Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 17 de 17
1.
Toxicon ; 147: 54-57, 2018 Jun 01.
Article En | MEDLINE | ID: mdl-29042311

Botulinum neurotoxins (BoNTs) are the most potent toxic proteins to mankind known but applied in low doses trigger a localized muscle paralysis that is beneficial for the therapy of several neurological disorders and aesthetic treatment. The paralytic effect is generated by the enzymatic activity of the light chain (LC) that cleaves specifically one of the SNARE proteins responsible for neurotransmitter exocytosis. The activity of the LC in a BoNT-containing therapeutic can be compromised by denaturing agents present during manufacturing and/or in the cell. Stabilization of the LC by reducing vulnerability towards denaturants would thus be advantageous for the development of BoNT-based therapeutics. In this work, we focused on increasing the stability of LC of BoNT/A1 (LC/A1) towards oxidative stress. We tackled this task by rational design of mutations at cysteine and methionine LC/A1 sites. Designed mutants showed improved oxidative stability in vitro and equipotency to wildtype toxin in vivo. Our results suggest that suitable modification of the catalytic domain can lead to more stable BoNTs without impairing their therapeutic efficacy.


Botulinum Toxins/genetics , Botulinum Toxins/toxicity , Clostridioides difficile/metabolism , Animals , Botulinum Toxins/chemistry , Botulinum Toxins/metabolism , Clostridioides difficile/genetics , Mice , Models, Molecular , Motor Activity/drug effects , Mutation , Oxidative Stress , Paralysis/chemically induced , Protein Conformation
2.
Toxicon ; 139: 101-108, 2017 Dec 01.
Article En | MEDLINE | ID: mdl-28918229

Botulinum neurotoxins (BoNTs) are classified by their antigenic properties into seven serotypes (A-G) and in addition by their corresponding subtypes. They are further characterized by divergent onset and duration of effect. Injections of low doses of botulinum neurotoxins cause localized muscle paralysis that is beneficial for the treatment of several medical disorders and aesthetic indications. Optimizing the therapeutic properties could offer new treatment opportunities. This report describes a rational design approach to modify the pharmacological properties by mutations in the C-terminus of BoNT/A1 light chain (LC). Toxins with C-terminal modified LC's displayed an altered onset and duration of the paralytic effect in vivo. The level of effect was dependent on the kind of the mutation in the sequence of the C-terminus. A mutant with three mutations (T420E F423M Y426F) revealed a faster onset and a shorter duration than BoNT/A1 wild type (WT). It could be shown that the C-terminus of BoNT/A1-Lc controls both onset and duration of effect. Thus, it is possible to create a mutated BoNT/A1 with different pharmacological properties which might be useful in the therapy of new indications. This strategy opens the way to design BoNT variants with novel and useful properties.


Botulinum Toxins, Type A/pharmacology , Drug Design , Neurotoxins/pharmacology , Animals , Botulinum Toxins, Type A/pharmacokinetics , Escherichia coli , Injections, Intramuscular , Mice , Muscle, Skeletal/drug effects , Neurotoxins/pharmacokinetics , Paralysis/chemically induced , Structure-Activity Relationship , Time Factors
3.
J Cell Sci ; 123(Pt 10): 1652-62, 2010 May 15.
Article En | MEDLINE | ID: mdl-20406886

Functional recovery and regeneration of corticospinal tract (CST) fibers following spinal cord injury by compression or dorsal hemisection in mice was monitored after application of the enzyme-deficient Clostridium botulinum C3-protein-derived 29-amino-acid fragment C3bot(154-182). This peptide significantly improved locomotor restoration in both injury models as assessed by the open-field Basso Mouse Scale for locomotion test and Rotarod treadmill experiments. These data were supported by tracing studies showing an enhanced regenerative growth of CST fibers in treated animals as visualized by anterograde tracing. Additionally, C3bot(154-182) stimulated regenerative growth of raphespinal fibers and improved serotonergic input to lumbar alpha-motoneurons. These in vivo data were confirmed by in vitro data, showing an enhanced axon outgrowth of alpha-motoneurons and hippocampal neurons cultivated on normal or growth-inhibitory substrates after application of C3bot(154-182). The observed effects were probably caused by a non-enzymatic downregulation of active RhoA by the C3 peptide as indicated by pull-down experiments. By contrast, C3bot(154-182) did not induce neurite outgrowth in primary cultures of dorsal root ganglion cells. In conclusion, C3bot(154-182) represents a novel, promising tool to foster axonal protection and/or repair, as well as functional recovery after traumatic CNS injury.


