Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 17 de 17
1.
Cell Rep ; 43(6): 114291, 2024 May 31.
Article En | MEDLINE | ID: mdl-38823017

Atoh7 is transiently expressed in retinal progenitor cells (RPCs) and is required for retinal ganglion cell (RGC) differentiation. In humans, a deletion in a distal non-coding regulatory region upstream of ATOH7 is associated with optic nerve atrophy and blindness. Here, we functionally interrogate the significance of the Atoh7 regulatory landscape to retinogenesis in mice. Deletion of the Atoh7 enhancer structure leads to RGC deficiency, optic nerve hypoplasia, and retinal blood vascular abnormalities, phenocopying inactivation of Atoh7. Further, loss of the Atoh7 remote enhancer impacts ipsilaterally projecting RGCs and disrupts proper axonal projections to the visual thalamus. Deletion of the Atoh7 remote enhancer is also associated with the dysregulation of axonogenesis genes, including the derepression of the axon repulsive cue Robo3. Our data provide insights into how Atoh7 enhancer elements function to promote RGC development and optic nerve formation and highlight a key role of Atoh7 in the transcriptional control of axon guidance molecules.

2.
Sci Rep ; 13(1): 5592, 2023 04 05.
Article En | MEDLINE | ID: mdl-37019993

As part of the central nervous system (CNS), retinal ganglion cells (RGCs) and their axons are the only neurons in the retina that transmit visual signals from the eye to the brain via the optic nerve (ON). Unfortunately, they do not regenerate upon injury in mammals. In ON trauma, retinal microglia (RMG) become activated, inducing inflammatory responses and resulting in axon degeneration and RGC loss. Since aldose reductase (AR) is an inflammatory response mediator highly expressed in RMG, we investigated if pharmacological inhibition of AR can attenuate ocular inflammation and thereby promote RGC survival and axon regeneration after ON crush (ONC). In vitro, we discovered that Sorbinil, an AR inhibitor, attenuates BV2 microglia activation and migration in the lipopolysaccharide (LPS) and monocyte chemoattractant protein-1 (MCP-1) treatments. In vivo, Sorbinil suppressed ONC-induced Iba1 + microglia/macrophage infiltration in the retina and ON and promoted RGC survival. Moreover, Sorbinil restored RGC function and delayed axon degeneration one week after ONC. RNA sequencing data revealed that Sorbinil protects the retina from ONC-induced degeneration by suppressing inflammatory signaling. In summary, we report the first study demonstrating that AR inhibition transiently protects RGC and axon from degeneration, providing a potential therapeutic strategy for optic neuropathies.


Optic Atrophy , Optic Nerve Injuries , Animals , Microglia , Axons/physiology , Aldehyde Reductase , Nerve Regeneration , Retina , Optic Nerve Injuries/pathology , Optic Atrophy/pathology , Nerve Degeneration/pathology , Mammals
3.
Development ; 149(8)2022 04 15.
Article En | MEDLINE | ID: mdl-34528064

Visual information is transmitted from the eye to the brain along the optic nerve, a structure composed of retinal ganglion cell (RGC) axons. The optic nerve is highly vulnerable to damage in neurodegenerative diseases, such as glaucoma, and there are currently no FDA-approved drugs or therapies to protect RGCs from death. Zebrafish possess remarkable neuroprotective and regenerative abilities. Here, utilizing an optic nerve transection (ONT) injury and an RNA-seq-based approach, we identify genes and pathways active in RGCs that may modulate their survival. Through pharmacological perturbation, we demonstrate that Jak/Stat pathway activity is required for RGC survival after ONT. Furthermore, we show that immune responses directly contribute to RGC death after ONT; macrophages/microglia are recruited to the retina and blocking neuroinflammation or depleting these cells after ONT rescues survival of RGCs. Taken together, these data support a model in which crosstalk between macrophages/microglia and RGCs, mediated by Jak/Stat pathway activity, regulates RGC survival after optic nerve injury.


