1
|
Handy NB, Xu Y, Moon D, Sowizral JJ, Moon E, Ho M, Wilson BA. Hierarchical determinants in cytotoxic necrotizing factor (CNF) toxins driving Rho G-protein deamidation versus transglutamination. mBio 2024; 15:e0122124. [PMID: 38920360 PMCID: PMC11253639 DOI: 10.1128/mbio.01221-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2024] [Accepted: 05/22/2024] [Indexed: 06/27/2024] Open
Abstract
The cytotoxic necrotizing factor (CNF) family of AB-type bacterial protein toxins catalyze two types of modification on their Rho GTPase substrates: deamidation and transglutamination. It has been established that E. coli CNF1 and its close homolog proteins catalyze primarily deamidation and Bordetella dermonecrotic toxin (DNT) catalyzes primarily transglutamination. The rapidly expanding microbial genome sequencing data have revealed that there are at least 13 full-length variants of CNF1 homologs. CNFx from E. coli strain GN02091 is the most distant from all other members of the CNF family with 50%-55% sequence identity at the protein level and 0.45-0.52 nucleotide substitutions per site at the DNA level. CNFx modifies RhoA, Rac1, and Cdc42, and like CNF1, activates downstream SRE-dependent mitogenic signaling pathways in human HEK293T cells, but at a 1,000-fold higher EC50 value. Unlike other previously characterized CNF toxins, CNFx modifies Rho proteins primarily through transglutamination, as evidenced by gel-shift assay and confirmed by MALDI mass spectral analysis, when coexpressed with Rho-protein substrates in E. coli BL21 cells or through direct treatment of HEK293T cells. A comparison of CNF1 and CNFx sequences identified two critical active-site residues corresponding to positions 832 and 862 in CNF1. Reciprocal site-specific mutations at these residues in each toxin revealed hierarchical rules that define the preference for deamidase versus a transglutaminase activity in CNFs. An additional unique Cys residue at the C-terminus of CNFx was also discovered to be critical for retarding cargo delivery.IMPORTANCECytotoxic necrotizing factor (CNF) toxins not only play important virulence roles in pathogenic E. coli and other bacterial pathogens, but CNF-like genes have also been found in an expanding number of genomes from clinical isolates. Harnessing the power of evolutionary relationships among the CNF toxins enabled the deciphering of the hierarchical active-site determinants that define whether they modify their Rho GTPase substrates through deamidation or transglutamination. With our finding that a distant CNF variant (CNFx) unlike other known CNFs predominantly transglutaminates its Rho GTPase substrates, the paradigm of "CNFs deamidate and DNTs transglutaminate" could finally be attributed to two critical amino acid residues within the active site other than the previously identified catalytic Cys-His dyad residues. The significance of our approach and research findings is that they can be applied to deciphering enzyme reaction determinants and substrate specificities for other bacterial proteins in the development of precision therapeutic strategies.
Collapse
Affiliation(s)
- Nicholas B. Handy
- Department of Microbiology, School of Molecular and Cellular Biology, University of Illinois at Urbana-Champaign, Urbana, Illinois, USA
| | - Yiting Xu
- Department of Microbiology, School of Molecular and Cellular Biology, University of Illinois at Urbana-Champaign, Urbana, Illinois, USA
| | - Damee Moon
- Department of Microbiology, School of Molecular and Cellular Biology, University of Illinois at Urbana-Champaign, Urbana, Illinois, USA
| | - Jacob J. Sowizral
- Department of Microbiology, School of Molecular and Cellular Biology, University of Illinois at Urbana-Champaign, Urbana, Illinois, USA
| | - Eric Moon
- Department of Microbiology, School of Molecular and Cellular Biology, University of Illinois at Urbana-Champaign, Urbana, Illinois, USA
| | - Mengfei Ho
- Department of Microbiology, School of Molecular and Cellular Biology, University of Illinois at Urbana-Champaign, Urbana, Illinois, USA
| | - Brenda A. Wilson
- Department of Microbiology, School of Molecular and Cellular Biology, University of Illinois at Urbana-Champaign, Urbana, Illinois, USA
| |
Collapse
|
2
|
The Cytotoxic Necrotizing Factors (CNFs)-A Family of Rho GTPase-Activating Bacterial Exotoxins. Toxins (Basel) 2021; 13:toxins13120901. [PMID: 34941738 PMCID: PMC8709095 DOI: 10.3390/toxins13120901] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2021] [Revised: 11/29/2021] [Accepted: 12/01/2021] [Indexed: 11/17/2022] Open
Abstract
The cytotoxic necrotizing factors (CNFs) are a family of Rho GTPase-activating single-chain exotoxins that are produced by several Gram-negative pathogenic bacteria. Due to the pleiotropic activities of the targeted Rho GTPases, the CNFs trigger multiple signaling pathways and host cell processes with diverse functional consequences. They influence cytokinesis, tissue integrity, cell barriers, and cell death, as well as the induction of inflammatory and immune cell responses. This has an enormous influence on host-pathogen interactions and the severity of the infection. The present review provides a comprehensive insight into our current knowledge of the modular structure, cell entry mechanisms, and the mode of action of this class of toxins, and describes their influence on the cell, tissue/organ, and systems levels. In addition to their toxic functions, possibilities for their use as drug delivery tool and for therapeutic applications against important illnesses, including nervous system diseases and cancer, have also been identified and are discussed.
Collapse
|
3
|
Carlini F, Maroccia Z, Fiorentini C, Travaglione S, Fabbri A. Effects of the Escherichia coli Bacterial Toxin Cytotoxic Necrotizing Factor 1 on Different Human and Animal Cells: A Systematic Review. Int J Mol Sci 2021; 22:ijms222212610. [PMID: 34830494 PMCID: PMC8621085 DOI: 10.3390/ijms222212610] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2021] [Revised: 11/19/2021] [Accepted: 11/20/2021] [Indexed: 12/13/2022] Open
Abstract
Cytotoxic necrotizing factor 1 (CNF1) is a bacterial virulence factor, the target of which is represented by Rho GTPases, small proteins involved in a huge number of crucial cellular processes. CNF1, due to its ability to modulate the activity of Rho GTPases, represents a widely used tool to unravel the role played by these regulatory proteins in different biological processes. In this review, we summarized the data available in the scientific literature concerning the observed in vitro effects induced by CNF1. An article search was performed on electronic bibliographic resources. Screenings were performed of titles, abstracts, and full-texts according to PRISMA guidelines, whereas eligibility criteria were defined for in vitro studies. We identified a total of 299 records by electronic article search and included 76 original peer-reviewed scientific articles reporting morphological or biochemical modifications induced in vitro by soluble CNF1, either recombinant or from pathogenic Escherichia coli extracts highly purified with chromatographic methods. Most of the described CNF1-induced effects on cultured cells are ascribable to the modulating activity of the toxin on Rho GTPases and the consequent effects on actin cytoskeleton organization. All in all, the present review could be a prospectus about the CNF1-induced effects on cultured cells reported so far.
Collapse
Affiliation(s)
- Francesca Carlini
- Department of Cardiovascular, Endocrine-Metabolic Diseases and Ageing, Istituto Superiore di Sanità, 00161 Rome, Italy; (F.C.); (Z.M.); (S.T.)
| | - Zaira Maroccia
- Department of Cardiovascular, Endocrine-Metabolic Diseases and Ageing, Istituto Superiore di Sanità, 00161 Rome, Italy; (F.C.); (Z.M.); (S.T.)
| | - Carla Fiorentini
- Associazione Ricerca Terapie Oncologiche Integrate, ARTOI, 00165 Rome, Italy;
| | - Sara Travaglione
- Department of Cardiovascular, Endocrine-Metabolic Diseases and Ageing, Istituto Superiore di Sanità, 00161 Rome, Italy; (F.C.); (Z.M.); (S.T.)
| | - Alessia Fabbri
- Department of Cardiovascular, Endocrine-Metabolic Diseases and Ageing, Istituto Superiore di Sanità, 00161 Rome, Italy; (F.C.); (Z.M.); (S.T.)
- Correspondence: ; Tel.: +39-06-4990-2939
| |
Collapse
|
4
|
Morgan RN, Saleh SE, Farrag HA, Aboulwafa MM. Bacterial cyclomodulins: types and roles in carcinogenesis. Crit Rev Microbiol 2021; 48:42-66. [PMID: 34265231 DOI: 10.1080/1040841x.2021.1944052] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
Various studies confirmed that bacterial infections contribute to carcinogenesis through the excessive accumulation of reactive oxygen species (ROS) and the expression of toxins that disrupt the cell cycle phases, cellular regulatory mechanisms and stimulate the production of tumorigenic inflammatory mediators. These toxins mimic carcinogens which act upon key cellular targets and result in mutations and genotoxicities. The cyclomodulins are bacterial toxins that incur cell cycle modulating effects rendering the expressing bacterial species of high carcinogenic potentiality. They are either cellular proliferating or cell cycle arrest cyclomodulins. Notably, cyclomodulins expressing bacterial species have been linked to different human carcinomas. For instance, Escherichia coli species producing the colibactin were highly prevalent among colorectal carcinoma patients, CagA+ Helicobacter pylori species were associated with MALT lymphomas and gastric carcinomas and Salmonella species producing CdtB were linked to hepatobiliary carcinomas. These species stimulated the overgrowth of pre-existing carcinomas and induced hyperplasia in in vivo animal models suggesting a role for the cyclomodulins in carcinogenesis. Wherefore, the prevalence and mode of action of these toxins were the focus of many researchers and studies. This review discusses different types of bacterial cyclomodulins highlighting their mode of action and possible role in carcinogenesis.
