Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 112
Filter
Add more filters











Publication year range
1.
Physiol Res ; 72(1): 87-97, 2023 03 08.
Article in English | MEDLINE | ID: mdl-36545878

ABSTRACT

Carnosine is a performance-enhancing food supplement with a potential to modulate muscle energy metabolism and toxic metabolites disposal. In this study we explored interrelations between carnosine supplementation (2 g/day, 12 weeks) induced effects on carnosine muscle loading and parallel changes in (i) muscle energy metabolism, (ii) serum albumin glycation and (iii) reactive carbonyl species sequestering in twelve (M/F=10/2) sedentary, overweight-to-obese (BMI: 30.0+/-2.7 kg/m2) adults (40.1+/-6.2 years). Muscle carnosine concentration (Proton Magnetic Resonance Spectroscopy; 1H-MRS), dynamics of muscle energy metabolism (Phosphorus Magnetic Resonance Spectroscopy; 31P-MRS), body composition (Magnetic Resonance Imaging; MRI), resting energy expenditure (indirect calorimetry), glucose tolerance (oGTT), habitual physical activity (accelerometers), serum carnosine and carnosinase-1 content/activity (ELISA), albumin glycation, urinary carnosine and carnosine-propanal concentration (mass spectrometry) were measured. Supplementation-induced increase in muscle carnosine was paralleled by improved dynamics of muscle post-exercise phosphocreatine recovery, decreased serum albumin glycation and enhanced urinary carnosine-propanal excretion (all p<0.05). Magnitude of supplementation-induced muscle carnosine accumulation was higher in individuals with lower baseline muscle carnosine, who had lower BMI, higher physical activity level, lower resting intramuscular pH, but similar muscle mass and dietary protein preference. Level of supplementation-induced increase in muscle carnosine correlated with reduction of protein glycation, increase in reactive carbonyl species sequestering, and acceleration of muscle post-exercise phosphocreatine recovery.


Subject(s)
Carnosine , Humans , Adult , Carnosine/metabolism , Carnosine/pharmacology , Maillard Reaction , Phosphocreatine/metabolism , Muscle, Skeletal/metabolism , Dietary Supplements
2.
J Artif Organs ; 24(3): 307-311, 2021 Sep.
Article in English | MEDLINE | ID: mdl-33846899

ABSTRACT

Allograft failure secondary to rejection commonly requires a multimodal treatment, ultimately including mechanical circulatory support. A few case reports have demonstrated the use of Impella-devices due to its assumed favorable safety profile in this fragile cohort. However, this treatment option does not play a role in choice of anti-rejective therapy in clinical routine up to date. We summarize our institutional experiences and literature mini-review on Impella-based treatment strategies in allograft rejection after heart transplantation. In all seven cases, three from our institution and four reported in the literature, Impella-based therapies led to hemodynamic stabilization in allograft failure secondary to rejection. Adverse events included hemolysis, non-fatal bleeding and in one patient a relevant aortic valve insufficiency occurred. All patients showed an improvement of allograft function. Two patients died in context of severe immunosuppression or late secondary organ failure. Based on the limited available data, we propose that Impella-mediated mechanical unloading represents a valuable option for hemodynamic stabilization in severe allograft failure due to rejection, enabling an initiation of causal therapy and thereby potentially representing an opportunity to prevent mortality. Furthermore, we hypothesize it might add to the traditional therapeutic approaches by facilitating recovery by decompressing the myocardium in allograft rejection.


Subject(s)
Heart Failure , Heart Transplantation , Heart-Assist Devices , Allografts , Graft Rejection , Heart Failure/etiology , Heart Failure/surgery , Humans , Treatment Outcome
3.
Brain ; 135(Pt 7): 2022-31, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22586289

ABSTRACT

Cerebral folate transport deficiency is an inherited brain-specific folate transport defect that is caused by mutations in the folate receptor 1 gene coding for folate receptor alpha (FRα). This genetic defect gives rise to a progressive neurological disorder with late infantile onset. We screened 72 children with low 5-methyltetrahydrofolate concentrations in the cerebrospinal fluid and neurological symptoms that developed after infancy. We identified nucleotide alterations in the folate receptor 1 gene in 10 individuals who shared developmental regression, ataxia, profound cerebral hypomyelination and cerebellar atrophy. We found four novel pathogenic alleles, one splice mutation and three missense mutations. Heterologous expression of the missense mutations, including previously described mutants, revealed minor decrease in protein expression but loss of cell surface localization, mistargeting to intracellular compartments and thus absence of cellular binding of folic acid. These results explain the functional loss of folate receptor alpha for all detected folate receptor 1 mutations. Three individuals presenting a milder clinical phenotype revealed very similar biochemical and brain imaging data but partially shared pathogenic alleles with more severely affected patients. Thus, our studies suggest that different clinical severities do not necessarily correlate with residual function of folate receptor alpha mutants and indicate that additional factors contribute to the clinical phenotype in cerebral folate transport deficiency.


Subject(s)
Folate Receptor 1/metabolism , Folic Acid Deficiency/genetics , Folic Acid/metabolism , Mutation/genetics , Adolescent , Alleles , Animals , CHO Cells , Child , Child, Preschool , Cricetinae , Female , Fibroblasts/metabolism , Folate Receptor 1/genetics , Folic Acid Deficiency/diagnosis , Hep G2 Cells , Humans , Magnetic Resonance Imaging/methods , Male , Phenotype , Protein Transport/genetics , Tetrahydrofolates/cerebrospinal fluid
4.
J Chromatogr B Analyt Technol Biomed Life Sci ; 877(13): 1344-51, 2009 May 01.
Article in English | MEDLINE | ID: mdl-19109081

ABSTRACT

Proteomic approaches are used to identify biomarkers, to monitor pathological changes inside of cells and for a better diseases diagnosis. Comparable changes in protein homeostasis also occur in differentiating cells and proteomic techniques should be suitable to identify biomarkers that indicate different steps of cellular development. The C3 exoenzyme from Clostridium botulinum (C3bot) inactivates Rho GTPases and induces morphological cellular changes like cell rounding and neurite outgrowth [G. Ahnert-Hilger, M. Höltje, G. Grosse, G. Pickert, C. Mucke, B. Nixdorf-Bergweiler, P. Boquet, F. Hofmann, I. Just, J. Neurochem. 90 (2004) 9]. To investigate these observations further a comparative proteomic approach has been chosen to elucidate C3bot effects in the neuroblastoma cell line model SH-SY5Y. The screening method applied for biomarker detection was based on the stable isotope approach isobaric tagging for relative and absolute quantification (iTRAQ). Proteins of C3bot-treated and untreated cells were digested and peptides were labeled by the iTRAQ reagent, combined, and separated by means of a two-dimensional nano-HPLC system. Peptide analysis was performed in a MALDI-TOF/TOF mass spectrometer. Identification and quantification of peptides and their corresponding proteins were accomplished by MS/MS spectra analysis. Overall, five replicate measurements identified 355 different proteins of which 235 were accessible for quantification. C3bot altered the concentration of 55 proteins (at least 1.3-fold) and several proteins were identified as possible biomarker candidates that indicate C3bot-induced cellular changes.


Subject(s)
ADP Ribose Transferases/pharmacology , Biomarkers/metabolism , Botulinum Toxins/pharmacology , Chromatography, High Pressure Liquid/methods , Clostridium botulinum/enzymology , Neurons/drug effects , Proteomics , Tandem Mass Spectrometry/methods , Cell Line, Tumor , Humans , Neurons/metabolism , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization/methods
5.
Curr Top Microbiol Immunol ; 291: 113-45, 2005.
Article in English | MEDLINE | ID: mdl-15981462

ABSTRACT

Rho proteins are master regulators of a large array of cellular functions, including control of cell morphology, cell migration and polarity, transcriptional activation, and cell cycle progression. They are the eukaryotic targets of various bacterial protein toxins and effectors, which activate or inactivate the GTPases. Here Rho-inactivating toxins and effectors are reviewed, including the families of large clostridial cytotoxins and C3-like transferases, which inactivate Rho GTPases by glucosylation and ADP-ribosylation, respectively.


Subject(s)
Bacterial Toxins/toxicity , Clostridium/pathogenicity , Virulence Factors/toxicity , rho GTP-Binding Proteins/antagonists & inhibitors , ADP Ribose Transferases/metabolism , Animals , Bacterial Toxins/chemistry , Bacterial Toxins/metabolism , Glycosylation , Humans , Virulence Factors/chemistry , Virulence Factors/metabolism , rho GTP-Binding Proteins/metabolism
6.
Article in English | MEDLINE | ID: mdl-15449191

ABSTRACT

The large clostridial cytotoxins are a family of structurally and functionally related exotoxins from Clostridium difficile (toxins A and B), C. sordellii (lethal and hemorrhagic toxin) and C. novyi (alpha-toxin). The exotoxins are major pathogenicity factors which in addition to their in vivo effects are cytotoxic to cultured cell lines causing reorganization of the cytoskeleton accompanied by morphological changes. The exotoxins are single-chain protein toxins, which are constructed of three domains: receptor-binding, translocation and catalytic domain. These domains reflect the self-mediated cell entry via receptor-mediated endocytosis, translocation into the cytoplasm, and execution of their cytotoxic activity by an inherent enzyme activity. Enzymatically, the toxins catalyze the transfer of a glucosyl moiety from UDP-glucose to the intracellular target proteins which are the Rho and Ras GTPases. The covalent attachment of the glucose moiety to a conserved threonine within the effector region of the GTPases renders the Rho-GTPases functionally inactive. Whereas the molecular mode of cytotoxic effects is fully understood, the mechanisms leading to inflammatory processes in the context of disease (e.g., antibiotic-associated pseudomembranous colitis caused by Clostridium difficile) are less clear.


Subject(s)
Bacterial Toxins/chemistry , Bacterial Toxins/pharmacology , Clostridium/chemistry , Cytotoxins/chemistry , Cytotoxins/pharmacology , Glucosyltransferases/metabolism , Glycosylation
7.
J Neurochem ; 90(1): 9-18, 2004 Jul.
Article in English | MEDLINE | ID: mdl-15198662

ABSTRACT

Formation of neurites and their differentiation into axons and dendrites requires precisely controlled changes in the cytoskeleton. While small GTPases of the Rho family appear to be involved in this regulation, it is still unclear how Rho function affects axonal and dendritic growth during development. Using hippocampal neurones at defined states of differentiation, we have dissected the function of RhoA in axonal and dendritic growth. Expression of a dominant negative RhoA variant inhibited axonal growth, whereas dendritic growth was promoted. The opposite phenotype was observed when a constitutively active RhoA variant was expressed. Inactivation of Rho by C3-catalysed ADP-ribosylation using C3 isoforms (Clostridium limosum, C3(lim) or Staphylococcus aureus, C3(stau2)), diminished axonal branching. By contrast, extracellularly applied nanomolar concentrations of C3 from C. botulinum (C3(bot)) or enzymatically dead C3(bot) significantly increased axon growth and axon branching. Taken together, axonal development requires activation of RhoA, whereas dendritic development benefits from its inactivation. However, extracellular application of enzymatically active or dead C3(bot) exclusively promotes axonal growth and branching suggesting a novel neurotrophic function of C3 that is independent from its enzymatic activity.


Subject(s)
Axons/physiology , Dendrites/physiology , Hippocampus/physiology , Neurons/physiology , rho GTP-Binding Proteins/metabolism , ADP Ribose Transferases/genetics , ADP Ribose Transferases/metabolism , ADP Ribose Transferases/pharmacology , Adenosine Diphosphate Ribose/metabolism , Animals , Axons/drug effects , Axons/enzymology , Botulinum Toxins/genetics , Botulinum Toxins/metabolism , Botulinum Toxins/pharmacology , Cells, Cultured , Dendrites/drug effects , Dendrites/enzymology , Gene Transfer Techniques , Genes, Dominant , Hippocampus/enzymology , Hippocampus/ultrastructure , Isoenzymes/genetics , Isoenzymes/metabolism , Isoenzymes/pharmacology , Mice , Mice, Inbred Strains , Neurons/enzymology , Neurons/ultrastructure , Phenotype , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Recombinant Proteins/pharmacology , rho GTP-Binding Proteins/antagonists & inhibitors , rho GTP-Binding Proteins/genetics
8.
Am J Physiol Gastrointest Liver Physiol ; 285(5): G1049-55, 2003 Nov.
Article in English | MEDLINE | ID: mdl-12801886

ABSTRACT

This study investigated whether toxin B of Clostridium difficile can activate human submucosal neurons and the involved pathways. Isolated segments of human colon were placed in organ culture for 3 h in the presence of toxin B or IL-1beta. Whole mounts of internal submucosal plexus were stained with antibodies against c-Fos, neuron-specific enolase (NSE), vasoactive intestinal polypeptide (VIP), and substance P (SP). The membrane potential (Vm) response of submucosal neurons to local application of toxin B and IL-1beta was determined by a multisite optical recording technique. Toxin B (0.1 to 10 ng/ml) increased the proportion of c-Fos-positive neurons dose dependently compared with the control. In the presence of toxin B (10 ng/ml), most c-Fos-positive neurons were immunoreactive for VIP (79.8 +/- 22.5%) but only 19.4 +/- 14.0% for SP. Toxin B induced a rapid rise in IL-1beta mRNA level and a sixfold increase in IL-1beta protein in supernatant after 3 h of incubation. c-Fos expression induced by toxin B was reduced dose dependently by IL-1 receptor antagonist (0.1-10 ng/ml). IL-1beta significantly increased c-Fos expression in submucosal neurons compared with the control (34.2 +/- 10.1 vs. 5.1 +/- 1.3% of NSE neurons). Microejection of toxin B had no effect on the Vm of enteric neurons. Evidence of a direct excitatory effect of IL-1beta on Vm was detected in a minority of enteric neurons. Therefore, toxin B of C. difficile activates VIP-positive submucosal neurons, at least in part, via an indirect IL-1beta-dependent pathway.


Subject(s)
Bacterial Proteins , Bacterial Toxins/pharmacology , Colon/innervation , Interleukin-1/physiology , Neurons/physiology , Submucous Plexus/physiology , Vasoactive Intestinal Peptide/metabolism , Aged , Humans , Immunohistochemistry , In Vitro Techniques , Interleukin-1/pharmacology , Membrane Potentials/drug effects , Neurons/drug effects , Neurons/metabolism , Proto-Oncogene Proteins c-fos/metabolism , Submucous Plexus/cytology , Submucous Plexus/drug effects , Submucous Plexus/metabolism
9.
Int J Med Microbiol ; 291(4): 243-50, 2001 Sep.
Article in English | MEDLINE | ID: mdl-11680784

ABSTRACT

The Rho GTPases, which belong to the Ras superfamily of low-molecular-mass GTP-binding proteins, are the preferred intracellular targets of bacterial protein toxins. The Rho GTPases RhoA/B/C, Rac1/2 and Cdc42 are the master regulators of the actin cytoskeleton. Clostridium difficile toxins A and B, the causative agents of the antibiotic-associated pseudomembranous colitis, are intracellularly acting cytotoxins which mono-glucosylate the Rho GTPases. Clostridium botulinum C3 toxin, which is not related to the clostridial neurotoxins, catalyses ADP-ribosylation of RhoA/B/C but not of other Rho GTPases. Glucosylation as well as ADP-ribosylation result in functional inactivation of Rho causing disassembly of the actin cytoskeleton.


Subject(s)
Bacterial Toxins/pharmacology , rho GTP-Binding Proteins/antagonists & inhibitors , Animals , Botulism/microbiology , Botulism/physiopathology , Cells, Cultured , Clostridioides difficile/metabolism , Clostridioides difficile/pathogenicity , Clostridium botulinum/metabolism , Clostridium botulinum/pathogenicity , Enterocolitis, Pseudomembranous/microbiology , Enterocolitis, Pseudomembranous/physiopathology , Humans
10.
Pflugers Arch ; 442(5): 675-87, 2001 Aug.
Article in English | MEDLINE | ID: mdl-11512023

ABSTRACT

Clostridium botulinum C2 toxin (C2 toxin) and purified ADP-ribosylated-alpha-actin (ADP-r-alpha-actin) cause specific actin depolymerisation in living cells. This effect was used to investigate the actin microfilament system with particular emphasis on cell-cell adhesion and plasma membrane integrity in endothelial cells. C2 toxin caused time- and dose-dependent (15-100 ng/ml) changes in endothelial surface morphology (investigated by atomic force microscopy), intercellular gap formation and cell detachment under shear stress. Low concentrations of C2 toxin (1.5 ng/ml), however, did not induce cell detachment but inhibited shear stress-dependent cell alignment. Gap formation as well as cell loss under shear stress was also observed in cells microinjected with purified ADP-r-alpha-actin. Intercellular gap formation was mediated by increased alpha-catenin solubility (40%) due to actin filament depolymerisation. Disintegration of plasma membranes (measured by LDH release) and cell fragmentation during simultaneous exposure to shear stress and C2 toxin were due to a loss of more than 50% of membrane-associated actin. These data show that small disturbances in actin dynamics inhibit shear stress-dependent cell alignment; that depolymerisation of actin filaments increases the solubility of alpha-catenin, thus resulting in cell dissociation and that actin filaments of the membrane cytoskeleton are required to protect the cells from haemodynamic injury such as shear stress. Together, the study shows a heterogeneous regulation of actin filament dynamics at subcellular locations. Junction-associated actin filaments displayed the highest sensitivity whereas stress fibres were far more stable.


Subject(s)
Actin Cytoskeleton/physiology , Actins/metabolism , Cell Adhesion/physiology , Endothelium, Vascular/cytology , Intercellular Junctions/metabolism , Stress, Mechanical , Animals , Botulinum Toxins/pharmacology , Cadherins/metabolism , Cell Fractionation , Cell Membrane/metabolism , Cell Surface Extensions/metabolism , Cells, Cultured , Cytoskeletal Proteins/metabolism , Endothelium, Vascular/drug effects , Endothelium, Vascular/metabolism , Immunoblotting , Microinjections , Microscopy, Atomic Force , Poly(ADP-ribose) Polymerases/metabolism , Polymers/metabolism , Swine , alpha Catenin
11.
J Biol Chem ; 276(12): 9537-42, 2001 Mar 23.
Article in English | MEDLINE | ID: mdl-11124969

ABSTRACT

Clostridium botulinum C3 is the prototype of the family of the C3-like transferases that ADP-ribosylate exclusively RhoA, -B and -C. The ADP-ribose at Asn-41 results in functional inactivation of Rho reflected by disaggregation of the actin cytoskeleton. We report on a new C3-like transferase produced by a pathogenic Staphylococcus aureus strain. The transferase designated C3(Stau) was cloned from the genomic DNA. At the amino acid level, C3(Stau) revealed an identity of 35% to C3 from C. botulinum and Clostridium limosum exoenzyme, respectively, and of 78% to EDIN from S. aureus. In addition to RhoA, which is the target of the other C3-like transferases, C3(Stau) modified RhoE and Rnd3. RhoE was ADP-ribosylated at Asn-44, which is equivalent to Asn-41 of RhoA. RhoE and Rnd3 are members of the Rho subfamily, which are deficient in intrinsic GTPase activity and possess a RhoA antagonistic cell function. The protein substrate specificity found with recombinant Rho proteins was corroborated by expression of RhoE in Xenopus laevis oocytes showing that RhoE was also modified in vivo by C3(Stau) but not by C3 from C. botulinum. The poor cell accessibility of C3(Stau) was overcome by generation of a chimeric toxin recruiting the cell entry machinery of C. botulinum C2 toxin. The chimeric C3(Stau) caused the same morphological and cytoskeletal changes as the chimeric C. botulinum C3. C3(Stau) is a new member of the family of the C3-like transferases but is also the prototype of a subfamily of RhoE/Rnd modifying transferases.


Subject(s)
ADP Ribose Transferases/metabolism , Botulinum Toxins , GTPase-Activating Proteins/metabolism , Poly(ADP-ribose) Polymerases/metabolism , Staphylococcus aureus/enzymology , Amino Acid Sequence , Base Sequence , DNA, Bacterial , Molecular Sequence Data , Sequence Homology, Amino Acid
12.
EMBO J ; 19(23): 6361-70, 2000 Dec 01.
Article in English | MEDLINE | ID: mdl-11101509

ABSTRACT

The pathogenic bacterium Helicobacter pylori produces the cytotoxin VacA, which is implicated in the genesis of gastric epithelial lesions. By transfect ing HEp-2 cells with DNAs encoding either the N-terminal (p34) or the C-terminal (p58) fragment of VacA, p34 was found localized specifically to mitochondria, whereas p58 was cytosolic. Incubated in vitro with purified mitochondria, VacA and p34 but not p58 translocated into the mitochondria. Microinjection of DNAs encoding VacA-GFP and p34-GFP, but not GFP-VacA or GFP-p34, induced cell death by apoptosis. Transient transfection of HeLa cells with p34-GFP or VacA-GFP induced the release of cytochrome c from mitochondria and activated the executioner caspase 3, as determined by the cleavage of poly(ADP-ribose) polymerase (PARP). PARP cleavage was antagonized specifically by co-transfection of DNA encoding Bcl-2, known to block mitochondria-dependent apoptotic signals. The relevance of these observations to the in vivo mechanism of VacA action was supported by the fact that purified activated VacA applied externally to cells induced cytochrome c release into the cytosol.


