1
|
Tozzi M, Fiore A, Travaglione S, Marcon F, Rainaldi G, Germinario EAP, Laterza I, Donati S, Macchia D, Spada M, Leoni O, Quattrini MC, Pietraforte D, Tomasoni S, Torrigiani F, Verin R, Matarrese P, Gambardella L, Spadaro F, Carollo M, Pietrantoni A, Carlini F, Panebianco C, Pazienza V, Colella F, Lucchetti D, Sgambato A, Sistigu A, Moschella F, Guidotti M, Vincentini O, Maroccia Z, Biffoni M, De Angelis R, Bracci L, Fabbri A. E. Coli cytotoxic necrotizing factor-1 promotes colorectal carcinogenesis by causing oxidative stress, DNA damage and intestinal permeability alteration. J Exp Clin Cancer Res 2025; 44:29. [PMID: 39876002 PMCID: PMC11776187 DOI: 10.1186/s13046-024-03271-w] [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: 11/23/2024] [Accepted: 12/31/2024] [Indexed: 01/30/2025] Open
Abstract
BACKGROUND Bacterial toxins are emerging as promising hallmarks of colorectal cancer (CRC) pathogenesis. In particular, Cytotoxic Necrotizing Factor 1 (CNF1) from E. coli deserves special consideration due to the significantly higher prevalence of this toxin gene in CRC patients with respect to healthy subjects, and to the numerous tumor-promoting effects that have been ascribed to the toxin in vitro. Despite this evidence, a definitive causal link between CNF1 and CRC was missing. Here we investigated whether CNF1 plays an active role in CRC onset by analyzing pro-carcinogenic key effects specifically induced by the toxin in vitro and in vivo. METHODS Viability assays, confocal microscopy of γH2AX and 53BP1 molecules and cytogenetic analysis were carried out to assess CNF1-induced genotoxicity on non-neoplastic intestinal epithelial cells. Caco-2 monolayers and 3D Caco-2 spheroids were used to evaluate permeability alterations specifically induced by CNF1, either in the presence or in the absence of inflammation. In vivo, an inflammatory bowel disease (IBD) model was exploited to evaluate the carcinogenic potential of CNF1. Immunohistochemistry and immunofluorescence stainings of formalin-fixed paraffin-embedded (FFPE) colon tissue were carried out as well as fecal microbiota composition analysis by 16 S rRNA gene sequencing. RESULTS CNF1 induces the release of reactive oxidizing species and chromosomal instability in non-neoplastic intestinal epithelial cells. In addition, CNF1 modifies intestinal permeability by directly altering tight junctions' distribution in 2D Caco-2 monolayers, and by hindering the differentiation of 3D Caco-2 spheroids with an irregular arrangement of these junctions. In vivo, repeated intrarectal administration of CNF1 induces the formation of dysplastic aberrant crypt foci (ACF), and produces the formation of colorectal adenomas in an IBD model. These effects are accompanied by the increased neutrophilic infiltration in colonic tissue, by a mixed pro-inflammatory and anti-inflammatory cytokine milieu, and by the pro-tumoral modulation of the fecal microbiota. CONCLUSIONS Taken together, our results support the hypothesis that the CNF1 toxin from E. coli plays an active role in colorectal carcinogenesis. Altogether, these findings not only add new knowledge to the contribution of bacterial toxins to CRC, but also pave the way to the implementation of current screening programs and preventive strategies.
Collapse
Affiliation(s)
- Michela Tozzi
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, Rome, Italy
| | - Alessia Fiore
- Department of Cardiovascular, Endocrine-Metabolic Diseases and Aging, Istituto Superiore di Sanità, Rome, Italy
| | - Sara Travaglione
- Department of Cardiovascular, Endocrine-Metabolic Diseases and Aging, Istituto Superiore di Sanità, Rome, Italy
| | - Francesca Marcon
- Department of Environment and Health, Istituto Superiore di Sanità, Rome, Italy
| | - Gabriella Rainaldi
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, Rome, Italy
| | - Elena Angela Pia Germinario
- Department of Cardiovascular, Endocrine-Metabolic Diseases and Aging, Istituto Superiore di Sanità, Rome, Italy
| | - Ilenia Laterza
- Department of Cardiovascular, Endocrine-Metabolic Diseases and Aging, Istituto Superiore di Sanità, Rome, Italy
| | - Simona Donati
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, Rome, Italy
| | - Daniele Macchia
- Center of Animal Research and Welfare, Istituto Superiore di Sanità, Rome, Italy
| | - Massimo Spada
- Center of Animal Research and Welfare, Istituto Superiore di Sanità, Rome, Italy
| | - Omar Leoni
- Center of Animal Research and Welfare, Istituto Superiore di Sanità, Rome, Italy
| | | | | | - Sofia Tomasoni
- Department of Comparative Biomedicine and Food Science, BCA-University of Padua, Legnaro, PD, Italy
| | - Filippo Torrigiani
- Department of Comparative Biomedicine and Food Science, BCA-University of Padua, Legnaro, PD, Italy
| | - Ranieri Verin
- Department of Comparative Biomedicine and Food Science, BCA-University of Padua, Legnaro, PD, Italy
| | - Paola Matarrese
- Center for Gender-Specific Medicine, Istituto Superiore di Sanità, Rome, Italy
| | | | | | - Maria Carollo
- Core Facilities, Istituto Superiore di Sanità, Rome, Italy
| | | | - Francesca Carlini
- Department of Cardiovascular, Endocrine-Metabolic Diseases and Aging, Istituto Superiore di Sanità, Rome, Italy
| | - Concetta Panebianco
- Division of Gastroenterology, Fondazione IRCCS "Casa Sollievo della Sofferenza", San Giovanni Rotondo, FG, Italy
| | - Valerio Pazienza
- Division of Gastroenterology, Fondazione IRCCS "Casa Sollievo della Sofferenza", San Giovanni Rotondo, FG, Italy
| | - Filomena Colella
- Multiplex Spatial Profiling Center, Fondazione Policlinico Universitario "A. Gemelli" - IRCCS, Rome, Italy
| | - Donatella Lucchetti
- Multiplex Spatial Profiling Center, Fondazione Policlinico Universitario "A. Gemelli" - IRCCS, Rome, Italy
- Department of Translational Medicine and Surgery, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Alessandro Sgambato
- Multiplex Spatial Profiling Center, Fondazione Policlinico Universitario "A. Gemelli" - IRCCS, Rome, Italy
- Department of Translational Medicine and Surgery, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Antonella Sistigu
- Department of Translational Medicine and Surgery, Università Cattolica del Sacro Cuore, Rome, Italy
- Fondazione Policlinico Universitario "A. Gemelli" - IRCCS, Rome, Italy
| | - Federica Moschella
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, Rome, Italy
| | - Marco Guidotti
- Department of Food Safety, Nutrition and Veterinary Public Health, Istituto Superiore di Sanità, Rome, Italy
| | - Olimpia Vincentini
- Department of Food Safety, Nutrition and Veterinary Public Health, Istituto Superiore di Sanità, Rome, Italy
| | - Zaira Maroccia
- Department of Cardiovascular, Endocrine-Metabolic Diseases and Aging, Istituto Superiore di Sanità, Rome, Italy
| | - Mauro Biffoni
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, Rome, Italy
| | - Roberta De Angelis
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, Rome, Italy
| | - Laura Bracci
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, Rome, Italy.
