Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 53
Filter
1.
Nature ; 617(7961): 616-622, 2023 May.
Article in English | MEDLINE | ID: mdl-36972684

ABSTRACT

Steroid hormone receptors are ligand-binding transcription factors essential for mammalian physiology. The androgen receptor (AR) binds androgens mediating gene expression for sexual, somatic and behavioural functions, and is involved in various conditions including androgen insensitivity syndrome and prostate cancer1. Here we identified functional mutations in the formin and actin nucleator DAAM2 in patients with androgen insensitivity syndrome. DAAM2 was enriched in the nucleus, where its localization correlated with that of the AR to form actin-dependent transcriptional droplets in response to dihydrotestosterone. DAAM2 AR droplets ranged from 0.02 to 0.06 µm3 in size and associated with active RNA polymerase II. DAAM2 polymerized actin directly at the AR to promote droplet coalescence in a highly dynamic manner, and nuclear actin polymerization is required for prostate-specific antigen expression in cancer cells. Our data uncover signal-regulated nuclear actin assembly at a steroid hormone receptor necessary for transcription.


Subject(s)
Actins , Formins , Nuclear Proteins , Receptors, Androgen , Transcription, Genetic , Humans , Actins/metabolism , Androgen-Insensitivity Syndrome/genetics , Androgen-Insensitivity Syndrome/metabolism , Androgens/pharmacology , Androgens/metabolism , Formins/metabolism , Gene Expression Regulation/drug effects , Nuclear Proteins/metabolism , Polymerization/drug effects , Prostate-Specific Antigen/metabolism , Prostatic Neoplasms/genetics , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , Receptors, Androgen/metabolism , RNA Polymerase II/metabolism , Signal Transduction/drug effects , Steroids/metabolism , Steroids/pharmacology , Testosterone/analogs & derivatives , Transcription, Genetic/drug effects
2.
EMBO J ; 39(23): e107086, 2020 12 01.
Article in English | MEDLINE | ID: mdl-33210291

ABSTRACT

Maintenance of the mature blood cells requires controlled cell fate decisions by hematopoietic stem and progenitor cells (HSPCs). While our knowledge of the gene expression changes that facilitate differentiation has made a leap forward, less is known about the cellular triggers that induce them. Biedzinski et al (2020) now uncover a new intracellular mechanism that drives myeloid differentiation: Microtubule bundles squeeze the nucleus of HSPCs and form large invaginations, thus causing changes in chromatin organization. These microtubule-induced nuclear shape changes result in gene expression profiles that favor myeloid differentiation.


Subject(s)
Hematopoietic Stem Cell Transplantation , Hematopoietic Stem Cells , Cell Differentiation , Constriction , Gene Expression , Microtubules
3.
Development ; 148(1)2021 01 07.
Article in English | MEDLINE | ID: mdl-33199445

ABSTRACT

Anterior mesoderm (AM) and definitive endoderm (DE) progenitors represent the earliest embryonic cell types that are specified during germ layer formation at the primitive streak (PS) of the mouse embryo. Genetic experiments indicate that both lineages segregate from Eomes-expressing progenitors in response to different Nodal signaling levels. However, the precise spatiotemporal pattern of the emergence of these cell types and molecular details of lineage segregation remain unexplored. We combined genetic fate labeling and imaging approaches with single-cell RNA sequencing (scRNA-seq) to follow the transcriptional identities and define lineage trajectories of Eomes-dependent cell types. Accordingly, all cells moving through the PS during the first day of gastrulation express Eomes AM and DE specification occurs before cells leave the PS from Eomes-positive progenitors in a distinct spatiotemporal pattern. ScRNA-seq analysis further suggested the immediate and complete separation of AM and DE lineages from Eomes-expressing cells as last common bipotential progenitor.


