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










Publication year range
1.
Cell Rep ; 42(9): 113068, 2023 09 26.
Article in English | MEDLINE | ID: mdl-37656624

ABSTRACT

Primary somatosensory axons stop regenerating as they re-enter the spinal cord, resulting in incurable sensory loss. What arrests them has remained unclear. We previously showed that axons stop by forming synaptic contacts with unknown non-neuronal cells. Here, we identified these cells in adult mice as oligodendrocyte precursor cells (OPCs). We also found that only a few axons stop regenerating by forming dystrophic endings, exclusively at the CNS:peripheral nervous system (PNS) borderline where OPCs are absent. Most axons stop in contact with a dense network of OPC processes. Live imaging, immuno-electron microscopy (immuno-EM), and OPC-dorsal root ganglia (DRG) co-culture additionally suggest that axons are rapidly immobilized by forming synapses with OPCs. Genetic OPC ablation enables many axons to continue regenerating deep into the spinal cord. We propose that sensory axons stop regenerating by encountering OPCs that induce presynaptic differentiation. Our findings identify OPCs as a major regenerative barrier that prevents intraspinal restoration of sensory circuits following spinal root injury.


Subject(s)
Oligodendrocyte Precursor Cells , Mice , Animals , Spinal Cord/physiology , Axons/physiology , Spinal Nerve Roots , Ganglia, Spinal/physiology , Nerve Regeneration/physiology
2.
Elife ; 102021 05 04.
Article in English | MEDLINE | ID: mdl-33942723

ABSTRACT

A major barrier to intraspinal regeneration after dorsal root (DR) injury is the DR entry zone (DREZ), the CNS/PNS interface. DR axons stop regenerating at the DREZ, even if regenerative capacity is increased by a nerve conditioning lesion. This potent blockade has long been attributed to myelin-associated inhibitors and (CSPGs), but incomplete lesions and conflicting reports have prevented conclusive agreement. Here, we evaluated DR regeneration in mice using novel strategies to facilitate complete lesions and analyses, selective tracing of proprioceptive and mechanoreceptive axons, and the first simultaneous targeting of Nogo/Reticulon-4, MAG, OMgp, CSPGs, and GDNF. Co-eliminating myelin inhibitors and CSPGs elicited regeneration of only a few conditioning-lesioned DR axons across the DREZ. Their absence, however, markedly and synergistically enhanced regeneration of GDNF-stimulated axons, highlighting the importance of sufficiently elevating intrinsic growth capacity. We also conclude that myelin inhibitors and CSPGs are not the primary mechanism stopping axons at the DREZ.


Subject(s)
Axons/physiology , Glial Cell Line-Derived Neurotrophic Factor/genetics , Myelin Sheath/metabolism , Spinal Cord/cytology , Spinal Nerve Roots/pathology , Animals , Female , Male , Mice , Mice, Inbred C57BL , Mice, Knockout
3.
eNeuro ; 4(6)2017.
Article in English | MEDLINE | ID: mdl-29218323

ABSTRACT

The dendritic tree is a key determinant of neuronal information processing. In the motor system, the dendritic tree of spinal cord neurons undergoes dramatic remodeling in an activity-dependent manner during early postnatal life. This leads to the proper segmental spinal cord connectivity that subserves normal locomotor behavior. One molecular system driving the establishment of dendrite architecture of mammalian motor neurons relies on AMPA receptors (AMPA-Rs) assembled with the GluA1 subunit, and this occurs in an NMDA receptor (NMDA-R)-independent manner. The dendrite growth promoting activity of GluA1-containing AMPA-Rs depends on its intracellular binding partner, SAP97, and SAP97's PDZ3 domain. We show here that cysteine-rich interactor of PDZ3 (CRIPT) is a bona fide SAP97 PDZ3-domain binding partner, localizes to synapses with GluA1 and SAP97 along the dendritic tree, and is a determinant of the dendritic growth of mammalian spinal cord neurons. We further show that CRIPT has a well-conserved ortholog in the nematode, Caenorhabditis elegans, and animals lacking CRIPT display decreased dendrite branching of the well-studied PVD neuron in vivo. The lack of CRIPT leads to a selective defect in touch perception, and this is rescued by expression of wild-type (WT) human CRIPT (hCRIPT) in the nervous system. This work brings new light into the molecular machinery that drives dendritic growth during development and may prove relevant to the promotion of nervous system plasticity following insult.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Dendrites , Membrane Proteins/metabolism , Neurogenesis/physiology , Spinal Cord/growth & development , Spinal Cord/metabolism , Animals , Caenorhabditis elegans , Discs Large Homolog 1 Protein , HEK293 Cells , Humans , Rats
4.
Elife ; 62017 01 26.
Article in English | MEDLINE | ID: mdl-28124973

