Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 53
Filter
1.
Mol Cell ; 81(12): 2507-2519, 2021 06 17.
Article in English | MEDLINE | ID: mdl-34107306

ABSTRACT

Protein homeostasis mechanisms are fundamentally important to match cellular needs and to counteract stress conditions. A fundamental challenge is to understand how defective proteins are recognized and extracted from cellular organelles to be degraded in the cytoplasm. The endoplasmic reticulum (ER)-associated degradation (ERAD) pathway is the best-understood organellar protein quality control system. Here, we review new insights into the mechanism of recognition and retrotranslocation of client proteins in ERAD. In addition to the membrane-integral ERAD E3 ubiquitin ligases, we highlight one protein family that is remarkably often involved in various aspects of membrane protein quality control and protein dislocation: the rhomboid superfamily, which includes derlins and intramembrane serine proteases. Rhomboid-like proteins have been found to control protein homeostasis in the ER, but also in other eukaryotic organelles and in bacteria, pointing toward conserved principles of membrane protein quality control across organelles and evolution.


Subject(s)
Endoplasmic Reticulum-Associated Degradation/physiology , Proteostasis/physiology , Endoplasmic Reticulum/metabolism , Endoplasmic Reticulum/physiology , Eukaryotic Cells/metabolism , Humans , Membrane Proteins/metabolism , Proteasome Endopeptidase Complex/metabolism , Proteolysis , Ubiquitin-Protein Ligases/metabolism
2.
Mol Cell ; 69(2): 161-162, 2018 01 18.
Article in English | MEDLINE | ID: mdl-29351840

ABSTRACT

Defective ER-resident membrane proteins need to be ejected into the cytoplasm in order to be degraded by the proteasome, but the exact mechanism remains unclear. In this issue of Molecular Cell, Neal et al. (2018) reveal that the rhomboid pseudoprotease Dfm1 defines the central ERAD component for membrane protein dislocation.


Subject(s)
Endoplasmic Reticulum-Associated Degradation , Membrane Proteins , Endoplasmic Reticulum , Proteasome Endopeptidase Complex
3.
J Biol Chem ; 300(2): 105644, 2024 Feb.
Article in English | MEDLINE | ID: mdl-38218226

ABSTRACT

Intramembrane proteolysis regulates important processes such as signaling and transcriptional and posttranslational abundance control of proteins with key functions in metabolic pathways. This includes transcriptional control of mevalonate pathway genes, thereby ensuring balanced biosynthesis of cholesterol and other isoprenoids. Our work shows that, at high cholesterol levels, signal peptide peptidase (SPP) cleaves squalene synthase (SQS), an enzyme that defines the branching point for allocation of isoprenoids to the sterol and nonsterol arms of the mevalonate pathway. This intramembrane cleavage releases SQS from the membrane and targets it for proteasomal degradation. Regulation of this mechanism is achieved by the E3 ubiquitin ligase TRC8 that, in addition to ubiquitinating SQS in response to cholesterol levels, acts as an allosteric activator of SPP-catalyzed intramembrane cleavage of SQS. Cellular cholesterol levels increase in the absence of SPP activity. We infer from these results that, SPP-TRC8 mediated abundance control of SQS acts as a regulation step within the mevalonate pathway.


Subject(s)
Farnesyl-Diphosphate Farnesyltransferase , Mevalonic Acid , Aspartic Acid Endopeptidases , Cholesterol/metabolism , Farnesyl-Diphosphate Farnesyltransferase/genetics , Farnesyl-Diphosphate Farnesyltransferase/metabolism , Mevalonic Acid/metabolism , Terpenes , HEK293 Cells , Humans
4.
EMBO J ; 39(10): e105012, 2020 05 18.
Article in English | MEDLINE | ID: mdl-32338387

ABSTRACT

Rhomboid intramembrane serine proteases are present in all kingdoms of life, but as we do not know their substrates in many species, it remains puzzling why rhomboids are among the most-conserved integral membrane proteins. Two new studies in The EMBO Journal by Began et al and Liu et al now link bacterial rhomboid proteases to membrane protein degradation, showing striking similarities to what is known about eukaryotic rhomboid family proteins, thus pointing toward a conserved membrane surveillance mechanism.


Subject(s)
Bacterial Proteins , Membrane Proteins , ATPases Associated with Diverse Cellular Activities , Bacteria/metabolism , Bacterial Proteins/metabolism , Endoplasmic Reticulum-Associated Degradation , Licensure , Membrane Proteins/metabolism , Membranes/metabolism , Peptide Hydrolases/metabolism
5.
J Biol Chem ; 298(9): 102321, 2022 09.
Article in English | MEDLINE | ID: mdl-35921890

ABSTRACT

The intramembrane protease PARL acts as a crucial mitochondrial safeguard by cleaving the mitophagy regulators PINK1 and PGAM5. Depending on the stress level, PGAM5 can either stimulate cell survival or cell death. In contrast to PINK1, which is constantly cleaved in healthy mitochondria and only active when the inner mitochondrial membrane is depolarized, PGAM5 processing is inversely regulated. However, determinants of PGAM5 that indicate it as a conditional substrate for PARL have not been rigorously investigated, and it is unclear how uncoupling the mitochondrial membrane potential affects its processing compared to that of PINK1. Here, we show that several polar transmembrane residues in PGAM5 distant from the cleavage site serve as determinants for its PARL-catalyzed cleavage. Our NMR analysis indicates that a short N-terminal amphipathic helix, followed by a kink and a C-terminal transmembrane helix harboring the scissile peptide bond are key for a productive interaction with PARL. Furthermore, we also show that PGAM5 is stably inserted into the inner mitochondrial membrane until uncoupling the membrane potential triggers its disassembly into monomers, which are then cleaved by PARL. In conclusion, we propose a model in which PGAM5 is slowly processed by PARL-catalyzed cleavage that is influenced by multiple hierarchical substrate features, including a membrane potential-dependent oligomeric switch.


Subject(s)
Homeostasis , Metalloproteases , Mitochondria , Mitochondrial Proteins , Phosphoprotein Phosphatases , Proteolysis , HeLa Cells , Humans , Metalloproteases/metabolism , Mitochondria/enzymology , Mitochondrial Proteins/metabolism , Peptides/metabolism , Phosphoprotein Phosphatases/metabolism , Protein Kinases/metabolism
6.
J Biol Chem ; 296: 100383, 2021.
Article in English | MEDLINE | ID: mdl-33556373

ABSTRACT

The rhomboid protease PARL is a critical regulator of mitochondrial homeostasis through its cleavage of substrates such as PINK1, PGAM5, and Smac/Diablo, which have crucial roles in mitochondrial quality control and apoptosis. However, the catalytic properties of PARL, including the effect of lipids on the protease, have never been characterized in vitro. To address this, we isolated human PARL expressed in yeast and used FRET-based kinetic assays to measure proteolytic activity in vitro. We show that PARL activity in detergent is enhanced by cardiolipin, a lipid enriched in the mitochondrial inner membrane. Significantly higher turnover rates were observed for PARL reconstituted in proteoliposomes, with Smac/Diablo being cleaved most rapidly at a rate of 1 min-1. In contrast, PGAM5 is cleaved with the highest efficiency (kcat/KM) compared with PINK1 and Smac/Diablo. In proteoliposomes, a truncated ß-cleavage form of PARL, a physiological form known to affect mitochondrial fragmentation, is more active than the full-length enzyme for hydrolysis of PINK1, PGAM5, and Smac/Diablo. Multiplex profiling of 228 peptides reveals that PARL prefers substrates with a bulky side chain such as Phe in P1, which is distinct from the preference for small side chain residues typically found with bacterial rhomboid proteases. This study using recombinant PARL provides fundamental insights into its catalytic activity and substrate preferences that enhance our understanding of its role in mitochondrial function and has implications for specific inhibitor design.


Subject(s)
Metalloproteases/metabolism , Metalloproteases/physiology , Mitochondrial Proteins/metabolism , Mitochondrial Proteins/physiology , Apoptosis Regulatory Proteins/metabolism , Catalytic Domain , Endopeptidases/metabolism , HEK293 Cells , HeLa Cells , Humans , Metalloproteases/genetics , Mitochondria/metabolism , Mitochondrial Membranes/metabolism , Mitochondrial Proteins/genetics , Peptide Hydrolases/metabolism , Protein Kinases/genetics , Protein Kinases/metabolism , Proteolysis
7.
EMBO J ; 37(15)2018 08 01.
Article in English | MEDLINE | ID: mdl-29976761

ABSTRACT

Proteolytic removal of membrane protein ectodomains (ectodomain shedding) is a post-translational modification that controls levels and function of hundreds of membrane proteins. The contributing proteases, referred to as sheddases, act as important molecular switches in processes ranging from signaling to cell adhesion. When deregulated, ectodomain shedding is linked to pathologies such as inflammation and Alzheimer's disease. While proteases of the "a disintegrin and metalloprotease" (ADAM) and "beta-site APP cleaving enzyme" (BACE) families are widely considered as sheddases, in recent years a much broader range of proteases, including intramembrane and soluble proteases, were shown to catalyze similar cleavage reactions. This review demonstrates that shedding is a fundamental process in cell biology and discusses the current understanding of sheddases and their substrates, molecular mechanisms and cellular localizations, as well as physiological functions of protein ectodomain shedding. Moreover, we provide an operational definition of shedding and highlight recent conceptual advances in the field. While new developments in proteomics facilitate substrate discovery, we expect that shedding is not a rare exception, but rather the rule for many membrane proteins, and that many more interesting shedding functions await discovery.


Subject(s)
Cell Membrane/metabolism , Membrane Proteins/metabolism , Protein Domains/physiology , Protein Processing, Post-Translational/physiology , Proteolysis , ADAM Proteins/metabolism , Amyloid Precursor Protein Secretases/metabolism , Animals , Aspartic Acid Endopeptidases/metabolism , Humans , Signal Transduction
8.
J Cell Sci ; 133(6)2020 03 26.
Article in English | MEDLINE | ID: mdl-32005703

ABSTRACT

The endoplasmic reticulum (ER)-resident intramembrane rhomboid protease RHBDL4 generates metastable protein fragments and together with the ER-associated degradation (ERAD) machinery provides a clearance mechanism for aberrant and surplus proteins. However, the endogenous substrate spectrum and with that the role of RHBDL4 in physiological ERAD is mainly unknown. Here, we use a substrate trapping approach in combination with quantitative proteomics to identify physiological RHBDL4 substrates. This revealed oligosaccharyltransferase (OST) complex subunits such as the catalytic active subunit STT3A as substrates for the RHBDL4-dependent ERAD pathway. RHBDL4-catalysed cleavage inactivates OST subunits by triggering dislocation into the cytoplasm and subsequent proteasomal degradation. RHBDL4 thereby controls the abundance and activity of OST, suggesting a novel link between the ERAD machinery and glycosylation tuning.


Subject(s)
Endoplasmic Reticulum-Associated Degradation , Hexosyltransferases , Membrane Proteins , Hexosyltransferases/genetics , Membrane Proteins/genetics , Membrane Proteins/metabolism , Peptide Hydrolases/metabolism
9.
FASEB J ; 35(3): e21380, 2021 03.
Article in English | MEDLINE | ID: mdl-33566379

ABSTRACT

Interleukin-11 (IL-11) is a pleiotropic cytokine with both pro- and anti-inflammatory properties. It activates its target cells via binding to the membrane-bound IL-11 receptor (IL-11R), which then recruits a homodimer of the ubiquitously expressed, signal-transducing receptor gp130. Besides this classic signaling pathway, IL-11 can also bind to soluble forms of the IL-11R (sIL-11R), and IL-11/sIL-11R complexes activate cells via the induction of gp130 homodimerization (trans-signaling). We have previously reported that the metalloprotease ADAM10 cleaves the membrane-bound IL-11R and thereby generates sIL-11R. In this study, we identify the rhomboid intramembrane protease RHBDL2 as a so far unrecognized alternative sheddase that can efficiently trigger IL-11R secretion. We determine the cleavage site used by RHBDL2, which is located in the extracellular part of the receptor in close proximity to the plasma membrane, between Ala-370 and Ser-371. Furthermore, we identify critical amino acid residues within the transmembrane helix that are required for IL-11R proteolysis. We also show that ectopically expressed RHBDL2 is able to cleave the IL-11R within the early secretory pathway and not only at the plasma membrane, indicating that its subcellular localization plays a central role in controlling its activity. Moreover, RHBDL2-derived sIL-11R is biologically active and able to perform IL-11 trans-signaling. Finally, we show that the human mutation IL-11R-A370V does not impede IL-11 classic signaling, but prevents RHBDL2-mediated IL-11R cleavage.


Subject(s)
Interleukin-11/physiology , Receptors, Interleukin-11/metabolism , Serine Endopeptidases/physiology , HEK293 Cells , HeLa Cells , Humans , Proteolysis , Receptors, Interleukin-11/chemistry , Signal Transduction/physiology
10.
Mol Cell ; 56(5): 630-40, 2014 Dec 04.
Article in English | MEDLINE | ID: mdl-25454947

ABSTRACT

Proteolysis by aspartyl intramembrane proteases such as presenilin and signal peptide peptidase (SPP) underlies many cellular processes in health and disease. Saccharomyces cerevisiae encodes a homolog that we named yeast presenilin fold 1 (Ypf1), which we verify to be an SPP-type protease that localizes to the endoplasmic reticulum (ER). Our work shows that Ypf1 functionally interacts with the ER-associated degradation (ERAD) factors Dfm1 and Doa10 to regulate the abundance of nutrient transporters by degradation. We demonstrate how this noncanonical branch of the ERAD pathway, which we termed "ERAD regulatory" (ERAD-R), responds to ligand-mediated sensing as a trigger. More generally, we show that Ypf1-mediated posttranslational regulation of plasma membrane transporters is indispensible for early sensing and adaptation to nutrient depletion. The combination of systematic analysis alongside mechanistic details uncovers a broad role of intramembrane proteolysis in regulating secretome dynamics.


Subject(s)
Endoplasmic Reticulum/metabolism , Peptide Hydrolases/genetics , Peptide Hydrolases/metabolism , Saccharomyces cerevisiae Proteins/genetics , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae/enzymology , Cell Membrane/metabolism , Endoplasmic Reticulum-Associated Degradation , Gene Expression Regulation, Fungal , Membrane Proteins/metabolism , Membrane Transport Proteins/metabolism , Phylogeny , Saccharomyces cerevisiae/physiology , Sequence Alignment , Ubiquitin-Protein Ligases/metabolism , Zinc/metabolism
11.
J Cell Sci ; 132(16)2019 08 15.
Article in English | MEDLINE | ID: mdl-31416853

ABSTRACT

Over the last two decades, a group of unusual proteases, so-called intramembrane proteases, have become increasingly recognized for their unique ability to cleave peptide bonds within cellular membranes. They are found in all kingdoms of life and fulfil versatile functions ranging from protein maturation, to activation of signalling molecules, to protein degradation. In this Cell Science at a Glance article and the accompanying poster, we focus on intramembrane proteases in mammalian cells. By comparing intramembrane proteases in different cellular organelles, we set out to review their functions within the context of the roles of individual cellular compartments. Additionally, we exemplify their mode of action in relation to known substrates by distinguishing cleavage events that promote degradation of substrate from those that release active domains from the membrane bilayer.


Subject(s)
Cell Membrane/metabolism , Membrane Proteins/metabolism , Peptide Hydrolases/metabolism , Proteolysis , Animals , Humans
12.
Nat Methods ; 15(8): 598-600, 2018 08.
Article in English | MEDLINE | ID: mdl-29988096

ABSTRACT

Here we describe a C-SWAT library for high-throughput tagging of Saccharomyces cerevisiae open reading frames (ORFs). In 5,661 strains, we inserted an acceptor module after each ORF that can be efficiently replaced with tags or regulatory elements. We validated the library with targeted sequencing and tagged the proteome with bright fluorescent proteins to quantify the effect of heterologous transcription terminators on protein expression and to localize previously undetected proteins.


Subject(s)
Genome, Fungal , Genomic Library , Saccharomyces cerevisiae/genetics , DNA, Fungal/genetics , High-Throughput Nucleotide Sequencing , Open Reading Frames , Proteome/genetics , Proteomics , Saccharomyces cerevisiae Proteins/genetics , Sequence Analysis, DNA , Sequence Tagged Sites
13.
EMBO Rep ; 20(3)2019 03.
Article in English | MEDLINE | ID: mdl-30733280

ABSTRACT

Signal peptide peptidase (SPP) and the four homologous SPP-like (SPPL) proteases constitute a family of intramembrane aspartyl proteases with selectivity for type II-oriented transmembrane segments. Here, we analyse the physiological function of the orphan protease SPPL2c, previously considered to represent a non-expressed pseudogene. We demonstrate proteolytic activity of SPPL2c towards selected tail-anchored proteins. Despite shared ER localisation, SPPL2c and SPP exhibit distinct, though partially overlapping substrate spectra and inhibitory profiles, and are organised in different high molecular weight complexes. Interestingly, SPPL2c is specifically expressed in murine and human testis where it is primarily localised in spermatids. In mice, SPPL2c deficiency leads to a partial loss of elongated spermatids and reduced motility of mature spermatozoa, but preserved fertility. However, matings of male and female SPPL2c-/- mice exhibit reduced litter sizes. Using proteomics we identify the sarco/endoplasmic reticulum Ca2+-ATPase (SERCA2)-regulating protein phospholamban (PLN) as a physiological SPPL2c substrate. Accumulation of PLN correlates with a decrease in intracellular Ca2+ levels in elongated spermatids that likely contribute to the compromised male germ cell differentiation and function of SPPL2c-/- mice.


Subject(s)
Aspartic Acid Endopeptidases/metabolism , Calcium-Binding Proteins/metabolism , Cell Membrane/enzymology , Germ Cells/metabolism , Membrane Proteins/metabolism , Amino Acid Sequence , Animals , Aspartic Acid Endopeptidases/chemistry , Calcium/metabolism , Endoplasmic Reticulum/metabolism , Female , HEK293 Cells , HeLa Cells , Homeostasis , Humans , Male , Membrane Proteins/chemistry , Mice , Organ Specificity , Spermatids/metabolism , Substrate Specificity , Testis/enzymology
14.
J Biol Chem ; 294(8): 2786-2800, 2019 02 22.
Article in English | MEDLINE | ID: mdl-30578301

ABSTRACT

The endoplasmic reticulum (ER), as a multifunctional organelle, plays crucial roles in lipid biosynthesis and calcium homeostasis as well as the synthesis and folding of secretory and membrane proteins. Therefore, it is of high importance to maintain ER homeostasis and to adapt ER function and morphology to cellular needs. Here, we show that signal peptide peptidase (SPP) modulates the ER shape through degradation of morphogenic proteins. Elevating SPP activity induces rapid rearrangement of the ER and formation of dynamic ER clusters. Inhibition of SPP activity rescues the phenotype without the need for new protein synthesis, and this rescue depends on a pre-existing pool of proteins in the Golgi. With the help of organelle proteomics, we identified certain membrane proteins to be diminished upon SPP expression and further show that the observed morphology changes depend on SPP-mediated cleavage of ER morphogenic proteins, including the SNARE protein syntaxin-18. Thus, we suggest that SPP-mediated protein abundance control by a regulatory branch of ER-associated degradation (ERAD-R) has a role in shaping the early secretory pathway.


Subject(s)
Aspartic Acid Endopeptidases/metabolism , Endoplasmic Reticulum-Associated Degradation , Endoplasmic Reticulum/metabolism , Golgi Apparatus/metabolism , Organelles/metabolism , Qa-SNARE Proteins/metabolism , HEK293 Cells , Humans , Proteolysis , Proteomics
15.
Mol Cell ; 47(4): 558-69, 2012 Aug 24.
Article in English | MEDLINE | ID: mdl-22795130

ABSTRACT

The ER-associated degradation (ERAD) pathway serves as an important cellular safeguard by directing incorrectly folded and unassembled proteins from the ER to the proteasome. Still, however, little is known about the components mediating ERAD of membrane proteins. Here we show that the evolutionary conserved rhomboid family protein RHBDL4 is a ubiquitin-dependent ER-resident intramembrane protease that is upregulated upon ER stress. RHBDL4 cleaves single-spanning and polytopic membrane proteins with unstable transmembrane helices, leading to their degradation by the canonical ERAD machinery. RHBDL4 specifically binds the AAA+-ATPase p97, suggesting that proteolytic processing and dislocation into the cytosol are functionally linked. The phylogenetic relationship between rhomboids and the ERAD factor derlin suggests that substrates for intramembrane proteolysis and protein dislocation are recruited by a shared mechanism.


Subject(s)
Endoplasmic Reticulum-Associated Degradation/physiology , Endoplasmic Reticulum/metabolism , Membrane Proteins/metabolism , Peptide Hydrolases/metabolism , Ubiquitin/metabolism , Adenosine Triphosphatases/genetics , Adenosine Triphosphatases/metabolism , Amino Acid Sequence , Cells, Cultured , Cytosol/metabolism , Endoplasmic Reticulum/genetics , Endoplasmic Reticulum-Associated Degradation/genetics , HEK293 Cells , Humans , Membrane Proteins/genetics , Molecular Sequence Data , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Peptide Hydrolases/genetics , Phylogeny , Proteasome Endopeptidase Complex/metabolism , Protein Binding , Protein Structure, Tertiary , Protein Transport , Proteolysis , Receptors, Antigen, T-Cell/genetics , Receptors, Antigen, T-Cell/metabolism , Stress, Physiological/genetics , Stress, Physiological/physiology , Ubiquitin/genetics
16.
Semin Cell Dev Biol ; 60: 29-37, 2016 12.
Article in English | MEDLINE | ID: mdl-27378062

ABSTRACT

Rhomboids, proteases containing an unusual membrane-integral serine protease active site, were first identified in Drosophila, where they fulfill an essential role in epidermal growth factor receptor signaling, by cleaving membrane-tethered growth factor precursors. It has recently become apparent that eukaryotic genomes harbor conserved catalytically inactive rhomboid protease homologs, including derlins and iRhoms. Here we highlight how loss of proteolytic activity was followed in evolution by impressive functional diversification, enabling these pseudoproteases to fulfill crucial roles within the secretory pathway, including protein degradation, trafficking regulation, and inflammatory signaling. We distil the current understanding of the roles of rhomboid pseudoproteases in development and disease. Finally, we address mechanistically how versatile features of proteolytically active rhomboids have been elaborated to serve the sophisticated functions of their pseudoprotease cousins. By comparing functional and structural clues, we highlight common principles shared by the rhomboid superfamily, and make mechanistic predictions.


Subject(s)
Disease , Health , Mitochondrial Proteins/metabolism , Animals , Humans , Models, Biological , Peptide Hydrolases/metabolism , Protein Transport
17.
EMBO J ; 33(21): 2492-506, 2014 Nov 03.
Article in English | MEDLINE | ID: mdl-25239945

ABSTRACT

Signal peptide peptidase (SPP) catalyzes intramembrane proteolysis of signal peptides at the endoplasmic reticulum (ER), but has also been suggested to play a role in ER-associated degradation (ERAD). Here, we show that SPP forms a complex with the ERAD factor Derlin1 and the E3 ubiquitin ligase TRC8 to cleave the unfolded protein response (UPR) regulator XBP1u. Cleavage occurs within a so far unrecognized type II transmembrane domain, which renders XBP1u as an SPP substrate through specific sequence features. Additionally, Derlin1 acts in the complex as a substrate receptor by recognizing the luminal tail of XBP1u. Remarkably, this interaction of Derlin1 with XBP1u obviates the need for ectodomain shedding prior to SPP cleavage, commonly required for intramembrane cuts. Furthermore, we show that XBP1u inhibits the UPR transcription factor XBP1s by targeting it toward proteasomal degradation. Thus, we identify an ERAD complex that controls the abundance of XBP1u and thereby tunes signaling through the UPR.


Subject(s)
DNA-Binding Proteins/metabolism , Endoplasmic Reticulum-Associated Degradation/physiology , Membrane Proteins/metabolism , Proteasome Endopeptidase Complex/metabolism , Proteolysis , Serine Endopeptidases/metabolism , Transcription Factors/metabolism , DNA-Binding Proteins/genetics , HEK293 Cells , Humans , Membrane Proteins/genetics , Proteasome Endopeptidase Complex/genetics , Receptors, Cell Surface/genetics , Receptors, Cell Surface/metabolism , Regulatory Factor X Transcription Factors , Serine Endopeptidases/genetics , Transcription Factors/genetics , X-Box Binding Protein 1
18.
Biochim Biophys Acta ; 1864(10): 1363-71, 2016 10.
Article in English | MEDLINE | ID: mdl-27426920

ABSTRACT

We describe in detail the usage of leucine metabolic labelling in yeast in order to monitor quantitative proteome alterations, e.g. upon removal of a protease. Since laboratory yeast strains are typically leucine auxotroph, metabolic labelling with trideuterated leucine (d3-leucine) is a straightforward, cost-effective, and ubiquitously applicable strategy for quantitative proteomic studies, similar to the widely used arginine/lysine metabolic labelling method for mammalian cells. We showcase the usage of advanced peptide quantification using the FeatureFinderMultiplex algorithm (part of the OpenMS software package) for robust and reliable quantification. Furthermore, we present an OpenMS bioinformatics data analysis workflow that combines accurate quantification with high proteome coverage. In order to enable visualization, peptide-mapping, and sharing of quantitative proteomic data, especially for membrane-spanning and cell-surface proteins, we further developed the web-application Proteator (http://proteator.appspot.com). Due to its simplicity and robustness, we expect metabolic leucine labelling in yeast to be of great interest to the research community. As an exemplary application, we show the identification of the copper transporter Ctr1 as a putative substrate of the ER-intramembrane protease Ypf1 by yeast membrane proteomics using d3-leucine isotopic labelling.


Subject(s)
Endoplasmic Reticulum/metabolism , Leucine/metabolism , Membrane Proteins/metabolism , Membranes/metabolism , Peptide Hydrolases/metabolism , Proteome/metabolism , Yeasts/metabolism , Computational Biology/methods , Fungal Proteins/metabolism , Isotope Labeling/methods , Peptide Mapping/methods , Peptides/metabolism , Proteomics/methods
19.
Biochim Biophys Acta ; 1828(12): 2840-8, 2013 Dec.
Article in English | MEDLINE | ID: mdl-23562403

ABSTRACT

From proteases that cleave peptide bonds in the plane of the membrane, rhomboids have evolved into a heterogeneous superfamily with a wide range of different mechanistic properties. In mammals 14 family members have been annotated based on a shared conserved membrane-integral rhomboid core domain, including intramembrane serine proteases and diverse proteolytically inactive homologues. While the function of rhomboid proteases is the proteolytic release of membrane-tethered factors, rhomboid pseudoproteases including iRhoms and derlins interact with their clients without cleaving them. It has become evident that specific recognition of membrane protein substrates and clients by the rhomboid fold reflects a spectrum of cellular functions ranging from growth factor activation, trafficking control to membrane protein degradation. This review summarizes recent progress on rhomboid family proteins in the mammalian secretory pathway and raises the question whether they can be seen as new drug targets for inflammatory diseases and cancer. This article is part of a special issue entitled: Intramembrane Proteases.


Subject(s)
Inflammation/metabolism , Membrane Proteins/metabolism , Neoplasms/metabolism , Serine Endopeptidases/metabolism , Serine Proteases/metabolism , Signal Transduction , ADAM Proteins/genetics , ADAM Proteins/metabolism , ADAM17 Protein , Animals , ErbB Receptors/genetics , ErbB Receptors/metabolism , Gene Expression Regulation , Humans , Membrane Proteins/chemistry , Membrane Proteins/genetics , Phylogeny , Proteolysis , Serine Endopeptidases/chemistry , Serine Endopeptidases/genetics , Serine Proteases/chemistry , Serine Proteases/genetics , Substrate Specificity
20.
J Immunol ; 188(6): 2794-804, 2012 Mar 15.
Article in English | MEDLINE | ID: mdl-22345649

ABSTRACT

Human CMV (HCMV)-encoded NK cell-evasion functions include an MHC class I homolog (UL18) with high affinity for the leukocyte inhibitory receptor-1 (CD85j, ILT2, or LILRB1) and a signal peptide (SP(UL40)) that acts by upregulating cell surface expression of HLA-E. Detailed characterization of SP(UL40) revealed that the N-terminal 14 aa residues bestowed TAP-independent upregulation of HLA-E, whereas C region sequences delayed processing of SP(UL40) by a signal peptide peptidase-type intramembrane protease. Most significantly, the consensus HLA-E-binding epitope within SP(UL40) was shown to promote cell surface expression of both HLA-E and gpUL18. UL40 was found to possess two transcription start sites, with utilization of the downstream site resulting in translation being initiated within the HLA-E-binding epitope (P2). Remarkably, this truncated SP(UL40) was functional and retained the capacity to upregulate gpUL18 but not HLA-E. Thus, our findings identify an elegant mechanism by which an HCMV signal peptide differentially regulates two distinct NK cell-evasion pathways. Moreover, we describe a natural SP(UL40) mutant that provides a clear example of an HCMV clinical virus with a defect in an NK cell-evasion function and exemplifies issues that confront the virus when adapting to immunogenetic diversity in the host.


Subject(s)
Capsid Proteins/metabolism , Histocompatibility Antigens Class I/metabolism , Immune Evasion/immunology , Killer Cells, Natural/immunology , Viral Proteins/metabolism , Amino Acid Sequence , Blotting, Northern , Blotting, Western , Capsid Proteins/genetics , Capsid Proteins/immunology , Cell Membrane/immunology , Cell Membrane/metabolism , Cell Separation , Cytomegalovirus/genetics , Cytomegalovirus/immunology , Cytomegalovirus/metabolism , Cytomegalovirus Infections , Flow Cytometry , Histocompatibility Antigens Class I/genetics , Histocompatibility Antigens Class I/immunology , Humans , Killer Cells, Natural/metabolism , Molecular Sequence Data , Reverse Transcriptase Polymerase Chain Reaction , Viral Proteins/genetics , Viral Proteins/immunology , HLA-E Antigens
SELECTION OF CITATIONS
SEARCH DETAIL