Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
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.
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
4.
Autophagy ; 17(10): 3082-3095, 2021 10.
Article in English | MEDLINE | ID: mdl-33218264

ABSTRACT

Mutations in the coding sequence of human TECPR2 were recently linked to spastic paraplegia type 49 (SPG49), a hereditary neurodegenerative disorder involving intellectual disability, autonomic-sensory neuropathy, chronic respiratory disease and decreased pain sensitivity. Here, we report the generation of a novel CRISPR-Cas9 tecpr2 knockout (tecpr2-/-) mouse that exhibits behavioral pathologies observed in SPG49 patients. tecpr2-/- mice develop neurodegenerative patterns in an age-dependent manner, manifested predominantly as neuroaxonal dystrophy in the gracile (GrN) and cuneate nuclei (CuN) of the medulla oblongata in the brainstem and dorsal white matter column of the spinal cord. Age-dependent correlation with accumulation of autophagosomes suggests compromised targeting to lysosome. Taken together, our findings establish the tecpr2 knockout mouse as a potential model for SPG49 and ascribe a new role to TECPR2 in macroautophagy/autophagy-related neurodegenerative disorders.


Subject(s)
Autophagosomes , Carrier Proteins , Nerve Tissue Proteins , Neuroaxonal Dystrophies , Animals , Autophagosomes/metabolism , Autophagy/genetics , Carrier Proteins/genetics , Humans , Mice , Mice, Knockout , Nerve Tissue Proteins/genetics
5.
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
6.
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
7.
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
8.
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
9.
Hippocampus ; 26(5): 658-67, 2016 May.
Article in English | MEDLINE | ID: mdl-26540287

ABSTRACT

Sprouty (Spry) proteins play a key role as negative feedback inhibitors of the Ras/Raf/MAPK/ERK pathway downstream of various receptor tyrosine kinases. Among the four Sprouty isoforms, Spry2 and Spry4 are expressed in the hippocampus. In this study, possible effects of Spry2 and Spry4 hypomorphism on neurodegeneration and seizure thresholds in a mouse model of epileptogenesis was analyzed. The Spry2/4 hypomorphs exhibited stronger ERK activation which was limited to the CA3 pyramidal cell layer and to the hilar region. The seizure threshold of Spry2/4(+/-) mice was significantly reduced at naive state but no difference to wildtype mice was observed 1 month following KA treatment. Histomorphological analysis revealed that dentate granule cell dispersion (GCD) was diminished in Spry2/4(+/-) mice in the subchronic phase after KA injection. Neuronal degeneration was reduced in CA1 and CA3 principal neuron layers as well as in scattered neurons of the contralateral CA1 and hilar regions. Moreover, Spry2/4 reduction resulted in enhanced survival of somatostatin and neuropeptide Y expressing interneurons. GFAP staining intensity and number of reactive astrocytes markedly increased in lesioned areas of Spry2/4(+/-) mice as compared with wildtype mice. Taken together, although the seizure threshold is reduced in naive Spry2/4(+/-) mice, neurodegeneration and GCD is mitigated following KA induced hippocampal lesions, identifying Spry proteins as possible pharmacological targets in brain injuries resulting in neurodegeneration. The present data are consistent with the established functions of the ERK pathway in astrocyte proliferation as well as protection from neuronal cell death and suggest a novel role of Spry proteins in the migration of differentiated neurons.


Subject(s)
Brain Injuries/pathology , Gliosis/pathology , Hippocampus/pathology , Intracellular Signaling Peptides and Proteins/deficiency , Membrane Proteins/deficiency , Nerve Tissue Proteins/deficiency , Neurons/pathology , Analysis of Variance , Animals , Brain Injuries/chemically induced , Brain Injuries/complications , Cell Survival/drug effects , Cell Survival/genetics , Disease Models, Animal , Excitatory Amino Acid Agonists/toxicity , Female , Gene Expression Regulation/drug effects , Gene Expression Regulation/genetics , Gliosis/chemically induced , Hippocampus/drug effects , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/metabolism , Kainic Acid/toxicity , Male , Membrane Proteins/genetics , Mice , Mice, Transgenic , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Neurons/metabolism , Pentylenetetrazole/toxicity , Protein Serine-Threonine Kinases , RNA, Messenger/metabolism , Seizures/chemically induced , Signal Transduction/drug effects , Signal Transduction/genetics
10.
Dev Neurobiol ; 75(3): 217-31, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25104556

ABSTRACT

Sprouty (Spry) proteins are negative feedback inhibitors of receptor tyrosine kinase signaling. Downregulation of Spry2 has been demonstrated to promote elongative axon growth of cultured peripheral and central neurons. Here, we analyzed Spry2 global knockout mice with respect to axon outgrowth in vitro and peripheral axon regeneration in vivo. Neurons dissociated from adult Spry2 deficient sensory ganglia revealed stronger extracellular signal-regulated kinase activation and enhanced axon outgrowth. Prominent axon elongation was observed in heterozygous Spry2(+/-) neuron cultures, whereas homozygous Spry2(-/-) neurons predominantly exhibited a branching phenotype. Following sciatic nerve crush, Spry2(+/-) mice recovered faster in motor but not sensory testing paradigms (Spry2(-/-) mice did not tolerate anesthesia required for nerve surgery). We attribute the improvement in the rotarod test to higher numbers of myelinated fibers in the regenerating sciatic nerve, higher densities of motor endplates in hind limb muscles and increased levels of GAP-43 mRNA, a downstream target of extracellular regulated kinase signaling. Conversely, homozygous Spry2(-/-) mice revealed enhanced mechanosensory function (von Frey's test) that was accompanied by an increased innervation of the epidermis, elevated numbers of nonmyelinated axons and more IB4-positive neurons in dorsal root ganglia. The present results corroborate the functional significance of receptor tyrosine kinase signaling inhibitors for axon outgrowth during development and nerve regeneration and propose Spry2 as a novel potential target for pharmacological inhibition to accelerate long-distance axon regeneration in injured peripheral nerves.


Subject(s)
Axons/physiology , Intracellular Signaling Peptides and Proteins/deficiency , Membrane Proteins/deficiency , Nerve Regeneration/genetics , Neurons/metabolism , Animals , GAP-43 Protein/metabolism , Heterozygote , Homozygote , Intracellular Signaling Peptides and Proteins/metabolism , Membrane Proteins/metabolism , Mice , Mice, 129 Strain , Mice, Inbred BALB C , Mice, Knockout , Motor Activity/physiology , Nerve Regeneration/physiology , Protein Serine-Threonine Kinases , Recovery of Function/physiology , Sciatic Nerve/injuries
11.
Neurosci Lett ; 566: 280-5, 2014 Apr 30.
Article in English | MEDLINE | ID: mdl-24631569

ABSTRACT

Intramuscular injection of the calpain inhibitor leupeptin promotes peripheral nerve regeneration in primates (Badalamente et al., 1989 [13]), and direct positive effects of leupeptin on axon outgrowth were observed in vitro (Hausott et al., 2012 [12]). In this study, we applied leupeptin (2mg/ml) directly to collagen-filled nerve conduits in the rat sciatic nerve transection model. Analysis of myelinated axons and retrogradely labeled motoneurons as well as functional 'CatWalk' video analysis did not reveal significant differences between vehicle controls and leupeptin treated animals. Therefore, leupeptin does not improve nerve regeneration via protease inhibition in regrowing axons or in surrounding Schwann cells following a single application to a peripheral nerve conduit suggesting indirect effects on motor endplate integrity if applied systemically.


Subject(s)
Calpain/antagonists & inhibitors , Cysteine Proteinase Inhibitors/pharmacology , Leupeptins/pharmacology , Nerve Regeneration/drug effects , Sciatic Nerve/drug effects , Action Potentials , Animals , Cysteine Proteinase Inhibitors/administration & dosage , Leupeptins/administration & dosage , Male , Muscle, Skeletal/drug effects , Muscle, Skeletal/innervation , Neural Conduction , Rats, Sprague-Dawley , Sciatic Nerve/injuries , Sciatic Nerve/physiopathology
12.
Neurochem Res ; 39(2): 403-7, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24407843

ABSTRACT

Recently, a new potent protein kinase inhibitor, SC82510, was identified acting on DRAK2 and stimulating axon outgrowth at low concentrations. DRAK is the Drosophila homologue of death-associated protein kinase that phosphorylates myosin-II regulatory light chain in a similar fashion as ROCK, the downstream target of RhoA mediating axon outgrowth inhibition. While higher concentrations of this novel compound exhibited toxic effects, significant promotion of process outgrowth of PC12 cells and of adult primary neurons was observed at 1 nM which could be further enhanced by addition of a neuronal growth factor (FGF-2). Unlike the effects of ROCK inhibitors on axon outgrowth that stimulate both, elongation and branching, SC82510 primarily promoted axon branching, whereas axon elongation was not increased in this cell culture model of peripheral axon regeneration.


Subject(s)
Protein Kinase Inhibitors/pharmacology , Sensory Receptor Cells/drug effects , Animals , Culture Media , In Vitro Techniques , PC12 Cells , Rats
13.
Biochim Biophys Acta ; 1833(12): 3166-3175, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24036102

ABSTRACT

The Transient Receptor Potential Vanilloid 1 (TRPV1, vanilloid receptor 1) ion channel plays a key role in the perception of thermal and inflammatory pain, however, its molecular environment in dorsal root ganglia (DRG) is largely unexplored. Utilizing a panel of sequence-directed antibodies against TRPV1 protein and mouse DRG membranes, the channel complex from mouse DRG was detergent-solubilized, isolated by immunoprecipitation and subsequently analyzed by mass spectrometry. A number of potential TRPV1 interaction partners were identified, among them cytoskeletal proteins, signal transduction molecules, and established ion channel subunits. Based on stringent specificity criteria, the voltage-gated K(+) channel beta 2 subunit (Kvß2), an accessory subunit of voltage-gated K(+) channels, was identified of being associated with native TRPV1 channels. Reverse co-immunoprecipitation and antibody co-staining experiments confirmed TRPV1/Kvß2 association. Biotinylation assays in the presence of Kvß2 demonstrated increased cell surface expression levels of TRPV1, while patch-clamp experiments resulted in a significant increase of TRPV1 sensitivity to capsaicin. Our work shows, for the first time, the association of a Kvß subunit with TRPV1 channels, and suggests that such interaction may play a role in TRPV1 channel trafficking to the plasma membrane.


Subject(s)
Protein Subunits/metabolism , Shaker Superfamily of Potassium Channels/metabolism , TRPV Cation Channels/metabolism , Animals , Biotinylation , Cell Membrane/metabolism , HEK293 Cells , Humans , Immunoprecipitation , Mass Spectrometry , Mice , Mice, Knockout , Patch-Clamp Techniques , Protein Binding , Rats , Recombinant Proteins/metabolism , Shaker Superfamily of Potassium Channels/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL
...