ABSTRACT

Nuclear exclusion of the transcriptional regulators and potent oncoproteins, YAP/TAZ, is considered necessary for adult tissue homeostasis. Here we show that nuclear YAP/TAZ are essential regulators of peripheral nerve development and myelin maintenance. To proliferate, developing Schwann cells (SCs) require YAP/TAZ to enter S-phase and, without them, fail to generate sufficient SCs for timely axon sorting. To differentiate, SCs require YAP/TAZ to upregulate Krox20 and, without them, completely fail to myelinate, resulting in severe peripheral neuropathy. Remarkably, in adulthood, nuclear YAP/TAZ are selectively expressed by myelinating SCs, and conditional ablation results in severe peripheral demyelination and mouse death. YAP/TAZ regulate both developmental and adult myelination by driving TEAD1 to activate Krox20. Therefore, YAP/TAZ are crucial for SCs to myelinate developing nerve and to maintain myelinated nerve in adulthood. Our study also provides a new insight into the role of nuclear YAP/TAZ in homeostatic maintenance of an adult tissue.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Myelin Sheath/metabolism , Phosphoproteins/metabolism , Schwann Cells/physiology , Transcription Factors/metabolism , Acyltransferases , Animals , Cell Cycle Proteins , Cell Differentiation , Cell Proliferation , Mice , YAP-Signaling Proteins
5.
Exp Neurol ; 283(Pt A): 73-84, 2016 09.
Article in English | MEDLINE | ID: mdl-27264357

ABSTRACT

Although previous studies have identified several strategies to stimulate regeneration of CNS axons, extensive regeneration and functional recovery have remained a major challenge, particularly for large diameter myelinated axons. Within the CNS, myelin is thought to inhibit axon regeneration, while modulating activity of the mTOR pathway promotes regeneration of injured axons. In this study, we examined NT-3 mediated regeneration of sensory axons through the dorsal root entry zone in a triple knockout of myelin inhibitory proteins or after activation of mTOR using a constitutively active (ca) Rheb in DRG neurons to determine the influence of environmental inhibitory or activation of intrinsic growth pathways could enhance NT-3-mediate regeneration. Loss of myelin inhibitory proteins showed modest enhancement of sensory axon regeneration. In mTOR studies, we found a dramatic age related decrease in the mTOR activation as determined by phosphorylation of the downstream marker S6 ribosomal subunit. Expression of caRheb within adult DRG neurons in vitro increased S6 phosphorylation and doubled the overall length of neurite outgrowth, which was reversed in the presence of rapamycin. In adult female rats, combined expression of caRheb in DRG neurons and NT-3 within the spinal cord increased regeneration of sensory axons almost 3 fold when compared to NT-3 alone. Proprioceptive assessment using a grid runway indicates functionally significant regeneration of large-diameter myelinated sensory afferents. Our results indicate that caRheb-induced increase in mTOR activation enhances neurotrophin-3 induced regeneration of large-diameter myelinated axons.


Subject(s)
Gene Expression Regulation/physiology , Nerve Regeneration/physiology , Neurotrophin 3/metabolism , Signal Transduction/physiology , Somatosensory Disorders/metabolism , TOR Serine-Threonine Kinases/metabolism , Animals , Animals, Newborn , Cells, Cultured , Disease Models, Animal , Embryo, Mammalian , Female , Ganglia, Spinal/cytology , Gene Expression Regulation/drug effects , Hyperalgesia/physiopathology , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Myelin-Associated Glycoprotein/deficiency , Myelin-Associated Glycoprotein/genetics , Neurotrophin 3/genetics , Neurotrophin 3/therapeutic use , Nogo Proteins/deficiency , Nogo Proteins/genetics , Rats , Rats, Sprague-Dawley , Sirolimus/pharmacology , Somatosensory Disorders/pathology , Somatosensory Disorders/physiopathology , Spinal Cord Injuries/drug therapy , Spinal Cord Injuries/pathology , Spinal Cord Injuries/physiopathology , Spinal Cord Injuries/therapy
6.
J Neurosci ; 35(24): 9088-105, 2015 Jun 17.
Article in English | MEDLINE | ID: mdl-26085633

ABSTRACT

Mutant genes that underlie Mendelian forms of amyotrophic lateral sclerosis (ALS) and biochemical investigations of genetic disease models point to potential driver pathophysiological events involving endoplasmic reticulum (ER) stress and autophagy. Several steps in these cell biological processes are known to be controlled physiologically by small ADP-ribosylation factor (ARF) signaling. Here, we investigated the role of ARF guanine nucleotide exchange factors (GEFs), cytohesins, in models of ALS. Genetic or pharmacological inhibition of cytohesins protects motor neurons in vitro from proteotoxic insults and rescues locomotor defects in a Caenorhabditis elegans model of disease. Cytohesins form a complex with mutant superoxide dismutase 1 (SOD1), a known cause of familial ALS, but this is not associated with a change in GEF activity or ARF activation. ER stress evoked by mutant SOD1 expression is alleviated by antagonism of cytohesin activity. In the setting of mutant SOD1 toxicity, inhibition of cytohesin activity enhances autophagic flux and reduces the burden of misfolded SOD1. These observations suggest that targeting cytohesins may have potential benefits for the treatment of ALS.


Subject(s)
Caenorhabditis elegans Proteins/antagonists & inhibitors , Caenorhabditis elegans Proteins/genetics , Disease Models, Animal , Guanine Nucleotide Exchange Factors/antagonists & inhibitors , Guanine Nucleotide Exchange Factors/genetics , Motor Neuron Disease/genetics , Amyotrophic Lateral Sclerosis/genetics , Amyotrophic Lateral Sclerosis/metabolism , Animals , Caenorhabditis elegans , Caenorhabditis elegans Proteins/biosynthesis , Cells, Cultured , GTPase-Activating Proteins/antagonists & inhibitors , GTPase-Activating Proteins/biosynthesis , GTPase-Activating Proteins/genetics , Guanine Nucleotide Exchange Factors/biosynthesis , HeLa Cells , Humans , Mice , Models, Genetic , Rats , Rats, Sprague-Dawley , Superoxide Dismutase/biosynthesis , Superoxide Dismutase/genetics
7.
Hum Mol Genet ; 20(21): 4116-31, 2011 Nov 01.
Article in English | MEDLINE | ID: mdl-21816949

ABSTRACT

Brain-derived neurotrophic factor (BDNF) and its receptor tropomyosin-related kinase B (TrkB) are widely expressed in the vertebrate nervous system and play a central role in mature neuronal function. In vitro BDNF/TrkB signaling promotes neuronal survival and can help neurons resist toxic insults. Paradoxically, BDNF/TrkB signaling has also been shown, under certain in vitro circumstances, to render neurons vulnerable to insults. We show here that in vivo conditional deletion of TrkB from mature motor neurons attenuates mutant superoxide dismutase 1 (SOD1) toxicity. Mutant SOD1 mice lacking motor neuron TrkB live a month longer than controls and retain motor function for a longer period, particularly in the early phase of the disease. These effects are subserved by slowed motor neuron loss, persistence of neuromuscular junction integrity and reduced astrocytic and microglial reactivity within the spinal cord. These results suggest that manipulation of BDNF/TrkB signaling might have therapeutic efficacy in motor neuron diseases.


Subject(s)
Motor Neuron Disease/enzymology , Motor Neuron Disease/pathology , Motor Neurons/metabolism , Motor Neurons/pathology , Mutation/genetics , Receptor, trkB/metabolism , Superoxide Dismutase/genetics , Amino Acid Substitution , Animals , Axons/metabolism , Axons/pathology , Denervation , Disease Progression , Ganglion Cysts/metabolism , Ganglion Cysts/pathology , Gene Deletion , Inclusion Bodies/metabolism , Inflammation/complications , Inflammation/pathology , Inflammation/physiopathology , Integrases/metabolism , Interneurons/metabolism , Interneurons/pathology , Longevity , Mice , Mice, Knockout , Motor Activity , Motor Neuron Disease/complications , Motor Neuron Disease/physiopathology , Neuromuscular Junction/metabolism , Neuromuscular Junction/pathology , Recombination, Genetic/genetics , Spinal Cord/metabolism , Spinal Cord/pathology , Superoxide Dismutase-1 , Ubiquitin/metabolism , Ubiquitination , Vesicular Acetylcholine Transport Proteins/metabolism
8.
J Cell Biol ; 180(1): 187-203, 2008 Jan 14.
Article in English | MEDLINE | ID: mdl-18195107

ABSTRACT

Integrin binding to matrix proteins such as fibronectin (FN) leads to formation of focal adhesion (FA) cellular contact sites that regulate migration. RhoA GTPases facilitate FA formation, yet FA-associated RhoA-specific guanine nucleotide exchange factors (GEFs) remain unknown. Here, we show that proline-rich kinase-2 (Pyk2) levels increase upon loss of focal adhesion kinase (FAK) in mouse embryonic fibroblasts (MEFs). Additionally, we demonstrate that Pyk2 facilitates deregulated RhoA activation, elevated FA formation, and enhanced cell proliferation by promoting p190RhoGEF expression. In normal MEFs, p190RhoGEF knockdown inhibits FN-associated RhoA activation, FA formation, and cell migration. Knockdown of p190RhoGEF-related GEFH1 does not affect FA formation in FAK(-/-) or normal MEFs. p190RhoGEF overexpression enhances RhoA activation and FA formation in MEFs dependent on FAK binding and associated with p190RhoGEF FA recruitment and tyrosine phosphorylation. These studies elucidate a compensatory function for Pyk2 upon FAK loss and identify the FAK-p190RhoGEF complex as an important integrin-proximal regulator of FA formation during FN-stimulated cell motility.


Subject(s)
Cell Movement/physiology , Focal Adhesion Kinase 1/metabolism , Focal Adhesion Kinase 2/metabolism , Focal Adhesions/physiology , ras-GRF1/metabolism , rhoA GTP-Binding Protein/metabolism , Animals , Cell Proliferation , Focal Adhesion Kinase 1/genetics , Focal Adhesion Kinase 2/genetics , Gene Expression Regulation , Mice , Paxillin/metabolism , Phosphorylation , Tyrosine/metabolism , ras-GRF1/genetics
9.
Hum Mol Genet ; 16(24): 3103-16, 2007 Dec 15.
Article in English | MEDLINE | ID: mdl-17881652

ABSTRACT

Mutations in neurofilament light (NFL) subunit and small heat-shock protein B1 (HSPB1) cause autosomal-dominant axonal Charcot-Marie-Tooth disease type 2E (CMT2E) and type 2F (CMT2F). Previous studies have shown that CMT mutations in NFL and HSPB1 disrupt NF assembly and cause aggregation of NFL protein. In this study, we investigate the role of aggregation of NFL protein in the neurotoxicity of CMT mutant NFL and CMT mutant HSPB1 in motor neurons. We find that expression of CMT mutant NFL leads to progressive degeneration and loss of neuronal viability of cultured motor neurons. Degenerating motor neurons show fragmentation and loss of neuritic processes associated with disruption of NF network and aggregation of NFL protein. Co-expression of wild-type HSPB1 diminishes aggregation of CMT mutant NFL, induces reversal of CMT mutant NFL aggregates and reduces CMT mutant NFL-induced loss of motor neuron viability. Like CMT mutant NFL, expression of S135F CMT mutant HSPB1 also leads to progressive degeneration of motor neurons with disruption of NF network and aggregation of NFL protein. Further studies show that wild-type and S135F mutant HSPB1 associate with wild-type and CMT mutant NFL and that S135F mutant HSPB1 has dominant effect on disruption of NF assembly and aggregation of NFL protein. Finally, we show that deletion of NFL markedly reduces degeneration and loss of motor neuron viability induced by S135F mutant HSPB1. Together, our data support the view that disruption of NF network with aggregation of NFL is a common triggering event of motor neuron degeneration in CMT2E and CMT2F disease.


Subject(s)
Charcot-Marie-Tooth Disease/genetics , Heat-Shock Proteins/genetics , Motor Neurons/pathology , Mutation , Neoplasm Proteins/genetics , Nerve Degeneration/pathology , Nerve Net/chemistry , Neurofilament Proteins/genetics , Neurofilament Proteins/metabolism , Animals , Cell Survival , Cells, Cultured , Charcot-Marie-Tooth Disease/pathology , Embryo, Mammalian , HSP27 Heat-Shock Proteins , Inclusion Bodies/metabolism , Inclusion Bodies/pathology , Mice , Mice, Knockout , Molecular Chaperones , Mutant Proteins/physiology , Mutation/physiology , Nerve Degeneration/genetics , Nerve Degeneration/metabolism , Nerve Net/metabolism , Neurofilament Proteins/chemistry , Signal Transduction/physiology
10.
Hum Mol Genet ; 14(23): 3643-59, 2005 Dec 01.
Article in English | MEDLINE | ID: mdl-16236762

ABSTRACT

Abnormal protein aggregation is emerging as a common theme in the pathogenesis of neurodegenerative disease. Our previous studies have shown that overexpression of untranslated light neurofilament (NF-L) RNA causes motor neuron degeneration in transgenic mice, leads to accumulation of ubiquitinated aggregates in degenerating cultured motor neurons and triggers aggregation of NF-L protein and co-aggregation of mutant SOD1 protein in neuronal cells. Here, we report that p190RhoGEF, an RNA-binding protein that binds to a destabilizing element in NF-L mRNA, is involved in aggregation of NF-L protein and is implicated in the pathogenesis of motor neuron degeneration. We show that p190RhoGEF co-aggregates with unassembled NF-L protein and that co-aggregation is associated with down-regulation of parent NF-L mRNA in neuronal cells. Co-expression of NF-M increases NF assembly and reduces RNA-triggered aggregation as well as loss of solubility of NF-L protein. siRNA-induced down-regulation of p190RhoGEF not only reduces aggregation and promotes assembly of NF-L and NF-M, but also causes reversal of aggregation and recovery of NF assembly in transfected cells. Examination of transgenic models of motor neuron disease shows that prominent aggregates of p190RhoGEF and NF-L and down-regulation of NF-L expression occur in degenerating motor neurons of mice expressing untranslated NF-L RNA or a G93A mutant SOD1 transgene. Moreover, aggregates of p190RhoGEF and NF-L appear as early pathological changes in presymptomatic G93A mutant SOD1 transgenic mice. Together, the findings indicate that p190RhoGEF is involved in aggregation of NF-L protein and support a working hypothesis that aggregation of p190RhoGEF and NF-L is an upstream event triggering neurotoxicity in motor neuron disease.


