Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 84
Filter
Add more filters










Publication year range
1.
Sci Adv ; 9(30): eadi0286, 2023 07 28.
Article in English | MEDLINE | ID: mdl-37506203

ABSTRACT

Polypyrimidine tract binding protein 1 (PTBP1) is thought to be expressed only at embryonic stages in central neurons. Its down-regulation triggers neuronal differentiation in precursor and non-neuronal cells, an approach recently tested for generation of neurons de novo for amelioration of neurodegenerative disorders. Moreover, PTBP1 is replaced by its paralog PTBP2 in mature central neurons. Unexpectedly, we found that both proteins are coexpressed in adult sensory and motor neurons, with PTBP2 restricted mainly to the nucleus, while PTBP1 also shows axonal localization. Levels of axonal PTBP1 increased markedly after peripheral nerve injury, and it associates in axons with mRNAs involved in injury responses and nerve regeneration, including importin ß1 (KPNB1) and RHOA. Perturbation of PTBP1 affects local translation in axons, nociceptor neuron regeneration and both thermal and mechanical sensation. Thus, PTBP1 has functional roles in adult axons. Hence, caution is required before considering targeting of PTBP1 for therapeutic purposes.


Subject(s)
Axons , Nerve Regeneration , Neurons , Peripheral Nerve Injuries , Adult , Humans , Axons/metabolism , Heterogeneous-Nuclear Ribonucleoproteins/genetics , Heterogeneous-Nuclear Ribonucleoproteins/metabolism , Interneurons/metabolism , Nerve Regeneration/genetics , Neurons/metabolism , Peripheral Nerve Injuries/genetics , Peripheral Nerve Injuries/metabolism
2.
J Cell Sci ; 136(5)2023 03 01.
Article in English | MEDLINE | ID: mdl-36218033

ABSTRACT

Size homeostasis is a fundamental process in biology and is particularly important for large cells such as neurons. We previously proposed a motor-dependent length-sensing mechanism wherein reductions in microtubule motor levels would be expected to accelerate neuronal growth, and validated this prediction in dynein heavy chain 1 Loa mutant (Dync1h1Loa) sensory neurons. Here, we describe a new mouse model with a conditional deletion allele of exons 24 and 25 in Dync1h1. Homozygous Islet1-Cre-mediated deletion of Dync1h1 (Isl1-Dync1h1-/-), which deletes protein from the motor and sensory neurons, is embryonic lethal, but heterozygous animals (Isl1-Dync1h1+/-) survive to adulthood with ∼50% dynein expression in targeted cells. Isl1-Dync1h1+/- sensory neurons reveal accelerated growth, as previously reported in Dync1h1Loa neurons. Moreover, Isl1-Dync1h1+/- mice show mild impairments in gait, proprioception and tactile sensation, similar to what is seen in Dync1h1Loa mice, confirming that specific aspects of the Loa phenotype are due to reduced dynein levels. Isl1-Dync1h1+/- mice also show delayed recovery from peripheral nerve injury, likely due to reduced injury signal delivery from axonal lesion sites. Thus, conditional deletion of Dync1h1 exons 24 and 25 enables targeted studies of the role of dynein in neuronal growth.


Subject(s)
Cytoplasmic Dyneins , Dyneins , Mice , Animals , Dyneins/genetics , Dyneins/metabolism , Cytoplasmic Dyneins/genetics , Cytoplasmic Dyneins/metabolism , Alleles , Mutation , Sensory Receptor Cells/metabolism
3.
Mol Cell Proteomics ; 21(11): 100418, 2022 11.
Article in English | MEDLINE | ID: mdl-36180036

ABSTRACT

Importin ß1 (KPNB1) is a nucleocytoplasmic transport factor with critical roles in both cytoplasmic and nucleocytoplasmic transport, hence there is keen interest in the characterization of its subcellular interactomes. We found limited efficiency of BioID in the detection of importin complex cargos and therefore generated a highly specific and sensitive anti-KPNB1 monoclonal antibody to enable biotinylation by antibody recognition analysis of importin ß1 interactomes. The monoclonal antibody recognizes an epitope comprising residues 301-320 of human KPBN1 and strikingly is highly specific for cytoplasmic KPNB1 in diverse applications, with little reaction with KPNB1 in the nucleus. Biotinylation by antibody recognition with this novel antibody revealed numerous new interactors of importin ß1, expanding the KPNB1 interactome to cytoplasmic and signaling complexes that highlight potential new functions for the importins complex beyond nucleocytoplasmic transport. Data are available via ProteomeXchange with identifier PXD032728.


Subject(s)
Antibodies, Monoclonal , Karyopherins , Humans , Karyopherins/metabolism , Antibodies, Monoclonal/metabolism , beta Karyopherins/metabolism , Active Transport, Cell Nucleus , Cytoplasm/metabolism , Cell Nucleus/metabolism
4.
Neural Regen Res ; 17(12): 2664-2665, 2022 Dec.
Article in English | MEDLINE | ID: mdl-35662201
5.
EMBO J ; 40(20): e107158, 2021 10 18.
Article in English | MEDLINE | ID: mdl-34515347

ABSTRACT

Nucleolin is a multifunctional RNA Binding Protein (RBP) with diverse subcellular localizations, including the nucleolus in all eukaryotic cells, the plasma membrane in tumor cells, and the axon in neurons. Here we show that the glycine arginine rich (GAR) domain of nucleolin drives subcellular localization via protein-protein interactions with a kinesin light chain. In addition, GAR sequences mediate plasma membrane interactions of nucleolin. Both these modalities are in addition to the already reported involvement of the GAR domain in liquid-liquid phase separation in the nucleolus. Nucleolin transport to axons requires the GAR domain, and heterozygous GAR deletion mice reveal reduced axonal localization of nucleolin cargo mRNAs and enhanced sensory neuron growth. Thus, the GAR domain governs axonal transport of a growth controlling RNA-RBP complex in neurons, and is a versatile localization determinant for different subcellular compartments. Localization determination by GAR domains may explain why GAR mutants in diverse RBPs are associated with neurodegenerative disease.


Subject(s)
Cell Nucleolus/metabolism , Ganglia, Spinal/metabolism , Kinesins/metabolism , Neurons/metabolism , Phosphoproteins/chemistry , RNA-Binding Proteins/chemistry , Sciatic Nerve/metabolism , Amino Acid Sequence , Animals , Axonal Transport/genetics , Cell Line, Tumor , Cell Nucleolus/ultrastructure , Ganglia, Spinal/cytology , Gene Expression , HEK293 Cells , HeLa Cells , Humans , Kinesins/genetics , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mutation , Neurons/cytology , Phosphoproteins/genetics , Phosphoproteins/metabolism , Primary Cell Culture , Protein Domains , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism , Sciatic Nerve/cytology , Nucleolin
6.
Cell Rep Med ; 2(5): 100281, 2021 05 18.
Article in English | MEDLINE | ID: mdl-34095883

ABSTRACT

Anxiety and stress-related conditions represent a significant health burden in modern society. Unfortunately, most anxiolytic drugs are prone to side effects, limiting their long-term usage. Here, we employ a bioinformatics screen to identify drugs for repurposing as anxiolytics. Comparison of drug-induced gene-expression profiles with the hippocampal transcriptome of an importin α5 mutant mouse model with reduced anxiety identifies the hypocholesterolemic agent ß-sitosterol as a promising candidate. ß-sitosterol activity is validated by both intraperitoneal and oral application in mice, revealing it as the only clear anxiolytic from five closely related phytosterols. ß-sitosterol injection reduces the effects of restraint stress, contextual fear memory, and c-Fos activation in the prefrontal cortex and dentate gyrus. Moreover, synergistic anxiolysis is observed when combining sub-efficacious doses of ß-sitosterol with the SSRI fluoxetine. These preclinical findings support further development of ß-sitosterol, either as a standalone anxiolytic or in combination with low-dose SSRIs.


Subject(s)
Anti-Anxiety Agents/pharmacology , Anxiety Disorders/drug therapy , Anxiety/drug therapy , Sitosterols/pharmacology , Animals , Fear/drug effects , Fluoxetine/pharmacology , Mice, Inbred C57BL , Motor Activity/drug effects , Pharmaceutical Preparations/metabolism , Prefrontal Cortex/drug effects , Proto-Oncogene Proteins c-fos/pharmacology , Selective Serotonin Reuptake Inhibitors/pharmacology , Tranquilizing Agents/pharmacology
7.
Curr Biol ; 30(24): 4882-4895.e6, 2020 12 21.
Article in English | MEDLINE | ID: mdl-33065005

ABSTRACT

The main limitation on axon regeneration in the peripheral nervous system (PNS) is the slow rate of regrowth. We recently reported that nerve regeneration can be accelerated by axonal G3BP1 granule disassembly, releasing axonal mRNAs for local translation to support axon growth. Here, we show that G3BP1 phosphorylation by casein kinase 2α (CK2α) triggers G3BP1 granule disassembly in injured axons. CK2α activity is temporally and spatially regulated by local translation of Csnk2a1 mRNA in axons after injury, but this requires local translation of mTor mRNA and buffering of the elevated axonal Ca2+ that occurs after axotomy. CK2α's appearance in axons after PNS nerve injury correlates with disassembly of axonal G3BP1 granules as well as increased phospho-G3BP1 and axon growth, although depletion of Csnk2a1 mRNA from PNS axons decreases regeneration and increases G3BP1 granules. Phosphomimetic G3BP1 shows remarkably decreased RNA binding in dorsal root ganglion (DRG) neurons compared with wild-type and non-phosphorylatable G3BP1; combined with other studies, this suggests that CK2α-dependent G3BP1 phosphorylation on Ser 149 after axotomy releases axonal mRNAs for translation. Translation of axonal mRNAs encoding some injury-associated proteins is known to be increased with Ca2+ elevations, and using a dual fluorescence recovery after photobleaching (FRAP) reporter assay for axonal translation, we see that translational specificity switches from injury-associated protein mRNA translation to CK2α translation with endoplasmic reticulum (ER) Ca2+ release versus cytoplasmic Ca2+ chelation. Our results point to axoplasmic Ca2+ concentrations as a determinant for the temporal specificity of sequential translational activation of different axonal mRNAs as severed axons transition from injury to regenerative growth.


Subject(s)
Axons/physiology , Casein Kinase II/metabolism , DNA Helicases/metabolism , Nerve Regeneration/genetics , Peripheral Nerve Injuries/physiopathology , Poly-ADP-Ribose Binding Proteins/metabolism , RNA Helicases/metabolism , RNA Recognition Motif Proteins/metabolism , Animals , Calcium/metabolism , Casein Kinase II/genetics , Cytoplasmic Granules/metabolism , DNA Helicases/genetics , Disease Models, Animal , Endoplasmic Reticulum/metabolism , Ganglia, Spinal/cytology , Ganglia, Spinal/injuries , Ganglia, Spinal/physiology , Humans , Male , Mice , Mice, Knockout , Models, Animal , Peripheral Nerve Injuries/pathology , Phosphorylation , Poly-ADP-Ribose Binding Proteins/genetics , Protein Biosynthesis/physiology , RNA Helicases/genetics , RNA Recognition Motif Proteins/genetics , RNA, Messenger/metabolism , Rats , TOR Serine-Threonine Kinases/genetics
8.
Science ; 369(6505): 842-846, 2020 08 14.
Article in English | MEDLINE | ID: mdl-32792398

ABSTRACT

How is neuropathic pain regulated in peripheral sensory neurons? Importins are key regulators of nucleocytoplasmic transport. In this study, we found that importin α3 (also known as karyopherin subunit alpha 4) can control pain responsiveness in peripheral sensory neurons in mice. Importin α3 knockout or sensory neuron-specific knockdown in mice reduced responsiveness to diverse noxious stimuli and increased tolerance to neuropathic pain. Importin α3-bound c-Fos and importin α3-deficient neurons were impaired in c-Fos nuclear import. Knockdown or dominant-negative inhibition of c-Fos or c-Jun in sensory neurons reduced neuropathic pain. In silico screens identified drugs that mimic importin α3 deficiency. These drugs attenuated neuropathic pain and reduced c-Fos nuclear localization. Thus, perturbing c-Fos nuclear import by importin α3 in peripheral neurons can promote analgesia.


Subject(s)
Chronic Pain/physiopathology , Neuralgia/physiopathology , Sensory Receptor Cells/physiology , alpha Karyopherins/physiology , Active Transport, Cell Nucleus , Animals , Benzophenones/pharmacology , Chronic Pain/genetics , Gene Expression Profiling , Gene Knockdown Techniques , Isoxazoles/pharmacology , Mice , Mice, Inbred C57BL , Neuralgia/genetics , Proto-Oncogene Proteins c-fos/antagonists & inhibitors , Proto-Oncogene Proteins c-fos/metabolism , Transcription Factor AP-1/metabolism , alpha Karyopherins/genetics
9.
Neurobiol Dis ; 140: 104816, 2020 07.
Article in English | MEDLINE | ID: mdl-32088381

ABSTRACT

The cytoplasmic dynein motor complex transports essential signals and organelles from the cell periphery to the perinuclear region, hence is critical for the survival and function of highly polarized cells such as neurons. Dynein Light Chain Roadblock-Type 1 (DYNLRB1) is thought to be an accessory subunit required for specific cargos, but here we show that it is essential for general dynein-mediated transport and sensory neuron survival. Homozygous Dynlrb1 null mice are not viable and die during early embryonic development. Furthermore, heterozygous or adult knockdown animals display reduced neuronal growth, and selective depletion of Dynlrb1 in proprioceptive neurons compromises their survival. Conditional depletion of Dynlrb1 in sensory neurons causes deficits in several signaling pathways, including ß-catenin subcellular localization, and a severe impairment in the axonal transport of both lysosomes and retrograde signaling endosomes. Hence, DYNLRB1 is an essential component of the dynein complex, and given dynein's critical functions in neuronal physiology, DYNLRB1 could have a prominent role in the etiology of human neurodegenerative diseases.


Subject(s)
Axonal Transport/physiology , Dyneins/metabolism , Sensory Receptor Cells/metabolism , Animals , Carrier Proteins/metabolism , Cell Survival , Cells, Cultured , Dyneins/genetics , Lysosomes/metabolism , Male , Mice , Neurogenesis , Organelles/metabolism , Transfection
10.
BMC Biol ; 17(1): 36, 2019 04 29.
Article in English | MEDLINE | ID: mdl-31035993

ABSTRACT

Individual cell types have characteristic sizes, suggesting that size sensing mechanisms may coordinate transcription, translation, and metabolism with cell growth rates. Two types of size-sensing mechanisms have been proposed: spatial sensing of the location or dimensions of a signal, subcellular structure or organelle; or titration-based sensing of the intracellular concentrations of key regulators. Here we propose that size sensing in animal cells combines both titration and spatial sensing elements in a dynamic mechanism whereby microtubule motor-dependent localization of RNA encoding importin ß1 and mTOR, coupled with regulated local protein synthesis, enable cytoskeleton length sensing for cell growth regulation.


Subject(s)
Cell Physiological Phenomena/physiology , Cell Size , Feedback, Physiological , Animals , Models, Biological
11.
Neuron ; 102(3): 507-509, 2019 05 08.
Article in English | MEDLINE | ID: mdl-31071280

ABSTRACT

In this issue of Neuron, Crerar et al. (2019) found Tp53inp2 as a highly expressed RNA in sympathetic neuron axons. Strikingly, its long 3' UTR ensures that Tp53inp2 is not translated in axons, and the untranslated RNA affects neuronal growth by interacting with neurotrophin receptors.


Subject(s)
Nerve Growth Factor , RNA , Axons , RNA, Messenger , Signal Transduction
12.
Neurosci Res ; 139: 26-36, 2019 Feb.
Article in English | MEDLINE | ID: mdl-30321567

ABSTRACT

Neurons convey signals over long distances, for example motor neurons and sensory neurons project axons up to a meter long in humans. To this end, a sophisticated network of long-range signaling mechanisms enables communication between neuronal processes and somata. These mechanisms are activated during axonal injury and have essential roles both for sensing the injury and regulating subsequent regeneration. Here we survey the role of one such mechanism, axonal translation, which contributes to both retrograde injury signaling and as a source of proteins for regenerating axons. The nature of the axonal synthesis machinery has become progressively clearer over the past decade. A large number of axonally localized mRNAs have been identified, which cover a wide spectrum of protein families; and axonal ribosomes have been detected, even though their origin is still subject to debate. Various kinase pathways, most prominently mTOR, have been implicated in driving local translation in axons. Finally, new technologies are becoming available to visualize axonal translation and enable proteomic analyses. These technological improvements offer new avenues towards comprehensive characterization of the axonal translational machinery.


Subject(s)
Axons/metabolism , Ganglia, Spinal/metabolism , Nerve Regeneration/physiology , Sensory Receptor Cells/metabolism , Animals , Humans , RNA, Messenger/metabolism , TOR Serine-Threonine Kinases/metabolism
13.
Cell Rep ; 25(11): 3169-3179.e7, 2018 12 11.
Article in English | MEDLINE | ID: mdl-30540948

ABSTRACT

Importins mediate transport from synapse to soma and from cytoplasm to nucleus, suggesting that perturbation of importin-dependent pathways should have significant neuronal consequences. A behavioral screen on five importin α knockout lines revealed that reduced expression of importin α5 (KPNA1) in hippocampal neurons specifically decreases anxiety in mice. Re-expression of importin α5 in ventral hippocampus of knockout animals increased anxiety behaviors to wild-type levels. Hippocampal neurons lacking importin α5 reveal changes in presynaptic plasticity and modified expression of MeCP2-regulated genes, including sphingosine kinase 1 (Sphk1). Knockout of importin α5, but not importin α3 or α4, reduces MeCP2 nuclear localization in hippocampal neurons. A Sphk1 blocker reverses anxiolysis in the importin α5 knockout mouse, while pharmacological activation of sphingosine signaling has robust anxiolytic effects in wild-type animals. Thus, importin α5 influences sphingosine-sensitive anxiety pathways by regulating MeCP2 nuclear import in hippocampal neurons.


Subject(s)
Anxiety/metabolism , Methyl-CpG-Binding Protein 2/metabolism , Phosphotransferases (Alcohol Group Acceptor)/metabolism , alpha Karyopherins/metabolism , Animals , Anti-Anxiety Agents/pharmacology , Behavior, Animal , Carbolines/pharmacology , Hippocampus/pathology , Mice, Knockout , Neurons/metabolism , Phenotype , Synapses/metabolism , Transcription, Genetic , alpha Karyopherins/deficiency
14.
Mol Omics ; 14(6): 380-388, 2018 12 03.
Article in English | MEDLINE | ID: mdl-30338329

ABSTRACT

Compartmentalized translation allows rapid synthesis of proteins in targeted cellular locations. Microarray and RNA sequencing combined with physical subcellular separation methods have enabled extensive charting of subcellular transcriptomes. However, the extent of translating these local messages into protein remains relatively understudied at the genome-wide level. Here we review omics methods currently available for these studies, placing special attention on methods allowing cell-specific and subcellularly restricted analysis.


Subject(s)
Genomics , Protein Biosynthesis , Animals , Gene Expression Profiling/methods , Genomics/methods , Humans , Intracellular Space , Proteomics/methods , Transcriptome
15.
Nat Commun ; 9(1): 3358, 2018 08 22.
Article in English | MEDLINE | ID: mdl-30135423

ABSTRACT

Critical functions of intra-axonally synthesized proteins are thought to depend on regulated recruitment of mRNA from storage depots in axons. Here we show that axotomy of mammalian neurons induces translation of stored axonal mRNAs via regulation of the stress granule protein G3BP1, to support regeneration of peripheral nerves. G3BP1 aggregates within peripheral nerve axons in stress granule-like structures that decrease during regeneration, with a commensurate increase in phosphorylated G3BP1. Colocalization of G3BP1 with axonal mRNAs is also correlated with the growth state of the neuron. Disrupting G3BP functions by overexpressing a dominant-negative protein activates intra-axonal mRNA translation, increases axon growth in cultured neurons, disassembles axonal stress granule-like structures, and accelerates rat nerve regeneration in vivo.


Subject(s)
Axons/metabolism , Cytoplasmic Granules/metabolism , Nerve Regeneration/physiology , Poly-ADP-Ribose Binding Proteins/metabolism , RNA, Messenger/metabolism , Animals , Cells, Cultured , Female , Fluorescence Recovery After Photobleaching , HEK293 Cells , Humans , Male , Mice , Microscopy, Fluorescence , NIH 3T3 Cells , Nerve Regeneration/genetics , Poly-ADP-Ribose Binding Proteins/genetics , RNA, Messenger/genetics , Rats , Rats, Sprague-Dawley
16.
Mol Cell Proteomics ; 17(11): 2091-2106, 2018 11.
Article in English | MEDLINE | ID: mdl-30038033

ABSTRACT

mRNA translation in axons enables neurons to introduce new proteins at sites distant from their cell body. mRNA-protein interactions drive this post-transcriptional regulation, yet knowledge of RNA binding proteins (RBP) in axons is limited. Here we used proteomics to identify RBPs interacting with the axonal localizing motifs of Nrn1, Hmgb1, Actb, and Gap43 mRNAs, revealing many novel RBPs in axons. Interestingly, no RBP is shared between all four RNA motifs, suggesting graded and overlapping specificities of RBP-mRNA pairings. A systematic assessment of axonal mRNAs interacting with hnRNP H1, hnRNP F, and hnRNP K, proteins that bound with high specificity to Nrn1 and Hmgb1, revealed that axonal mRNAs segregate into axon growth-associated RNA regulons based on hnRNP interactions. Axotomy increases axonal transport of hnRNPs H1, F, and K, depletion of these hnRNPs decreases axon growth and reduces axonal mRNA levels and axonal protein synthesis. Thus, subcellular hnRNP-interacting RNA regulons support neuronal growth and regeneration.


Subject(s)
Axons/metabolism , Heterogeneous-Nuclear Ribonucleoproteins/metabolism , Nucleotide Motifs/genetics , RNA, Messenger/genetics , Regulon/genetics , 5' Untranslated Regions/genetics , Animals , Axonal Transport/genetics , GAP-43 Protein/genetics , GAP-43 Protein/metabolism , GPI-Linked Proteins/genetics , GPI-Linked Proteins/metabolism , HMGB1 Protein/genetics , HMGB1 Protein/metabolism , Heterogeneous-Nuclear Ribonucleoproteins/genetics , Male , Neuropeptides/genetics , Neuropeptides/metabolism , Protein Binding , Protein Biosynthesis , RNA Transport/genetics , RNA, Messenger/metabolism , Rats, Sprague-Dawley
17.
eNeuro ; 5(2)2018.
Article in English | MEDLINE | ID: mdl-29756027

ABSTRACT

Transcriptional events leading to outgrowth of neuronal axons have been intensively studied, but the role of translational regulation in this process is not well understood. Here, we use translatome analyses by ribosome pull-down and protein synthesis characterization by metabolic isotopic labeling to study nerve injury and axon outgrowth proteomes in rodent dorsal root ganglia (DRGs) and sensory neurons. We identify over 1600 gene products that are primarily translationally regulated in DRG neurons after nerve injury, many of which contain a 5'UTR cytosine-enriched regulator of translation (CERT) motif, implicating the translation initiation factor Eif4e in the injury response. We further identified approximately 200 proteins that undergo robust de novo synthesis in the initial stages of axon growth. ApoE is one of the highly synthesized proteins in neurons, and its receptor binding inhibition or knockout affects axon outgrowth. These findings provide a resource for future analyses of the role of translational regulation in neuronal injury responses and axon extension.


Subject(s)
Axons/metabolism , Ganglia, Spinal/metabolism , Gene Expression Regulation/genetics , Nerve Regeneration/genetics , Neuronal Outgrowth/genetics , Peripheral Nerve Injuries/genetics , Protein Biosynthesis/genetics , Sensory Receptor Cells/metabolism , Animals , Cell Culture Techniques , Male , Mice , Mice, Inbred C57BL , Proteomics , Rats , Rats, Wistar
18.
Nat Cell Biol ; 20(9): 1098, 2018 Sep.
Article in English | MEDLINE | ID: mdl-29520084

ABSTRACT

In the version of this Article originally published, the affiliations for Roland A. Fleck and José Antonio Del Río were incorrect due to a technical error that resulted in affiliations 8 and 9 being switched. The correct affiliations are: Roland A. Fleck: 8Centre for Ultrastructural Imaging, Kings College London, London, UK. José Antonio Del Río: 2Cellular and Molecular Neurobiotechnology, Institute for Bioengineering of Catalonia, Barcelona, Spain; 9Department of Cell Biology, Physiology and Immunology, Facultat de Biologia, Universitat de Barcelona, Barcelona, Spain; 10Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Barcelona, Spain. This has now been amended in all online versions of the Article.

19.
Science ; 359(6382): 1416-1421, 2018 03 23.
Article in English | MEDLINE | ID: mdl-29567716

ABSTRACT

How is protein synthesis initiated locally in neurons? We found that mTOR (mechanistic target of rapamycin) was activated and then up-regulated in injured axons, owing to local translation of mTOR messenger RNA (mRNA). This mRNA was transported into axons by the cell size-regulating RNA-binding protein nucleolin. Furthermore, mTOR controlled local translation in injured axons. This included regulation of its own translation and that of retrograde injury signaling molecules such as importin ß1 and STAT3 (signal transducer and activator of transcription 3). Deletion of the mTOR 3' untranslated region (3'UTR) in mice reduced mTOR in axons and decreased local translation after nerve injury. Both pharmacological inhibition of mTOR in axons and deletion of the mTOR 3'UTR decreased proprioceptive neuronal survival after nerve injury. Thus, mRNA localization enables spatiotemporal control of mTOR pathways regulating local translation and long-range intracellular signaling.


Subject(s)
Axons/metabolism , Ganglia, Spinal/injuries , Protein Biosynthesis , Sciatic Nerve/injuries , TOR Serine-Threonine Kinases/biosynthesis , 3' Untranslated Regions , Animals , Cell Size , Mice , Mice, Inbred Strains , Phosphoproteins/metabolism , RNA, Messenger/metabolism , RNA-Binding Proteins/metabolism , Rats , Rats, Inbred BB , Rats, Sprague-Dawley , Signal Transduction , TOR Serine-Threonine Kinases/genetics , Nucleolin
20.
Nat Cell Biol ; 20(3): 307-319, 2018 03.
Article in English | MEDLINE | ID: mdl-29434374

ABSTRACT

Reactive oxygen species (ROS) contribute to tissue damage and remodelling mediated by the inflammatory response after injury. Here we show that ROS, which promote axonal dieback and degeneration after injury, are also required for axonal regeneration and functional recovery after spinal injury. We find that ROS production in the injured sciatic nerve and dorsal root ganglia requires CX3CR1-dependent recruitment of inflammatory cells. Next, exosomes containing functional NADPH oxidase 2 complexes are released from macrophages and incorporated into injured axons via endocytosis. Once in axonal endosomes, active NOX2 is retrogradely transported to the cell body through an importin-ß1-dynein-dependent mechanism. Endosomal NOX2 oxidizes PTEN, which leads to its inactivation, thus stimulating PI3K-phosporylated (p-)Akt signalling and regenerative outgrowth. Challenging the view that ROS are exclusively involved in nerve degeneration, we propose a previously unrecognized role of ROS in mammalian axonal regeneration through a NOX2-PI3K-p-Akt signalling pathway.


Subject(s)
Axons/enzymology , Exosomes/enzymology , Ganglia, Spinal/enzymology , NADPH Oxidase 2/metabolism , Nerve Degeneration , Nerve Regeneration , Peripheral Nerve Injuries/enzymology , Reactive Oxygen Species/metabolism , Sciatic Nerve/enzymology , Spinal Cord Injuries/enzymology , Animals , Axons/pathology , CX3C Chemokine Receptor 1/metabolism , Cell Line , Disease Models, Animal , Dyneins/metabolism , Endocytosis , Endosomes/enzymology , Endosomes/pathology , Exosomes/pathology , Ganglia, Spinal/injuries , Ganglia, Spinal/pathology , Macrophages/enzymology , Macrophages/pathology , Mice, Inbred C57BL , Mice, Knockout , NADPH Oxidase 2/deficiency , NADPH Oxidase 2/genetics , Nuclear Proteins/metabolism , PTEN Phosphohydrolase/metabolism , Peripheral Nerve Injuries/genetics , Peripheral Nerve Injuries/pathology , Peripheral Nerve Injuries/physiopathology , Phosphatidylinositol 3-Kinase/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Sciatic Nerve/injuries , Sciatic Nerve/pathology , Sciatic Nerve/physiopathology , Signal Transduction , Spinal Cord Injuries/genetics , Spinal Cord Injuries/pathology , Spinal Cord Injuries/physiopathology , beta Karyopherins
SELECTION OF CITATIONS
SEARCH DETAIL
...