Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 23
1.
Autophagy ; 19(4): 1277-1292, 2023 04.
Article En | MEDLINE | ID: mdl-36131358

How macroautophagy/autophagy influences neurofilament (NF) proteins in neurons, a frequent target in neurodegenerative diseases and injury, is not known. NFs in axons have exceptionally long half-lives in vivo enabling formation of large stable supporting networks, but they can be rapidly degraded during Wallerian degeneration initiated by a limited calpain cleavage. Here, we identify autophagy as a previously unrecognized pathway for NF subunit protein degradation that modulates constitutive and inducible NF turnover in vivo. Levels of NEFL/NF-L, NEFM/NF-M, and NEFH/NF-H subunits rise substantially in neuroblastoma (N2a) cells after blocking autophagy either with the phosphatidylinositol 3-kinase (PtdIns3K) inhibitor 3-methyladenine (3-MA), by depleting ATG5 expression with shRNA, or by using both treatments. In contrast, activating autophagy with rapamycin significantly lowers NF levels in N2a cells. In the mouse brain, NF subunit levels increase in vivo after intracerebroventricular infusion of 3-MA. Furthermore, using tomographic confocal microscopy, immunoelectron microscopy, and biochemical fractionation, we demonstrate the presence of NF proteins intra-lumenally within autophagosomes (APs), autolysosomes (ALs), and lysosomes (LYs). Our findings establish a prominent role for autophagy in NF proteolysis. Autophagy may regulate axon cytoskeleton size and responses of the NF cytoskeleton to injury and disease.


Autophagy , Intermediate Filaments , Mice , Animals , Autophagy/physiology , Proteolysis , Intermediate Filaments/metabolism , Neurofilament Proteins/genetics , Neurofilament Proteins/metabolism , Neurons/metabolism
2.
Autophagy ; 15(3): 543-557, 2019 03.
Article En | MEDLINE | ID: mdl-30269645

Autophagy-lysosome pathway (ALP) disruption is considered pathogenic in multiple neurodegenerative diseases; however, current methods are inadequate to investigate macroautophagy/autophagy flux in brain in vivo and its therapeutic modulation. Here, we describe a novel autophagy reporter mouse (TRGL6) stably expressing a dual-fluorescence-tagged LC3 (tfLC3, mRFP-eGFP-LC3) by transgenesis selectively in neurons. The tfLC3 probe distributes widely in the central nervous system, including spinal cord. Expression levels were similar to endogenous LC3 and induced no detectable ALP changes. This ratiometric reporter registers differential pH-dependent changes in color as autophagosomes form, fuse with lysosomes, acidify, and degrade substrates within autolysosomes. We confirmed predicted changes in neuronal autophagy flux following specific experimental ALP perturbations. Furthermore, using a third fluorescence label in TRGL6 brains to identify lysosomes by immunocytochemistry, we validated a novel procedure to detect defective autolysosomal acidification in vivo. Thus, TRGL6 mice represent a unique tool to investigate in vivo ALP dynamics in specific neuron populations in relation to neurological diseases, aging, and disease modifying agents. Abbreviations: ACTB: actin, beta; AD: Alzheimer disease; AL: autolysosomes; ALP: autophagy-lysosome pathway; AP: autophagosome; APP: amyloid beta (Abeta) precursor protein; ATG5: autophagy related 5; ATG7: autophagy related 7; AV: autophagic vacuoles; CNS: central nervous system; CTSD: cathepsin D; CQ: chloroquine; DMEM: Dulbecco's modified Eagle's medium; GFP: green fluorescent protein; GABARAP: gamma-aminobutyric acid receptor associated protein; GABARAPL2/GATE16: gamma-aminobutyric acid (GABA) receptor-associated protein-like 2; ICC: immunocytochemistry; ICV: intra-cerebroventricular; LAMP2: lysosomal-associated membrane protein 2; Leup: leupeptin; LY: lysosomes; MAP1LC3/LC3: microtubule-associated protein 1 light chain 3; MTOR: mechanistic target of rapamycin kinase; RBFOX3/NeuN: RNA binding protein, fox-1 homolog (C. elegans) 3; RFP: red fluorescent protein; RPS6KB1: ribosomal protein S6 kinase, polypeptide 1; SDS-PAGE: sodium dodecyl sulfate-polyacrylamide gel electrophoresis; SQSTM1: sequestosome 1; tfLC3: mRFP-eGFP-LC3; TRGL6: Thy1 mRFP eGFP LC3-line 6; PCR: polymerase chain reaction; PD: Parkinson disease.


Autophagy , Brain/metabolism , Lysosomes/chemistry , Microtubule-Associated Proteins/genetics , Neurons/metabolism , Animals , Autophagosomes/drug effects , Autophagosomes/metabolism , Autophagy/drug effects , Autophagy/genetics , Brain/cytology , Brain Chemistry , Cells, Cultured , Chloroquine/pharmacology , Fluorescent Dyes , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Luminescent Proteins/genetics , Luminescent Proteins/metabolism , Lysosomes/drug effects , Lysosomes/metabolism , Mice , Mice, Inbred C57BL , Mice, Transgenic , Microtubule-Associated Proteins/metabolism , Morpholines/pharmacology , Neurons/chemistry , Neurons/cytology , Neurons/drug effects , Red Fluorescent Protein
3.
Transl Psychiatry ; 8(1): 167, 2018 08 24.
Article En | MEDLINE | ID: mdl-30143609

Neurofilament (NFL) proteins have recently been found to play unique roles in synapses. NFL is known to interact with the GluN1 subunit of N-methyl-D-aspartic acid (NMDAR) and be reduced in schizophrenia though functional consequences are unknown. Here we investigated whether the interaction of NFL with GluN1 modulates synaptic transmission and schizophrenia-associated behaviors. The interaction of NFL with GluN1 was assessed by means of molecular, pharmacological, electrophysiological, magnetic resonance spectroscopy (MRS), and schizophrenia-associated behavior analyses. NFL deficits cause an NMDAR hypofunction phenotype including abnormal hippocampal function, as seen in schizophrenia. NFL-/- deletion in mice reduces dendritic spines and GluN1 protein levels, elevates ubiquitin-dependent turnover of GluN1 and hippocampal glutamate measured by MRS, and depresses hippocampal long-term potentiation. NMDAR-related behaviors are also impaired, including pup retrieval, spatial and social memory, prepulse inhibition, night-time activity, and response to NMDAR antagonist, whereas motor deficits are minimal. Importantly, partially lowering NFL in NFL+/- mice to levels seen regionally in schizophrenia, induced similar but milder NMDAR-related synaptic and behavioral deficits. Our findings support an emerging view that central nervous system neurofilament subunits including NFL in the present report, serve distinctive, critical roles in synapses relevant to neuropsychiatric diseases.


Dendritic Spines/metabolism , Nerve Tissue Proteins/physiology , Neurofilament Proteins/physiology , Receptors, N-Methyl-D-Aspartate/physiology , Synapses/physiology , Synaptic Transmission/physiology , Animals , Behavior, Animal , Female , Glutamic Acid/metabolism , Hippocampus/pathology , Magnetic Resonance Spectroscopy , Male , Mice , Mice, Knockout
4.
Article En | MEDLINE | ID: mdl-28373358

SUMMARYNeurofilaments (NFs) are unique among tissue-specific classes of intermediate filaments (IFs) in being heteropolymers composed of four subunits (NF-L [neurofilament light]; NF-M [neurofilament middle]; NF-H [neurofilament heavy]; and α-internexin or peripherin), each having different domain structures and functions. Here, we review how NFs provide structural support for the highly asymmetric geometries of neurons and, especially, for the marked radial expansion of myelinated axons crucial for effective nerve conduction velocity. NFs in axons extensively cross-bridge and interconnect with other non-IF components of the cytoskeleton, including microtubules, actin filaments, and other fibrous cytoskeletal elements, to establish a regionally specialized network that undergoes exceptionally slow local turnover and serves as a docking platform to organize other organelles and proteins. We also discuss how a small pool of oligomeric and short filamentous precursors in the slow phase of axonal transport maintains this network. A complex pattern of phosphorylation and dephosphorylation events on each subunit modulates filament assembly, turnover, and organization within the axonal cytoskeleton. Multiple factors, and especially turnover rate, determine the size of the network, which can vary substantially along the axon. NF gene mutations cause several neuroaxonal disorders characterized by disrupted subunit assembly and NF aggregation. Additional NF alterations are associated with varied neuropsychiatric disorders. New evidence that subunits of NFs exist within postsynaptic terminal boutons and influence neurotransmission suggests how NF proteins might contribute to normal synaptic function and neuropsychiatric disease states.


Intermediate Filaments/physiology , Neurofilament Proteins/physiology , Animals , Biomarkers/metabolism , Humans , Intermediate Filaments/metabolism , Mental Disorders/physiopathology , Neurofilament Proteins/metabolism , Organelles/metabolism , Protein Processing, Post-Translational
5.
J Neurochem ; 137(2): 253-65, 2016 Apr.
Article En | MEDLINE | ID: mdl-26756888

Amyotrophic lateral sclerosis (ALS) is a progressive motor neuron disease with a poorly understood cause and no effective treatment. Given that calpains mediate neurodegeneration in other pathological states and are abnormally activated in ALS, we investigated the possible ameliorative effects of inhibiting calpain over-activation in hSOD1(G93A) transgenic (Tg) mice in vivo by neuron-specific over-expression of calpastatin (CAST), the highly selective endogenous inhibitor of calpains. Our data indicate that over-expression of CAST in hSOD1(G93A) mice, which lowered calpain activation to levels comparable to wild-type mice, inhibited the abnormal breakdown of cytoskeletal proteins (spectrin, MAP2 and neurofilaments), and ameliorated motor axon loss. Disease onset in hSOD1(G93A) /CAST mice compared to littermate hSOD1(G93A) mice is delayed, which accounts for their longer time of survival. We also find that neuronal over-expression of CAST in hSOD1(G93A) transgenic mice inhibited production of putative neurotoxic caspase-cleaved tau and activation of Cdk5, which have been implicated in neurodegeneration in ALS models, and also reduced the formation of SOD1 oligomers. Our data indicate that inhibition of calpain with CAST is neuroprotective in an ALS mouse model. CAST (encoding calpastatin) inhibits hyperactivated calpain to prevent motor neuron disease operating through a cascade of events as indicated in the schematic, with relevance to amyotrophic lateral sclerosis (ALS). We propose that over-expression of CAST in motor neurons of hSOD1(G93A) mice inhibits activation of CDK5, breakdown of cytoskeletal proteins (NFs, MAP2 and Tau) and regulatory molecules (Cam Kinase IV, Calcineurin A), and disease-causing proteins (TDP-43, α-Synuclein and Huntingtin) to prevent neuronal loss and delay neurological deficits. In our experiments, CAST could also inhibit cleavage of Bid, Bax, AIF to prevent mitochondrial, ER and lysosome-mediated cell death mechanisms. Similarly, CAST over-expression in neurons attenuated pathological effects of TDP-43, α-synuclein and Huntingtin. These results suggest a potential value of specific small molecule inhibitors of calpains in delaying the development of ALS. Read the Editorial Highlight for this article on page 140.


Calcium-Binding Proteins/pharmacology , Cysteine Proteinase Inhibitors/pharmacology , Motor Neurons/drug effects , Superoxide Dismutase/genetics , Age Factors , Amyotrophic Lateral Sclerosis/drug therapy , Amyotrophic Lateral Sclerosis/genetics , Amyotrophic Lateral Sclerosis/pathology , Animals , Axons/pathology , Calpain/metabolism , Cell Death/drug effects , Cell Death/genetics , Cyclin-Dependent Kinase 5/metabolism , Cytoskeletal Proteins/metabolism , Disease Models, Animal , Disease Progression , Gene Expression Regulation , Humans , Mice , Mice, Transgenic , Motor Activity/drug effects , Motor Activity/genetics , Motor Neurons/cytology , Motor Neurons/metabolism , Nerve Degeneration/drug therapy , Nerve Degeneration/genetics , Nerve Degeneration/pathology , Spinal Cord/cytology
6.
PLoS One ; 10(7): e0133848, 2015.
Article En | MEDLINE | ID: mdl-26208164

The axonal cytoskeleton of neurofilament (NF) is a long-lived network of fibrous elements believed to be a stationary structure maintained by a small pool of transported cytoskeletal precursors. Accordingly, it may be predicted that NF content in axons can vary independently from the transport rate of NF. In the present report, we confirm this prediction by showing that human NFH transgenic mice and transgenic mice expressing human NFL Ser55 (Asp) develop nearly identical abnormal patterns of NF accumulation and distribution in association with opposite changes in NF slow transport rates. We also show that the rate of NF transport in wild-type mice remains constant along a length of the optic axon where NF content varies 3-fold. Moreover, knockout mice lacking NFH develop even more extreme (6-fold) proximal to distal variation in NF number, which is associated with a normal wild-type rate of NF transport. The independence of regional NF content and NF transport is consistent with previous evidence suggesting that the rate of incorporation of transported NF precursors into a metabolically stable stationary cytoskeletal network is the major determinant of axonal NF content, enabling the generation of the striking local variations in NF number seen along axons.


Axonal Transport , Axons/metabolism , Intermediate Filaments/metabolism , Animals , Female , Humans , Male , Mice , Mice, Knockout , Mice, Transgenic , Optic Nerve/metabolism , Phosphorylation
7.
J Neurosci ; 34(28): 9222-34, 2014 Jul 09.
Article En | MEDLINE | ID: mdl-25009256

Tau pathogenicity in Alzheimer's disease and other tauopathies is thought to involve the generation of hyperphosphorylated, truncated, and oligomeric tau species with enhanced neurotoxicity, although the generative mechanisms and the implications for disease therapy are not well understood. Here, we report a striking rescue from mutant tau toxicity in the JNPL3 mouse model of tauopathy. We show that pathological activation of calpains gives rise to a range of potentially toxic forms of tau, directly, and by activating cdk5. Calpain overactivation in brains of these mice is accelerated as a result of the marked depletion of the endogenous calpain inhibitor, calpastatin. When levels of this inhibitor are restored in neurons of JNPL3 mice by overexpressing calpastatin, tauopathy is prevented, including calpain-mediated breakdown of cytoskeletal proteins, cdk5 activation, tau hyperphosphorylation, formation of potentially neurotoxic tau fragments by either calpain or caspase-3, and tau oligomerization. Calpastatin overexpression also prevents loss of motor axons, delays disease onset, and extends survival of JNPL3 mice by 3 months to within the range of normal lifespan. Our findings support the therapeutic promise of highly specific calpain inhibition in the treatment of tauopathies and other neurodegenerative states.