| | - Alessia Fabbri
- Department of Cardiovascular, Endocrine-Metabolic Diseases and Aging, Istituto Superiore di Sanità, Rome, Italy
| |
Collapse
|
2
|
Jeon S, Jeon Y, Lim JY, Kim Y, Cha B, Kim W. Emerging regulatory mechanisms and functions of biomolecular condensates: implications for therapeutic targets. Signal Transduct Target Ther 2025; 10:4. [PMID: 39757214 DOI: 10.1038/s41392-024-02070-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2024] [Revised: 10/01/2024] [Accepted: 11/06/2024] [Indexed: 01/07/2025] Open
Abstract
Cells orchestrate their processes through complex interactions, precisely organizing biomolecules in space and time. Recent discoveries have highlighted the crucial role of biomolecular condensates-membrane-less assemblies formed through the condensation of proteins, nucleic acids, and other molecules-in driving efficient and dynamic cellular processes. These condensates are integral to various physiological functions, such as gene expression and intracellular signal transduction, enabling rapid and finely tuned cellular responses. Their ability to regulate cellular signaling pathways is particularly significant, as it requires a careful balance between flexibility and precision. Disruption of this balance can lead to pathological conditions, including neurodegenerative diseases, cancer, and viral infections. Consequently, biomolecular condensates have emerged as promising therapeutic targets, with the potential to offer novel approaches to disease treatment. In this review, we present the recent insights into the regulatory mechanisms by which biomolecular condensates influence intracellular signaling pathways, their roles in health and disease, and potential strategies for modulating condensate dynamics as a therapeutic approach. Understanding these emerging principles may provide valuable directions for developing effective treatments targeting the aberrant behavior of biomolecular condensates in various diseases.
Collapse
Affiliation(s)
- Soyoung Jeon
- Department of Life Science, University of Seoul, Seoul, South Korea
| | - Yeram Jeon
- Department of Life Science, University of Seoul, Seoul, South Korea
| | - Ji-Youn Lim
- New Drug Development Center, Daegu-Gyeongbuk Medical Innovation Foundation, Daegu, South Korea
| | - Yujeong Kim
- Department of Life Science, University of Seoul, Seoul, South Korea
| | - Boksik Cha
- New Drug Development Center, Daegu-Gyeongbuk Medical Innovation Foundation, Daegu, South Korea.
| | - Wantae Kim
- Department of Life Science, University of Seoul, Seoul, South Korea.
| |
Collapse
|
3
|
Danov A, Pollin I, Moon E, Ho M, Wilson BA, Papathanos PA, Kaplan T, Levy A. Identification of novel toxins associated with the extracellular contractile injection system using machine learning. Mol Syst Biol 2024; 20:859-879. [PMID: 39069594 PMCID: PMC11297309 DOI: 10.1038/s44320-024-00053-6] [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: 01/24/2024] [Revised: 06/06/2024] [Accepted: 06/27/2024] [Indexed: 07/30/2024] Open
Abstract
Secretion systems play a crucial role in microbe-microbe or host-microbe interactions. Among these systems, the extracellular contractile injection system (eCIS) is a unique bacterial and archaeal extracellular secretion system that injects protein toxins into target organisms. However, the specific proteins that eCISs inject into target cells and their functions remain largely unknown. Here, we developed a machine learning classifier to identify eCIS-associated toxins (EATs). The classifier combines genetic and biochemical features to identify EATs. We also developed a score for the eCIS N-terminal signal peptide to predict EAT loading. Using the classifier we classified 2,194 genes from 950 genomes as putative EATs. We validated four new EATs, EAT14-17, showing toxicity in bacterial and eukaryotic cells, and identified residues of their respective active sites that are critical for toxicity. Finally, we show that EAT14 inhibits mitogenic signaling in human cells. Our study provides insights into the diversity and functions of EATs and demonstrates machine learning capability of identifying novel toxins. The toxins can be employed in various applications dependently or independently of eCIS.
Collapse
Affiliation(s)
- Aleks Danov
- Department of Plant Pathology and Microbiology, Institute of Environmental Sciences, The Robert H. Smith Faculty of Agriculture, Food & Environment, The Hebrew University of Jerusalem, Rehovot, 7610001, Israel
| | - Inbal Pollin
- Department of Plant Pathology and Microbiology, Institute of Environmental Sciences, The Robert H. Smith Faculty of Agriculture, Food & Environment, The Hebrew University of Jerusalem, Rehovot, 7610001, Israel
| | - Eric Moon
- Department of Microbiology, University of Illinois Urbana-Champaign, 601 South Goodwin Ave, Urbana, 61801, IL, USA
| | - Mengfei Ho
- Department of Microbiology, University of Illinois Urbana-Champaign, 601 South Goodwin Ave, Urbana, 61801, IL, USA
| | - Brenda A Wilson
- Department of Microbiology, University of Illinois Urbana-Champaign, 601 South Goodwin Ave, Urbana, 61801, IL, USA
| | - Philippos A Papathanos
- Department of Entomology, Institute of Environmental Sciences, The Robert H. Smith Faculty of Agriculture, Food & Environment, The Hebrew University of Jerusalem, Rehovot, 7610001, Israel
| | - Tommy Kaplan
- School of Computer Science and Engineering, The Hebrew University of Jerusalem, Jerusalem, Israel
- Department of Developmental Biology and Cancer Research, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Asaf Levy
- Department of Plant Pathology and Microbiology, Institute of Environmental Sciences, The Robert H. Smith Faculty of Agriculture, Food & Environment, The Hebrew University of Jerusalem, Rehovot, 7610001, Israel.
| |
Collapse
|
4
|
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
|
5
|
Popoff MR. Overview of Bacterial Protein Toxins from Pathogenic Bacteria: Mode of Action and Insights into Evolution. Toxins (Basel) 2024; 16:182. [PMID: 38668607 PMCID: PMC11054074 DOI: 10.3390/toxins16040182] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2024] [Revised: 03/29/2024] [Accepted: 03/30/2024] [Indexed: 04/29/2024] Open
Abstract
Bacterial protein toxins are secreted by certain bacteria and are responsible for mild to severe diseases in humans and animals. They are among the most potent molecules known, which are active at very low concentrations. Bacterial protein toxins exhibit a wide diversity based on size, structure, and mode of action. Upon recognition of a cell surface receptor (protein, glycoprotein, and glycolipid), they are active either at the cell surface (signal transduction, membrane damage by pore formation, or hydrolysis of membrane compound(s)) or intracellularly. Various bacterial protein toxins have the ability to enter cells, most often using an endocytosis mechanism, and to deliver the effector domain into the cytosol, where it interacts with an intracellular target(s). According to the nature of the intracellular target(s) and type of modification, various cellular effects are induced (cell death, homeostasis modification, cytoskeleton alteration, blockade of exocytosis, etc.). The various modes of action of bacterial protein toxins are illustrated with representative examples. Insights in toxin evolution are discussed.
Collapse
Affiliation(s)
- Michel R Popoff
- Unité des Toxines Bactériennes, Institut Pasteur, Université Paris Cité, CNRS UMR 2001 INSERM U1306, F-75015 Paris, France
| |
Collapse
|
6
|
Chat H, Dalmasso G, Godfraind C, Bonnin V, Beyrouthy R, Bonnet M, Barnich N, Mettouchi A, Lemichez E, Bonnet R, Delmas J. Cytotoxic necrotizing factor 1 hinders colon tumorigenesis induced by colibactin-producing Escherichia coli in ApcMin/+ mice. Gut Microbes 2023; 15:2229569. [PMID: 37417545 PMCID: PMC10332217 DOI: 10.1080/19490976.2023.2229569] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/24/2022] [Accepted: 06/21/2023] [Indexed: 07/08/2023] Open
Abstract
Colorectal cancer (CRC) patients are frequently colonized by colibactin-producing Escherichia coli (CoPEC) (>40%), which enhances tumorigenesis in mouse models of CRC. We observed that 50% of CoPEC also contains the cnf1 gene, which encodes cytotoxic necrotizing factor-1 (CNF1), an enhancer of the eukaryotic cell cycle. The impact of its co-occurrence with colibactin (Clb) has not yet been investigated. We evaluated the impact of CNF1 on colorectal tumorigenesis using human colonic epithelial HT-29 cells and CRC-susceptible ApcMin/+ mice inoculated with the CoPEC 21F8 clinical strain (Clb+Cnf+) or 21F8 isogenic mutants (Clb+Cnf-, Clb-Cnf+ and Clb-Cnf-). Infection with the Clb+Cnf- strain induced higher levels of inflammatory cytokines and senescence markers both in vitro and in vivo compared to those induced by infection with the Clb+Cnf+ strain. In contrast, the Clb+Cnf- and Clb+Cnf+ strains generated similar levels of DNA damage in HT-29 cells and in colonic murine tissues. Furthermore, the ApcMin/+ mice inoculated with the Clb+Cnf- strain developed significantly more tumors than the mice inoculated with the Clb+Cnf+ strain or the isogenic mutants, and the composition of their microbiota was changed. Finally, rectal administration of the CNF1 protein in ApcMin/+ mice inoculated with the Clb+Cnf- strain significantly decreased tumorigenesis and inflammation. Overall, this study provides evidence that CNF1 decreases the carcinogenic effects of CoPEC in ApcMin/+ mice by decreasing CoPEC-induced cellular senescence and inflammation.
Collapse
Affiliation(s)
- Héloïse Chat
- Centre de Recherche en Nutrition Humaine Auvergne, University Clermont Auvergne, Inserm U1071, INRAE USC 1382, Microbes, Intestin, Inflammation et Susceptibilité de l’Hôte (M2iSH), Clermont-Ferrand, France
| | - Guillaume Dalmasso
- Centre de Recherche en Nutrition Humaine Auvergne, University Clermont Auvergne, Inserm U1071, INRAE USC 1382, Microbes, Intestin, Inflammation et Susceptibilité de l’Hôte (M2iSH), Clermont-Ferrand, France
| | - Catherine Godfraind
- Centre de Recherche en Nutrition Humaine Auvergne, University Clermont Auvergne, Inserm U1071, INRAE USC 1382, Microbes, Intestin, Inflammation et Susceptibilité de l’Hôte (M2iSH), Clermont-Ferrand, France
- Neuropathology Unit, University Hospital of Clermont-Ferrand, Clermont-Ferrand, France
| | - Virginie Bonnin
- Centre de Recherche en Nutrition Humaine Auvergne, University Clermont Auvergne, Inserm U1071, INRAE USC 1382, Microbes, Intestin, Inflammation et Susceptibilité de l’Hôte (M2iSH), Clermont-Ferrand, France
| | - Racha Beyrouthy
- Centre de Recherche en Nutrition Humaine Auvergne, University Clermont Auvergne, Inserm U1071, INRAE USC 1382, Microbes, Intestin, Inflammation et Susceptibilité de l’Hôte (M2iSH), Clermont-Ferrand, France
| | - Mathilde Bonnet
- Centre de Recherche en Nutrition Humaine Auvergne, University Clermont Auvergne, Inserm U1071, INRAE USC 1382, Microbes, Intestin, Inflammation et Susceptibilité de l’Hôte (M2iSH), Clermont-Ferrand, France
- Institut Universitaire de Technologie, University Clermont Auvergne, Clermont-Ferrand, France
| | - Nicolas Barnich
- Centre de Recherche en Nutrition Humaine Auvergne, University Clermont Auvergne, Inserm U1071, INRAE USC 1382, Microbes, Intestin, Inflammation et Susceptibilité de l’Hôte (M2iSH), Clermont-Ferrand, France
- Institut Universitaire de Technologie, University Clermont Auvergne, Clermont-Ferrand, France
| | - Amel Mettouchi
- Institut Pasteur, University of Paris, CNRS UMR2001, Paris, France
| | | | - Richard Bonnet
- Centre de Recherche en Nutrition Humaine Auvergne, University Clermont Auvergne, Inserm U1071, INRAE USC 1382, Microbes, Intestin, Inflammation et Susceptibilité de l’Hôte (M2iSH), Clermont-Ferrand, France
- Department of Bacteriology, University Hospital of Clermont-Ferrand, Clermont-Ferrand, France
| | - Julien Delmas
- Centre de Recherche en Nutrition Humaine Auvergne, University Clermont Auvergne, Inserm U1071, INRAE USC 1382, Microbes, Intestin, Inflammation et Susceptibilité de l’Hôte (M2iSH), Clermont-Ferrand, France
- Department of Bacteriology, University Hospital of Clermont-Ferrand, Clermont-Ferrand, France
| |
Collapse
|
7
|
Kubatzky KF. Pasteurella multocida toxin - lessons learned from a mitogenic toxin. Front Immunol 2022; 13:1058905. [PMID: 36591313 PMCID: PMC9800868 DOI: 10.3389/fimmu.2022.1058905] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2022] [Accepted: 11/29/2022] [Indexed: 12/23/2022] Open
Abstract
The gram-negative, zoonotic bacterium Pasteurella multocida was discovered in 1880 and found to be the causative pathogen of fowl cholera. Pasteurella-related diseases can be found in domestic and wild life animals such as buffalo, sheep, goat, deer and antelope, cats, dogs and tigers and cause hemorrhagic septicemia in cattle, rhinitis or pneumonia in rabbits or fowl cholera in poultry and birds. Pasteurella multocida does not play a major role in the immune-competent human host, but can be found after animal bites or in people with close contact to animals. Toxigenic strains are most commonly found in pigs and express a phage-encoded 146 kDa protein, the Pasteurella multocida toxin (PMT). Toxin-expressing strains cause atrophic rhinitis where nasal turbinate bones are destroyed through the inhibition of bone building osteoblasts and the activation of bone resorbing osteoclasts. After its uptake through receptor-mediated endocytosis, PMT specifically targets the alpha subunit of several heterotrimeric G proteins and constitutively activates them through deamidation of a glutamine residue to glutamate in the alpha subunit. This results in cytoskeletal rearrangement, proliferation, differentiation and survival of cells. Because of the toxin's mitogenic effects, it was suggested that it might have carcinogenic properties, however, no link between Pasteurella infections and cell transformation could be established, neither in tissue culture models nor through epidemiological data. In the recent years it was shown that the toxin not only affects bone, but also the heart as well as basically all cells of innate and adaptive immunity. During the last decade the focus of research shifted from signal transduction processes to understanding how the bacteria might benefit from a bone-destroying toxin. The primary function of PMT seems to be the modulation of immune cell activation which at the same time creates an environment permissive for osteoclast formation. While the disease is restricted to pigs, the implications of the findings from PMT research can be used to explore human diseases and have a high translational potential. In this review our current knowledge will be summarized and it will be discussed what can be learned from using PMT as a tool to understand human pathologies.
Collapse
Affiliation(s)
- Katharina F. Kubatzky
- Department of Infectious Diseases, Medical Microbiology and Hygiene, Heidelberg University, Heidelberg, Germany
| |
Collapse
|
8
|
Markelova NN, Semenova EF, Sineva ON, Sadykova VS. The Role of Cyclomodulins and Some Microbial Metabolites in Bacterial Microecology and Macroorganism Carcinogenesis. Int J Mol Sci 2022; 23:ijms231911706. [PMID: 36233008 PMCID: PMC9570213 DOI: 10.3390/ijms231911706] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2022] [Revised: 09/24/2022] [Accepted: 09/26/2022] [Indexed: 11/16/2022] Open
Abstract
A number of bacteria that colonize the human body produce toxins and effectors that cause changes in the eukaryotic cell cycle—cyclomodulins and low-molecular-weight compounds such as butyrate, lactic acid, and secondary bile acids. Cyclomodulins and metabolites are necessary for bacteria as adaptation factors—which are influenced by direct selection—to the ecological niches of the host. In the process of establishing two-way communication with the macroorganism, these compounds cause limited damage to the host, despite their ability to disrupt key processes in eukaryotic cells, which can lead to pathological changes. Possible negative consequences of cyclomodulin and metabolite actions include their potential role in carcinogenesis, in particular, with the ability to cause DNA damage, increase genome instability, and interfere with cancer-associated regulatory pathways. In this review, we aim to examine cyclomodulins and bacterial metabolites as important factors in bacterial survival and interaction with the host organism to show their heterogeneous effect on oncogenesis depending on the surrounding microenvironment, pathological conditions, and host genetic background.
