Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 24
Filter
Add more filters










Publication year range
1.
Methods Mol Biol ; 1321: 307-22, 2015.
Article in English | MEDLINE | ID: mdl-26082231

ABSTRACT

Some of the most important and interesting molecules in metazoan biology are glycoproteins. The importance of the carbohydrate component of these structures is often revealed by the disease phenotypes that manifest when the biosynthesis of particular glycoforms is disrupted. On the other hand, the presence of large amounts of carbohydrate can often hinder the structural and functional analysis of glycoproteins. There are often good reasons, therefore, for wanting to engineer and predefine the N-glycans present on glycoproteins, e.g., in order to characterize the functions of the glycans or facilitate their subsequent removal. Here, we describe in detail two distinct ways in which to usefully interfere with oligosaccharide processing, one involving the use of specific processing inhibitors, and the other the selection of cell lines mutated at gene loci that control oligosaccharide processing, using cytotoxic lectins. Both approaches have the capacity for controlled, radical alteration of oligosaccharide processing in eukaryotic cells used for heterologous protein expression, and have great utility in the structural analysis of glycoproteins.


Subject(s)
Eukaryotic Cells/metabolism , Glycoproteins/metabolism , Lectins/metabolism , Polysaccharides/metabolism , Animals , Glycosylation , Humans , Oligosaccharides/metabolism
2.
Toxins (Basel) ; 7(1): 49-65, 2015 Jan 09.
Article in English | MEDLINE | ID: mdl-25584427

ABSTRACT

The heterodimeric plant toxin ricin binds exposed galactosyls at the cell surface of target mammalian cells, and, following endocytosis, is transported in vesicular carriers to the endoplasmic reticulum (ER). Subsequently, the cell-binding B chain (RTB) and the catalytic A chain (RTA) are separated reductively, RTA embeds in the ER membrane and then retrotranslocates (or dislocates) across this membrane. The protein conducting channels used by RTA are usually regarded as part of the ER-associated protein degradation system (ERAD) that removes misfolded proteins from the ER for destruction by the cytosolic proteasomes. However, unlike ERAD substrates, cytosolic RTA avoids destruction and folds into a catalytic conformation that inactivates its target ribosomes. Protein synthesis ceases, and subsequently the cells die apoptotically. This raises questions about how this protein avoids the pathways that are normally sanctioned for ER-dislocating substrates. In this review we focus on the molecular events that occur with non-tagged ricin and its isolated subunits at the ER-cytosol interface. This focus reveals that intra-membrane interactions of RTA may control its fate, an area that warrants further investigation.


Subject(s)
Ricin/metabolism , Animals , Cytosol/metabolism , Endoplasmic Reticulum/metabolism , Golgi Apparatus/metabolism , Protein Transport , Ricin/chemistry
3.
Biochem J ; 453(3): 435-45, 2013 Aug 01.
Article in English | MEDLINE | ID: mdl-23617410

ABSTRACT

The plant cytotoxin ricin enters mammalian cells by receptor-mediated endocytosis, undergoing retrograde transport to the ER (endoplasmic reticulum) where its catalytic A chain (RTA) is reductively separated from the holotoxin to enter the cytosol and inactivate ribosomes. The currently accepted model is that the bulk of ER-dislocated RTA is degraded by proteasomes. We show in the present study that the proteasome has a more complex role in ricin intoxication than previously recognized, that the previously reported increase in sensitivity of mammalian cells to ricin in the presence of proteasome inhibitors simply reflects toxicity of the inhibitors themselves, and that RTA is a very poor substrate for proteasomal degradation. Denatured RTA and casein compete for a binding site on the regulatory particle of the 26S proteasome, but their fates differ. Casein is degraded, but the mammalian 26S proteasome AAA (ATPase associated with various cellular activities)-ATPase subunit RPT5 acts as a chaperone that prevents aggregation of denatured RTA and stimulates recovery of catalytic RTA activity in vitro. Furthermore, in vivo, the ATPase activity of Rpt5p is required for maximal toxicity of RTA dislocated from the Saccharomyces cerevisiae ER. The results of the present study implicate RPT5/Rpt5p in the triage of substrates in which either activation (folding) or inactivation (degradation) pathways may be initiated.


Subject(s)
Proteasome Endopeptidase Complex/metabolism , Ricin/metabolism , Animals , Caseins/chemistry , Caseins/metabolism , Cattle , Endoplasmic Reticulum/metabolism , HeLa Cells , Humans , Ricin/chemistry , Saccharomyces cerevisiae/metabolism , Signal Transduction
4.
PLoS One ; 7(7): e41119, 2012.
Article in English | MEDLINE | ID: mdl-22829918

ABSTRACT

BACKGROUND: Escherichia coli Shiga-like toxin 1 normally traffics to the endoplasmic reticulum (ER) in sensitive mammalian cells from where the catalytic A chain (SLTxA1) dislocates to the cytosol to inactivate ribosomes. Currently, no molecular details of the dislocation process are available. To investigate the mechanism of the dislocation step we expressed SLTxA1 in the ER of Saccharomyces cerevisiae. METHODOLOGY AND PRINCIPAL FINDINGS: Using a combination of growth studies and biochemical tracking in yeast knock-out strains we show that SLTxA1 follows an ER-associated degradation (ERAD) pathway to enter the cytosol in a step mediated by the transmembrane Hrd1p ubiquitin ligase complex. ER-to-cytosol dislocation of the bulk population of SLTxA1 requires Cdc48p and its ubiquitin-handling co-factor Npl4p, and this population of toxin is terminally dispatched by proteasomal degradation. A small sub-population of SLTxA1 uncouples from this classical ERAD pathway and recovers catalytic activity in the cytosol. The pathway that leads to toxicity is also Hrd1p-dependent but, unlike that for the related ricin A chain toxin, SLTxA1 dislocation does require the catalytic cysteine of Hrd1p. However it does not depend on canonical ubiquitylation since toxin variants lacking endogenous lysyl residues also utilize this pathway, and furthermore there is no requirement for a number of Cdc48p co-factors. CONCLUSIONS AND SIGNIFICANCE: The fraction of SLTxA1 that disengages from the ERAD pathway thus does so upstream of Cdc48p interactions and downstream of Hrd1p interactions, in a step that possibly involves de-ubiquitylation. Mechanistically therefore, the dislocation of this toxin is quite distinct from that of conventional ERAD substrates that are normally degraded, and the toxins partially characterised to date that do not require the catalytic cysteine of the major Hrd1p component of the dislocation apparatus.


Subject(s)
Cytosol/metabolism , Endoplasmic Reticulum/metabolism , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae/metabolism , Shiga Toxins/metabolism , Ubiquitin-Protein Ligases/metabolism , Endoplasmic Reticulum/genetics , Protein Transport/genetics , Protein Transport/physiology , Ribosomes/metabolism , Saccharomyces cerevisiae Proteins/genetics , Ubiquitin-Protein Ligases/genetics
5.
Curr Top Microbiol Immunol ; 357: 19-40, 2012.
Article in English | MEDLINE | ID: mdl-21761287

ABSTRACT

A number of protein toxins bind at the surface of mammalian cells and after endocytosis traffic to the endoplasmic reticulum, where the toxic A chains are liberated from the holotoxin. The free A chains are then dislocated, or retrotranslocated, across the ER membrane into the cytosol. Here, in contrast to ER substrates destined for proteasomal destruction, they undergo folding to a catalytic conformation and subsequently inactivate their cytosolic targets. These toxins therefore provide toxic probes for testing the molecular requirements for retrograde trafficking, the ER processes that prepare the toxic A chains for transmembrane transport, the dislocation step itself and for the post-dislocation folding that results in catalytic activity. We describe here the dislocation of ricin A chain and Shiga toxin A chain, but also consider cholera toxin which bears a superficial structural resemblance to Shiga toxin. Recent studies not only describe how these proteins breach the ER membrane, but also reveal aspects of a fundamental cell biological process, that of ER-cytosol dislocation.


Subject(s)
Cytosol/metabolism , Endoplasmic Reticulum/metabolism , Ricin/metabolism , Shiga Toxin/metabolism , Intracellular Membranes/metabolism , Protein Transport , Ricin/chemistry , Shiga Toxin/chemistry
6.
Toxins (Basel) ; 3(7): 787-801, 2011 07.
Article in English | MEDLINE | ID: mdl-22069740

ABSTRACT

Ricin is a heterodimeric plant protein that is potently toxic to mammalian and many other eukaryotic cells. It is synthesized and stored in the endosperm cells of maturing Ricinus communis seeds (castor beans). The ricin family has two major members, both, lectins, collectively known as Ricinus communis agglutinin ll (ricin) and Ricinus communis agglutinin l (RCA). These proteins are stored in vacuoles within the endosperm cells of mature Ricinus seeds and they are rapidly broken down by hydrolysis during the early stages of post-germinative growth. Both ricin and RCA traffic within the plant cell from their site of synthesis to the storage vacuoles, and when they intoxicate mammalian cells they traffic from outside the cell to their site of action. In this review we will consider both of these trafficking routes.


Subject(s)
Endoplasmic Reticulum/metabolism , Ricin/biosynthesis , Ricinus/genetics , Vacuoles/metabolism , Animals , Mammals/metabolism , Plant Lectins/genetics , Plant Lectins/metabolism , Protein Transport , Ricin/metabolism , Ricinus/metabolism
7.
Traffic ; 12(11): 1552-62, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21801289

ABSTRACT

We screened a panel of compounds derived from Exo2 - a drug that perturbs post-Golgi compartments and trafficking in mammalian cells - for their effect on the secretory pathway in Arabidopsis root epidermal cells. While Exo2 and most related compounds had no significant effect, one Exo2 derivative, named LG8, induced severe morphological alterations in both the Golgi (at high concentrations) and the endoplasmic reticulum (ER). LG8 causes the ER to form foci of interconnecting tubules, which at the ultrastructural level appear similar to those previously reported in Arabidopsis roots after treatment with the herbicide oryzalin. In cotyledonary leaves, LG8 causes redistribution of a trans Golgi network (TGN) marker to the vacuole. LG8 affects the anterograde secretory pathway by inducing secretion of vacuolar cargo and preventing the brassinosteroid receptor BRI1 from reaching the plasma membrane. Uptake and arrival at the TGN of the endocytic marker FM4-64 is not affected. Unlike the ADP ribosylation factor-GTP exchange factor (ARF-GEF) inhibitor brefeldin A (BFA), LG8 affects these post-Golgi events without causing the formation of BFA bodies. Up to concentrations of 50 µm, the effects of LG8 are reversible.


Subject(s)
Arabidopsis/drug effects , Benzaldehydes/pharmacology , Endoplasmic Reticulum/drug effects , Golgi Apparatus/drug effects , Pyrimidines/pharmacology , Vacuoles/drug effects , trans-Golgi Network/drug effects , ADP-Ribosylation Factors/metabolism , Arabidopsis/metabolism , Arabidopsis Proteins/metabolism , Brefeldin A/pharmacology , Cell Membrane/drug effects , Cell Membrane/metabolism , Dinitrobenzenes/pharmacology , Endoplasmic Reticulum/metabolism , Endoplasmic Reticulum/ultrastructure , Endosomes/drug effects , Endosomes/metabolism , Golgi Apparatus/metabolism , Golgi Apparatus/ultrastructure , Plant Roots/drug effects , Plant Roots/metabolism , Protein Kinases/metabolism , Protein Transport , Pyridinium Compounds/metabolism , Quaternary Ammonium Compounds/metabolism , Secretory Pathway/drug effects , Sulfanilamides/pharmacology , Vacuoles/metabolism , trans-Golgi Network/metabolism
8.
PLoS One ; 6(7): e22713, 2011.
Article in English | MEDLINE | ID: mdl-21799938

ABSTRACT

BACKGROUND: The small molecule Eeyarestatin I (ESI) inhibits the endoplasmic reticulum (ER)-cytosol dislocation and subsequent degradation of ERAD (ER associated protein degradation) substrates. Toxins such as ricin and Shiga/Shiga-like toxins (SLTx) are endocytosed and trafficked to the ER. Their catalytic subunits are thought to utilise ERAD-like mechanisms to dislocate from the ER into the cytosol, where a proportion uncouples from the ERAD process, recovers a catalytic conformation and destroys their cellular targets. We therefore investigated ESI as a potential inhibitor of toxin dislocation. METHODOLOGY AND PRINCIPAL FINDINGS: Using cytotoxicity measurements, we found no role for ES(I) as an inhibitor of toxin dislocation from the ER, but instead found that for SLTx, ESI treatment of cells was protective by reducing the rate of toxin delivery to the ER. Microscopy of the trafficking of labelled SLTx and its B chain (lacking the toxic A chain) showed a delay in its accumulation at a peri-nuclear location, confirmed to be the Golgi by examination of SLTx B chain metabolically labelled in the trans-Golgi cisternae. The drug also reduced the rate of endosomal trafficking of diphtheria toxin, which enters the cytosol from acidified endosomes, and delayed the Golgi-specific glycan modifications and eventual plasma membrane appearance of tsO45 VSV-G protein, a classical marker for anterograde trafficking. CONCLUSIONS AND SIGNIFICANCE: ESI acts on one or more components that function during vesicular transport, whilst at least one retrograde trafficking pathway, that of ricin, remains unperturbed.


Subject(s)
Hydrazones/pharmacology , Hydroxyurea/analogs & derivatives , Intracellular Space/drug effects , Intracellular Space/metabolism , Biological Transport/drug effects , Cytosol/drug effects , Cytosol/metabolism , Diphtheria Toxin/metabolism , Diphtheria Toxin/toxicity , Endoplasmic Reticulum/drug effects , Endoplasmic Reticulum/metabolism , HeLa Cells , Humans , Hydroxyurea/pharmacology , Intracellular Membranes/drug effects , Intracellular Membranes/metabolism , Membrane Glycoproteins/metabolism , Ricin/metabolism , Ricin/toxicity , Shiga Toxin/metabolism , Shiga Toxin/toxicity , Time Factors , Viral Envelope Proteins/metabolism
9.
J Biomol Screen ; 16(4): 436-42, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21364088

ABSTRACT

The ribosome-inhibiting toxin ricin binds exposed ß1→4 linked galactosyls on multiple glycolipids and glycoproteins on the cell surface of most eukaryotic cells. After endocytosis, internal cell trafficking is promiscuous, with only a small proportion of ricin proceeding down a productive (cytotoxic) trafficking route to the endoplasmic reticulum (ER). Here, the catalytic ricin A chain traverses the membrane to inactivate the cytosolic ribosomes, which can be monitored by measuring reduction in protein biosynthetic capacity or cell viability. Although some markers have been discovered for the productive pathway, many molecular details are lacking. To identify a more comprehensive set of requirements for ricin intoxication, the authors have developed an RNAi screen in Drosophila S2 cells, screening in parallel the effects of individual RNAi treatments alone and when combined with a ricin challenge. Initial screening of 806 gene knockdowns has revealed a number of candidates for both productive and nonproductive ricin trafficking, including proteins required for transport to the Golgi, plus potential toxin interactors within the ER and cytosol.


Subject(s)
Drosophila melanogaster/drug effects , Drosophila melanogaster/genetics , RNA Interference , Ricin/pharmacology , Animals , Cells, Cultured , Chemical Warfare Agents/pharmacology , Drosophila melanogaster/cytology , Drug Resistance/genetics , Gene Library , High-Throughput Screening Assays
10.
Traffic ; 11(12): 1537-51, 2010 Dec.
Article in English | MEDLINE | ID: mdl-20854417

ABSTRACT

Brefeldin A-mediated inhibition of ADP ribosylation factor (Arf) GTPases and their guanine nucleotide exchange factors, Arf-GEFs, has been a cornerstone of membrane trafficking research for many years. Brefeldin A (BFA) is relatively non-selective inhibiting at least three targets in human cells, Golgi brefeldin A resistance factor 1 (GBF1), brefeldin A inhibited guanine nucleotide exchange factor 1 (BIG1) and brefeldin A inhibited guanine nucleotide exchange factor 2 (BIG2). Here, we show that the previously described compound Exo2 acts through inhibition of Arf-GEF function, but causes other phenotypic changes that are not GBF1 related. We describe the engineering of Exo2 to produce LG186, a more selective, reversible inhibitor of Arf-GEF function. Using multiple-cell-based assays and GBF1 mutants, our data are most consistent with LG186 acting by selective inhibition of GBF1. Unlike other Arf-GEF and reported GBF1 inhibitors including BFA, Exo2 and Golgicide A, LG186 induces disassembly of the Golgi stack in both human and canine cells.


Subject(s)
Golgi Apparatus/drug effects , Guanine Nucleotide Exchange Factors/antagonists & inhibitors , Heterocyclic Compounds, 3-Ring/pharmacology , Hydrazones/pharmacology , ADP-Ribosylation Factor 1/antagonists & inhibitors , Amino Acid Sequence , Animals , Benzaldehydes/pharmacology , Brefeldin A/pharmacology , Cell Line , Dogs , Golgi Apparatus/metabolism , Guanine Nucleotide Exchange Factors/chemistry , Guanine Nucleotide Exchange Factors/metabolism , Heterocyclic Compounds, 3-Ring/chemical synthesis , Humans , Hydrazones/chemical synthesis , Molecular Sequence Data , Protein Conformation , Pyridines/pharmacology , Pyrimidines/pharmacology , Quinolines/pharmacology
11.
Mol Biosyst ; 6(10): 2030-8, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20697620

ABSTRACT

The small molecule 4-hydroxy-3-methoxybenzaldehyde (5,6,7,8-tetrahydro[1]benzothieno[2,3-d]pyrimidin-4-yl)hydrazone (Exo2) stimulates morphological changes at the mammalian Golgi and trans-Golgi network that are virtually indistinguishable from those induced by brefeldin A. Both brefeldin A and Exo2 protect cells from intoxication by Shiga(-like) toxins by acting on other targets that operate at the early endosome, but do so at the cost of high toxicity to target cells. The advantage of Exo2 is that it is much more amenable to chemical modification and here we report a range of Exo2 analogues produced by modifying the tetrahydrobenzothienopyrimidine core, the vanillin moiety and the hydrazone bond that links these two. These compounds were examined for the morphological changes they stimulated at the Golgi stack, the trans-Golgi network and the transferrin receptor-positive early endosomes and this activity correlated with their inherent toxicity towards the protein manufacturing ability of the cell and their protective effect against toxin challenge. We have developed derivatives that can separate organelle morphology, target specificity, innate toxicity and toxin protection. Our results provide unique compounds with low toxicity and enhanced specificity to unpick the complexity of membrane trafficking networks.


Subject(s)
Benzaldehydes/pharmacology , Biological Transport/drug effects , Pyrimidines/pharmacology , HeLa Cells , Humans
12.
Mol Biol Cell ; 21(15): 2543-54, 2010 Aug 01.
Article in English | MEDLINE | ID: mdl-20519439

ABSTRACT

We report that a toxic polypeptide retaining the potential to refold upon dislocation from the endoplasmic reticulum (ER) to the cytosol (ricin A chain; RTA) and a misfolded version that cannot (termed RTA(Delta)), follow ER-associated degradation (ERAD) pathways in Saccharomyces cerevisiae that substantially diverge in the cytosol. Both polypeptides are dislocated in a step mediated by the transmembrane Hrd1p ubiquitin ligase complex and subsequently degraded. Canonical polyubiquitylation is not a prerequisite for this interaction because a catalytically inactive Hrd1p E3 ubiquitin ligase retains the ability to retrotranslocate RTA, and variants lacking one or both endogenous lysyl residues also require the Hrd1p complex. In the case of native RTA, we established that dislocation also depends on other components of the classical ERAD-L pathway as well as an ongoing ER-Golgi transport. However, the dislocation pathways deviate strikingly upon entry into the cytosol. Here, the CDC48 complex is required only for RTA(Delta), although the involvement of individual ATPases (Rpt proteins) in the 19S regulatory particle (RP) of the proteasome, and the 20S catalytic chamber itself, is very different for the two RTA variants. We conclude that cytosolic ERAD components, particularly the proteasome RP, can discriminate between structural features of the same substrate.


Subject(s)
Endoplasmic Reticulum/metabolism , Protein Folding , Ricin/chemistry , Ricin/metabolism , Cytosol/metabolism , Gene Deletion , Gene Library , Golgi Apparatus/metabolism , Lysine/metabolism , Models, Biological , Molecular Chaperones/metabolism , Mutant Proteins/chemistry , Mutant Proteins/metabolism , Proteasome Endopeptidase Complex/metabolism , Protein Processing, Post-Translational , Protein Transport , Saccharomyces cerevisiae/cytology , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism , Saccharomyces cerevisiae Proteins/metabolism , Ubiquitination
13.
Curr Opin Drug Discov Devel ; 13(1): 86-95, 2010 Jan.
Article in English | MEDLINE | ID: mdl-20047149

ABSTRACT

A significant number of therapeutic targets reside inside cells and intracellular organelles. Therapeutics therefore must be able to gain access to cellular compartments, and be able to interact specifically with a given molecule to exert a desired effect. Many naturally occurring toxins perform such targeting with apparent ease, making them excellent paradigms for the delivery of therapeutics to the cell interior. By studying the mechanisms of cell entry, trafficking and modes of toxicity of these model delivery vectors, researchers can decipher how cells transport both endogenous molecules and exogenously applied therapeutics inside cells. Perhaps more importantly, the exploitation of cell binding and trafficking motifs could allow a therapeutic to target specifically, traffic within and escape from cellular compartments; in addition, toxic domains can be used to disrupt cell function specifically for therapeutic purposes. This review provides an overview of recent developments in the understanding of toxin targeting and trafficking, and discusses how these developments could result in opportunities for the design of more specific and efficient systems for therapeutic targeting.


Subject(s)
Cells/metabolism , Drug Delivery Systems , Toxins, Biological/metabolism , Animals , Cells/ultrastructure , Endoplasmic Reticulum/metabolism , Humans , Models, Biological , Nanoparticles , Plasma/metabolism , Protein Transport/drug effects , Protein Transport/physiology , Small Molecule Libraries
14.
Eur J Med Chem ; 45(1): 275-83, 2010 Jan.
Article in English | MEDLINE | ID: mdl-19883956

ABSTRACT

The oxidative cyclisation of a range of benzothieno[2,3-d]pyrimidine hydrazones (7a-j) to the 1,2,4-triazolo[4,3-c]pyrimidines (8a-j) catalysed by lithium iodide or to the 1,2,4-triazolo[1,5-c]pyrimidines (10a-j) with sodium carbonate is presented. A complementary synthesis of the 1,2,4-triazolo[1,5-c]pyrimidines starting from the amino imine 11 is also reported. The effect of these compounds on Shiga toxin (STx) trafficking in HeLa cells and comparison to the previously reported Exo2 is also detailed.


Subject(s)
Hydrazones/chemistry , Hydrazones/pharmacology , Shiga Toxin/metabolism , Triazoles/chemistry , Triazoles/pharmacology , Benzaldehydes/chemistry , Benzaldehydes/pharmacology , Biological Transport/drug effects , Cyclization , HeLa Cells , Humans , Oxidation-Reduction , Pyrimidines/chemistry , Pyrimidines/pharmacology
15.
J Biol Chem ; 284(32): 21684-95, 2009 Aug 07.
Article in English | MEDLINE | ID: mdl-19465480

ABSTRACT

Disruption of Golgi alpha-mannosidase II activity can result in type II congenital dyserythropoietic anemia and induce lupus-like autoimmunity in mice. Here, we isolated a mutant human embryonic kidney (HEK) 293T cell line called Lec36, which displays sensitivity to ricin that lies between the parental HEK 293T cells, in which the secreted and membrane-expressed proteins are dominated by complex-type glycosylation, and 293S Lec1 cells, which produce only oligomannose-type N-linked glycans. Stem cell marker 19A was transiently expressed in the HEK 293T Lec36 cells and in parental HEK 293T cells with and without the potent Golgi alpha-mannosidase II inhibitor, swainsonine. Negative ion nano-electrospray ionization mass spectra of the 19A N-linked glycans from HEK 293T Lec36 and swainsonine-treated HEK 293T cells were qualitatively indistinguishable and, as shown by collision-induced dissociation spectra, were dominated by hybrid-type glycosylation. Nucleotide sequencing revealed mutations in each allele of MAN2A1, the gene encoding Golgi alpha-mannosidase II: a point mutation that mapped to the active site was found in one allele, and an in-frame deletion of 12 nucleotides was found in the other allele. Expression of the wild type but not the mutant MAN2A1 alleles in Lec36 cells restored processing of the 19A reporter glycoprotein to complex-type glycosylation. The Lec36 cell line will be useful for expressing therapeutic glycoproteins with hybrid-type glycans and as a sensitive host for detecting mutations in human MAN2A1 causing type II congenital dyserythropoietic anemia.


Subject(s)
Cell Line , Golgi Apparatus/metabolism , Mutation , alpha-Mannosidase/genetics , Alleles , DNA, Complementary/metabolism , Glycosylation , Humans , Models, Biological , Mutagenesis , Nucleotides/chemistry , Oligonucleotides/chemistry , Polysaccharides/chemistry , Sequence Analysis, DNA , Spectrometry, Mass, Electrospray Ionization/methods
16.
Proc Natl Acad Sci U S A ; 105(45): 17408-13, 2008 Nov 11.
Article in English | MEDLINE | ID: mdl-18988734

ABSTRACT

The plant cytotoxin ricin enters target mammalian cells by receptor-mediated endocytosis and undergoes retrograde transport to the endoplasmic reticulum (ER). Here, its catalytic A chain (RTA) is reductively separated from the cell-binding B chain, and free RTA enters the cytosol where it inactivates ribosomes. Cytosolic entry requires unfolding of RTA and dislocation across the ER membrane such that it arrives in the cytosol in a vulnerable, nonnative conformation. Clearly, for such a dislocated toxin to become active, it must avoid degradation and fold to a catalytic conformation. Here, we show that, in vitro, Hsc70 prevents aggregation of heat-treated RTA, and that RTA catalytic activity is recovered after chaperone treatment. A combination of pharmacological inhibition and cochaperone expression reveals that, in vivo, cytosolic RTA is scrutinized sequentially by the Hsc70 and Hsp90 cytosolic chaperone machineries, and that its eventual fate is determined by the balance of activities of cochaperones that regulate Hsc70 and Hsp90 functions. Cytotoxic activity follows Hsc70-mediated escape of RTA from an otherwise destructive pathway facilitated by Hsp90. We demonstrate a role for cytosolic chaperones, proteins typically associated with folding nascent proteins, assembling multimolecular protein complexes and degrading cytosolic and stalled, cotranslocational clients, in a toxin triage, in which both toxin folding and degradation are initiated from chaperone-bound states.


Subject(s)
Cytosol/metabolism , Endoplasmic Reticulum/metabolism , HSC70 Heat-Shock Proteins/metabolism , Molecular Chaperones/metabolism , Ricin/metabolism , Electrophoresis, Polyacrylamide Gel , HSP90 Heat-Shock Proteins/metabolism , HeLa Cells , Humans , Protein Conformation , Ribosomes/metabolism , Ricin/toxicity , Ubiquitination
17.
Biochem J ; 414(3): 471-84, 2008 Sep 15.
Article in English | MEDLINE | ID: mdl-18522538

ABSTRACT

The small-molecule inhibitor Exo2 {4-hydroxy-3-methoxy-(5,6,7,8-tetrahydrol[1]benzothieno[2,3-d]pyrimidin-4-yl)hydraz-one benzaldehyde} has been reported to disrupt the Golgi apparatus completely and to stimulate Golgi-ER (endoplasmic reticulum) fusion in mammalian cells, akin to the well-characterized fungal toxin BFA (brefeldin A). It has also been reported that Exo2 does not affect the integrity of the TGN (trans-Golgi network), or the direct retrograde trafficking of the glycolipid-binding cholera toxin from the TGN to the ER lumen. We have examined the effects of BFA and Exo2, and found that both compounds are indistinguishable in their inhibition of anterograde transport and that both reagents significantly disrupt the morphology of the TGN in HeLa and in BS-C-1 cells. However, Exo2, unlike BFA, does not induce tubulation and merging of the TGN and endosomal compartments. Furthermore, and in contrast with its effects on cholera toxin, Exo2 significantly perturbs the delivery of Shiga toxin to the ER. Together, these results suggest that the likely target(s) of Exo2 operate at the level of the TGN, the Golgi and a subset of early endosomes, and thus Exo2 provides a more selective tool than BFA for examining membrane trafficking in mammalian cells.


Subject(s)
Benzaldehydes/pharmacology , Endosomes/metabolism , Pyrimidines/pharmacology , Shiga Toxin/metabolism , trans-Golgi Network/metabolism , Animals , Benzaldehydes/metabolism , Brefeldin A/pharmacology , Chlorocebus aethiops , Endoplasmic Reticulum , Endosomes/drug effects , Fluorescent Antibody Technique , Green Fluorescent Proteins/metabolism , HeLa Cells , Humans , Protein Transport/drug effects , Pyrimidines/metabolism , Vero Cells , trans-Golgi Network/drug effects
18.
J Cell Biol ; 177(6): 1133-43, 2007 Jun 18.
Article in English | MEDLINE | ID: mdl-17562788

ABSTRACT

Rab family guanosine triphosphatases (GTPases) together with their regulators define specific pathways of membrane traffic within eukaryotic cells. In this study, we have investigated which Rab GTPase-activating proteins (GAPs) can interfere with the trafficking of Shiga toxin from the cell surface to the Golgi apparatus and studied transport of the epidermal growth factor (EGF) from the cell surface to endosomes. This screen identifies 6 (EVI5, RN-tre/USP6NL, TBC1D10A-C, and TBC1D17) of 39 predicted human Rab GAPs as specific regulators of Shiga toxin but not EGF uptake. We show that Rab43 is the target of RN-tre and is required for Shiga toxin uptake. In contrast, RabGAP-5, a Rab5 GAP, was unique among the GAPs tested and reduced the uptake of EGF but not Shiga toxin. These results suggest that Shiga toxin trafficking to the Golgi is a multistep process controlled by several Rab GAPs and their target Rabs and that this process is discrete from ligand-induced EGF receptor trafficking.


Subject(s)
Epidermal Growth Factor/metabolism , GTPase-Activating Proteins/physiology , Shiga Toxin/metabolism , rab GTP-Binding Proteins/physiology , Endosomes/metabolism , Golgi Apparatus/metabolism , Humans , Protein Transport
19.
Adv Drug Deliv Rev ; 58(15): 1581-96, 2006 Dec 30.
Article in English | MEDLINE | ID: mdl-17118486

ABSTRACT

There is a vast number of bacterial and plant toxins that affect cytosolic targets in mammalian cells, and whether the purpose of the toxin is to act as a defence mechanism against predators, or to cause deliberate cell death in order to form an environment more suitable for bacterial growth, each of these toxins must cross a lipid membrane barrier in order to exert their effect. This review looks at the methods employed by bacterial and plant toxins in order to reach their target. We examine the trafficking methods involved in toxin transport throughout the cell, highlighting the proteins necessary for the toxins movement, and noting how many of the toxins hijack the cells own trafficking and protein processing machinery in order to reach their goals. Studying the trafficking of toxins has led to a greater understanding of retrograde transport, a process which has key relevance to the correct intracellular delivery of pharmacological agents.


Subject(s)
Bacterial Toxins/metabolism , Cells/metabolism , Mammals/metabolism , Animals , Cell Membrane/metabolism , Endoplasmic Reticulum/metabolism , Endosomes/metabolism , Humans
20.
Virol J ; 3: 26, 2006 Apr 07.
Article in English | MEDLINE | ID: mdl-16603059

ABSTRACT

A model has been presented for retrograde transport of certain toxins and viruses from the cell surface to the ER that suggests an obligatory interaction with a glycolipid receptor at the cell surface. Here we review studies on the ER trafficking cholera toxin, Shiga and Shiga-like toxins, Pseudomonas exotoxin A and ricin, and compare the retrograde routes followed by these protein toxins to those of the ER trafficking SV40 and polyoma viruses. We conclude that there is in fact no obligatory requirement for a glycolipid receptor, nor even with a protein receptor in a lipid-rich environment. Emerging data suggests instead that there is no common pathway utilised for retrograde transport by all of these pathogens, the choice of route being determined by the particular receptor utilised.


Subject(s)
Bacterial Toxins/metabolism , Endoplasmic Reticulum/metabolism , Polyomavirus/pathogenicity , Protein Transport , Simian virus 40/pathogenicity , Animals , Cytosol/metabolism , Humans , Mice , Polyomavirus/metabolism , Receptors, Cell Surface/metabolism , Receptors, Virus/metabolism , Simian virus 40/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...