Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters










Publication year range
1.
Invest Ophthalmol Vis Sci ; 64(14): 8, 2023 11 01.
Article in English | MEDLINE | ID: mdl-37934159

ABSTRACT

Purpose: The proliferative and neurogenic potential of retinal Müller glia after injury varies widely across species. To identify the endogenous mechanisms regulating the proliferative response of mammalian Müller glia, we comparatively analyzed the expression and function of nestin, an intermediate filament protein established as a neural stem cell marker, in the mouse and rat retinas after injury. Methods: Nestin expression in the retinas of C57BL/6 mice and Wistar rats after methyl methanesulfonate (MMS)-induced photoreceptor injury was examined by immunofluorescence and Western blotting. Adeno-associated virus (AAV)-delivered control and nestin short hairpin RNA (shRNA) were intravitreally injected to rats and Müller glia proliferation after MMS-induced injury was analyzed by BrdU incorporation and immunofluorescence. Photoreceptor removal and microglia/macrophage infiltration were also analyzed by immunofluorescence. Results: Rat Müller glia re-entered the cell cycle and robustly upregulated nestin after injury whereas Müller glia proliferation and nestin upregulation were not observed in mice. In vivo knockdown of nestin in the rat retinas inhibited Müller glia proliferation while transiently stimulating microglia/macrophage infiltration and phagocytic removal of dead photoreceptors. Conclusions: Our findings suggest a critical role for nestin in the regulation of Müller glia proliferation after retinal injury and highlight the importance of cross species analysis to identify the molecular mechanisms regulating the injury responses of the mammalian retina.


Subject(s)
Eye Injuries , Nestin , Neuroglia , Animals , Mice , Rats , Cell Proliferation , Methyl Methanesulfonate , Mice, Inbred C57BL , Rats, Wistar , Neuroglia/cytology
2.
Sci Rep ; 10(1): 1488, 2020 01 30.
Article in English | MEDLINE | ID: mdl-32001733

ABSTRACT

Müller glia, the principal glial cell type in the retina, have the potential to reenter the cell cycle after retinal injury. In mammals, proliferation of Müller glia is followed by gliosis, but not regeneration of neurons. Retinal injury is also accompanied by phagocytic removal of degenerated cells. We here investigated the possibility that proliferation and gliosis of Müller glia and phagocytosis of degenerated cells may be regulated by the same molecular pathways. After N-methyl-N-nitrosourea-induced retinal injury, degenerated photoreceptors were eliminated prior to the infiltration of microglia/macrophages into the outer nuclear layer, almost in parallel with cell cycle reentry of Müller glia. Inhibition of microglia/macrophage activation with minocycline did not affect the photoreceptor clearance. Accumulation of lysosomes and rhodopsin-positive photoreceptor debris within the cytoplasm of Müller glia indicated that Müller glia phagocytosed most photoreceptor debris. Pharmacological inhibition of phosphatidylserine and Rac1, key regulators of the phagocytic pathway, prevented cell cycle reentry, migration, upregulation of glial fibrillary acidic protein, and phagocytic activity of Müller glia. These data provide evidence that phosphatidylserine and Rac1 may contribute to the crosstalk between different signaling pathways activated in Müller glia after injury.


Subject(s)
Phosphatidylserines/metabolism , Retina/injuries , rac1 GTP-Binding Protein/metabolism , Animals , Cell Cycle , Cell Line , Cell Proliferation , Disease Models, Animal , Ependymoglial Cells/metabolism , Ependymoglial Cells/pathology , Gliosis/metabolism , Gliosis/pathology , Male , Phagocytosis , Photoreceptor Cells, Vertebrate/metabolism , Photoreceptor Cells, Vertebrate/pathology , Rats , Rats, Wistar , Retina/metabolism , Retina/pathology , Signal Transduction
3.
Neurosci Res ; 139: 58-62, 2019 Feb.
Article in English | MEDLINE | ID: mdl-30194028

ABSTRACT

Pyridoxal, an active form of vitamin B6, is known to inhibit formation of advanced glycation end-products and protect tissues from diabetic complications. Here we identified that pyridoxal is a required component for establishing Schwann cell myelination in our Schwann cell-dorsal root ganglion neuron co-culture system. When the co-culture was maintained without pyridoxal, carboxymethylation of collapsin response mediator protein 2 (CRMP2) became detectable. Carboxymethylation decreased the affinity of CRMP2 to bind with microtubules, indicating that carboxymethylation affected CRMP2 function. These results suggest that carboxymethylation of CRMP2 may be an indicator of dysfunction caused by glycation which is observed in pathological conditions, including diabetic neuropathy.


Subject(s)
Ganglia, Spinal/metabolism , Myelin Sheath/pathology , Neurons/metabolism , Schwann Cells/metabolism , Animals , Diabetic Neuropathies/metabolism , Diabetic Neuropathies/pathology , Humans , Intercellular Signaling Peptides and Proteins/metabolism , Nerve Tissue Proteins/metabolism
4.
Neural Dev ; 12(1): 17, 2017 Sep 20.
Article in English | MEDLINE | ID: mdl-28931408

ABSTRACT

BACKGROUND: Cyclin-dependent kinase (CDK) inhibitors play an important role in regulating cell cycle progression, cell cycle exit and cell differentiation. p27KIP1 (p27), one of the major CDK inhibitors in the retina, has been shown to control the timing of cell cycle exit of retinal progenitors. However, the precise role of this protein in retinal development remains largely unexplored. We thus analyzed p27-deficient mice to characterize the effects of p27 loss on proliferation, differentiation, and survival of retinal cells. METHODS: Expression of p27 in the developing and mature mouse retina was analyzed by immunohistochemistry using antibodies against p27 and cell type-specific markers. Cell proliferation and differentiation were examined in the wild-type and p27-deficient retinas by immunohistochemistry using various cell cycle and differentiation markers. RESULTS: All postmitotic retinal cell types expressed p27 in the mouse retinas. p27 loss caused extension of the period of proliferation in the developing retinas. This extra proliferation was mainly due to ectopic cell cycle reentry of differentiating cells including bipolar cells, Müller glial cells and cones, rather than persistent division of progenitors as previously suggested. Aberrant cell cycle activity of cones was followed by cone death resulting in a significant reduction in cone number in the mature p27-deficient retinas. CONCLUSIONS: Although expressed in all retinal cell types, p27 is required to maintain the quiescence of specific cell types including bipolar cells, Müller glia, and cones while it is dispensable for preventing cell cycle reentry in other cell types.


Subject(s)
Cell Proliferation/physiology , Cyclin-Dependent Kinase Inhibitor p27/metabolism , Neurogenesis/physiology , Retina/cytology , Retina/embryology , Animals , Cell Differentiation/physiology , Mice , Mice, Knockout , Retina/metabolism
5.
Sci Rep ; 6: 29856, 2016 07 19.
Article in English | MEDLINE | ID: mdl-27432639

ABSTRACT

Rapid saltatory nerve conduction is facilitated by myelin structure, which is produced by Schwann cells (SC) in the peripheral nervous system (PNS). Proper development and degeneration/regeneration after injury requires regulated phenotypic changes of SC. We have previously shown that glutamate can induce SC proliferation in culture. Here we show that glutamate signals through metabotropic glutamate receptor 2 (mGluR2) to induce Erk phosphorylation in SC. mGluR2-elicited Erk phosphorylation requires ErbB2/3 receptor tyrosine kinase phosphorylation to limit the signaling cascade that promotes phosphorylation of Erk, but not Akt. We found that Gßγ and Src are involved in subcellular signaling downstream of mGluR2. We also found that glutamate can transform myelinating SC to proliferating SC, while inhibition of mGluR2 signaling can inhibit demyelination of injured nerves in vivo. These data suggest pathophysiological significance of mGluR2 signaling in PNS and its possible therapeutic importance to combat demyelinating disorders including Charcot-Marie-Tooth disease.


Subject(s)
Glutamic Acid/genetics , Peripheral Nerve Injuries/genetics , Receptors, Metabotropic Glutamate/genetics , Schwann Cells/metabolism , Animals , Cell Differentiation/genetics , Cell Proliferation/genetics , Glutamic Acid/metabolism , Humans , MAP Kinase Signaling System/genetics , Mice , Myelin Sheath/genetics , Myelin Sheath/metabolism , Peripheral Nerve Injuries/pathology , Peripheral Nervous System/injuries , Peripheral Nervous System/metabolism , Peripheral Nervous System/pathology , Phosphorylation , Proto-Oncogene Proteins c-akt/genetics , Rats , Receptor, ErbB-2/genetics , Schwann Cells/pathology , Sciatic Nerve/injuries , Sciatic Nerve/pathology
6.
Invest Ophthalmol Vis Sci ; 57(3): 1169-82, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26975029

ABSTRACT

PURPOSE: Müller glia, the principal glial cell type in the retina, have the potential to proliferate and regenerate neurons after retinal damage. However, unlike the situation in fish and birds, this capacity of Müller glia is extremely limited in mammals. To gain new insights into the mechanisms that hamper retinal regeneration in mammals, we examined the cell cycle progression and DNA damage response in Müller glia after retinal damage. METHODS: Expression of cell cycle-related proteins and DNA damage response were analyzed in adult rat and mouse retinas after N-methyl-N-nitrosourea (MNU)- or N-methyl-D-aspartate (NMDA)-induced retinal damage. Zebrafish and postnatal rat retinas were also investigated for comparison. Analysis was conducted by using immunofluorescence, Western blotting, and quantitative real-time polymerase chain reaction. RESULTS: In the rat retina, most Müller glia reentered the cell cycle after MNU-induced photoreceptor damage while no proliferative response was observed in the mouse model. Cell cycle reentry of rat Müller glia was accompanied by DNA damage response including the phosphorylation of the histone variant H2AX and upregulation of p53 and p21. The DNA damage response was also observed in rat Müller glia after NMDA-induced loss of inner retinal neurons, but not in zebrafish Müller glia or rat retinal progenitor cells. CONCLUSIONS: Our findings suggest that the DNA damage response induced by unscheduled cell cycle reentry may be one of the mechanisms that limit the proliferative and regenerative capacity of Müller glia in the mammalian retina.


Subject(s)
Cyclin D1/genetics , DNA Damage/genetics , Ependymoglial Cells/pathology , Gene Expression Regulation , Photoreceptor Cells/pathology , Retinal Diseases/genetics , Retinal Neurons/pathology , Animals , Blotting, Western , Cell Count , Cell Cycle , Cell Proliferation , Cyclin D1/biosynthesis , Cyclin-Dependent Kinase 4/biosynthesis , Cyclin-Dependent Kinase 4/genetics , Disease Models, Animal , Ependymoglial Cells/metabolism , Immunohistochemistry , In Situ Nick-End Labeling , Male , Mice , Mice, Inbred C57BL , Nerve Regeneration/genetics , Photoreceptor Cells/metabolism , RNA/genetics , Rats , Rats, Wistar , Real-Time Polymerase Chain Reaction , Retina/metabolism , Retina/pathology , Retinal Diseases/metabolism , Retinal Diseases/pathology , Retinal Neurons/metabolism
7.
Nat Cell Biol ; 13(12): 1415-23, 2011 Nov 06.
Article in English | MEDLINE | ID: mdl-22057101

ABSTRACT

Wallerian degeneration is observed in many neurological disorders, and it is therefore important to elucidate the axonal degeneration mechanism to prevent, and further develop treatment for, such diseases. The ubiquitin-proteasome system (UPS) has been implicated in Wallerian degeneration, but the underlying molecular mechanism remains unclear. Here we show that ZNRF1, an E3 ligase, promotes Wallerian degeneration by targeting AKT to degrade through the UPS. AKT phosphorylates glycogen synthase kinase-3ß (GSK3B), and thereby inactivates it in axons. AKT overexpression significantly delays axonal degeneration. Overexpression of the active (non-phosphorylated) form of GSK3B induces CRMP2 phosphorylation, which is required for the microtubule reorganization observed in the degenerating axon. The inhibition of GSK3B and the overexpression of non-phosphorylated CRMP2 both protected axons from Wallerian degeneration. These findings indicate that the ZNRF1-AKT-GSK3B-CRMP2 pathway plays an important role in controlling Wallerian degeneration.


Subject(s)
Glycogen Synthase Kinase 3/physiology , Intercellular Signaling Peptides and Proteins/metabolism , Nerve Tissue Proteins/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Ubiquitin-Protein Ligases/physiology , Wallerian Degeneration/metabolism , Animals , Axons/enzymology , Axons/metabolism , Cells, Cultured , Female , Glycogen Synthase Kinase 3 beta , HEK293 Cells , Humans , Mice , Phosphorylation/physiology , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Signal Transduction/physiology , Ubiquitin-Protein Ligases/metabolism
8.
J Neurosci ; 30(4): 1204-12, 2010 Jan 27.
Article in English | MEDLINE | ID: mdl-20107048

ABSTRACT

Rapid saltatory nerve conduction is facilitated by myelin structure, which is composed of Schwann cells in the peripheral nervous system. Schwann cells drastically change their phenotype following peripheral nerve injury. These phenotypic changes are required for efficient degeneration/regeneration. We previously identified ZNRF1 as an E3 ubiquitin ligase containing a RING finger motif, whose expression is upregulated in the Schwann cells following nerve injury. This suggested that posttranscriptional regulation of protein expression in Schwann cells may be involved in their phenotypic changes during nerve degeneration/regeneration. Here we report the identification of glutamine synthetase (GS), an enzyme that synthesizes glutamine using glutamate and ammonia, as a substrate for E3 activity of ZNRF1 in Schwann cells. GS is known to be highly expressed in differentiated Schwann cells, but its functional significance has remained unclear. We found that during nerve degeneration/regeneration, GS expression is controlled mostly by ZNRF1-dependent proteasomal degradation. We also found that Schwann cells increase oxidative stress upon initiation of nerve degeneration, which promotes carbonylation and subsequent degradation of GS. Surprisingly, we discovered that GS expression regulates Schwann cell differentiation; i.e., increased GS expression promotes myelination via its enzymatic activity. Among the substrates and products of GS, increased glutamate concentration inhibited myelination and yet promoted Schwann cell proliferation by activating metabotropic glutamate receptor signaling. This would suggest that GS may exert its effect on Schwann cell differentiation by regulating glutamate concentration. These results indicate that the ZNRF1-GS system may play an important role in correlating Schwann cell metabolism with its differentiation.


Subject(s)
Cell Differentiation/physiology , Glutamate-Ammonia Ligase/metabolism , Nerve Regeneration/physiology , Peripheral Nervous System Diseases/enzymology , Proteasome Endopeptidase Complex/metabolism , Schwann Cells/enzymology , Animals , Animals, Newborn , Carrier Proteins/metabolism , Cell Line , Cells, Cultured , Disease Models, Animal , Glutamine/biosynthesis , Humans , Mice , Myelin Sheath/metabolism , Oxidative Stress/physiology , Peripheral Nervous System Diseases/physiopathology , Rats , Rats, Sprague-Dawley , Receptors, Metabotropic Glutamate/metabolism , Schwann Cells/cytology , Ubiquitin-Protein Ligases/metabolism
9.
Biochem Biophys Res Commun ; 347(3): 814-20, 2006 Sep 01.
Article in English | MEDLINE | ID: mdl-16872923

ABSTRACT

Localization of CaMKIIalpha in lipid rafts was demonstrated in both cultured neurons and mammalian cells transfected with plasmid with an insert of CaMKIIalpha cDNA by using sucrose gradient centrifugation and the sensitivity to a cholesterol-extractor, methyl-beta-cyclodextrin. CaMKIIalpha was targeted to lipid rafts possibly through protein-protein interactions via at least three domains (a.a. 261-309, 371-420, and 421-478). The multimeric structure of the full-length molecule also appeared to contribute to efficient lipid raft-targeting. Acylation of CaMKIIalpha did not appear to be a mechanism for the targeting.


Subject(s)
Calcium-Calmodulin-Dependent Protein Kinases/metabolism , Membrane Microdomains/enzymology , Animals , COS Cells , Calcium-Calmodulin-Dependent Protein Kinase Type 2 , Calcium-Calmodulin-Dependent Protein Kinases/genetics , Cell Fractionation , Chlorocebus aethiops , Hydroxylamine/pharmacology , Membrane Microdomains/drug effects , Phosphothreonine/metabolism , Protein Binding , Rats
10.
Eur J Neurosci ; 23(11): 2864-76, 2006 Jun.
Article in English | MEDLINE | ID: mdl-16819975

ABSTRACT

We cloned here a full-length cDNA of Dem26[Tian et al. (1999)Mol. Brain Res., 72, 147-157], a member of the low-density lipoprotein (LDL) receptor gene family from the rat brain. We originally named the corresponding protein synaptic LDL receptor-related protein (synLRP) [Tian et al. (2002) Soc. Neurosci. Abstr., 28, 405] and have renamed it LRP4 to accord it systematic nomenclature (GenBank(TM) accession no. AB073317). LRP4 protein interacted with postsynaptic scaffold proteins such as postsynaptic density (PSD)-95 via its C-terminal tail sequence, and associated with N-methyl-D-aspartate (NMDA)-type glutamate receptor subunit. The mRNA of LRP4 was localized to dendrites, as well as somas, of neuronal cells, and the full-length protein of 250 kDa was highly concentrated in the brain and localized to various subcellular compartments in the brain, including synaptic fractions. Immunocytochemical study using cultured cortical neurons suggested surface localization in the neuronal cells both in somas and dendrites. Ca(2+)/calmodulin-dependent protein kinase II (CaMKII) phosphorylated the C-terminal cytoplasmic region of LRP4 at Ser1887 and Ser1900, and the phosphorylation at the latter site suppressed the interaction of the protein with PSD-95 and synapse-associated protein 97 (SAP97). These findings suggest a postsynaptic role for LRP4, a putative endocytic multiligand receptor, and a mechanism in which CaMKII regulates PDZ-dependent protein-protein interactions and receptor dynamics.


Subject(s)
Calcium-Calmodulin-Dependent Protein Kinases/physiology , Low Density Lipoprotein Receptor-Related Protein-1/metabolism , Nerve Tissue Proteins/metabolism , Animals , Animals, Newborn , Blotting, Northern/methods , Brain/cytology , Calcium-Calmodulin-Dependent Protein Kinase Type 2 , Cells, Cultured , Chlorocebus aethiops , Cloning, Molecular/methods , Electrophoresis, Polyacrylamide Gel , Electrophoretic Mobility Shift Assay , Immunohistochemistry/methods , Immunoprecipitation/methods , In Situ Hybridization/methods , Low Density Lipoprotein Receptor-Related Protein-1/chemistry , Low Density Lipoprotein Receptor-Related Protein-1/genetics , Phosphorylation , Protein Binding/drug effects , Protein Binding/physiology , Protein Structure, Tertiary/physiology , Rats , Rats, Wistar , Serine/metabolism , Time Factors , Transfection/methods
11.
J Biol Chem ; 279(28): 29461-8, 2004 Jul 09.
Article in English | MEDLINE | ID: mdl-15105416

ABSTRACT

Targeting of neuronal nitric-oxide synthase (nNOS) to appropriate sites in a cell is mediated by interactions with its PDZ domain and plays an important role in specifying the sites of reaction of nitric oxide (NO) in the central nervous system. Here we report the identification and characterization of a novel nNOS-interacting DHHC domain-containing protein with dendritic mRNA (NIDD) (GenBank accession number AB098078), which increases nNOS enzyme activity by targeting the nNOS to the synaptic plasma membrane in a PDZ domain-dependent manner. The deduced NIDD protein consisted of 392 amino acid residues and possessed five transmembrane segments, a zinc finger DHHC domain, and a PDZ-binding motif (-EDIV) at its C-terminal tail. In vitro pull-down assays suggested that the C-terminal tail region of NIDD specifically interacted with the PDZ domain of nNOS. The PDZ dependence was confirmed by an experiment using a deletion mutant, and the interaction was further confirmed by co-sedimentation assays using COS-7 cells transfected with NIDD and nNOS. Both NIDD and nNOS were enriched in synaptosome and synaptic plasma membrane fractions and were present in the lipid raft and postsynaptic density fractions in the rat brain. Co-localization of these proteins was also observed by double staining of the proteins in cultured cortical neurons. Thus, NIDD and nNOS were co-localized in the brain, although the colocalizing regions were restricted, as indicated by the distribution of their mRNA expression. Most important, co-transfection of NIDD and nNOS increased NO-producing nNOS activity. These results suggested that NIDD plays an important role in the regulation of the NO signaling pathway at postsynaptic sites through targeting of nNOS to the postsynaptic membrane.


Subject(s)
Carrier Proteins/metabolism , Nerve Tissue Proteins/metabolism , Nitric Oxide Synthase/metabolism , Synaptic Membranes/metabolism , Amino Acid Sequence , Animals , COS Cells , Carrier Proteins/genetics , Cell Fractionation , Cells, Cultured , Cerebral Cortex/cytology , Chlorocebus aethiops , In Situ Hybridization , Male , Molecular Sequence Data , Nerve Tissue Proteins/genetics , Neurons/cytology , Neurons/metabolism , Nitric Oxide Synthase/genetics , Nitric Oxide Synthase Type I , Protein Structure, Tertiary , RNA, Messenger/metabolism , Rats , Rats, Wistar , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Signal Transduction/physiology , Synaptic Membranes/chemistry , Tissue Distribution
SELECTION OF CITATIONS
SEARCH DETAIL
...