Collapse
Affiliation(s)
- Natalia N. Markelova
- Gause Institute of New Antibiotics, ul. Bolshaya Pirogovskaya, 11, 119021 Moscow, Russia
- Correspondence: (N.N.M.); (V.S.S.)
| | - Elena F. Semenova
- Institute of Biochemical Technology, Ecology and Pharmacy, V.I. Vernadsky Crimean Federal University, 295007 Simferopol, Russia
| | - Olga N. Sineva
- Gause Institute of New Antibiotics, ul. Bolshaya Pirogovskaya, 11, 119021 Moscow, Russia
| | - Vera S. Sadykova
- Gause Institute of New Antibiotics, ul. Bolshaya Pirogovskaya, 11, 119021 Moscow, Russia
- Correspondence: (N.N.M.); (V.S.S.)
| |
Collapse
|
9
|
Wu Y, Mahtal N, Paillares E, Swistak L, Sagadiev S, Acharya M, Demeret C, Werf SVD, Guivel-Benhassine F, Schwartz O, Petracchini S, Mettouchi A, Caramelle L, Couvineau P, Thai R, Barbe P, Keck M, Brodin P, Machelart A, Sencio V, Trottein F, Sachse M, Chicanne G, Payrastre B, Ville F, Kreis V, Popoff MR, Johannes L, Cintrat JC, Barbier J, Gillet D, Lemichez E. C910 chemical compound inhibits the traffiking of several bacterial AB toxins with cross-protection against influenza virus. iScience 2022; 25:104537. [PMID: 35769882 PMCID: PMC9234246 DOI: 10.1016/j.isci.2022.104537] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2021] [Revised: 04/20/2022] [Accepted: 06/01/2022] [Indexed: 11/23/2022] Open
Abstract
The development of anti-infectives against a large range of AB-like toxin-producing bacteria includes the identification of compounds disrupting toxin transport through both the endolysosomal and retrograde pathways. Here, we performed a high-throughput screening of compounds blocking Rac1 proteasomal degradation triggered by the Cytotoxic Necrotizing Factor-1 (CNF1) toxin, which was followed by orthogonal screens against two toxins that hijack the endolysosomal (diphtheria toxin) or retrograde (Shiga-like toxin 1) pathways to intoxicate cells. This led to the identification of the molecule C910 that induces the enlargement of EEA1-positive early endosomes associated with sorting defects of CNF1 and Shiga toxins to their trafficking pathways. C910 protects cells against eight bacterial AB toxins and the CNF1-mediated pathogenic Escherichia coli invasion. Interestingly, C910 reduces influenza A H1N1 and SARS-CoV-2 viral infection in vitro. Moreover, parenteral administration of C910 to mice resulted in its accumulation in lung tissues and a reduction in lethal influenza infection.
Collapse
Affiliation(s)
- Yu Wu
- Unité des Toxines Bactériennes, UMR CNRS 6047, Inserm U1306, Institut Pasteur, 25 rue du Dr Roux, 75724 Paris, France
| | - Nassim Mahtal
- Université Paris-Saclay, CEA, INRAE, Département Médicaments et Technologies pour la Santé, SIMoS, 91191 Gif-sur-Yvette, France
- Université Paris-Saclay, CEA, INRAE, Département Médicaments et Technologies pour la Santé, SCBM, 91191 Gif-sur-Yvette, France
| | - Eléa Paillares
- Unité des Toxines Bactériennes, UMR CNRS 6047, Inserm U1306, Institut Pasteur, 25 rue du Dr Roux, 75724 Paris, France
- Université Paris Cité, 75006 Paris, France
| | - Léa Swistak
- Unité des Toxines Bactériennes, UMR CNRS 6047, Inserm U1306, Institut Pasteur, 25 rue du Dr Roux, 75724 Paris, France
- Université Paris Cité, 75006 Paris, France
| | - Sara Sagadiev
- Seattle Children’s Research Institute, Jack R MacDonald Building, 1900 9th Avenue, Seattle, WA 98101, USA
| | - Mridu Acharya
- Seattle Children’s Research Institute, Jack R MacDonald Building, 1900 9th Avenue, Seattle, WA 98101, USA
| | - Caroline Demeret
- Unité Génétique Moléculaire des Virus à ARN, UMR 3569 CNRS, Université de Paris, Département de Virologie, Institut Pasteur, 28 rue du Dr Roux, 75724 Paris, France
| | - Sylvie Van Der Werf
- Unité Génétique Moléculaire des Virus à ARN, UMR 3569 CNRS, Université de Paris, Département de Virologie, Institut Pasteur, 28 rue du Dr Roux, 75724 Paris, France
| | - Florence Guivel-Benhassine
- Unité virus et immunité, Département de Virologie, Institut Pasteur, 28 rue du Dr Roux, 75724 Paris, France
| | - Olivier Schwartz
- Unité virus et immunité, Département de Virologie, Institut Pasteur, 28 rue du Dr Roux, 75724 Paris, France
| | - Serena Petracchini
- Unité des Toxines Bactériennes, UMR CNRS 6047, Inserm U1306, Institut Pasteur, 25 rue du Dr Roux, 75724 Paris, France
- Université Paris Cité, 75006 Paris, France
| | - Amel Mettouchi
- Unité des Toxines Bactériennes, UMR CNRS 6047, Inserm U1306, Institut Pasteur, 25 rue du Dr Roux, 75724 Paris, France
| | - Lucie Caramelle
- Université Paris-Saclay, CEA, INRAE, Département Médicaments et Technologies pour la Santé, SIMoS, 91191 Gif-sur-Yvette, France
| | - Pierre Couvineau
- Université Paris-Saclay, CEA, INRAE, Département Médicaments et Technologies pour la Santé, SIMoS, 91191 Gif-sur-Yvette, France
| | - Robert Thai
- Université Paris-Saclay, CEA, INRAE, Département Médicaments et Technologies pour la Santé, SIMoS, 91191 Gif-sur-Yvette, France
| | - Peggy Barbe
- Université Paris-Saclay, CEA, INRAE, Département Médicaments et Technologies pour la Santé, SIMoS, 91191 Gif-sur-Yvette, France
| | - Mathilde Keck
- Université Paris-Saclay, CEA, INRAE, Département Médicaments et Technologies pour la Santé, SIMoS, 91191 Gif-sur-Yvette, France
| | - Priscille Brodin
- Centre d’Infection et d’Immunité de Lille, Inserm U1019, CNRS UMR 9017, University of Lille, CHU Lille- Institut Pasteur de Lille, 59000 Lille, France
| | - Arnaud Machelart
- Centre d’Infection et d’Immunité de Lille, Inserm U1019, CNRS UMR 9017, University of Lille, CHU Lille- Institut Pasteur de Lille, 59000 Lille, France
| | - Valentin Sencio
- Centre d’Infection et d’Immunité de Lille, Inserm U1019, CNRS UMR 9017, University of Lille, CHU Lille- Institut Pasteur de Lille, 59000 Lille, France
| | - François Trottein
- Centre d’Infection et d’Immunité de Lille, Inserm U1019, CNRS UMR 9017, University of Lille, CHU Lille- Institut Pasteur de Lille, 59000 Lille, France
| | - Martin Sachse
- Unité Technologie et service BioImagerie Ultrastructurale, Institut Pasteur, 28 rue du Dr Roux, 75724 Paris, France
| | - Gaëtan Chicanne
- Inserm, UMR1297 and Université Toulouse III Paul Sabatier, I2MC, 31024 Toulouse, France
| | - Bernard Payrastre
- Inserm, UMR1297 and Université Toulouse III Paul Sabatier, I2MC, 31024 Toulouse, France
| | - Florian Ville
- Université Paris-Saclay, CEA, INRAE, Département Médicaments et Technologies pour la Santé, SIMoS, 91191 Gif-sur-Yvette, France
- Université Paris-Saclay, CEA, INRAE, Département Médicaments et Technologies pour la Santé, SCBM, 91191 Gif-sur-Yvette, France
| | - Victor Kreis
- Université Paris-Saclay, CEA, INRAE, Département Médicaments et Technologies pour la Santé, SIMoS, 91191 Gif-sur-Yvette, France
| | - Michel-Robert Popoff
- Unité des Toxines Bactériennes, UMR CNRS 6047, Inserm U1306, Institut Pasteur, 25 rue du Dr Roux, 75724 Paris, France
| | - Ludger Johannes
- Institut Curie, PSL Research University, Cellular and Chemical Biology unit, Endocytic Trafficking and Intracellular Delivery team, U1143 INSERM, UMR3666 CNRS, 26 rue d'Ulm, 75248 Paris, France
| | - Jean-Christophe Cintrat
- Université Paris-Saclay, CEA, INRAE, Département Médicaments et Technologies pour la Santé, SCBM, 91191 Gif-sur-Yvette, France
| | - Julien Barbier
- Université Paris-Saclay, CEA, INRAE, Département Médicaments et Technologies pour la Santé, SIMoS, 91191 Gif-sur-Yvette, France
| | - Daniel Gillet
- Université Paris-Saclay, CEA, INRAE, Département Médicaments et Technologies pour la Santé, SIMoS, 91191 Gif-sur-Yvette, France
| | - Emmanuel Lemichez
- Unité des Toxines Bactériennes, UMR CNRS 6047, Inserm U1306, Institut Pasteur, 25 rue du Dr Roux, 75724 Paris, France
| |
Collapse
|
10
|
Interactions of Bacterial Toxin CNF1 and Host JAK1/2 Driven by Liquid-Liquid Phase Separation Enhance Macrophage Polarization. mBio 2022; 13:e0114722. [PMID: 35766380 PMCID: PMC9426534 DOI: 10.1128/mbio.01147-22] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Urinary tract infections (UTIs) are a global public health concern, which is mainly caused by uropathogenic Escherichia coli (UPEC). Cytotoxic necrotizing factor 1 (CNF1) is a key UPEC toxin and regulates multiple host cellular processes through activating the Rho GTPases; however, the effect of CNF1 on macrophage polarization remains unknown. Here, we found that CNF1 promoted M1 macrophage polarization through regulating NF-κB and JAK-STAT1 signaling pathways in kidney at an early stage of acute UTIs. Notably, we identified CNF1 could directly interact with JAK1/2 through its domain without Rho GTPases activation, which induced JAK1/2 phosphorylation, subsequent STAT1 activation and M1 polarization. Moreover, CNF1 exhibited liquid-liquid phase separation (LLPS) to induce a CNF1-JAK1/2 complex, promoting macrophage reprogramming. These findings highlight the LLPS-dependent and Rho GTPase-independent effect of CNF1 as an adaptor on interfering with host cell signals.
Collapse
|
11
|
Aravind L, Iyer LM, Burroughs AM. Discovering Biological Conflict Systems Through Genome Analysis: Evolutionary Principles and Biochemical Novelty. Annu Rev Biomed Data Sci 2022; 5:367-391. [PMID: 35609893 DOI: 10.1146/annurev-biodatasci-122220-101119] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Biological replicators, from genes within a genome to whole organisms, are locked in conflicts. Comparative genomics has revealed a staggering diversity of molecular armaments and mechanisms regulating their deployment, collectively termed biological conflict systems. These encompass toxins used in inter- and intraspecific interactions, self/nonself discrimination, antiviral immune mechanisms, and counter-host effectors deployed by viruses and intragenomic selfish elements. These systems possess shared syntactical features in their organizational logic and a set of effectors targeting genetic information flow through the Central Dogma, certain membranes, and key molecules like NAD+. These principles can be exploited to discover new conflict systems through sensitive computational analyses. This has led to significant advances in our understanding of the biology of these systems and furnished new biotechnological reagents for genome editing, sequencing, and beyond. We discuss these advances using specific examples of toxins, restriction-modification, apoptosis, CRISPR/second messenger-regulated systems, and other enigmatic nucleic acid-targeting systems. Expected final online publication date for the Annual Review of Biomedical Data Science, Volume 5 is August 2022. Please see http://www.annualreviews.org/page/journal/pubdates for revised estimates.
