Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters










Database
Publication year range
1.
Cell Struct Funct ; 48(2): 211-221, 2023 Nov 03.
Article in English | MEDLINE | ID: mdl-37766570

ABSTRACT

Secretory pathway proteins are cotranslationally translocated into the endoplasmic reticulum (ER) of metazoan cells through the protein channel, translocon. Given that there are far fewer translocons than ribosomes in a cell, it is essential that secretory protein-translating ribosomes only occupy translocons transiently. Therefore, if translocons are obstructed by ribosomes stalled or slowed in translational elongation, it possibly results in deleterious consequences to cellular function. Hence, we investigated how translocon clogging by stalled ribosomes affects mammalian cells. First, we constructed ER-destined translational arrest proteins (ER-TAP) as an artificial protein that clogged the translocon in the ER membrane. Here, we show that the translocon clogging by ER-TAP expression activates triage of signal sequences (SS) in which secretory pathway proteins harboring highly efficient SS are preferentially translocated into the ER lumen. Interestingly, the translocon obstructed status specifically activates inositol requiring enzyme 1α (IRE1α) but not protein kinase R-like ER kinase (PERK). Given that the IRE1α-XBP1 pathway mainly induces the translocon components, our discovery implies that lowered availability of translocon activates IRE1α, which induces translocon itself. This results in rebalance between protein influx into the ER and the cellular translocation capacity.Key words: endoplasmic reticulum, translocation capacity, translocon clogging, IRE1, signal sequence.


Subject(s)
Endoribonucleases , Protein Serine-Threonine Kinases , Animals , Endoribonucleases/metabolism , Protein Serine-Threonine Kinases/metabolism , Protein Sorting Signals , Triage , Endoplasmic Reticulum Stress , Mammals/metabolism
2.
EMBO J ; 39(24): e103303, 2020 12 15.
Article in English | MEDLINE | ID: mdl-33215740

ABSTRACT

HOIP, the catalytic component of the linear ubiquitin chain assembly complex (LUBAC), is a critical regulator of inflammation. However, how HOIP itself is regulated to control inflammatory responses is unclear. Here, we discover that site-specific ubiquitination of K784 within human HOIP promotes tumor necrosis factor (TNF)-induced inflammatory signaling. A HOIP K784R mutant is catalytically active but shows reduced induction of an NF-κB reporter relative to wild-type HOIP. HOIP K784 is evolutionarily conserved, equivalent to HOIP K778 in mice. We generated HoipK778R/K778R knock-in mice, which show no overt developmental phenotypes; however, in response to TNF, HoipK778R/K778R mouse embryonic fibroblasts display mildly suppressed NF-κB activation and increased apoptotic markers. On the other hand, HOIP K778R enhances the TNF-induced formation of TNFR complex II and an interaction between TNFR complex II and LUBAC. Loss of the LUBAC component SHARPIN leads to embryonic lethality in HoipK778R/K778R mice, which is rescued by knockout of TNFR1. We propose that site-specific ubiquitination of HOIP regulates a LUBAC-dependent switch between survival and apoptosis in TNF signaling.


Subject(s)
Apoptosis/drug effects , Signal Transduction/drug effects , Ubiquitin-Protein Ligases/metabolism , Ubiquitination/drug effects , Animals , Female , Gene Knock-In Techniques , HEK293 Cells , Humans , Male , Mice , NF-kappa B/metabolism , Receptors, Tumor Necrosis Factor, Type I/genetics , Receptors, Tumor Necrosis Factor, Type I/metabolism , Receptors, Tumor Necrosis Factor, Type II , Transcriptome , Ubiquitin/metabolism , Ubiquitin-Protein Ligases/genetics , Ubiquitin-Protein Ligases/pharmacology
3.
Science ; 357(6350): 472-475, 2017 08 04.
Article in English | MEDLINE | ID: mdl-28774922

ABSTRACT

Many nascent proteins are assembled into multiprotein complexes of defined stoichiometry. Imbalances in the synthesis of individual subunits result in orphans. How orphans are selectively eliminated to maintain protein homeostasis is poorly understood. Here, we found that the conserved ubiquitin-conjugating enzyme UBE2O directly recognized juxtaposed basic and hydrophobic patches on unassembled proteins to mediate ubiquitination without a separate ubiquitin ligase. In reticulocytes, where UBE2O is highly up-regulated, unassembled α-globin molecules that failed to assemble with ß-globin were selectively ubiquitinated by UBE2O. In nonreticulocytes, ribosomal proteins that did not engage nuclear import factors were targets for UBE2O. Thus, UBE2O is a self-contained quality control factor that comprises substrate recognition and ubiquitin transfer activities within a single protein to efficiently target orphans of multiprotein complexes for degradation.


Subject(s)
Multiprotein Complexes/metabolism , Proteolysis , Ribosomal Proteins/metabolism , Ubiquitin-Conjugating Enzymes/metabolism , Ubiquitination , Active Transport, Cell Nucleus , Cytosol/enzymology , HEK293 Cells , Humans , Hydrophobic and Hydrophilic Interactions , Reticulocytes/enzymology , SEC Translocation Channels/metabolism , Ubiquitin/metabolism , alpha-Globins/metabolism , beta-Globins/metabolism
4.
Proc Natl Acad Sci U S A ; 113(40): E5886-E5895, 2016 10 04.
Article in English | MEDLINE | ID: mdl-27651490

ABSTRACT

Unconventional mRNA splicing on the endoplasmic reticulum (ER) membrane is the sole conserved mechanism in eukaryotes to transmit information regarding misfolded protein accumulation to the nucleus to activate the stress response. In metazoans, the unspliced form of X-box-binding protein 1 (XBP1u) mRNA is recruited to membranes as a ribosome nascent chain (RNC) complex for efficient splicing. We previously reported that both hydrophobic (HR2) and translational pausing regions of XBP1u are important for the recruitment of its own mRNA to membranes. However, its precise location and the molecular mechanism of translocation are unclear. We show that XBP1u-RNC is specifically recruited to the ER membrane in an HR2- and translational pausing-dependent manner by immunostaining, fluorescent recovery after photobleaching, and biochemical analyses. Notably, translational pausing during XBP1u synthesis is indispensable for the recognition of HR2 by the signal recognition particle (SRP), resulting in efficient ER-specific targeting of the complex, similar to secretory protein targeting to the ER. On the ER, the XBP1u nascent chain is transferred from the SRP to the translocon; however, it cannot pass through the translocon or insert into the membrane. Therefore, our results support a noncanonical mechanism by which mRNA substrates are recruited to the ER for unconventional splicing.


Subject(s)
Endoplasmic Reticulum/metabolism , Protein Biosynthesis , Signal Recognition Particle/metabolism , Signal Transduction , X-Box Binding Protein 1/genetics , Amino Acid Sequence , Animals , COS Cells , Chlorocebus aethiops , HEK293 Cells , HeLa Cells , Humans , Intracellular Membranes/metabolism , Models, Biological , Nuclear Localization Signals/metabolism , Protein Binding , Protein Transport , RNA Splicing , RNA, Messenger/genetics , RNA, Messenger/metabolism , X-Box Binding Protein 1/chemistry , X-Box Binding Protein 1/metabolism
6.
Nucleic Acids Res ; 39(12): 5245-54, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21398633

ABSTRACT

Upon endoplasmic reticulum (ER) stress, mammalian cells induce the synthesis of a transcriptional activator XBP1s to alleviate the stress. Under unstressed conditions, the messenger RNA (mRNA) for XBP1s exists in the cytosol as an unspliced precursor form, XBP1u mRNA. Thus, its intron must be removed for the synthesis of XBP1s. Upon ER stress, a stress sensor IRE1α cleaves XBP1u mRNA to initiate the unconventional splicing of XBP1u mRNA on the ER membrane. The liberated two exons are ligated to form the mature XBP1s mRNA. However, the mechanism of this splicing is still obscure mainly because the enzyme that joins XBP1s mRNA halves is unknown. Here, we reconstituted the whole splicing reaction of XBP1u mRNA in vitro. Using this assay, we showed that, consistent with the in vivo studies, mammalian cytosol indeed had RNA ligase that could join XBP1s mRNA halves. Further, the cleavage of XBP1u mRNA with IRE1α generated 2', 3'-cyclic phosphate structure at the cleavage site. Interestingly, this phosphate was incorporated into XBP1s mRNA by the enzyme in the cytosol to ligate the two exons. These studies reveal the utility of the assay system and the unique properties of the mammalian cytosolic enzyme that can promote the splicing of XBP1u mRNA.


Subject(s)
DNA-Binding Proteins/genetics , RNA Splicing , RNA, Messenger/metabolism , Transcription Factors/genetics , DNA-Binding Proteins/metabolism , Endoribonucleases/metabolism , Exons , Phosphates/chemistry , Protein Serine-Threonine Kinases/metabolism , RNA Ligase (ATP)/isolation & purification , RNA Ligase (ATP)/metabolism , RNA, Messenger/chemistry , Regulatory Factor X Transcription Factors , Transcription Factors/metabolism
7.
Science ; 331(6017): 586-9, 2011 Feb 04.
Article in English | MEDLINE | ID: mdl-21233347

ABSTRACT

Upon endoplasmic reticulum (ER) stress, an endoribonuclease, inositol-requiring enzyme-1α, splices the precursor unspliced form of X-box-binding protein 1 messenger RNA (XBP1u mRNA) on the ER membrane to yield an active transcription factor (XBP1s), leading to the alleviation of the stress. The nascent peptide encoded by XBP1u mRNA drags the mRNA-ribosome-nascent chain (R-RNC) complex to the membrane for efficient cytoplasmic splicing. We found that translation of the XBP1u mRNA was briefly paused to stabilize the R-RNC complex. Mutational analysis of XBP1u revealed an evolutionarily conserved peptide module at the carboxyl terminus that was responsible for the translational pausing and was required for the efficient targeting and splicing of the XBP1u mRNA. Thus, translational pausing may be used for unexpectedly diverse cellular processes in mammalian cells.


Subject(s)
Cytoplasm/metabolism , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Endoplasmic Reticulum/metabolism , Protein Biosynthesis , RNA Splicing , RNA, Messenger/genetics , Transcription Factors/genetics , Transcription Factors/metabolism , Cell Line , DNA-Binding Proteins/chemistry , Endoribonucleases/metabolism , Humans , Hydrophobic and Hydrophilic Interactions , Intracellular Membranes/metabolism , Protein Serine-Threonine Kinases/metabolism , RNA, Messenger/metabolism , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/metabolism , Regulatory Factor X Transcription Factors , Ribosomes/metabolism , Transcription Factors/chemistry , X-Box Binding Protein 1
8.
Mol Cell ; 34(2): 191-200, 2009 Apr 24.
Article in English | MEDLINE | ID: mdl-19394296

ABSTRACT

Endoplasmic reticulum (ER) stress triggers the cytoplasmic splicing of XBP1 mRNA by the transmembrane endoribonuclease IRE1alpha, resulting in activation of the unfolded protein response, which maintains ER homeostasis. We show that the unspliced XBP1 (XBP1u) mRNA is localized to the membrane, although its product is neither a secretory nor a membrane protein and is released to the cytosol after splicing. Biochemical and mutagenic analyses demonstrated that membrane localization of XBP1u mRNA required its in-frame translation. An insertional frame-shift mutation greatly diminished both membrane localization and splicing of the XBP1u mRNA. Furthermore, membrane localization was compromised by puromycin treatment and required a hydrophobic region within XBP1u. These data demonstrate that the nascent XBP1u polypeptide recruits its own mRNA to the membrane. This system serves to enhance cytoplasmic splicing and could facilitate a more rapid response to ER stress, and represents a unique way of cotranslational protein targeting coupled to mRNA maturation.


Subject(s)
Cytoplasm/metabolism , DNA-Binding Proteins/metabolism , Intracellular Membranes/metabolism , RNA Splicing , RNA, Messenger/metabolism , Transcription Factors/metabolism , Animals , Base Sequence , DNA-Binding Proteins/analysis , DNA-Binding Proteins/chemistry , DNA-Binding Proteins/genetics , Endoribonucleases/metabolism , Endoribonucleases/physiology , Humans , Mice , Molecular Sequence Data , Protein Serine-Threonine Kinases/metabolism , Protein Serine-Threonine Kinases/physiology , Protein Structure, Tertiary , Protein Transport , RNA, Messenger/analysis , Regulatory Factor X Transcription Factors , Ribosomes/metabolism , Transcription Factors/analysis , Transcription Factors/chemistry , Transcription Factors/genetics , X-Box Binding Protein 1
SELECTION OF CITATIONS
SEARCH DETAIL
...