Collapse
Affiliation(s)
- Radwa N Morgan
- Drug radiation research Department, Egyptian Atomic Energy Authority (EAEA), National Center for Radiation Research and Technology (NCRRT), Cairo, Egypt
| | - Sarra E Saleh
- Faculty of Pharmacy, Microbiology and Immunology Department, Ain Shams University, Cairo, Egypt
| | - Hala A Farrag
- Drug radiation research Department, Egyptian Atomic Energy Authority (EAEA), National Center for Radiation Research and Technology (NCRRT), Cairo, Egypt
| | - Mohammad M Aboulwafa
- Faculty of Pharmacy, Microbiology and Immunology Department, Ain Shams University, Cairo, Egypt.,Faculty of Pharmacy, King Salman International University, Ras-Sedr, Egypt
| |
Collapse
|
5
|
Ho M, Mettouchi A, Wilson BA, Lemichez E. CNF1-like deamidase domains: common Lego bricks among cancer-promoting immunomodulatory bacterial virulence factors. Pathog Dis 2018; 76:4992304. [PMID: 29733372 DOI: 10.1093/femspd/fty045] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2018] [Accepted: 05/01/2018] [Indexed: 12/28/2022] Open
Abstract
Alterations of the cellular proteome over time due to spontaneous or toxin-mediated enzymatic deamidation of glutamine (Gln) and asparagine (Asn) residues contribute to bacterial infection and might represent a source of aging-related diseases. Here, we put into perspective what is known about the mode of action of the CNF1 toxin from pathogenic Escherichia coli, a paradigm of bacterial deamidases that activate Rho GTPases, to illustrate the importance of determining whether exposure to these factors are risk factors in the etiology age-related diseases, such as cancer. In particular, through in silico analysis of the distribution of the CNF1-like deamidase active site Gly-Cys-(Xaa)n-His sequence motif in bacterial genomes, we unveil the wide distribution of the super-family of CNF-like toxins and CNF-like deamidase domains among members of the Enterobacteriacae and in association with a large variety of toxin delivery systems. We extent our discussion with recent findings concerning cellular systems that control activated Rac1 GTPase stability and provide protection against cancer. These findings point to the urgency for developing holistic approaches toward personalized medicine that include monitoring for asymptomatic carriage of pathogenic toxin-producing bacteria and that ultimately might lead to improved public health and increased lifespans.
Collapse
Affiliation(s)
- Mengfei Ho
- Department of Microbiology, School of Molecular and Cellular Biology, University of Illinois at Urbana-Champaign, Illinois 61801, USA
| | - Amel Mettouchi
- Bacterial Toxins Unit, Department of Microbiology, Institut Pasteur, 25 Rue du Docteur Roux, 75724 Paris, France
| | - Brenda A Wilson
- Department of Microbiology, School of Molecular and Cellular Biology, University of Illinois at Urbana-Champaign, Illinois 61801, USA
| | - Emmanuel Lemichez
- Bacterial Toxins Unit, Department of Microbiology, Institut Pasteur, 25 Rue du Docteur Roux, 75724 Paris, France
| |
Collapse
|
6
|
Haywood EE, Ho M, Wilson BA. Modular domain swapping among the bacterial cytotoxic necrotizing factor (CNF) family for efficient cargo delivery into mammalian cells. J Biol Chem 2018; 293:3860-3870. [PMID: 29371399 DOI: 10.1074/jbc.ra117.001381] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2017] [Revised: 01/11/2018] [Indexed: 11/06/2022] Open
Abstract
Modular AB-type bacterial protein toxins target mammalian host cells with high specificity and deliver their toxic cargo into the cytosol. Hence, these toxins are being explored as agents for targeted cytosolic delivery in biomedical and research applications. The cytotoxic necrotizing factor (CNF) family is unique among these toxins in that their homologous sequences are found in a wide array of bacteria, and their activity domains are packaged in various delivery systems. Here, to study how CNF cargo and delivery modules can be assembled for efficient cytosolic delivery, we generated chimeric toxins by swapping functional domains among CNF1, CNF2, CNF3, and CNFy. Chimeras with a CNFy delivery vehicle were more stably expressed, but were less efficient at cargo delivery into HEK293-T cells. We also found that CNFy cargo is the most universally compatible and that CNF3 delivery vehicle is the most flexible and efficient at delivering cargo. These findings suggest that domains within proteins can be swapped and accommodate each other for efficient function and that an individual domain could be engineered for compatibility with multiple partner domains. We anticipate that our insights could help inform chemical biology approaches to develop toxin-based cargo-delivery platforms for cytosolic cargo delivery of therapeutics or molecular probes into mammalian cells.
Collapse
Affiliation(s)
- Elizabeth E Haywood
- From the Department of Microbiology, School of Molecular and Cellular Biology, University of Illinois Urbana-Champaign, Urbana, Illinois 61801
| | - Mengfei Ho
- From the Department of Microbiology, School of Molecular and Cellular Biology, University of Illinois Urbana-Champaign, Urbana, Illinois 61801
| | - Brenda A Wilson
- From the Department of Microbiology, School of Molecular and Cellular Biology, University of Illinois Urbana-Champaign, Urbana, Illinois 61801
| |
Collapse
|
7
|
Antibodies against hemolysin and cytotoxic necrotizing factor type 1 (CNF1) reduce bladder inflammation in a mouse model of urinary tract infection with toxigenic uropathogenic Escherichia coli. Infect Immun 2015; 83:1661-73. [PMID: 25667267 DOI: 10.1128/iai.02848-14] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Uropathogenic Escherichia coli (UPEC) is the leading cause of cystitis. Cytotoxic necrotizing factor 1 (CNF1) and hemolysin (Hly) are toxins made by approximately 50% of UPEC isolates. CNF1 and Hly contribute to the robust inflammatory response in the bladders of mice challenged with UPEC strain CP9. We hypothesized that antibodies against CNF1 and/or Hly would reduce cystitis caused by CP9. To test this theory, we immunized female C3H/HeOuJ mice subcutaneously with a genetically derived Hly toxoid or genetically derived CNF1 toxoid plus sublethal doses of CNF1. We collected serum and observed increasing titers of specific and neutralizing antibodies against Hly or CNF1 over time. We challenged the mice intraurethrally with CP9 and euthanized them 24 h later. We observed 10-fold lower bacterial titers in the urine of Hly-immunized mice than in that of sham-immunized mice but no difference in kidney bacterial titers. Immunized mice also exhibited significantly less cystitis than sham-immunized mice. In CNF1-vaccinated mice, we detected neither a difference in urine or kidney bacterial titers nor a reduction in the severity of cystitis versus that of sham-immunized mice. We then passively administered an anti-CNF1 monoclonal antibody intraperitoneally to female C3H/HeOuJ mice prior to intraurethral challenge with CP9. Upon challenge, we noted no difference in colonization of the urine or kidney; however, cystitis was reduced significantly in mice treated with the anti-CNF1 antibody versus that in the bladders of mice given an isotype control antibody. Taken together, our data demonstrate that antibodies against CNF1 or Hly reduce the bladder pathology caused by UPEC.
Collapse
|
8
|
What a difference a Dalton makes: bacterial virulence factors modulate eukaryotic host cell signaling systems via deamidation. Microbiol Mol Biol Rev 2014; 77:527-39. [PMID: 24006474 DOI: 10.1128/mmbr.00013-13] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Pathogenic bacteria commonly deploy enzymes to promote virulence. These enzymes can modulate the functions of host cell targets. While the actions of some enzymes can be very obvious (e.g., digesting plant cell walls), others have more subtle activities. Depending on the lifestyle of the bacteria, these subtle modifications can be crucially important for pathogenesis. In particular, if bacteria rely on a living host, subtle mechanisms to alter host cellular function are likely to dominate. Several bacterial virulence factors have evolved to use enzymatic deamidation as a subtle posttranslational mechanism to modify the functions of host protein targets. Deamidation is the irreversible conversion of the amino acids glutamine and asparagine to glutamic acid and aspartic acid, respectively. Interestingly, all currently characterized bacterial deamidases affect the function of the target protein by modifying a single glutamine residue in the sequence. Deamidation of target host proteins can disrupt host signaling and downstream processes by either activating or inactivating the target. Despite the subtlety of this modification, it has been shown to cause dramatic, context-dependent effects on host cells. Several crystal structures of bacterial deamidases have been solved. All are members of the papain-like superfamily and display a cysteine-based catalytic triad. However, these proteins form distinct structural subfamilies and feature combinations of modular domains of various functions. Based on the diverse pathogens that use deamidation as a mechanism to promote virulence and the recent identification of multiple deamidases, it is clear that this enzymatic activity is emerging as an important and widespread feature in bacterial pathogenesis.
