Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Wolfgang Fischer is active.

Publication


Featured researches published by Wolfgang Fischer.


Molecular Microbiology | 2003

A novel sheathed surface organelle of the Helicobacter pylori cag type IV secretion system

Manfred Rohde; J. Puls; Renate Buhrdorf; Wolfgang Fischer; Rainer Haas

Type I strains of Helicobacter pylori (Hp) use a type IV secretion system (T4SS), encoded by the cag pathogenicity island (cag‐PAI), to deliver the bacterial protein CagA into eukaryotic cells and to induce interleukin‐8 secretion. Translocated CagA is activated by tyrosine phosphorylation involving Src‐family kinases. The mechanism and structural basis for type IV protein secretion is not well understood. We describe here, by confocal laser scanning microscopy and field emission scanning electron microscopy, a novel filamentous surface organelle which is part of the Hp T4SS. The organelle is often located at one bacterial pole but can be induced by cell contact also along the lateral side of the bacteria. It consists of a rigid needle, covered focally or completely by HP0527 (Cag7 or CagY), a VirB10‐homologous protein. HP0527 is also clustered in the outer membrane. The VirB7‐homologous protein HP0532 is found at the base of this organelle. These observations demonstrate for the first time by microscopic techniques a complex T4SS‐associated, sheathed surface organelle reminiscent to the needle structures of bacterial type III secretion systems.


PLOS Pathogens | 2009

Helicobacter pylori Type IV Secretion Apparatus Exploits β1 Integrin in a Novel RGD-Independent Manner

Luisa F. Jiménez-Soto; Stefan Kutter; Xaver Sewald; Claudia Ertl; Evelyn Weiss; Ulrike Kapp; Manfred Rohde; Torsten Pirch; Kirsten Jung; S. Francesco Retta; Laurent Terradot; Wolfgang Fischer; Rainer Haas

Translocation of the Helicobacter pylori (Hp) cytotoxin-associated gene A (CagA) effector protein via the cag-Type IV Secretion System (T4SS) into host cells is a major risk factor for severe gastric diseases, including gastric cancer. However, the mechanism of translocation and the requirements from the host cell for that event are not well understood. The T4SS consists of inner- and outer membrane-spanning Cag protein complexes and a surface-located pilus. Previously an arginine-glycine-aspartate (RGD)-dependent typical integrin/ligand type interaction of CagL with α5β1 integrin was reported to be essential for CagA translocation. Here we report a specific binding of the T4SS-pilus-associated components CagY and the effector protein CagA to the host cell β1 Integrin receptor. Surface plasmon resonance measurements revealed that CagA binding to α5β1 integrin is rather strong (dissociation constant, KD of 0.15 nM), in comparison to the reported RGD-dependent integrin/fibronectin interaction (KD of 15 nM). For CagA translocation the extracellular part of the β1 integrin subunit is necessary, but not its cytoplasmic domain, nor downstream signalling via integrin-linked kinase. A set of β1 integrin-specific monoclonal antibodies directed against various defined β1 integrin epitopes, such as the PSI, the I-like, the EGF or the β-tail domain, were unable to interfere with CagA translocation. However, a specific antibody (9EG7), which stabilises the open active conformation of β1 integrin heterodimers, efficiently blocked CagA translocation. Our data support a novel model in which the cag-T4SS exploits the β1 integrin receptor by an RGD-independent interaction that involves a conformational switch from the open (extended) to the closed (bent) conformation, to initiate effector protein translocation.


The EMBO Journal | 2003

VirB11 ATPases are dynamic hexameric assemblies: New insights into bacterial type IV secretion

Savvas N. Savvides; Hye-Jeong Yeo; Moriah R. Beck; Franca Blaesing; Rudi Lurz; Erich Lanka; Renate Buhrdorf; Wolfgang Fischer; Rainer Haas; Gabriel Waksman

The coupling of ATP binding/hydrolysis to macromolecular secretion systems is crucial to the pathogenicity of Gram‐negative bacteria. We reported previously the structure of the ADP‐bound form of the hexameric traffic VirB11 ATPase of the Helicobacter pylori type IV secretion system (named HP0525), and proposed that it functions as a gating molecule at the inner membrane, cycling through closed and open forms regulated by ATP binding/hydrolysis. Here, we combine crystal structures with analytical ultracentrifugation experiments to show that VirB11 ATPases indeed function as dynamic hexameric assemblies. In the absence of nucleotide, the N‐terminal domains exhibit a collection of rigid‐body conformations. Nucleotide binding ‘locks’ the hexamer into a symmetric and compact structure. We propose that VirB11s use the mechanical leverage generated by such nucleotide‐dependent conformational changes to facilitate the export of substrates or the assembly of the type IV secretion apparatus. Bio chemical characterization of mutant forms of HP0525 coupled with electron microscopy and in vivo assays support such hypothesis, and establish the relevance of VirB11s ATPases as drug targets against pathogenic bacteria.


