Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
1.
Molecules ; 26(12)2021 Jun 11.
Article in English | MEDLINE | ID: mdl-34208277

ABSTRACT

In human cells, one-third of all polypeptides enter the secretory pathway at the endoplasmic reticulum (ER). The specificity and efficiency of this process are guaranteed by targeting of mRNAs and/or polypeptides to the ER membrane. Cytosolic SRP and its receptor in the ER membrane facilitate the cotranslational targeting of most ribosome-nascent precursor polypeptide chain (RNC) complexes together with the respective mRNAs to the Sec61 complex in the ER membrane. Alternatively, fully synthesized precursor polypeptides are targeted to the ER membrane post-translationally by either the TRC, SND, or PEX19/3 pathway. Furthermore, there is targeting of mRNAs to the ER membrane, which does not involve SRP but involves mRNA- or RNC-binding proteins on the ER surface, such as RRBP1 or KTN1. Traditionally, the targeting reactions were studied in cell-free or cellular assays, which focus on a single precursor polypeptide and allow the conclusion of whether a certain precursor can use a certain pathway. Recently, cellular approaches such as proximity-based ribosome profiling or quantitative proteomics were employed to address the question of which precursors use certain pathways under physiological conditions. Here, we combined siRNA-mediated depletion of putative mRNA receptors in HeLa cells with label-free quantitative proteomics and differential protein abundance analysis to characterize RRBP1- or KTN1-involving precursors and to identify possible genetic interactions between the various targeting pathways. Furthermore, we discuss the possible implications on the so-called TIGER domains and critically discuss the pros and cons of this experimental approach.


Subject(s)
Carrier Proteins/metabolism , Endoplasmic Reticulum/metabolism , Intracellular Membranes/metabolism , Membrane Proteins/metabolism , RNA, Messenger/metabolism , Carrier Proteins/genetics , HeLa Cells , Humans , Membrane Proteins/genetics , Proteome/analysis , Proteome/metabolism , RNA, Messenger/genetics
2.
FEBS J ; 287(21): 4612-4640, 2020 11.
Article in English | MEDLINE | ID: mdl-32133789

ABSTRACT

In mammalian cells, one-third of all polypeptides are integrated into the membrane or translocated into the lumen of the endoplasmic reticulum (ER) via the Sec61 channel. While the Sec61 complex facilitates ER import of most precursor polypeptides, the Sec61-associated Sec62/Sec63 complex supports ER import in a substrate-specific manner. So far, mainly posttranslationally imported precursors and the two cotranslationally imported precursors of ERj3 and prion protein were found to depend on the Sec62/Sec63 complex in vitro. Therefore, we determined the rules for engagement of Sec62/Sec63 in ER import in intact human cells using a recently established unbiased proteomics approach. In addition to confirming ERj3, we identified 22 novel Sec62/Sec63 substrates under these in vivo-like conditions. As a common feature, those previously unknown substrates share signal peptides (SP) with comparatively longer but less hydrophobic hydrophobic region of SP and lower carboxy-terminal region of SP (C-region) polarity. Further analyses with four substrates, and ERj3 in particular, revealed the combination of a slowly gating SP and a downstream translocation-disruptive positively charged cluster of amino acid residues as decisive for the Sec62/Sec63 requirement. In the case of ERj3, these features were found to be responsible for an additional immunoglobulin heavy-chain binding protein (BiP) requirement and to correlate with sensitivity toward the Sec61-channel inhibitor CAM741. Thus, the human Sec62/Sec63 complex may support Sec61-channel opening for precursor polypeptides with slowly gating SPs by direct interaction with the cytosolic amino-terminal peptide of Sec61α or via recruitment of BiP and its interaction with the ER-lumenal loop 7 of Sec61α. These novel insights into the mechanism of human ER protein import contribute to our understanding of the etiology of SEC63-linked polycystic liver disease. DATABASES: The mass spectrometry proteomics data have been deposited to the ProteomeXchange Consortium via the PRIDE partner repository (http://www.ebi.ac.uk/pride/archive/projects/Identifiers) with the dataset identifiers: PXD008178, PXD011993, and PXD012078. Supplementary information was deposited at Mendeley Data (https://data.mendeley.com/datasets/6s5hn73jcv/2).


Subject(s)
Endoplasmic Reticulum/metabolism , Membrane Transport Proteins/metabolism , Molecular Chaperones/metabolism , Protein Sorting Signals , RNA-Binding Proteins/metabolism , Animals , HEK293 Cells , HSP40 Heat-Shock Proteins/metabolism , HeLa Cells , Humans , Membrane Transport Proteins/genetics , Mice, Inbred C57BL , Mice, Knockout , Molecular Chaperones/genetics , Protein Transport , Proteome/metabolism , Proteomics/methods , RNA-Binding Proteins/genetics , SEC Translocation Channels/genetics , SEC Translocation Channels/metabolism , Substrate Specificity
3.
Nat Commun ; 9(1): 3765, 2018 09 14.
Article in English | MEDLINE | ID: mdl-30217974

ABSTRACT

In mammalian cells, one-third of all polypeptides are transported into or across the ER membrane via the Sec61 channel. While the Sec61 complex facilitates translocation of all polypeptides with amino-terminal signal peptides (SP) or transmembrane helices, the Sec61-auxiliary translocon-associated protein (TRAP) complex supports translocation of only a subset of precursors. To characterize determinants of TRAP substrate specificity, we here systematically identify TRAP-dependent precursors by analyzing cellular protein abundance changes upon TRAP depletion using quantitative label-free proteomics. The results are validated in independent experiments by western blotting, quantitative RT-PCR, and complementation analysis. The SPs of TRAP clients exhibit above-average glycine-plus-proline content and below-average hydrophobicity as distinguishing features. Thus, TRAP may act as SP receptor on the ER membrane's cytosolic face, recognizing precursor polypeptides with SPs of high glycine-plus-proline content and/or low hydrophobicity, and triggering substrate-specific opening of the Sec61 channel through interactions with the ER-lumenal hinge of Sec61α.


Subject(s)
Calcium-Binding Proteins/metabolism , Endoplasmic Reticulum/metabolism , Membrane Glycoproteins/metabolism , Protein Sorting Signals , Protein Transport , Receptors, Cytoplasmic and Nuclear/metabolism , Receptors, Peptide/metabolism , SEC Translocation Channels/metabolism , Blotting, Western , Glycine , HeLa Cells , Humans , Hydrophobic and Hydrophilic Interactions , Proline , Proteomics , Real-Time Polymerase Chain Reaction , Substrate Specificity
4.
Nat Commun ; 9(1): 3489, 2018 08 28.
Article in English | MEDLINE | ID: mdl-30154480

ABSTRACT

To fulfill its role in protein biogenesis, the endoplasmic reticulum (ER) depends on the Hsp70-type molecular chaperone BiP, which requires a constant ATP supply. However, the carrier that catalyzes ATP uptake into the ER was unknown. Here, we report that our screen of gene expression datasets for member(s) of the family of solute carriers that are co-expressed with BiP and are ER membrane proteins identifies SLC35B1 as a potential candidate. Heterologous expression of SLC35B1 in E. coli reveals that SLC35B1 is highly specific for ATP and ADP and acts in antiport mode. Moreover, depletion of SLC35B1 from HeLa cells reduces ER ATP levels and, as a consequence, BiP activity. Thus, human SLC35B1 may provide ATP to the ER and was named AXER (ATP/ADP exchanger in the ER membrane). Furthermore, we propose an ER to cytosol low energy response regulatory axis (termed lowER) that appears as central for maintaining ER ATP supply.


Subject(s)
Endoplasmic Reticulum/metabolism , Membrane Proteins/metabolism , Mitochondrial Membranes/metabolism , Adenosine Diphosphate/metabolism , Adenosine Triphosphate/metabolism , Amino Acid Sequence , Biological Transport/physiology , Cytosol/metabolism , Electrophoresis, Polyacrylamide Gel , HeLa Cells , Humans , Membrane Proteins/chemistry , Molecular Sequence Data , Monosaccharide Transport Proteins/chemistry , Monosaccharide Transport Proteins/genetics , Monosaccharide Transport Proteins/metabolism , Real-Time Polymerase Chain Reaction , Sequence Homology, Amino Acid
5.
Semin Cell Dev Biol ; 76: 191-200, 2018 04.
Article in English | MEDLINE | ID: mdl-28923515

ABSTRACT

Mitochondria are complex double-membrane-bound organelles of eukaryotic cells that function as energy-converting powerhouses, metabolic factories and signaling centers. The outer membrane controls the exchange of material and information with other cellular compartments. The inner membrane provides an extended, highly folded surface for selective transport and energy-coupling reactions. It can be divided into an inner boundary membrane and tubular or lamellar cristae membranes that accommodate the oxidative phosphorylation units. Outer membrane, inner boundary membrane and cristae come together at crista junctions, where the mitochondrial contact site and cristae organizing system (MICOS) acts as a membrane-shaping and -connecting scaffold. This peculiar architecture is of pivotal importance for multiple mitochondrial functions. Many elaborate studies in the past years have shed light on the subunit composition and organization of MICOS. In this review article, we summarize these insights and then move on to discuss exciting recent discoveries on the integrative functions of MICOS. Multi-faceted connections to other major players of mitochondrial biogenesis and physiology, like the protein import machineries, the oxidative phosphorylation system, carrier proteins and phospholipid biosynthesis enzymes, are currently emerging. Therefore, we propose that MICOS acts as a central hub in mitochondrial membrane architecture and functionality.


Subject(s)
Mitochondria/genetics , Humans , Mitochondria/metabolism , Signal Transduction
6.
FEBS Lett ; 591(20): 3211-3224, 2017 10.
Article in English | MEDLINE | ID: mdl-28862756

ABSTRACT

Recently, understanding of protein targeting to the endoplasmic reticulum (ER) was expanded by the discovery of multiple pathways that function in parallel to the signal recognition particle (SRP). Guided entry of tail-anchored proteins and SRP independent (SND) are two such targeting pathways described in yeast. So far, no human SND component is functionally characterized. Here, we report hSnd2 as the first constituent of the human SND pathway able to support substrate-specific protein targeting to the ER. Similar to its yeast counterpart, hSnd2 is assumed to function as a membrane-bound receptor preferentially targeting precursors carrying C-terminal transmembrane domains. Our genetic and physical interaction studies show that hSnd2 is part of a complex network of targeting and translocation that is dynamically regulated.


Subject(s)
Endoplasmic Reticulum/metabolism , Membrane Proteins/genetics , Nuclear Proteins/genetics , Protein Subunits/genetics , Receptors, Cytoplasmic and Nuclear/genetics , Receptors, Peptide/genetics , SEC Translocation Channels/genetics , Saccharomyces cerevisiae Proteins/genetics , Amino Acid Sequence , Animals , Gene Expression Regulation , HeLa Cells , Humans , Membrane Proteins/antagonists & inhibitors , Membrane Proteins/metabolism , Nuclear Proteins/metabolism , Peptides/chemical synthesis , Peptides/metabolism , Protein Binding , Protein Isoforms/antagonists & inhibitors , Protein Isoforms/genetics , Protein Isoforms/metabolism , Protein Precursors/genetics , Protein Precursors/metabolism , Protein Sorting Signals/genetics , Protein Subunits/metabolism , Protein Transport , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Rabbits , Receptors, Cytoplasmic and Nuclear/metabolism , Receptors, Peptide/metabolism , SEC Translocation Channels/metabolism , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism , Saccharomyces cerevisiae Proteins/antagonists & inhibitors , Saccharomyces cerevisiae Proteins/metabolism , Signal Recognition Particle , Signal Transduction , Substrate Specificity
8.
Nature ; 540(7631): 134-138, 2016 11 30.
Article in English | MEDLINE | ID: mdl-27905431

ABSTRACT

In eukaryotes, up to one-third of cellular proteins are targeted to the endoplasmic reticulum, where they undergo folding, processing, sorting and trafficking to subsequent endomembrane compartments. Targeting to the endoplasmic reticulum has been shown to occur co-translationally by the signal recognition particle (SRP) pathway or post-translationally by the mammalian transmembrane recognition complex of 40 kDa (TRC40) and homologous yeast guided entry of tail-anchored proteins (GET) pathways. Despite the range of proteins that can be catered for by these two pathways, many proteins are still known to be independent of both SRP and GET, so there seems to be a critical need for an additional dedicated pathway for endoplasmic reticulum relay. We set out to uncover additional targeting proteins using unbiased high-content screening approaches. To this end, we performed a systematic visual screen using the yeast Saccharomyces cerevisiae, and uncovered three uncharacterized proteins whose loss affected targeting. We suggest that these proteins work together and demonstrate that they function in parallel with SRP and GET to target a broad range of substrates to the endoplasmic reticulum. The three proteins, which we name Snd1, Snd2 and Snd3 (for SRP-independent targeting), can synthetically compensate for the loss of both the SRP and GET pathways, and act as a backup targeting system. This explains why it has previously been difficult to demonstrate complete loss of targeting for some substrates. Our discovery thus puts in place an essential piece of the endoplasmic reticulum targeting puzzle, highlighting how the targeting apparatus of the eukaryotic cell is robust, interlinked and flexible.


Subject(s)
Endoplasmic Reticulum/metabolism , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae/cytology , Saccharomyces cerevisiae/metabolism , HEK293 Cells , Humans , Membrane Proteins/metabolism , Phosphate Transport Proteins/metabolism , Protein Domains , Protein Sorting Signals , Protein Transport , Ribosomal Proteins/metabolism , Signal Recognition Particle/metabolism
9.
Nat Cell Biol ; 18(11): 1173-1184, 2016 11.
Article in English | MEDLINE | ID: mdl-27749824

ABSTRACT

The endoplasmic reticulum (ER) is a site of protein biogenesis in eukaryotic cells. Perturbing ER homeostasis activates stress programs collectively called the unfolded protein response (UPR). The UPR enhances production of ER-resident chaperones and enzymes to reduce the burden of misfolded proteins. On resolution of ER stress, ill-defined, selective autophagic programs remove excess ER components. Here we identify Sec62, a constituent of the translocon complex regulating protein import in the mammalian ER, as an ER-resident autophagy receptor. Sec62 intervenes during recovery from ER stress to selectively deliver ER components to the autolysosomal system for clearance in a series of events that we name recovER-phagy. Sec62 contains a conserved LC3-interacting region in the C-terminal cytosolic domain that is required for its function in recovER-phagy, but is dispensable for its function in the protein translocation machinery. Our results identify Sec62 as a critical molecular component in maintenance and recovery of ER homeostasis.


Subject(s)
Endoplasmic Reticulum Stress/physiology , Endoplasmic Reticulum/metabolism , Membrane Transport Proteins/metabolism , Animals , Autophagy , Homeostasis , Humans , Mice , Molecular Chaperones/metabolism , Protein Biosynthesis/physiology , Protein Transport/physiology , Unfolded Protein Response/physiology
10.
J Biol Chem ; 290(30): 18621-35, 2015 Jul 24.
Article in English | MEDLINE | ID: mdl-26085089

ABSTRACT

In mammalian cells, signal peptide-dependent protein transport into the endoplasmic reticulum (ER) is mediated by a dynamic polypeptide-conducting channel, the heterotrimeric Sec61 complex. Previous work has characterized the Sec61 complex as a potential ER Ca(2+) leak channel in HeLa cells and identified ER lumenal molecular chaperone immunoglobulin heavy-chain-binding protein (BiP) as limiting Ca(2+) leakage via the open Sec61 channel by facilitating channel closing. This BiP activity involves binding of BiP to the ER lumenal loop 7 of Sec61α in the vicinity of tyrosine 344. Of note, the Y344H mutation destroys the BiP binding site and causes pancreatic ß-cell apoptosis and diabetes in mice. Here, we systematically depleted HeLa cells of the BiP co-chaperones by siRNA-mediated gene silencing and used live cell Ca(2+) imaging to monitor the effects on ER Ca(2+) leakage. Depletion of either one of the ER lumenal BiP co-chaperones, ERj3 and ERj6, but not the ER membrane-resident co-chaperones (such as Sec63 protein, which assists BiP in Sec61 channel opening) led to increased Ca(2+) leakage via Sec6 complex, thereby phenocopying the effect of BiP depletion. Thus, BiP facilitates Sec61 channel closure (i.e. limits ER Ca(2+) leakage) via the Sec61 channel with the help of ERj3 and ERj6. Interestingly, deletion of ERj6 causes pancreatic ß-cell failure and diabetes in mice and humans. We suggest that co-chaperone-controlled gating of the Sec61 channel by BiP is particularly important for cells, which are highly active in protein secretion, and that breakdown of this regulatory mechanism can cause apoptosis and disease.


Subject(s)
Diabetes Mellitus/genetics , Endoplasmic Reticulum/metabolism , HSP40 Heat-Shock Proteins/metabolism , Heat-Shock Proteins/metabolism , Membrane Proteins/metabolism , Animals , Binding Sites , Calcium/metabolism , Calcium Signaling/genetics , Diabetes Mellitus/metabolism , Diabetes Mellitus/pathology , Endoplasmic Reticulum Chaperone BiP , Gene Silencing , HSP40 Heat-Shock Proteins/genetics , HeLa Cells , Heat-Shock Proteins/genetics , Humans , Insulin-Secreting Cells/metabolism , Insulin-Secreting Cells/pathology , Membrane Proteins/genetics , Mice , Protein Binding , Protein Transport , SEC Translocation Channels
11.
Am J Hum Genet ; 95(6): 689-97, 2014 Dec 04.
Article in English | MEDLINE | ID: mdl-25466870

ABSTRACT

Diabetes mellitus and neurodegeneration are common diseases for which shared genetic factors are still only partly known. Here, we show that loss of the BiP (immunoglobulin heavy-chain binding protein) co-chaperone DNAJC3 leads to diabetes mellitus and widespread neurodegeneration. We investigated three siblings with juvenile-onset diabetes and central and peripheral neurodegeneration, including ataxia, upper-motor-neuron damage, peripheral neuropathy, hearing loss, and cerebral atrophy. Exome sequencing identified a homozygous stop mutation in DNAJC3. Screening of a diabetes database with 226,194 individuals yielded eight phenotypically similar individuals and one family carrying a homozygous DNAJC3 deletion. DNAJC3 was absent in fibroblasts from all affected subjects in both families. To delineate the phenotypic and mutational spectrum and the genetic variability of DNAJC3, we analyzed 8,603 exomes, including 506 from families affected by diabetes, ataxia, upper-motor-neuron damage, peripheral neuropathy, or hearing loss. This analysis revealed only one further loss-of-function allele in DNAJC3 and no further associations in subjects with only a subset of the features of the main phenotype. Our findings demonstrate that loss-of-function DNAJC3 mutations lead to a monogenic, recessive form of diabetes mellitus in humans. Moreover, they present a common denominator for diabetes and widespread neurodegeneration. This complements findings from mice in which knockout of Dnajc3 leads to diabetes and modifies disease in a neurodegenerative model of Marinesco-Sjögren syndrome.


Subject(s)
Diabetes Mellitus, Type 1/genetics , Gene Expression Regulation , HSP40 Heat-Shock Proteins/genetics , Heat-Shock Proteins/genetics , Multiple System Atrophy/genetics , Adolescent , Adult , Ataxia/genetics , Diabetes Mellitus, Type 1/diagnostic imaging , Endoplasmic Reticulum Chaperone BiP , Exome/genetics , Female , Fibroblasts , HSP40 Heat-Shock Proteins/metabolism , Homozygote , Humans , Male , Models, Molecular , Multiple System Atrophy/diagnostic imaging , Mutation , Pedigree , Phenotype , Radiography , Sequence Analysis, DNA , Young Adult
12.
Nat Commun ; 5: 3072, 2014.
Article in English | MEDLINE | ID: mdl-24407213

ABSTRACT

In mammalian cells, proteins are typically translocated across the endoplasmic reticulum (ER) membrane in a co-translational mode by the ER protein translocon, comprising the protein-conducting channel Sec61 and additional complexes involved in nascent chain processing and translocation. As an integral component of the translocon, the oligosaccharyl-transferase complex (OST) catalyses co-translational N-glycosylation, one of the most common protein modifications in eukaryotic cells. Here we use cryoelectron tomography, cryoelectron microscopy single-particle analysis and small interfering RNA-mediated gene silencing to determine the overall structure, oligomeric state and position of OST in the native ER protein translocon of mammalian cells in unprecedented detail. The observed positioning of OST in close proximity to Sec61 provides a basis for understanding how protein translocation into the ER and glycosylation of nascent proteins are structurally coupled. The overall spatial organization of the native translocon, as determined here, serves as a reliable framework for further hypothesis-driven studies.


Subject(s)
Endoplasmic Reticulum/chemistry , Hexosyltransferases/chemistry , Membrane Proteins/chemistry , Transcription Factors/chemistry , Cells, Cultured , Cryoelectron Microscopy , Endoplasmic Reticulum/physiology , Gene Silencing/physiology , Glycosylation , HeLa Cells , Hexosyltransferases/physiology , Humans , Membrane Proteins/physiology , Protein Transport/physiology , RNA, Small Interfering/physiology , SEC Translocation Channels , Transcription Factors/physiology
13.
BMC Cancer ; 13: 574, 2013 Dec 05.
Article in English | MEDLINE | ID: mdl-24304694

ABSTRACT

BACKGROUND: Tumor cells benefit from their ability to avoid apoptosis and invade other tissues. The endoplasmic reticulum (ER) membrane protein Sec62 is a key player in these processes. Sec62 is essential for cell migration and protects tumor cells against thapsigargin-induced ER stress, which are both linked to cytosolic Ca²âº. SEC62 silencing leads to elevated cytosolic Ca²âº and increased ER Ca²âº leakage after thapsigargin treatment. Sec62 protein levels are significantly increased in different tumors, including prostate, lung and thyroid cancer. METHODS: In lung cancer, the influence of Sec62 protein levels on patient survival was analyzed using the Kaplan-Meier method and log-rank test. To elucidate the underlying pathophysiological functions of Sec62, Ca²âº imaging techniques, real-time cell analysis and cell migration assays were performed. The effects of treatment with the calmodulin antagonists, trifluoperazine (TFP) and ophiobolin A, on cellular Ca²âº homeostasis, cell growth and cell migration were compared with the effects of siRNA-mediated Sec62 depletion or the expression of a mutated SEC62 variant in vitro. Using Biacore analysis we examined the Ca²âº-sensitive interaction of Sec62 with the Sec61 complex. RESULTS: Sec62 overproduction significantly correlated with reduced patient survival. Therefore, Sec62 is not only a predictive marker for this type of tumor, but also an interesting therapeutic target. The present study suggests a regulatory function for Sec62 in the major Ca²âº leakage channel in the ER, Sec61, by a direct and Ca²âº-sensitive interaction. A Ca²âº-binding motif in Sec62 is essential for its molecular function. Treatment of cells with calmodulin antagonists mimicked Sec62 depletion by inhibiting cell migration and rendering the cells sensitive to thapsigargin treatment. CONCLUSIONS: Targeting tumors that overproduce Sec62 with calmodulin antagonists in combination with targeted thapsigargin analogues may offer novel personalized therapeutic options.


Subject(s)
Calmodulin/antagonists & inhibitors , Cell Movement/drug effects , Endoplasmic Reticulum Stress/drug effects , Membrane Transport Proteins/genetics , Sesterterpenes/pharmacology , Trifluoperazine/pharmacology , Amino Acid Motifs , Amino Acid Sequence , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Calcium/metabolism , Calcium Signaling , Calmodulin/metabolism , Carcinoma, Non-Small-Cell Lung/metabolism , Carcinoma, Non-Small-Cell Lung/mortality , Cell Proliferation , Gene Expression , HEK293 Cells , HeLa Cells , Homeostasis , Humans , Kaplan-Meier Estimate , Lung Neoplasms/metabolism , Lung Neoplasms/mortality , Membrane Transport Proteins/chemistry , Membrane Transport Proteins/metabolism , Molecular Sequence Data , Phenotype , Prognosis , RNA Interference , RNA, Small Interfering/genetics
14.
Methods Mol Biol ; 1033: 285-99, 2013.
Article in English | MEDLINE | ID: mdl-23996184

ABSTRACT

The development of small-interfering RNA (siRNA)-mediated gene-silencing strategies has made it possible to study the transport of precursors of soluble and membrane proteins into the endoplasmic reticulum (ER) of human cells. In these approaches, a certain target gene is silenced in the cell type of choice, followed by analysis of the effect of this silencing on the biogenesis of a single or set of precursor polypeptide(s) in cell culture or in cell-free assays involving semi-permeabilized cells and in vitro translations systems. These approaches allow for functional analysis of components of the ER-resident protein transport machinery as well as the elucidation of their potential cell-type variations and regulatory mechanisms. The gene-silencing and subsequent plasmid-based complementation carries the additional benefit of facilitating analysis of the consequences of disease-linked mutations in ER transport components.


Subject(s)
Endoplasmic Reticulum/metabolism , Proteins/metabolism , Animals , Biological Transport , Cell Culture Techniques , Cell Line , Gene Silencing , Humans , Mice , Permeability , Protein Transport , Proteins/genetics , RNA Interference
15.
EMBO J ; 31(15): 3282-96, 2012 Aug 01.
Article in English | MEDLINE | ID: mdl-22796945

ABSTRACT

In mammalian cells, signal peptide-dependent protein transport into the endoplasmic reticulum (ER) is mediated by a dynamic protein-conducting channel, the Sec61 complex. Previous work has characterized the Sec61 channel as a potential ER Ca(2+) leak channel and identified calmodulin as limiting Ca(2+) leakage in a Ca(2+)-dependent manner by binding to an IQ motif in the cytosolic aminoterminus of Sec61α. Here, we manipulated the concentration of the ER lumenal chaperone BiP in cells in different ways and used live cell Ca(2+) imaging to monitor the effects of reduced levels of BiP on ER Ca(2+) leakage. Regardless of how the BiP concentration was lowered, the absence of available BiP led to increased Ca(2+) leakage via the Sec61 complex. When we replaced wild-type Sec61α with mutant Sec61αY344H in the same model cell, however, Ca(2+) leakage from the ER increased and was no longer affected by manipulation of the BiP concentration. Thus, BiP limits ER Ca(2+) leakage through the Sec61 complex by binding to the ER lumenal loop 7 of Sec61α in the vicinity of tyrosine 344.


Subject(s)
Calcium/metabolism , Endoplasmic Reticulum/metabolism , Heat-Shock Proteins/physiology , Ion Channel Gating , Membrane Proteins/metabolism , Amino Acid Sequence , Calcium Signaling/drug effects , Calcium Signaling/genetics , Endoplasmic Reticulum/drug effects , Endoplasmic Reticulum/physiology , Endoplasmic Reticulum Chaperone BiP , Gene Silencing/physiology , HeLa Cells , Heat-Shock Proteins/antagonists & inhibitors , Heat-Shock Proteins/genetics , Heat-Shock Proteins/metabolism , Humans , Ion Channel Gating/drug effects , Ion Channel Gating/genetics , Ion Transport/drug effects , Ion Transport/genetics , Membrane Proteins/chemistry , Membrane Proteins/genetics , Models, Molecular , Molecular Sequence Data , Protein Binding/drug effects , Protein Binding/genetics , Protein Binding/physiology , Protein Folding/drug effects , RNA, Small Interfering/pharmacology , SEC Translocation Channels
16.
J Cell Sci ; 125(Pt 8): 1958-69, 2012 Apr 15.
Article in English | MEDLINE | ID: mdl-22375059

ABSTRACT

Co-translational transport of polypeptides into the endoplasmic reticulum (ER) involves the Sec61 channel and additional components such as the ER lumenal Hsp70 BiP and its membrane-resident co-chaperone Sec63p in yeast. We investigated whether silencing the SEC61A1 gene in human cells affects co- and post-translational transport of presecretory proteins into the ER and post-translational membrane integration of tail-anchored proteins. Although silencing the SEC61A1 gene in HeLa cells inhibited co- and post-translational transport of signal-peptide-containing precursor proteins into the ER of semi-permeabilized cells, silencing the SEC61A1 gene did not affect transport of various types of tail-anchored protein. Furthermore, we demonstrated, with a similar knockdown approach, a precursor-specific involvement of mammalian Sec63 in the initial phase of co-translational protein transport into the ER. By contrast, silencing the SEC62 gene inhibited only post-translational transport of a signal-peptide-containing precursor protein.


Subject(s)
DNA Helicases/metabolism , Endoplasmic Reticulum/metabolism , Membrane Proteins/metabolism , Membrane Transport Proteins/metabolism , Peptides/metabolism , Animals , DNA Helicases/genetics , Endoplasmic Reticulum/genetics , Gene Silencing , HeLa Cells , Humans , Membrane Proteins/genetics , Membrane Transport Proteins/genetics , Mice , Molecular Chaperones , NIH 3T3 Cells , Protein Precursors/genetics , Protein Precursors/metabolism , Protein Processing, Post-Translational , Protein Transport , RNA-Binding Proteins , SEC Translocation Channels
SELECTION OF CITATIONS
SEARCH DETAIL
...