Subject(s)
Carrier Proteins/metabolism , Motor Neuron Disease/metabolism , Nerve Degeneration/metabolism , Neurofilament Proteins/metabolism , RNA Stability , RNA-Binding Proteins/metabolism , Animals , Carrier Proteins/analysis , Carrier Proteins/genetics , DNA-Binding Proteins , GTPase-Activating Proteins , Mice , Mice, Transgenic , Motor Neuron Disease/genetics , Motor Neuron Disease/pathology , Motor Neurons/chemistry , Motor Neurons/metabolism , Nerve Degeneration/genetics , Neurofilament Proteins/analysis , Neurofilament Proteins/genetics , RNA Interference , RNA, Messenger/metabolism , RNA-Binding Proteins/analysis , RNA-Binding Proteins/genetics , Repressor Proteins , Solubility
11.
Hum Mol Genet ; 14(18): 2629-40, 2005 Sep 15.
Article in English | MEDLINE | ID: mdl-16079151

ABSTRACT

Mutations in Cu/Zn superoxide dismutase (SOD1) cause approximately 20% of familial amyotrophic lateral sclerosis by a toxic gain of function; however, the precise mechanisms remain unclear. Here, we report the identification of HoxB2, a homeodomain-containing transcription factor, as a G93A mutant SOD1 interactive protein in a yeast two-hybrid screen. We show that HoxB2 co-precipitates and co-localizes with mutant SOD1 in neuronal cell lines, as well as in brain and spinal cord of G93A mutant SOD1 transgenic mice. Mutagenesis further shows that this interaction is mediated by the central homeodomain of HoxB2. In motor neuron-like NSC-34 cells, overexpression of HoxB2 or its homeodomain decreases the insolubility of mutant SOD1 and inhibits G93A or G86R mutant SOD1-induced neuronal cell death. In human and mouse tissues, we show that expression of HoxB2 persists in adult spinal cord and is primarily localized in nuclei of motor neurons. In G93A transgenic mice, HoxB2 co-localizes with mutant SOD1 and is redistributed to perikarya and proximal neurites of motor neurons. In addition, there is progressive accumulation of HoxB2 and mutant SOD1 as punctate inclusions in the neuropil surrounding motor neurons. Taken together, our findings demonstrate that interaction of HoxB2 with mutant SOD1 occurs in motor neurons of G93A mutant SOD1 transgenic mice and suggest that this interaction may modulate the neurotoxicity of mutant SOD1.


Subject(s)
Amyotrophic Lateral Sclerosis/genetics , Gene Expression , Homeodomain Proteins/metabolism , Superoxide Dismutase/metabolism , Transcription Factors/metabolism , Animals , Apoptosis/genetics , Blotting, Western , Cell Line , Fluorescent Antibody Technique , Homeodomain Proteins/genetics , Humans , Immunohistochemistry , Immunoprecipitation , Mice , Mice, Transgenic , Motor Neurons/metabolism , Mutagenesis, Site-Directed , Mutation/genetics , Oligonucleotides , Reverse Transcriptase Polymerase Chain Reaction , Superoxide Dismutase/genetics , Superoxide Dismutase-1 , Transcription Factors/genetics , Two-Hybrid System Techniques
12.
J Neurosci ; 25(17): 4353-64, 2005 Apr 27.
Article in English | MEDLINE | ID: mdl-15858061

ABSTRACT

A 68 nucleotide segment of the light neurofilament (NF-L) mRNA, spanning the translation termination signal, participates in regulating the stability of the transcript in vivo. Aldolases A and C, but not B, interact specifically with this segment of the transcript in vitro. Aldolases A and C are glycolytic enzymes expressed in neural cells, and their mRNA binding activity represents a novel function of these isozymes. This unsuspected new activity was first uncovered by Northwestern blotting of a brainstem/spinal cord cDNA library. It was confirmed by two-dimensional fractionation of mouse brain cytosol followed by Northwestern hybridization and protein sequencing. Both neuronal aldolases interact specifically with the NF-L but not the heavy neurofilament mRNA, and their binding to the transcript excludes the poly(A)-binding protein (PABP) from the complex. Constitutive ectopic expression of aldolases A and C accelerates the decay of a neurofilament transgene (NF-L) driven by a tetracycline inducible system. In contrast, mutant transgenes lacking mRNA sequence for aldolase binding are stabilized. Our findings strongly suggest that aldolases A and C are regulatory components of a light neurofilament mRNA complex that modulates the stability of NF-L mRNA. This modulation likely involves endonucleolytic cleavage and a competing interaction with the PABP. Interactions of aldolases A and C in NF-L expression may be linked to regulatory pathways that maintain the highly asymmetrical form and function of large neurons.