Collapse
Affiliation(s)
- L Aravind
- National Center for Biotechnology Information, National Library of Medicine, National Institutes of Health, Bethesda, Maryland, USA;
| | - Lakshminarayan M Iyer
- National Center for Biotechnology Information, National Library of Medicine, National Institutes of Health, Bethesda, Maryland, USA;
| | - A Maxwell Burroughs
- National Center for Biotechnology Information, National Library of Medicine, National Institutes of Health, Bethesda, Maryland, USA;
| |
Collapse
|
12
|
Tsoumtsa Meda LL, Landraud L, Petracchini S, Descorps-Declere S, Perthame E, Nahori MA, Ramirez Finn L, Ingersoll MA, Patiño-Navarrete R, Glaser P, Bonnet R, Dussurget O, Denamur E, Mettouchi A, Lemichez E. The cnf1 gene is associated with an expanding Escherichia coli ST131 H30Rx/C2 subclade and confers a competitive advantage for gut colonization. Gut Microbes 2022; 14:2121577. [PMID: 36154446 PMCID: PMC9519008 DOI: 10.1080/19490976.2022.2121577] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/27/2022] [Accepted: 08/30/2022] [Indexed: 02/04/2023] Open
Abstract
Epidemiological projections point to acquisition of ever-expanding multidrug resistance (MDR) by Escherichia coli, a commensal of the digestive tract and a source of urinary tract pathogens. Bioinformatics analyses of a large collection of E. coli genomes from EnteroBase, enriched in clinical isolates of worldwide origins, suggest the Cytotoxic Necrotizing Factor 1 (CNF1)-toxin encoding gene, cnf1, is preferentially distributed in four common sequence types (ST) encompassing the pandemic E. coli MDR lineage ST131. This lineage is responsible for a majority of extraintestinal infections that escape first-line antibiotic treatment, with known enhanced capacities to colonize the gastrointestinal tract. Statistical projections based on this dataset point to a global expansion of cnf1-positive multidrug-resistant ST131 strains from subclade H30Rx/C2, accounting for a rising prevalence of cnf1-positive strains in ST131. Despite the absence of phylogeographical signals, cnf1-positive isolates segregated into clusters in the ST131-H30Rx/C2 phylogeny, sharing a similar profile of virulence factors and the same cnf1 allele. The suggested dominant expansion of cnf1-positive strains in ST131-H30Rx/C2 led us to uncover the competitive advantage conferred by cnf1 for gut colonization to the clinical strain EC131GY ST131-H30Rx/C2 versus cnf1-deleted isogenic strain. Complementation experiments showed that colon tissue invasion was compromised in the absence of deamidase activity on Rho GTPases by CNF1. Hence, gut colonization factor function of cnf1 was confirmed for another clinical strain ST131-H30Rx/C2. In addition, functional analysis of the cnf1-positive clinical strain EC131GY ST131-H30Rx/C2 and a cnf1-deleted isogenic strain showed no detectable impact of the CNF1 gene on bacterial fitness and inflammation during the acute phase of bladder monoinfection. Together these data argue for an absence of role of CNF1 in virulence during UTI, while enhancing gut colonization capacities of ST131-H30Rx/C2 and suggested expansion of cnf1-positive MDR isolates in subclade ST131-H30Rx/C2.
Collapse
Affiliation(s)
- Landry L. Tsoumtsa Meda
- Institut Pasteur, Université Paris Cité, CNRS UMR6047, INSERM U1306, Unité des Toxines Bactériennes, Département de Microbiologie, Paris, France
| | - Luce Landraud
- Université Paris Cité et Université Sorbonne Paris Nord, INSERM U1137, IAME, Paris, France
- Laboratoire Microbiologie-hygiène, AP-HP, Hôpital Louis Mourier, Colombes, France
| | - Serena Petracchini
- Institut Pasteur, Université Paris Cité, CNRS UMR6047, INSERM U1306, Unité des Toxines Bactériennes, Département de Microbiologie, Paris, France
| | - Stéphane Descorps-Declere
- Institut Pasteur, Université Paris Cité, CNRS UMR6047, INSERM U1306, Unité des Toxines Bactériennes, Département de Microbiologie, Paris, France
- Institut Pasteur, Université Paris Cité, Bioinformatics and Biostatistics Hub, Paris, France
| | - Emeline Perthame
- Institut Pasteur, Université Paris Cité, Bioinformatics and Biostatistics Hub, Paris, France
| | - Marie-Anne Nahori
- Institut Pasteur, Université Paris Cité, CNRS UMR6047, INSERM U1306, Unité des Toxines Bactériennes, Département de Microbiologie, Paris, France
| | - Laura Ramirez Finn
- Institut Pasteur, Department of Immunology, Mucosal Inflammation and Immunity group, Paris, France
- Université Paris Cité, Institut Cochin, CNRS UMR8104, INSERM U1016, Paris, France
| | - Molly A. Ingersoll
- Institut Pasteur, Department of Immunology, Mucosal Inflammation and Immunity group, Paris, France
- Université Paris Cité, Institut Cochin, CNRS UMR8104, INSERM U1016, Paris, France
| | - Rafael Patiño-Navarrete
- Institut Pasteur, Université Paris Cité, CNRS UMR6047, Unité Ecologie et Evolution de la Résistance aux Antibiotiques, Département de Microbiologie, Paris, France
| | - Philippe Glaser
- Institut Pasteur, Université Paris Cité, CNRS UMR6047, Unité Ecologie et Evolution de la Résistance aux Antibiotiques, Département de Microbiologie, Paris, France
| | - Richard Bonnet
- UMR INSERM U1071, INRA USC-2018, Université Clermont Auvergne, Clermont-Ferrand, France
- Centre National de Référence de la Résistance aux Antibiotiques, Centre Hospitalier Universitaire, Clermont-Ferrand, France
| | - Olivier Dussurget
- Institut Pasteur, Université Paris Cité, CNRS UMR6047, Unité de Recherche Yersinia, Département de Microbiologie, Paris, France
| | - Erick Denamur
- Université Paris Cité et Université Sorbonne Paris Nord, INSERM U1137, IAME, Paris, France
- AP-HP, Laboratoire de Génétique Moléculaire, Hôpital Bichat, Paris, France
| | - Amel Mettouchi
- Institut Pasteur, Université Paris Cité, CNRS UMR6047, INSERM U1306, Unité des Toxines Bactériennes, Département de Microbiologie, Paris, France
| | - Emmanuel Lemichez
- Institut Pasteur, Université Paris Cité, CNRS UMR6047, INSERM U1306, Unité des Toxines Bactériennes, Département de Microbiologie, Paris, France
| |
Collapse
|
13
|
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
|
14
|
Conformational Insights into the Control of CNF1 Toxin Activity by Peptidyl-Prolyl Isomerization: A Molecular Dynamics Perspective. Int J Mol Sci 2021; 22:ijms221810129. [PMID: 34576292 PMCID: PMC8467853 DOI: 10.3390/ijms221810129] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2021] [Revised: 09/11/2021] [Accepted: 09/14/2021] [Indexed: 12/18/2022] Open
Abstract
The cytotoxic necrotizing factor 1 (CNF1) toxin from uropathogenic Escherichia coli constitutively activates Rho GTPases by catalyzing the deamidation of a critical glutamine residue located in the switch II (SWII). In crystallographic structures of the CNF1 catalytic domain (CNF1CD), surface-exposed P768 and P968 peptidyl-prolyl imide bonds (X-Pro) adopt an unusual cis conformation. Here, we show that mutation of each proline residue into glycine abrogates CNF1CD in vitro deamidase activity, while mutant forms of CNF1 remain functional on RhoA in cells. Using molecular dynamics simulations coupled to protein-peptide docking, we highlight the long-distance impact of peptidyl-prolyl cis-trans isomerization on the network of interactions between the loops bordering the entrance of the catalytic cleft. The energetically favorable isomerization of P768 compared with P968, induces an enlargement of loop L1 that fosters the invasion of CNF1CD catalytic cleft by a peptide encompassing SWII of RhoA. The connection of the P968 cis isomer to the catalytic cysteine C866 via a ladder of stacking interactions is alleviated along the cis-trans isomerization. Finally, the cis-trans conversion of P768 favors a switch of the thiol side chain of C866 from a resting to an active orientation. The long-distance impact of peptidyl-prolyl cis-trans isomerizations is expected to have implications for target modification.
Collapse
|
15
|
Chaoprasid P, Lukat P, Mühlen S, Heidler T, Gazdag E, Dong S, Bi W, Rüter C, Kirchenwitz M, Steffen A, Jänsch L, Stradal TEB, Dersch P, Blankenfeldt W. Crystal structure of bacterial cytotoxic necrotizing factor CNF Y reveals molecular building blocks for intoxication. EMBO J 2021; 40:e105202. [PMID: 33410511 PMCID: PMC7883292 DOI: 10.15252/embj.2020105202] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2020] [Revised: 11/12/2020] [Accepted: 11/23/2020] [Indexed: 12/30/2022] Open
Abstract
Cytotoxic necrotizing factors (CNFs) are bacterial single-chain exotoxins that modulate cytokinetic/oncogenic and inflammatory processes through activation of host cell Rho GTPases. To achieve this, they are secreted, bind surface receptors to induce endocytosis and translocate a catalytic unit into the cytosol to intoxicate host cells. A three-dimensional structure that provides insight into the underlying mechanisms is still lacking. Here, we determined the crystal structure of full-length Yersinia pseudotuberculosis CNFY . CNFY consists of five domains (D1-D5), and by integrating structural and functional data, we demonstrate that D1-3 act as export and translocation module for the catalytic unit (D4-5) and for a fused β-lactamase reporter protein. We further found that D4, which possesses structural similarity to ADP-ribosyl transferases, but had no equivalent catalytic activity, changed its position to interact extensively with D5 in the crystal structure of the free D4-5 fragment. This liberates D5 from a semi-blocked conformation in full-length CNFY , leading to higher deamidation activity. Finally, we identify CNF translocation modules in several uncharacterized fusion proteins, which suggests their usability as a broad-specificity protein delivery tool.
Collapse
Affiliation(s)
- Paweena Chaoprasid
- Institute of InfectiologyCenter for Molecular Biology of Inflammation (ZMBE)University of MünsterMünsterGermany
- Molecular Infection BiologyHelmholtz Centre for Infection ResearchBraunschweigGermany
| | - Peer Lukat
- Structure and Function of ProteinsHelmholtz Centre for Infection ResearchBraunschweigGermany
| | - Sabrina Mühlen
- Institute of InfectiologyCenter for Molecular Biology of Inflammation (ZMBE)University of MünsterMünsterGermany
- Molecular Infection BiologyHelmholtz Centre for Infection ResearchBraunschweigGermany
- Deutsches Zentrum für InfektionsforschungBraunschweigGermany
| | - Thomas Heidler
- Molecular Structural BiologyHelmholtz Centre for Infection ResearchBraunschweigGermany
| | - Emerich‐Mihai Gazdag
- Structure and Function of ProteinsHelmholtz Centre for Infection ResearchBraunschweigGermany
| | - Shuangshuang Dong
- Structure and Function of ProteinsHelmholtz Centre for Infection ResearchBraunschweigGermany
| | - Wenjie Bi
- Cellular ProteomicsHelmholtz Centre for Infection ResearchBraunschweigGermany
| | - Christian Rüter
- Institute of InfectiologyCenter for Molecular Biology of Inflammation (ZMBE)University of MünsterMünsterGermany
| | - Marco Kirchenwitz
- Cell BiologyHelmholtz Centre for Infection ResearchBraunschweigGermany
| | - Anika Steffen
- Cell BiologyHelmholtz Centre for Infection ResearchBraunschweigGermany
| | - Lothar Jänsch
- Cellular ProteomicsHelmholtz Centre for Infection ResearchBraunschweigGermany
- Institute of ZoologyTechnische Universität BraunschweigBraunschweigGermany
| | - Theresia E B Stradal
- Cell BiologyHelmholtz Centre for Infection ResearchBraunschweigGermany
- Institute of ZoologyTechnische Universität BraunschweigBraunschweigGermany
| | - Petra Dersch
- Institute of InfectiologyCenter for Molecular Biology of Inflammation (ZMBE)University of MünsterMünsterGermany
- Molecular Infection BiologyHelmholtz Centre for Infection ResearchBraunschweigGermany
- Deutsches Zentrum für InfektionsforschungBraunschweigGermany
- Institute of MicrobiologyTechnische Universität BraunschweigBraunschweigGermany
| | - Wulf Blankenfeldt
- Structure and Function of ProteinsHelmholtz Centre for Infection ResearchBraunschweigGermany
- Institute for Biochemistry, Biotechnology and BioinformaticsTechnische Universität BraunschweigBraunschweigGermany
| |
Collapse
|
16
|
Mohseni AH, Taghinezhad-S S, Fu X. Gut microbiota-derived metabolites and colorectal cancer: New insights and updates. Microb Pathog 2020; 149:104569. [DOI: 10.1016/j.micpath.2020.104569] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2020] [Revised: 10/04/2020] [Accepted: 10/12/2020] [Indexed: 12/16/2022]
|