ADP Ribose Transferases/pharmacology , Botulinum Toxins/pharmacology , Clostridium botulinum/metabolism , Motor Neurons/drug effects , Nerve Regeneration , Peptide Fragments/pharmacology , Spinal Cord Injuries/physiopathology , Spinal Cord/drug effects , Animals , Cell Growth Processes/drug effects , Cells, Cultured , Humans , Mice , Mice, Inbred BALB C , Motor Activity/drug effects , Motor Neurons/metabolism , Motor Neurons/pathology , Pyramidal Tracts/drug effects , Pyramidal Tracts/physiology , Recovery of Function , Serotonin/genetics , Serotonin/metabolism , Spinal Cord/metabolism , Spinal Cord/pathology , Spinal Cord/surgery , Spinal Cord Injuries/drug therapy , rho GTP-Binding Proteins/genetics , rho GTP-Binding Proteins/metabolism , rhoA GTP-Binding Protein
4.
PLoS One ; 5(3): e9887, 2010 Mar 25.
Article En | MEDLINE | ID: mdl-20360858

The intracellular bacterium Chlamydia trachomatis causes infections of urogenital tract, eyes or lungs. Alignment reveals homology of CT166, a putative effector protein of urogenital C. trachomatis serovars, with the N-terminal glucosyltransferase domain of clostridial glucosylating toxins (CGTs). CGTs contain an essential DXD-motif and mono-glucosylate GTP-binding proteins of the Rho/Ras families, the master regulators of the actin cytoskeleton. CT166 is preformed in elementary bodies of C. trachomatis D and is detected in the host-cell shortly after infection. Infection with high MOI of C. trachomatis serovar D containing the CT166 ORF induces actin re-organization resulting in cell rounding and a decreased cell diameter. A comparable phenotype was observed in HeLa cells treated with the Rho-GTPase-glucosylating Toxin B from Clostridium difficile (TcdB) or HeLa cells ectopically expressing CT166. CT166 with a mutated DXD-motif (CT166-mut) exhibited almost unchanged actin dynamics, suggesting that CT166-induced actin re-organization depends on the glucosyltransferase motif of CT166. The cytotoxic necrotizing factor 1 (CNF1) from E. coli deamidates and thereby activates Rho-GTPases and transiently protects them against TcdB-induced glucosylation. CNF1-treated cells were found to be protected from TcdB- and CT166-induced actin re-organization. CNF1 treatment as well as ectopic expression of non-glucosylable Rac1-G12V, but not RhoA-G14A, reverted CT166-induced actin re-organization, suggesting that CT166-induced actin re-organization depends on the glucosylation of Rac1. In accordance, over-expression of CT166-mut diminished TcdB induced cell rounding, suggesting shared substrates. Cell rounding induced by high MOI infection with C. trachomatis D was reduced in cells expressing CT166-mut or Rac1-G12V, and in CNF1 treated cells. These observations indicate that the cytopathic effect of C. trachomatis D is mediated by CT166 induced Rac1 glucosylation. Finally, chlamydial uptake was impaired in CT166 over-expressing cells. Our data strongly suggest CT166's participation as an effector protein during host-cell entry, ensuring a balanced uptake into host-cells by interfering with Rac-dependent cytoskeletal changes.


Bacterial Proteins/metabolism , Chlamydia trachomatis/metabolism , Glucosyltransferases/physiology , rac GTP-Binding Proteins/metabolism , Actins/chemistry , Amino Acid Motifs , Glucose/chemistry , Glucosyltransferases/chemistry , Glucosyltransferases/metabolism , HeLa Cells , Humans , Mutation , Open Reading Frames , Phenotype , Recombinant Proteins/chemistry , rac1 GTP-Binding Protein/biosynthesis , rhoA GTP-Binding Protein/biosynthesis
5.
Biochemistry ; 48(8): 1785-92, 2009 Mar 03.
Article En | MEDLINE | ID: mdl-19199813