Subject(s)
Bacterial Proteins/metabolism , Cytochrome c Group/metabolism , Mitochondria/metabolism , Animals , Apoptosis , Caspase 3 , Caspases/metabolism , Cell Line , Cell Nucleus/metabolism , Cytosol/metabolism , Digitonin/pharmacology , Electrophoresis, Polyacrylamide Gel , Fluorescent Antibody Technique , Green Fluorescent Proteins , HeLa Cells , Humans , Immunohistochemistry , Luminescent Proteins/metabolism , Microscopy, Electron , Poly(ADP-ribose) Polymerases/metabolism , Proto-Oncogene Proteins c-bcl-2/metabolism , Rabbits , Reticulocytes/metabolism , Stomach Diseases/metabolism , Transfection
15.
J Mol Biol ; 301(5): 1091-5, 2000 Sep 01.
Article in English | MEDLINE | ID: mdl-10966807

ABSTRACT

Mono-glucosylation of Ha-Ras by Clostridium sordellii lethal toxin at effector region threonine 35 has diverse effects on the Ras GTPase cycle, the dominant one of which is the inhibition of Ras-Raf coupling, leading to complete blockade of Ras downstream signaling. To understand the structural basis of the functional consequences of glucosylation, the X-ray crystal structure of glucosylated Ras-GDP was compared with that of non-modified Ras. Glucosylated Ras exhibits a different crystal packing but the overall three-dimensional structure is not altered. The glucose group does not affect the conformation of the effector loop. Due to steric constraints, the glucose moiety prevents the formation of the GTP conformation of the effector loop, which is a prerequisite for binding to the Raf-kinase. The X-ray crystal data also revealed the alpha-anomeric configuration of the bound glucose, indicating that the glucose transfer proceeds under retention of the C-1 configuration of the d-alpha-glucose. Therefore, glucosylation preserves the inactive conformation of the effector loop independently of the nucleotide occupancy, leading to a complete inhibition of downstream signaling of Ras.


Subject(s)
Bacterial Toxins/metabolism , Clostridium , Cytotoxins/metabolism , Glucose/metabolism , ras Proteins/chemistry , ras Proteins/metabolism , Bacterial Toxins/toxicity , Crystallography, X-Ray , Cytotoxins/toxicity , Guanosine Diphosphate/metabolism , Models, Molecular , Protein Conformation/drug effects , Signal Transduction/drug effects , Structure-Activity Relationship , Threonine/metabolism , ras Proteins/antagonists & inhibitors
16.
Biol Chem ; 381(5-6): 421-6, 2000.
Article in English | MEDLINE | ID: mdl-10937872

ABSTRACT

Several bacterial toxins target Rho GTPases, which constitute molecular switches in several signaling processes and master regulators of the actin cytoskeleton. The biological activities of Rho GTPases are blocked by C3-like transferases, which ADP-ribosylate Rho at Asn41, but not Rac or Cdc42. Large clostridial cytotoxins (e. g., Clostridium difficile toxin A and B) glucosylate Rho GTPases at Thr37 (Rho) or Thr35 (Rac/Cdc42), thereby inhibiting Rho functions by preventing effector coupling. The 'injected' toxins ExoS, YopE and SptP from Pseudomonas aeruginosa, Yersinia and Salmonella ssp., respectively, which are transferred into the eukaryotic target cells by the type-III secretion system, inhibit Rho functions by acting as Rho GAP proteins. Rho GTPases are activated by the cytotoxic necrotizing factors CNF1 and CNF2 from Escherichia coli and by the dermonecrotizing toxin DNT from B. bronchiseptica. These toxins deamidate/transglutaminate Gln63 of Rho to block the intrinsic and GAP-stimulated GTP hydrolysis, thereby constitutively activating the GTPases. Rho GTPases are also activated by SopE, a type-III system injected protein from Salmonella ssp., that acts as a GEF protein.