Subject(s)
Fructose-Bisphosphate Aldolase/physiology , Gene Expression Regulation/physiology , Neurofilament Proteins/metabolism , Animals , Blotting, Northern/methods , Blotting, Western/methods , Brain/metabolism , Cell Line , Chlorocebus aethiops , Chromatography, High Pressure Liquid/methods , Cloning, Molecular/methods , Electrophoresis, Gel, Two-Dimensional/methods , Electrophoretic Mobility Shift Assay/methods , Gene Expression/physiology , Gene Library , Humans , Immunoprecipitation/methods , Mice , Molecular Sequence Data , Molecular Weight , Neurofilament Proteins/genetics , Peptide Mapping/methods , Protein Biosynthesis/physiology , RNA, Messenger/metabolism , Recombinant Fusion Proteins , Reverse Transcriptase Polymerase Chain Reaction/methods , Transfection/methods
13.
J Neurosci ; 24(11): 2716-26, 2004 Mar 17.
Article in English | MEDLINE | ID: mdl-15028764

ABSTRACT

The pathogenesis of neurodegenerative diseases is believed to involve abnormal aggregation of proteins, but the mechanisms initiating protein aggregation are unclear. Here we report a novel phenomenon that could be instrumental in triggering protein aggregation in neurodegenerative diseases. We show that the 3' untranslated region (3'UTR) of a light neurofilament (NF-L) transcript enhances the reactivity of its own translated product and leads to loss of solubility and aggregation of NF-L protein and to coaggregation of mutant superoxide dismutase 1 (SOD1) protein. Full-length mouse NF-L cDNAs, with and without NF-L 3'UTR, were fused to the C terminus of a green fluorescent protein (GFP) reporter gene, and the GFP-tagged NF-L proteins were examined in transfected Neuro2a cells. The GFP-tagged NF-L protein expressed from the transgene containing NF-L 3'UTR, but not from the transgene lacking NF-L 3'UTR, colocalizes with endogenous heavy neurofilament protein and, at high-level expression, leads to loss of solubility and aggregation of GFP-tagged NF-L protein. Aggregation of GFP-tagged NF-L protein triggers coaggregation and loss of solubility of coexpressed DsRed-tagged mutant (G93A) SOD1 protein but not wild-type SOD1 protein. Deletional mutagenesis maps the RNA sequence causing aggregation of GFP-tagged NF-L protein to the proximal 45 nucleotides of NF-L 3'UTR. This is the site of a major destabilizing element in NF-L RNA and binding site for RNA-binding proteins. Our findings support a working model whereby NF-L RNA, or cognate RNA-binding factors, enhances the reactivity of NF-L protein and provides a triggering mechanism leading to aggregation of NF-L and other proteins in neurodegenerative diseases.


Subject(s)
3' Untranslated Regions/physiology , Neurofilament Proteins/genetics , Neurofilament Proteins/metabolism , Neurons/metabolism , RNA, Messenger/physiology , Superoxide Dismutase/metabolism , 3' Untranslated Regions/pharmacology , Animals , Cell Line , Genes, Reporter/genetics , Green Fluorescent Proteins , Heat-Shock Proteins/metabolism , Luminescent Proteins/genetics , Macromolecular Substances , Mice , Neuroblastoma/metabolism , Neurodegenerative Diseases/etiology , Neurodegenerative Diseases/metabolism , Neurons/drug effects , Protein Binding/drug effects , Protein Binding/genetics , Protein Binding/physiology , RNA, Messenger/metabolism , RNA, Messenger/pharmacology , Receptors, Androgen/genetics , Receptors, Androgen/metabolism , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Solubility , Superoxide Dismutase/genetics , Superoxide Dismutase-1 , Transfection , Transgenes , Trinucleotide Repeat Expansion , Ubiquitins/metabolism
14.
J Neuropathol Exp Neurol ; 62(9): 936-50, 2003 Sep.
Article in English | MEDLINE | ID: mdl-14533783

ABSTRACT

The mechanisms whereby mutant gene expression triggers neurodegeneration are poorly understood but have generally been attributed to translated gene products. We now demonstrate direct neuropathic effects of untranslated RNA on cultured motor neurons. We show that expression of untranslated light neurofilament (NF-L) RNA sequence in the 3'UTR of an EGFP transgene (pEGFP/NF-L RNA) or in a separate expression vector (pRc/NF-L RNA) causes dose-dependent, neuron-specific motor neuron degeneration. Neither unfused EGFP protein (pEGFP/wt) nor EGFP-tagged NF-L protein (pEGFP/NF-L protein) has similar neuropathic effects. The findings are the first demonstration of a direct RNA-mediated neurotoxic effect. Moreover, the resulting neuropathological changes show that untranslated RNA can lead to early degeneration of neuritic processes and accumulations of ubiquitinated aggregates in the perikarya and nuclei of degenerating motor neurons. The latter findings are hallmark neuropathological features of neurodegenerative diseases and their occurrence as a result of altered RNA expression raises the prospects of an RNA-mediated component in the pathogenesis of neurodegenerative states.


Subject(s)
Motor Neurons/metabolism , Nerve Degeneration/metabolism , Neurofilament Proteins/biosynthesis , RNA/biosynthesis , Ubiquitins/biosynthesis , Animals , Cell Aggregation/physiology , Cell Survival/physiology , Cells, Cultured , Gene Expression Regulation/physiology , Mice , Motor Neurons/pathology , Nerve Degeneration/pathology , Neurofilament Proteins/genetics , RNA/metabolism , Spinal Cord/metabolism , Spinal Cord/pathology , Ubiquitins/genetics
15.
Brain Res Mol Brain Res ; 117(1): 27-38, 2003 Sep 10.
Article in English | MEDLINE | ID: mdl-14499478

ABSTRACT

p190RhoGEF is a large multi-functional protein with guanine nucleotide exchange (GEF) activity. The C-terminal region of p190RhoGEF is a highly interactive domain that binds multiple factors, including proteins with anti-apoptotic activities. We now report that transfection of EGFP-tagged p190RhoGEF protects Neuro 2a cells from stress-induced apoptosis and that anti-apoptotic activity is localized to cytoplasmic retention sequences (CRS-1 and CRS-2) in the C-terminal region of p190RhoGEF. Cytoplasmic retention is conferred to an EGFP fluorescent marker when fused to either CRS-1 or CRS-2. Both cytoplasmic retention and anti-apoptotic activity are lost by deleting CRS-1 and CRS-2 in the p190RhoGEF sequence and can be recovered by restoring either CRS-1 or CRS-2 to the EGFP-tagged sequence. Since the CRS-1 and CRS-2 contain the JIP-1 and 14-3-3 binding sites, we propose that anti-apoptotic activity may be conferred by the binding of p190RhoGEF to JIP-1 or 14-3-3, possibly by altering their interactive properties or nucleocytoplasmic movements. Taken together, our findings support a model whereby multiple interactions of p190RhoGEF confer homeostatic properties to differentiated neurons and may link neuronal homeostasis to the regulation of NF-L expression.


Subject(s)
Apoptosis/physiology , Cytoplasm/metabolism , Guanine Nucleotide Exchange Factors/metabolism , Intracellular Signaling Peptides and Proteins , Luminescent Proteins/metabolism , Nuclear Proteins/metabolism , Animals , Binding Sites , Blotting, Western , Carrier Proteins/metabolism , Cell Aggregation , Cell Death , Cell Line , DNA-Binding Proteins , GTPase-Activating Proteins , Glutathione Transferase/genetics , Green Fluorescent Proteins , In Situ Nick-End Labeling/methods , Microscopy, Confocal , Peptide Fragments/metabolism , Precipitin Tests/methods , Protein Binding , Proto-Oncogene Proteins c-myc/metabolism , Repressor Proteins , Sequence Homology, Amino Acid , Transfection , Two-Hybrid System Techniques
16.
J Biol Chem ; 278(27): 24865-73, 2003 Jul 04.
Article in English | MEDLINE | ID: mdl-12702722

ABSTRACT

Focal adhesion kinase (FAK) is a protein-tyrosine kinase that associates with multiple cell surface receptors and signaling proteins through which it can modulate the activity of several intracellular signaling pathways. FAK activity can influence the formation of distinct actin cytoskeletal structures such as lamellipodia and stress fibers in part through effects on small Rho GTPases, although the molecular interconnections of these events are not well defined. Here, we report that FAK interacts with p190RhoGEF, a RhoA-specific GDP/GTP exchange factor, in neuronal cells and in brain tissue extracts by co-immunoprecipitation and co-localization analyses. Using a two-hybrid assay and deletion mutagenesis, the binding site of the FAK C-terminal focal adhesion targeting (FAT) domain was identified within the C-terminal coiled-coil domain of p190RhoGEF. Binding was independent of a LD-like binding motif within p190RhoGEF, yet FAK association was disrupted by a mutation (Leu-1034 to Ser) that weakens the helical bundle structure of the FAK FAT domain. Neuro-2a cell binding to laminin increased endogenous FAK and p190RhoGEF tyrosine phosphorylation, and co-transfection of a dominant-negative inhibitor of FAK activity, termed FRNK, inhibited lamininstimulated p190RhoGEF tyrosine phosphorylation and p21 RhoA GTP binding. Overexpression of FAK in Neuro-2a cells increased both endogenous p190RhoGEF tyrosine phosphorylation and RhoA activity, whereas these events were inhibited by FRNK co-expression. Because insulin-like growth factor 1 treatment of Neuro-2a cells increased FAK tyrosine phosphorylation and enhanced p190RhoGEF-mediated activation of RhoA, our results support the conclusion that FAK association with p190RhoGEF functions as a signaling pathway downstream of integrins and growth factor receptors to stimulate Rho activity.


Subject(s)
Guanine Nucleotide Exchange Factors/metabolism , Nuclear Proteins/metabolism , Protein-Tyrosine Kinases/metabolism , Signal Transduction , Animals , DNA-Binding Proteins , Focal Adhesion Kinase 1 , Focal Adhesion Protein-Tyrosine Kinases , GTPase-Activating Proteins , Mice , Protein Binding , Repressor Proteins , Tumor Cells, Cultured , rhoA GTP-Binding Protein/metabolism
17.
J Biol Chem ; 277(45): 42701-5, 2002 Nov 08.
Article in English | MEDLINE | ID: mdl-12215442

ABSTRACT

The enhancement of RNA-mediated motor neuron degeneration in transgenic mice by mutating a major mRNA instability determinant in a light neurofilament (NF-L) transgene implicates cognate RNA binding factors in the pathogenesis of motor neuron degeneration. p190RhoGEF is a neuron-enriched guanine exchange factor (GEF) that binds to the NF-L-destabilizing element, to c-Jun N-terminal kinase-interactive protein-1 (JIP-1), and to 14-3-3 and may link neurofilament expression to pathways affecting neuronal homeostasis. This study was undertaken to identify additional RNA species that bind p190RhoGEF and could affect interactions of the exchange factor with NF-L transcripts. The C-terminal domain of p190RhoGEF, containing the RNA-binding site, was expressed as a glutathione S-transferase fusion protein and was used as an affinity probe to isolate interactive RNAs in rat brain extracts. As expected, NF-L mRNA was identified as an RNA specie eluted from the affinity column. In addition, BC1 RNA was also found enriched in the bound RNA fraction. BC1 is a 152-nucleotide RNA that is highly expressed but untranslated in differentiated neurons. We show that BC1 and NF-L mRNA bind to a similar site in the C-terminal domain of p190RhoGEF, and their bindings to p190RhoGEF are readily cross-competed. Moreover, we identify a novel binding site in BC1 to account for its interaction with p190RhoGEF. The findings suggest a novel role of BC1 in differentiated neurons involving RNA-protein interactions of p190RhoGEF.


Subject(s)
Brain/physiology , Guanine Nucleotide Exchange Factors/metabolism , Intermediate Filaments/metabolism , Neurofilament Proteins/genetics , Neurons/physiology , Nuclear Proteins/metabolism , RNA, Messenger/genetics , RNA, Small Cytoplasmic/genetics , Animals , Base Sequence , Binding Sites , DNA-Binding Proteins , GTPase-Activating Proteins , Glutathione Transferase/genetics , Mice , Mice, Transgenic , Molecular Sequence Data , Nerve Degeneration/genetics , RNA Probes , RNA, Messenger/metabolism , RNA, Small Cytoplasmic/chemistry , RNA, Small Cytoplasmic/metabolism , Recombinant Fusion Proteins/metabolism , Repressor Proteins , Transcription, Genetic
18.
J Neurosci ; 22(17): 7662-70, 2002 Sep 01.
Article in English | MEDLINE | ID: mdl-12196589

ABSTRACT

Studies of experimental motor neuron degeneration attributable to expression of neurofilament light chain (NF-L) transgenes have raised the possibility that the neuropathic effects result from overexpression of NF-L mRNA, independent of NF-L protein effects (Cañete-Soler et al., 1999). The present study was undertaken to test for an RNA-mediated pathogenesis. Transgenic mice were derived using either an enhanced green fluorescent protein reporter construct or modified chimeric constructs that differ only in their 3' untranslated regions (UTRs). Motor function and spinal cord histology were normal in mice expressing the unmodified reporter transgene. In mice expressing a chimeric transgene in which sequence of NF-L 3' UTR was inserted into the 3' UTR of the reporter transgene, we observed growth retardation and reduced kinetic activity during postnatal development. Older mice developed impairment of motor function and atrophy of nerve fibers in the ventral roots. A similar but more severe phenotype was observed when the chimeric transgene contained a 36 bp c-myc insert in an mRNA destabilizing element of the NF-L sequence. Our results suggest that neuropathic effects of overexpressing NF-L can occur at the level of transgene RNA and are mediated by sequences in the NF-L 3' UTR.


Subject(s)
3' Untranslated Regions/genetics , Growth Disorders/genetics , Motor Neuron Disease/physiopathology , Motor Neurons/metabolism , Neurofilament Proteins/genetics , Animals , Behavior, Animal , Body Weight/genetics , COS Cells , Genes, Reporter , Genes, myc/genetics , Growth Disorders/pathology , Luminescent Proteins/genetics , Mice , Mice, Transgenic , Motor Activity/genetics , Motor Neuron Disease/genetics , Motor Neuron Disease/pathology , Motor Neurons/drug effects , Motor Neurons/pathology , Mutation , Neurofilament Proteins/biosynthesis , Neurofilament Proteins/pharmacology , Phenotype , RNA, Messenger/genetics , RNA, Messenger/metabolism , Transgenes/physiology , Vacuoles/pathology
SELECTION OF CITATIONS
SEARCH DETAIL
...