Subject(s)
Cell Lineage , Endoderm/cytology , Endoderm/metabolism , Gastrulation , Mesoderm/cytology , Mesoderm/metabolism , Alleles , Animals , Basic Helix-Loop-Helix Transcription Factors/genetics , Cell Differentiation , Epithelial Cells/metabolism , Gene Expression Regulation, Developmental , Germ Layers/cytology , Mice , Models, Biological , Primitive Streak/embryology , Primitive Streak/metabolism , Stem Cells/metabolism , T-Box Domain Proteins/metabolism , Time Factors , Transcription, Genetic
4.
J Infect Dis ; 225(7): 1296-1300, 2022 04 01.
Article in English | MEDLINE | ID: mdl-33011801

ABSTRACT

Clostridioides difficile infection (CDI) represents a significant burden on the health care system, one that is exacerbated by the emergence of binary toxin (CDT)-producing hypervirulent C. difficile strains. Previous work from our laboratory has shown that Toll-like receptor 2 (TLR2) recognizes CDT to induce inflammation. Here we explore the interactions of CDT with TLR2 and the impact on host immunity during CDI. We found that the TLR2/6 heterodimer, not TLR2/1, is responsible for CDT recognition, and that gene pathways including nuclear factor-κB and MAPK downstream of TLR2/6 are upregulated in mice with intact TLR2/6 signaling during CDI.


Subject(s)
Clostridioides difficile , Clostridium Infections , Animals , Antibodies, Bacterial , Mice , NF-kappa B , Toll-Like Receptor 2/genetics , Toll-Like Receptor 6
5.
Annu Rev Microbiol ; 71: 281-307, 2017 09 08.
Article in English | MEDLINE | ID: mdl-28657883

ABSTRACT

Clostridium difficile is the cause of antibiotics-associated diarrhea and pseudomembranous colitis. The pathogen produces three protein toxins: C. difficile toxins A (TcdA) and B (TcdB), and C. difficile transferase toxin (CDT). The single-chain toxins TcdA and TcdB are the main virulence factors. They bind to cell membrane receptors and are internalized. The N-terminal glucosyltransferase and autoprotease domains of the toxins translocate from low-pH endosomes into the cytosol. After activation by inositol hexakisphosphate (InsP6), the autoprotease cleaves and releases the glucosyltransferase domain into the cytosol, where GTP-binding proteins of the Rho/Ras family are mono-O-glucosylated and, thereby, inactivated. Inactivation of Rho proteins disturbs the organization of the cytoskeleton and affects multiple Rho-dependent cellular processes, including loss of epithelial barrier functions, induction of apoptosis, and inflammation. CDT, the third C. difficile toxin, is a binary actin-ADP-ribosylating toxin that causes depolymerization of actin, thereby inducing formation of the microtubule-based protrusions. Recent progress in understanding of the toxins' actions include insights into the toxin structures, their interaction with host cells, and functional consequences of their actions.


Subject(s)
ADP Ribose Transferases/toxicity , Bacterial Proteins/toxicity , Bacterial Toxins/toxicity , Clostridioides difficile/metabolism , Enterotoxins/toxicity , Epithelial Cells/drug effects , Virulence Factors/toxicity , ADP Ribose Transferases/metabolism , Animals , Bacterial Proteins/metabolism , Bacterial Toxins/metabolism , Cytoskeleton/drug effects , Endocytosis , Enterotoxins/metabolism , Epithelial Cells/physiology , Humans , Microtubules/drug effects , Virulence Factors/metabolism
6.
EMBO Rep ; 21(11): e50758, 2020 11 05.
Article in English | MEDLINE | ID: mdl-32959960

ABSTRACT

The actin cytoskeleton operates in a multitude of cellular processes including cell shape and migration, mechanoregulation, and membrane or organelle dynamics. However, its filamentous properties and functions inside the mammalian cell nucleus are less well explored. We previously described transient actin assembly at mitotic exit that promotes nuclear expansion during chromatin decondensation. Here, we identify non-muscle α-actinin 4 (ACTN4) as a critical regulator to facilitate F-actin reorganization and bundling during postmitotic nuclear expansion. ACTN4 binds to nuclear actin filament structures, and ACTN4 clusters associate with nuclear F-actin in a highly dynamic fashion. ACTN4 but not ACTN1 is required for proper postmitotic nuclear volume expansion, mediated by its actin-binding domain. Using super-resolution imaging to quantify actin filament numbers and widths in individual nuclei, we find that ACTN4 is necessary for postmitotic nuclear actin reorganization and actin filament bundling. Our findings uncover a nuclear cytoskeletal function for ACTN4 to control nuclear size and chromatin organization during mitotic cell division.


Subject(s)
Actinin , Actins , Actin Cytoskeleton , Actinin/genetics , Actins/genetics , Animals , Cell Nucleus , Cytoskeleton
7.
FASEB J ; 34(5): 6244-6261, 2020 05.
Article in English | MEDLINE | ID: mdl-32190927

ABSTRACT

The human pathogenic bacterium Clostridioides difficile produces two exotoxins TcdA and TcdB, which inactivate Rho GTPases thereby causing C. difficile-associated diseases (CDAD) including life-threatening pseudomembranous colitis. Hypervirulent strains produce additionally the binary actin ADP-ribosylating toxin CDT. These strains are hallmarked by more severe forms of CDAD and increased frequency and severity. Once in the cytosol, the toxins act as enzymes resulting in the typical clinical symptoms. Therefore, targeting and inactivation of the released toxins are of peculiar interest. Prompted by earlier findings that human α-defensin-1 neutralizes TcdB, we investigated the effects of the defensin on all three C. difficile toxins. Inhibition of TcdA, TcdB, and CDT was demonstrated by analyzing toxin-induced changes in cell morphology, substrate modification, and decrease in transepithelial electrical resistance. Application of α-defensin-1 protected cells and human intestinal organoids from the cytotoxic effects of TcdA, TcdB, CDT, and their combination which is attributed to a direct interaction between the toxins and α-defensin-1. In mice, the application of α-defensin-1 reduced the TcdA-induced damage of intestinal loops in vivo. In conclusion, human α-defensin-1 is a specific and potent inhibitor of the C. difficile toxins and a promising agent to develop novel therapeutic options against C. difficile infections.


Subject(s)
ADP Ribose Transferases/toxicity , Anti-Infective Agents/metabolism , Bacterial Proteins/toxicity , Bacterial Toxins/toxicity , Enterotoxins/toxicity , Intestinal Mucosa/drug effects , Organoids/drug effects , Peptide Fragments/metabolism , alpha-Defensins/metabolism , ADP Ribose Transferases/metabolism , Animals , Bacterial Proteins/metabolism , Bacterial Toxins/metabolism , Enterotoxins/metabolism , Humans , Intestinal Mucosa/metabolism , Intestinal Mucosa/pathology , Male , Mice , Organoids/metabolism , Organoids/pathology
8.
PLoS Pathog ; 14(8): e1007248, 2018 08.
Article in English | MEDLINE | ID: mdl-30102745

ABSTRACT

Salmonella enterica serotype Typhimurium (S. Typhimurium) is one of the most frequent causes of food-borne illness in humans and usually associated with acute self-limiting gastroenteritis. However, in immunocompromised patients, the pathogen can disseminate and lead to severe systemic diseases. S. Typhimurium are facultative intracellular bacteria. For uptake and intracellular life, Salmonella translocate numerous effector proteins into host cells using two type-III secretion systems (T3SS), which are encoded within Salmonella pathogenicity islands 1 (SPI-1) and 2 (SPI-2). While SPI-1 effectors mainly promote initial invasion, SPI-2 effectors control intracellular survival and proliferation. Here, we elucidate the mode of action of Salmonella SPI-2 effector SseI, which is involved in control of systemic dissemination of S. Typhimurium. SseI deamidates a specific glutamine residue of heterotrimeric G proteins of the Gαi family, resulting in persistent activation of the G protein. Gi activation inhibits cAMP production and stimulates PI3-kinase γ by Gαi-released Gßγ subunits, resulting in activation of survival pathways by phosphorylation of Akt and mTOR. Moreover, SseI-induced deamidation leads to non-polarized activation of Gαi and, thereby, to loss of directed migration of dendritic cells.


Subject(s)
Bacterial Proteins/physiology , Chemotaxis , GTP-Binding Protein alpha Subunits, Gi-Go/metabolism , Salmonella typhimurium , Type III Secretion Systems/physiology , Animals , Bacterial Proteins/genetics , Cell Survival/genetics , Chemotaxis/genetics , Deamination/genetics , Female , GTP-Binding Protein alpha Subunits, Gi-Go/chemistry , HEK293 Cells , HeLa Cells , Host-Pathogen Interactions/genetics , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Protein Multimerization/genetics , Protein Processing, Post-Translational/genetics , RAW 264.7 Cells , Salmonella Infections/metabolism , Salmonella Infections/pathology , Salmonella typhimurium/genetics , Salmonella typhimurium/metabolism , Type III Secretion Systems/genetics , Type III Secretion Systems/metabolism
9.
Proc Natl Acad Sci U S A ; 113(28): 7870-5, 2016 07 12.
Article in English | MEDLINE | ID: mdl-27339141

ABSTRACT

Hypervirulent Clostridium difficile strains, which are associated with increased morbidity and mortality, produce the actin-ADP ribosylating toxin Clostridium difficile transferase (CDT). CDT depolymerizes actin, causes formation of microtubule-based protrusions, and increases pathogen adherence. Here, we show that septins (SEPT) are essential for CDT-induced protrusion formation. SEPT2, -6, -7, and -9 accumulate at predetermined protrusion sites and form collar-like structures at the base of protrusions. The septin inhibitor forchlorfenuron or knockdown of septins inhibits protrusion formation. At protrusion sites, septins colocalize with the GTPase Cdc42 (cell division control protein 42) and its effector Borg (binder of Rho GTPases), which act as up-stream regulators of septin polymerization. Precipitation and surface plasmon resonance studies revealed high-affinity binding of septins to the microtubule plus-end tracking protein EB1, thereby guiding incoming microtubules. The data suggest that CDT usurps conserved regulatory principles involved in microtubule-membrane interaction, depending on septins, Cdc42, Borgs, and restructuring of the actin cytoskeleton.


Subject(s)
Bacterial Toxins/pharmacology , Clostridioides difficile/enzymology , Microtubules/drug effects , Septins/metabolism , Animals , Caco-2 Cells , Dogs , GTP-Binding Protein Regulators/metabolism , HeLa Cells , Humans , Madin Darby Canine Kidney Cells , Mice, Inbred C57BL , Transferases/metabolism , cdc42 GTP-Binding Protein/metabolism
10.
Curr Top Microbiol Immunol ; 399: 35-51, 2017.
Article in English | MEDLINE | ID: mdl-27726005

ABSTRACT

A large group of bacterial protein toxins, including binary ADP-ribosylating toxins, modify actin at arginine-177, thereby actin polymerization is blocked and the actin cytoskeleton is redistributed. Modulation of actin functions largely affects other components of the cytoskeleton, especially microtubules and septins. Here, recent findings about the functional interconnections of the actin cytoskeleton with microtubules and septins, affected by bacterial toxins, are reviewed.


Subject(s)
Actin Cytoskeleton/metabolism , Bacterial Proteins/metabolism , Bacterial Toxins/metabolism , Clostridium Infections/metabolism , Clostridium/metabolism , Septins/metabolism , Actin Cytoskeleton/chemistry , Animals , Bacterial Proteins/genetics , Cell Shape , Clostridium/genetics , Clostridium Infections/microbiology , Humans , Polymerization , Septins/chemistry
11.
Cell Microbiol ; 19(1)2017 01.
Article in English | MEDLINE | ID: mdl-27341322

ABSTRACT

Intoxication of eukaryotic cells by Photorhabdus luminescens toxin TccC3 induces cell rounding and detachment from the substratum within a few hours and compromises a number of cell functions like phagocytosis. Here, we used morphological and biochemical procedures to analyse the mechanism of TccC3 intoxication. Life imaging of TccC3-intoxicated HeLa cells transfected with AcGFP-actin shows condensation of F-actin into large aggregates. Life cell total internal reflection fluorescence (TIRF) microscopy of identically treated HeLa cells confirmed the formation of actin aggregates but also disassembly of F-actin stress fibres. Recombinant TccC3 toxin ADP-ribosylates purified skeletal and non-muscle actin at threonine148 leading to a strong propensity to polymerize and F-actin bundle formation as shown by TIRF and electron microscopy. Native gel electrophoresis shows strongly reduced binding of Thr148-ADP-ribosylated actin to the severing proteins gelsolin and its fragments G1 and G1-3, and to ADF/cofilin. Complexation of actin with these proteins inhibits its ADP-ribosylation. TIRF microscopy demonstrates rapid polymerization of Thr148-ADP-ribosylated actin to curled F-actin bundles even in the presence of thymosin ß4, gelsolin or G1-3. Thr148-ADP-ribosylated F-actin cannot be depolymerized by gelsolin or G1-3 as verified by TIRF, co-sedimentation and electron microscopy and shows reduced treadmilling as indicated by a lack of stimulation of its ATPase activity after addition of cofilin-1.


Subject(s)
Actins/metabolism , Adenosine Diphosphate/metabolism , Bacterial Toxins/metabolism , Photorhabdus/metabolism , Protein Aggregation, Pathological , Epithelial Cells/drug effects , HeLa Cells , Humans , Microscopy, Electron , Microscopy, Fluorescence
12.
Anaerobe ; 53: 21-29, 2018 Oct.
Article in English | MEDLINE | ID: mdl-29524654

ABSTRACT

Clostridium difficile infection causes antibiotics-associated diarrhea and pseudomembranous colitis. Major virulence factors of C. difficile are the Rho-glucosylating toxins TcdA and TcdB. In addition, many, so-called hypervirulent C. difficile strains produce the binary actin-ADP-ribosylating toxin CDT. CDT causes depolymerization of F-actin and rearrangement of the actin cytoskeleton. Thereby, many cellular functions, which depend on actin, are altered. CDT disturbs the dynamic balance between actin and microtubules in target cells. The toxin increases microtubule polymerization and induces the formation of microtubule-based protrusions at the plasma membrane of target cells. Moreover, CDT causes a redistribution of vesicles from the basolateral side to the apical side, where extracellular matrix proteins are released. These processes may increase the adherence of clostridia to target cells. Here, we review the effects of the action of CDT on the actin cytoskeleton and on the microtubule system.


Subject(s)
ADP Ribose Transferases/metabolism , Bacterial Proteins/metabolism , Clostridioides difficile/metabolism , Cytoskeleton/drug effects , Virulence Factors/metabolism , Actins/metabolism , Bacterial Adhesion , Clostridioides difficile/physiology , Cytoplasmic Vesicles/drug effects , Cytoplasmic Vesicles/metabolism , Humans , Microtubules/drug effects , Microtubules/metabolism
13.
Handb Exp Pharmacol ; 235: 179-206, 2017.
Article in English | MEDLINE | ID: mdl-27316913

ABSTRACT

Actin and the actin cytoskeleton play fundamental roles in host-pathogen interactions. Proper function of the actin cytoskeleton is crucial for innate and acquired immune defense. Bacterial toxins attack the actin cytoskeleton by targeting regulators of actin. Moreover, actin is directly modified by various bacterial protein toxins and effectors, which cause ADP-ribosylation or cross-linking of actin. Modification of actin can result in inhibition or stimulation of actin polymerization. Toxins, acting directly on actin, are reviewed.


Subject(s)
Actins/chemistry , Adenosine Diphosphate Ribose/chemistry , Bacterial Toxins/pharmacology , Host-Pathogen Interactions , Actin Cytoskeleton/chemistry , Animals , Cross-Linking Reagents/chemistry , Humans , Polymerization
14.
Proc Natl Acad Sci U S A ; 111(6): 2313-8, 2014 Feb 11.
Article in English | MEDLINE | ID: mdl-24469807

ABSTRACT

Clostridium difficile causes antibiotic-associated diarrhea and pseudomembranous colitis by the actions of Rho-glucosylating toxins A and B. Recently identified hypervirulent strains, which are associated with increased morbidity and mortality, additionally produce the actin-ADP-ribosylating toxin C. difficile transferase (CDT). CDT depolymerizes actin, causes formation of microtubule-based protrusions, and increases pathogen adherence. Here we show that CDT-induced protrusions allow vesicle traffic and contain endoplasmic reticulum tubules, connected to microtubules via the calcium sensor Stim1. The toxin reroutes Rab11-positive vesicles containing fibronectin, which is involved in bacterial adherence, from basolateral to the apical membrane sides in a microtubule- and Stim1-dependent manner. The data yield a model of C. difficile adherence regulated by actin depolymerization, microtubule restructuring, subsequent Stim1-dependent Ca(2+) signaling, vesicle rerouting, and secretion of ECM proteins to increase bacterial adherence.


Subject(s)
Bacterial Adhesion , Bacterial Toxins/toxicity , Clostridioides difficile/pathogenicity , Enterotoxins/toxicity , Microtubules/drug effects , Biological Transport , Caco-2 Cells , Calcium Signaling , Clostridioides difficile/metabolism , Endoplasmic Reticulum/drug effects , Fibronectins/metabolism , Humans
15.
PLoS Pathog ; 10(1): e1003884, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24453976

ABSTRACT

The Cytotoxic Necrotizing Factor 1 (CNF1) is a protein toxin which is a major virulence factor of pathogenic Escherichia coli strains. Here, we identified the Lutheran (Lu) adhesion glycoprotein/basal cell adhesion molecule (BCAM) as cellular receptor for CNF1 by co-precipitation of cell surface molecules with tagged toxin. The CNF1-Lu/BCAM interaction was verified by direct protein-protein interaction analysis and competition studies. These studies revealed amino acids 720 to 1014 of CNF1 as the binding site for Lu/BCAM. We suggest two cell interaction sites in CNF1: first the N-terminus, which binds to p37LRP as postulated before. Binding of CNF1 to p37LRP seems to be crucial for the toxin's action. However, it is not sufficient for the binding of CNF1 to the cell surface. A region directly adjacent to the catalytic domain is a high affinity interaction site for Lu/BCAM. We found Lu/BCAM to be essential for the binding of CNF1 to cells. Cells deficient in Lu/BCAM but expressing p37LRP could not bind labeled CNF1. Therefore, we conclude that LRP and Lu/BCAM are both required for toxin action but with different functions.


Subject(s)
Bacterial Toxins/metabolism , Cell Adhesion Molecules/metabolism , Escherichia coli Proteins/metabolism , Escherichia coli/metabolism , Escherichia coli/pathogenicity , Lutheran Blood-Group System/metabolism , Virulence Factors/metabolism , Bacterial Toxins/genetics , Cell Adhesion Molecules/immunology , Escherichia coli/genetics , Escherichia coli Proteins/genetics , HEK293 Cells , HeLa Cells , Humans , Lutheran Blood-Group System/immunology , Protein Binding , Protein Structure, Tertiary , Virulence Factors/genetics
16.
FASEB J ; 29(7): 2789-802, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25782990

ABSTRACT

The bacterial toxin Photorhabdus asymbiotica toxin (PaTox) modifies Rho proteins by tyrosine GlcNAcylation and heterotrimeric Gα proteins by deamidation. Inactivation of Rho proteins results in F-actin disassembly in host cells. Here, we analyzed the subcellular distribution of PaTox and show that the glycosyltransferase domain of PaTox associates with the negatively charged inner surface of the plasma membrane. Localization studies with site-directed mutants, liposome precipitation analysis, lipid overlay assays, and confocal time-lapse microscopy revealed that a patch of positively charged lysine and arginine residues located on helix α1 of the glycosyltransferase is essential for membrane attachment. Using a helix1 deletion mutant, we show that plasma membrane localization of PaTox is essential for cytotoxicity and proved this by substitution of helix1 by an N-terminal myristoylation signal peptide, which restored plasma membrane localization and cytotoxicity. Furthermore, we also show that the intracellular deamidase activity of PaTox depends on the presence of the membrane localization domain. Comparison of PaTox membrane-binding domain with the 4-helix-bundle membrane-binding domain of Pasteurella multocida toxin, Vibrio cholerae multifunctional autoprocessing repeats-in-toxin, and clostridial glucosylating toxins revealed similar spatial geometry and charge distribution but different structural topology, indicating convergent evolution of toxin domains for optimized host target interaction.


Subject(s)
Bacterial Toxins/toxicity , Photorhabdus/pathogenicity , Amino Acid Sequence , Bacterial Toxins/chemistry , Bacterial Toxins/genetics , Cell Membrane/drug effects , Cell Membrane/metabolism , Genes, Bacterial , HeLa Cells , Humans , Membrane Lipids/metabolism , Models, Molecular , Molecular Sequence Data , Mutagenesis, Site-Directed , Phospholipids/metabolism , Photorhabdus/chemistry , Photorhabdus/genetics , Protein Conformation , Protein Structure, Secondary , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/toxicity , Sequence Deletion , Static Electricity , rho GTP-Binding Proteins/metabolism
17.
Cell Microbiol ; 17(4): 579-94, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25338512

ABSTRACT

PTC3 and PTC5 are tripartite Tc (toxin complex) toxins from Photorhabdus luminescens, which consist of the binding component TcdA1, the linker component TcdB2 and the enzyme components TccC3 and TccC5 respectively. While PTC5 adenosine diphosphate (ADP)-ribosylates Rho proteins at Gln61/63 resulting in constitutive activation of the GTPases, PTC3 ADP-ribosylates actin at Thr148 thereby inducing actin polymerization. Here, we identified amino acids involved in ADP-ribosyltransferase activity of TccC3 and TccC5 and analysed the substrate specificity of Rho-activating TccC5. We compared the time dependency of Rho protein activation by PTC5 in HeLa cells with the effects of Escherichia coli cytotoxic necrotizing factor 1, which activates Rho GTPases by deamidation of Gln61/63. Using a luciferase reporter assay, we show that PTC5 and PTC3 stimulated gene transcription via myocardin-related transcription factor A (also called MAL) and AP1. MAL activation by PTC5 involved Rho kinase and formins. Activation of AP1 by PTC5 occurred via two MAP kinase pathways involving extracellular signal-regulated kinase and Jun kinase respectively.


Subject(s)
ADP Ribose Transferases/metabolism , Actins/metabolism , Bacterial Toxins/metabolism , Photorhabdus/enzymology , Trans-Activators/metabolism , Transcription Factor AP-1/metabolism , rho GTP-Binding Proteins/metabolism , Genes, Reporter , HeLa Cells , Humans , Luciferases/analysis , Luciferases/genetics , Protein Processing, Post-Translational , Transcription, Genetic
18.
Cell Microbiol ; 16(4): 490-503, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24138221

ABSTRACT

TccC3 and TccC5 from Photorhabdus luminescens are ADP-ribosyltransferases, which modify actin and Rho GTPases, respectively, thereby inducing polymerization and clustering of actin. The bacterial proteins are components of the Photorhabdus toxin complexes, consisting of the binding and translocation component TcdA1, a proposed linker component TcdB2 and the enzymatic component TccC3/5. While the action of the toxins on target proteins is clearly defined, uptake and translocation of the toxins into the cytosol of target cells are not well understood. Here we show by using pharmacological inhibitors that heat shock protein 90 (Hsp90) and peptidyl prolyl cis/trans isomerases (PPIases) including cyclophilins and FK506-binding proteins (FKBPs) facilitate the uptake of the ADP-ribosylating toxins into the host cell cytosol. Inhibition of Hsp90 and/or PPIases resulted in decreased intoxication of target cells by Photorhabdus toxin complexes determined by cell rounding and reduction of transepithelial electrical resistance of cell monolayers. ADP-ribosyltransferase activity of toxins and toxin-induced pore formation were notimpaired by the inhibitors of Hsp90 and PPIases. The Photorhabdus toxins interacted with Hsp90, FKBP51, Cyp40 and CypA, suggesting a role of these host cell factors in translocation and/or refolding of the ADP-ribosyltransferases.


Subject(s)
ADP Ribose Transferases/metabolism , Bacterial Toxins/metabolism , HSP90 Heat-Shock Proteins/metabolism , Host-Pathogen Interactions , Peptidylprolyl Isomerase/metabolism , Photorhabdus/enzymology , Protein Transport
19.
Proc Natl Acad Sci U S A ; 108(39): 16422-7, 2011 Sep 27.
Article in English | MEDLINE | ID: mdl-21930894

ABSTRACT

Clostridium difficile infection (CDI) causes antibiotic-associated diarrhea and pseudomembranous colitis. Hypervirulent strains of the pathogen, which are responsible for increased morbidity and mortality of CDI, produce the binary actin-ADP ribosylating toxin Clostridium difficile transferase (CDT) in addition to the Rho-glucosylating toxins A and B. CDT depolymerizes the actin cytoskeleton, increases adherence and colonization of Clostridia by induction of microtubule-based cell protrusions and, eventually, causes death of target cells. Using a haploid genetic screen, we identified the lipolysis-stimulated lipoprotein receptor as the membrane receptor for CDT uptake by target cells. Moreover, we show that Clostridium perfringens iota toxin, which is a related binary actin-ADP ribosylating toxin, enters target cells via the lipolysis-stimulated lipoprotein receptor. Identification of the toxin receptors is essential for understanding of the toxin uptake and provides a most valuable basis for antitoxin strategies.


Subject(s)
Clostridioides difficile/metabolism , Enterotoxins/metabolism , Receptors, LDL/metabolism , Transferases/metabolism , Clostridioides difficile/enzymology , Clostridioides difficile/genetics , Haploidy , HeLa Cells , Humans
20.
Nat Commun ; 15(1): 446, 2024 Jan 10.
Article in English | MEDLINE | ID: mdl-38199985

ABSTRACT

Patients with corticosteroid-refractory acute graft-versus-host disease (aGVHD) have a low one-year survival rate. Identification and validation of novel targetable kinases in patients who experience corticosteroid-refractory-aGVHD may help improve outcomes. Kinase-specific proteomics of leukocytes from patients with corticosteroid-refractory-GVHD identified rho kinase type 1 (ROCK1) as the most significantly upregulated kinase. ROCK1/2 inhibition improved survival and histological GVHD severity in mice and was synergistic with JAK1/2 inhibition, without compromising graft-versus-leukemia-effects. ROCK1/2-inhibition in macrophages or dendritic cells prior to transfer reduced GVHD severity. Mechanistically, ROCK1/2 inhibition or ROCK1 knockdown interfered with CD80, CD86, MHC-II expression and IL-6, IL-1ß, iNOS and TNF production in myeloid cells. This was accompanied by impaired T cell activation by dendritic cells and inhibition of cytoskeletal rearrangements, thereby reducing macrophage and DC migration. NF-κB signaling was reduced in myeloid cells following ROCK1/2 inhibition. In conclusion, ROCK1/2 inhibition interferes with immune activation at multiple levels and reduces acute GVHD while maintaining GVL-effects, including in corticosteroid-refractory settings.


Subject(s)
Graft vs Host Disease , rho-Associated Kinases , Humans , Animals , Mice , rho-Associated Kinases/genetics , Graft vs Host Disease/drug therapy , Signal Transduction , NF-kappa B , Adrenal Cortex Hormones/pharmacology , Adrenal Cortex Hormones/therapeutic use
SELECTION OF CITATIONS
SEARCH DETAIL