FEBS Journal | 2011

Assembly and molecular mode of action of the Helicobacter pylori Cag type IV secretion apparatus

Wolfgang Fischer

Bacterial type IV secretion systems (T4SS) form supramolecular protein complexes that are capable of transporting DNA or protein substrates across the bacterial cell envelope and, in many cases, also across eukaryotic target cell membranes. Because of these characteristics, they are often used by pathogenic bacteria for the injection of host cell‐modulating virulence factors. One example is the human pathogen Helicobacteru2003pylori, which uses the Cag‐T4SS to induce a pro‐inflammatory response and multiple cytoskeletal and gene regulatory effects in gastric epithelial cells. Work in recent years has shown that the Cag‐T4SS exhibits marked differences in relation to other systems, both with respect to the composition of its secretion apparatus and the molecular details of its secretion mechanisms. This review describes the molecular properties of the Cag‐T4SS and compares these with prototypical systems of this family of protein transporters.


Journal of Experimental Medicine | 2003

Plasticity of repetitive DNA sequences within a bacterial (type IV) secretion system component

Rahul A. Aras; Wolfgang Fischer; Guillermo I. Perez-Perez; Marialuisa Crosatti; Takafumi Ando; Rainer Haas; Martin J. Blaser

DNA rearrangement permits bacteria to regulate gene content and expression. In Helicobacter pylori, cagY, which contains an extraordinary number of direct DNA repeats, encodes a surface-exposed subunit of a (type IV) bacterial secretory system. Examining potential DNA rearrangements involving the cagY repeats indicated that recombination events invariably yield in-frame open reading frames, producing alternatively expressed genes. In individual hosts, H. pylori cell populations include strains that produce CagY proteins that differ in size, due to the predicted in-frame deletions or duplications, and elicit minimal or no host antibody recognition. Using repetitive DNA, H. pylori rearrangements in a host-exposed subunit of a conserved bacterial secretion system may permit a novel form of antigenic evasion.


Molecular Microbiology | 2006

A C-terminal translocation signal is necessary, but not sufficient for type IV secretion of the Helicobacter pylori CagA protein

Sabine Hohlfeld; Isabelle Pattis; J. Puls; Gregory V. Plano; Rainer Haas; Wolfgang Fischer

Type IV secretion systems are increasingly recognized as important virulence determinants of Gram‐negative bacterial pathogens. While the examination of several type IV‐secreted proteins suggested that their secretion depends on C‐terminal signals, the nature of these signals and their conservation among different systems remain unclear. Here, we have characterized the secretion signal of the Helicobacter pylori CagA protein, which is translocated by the Cag type IV secretion apparatus into eucaryotic cells. The production of fusion proteins of CagA and green fluorescent protein (GFP) did not result in translocation of GFP to epithelial cells, but a fusion of GFP with the CagA C‐terminus exerted a dominant‐negative effect upon wild‐type CagA translocation. We show that CagA translocation depends on the presence of its 20 C‐terminal amino acids, containing an array of positively charged residues. Interestingly, these positive charges are neither necessary nor sufficient for CagA translocation, but replacing the C‐terminal region of CagA with that of other type IV‐secreted proteins reconstitutes CagA translocation competence. Using a novel type IV translocation assay with a phosphorylatable peptide tag, we show that removal of the N‐terminal part of the CagA protein renders the protein translocation‐incompetent as well. Thus, the Cag type IV secretion system seems to diverge from other systems not only with respect to its composition and architecture, but also in terms of substrate recognition and transport.


Current Topics in Microbiology and Immunology | 2009

Virulence Mechanisms and Persistence Strategies of the Human Gastric Pathogen Helicobacter pylori

Wolfgang Fischer; Sandra Prassl; Rainer Haas

The human gastric pathogen Helicobacter pylori is able to establish an infection in a hostile environment with virtually no competitors. For this purpose, it has elaborated a set of colonization factors which facilitate both survival under acid exposure, motility and orientation in a highly viscous mucus layer, and adherence to epithelial surfaces. A more intimate interaction with gastric epithelia provides the basis to influence gene expression profiles as well as morphological transitions via signaling cascades or via direct activities of virulence factors. H. pylori is also one of the most genetically diverse of organisms, and variations are not only found in outer membrane adhesins, but also in two major virulence factors, the VacA cytotoxin and the cag pathogenicity island. Both factors are able to target different cell types and different interaction partners to induce a wide range of possible cellular effects. Despite the fact that H. pylori elicits a strong inflammatory response, the immune system fails to clear the infection, suggesting that immune evasion strategies are used. The mechanisms for immune evasion include the induction of a strongly polarized immune response, a modulation of phagocytosis and neutrophil function, and an inhibition of lymphocyte proliferation. Prolonged inflammation and direct action of bacterial factors may lead to impairment of gland function and eventually to carcinogenesis.


Infection and Immunity | 2001

Outer Membrane Targeting of Passenger Proteins by the Vacuolating Cytotoxin Autotransporter of Helicobacter pylori

Wolfgang Fischer; Renate Buhrdorf; Elke Gerland; Rainer Haas

ABSTRACT Helicobacter pylori produces a number of proteins associated with the outer membrane, including adhesins and the vacuolating cytotoxin. These proteins are supposed to integrate into the outer membrane by β-barrel structures, characteristic of the family of autotransporter proteins. By using the SOMPES (shuttle vector-based outer membrane protein expression) system for outer membrane protein production, we were able to functionally express inH. pylori the cholera toxin B subunit genetically fused to the C-terminal VacA domain. We demonstrate that the fusion protein is translocated to the H. pylori outer membrane and that the CtxB domain is exposed on the H. pylori surface. Thus, we provide the first experimental evidence that the C-terminal β-domain of VacA can transport a foreign passenger protein to theH. pylori surface and hence acts as a functional autotransporter.


Infection and Immunity | 2006

Measurement of Effector Protein Injection by Type III and Type IV Secretion Systems by Using a 13-Residue Phosphorylatable Glycogen Synthase Kinase Tag

Julie Torruellas Garcia; Franco Ferracci; Michael W. Jackson; Sabrina S. Joseph; Isabelle Pattis; Lisa R. W. Plano; Wolfgang Fischer; Gregory V. Plano

ABSTRACT Numerous bacterial pathogens use type III secretion systems (T3SSs) or T4SSs to inject or translocate virulence proteins into eukaryotic cells. Several different reporter systems have been developed to measure the translocation of these proteins. In this study, a peptide tag-based reporter system was developed and used to monitor the injection of T3S and T4S substrates. The glycogen synthase kinase (GSK) tag is a 13-residue phosphorylatable peptide tag derived from the human GSK-3β kinase. Translocation of a GSK-tagged protein into a eukaryotic cell results in host cell protein kinase-dependent phosphorylation of the tag, which can be detected with phosphospecific GSK-3β antibodies. A series of expression plasmids encoding Yop-GSK fusion proteins were constructed to evaluate the ability of the GSK tag to measure the injection of Yops by the Yersinia pestis T3SS. GSK-tagged YopE, YopH, LcrQ, YopK, YopN, and YopJ were efficiently phosphorylated when translocated into HeLa cells. Similarly, the injection of GSK-CagA by the Helicobacter pylori T4SS into different cell types was measured via phosphorylation of the GSK tag. The GSK tag provides a simple method to monitor the translocation of T3S and T4S substrates.


PLOS ONE | 2010

Helicobacter pylori-Induced Histone Modification, Associated Gene Expression in Gastric Epithelial Cells, and Its Implication in Pathogenesis

Song-Ze Ding; Wolfgang Fischer; Maria Kaparakis-Liaskos; George Liechti; D. Scott Merrell; Patrick A. Grant; Richard L. Ferrero; Sheila E. Crowe; Rainer Haas; Masanori Hatakeyama; Joanna B. Goldberg

Histone modifications are critical in regulating gene expression, cell cycle, cell proliferation, and development. Relatively few studies have investigated whether Helicobacter pylori, the major cause of human gastric diseases, affects histone modification. We therefore investigated the effects of H. pylori infection on histone modifications in a global and promoter-specific manner in gastric epithelial cells. Infection of gastric epithelial cells by wild-type H. pylori induced time- and dose-dependent dephosphorylation of histone H3 at serine 10 (H3 Ser10) and decreased acetylation of H3 lysine 23, but had no effects on seven other specific modifications. Different cag pathogenicity island (PAI)-containing-clinical isolates showed similar abilities to induce H3 Ser10 dephosphorylation. Mutation of cagA, vacA, nonphosphorylateable CagA mutant cagAEPISA, or disruption of the flagella showed no effects, while deletion of the entire cagPAI restored the H3 Ser10 phosphorylation to control levels. Analysis of 27 cagPAI mutants indicated that the genes that caused H3 Ser10 dephosphorylation were similar to those that were previously found to induce interleukin-8, irrespective of CagA translocation. This effect was independent of ERK or p38 pathways and type I interferon signaling. Additionally, c-Jun and hsp70 gene expression was associated with this histone modification. These results demonstrate that H. pylori alters histone modification and host response via a cagA-, vacA-independent, but cagPAI-dependent mechanisms, which contribute to its persistent infection and pathogenesis.

Collaboration


Dive into the Wolfgang Fischer's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Manfred Rohde

University of Göttingen

View shared research outputs
Top Co-Authors

Avatar

Michael Naumann

Otto-von-Guericke University Magdeburg

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge