Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 177
Filter
1.
FEBS Lett ; 597(10): 1355-1362, 2023 05.
Article in English | MEDLINE | ID: mdl-36520514

ABSTRACT

Human growth hormone (hGH) is a four-helix bundle protein of considerable pharmacological interest. Recombinant hGH is produced in bacteria, yet little is known about its folding during expression in Escherichia coli. We have studied the cotranslational folding of hGH using force profile analysis (FPA), both during in vitro translation in the absence and presence of the chaperone trigger factor (TF), and when expressed in E. coli. We find that the main folding transition starts before hGH is completely released from the ribosome, and that it can interact with TF and possibly other chaperones.


Subject(s)
Escherichia coli Proteins , Human Growth Hormone , Humans , Escherichia coli/genetics , Escherichia coli/metabolism , Protein Folding , Human Growth Hormone/genetics , Human Growth Hormone/metabolism , Protein Biosynthesis , Molecular Chaperones/genetics , Molecular Chaperones/metabolism , Escherichia coli Proteins/genetics , Escherichia coli Proteins/metabolism
2.
Proc Natl Acad Sci U S A ; 119(35): e2205810119, 2022 08 30.
Article in English | MEDLINE | ID: mdl-35994672

ABSTRACT

In recent years, it has become clear that many homo- and heterodimeric cytoplasmic proteins in both prokaryotic and eukaryotic cells start to dimerize cotranslationally (i.e., while at least one of the two chains is still attached to the ribosome). Whether this is also possible for integral membrane proteins is, however, unknown. Here, we apply force profile analysis (FPA)-a method where a translational arrest peptide (AP) engineered into the polypeptide chain is used to detect force generated on the nascent chain during membrane insertion-to demonstrate cotranslational interactions between a fully membrane-inserted monomer and a nascent, ribosome-tethered monomer of the Escherichia coli inner membrane protein EmrE. Similar cotranslational interactions are also seen when the two monomers are fused into a single polypeptide. Further, we uncover an apparent intrachain interaction between E14 in transmembrane helix 1 (TMH1) and S64 in TMH3 that forms at a precise nascent chain length during cotranslational membrane insertion of an EmrE monomer. Like soluble proteins, inner membrane proteins thus appear to be able to both start to fold and start to dimerize during the cotranslational membrane insertion process.


Subject(s)
Antiporters , Escherichia coli Proteins , Escherichia coli , Antiporters/metabolism , Escherichia coli/genetics , Escherichia coli/metabolism , Escherichia coli Proteins/metabolism , Peptides/metabolism , Protein Biosynthesis , Protein Folding
3.
FEBS Lett ; 596(8): 1004-1012, 2022 04.
Article in English | MEDLINE | ID: mdl-35038773

ABSTRACT

During SecYEG-mediated cotranslational insertion of membrane proteins, transmembrane helices (TMHs) first make contact with the membrane when their N-terminal end is ~ 45 residues away from the peptidyl transferase centre. However, we recently uncovered instances where the first contact is delayed by up to ~ 10 residues. Here, we recapitulate these effects using a model TMH fused to two short segments from the Escherichia coli inner membrane protein BtuC: a positively charged loop and a re-entrant loop. We show that the critical residues are two Arg residues in the positively charged loop and four hydrophobic residues in the re-entrant loop. Thus, both electrostatic and hydrophobic interactions involving sequence elements that are not part of a TMH can impact the way the latter behaves during membrane insertion.


Subject(s)
Escherichia coli Proteins , Cell Membrane/metabolism , Escherichia coli/genetics , Escherichia coli/metabolism , Escherichia coli Proteins/metabolism , Hydrophobic and Hydrophilic Interactions , Membrane Proteins/metabolism , Protein Structure, Secondary
4.
Nat Biotechnol ; 40(7): 1023-1025, 2022 07.
Article in English | MEDLINE | ID: mdl-34980915

ABSTRACT

Signal peptides (SPs) are short amino acid sequences that control protein secretion and translocation in all living organisms. SPs can be predicted from sequence data, but existing algorithms are unable to detect all known types of SPs. We introduce SignalP 6.0, a machine learning model that detects all five SP types and is applicable to metagenomic data.


Subject(s)
Language , Protein Sorting Signals , Algorithms , Amino Acid Sequence , Protein Sorting Signals/genetics , Proteins
5.
J Chem Theory Comput ; 18(3): 1905-1914, 2022 Mar 08.
Article in English | MEDLINE | ID: mdl-34881571

ABSTRACT

The ribosome stalling mechanism is a crucial biological process, yet its atomistic underpinning is still elusive. In this framework, the human XBP1u translational arrest peptide (AP) plays a central role in regulating the unfolded protein response (UPR) in eukaryotic cells. Here, we report multimicrosecond all-atom molecular dynamics simulations designed to probe the interactions between the XBP1u AP and the mammalian ribosome exit tunnel, both for the wild type AP and for four mutant variants of different arrest potencies. Enhanced sampling simulations allow investigating the AP release process of the different variants, shedding light on this complex mechanism. The present outcomes are in qualitative/quantitative agreement with available experimental data. In conclusion, we provide an unprecedented atomistic picture of this biological process and clear-cut insights into the key AP-ribosome interactions.


Subject(s)
Peptides , Ribosomes , Animals , Cytosol , Humans , Mammals , Molecular Dynamics Simulation , Peptides/chemistry , Ribosomes/chemistry
6.
Nucleic Acids Res ; 49(16): 9539-9547, 2021 09 20.
Article in English | MEDLINE | ID: mdl-34403461

ABSTRACT

In Escherichia coli, elevated levels of free l-tryptophan (l-Trp) promote translational arrest of the TnaC peptide by inhibiting its termination. However, the mechanism by which translation-termination by the UGA-specific decoding release factor 2 (RF2) is inhibited at the UGA stop codon of stalled TnaC-ribosome-nascent chain complexes has so far been ambiguous. This study presents cryo-EM structures for ribosomes stalled by TnaC in the absence and presence of RF2 at average resolutions of 2.9 and 3.5 Å, respectively. Stalled TnaC assumes a distinct conformation composed of two small α-helices that act together with residues in the peptide exit tunnel (PET) to coordinate a single L-Trp molecule. In addition, while the peptidyl-transferase center (PTC) is locked in a conformation that allows RF2 to adopt its canonical position in the ribosome, it prevents the conserved and catalytically essential GGQ motif of RF2 from adopting its active conformation in the PTC. This explains how translation of the TnaC peptide effectively allows the ribosome to function as a L-Trp-specific small-molecule sensor that regulates the tnaCAB operon.


Subject(s)
Escherichia coli Proteins/ultrastructure , Peptide Termination Factors/ultrastructure , Protein Biosynthesis , Ribosomes/ultrastructure , Codon, Terminator/genetics , Cryoelectron Microscopy , Escherichia coli/genetics , Escherichia coli/ultrastructure , Escherichia coli Proteins/chemistry , Escherichia coli Proteins/genetics , Peptide Termination Factors/chemistry , Peptide Termination Factors/genetics , Protein Conformation , Protein Conformation, alpha-Helical , Ribosomes/genetics , Tryptophan/genetics
8.
Annu Rev Biochem ; 90: 503-505, 2021 06 20.
Article in English | MEDLINE | ID: mdl-34153216

ABSTRACT

This volume of the Annual Review of Biochemistry contains three reviews on membrane channel proteins: the first by Szczot et al., titled The Form and Function of PIEZO2; the second by Ruprecht & Kunji, titled Structural Mechanism of Transport of Mitochondrial Carriers; and the third by McIlwain et al., titled Membrane Exporters of Fluoride Ion. These reviews provide nice illustrations of just how far evolution has been able to play with the basic helix-bundle architecture of integral membrane proteins to produce membrane channels and transporters of widely different functions.


Subject(s)
Ion Channels/chemistry , Ion Channels/metabolism , Adenosine Diphosphate/metabolism , Adenosine Triphosphate/metabolism , Fluorides/metabolism
9.
Commun Biol ; 4(1): 523, 2021 05 05.
Article in English | MEDLINE | ID: mdl-33953328

ABSTRACT

Proteins commonly fold co-translationally at the ribosome, while the nascent chain emerges from the ribosomal exit tunnel. Protein domains that are sufficiently small can even fold while still located inside the tunnel. However, the effect of the tunnel on the folding dynamics of these domains is not well understood. Here, we combine optical tweezers with single-molecule FRET and molecular dynamics simulations to investigate folding of the small zinc-finger domain ADR1a inside and at the vestibule of the ribosomal tunnel. The tunnel is found to accelerate folding and stabilize the folded state, reminiscent of the effects of chaperonins. However, a simple mechanism involving stabilization by confinement does not explain the results. Instead, it appears that electrostatic interactions between the protein and ribosome contribute to the observed folding acceleration and stabilization of ADR1a.


Subject(s)
DNA-Binding Proteins/chemistry , Molecular Dynamics Simulation , Protein Biosynthesis , Protein Folding , Ribosomes/chemistry , Saccharomyces cerevisiae Proteins/chemistry , Saccharomyces cerevisiae/metabolism , Transcription Factors/chemistry , DNA-Binding Proteins/metabolism , Protein Domains , Ribosomes/metabolism , Saccharomyces cerevisiae Proteins/metabolism , Transcription Factors/metabolism
10.
J Mol Biol ; 433(15): 167047, 2021 07 23.
Article in English | MEDLINE | ID: mdl-33989648

ABSTRACT

In Gram-negative bacteria, periplasmic domains in inner membrane proteins are cotranslationally translocated across the inner membrane through the SecYEG translocon. To what degree such domains also start to fold cotranslationally is generally difficult to determine using currently available methods. Here, we apply Force Profile Analysis (FPA) - a method where a translational arrest peptide is used to detect folding-induced forces acting on the nascent polypeptide - to follow the cotranslational translocation and folding of the large periplasmic domain of the E. coli inner membrane protease LepB in vivo. Membrane insertion of LepB's two N-terminal transmembrane helices is initiated when their respective N-terminal ends reach 45-50 residues away from the peptidyl transferase center (PTC) in the ribosome. The main folding transition in the periplasmic domain involves all but the ~15 most C-terminal residues of the protein and happens when the C-terminal end of the folded part is ~70 residues away from the PTC; a smaller putative folding intermediate is also detected. This implies that wildtype LepB folds post-translationally in vivo, and shows that FPA can be used to study both co- and post-translational protein folding in the periplasm.


Subject(s)
Escherichia coli/metabolism , Membrane Proteins/chemistry , Membrane Proteins/metabolism , SEC Translocation Channels/metabolism , Serine Endopeptidases/chemistry , Serine Endopeptidases/metabolism , Escherichia coli/chemistry , Escherichia coli/genetics , Escherichia coli Proteins/chemistry , Escherichia coli Proteins/genetics , Escherichia coli Proteins/metabolism , Membrane Proteins/genetics , Models, Molecular , Mutation , Protein Biosynthesis , Protein Conformation , Protein Folding , Serine Endopeptidases/genetics
11.
Elife ; 102021 02 08.
Article in English | MEDLINE | ID: mdl-33554862

ABSTRACT

We follow the cotranslational biosynthesis of three multispanning Escherichia coli inner membrane proteins in vivo using high-resolution force profile analysis. The force profiles show that the nascent chain is subjected to rapidly varying pulling forces during translation and reveal unexpected complexities in the membrane integration process. We find that an N-terminal cytoplasmic domain can fold in the ribosome exit tunnel before membrane integration starts, that charged residues and membrane-interacting segments such as re-entrant loops and surface helices flanking a transmembrane helix (TMH) can advance or delay membrane integration, and that point mutations in an upstream TMH can affect the pulling forces generated by downstream TMHs in a highly position-dependent manner, suggestive of residue-specific interactions between TMHs during the integration process. Our results support the 'sliding' model of translocon-mediated membrane protein integration, in which hydrophobic segments are continually exposed to the lipid bilayer during their passage through the SecYEG translocon.


Subject(s)
Escherichia coli Proteins/genetics , Escherichia coli/genetics , Protein Biosynthesis , Amino Acid Motifs , Cell Membrane/chemistry , Cell Membrane/genetics , Cell Membrane/metabolism , Escherichia coli/chemistry , Escherichia coli/metabolism , Escherichia coli Proteins/chemistry , Escherichia coli Proteins/metabolism , Protein Domains
12.
Protein Sci ; 29(10): 2028-2037, 2020 10.
Article in English | MEDLINE | ID: mdl-32790204

ABSTRACT

Cotranslational protein folding studies using Force Profile Analysis, a method where the SecM translational arrest peptide is used to detect folding-induced forces acting on the nascent polypeptide, have so far been limited mainly to small domains of cytosolic proteins that fold in close proximity to the translating ribosome. In this study, we investigate the cotranslational folding of the periplasmic, disulfide bond-containing Escherichia coli protein alkaline phosphatase (PhoA) in a wild-type strain background and a strain background devoid of the periplasmic thiol: disulfide interchange protein DsbA. We find that folding-induced forces can be transmitted via the nascent chain from the periplasm to the polypeptide transferase center in the ribosome, a distance of ~160 Å, and that PhoA appears to fold cotranslationally via at least two disulfide-stabilized folding intermediates. Thus, Force Profile Analysis can be used to study cotranslational folding of proteins in an extra-cytosolic compartment, like the periplasm.


Subject(s)
Alkaline Phosphatase/biosynthesis , Escherichia coli Proteins/biosynthesis , Escherichia coli/enzymology , Periplasm/enzymology , Protein Biosynthesis , Protein Folding , Alkaline Phosphatase/genetics , Escherichia coli/genetics , Escherichia coli Proteins/genetics , Periplasm/genetics
13.
Proc Natl Acad Sci U S A ; 117(25): 14119-14126, 2020 06 23.
Article in English | MEDLINE | ID: mdl-32513720

ABSTRACT

Proteins synthesized in the cell can begin to fold during translation before the entire polypeptide has been produced, which may be particularly relevant to the folding of multidomain proteins. Here, we study the cotranslational folding of adjacent domains from the cytoskeletal protein α-spectrin using force profile analysis (FPA). Specifically, we investigate how the cotranslational folding behavior of the R15 and R16 domains are affected by their neighboring R14 and R16, and R15 and R17 domains, respectively. Our results show that the domains impact each other's folding in distinct ways that may be important for the efficient assembly of α-spectrin, and may reduce its dependence on chaperones. Furthermore, we directly relate the experimentally observed yield of full-length protein in the FPA assay to the force exerted by the folding protein in piconewtons. By combining pulse-chase experiments to measure the rate at which the arrested protein is converted into full-length protein with a Bell model of force-induced rupture, we estimate that the R16 domain exerts a maximal force on the nascent chain of ∼15 pN during cotranslational folding.


Subject(s)
Protein Folding , Spectrin/chemistry , Escherichia coli , Molecular Dynamics Simulation , Protein Biosynthesis , Protein Domains , Spectrin/genetics , Spectrin/metabolism
14.
FEBS J ; 287(13): 2744-2762, 2020 07.
Article in English | MEDLINE | ID: mdl-31821735

ABSTRACT

The malarial parasite Plasmodium exports its own proteins to the cell surfaces of red blood cells (RBCs) during infection. Examples of exported proteins include members of the repetitive interspersed family (RIFIN) and subtelomeric variable open reading frame (STEVOR) family of proteins from Plasmodium falciparum. The presence of these parasite-derived proteins on surfaces of infected RBCs triggers the adhesion of infected cells to uninfected cells (rosetting) and to the vascular endothelium potentially obstructing blood flow. While there is a fair amount of information on the localization of these proteins on the cell surfaces of RBCs, less is known about how they can be exported to the membrane and the topologies they can adopt during the process. The first step of export is plausibly the cotranslational insertion of proteins into the endoplasmic reticulum (ER) of the parasite, and here, we investigate the insertion of three RIFIN and two STEVOR proteins into the ER membrane. We employ a well-established experimental system that uses N-linked glycosylation of sites within the protein as a measure to assess the extent of membrane insertion and the topology it assumes when inserted into the ER membrane. Our results indicate that for all the proteins tested, transmembranes (TMs) 1 and 3 integrate into the membrane, so that the protein assumes an overall topology of Ncyt-Ccyt. We also show that the segment predicted to be TM2 for each of the proteins likely does not reside in the membrane, but is translocated to the lumen.


Subject(s)
Antigens, Protozoan/chemistry , Antigens, Protozoan/metabolism , Cell Membrane/metabolism , Membrane Proteins/chemistry , Membrane Proteins/metabolism , Plasmodium falciparum/physiology , Protozoan Proteins/chemistry , Protozoan Proteins/metabolism , Cell Membrane/chemistry , Endoplasmic Reticulum , HEK293 Cells , Humans , Protein Conformation
15.
FEBS Lett ; 594(6): 1081-1087, 2020 03.
Article in English | MEDLINE | ID: mdl-31764998

ABSTRACT

The TIM23 complex mediates membrane insertion of presequence-containing mitochondrial proteins via a stop-transfer mechanism. Stop-transfer signals consist of hydrophobic transmembrane segments and flanking charges. Mgr2 functions as a lateral gatekeeper of the TIM23 complex. However, it remains elusive which features of stop-transfer signals are discriminated by Mgr2. To determine the effects of Mgr2 on the TIM23-mediated stop-transfer pathway, we measured membrane insertion of model transmembrane segments of varied hydrophobicity and flanking charges in Mgr2-deletion or -overexpression yeast strains. We found that upon deletion of Mgr2, the threshold hydrophobicity for membrane insertion, as well as the requirement for matrix-facing positive charges, is reduced. These results imply that the Mgr2-mediated gatekeeper function is important for controlling membrane sorting of marginal stop-transfer signals.


Subject(s)
Membrane Proteins/metabolism , Membrane Transport Proteins/metabolism , Mitochondria/metabolism , Mitochondrial Membranes/metabolism , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae/metabolism , Membrane Proteins/genetics , Membrane Transport Proteins/genetics , Mitochondria/genetics , Mitochondrial Precursor Protein Import Complex Proteins , Protein Transport , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae Proteins/genetics
16.
Nature ; 574(7780): 634, 2019 10.
Article in English | MEDLINE | ID: mdl-31664210
17.
Life Sci Alliance ; 2(5)2019 10.
Article in English | MEDLINE | ID: mdl-31570514

ABSTRACT

In bioinformatics, machine learning methods have been used to predict features embedded in the sequences. In contrast to what is generally assumed, machine learning approaches can also provide new insights into the underlying biology. Here, we demonstrate this by presenting TargetP 2.0, a novel state-of-the-art method to identify N-terminal sorting signals, which direct proteins to the secretory pathway, mitochondria, and chloroplasts or other plastids. By examining the strongest signals from the attention layer in the network, we find that the second residue in the protein, that is, the one following the initial methionine, has a strong influence on the classification. We observe that two-thirds of chloroplast and thylakoid transit peptides have an alanine in position 2, compared with 20% in other plant proteins. We also note that in fungi and single-celled eukaryotes, less than 30% of the targeting peptides have an amino acid that allows the removal of the N-terminal methionine compared with 60% for the proteins without targeting peptide. The importance of this feature for predictions has not been highlighted before.


Subject(s)
Computational Biology/methods , Peptides/analysis , Peptides/genetics , Amino Acid Sequence , Chloroplasts/genetics , Chloroplasts/metabolism , Deep Learning , Fungi/genetics , Fungi/metabolism , Methionine/metabolism , Protein Sorting Signals , Thylakoids/genetics , Thylakoids/metabolism
18.
Nat Chem Biol ; 15(10): 945-948, 2019 10.
Article in English | MEDLINE | ID: mdl-31501590

ABSTRACT

Helical membrane proteins are typically assumed to attain stable transmembrane topologies immediately upon co-translational membrane insertion. Here we show that unassembled monomers of the small multidrug resistance (SMR) family exist in a dynamic equilibrium where the N-terminal transmembrane helix flips in and out of the membrane, with rates that depend on dimerization and the polypeptide sequence. Thus, membrane topology can display rapid dynamics in vivo and can be regulated by post-translational assembly.


Subject(s)
Antiporters/metabolism , Escherichia coli Proteins/chemistry , Escherichia coli/physiology , Membrane Proteins/chemistry , Amino Acid Sequence , Animals , Anti-Bacterial Agents/pharmacology , Antiporters/genetics , Drug Resistance, Multiple, Bacterial , Escherichia coli/drug effects , Escherichia coli Proteins/genetics , Escherichia coli Proteins/metabolism , Gene Expression Regulation, Bacterial , Genetic Variation , Membrane Proteins/metabolism , Plasmids , Protein Conformation
19.
Elife ; 82019 06 27.
Article in English | MEDLINE | ID: mdl-31246176

ABSTRACT

XBP1u, a central component of the unfolded protein response (UPR), is a mammalian protein containing a functionally critical translational arrest peptide (AP). Here, we present a 3 Å cryo-EM structure of the stalled human XBP1u AP. It forms a unique turn in the ribosomal exit tunnel proximal to the peptidyl transferase center where it causes a subtle distortion, thereby explaining the temporary translational arrest induced by XBP1u. During ribosomal pausing the hydrophobic region 2 (HR2) of XBP1u is recognized by SRP, but fails to efficiently gate the Sec61 translocon. An exhaustive mutagenesis scan of the XBP1u AP revealed that only 8 out of 20 mutagenized positions are optimal; in the remaining 12 positions, we identify 55 different mutations increase the level of translational arrest. Thus, the wildtype XBP1u AP induces only an intermediate level of translational arrest, allowing efficient targeting by SRP without activating the Sec61 channel.


Subject(s)
Ribosomes/metabolism , X-Box Binding Protein 1/chemistry , X-Box Binding Protein 1/genetics , Amino Acid Sequence , Animals , Biomechanical Phenomena , DNA Mutational Analysis , Endoribonucleases/metabolism , Humans , Models, Molecular , Mutagenesis , Peptides/chemistry , Peptidyl Transferases/metabolism , Protein Binding , Protein Serine-Threonine Kinases/metabolism , Protein Stability , Rabbits , Ribosomes/ultrastructure , SEC Translocation Channels/chemistry , SEC Translocation Channels/metabolism , Signal Recognition Particle/metabolism , Signal Transduction , Unfolded Protein Response , X-Box Binding Protein 1/ultrastructure
20.
J Mol Biol ; 431(14): 2567-2580, 2019 06 28.
Article in English | MEDLINE | ID: mdl-31100385

ABSTRACT

Signal recognition particle (SRP) recognizes signal sequences of secretory proteins and targets them to the endoplasmic reticulum membrane for translocation. Many human diseases are connected with defects in signal sequences. The current dogma states that the molecular basis of the disease-associated mutations in the secretory proteins is connected with defects in their transport. Here, we demonstrate for several secretory proteins with disease-associated mutations that the molecular mechanism is different from the dogma. Positively charged or helix-breaking mutations in the signal sequence hydrophobic core prevent synthesis of the aberrant proteins and lead to degradation of their mRNAs. The degree of mRNA depletion depends on the location and severity of the mutation in the signal sequence and correlates with inhibition of SRP interaction. Thus, SRP protects secretory protein mRNAs from degradation. The data demonstrate that if disease-associated mutations obstruct SRP interaction, they lead to silencing of the mutated protein expression.


Subject(s)
Disease/genetics , Mutation , Proteins/antagonists & inhibitors , Proteins/metabolism , RNA, Messenger/metabolism , Signal Recognition Particle/antagonists & inhibitors , Signal Recognition Particle/metabolism , HeLa Cells , Humans , Mutant Proteins/antagonists & inhibitors , Mutant Proteins/genetics , Mutant Proteins/metabolism , Proteins/genetics , RNA, Messenger/genetics , Signal Recognition Particle/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...