Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 58
Filter
1.
Cell ; 177(2): 272-285.e16, 2019 04 04.
Article in English | MEDLINE | ID: mdl-30853216

ABSTRACT

Proper brain function requires high-precision neuronal expansion and wiring, processes controlled by the transmembrane Roundabout (Robo) receptor family and their Slit ligands. Despite their great importance, the molecular mechanism by which Robos' switch from "off" to "on" states remains unclear. Here, we report a 3.6 Å crystal structure of the intact human Robo2 ectodomain (domains D1-8). We demonstrate that Robo cis dimerization via D4 is conserved through hRobo1, 2, and 3 and the C. elegans homolog SAX-3 and is essential for SAX-3 function in vivo. The structure reveals two levels of auto-inhibition that prevent premature activation: (1) cis blocking of the D4 dimerization interface and (2) trans interactions between opposing Robo receptors that fasten the D4-blocked conformation. Complementary experiments in mouse primary neurons and C. elegans support the auto-inhibition model. These results suggest that Slit stimulation primarily drives the release of Robo auto-inhibition required for dimerization and activation.


Subject(s)
Receptors, Immunologic/metabolism , Receptors, Immunologic/ultrastructure , Animals , Axons/metabolism , COS Cells , Caenorhabditis elegans/metabolism , Carrier Proteins , Chlorocebus aethiops , HEK293 Cells , Humans , Mice , Mice, Inbred ICR , Nerve Tissue Proteins/metabolism , Neurons/metabolism , Primary Cell Culture , Signal Transduction , Roundabout Proteins
2.
EMBO J ; 40(17): e107586, 2021 09 01.
Article in English | MEDLINE | ID: mdl-34190355

ABSTRACT

Amyotrophic lateral sclerosis (ALS) is a fatal non-cell-autonomous neurodegenerative disease characterized by the loss of motor neurons (MNs). Mutations in CRMP4 are associated with ALS in patients, and elevated levels of CRMP4 are suggested to affect MN health in the SOD1G93A -ALS mouse model. However, the mechanism by which CRMP4 mediates toxicity in ALS MNs is poorly understood. Here, by using tissue from human patients with sporadic ALS, MNs derived from C9orf72-mutant patients, and the SOD1G93A -ALS mouse model, we demonstrate that subcellular changes in CRMP4 levels promote MN loss in ALS. First, we show that while expression of CRMP4 protein is increased in cell bodies of ALS-affected MN, CRMP4 levels are decreased in the distal axons. Cellular mislocalization of CRMP4 is caused by increased interaction with the retrograde motor protein, dynein, which mediates CRMP4 transport from distal axons to the soma and thereby promotes MN loss. Blocking the CRMP4-dynein interaction reduces MN loss in human-derived MNs (C9orf72) and in ALS model mice. Thus, we demonstrate a novel CRMP4-dependent retrograde death signal that underlies MN loss in ALS.


Subject(s)
Amyotrophic Lateral Sclerosis/metabolism , Axonal Transport , Nerve Tissue Proteins/metabolism , Amyotrophic Lateral Sclerosis/genetics , Animals , Axons/metabolism , Cell Death , Cell Line , Cells, Cultured , Dyneins/metabolism , Mice , Mice, Inbred C57BL , Motor Neurons/metabolism , Motor Neurons/pathology , Nerve Tissue Proteins/genetics , Signal Transduction , Superoxide Dismutase-1/genetics
3.
J Cell Sci ; 135(16)2022 08 15.
Article in English | MEDLINE | ID: mdl-35833493

ABSTRACT

Nuclear-encoded mitochondrial protein mRNAs have been found to be localized and locally translated within neuronal processes. However, the mechanism of transport for those mRNAs to distal locations is not fully understood. Here, we describe axonal co-transport of Cox7c with mitochondria. Fractionation analysis and single-molecule fluorescence in situ hybridization (smFISH) assay revealed that endogenous mRNA encoding Cox7c was preferentially associated with mitochondria in a mouse neuronal cell line and within mouse primary motor neuron axons, whereas other mRNAs that do not encode mitochondrial protein were much less associated. Live-cell imaging of MS2-tagged Cox7c mRNA further confirmed the preferential colocalization and co-transport of Cox7c mRNA with mitochondria in motor neuron axons. Intriguingly, the coding region, rather than the 3' untranslated region (UTR), was the key domain for the co-transport. Our results reveal that Cox7c mRNA can be transported with mitochondria along significant distances and that its coding region is a major recognition feature. This is consistent with the idea that mitochondria can play a vital role in spatial regulation of the axonal transcriptome at distant neuronal sites.


Subject(s)
Axons , Electron Transport Complex IV/metabolism , Mitochondria , 3' Untranslated Regions/genetics , Animals , Axons/metabolism , In Situ Hybridization, Fluorescence , Mice , Mitochondria/metabolism , Mitochondrial Proteins/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism
4.
Cell ; 135(3): 414-5, 2008 Oct 31.
Article in English | MEDLINE | ID: mdl-18984153

ABSTRACT

The induction of long-term potentiation (LTP) leads to an increase in the density of AMPA receptors at dendritic spines. New work by Wang et al. (2008) reveals the mechanism by which myosin Vb regulates the intracellular trafficking of AMPA receptors from recycling endosomes to synaptic sites during LTP.


Subject(s)
Long-Term Potentiation , Myosins/metabolism , Receptors, AMPA/metabolism , Animals , Dendritic Spines/metabolism , Endosomes , Signal Transduction
5.
EMBO Rep ; 21(3): e48512, 2020 03 04.
Article in English | MEDLINE | ID: mdl-31919978

ABSTRACT

Regulation of axon guidance and pruning of inappropriate synapses by class 3 semaphorins are key to the development of neural circuits. Collapsin response mediator protein 2 (CRMP2) has been shown to regulate axon guidance by mediating semaphorin 3A (Sema3A) signaling; however, nothing is known about its role in synapse pruning. Here, using newly generated crmp2-/- mice we demonstrate that CRMP2 has a moderate effect on Sema3A-dependent axon guidance in vivo, and its deficiency leads to a mild defect in axon guidance in peripheral nerves and the corpus callosum. Surprisingly, crmp2-/- mice display prominent defects in stereotyped axon pruning in hippocampus and visual cortex and altered dendritic spine remodeling, which is consistent with impaired Sema3F signaling and with models of autism spectrum disorder (ASD). We demonstrate that CRMP2 mediates Sema3F signaling in primary neurons and that crmp2-/- mice display ASD-related social behavior changes in the early postnatal period as well as in adults. Together, we demonstrate that CRMP2 mediates Sema3F-dependent synapse pruning and its dysfunction shares histological and behavioral features of ASD.


Subject(s)
Autism Spectrum Disorder , Intercellular Signaling Peptides and Proteins/genetics , Membrane Proteins/physiology , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/physiology , Semaphorins , Animals , Dendritic Spines , Mice , Mice, Knockout , Neuronal Plasticity , Neurons , Signal Transduction
6.
J Cell Sci ; 132(23)2019 12 02.
Article in English | MEDLINE | ID: mdl-31722980

ABSTRACT

The neuromuscular junction (NMJ) is the largest, most-complex synapse in the human body. Motor neuron (MN) diseases, such as amyotrophic lateral sclerosis (ALS), specifically target MNs and the NMJs. However, little is known about the reasons for MN-selective neuronal and synaptic vulnerability in MN diseases. Here, utilizing a compartmental microfluidic in vitro co-culture system, we provide a possible explanation for why the NMJ, other than its unusual dimensions, differs from other synapses. By using live-imaging techniques, we discovered that cultured MNs display higher axonal and synaptic mitochondrial immobility compared with sympathetic neurons (SNs), leading to a profound enrichment of mitochondria only in the MN NMJ. Furthermore, by employing a synaptic ATP sensor, we show that mitochondrial respiration is the key contributor to ATP production in MN NMJs but not in SN synapses. Taken together, our data suggest that mitochondrial localization underlies the unique and specific qualities of MN NMJs. Our findings shed light on the role of mitochondria in MN and NMJ maintenance, and possibly indicate how mitochondria may serve as a source for selective MN vulnerability in neurodegenerative diseases.This article has an associated First Person interview with the first author of the paper.


Subject(s)
Adenosine Triphosphate/metabolism , Amyotrophic Lateral Sclerosis/metabolism , Neuromuscular Junction/metabolism , Animals , Axons/metabolism , Coculture Techniques , Female , Fluorescent Antibody Technique , Humans , Male , Mice , Microscopy, Fluorescence , Mitochondria/metabolism , Motor Neurons/metabolism , Muscle, Skeletal/metabolism , Myocytes, Cardiac/metabolism , Plasmids/genetics
7.
Biophys J ; 117(5): 810-816, 2019 09 03.
Article in English | MEDLINE | ID: mdl-31326106

ABSTRACT

The arrangement of receptors in the plasma membrane strongly affects the ability of a cell to sense its environment both in terms of sensitivity and in terms of spatial resolution. The spatial and temporal arrangement of the receptors is affected in turn by the mechanical properties and the structure of the cell membrane. Here, we focus on characterizing the flow of the membrane in response to the motion of a protein embedded in it. We do so by measuring the correlated diffusion of extracellularly tagged transmembrane neurotrophin receptors TrkB and p75 on transfected neuronal cells. In accord with previous reports, we find that the motion of single receptors exhibits transient confinement to submicron domains. We confirm predictions based on hydrodynamics of fluid membranes, finding long-range correlations in the motion of the receptors in the plasma membrane. However, we discover that these correlations do not persist for long ranges, as predicted, but decay exponentially, with a typical decay length on the scale of the average confining domain size.