Behavior, Animal/drug effects , Calcium-Binding Proteins/administration & dosage , Calpain/antagonists & inhibitors , Longevity/drug effects , Tauopathies/prevention & control , Tauopathies/physiopathology , Animals , Calpain/metabolism , Cysteine Proteinase Inhibitors/administration & dosage , Female , Male , Mice , Mice, Transgenic , Nerve Degeneration/pathology , Nerve Degeneration/physiopathology , Nerve Degeneration/prevention & control , Survival Rate , Tauopathies/pathology , Treatment Outcome , tau Proteins/drug effects , tau Proteins/genetics
8.
Hum Mol Genet ; 23(15): 3975-89, 2014 Aug 01.
Article En | MEDLINE | ID: mdl-24619358

Lewy bodies, a pathological hallmark of Parkinson's disease (PD), contain aggregated alpha-synuclein (αSyn), which is found in several modified forms and can be discovered phosphorylated, ubiquitinated and truncated. Aggregation-prone truncated species of αSyn caused by aberrant cleavage of this fibrillogenic protein are hypothesized to participate in its sequestration into inclusions subsequently leading to synaptic dysfunction and neuronal death. Here, we investigated the role of calpain cleavage of αSyn in vivo by generating two opposing mouse models. We crossed into human [A30P]αSyn transgenic (i) mice deficient for calpastatin, a calpain-specific inhibitor, thus enhancing calpain activity (SynCAST(-)) and (ii) mice overexpressing human calpastatin leading to reduced calpain activity (SynCAST(+)). As anticipated, a reduced calpain activity led to a decreased number of αSyn-positive aggregates, whereas loss of calpastatin led to increased truncation of αSyn in SynCAST(-). Furthermore, overexpression of calpastatin decreased astrogliosis and the calpain-dependent degradation of synaptic proteins, potentially ameliorating the observed neuropathology in [A30P]αSyn and SynCAST(+) mice. Overall, our data further support a crucial role of calpains, particularly of calpain 1, in the pathogenesis of PD and in disease-associated aggregation of αSyn, indicating a therapeutic potential of calpain inhibition in PD.


Calcium-Binding Proteins/genetics , Calpain/genetics , Parkinson Disease/genetics , Protein Aggregation, Pathological/genetics , alpha-Synuclein/genetics , Animals , Calcium-Binding Proteins/metabolism , Calpain/antagonists & inhibitors , Calpain/metabolism , Disease Models, Animal , Gene Expression Regulation , Humans , Lewy Bodies/metabolism , Lewy Bodies/pathology , Mice , Mice, Transgenic , Neurons/metabolism , Neurons/pathology , Parkinson Disease/metabolism , Parkinson Disease/pathology , Protein Aggregates , Protein Aggregation, Pathological/metabolism , Protein Aggregation, Pathological/pathology , Proteolysis , Signal Transduction , Synapses/metabolism , Synapses/pathology , alpha-Synuclein/metabolism
9.
PLoS One ; 7(9): e44320, 2012.
Article En | MEDLINE | ID: mdl-23028520

Newly synthesized neurofilaments or protofilaments are incorporated into a highly stable stationary cytoskeleton network as they are transported along axons. Although the heavily phosphorylated carboxyl-terminal tail domains of the heavy and medium neurofilament (NF) subunits have been proposed to contribute to this process and particularly to stability of this structure, their function is still obscure. Here we show in NF-H/M tail deletion [NF-(H/M)(tailΔ)] mice that the deletion of both of these domains selectively lowers NF levels 3-6 fold along optic axons without altering either rates of subunit synthesis or the rate of slow axonal transport of NF. Pulse labeling studies carried out over 90 days revealed a significantly faster rate of disappearance of NF from the stationary NF network of optic axons in NF-(H/M)(tailΔ) mice. Faster NF disappearance was accompanied by elevated levels of NF-L proteolytic fragments in NF-(H/M)(tailΔ) axons. We conclude that NF-H and NF-M C-terminal domains do not normally regulate NF transport rates as previously proposed, but instead increase the proteolytic resistance of NF, thereby stabilizing the stationary neurofilament cytoskeleton along axons.


Axons/metabolism , Neurofilament Proteins/metabolism , Animals , Female , Immunoblotting , Male , Mice , Neurofilament Proteins/genetics , Retinal Ganglion Cells/metabolism
11.
J Neurosci ; 32(25): 8501-8, 2012 Jun 20.
Article En | MEDLINE | ID: mdl-22723690

Peripherin, a neuronal intermediate filament protein implicated in neurodegenerative disease, coexists with the neurofilament triplet proteins [neurofilament light (NFL), medium (NFM), and heavy (NFH) chain] but has an unknown function. The earlier peak expression of peripherin than the triplet during brain development and its ability to form homopolymers, unlike the triplet, which are obligate heteropolymers, have supported a widely held view that peripherin and neurofilament triplets form separate filament systems. However, here, we demonstrate that, despite a postnatal decline in expression, peripherin is as abundant as the triplet in the adult PNS and exists in a relatively fixed stoichiometry with these subunits. Peripherin exhibits a distribution pattern identical to those of triplet proteins in sciatic axons and colocalizes with NFL on single neurofilaments by immunogold electron microscopy. Peripherin also coassembles into a single network of filaments containing NFL, NFM, and NFH with and without α-internexin in quadruple- or quintuple-transfected SW13vim(-) cells. Genetically deleting NFL in mice dramatically reduces peripherin content in sciatic axons. Moreover, peripherin mutations has been shown to disrupt the neurofilament network in transfected SW13vim(-) cells. These data show that peripherin and the neurofilament proteins are functionally interdependent. The results strongly support the view that, rather than forming an independent structure, peripherin is a subunit of neurofilaments in the adult PNS. Our findings provide a basis for its close relationship with neurofilaments in PNS diseases associated with neurofilament accumulation.


Axons/metabolism , Central Nervous System/metabolism , Intermediate Filament Proteins/metabolism , Membrane Glycoproteins/metabolism , Nerve Tissue Proteins/metabolism , Neurofilament Proteins/metabolism , Peripheral Nervous System/metabolism , Animals , Antibodies, Monoclonal , Axons/ultrastructure , Blotting, Western , Central Nervous System/cytology , Central Nervous System/ultrastructure , Electrophoresis, Polyacrylamide Gel , Female , Fluorescent Antibody Technique , Immunohistochemistry , Male , Mice , Mice, Inbred C57BL , Microscopy, Immunoelectron , Peripheral Nervous System/cytology , Peripheral Nervous System/ultrastructure , Peripherins , Sciatic Nerve/cytology , Sciatic Nerve/metabolism , Transfection
12.
Neurobiol Aging ; 33(6): 1125.e9-18, 2012 Jun.
Article En | MEDLINE | ID: mdl-22206846

We report that neuronal overexpression of the endogenous inhibitor of calpains, calpastatin (CAST), in a mouse model of human Alzheimer's disease (AD) ß-amyloidosis, the APP23 mouse, reduces ß-amyloid (Aß) pathology and Aß levels when comparing aged, double transgenic (tg) APP23/CAST with APP23 mice. Concurrent with Aß plaque deposition, aged APP23/CAST mice show a decrease in the steady-state brain levels of the amyloid precursor protein (APP) and APP C-terminal fragments (CTFs) when compared with APP23 mice. This CAST-dependent decrease in APP metabolite levels was not observed in single tg CAST mice expressing endogenous APP or in younger, Aß plaque predepositing APP23/CAST mice. We also determined that the CAST-mediated inhibition of calpain activity in the brain is greater in the CAST mice with Aß pathology than in non-APP tg mice, as demonstrated by a decrease in calpain-mediated cytoskeleton protein cleavage. Moreover, aged APP23/CAST mice have reduced extracellular signal-regulated kinase 1/2 (ERK1/2) activity and tau phosphorylation when compared with APP23 mice. In summary, in vivo calpain inhibition mediated by CAST transgene expression reduces Aß pathology in APP23 mice, with our findings further suggesting that APP metabolism is modified by CAST overexpression as the mice develop Aß pathology. Our results indicate that the calpain system in neurons is more responsive to CAST inhibition under conditions of Aß pathology, suggesting that in the disease state neurons may be more sensitive to the therapeutic use of calpain inhibitors.


Amyloid beta-Peptides/metabolism , Amyloid beta-Protein Precursor/metabolism , Brain/metabolism , Calcium-Binding Proteins/physiology , Amyloid beta-Peptides/genetics , Amyloid beta-Protein Precursor/genetics , Animals , Brain/pathology , Female , Humans , Mice , Mice, Inbred C57BL , Mice, Transgenic
13.
PLoS One ; 6(2): e17087, 2011 Feb 16.
Article En | MEDLINE | ID: mdl-21359212

The neurofilament light subunit (NF-L) binds to myosin Va (Myo Va) in neurons but the sites of interaction and functional significance are not clear. We show by deletion analysis that motor domain of Myo Va binds to the NF-L rod domain that forms the NF backbone. Loss of NF-L and Myo Va binding from axons significantly reduces the axonal content of ER, and redistributes ER to the periphery of axon. Our data are consistent with a novel function for NFs as a scaffold in axons for maintaining the content and proper distribution of vesicular organelles, mediated in part by Myo Va. Based on observations that the Myo Va motor domain binds to intermediate filament (IF) proteins of several classes, Myo Va interactions with IFs may serve similar roles in organizing organelle topography in different cell types.