Collapse
|
9
|
Aktories K, Schmidt G, Lang AE. Photorhabdus luminescens toxins TccC3 and TccC5: insecticidal ADP-ribosyltransferases that modify threonine and glutamine. Curr Top Microbiol Immunol 2014; 384:53-67. [PMID: 24908144 DOI: 10.1007/82_2014_382] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
The ADP-ribosyltransferases TccC3 and TccC5 are the biologically active TcC components of the tripartite Photorhabdus luminescens Tc toxin, which consist of TcA, TcB, and TcC components. TcA is the binding and membrane translocation component. TcB is a functional linker between TcC and TcA and also involved in the translocation of the toxin. While TccC3 ADP-ribosylates actin at threonine 148, TccC5 modifies Rho proteins at glutamine 61/63. Both modifications result in major alteration of the actin cytoskeleton. Here we discuss structure and function of the Tc toxin and compare its ADP-ribosyltransferase activities with other types of actin and Rho modifying toxins.
Collapse
Affiliation(s)
- Klaus Aktories
- Institut für Experimentelle und Klinische Pharmakologie und Toxikologie, Albert-Ludwigs-Universität Freiburg, Albertstr. 25, 79104, Freiburg, Germany,
| | | | | |
Collapse
|
10
|
Piteau M, Papatheodorou P, Schwan C, Schlosser A, Aktories K, Schmidt G. Lu/BCAM adhesion glycoprotein is a receptor for Escherichia coli Cytotoxic Necrotizing Factor 1 (CNF1). PLoS Pathog 2014; 10:e1003884. [PMID: 24453976 PMCID: PMC3894216 DOI: 10.1371/journal.ppat.1003884] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2013] [Accepted: 11/27/2013] [Indexed: 01/05/2023] Open
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.
Collapse
Affiliation(s)
- Marianne Piteau
- Institute for Experimental and Clinical Pharmacology and Toxicology, Albert-Ludwigs-University of Freiburg, Freiburg, Germany
- Biological Faculty, Albert-Ludwigs-University of Freiburg, Freiburg, Germany
| | - Panagiotis Papatheodorou
- Institute for Experimental and Clinical Pharmacology and Toxicology, Albert-Ludwigs-University of Freiburg, Freiburg, Germany
| | - Carsten Schwan
- Institute for Experimental and Clinical Pharmacology and Toxicology, Albert-Ludwigs-University of Freiburg, Freiburg, Germany
| | - Andreas Schlosser
- Rudolf-Virchow-Zentrum für Experimentelle Biomedizin, Universität Würzburg, Würzburg, Germany
| | - Klaus Aktories
- Institute for Experimental and Clinical Pharmacology and Toxicology, Albert-Ludwigs-University of Freiburg, Freiburg, Germany
- BIOSS (Centre for Biological Signalling Studies), Freiburg, Germany
| | - Gudula Schmidt
- Institute for Experimental and Clinical Pharmacology and Toxicology, Albert-Ludwigs-University of Freiburg, Freiburg, Germany
| |
Collapse
|
11
|
Singh MK, Ren F, Giesemann T, Dal Bosco C, Pasternak TP, Blein T, Ruperti B, Schmidt G, Aktories K, Molendijk AJ, Palme K. Modification of plant Rac/Rop GTPase signalling using bacterial toxin transgenes. THE PLANT JOURNAL : FOR CELL AND MOLECULAR BIOLOGY 2013; 73:314-24. [PMID: 23020817 DOI: 10.1111/tpj.12040] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/19/2012] [Revised: 09/20/2012] [Accepted: 09/27/2012] [Indexed: 05/23/2023]
Abstract
Bacterial protein toxins which modify Rho GTPase are useful for the analysis of Rho signalling in animal cells, but these toxins cannot be taken up by plant cells. We demonstrate in vitro deamidation of Arabidopsis Rop4 by Escherichia coli Cytotoxic Necrotizing Factor 1 (CNF1) and glucosylation by Clostridium difficile toxin B. Expression of the catalytic domain of CNF1 caused modification and activation of co-expressed Arabidopsis Rop4 GTPase in tobacco leaves, resulting in hypersensitive-like cell death. By contrast, the catalytic domain of toxin B modified and inactivated co-expressed constitutively active Rop4, blocking the hypersensitive response caused by over-expression of active Rops. In transgenic Arabidopsis, both CNF1 and toxin B inhibited Rop-dependent polar morphogenesis of leaf epidermal cells. Toxin B expression also inhibited Rop-dependent morphogenesis of root hairs and trichome branching, and resulted in root meristem enlargement and dwarf growth. Our results show that CNF1 and toxin B transgenes are effective tools in Rop GTPase signalling studies.
Collapse
Affiliation(s)
- Manoj K Singh
- Faculty of Biology, Institute for Biology II/Botany, Albert-Ludwigs-University of Freiburg, Schänzlestrasse 1, 79104, Freiburg, Germany
| | - Fugang Ren
- Faculty of Biology, Institute for Biology II/Botany, Albert-Ludwigs-University of Freiburg, Schänzlestrasse 1, 79104, Freiburg, Germany
| | - Torsten Giesemann
- Institute for Clinical and Experimental Pharmacology and Toxicology, Albert-Ludwigs-University of Freiburg, Albertstrasse 25, 79104, Freiburg, Germany
| | - Cristina Dal Bosco
- Faculty of Biology, Institute for Biology II/Botany, Albert-Ludwigs-University of Freiburg, Schänzlestrasse 1, 79104, Freiburg, Germany
| | - Taras P Pasternak
- Faculty of Biology, Institute for Biology II/Botany, Albert-Ludwigs-University of Freiburg, Schänzlestrasse 1, 79104, Freiburg, Germany
| | - Thomas Blein
- Faculty of Biology, Institute for Biology II/Botany, Albert-Ludwigs-University of Freiburg, Schänzlestrasse 1, 79104, Freiburg, Germany
| | - Benedetto Ruperti
- Department of Environmental Agronomy and Crop Science, University of Padova, Via Romea, 16, Agripolis, 35020, Legnaro, Padova, Italy
| | - Gudula Schmidt
- Institute for Clinical and Experimental Pharmacology and Toxicology, Albert-Ludwigs-University of Freiburg, Albertstrasse 25, 79104, Freiburg, Germany
| | - Klaus Aktories
- Institute for Clinical and Experimental Pharmacology and Toxicology, Albert-Ludwigs-University of Freiburg, Albertstrasse 25, 79104, Freiburg, Germany
| | - Arthur J Molendijk
- Faculty of Biology, Institute for Biology II/Botany, Albert-Ludwigs-University of Freiburg, Schänzlestrasse 1, 79104, Freiburg, Germany
- Freiburg Initiative for Systems Biology (FRISYS), Albert-Ludwigs-University of Freiburg, Schänzlestrasse 1, 79104, Freiburg, Germany
| | - Klaus Palme
- Faculty of Biology, Institute for Biology II/Botany, Albert-Ludwigs-University of Freiburg, Schänzlestrasse 1, 79104, Freiburg, Germany
- Freiburg Initiative for Systems Biology (FRISYS), Albert-Ludwigs-University of Freiburg, Schänzlestrasse 1, 79104, Freiburg, Germany
- Centre of Biological Systems Analysis, Albert-Ludwigs-University of Freiburg, Habsburgerstrasse 49, 79104, Freiburg, Germany
- Freiburg Institute for Advanced Sciences (FRIAS), Albert-Ludwigs-University of Freiburg, Albertstrasse 19, 79104, Freiburg, Germany
- Centre for Biological Signalling Studies (BIOSS), Albert-Ludwigs-University of Freiburg, Albertstrasse 19, 79104, Freiburg, Germany
| |
Collapse
|
12
|
Reipschläger S, Kubatzky K, Taromi S, Burger M, Orth J, Aktories K, Schmidt G. Toxin-induced RhoA activity mediates CCL1-triggered signal transducers and activators of transcription protein signaling. J Biol Chem 2012; 287:11183-94. [PMID: 22311973 DOI: 10.1074/jbc.m111.313395] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
RhoA is reportedly involved in signal transducers and activators of transcription (STAT)-dependent transcription. However, the pathway connecting the GTPase and STAT signaling has not been characterized. Here, we made use of bacterial toxins, which directly activate Rho GTPases to analyze this pathway. Cytotoxic necrotizing factors (CNFs) are produced by pathogenic Escherichia coli strains and by Yersinia pseudotuberculosis. They activate small GTPases of the Rho family by deamidation of a glutamine, which is crucial for GTP hydrolysis. We show that RhoA activation leads to phosphorylation and activation of STAT3 and identify signal proteins involved in this pathway. RhoA-dependent STAT3 stimulation requires ROCK and Jun kinase activation as well as AP1-induced protein synthesis. The secretion of one or more factors activates the JAK-STAT pathway in an auto/paracrine manner. We identify CCL1/I-309 as an essential cytokine, which is produced and secreted upon RhoA activation and which is able to activate STAT3-dependent signaling pathways.
Collapse
Affiliation(s)
- Simone Reipschläger
- Institute for Experimental and Clinical Pharmacology and Toxicology, Albert-Ludwigs-University of Freiburg, Albert-Str. 25, 79104 Freiburg, Germany
| | | | | | | | | | | | | |
Collapse
|
13
|
Pasteurella multocida toxin interaction with host cells: entry and cellular effects. Curr Top Microbiol Immunol 2012; 361:93-111. [PMID: 22552700 PMCID: PMC4408768 DOI: 10.1007/82_2012_219] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
The mitogenic dermonecrotic toxin from Pasteurella multocida (PMT) is a 1285-residue multipartite protein that belongs to the A-B family of bacterial protein toxins. Through its G-protein-deamidating activity on the α subunits of heterotrimeric G(q)-, G(i)- and G(12/13)-proteins, PMT potently stimulates downstream mitogenic, calcium, and cytoskeletal signaling pathways. These activities lead to pleiotropic effects in different cell types, which ultimately result in cellular proliferation, while inhibiting cellular differentiation, and account for the myriad of physiological outcomes observed during infection with toxinogenic strains of P. multocida.
Collapse
|
14
|
Orth JHC, Aktories K. Molecular biology of Pasteurella multocida toxin. Curr Top Microbiol Immunol 2012; 361:73-92. [PMID: 22371145 DOI: 10.1007/82_2012_201] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Pasteurella multocida toxin (PMT) is the causative agent of progressive atrophic rhinitis in swine. The 146 kDa single-chain toxin harbours discrete domains important for receptor binding, internalisation and biological activity. The molecular basis of the toxin's activity is the deamidation of a specific glutamine residue in the α-subunit of heterotrimeric G proteins. This results in an inhibition of the inherent GTPase activity leading to a constitutively active phenotype of the G protein. Due to the ability of the toxin to act on various families of heterotrimeric G proteins, a large subset of signal transduction pathways is stimulated.
Collapse
Affiliation(s)
- Joachim H C Orth
- Institut für Experimentelle und Klinische Pharmakologie und Toxikologie, Albert-Ludwigs-Universität Freiburg, Albertstr. 25, 79104, Freiburg, Germany.
| | | |
Collapse
|
15
|
Brothers MC, Ho M, Maharjan R, Clemons NC, Bannai Y, Waites MA, Faulkner MJ, Kuhlenschmidt TB, Kuhlenschmidt MS, Blanke SR, Rienstra CM, Wilson BA. Membrane interaction of Pasteurella multocida toxin involves sphingomyelin. FEBS J 2011; 278:4633-48. [PMID: 21951695 PMCID: PMC3220749 DOI: 10.1111/j.1742-4658.2011.08365.x] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Pasteurella multocida toxin (PMT) is an AB toxin that causes pleiotropic effects in targeted host cells. The N-terminus of PMT (PMT-N) is considered to harbor the membrane receptor binding and translocation domains responsible for mediating cellular entry and delivery of the C-terminal catalytic domain into the host cytosol. Previous studies have implicated gangliosides as the host receptors for PMT binding. To gain further insight into the binding interactions involved in PMT binding to cell membranes, we explored the role of various membrane components in PMT binding, utilizing four different approaches: (a) TLC-overlay binding experiments with (125) I-labeled PMT, PMT-N or the C-terminus of PMT; (b) pull-down experiments using reconstituted membrane liposomes with full-length PMT; (c) surface plasmon resonance analysis of PMT-N binding to reconstituted membrane liposomes; (d) and surface plasmon resonance analysis of PMT-N binding to HEK-293T cell membranes without or with sphingomyelinase, phospholipase D or trypsin treatment. The results obtained revealed that, in our experimental system, full-length PMT and PMT-N did not bind to gangliosides, including monoasialogangliosides GM(1) , GM(2) or GM(3) , but instead bound to membrane phospholipids, primarily the abundant sphingophospholipid sphingomyelin or phosphatidylcholine with other lipid components. Collectively, these studies demonstrate the importance of sphingomyelin for PMT binding to membranes and suggest the involvement of a protein co-receptor.
Collapse
Affiliation(s)
| | - Mengfei Ho
- Department of Microbiology, University of Illinois, Urbana, IL 61801, USA
- Institute for Genomic Biology, University of Illinois, Urbana, IL 61801, USA
| | - Ram Maharjan
- Department of Microbiology, University of Illinois, Urbana, IL 61801, USA
| | - Nathan C. Clemons
- Department of Microbiology, University of Illinois, Urbana, IL 61801, USA
| | - Yuka Bannai
- Department of Microbiology, University of Illinois, Urbana, IL 61801, USA
| | - Mark A. Waites
- Department of Microbiology, University of Illinois, Urbana, IL 61801, USA
| | | | | | | | - Steven R. Blanke
- Department of Microbiology, University of Illinois, Urbana, IL 61801, USA
- Institute for Genomic Biology, University of Illinois, Urbana, IL 61801, USA
| | - Chad M. Rienstra
- Department of Chemistry, University of Illinois, Urbana, IL 61801, USA
- Department of Biochemistry, University of Illinois, Urbana, IL 61801, USA
| | - Brenda A. Wilson
- Department of Microbiology, University of Illinois, Urbana, IL 61801, USA
- Institute for Genomic Biology, University of Illinois, Urbana, IL 61801, USA
| |
Collapse
|
16
|
Abstract
Many bacterial pathogens produce protein toxins to outmanoeuvre the immune system of the host. Some of these proteins target regulatory GTPases such as those belonging to the RHO family, which control the actin cytoskeleton of the host cell. In this Review, I discuss a diversity of mechanisms that are used by bacterial effectors and toxins to modulate the activity of host GTPases, with a focus on covalent modifications such as ADP-ribosylation, glucosylation, adenylylation, proteolysis, deamidation and transglutamination.
Collapse
Affiliation(s)
- Klaus Aktories
- Institut für Experimentelle und Klinische Pharmakologie und Toxikologie, Albert-Ludwigs-Universität Freiburg, Albertstr. 25, 79104 Freiburg, Germany.
| |
Collapse
|
17
|
Knust Z, Schmidt G. Cytotoxic Necrotizing Factors (CNFs)-A Growing Toxin Family. Toxins (Basel) 2011; 2:116-27. [PMID: 22069550 PMCID: PMC3206620 DOI: 10.3390/toxins2010116] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2009] [Revised: 01/15/2010] [Accepted: 01/20/2010] [Indexed: 12/25/2022] Open
Abstract
The Escherichia coli Cytotoxic Necrotizing Factors, CNF1, CNF2, CNF3 and CNFY from Yersinia pseudotuberculosis belong to a family of deamidating toxins. CNFs deamidate glutamine 63/61 in the switch II region of Rho GTPases that is essential for GTP hydrolysing activity. Deamidation leads to constitutive activation of Rho GTPases. However, cellular mechanisms like proteasomal degradation of the activated Rho proteins restrict the action of the GTPases. This review describes the differences between the toxin family members concerning expression, cellular entry and substrate specificity.
Collapse
Affiliation(s)
- Zeynep Knust
- Institut für Experimentelle und Klinische Pharmakologie und Toxikologie, Albert-Ludwigs-Universität Freiburg, Albertstr. 25, 79104 Freiburg, Germany.
| | | |
Collapse
|
18
|
Abstract
Small GTPases of the Rho protein family are master regulators of the actin cytoskeleton and are targeted by potent virulence factors of several pathogenic bacteria. Their dysfunctional regulation can lead to severe human pathologies. Both host and bacterial factors can activate or inactivate Rho proteins by direct post-translational modifications: such as deamidation and transglutamination for activation, or ADP-ribosylation, glucosylation, adenylylation and phosphorylation for inactivation. We review and compare these unconventional ways in which both host cells and bacterial pathogens regulate Rho proteins.
Collapse
|
19
|
Thalmann J, Janik K, May M, Sommer K, Ebeling J, Hofmann F, Genth H, Klos A. Actin re-organization induced by Chlamydia trachomatis serovar D--evidence for a critical role of the effector protein CT166 targeting Rac. PLoS One 2010; 5:e9887. [PMID: 20360858 PMCID: PMC2845625 DOI: 10.1371/journal.pone.0009887] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2009] [Accepted: 02/26/2010] [Indexed: 11/18/2022] Open
Abstract
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.
Collapse
Affiliation(s)
- Jessica Thalmann
- Institute for Medical Microbiology and Hospital Epidemiology, Hannover Medical School, Hannover, Germany
| | | | | | | | | | | | | | | |
Collapse
|