Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 20
1.
Fish Shellfish Immunol ; 147: 109404, 2024 Apr.
Article En | MEDLINE | ID: mdl-38325590

Cardiomyopathy syndrome (CMS) caused by piscine myocarditis virus (PMCV) is a severe cardiac disease in Atlantic salmon (Salmo salar) and one of the leading causes of morbidity and mortality in the Norwegian aquaculture industry. Previous research suggest a variation in individual susceptibility to develop severe disease, however the role of the immune response in determining individual outcome of CMS is poorly understood particularly in cases where fish are also challenged by stress. The present study's aim was therefore to characterize cardiac transcriptional responses to PMCV infection in Atlantic salmon responding to infection under stressful conditions with a high versus low degree of histopathological damage. The study was performed as a large-scale controlled experiment of Atlantic salmon smolts from pre-challenge to 12 weeks post infection (wpi) with PMCV, during which fish were exposed to intermittent stressors. RNA sequencing (RNAseq) was used to compare the heart transcriptome of high responders (HR) with atrium histopathology score '4' and low responders (LR) with score '0.5' at 12 wpi. A high-throughput quantitative PCR (qPCR) analysis was used to compare immune gene transcription between individuals sampled at 6, 9 and 12 wpi. Based on RNAseq and qPCR results, RNAscope in situ hybridization (ISH) was performed for visualization of IFN-γ - and IFNb producing immune cells in affected heart tissue. Compared to LR, the transcription of 1592 genes was increased in HR at 12 wpi. Of these genes, around. 40 % were immune-related, including various chemokines, key antiviral response molecules, and genes. associated with a Th1 pro-inflammatory immune response. Further, the qPCR analysis confirmed. increased immune gene transcription in HR at both 9 and 12 wpi, despite a decrease in PMCV. transcription between these time points. Interestingly, increased IFNb transcription in HR suggests the. presence of high-quantity IFN secreting cells in the hearts of these individuals. Indeed, RNAscope. confirmed the presence of IFN-γ and IFNb-positive cells in the heart ventricle of HR but not LR. To conclude, our data indicate that in severe outcomes of PMCV infection various chemokines attract leucocytes to the salmon heart, including IFN-γ and IFNb-secreting cells, and that these cells play important roles in maintaining persistent antiviral responses and a sustained host immunopathology despite decreasing heart viral transcription.


Cardiomyopathies , Fish Diseases , Salmo salar , Totiviridae , Animals , Totiviridae/genetics , Cardiomyopathies/genetics , Adaptive Immunity , Chemokines , Antiviral Agents
2.
Toxins (Basel) ; 14(5)2022 05 22.
Article En | MEDLINE | ID: mdl-35622606

The compound EACC (ethyl (2-(5-nitrothiophene-2-carboxamido) thiophene-3-carbonyl) carbamate) was recently reported to inhibit fusion of autophagosomes with lysosomes in a reversible manner by inhibiting recruitment of syntaxin 17 to autophagosomes. We report here that this compound also provides a strong protection against the protein toxin ricin as well as against other plant toxins such as abrin and modeccin. The protection did not seem to be caused by inhibition of endocytosis and retrograde transport, but rather by inhibited release of the enzymatically active A-moiety to the cytosol. The TANK-binding kinase 1 (TBK1) has been reported to phosphorylate syntaxin 17 and be required for initiation of autophagy. The inhibitor of TBK1, MRT68601, induced in itself a strong sensitization to ricin, apparently by increasing transport to the Golgi apparatus. Importantly, MRT68601 increased Golgi transport of ricin even in the presence of EACC, but EACC was still able to inhibit intoxication, supporting the idea that EACC protects at a late step along the retrograde pathway. These results also indicate that phosphorylation of syntaxin 17 is not required for the protection observed.


Abrin , Ricin , Autophagy , Lysosomes , Qa-SNARE Proteins , Ricin/toxicity
3.
J Mol Biol ; 433(15): 167086, 2021 07 23.
Article En | MEDLINE | ID: mdl-34089718

Ricin toxin kills mammalian cells with notorious efficiency. The toxin's B subunit (RTB) is a Gal/GalNAc-specific lectin that attaches to cell surfaces and promotes retrograde transport of ricin's A subunit (RTA) to the trans Golgi network (TGN) and endoplasmic reticulum (ER). RTA is liberated from RTB in the ER and translocated into the cell cytoplasm, where it functions as a ribosome-inactivating protein. While antibodies against ricin's individual subunits have been reported, we now describe seven alpaca-derived, single-domain antibodies (VHHs) that span the RTA-RTB interface, including four Tier 1 VHHs with IC50 values <1 nM. Crystal structures of each VHH bound to native ricin holotoxin revealed three different binding modes, based on contact with RTA's F-G loop (mode 1), RTB's subdomain 2γ (mode 2) or both (mode 3). VHHs in modes 2 and 3 were highly effective at blocking ricin attachment to HeLa cells and immobilized asialofetuin, due to framework residues (FR3) that occupied the 2γ Gal/GalNAc-binding pocket and mimic ligand. The four Tier 1 VHHs also interfered with intracellular functions of RTB, as they neutralized ricin in a post-attachment cytotoxicity assay (e.g., the toxin was bound to cell surfaces before antibody addition) and reduced the efficiency of toxin transport to the TGN. We conclude that the RTA-RTB interface is a target of potent toxin-neutralizing antibodies that interfere with both extracellular and intracellular events in ricin's cytotoxic pathway.


Antibodies, Neutralizing/pharmacology , Cytoplasm/metabolism , Endoplasmic Reticulum/metabolism , Golgi Apparatus/metabolism , Ricin/chemistry , Animals , Chlorocebus aethiops , Crystallography, X-Ray , HeLa Cells , Humans , Models, Molecular , Protein Conformation , Ricin/immunology , Single-Domain Antibodies/pharmacology , THP-1 Cells , Vero Cells
4.
Toxins (Basel) ; 13(6)2021 05 25.
Article En | MEDLINE | ID: mdl-34070659

Protein toxins secreted by bacteria and found in plants can be threats to human health. However, their extreme toxicity can also be exploited in different ways, e.g., to produce hybrid toxins directed against cancer cells and to study transport mechanisms in cells. Investigations during the last decades have shown how powerful these molecules are as tools in cell biological research. Here, we first present a partly historical overview, with emphasis on Shiga toxin and ricin, of how such toxins have been used to characterize processes and proteins of importance for their trafficking. In the second half of the article, we describe how one can now use toxins to investigate the role of lipid classes for intracellular transport. In recent years, it has become possible to quantify hundreds of lipid species using mass spectrometry analysis. Thus, it is also now possible to explore the importance of lipid species in intracellular transport. The detailed analyses of changes in lipids seen under conditions of inhibited toxin transport reveal previously unknown connections between syntheses of lipid classes and demonstrate the ability of cells to compensate under given conditions.


Ricin/metabolism , Shiga Toxin/metabolism , Animals , Biological Transport , Cell Count , Endocytosis , Endosomes/metabolism , Golgi Apparatus/metabolism , Humans , Membrane Microdomains/metabolism , Protein Transport , Trihexosylceramides/analysis
5.
Cell Mol Life Sci ; 78(3): 985-1009, 2021 Feb.
Article En | MEDLINE | ID: mdl-32447426

The membrane lipids diacylglycerol (DAG) and phosphatidic acid (PA) are important second messengers that can regulate membrane transport by recruiting proteins to the membrane and by altering biophysical membrane properties. DAG and PA are involved in the transport from the Golgi apparatus to endosomes, and we have here investigated whether changes in these lipids might be important for regulation of transport to the Golgi using the protein toxin ricin. Modulation of DAG and PA levels using DAG kinase (DGK) and phospholipase D (PLD) inhibitors gave a strong increase in retrograde ricin transport, but had little impact on ricin recycling or degradation. Inhibitor treatment strongly affected the endosome morphology, increasing endosomal tubulation and size. Furthermore, ricin was present in these tubular structures together with proteins known to regulate retrograde transport. Using siRNA to knock down different isoforms of PLD and DGK, we found that several isoforms of PLD and DGK are involved in regulating ricin transport to the Golgi. Finally, by performing lipidomic analysis we found that the DGK inhibitor gave a weak, but expected, increase in DAG levels, while the PLD inhibitor gave a strong and unexpected increase in DAG levels, showing that it is important to perform lipidomic analysis when using inhibitors of lipid metabolism.


Diacylglycerol Kinase/metabolism , Endosomes/metabolism , Golgi Apparatus/metabolism , Phospholipase D/metabolism , Cell Line, Tumor , Diacylglycerol Kinase/antagonists & inhibitors , Diacylglycerol Kinase/genetics , Diglycerides/metabolism , Endocytosis/drug effects , Enzyme Inhibitors/pharmacology , Humans , Lipidomics/methods , Lipids/analysis , Phospholipase D/antagonists & inhibitors , Phospholipase D/genetics , Protein Isoforms/antagonists & inhibitors , Protein Isoforms/genetics , Protein Isoforms/metabolism , Protein Transport/drug effects , Proteolysis/drug effects , Pyrimidinones/pharmacology , RNA Interference , RNA, Small Interfering/metabolism , Ricin/metabolism , Thiazoles/pharmacology
6.
Cancer Metastasis Rev ; 39(2): 343-360, 2020 06.
Article En | MEDLINE | ID: mdl-32314087

Several studies have demonstrated interactions between the two leaflets in membrane bilayers and the importance of specific lipid species for such interaction and membrane function. We here discuss these investigations with a focus on the sphingolipid and cholesterol-rich lipid membrane domains called lipid rafts, including the small flask-shaped invaginations called caveolae, and the importance of such membrane structures in cell biology and cancer. We discuss the possible interactions between the very long-chain sphingolipids in the outer leaflet of the plasma membrane and the phosphatidylserine species PS 18:0/18:1 in the inner leaflet and the importance of cholesterol for such interactions. We challenge the view that lipid rafts contain a large fraction of lipids with two saturated fatty acyl groups and argue that it is important in future studies of membrane models to use asymmetric membrane bilayers with lipid species commonly found in cellular membranes. We also discuss the need for more quantitative lipidomic studies in order to understand membrane function and structure in general, and the importance of lipid rafts in biological systems. Finally, we discuss cancer-related changes in lipid rafts and lipid composition, with a special focus on changes in glycosphingolipids and the possibility of using lipid therapy for cancer treatment.


Glycosphingolipids/metabolism , Membrane Microdomains/metabolism , Neoplasms/metabolism , Animals , Cell Membrane/metabolism , Cell Membrane/pathology , Cholesterol/metabolism , Humans , Lipid Bilayers/metabolism , Lipid Metabolism , Membrane Microdomains/pathology , Neoplasms/drug therapy , Neoplasms/pathology , Phosphatidylserines/metabolism , Sphingolipids/metabolism
7.
Histochem Cell Biol ; 150(2): 107-118, 2018 Aug.
Article En | MEDLINE | ID: mdl-29774430

This article aims at providing an update on the complexity of clathrin-independent endocytosis. It is now almost 30 years since we first wrote a review about its existence; at that time many people believed that with the exception of macropinocytosis, which will only be briefly mentioned in this review, all uptake could be accounted for by clathrin-dependent endocytosis. Now it is generally accepted that there are different clathrin-independent mechanisms, some of them regulated by ligands and membrane lipid composition. They can be both dynamin-dependent and -independent, meaning that the uptake cannot be accounted for by caveolae and other dynamin-dependent processes such as tubular structures that can be induced by toxins, e.g. Shiga toxin, or the fast endophilin mediated endocytosis recently described. Caveolae seem to be mostly quite stable structures with other functions than endocytosis, but evidence suggests that they may have cell-type dependent functions. Although several groups have been working on endocytic mechanisms for years, and new advanced methods have improved our ability to study mechanistic details, there are still a number of important questions we need to address, such as: How many endocytic mechanisms does a cell have? How quantitatively important are they? What about the complexity in polarized cells where clathrin-independent endocytosis is differentially regulated on the apical and basolateral poles? These questions are not easy to answer since one and the same molecule may contribute to more than one process, and manipulating one mechanism can affect another. Also, several inhibitors of endocytic processes commonly used turn out to be less specific than originally thought. We will here describe the current view of clathrin-independent endocytic processes and the challenges in studying them.


Clathrin/metabolism , Endocytosis , Humans
8.
Toxins (Basel) ; 9(2)2017 02 03.
Article En | MEDLINE | ID: mdl-28165371

Shiga toxins consist of an A-moiety and five B-moieties able to bind the neutral glycosphingolipid globotriaosylceramide (Gb3) on the cell surface. To intoxicate cells efficiently, the toxin A-moiety has to be cleaved by furin and transported retrogradely to the Golgi apparatus and to the endoplasmic reticulum. The enzymatically active part of the A-moiety is then translocated to the cytosol, where it inhibits protein synthesis and in some cell types induces apoptosis. Protection of cells can be provided either by inhibiting binding of the toxin to cells or by interfering with any of the subsequent steps required for its toxic effect. In this article we provide a brief overview of the interaction of Shiga toxins with cells, describe some compounds and conditions found to protect cells against Shiga toxins, and discuss whether they might also provide protection in animals and humans.


