Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 42
Filter
Add more filters










Publication year range
1.
Methods Enzymol ; 686: 143-163, 2023.
Article in English | MEDLINE | ID: mdl-37532398

ABSTRACT

The N-degron pathways are a set of proteolytic systems that relate the half-life of a protein to its N-terminal (Nt) residue. In Escherichia coli the principal N-degron pathway is known as the Leu/N-degron pathway. Proteins degraded by this pathway contain an Nt degradation signal (N-degron) composed of an Nt primary destabilizing (Nd1) residue (Leu, Phe, Trp or Tyr). All Leu/N-degron substrates are recognized by the adaptor protein, ClpS and delivered to the ClpAP protease for degradation. Although many components of the pathway are well defined, the physiological role of this pathway remains poorly understood. To address this gap in knowledge we developed a biospecific affinity chromatography technique to isolate physiological substrates of the Leu/N-degron pathway. In this chapter we describe the use of peptide arrays to determine the binding specificity of ClpS. We demonstrate how the information obtained from the peptide array, when coupled with ClpS affinity chromatography, can be used to specifically elute physiological Leu/N-degron ligands from a bacterial lysate. These techniques are illustrated using E. coli ClpS (EcClpS), but both are broadly suitable for application to related N-recognins and systems, not only for the determination of N-recognin specificity, but also for the identification of natural Leu/N-degron ligands from various bacterial and plant species that contain ClpS homologs.


Subject(s)
Escherichia coli , Peptides , Escherichia coli/genetics , Escherichia coli/metabolism , Ligands , Protein Binding , Peptides/chemistry , Proteolysis , Peptide Hydrolases/metabolism , Substrate Specificity
3.
Commun Biol ; 3(1): 646, 2020 11 06.
Article in English | MEDLINE | ID: mdl-33159171

ABSTRACT

Over a decade ago Polymerase δ interacting protein of 38 kDa (PDIP38) was proposed to play a role in DNA repair. Since this time, both the physiological function and subcellular location of PDIP38 has remained ambiguous and our present understanding of PDIP38 function has been hampered by a lack of detailed biochemical and structural studies. Here we show, that human PDIP38 is directed to the mitochondrion in a membrane potential dependent manner, where it resides in the matrix compartment, together with its partner protein CLPX. Our structural analysis revealed that PDIP38 is composed of two conserved domains separated by an α/ß linker region. The N-terminal (YccV-like) domain of PDIP38 forms an SH3-like ß-barrel, which interacts specifically with CLPX, via the adaptor docking loop within the N-terminal Zinc binding domain of CLPX. In contrast, the C-terminal (DUF525) domain forms an immunoglobin-like ß-sandwich fold, which contains a highly conserved putative substrate binding pocket. Importantly, PDIP38 modulates the substrate specificity of CLPX and protects CLPX from LONM-mediated degradation, which stabilises the cellular levels of CLPX. Collectively, our findings shed new light on the mechanism and function of mitochondrial PDIP38, demonstrating that PDIP38 is a bona fide adaptor protein for the mitochondrial protease, CLPXP.


Subject(s)
Endopeptidase Clp/metabolism , Mitochondria/metabolism , Nuclear Proteins/metabolism , Endopeptidase Clp/genetics , Gene Expression Regulation , HeLa Cells , Humans , Nuclear Proteins/genetics , Recombinant Proteins
4.
Biomolecules ; 10(4)2020 04 16.
Article in English | MEDLINE | ID: mdl-32316259

ABSTRACT

In Escherichia coli, SigmaS (σS) is the master regulator of the general stress response. The cellular levels of σS are controlled by transcription, translation and protein stability. The turnover of σS, by the AAA+ protease (ClpXP), is tightly regulated by a dedicated adaptor protein, termed RssB (Regulator of Sigma S protein B)-which is an atypical member of the response regulator (RR) family. Currently however, the molecular mechanism of σS recognition and delivery by RssB is only poorly understood. Here we describe the crystal structures of both RssB domains (RssBN and RssBC) and the SAXS analysis of full-length RssB (both free and in complex with σS). Together with our biochemical analysis we propose a model for the recognition and delivery of σS by this essential adaptor protein. Similar to most bacterial RRs, the N-terminal domain of RssB (RssBN) comprises a typical mixed (ßα)5-fold. Although phosphorylation of RssBN (at Asp58) is essential for high affinity binding of σS, much of the direct binding to σS occurs via the C-terminal effector domain of RssB (RssBC). In contrast to most RRs the effector domain of RssB forms a ß-sandwich fold composed of two sheets surrounded by α-helical protrusions and as such, shares structural homology with serine/threonine phosphatases that exhibit a PPM/PP2C fold. Our biochemical data demonstrate that this domain plays a key role in both substrate interaction and docking to the zinc binding domain (ZBD) of ClpX. We propose that RssB docking to the ZBD of ClpX overlaps with the docking site of another regulator of RssB, the anti-adaptor IraD. Hence, we speculate that docking to ClpX may trigger release of its substrate through activation of a "closed" state (as seen in the RssB-IraD complex), thereby coupling adaptor docking (to ClpX) with substrate release. This competitive docking to RssB would prevent futile interaction of ClpX with the IraD-RssB complex (which lacks a substrate). Finally, substrate recognition by RssB appears to be regulated by a key residue (Arg117) within the α5 helix of the N-terminal domain. Importantly, this residue is not directly involved in σS interaction, as σS binding to the R117A mutant can be restored by phosphorylation. Likewise, R117A retains the ability to interact with and activate ClpX for degradation of σS, both in the presence and absence of acetyl phosphate. Therefore, we propose that this region of RssB (the α5 helix) plays a critical role in driving interaction with σS at a distal site.


Subject(s)
ATPases Associated with Diverse Cellular Activities/metabolism , DNA-Binding Proteins/metabolism , Endopeptidase Clp/metabolism , Escherichia coli Proteins/metabolism , Escherichia coli/metabolism , Molecular Chaperones/metabolism , Transcription Factors/metabolism , ATPases Associated with Diverse Cellular Activities/chemistry , DNA-Binding Proteins/chemistry , Endopeptidase Clp/chemistry , Escherichia coli Proteins/chemistry , Models, Molecular , Molecular Chaperones/chemistry , Mutation/genetics , Phosphorylation , Protein Binding , Protein Domains , Scattering, Small Angle , Sigma Factor/chemistry , Sigma Factor/metabolism , Transcription Factors/chemistry , X-Ray Diffraction
5.
Sci Rep ; 9(1): 18019, 2019 12 02.
Article in English | MEDLINE | ID: mdl-31792243

ABSTRACT

The ClpP protease is found in all kingdoms of life, from bacteria to humans. In general, this protease forms a homo-oligomeric complex composed of 14 identical subunits, which associates with its cognate ATPase in a symmetrical manner. Here we show that, in contrast to this general architecture, the Clp protease from Mycobacterium smegmatis (Msm) forms an asymmetric hetero-oligomeric complex ClpP1P2, which only associates with its cognate ATPase through the ClpP2 ring. Our structural and functional characterisation of this complex demonstrates that asymmetric docking of the ATPase component is controlled by both the composition of the ClpP1 hydrophobic pocket (Hp) and the presence of a unique C-terminal extension in ClpP1 that guards this Hp. Our structural analysis of MsmClpP1 also revealed openings in the side-walls of the inactive tetradecamer, which may represent sites for product egress.


Subject(s)
Bacterial Proteins/ultrastructure , Endopeptidase Clp/ultrastructure , Mycobacterium smegmatis/metabolism , Protein Multimerization , Protein Subunits/metabolism , Adenosine Triphosphatases/metabolism , Adenosine Triphosphatases/ultrastructure , Bacterial Proteins/metabolism , Crystallography, X-Ray , Endopeptidase Clp/metabolism , Molecular Docking Simulation , Protein Structure, Quaternary , Proteolysis
6.
Sci Rep ; 8(1): 12862, 2018 08 27.
Article in English | MEDLINE | ID: mdl-30150665

ABSTRACT

The maintenance of mitochondrial protein homeostasis (proteostasis) is crucial for correct cellular function. Recently, several mutations in the mitochondrial protease CLPP have been identified in patients with Perrault syndrome 3 (PRLTS3). These mutations can be arranged into two groups, those that cluster near the docking site (hydrophobic pocket, Hp) for the cognate unfoldase CLPX (i.e. T145P and C147S) and those that are adjacent to the active site of the peptidase (i.e. Y229D). Here we report the biochemical consequence of mutations in both regions. The Y229D mutant not only inhibited CLPP-peptidase activity, but unexpectedly also prevented CLPX-docking, thereby blocking the turnover of both peptide and protein substrates. In contrast, Hp mutations cause a range of biochemical defects in CLPP, from no observable change to CLPP activity for the C147S mutant, to dramatic disruption of most activities for the "gain-of-function" mutant T145P - including loss of oligomeric assembly and enhanced peptidase activity.


Subject(s)
Endopeptidase Clp/genetics , Genetic Association Studies , Genetic Predisposition to Disease , Genetic Variation , Gonadal Dysgenesis, 46,XX/diagnosis , Gonadal Dysgenesis, 46,XX/genetics , Hearing Loss, Sensorineural/diagnosis , Hearing Loss, Sensorineural/genetics , Endopeptidase Clp/chemistry , Endopeptidase Clp/metabolism , Gonadal Dysgenesis, 46,XX/metabolism , Hearing Loss, Sensorineural/metabolism , Humans , Mitochondria/genetics , Mitochondria/metabolism , Models, Molecular , Mutation , Protein Conformation
7.
Proc Natl Acad Sci U S A ; 115(12): 2982-2987, 2018 03 20.
Article in English | MEDLINE | ID: mdl-29514959

ABSTRACT

Succinate:quinone oxidoreductase (SQR) functions in energy metabolism, coupling the tricarboxylic acid cycle and electron transport chain in bacteria and mitochondria. The biogenesis of flavinylated SdhA, the catalytic subunit of SQR, is assisted by a highly conserved assembly factor termed SdhE in bacteria via an unknown mechanism. By using X-ray crystallography, we have solved the structure of Escherichia coli SdhE in complex with SdhA to 2.15-Å resolution. Our structure shows that SdhE makes a direct interaction with the flavin adenine dinucleotide-linked residue His45 in SdhA and maintains the capping domain of SdhA in an "open" conformation. This displaces the catalytic residues of the succinate dehydrogenase active site by as much as 9.0 Å compared with SdhA in the assembled SQR complex. These data suggest that bacterial SdhE proteins, and their mitochondrial homologs, are assembly chaperones that constrain the conformation of SdhA to facilitate efficient flavinylation while regulating succinate dehydrogenase activity for productive biogenesis of SQR.


Subject(s)
Electron Transport Complex II/metabolism , Escherichia coli Proteins/chemistry , Flavoproteins/chemistry , Bacterial Proteins , Crystallization , Crystallography, X-Ray , Electron Transport Complex II/genetics , Escherichia coli , Escherichia coli Proteins/ultrastructure , Flavoproteins/ultrastructure , Models, Molecular , Protein Binding , Protein Conformation , Protein Domains , Strobilurins
8.
FEBS Lett ; 592(1): 15-23, 2018 01.
Article in English | MEDLINE | ID: mdl-29197082

ABSTRACT

The pupylation of cellular proteins plays a crucial role in the degradation cascade via the Pup-Proteasome system (PPS). It is essential for the survival of Mycobacterium smegmatis under nutrient starvation and, as such, the activity of many components of the pathway is tightly regulated. Here, we show that Pup, like ubiquitin, can form polyPup chains primarily through K61 and that this form of Pup inhibits the ATPase-mediated turnover of pupylated substrates by the 20S proteasome. Similarly, the autopupylation of PafA (the sole Pup ligase found in mycobacteria) inhibits its own enzyme activity; hence, pupylation of PafA may act as a negative feedback mechanism to prevent substrate pupylation under specific cellular conditions.


Subject(s)
Bacterial Proteins/metabolism , Mycobacterium smegmatis/metabolism , Proteasome Endopeptidase Complex/metabolism , Adenosine Triphosphatases/metabolism , Amino Acid Substitution , Bacterial Proteins/chemistry , Bacterial Proteins/genetics , Lysine/chemistry , Mutagenesis, Site-Directed , Mycobacterium smegmatis/genetics , Protein Processing, Post-Translational , Proteolysis , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Substrate Specificity , Ubiquitin-Protein Ligase Complexes/chemistry , Ubiquitin-Protein Ligase Complexes/genetics , Ubiquitin-Protein Ligase Complexes/metabolism
9.
FEBS Lett ; 590(19): 3397-3406, 2016 Oct.
Article in English | MEDLINE | ID: mdl-27588721

ABSTRACT

The N-end rule is a conserved protein degradation pathway that relates the metabolic stability of a protein to the identity of its N-terminal residue. Proteins bearing a destabilising N-terminal residue (N-degron) are recognised by specialised components of the pathway (N-recognins) and degraded by cellular proteases. In bacteria, the N-recognin ClpS is responsible for the specific recognition of proteins bearing an N-terminal destabilising residue such as leucine, phenylalanine, tyrosine or tryptophan. In this study, we show that the putative apicoplast N-recognin from Plasmodium falciparum (PfClpS), in contrast to its bacterial homologues, exhibits an expanded substrate specificity that includes recognition of the branched chain amino acid isoleucine.


Subject(s)
Endopeptidase Clp/metabolism , Neoplasm Proteins/metabolism , Plasmodium/enzymology , Protozoan Proteins/metabolism , Endopeptidase Clp/chemistry , Isoleucine/metabolism , Neoplasm Proteins/chemistry , Protein Domains , Proteolysis , Protozoan Proteins/chemistry , Substrate Specificity
10.
Sci Rep ; 5: 17397, 2015 Dec 02.
Article in English | MEDLINE | ID: mdl-26627475

ABSTRACT

Maintenance of mitochondrial protein homeostasis is critical for proper cellular function. Under normal conditions resident molecular chaperones and proteases maintain protein homeostasis within the organelle. Under conditions of stress however, misfolded proteins accumulate leading to the activation of the mitochondrial unfolded protein response (UPR(mt)). While molecular chaperone assisted refolding of proteins in mammalian mitochondria has been well documented, the contribution of AAA+ proteases to the maintenance of protein homeostasis in this organelle remains unclear. To address this gap in knowledge we examined the contribution of human mitochondrial matrix proteases, LONM and CLPXP, to the turnover of OTC-∆, a folding incompetent mutant of ornithine transcarbamylase, known to activate UPR(mt). Contrary to a model whereby CLPXP is believed to degrade misfolded proteins, we found that LONM, and not CLPXP is responsible for the turnover of OTC-∆ in human mitochondria. To analyse the conformational state of proteins that are recognised by LONM, we examined the turnover of unfolded and aggregated forms of malate dehydrogenase (MDH) and OTC. This analysis revealed that LONM specifically recognises and degrades unfolded, but not aggregated proteins. Since LONM is not upregulated by UPR(mt), this pathway may preferentially act to promote chaperone mediated refolding of proteins.


Subject(s)
Mitochondria/metabolism , Mitochondrial Proteins/metabolism , Protease La/metabolism , Protein Aggregates , Proteolysis , Unfolded Protein Response , Animals , Endopeptidase Clp/genetics , Endopeptidase Clp/metabolism , HeLa Cells , Humans , Mitochondria/genetics , Mitochondrial Proteins/genetics , Protease La/genetics , Rats
11.
Front Mol Biosci ; 2: 15, 2015.
Article in English | MEDLINE | ID: mdl-25988182

ABSTRACT

In Escherichia coli, σ(S) is the master regulator of the general stress response. The level of σ(S) changes in response to multiple stress conditions and it is regulated at many levels including protein turnover. In the absence of stress, σ(S) is rapidly degraded by the AAA+ protease, ClpXP in a regulated manner that depends on the adaptor protein RssB. This two-component response regulator mediates the recognition of σ(S) and its delivery to ClpXP. The turnover of σ(S) however, can be inhibited in a stress specific manner, by one of three anti-adaptor proteins. Each anti-adaptor binds to RssB and inhibits its activity, but how this is achieved is not fully understood at a molecular level. Here, we describe details of the interaction between each anti-adaptor and RssB that leads to the stabilization of σ(S). By defining the domains of RssB using partial proteolysis we demonstrate that each anti-adaptor uses a distinct mode of binding to inhibit RssB activity. IraD docks specifically to the N-terminal domain of RssB, IraP interacts primarily with the C-terminal domain, while IraM interacts with both domains. Despite these differences in binding, we propose that docking of each anti-adaptor induces a conformational change in RssB, which resembles the inactive dimer of RssB. This dimer-like state of RssB not only prevents substrate binding but also triggers substrate release from a pre-bound complex.

12.
FASEB J ; 28(4): 1794-804, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24414418

ABSTRACT

Mutations in succinate dehydrogenase (SDH) subunits and assembly factors cause a range of clinical conditions. One such condition, hereditary paraganglioma 2 (PGL2), is caused by a G78R mutation in the assembly factor SDH5. Although SDH5(G78R) is deficient in its ability to promote SDHA flavinylation, it has remained unclear whether impairment to its import, structure, or stability contributes to its loss of function. Using import-chase analysis in human mitochondria isolated from HeLa cells, we found that the import and maturation of human SDH5(G78R) was normal, while its stability was reduced significantly, with ~25% of the protein remaining after 180 min compared to ~85% for the wild-type protein. Notably, the metabolic stability of SDH5(G78R) was restored to wild-type levels by depleting mitochondrial LON (LONM). Degradation of SDH5(G78R) by LONM was confirmed in vitro; however, in contrast to the in organello analysis, wild-type SDH5 was also rapidly degraded by LONM. SDH5 instability was confirmed in SDHA-depleted mitochondria. Blue native PAGE showed that imported SDH5(G78R) formed a transient complex with SDHA; however, this complex was stabilized in LONM depleted mitochondria. These data demonstrate that SDH5 is protected from LONM-mediated degradation in mitochondria by its stable interaction with SDHA, a state that is dysregulated in PGL2.


Subject(s)
Electron Transport Complex II/metabolism , Mitochondria/metabolism , Mitochondrial Proteins/metabolism , Paraganglioma/metabolism , Protease La/metabolism , Proteostasis Deficiencies/metabolism , Electron Transport Complex II/genetics , Enzyme Stability/genetics , Flavin-Adenine Dinucleotide/metabolism , HeLa Cells , Humans , Immunoblotting , Mitochondrial Proteins/genetics , Paraganglioma/genetics , Protease La/genetics , Protein Binding , Protein Subunits/genetics , Protein Subunits/metabolism , Proteostasis Deficiencies/genetics , Substrate Specificity
13.
J Mol Microbiol Biotechnol ; 23(4-5): 335-44, 2013.
Article in English | MEDLINE | ID: mdl-23920496

ABSTRACT

Targeted protein degradation is crucial for the correct function and maintenance of a cell. In bacteria, this process is largely performed by a handful of ATP-dependent machines, which generally consist of two components - an unfoldase and a peptidase. In some cases, however, substrate recognition by the protease may be regulated by specialized delivery factors (known as adaptor proteins). Our detailed understanding of how these machines are regulated to prevent uncontrolled degradation within a cell has permitted the identification of novel antimicrobials that dysregulate these machines, as well as the development of tunable degradation systems that have applications in biotechnology. Here, we focus on the physiological role of the ClpP peptidase in bacteria, its role as a novel antibiotic target and the use of protein degradation as a biotechnological approach to artificially control the expression levels of a protein of interest.


Subject(s)
Bacteria/metabolism , Bacterial Proteins/metabolism , Endopeptidase Clp/metabolism , Gene Expression Regulation, Bacterial , Proteolysis , Biomedical Technology/methods , Biotechnology/methods
14.
Subcell Biochem ; 66: 223-63, 2013.
Article in English | MEDLINE | ID: mdl-23479443

ABSTRACT

Mitochondria are specialised organelles that are structurally and functionally integrated into cells in the vast majority of eukaryotes. They are the site of numerous enzymatic reactions, some of which are essential for life. The double lipid membrane of the mitochondrion, that spatially defines the organelle and is necessary for some functions, also creates a physical but semi-permeable barrier to the rest of the cell. Thus to ensure the biogenesis, regulation and maintenance of a functional population of proteins, an autonomous protein handling network within mitochondria is required. This includes resident mitochondrial protein translocation machinery, processing peptidases, molecular chaperones and proteases. This review highlights the contribution of proteases of the AAA+ superfamily to protein quality and activity control within the mitochondrion. Here they are responsible for the degradation of unfolded, unassembled and oxidatively damaged proteins as well as the activity control of some enzymes. Since most knowledge about these proteases has been gained from studies in the eukaryotic microorganism Saccharomyces cerevisiae, much of the discussion here centres on their role in this organism. However, reference is made to mitochondrial AAA+ proteases in other organisms, particularly in cases where they play a unique role such as the mitochondrial unfolded protein response. As these proteases influence mitochondrial function in both health and disease in humans, an understanding of their regulation and diverse activities is necessary.