Clostridium sordellii lethal toxin (TcsL) belongs to the family of clostridial glucosylating toxins. TcsL exhibits glucosyltransferase activity to inactivate Rho and Ras proteins. On cultured cells, TcsL causes actin reorganization ("cytopathic effect") and apoptotic cell death ("cytotoxic effect"). This study is based on the concept that the cytotoxic effects of TcsL depend on the glucosylation of critical substrate proteins rather than on the glucosyltransferase activity per se. The cytotoxic effects of TcsL depend on the glucosyltransferase activity of TcsL, as neither chemically inactivated TcsL nor a glucosyltransferase-deficient mutant version of TcsL caused it. The TcsL homologous toxin B from Clostridium difficile serotype F strain 1470 (TcdBF) also failed to cause cytotoxic effects. Correlation of the toxins' respective protein substrate specificities highlighted (H/K/N)Ras as critical substrate proteins for the cytotoxic effects. (H/K/N)Ras are critical upstream regulators of phosphatidylinositide 3'-OH kinase (PI3K)/Akt survival signaling. Tauroursodeoxycholic acid (TUDCA) classified to activate PI3K/Akt signaling downstream of apoptosis-inducing stimuli prevented the cytotoxic effects of TcsL. In conclusion, (H/K/N)Ras glucosylation and subsequent inhibition of PI3K/Akt signaling are critical for the cytotoxic effects of TcsL.


Bacterial Toxins/toxicity , Leukemia, Basophilic, Acute/pathology , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction/drug effects , Animals , Bacterial Toxins/chemistry , Caspase 3/metabolism , Cell Death/drug effects , Cell Line, Tumor , Cell Survival/drug effects , Glycosylation/drug effects , Glycosyltransferases/metabolism , Leukemia, Basophilic, Acute/enzymology , Phosphoinositide-3 Kinase Inhibitors , Protein Structure, Tertiary , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Rats , Taurochenodeoxycholic Acid/pharmacology
6.
FASEB J ; 23(4): 1115-26, 2009 Apr.
Article En | MEDLINE | ID: mdl-19047066

Small GTPases of the Rho family play versatile roles in the formation and development of axons and dendrites, effects often studied by the Rho-inactivating C3 transferase (C3bot) from Clostridium botulinum. Recently, we reported that transferase-deficient C3bot also exerted axonotrophic activity. Using overlapping peptides from the C3bot sequence, we identified a small peptide of 29 amino acids (covering residues 154-182) from the C-terminal region of C3bot that promotes both axonal and dendritic growth, as well as branching of hippocampal neurons, at submicromolar concentrations. Several C3bot constructs, including the short peptide, enhanced the number of axonal segments from mid- to higher-order segments. C3bot(154-182) also increased the number of synaptophysin-expressing terminals, up-regulated various synaptic proteins, and functionally increased the glutamate uptake. Staining against the vesicular glutamate and GABA transporters further revealed that the effect was attributable to a higher number of glutamatergic and GABAergic inputs on proximal dendrites of enhanced green fluorescent protein (EGFP)-transfected neurons. Using organotypical slice cultures, we also detected trophic effects of C3bot(154-182) on length and density of outgrowing fibers from the entorhinal cortex that were comparable to the effects elicited by full-length C3bot. In addition, an enhanced reinnervation was observed in a hippocampal-entorhinal lesion model. In summary, the neurotrophic effect of C3bot is executed by a C-terminal peptide fragment covering aa 154-182 of C3; it triggers dendritic and axonal growth and branching as well as increased synaptic connectivity. In contrast to full-length C3, this C3 peptide selectively acts on neurons but not on glial cells.


ADP Ribose Transferases/metabolism , Amino Acids/pharmacology , Axons/physiology , Botulinum Toxins/metabolism , Dendrites/physiology , Neurons/physiology , ADP Ribose Transferases/chemistry , ADP Ribose Transferases/genetics , Amino Acids/chemistry , Animals , Axons/drug effects , Axons/metabolism , Biomarkers/metabolism , Botulinum Toxins/chemistry , Botulinum Toxins/genetics , Cells, Cultured , Clostridium botulinum/genetics , Clostridium botulinum/metabolism , Coculture Techniques , Dendrites/drug effects , Dendrites/genetics , Dendrites/metabolism , Embryo, Mammalian , Glutathione Transferase/metabolism , Green Fluorescent Proteins/metabolism , Hippocampus/cytology , Hippocampus/embryology , Immunohistochemistry , Mice , Mice, Inbred Strains , Microtubule-Associated Proteins/metabolism , Molecular Weight , Neurofilament Proteins/chemistry , Neurofilament Proteins/metabolism , Neurons/drug effects , Neurons/metabolism , Recombinant Proteins/chemistry , Recombinant Proteins/isolation & purification , Recombinant Proteins/metabolism , Time Factors
7.
Glia ; 56(11): 1162-75, 2008 Aug 15.
Article En | MEDLINE | ID: mdl-18442097

Successful regeneration in the central nervous system crucially depends on the adequate environment. Microglia as brain immune-competent cells importantly contribute to this task by producing pro- and anti-inflammatory mediators. Any environmental change transforms these cells towards an activated phenotype, leading to major morphological, transcriptional and functional alterations. Rho GTPases affect multiple cellular properties, including the cytoskeleton, and C3 proteins are widely used to study their involvement. Especially C3bot from Clostridium botulinum has been considered to promote neuronal regeneration by changing Rho activity. Yet C3bot may exert cellular influences through alternative mechanisms. To determine the role of Rho-dependent pathways in microglia we investigated the influence of C3bot on functional properties of cultivated primary mouse microglial cells. Nanomolar concentrations of C3bot transformed microglia towards an activated phenotype and triggered the release of nitric oxide and several proinflammatory cyto- and chemokines. These inductions were not mediated by the ROCK-kinase pathway, since its selective inhibitors Y27632 and H1152 had no effect. C3-induced and Rho-mediated NO release was instead found to be under the control of NFkappaB, as revealed by treatment with the NFkappaB inhibitor PDTC. Thus, C3bot induces a proinflammatory response in microglia resembling the classical proinflammatory phenotype elicited by bacterial LPS. The findings are relevant for the use of C3bot in regenerative approaches.


ADP Ribose Transferases/physiology , Botulinum Toxins/physiology , Clostridium botulinum/enzymology , Inflammation Mediators/physiology , Microglia/enzymology , Microglia/pathology , Signal Transduction/physiology , rho GTP-Binding Proteins/antagonists & inhibitors , Animals , Cells, Cultured , Dose-Response Relationship, Drug , Inflammation/enzymology , Inflammation/microbiology , Lipopolysaccharides/pharmacology , Mice , Microglia/metabolism , Nitric Oxide/metabolism , Nitric Oxide Synthase Type II/metabolism , Phenotype , rho GTP-Binding Proteins/physiology
8.
J Biol Chem ; 283(14): 9289-99, 2008 Apr 04.
Article En | MEDLINE | ID: mdl-18234672

Inhibition of Rho activity by Clostridium botulinum C3 transferase (C3bot) versatily changes functional properties of neural cells. Using cultivated mouse astrocytes, we show here that C3bot increases both uptake and secretion of glutamate. The enhanced glutamate uptake is initiated by an NFkappaB-dependent up-regulation of the glial glutamate transporter 1 that is efficaciously sorted to the plasma membrane. The increase in cytosolic glutamate concentration promotes vesicular glutamate storage in astrocytes treated with C3bot. Parallel to the increased storage, C3-induced impairment of Rho-dependent pathways strongly enhances Ca(2+)-dependent secretion of glutamate. This is accompanied by higher levels of the SNARE protein synaptobrevin. Synaptobrevin inactivation by botulinum neurotoxin D almost completely inhibits Ca(2+)-dependent glutamate secretion triggered by C3bot, indicating that the enhanced release of glutamate mainly originates from exocytosis. In addition, C3bot increases the exocytosis/endocytosis turnover, as analyzed by the stimulated accumulation of the fluorescent dye AM1-43. The release of glutamine, the main metabolite of glutamate, is only moderately affected by C3bot. In conclusion, inhibition of Rho-dependent pathways shifts astrocytes to a secretory active stage in which they may modulate neuronal excitability.


ADP Ribose Transferases/pharmacology , Astrocytes/metabolism , Botulinum Toxins/pharmacology , Glutamic Acid/metabolism , Secretory Vesicles/metabolism , rho GTP-Binding Proteins/metabolism , Animals , Astrocytes/cytology , Calcium/metabolism , Cell Membrane/metabolism , Cells, Cultured , Endocytosis/drug effects , Excitatory Amino Acid Transporter 2/metabolism , Exocytosis/drug effects , Mice , NF-kappa B/metabolism , Pyridinium Compounds/pharmacology , Quaternary Ammonium Compounds/pharmacology , R-SNARE Proteins/metabolism , Up-Regulation/drug effects
9.
J Biol Chem ; 282(22): 16423-33, 2007 Jun 01.
Article En | MEDLINE | ID: mdl-17389603

The repulsive guidance molecule RGMa has been shown to induce outgrowth inhibition of neurites by interacting with the transmembrane receptor neogenin. Here we show that RGMa-induced growth cone collapse is mediated by activation of the small GTPase RhoA, its downstream effector Rho kinase and PKC. In contrast to DRG cultures from neogenin-/- mice, in which no RGMa-mediated growth cone collapse and activation of RhoA occurred, treatment of wild type DRG neurites with soluble RGMa led to a marked activation of RhoA within 3 min followed by collapse, but left Rac1 and Cdc42 unaffected. Furthermore, preincubation of DRG axons with the bone morphogenetic protein (BMP) antagonist noggin had no effect on RGMa-mediated growth cone collapse, implying that the role of RGM in axonal guidance is neogenin- and not BMP receptor-dependent. Pretreatment with 1) C3-transferase, a specific inhibitor of the Rho GTPase; 2) Y-27632, a specific inhibitor of Rho kinase; and 3) Gö6976, the general PKC inhibitor, strongly inhibited the collapse rate of PC12 neurites. Growth cone collapse induced by RGMa was abolished by the expression of dominant negative RhoA, but not by dominant negative Rac1. In contrast to RGMa, netrin-1 induced no growth cone retraction but instead reduced RGMa-mediated growth cone collapse. These results suggest that activation of RhoA, Rho kinase, and PKC are physiologically relevant and important elements of the RGMa-mediated neogenin signal transduction pathway involved in axonal guidance.


Intracellular Signaling Peptides and Proteins/metabolism , Membrane Proteins/metabolism , Nerve Tissue Proteins/metabolism , Neurites/metabolism , Protein Kinase C/metabolism , Protein Serine-Threonine Kinases/metabolism , Signal Transduction/physiology , rhoA GTP-Binding Protein/metabolism , Animals , Bone Morphogenetic Protein Receptors/antagonists & inhibitors , Bone Morphogenetic Protein Receptors/metabolism , Bone Morphogenetic Proteins/antagonists & inhibitors , Bone Morphogenetic Proteins/pharmacology , Carrier Proteins/metabolism , GPI-Linked Proteins , Intracellular Signaling Peptides and Proteins/antagonists & inhibitors , Mice , Mice, Knockout , Nerve Tissue Proteins/deficiency , Nerve Tissue Proteins/pharmacology , Neuropeptides/metabolism , PC12 Cells , Protein Kinase C/antagonists & inhibitors , Protein Kinase Inhibitors/pharmacology , Protein Serine-Threonine Kinases/antagonists & inhibitors , Rats , Signal Transduction/drug effects , rac GTP-Binding Proteins/metabolism , rac1 GTP-Binding Protein/pharmacology , rho-Associated Kinases , rhoA GTP-Binding Protein/antagonists & inhibitors
10.
FEBS Lett ; 580(14): 3565-9, 2006 Jun 12.
Article En | MEDLINE | ID: mdl-16730714

Mono-glucosylation of Rho, Rac, and Cdc42 by Clostridium difficile toxin B (TcdB) induces changes of actin dynamics and apoptosis. When fibroblasts were treated with TcdB, an apparent decrease of the cellular Rac1 level was observed when applying anti-Rac1(Mab 102). This decrease was not based on degradation as inhibition of the proteasome by lactacystin did not stabilise cellular Rac1 levels. The application of anti-Rac1 (Mab 23A8) showed that the cellular Rac1 level slightly increased in TcdB-treated fibroblasts; thus, the apparent loss of cellular Rac1 was not due to degradation but due to impaired recognition of glucosylated Rac1 by anti-Rac1 (Mab 102). In contrast, recognition of RhoA by anti-RhoA (Mab 26C4) and Cdc42 by anti-Cdc42 (Mab 44) was not altered by glucosylation; a transient decrease of cellular RhoA and Cdc42 in TcdB-treated fibroblasts was indeed due to proteasomal degradation, as inhibition of the proteasome by lactacystin stabilised both cellular RhoA and Cdc42 levels. The finding that the apparent decrease of Rac1 reflects Rac1 glucosylation offers a valuable tool to determine Rac1 glucosylation.


Bacterial Proteins/metabolism , Bacterial Toxins/metabolism , rho GTP-Binding Proteins/metabolism , Animals , Bacterial Proteins/pharmacology , Bacterial Toxins/pharmacology , Cell-Free System , Fibroblasts/drug effects , Fibroblasts/metabolism , Glycosylation , Mice , NIH 3T3 Cells
11.
J Neurochem ; 95(5): 1237-48, 2005 Dec.
Article En | MEDLINE | ID: mdl-16150054

Small Rho GTPases are key regulators of the cytoskeleton in a great variety of cells. Rho function mediates morphological changes as well as locomotor activity. Using astrocyte cultures established from neonatal mice we investigated the role of Rho in process formation during astrocyte stellation. Using a scratch-wound model, we examined the impact of Rho on a variety of morphological and functional variables such as stellation and migratory activity during wound healing. C3 proteins are widely used to study cellular Rho functions. In addition, C3 derived from Clostridium botulinum (C3bot) is considered selectively to promote neuronal regeneration. Because the latter requires a balanced activity of neurones and glial cells, the effects of C3 protein on glial cells such as astrocytes have to be considered carefully. Low nanomolar concentrations of C3 proteins significantly promoted process outgrowth and increased process branching. Besides enzymatic inactivation of Rho by ADP-ribosylation, changes in protein levels of the various Rho GTPases may also contribute to the observed effects. Furthermore, incubation of scratch-wounded astrocyte cultures with C3bot accelerated wound healing. By inhibiting the Rho downstream effector ROCK with the selective inhibitor Y27632 we were able to demonstrate that the accelerated wound closure resulted from both enhanced polarized process formation and increased migratory activity of astrocytes into the lesion site. These results suggest that Rho negatively regulates astrocytic process growth and migratory responses after injury and that its inactivation by C3bot in nanomolar concentrations promotes astrocyte migration.


Astrocytes/cytology , Cell Movement/physiology , Wound Healing/physiology , rho GTP-Binding Proteins/physiology , Amides/pharmacology , Animals , Animals, Newborn , Astrocytes/drug effects , Brain/cytology , Cell Movement/drug effects , Cell Proliferation/drug effects , Cell Size/drug effects , Cell Survival/drug effects , Cell Survival/physiology , Cells, Cultured , Complement C3/pharmacology , Dose-Response Relationship, Drug , Enzyme Inhibitors/pharmacology , Fluorescent Antibody Technique/methods , Gene Expression Regulation/drug effects , Gene Expression Regulation/physiology , Glial Fibrillary Acidic Protein/metabolism , Indoles , L-Lactate Dehydrogenase , Lectins/metabolism , Liposomes/pharmacology , Mice , Pyridines/pharmacology , Recombinant Proteins/pharmacology , Tetrazolium Salts , Time Factors , Wound Healing/drug effects , cdc42 GTP-Binding Protein/metabolism
12.
J Immunol ; 175(5): 2960-7, 2005 Sep 01.
Article En | MEDLINE | ID: mdl-16116182

Sphingosine-1-phosphate (S1P) represents a potent modulator of diverse cellular activities, including lymphocyte trafficking and maintenance of lymphocyte homeostasis. The five known receptors for S1P (S1P(1-5)) belong to the family of G protein-coupled receptors. Upon binding S1P, they act downstream via heterotrimeric G proteins on members of the small GTPase family (Cdc42/Rac/Rho), evoking a S1P receptor-dependent activation pattern of Cdc42, Rac, and Rho, respectively. This, in turn, triggers cytoskeletal rearrangements determining cellular morphology and movement. In this study we investigated the effects of S1P on murine dendritic cells (DC). Mature DC, but not immature in vitro differentiated DC, were found to migrate to S1P, a phenomenon that correlated to the up-regulation of S1P1 and S1P3 in maturing DC. The same pattern of S1P receptor regulation could be observed in vivo on skin DC after their activation and migration into the lymph node. The migration-inducing effect of S1P could be severely hampered by application of the S1P analogon FTY720 in vitro and in vivo. A similar, yet more pronounced, block was observed upon preventing Cdc42/Rac and/or Rho activation by specific inhibitors. These results suggest that S1P-mediated signaling plays a pivotal role in the life cycle of DC.


Dendritic Cells/drug effects , Lysophospholipids/pharmacology , Sphingosine/analogs & derivatives , Animals , Cell Differentiation , Cell Movement/drug effects , Dendritic Cells/physiology , Fingolimod Hydrochloride , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Propylene Glycols/pharmacology , Receptors, Lysosphingolipid/analysis , Receptors, Lysosphingolipid/physiology , Signal Transduction , Sphingosine/pharmacology
13.
Microb Pathog ; 38(2-3): 77-83, 2005.
Article En | MEDLINE | ID: mdl-15748809

Toxins A and B from Clostridium difficile are single-chain proteins of 308,000 and 270,000 Da, respectively. They possess transferase activity to monoglucosylate proteins of the Rho GTPase family whereby Rho, Rac, and Cdc42 are the canonical substrates. For application of these toxins as specific Rho GTPase inhibitors the highest possible purity is of crucial interest. We, therefore, expressed recombinant His-tagged toxin A using the Bacillus megaterium expression system. Specific antisera raised against the native toxin A from C. difficile and the recombinant toxin, respectively, showed identical sensitivity and specificity in Western blot and ELISA analyses towards both toxins. By comparison of both toxins in functional studies we showed that the recombinant toxin was about two times more cytotoxic than the native toxin, and the glucosyltransferase-activity of the recombinant toxin was even 10-fold increased. However, recombinant toxin A showed one essential difference to the classically purified one. The reported transferase-independent effect of toxin A to release cytochrome c from isolated mitochondria was not exhibited by the recombinant toxin A. This putative mitochondrial effect decreased with increased purity of toxin A, and was absent with recombinant toxin, strongly suggesting an clostridial contamination responsible. In summary, we tested the recombinant toxin A to be at least an adequate substitute for the native toxin, bearing the advantage of a rapid single-step purification and the absence of biological active contaminations.


Bacterial Toxins/metabolism , Bacterial Toxins/toxicity , Clostridioides difficile/enzymology , Enterotoxins/metabolism , Enterotoxins/toxicity , Animals , Antibodies, Bacterial/immunology , Bacillus megaterium/genetics , Bacillus megaterium/metabolism , Bacterial Toxins/genetics , Bacterial Toxins/immunology , Blotting, Western , Cells, Cultured , Cloning, Molecular , Cytochromes c/analysis , Enterotoxins/genetics , Enterotoxins/immunology , Enzyme-Linked Immunosorbent Assay , Glucosyltransferases/genetics , Glucosyltransferases/immunology , Glucosyltransferases/isolation & purification , Glucosyltransferases/metabolism , Glucosyltransferases/toxicity , Humans , Mice , Mitochondria/drug effects , NIH 3T3 Cells , Recombinant Proteins/genetics , Recombinant Proteins/immunology , Recombinant Proteins/metabolism , Recombinant Proteins/toxicity
14.
J Thorac Cardiovasc Surg ; 128(6): 900-6, 2004 Dec.
Article En | MEDLINE | ID: mdl-15573075

OBJECTIVE: We sought to grow in vitro functional smooth muscle cells, chondrocytes, and respiratory epithelium on a biologic, directly vascularized matrix as a scaffold for tracheal tissue engineering. METHODS: Ten- to 15-cm-long free jejunal segments with their own vascular pedicle were harvested and acellularized from donor pigs (n = 10) and used as a vascular matrix. Autologous costal chondrocytes, smooth muscle cells, and respiratory epithelium and endothelial progenitor cells were first cultured in vitro and then disseminated on the previously acellularized vascular matrix. Histologic, immunohistologic, molecular imaging, and Western blotting studies were then performed to assess cell viability. RESULTS: The endothelial progenitor cells re-endothelialized the matrix to such an extent that endothelial cell viability was uniformly documented through 2-(18F)-fluoro-2'-deoxyglucose positron emission tomography. This vascularized scaffold was seeded with functional (according to Western blot analysis) smooth muscle cells and successfully reseeded with viable ciliated respiratory epithelium. Chondrocyte growth and production of extracellular cartilaginous matrix was observed as soon as 2 weeks after their culture. CONCLUSIONS: The fundamental elements for a bioartificial trachea were successfully engineered in vitro in a direct vascularized 10- to 15-cm-long bioartificial matrix. Future experimental work will be directed to give them a 3-dimensional aspect and a biomechanical profile of a functioning trachea.


Bioartificial Organs , Tissue Engineering , Trachea , Animals , Cells, Cultured , Immunohistochemistry , Myocytes, Smooth Muscle/physiology , Stem Cells/metabolism , Swine
15.
Biochem Biophys Res Commun ; 307(3): 584-8, 2003 Aug 01.
Article En | MEDLINE | ID: mdl-12893263

Pathogenic Clostridium difficile produces two major protein toxins, toxin A and toxin B. We used the Bacillus megaterium expression system for expression of recombinant toxin A. The construct for the toxin A gene was obtained by the following cloning strategy: the gene for toxin A was generated in three parts, each of them ligated into a cloning vector. The three parts were sequentially fused to the complete gene. The holotoxin gene was ligated into the expression vector pWH1520. This vector was modified to generate a toxin with a C-terminally located His-tag. Gene expression in the B. megaterium system resulted in an approximate 300 kDa protein, which was identified by specific antibody as toxin A. Recombinant, His-tagged toxin A was purified by Ni(2+) as well as thyroglobulin affinity chromatography. Characterization of the recombinant toxin A showed identical cytotoxicity and in vitro-glucosyltransferase activity as the native toxin A from C. difficile.


Bacillus megaterium/genetics , Bacterial Toxins/genetics , Enterotoxins/genetics , Animals , Bacterial Toxins/metabolism , Bacterial Toxins/toxicity , Cell Line , Enterotoxins/metabolism , Enterotoxins/toxicity , Gene Expression , Genetic Vectors , Recombinant Proteins/isolation & purification , Recombinant Proteins/metabolism , Recombinant Proteins/toxicity
16.
Biochem Biophys Res Commun ; 300(3): 706-11, 2003 Jan 17.
Article En | MEDLINE | ID: mdl-12507507

Clostridium difficile toxin A, the chief pathogenicity factor of the antibiotic-associated pseudomembranous colitis, is an intracellular acting cytotoxin that reaches its targets, the Rho GTPases, after receptor-mediated endocytosis. The C-terminal part, constructed of repetitive peptide elements, is thought to bind to a lot of carbohydrate containing receptor molecules to induce clustering and endocytosis. To study which part of the receptor-binding domain is in charge of addressing toxin A into the target cells, we studied the functional, i.e., endocytosis-inducing, binding of toxin A. By a competition assay between various receptor-binding fragments of toxin A and the holotoxin A we found that the complete receptor-binding domain, encompassing the entire repetitive elements, but not parts of it, is necessary for binding-induced endocytosis. The receptor binding domain itself shows weaker competition with holotoxin A than the fragment consisting of receptor-binding domain plus intermediary part of the toxin. All toxin A fragments that compete with holotoxin A are capable of inducing their own endocytosis. Thus, the entire receptor-binding domain, covering the C-terminal third of the toxin A molecule, is responsible for cell uptake of toxin A and the intermediary part contributes to the correct folding and assembly of the repetitive domains.


Bacterial Toxins/chemistry , Endocytosis/physiology , Enterotoxins/chemistry , Receptors, Cell Surface/metabolism , Bacterial Toxins/metabolism , Bacterial Toxins/toxicity , Binding, Competitive/physiology , Cell Count , Cell Survival/drug effects , Endocytosis/drug effects , Enterotoxins/metabolism , Enterotoxins/toxicity , HT29 Cells , Humans , Peptide Fragments/chemistry , Peptide Fragments/metabolism , Peptide Fragments/toxicity , Protein Binding/physiology , Protein Structure, Tertiary/physiology , Temperature
17.
J Cell Biol ; 160(3): 375-85, 2003 Feb 03.
Article En | MEDLINE | ID: mdl-12551955

Neutrophils exposed to chemoattractants polarize and accumulate polymerized actin at the leading edge. In neutrophil-like HL-60 cells, this asymmetry depends on a positive feedback loop in which accumulation of a membrane lipid, phosphatidylinositol (PI) 3,4,5-trisphosphate (PI[3,4,5]P3), leads to activation of Rac and/or Cdc42, and vice versa. We now report that Rac and Cdc42 play distinct roles in regulating this asymmetry. In the absence of chemoattractant, expression of constitutively active Rac stimulates accumulation at the plasma membrane of actin polymers and of GFP-tagged fluorescent probes for PI(3,4,5)P3 (the PH domain of Akt) and activated Rac (the p21-binding domain of p21-activated kinase). Dominant negative Rac inhibits chemoattractant-stimulated accumulation of actin polymers and membrane translocation of both fluorescent probes and attainment of morphologic polarity. Expression of constitutively active Cdc42 or of two different protein inhibitors of Cdc42 fails to mimic effects of the Rac mutants on actin or PI(3,4,5)P3. Instead, Cdc42 inhibitors prevent cells from maintaining a persistent leading edge and frequently induce formation of multiple, short lived leading edges containing actin polymers, PI(3,4,5)P3, and activated Rac. We conclude that Rac plays a dominant role in the PI(3,4,5)P3-dependent positive feedback loop required for forming a leading edge, whereas location and stability of the leading edge are regulated by Cdc42.


Cell Polarity/physiology , Chemotaxis, Leukocyte/physiology , Inositol Phosphates/metabolism , N-Formylmethionine Leucyl-Phenylalanine/analogs & derivatives , Neutrophils/metabolism , Pseudopodia/metabolism , cdc42 GTP-Binding Protein/metabolism , rac GTP-Binding Proteins/metabolism , Actins/drug effects , Actins/metabolism , Animals , Bacterial Toxins/pharmacology , COS Cells , Cell Membrane/drug effects , Cell Membrane/metabolism , Feedback, Physiological/drug effects , Feedback, Physiological/genetics , Green Fluorescent Proteins , HL-60 Cells , Humans , Inositol Phosphates/genetics , Luminescent Proteins , N-Formylmethionine Leucyl-Phenylalanine/pharmacology , Neutrophils/cytology , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Protein Transport/drug effects , Protein Transport/physiology , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-akt , Pseudopodia/ultrastructure , Recombinant Fusion Proteins , cdc42 GTP-Binding Protein/genetics , p21-Activated Kinases , rac GTP-Binding Proteins/genetics
...