Subject(s)
Cell Membrane/physiology , Rheology , Animals , Diffusion , Fluorescent Dyes/metabolism , HEK293 Cells , Humans , Membrane Proteins/metabolism , Models, Biological , Neurons/metabolism , Receptors, Nerve Growth Factor/metabolism
8.
Biophys J ; 117(2): 185-192, 2019 07 23.
Article in English | MEDLINE | ID: mdl-31280841

ABSTRACT

Diffusion plays a crucial role in many biological processes including signaling, cellular organization, transport mechanisms, and more. Direct observation of molecular movement by single-particle-tracking experiments has contributed to a growing body of evidence that many cellular systems do not exhibit classical Brownian motion but rather anomalous diffusion. Despite this evidence, characterization of the physical process underlying anomalous diffusion remains a challenging problem for several reasons. First, different physical processes can exist simultaneously in a system. Second, commonly used tools for distinguishing between these processes are based on asymptotic behavior, which is experimentally inaccessible in most cases. Finally, an accurate analysis of the diffusion model requires the calculation of many observables because different transport modes can result in the same diffusion power-law α, which is typically obtained from the mean-square displacements (MSDs). The outstanding challenge in the field is to develop a method to extract an accurate assessment of the diffusion process using many short trajectories with a simple scheme that is applicable at the nonexpert level. Here, we use deep learning to infer the underlying process resulting in anomalous diffusion. We implement a neural network to classify single-particle trajectories by diffusion type: Brownian motion, fractional Brownian motion and continuous time random walk. Further, we demonstrate the applicability of our network architecture for estimating the Hurst exponent for fractional Brownian motion and the diffusion coefficient for Brownian motion on both simulated and experimental data. These networks achieve greater accuracy than time-averaged MSD analysis on simulated trajectories while only requiring as few as 25 steps. When tested on experimental data, both net and ensemble MSD analysis converge to similar values; however, the net needs only half the number of trajectories required for ensemble MSD to achieve the same confidence interval. Finally, we extract diffusion parameters from multiple extremely short trajectories (10 steps) using our approach.


Subject(s)
Deep Learning , Single Molecule Imaging , Computer Simulation , Diffusion , Models, Biological
9.
Traffic ; 18(7): 415-424, 2017 07.
Article in English | MEDLINE | ID: mdl-28393448

ABSTRACT

Neurons are highly polarized cells, possessing long axons that can extend to more than 1-m long in adult humans. In order to survive and maintain proper functions, neurons have to respond accurately in both space and time to intracellular or intercellular cues. The regulation of these comprehensive responses involves ligand-receptor interactions, trafficking and local protein synthesis. Alterations in these mechanisms can lead to cellular dysfunction and disease. Although studies on the transport and localization of signalling endosomes along the axon have shed light on some central pathways of neuronal survival and growth as well as synapse function, little is known about the spatiotemporal mechanisms that allow the same molecule to signal differently at diverse subcellular locations and in specific neuronal populations. In this review, we will provide an overview of retrograde axonal signalling mechanisms and discuss new advances in our understanding of the spatial-specific regulation of neuronal signalling and functions, mechanisms that allow the same signal to have a different effect in another subcellular location.


Subject(s)
Axonal Transport/physiology , Axons/physiology , Neurons/physiology , Signal Transduction/physiology , Animals , Axons/metabolism , Humans , Nerve Growth Factors/metabolism , Neurons/metabolism , Protein Transport , Receptors, Growth Factor/metabolism
10.
J Neurosci ; 38(24): 5478-5494, 2018 06 13.
Article in English | MEDLINE | ID: mdl-29773756

ABSTRACT

Axon degeneration and disruption of neuromuscular junctions (NMJs) are key events in amyotrophic lateral sclerosis (ALS) pathology. Although the disease's etiology is not fully understood, it is thought to involve a non-cell-autonomous mechanism and alterations in RNA metabolism. Here, we identified reduced levels of miR126-5p in presymptomatic ALS male mice models, and an increase in its targets: axon destabilizing Type 3 Semaphorins and their coreceptor Neuropilins. Using compartmentalized in vitro cocultures, we demonstrated that myocytes expressing diverse ALS-causing mutations promote axon degeneration and NMJ dysfunction, which were inhibited by applying Neuropilin1 blocking antibody. Finally, overexpressing miR126-5p is sufficient to transiently rescue axon degeneration and NMJ disruption both in vitro and in vivo Thus, we demonstrate a novel mechanism underlying ALS pathology, in which alterations in miR126-5p facilitate a non-cell-autonomous mechanism of motor neuron degeneration in ALS.SIGNIFICANCE STATEMENT Despite some progress, currently no effective treatment is available for amyotrophic lateral sclerosis (ALS). We suggest a novel regulatory role for miR126-5p in ALS and demonstrate, for the first time, a mechanism by which alterations in miR126-5p contribute to axon degeneration and NMJ disruption observed in ALS. We show that miR126-5p is altered in ALS models and that it can modulate Sema3 and NRP protein expression. Furthermore, NRP1 elevations in motor neurons and muscle secretion of Sema3A contribute to axon degeneration and NMJ disruption in ALS. Finally, overexpressing miR126-5p is sufficient to transiently rescue NMJ disruption and axon degeneration both in vitro and in vivo.


Subject(s)
Amyotrophic Lateral Sclerosis/metabolism , MicroRNAs/metabolism , Nerve Degeneration/metabolism , Amyotrophic Lateral Sclerosis/genetics , Animals , Axons/metabolism , Axons/pathology , Down-Regulation , Gene Expression Regulation , Humans , Mice , MicroRNAs/genetics , Nerve Degeneration/genetics , Nerve Degeneration/pathology , Neuromuscular Junction/metabolism , Neuromuscular Junction/pathology , Neuropilin-1/biosynthesis , Neuropilin-1/genetics , Semaphorin-3A/biosynthesis , Semaphorin-3A/genetics
11.
PLoS Genet ; 12(12): e1006486, 2016 Dec.
Article in English | MEDLINE | ID: mdl-27997532

ABSTRACT

Familial Dysautonomia (FD) is a neurodegenerative disease in which aberrant tissue-specific splicing of IKBKAP exon 20 leads to reduction of IKAP protein levels in neuronal tissues. Here we generated a conditional knockout (CKO) mouse in which exon 20 of IKBKAP is deleted in the nervous system. The CKO FD mice exhibit developmental delays, sensory abnormalities, and less organized dorsal root ganglia (DRGs) with attenuated axons compared to wild-type mice. Furthermore, the CKO FD DRGs show elevated HDAC6 levels, reduced acetylated α-tubulin, unstable microtubules, and impairment of axonal retrograde transport of nerve growth factor (NGF). These abnormalities in DRG properties underlie neuronal degeneration and FD symptoms. Phosphatidylserine treatment decreased HDAC6 levels and thus increased acetylation of α-tubulin. Further PS treatment resulted in recovery of axonal outgrowth and enhanced retrograde axonal transport by decreasing histone deacetylase 6 (HDAC6) levels and thus increasing acetylation of α-tubulin levels. Thus, we have identified the molecular pathway that leads to neurodegeneration in FD and have demonstrated that phosphatidylserine treatment has the potential to slow progression of neurodegeneration.


Subject(s)
Axonal Transport/drug effects , Dysautonomia, Familial/genetics , Histone Deacetylases/genetics , Phosphatidylserines/administration & dosage , Tubulin/genetics , Alternative Splicing/genetics , Animals , Axonal Transport/genetics , Axons/drug effects , Disease Models, Animal , Dysautonomia, Familial/drug therapy , Dysautonomia, Familial/pathology , Exons/genetics , Ganglia, Spinal/growth & development , Ganglia, Spinal/pathology , Histone Deacetylase 6 , Histone Deacetylases/biosynthesis , Humans , Mice , Mice, Knockout , Nerve Degeneration/drug therapy , Nerve Degeneration/genetics , Nerve Degeneration/pathology , Nerve Growth Factor/genetics , Neurons/drug effects , Neurons/metabolism , Neurons/pathology , Phosphatidylserines/metabolism , Signal Transduction/drug effects , Signal Transduction/genetics
12.
Mol Cell Proteomics ; 15(2): 409-25, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26657538

ABSTRACT

Neurodegenerative diseases, such as Alzheimer's diseases (AD), are becoming more prevalent as the population ages. However, the mechanisms that lead to synapse destabilization and neuron death remain elusive. The advent of proteomics has allowed for high-throughput screening methods to search for biomarkers that could lead to early diagnosis and treatment and to identify alterations in the cellular proteome that could provide insight into disease etiology and possible treatment avenues. In this review, we have concentrated mainly on the findings that are related to how and whether proteomics studies have contributed to two aspects of AD research, the development of biomarkers for clinical diagnostics, and the recognition of proteins that can help elucidate the pathways leading to AD brain pathology. As a result of these studies, several candidate cerebrospinal fluid biomarkers are now available for further validation in different AD cohorts. Studies in AD brain and AD transgenic models support the notion that oxidative damage results in the alterations of metabolic enzymes and that mitochondrial dysfunction is central to AD neuropathology.


Subject(s)
Alzheimer Disease/genetics , Nerve Degeneration/genetics , Proteomics , Synapses/genetics , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Animals , Biomarkers/metabolism , Brain/metabolism , Brain/pathology , Humans , Mice , Mice, Transgenic , Nerve Degeneration/metabolism , Nerve Degeneration/pathology , Neurons/metabolism , Neurons/pathology , Proteome/genetics , Synapses/pathology
13.
Mol Cell Proteomics ; 15(2): 506-22, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26598648

ABSTRACT

Synapse disruption takes place in many neurodegenerative diseases, including amyotrophic lateral sclerosis (ALS). However, the mechanistic understanding of this process is still limited. We set out to study a possible role for dynein in synapse integrity. Cytoplasmic dynein is a multisubunit intracellular molecule responsible for diverse cellular functions, including long-distance transport of vesicles, organelles, and signaling factors toward the cell center. A less well-characterized role dynein may play is the spatial clustering and anchoring of various factors including mRNAs in distinct cellular domains such as the neuronal synapse. Here, in order to gain insight into dynein functions in synapse integrity and disruption, we performed a screen for novel dynein interactors at the synapse. Dynein immunoprecipitation from synaptic fractions of the ALS model mSOD1(G93A) and wild-type controls, followed by mass spectrometry analysis on synaptic fractions of the ALS model mSOD1(G93A) and wild-type controls, was performed. Using advanced network analysis, we identified Staufen1, an RNA-binding protein required for the transport and localization of neuronal RNAs, as a major mediator of dynein interactions via its interaction with protein phosphatase 1-beta (PP1B). Both in vitro and in vivo validation assays demonstrate the interactions of Staufen1 and PP1B with dynein, and their colocalization with synaptic markers was altered as a result of two separate ALS-linked mutations: mSOD1(G93A) and TDP43(A315T). Taken together, we suggest a model in which dynein's interaction with Staufen1 regulates mRNA localization along the axon and the synapses, and alterations in this process may correlate with synapse disruption and ALS toxicity.


Subject(s)
Amyotrophic Lateral Sclerosis/genetics , Cytoplasmic Dyneins/genetics , Proteomics , RNA-Binding Proteins/biosynthesis , Amyotrophic Lateral Sclerosis/metabolism , Amyotrophic Lateral Sclerosis/pathology , Animals , Axons/metabolism , Axons/pathology , Cytoplasmic Dyneins/metabolism , Disease Models, Animal , Humans , Mice , Motor Neurons/metabolism , Motor Neurons/pathology , Mutation , RNA-Binding Proteins/genetics , Synapses/genetics , Synapses/metabolism , Synapses/pathology , Synaptosomes/metabolism , Synaptosomes/pathology
14.
J Cell Sci ; 128(6): 1241-52, 2015 Mar 15.
Article in English | MEDLINE | ID: mdl-25632161

ABSTRACT

Bidirectional molecular communication between the motoneuron and the muscle is vital for neuromuscular junction (NMJ) formation and maintenance. The molecular mechanisms underlying such communication are of keen interest and could provide new targets for intervention in motoneuron disease. Here, we developed a microfluidic platform with motoneuron cell bodies on one side and muscle cells on the other, connected by motor axons extending through microgrooves to form functional NMJs. Using this system, we were able to differentiate between the proximal and distal effects of oxidative stress and glial-derived neurotrophic factor (GDNF), demonstrating a dying-back degeneration and retrograde transmission of pro-survival signaling, respectively. Furthermore, we show that GDNF acts differently on motoneuron axons versus soma, promoting axonal growth and innervation only when applied locally to axons. Finally, we track for the first time the retrograde transport of secreted GDNF from muscle to neuron. Thus, our data suggests spatially distinct effects of GDNF--facilitating growth and muscle innervation at axon terminals and survival pathways in the soma.


Subject(s)
Axons/metabolism , Glial Cell Line-Derived Neurotrophic Factor/metabolism , Microfluidics , Motor Neurons/metabolism , Muscle, Skeletal/metabolism , Neuromuscular Junction/physiology , Proto-Oncogene Proteins c-akt/metabolism , Animals , Calcium/metabolism , Cells, Cultured , Coculture Techniques , Immunoenzyme Techniques , Microscopy, Fluorescence , Motor Neurons/cytology , Muscle, Skeletal/cytology , Oxidative Stress , Phosphorylation , Spinal Cord/cytology , Spinal Cord/metabolism
15.
J Virol ; 89(18): 9591-600, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26157129

ABSTRACT

UNLABELLED: Rabies virus (RABV) polymerase L together with phosphoprotein P forms the PL polymerase complex that is essential for replication and transcription. However, its exact mechanism of action, interactions with cellular factors, and intracellular distribution are yet to be understood. Here by imaging a fluorescently tagged polymerase (mCherry-RABV-L), we show that L accumulates at acetylated and reorganized microtubules (MT). In silico analysis revealed a dynein light chain 1 (DLC1) binding motif in L that could mediate MT binding through dynein motors. As DLC1 binding by polymerase cofactor P is known, we compared the impact of the DLC1-binding motifs in P and L. Viruses with mutations in the respective motifs revealed that both motifs are required for efficient primary transcription, indicating that DLC1 acts as a transcription enhancer by binding to both P and L. Notably, also the levels of cellular DLC1 protein were regulated by both motifs, suggesting regulation of the DLC1 gene expression by both P and L. Finally, disruption of the motif in L resulted in a cell-type-specific loss of MT localization, demonstrating that DLC1 is involved in L-mediated cytoskeleton reorganization. Overall, we conclude that DLC1 acts as a transcription factor that stimulates primary RABV transcription by binding to both P and L. We further conclude that L influences MT organization and posttranslational modification, suggesting a model in which MT manipulation by L contributes to efficient intracellular transport of virus components and thus may serve as an important step in virus replication. IMPORTANCE: Regulation of rabies virus polymerase complex by viral and cellular factors thus far has not been fully understood. Although cellular dynein light chain 1 (DLC1) has been reported to increase primary transcription by binding to polymerase cofactor phosphoprotein P, the detailed mechanism is unknown, and it is also not known whether the large enzymatic polymerase subunit L is involved. By fluorescence microscopy analysis of fluorescence-tagged rabies virus L, in silico identification of a potential DLC1 binding site in L, and characterization of recombinant rabies virus mutants, we show that a DLC1 binding motif in L is involved in cytoskeleton localization and reorganization, primary transcription regulation by DLC1, and regulation of cellular DLC1 gene expression. By providing evidence for a direct contribution of a DLC1 binding motif in L, our data significantly increase the understanding of rabies virus polymerase regulation and host manipulation by the virus as well.


Subject(s)
Cytoplasmic Dyneins/metabolism , DNA-Directed RNA Polymerases/metabolism , Rabies virus/physiology , Transcription Factors/metabolism , Transcription, Genetic/physiology , Viral Proteins/metabolism , Virus Replication/physiology , Amino Acid Motifs , Cell Line, Tumor , Cytoplasmic Dyneins/genetics , DNA-Directed RNA Polymerases/genetics , HEK293 Cells , Humans , Transcription Factors/genetics , Viral Proteins/genetics
16.
PLoS Pathog ; 10(8): e1004348, 2014 Aug.
Article in English | MEDLINE | ID: mdl-25165859

ABSTRACT

Rabies virus (RABV) is a neurotropic virus that depends on long distance axonal transport in order to reach the central nervous system (CNS). The strategy RABV uses to hijack the cellular transport machinery is still not clear. It is thought that RABV interacts with membrane receptors in order to internalize and exploit the endosomal trafficking pathway, yet this has never been demonstrated directly. The p75 Nerve Growth Factor (NGF) receptor (p75NTR) binds RABV Glycoprotein (RABV-G) with high affinity. However, as p75NTR is not essential for RABV infection, the specific role of this interaction remains in question. Here we used live cell imaging to track RABV entry at nerve terminals and studied its retrograde transport along the axon with and without the p75NTR receptor. First, we found that NGF, an endogenous p75NTR ligand, and RABV, are localized in corresponding domains along nerve tips. RABV and NGF were internalized at similar time frames, suggesting comparable entry machineries. Next, we demonstrated that RABV could internalize together with p75NTR. Characterizing RABV retrograde movement along the axon, we showed the virus is transported in acidic compartments, mostly with p75NTR. Interestingly, RABV is transported faster than NGF, suggesting that RABV not only hijacks the transport machinery but can also manipulate it. Co-transport of RABV and NGF identified two modes of transport, slow and fast, that may represent a differential control of the trafficking machinery by RABV. Finally, we determined that p75NTR-dependent transport of RABV is faster and more directed than p75NTR-independent RABV transport. This fast route to the neuronal cell body is characterized by both an increase in instantaneous velocities and fewer, shorter stops en route. Hence, RABV may employ p75NTR-dependent transport as a fast mechanism to facilitate movement to the CNS.


Subject(s)
Axonal Transport/physiology , Axons/virology , Rabies virus/pathogenicity , Rabies/metabolism , Receptors, Nerve Growth Factor/metabolism , Animals , HEK293 Cells , Humans , Mice , Mice, Inbred ICR , Microfluidic Analytical Techniques , Rabies/parasitology
17.
Hum Mol Genet ; 22(23): 4720-5, 2013 Dec 01.
Article in English | MEDLINE | ID: mdl-23836781

ABSTRACT

Amyotrophic lateral sclerosis (ALS) is a fatal neurodegenerative disorder of motor neurons. Although most cases of ALS are sporadic (sALS) and of unknown etiology, there are also inherited familial ALS (fALS) cases that share a phenotype similar to sALS pathological and clinical phenotype. In this study, we have identified two new potential genetic ALS biomarkers in human bone marrow mesenchymal stem cells (hMSC) obtained from sALS patients, namely the TDP-43 (TAR DNA-binding protein 43) and SLPI (secretory leukocyte protease inhibitor). Together with the previously discovered ones-CyFIP2 and RbBP9, we investigated whether these four potential ALS biomarkers may be differentially expressed in tissues obtained from mutant SOD1(G93A) transgenic mice, a model that is relevant for at least 20% of the fALS cases. Quantitative real-time PCR analysis of brain, spinal cord and muscle tissues of the mSOD1(G93A) and controls at various time points during the progression of the neurological disease showed differential expression of the four identified biomarkers in correlation with (i) the tissue type, (ii) the stage of the disease and (iii) the gender of the animals, creating thus a novel spatiotemporal molecular signature of ALS. The biomarkers detected in the fALS animal model were homologous to those that were identified in hMSC of our sALS cases. These results support the possibility of a molecular link between sALS and fALS and may indicate common pathogenetic mechanisms involved in both types of ALS. Moreover, these results may pave the path for using the mSOD1(G93A) mouse model and these biomarkers as molecular beacons to evaluate the effects of novel drugs/treatments in ALS.


Subject(s)
Adaptor Proteins, Signal Transducing/genetics , Amyotrophic Lateral Sclerosis/pathology , Biomarkers/metabolism , Cell Cycle Proteins/genetics , DNA-Binding Proteins/genetics , Secretory Leukocyte Peptidase Inhibitor/genetics , Superoxide Dismutase/genetics , Adaptor Proteins, Signal Transducing/metabolism , Adult , Amyotrophic Lateral Sclerosis/genetics , Animals , Brain/metabolism , Brain/pathology , Cell Cycle Proteins/metabolism , Cells, Cultured , DNA-Binding Proteins/metabolism , Disease Models, Animal , Disease Progression , Female , Humans , Male , Mesenchymal Stem Cells/metabolism , Mesenchymal Stem Cells/pathology , Mice , Mice, Transgenic , Middle Aged , Muscles/metabolism , Muscles/pathology , Secretory Leukocyte Peptidase Inhibitor/metabolism , Spinal Cord/metabolism , Spinal Cord/pathology , Superoxide Dismutase/metabolism , Young Adult
18.
J Virol ; 88(24): 14172-83, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25275124

ABSTRACT

UNLABELLED: Rabies virus (RABV) spread is widely accepted to occur only by retrograde axonal transport. However, examples of anterograde RABV spread in peripheral neurons such as dorsal root ganglion (DRG) neurons indicated a possible bidirectional transport by an uncharacterized mechanism. Here, we analyzed the axonal transport of fluorescence-labeled RABV in DRG neurons by live-cell microscopy. Both entry-related retrograde transport of RABV after infection at axon endings and postreplicative transport of newly formed virus were visualized in compartmentalized DRG neuron cultures. Whereas entry-related transport at 1.5 µm/s occurred only retrogradely, after 2 days of infection, multiple particles were observed in axons moving in both the anterograde and retrograde directions. The dynamics of postreplicative retrograde transport (1.6 µm/s) were similar to those of entry-related retrograde transport. In contrast, anterograde particle transport at 3.4 µm/s was faster, indicating active particle transport. Interestingly, RABV missing the glycoproteins did not move anterogradely within the axon. Thus, anterograde RABV particle transport depended on the RABV glycoprotein. Moreover, colocalization of green fluorescent protein (GFP)-labeled ribonucleoproteins (RNPs) and glycoprotein in distal axonal regions as well as cotransport of labeled RNPs with membrane-anchored mCherry reporter confirmed that either complete enveloped virus particles or vesicle associated RNPs were transported. Our data show that anterograde RABV movement in peripheral DRG neurons occurs by active motor protein-dependent transport. We propose two models for postreplicative long-distance transport in peripheral neurons: either transport of complete virus particles or cotransport of RNPs and G-containing vesicles through axons to release virus at distal sites of infected DRG neurons. IMPORTANCE: Rabies virus retrograde axonal transport by dynein motors supports virus spread over long distances and lethal infection of the central nervous system. Though active rabies virus transport has been widely accepted to be unidirectional, evidence for anterograde spread in peripheral neurons supports the hypothesis that in some neurons RABV also enters the anterograde pathway by so-far unknown mechanisms. By live microscopy we visualized fast anterograde axonal transport of rabies virus. The velocities exceeded those of retrograde movements, suggesting that active, most likely kinesin-dependent transport machineries are involved. Dependency of anterograde transport on the expression of virus glycoprotein G and cotransport with vesicles further suggest that complete enveloped virus particles or cotransport of virus ribonucleoprotein and G-containing vesicles occurred. These data provide the first insight in the mechanism of anterograde rabies virus transport and substantially contribute to the understanding of RABV replication and spread of newly formed virus in peripheral neurons.


Subject(s)
Axonal Transport , Ganglia, Spinal/virology , Glycoproteins/metabolism , Neurons/virology , Rabies virus/physiology , Virion/metabolism , Animals , Cells, Cultured , Female , Microscopy, Fluorescence , Microscopy, Video , Rats, Sprague-Dawley , Staining and Labeling
19.
Mol Ther ; 22(3): 554-566, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24100640

ABSTRACT

Adeno-associated virus (AAV) vectors can move along axonal pathways after brain injection, resulting in transduction of distal brain regions. This can enhance the spread of therapeutic gene transfer and improve treatment of neurogenetic disorders that require global correction. To better understand the underlying cellular mechanisms that drive AAV trafficking in neurons, we investigated the axonal transport of dye-conjugated AAV9, utilizing microfluidic primary neuron cultures that isolate cell bodies from axon termini and permit independent analysis of retrograde and anterograde axonal transport. After entry, AAV was trafficked into nonmotile early and recycling endosomes, exocytic vesicles, and a retrograde-directed late endosome/lysosome compartment. Rab7-positive late endosomes/lysosomes that contained AAV were highly motile, exhibiting faster retrograde velocities and less pausing than Rab7-positive endosomes without virus. Inhibitor experiments indicated that the retrograde transport of AAV within these endosomes is driven by cytoplasmic dynein and requires Rab7 function, whereas anterograde transport of AAV is driven by kinesin-2 and exhibits unusually rapid velocities. Furthermore, increasing AAV9 uptake by neuraminidase treatment significantly enhanced virus transport in both directions. These findings provide novel insights into AAV trafficking within neurons, which should enhance progress toward the utilization of AAV for improved distribution of transgene delivery within the brain.


Subject(s)
Axonal Transport , Dependovirus/physiology , Dyneins/metabolism , Kinesins/metabolism , Neurons/virology , rab GTP-Binding Proteins/metabolism , Animals , Cells, Cultured , Endosomes/metabolism , Neuraminidase/pharmacology , Neurons/metabolism , Rats , rab7 GTP-Binding Proteins
20.
J Biol Chem ; 288(39): 27812-24, 2013 Sep 27.
Article in English | MEDLINE | ID: mdl-23960070

ABSTRACT

Cytoplasmic dynein is well characterized as an organelle motor, but dynein also acts to tether and stabilize dynamic microtubule plus-ends in vitro. Here we identify a novel and direct interaction between dynein and the 180-kDa isoform of the neural cell adhesion molecule (NCAM). Optical trapping experiments indicate that dynein bound to beads via the NCAM180 interaction domain can tether projecting microtubule plus-ends. Live cell assays indicate that the NCAM180-dependent recruitment of dynein to the cortex leads to the selective stabilization of microtubules projecting to NCAM180 patches at the cell periphery. The dynein-NCAM180 interaction also enhances cell-cell adhesion in heterologous cell assays. Dynein and NCAM180 co-precipitate from mouse brain extract and from synaptosomal fractions, consistent with an endogenous interaction in neurons. Thus, we examined microtubule dynamics and synaptic density in primary cortical neurons. We find that depletion of NCAM, inhibition of the dynein-NCAM180 interaction, or dampening of microtubule dynamics with low dose nocodazole all result in significantly decreased in synaptic density. Based on these observations, we propose a working model for the role of dynein at the synapse, in which the anchoring of the motor to the cortex via binding to an adhesion molecule mediates the tethering of dynamic microtubule plus-ends to potentiate synaptic stabilization.


Subject(s)
Dyneins/chemistry , Microtubules/metabolism , Neural Cell Adhesion Molecules/metabolism , Neurons/metabolism , Synapses/metabolism , Animals , COS Cells , Chlorocebus aethiops , Cytoplasm/metabolism , Cytoplasmic Dyneins/chemistry , HeLa Cells , Humans , Mice , Protein Binding , Synaptosomes/metabolism , Two-Hybrid System Techniques
SELECTION OF CITATIONS
SEARCH DETAIL