Antidotes/pharmacology , Bacterial Proteins/antagonists & inhibitors , Dysentery, Bacillary/prevention & control , Hemolytic-Uremic Syndrome/prevention & control , Shiga Toxins/antagonists & inhibitors , Shiga-Toxigenic Escherichia coli/drug effects , Shigella dysenteriae/drug effects , Animals , Apoptosis , Bacterial Proteins/chemistry , Bacterial Proteins/metabolism , Dysentery, Bacillary/metabolism , Dysentery, Bacillary/microbiology , Hemolytic-Uremic Syndrome/metabolism , Hemolytic-Uremic Syndrome/microbiology , Host-Pathogen Interactions , Humans , Protein Biosynthesis , Protein Conformation , Protein Transport , Shiga Toxins/chemistry , Shiga Toxins/metabolism , Shiga-Toxigenic Escherichia coli/metabolism , Shiga-Toxigenic Escherichia coli/pathogenicity , Shigella dysenteriae/metabolism , Shigella dysenteriae/pathogenicity , Structure-Activity Relationship , Trihexosylceramides/metabolism
9.
Traffic ; 18(3): 176-191, 2017 03.
Article En | MEDLINE | ID: mdl-28067430

In this study, we have investigated how clathrin-dependent endocytosis is affected by exogenously added lysophospholipids (LPLs). Addition of LPLs with large head groups strongly inhibits transferrin (Tf) endocytosis in various cell lines, while LPLs with small head groups do not. Electron and total internal reflection fluorescence microscopy (EM and TIRF) reveal that treatment with lysophosphatidylinositol (LPI) with the fatty acyl group C18:0 leads to reduced numbers of invaginated clathrin-coated pits (CCPs) at the plasma membrane, fewer endocytic events per membrane area and increased lifetime of CCPs. Also, endocytosis of Tf becomes dependent on actin upon LPI treatment. Thus, our results demonstrate that one can regulate the kinetics and properties of clathrin-dependent endocytosis by addition of LPLs in a head group size- and fatty acyl-dependent manner. Furthermore, studies performed with optical tweezers show that less force is required to pull membrane tubules outwards from the plasma membrane when LPI is added to the cells. The results are in agreement with the notion that insertion of LPLs with large head groups creates a positive membrane curvature which might have a negative impact on events that require plasma membrane invagination, while it may facilitate membrane bending toward the cell exterior.


Clathrin/metabolism , Coated Pits, Cell-Membrane/metabolism , Endocytosis/physiology , Lysophospholipids/metabolism , Actins/metabolism , Cell Line, Tumor , Cell Membrane/metabolism , Cell Membrane/physiology , HeLa Cells , Humans , Transferrin/metabolism
10.
Oncotarget ; 7(52): 86871-86888, 2016 Dec 27.
Article En | MEDLINE | ID: mdl-27894086

2-hydroxyoleic acid (OHOA, Minerval®) is an example of a substance used for membrane lipid therapy, where the cellular membranes rather than specific proteins constitute the therapeutical target. OHOA is thought to mediate its anti-tumor effect by affecting the biophysical properties of membranes, which leads to altered recruitment and activation of amphitropic proteins, altered cellular signaling, and eventual cell death. Little is known about the initial signaling events upon treatment with OHOA, and whether the altered membrane properties would have any impact on the dynamic intracellular transport system. In the present study we demonstrate that treatment with OHOA led to a rapid release of intracellular calcium and activation of multiple signaling pathways in HeLa cells, including the PI3K-AKT1-MTOR pathway and several MAP kinases, in a process independent of the EGFR. By lipidomics we confirmed that OHOA was incorporated into several lipid classes. Concomitantly, OHOA potently increased retrograde transport of the plant toxin ricin from endosomes to the Golgi and further to the endoplasmic reticulum. The OHOA-stimulated ricin transport seemed to require several amphitropic proteins, including Src, phospholipase C, protein kinase C, and also Ca2+/calmodulin. Interestingly, OHOA induced a slight increase in endosomal localization of the retromer component VPS35. Thus, our data show that addition of a lipid known to alter membrane properties not only affects signaling, but also intracellular transport.


Calcium/metabolism , Cell Membrane/metabolism , Oleic Acids/pharmacology , Signal Transduction/drug effects , Antineoplastic Agents/pharmacology , Cell Line, Tumor , Endoplasmic Reticulum/metabolism , Endosomes/metabolism , Golgi Apparatus/metabolism , HeLa Cells , Humans , Lipid Metabolism/drug effects , Membrane Fluidity/drug effects , Protein Transport/drug effects , Ricin/metabolism , Ricin/pharmacology
11.
Oncotarget ; 7(48): 79885-79900, 2016 Nov 29.
Article En | MEDLINE | ID: mdl-27829218

2-fluoro-2-deoxy-D-glucose (FDG), labeled with 18F radioisotope, is the most common imaging agent used for positron emission tomography (PET) in oncology. However, little is known about the cellular effects of FDG. Another glucose analogue, 2-deoxy-D-glucose (2DG), has been shown to affect many cellular functions, including intracellular transport and lipid metabolism, and has been found to improve the efficacy of cancer chemotherapeutic agents in vivo. Thus, in the present study, we have investigated cellular effects of FDG with the focus on changes in cellular lipids and intracellular transport. By quantifying more than 200 lipids from 17 different lipid classes in HEp-2 cells and by analyzing glycosphingolipids from MCF-7, HT-29 and HBMEC cells, we have discovered that FDG treatment inhibits glucosylceramide synthesis and thus reduces cellular levels of glycosphingolipids. In addition, in HEp-2 cells the levels and/or species composition of other lipid classes, namely diacylglycerols, phosphatidic acids and phosphatidylinositols, were found to change upon treatment with FDG. Furthermore, we show here that FDG inhibits retrograde Shiga toxin transport and is much more efficient in protecting cells against the toxin than 2DG. In summary, our data reveal novel effects of FDG on cellular transport and glycosphingolipid metabolism, which suggest a potential clinical application of FDG as an adjuvant for cancer chemotherapy.


Fluorodeoxyglucose F18/pharmacology , Lipid Metabolism/drug effects , Metabolome/drug effects , Biological Transport/drug effects , Biological Transport/radiation effects , Cells, Cultured , Endocytosis/drug effects , Endocytosis/radiation effects , Endoplasmic Reticulum/drug effects , Endoplasmic Reticulum/metabolism , Endoplasmic Reticulum/radiation effects , Golgi Apparatus/drug effects , Golgi Apparatus/metabolism , Golgi Apparatus/radiation effects , HT29 Cells , Humans , Lipid Metabolism/radiation effects , MCF-7 Cells , Metabolome/radiation effects , Protein Transport/drug effects , Protein Transport/radiation effects , Shiga Toxin/metabolism
12.
Sci Rep ; 6: 30336, 2016 07 26.
Article En | MEDLINE | ID: mdl-27458147

Shiga toxin (Stx), an AB5 toxin, binds specifically to the neutral glycosphingolipid Gb3 at the cell surface before being transported into cells. We here demonstrate that addition of conical lysophospholipids (LPLs) with large head groups inhibit Stx binding to cells whereas LPLs with small head groups do not. Lysophosphatidylinositol (LPI 18:0), the most efficient LPL with the largest head group, was selected for in-depth investigations to study how the binding of Stx is regulated. We show that the inhibition of Stx binding by LPI is reversible and possibly regulated by cholesterol since addition of methyl-ß-cyclodextrin (mßCD) reversed the ability of LPI to inhibit binding. LPI-induced inhibition of Stx binding is independent of signalling and membrane turnover as it occurs in fixed cells as well as after depletion of cellular ATP. Furthermore, data obtained with fluorescent membrane dyes suggest that LPI treatment has a direct effect on plasma membrane lipid packing with shift towards a liquid disordered phase in the outer leaflet, while lysophosphoethanolamine (LPE), which has a small head group, does not. In conclusion, our data show that cellular treatment with conical LPLs with large head groups changes intrinsic properties of the plasma membrane and modulates Stx binding to Gb3.


Lysophospholipids/pharmacology , Neutral Glycosphingolipids/metabolism , Shiga Toxin/pharmacology , Cell Membrane/drug effects , Cell Membrane/metabolism , HeLa Cells , Humans , Lysophospholipids/chemistry , Protein Binding , beta-Cyclodextrins/pharmacology
13.
J Mol Biol ; 428(24 Pt A): 4856-4866, 2016 12 04.
Article En | MEDLINE | ID: mdl-27363608

In this study, we used water-soluble stable mass isotope precursors to measure the turnover of endogenous ceramide (Cer) and glycosphingolipids (GSLs) in HEp-2 cells. Cells incubated in the presence of [13C-U]glucose showed rapid incorporation of hexose residues with an increased mass of 6Da into GSLs. Different turnover rates of GSL classes and their molecular species were observed. Approximately 30% of the glucosylceramide, 50% of the lactosylceramide, and 50% of the globotriaosylceramide species showed a much slower turnover than the rest. This demonstrates the existence of different lipid pools, where a certain fraction of species survived for a long time in the cells. The species with the shortest N-amidated fatty acyl groups (C16:0 and C18:0) showed a more rapid turnover than those with the longest N-amidated fatty acids (C24:0 and C24:1). Experiments with addition of [13C-U]serine were performed to study de novo synthesis of Cer from serine and palmitoyl-CoA. These experiments revealed that de novo synthesis contributes to a minor extent to the total synthesis of new sphingolipids and showed that there is a more rapid formation of the longest Cer species (C24:0 and C24:1) than of the shortest species (C16:0), that is, the opposite as observed for the GSLs in the experiments with [13C-U]glucose. In conclusion, this FLUX lipidomics experimental approach with the addition of [13C-U]glucose to cells allows us to not only study the total turnover but also permit observations of lipid intermediates and metabolic flow of endogenous GSL species at the molecular lipid level.


Epithelial Cells/metabolism , Glycosphingolipids/metabolism , Isotope Labeling/methods , Isotopes/metabolism , Carbon Isotopes/metabolism , Cell Line , Glucose/metabolism , Humans , Serine/metabolism
14.
Cell Mol Life Sci ; 73(6): 1301-16, 2016 Mar.
Article En | MEDLINE | ID: mdl-26407609

Glycosphingolipids (GSLs) are predominantly found in the outer leaflet of the plasma membrane, where they play a role in important processes such as cell adhesion, migration and signaling. However, by which mechanisms GSLs regulate these processes remains elusive. In this study, we therefore took advantage of the fact that some GSLs also serve as receptors for certain protein toxins, which rely on receptor binding for internalization and intoxication. Here, we demonstrate that Shiga and cholera toxins, which both possess multivalent GSL-binding capacity, induce dissociation of the cytosolic cPLA2α-AnxA1 complex in HeLa and HMEC-1 cells. The dissociation is mediated through an increase in cytosolic calcium levels and activation of the tyrosine kinase Syk. Ricin, a protein toxin that does not cross-link surface molecules, has no effect on the same complex. Importantly, we find that antibody-mediated cross-linking of Gb3 and GM1, the GSL receptors for Shiga and cholera toxin, respectively, also induces dissociation. These data demonstrate that cross-linking of GSLs at the plasma membrane mediates the intracellular signaling events resulting in dissociation of the complex. After dissociation, cPLA2α and AnxA1 are translocated to intracellular membranes where they are known to function in regulating membrane transport processes. In conclusion, we have characterized a novel mechanism for cell surface-induced initiation of intracellular signaling and transport events.


Annexin A1/metabolism , Cell Membrane/metabolism , Cholera Toxin/metabolism , Glycosphingolipids/metabolism , Group IV Phospholipases A2/metabolism , Shiga Toxin/metabolism , Calcium/metabolism , Cell Line , Cytosol/metabolism , HeLa Cells , Humans , Intracellular Signaling Peptides and Proteins/metabolism , Protein-Tyrosine Kinases/metabolism , Signal Transduction , Syk Kinase
15.
Biochem J ; 470(1): 23-37, 2015 Aug 15.
Article En | MEDLINE | ID: mdl-26251444

2-Deoxy-D-glucose (2DG) is a structural analogue of glucose with well-established applications as an inhibitor of glycolysis and N-glycosylation. Importantly, 2DG has been shown to improve the efficacy of several cancer chemotherapeutic agents in vivo and thus it is in clinical studies in combination with chemotherapy and radiotherapy. However, although 2DG has been demonstrated to modulate many cellular functions, including autophagy, apoptosis and cell cycle control, little is known about the effects of 2DG on intracellular transport, which is of great importance when predicting the effects of 2DG on therapeutic agents. In addition to proteins, lipids play important roles in cellular signalling and in controlling cellular trafficking. We have, in the present study, investigated the effects of 2DG on cellular lipid composition and by use of protein toxins we have studied 2DG-mediated changes in intracellular trafficking. By quantifying more than 200 individual lipid species from 17 different lipid classes, we have found that 2DG treatment changes the levels and/or species composition of several lipids, such as phosphatidylinositol (PI), diacylglycerol (DAG), cholesteryl ester (CE), ceramide (Cer) and lysophospho-lipids. Moreover, 2DG becomes incorporated into the carbohydrate moiety of glycosphingolipids (GSLs). In addition, we have discovered that 2DG protects cells against Shiga toxins (Stxs) and inhibits release of the cytotoxic StxA1 moiety in the endoplasmic reticulum (ER). The data indicate that the 2DG-induced protection against Stx is independent of inhibition of glycolysis or N-glycosylation, but rather mediated via the depletion of Ca(2+) from cellular reservoirs by 2DG. In conclusion, our results reveal novel actions of 2DG on cellular lipids and Stx toxicity.


Cytoprotection/drug effects , Deoxyglucose/pharmacology , Membrane Lipids/metabolism , Shiga Toxins/toxicity , Cell Line , Cytoprotection/physiology , Humans
16.
Prog Lipid Res ; 54: 1-13, 2014 Apr.
Article En | MEDLINE | ID: mdl-24462587

The plant toxin ricin and the bacterial toxin Shiga toxin both belong to a group of protein toxins having one moiety that binds to the cell surface, and another, enzymatically active moiety, that enters the cytosol and inhibits protein synthesis by inactivating ribosomes. Both toxins travel all the way from the cell surface to endosomes, the Golgi apparatus and the ER before the ribosome-inactivating moiety enters the cytosol. Shiga toxin binds to the neutral glycosphingolipid Gb3 at the cell surface and is therefore dependent on this lipid for transport into the cells, whereas ricin binds both glycoproteins and glycolipids with terminal galactose. The different steps of transport used by these toxins have specific requirements for lipid species, and with the recent developments in mass spectrometry analysis of lipids and microscopical and biochemical dissection of transport in cells, we are starting to see the complexity of endocytosis and intracellular transport. In this article we describe lipid requirements and the consequences of lipid changes for the entry and intoxication with ricin and Shiga toxin. These toxins can be a threat to human health, but can also be exploited for diagnosis and therapy, and have proven valuable as tools to study intracellular transport.


Cells/metabolism , Lipid Metabolism , Proteins/metabolism , Toxins, Biological/metabolism , Animals , Cells/cytology , Endocytosis , Humans , Protein Transport
17.
Cell Mol Life Sci ; 71(6): 1097-116, 2014 Mar.
Article En | MEDLINE | ID: mdl-23921715

Cell density is one of the extrinsic factors to which cells adapt their physiology when grown in culture. However, little is known about the molecular changes which occur during cell growth and how cellular responses are then modulated. In many cases, inhibitors, drugs or growth factors used for in vitro studies change the rate of cell proliferation, resulting in different cell densities in control and treated samples. Therefore, for a comprehensive data analysis, it is essential to understand the implications of cell density on the molecular level. In this study, we have investigated how lipid composition changes during cell growth, and the consequences it has for transport of Shiga toxin. By quantifying 308 individual lipid species from 17 different lipid classes, we have found that the levels and species distribution of several lipids change during cell growth, with the major changes observed for diacylglycerols, phosphatidic acids, cholesterol esters, and lysophosphatidylethanolamines. In addition, there is a reduced binding and retrograde transport of Shiga toxin in high density cells which lead to reduced intoxication by the toxin. In conclusion, our data provide novel information on how lipid composition changes during cell growth in culture, and how these changes can modulate intracellular trafficking.


Cell Count , Lipid Metabolism , Syntaxin 1/metabolism , Cell Culture Techniques , Cell Line, Tumor , Cholesterol/metabolism , Diglycerides/metabolism , Globosides/metabolism , Glycosphingolipids/metabolism , HeLa Cells , Hep G2 Cells , Humans , Lysophospholipids/metabolism , Phosphatidic Acids/metabolism , Protein Transport , Shiga Toxin/metabolism , Syntaxin 1/genetics , Trihexosylceramides/metabolism
18.
Biochim Biophys Acta ; 1820(7): 870-7, 2012 Jul.
Article En | MEDLINE | ID: mdl-22507270

BACKGROUND: Blebbistatin is a new inhibitor of cell motility. It is used to study dynamics of cytokinesis machinery in cells. However, the potential of this inhibitor as an anticancer agent has not been studied so far. METHODS: Cytotoxicity of blebbistatin was evaluated in five human cell lines, FEMX-I melanoma, U87 glioma, androgen independent Du145 and androgen sensitive LNCaP prostate adenocarcinoma, and F11-hTERT immortalized fibroblasts. Phototoxicity of blebbistatin was assessed in these cell lines after their exposure to a blue light (390-470 nm). Photostability of blebbistatin and its reactive oxygen species (ROS) generating properties were measured during irradiation with the blue light. RESULTS: Blebbistatin at a concentration range of 10-200 µmol/L was toxic to all studied cells. Toxic concentrations (TC) were about 10-25 µmol/L corresponding to TC10, 50-100 µmol/L to TC50 and 140-190 µmol/L to TC90. Only for the U87 glioma cells TC90 could not be measured as the highest studied concentration of 200 µmol/L gave around 70% toxicity. However, after exposure to the blue light blebbistatin exhibited phototoxicity on the cells, with a cytotoxicity enhancement ratio that was greatest for the FEMX-I cells (about 9) followed by LNCaP (5), Du145 (3), U87 (2) and F11-hTERT (1.7) cells. CONCLUSIONS: Blebbistatin inhibits cell motility and viability. Under exposure to the blue light blebbistatin exhibits photodynamic action on human cancer cells. During the irradiation blebbistatin oxidizes dihydrorhodamine 123 but not Singlet Oxygen Sensor Green. GENERAL SIGNIFICANCE: Our findings offer new possibilities for blebbistatin as a potential anticancer and photodynamic agent.


Heterocyclic Compounds, 4 or More Rings/pharmacology , Light , Myosins/antagonists & inhibitors , Photosensitizing Agents/pharmacology , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/pathology , Apoptosis/drug effects , Cell Movement/drug effects , Cell Survival/drug effects , Humans , Male , Myosins/metabolism , Oxidation-Reduction , Prostatic Neoplasms/metabolism , Reactive Oxygen Species/metabolism , Tumor Cells, Cultured
19.
Int J Nanomedicine ; 6: 2437-44, 2011.
Article En | MEDLINE | ID: mdl-22072879

BACKGROUND: In novel treatment approaches, therapeutics should be designed to target cancer stem cells (CSCs). Quantum dots (QDs) are a promising new tool in fighting against cancer. However, little is known about accumulation and cytotoxicity of QDs in CSCs. METHODS: Accumulation and cytotoxicity of CdTe-MPA (mercaptopropionic acid) QDs in CSCs were assessed using flow cytometry and fluorescence-activated cell sorting techniques as well as a colorimetric cell viability assay. RESULTS: We investigated the expression of two cell surface-associated glycoproteins, CD44 and CD133, in four different cancer cell lines (glioblastoma, melanoma, pancreatic, and prostate adenocarcinoma). Only the melanoma cells were positive to both markers of CD44 and CD133, whereas the other cells were only CD44-positive. The QDs accumulated to a similar extent in all subpopulations of the melanoma cells. The phenotypical response after QD treatment was compared with the response after ionizing radiation treatment. The percentage of the CD44(high-)CD133(high) subpopulation decreased from 72% to 55%-58% for both treatments. The stem-like subpopulation CD44(high)CD133(low/-) increased from 26%-28% in the untreated melanoma cells to 36%-40% for both treatments. CONCLUSION: Treatment of melanoma cells with QDs results in an increase of stem-like cell subpopulations. The changes in phenotype distribution of the melanoma cells after the treatment with QDs are comparable with the changes after ionizing radiation.


Antigens, CD/biosynthesis , Cadmium Compounds/administration & dosage , Glycoproteins/biosynthesis , Melanoma/therapy , Quantum Dots , Tellurium/administration & dosage , 3-Mercaptopropionic Acid/administration & dosage , 3-Mercaptopropionic Acid/chemistry , 3-Mercaptopropionic Acid/pharmacokinetics , AC133 Antigen , Biomarkers, Tumor , Cadmium Compounds/chemistry , Cadmium Compounds/pharmacokinetics , Cell Line, Tumor , Cell Survival/drug effects , Flow Cytometry , Glioblastoma/immunology , Glioblastoma/pathology , Glioblastoma/therapy , Humans , Hyaluronan Receptors/biosynthesis , Male , Melanoma/immunology , Melanoma/metabolism , Melanoma/pathology , Neoplastic Stem Cells/pathology , Pancreatic Neoplasms/immunology , Pancreatic Neoplasms/pathology , Pancreatic Neoplasms/therapy , Peptides , Phenotype , Prostatic Neoplasms/immunology , Prostatic Neoplasms/pathology , Prostatic Neoplasms/therapy , Tellurium/chemistry , Tellurium/pharmacokinetics
20.
Int J Nanomedicine ; 6: 1875-88, 2011.
Article En | MEDLINE | ID: mdl-21931483

Quantum dots have emerged with great promise for biological applications as fluorescent markers for immunostaining, labels for intracellular trafficking, and photosensitizers for photodynamic therapy. However, upon entry into a cell, quantum dots are trapped and their fluorescence is quenched in endocytic vesicles such as endosomes and lysosomes. In this study, the photophysical properties of quantum dots were investigated in liposomes as an in vitro vesicle model. Entrapment of quantum dots in liposomes decreases their fluorescence lifetime and intensity. Generation of free radicals by liposomal quantum dots is inhibited compared to that of free quantum dots. Nevertheless, quantum dot fluorescence lifetime and intensity increases due to photolysis of liposomes during irradiation. In addition, protein adsorption on the quantum dot surface and the acidic environment of vesicles also lead to quenching of quantum dot fluorescence, which reappears during irradiation. In conclusion, the in vitro model of phospholipid vesicles has demonstrated that those quantum dots that are fated to be entrapped in endocytic vesicles lose their fluorescence and ability to act as photosensitizers.


Quantum Dots , Fluorescence , Free Radicals , Liposomes , Microscopy, Electron, Transmission , Microscopy, Fluorescence , Nanomedicine , Optical Phenomena , Particle Size , Phospholipids , Photochemotherapy , Photolysis , Photosensitizing Agents
...