Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 77
Filter
Add more filters










Publication year range
1.
Methods Mol Biol ; 1877: 131-149, 2019.
Article in English | MEDLINE | ID: mdl-30536003

ABSTRACT

The Bcl-2 family of proteins regulates mitochondrial outer membrane permeability thereby making life or death decisions for cells. Most of Bcl-2 proteins contain hydrophobic regions that are embedded in intracellular membranes such as mitochondria. These membrane proteins are difficult to express and purify thereby preluding biochemical and biophysical characterizations. Here, we describe a photocrosslinking approach based on in vitro synthesis of Bcl-2 proteins with photoreactive amino acid analogs incorporated at specific locations. These photoreactive proteins are reconstituted into liposomal membranes with defined phospholipids or mitochondrial membranes isolated from animals, and their interactions with other Bcl-2 proteins are detected by photocrosslinking.


Subject(s)
Protein Interaction Domains and Motifs/physiology , Proto-Oncogene Proteins c-bcl-2/metabolism , Amino Acids/metabolism , Animals , Humans , Liposomes/metabolism , Membrane Proteins/metabolism , Mice , Mitochondria/metabolism , Mitochondrial Membranes/metabolism , Permeability , Phospholipids/metabolism
2.
Traffic ; 17(2): 117-30, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26572236

ABSTRACT

The long-standing paradigm that all peroxisomal proteins are imported post-translationally into pre-existing peroxisomes has been challenged by the detection of peroxisomal membrane proteins (PMPs) inside the endoplasmic reticulum (ER). In mammals, the mechanisms of ER entry and exit of PMPs are completely unknown. We show that the human PMP PEX3 inserts co-translationally into the mammalian ER via the Sec61 translocon. Photocrosslinking and fluorescence spectroscopy studies demonstrate that the N-terminal transmembrane segment (TMS) of ribosome-bound PEX3 is recognized by the signal recognition particle (SRP). Binding to SRP is a prerequisite for targeting of the PEX3-containing ribosome•nascent chain complex (RNC) to the translocon, where an ordered multistep pathway integrates the nascent chain into the membrane adjacent to translocon proteins Sec61α and TRAM. This insertion of PEX3 into the ER is physiologically relevant because PEX3 then exits the ER via budding vesicles in an ATP-dependent process. This study identifies early steps in human peroxisomal biogenesis by demonstrating sequential stages of PMP passage through the mammalian ER.


Subject(s)
Endoplasmic Reticulum/metabolism , Lipoproteins/metabolism , Membrane Proteins/metabolism , Adaptor Proteins, Signal Transducing/metabolism , Humans , Intracellular Membranes/metabolism , Peroxins , Peroxisomes/metabolism , Protein Transport/physiology , Ribosomes/metabolism , Signal Recognition Particle/metabolism
3.
J Biol Chem ; 290(29): 17733-17744, 2015 Jul 17.
Article in English | MEDLINE | ID: mdl-26032415

ABSTRACT

The majority of cholesterol-dependent cytolysins (CDCs) utilize cholesterol as a membrane receptor, whereas a small number are restricted to the GPI-anchored protein CD59 for initial membrane recognition. Two cholesterol-binding CDCs, perfringolysin O (PFO) and streptolysin O (SLO), were found to exhibit strikingly different binding properties to cholesterol-rich natural and synthetic membranes. The structural basis for this difference was mapped to one of the loops (L3) in the membrane binding interface that help anchor the toxin monomers to the membrane after receptor (cholesterol) binding by the membrane insertion of its amino acid side chains. A single point mutation in this loop conferred the binding properties of SLO to PFO and vice versa. Our studies strongly suggest that changing the side chain structure of this loop alters its equilibrium between membrane-inserted and uninserted states, thereby affecting the overall binding affinity and total bound toxin. Previous studies have shown that the lipid environment of cholesterol has a dramatic effect on binding and activity. Combining this data with the results of our current studies on L3 suggests that the structure of this loop has evolved in the different CDCs to preferentially direct binding to cholesterol in different lipid environments. Finally, the efficiency of ß-barrel pore formation was inversely correlated with the increased binding and affinity of the PFO L3 mutant, suggesting that selection of a compatible lipid environment impacts the efficiency of membrane insertion of the ß-barrel pore.


Subject(s)
Bacterial Physiological Phenomena , Bacterial Toxins/metabolism , Cell Membrane/microbiology , Cholesterol/metabolism , Cytotoxins/metabolism , Hemolysin Proteins/metabolism , Streptolysins/metabolism , Amino Acid Sequence , Animals , Bacterial Proteins/chemistry , Bacterial Proteins/metabolism , Bacterial Toxins/chemistry , Cell Line , Cell Membrane/metabolism , Cytotoxins/chemistry , Hemolysin Proteins/chemistry , Liposomes/metabolism , Mice , Models, Molecular , Protein Binding , Protein Structure, Secondary , Streptolysins/chemistry
4.
J Mol Biol ; 427(6 Pt A): 1191-201, 2015 Mar 27.
Article in English | MEDLINE | ID: mdl-24979680

ABSTRACT

The signal recognition particle (SRP) cotranslationally recognizes signal sequences of secretory proteins and targets ribosome-nascent chain complexes to the SRP receptor in the endoplasmic reticulum membrane, initiating translocation of the nascent chain through the Sec61 translocon. Although signal sequences do not have homology, they have similar structural regions: a positively charged N-terminus, a hydrophobic core and a more polar C-terminal region that contains the cleavage site for the signal peptidase. Here, we have used site-specific photocrosslinking to study SRP-signal sequence interactions. A photoreactive probe was incorporated into the middle of wild-type or mutated signal sequences of the secretory protein preprolactin by in vitro translation of mRNAs containing an amber-stop codon in the signal peptide in the presence of the N(ε)-(5-azido-2 nitrobenzoyl)-Lys-tRNA(amb) amber suppressor. A homogeneous population of SRP-ribosome-nascent chain complexes was obtained by the use of truncated mRNAs in translations performed in the presence of purified canine SRP. Quantitative analysis of the photoadducts revealed that charged residues at the N-terminus of the signal sequence or in the early part of the mature protein have only a mild effect on the SRP-signal sequence association. However, deletions of amino acid residues in the hydrophobic portion of the signal sequence severely affect SRP binding. The photocrosslinking data correlate with targeting efficiency and translocation across the membrane. Thus, the hydrophobic core of the signal sequence is primarily responsible for its recognition and binding by SRP, while positive charges fine-tune the SRP-signal sequence affinity and targeting to the translocon.


Subject(s)
Endoplasmic Reticulum/metabolism , Protein Sorting Signals , Signal Recognition Particle/metabolism , Animals , Dogs , Membrane Proteins/metabolism , Prolactin/chemistry , Prolactin/metabolism , Protein Binding/genetics , Protein Interaction Domains and Motifs/genetics , Protein Precursors/chemistry , Protein Precursors/metabolism , Protein Sorting Signals/genetics , Protein Transport , RNA, Transfer/metabolism , SEC Translocation Channels , Sequence Deletion
5.
Cell ; 156(1-2): 146-57, 2014 Jan 16.
Article in English | MEDLINE | ID: mdl-24439374

ABSTRACT

Misfolded proteins are often cytotoxic, unless cellular systems prevent their accumulation. Data presented here uncover a mechanism by which defects in secretory proteins lead to a dramatic reduction in their mRNAs and protein expression. When mutant signal sequences fail to bind to the signal recognition particle (SRP) at the ribosome exit site, the nascent chain instead contacts Argonaute2 (Ago2), and the mutant mRNAs are specifically degraded. Severity of signal sequence mutations correlated with increased proximity of Ago2 to nascent chain and mRNA degradation. Ago2 knockdown inhibited degradation of the mutant mRNA, while overexpression of Ago2 or knockdown of SRP54 promoted degradation of secretory protein mRNA. The results reveal a previously unappreciated general mechanism of translational quality control, in which specific mRNA degradation preemptively regulates aberrant protein production (RAPP).


Subject(s)
Protein Biosynthesis , Protein Folding , RNA Stability , Signal Recognition Particle/metabolism , Amino Acid Sequence , Animals , Argonaute Proteins/metabolism , Dogs , HeLa Cells , Humans , Molecular Sequence Data , RNA, Messenger/genetics , RNA, Messenger/metabolism , Sequence Alignment
6.
Nat Struct Mol Biol ; 20(8): 965-72, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23832274

ABSTRACT

Tim23, the central subunit of the TIM23 protein-translocation complex, forms a voltage-gated channel in the mitochondrial inner membrane (MIM), an energy-conserving membrane that generates a proton-motive force to drive vital processes. Using high-resolution fluorescence mapping of a channel-facing transmembrane segment (TMS2) of Tim23 from Saccharomyces cerevisiae, we demonstrate that changes in the energized state of the MIM cause marked structural alterations in the channel region. In an energized membrane, TMS2 forms a continuous α-helix that is inaccessible to the aqueous intermembrane space (IMS). Upon depolarization, the helical periodicity of TMS2 is disrupted, and the channel becomes exposed to the IMS. Kinetic measurements confirm that changes in TMS2 conformation coincide with depolarization. These results reveal how the energized state of the membrane drives functionally relevant structural dynamics in membrane proteins coupled to processes such as channel gating.


Subject(s)
Membrane Transport Proteins/chemistry , Mitochondrial Membranes/metabolism , Models, Molecular , Multiprotein Complexes/chemistry , Proton-Motive Force/physiology , Saccharomyces cerevisiae Proteins/chemistry , Amino Acid Sequence , Cloning, Molecular , Kinetics , Membrane Transport Proteins/metabolism , Microscopy, Fluorescence , Mitochondrial Precursor Protein Import Complex Proteins , Molecular Sequence Data , Multiprotein Complexes/metabolism , Protein Structure, Secondary , Saccharomyces cerevisiae Proteins/metabolism , Sequence Alignment
7.
Nat Chem Biol ; 9(6): 383-9, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23563525

ABSTRACT

Perfringolysin O (PFO), a bacterial cholesterol-dependent cytolysin, binds a mammalian cell membrane, oligomerizes into a circular prepore complex (PPC) and forms a 250-Å transmembrane ß-barrel pore in the cell membrane. Each PFO monomer has two sets of three short α-helices that unfold and ultimately refold into two transmembrane ß-hairpin (TMH) components of the membrane-embedded ß-barrel. Interstrand disulfide-bond scanning revealed that ß-strands in a fully assembled PFO ß-barrel were strictly aligned and tilted at 20° to the membrane perpendicular. In contrast, in a low temperature-trapped PPC intermediate, the TMHs were unfolded and had sufficient freedom of motion to interact transiently with each other, yet the TMHs were not aligned or stably hydrogen bonded. The PFO PPC-to-pore transition therefore converts TMHs in a dynamic folding intermediate far above the membrane into TMHs that are hydrogen bonded to those of adjacent subunits in the bilayer-embedded ß-barrel.


Subject(s)
Bacterial Toxins/chemistry , Disulfides , Hemolysin Proteins/chemistry , Cell Membrane/metabolism , Cholesterol/chemistry , Clostridium perfringens/metabolism , Cross-Linking Reagents/chemistry , Dimerization , Escherichia coli/metabolism , Liposomes/chemistry , Molecular Conformation , Mutation , Protein Binding , Protein Structure, Secondary , Temperature , Trypsin/chemistry
8.
Mol Cell ; 48(3): 398-408, 2012 Nov 09.
Article in English | MEDLINE | ID: mdl-23022384

ABSTRACT

Most membrane proteins are integrated cotranslationally into the ER membrane at the translocon, where nonpolar nascent protein transmembrane segments (TMSs) are widely believed to partition directly into the nonpolar membrane interior. However, a FRET approach that monitors the separation between a fluorescent-labeled TMS and fluorescent phospholipids diffusing in the bulk lipid reveals that TMSs do not immediately enter the lipid phase of the membrane. Instead, TMSs are retained at the translocon by protein-protein interactions until their release into bulk lipid is triggered by translation termination or, in some cases, by the arrival of another nascent chain TMS at a translocon. Nascent chain status and structural elements therefore dictate the timing of TMS release into the lipid phase by altering TMS and flanking sequence interactions with translocons, ribosomes, and associated proteins, thereby controlling when successive TMSs assemble in the bilayer and TMS-delineated loops fold.


Subject(s)
Endoplasmic Reticulum/metabolism , Fluorescence Resonance Energy Transfer/methods , Membrane Proteins/metabolism , Phospholipids/metabolism , Electrophoresis, Polyacrylamide Gel , Models, Biological , Protein Biosynthesis , Protein Transport , RNA, Transfer/metabolism , Ribosomes/metabolism , Temperature , Time Factors
9.
J Biol Chem ; 287(29): 24534-43, 2012 Jul 13.
Article in English | MEDLINE | ID: mdl-22645132

ABSTRACT

The assembly of the cholesterol-dependent cytolysin (CDC) oligomeric pore complex requires a complex choreography of secondary and tertiary structural changes in domain 3 (D3) of the CDC monomer structure. A point mutation was identified in the archetype CDC, perfringolysin O, that blocks detectable D3 structural changes and traps the membrane-bound monomers in an early and reversible stage of oligomer assembly. Using this and other mutants we show that specific D3 structural changes are propagated from one membrane-bound monomer to another. Propagation of these structural changes results in the exposure of a ß-strand in D3 that allows it to pair and form edge-on interactions with a second ß-strand of a free membrane-bound monomer. Pairing of these strands establishes the final geometry of the pore complex and is necessary to drive the formation of the ß-barrel pore. These studies provide new insights into how structural information is propagated between membrane-bound monomers of a self-assembling system and the interactions that establish the geometry of the final pore complex.


Subject(s)
Cholesterol/metabolism , Perforin/chemistry , Perforin/metabolism , Cells, Cultured , Electrophoresis, Polyacrylamide Gel , Fluorescence Resonance Energy Transfer , Hemolysis , Humans , Microscopy, Electron , Perforin/genetics , Point Mutation/genetics , Protein Structure, Secondary , Protein Structure, Tertiary
10.
J Mol Biol ; 416(4): 518-33, 2012 Mar 02.
Article in English | MEDLINE | ID: mdl-22244852

ABSTRACT

The fungal arginine attenuator peptide (AAP) is a regulatory peptide that controls ribosome function. As a nascent peptide within the ribosome exit tunnel, it acts to stall ribosomes in response to arginine (Arg). We used three approaches to probe the molecular basis for stalling. First, PEGylation assays revealed that the AAP did not undergo overall compaction in the tunnel in response to Arg. Second, site-specific photocross-linking showed that Arg altered the conformation of the wild-type AAP, but not of nonfunctional mutants, with respect to the tunnel. Third, using time-resolved spectral measurements with a fluorescent probe placed in the nascent AAP, we detected sequence-specific changes in the disposition of the AAP near the peptidyltransferase center in response to Arg. These data provide evidence that an Arg-induced change in AAP conformation and/or environment in the ribosome tunnel is important for stalling.


Subject(s)
Arginine/chemistry , Carbon-Nitrogen Ligases with Glutamine as Amide-N-Donor/chemistry , Fungal Proteins/chemistry , Peptide Fragments/chemistry , Ribosomal Proteins/chemistry , Ribosomes/chemistry , Amino Acid Sequence , Base Sequence , Carbon-Nitrogen Ligases with Glutamine as Amide-N-Donor/genetics , Molecular Sequence Data , Mutation , Neurospora/chemistry , Open Reading Frames , Peptide Fragments/genetics , Protein Conformation
11.
J Cell Biol ; 195(1): 55-70, 2011 Oct 03.
Article in English | MEDLINE | ID: mdl-21949410

ABSTRACT

Multi-spanning membrane protein loops are directed alternately into the cytosol or ER lumen during cotranslational integration. Nascent chain exposure is switched after a newly synthesized transmembrane segment (TMS) enters the ribosomal tunnel. FRET measurements revealed that each TMS is initially extended, but folds into a compact conformation after moving 6-7 residues from the peptidyltransferase center, irrespective of loop size. The ribosome-induced folding of each TMS coincided with its photocrosslinking to ribosomal protein L17 and an inversion of compartmental exposure. This correlation indicates that successive TMSs fold and bind at a specific ribosomal tunnel site that includes L17, thereby triggering structural rearrangements of multiple components in and on both sides of the ER membrane, most likely via TMS-dependent L17 and/or rRNA conformational changes transmitted to the surface. Thus, cyclical changes at the membrane during integration are initiated by TMS folding, even though nascent chain conformation and location vary dynamically in the ribosome tunnel. Nascent chains therefore control their own trafficking.


Subject(s)
Membrane Proteins/biosynthesis , Protein Biosynthesis/physiology , Protein Folding , Ribosomal Proteins/metabolism , Ribosomes/metabolism , Humans , Saccharomyces cerevisiae
12.
J Cell Biol ; 195(1): 41-54, 2011 Oct 03.
Article in English | MEDLINE | ID: mdl-21949411

ABSTRACT

During cotranslational integration of a eukaryotic multispanning polytopic membrane protein (PMP), its hydrophilic loops are alternately directed to opposite sides of the ER membrane. Exposure of fluorescently labeled nascent PMP to the cytosol or ER lumen was detected by collisional quenching of its fluorescence by iodide ions localized in the cytosol or lumen. PMP loop exposure to the cytosol or lumen was controlled by structural rearrangements in the ribosome, translocon, and associated proteins that occurred soon after a nascent chain transmembrane segment (TMS) entered the ribosomal tunnel. Each successive TMS, although varying in length, sequence, hydrophobicity, and orientation, reversed the structural changes elicited by its predecessor, irrespective of loop size. Fluorescence lifetime data revealed that TMSs occupied a more nonpolar environment than secretory proteins inside the aqueous ribosome tunnel, which suggests that TMS recognition by the ribosome involves hydrophobic interactions. Importantly, the TMS-triggered structural rearrangements that cycle nascent chain exposure between cytosolic and lumenal occur without compromising the permeability barrier of the ER membrane.


Subject(s)
Cell Membrane/metabolism , Cytosol/metabolism , Endoplasmic Reticulum/metabolism , Membrane Proteins/biosynthesis , Protein Biosynthesis/physiology , Ribosomes/metabolism , Cell Membrane/genetics , Endoplasmic Reticulum/genetics , Humans , Hydrophobic and Hydrophilic Interactions , Membrane Proteins/genetics , Protein Structure, Secondary , Ribosomes/genetics , Saccharomyces cerevisiae
13.
Cell ; 146(1): 134-47, 2011 Jul 08.
Article in English | MEDLINE | ID: mdl-21729785

ABSTRACT

In eukaryotic cells, the ribosome-Sec61 translocon complex (RTC) establishes membrane protein topology by cotranslationally partitioning nascent polypeptides into the cytosol, ER lumen, and lipid bilayer. Using photocrosslinking, collisional quenching, cysteine accessibility, and protease protection, we show that a canonical type II signal anchor (SA) acquires its topology through four tightly coupled and mechanistically distinct steps: (1) head-first insertion into Sec61α, (2) nascent chain accumulation within the RTC, (3) inversion from type I to type II topology, and (4) stable translocation of C-terminal flanking residues. Progression through each stage is induced by incremental increases in chain length and involves abrupt changes in the molecular environment of the SA. Importantly, type II SA inversion deviates from a type I SA at an unstable intermediate whose topology is controlled by dynamic interactions between the ribosome and translocon. Thus, the RTC coordinates SA topogenesis within a protected environment via sequential energetic transitions of the TM segment.


Subject(s)
Membrane Proteins/metabolism , Protein Biosynthesis , Ribosomes/metabolism , Animals , Cell-Free System , Dogs , Endoplasmic Reticulum/metabolism , Microsomes/metabolism , Protein Sorting Signals , Rabbits , SEC Translocation Channels
14.
Biochem J ; 437(1): 149-55, 2011 Jul 01.
Article in English | MEDLINE | ID: mdl-21466505

ABSTRACT

A cpSRP [chloroplast SRP (signal recognition particle)] comprising cpSRP54 and cpSRP43 subunits mediates the insertion of light-harvesting proteins into the thylakoid membrane. We dissected its interaction with a full-length membrane protein substrate in aqueous solution by insertion of site-specific photo-activatable cross-linkers into in vitro-synthesized Lhcb1 (major light-harvesting chlorophyll-binding protein of photosystem II). We show that Lhcb1 residues 166-176 cross-link specifically to the cpSRP43 subunit. Some cross-link positions within Lhcb1 are in the 'L18' peptide required for targeting of cpSRP substrates, whereas other cross-linking positions define a new targeting signal in the third transmembrane span. Lhcb1 was not found to cross-link to cpSRP54 at any position, and cross-linking to cpSRP43 is unaffected by the absence of cpSRP54. cpSRP43 thus effectively binds substrates autonomously, and its ability to independently bind an extended 20+-residue substrate region highlights a major difference with other SRP types where the SRP54 subunit binds to hydrophobic target sequences. The results also show that cpSRP43 can bind to a hydrophobic, three-membrane span, substrate in aqueous solution, presumably reflecting a role for cpSRP in the chloroplast stroma. This mode of action, and the specificity of the cpSRP43-substrate interaction, may be associated with cpSRP's unique post-translational mode of action.


Subject(s)
Chloroplasts/metabolism , Membrane Proteins/metabolism , Plant Proteins/metabolism , Signal Recognition Particle/chemistry , Signal Recognition Particle/metabolism , Thylakoids/metabolism , Amino Acid Sequence , Arabidopsis/metabolism , Arabidopsis Proteins/chemistry , Arabidopsis Proteins/metabolism , Binding Sites , Light-Harvesting Protein Complexes/chemistry , Light-Harvesting Protein Complexes/metabolism , Membrane Proteins/chemistry , Molecular Sequence Data , Plant Proteins/chemistry , Protein Conformation , Protein Structure, Tertiary , Solutions/metabolism
15.
Mol Cell ; 41(6): 682-92, 2011 Mar 18.
Article in English | MEDLINE | ID: mdl-21419343

ABSTRACT

The mechanism by which protein folding is coupled to biosynthesis is a critical, but poorly understood, aspect of protein conformational diseases. Here we use fluorescence resonance energy transfer (FRET) to characterize tertiary structural transitions of nascent polypeptides and show that the first nucleotide-binding domain (NBD1) of human CFTR, whose folding is defective in cystic fibrosis, folds via a cotranslational multistep pathway as it is synthesized on the ribosome. Folding begins abruptly as NBD1 residues 389-500 emerge from the ribosome exit tunnel, initiating compaction of a small, N-terminal α/ß-subdomain. Real-time kinetics of synchronized nascent chains revealed that subdomain folding is rapid, occurs coincident with synthesis, and is facilitated by direct ATP binding to the nascent polypeptide. These findings localize the major CF defect late in the NBD1 folding pathway and establish a paradigm wherein a cellular ligand promotes vectorial domain folding by facilitating an energetically favored local peptide conformation.


Subject(s)
Cystic Fibrosis Transmembrane Conductance Regulator/chemistry , Cystic Fibrosis Transmembrane Conductance Regulator/metabolism , Protein Folding , Protein Structure, Tertiary , Ribosomes/metabolism , Adenosine Triphosphate/metabolism , Binding Sites , Cystic Fibrosis Transmembrane Conductance Regulator/genetics , Fluorescence Resonance Energy Transfer , Humans , Ligands , Models, Molecular , Protein Binding , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism
16.
J Mol Biol ; 406(4): 571-82, 2011 Mar 04.
Article in English | MEDLINE | ID: mdl-21237175

ABSTRACT

The translocating chain-associating membrane protein (TRAM) is a glycoprotein involved in the translocation of secreted proteins into the endoplasmic reticulum (ER) lumen and in the insertion of integral membrane proteins into the lipid bilayer. As a major step toward elucidating the structure of the functional ER translocation/insertion machinery, we have characterized the membrane integration mechanism and the transmembrane topology of TRAM using two approaches: photocross-linking and truncated C-terminal reporter tag fusions. Our data indicate that TRAM is recognized by the signal recognition particle and translocon components, and suggest a membrane topology with eight transmembrane segments, including several poorly hydrophobic segments. Furthermore, we studied the membrane insertion capacity of these poorly hydrophobic segments into the ER membrane by themselves. Finally, we confirmed the main features of the proposed membrane topology in mammalian cells expressing full-length TRAM.


Subject(s)
Intracellular Membranes/chemistry , Intracellular Membranes/metabolism , Membrane Glycoproteins/chemistry , Membrane Glycoproteins/metabolism , Animals , Endoplasmic Reticulum/chemistry , Endoplasmic Reticulum/metabolism , Mice , Models, Biological , Models, Molecular , Protein Conformation , Protein Structure, Secondary , Protein Structure, Tertiary
17.
RNA ; 16(8): 1660-72, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20581130

ABSTRACT

Amber suppressor tRNAs are widely used to incorporate nonnatural amino acids into proteins to serve as probes of structure, environment, and function. The utility of this approach would be greatly enhanced if multiple probes could be simultaneously incorporated at different locations in the same protein without other modifications. Toward this end, we have developed amber, opal, and ochre suppressor tRNAs derived from Escherichia coli, and yeast tRNA(Cys) that incorporate a chemically modified cysteine residue with high selectivity at the cognate UAG, UGA, and UAA stop codons in an in vitro translation system. These synthetic tRNAs were aminoacylated in vitro, and the labile aminoacyl bond was stabilized by covalently attaching a fluorescent dye to the cysteine sulfhydryl group. Readthrough efficiency (amber > opal > ochre) was substantially improved by eRF1/eRF3 inhibition with an RNA aptamer, thus overcoming an intrinsic hierarchy in stop codon selection that limits UGA and UAA termination suppression in higher eukaryotic translation systems. This approach now allows concurrent incorporation of two different modified amino acids at amber and opal codons with a combined apparent readthrough efficiency of up to 25% when compared with the parent protein lacking a stop codon. As such, it significantly expands the possibilities for incorporating nonnative amino acids for protein structure/function studies.


Subject(s)
Amino Acids/genetics , Amino Acids/metabolism , RNA, Transfer , Amber , Aminoacylation/genetics , Antineoplastic Combined Chemotherapy Protocols , Asparaginase , Base Pairing , Codon, Terminator , Cysteine/genetics , Cysteine/metabolism , Doxorubicin , Escherichia coli/genetics , Escherichia coli/metabolism , Eukaryota , Prednisone , Proteins/genetics , Proteins/metabolism , RNA, Transfer/chemistry , RNA, Transfer/genetics , RNA, Transfer/metabolism , Vincristine
18.
J Biol Chem ; 285(23): 17614-27, 2010 Jun 04.
Article in English | MEDLINE | ID: mdl-20382739

ABSTRACT

Interactions of Bcl-2 family proteins regulate permeability of the mitochondrial outer membrane and apoptosis. In particular, Bax forms an oligomer that permeabilizes the membrane. To map the interface of the Bax oligomer we used Triton X-100 as a membrane surrogate and performed site-specific photocross-linking. Bax-specific adducts were formed through photo-reactive probes at multiple sites that can be grouped into two surfaces. The first surface overlaps with the BH1-3 groove formed by Bcl-2 Homology motif 1, 2, and 3; the second surface is a rear pocket located on the opposite side of the protein from the BH1-3 groove. Further cross-linking experiments using Bax BH3 peptides and mutants demonstrated that the two surfaces interact with their counterparts in neighboring proteins to form two separated interfaces and that interaction at the BH1-3 groove primes the rear pocket for further interaction. Therefore, Bax oligomerization proceeds through a series of interactions that occur at separate, yet allosterically, coupled interfaces.


Subject(s)
Apoptosis , bcl-2-Associated X Protein/metabolism , Allosteric Site , Amino Acid Motifs , Biochemistry/methods , Cross-Linking Reagents/chemistry , Detergents/pharmacology , Humans , Mutation , Octoxynol/pharmacology , Peptides/chemistry , Plasmids/metabolism , Protein Binding , Protein Structure, Tertiary , Proto-Oncogene Proteins c-bcl-2/chemistry
19.
RNA ; 16(5): 969-79, 2010 May.
Article in English | MEDLINE | ID: mdl-20348448

ABSTRACT

The signal recognition particle (SRP) is a ubiquitous cytoplasmic ribonucleoprotein complex required for the cotranslational targeting of proteins to the endoplasmic reticulum (ER). In eukaryotes, SRP has to arrest the elongation of the nascent chains during targeting to ensure efficient translocation of the preprotein, and this function of SRP is dependent on SRP9/14. Here we present the results of a mutational study on the human protein h9/14 that identified and characterized regions and single residues essential for elongation arrest activity. Effects of the mutations were assessed both in cell-free translation/translocation assays and in cultured mammalian cells. We identified two patches of basic amino acid residues that are essential for activity, whereas the internal loop of SRP14 was found to be dispensable. One patch of important basic residues comprises the previously identified basic pentapetide KRDKK, which can be substituted by four lysines without loss of function. The other patch includes three lysines in the solvent-accessible alpha2 of h9. All essential residues are located in proximity in SRP9/14 and their basic character suggests that they serve as a positively charged platform for interactions with ribosomal RNA. In addition, they can all be lysines consistent with the hypothesis that they recognize their target(s) via electrostatic contacts, most likely with the phosphate backbone, as opposed to contacts with specific bases.


Subject(s)
Signal Recognition Particle/chemistry , Signal Recognition Particle/metabolism , Amino Acid Sequence , Amino Acid Substitution , Base Sequence , Cell Line , Conserved Sequence , Genetic Complementation Test , Humans , In Vitro Techniques , Models, Molecular , Molecular Sequence Data , Mutagenesis, Site-Directed , Peptide Chain Elongation, Translational , Protein Multimerization , Protein Structure, Tertiary , RNA, Small Interfering/genetics , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Sequence Homology, Amino Acid , Signal Recognition Particle/genetics , Static Electricity
20.
J Virol ; 84(11): 5520-7, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20335263

ABSTRACT

Plant viral infection and spread depends on the successful introduction of a virus into a cell of a compatible host, followed by replication and cell-to-cell transport. The movement proteins (MPs) p8 and p9 of Turnip crinkle virus are required for cell-to-cell movement of the virus. We have examined the membrane association of p9 and found that it is an integral membrane protein with a defined topology in the endoplasmic reticulum (ER) membrane. Furthermore, we have used a site-specific photo-cross-linking strategy to study the membrane integration of the protein at the initial stages of its biosynthetic process. This process is cotranslational and proceeds through the signal recognition particle and the translocon complex.


Subject(s)
Carmovirus/chemistry , Membrane Proteins/metabolism , Plant Viral Movement Proteins/metabolism , Endoplasmic Reticulum/chemistry , Signal Recognition Particle
SELECTION OF CITATIONS
SEARCH DETAIL
...