Subject(s)
Bacterial Proteins/metabolism , Gene Expression Regulation, Bacterial , Homeostasis/physiology , Mitochondria/metabolism , Mitochondrial Proteins/metabolism , Peptide Hydrolases/metabolism , Protein Biosynthesis , Proteolysis
15.
Mol Biol Cell ; 23(20): 3957-69, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22918950

ABSTRACT

The intermembrane space of mitochondria accommodates the essential mitochondrial intermembrane space assembly (MIA) machinery that catalyzes oxidative folding of proteins. The disulfide bond formation pathway is based on a relay of reactions involving disulfide transfer from the sulfhydryl oxidase Erv1 to Mia40 and from Mia40 to substrate proteins. However, the substrates of the MIA typically contain two disulfide bonds. It was unclear what the mechanisms are that ensure that proteins are released from Mia40 in a fully oxidized form. In this work, we dissect the stage of the oxidative folding relay, in which Mia40 binds to its substrate. We identify dynamics of the Mia40-substrate intermediate complex. Our experiments performed in a native environment, both in organello and in vivo, show that Erv1 directly participates in Mia40-substrate complex dynamics by forming a ternary complex. Thus Mia40 in cooperation with Erv1 promotes the formation of two disulfide bonds in the substrate protein, ensuring the efficiency of oxidative folding in the intermembrane space of mitochondria.


Subject(s)
Mitochondrial Membrane Transport Proteins/metabolism , Mitochondrial Proteins/metabolism , Oxidoreductases Acting on Sulfur Group Donors/metabolism , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae/metabolism , Binding Sites , Cysteine/genetics , Mitochondrial Membrane Transport Proteins/chemistry , Mitochondrial Membranes/metabolism , Mitochondrial Precursor Protein Import Complex Proteins , Multiprotein Complexes/metabolism , Mutation/genetics , Oxidation-Reduction , Phenotype , Protein Binding , Protein Conformation , Protein Precursors/metabolism , Saccharomyces cerevisiae/growth & development , Saccharomyces cerevisiae Proteins/chemistry , Substrate Specificity
16.
J Struct Biol ; 179(2): 193-201, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22710082

ABSTRACT

The mitochondrial matrix of mammalian cells contains several different ATP-dependent proteases, including CLPXP, some of which contribute to protein maturation and quality control. Currently however, the substrates and the physiological roles of mitochondrial CLPXP in humans, has remained elusive. Similarly, the mechanism by which these ATP-dependent proteases recognize their substrates currently remains unclear. Here we report the characterization of a Walker B mutation in human CLPX, in which the highly conserved glutamate was replaced with alanine. This mutant protein exhibits improved interaction with the model unfolded substrate casein and several putative physiological substrates in vitro. Although this mutant lacks ATPase activity, it retains the ability to mediate casein degradation by hCLPP, in a fashion similar to the small molecule ClpP-activator, ADEP. Our functional dissection of hCLPX structure, also identified that most model substrates are recognized by the N-terminal domain, although some substrates bypass this step and dock, directly to the pore-1 motif. Collectively these data reveal, that despite the difference between bacterial and human CLPXP complexes, human CLPXP exhibits a similar mode of substrate recognition and is deregulated by ADEPs.


Subject(s)
Endopeptidase Clp/metabolism , Animals , Endopeptidase Clp/genetics , Humans , Mutation , Protein Binding , Substrate Specificity
17.
Mol Cell ; 44(5): 811-8, 2011 Dec 09.
Article in English | MEDLINE | ID: mdl-22152483

ABSTRACT

The mitochondrial inner membrane harbors the complexes of the respiratory chain and translocase complexes for precursor proteins. We have identified a further subunit of the carrier translocase (TIM22 complex) that surprisingly is identical to subunit 3 of respiratory complex II, succinate dehydrogenase (Sdh3). The membrane-integral protein Sdh3 plays specific functions in electron transfer in complex II. We show by genetic and biochemical approaches that Sdh3 also plays specific functions in the TIM22 complex. Sdh3 forms a subcomplex with Tim18 and is involved in biogenesis and assembly of the membrane-integral subunits of the TIM22 complex. We conclude that the assembly of Sdh3 with different partner proteins, Sdh4 and Tim18, recruits it to two different mitochondrial membrane complexes with functions in bioenergetics and protein biogenesis, respectively.


Subject(s)
Electron Transport , Mitochondrial Membrane Transport Proteins/metabolism , Mitochondrial Membranes/metabolism , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae/metabolism , Succinate Dehydrogenase/metabolism , Electron Transport Complex II/metabolism , Mitochondrial Membranes/enzymology , Mitochondrial Precursor Protein Import Complex Proteins , Saccharomyces cerevisiae/cytology , Saccharomyces cerevisiae/enzymology
18.
IUBMB Life ; 63(11): 955-63, 2011 Nov.
Article in English | MEDLINE | ID: mdl-22031494

ABSTRACT

In the crowded environment of a cell, the protein quality control machinery, such as molecular chaperones and proteases, maintains a population of folded and hence functional proteins. The accumulation of unfolded proteins in a cell is particularly harmful as it not only reduces the concentration of active proteins but also overburdens the protein quality control machinery, which in turn, can lead to a significant increase in nonproductive folding and protein aggregation. To circumvent this problem, cells use heat shock and unfolded protein stress response pathways, which essentially sense the change to protein homeostasis upregulating protein quality control factors that act to restore the balance. Interestingly, several stress response pathways are proteolytically controlled. In this review, we provide a brief summary of targeted protein degradation by AAA+ proteases and focus on the role of ClpXP proteases, particularly in the signaling pathway of the Escherichia coli extracellular stress response and the mitochondrial unfolded protein response.


Subject(s)
Endopeptidase Clp/metabolism , Escherichia coli Proteins/metabolism , Escherichia coli/metabolism , Mitochondria/metabolism , Unfolded Protein Response , Animals , Cell Wall/metabolism , Escherichia coli/enzymology , Humans , Mitochondria/enzymology , Proteolysis , Signal Transduction , Stress, Physiological
19.
Biochem Cell Biol ; 88(1): 97-108, 2010 Feb.
Article in English | MEDLINE | ID: mdl-20130683

ABSTRACT

In eukaryotes, mitochondria are required for the proper function of the cell and as such the maintenance of proteins within this organelle is crucial. One class of proteins, collectively known as the AAA+ (ATPases associated with various cellular activities) superfamily, make a number of important contributions to mitochondrial protein homeostasis. In this organelle, they contribute to the maturation and activation of proteins, general protein quality control, respiratory chain complex assembly, and mitochondrial DNA maintenance and integrity. To achieve such diverse functions this group of ATP-dependent unfoldases utilize the energy from ATP hydrolysis to modulate the structure of proteins via unique domains and (or) associated functional components. In this review, we describe the current status of knowledge regarding the known mitochondrial AAA+ proteins and their role in this organelle.


Subject(s)
Adenosine Triphosphatases/chemistry , Adenosine Triphosphatases/metabolism , Mitochondria/metabolism , Mitochondrial Proteins/metabolism , Adenosine Triphosphatases/genetics , Humans , Hydrolysis , Mitochondria/genetics , Mitochondrial Proteins/genetics , Molecular Chaperones/genetics , Molecular Chaperones/metabolism , Protein Structure, Secondary , Protein Structure, Tertiary
20.
Mol Cell Biol ; 30(1): 307-18, 2010 Jan.
Article in English | MEDLINE | ID: mdl-19884344

ABSTRACT

Mitochondrial import of cleavable preproteins occurs at translocation contact sites, where the translocase of the outer membrane (TOM) associates with the presequence translocase of the inner membrane (TIM23) in a supercomplex. Different views exist on the mechanism of how TIM23 mediates preprotein sorting to either the matrix or inner membrane. On the one hand, two TIM23 forms were proposed, a matrix transport form containing the presequence translocase-associated motor (PAM; TIM23-PAM) and a sorting form containing Tim21 (TIM23(SORT)). On the other hand, it was reported that TIM23 and PAM are permanently associated in a single-entity translocase. We have accumulated distinct transport intermediates of preproteins to analyze the translocases in their active, preprotein-carrying state. We identified two different forms of active TOM-TIM23 supercomplexes, TOM-TIM23(SORT) and TOM-TIM23-PAM. These two supercomplexes do not represent separate pathways but are in dynamic exchange during preprotein translocation and sorting. Depending on the signals of the preproteins, switches between the different forms of supercomplex and TIM23 are required for the completion of preprotein import.


Subject(s)
Carrier Proteins/physiology , Membrane Transport Proteins/physiology , Mitochondrial Proteins/physiology , Saccharomyces cerevisiae Proteins/physiology , Saccharomyces cerevisiae/metabolism , Mitochondria/metabolism , Mitochondrial Membrane Transport Proteins/metabolism , Mitochondrial Precursor Protein Import Complex Proteins , Multiprotein Complexes , Protein Sorting Signals , Protein Transport , Saccharomyces cerevisiae Proteins/metabolism , Tetrahydrofolate Dehydrogenase/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...