductions in FluoroGold (FG) retrograde transport. Surprisingly, there are also large numbers of Sncg-expressing cells without any FG labeling, and among these many that have a marker previously associated with disconnected RGCs, accumulation of phosphorylated neurofilaments in their somas. These same diseased retinas also have large numbers of RGCs that maintain the intraocular portion while losing the optic nerve portion of their axons, and these disconnected axons terminate within the optic nerve head. Our data support the view that RGC degeneration in glaucoma has two separable stages: the first involves atrophy of RGCs, whereas the second involves an insult to axons, which causes the degeneration of axon portions distal to the optic nerve head but does not cause the immediate degeneration of intraretinal portions of axons or the immediate death of RGCs.


Subject(s)
Axons/pathology , Down-Regulation/physiology , Glaucoma/pathology , Optic Disk/pathology , Optic Disk/physiopathology , Retinal Ganglion Cells/physiology , Age Factors , Animals , Axotomy/methods , Disease Models, Animal , Glaucoma/complications , Mice , Mice, Inbred C57BL , Mice, Inbred DBA , Nerve Degeneration/etiology , Nerve Degeneration/physiopathology , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , RNA, Messenger/metabolism , Species Specificity , Stilbamidines/metabolism , gamma-Synuclein/genetics , gamma-Synuclein/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL