Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
Add more filters











Publication year range
1.
Nucleic Acids Res ; 47(15): 7870-7885, 2019 09 05.
Article in English | MEDLINE | ID: mdl-31226208

ABSTRACT

Long interspersed elements-1 (LINE-1, L1) are retrotransposons that hold the capacity of self-propagation in the genome with potential mutagenic outcomes. How somatic cells restrict L1 activity and how this process becomes dysfunctional during aging and in cancer cells is poorly understood. L1s are enriched at lamin-associated domains, heterochromatic regions of the nuclear periphery. Whether this association is necessary for their repression has been elusive. Here we show that the sirtuin family member SIRT7 participates in the epigenetic transcriptional repression of L1 genome-wide in both mouse and human cells. SIRT7 depletion leads to increased L1 expression and retrotransposition. Mechanistically, we identify a novel interplay between SIRT7 and Lamin A/C in L1 repression. Our results demonstrate that SIRT7-mediated H3K18 deacetylation regulates L1 expression and promotes L1 association with elements of the nuclear lamina. The failure of such activity might contribute to the observed genome instability and compromised viability in SIRT7 knockout mice. Overall, our results reveal a novel function of SIRT7 on chromatin organization by mediating the anchoring of L1 to the nuclear envelope, and a new functional link of the nuclear lamina with transcriptional repression.


Subject(s)
Genome , Lamin Type A/genetics , Long Interspersed Nucleotide Elements , Sirtuins/genetics , Transcription, Genetic , Animals , Cell Line , Cell Line, Tumor , Chromatin Immunoprecipitation , Epigenesis, Genetic , Fibroblasts/cytology , Fibroblasts/metabolism , Heterochromatin/chemistry , Heterochromatin/metabolism , Histones/genetics , Histones/metabolism , Humans , K562 Cells , Lamin Type A/metabolism , Liver/cytology , Liver/metabolism , Male , Mice , Mice, Knockout , Myocardium/cytology , Myocardium/metabolism , Nuclear Lamina/metabolism , Nuclear Lamina/ultrastructure , Sirtuins/deficiency , Sirtuins/metabolism , Testis/cytology , Testis/metabolism
2.
Cell Rep ; 21(8): 2048-2057, 2017 Nov 21.
Article in English | MEDLINE | ID: mdl-29166597

ABSTRACT

HP1 is a structural component of heterochromatin. Mammalian HP1 isoforms HP1α, HP1ß, and HP1γ play different roles in genome stability, but their precise role in heterochromatin structure is unclear. Analysis of Hp1α-/-, Hp1ß-/-, and Hp1γ-/- MEFs show that HP1 proteins have both redundant and unique functions within pericentric heterochromatin (PCH) and also act globally throughout the genome. HP1α confines H4K20me3 and H3K27me3 to regions within PCH, while its absence results in a global hyper-compaction of chromatin associated with a specific pattern of mitotic defects. In contrast, HP1ß is functionally associated with Suv4-20h2 and H4K20me3, and its loss induces global chromatin decompaction and an abnormal enrichment of CTCF in PCH and other genomic regions. Our work provides insight into the roles of HP1 proteins in heterochromatin structure and genome stability.


Subject(s)
Chromosomal Proteins, Non-Histone/genetics , Chromosomal Proteins, Non-Histone/metabolism , Amino Acid Sequence/genetics , Animals , Chromatin/metabolism , Chromobox Protein Homolog 5 , HeLa Cells , Humans , Mammals/metabolism , Protein Binding/genetics , Protein Binding/immunology , Protein Isoforms/genetics , Protein Isoforms/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism
3.
Epigenetics ; 12(2): 166-175, 2017 02.
Article in English | MEDLINE | ID: mdl-28059589

ABSTRACT

The presence of H3K9me3 and heterochromatin protein 1 (HP1) are hallmarks of heterochromatin conserved in eukaryotes. The spreading and maintenance of H3K9me3 is effected by the functional interplay between the H3K9me3-specific histone methyltransferase Suv39h1 and HP1. This interplay is complex in mammals because the three HP1 isoforms, HP1α, ß, and γ, are thought to play a redundant role in Suv39h1-dependent deposition of H3K9me3 in pericentric heterochromatin (PCH). Here, we demonstrate that despite this redundancy, HP1α and, to a lesser extent, HP1γ have a closer functional link to Suv39h1, compared to HP1ß. HP1α and γ preferentially interact in vivo with Suv39h1, regulate its dynamics in heterochromatin, and increase Suv39h1 protein stability through an inhibition of MDM2-dependent Suv39h1-K87 polyubiquitination. The reverse is also observed, where Suv39h1 increases HP1α stability compared HP1ß and γ. The interplay between Suv39h1 and HP1 isoforms appears to be relevant under genotoxic stress. Specifically, loss of HP1α and γ isoforms inhibits the upregulation of Suv39h1 and H3K9me3 that is observed under stress conditions. Reciprocally, Suv39h1 deficiency abrogates stress-dependent upregulation of HP1α and γ, and enhances HP1ß levels. Our work defines a specific role for HP1 isoforms in regulating Suv39h1 function under stress via a feedback mechanism that likely regulates heterochromatin formation.


Subject(s)
Chromosomal Proteins, Non-Histone/metabolism , DNA Damage , Feedback, Physiological , Methyltransferases/genetics , Repressor Proteins/genetics , Cell Line , Chromatin Assembly and Disassembly , Chromobox Protein Homolog 5 , Chromosomal Proteins, Non-Histone/genetics , Histones/metabolism , Humans , Methyltransferases/metabolism , Protein Binding , Protein Isoforms/genetics , Protein Isoforms/metabolism , Protein Stability , Repressor Proteins/metabolism , Ubiquitination
4.
EMBO J ; 35(14): 1488-503, 2016 07 15.
Article in English | MEDLINE | ID: mdl-27225932

ABSTRACT

Sirtuins, a family of protein deacetylases, promote cellular homeostasis by mediating communication between cells and environment. The enzymatic activity of the mammalian sirtuin SIRT7 targets acetylated lysine in the N-terminal tail of histone H3 (H3K18Ac), thus modulating chromatin structure and transcriptional competency. SIRT7 deletion is associated with reduced lifespan in mice through unknown mechanisms. Here, we show that SirT7-knockout mice suffer from partial embryonic lethality and a progeroid-like phenotype. Consistently, SIRT7-deficient cells display increased replication stress and impaired DNA repair. SIRT7 is recruited in a PARP1-dependent manner to sites of DNA damage, where it modulates H3K18Ac levels. H3K18Ac in turn affects recruitment of the damage response factor 53BP1 to DNA double-strand breaks (DSBs), thereby influencing the efficiency of non-homologous end joining (NHEJ). These results reveal a direct role for SIRT7 in DSB repair and establish a functional link between SIRT7-mediated H3K18 deacetylation and the maintenance of genome integrity.


Subject(s)
DNA Damage , DNA End-Joining Repair , DNA/metabolism , Sirtuins/metabolism , Animals , Mice, Inbred C57BL , Mice, Knockout
5.
Prostate ; 74(15): 1551-9, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25175909

ABSTRACT

BACKGROUND: Biomarkers predicting tumor response are important to emerging targeted therapeutics. Complimentary methods to assess and understand genetic changes and heterogeneity within only few cancer cells in tissue will be a valuable addition for assessment of tumors such as prostate cancer that often have insufficient tumor for next generation sequencing in a single biopsy core. METHODS: Using confocal microscopy to identify cell-to-cell relationships in situ, we studied the most common gene rearrangement in prostate cancer (TMPRSS2 and ERG) and the tumor suppressor CHD1 in 56 patients who underwent radical prostatectomy. RESULTS: Wild type ERG was found in 22 of 56 patients; ERG copy number was increased in 10/56, and ERG rearrangements confirmed in 24/56 patients. In 24 patients with ERG rearrangements, the mechanisms of rearrangement were heterogeneous, with deletion in 14/24, a split event in 7/24, and both deletions and split events in the same tumor focus in 3/24 patients. Overall, 14/45 (31.1%) of patients had CHD1 deletion, with the majority of patients with CHD1 deletions (13/14) correlating with ERG-rearrangement negative status (P < 0.001). CONCLUSIONS: These results demonstrate the ability of confocal microscopy and FISH to identify the cell-to-cell differences in common gene fusions such as TMPRSS2-ERG that may arise independently within the same tumor focus. These data support the need to study complimentary approaches to assess genetic changes that may stratify therapy based on predicted sensitivities.


Subject(s)
DNA Helicases/genetics , DNA-Binding Proteins/genetics , Prostate/pathology , Prostatic Neoplasms/genetics , Serine Endopeptidases/genetics , Trans-Activators/genetics , Aged , Gene Expression Profiling , Gene Rearrangement , Genetic Heterogeneity , Humans , In Situ Hybridization, Fluorescence , Male , Microscopy, Confocal , Middle Aged , Prostatic Neoplasms/pathology , Transcriptional Regulator ERG
6.
Genes Dev ; 27(6): 639-53, 2013 Mar 15.
Article in English | MEDLINE | ID: mdl-23468428

ABSTRACT

The establishment of the epigenetic mark H4K20me1 (monomethylation of H4K20) by PR-Set7 during G2/M directly impacts S-phase progression and genome stability. However, the mechanisms involved in the regulation of this event are not well understood. Here we show that SirT2 regulates H4K20me1 deposition through the deacetylation of H4K16Ac (acetylation of H4K16) and determines the levels of H4K20me2/3 throughout the cell cycle. SirT2 binds and deacetylates PR-Set7 at K90, modulating its chromatin localization. Consistently, SirT2 depletion significantly reduces PR-Set7 chromatin levels, alters the size and number of PR-Set7 foci, and decreases the overall mitotic deposition of H4K20me1. Upon stress, the interaction between SirT2 and PR-Set7 increases along with the H4K20me1 levels, suggesting a novel mitotic checkpoint mechanism. SirT2 loss in mice induces significant defects associated with defective H4K20me1-3 levels. Accordingly, SirT2-deficient animals exhibit genomic instability and chromosomal aberrations and are prone to tumorigenesis. Our studies suggest that the dynamic cross-talk between the environment and the genome during mitosis determines the fate of the subsequent cell cycle.


Subject(s)
Cell Cycle/physiology , Genomic Instability , Sirtuin 2/metabolism , Acetylation , Amino Acid Sequence , Animals , Cell Transformation, Neoplastic/genetics , Chromatin/metabolism , DNA Damage/genetics , Gene Knockout Techniques , HeLa Cells , Histone-Lysine N-Methyltransferase/metabolism , Histones/metabolism , Humans , M Phase Cell Cycle Checkpoints/physiology , Methylation , Mice , Mice, Knockout , Mitosis , Protein Binding , Sirtuin 2/genetics
7.
Behav Brain Res ; 223(1): 233-8, 2011 Sep 30.
Article in English | MEDLINE | ID: mdl-21545813

ABSTRACT

Since zinc transporter ZnT3 is localized to the hippocampus and perirhinal cortex, we used ZnT3 knockout mice (KO) to analyze the role of ZnT3 in memory and behavior dependent on these brain regions. ZnT3KO mice were normal in initial learning in the standard water maze but had difficulty finding a second platform location. The mutants showed increased social interaction but were deficient in social and object recognition memory. These data suggest that ZnT3 is involved in certain types of spatial memory and behavior dependent on the hippocampus and perirhinal cortex.


Subject(s)
Carrier Proteins/physiology , Hippocampus/physiology , Membrane Proteins/physiology , Memory/physiology , Temporal Lobe/physiology , Animals , Carrier Proteins/genetics , Cation Transport Proteins , Interpersonal Relations , Male , Maze Learning/physiology , Membrane Proteins/genetics , Membrane Transport Proteins , Mice , Mice, Inbred C57BL , Mice, Knockout
8.
Mol Cell ; 42(2): 210-23, 2011 Apr 22.
Article in English | MEDLINE | ID: mdl-21504832

ABSTRACT

Sirtuins are NAD-dependent deacetylases that sense oxidative stress conditions and promote a protective cellular response. The Sirtuin SirT1 is involved in facultative heterochromatin formation through an intimate functional relationship with the H3K9me3 methyltransferase Suv39h1, a chromatin organization protein. However, SirT1 also regulates Suv39h1-dependent constitutive heterochromatin (CH) through an unknown mechanism; interestingly, SirT1 does not significantly localize in these regions. Herein, we report that SirT1 controls global levels of Suv39h1 by increasing its half-life through inhibition of Suv39h1 lysine 87 polyubiquitination by the E3-ubiquitin ligase MDM2. This in turn increases Suv39h1 turnover in CH and ensures genome integrity. Stress conditions that lead to SirT1 upregulation, such as calorie restriction, also induce higher levels of Suv39h1 in a SirT1-dependent manner in vivo. These observations reflect a direct link between oxidative stress response and Suv39h1 and support a dynamic view of heterochromatin, in which its structure adapts to cell physiology.


Subject(s)
Chromatin Assembly and Disassembly , Genomic Instability , Heterochromatin/metabolism , Methyltransferases/metabolism , Oxidative Stress , Repressor Proteins/metabolism , Sirtuin 1/metabolism , Amino Acid Sequence , Animals , Caloric Restriction , Enzyme Stability , HEK293 Cells , Half-Life , HeLa Cells , Humans , Lysine , Male , Methyltransferases/genetics , Mice , Molecular Sequence Data , Mutation , NIH 3T3 Cells , Polyubiquitin/metabolism , Proto-Oncogene Proteins c-mdm2/metabolism , RNA Interference , Rats , Rats, Sprague-Dawley , Recombinant Fusion Proteins/metabolism , Repressor Proteins/genetics , Sirtuin 1/genetics , Time Factors , Transfection , Ubiquitination
9.
J Neurotrauma ; 28(6): 1063-76, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21443453

ABSTRACT

RhoA is a key regulator of the actin cytoskeleton that is upregulated after spinal cord injury (SCI). We analyzed different methods for siRNA delivery and developed siRNAs targeting RhoA (siRhoA) for SCI treatment. Cy 3.5-labeled siRNA delivered at the time of SCI yielded fluorescence in several cell types in the injury site. Intraspinal injections of chemically stabilized siRhoA into the spinal cord of injured rats reduced RhoA protein levels after 1 week and improved hindlimb walking over 6 weeks. To explore a less invasive route, we tested intrathecal injection of Cy 3.5-labeled siRNA via lumbar puncture 1 day after SCI, which resulted in robust uptake in the T9-T10 injury site. Lumbar injection of siRhoA 1 day after SCI reduced RhoA mRNA and protein levels 3 days after injection. Although siRhoA treatment did not yield significant improvement in locomotion, it decreased tactile hypersensitivity significantly compared to controls. Histological analysis at 8 weeks showed significant improvement in white matter sparing with siRhoA compared to control siRNA. siRhoA treatment also resulted in less accumulation of ED1+macrophages, increased PKC-γ immunoreactivity in the corticospinal tract rostral to the injury site, and increased serotonergic fiber growth 12 mm caudal to the contusion site. The ability of siRhoA to preserve white matter and promote serotonergic axonal regrowth caudal to the injury site is likely to suppress allodynia. This provides justification for considering clinical development of RhoA inhibitors to treat SCI sub-acutely to reduce allodynia, which occurs frequently in SCI patients.


Subject(s)
Genetic Therapy/methods , Hyperalgesia/therapy , RNA, Small Interfering/administration & dosage , Serotonin/physiology , Spinal Cord Injuries/therapy , rhoA GTP-Binding Protein/administration & dosage , Animals , Disease Models, Animal , Female , Hyperalgesia/genetics , Injections, Spinal , Nerve Regeneration/genetics , RNA, Small Interfering/genetics , Rats , Rats, Sprague-Dawley , Spinal Cord Injuries/genetics , Up-Regulation/physiology , rhoA GTP-Binding Protein/antagonists & inhibitors , rhoA GTP-Binding Protein/genetics
10.
Exp Neurol ; 229(1): 72-9, 2011 May.
Article in English | MEDLINE | ID: mdl-20850435

ABSTRACT

Olfactory ensheathing cells (OEC), which normally associate closely with but do not myelinate axons in situ, myelinate axons in the adult mammalian spinal cord. They are of clinical interest as candidate cells for autologous transplantation but the ability of OEC to myelinate axons in vitro has been controversial. To clarify this issue, we isolated OEC from olfactory bulbs (OB) of juvenile and adult rats expressing GFP and analyzed their ability to myelinate axons. Using a well-defined assay for myelination of dorsal root ganglia (DRG) axons in culture, we found that OEC from juvenile pups associated with and then myelinated DRG axons. OEC assembled into bundles with the axons by 1week and required more than a week before myelination on axons was detected. In contrast, rat Schwann cells did not bundle axons and they formed P0(+) and MBP(+) myelin segments after as little as 1week. Most of the OEC in culture exhibited staining for calponin, a marker that was not found on Schwann cells in culture, whereas in both OEC and Schwann cell populations nearly all cells were positive for p75NTR and GFAP. These results confirm previous reports showing only subtle immunological differences between Schwann cells and OEC. Besides differences in the rate of myelination, we detected two additional functional differences in the interactions of OEC and Schwann cells with DRG axons. First, the diameter of OEC generated myelin was greater than for Schwann cell myelin on DRG axons. Second, OEC but not Schwann cells myelinated DRG axons in the absence of vitamin C. OEC isolated from adult OB were also found to bundle and myelinate DRG axons but the latter occurred only after incubation times of at least 3weeks. The results indicate that adult OEC require longer incubation times than juvenile OEC to myelinate axons and suggest that patterns of myelination by OEC and Schwann cells are distinguishable at least on axons in vitro. This article is part of a Special Issue entitled: Understanding olfactory ensheathing glia and their prospect for nervous system repair.


Subject(s)
Axons/physiology , Ganglia, Spinal/physiology , Myelin Sheath/physiology , Nerve Fibers, Myelinated/physiology , Olfactory Bulb/physiology , Age Factors , Animals , Animals, Newborn , Cells, Cultured , Ganglia, Spinal/cytology , Olfactory Bulb/cytology , Rats , Rats, Sprague-Dawley
11.
J Neurotrauma ; 26(7): 979-93, 2009 Jul.
Article in English | MEDLINE | ID: mdl-19257808

ABSTRACT

Cell-based therapy has been widely evaluated in spinal cord injury (SCI) animal models and shown to improve functional recovery. However, host response to cell transplants at gene expression level is rarely discussed. We reported previously that acute transplantation of radial glial cells RG3.6 following SCI promoted early locomotion improvement within 1 week post-injury. To identify rapid molecular changes induced by RG3.6 transplantation in the host tissue, distal spinal cord segments were subjected to microarray analysis. Although RG3.6 transplantation, reduced activity of macrophages as early as 1-2 weeks post-injury, the expression levels of inflammatory genes (e.g., IL-6, MIP-2, MCP-1) were not decreased by RG3.6 treatment as compared to medium or other cell controls at 6-12 h post-injury. However, genes associated with tissue protection (Hsp70 and Hsp32) and neural cell development (Foxg1, Top2a, Sox11, Nkx2.2, Vimentin) were found to be significantly up-regulated by RG3.6 transplants. Foxg1 was the most highly induced gene in the RG3.6-treated spinal cords, and its expression by immunocytochemistry was confirmed in the host tissue. Moreover, RG3.6 treatment boosted the number of Nkx2.2 cells in the spinal cord, and these cells frequently co-expressed NG2, which marks progenitor cells. Taken together, these results demonstrate that radial glial transplants induced rapid and specific gene expression in the injured host tissue, and suggest that these early responses are associated with mechanisms of tissue protection and activation of endogenous neural progenitor cells.


Subject(s)
Neuroglia/transplantation , Spinal Cord Injuries/genetics , Spinal Cord Injuries/therapy , Spinal Cord/metabolism , Analysis of Variance , Animals , Female , Fluorescent Antibody Technique , Gene Expression , Gene Expression Profiling , Homeobox Protein Nkx-2.2 , Laminectomy , Neurogenesis/genetics , Oligonucleotide Array Sequence Analysis , Rats , Rats, Sprague-Dawley , Reverse Transcriptase Polymerase Chain Reaction , Spinal Cord Injuries/metabolism
12.
J Neurotrauma ; 25(4): 334-49, 2008 Apr.
Article in English | MEDLINE | ID: mdl-18373483

ABSTRACT

Chondroitin sulfate proteoglycans (CSPGs) inhibit axonal growth, and treatment with chondroitinase ABC promotes axonal regeneration in some models of central nervous system (CNS) injury. The aims of this study were (1) to compare the spatiotemporal appearance of CSPG expression between spinal cord contusion and hemisection models, and (2) to evaluate chondroitinase treatment effects on axonal regrowth in the two injury models. After hemisection, CSPG-immunoreactivity (IR) in the injury site rose to peak levels at 18 days but then decreased dramatically by 49 days; in contrast, CSPG-IR remained high for at least 49 days after contusion. After hemisection, many anterogradely labeled corticospinal tract (CST) axons remained close to CSPG-rich lesion sites, but after contusion, most CST axons retracted by approximately 1 mm rostral from the rostral-most CSPG-rich cyst. Intraspinal injection of chondroitinase at 0, 1, 2, and 4 weeks following injury dramatically reduced CSPG-IR in both injury models within 4 days, and CSPG-IR remained low for at least 3 weeks. After the chondroitinase treatment, many axons grew around the lesion site in hemisected spinal cords but not in contused spinal cords. We propose that improved axonal growth in hemisected spinal cords is due to decreased inhibition resulting from degradation of CSPGs located adjacent to severed CST axons. However, in spinal cord contusions, retracted CST axons fail to grow across gliotic regions that surround CSPG-rich injury sites despite efficient degradation with chondroitinase, suggesting that other inhibitors of axonal growth persist in the gliotic regions.


Subject(s)
Axons/physiology , Chondroitin ABC Lyase/administration & dosage , Chondroitin Sulfates/metabolism , Nerve Regeneration/physiology , Pyramidal Tracts/physiopathology , Spinal Cord Injuries/drug therapy , Animals , Dose-Response Relationship, Drug , Female , Injections, Spinal , Pyramidal Tracts/metabolism , Pyramidal Tracts/pathology , Rats , Rats, Sprague-Dawley , Spinal Cord Injuries/metabolism , Spinal Cord Injuries/physiopathology , Time Factors
13.
Dev Biol ; 311(2): 408-22, 2007 Nov 15.
Article in English | MEDLINE | ID: mdl-17936266

ABSTRACT

The cell adhesion molecule neurofascin (NF) has a major neuronal isoform (NF186) containing a mucin-like domain followed by a fifth fibronectin type III repeat while these domains are absent from glial NF155. Neuronal NF isoforms lacking one or both of these domains are expressed transiently in embryonic dorsal root ganglia (DRG). These two domains are co-expressed in mature NF186, which peaks in expression prior to birth and then persists almost exclusively at nodes of Ranvier on myelinated axons. In contrast, glial NF155 is only detected postnatally with the onset of myelination. All these forms of NF bound homophilically and to Schwann cells but only the mature NF186 isoform inhibits cell adhesion, and this activity may be important in formation of the node of Ranvier. Schwann cells deficient in NF155 myelinated DRG axons in a delayed manner and they showed significantly decreased clustering of both NF and Caspr in regions where paranodes normally form. The combined results suggest that NF186 is expressed prenatally on DRG neurons and it may modulate their adhesive interactions with Schwann cells, which express NF155 postnatally and require it for development of axon-glial paranodal junctions.


Subject(s)
Alternative Splicing , Cell Adhesion Molecules/metabolism , Ganglia, Spinal , Nerve Growth Factors/metabolism , Neuroglia/metabolism , Neurons/metabolism , Protein Isoforms/metabolism , Animals , Cell Adhesion/physiology , Cell Adhesion Molecules/genetics , Cell Adhesion Molecules, Neuronal/genetics , Cell Adhesion Molecules, Neuronal/metabolism , Cells, Cultured , Coculture Techniques , Embryo, Mammalian/anatomy & histology , Embryo, Mammalian/physiology , Fibronectins/genetics , Fibronectins/metabolism , Ganglia, Spinal/cytology , Ganglia, Spinal/embryology , Ganglia, Spinal/growth & development , Humans , Myelin Sheath/physiology , Nerve Growth Factors/genetics , Neuroglia/cytology , Neurons/cytology , Protein Isoforms/genetics , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Rats , Schwann Cells/cytology , Schwann Cells/metabolism
14.
Dev Biol ; 293(1): 1-12, 2006 May 01.
Article in English | MEDLINE | ID: mdl-16566914

ABSTRACT

The Ig cell adhesion molecules (CAM) neurofascin (NF) and Nr-CAM are localized at developing nodes of Ranvier in peripheral myelinated axons prior to clustering of Na+ channels. Different isoforms of NF are expressed on neurons and glia, and NF binding on both cells has been suggested to play roles in node and paranode formation. To clarify the role of NF further, we analyzed effects of NF-Fc fusion proteins in Schwann cell-DRG neuron myelinating cocultures. NF-Fc significantly inhibited nodal clustering of Na+ channels, ankyrin G, and betaIV spectrin, and modestly reduced Caspr clustering at paranodal junctions; it did not significantly affect lengths or numbers of myelin-positive segments, axon initial segments, or accumulations of phosphorylated-ERM proteins in Schwann cell nodal microvilli. NF-Fc binds to Schwann cells but little or no binding to DRG neurons was detected. The results suggest a critical early role for axonal NF in clustering of Na+ channels at nodes of Ranvier via interactions with receptors on Schwann cells.


Subject(s)
Ankyrins/metabolism , Cell Adhesion Molecules/physiology , Nerve Growth Factors/physiology , Nerve Tissue Proteins/metabolism , Ranvier's Nodes/metabolism , Sodium Channels/metabolism , Spectrin/metabolism , Animals , Axons/metabolism , Axons/physiology , Cell Adhesion Molecules/biosynthesis , Cell Communication/physiology , Coculture Techniques , Growth Inhibitors/genetics , Growth Inhibitors/physiology , Nerve Growth Factors/biosynthesis , Rats , Receptors, Fc/genetics , Recombinant Fusion Proteins/biosynthesis , Schwann Cells/metabolism
15.
Mol Cell Neurosci ; 30(1): 137-48, 2005 Sep.
Article in English | MEDLINE | ID: mdl-16061393

ABSTRACT

Neurofascin (NF) is a neural cell adhesion molecule in the L1-family containing six Ig domains and multiple fibronectin type III (FnIII) repeats in its extracellular region. NF has many splicing variants and two of these are exemplars that have different cellular patterns of expression during development. NF186, which is expressed on neurons, contains an unusual mucin-like region and NF155, which is expressed on glia, contains a unique FnIII repeat with an RGD motif. Analysis of Fc fusion proteins representing different extracellular regions of NF indicate that NF186 inhibits cell adhesion and neurite outgrowth, and the inhibition is associated with the region containing the mucin-like domain. NF155 promotes neural cell adhesion and neurite outgrowth, and the RGD motif in its third FnIII repeat is critical for cell spreading and neurite outgrowth. The results suggest that different splicing variants of NF expressed on neurons and glia play distinct roles during neural development.


Subject(s)
Cell Adhesion Molecules/genetics , Cell Adhesion Molecules/metabolism , Cell Adhesion/physiology , Nerve Growth Factors/genetics , Nerve Growth Factors/metabolism , Neurites/physiology , Alternative Splicing , Cell Adhesion Molecules/chemistry , Cell Line , Humans , Kidney/cytology , Mutagenesis , Nerve Growth Factors/chemistry , Neuroglia/cytology , Neuroglia/physiology , Neurons/physiology , Neurons/ultrastructure , Protein Structure, Tertiary , Ranvier's Nodes/physiology
16.
Exp Neurol ; 193(2): 394-410, 2005 Jun.
Article in English | MEDLINE | ID: mdl-15869942

ABSTRACT

Radial glial cells are neural stem cells (NSC) that are transiently found in the developing CNS. To study radial glia, we isolated clones following immortalization of E13.5 GFP rat neurospheres with v-myc. Clone RG3.6 exhibits polarized morphology and expresses the radial glial markers nestin and brain lipid binding protein. Both NSC and RG3.6 cells migrated extensively in the adult spinal cord. However, RG3.6 cells differentiated into astroglia slower than NSC, suggesting that immortalization can delay differentiation of radial glia. Following spinal cord contusion, implanted RG3.6 cells migrated widely in the contusion site and into spared white matter where they exhibited a highly polarized morphology. When injected immediately after injury, RG3.6 cells formed cellular bridges surrounding spinal cord lesion sites and extending into spared white matter regions in contrast to GFP fibroblasts that remained in the lesion site. Behavioral analysis indicated higher BBB scores in rats injected with RG3.6 cells than rats injected with fibroblasts or medium as early as 1 week after injury. Spinal cords transplanted with RG3.6 cells or dermal fibroblasts exhibited little accumulation of chondroitin sulfate proteoglycans (CSPG) including NG2 proteoglycans that are known to inhibit axonal growth. Reduced levels of CSPG were accompanied by little accumulation in the injury site of activated macrophages, which are a major source of CSPG. However, increased staining and organization of neurofilaments were found in injured rats transplanted with RG3.6 cells suggesting neuroprotection or regrowth. The combined results indicate that acutely transplanted radial glia can migrate to form bridges across spinal cord lesions in vivo and promote functional recovery following spinal cord injury by protecting against macrophages and secondary damage.


Subject(s)
Neuroglia/cytology , Recovery of Function/physiology , Spinal Cord Injuries/surgery , Stem Cell Transplantation , Stem Cells/physiology , Animals , Antigens/metabolism , Behavior, Animal , Carrier Proteins/metabolism , Cell Differentiation/physiology , Cells, Cultured , Chondroitin Sulfates/metabolism , Clone Cells/physiology , Ectodysplasins , Embryo, Mammalian , Fatty Acid-Binding Protein 7 , Fatty Acid-Binding Proteins , Female , Fibroblasts/physiology , Fibroblasts/transplantation , Fluorescent Antibody Technique/methods , Glial Fibrillary Acidic Protein/metabolism , Green Fluorescent Proteins/metabolism , Indoles/metabolism , Intermediate Filament Proteins/metabolism , Male , Membrane Proteins/metabolism , Motor Activity/physiology , Nerve Tissue Proteins/metabolism , Nestin , Neurofilament Proteins/metabolism , Prosencephalon/cytology , Prosencephalon/embryology , Proteoglycans/metabolism , Rats , Rats, Sprague-Dawley , Time Factors , Tubulin/metabolism
17.
Dev Biol ; 271(2): 225-38, 2004 Jul 15.
Article in English | MEDLINE | ID: mdl-15223331

ABSTRACT

Radial glia are among the first cells that develop in the embryonic central nervous system. They are progenitors of glia and neurons but their relationship with restricted precursors that are also derived from neuroepithelia is unclear. To clarify this issue, we analyzed expression of cell type specific markers (BLBP for radial glia, 5A5/E-NCAM for neuronal precursors and A2B5 for glial precursors) on cortical radial glia in vivo and their progeny in vitro. Clones of cortical cells initially expressing only BLBP gave rise to cells that were A2B5+ and eventually lost BLBP expression in vitro. BLBP is expressed in the rat neuroepithelium as early as E12.5 when there is little or no staining for A2B5 and 5A5. In E13.5-15.5 forebrain, A2B5 is spatially restricted co-localizing with a subset of the BLBP+ radial glia. Analysis of cells isolated acutely from embryonic cortices confirmed that BLBP expression could appear without, or together with, A2B5 or 5A5. The numbers of BLBP+/5A5+ cells decreased during neurogenesis while the numbers of BLBP+/A2B5+ cells remained high through the beginning of gliogenesis. The combined results demonstrate that spatially restricted subpopulations of radial glia along the dorsal-ventral axis acquire different markers for neuronal or glial precursors during CNS development.


Subject(s)
Cell Differentiation/physiology , Central Nervous System/embryology , Gene Expression Regulation, Developmental , Neuroglia/physiology , Neurons/physiology , Animals , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal/metabolism , Biomarkers , Carrier Proteins/immunology , Carrier Proteins/metabolism , Cells, Cultured , Central Nervous System/cytology , Fluorescent Antibody Technique , Neural Cell Adhesion Molecules/immunology , Neural Cell Adhesion Molecules/metabolism , Neuroglia/metabolism , Neurons/metabolism , Rats , Time Factors , Tumor Suppressor Proteins/immunology , Tumor Suppressor Proteins/metabolism
18.
J Neurobiol ; 51(3): 177-89, 2002 Jun 05.
Article in English | MEDLINE | ID: mdl-11984840

ABSTRACT

The neural cell adhesion molecule (CAM) L1 is a member of the immunoglobulin superfamily that has been implicated in neuronal adhesion, neurite outgrowth, and axon guidance. The clinical importance of L1 is illustrated by pathological mutations that lead to hydrocephalus, mental retardation, motor defects, and early mortality. The L1 gene is composed of 28 exons, including exons 2 and 27 that are spliced alternatively, and mutations in exon 2 are associated with severe neurological abnormalities in humans. To elucidate the role of L1 exon 2, a recombinant Fc fusion protein called Delta2L1 was constructed lacking the second exon in the extracellular domain of L1. When bound to fluorescent beads, L1 exhibited homophilic binding while Delta2L1 did not. However, L1 beads coaggregated with the Delta2L1 beads. Similarly, in cell binding studies, L1 bound to L1 and Delta2L1 did not bind to Delta2L1 but it bound moderately to L1. Given the reduced binding of Delta2L1, we tested its effect on neurons. By comparison to L1, a lower percentage of dissociated neurons extended neurites on Delta2L1, and there was a modest decrease in the length of the neurites that grew. Neurite outgrowth from reaggregated neurons was much less robust on Delta2L1 than on L1. The combined results indicate that Delta2L1 does not bind homophilically but it can interact with L1 containing exon 2. The reduced binding and neurite promoting activity of Delta2L1 provides an explanation for certain pathological mutations in L1 that lead to clinically apparent disease in the absence of the normal form of L1 in the nervous system.


Subject(s)
Alternative Splicing/physiology , Membrane Glycoproteins/genetics , Membrane Glycoproteins/metabolism , Neural Cell Adhesion Molecules/genetics , Neural Cell Adhesion Molecules/metabolism , Neurites/physiology , Animals , Cell Line , Cerebellum/cytology , Exons/genetics , Gene Expression , Genetic Vectors , Humans , Hydrocephalus/genetics , Kidney/cytology , Leukocyte L1 Antigen Complex , Mice , Microspheres , Neurons/metabolism , Neurons/ultrastructure , Protein Binding/genetics , Sindbis Virus/genetics , Transfection , X Chromosome
SELECTION OF CITATIONS
SEARCH DETAIL