Axons/metabolism , Endoplasmic Reticulum/metabolism , Myosin Heavy Chains/chemistry , Myosin Heavy Chains/metabolism , Myosin Heavy Chains/physiology , Myosin Type V/chemistry , Myosin Type V/metabolism , Myosin Type V/physiology , Neurofilament Proteins/metabolism , Animals , Axons/physiology , Intermediate Filaments/metabolism , Intermediate Filaments/physiology , Mice , Mice, Inbred C57BL , Mice, Knockout , Myosin Heavy Chains/genetics , Myosin Type V/genetics , Neurons/metabolism , Neurons/physiology , Protein Binding/physiology , Protein Structure, Tertiary/genetics , Protein Structure, Tertiary/physiology , Tissue Distribution
14.
J Neurosci ; 29(36): 11316-29, 2009 Sep 09.
Article En | MEDLINE | ID: mdl-19741138

The ultrastructural view of the axonal cytoskeleton as an extensively cross-linked network of neurofilaments (NFs) and other cytoskeletal polymers contrasts with the dynamic view suggested by axonal transport studies on cytoskeletal elements. Here we reconcile these perspectives by showing that neurons form a large NF network along axons which is unequivocally stationary, metabolically stable, and maintained by NFs and nonfilamentous subunit assemblies undergoing slow transport by intermittent rapid movements and pauses. In mouse primary cortical neurons transfected with EGFP-NFL, formation of this stationary NF network requires a critical level of NFs, which explains its absence in NF-poor developing neurons studied previously. Most NFs at proximal axon regions were in a stationary structure coexisting with a smaller pool of moving EGFP-NFL assemblies that were mainly nonfilamentous. Distally along the same axon, EGFP-labeled NFL was much less abundant, and we detected only short filaments moving bidirectionally by slow transport (rapid movements and pauses) as previously described. In living mice, >25% of radiolabeled newly synthesized NFs remained in optic axons after slowly transported NFs had exited. Retained NF remained fixed over several months in a nonuniform distribution and exhibited exceptionally slow turnover (t(1/2) >2.5 months), implying that, at steady state, >90% of NFs in mature optic axons comprise the stationary cytoskeleton and <10% are undergoing slow transport. These findings reconcile in vitro and in vivo axonal transport observations, showing that slowly transported NFs or subunit oligomers are precursors to a highly stable stationary cytoskeletal network that supports mature axons.


Axons/physiology , Cytoskeleton/physiology , Intermediate Filaments/physiology , Nerve Tissue Proteins/physiology , Animals , Axons/chemistry , Cells, Cultured , Cytoskeleton/chemistry , Intermediate Filaments/chemistry , Mice , Mice, Inbred C57BL , Nerve Tissue Proteins/analysis , Rats
15.
J Neurosci ; 29(5): 1277-84, 2009 Feb 04.
Article En | MEDLINE | ID: mdl-19193875

Neurofilament medium (NF-M) is essential for the acquisition of normal axonal caliber in response to a myelin-dependent "outside-in" trigger for radial axonal growth. Removal of the tail domain and lysine-serine-proline (KSP) repeats of NF-M, but not neurofilament heavy, produced axons with impaired radial growth and reduced conduction velocities. These earlier findings supported myelin-dependent phosphorylation of NF-M KSP repeats as an essential component of axonal growth. As a direct test of whether phosphorylation of NF-M KSP repeats is the target for the myelin-derived signal, gene replacement has now been used to produce mice in which all serines of NF-M's KSP repeats have been replaced with phosphorylation-incompetent alanines. This substitution did not alter accumulation of the neurofilaments or their subunits. Axonal caliber and motor neuron conduction velocity of mice expressing KSP phospho-incompetent NF-M were also indistinguishable from wild-type mice. Thus, phosphorylation of NF-M KSP repeats is not an essential component for the acquisition of normal axonal caliber mediated by myelin-dependent outside-in signaling.


Axons/physiology , Conserved Sequence , Lysine , Myelin Sheath/physiology , Neurofilament Proteins/physiology , Proline , Repetitive Sequences, Amino Acid , Serine , Alanine/genetics , Amino Acid Sequence , Amino Acid Substitution/genetics , Animals , Axons/metabolism , Axons/ultrastructure , Conserved Sequence/genetics , Gene Knock-In Techniques , Lysine/metabolism , Mice , Mice, Neurologic Mutants , Molecular Sequence Data , Myelin Sheath/genetics , Myelin Sheath/metabolism , Nerve Crush , Neural Pathways/physiology , Neurofilament Proteins/genetics , Neurofilament Proteins/metabolism , Phosphorylation/genetics , Proline/metabolism , Repetitive Sequences, Amino Acid/genetics , Serine/genetics
16.
J Neurosci ; 28(47): 12241-54, 2008 Nov 19.
Article En | MEDLINE | ID: mdl-19020018

Increased activity of calpains is implicated in synaptic dysfunction and neurodegeneration in Alzheimer's disease (AD). The molecular mechanisms responsible for increased calpain activity in AD are not known. Here, we demonstrate that disease progression is propelled by a marked depletion of the endogenous calpain inhibitor, calpastatin (CAST), from AD neurons, which is mediated by caspase-1, caspase-3, and calpains. Initial CAST depletion focally along dendrites coincides topographically with calpain II and ERK 1/2 activation, tau cleavage by caspase-3, and tau and neurofilament hyperphosphorylation. These same changes, together with cytoskeletal proteolysis and neuronal cell death, accompany CAST depletion after intrahippocampal kainic acid administration to mice, and are substantially reduced in mice overexpressing human CAST. Moreover, CAST reduction by shRNA in neuronal cells causes calpain-mediated death at levels of calcium-induced injury that are sublethal to cells normally expressing CAST. Our results strongly support a novel hypothesis that CAST depletion by multiple abnormally activated proteases accelerates calpain dysregulation in AD leading to cytoskeleton disruption and neurodegeneration. CAST mimetics may, therefore, be neuroprotective in AD.


Alzheimer Disease/metabolism , Calcium-Binding Proteins/metabolism , Cytoskeleton/metabolism , Nerve Degeneration/etiology , Aged , Aged, 80 and over , Alzheimer Disease/pathology , Animals , Brain/metabolism , Brain/pathology , Calcium-Binding Proteins/genetics , Calpain/metabolism , Case-Control Studies , Caspases/metabolism , Cell Death/drug effects , Cell Death/physiology , Cell Line, Transformed , Excitatory Amino Acid Agonists/toxicity , Extracellular Signal-Regulated MAP Kinases/metabolism , Female , Gene Expression Regulation/drug effects , Gene Expression Regulation/radiation effects , Hippocampus/drug effects , Humans , Kainic Acid/toxicity , Male , Mice , Mice, Transgenic , Middle Aged , Nerve Degeneration/metabolism , Nerve Degeneration/pathology , Postmortem Changes , RNA, Small Interfering/pharmacology , Transfection/methods
17.
J Neurosci ; 26(39): 10006-19, 2006 Sep 27.
Article En | MEDLINE | ID: mdl-17005864

Alpha-internexin, a neuronal intermediate filament protein implicated in neurodegenerative disease, coexists with the neurofilament (NF) triplet proteins (NF-L, NF-M, and NF-H) but has an unknown function. The earlier peak expression of alpha-internexin than the triplet during brain development and its ability to form homopolymers, unlike the triplet, which are obligate heteropolymers, have supported a widely held view that alpha-internexin and neurofilament triplet form separate filament systems. Here, we demonstrate, however, that despite a postnatal decline in expression, alpha-internexin is as abundant as the triplet in the adult CNS and exists in a relatively fixed stoichiometry with these subunits. Alpha-internexin exhibits transport and turnover rates identical to those of triplet proteins in optic axons and colocalizes with NF-M on single neurofilaments by immunogold electron microscopy. Alpha-internexin also coassembles with all three neurofilament proteins into a single network of filaments in quadruple-transfected SW13vim(-) cells. Genetically deleting NF-M alone or together with NF-H in mice dramatically reduces alpha-internexin transport and content in axons throughout the CNS. Moreover, deleting alpha-internexin potentiates the effects of NF-M deletion on NF-H and NF-L transport. Finally, overexpressing a NF-H-LacZ fusion protein in mice induces alpha-internexin and neurofilament triplet to aggregate in neuronal perikarya and greatly reduces their transport and content selectively in axons. Our data show that alpha-internexin and the neurofilament proteins are functionally interdependent. The results strongly support the view that alpha-internexin is a fourth subunit of neurofilaments in the adult CNS, providing a basis for its close relationship with neurofilaments in CNS diseases associated with neurofilament accumulation.


Axons/chemistry , Intermediate Filament Proteins/physiology , Intermediate Filaments/chemistry , Neurofilament Proteins/physiology , Animals , Axons/ultrastructure , Crosses, Genetic , Female , Intermediate Filament Proteins/analysis , Intermediate Filament Proteins/deficiency , Intermediate Filament Proteins/genetics , Intermediate Filament Proteins/ultrastructure , Intermediate Filaments/ultrastructure , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Microscopy, Confocal , Microscopy, Fluorescence , Microscopy, Immunoelectron , Multiprotein Complexes , Nerve Degeneration/metabolism , Nerve Degeneration/pathology , Neurofilament Proteins/analysis , Neurofilament Proteins/deficiency , Neurofilament Proteins/genetics , Neurofilament Proteins/ultrastructure , Protein Interaction Mapping , Protein Transport , Rats , Recombinant Fusion Proteins/analysis , Recombinant Fusion Proteins/physiology , Retinal Ganglion Cells/chemistry , Retinal Ganglion Cells/ultrastructure , Spinal Cord/chemistry , Spinal Cord/ultrastructure , Structure-Activity Relationship , Transfection
18.
Neurosci Lett ; 393(2-3): 264-8, 2006 Jan 30.
Article En | MEDLINE | ID: mdl-16266786

Phosphorylation of the carboxyl tail domains of the neurofilament heavy (NF-H) and middle molecular weight (NF-M) subunits has been proposed to regulate the axonal transport of neurofilaments. To test this hypothesis, we recently constructed mice lacking the extensively phosphorylated NF-H tail domain (NF-HtailDelta) and showed that the transport rate of neurofilaments in optic axons is unaltered in the absence of this domain [M.V. Rao, M.L. Garcia, Y. Miyazaki, T. Gotow, A. Yuan, S. Mattina, C.M. Ward, N.A. Calcutt, Y. Uchiyama, R.A. Nixon, D.W. Cleveland, Gene replacement in mice reveals that the heavily phosphorylated tail of neurofilament heavy subunit does not affect axonal caliber or the transit of cargoes in slow axonal transport, J. Cell Biol. 158 (2002) 681-693]. However, Shea et al. proposed that deletion of NF-H carboxyl-terminal region accelerates the transport of a subpopulation of neurofilaments based on minor differences between tail-deleted and control mice in our axonal transport analysis. Here, we present additional evidence that neurofilament transport rate is unchanged after deleting the phosphorylated NF-H tail domain, establishing unequivocally that the NF-H tail domain alone does not regulate the rate of neurofilament transport in optic axons in vivo. Possible roles for tail domains as cross-bridges between a neurofilament and its neighbors or other cytoskeletal elements is discussed.


Axonal Transport/physiology , Gene Deletion , Neurofilament Proteins/metabolism , Protein Subunits/metabolism , Animals , Axonal Transport/genetics , Methionine/metabolism , Mice , Mice, Transgenic , Neurofilament Proteins/chemistry , Neurofilament Proteins/genetics , Phosphorylation , Protein Structure, Tertiary , Protein Subunits/chemistry , Protein Subunits/genetics , Retinal Ganglion Cells/drug effects , Retinal Ganglion Cells/metabolism , Sulfur Isotopes/metabolism , Time Factors
19.
J Cell Biol ; 163(5): 1021-31, 2003 Dec 08.
Article En | MEDLINE | ID: mdl-14662746

The phosphorylated carboxyl-terminal "tail" domains of the neurofilament (NF) subunits, NF heavy (NF-H) and NF medium (NF-M) subunits, have been proposed to regulate axon radial growth, neurofilament spacing, and neurofilament transport rate, but direct in vivo evidence is lacking. Because deletion of the tail domain of NF-H did not alter these axonal properties (Rao, M.V., M.L. Garcia, Y. Miyazaki, T. Gotow, A. Yuan, S. Mattina, C.M. Ward, N.S. Calcutt, Y. Uchiyama, R.A. Nixon, and D.W. Cleveland. 2002. J. Cell Biol. 158:681-693), we investigated possible functions of the NF-M tail domain by constructing NF-M tail-deleted (NF-MtailDelta) mutant mice using an embryonic stem cell-mediated "gene knockin" approach that preserves normal ratios of the three neurofilament subunits. Mutant NF-MtailDelta mice exhibited severely inhibited radial growth of both motor and sensory axons. Caliber reduction was accompanied by reduced spacing between neurofilaments and loss of long cross-bridges with no change in neurofilament protein content. These observations define distinctive functions of the NF-M tail in regulating axon caliber by modulating the organization of the neurofilament network within axons. Surprisingly, the average rate of axonal transport of neurofilaments was unaltered despite these substantial effects on axon morphology. These results demonstrate that NF-M tail-mediated interactions of neurofilaments, independent of NF transport rate, are critical determinants of the size and cytoskeletal architecture of axons, and are mediated, in part, by the highly phosphorylated tail domain of NF-M.


Axons/metabolism , Cell Division/physiology , Cytoskeleton/metabolism , Neurofilament Proteins/metabolism , Protein Subunits/metabolism , Animals , Axonal Transport/physiology , Axons/ultrastructure , Cell Survival , Gene Targeting , Mice , Mice, Transgenic , Microtubules/metabolism , Molecular Weight , Neurofilament Proteins/chemistry , Neurofilament Proteins/genetics , Protein Structure, Tertiary , Protein Subunits/chemistry , Protein Subunits/genetics
20.
J Neurosci ; 23(28): 9452-8, 2003 Oct 15.
Article En | MEDLINE | ID: mdl-14561875

Neurofilament assembly requires at minimum the polymerization of neurofilament light chain (NF-L) with either neurofilament medium chain (NF-M) or neurofilament heavy chain (NF-H) subunits, but requirements for their axonal transport have long been controversial. Using a gene deletion approach, we generated mice containing only NF-L or NF-M. In vivo pulse radiolabeling analyses in retinal ganglion cell neurons revealed that NF-L alone is incapable of efficient transport, whereas nearly one-half of the normal level of NF-M is transported along optic axons in the absence of the other triplet subunits. Under these conditions, however, NF-M transport is completely abolished by deleting alpha-internexin. Our results strongly suggest that efficient neurofilament protein transport in vivo minimally requires hetero-oligomer formation. They also show that NF-M can partner with intermediate filament proteins other than the NF-H and NF-L subunits in neurons to support slow transport and possibly other functions of neuronal intermediate filaments.


Neurofilament Proteins/metabolism , Animals , Axonal Transport/physiology , Axons/metabolism , Axons/ultrastructure , Carrier Proteins/genetics , Carrier Proteins/metabolism , Female , Intermediate Filament Proteins , Intermediate Filaments/metabolism , Intermediate Filaments/ultrastructure , Macromolecular Substances , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Neurofilament Proteins/deficiency , Neurofilament Proteins/genetics , Protein Subunits/metabolism , Protein Transport/physiology
...