Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
Add more filters










Publication year range
1.
Curr Opin Chem Biol ; 66: 102102, 2022 02.
Article in English | MEDLINE | ID: mdl-34861482

ABSTRACT

Fluorescence microscopy revolutionized cell biology and changed requirements for dyes towards higher brightness, novel capacities, and specific targets. With the need for multiplexing assays in high-throughput methodologies, surface staining gained particular interest because it allows rapid application of exogenous stains to track cellular identity in mixed populations. Indeed, the last decade has enriched the toolbox of general lipid stains, fluorescent lipid analogues, sugar-binding lectins, and protein-specific antibodies enabling the first rationally designed plasma membrane-specific dyes. Still, multiple challenges exist, and the unique properties of each dye must be considered when selecting a staining approach for a specific application. Recent advances are also promising that future dyes will provide ultimate brightness and photostability in diverse colors and reduced sizes for high-resolution imaging.


Subject(s)
Fluorescent Dyes , Mammals , Animals , Cell Membrane/metabolism , Fluorescent Dyes/chemistry , Microscopy, Fluorescence/methods , Staining and Labeling
2.
Int J Mol Sci ; 23(1)2021 Dec 23.
Article in English | MEDLINE | ID: mdl-35008565

ABSTRACT

Looking at the variety of the thousands of different polypeptides that have been focused on in the research on the endoplasmic reticulum from the last five decades taught us one humble lesson: no one size fits all. Cells use an impressive array of components to enable the safe transport of protein cargo from the cytosolic ribosomes to the endoplasmic reticulum. Safety during the transit is warranted by the interplay of cytosolic chaperones, membrane receptors, and protein translocases that together form functional networks and serve as protein targeting and translocation routes. While two targeting routes to the endoplasmic reticulum, SRP (signal recognition particle) and GET (guided entry of tail-anchored proteins), prefer targeting determinants at the N- and C-terminus of the cargo polypeptide, respectively, the recently discovered SND (SRP-independent) route seems to preferentially cater for cargos with non-generic targeting signals that are less hydrophobic or more distant from the termini. With an emphasis on targeting routes and protein translocases, we will discuss those functional networks that drive efficient protein topogenesis and shed light on their redundant and dynamic nature in health and disease.


Subject(s)
Endoplasmic Reticulum/metabolism , Protein Transport/physiology , Proteins/metabolism , Animals , Cytosol/metabolism , Humans , Protein Sorting Signals/physiology , Signal Recognition Particle/metabolism
3.
J Allergy Clin Immunol ; 146(5): 1180-1193, 2020 11.
Article in English | MEDLINE | ID: mdl-32325141

ABSTRACT

BACKGROUND: The molecular cause of severe congenital neutropenia (SCN) is unknown in 30% to 50% of patients. SEC61A1 encodes the α-subunit of the Sec61 complex, which governs endoplasmic reticulum protein transport and passive calcium leakage. Recently, mutations in SEC61A1 were reported to be pathogenic in common variable immunodeficiency and glomerulocystic kidney disease. OBJECTIVE: Our aim was to expand the spectrum of SEC61A1-mediated disease to include autosomal dominant SCN. METHODS: Whole exome sequencing findings were validated, and reported mutations were compared by Western blotting, Ca2+ flux assays, differentiation of transduced HL-60 cells, in vitro differentiation of primary CD34 cells, quantitative PCR for unfolded protein response (UPR) genes, and single-cell RNA sequencing on whole bone marrow. RESULTS: We identified a novel de novo missense mutation in SEC61A1 (c.A275G;p.Q92R) in a patient with SCN who was born to nonconsanguineous Belgian parents. The mutation results in diminished protein expression, disturbed protein translocation, and an increase in calcium leakage from the endoplasmic reticulum. In vitro differentiation of CD34+ cells recapitulated the patient's clinical arrest in granulopoiesis. The impact of Q92R-Sec61α1 on neutrophil maturation was validated by using HL-60 cells, in which transduction reduced differentiation into CD11b+CD16+ cells. A potential mechanism for this defect is the uncontrolled initiation of the unfolded protein stress response, with single-cell analysis of primary bone marrow revealing perturbed UPR in myeloid precursors and in vitro differentiation of primary CD34+ cells revealing upregulation of CCAAT/enhancer-binding protein homologous protein and immunoglobulin heavy chain binding protein UPR-response genes. CONCLUSION: Specific mutations in SEC61A1 cause SCN through dysregulation of the UPR.


Subject(s)
Congenital Bone Marrow Failure Syndromes/genetics , Mutation/genetics , Neutropenia/congenital , Neutrophils/physiology , SEC Translocation Channels/genetics , Antigens, CD34/metabolism , Chromosome Disorders , Female , Genes, Dominant , HL-60 Cells , Humans , Neutropenia/genetics , Pedigree , Single-Cell Analysis , Unfolded Protein Response/genetics , Exome Sequencing , Young Adult
4.
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
5.
FEBS Lett ; 593(18): 2506-2524, 2019 09.
Article in English | MEDLINE | ID: mdl-31325177

ABSTRACT

Protein transport into the mammalian endoplasmic reticulum (ER) used to be seen as strictly cotranslational, that is temporarily and mechanistically coupled to protein synthesis. In the course of the last decades, however, several classes of precursors of soluble and membrane proteins were found to be post-translationally imported into the ER, without any involvement of the ribosome. The first such class to be identified were the small presecretory proteins; tail-anchored membrane proteins followed next. In both classes, the inherent address tag is released from the translating ribosome before the initiation of ER import, as part of the fully synthesized precursor. In small presecretory proteins, the information for ER targeting and -translocation via the polypeptide-conducting Sec61-channel is encoded by a classical N-terminal signal peptide, which is released from the ribsosome before targeting due to the small size of the full-length precursor. Here, we discuss the current state of research on targeting and translocation of small presecretory proteins into the mammalian ER. In closing, we present a unifying hypothesis for ER protein translocation in terms of an energy diagram for Sec61-channel gating.


Subject(s)
Endoplasmic Reticulum/metabolism , Membrane Proteins/metabolism , Humans , Membrane Proteins/chemistry , Protein Transport
6.
Biol Open ; 8(3)2019 Mar 05.
Article in English | MEDLINE | ID: mdl-30745438

ABSTRACT

The Sec61-complex as a dynamic polypeptide-conducting channel mediates protein transport into the human endoplasmic reticulum (ER) with the help of additional components. ER membrane resident Hsp40-type co-chaperone Sec63 as well as the ER lumenal Hsp70-type chaperone BiP were proposed to facilitate channel opening in a precursor-specific fashion. Here, we report on their rules of engagement in ER import of the prion protein (PrP) by addressing sixteen PrP-related variants which differ in their signal peptides and mature parts, respectively. Transport into the ER of semi-permeabilized human cells was analyzed upon depletion of the components by siRNA- or toxin-treatment. The results are consistent with the view of separate functions of BiP and Sec63 and strongly suggest that the co-chaperone/chaperone-pair facilitates Sec61 channel gating to the open state when precursor polypeptides with weak signal peptides in combination with detrimental features in the adjacent mature part were targeted. Thus, we expand the view of chaperone-mediated Sec61 channel gating by providing a novel example of a polybasic motif that interferes with signal peptide-mediated Sec61 channel gating. This article has an associated First Person interview with the first author of the paper.

7.
Cell Rep ; 23(5): 1373-1386, 2018 05 01.
Article in English | MEDLINE | ID: mdl-29719251

ABSTRACT

Protein transport into the mammalian endoplasmic reticulum (ER) is mediated by the heterotrimeric Sec61 channel. The signal recognition particle (SRP) and TRC systems and Sec62 have all been characterized as membrane-targeting components for small presecretory proteins, whereas Sec63 and the lumenal chaperone BiP act as auxiliary translocation components. Here, we report the transport requirements of two natural, small presecretory proteins and engineered variants using semipermeabilized human cells after the depletion of specific ER components. Our results suggest that hSnd2, Sec62, and SRP and TRC receptor each provide alternative targeting pathways for short secretory proteins and define rules of engagement for the actions of Sec63 and BiP during their membrane translocation. We find that the Sec62/Sec63 complex plus BiP can facilitate Sec61 channel opening, thereby allowing precursors that have weak signal peptides or other inhibitory features to translocate. A Sec61 inhibitor can mimic the effect of BiP depletion on Sec61 gating, suggesting that they both act at the same essential membrane translocation step.


Subject(s)
Endoplasmic Reticulum/metabolism , Heat-Shock Proteins/metabolism , Intracellular Membranes/metabolism , SEC Translocation Channels/metabolism , Signal Recognition Particle/metabolism , Animals , Endoplasmic Reticulum/genetics , Endoplasmic Reticulum Chaperone BiP , HeLa Cells , Heat-Shock Proteins/genetics , Humans , Mice , Protein Sorting Signals/physiology , Protein Transport/physiology , SEC Translocation Channels/genetics , Signal Recognition Particle/genetics
8.
J Allergy Clin Immunol ; 141(4): 1427-1438, 2018 04.
Article in English | MEDLINE | ID: mdl-28782633

ABSTRACT

BACKGROUND: Primary antibody deficiencies (PADs) are the most frequent primary immunodeficiencies in human subjects. The genetic causes of PADs are largely unknown. Sec61 translocon alpha 1 subunit (SEC61A1) is the major subunit of the Sec61 complex, which is the main polypeptide-conducting channel in the endoplasmic reticulum membrane. SEC61A1 is a target gene of spliced X-box binding protein 1 and strongly induced during plasma cell (PC) differentiation. OBJECTIVE: We identified a novel genetic defect and studied its pathologic mechanism in 11 patients from 2 unrelated families with PADs. METHODS: Whole-exome and targeted sequencing were conducted to identify novel genetic mutations. Functional studies were carried out ex vivo in primary cells of patients and in vitro in different cell lines to assess the effect of SEC61A1 mutations on B-cell differentiation and survival. RESULTS: We investigated 2 families with patients with hypogammaglobulinemia, severe recurrent respiratory tract infections, and normal peripheral B- and T-cell subpopulations. On in vitro stimulation, B cells showed an intrinsic deficiency to develop into PCs. Genetic analysis and targeted sequencing identified novel heterozygous missense (c.254T>A, p.V85D) and nonsense (c.1325G>T, p.E381*) mutations in SEC61A1, segregating with the disease phenotype. SEC61A1-V85D was deficient in cotranslational protein translocation, and it disturbed the cellular calcium homeostasis in HeLa cells. Moreover, SEC61A1-V85D triggered the terminal unfolded protein response in multiple myeloma cell lines. CONCLUSION: We describe a monogenic defect leading to a specific PC deficiency in human subjects, expanding our knowledge about the pathogenesis of antibody deficiencies.


Subject(s)
Immunologic Deficiency Syndromes/genetics , Mutation/genetics , Plasma Cells/pathology , SEC Translocation Channels/genetics , Agammaglobulinemia/genetics , Agammaglobulinemia/metabolism , Agammaglobulinemia/pathology , B-Lymphocytes/metabolism , B-Lymphocytes/pathology , Calcium/metabolism , Cell Differentiation/genetics , Cell Line , Cell Line, Tumor , Exome/genetics , HEK293 Cells , HeLa Cells , Heterozygote , Humans , Immunologic Deficiency Syndromes/metabolism , Plasma Cells/metabolism , Protein Transport/genetics , Respiratory Tract Infections/genetics , Respiratory Tract Infections/metabolism , Respiratory Tract Infections/pathology , T-Lymphocytes/metabolism , T-Lymphocytes/pathology , Unfolded Protein Response/genetics
9.
J Cell Sci ; 130(22): 3851-3861, 2017 Nov 15.
Article in English | MEDLINE | ID: mdl-29021347

ABSTRACT

Tail-anchored (TA) proteins are transmembrane proteins with a single C-terminal transmembrane domain, which functions as both their subcellular targeting signal and membrane anchor. We show that knockout of TRC40 in cultured human cells has a relatively minor effect on endogenous TA proteins, despite their apparent reliance on this pathway in vitro These findings support recent evidence that the canonical TRC40 pathway is not essential for TA protein biogenesis in vivo We therefore investigated the possibility that other ER-targeting routes can complement the TRC40 pathway and identified roles for both the SRP pathway and the recently described mammalian SND pathway in TA protein biogenesis. We conclude that, although TRC40 normally plays an important role in TA protein biogenesis, it is not essential, and speculate that alternative pathways for TA protein biogenesis, including those identified in this study, contribute to the redundancy of the TRC40 pathway.


Subject(s)
Arsenite Transporting ATPases/genetics , Protein Biosynthesis , Arsenite Transporting ATPases/metabolism , Biosynthetic Pathways , HeLa Cells , Humans , Membrane Proteins/biosynthesis , Membrane Proteins/genetics , Protein Transport
10.
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
11.
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
12.
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
13.
Biochem Cell Biol ; 92(6): 499-509, 2014 Dec.
Article in English | MEDLINE | ID: mdl-24934166

ABSTRACT

Protein transport into the human endoplasmic reticulum (ER) is relevant to the biogenesis of most soluble and membrane proteins of organelles, which are involved in endo- or exo-cytsosis. It involves amino-terminal signal peptides in the precursor polypeptides and various transport components in the cytosol plus the ER, and can occur co- or post-translationally. The two mechanisms merge at the level of the ER membrane, specifically at the level of the heterotrimeric Sec61 complex, which forms a dynamic polypeptide-conducting channel in the ER membrane. Since the mammalian ER is also the main intracellular calcium storage organelle, and the Sec61 complex is calcium permeable, the Sec61 complex is tightly regulated in its equilibrium between the closed and open conformations, or "gated", by ligands, such as signal peptides of the transport substrates and the ER lumenal Hsp70-type molecular chaperone BiP. Furthermore, BiP binding to the incoming polypeptide contributes to the efficiency and unidirectionality of transport. Recent insights into the structure and dynamic equilibrium of the Sec61 complex have various mechanistic as well as medical implications.


Subject(s)
Channelopathies/metabolism , Endoplasmic Reticulum/metabolism , Intracellular Membranes/metabolism , Membrane Proteins/metabolism , Multiprotein Complexes/metabolism , Channelopathies/genetics , Channelopathies/pathology , Endoplasmic Reticulum/genetics , Endoplasmic Reticulum/pathology , HSP70 Heat-Shock Proteins/genetics , HSP70 Heat-Shock Proteins/metabolism , Humans , Intracellular Membranes/pathology , Membrane Proteins/genetics , Multiprotein Complexes/genetics , Protein Sorting Signals , Protein Transport/genetics , SEC Translocation Channels
14.
PLoS One ; 8(10): e75394, 2013.
Article in English | MEDLINE | ID: mdl-24130708

ABSTRACT

The metazoan Sec61 translocon transports polypeptides into and across the membrane of the endoplasmic reticulum via two major routes, a well-established co-translational pathway and a post-translational alternative. We have used two model substrates to explore the elements of a secretory protein precursor that preferentially direct it towards a co- or post-translational pathway for ER translocation. Having first determined the capacity of precursors to enter ER derived microsomes post-translationally, we then exploited semi-permeabilized mammalian cells specifically depleted of key membrane components using siRNA to address their contribution to the membrane translocation process. These studies suggest precursor chain length is a key factor in the post-translational translocation at the mammalian ER, and identify Sec62 and Sec63 as important components acting on this route. This role for Sec62 and Sec63 is independent of the signal sequence that delivers the precursor to the ER. However, the signal sequence can influence the subsequent membrane translocation process, conferring sensitivity to a small molecule inhibitor and dictating reliance on the molecular chaperone BiP. Our data support a model where secretory protein precursors that fail to engage the signal recognition particle, for example because they are short, are delivered to the ER membrane via a distinct route that is dependent upon both Sec62 and Sec63. Although this requirement for Sec62 and Sec63 is unaffected by the specific signal sequence that delivers a precursor to the ER, this region can influence subsequent events, including both Sec61 mediated transport and the importance of BiP for membrane translocation. Taken together, our data suggest that an ER signal sequence can regulate specific aspects of Sec61 mediated membrane translocation at a stage following Sec62/Sec63 dependent ER delivery.


Subject(s)
Endoplasmic Reticulum/metabolism , Intracellular Membranes/metabolism , Protein Processing, Post-Translational/genetics , Protein Processing, Post-Translational/physiology , Protein Transport/physiology , RNA, Small Interfering
15.
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
16.
FEBS Lett ; 585(21): 3485-90, 2011 Nov 04.
Article in English | MEDLINE | ID: mdl-22001204

ABSTRACT

Cytosolic components and pathways have been identified that are involved in inserting tail-anchored (TA) membrane proteins into the yeast or mammalian endoplasmic reticulum (ER) membrane. Searching for regulatory mechanisms of TA protein biogenesis, we found that Ca(2+)-calmodulin (CaM) inhibits the insertion of TA proteins into mammalian ER membranes and that this inhibition is prevented by trifluoperazine, a CaM antagonist that interferes with substrate binding of Ca(2+)-CaM. The effects of Ca(2+)-CaM on cytochrome b(5) and Synaptobrevin 2 suggest a direct interaction between Ca(2+)-CaM and TA proteins. Thus, CaM appears to regulate TA insertion into the ER membrane in a Ca(2+) dependent manner.


Subject(s)
Calcium/pharmacology , Calmodulin/pharmacology , Endoplasmic Reticulum/drug effects , Endoplasmic Reticulum/metabolism , Intracellular Membranes/drug effects , Intracellular Membranes/metabolism , Membrane Proteins/metabolism , Animals , Cattle , Cytosol/drug effects , Cytosol/metabolism , Male , Membrane Proteins/chemistry , Microsomes/drug effects , Microsomes/metabolism , Protein Transport/drug effects , Rabbits
17.
J Gen Virol ; 92(Pt 12): 2734-2745, 2011 Dec.
Article in English | MEDLINE | ID: mdl-21832006

ABSTRACT

Release of herpes simplex virus type 1 (HSV-1) nucleocapsids from the host nucleus relies on the nuclear egress complex consisting of the two essential proteins pUL34 and pUL31. The cytoplasmically exposed N-terminal region of pUL34 interacts with pUL31, while a hydrophobic region followed by a short luminal part mediates membrane association. Based on its domain organization, pUL34 was postulated to be a tail-anchor (TA) protein. We performed a coupled in vitro transcription/translation assay to show that membrane insertion of pUL34 occurs post-translationally. Transient transfection and localization experiments in mammalian cells were combined with HSV-1 bacterial artificial chromosome mutagenesis to reveal the functional properties of the essential pUL34 TA. Our data show that a minimal tail length of 15 residues is sufficient for nuclear envelope targeting and pUL34 function. Permutations of the pUL34 TA with orthologous regions of human cytomegalovirus pUL50 or Epstein-Barr virus pBFRF1 as well as the heterologous HSV-1 TA proteins pUL56 or pUS9 or the cellular TA proteins Bcl-2 and Vamp2 revealed that nuclear egress tolerates TAs varying in sequence and hydrophobicity, while a non-α-helical membrane anchor failed to complement the pUL34 function. In conclusion, this study provides the first mechanistic insights into the particular role of the TA of pUL34 in membrane curving and capsid egress from the host nucleus.


Subject(s)
Herpesvirus 1, Human/genetics , Nuclear Envelope/chemistry , Nuclear Envelope/virology , Nuclear Proteins/metabolism , Viral Proteins/metabolism , Animals , Fluorescent Antibody Technique, Indirect/methods , Gene Deletion , Herpesvirus 1, Human/metabolism , Humans , Mutation , Nuclear Envelope/metabolism , Nuclear Proteins/genetics , Plasmids , Vero Cells , Viral Proteins/genetics , Virus Replication
SELECTION OF CITATIONS
SEARCH DETAIL
...