Subject(s)
Bacterial Toxins/metabolism , GTP Phosphohydrolases/metabolism
17.
J Biol Chem ; 275(22): 16478-83, 2000 Jun 02.
Article in English | MEDLINE | ID: mdl-10748216

ABSTRACT

The C3-like ADP-ribosyltransferases exhibit a very confined substrate specificity compared with other Rho-modifying bacterial toxins; they selectively modify the RhoA, -B, and -C isoforms but not other members of the Rho or Ras subfamilies. In this study, the amino acid residues involved in the RhoA substrate recognition by C3 from Clostridium botulinum are identified by applying mutational analyses of the nonsubstrate Rac. First, the minimum domain responsible for the recognition by C3 was identified as the N-terminal 90 residues. Second, the combination of the N-terminal basic amino acids ((Rho)Arg(5)-Lys(6)), the acid residues (Rho)Glu(47) and (Rho)Glu(54) only slightly increases ADP-ribosylation but fully restores the binding of the respective mutant Rac to C3. Third, the residues (Rho)Glu(40) and (Rho)Val(43) also participate in binding to C3 but they are mainly involved in the correct formation of the ternary complex between Rho, C3, and NAD(+). Thus, these six residues (Arg(5), Lys(6), Glu(40), Val(43), Glu(47), and Glu(54)) distributed over the N-terminal part of Rho are involved in the correct binding of Rho to C3. Mutant Rac harboring these residues shows a kinetic property with regard to ADP-ribosylation, which is identical with that of RhoA. Differences in the conformation of Rho given by the nucleotide occupancy have only minor effects on ADP-ribosylation.


Subject(s)
ADP Ribose Transferases/metabolism , Botulinum Toxins , Clostridium botulinum/enzymology , rhoA GTP-Binding Protein/metabolism , Adenosine Diphosphate Ribose/metabolism , Amino Acid Sequence , Molecular Sequence Data , Mutagenesis, Site-Directed , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Sequence Homology, Amino Acid , Substrate Specificity , rhoA GTP-Binding Protein/chemistry , rhoA GTP-Binding Protein/genetics
18.
Infect Immun ; 68(3): 1094-101, 2000 Mar.
Article in English | MEDLINE | ID: mdl-10678912

ABSTRACT

The family of the large clostridial cytotoxins, encompassing Clostridium difficile toxins A and B as well as the lethal and hemorrhagic toxins from Clostridium sordellii, monoglucosylate the Rho GTPases by transferring a glucose moiety from the cosubstrate UDP-glucose. Here we present a new detoxification procedure to block the enzyme activity by treatment with the reactive UDP-2', 3'-dialdehyde to result in alkylation of toxin A and B. Alkylation is likely to occur in the catalytic domain, because the native cosubstrate UDP-glucose completely protected the toxins from inactivation and the alkylated toxin competes with the native toxin at the cell receptor. Alkylated toxins are good antigens resulting in antibodies recognizing only the C-terminally located receptor binding domain, whereas formaldehyde treatment resulted in antibodies recognizing both the receptor binding domain and the catalytic domain, indicating that the catalytic domain is concealed under native conditions. Antibodies against the native catalytic domain (amino acids 1 through 546) and those holotoxin antibodies recognizing the catalytic domain inhibited enzyme activity. However, only antibodies against the receptor binding domain protected intact cells from the cytotoxic activity of toxin B, whereas antibodies against the catalytic domain were protective only when inside the cell.


Subject(s)
Antibodies, Bacterial/immunology , Bacterial Proteins , Bacterial Toxins/immunology , Clostridioides difficile/immunology , Animals , Antibodies, Bacterial/biosynthesis , Bacterial Toxins/antagonists & inhibitors , Bacterial Toxins/toxicity , Binding Sites , Clostridioides difficile/pathogenicity , HeLa Cells , Humans , Immune Sera/immunology , Immunization , Rabbits , Uridine Diphosphate/analogs & derivatives , Uridine Diphosphate/pharmacology
19.
Arterioscler Thromb Vasc Biol ; 20(1): 61-9, 2000 Jan.
Article in English | MEDLINE | ID: mdl-10634801

ABSTRACT

Three 3-hydroxy-3-methylglutaryl coenzyme A (HMG-CoA) reductase inhibitors (HCRIs), atorvastatin, pravastatin, and cerivastatin, inhibited phorbol ester-stimulated superoxide anion (O(2)(-)) formation in endothelium-intact segments of the rat aorta in a time- and concentration-dependent manner (maximum inhibition of 70% after 18 hours at 1 to 10 micromol/L). The HMG-CoA reductase product mevalonic acid (400 micromol/L) reversed the inhibitory effect of the HCRIs, which, conversely, was mimicked by inactivation of p21 Rac with Clostridium sordellii lethal toxin but not by inactivation of p21 Rho with Clostridium botulinum exoenzyme (C3). A mevalonate-sensitive inhibition of phorbol ester-stimulated O(2)(-) formation by atorvastatin was also observed in porcine cultured endothelial cells and in a murine macrophage cell line. In the rat aorta, no effect of the HCRIs on protein kinase C, NADPH oxidase, or superoxide dismutase (SOD) activity and expression was detected, whereas that of endothelial nitric oxide (NO) synthase was enhanced approximately 2-fold. Moreover, exposure of the segments to atorvastatin resulted in a significant improvement of endothelium-dependent NO-mediated relaxation, and this effect was abolished in the presence of SOD. Taken together, these findings suggest that in addition to augmenting endothelial NO synthesis, HCRIs inhibit endothelial O(2)(-) formation by preventing the isoprenylation of p21 Rac, which is critical for the assembly of NADPH oxidase after activation of protein kinase C. The resulting shift in the balance between NO and O(2)(-) in the endothelium improves endothelial function even in healthy blood vessels and therefore may provide a reasonable explanation for the beneficial effects of HCRIs in patients with coronary heart disease in addition to or as an alternative to the reduction in serum LDL cholesterol.


Subject(s)
Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacology , Nitric Oxide/metabolism , Superoxides/metabolism , Vasodilation/drug effects , Vasodilation/physiology , Animals , Aorta, Thoracic/drug effects , Aorta, Thoracic/metabolism , Atorvastatin , Base Sequence , Botulinum Toxins/pharmacology , DNA Primers/genetics , Endothelium, Vascular/drug effects , Endothelium, Vascular/metabolism , Heptanoic Acids/pharmacology , In Vitro Techniques , Male , Mevalonic Acid/pharmacology , NADPH Oxidases/metabolism , Nitric Oxide Synthase/genetics , Nitric Oxide Synthase/metabolism , Nitric Oxide Synthase Type III , Phorbol 12,13-Dibutyrate/pharmacology , Pravastatin/pharmacology , Protein Kinase C/metabolism , Pyridines/pharmacology , Pyrroles/pharmacology , Rats , Rats, Wistar , Superoxide Dismutase/genetics , Superoxide Dismutase/metabolism
20.
J Biol Chem ; 274(41): 29050-6, 1999 Oct 08.
Article in English | MEDLINE | ID: mdl-10506156

ABSTRACT

The small GTPases Rho, Rac, and Cdc42 are monoglucosylated at effector domain amino acid threonine 37/35 by Clostridium difficile toxins A and B. Glucosylation renders the Rho proteins inactive by inhibiting effector coupling. To understand the functional consequences, effects of glucosylation on subcellular distribution and cycling of Rho GTPases between cytosol and membranes were analyzed. In intact cells and in cell lysates, glucosylation leads to a translocation of the majority of RhoA GTPase to the membranes whereas a minor fraction is monomeric in the cytosol without being complexed with the guanine nucleotide dissociation inhibitor (GDI-1). Rho complexed with GDI-1 is not substrate for glucosylation, and modified Rho does not bind to GDI-1. However, a membranous factor inducing release of Rho from the GDI complex makes cytosolic Rho available as a substrate for glucosylation. The binding of glucosylated RhoA to the plasma membranes is saturable, competable with unmodified Rho-GTPgammaS guanosine 5'-O-(3-thiotriphosphate), and takes place at a membrane protein with a molecular mass of about 70 kDa. Membrane-bound glucosylated Rho is not extractable by GDI-1 as unmodified Rho is, leading to accumulation of modified Rho at membranous binding sites. Thus, in addition to effector coupling inhibition, glucosylation also inhibits Rho cycling between cytosol and membranes, a prerequisite for Rho activation.


Subject(s)
Bacterial Proteins , Cell Membrane/metabolism , Threonine/metabolism , rhoA GTP-Binding Protein/metabolism , 3T3 Cells , Animals , Bacterial Toxins/pharmacology , Cell Fractionation , Cytosol/metabolism , Enterotoxins/pharmacology , Glucose/metabolism , Glycosylation , Guanine Nucleotide Dissociation Inhibitors/metabolism , Mice , Protein Binding , Threonine/chemistry , rhoA GTP-Binding Protein/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL