Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 93
Filtrar
1.
J Cell Sci ; 135(9)2022 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-35543156

RESUMO

The heterotrimeric BAG6 complex coordinates the direct handover of newly synthesised tail-anchored (TA) membrane proteins from an SGTA-bound preloading complex to the endoplasmic reticulum (ER) delivery component TRC40. In contrast, defective precursors, including aberrant TA proteins, form a stable complex with this cytosolic protein quality control factor, enabling such clients to be either productively re-routed or selectively degraded. We identify the mitochondrial antiviral-signalling protein (MAVS) as an endogenous TA client of both SGTA and the BAG6 complex. Our data suggest that the BAG6 complex binds to a cytosolic pool of MAVS before its misinsertion into the ER membrane, from where it can subsequently be removed via ATP13A1-mediated dislocation. This BAG6-associated fraction of MAVS is dynamic and responds to the activation of an innate immune response, suggesting that BAG6 may modulate the pool of MAVS that is available for coordinating the cellular response to viral infection.


Assuntos
Retículo Endoplasmático , Chaperonas Moleculares , Antivirais , Citosol/metabolismo , Retículo Endoplasmático/metabolismo , Humanos , Proteínas de Membrana/metabolismo , Chaperonas Moleculares/metabolismo
2.
J Cell Sci ; 135(5)2022 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-34558621

RESUMO

Membrane proteins destined for lipid droplets (LDs), a major intracellular storage site for neutral lipids, are inserted into the endoplasmic reticulum (ER) and then trafficked to LDs where they reside in a hairpin loop conformation. Here, we show that LD membrane proteins can be delivered to the ER either co- or post-translationally and that their membrane-embedded region specifies pathway selection. The co-translational route for LD membrane protein biogenesis is insensitive to a small molecule inhibitor of the Sec61 translocon, Ipomoeassin F, and instead relies on the ER membrane protein complex (EMC) for membrane insertion. This route may even result in a transient exposure of the short N termini of some LD membrane proteins to the ER lumen, followed by putative topological rearrangements that would enable their transmembrane segment to form a hairpin loop and N termini to face the cytosol. Our study reveals an unexpected complexity to LD membrane protein biogenesis and identifies a role for the EMC during their co-translational insertion into the ER.


Assuntos
Gotículas Lipídicas , Proteínas de Membrana , Citosol/metabolismo , Retículo Endoplasmático/genética , Retículo Endoplasmático/metabolismo , Gotículas Lipídicas/metabolismo , Proteínas de Membrana/genética , Proteínas de Membrana/metabolismo , Canais de Translocação SEC/genética
3.
J Cell Sci ; 134(4)2021 02 19.
Artigo em Inglês | MEDLINE | ID: mdl-33468620

RESUMO

In order to produce proteins essential for their propagation, many pathogenic human viruses, including SARS-CoV-2, the causative agent of COVID-19 respiratory disease, commandeer host biosynthetic machineries and mechanisms. Three major structural proteins, the spike, envelope and membrane proteins, are amongst several SARS-CoV-2 components synthesised at the endoplasmic reticulum (ER) of infected human cells prior to the assembly of new viral particles. Hence, the inhibition of membrane protein synthesis at the ER is an attractive strategy for reducing the pathogenicity of SARS-CoV-2 and other obligate viral pathogens. Using an in vitro system, we demonstrate that the small molecule inhibitor ipomoeassin F (Ipom-F) potently blocks the Sec61-mediated ER membrane translocation and/or insertion of three therapeutic protein targets for SARS-CoV-2 infection; the viral spike and ORF8 proteins together with angiotensin-converting enzyme 2, the host cell plasma membrane receptor. Our findings highlight the potential for using ER protein translocation inhibitors such as Ipom-F as host-targeting, broad-spectrum antiviral agents.This article has an associated First Person interview with the first author of the paper.


Assuntos
Tratamento Farmacológico da COVID-19 , Glicoconjugados/farmacologia , SARS-CoV-2/efeitos dos fármacos , Glicoproteína da Espícula de Coronavírus/efeitos dos fármacos , Antivirais/farmacologia , COVID-19/virologia , Humanos , SARS-CoV-2/patogenicidade , Glicoproteína da Espícula de Coronavírus/metabolismo , Internalização do Vírus/efeitos dos fármacos
4.
PLoS Biol ; 18(9): e3000874, 2020 09.
Artigo em Inglês | MEDLINE | ID: mdl-32997663

RESUMO

Small membrane proteins represent a largely unexplored yet abundant class of proteins in pro- and eukaryotes. They essentially consist of a single transmembrane domain and are associated with stress response mechanisms in bacteria. How these proteins are inserted into the bacterial membrane is unknown. Our study revealed that in Escherichia coli, the 27-amino-acid-long model protein YohP is recognized by the signal recognition particle (SRP), as indicated by in vivo and in vitro site-directed cross-linking. Cross-links to SRP were also observed for a second small membrane protein, the 33-amino-acid-long YkgR. However, in contrast to the canonical cotranslational recognition by SRP, SRP was found to bind to YohP posttranslationally. In vitro protein transport assays in the presence of a SecY inhibitor and proteoliposome studies demonstrated that SRP and its receptor FtsY are essential for the posttranslational membrane insertion of YohP by either the SecYEG translocon or by the YidC insertase. Furthermore, our data showed that the yohP mRNA localized preferentially and translation-independently to the bacterial membrane in vivo. In summary, our data revealed that YohP engages an unique SRP-dependent posttranslational insertion pathway that is likely preceded by an mRNA targeting step. This further highlights the enormous plasticity of bacterial protein transport machineries.


Assuntos
Proteínas de Membrana/metabolismo , Processamento de Proteína Pós-Traducional , Partícula de Reconhecimento de Sinal/metabolismo , Sequência de Aminoácidos , Escherichia coli/metabolismo , Proteínas de Escherichia coli/química , Proteínas de Escherichia coli/genética , Proteínas de Escherichia coli/metabolismo , Modelos Biológicos , Ligação Proteica , Biossíntese de Proteínas , Transporte Proteico , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Canais de Translocação SEC/metabolismo
5.
Mol Microbiol ; 115(1): 28-40, 2021 01.
Artigo em Inglês | MEDLINE | ID: mdl-32798330

RESUMO

Eeyarestatin 1 (ES1) is an inhibitor of endoplasmic reticulum (ER) associated protein degradation, Sec61-dependent Ca2+ homeostasis and protein translocation into the ER. Recently, evidence was presented showing that a smaller analog of ES1, ES24, targets the Sec61-translocon, and captures it in an open conformation that is translocation-incompetent. We now show that ES24 impairs protein secretion and membrane protein insertion in Escherichia coli via the homologous SecYEG-translocon. Transcriptomic analysis suggested that ES24 has a complex mode of action, probably involving multiple targets. Interestingly, ES24 shows antibacterial activity toward clinically relevant strains. Furthermore, the antibacterial activity of ES24 is equivalent to or better than that of nitrofurantoin, a known antibiotic that, although structurally similar to ES24, does not interfere with SecYEG-dependent protein trafficking. Like nitrofurantoin, we find that ES24 requires activation by the NfsA and NfsB nitroreductases, suggesting that the formation of highly reactive nitroso intermediates is essential for target inactivation in vivo.


Assuntos
Hidrazonas/farmacologia , Hidroxiureia/análogos & derivados , Canais de Translocação SEC/metabolismo , Antibacterianos/metabolismo , Retículo Endoplasmático/metabolismo , Escherichia coli/metabolismo , Proteínas de Escherichia coli/metabolismo , Hidrazonas/química , Hidroxiureia/química , Hidroxiureia/farmacologia , Proteínas de Membrana/metabolismo , Nitrorredutases/metabolismo , Transporte Proteico/efeitos dos fármacos , Canais de Translocação SEC/efeitos dos fármacos
7.
EMBO Rep ; 21(5): e48835, 2020 05 06.
Artigo em Inglês | MEDLINE | ID: mdl-32216016

RESUMO

The endoplasmic reticulum (ER) is a major site for membrane protein synthesis in eukaryotes. The majority of integral membrane proteins are delivered to the ER membrane via the co-translational, signal recognition particle (SRP)-dependent route. However, tail-anchored proteins employ an alternative, post-translational route(s) that relies on distinct factors such as a cytosolic protein quality control component, SGTA. We now show that SGTA is selectively recruited to ribosomes synthesising a diverse range of membrane proteins, suggesting that its biosynthetic client base also includes precursors on the co-translational ER delivery pathway. Strikingly, SGTA is recruited to nascent membrane proteins before their transmembrane domain emerges from the ribosome. Hence, SGTA is ideally placed to capture these aggregation prone regions shortly after their synthesis. For nascent membrane proteins on the co-translational pathway, SGTA complements the role of SRP by reducing the co-translational ubiquitination of clients with multiple hydrophobic signal sequences. On this basis, we propose that SGTA acts to mask specific transmembrane domains located in complex membrane proteins until they can engage the ER translocon and become membrane inserted.


Assuntos
Chaperonas Moleculares , Precursores de Proteínas , Retículo Endoplasmático/metabolismo , Humanos , Chaperonas Moleculares/metabolismo , Precursores de Proteínas/metabolismo , Ribossomos/genética , Ribossomos/metabolismo , Partícula de Reconhecimento de Sinal/genética
8.
Molecules ; 27(14)2022 Jul 10.
Artigo em Inglês | MEDLINE | ID: mdl-35889292

RESUMO

The plant-derived macrocyclic resin glycoside ipomoeassin F (Ipom-F) binds to Sec61α and significantly disrupts multiple aspects of Sec61-mediated protein biogenesis at the endoplasmic reticulum, ultimately leading to cell death. However, extensive assessment of Ipom-F as a molecular tool and a therapeutic lead is hampered by its limited production scale, largely caused by intramolecular assembly of the macrocyclic ring. Here, using in vitro and/or in cellula biological assays to explore the first series of ring-opened analogues for the ipomoeassins, and indeed all resin glycosides, we provide clear evidence that macrocyclic integrity is not required for the cytotoxic inhibition of Sec61-dependent protein translocation by Ipom-F. Furthermore, our modeling suggests that open-chain analogues of Ipom-F can interact with multiple sites on the Sec61α subunit, most likely located at a previously identified binding site for mycolactone and/or the so-called lateral gate. Subsequent in silico-aided design led to the discovery of the stereochemically simplified analogue 3 as a potent, alternative lead compound that could be synthesized much more efficiently than Ipom-F and will accelerate future ipomoeassin research in chemical biology and drug discovery. Our work may also inspire further exploration of ring-opened analogues of other resin glycosides.


Assuntos
Antineoplásicos , Glicoconjugados , Antineoplásicos/química , Glicoconjugados/química , Glicosídeos/farmacologia , Canais de Translocação SEC/metabolismo
9.
Nat Rev Mol Cell Biol ; 10(4): 255-64, 2009 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-19305415

RESUMO

Correct protein function depends on delivery to the appropriate cellular or subcellular compartment. Following the initiation of protein synthesis in the cytosol, many bacterial and eukaryotic proteins must be integrated into or transported across a membrane to reach their site of function. Whereas in the post-translational delivery pathway ATP-dependent factors bind to completed polypeptides and chaperone them until membrane translocation is initiated, a GTP-dependent co-translational pathway operates to couple ongoing protein synthesis to membrane transport. These distinct pathways provide different solutions for the maintenance of proteins in a state that is competent for membrane translocation and their delivery for export from the cytosol.


Assuntos
Citosol/metabolismo , Células Procarióticas/metabolismo , Transporte Proteico , Animais , Células Eucarióticas/metabolismo , Biossíntese de Proteínas , Partícula de Reconhecimento de Sinal/metabolismo
10.
J Org Chem ; 85(24): 16226-16235, 2020 12 18.
Artigo em Inglês | MEDLINE | ID: mdl-33264019

RESUMO

Two new ring-size-varying analogues (2 and 3) of ipomoeassin F were synthesized and evaluated. Improved cytotoxicity (IC50: from 1.8 nM) and in vitro protein translocation inhibition (IC50: 35 nM) derived from ring expansion imply that the binding pocket of Sec61α (isoform 1) can accommodate further structural modifications, likely in the fatty acid portion. Streamlined preparation of the key diol intermediate 5 enabled gram-scale production, allowing us to establish that ipomoeassin F is biologically active in vivo (MTD: ∼3 mg/kg).


Assuntos
Glicoconjugados , Ensaios de Seleção de Medicamentos Antitumorais , Estrutura Molecular , Relação Estrutura-Atividade
11.
Glycobiology ; 29(7): 530-542, 2019 07 01.
Artigo em Inglês | MEDLINE | ID: mdl-30976784

RESUMO

The endoplasmic reticulum (ER) contains both α-glucosidases and α-mannosidases which process the N-linked oligosaccharides of newly synthesized glycoproteins and thereby facilitate polypeptide folding and glycoprotein quality control. By acting as structural mimetics, iminosugars can selectively inhibit these ER localized α-glycosidases, preventing N-glycan trimming and providing a molecular basis for their therapeutic applications. In this study, we investigate the effects of a panel of nine iminosugars on the actions of ER luminal α-glucosidase I and α-glucosidase II. Using ER microsomes to recapitulate authentic protein N-glycosylation and oligosaccharide processing, we identify five iminosugars that selectively inhibit N-glycan trimming. Comparison of their inhibitory activities in ER microsomes against their effects on purified ER α-glucosidase II, suggests that 3,7a-diepi-alexine acts as a selective inhibitor of ER α-glucosidase I. The other active iminosugars all inhibit α-glucosidase II and, having identified 1,4-dideoxy-1,4-imino-D-arabinitol (DAB) as the most effective of these compounds, we use in silico modeling to understand the molecular basis for this enhanced activity. Taken together, our work identifies the C-3 substituted pyrrolizidines casuarine and 3,7a-diepi-alexine as promising "second-generation" iminosugar inhibitors.


Assuntos
Arabinose/farmacologia , Retículo Endoplasmático/enzimologia , Inibidores de Glicosídeo Hidrolases/farmacologia , Imino Furanoses/farmacologia , Alcaloides de Pirrolizidina/farmacologia , Álcoois Açúcares/farmacologia , alfa-Glucosidases/metabolismo , Animais , Arabinose/química , Cães , Inibidores de Glicosídeo Hidrolases/química , Humanos , Imino Furanoses/química , Camundongos , Microssomos/efeitos dos fármacos , Microssomos/metabolismo , Alcaloides de Pirrolizidina/química , Álcoois Açúcares/química
12.
J Am Chem Soc ; 141(21): 8450-8461, 2019 05 29.
Artigo em Inglês | MEDLINE | ID: mdl-31059257

RESUMO

Ipomoeassin F is a potent natural cytotoxin that inhibits growth of many tumor cell lines with single-digit nanomolar potency. However, its biological and pharmacological properties have remained largely unexplored. Building upon our earlier achievements in total synthesis and medicinal chemistry, we used chemical proteomics to identify Sec61α (protein transport protein Sec61 subunit alpha isoform 1), the pore-forming subunit of the Sec61 protein translocon, as a direct binding partner of ipomoeassin F in living cells. The interaction is specific and strong enough to survive lysis conditions, enabling a biotin analogue of ipomoeassin F to pull down Sec61α from live cells, yet it is also reversible, as judged by several experiments including fluorescent streptavidin staining, delayed competition in affinity pulldown, and inhibition of TNF biogenesis after washout. Sec61α forms the central subunit of the ER protein translocation complex, and the binding of ipomoeassin F results in a substantial, yet selective, inhibition of protein translocation in vitro and a broad ranging inhibition of protein secretion in live cells. Lastly, the unique resistance profile demonstrated by specific amino acid single-point mutations in Sec61α provides compelling evidence that Sec61α is the primary molecular target of ipomoeassin F and strongly suggests that the binding of this natural product to Sec61α is distinctive. Therefore, ipomoeassin F represents the first plant-derived, carbohydrate-based member of a novel structural class that offers new opportunities to explore Sec61α function and to further investigate its potential as a therapeutic target for drug discovery.


Assuntos
Glicoconjugados/farmacologia , Canais de Translocação SEC/antagonistas & inibidores , Sítios de Ligação/efeitos dos fármacos , Glicoconjugados/química , Humanos , Estrutura Molecular , Transporte Proteico/efeitos dos fármacos , Canais de Translocação SEC/metabolismo
13.
J Cell Sci ; 130(7): 1307-1320, 2017 04 01.
Artigo em Inglês | MEDLINE | ID: mdl-28219954

RESUMO

Mycolactone is the exotoxin virulence factor produced by Mycobacterium ulcerans, the pathogen responsible for Buruli ulcer. The skin lesions and immunosuppression that are characteristic of this disease result from the action of mycolactone, which targets the Sec61 complex and inhibits the co-translational translocation of secretory proteins into the endoplasmic reticulum. In this study, we investigate the effect of mycolactone on the Sec61-dependent biogenesis of different classes of transmembrane protein (TMP). Our data suggest that the effect of mycolactone on TMP biogenesis depends on how the nascent chain initially engages the Sec61 complex. For example, the translocation of TMP lumenal domains driven by an N-terminal cleavable signal sequence is efficiently inhibited by mycolactone. In contrast, the effect of mycolactone on protein translocation that is driven solely by a non-cleavable signal anchor/transmembrane domain depends on which flanking region is translocated. For example, while translocation of the region N-terminal to a signal anchor/transmembrane domain is refractive to mycolactone, C-terminal translocation is efficiently inhibited. Our findings highlight the diversity of Sec61-dependent translocation and provide a molecular basis for understanding the effect of mycolactone on the biogenesis of different TMPs.


Assuntos
Macrolídeos/farmacologia , Proteínas de Membrana/metabolismo , Biossíntese de Proteínas/efeitos dos fármacos , Canais de Translocação SEC/metabolismo , Citosol/efeitos dos fármacos , Citosol/metabolismo , Retículo Endoplasmático/efeitos dos fármacos , Retículo Endoplasmático/metabolismo , Humanos , Proteínas de Membrana/química , Domínios Proteicos , Molécula 1 de Adesão de Célula Vascular/metabolismo
14.
J Cell Sci ; 130(22): 3851-3861, 2017 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-29021347

RESUMO

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


Assuntos
ATPases Transportadoras de Arsenito/genética , Biossíntese de Proteínas , ATPases Transportadoras de Arsenito/metabolismo , Vias Biossintéticas , Células HeLa , Humanos , Proteínas de Membrana/biossíntese , Proteínas de Membrana/genética , Transporte Proteico
15.
Biol Cell ; 110(11): 237-248, 2018 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-30055020

RESUMO

Infection with Mycobacterium ulcerans results in a necrotising skin disease known as a Buruli ulcer, the pathology of which is directly linked to the bacterial production of the toxin mycolactone. Recent studies have identified the protein translocation machinery of the endoplasmic reticulum (ER) membrane as the primary cellular target of mycolactone, and shown that the toxin binds to the core subunit of the Sec61 complex. Mycolactone binding strongly inhibits the capacity of the Sec61 translocon to transport newly synthesised membrane and secretory proteins into and across the ER membrane. Since the ER acts as the entry point for the mammalian secretory pathway, and hence regulates initial access to the entire endomembrane system, mycolactone-treated cells have a reduced ability to produce a range of proteins including secretory cytokines and plasma membrane receptors. The global effect of this molecular blockade of protein translocation at the ER is that the host is unable to mount an effective immune response to the underlying mycobacterial infection. Prolonged exposure to mycolactone is normally cytotoxic, since it triggers stress responses activating the transcription factor ATF4 and ultimately inducing apoptosis.


Assuntos
Úlcera de Buruli/etiologia , Úlcera de Buruli/microbiologia , Macrolídeos/toxicidade , Mycobacterium ulcerans/patogenicidade , Canais de Translocação SEC/antagonistas & inibidores , Animais , Retículo Endoplasmático/efeitos dos fármacos , Retículo Endoplasmático/metabolismo , Humanos , Macrolídeos/efeitos adversos , Macrolídeos/química , Modelos Biológicos , Transporte Proteico/efeitos dos fármacos , Canais de Translocação SEC/metabolismo
16.
BMC Biol ; 16(1): 76, 2018 07 11.
Artigo em Inglês | MEDLINE | ID: mdl-29996828

RESUMO

BACKGROUND: Protein quality control mechanisms are essential for cell health and involve delivery of proteins to specific cellular compartments for recycling or degradation. In particular, stray hydrophobic proteins are captured in the aqueous cytosol by a co-chaperone, the small glutamine-rich, tetratricopeptide repeat-containing protein alpha (SGTA), which facilitates the correct targeting of tail-anchored membrane proteins, as well as the sorting of membrane and secretory proteins that mislocalize to the cytosol and endoplasmic reticulum-associated degradation. Full-length SGTA has an unusual elongated dimeric structure that has, until now, evaded detailed structural analysis. The C-terminal region of SGTA plays a key role in binding a broad range of hydrophobic substrates, yet in contrast to the well-characterized N-terminal and TPR domains, there is a lack of structural information on the C-terminal domain. In this study, we present new insights into the conformation and organization of distinct domains of SGTA and show that the C-terminal domain possesses a conserved region essential for substrate processing in vivo. RESULTS: We show that the C-terminal domain region is characterized by α-helical propensity and an intrinsic ability to dimerize independently of the N-terminal domain. Based on the properties of different regions of SGTA that are revealed using cell biology, NMR, SAXS, Native MS, and EPR, we observe that its C-terminal domain can dimerize in the full-length protein and propose that this reflects a closed conformation of the substrate-binding domain. CONCLUSION: Our results provide novel insights into the structural complexity of SGTA and provide a new basis for mechanistic studies of substrate binding and release at the C-terminal region.


Assuntos
Proteínas de Transporte/química , Chaperonas Moleculares/química , Sequência de Aminoácidos , Animais , Células Cultivadas , Humanos , Interações Hidrofóbicas e Hidrofílicas , Imageamento por Ressonância Magnética/métodos , Espectroscopia de Ressonância Magnética/métodos , Ligação Proteica , Domínios Proteicos , Multimerização Proteica , Transporte Proteico , Espalhamento a Baixo Ângulo
17.
J Cell Sci ; 129(7): 1404-15, 2016 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-26869228

RESUMO

The virulence factor mycolactone is responsible for the immunosuppression and tissue necrosis that characterise Buruli ulcer, a disease caused by infection with Mycobacterium ulcerans In this study, we confirm that Sec61, the protein-conducting channel that coordinates entry of secretory proteins into the endoplasmic reticulum, is a primary target of mycolactone, and characterise the nature of its inhibitory effect. We conclude that mycolactone constrains the ribosome-nascent-chain-Sec61 complex, consistent with its broad-ranging perturbation of the co-translational translocation of classical secretory proteins. In contrast, the effect of mycolactone on the post-translational ribosome-independent translocation of short secretory proteins through the Sec61 complex is dependent on both signal sequence hydrophobicity and the translocation competence of the mature domain. Changes to protease sensitivity strongly suggest that mycolactone acts by inducing a conformational change in the pore-forming Sec61α subunit. These findings establish that mycolactone inhibits Sec61-mediated protein translocation and highlight differences between the co- and post-translational routes that the Sec61 complex mediates. We propose that mycolactone also provides a useful tool for further delineating the molecular mechanisms of Sec61-dependent protein translocation.


Assuntos
Úlcera de Buruli/patologia , Macrolídeos/metabolismo , Mycobacterium ulcerans/patogenicidade , Canais de Translocação SEC/antagonistas & inibidores , Canais de Translocação SEC/metabolismo , Animais , Retículo Endoplasmático/metabolismo , Humanos , Transporte Proteico/fisiologia , Ribossomos/metabolismo
18.
J Cell Sci ; 128(17): 3187-96, 2015 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-26169395

RESUMO

Rpn13 is an intrinsic ubiquitin receptor of the 26S proteasome regulatory subunit that facilitates substrate capture prior to degradation. Here we show that the C-terminal region of Rpn13 binds to the tetratricopeptide repeat (TPR) domain of SGTA, a cytosolic factor implicated in the quality control of mislocalised membrane proteins (MLPs). The overexpression of SGTA results in a substantial increase in steady-state MLP levels, consistent with an effect on proteasomal degradation. However, this effect is strongly dependent upon the interaction of SGTA with the proteasomal component Rpn13. Hence, overexpression of the SGTA-binding region of Rpn13 or point mutations within the SGTA TPR domain both inhibit SGTA binding to the proteasome and substantially reduce MLP levels. These findings suggest that SGTA can regulate the access of MLPs to the proteolytic core of the proteasome, implying that a protein quality control cycle that involves SGTA and the BAG6 complex can operate at the 19S regulatory particle. We speculate that the binding of SGTA to Rpn13 enables specific polypeptides to escape proteasomal degradation and/or selectively modulates substrate degradation.


Assuntos
Proteínas de Transporte/metabolismo , Moléculas de Adesão Celular/metabolismo , Glicoproteínas de Membrana/metabolismo , Complexo de Endopeptidases do Proteassoma/metabolismo , Proteólise , Animais , Proteínas de Transporte/genética , Moléculas de Adesão Celular/genética , Células HEK293 , Células HeLa , Humanos , Peptídeos e Proteínas de Sinalização Intracelular , Glicoproteínas de Membrana/genética , Camundongos , Chaperonas Moleculares/genética , Chaperonas Moleculares/metabolismo , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Mutação Puntual , Complexo de Endopeptidases do Proteassoma/genética , Estrutura Terciária de Proteína
19.
Biochem J ; 473(21): 3965-3978, 2016 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-27582497

RESUMO

LITAF (LPS-induced TNF-activating factor) is an endosome-associated integral membrane protein important for multivesicular body sorting. Several mutations in LITAF cause autosomal-dominant Charcot Marie Tooth disease type 1C. These mutations map to a highly conserved C-terminal region, termed the LITAF domain, which includes a 22 residue hydrophobic sequence and flanking cysteine-rich regions that contain peptide motifs found in zinc fingers. Although the LITAF domain is thought to be responsible for membrane integration, the membrane topology of LITAF has not been established. Here, we have investigated whether LITAF is a tail-anchored (TA) membrane-spanning protein or monotopic membrane protein. When translated in vitro, LITAF integrates poorly into ER-derived microsomes compared with Sec61ß, a bona fide TA protein. Furthermore, introduction of N-linked glycosylation reporters shows that neither the N-terminal nor C-terminal domains of LITAF translocate into the ER lumen. Expression in cells of an LITAF construct containing C-terminal glycosylation sites confirms that LITAF is not a TA protein in cells. Finally, an immunofluorescence-based latency assay showed that both the N- and C-termini of LITAF are exposed to the cytoplasm. Recombinant LITAF contains 1 mol/mol zinc, while mutation of predicted zinc-binding residues disrupts LITAF membrane association. Hence, we conclude that LITAF is a monotopic membrane protein whose membrane integration is stabilised by a zinc finger. The related human protein, CDIP1 (cell death involved p53 target 1), displays identical membrane topology, suggesting that this mode of membrane integration is conserved in LITAF family proteins.


Assuntos
Proteínas de Transporte/química , Proteínas de Transporte/metabolismo , Doença de Charcot-Marie-Tooth/metabolismo , Proteínas de Membrana/química , Proteínas de Membrana/metabolismo , Proteínas Nucleares/química , Proteínas Nucleares/metabolismo , Fatores de Transcrição/química , Fatores de Transcrição/metabolismo , Motivos de Aminoácidos , Sequência de Aminoácidos , Proteínas Reguladoras de Apoptose/química , Proteínas Reguladoras de Apoptose/metabolismo , Linhagem Celular , Células HeLa , Humanos , Microssomos/metabolismo , Dados de Sequência Molecular , Transporte Proteico
20.
J Cell Sci ; 127(Pt 13): 2898-909, 2014 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-24806960

RESUMO

BAG6 participates in protein quality control and, here, we address its role in endoplasmic-reticulum-associated degradation (ERAD) by using the polytopic membrane protein OpD, an opsin degron mutant. Both BAG6 knockdown and BAG6 overexpression delay OpD degradation; however, our data suggest that these two perturbations are mechanistically distinct. Hence, BAG6 knockdown correlates with reduced OpD polyubiquitylation, whereas BAG6 overexpression increases the level of polyubiquitylated OpD. The UBL- and BAG-domains of exogenous BAG6 are dispensable for OpD stabilisation and enhanced levels of polyubiquitylated OpD. Thus, although endogenous BAG6 normally promotes OpD degradation, exogenous BAG6 expression delays this process. We speculate that overexpressed BAG6 subunits might associate with the endogenous BAG6 complex, resulting in a dominant-negative effect that inhibits its function. Interestingly, cellular levels of BAG6 also correlate with total steady-state polyubiquitylation, with Rpn10 (officially known as PSMD4) overexpression showing a similar effect. These findings suggest that perturbations of the levels of ubiquitin-binding proteins can impact upon cellular ubiquitin homeostasis. We propose that exogenous BAG6 perturbs the function of the BAG6 complex at a stage subsequent to substrate recognition and polyubiquitylation, most likely the BAG6-dependent delivery of OpD to the proteasome.


Assuntos
Degradação Associada com o Retículo Endoplasmático , Chaperonas Moleculares/metabolismo , Complexo de Endopeptidases do Proteassoma/metabolismo , Ubiquitina/metabolismo , Células HeLa , Humanos , Chaperonas Moleculares/genética , Mutagênese Sítio-Dirigida , Opsinas/genética , Opsinas/metabolismo , Transfecção
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA