Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 4 de 4
Filter
Add more filters










Database
Language
Publication year range
1.
Nat Commun ; 14(1): 2704, 2023 05 17.
Article in English | MEDLINE | ID: mdl-37198183

ABSTRACT

In the early stage of bacterial translation, peptidyl-tRNAs frequently dissociate from the ribosome (pep-tRNA drop-off) and are recycled by peptidyl-tRNA hydrolase. Here, we establish a highly sensitive method for profiling of pep-tRNAs using mass spectrometry, and successfully detect a large number of nascent peptides from pep-tRNAs accumulated in Escherichia coli pthts strain. Based on molecular mass analysis, we found about 20% of the peptides bear single amino-acid substitutions of the N-terminal sequences of E. coli ORFs. Detailed analysis of individual pep-tRNAs and reporter assay revealed that most of the substitutions take place at the C-terminal drop-off site and that the miscoded pep-tRNAs rarely participate in the next round of elongation but dissociate from the ribosome. These findings suggest that pep-tRNA drop-off is an active mechanism by which the ribosome rejects miscoded pep-tRNAs in the early elongation, thereby contributing to quality control of protein synthesis after peptide bond formation.


Subject(s)
Escherichia coli , RNA, Transfer, Amino Acyl , Escherichia coli/genetics , Escherichia coli/metabolism , RNA, Transfer, Amino Acyl/metabolism , Ribosomes/metabolism , RNA, Transfer/genetics , RNA, Transfer/chemistry , Peptides/chemistry , Quality Control , Protein Biosynthesis
2.
Methods Mol Biol ; 2515: 89-97, 2022.
Article in English | MEDLINE | ID: mdl-35776347

ABSTRACT

Degeneration of axons is characteristic of many devastating diseases including amyotrophic lateral sclerosis (ALS). However, lack of an in vitro neuronal culture system that mimics damages on nerves and axonal tracts hampered development of effective treatments. Here, we describe a method to model degeneration of motor neuron axons using motor nerve organoids that are formed with human induced pluripotent stem cells. In this protocol, motor neuron axon degeneration can be rapidly induced with chemical damages. Neuroprotective effects of compounds can be examined using the degenerated axons. This motor neuron axon bundle degeneration model should facilitate future screening for drugs against diseases affecting axon fascicles.


Subject(s)
Amyotrophic Lateral Sclerosis , Induced Pluripotent Stem Cells , Nerve Tissue , Humans , Motor Neurons , Organoids
3.
J Vis Exp ; (163)2020 09 24.
Article in English | MEDLINE | ID: mdl-33044443

ABSTRACT

A fascicle of axons is one of the major structural motifs observed in the nervous system. Disruption of axon fascicles could cause developmental and neurodegenerative diseases. Although numerous studies of axons have been conducted, our understanding of formation and dysfunction of axon fascicles is still limited due to the lack of robust three-dimensional in vitro models. Here, we describe a step-by-step protocol for the rapid generation of a motor nerve organoid (MNO) from human induced pluripotent stem (iPS) cells in a microfluidic-based tissue culture chip. First, fabrication of chips used for the method is described. From human iPS cells, a motor neuron spheroid (MNS) is formed. Next, the differentiated MNS is transferred into the chip. Thereafter, axons spontaneously grow out of the spheroid and assemble into a fascicle within a microchannel equipped in the chip, which generates an MNO tissue carrying a bundle of axons extended from the spheroid. For the downstream analysis, MNOs can be taken out of the chip to be fixed for morphological analyses or dissected for biochemical analyses, as well as calcium imaging and multi-electrode array recordings. MNOs generated with this protocol can facilitate drug testing and screening and can contribute to understanding of mechanisms underlying development and diseases of axon fascicles.


Subject(s)
Motor Neurons/physiology , Organoids/physiology , Animals , Calcium/metabolism , Cell Differentiation , Dimethylpolysiloxanes/chemistry , Electrodes , Epoxy Compounds/chemistry , Humans , Induced Pluripotent Stem Cells/cytology , Microfluidics , Polymers/chemistry , Tissue Culture Techniques
4.
J Neurosci ; 38(21): 4985-4995, 2018 05 23.
Article in English | MEDLINE | ID: mdl-29712777

ABSTRACT

The mTOR signaling pathway regulates protein synthesis and diverse aspects of neuronal morphology that are important for brain development and function. To identify proteins controlled translationally by mTOR signaling, we performed ribosome profiling analyses in mouse cortical neurons and embryonic stem cells upon acute mTOR inhibition. Among proteins whose translation was significantly affected by mTOR inhibition selectively in neurons, we identified the cytoskeletal regulator protein palladin, which is localized within the cell body and axons in hippocampal neurons. Knockdown of palladin eliminated supernumerary axons induced by suppression of the tuberous sclerosis complex protein TSC1 in neurons, demonstrating that palladin regulates neuronal morphogenesis downstream of mTOR signaling. Our findings provide novel insights into an mTOR-dependent mechanism that controls neuronal morphogenesis through translational regulation.SIGNIFICANCE STATEMENT This study reports the discovery of neuron-specific protein translational responses to alterations of mTOR activity. By using ribosome profiling analysis, which can reveal the location and quantity of translating ribosomes on mRNAs, multiple aspects of protein translation were quantitatively analyzed in mouse embryonic stem cells and cortical neurons upon acute mTOR inhibition. Neurons displayed distinct patterns of ribosome occupancy for each codon and ribosome stalling during translation at specific positions of mRNAs. Importantly, the cytoskeletal regulator palladin was identified as a translational target protein of mTOR signaling in neurons. Palladin operates downstream of mTOR to modulate axon morphogenesis. This study identifies a novel mechanism of neuronal morphogenesis regulated by mTOR signaling through control of translation of the key protein palladin.


Subject(s)
Axons/physiology , Cytoskeletal Proteins/physiology , Morphogenesis/genetics , Morphogenesis/physiology , Phosphoproteins/physiology , Signal Transduction/physiology , TOR Serine-Threonine Kinases/physiology , Animals , Cells, Cultured , Cytoskeletal Proteins/genetics , Female , Gene Knockdown Techniques , Male , Mice , Mice, Inbred ICR , Phosphoproteins/genetics , Protein Biosynthesis , RNA Editing , Ribosomes/chemistry , Ribosomes/genetics , TOR Serine-Threonine Kinases/genetics , Tuberous Sclerosis Complex 1 Protein/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...