Immunity, Innate , Janus Kinases/immunology , Optic Nerve Injuries/immunology , Retinal Ganglion Cells/immunology , STAT Transcription Factors/immunology , Signal Transduction/immunology , Zebrafish Proteins/immunology , Zebrafish/immunology , Animals , Animals, Genetically Modified , Female , Janus Kinases/genetics , Male , Optic Nerve Injuries/genetics , STAT Transcription Factors/genetics , Signal Transduction/genetics , Zebrafish/genetics , Zebrafish Proteins/genetics
4.
PLoS Genet ; 17(11): e1009885, 2021 11.
Article En | MEDLINE | ID: mdl-34735454

Molecular insights into the selective vulnerability of retinal ganglion cells (RGCs) in optic neuropathies and after ocular trauma can lead to the development of novel therapeutic strategies aimed at preserving RGCs. However, little is known about what molecular contexts determine RGC susceptibility. In this study, we show the molecular mechanisms underlying the regional differential vulnerability of RGCs after optic nerve injury. We identified RGCs in the mouse peripheral ventrotemporal (VT) retina as the earliest population of RGCs susceptible to optic nerve injury. Mechanistically, the serotonin transporter (SERT) is upregulated on VT axons after injury. Utilizing SERT-deficient mice, loss of SERT attenuated VT RGC death and led to robust retinal axon regeneration. Integrin ß3, a factor mediating SERT-induced functions in other systems, is also upregulated in RGCs and axons after injury, and loss of integrin ß3 led to VT RGC protection and axon regeneration. Finally, RNA sequencing analyses revealed that loss of SERT significantly altered molecular signatures in the VT retina after optic nerve injury, including expression of the transmembrane protein, Gpnmb. GPNMB is rapidly downregulated in wild-type, but not SERT- or integrin ß3-deficient VT RGCs after injury, and maintaining expression of GPNMB in RGCs via AAV2 viruses even after injury promoted VT RGC survival and axon regeneration. Taken together, our findings demonstrate that the SERT-integrin ß3-GPNMB molecular axis mediates selective RGC vulnerability and axon regeneration after optic nerve injury.


Axons , Nerve Regeneration , Nervous System Diseases/metabolism , Retinal Ganglion Cells/cytology , Serotonin Plasma Membrane Transport Proteins/physiology , Animals , Mice , Mice, Inbred C57BL , Nervous System Diseases/pathology
5.
Nat Commun ; 12(1): 5040, 2021 08 19.
Article En | MEDLINE | ID: mdl-34413305

SMN is a ubiquitously expressed protein and is essential for life. SMN deficiency causes the neurodegenerative disease spinal muscular atrophy (SMA), the leading genetic cause of infant mortality. SMN interacts with itself and other proteins to form a complex that functions in the assembly of ribonucleoproteins. SMN is modified by SUMO (Small Ubiquitin-like Modifier), but whether sumoylation is required for the functions of SMN that are relevant to SMA pathogenesis is not known. Here, we show that inactivation of a SUMO-interacting motif (SIM) alters SMN sub-cellular distribution, the integrity of its complex, and its function in small nuclear ribonucleoproteins biogenesis. Expression of a SIM-inactivated mutant of SMN in a mouse model of SMA slightly extends survival rate with limited and transient correction of motor deficits. Remarkably, although SIM-inactivated SMN attenuates motor neuron loss and improves neuromuscular junction synapses, it fails to prevent the loss of sensory-motor synapses. These findings suggest that sumoylation is important for proper assembly and function of the SMN complex and that loss of this post-translational modification impairs the ability of SMN to correct selective deficits in the sensory-motor circuit of SMA mice.


Motor Neurons/metabolism , Muscular Atrophy, Spinal/pathology , Neurodegenerative Diseases/pathology , Ribonucleoproteins, Small Nuclear/metabolism , SMN Complex Proteins/metabolism , Sumoylation , Synapses/metabolism , Animals , Animals, Genetically Modified , Cells, Cultured , Disease Models, Animal , Humans , Mice , Motor Neurons/pathology , Muscular Atrophy, Spinal/metabolism , Neurodegenerative Diseases/metabolism , Synapses/pathology , Zebrafish
6.
J Comp Neurol ; 526(7): 1077-1096, 2018 05 01.
Article En | MEDLINE | ID: mdl-29322522

Prior to forming and refining synaptic connections, axons of projection neurons navigate long distances to their targets. While much is known about guidance cues for axon navigation through intermediate choice points, whether and how axons are organized within tracts is less clear. Here we analyze the organization of retinal ganglion cell (RGC) axons in the developing mouse retinogeniculate pathway. RGC axons are organized by both eye-specificity and topography in the optic nerve and tract: ipsilateral RGC axons are segregated from contralateral axons and are offset laterally in the tract relative to contralateral axon topographic position. To identify potential cell-autonomous factors contributing to the segregation of ipsilateral and contralateral RGC axons in the visual pathway, we assessed their fasciculation behavior in a retinal explant assay. Ipsilateral RGC neurites self-fasciculate more than contralateral neurites in vitro and maintain this difference in the presence of extrinsic chiasm cues. To further probe the role of axon self-association in circuit formation in vivo, we examined RGC axon organization and fasciculation in an EphB1-/- mutant, in which a subset of ipsilateral RGC axons aberrantly crosses the midline but targets the ipsilateral zone in the dorsal lateral geniculate nucleus on the opposite side. Aberrantly crossing axons retain their association with ipsilateral axons in the contralateral tract, indicating that cohort-specific axon affinity is maintained independently of guidance signals present at the midline. Our results provide a comprehensive assessment of RGC axon organization in the retinogeniculate pathway and suggest that axon self-association contributes to pre-target axon organization.


Axons/physiology , Optic Nerve/physiology , Retinal Ganglion Cells/cytology , Visual Pathways , Amino Acids/metabolism , Animals , Animals, Newborn , Embryo, Mammalian , Eye/cytology , Eye/innervation , Fasciculation , Functional Laterality , In Vitro Techniques , Intermediate Filaments/metabolism , Luminescent Proteins/genetics , Luminescent Proteins/metabolism , Mice , Mice, Inbred C57BL , Mice, Transgenic , Optic Nerve/embryology , Optic Nerve/growth & development , Receptor, EphB1/genetics , Receptor, EphB1/metabolism , Serotonin Plasma Membrane Transport Proteins/genetics , Serotonin Plasma Membrane Transport Proteins/metabolism , Visual Pathways/anatomy & histology , Visual Pathways/embryology , Visual Pathways/growth & development
7.
Neuron ; 94(6): 1043-1046, 2017 Jun 21.
Article En | MEDLINE | ID: mdl-28641102

In this issue of Neuron, Welsbie et al. (2017) and Norsworthy et al. (2017) implicate the transcription factor Sox11 as a key player after optic nerve injury-in DLK signaling of RGC cell death, and in RGC regeneration and survival but only in certain RGCs.


Axons , Retinal Ganglion Cells , Cell Death , Cell Survival , Humans , Nerve Regeneration , Optic Nerve , Optic Nerve Injuries , Signal Transduction
8.
Neuron ; 93(5): 1110-1125.e5, 2017 Mar 08.
Article En | MEDLINE | ID: mdl-28215559

Transcription factors control cell identity by regulating diverse developmental steps such as differentiation and axon guidance. The mammalian binocular visual circuit is comprised of projections of retinal ganglion cells (RGCs) to ipsilateral and contralateral targets in the brain. A transcriptional code for ipsilateral RGC identity has been identified, but less is known about the transcriptional regulation of contralateral RGC development. Here we demonstrate that SoxC genes (Sox4, 11, and 12) act on the progenitor-to-postmitotic transition to implement contralateral, but not ipsilateral, RGC differentiation, by binding to Hes5 and thus repressing Notch signaling. When SoxC genes are deleted in postmitotic RGCs, contralateral RGC axons grow poorly on chiasm cells in vitro and project ipsilaterally at the chiasm midline in vivo, and Plexin-A1 and Nr-CAM expression in RGCs is downregulated. These data implicate SoxC transcription factors in the regulation of contralateral RGC differentiation and axon guidance.


Axons/metabolism , Cell Differentiation/physiology , Retina/metabolism , Retinal Ganglion Cells/cytology , Retinal Ganglion Cells/metabolism , SOXC Transcription Factors/metabolism , Visual Pathways/metabolism , Animals , Axon Guidance/physiology , Mice, Transgenic , Nerve Tissue Proteins/metabolism , Optic Chiasm/metabolism , Retina/cytology
9.
Cell Rep ; 17(12): 3153-3164, 2016 12 20.
Article En | MEDLINE | ID: mdl-28009286

The retina of lower vertebrates grows continuously by integrating new neurons generated from progenitors in the ciliary margin zone (CMZ). Whether the mammalian CMZ provides the neural retina with retinal cells is controversial. Live imaging of embryonic retina expressing eGFP in the CMZ shows that cells migrate laterally from the CMZ to the neural retina where differentiated retinal ganglion cells (RGCs) reside. Because Cyclin D2, a cell-cycle regulator, is enriched in ventral CMZ, we analyzed Cyclin D2-/- mice to test whether the CMZ is a source of retinal cells. Neurogenesis is diminished in Cyclin D2 mutants, leading to a reduction of RGCs in the ventral retina. In line with these findings, in the albino retina, the decreased production of ipsilateral RGCs is correlated with fewer Cyclin D2+ cells. Together, these results implicate the mammalian CMZ as a neurogenic site that produces RGCs and whose proper generation depends on Cyclin D2 activity.


Cyclin D2/genetics , Neurogenesis/genetics , Retina/metabolism , Retinal Ganglion Cells/metabolism , Animals , Cell Differentiation/genetics , Cell Proliferation/genetics , Cilia/genetics , Cilia/metabolism , Humans , Mammals , Mice , Mice, Knockout , Retina/embryology , Retina/growth & development
10.
Elife ; 52016 09 20.
Article En | MEDLINE | ID: mdl-27648578

The regulation of protein degradation is essential for maintaining the appropriate environment to coordinate complex cell signaling events and to promote cellular remodeling. The Autophagy linked FYVE protein (Alfy), previously identified as a molecular scaffold between the ubiquitinated cargo and the autophagic machinery, is highly expressed in the developing central nervous system, indicating that this pathway may have yet unexplored roles in neurodevelopment. To examine this possibility, we used mouse genetics to eliminate Alfy expression. We report that this evolutionarily conserved protein is required for the formation of axonal tracts throughout the brain and spinal cord, including the formation of the major forebrain commissures. Consistent with a phenotype reflecting a failure in axon guidance, the loss of Alfy in mice disrupts localization of glial guidepost cells, and attenuates axon outgrowth in response to Netrin-1. These findings further support the growing indication that macroautophagy plays a key role in the developing CNS.


Brain/embryology , Neural Pathways/embryology , Neurons/physiology , Vesicular Transport Proteins/metabolism , Adaptor Proteins, Signal Transducing , Animals , Autophagy-Related Proteins , Gene Knockout Techniques , Mice, Inbred C57BL
11.
Cell Rep ; 16(2): 545-558, 2016 07 12.
Article En | MEDLINE | ID: mdl-27373155

Suboptimal axonal regeneration contributes to the consequences of nervous system trauma and neurodegenerative disease, but the intrinsic mechanisms that regulate axon growth remain unclear. We screened 50,400 small molecules for their ability to promote axon outgrowth on inhibitory substrata. The most potent hits were the statins, which stimulated growth of all mouse- and human-patient-derived neurons tested, both in vitro and in vivo, as did combined inhibition of the protein prenylation enzymes farnesyltransferase (PFT) and geranylgeranyl transferase I (PGGT-1). Compensatory sprouting of motor axons may delay clinical onset of amyotrophic lateral sclerosis (ALS). Accordingly, elevated levels of PGGT1B, which would be predicted to reduce sprouting, were found in motor neurons of early- versus late-onset ALS patients postmortem. The mevalonate-prenylation pathway therefore constitutes an endogenous brake on axonal growth, and its inhibition provides a potential therapeutic approach to accelerate neuronal regeneration in humans.


Neurites/physiology , Protein Prenylation , Amyotrophic Lateral Sclerosis/pathology , Animals , Cell Enlargement , Cells, Cultured , Humans , Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacology , Mevalonic Acid/metabolism , Mice , Motor Neurons/physiology , Nerve Regeneration
12.
Development ; 140(6): 1364-8, 2013 Mar.
Article En | MEDLINE | ID: mdl-23444362

We describe a clearing method for enhanced visualization of cell morphology and connections in neuronal and non-neuronal tissue. Using Clear(T) or Clear(T2), which are composed of formamide or formamide/polyethylene glycol, respectively, embryos, whole mounts and thick brain sections can be rapidly cleared with minimal volume changes. Unlike other available clearing techniques, these methods do not use detergents or solvents, and thus preserve lipophilic dyes, fluorescent tracers and immunohistochemical labeling, as well as fluorescent-protein labeling.


Cytological Techniques/methods , Dissection/methods , Indicators and Reagents , Neurons/cytology , Animals , Brain/cytology , Detergents/adverse effects , Embryo, Mammalian , Indicators and Reagents/pharmacology , Mice , Mice, Inbred C57BL , Models, Biological , Neurons/physiology , Solvents/adverse effects , Specimen Handling/methods , Staining and Labeling/methods , Tissue Fixation/methods
13.
Neuron ; 74(4): 676-90, 2012 May 24.
Article En | MEDLINE | ID: mdl-22632726

At the optic chiasm, retinal ganglion cells (RGCs) project ipsi- or contralaterally to establish the circuitry for binocular vision. Ipsilateral guidance programs have been characterized, but contralateral guidance programs are not well understood. Here, we identify a tripartite molecular system for contralateral RGC projections: Semaphorin6D (Sema6D) and Nr-CAM are expressed on midline radial glia and Plexin-A1 on chiasm neurons, and Plexin-A1 and Nr-CAM are also expressed on contralateral RGCs. Sema6D is repulsive to contralateral RGCs, but Sema6D in combination with Nr-CAM and Plexin-A1 converts repulsion to growth promotion. Nr-CAM functions as a receptor for Sema6D. Sema6D, Plexin-A1, and Nr-CAM are all required for efficient RGC decussation at the optic chiasm. These findings suggest a mechanism by which a complex of Sema6D, Nr-CAM, and Plexin-A1 at the chiasm midline alters the sign of Sema6D and signals Nr-CAM/Plexin-A1 receptors on RGCs to implement the contralateral RGC projection.


Cell Adhesion Molecules/metabolism , Nerve Tissue Proteins/metabolism , Optic Chiasm/metabolism , Receptors, Cell Surface/metabolism , Retinal Ganglion Cells/metabolism , Semaphorins/metabolism , Animals , Axons/metabolism , Cells, Cultured , Mice , Mice, Knockout , Optic Chiasm/cytology , Retina/cytology , Retina/metabolism , Retinal Ganglion Cells/cytology
14.
J Neurosci ; 30(10): 3709-14, 2010 Mar 10.
Article En | MEDLINE | ID: mdl-20220004

Necdin is a pleiotropic protein that promotes neuronal differentiation and survival. In mammals, the necdin gene on the maternal chromosome is silenced by genomic imprinting, and only the paternal necdin gene is expressed in virtually all postmitotic neurons. Necdin forms a complex with the homeodomain protein Dlx2 to enhance its transcriptional activity. Dlx2 plays a major role in controlling tangential migration of GABAergic interneurons from the basal forebrain to the neocortex. Here, we examined whether Dlx2-expressing interneurons migrate properly in vivo in mutant mice lacking the paternal necdin gene. In necdin-deficient mice at birth, the population of Dlx2-expressing cells significantly decreased in the neocortex but increased in the preoptic area. DiI-labeled cell migration assay using organotypic forebrain slice cultures revealed that the number of cells migrating from the medial ganglionic eminence into the neocortex was significantly reduced in necdin-deficient embryos. Furthermore, necdin-deficient mice had a decreased population of neocortical GABA-containing neurons and were highly susceptible to pentylenetetrazole-induced seizures. These results suggest that necdin promotes tangential migration of neocortical GABAergic interneurons during mammalian forebrain development.


Cell Movement/physiology , Interneurons/cytology , Interneurons/physiology , Neocortex/cytology , Neocortex/physiology , Nerve Tissue Proteins/deficiency , Nuclear Proteins/deficiency , Prosencephalon/cytology , Prosencephalon/physiology , Animals , Evolution, Molecular , Female , Interneurons/metabolism , Male , Mice , Mice, Inbred ICR , Mice, Knockout , Neocortex/metabolism , Nerve Tissue Proteins/genetics , Nuclear Proteins/genetics , Organ Culture Techniques , Prosencephalon/metabolism
15.
J Neurosci ; 26(46): 12003-13, 2006 Nov 15.
Article En | MEDLINE | ID: mdl-17108174

The cell cycle-regulatory transcription factor E2F1 induces apoptosis of postmitotic neurons in developmental and pathological situations. E2F1 transcriptionally activates many proapoptotic genes including the cyclin-dependent protein kinase cell division cycle 2 (Cdc2). Necdin is a potent mitotic suppressor expressed predominantly in postmitotic neurons and interacts with E2F1 to suppress E2F1-mediated gene transcription. The necdin gene NDN is maternally imprinted and expressed only from the paternal allele. Deletion of the paternal NDN is implicated in the pathogenesis of Prader-Willi syndrome, a genomic imprinting-associated neurodevelopmental disorder. Here, we show that paternally expressed necdin represses E2F1-dependent cdc2 gene transcription and attenuates apoptosis of postmitotic neurons. Necdin was abundantly expressed in differentiated cerebellar granule neurons (CGNs). Neuronal activity deprivation elevated the expression of both E2F1 and Cdc2 in primary CGNs prepared from mice at postnatal day 6, whereas the necdin levels remained unchanged. In chromatin immunoprecipitation analysis, endogenous necdin was associated with the cdc2 promoter containing an E2F-binding site in activity-deprived CGNs. After activity deprivation, CGNs underwent apoptosis, which was augmented in those prepared from mice defective in the paternal Ndn allele (Ndn(+m/-p)). The levels of cdc2 mRNA, protein, and kinase activity were significantly higher in Ndn(+m/-p) CGNs than in wild-type CGNs under activity-deprived conditions. Furthermore, the populations of Cdc2-immunoreactive and apoptotic cells were increased in the cerebellum in vivo of Ndn(+m/-p) mice. These results suggest that endogenous necdin attenuates neuronal apoptosis by suppressing the E2F1-Cdc2 system.


Apoptosis/genetics , CDC2 Protein Kinase/biosynthesis , Cerebellar Cortex/metabolism , Down-Regulation/physiology , Nerve Tissue Proteins/metabolism , Neurons/metabolism , Nuclear Proteins/metabolism , Animals , Animals, Newborn , Binding Sites/genetics , CDC2 Protein Kinase/genetics , Cell Count , Cells, Cultured , Cerebellar Cortex/cytology , Cerebellar Cortex/growth & development , E2F1 Transcription Factor/metabolism , Gene Expression Regulation, Developmental/physiology , Mice , Mice, Inbred ICR , Nerve Tissue Proteins/genetics , Neurons/cytology , Nuclear Proteins/genetics , Promoter Regions, Genetic/genetics , RNA, Messenger/metabolism , Regulatory Elements, Transcriptional/genetics
16.
J Neurosci ; 26(20): 5383-92, 2006 May 17.
Article En | MEDLINE | ID: mdl-16707790

Necdin, a member of the MAGE (melanoma antigen) protein family, is expressed predominantly in terminally differentiated neurons. The necdin gene NDN is maternally imprinted and expressed only from the paternal allele, the deficiency of which is implicated in the pathogenesis of the neurodevelopmental disorder Prader-Willi syndrome. Necdin binds to its homologous MAGE protein MAGE-D1 (also known as NRAGE or Dlxin-1), which interacts with Msx (msh homeobox) and Dlx (distal-less homeobox) family homeodomain transcription factors. Members of the Dlx homeobox gene family are involved in the differentiation and specification of forebrain GABAergic neurons. Here we demonstrate that necdin associates with Dlx homeodomain proteins via MAGE-D1 to promote the differentiation of GABAergic neurons in mouse embryonic forebrain. Immunohistochemical analysis revealed that necdin was coexpressed with Dlx2, Dlx5, or MAGE-D1 in a subpopulation of embryonic forebrain cells. Necdin bound to Dlx2 and Dlx5 via MAGE-D1 and enhanced Dlx2-dependent activation of the Wnt1 (wingless-type MMTV integration site family) promoter. Necdin significantly increased the populations of cells expressing the GABAergic neuron markers calbindin D-28k and glutamic acid decarboxylase when overexpressed by electroporation in cultured forebrain slices. In this assay, Dlx5N, a truncated Dlx5 mutant that competes with Dlx2 to bind MAGE-D1, diminished the effect of necdin on GABAergic neuron differentiation. Furthermore, mutant mice lacking the paternal necdin allele showed a significant reduction in the differentiation of forebrain GABAergic neurons in vivo and in vitro. These results suggest that paternally expressed necdin facilitates the differentiation and specification of GABAergic neurons in cooperation with Dlx homeodomain proteins.


Cell Differentiation/physiology , Homeodomain Proteins/metabolism , Nerve Tissue Proteins/metabolism , Neurons/metabolism , Nuclear Proteins/metabolism , Prosencephalon/embryology , Transcription Factors/metabolism , gamma-Aminobutyric Acid/metabolism , Animals , Calbindins , Calcium-Binding Proteins/metabolism , Cell Line , Cells, Cultured , Glutamate Decarboxylase/metabolism , Homeodomain Proteins/genetics , Humans , Mice , Mice, Inbred ICR , Mice, Knockout , Mutation/genetics , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , Nerve Tissue Proteins/genetics , Nuclear Proteins/genetics , Organ Culture Techniques , Promoter Regions, Genetic/genetics , Prosencephalon/metabolism , Protein Binding/physiology , S100 Calcium Binding Protein G , Transcription Factors/genetics , Wnt1 Protein/genetics , Wnt1 Protein/metabolism
17.
J Biol Chem ; 279(39): 40484-93, 2004 Sep 24.
Article En | MEDLINE | ID: mdl-15272023

Necdin is a potent growth suppressor that is expressed predominantly in postmitotic cells such as neurons and skeletal muscle cells. Necdin shows a significant homology to MAGE (melanoma antigen) family proteins, all of which contain a large homology domain. MAGE-D1 (NRAGE, Dlxin-1) interacts with the Dlx/Msx family homeodomain proteins via an interspersed hexapeptide repeat domain distinct from the homology domain. Here we report that necdin associates with the Msx homeodomain proteins via MAGE-D1 to modulate their function. In vitro binding and co-immunoprecipitation analyses revealed that MAGE-D1 directly interacted with necdin via the homology domain and Msx1 (or Msx2) via the repeat domain. A ternary complex of necdin, MAGE-D1, and Msx2 was formed in vitro, and an endogenous complex containing these three proteins was detected in differentiating embryonal carcinoma cells. Co-expression of necdin and MAGE-D1 released Msx-dependent transcriptional repression. C2C12 myoblast cells that were stably transfected with Msx2 cDNA showed a marked reduction in myogenic differentiation, and co-expression of necdin and MAGE-D1 canceled the Msx2-dependent repression. These results suggest that necdin and MAGE-D1 cooperate to modulate the function of Dlx/Msx homeodomain proteins in cellular differentiation.


DNA-Binding Proteins/metabolism , Nerve Tissue Proteins/physiology , Nuclear Proteins/physiology , Animals , Blotting, Western , COS Cells , Cell Differentiation , Cell Line , Cell Line, Tumor , DNA, Complementary/metabolism , Gene Deletion , Genetic Vectors , Homeodomain Proteins , Immunoblotting , Immunohistochemistry , Mice , Mice, Inbred ICR , Microscopy, Fluorescence , Mitosis , Models, Biological , Neoplasm Proteins/metabolism , Neurons/metabolism , Precipitin Tests , Protein Binding , Protein Structure, Tertiary , Tissue Distribution , Transcription, Genetic
...