Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 3 de 3
Filter
Add more filters










Database
Language
Publication year range
1.
J Biol Chem ; 298(3): 101647, 2022 03.
Article in English | MEDLINE | ID: mdl-35101451

ABSTRACT

The dual leucine zipper kinase (DLK) is a key regulator of axon regeneration and degeneration in response to neuronal injury; however, regulatory mechanisms of the DLK function via its interacting proteins are largely unknown. To better understand the molecular mechanism of DLK function, we performed yeast two-hybrid screening analysis and identified FK506-binding protein-like (FKBPL, also known as WAF-1/CIP1 stabilizing protein 39) as a DLK-binding protein. FKBPL binds to the kinase domain of DLK and inhibits its kinase activity. In addition, FKBPL induces DLK protein degradation through ubiquitin-dependent pathways. We further assessed other members in the FKBP protein family and found that FK506-binding protein 8 (FKBP8) also induced DLK degradation. We identified the lysine 271 residue in the kinase domain as a major site of DLK ubiquitination and SUMO3 conjugation and was thus responsible for regulating FKBP8-mediated proteasomal degradation that was inhibited by the substitution of the lysine 271 to arginine. FKBP8-mediated degradation of DLK is mediated by autophagy pathway because knockdown of Atg5 inhibited DLK destabilization. We show that in vivo overexpression of FKBP8 delayed the progression of axon degeneration and suppressed neuronal death after axotomy in sciatic and optic nerves. Taken together, this study identified FKBPL and FKBP8 as novel DLK-interacting proteins that regulate DLK stability via the ubiquitin-proteasome and lysosomal protein degradation pathways.


Subject(s)
Axons , MAP Kinase Kinase Kinases , Nerve Degeneration , Tacrolimus Binding Proteins , Axons/enzymology , Axons/metabolism , Axons/pathology , Leucine Zippers , Lysine/metabolism , MAP Kinase Kinase Kinases/metabolism , Nerve Degeneration/enzymology , Nerve Degeneration/metabolism , Nerve Degeneration/pathology , Nerve Regeneration , Tacrolimus Binding Proteins/metabolism , Ubiquitin/metabolism
2.
Int J Mol Sci ; 22(2)2021 Jan 06.
Article in English | MEDLINE | ID: mdl-33418850

ABSTRACT

Neurons are structurally unique and have dendrites and axons that are vulnerable to injury. Some neurons in the peripheral nervous system (PNS) can regenerate their axons after injuries. However, most neurons in the central nervous system (CNS) fail to do so, resulting in irreversible neurological disorders. To understand the mechanisms of axon regeneration, various experimental models have been utilized in vivo and in vitro. Here, we collate the key experimental models that revealed the important mechanisms regulating axon regeneration and degeneration in different systems. We also discuss the advantages of experimenting with the rodent model, considering the application of these findings in understanding human diseases and for developing therapeutic methods.


Subject(s)
Axons/metabolism , Models, Theoretical , Animals , Cytoskeletal Proteins/metabolism , Humans , Nerve Regeneration , Nervous System Diseases/metabolism , Nervous System Diseases/pathology , Neurons/cytology , Neurons/metabolism , Sciatic Nerve/injuries , Sciatic Nerve/physiology
3.
Proc Natl Acad Sci U S A ; 117(27): 15955-15966, 2020 07 07.
Article in English | MEDLINE | ID: mdl-32554499

ABSTRACT

Axon regeneration is regulated by a neuron-intrinsic transcriptional program that is suppressed during development but that can be reactivated following peripheral nerve injury. Here we identify Prom1, which encodes the stem cell marker prominin-1, as a regulator of the axon regeneration program. Prom1 expression is developmentally down-regulated, and the genetic deletion of Prom1 in mice inhibits axon regeneration in dorsal root ganglion (DRG) cultures and in the sciatic nerve, revealing the neuronal role of Prom1 in injury-induced regeneration. Elevating prominin-1 levels in cultured DRG neurons or in mice via adeno-associated virus-mediated gene delivery enhances axon regeneration in vitro and in vivo, allowing outgrowth on an inhibitory substrate. Prom1 overexpression induces the consistent down-regulation of cholesterol metabolism-associated genes and a reduction in cellular cholesterol levels in a Smad pathway-dependent manner, which promotes axonal regrowth. We find that prominin-1 interacts with the type I TGF-ß receptor ALK4, and that they synergistically induce phosphorylation of Smad2. These results suggest that Prom1 and cholesterol metabolism pathways are possible therapeutic targets for the promotion of neural recovery after injury.


Subject(s)
AC133 Antigen/metabolism , Axons/metabolism , Cholesterol/metabolism , Nerve Regeneration/physiology , Signal Transduction , Stem Cells/metabolism , AC133 Antigen/genetics , Activin Receptors, Type I , Animals , Axons/pathology , Cholesterol/genetics , Down-Regulation , Ganglia, Spinal/metabolism , Gene Deletion , Gene Expression Regulation , Mice , Mice, Knockout , Neurons/metabolism , Peripheral Nerve Injuries/metabolism , Receptor, Transforming Growth Factor-beta Type I/metabolism , Sciatic Nerve
SELECTION OF CITATIONS
SEARCH DETAIL
...