1
|
Jin J, Guo Q, Yan Z. The Role of Lutheran/Basal Cell Adhesion Molecule in Hematological Diseases and Tumors. Int J Mol Sci 2024; 25:7268. [PMID: 39000374 PMCID: PMC11242806 DOI: 10.3390/ijms25137268] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2024] [Revised: 06/21/2024] [Accepted: 06/27/2024] [Indexed: 07/16/2024] Open
Abstract
Cell adhesion is a dynamic process that plays a fundamental role in cell proliferation, maintenance, differentiation, and migration. Basal cell adhesion molecule (BCAM), also known as Lutheran (Lu), belongs to the immunoglobulin superfamily of cell adhesion molecules. Lu/BCAM, which is widely expressed in red blood cells, endothelial cells, smooth muscle cells and epithelial cells across various tissues, playing a crucial role in many cellular processes, including cell adhesion, cell motility and cell migration. Moreover, Lu/BCAM, dysregulated in many diseases, such as blood diseases and various types of cancer, may act as a biomarker and target for the treatment of these diseases. This review explores the significance of Lu/BCAM in cell adhesion and its potential as a novel target for treating hematological diseases and tumors.
Collapse
Affiliation(s)
| | | | - Zhibin Yan
- College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou 310018, China; (J.J.); (Q.G.)
| |
Collapse
|
2
|
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
|
3
|
Song Y, Fothergill LJ, Lee KS, Liu BY, Koo A, Perelis M, Diwakarla S, Callaghan B, Huang J, Wykosky J, Furness JB, Yeo GW. Stratification of enterochromaffin cells by single-cell expression analysis. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.08.24.554649. [PMID: 37662229 PMCID: PMC10473706 DOI: 10.1101/2023.08.24.554649] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/05/2023]
Abstract
Dynamic interactions between gut mucosal cells and the external environment are essential to maintain gut homeostasis. Enterochromaffin (EC) cells transduce both chemical and mechanical signals and produce 5-hydroxytryptamine (5-HT) to mediate disparate physiological responses. However, the molecular and cellular basis for functional diversity of ECs remains to be adequately defined. Here, we integrated single-cell transcriptomics with spatial image analysis to identify fourteen EC clusters that are topographically organized along the gut. Subtypes predicted to be sensitive to the chemical environment and mechanical forces were identified that express distinct transcription factors and hormones. A Piezo2+ population in the distal colon was endowed with a distinctive neuronal signature. Using a combination of genetic, chemogenetic and pharmacological approaches, we demonstrated Piezo2+ ECs are required for normal colon motility. Our study constructs a molecular map for ECs and offers a framework for deconvoluting EC cells with pleiotropic functions.
Collapse
Affiliation(s)
- Yan Song
- Department of Cellular and Molecular Medicine, University of California San Diego, La Jolla, CA 92093, United States
- Stem Cell Program, University of California San Diego, La Jolla, CA 92093, United States
- Institute for Genomic Medicine, University of California San Diego, La Jolla, CA 92093, United States
| | - Linda J. Fothergill
- Department of Anatomy & Physiology, University of Melbourne, Parkville, Victoria 3010, Australia
- Florey Institute of Neuroscience and Mental Health, Parkville, Victoria 3010, Australia
| | - Kari S. Lee
- Department of Cellular and Molecular Medicine, University of California San Diego, La Jolla, CA 92093, United States
- Stem Cell Program, University of California San Diego, La Jolla, CA 92093, United States
- Institute for Genomic Medicine, University of California San Diego, La Jolla, CA 92093, United States
| | - Brandon Y. Liu
- Department of Cellular and Molecular Medicine, University of California San Diego, La Jolla, CA 92093, United States
- Stem Cell Program, University of California San Diego, La Jolla, CA 92093, United States
- Institute for Genomic Medicine, University of California San Diego, La Jolla, CA 92093, United States
| | - Ada Koo
- Department of Anatomy & Physiology, University of Melbourne, Parkville, Victoria 3010, Australia
| | - Mark Perelis
- Department of Cellular and Molecular Medicine, University of California San Diego, La Jolla, CA 92093, United States
- Stem Cell Program, University of California San Diego, La Jolla, CA 92093, United States
- Institute for Genomic Medicine, University of California San Diego, La Jolla, CA 92093, United States
| | - Shanti Diwakarla
- Department of Anatomy & Physiology, University of Melbourne, Parkville, Victoria 3010, Australia
| | - Brid Callaghan
- Department of Anatomy & Physiology, University of Melbourne, Parkville, Victoria 3010, Australia
| | - Jie Huang
- Takeda Pharmaceuticals, San Diego, CA 92121, United States
| | - Jill Wykosky
- Takeda Pharmaceuticals, San Diego, CA 92121, United States
| | - John B. Furness
- Department of Anatomy & Physiology, University of Melbourne, Parkville, Victoria 3010, Australia
- Florey Institute of Neuroscience and Mental Health, Parkville, Victoria 3010, Australia
| | - Gene W. Yeo
- Department of Cellular and Molecular Medicine, University of California San Diego, La Jolla, CA 92093, United States
- Stem Cell Program, University of California San Diego, La Jolla, CA 92093, United States
- Institute for Genomic Medicine, University of California San Diego, La Jolla, CA 92093, United States
| |
Collapse
|
4
|
Lafrance AE, Chimalapati S, Garcia Rodriguez N, Kinch LN, Kaval KG, Orth K. Enzymatic Specificity of Conserved Rho GTPase Deamidases Promotes Invasion of Vibrio parahaemolyticus at the Expense of Infection. mBio 2022; 13:e0162922. [PMID: 35862776 PMCID: PMC9426531 DOI: 10.1128/mbio.01629-22] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2022] [Accepted: 06/17/2022] [Indexed: 11/23/2022] Open
Abstract
Vibrio parahaemolyticus is among the leading causes of bacterial seafood-borne acute gastroenteritis. Like many intracellular pathogens, V. parahaemolyticus invades host cells during infection by deamidating host small Rho GTPases. The Rho GTPase deamidating activity of VopC, a type 3 secretion system (T3SS) translocated effector, drives V. parahaemolyticus invasion. The intracellular pathogen uropathogenic Escherichia coli (UPEC) invades host cells by secreting a VopC homolog, the secreted toxin cytotoxic necrotizing factor 1 (CNF1). Because of the homology between VopC and CNF1, we hypothesized that topical application of CNF1 during V. parahaemolyticus infection could supplement VopC activity. Here, we demonstrate that CNF1 improves the efficiency of V. parahaemolyticus invasion, a bottleneck in V. parahaemolyticus infection, across a range of doses. CNF1 increases V. parahaemolyticus invasion independent of both VopC and the T3SS altogether but leaves a disproportionate fraction of intracellular bacteria unable to escape the endosome and complete their infection cycle. This phenomenon holds true in the presence or absence of VopC but is particularly pronounced in the absence of a T3SS. The native VopC, by contrast, promotes a far less efficient invasion but permits the majority of internalized bacteria to escape the endosome and complete their infection cycle. These studies highlight the significance of enzymatic specificity during infection, as virulence factors (VopC and CNF1 in this instance) with similarities in function (bacterial uptake), catalytic activity (deamidation), and substrates (Rho GTPases) are not sufficiently interchangeable for mediating a successful invasion for neighboring bacterial pathogens. IMPORTANCE Many species of intracellular bacterial pathogens target host small Rho GTPases to initiate invasion, including the human pathogens Vibrio parahaemolyticus and uropathogenic Escherichia coli (UPEC). The type three secretion system (T3SS) effector VopC of V. parahaemolyticus promotes invasion through the deamidation of Rac1 and CDC42 in the host, whereas the secreted toxin cytotoxic necrotizing factor 1 (CNF1) drives UPEC's internalization through the deamidation of Rac1, CDC42, and RhoA. Despite these similarities in the catalytic activity of CNF1 and VopC, we observed that the two enzymes were not interchangeable. Although CNF1 increased V. parahaemolyticus endosomal invasion, most intracellular V. parahaemolyticus aborted their infection cycle and remained trapped in endosomes. Our findings illuminate how the precise biochemical fine-tuning of T3SS effectors is essential for efficacious pathogenesis. Moreover, they pave the way for future investigations into the biochemical mechanisms underpinning V. parahaemolyticus endosomal escape and, more broadly, the regulation of successful pathogenesis.
Collapse
Affiliation(s)
- Alexander E. Lafrance
- Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Suneeta Chimalapati
- Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, Texas, USA
- Howard Hughes Medical Institute, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Nalleli Garcia Rodriguez
- Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Lisa N. Kinch
- Department of Biochemistry, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Karan Gautam Kaval
- Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, Texas, USA
- Howard Hughes Medical Institute, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Kim Orth
- Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, Texas, USA
- Howard Hughes Medical Institute, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| |
Collapse
|
5
|
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
|
6
|
Fabbri A, Bracci L. Immunomodulatory properties of CNF1 toxin from E. coli: implications for colorectal carcinogenesis. Am J Cancer Res 2022; 12:651-660. [PMID: 35261793 PMCID: PMC8899975] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Accepted: 01/27/2022] [Indexed: 06/14/2023] Open
Abstract
Colorectal cancer (CRC) is a leading cause of cancer death worldwide. The risk of developing CRC is influenced by both environmental and genetic factors. Recently, chronic inflammation and gut microbiota modifications have been associated with increased CRC risk. Escherichia coli belongs to the commensal intestinal flora and can become highly pathogenic following the acquisition of genes coding for virulence factors, such as the cytotoxic necrotizing factor type 1 (CNF1). Numerous reports highlight that, besides exerting direct effects on epithelial cells, CNF1 can also act on immune cells, modulating their responses and possibly contributing to disease development. In the present review, we summarized the key studies addressing the immunomodulatory functions of CNF1 and discussed the contribution that CNF1 can bring about to CRC through the creation of a pro-inflammatory microenvironment.
Collapse
Affiliation(s)
- Alessia Fabbri
- Department of Cardiovascular, Endocrine-Metabolic Diseases and Aging, Istituto Superiore di SanitàViale Regina Elena 299, Rome, Italy
| | - Laura Bracci
- Departement of Oncology and Molecular Medicine, Istituto Superiore di SanitàViale Regina Elena 299, Rome, Italy
| |
Collapse
|
7
|
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: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2021] [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
|
8
|
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: 5] [Impact Index Per Article: 1.7] [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
|
9
|
Kowarschik S, Schöllkopf J, Müller T, Tian S, Knerr J, Bakker H, Rein S, Dong M, Weber S, Grosse R, Schmidt G. Yersinia pseudotuberculosis cytotoxic necrotizing factor interacts with glycosaminoglycans. FASEB J 2021; 35:e21647. [PMID: 34165206 DOI: 10.1096/fj.202001630r] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2020] [Revised: 04/12/2021] [Accepted: 04/21/2021] [Indexed: 12/20/2022]
Abstract
The Cytotoxic Necrotizing Factor Y (CNFY) is produced by the gram-negative, enteric pathogen Yersinia pseudotuberculosis. The bacterial toxin belongs to a family of deamidases, which constitutively activate Rho GTPases, thereby balancing inflammatory processes. We identified heparan sulfate proteoglycans as essential host cell factors for intoxication with CNFY. Using flow cytometry, microscopy, knockout cell lines, pulsed electron-electron double resonance, and bio-layer interferometry, we studied the role of glucosaminoglycans in the intoxication process of CNFY. Especially the C-terminal part of CNFY, which encompasses the catalytic activity, binds with high affinity to heparan sulfates. CNFY binding with the N-terminal domain to a hypothetical protein receptor may support the interaction between the C-terminal domain and heparan sulfates, which seems sterically hindered in the full toxin. A second conformational change occurs by acidification of the endosome, probably allowing insertion of the hydrophobic regions of the toxin into the endosomal membrane. Our findings suggest that heparan sulfates play a major role for intoxication within the endosome, rather than being relevant for an interaction at the cell surface.
Collapse
Affiliation(s)
- Stefanie Kowarschik
- Institute for Experimental and Clinical Pharmacology and Toxicology, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Julian Schöllkopf
- Institute for Experimental and Clinical Pharmacology and Toxicology, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Thomas Müller
- Institute for Experimental and Clinical Pharmacology and Toxicology, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Songhai Tian
- Department of Urology, Boston Children's Hospital, Boston, MA, USA.,Department of Microbiology, Harvard Medical School, Boston, MA, USA
| | - Julian Knerr
- Institute for Experimental and Clinical Pharmacology and Toxicology, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Hans Bakker
- Hannover Medical School, Institute for Clinical Biochemistry, Hannover, Germany
| | - Stephan Rein
- Institute for Physical Chemistry, University of Freiburg, Freiburg, Germany
| | - Min Dong
- Department of Urology, Boston Children's Hospital, Boston, MA, USA.,Department of Microbiology, Harvard Medical School, Boston, MA, USA
| | - Stefan Weber
- Institute for Physical Chemistry, University of Freiburg, Freiburg, Germany
| | - Robert Grosse
- Institute for Experimental and Clinical Pharmacology and Toxicology, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Gudula Schmidt
- Institute for Experimental and Clinical Pharmacology and Toxicology, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| |
Collapse
|
10
|
Vannini E, Mori E, Tantillo E, Schmidt G, Caleo M, Costa M. CTX-CNF1 Recombinant Protein Selectively Targets Glioma Cells In Vivo. Toxins (Basel) 2021; 13:194. [PMID: 33800135 PMCID: PMC7998600 DOI: 10.3390/toxins13030194] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2021] [Revised: 02/24/2021] [Accepted: 02/25/2021] [Indexed: 01/11/2023] Open
Abstract
Current strategies for glioma treatment are only partly effective because of the poor selectivity for tumoral cells. Hence, the necessity to identify novel approaches is urgent. Recent studies highlighted the effectiveness of the bacterial protein cytotoxic necrotizing factor 1 (CNF1) in reducing tumoral mass, increasing survival of glioma-bearing mice and protecting peritumoral neural tissue from dysfunction. However, native CNF1 needs to be delivered into the brain, because of its incapacity to cross the blood-brain barrier (BBB) per se, thus hampering its clinical translation. To allow a non-invasive administration of CNF1, we here developed a chimeric protein (CTX-CNF1) conjugating CNF1 with chlorotoxin (CTX), a peptide already employed in clinics due to its ability of passing the BBB and selectively binding glioma cells. After systemic administration, we found that CTX-CNF1 is able to target glioma cells and significantly prolong survival of glioma-bearing mice. Our data point out the potentiality of CTX-CNF1 as a novel effective tool to treat gliomas.
Collapse
Affiliation(s)
- Eleonora Vannini
- Neuroscience Institute, National Research Council (CNR), via G. Moruzzi 1, 56124 Pisa, Italy; (E.T.); (M.C.); (M.C.)
- Fondazione Umberto Veronesi, 20122 Milan, Italy
| | | | - Elena Tantillo
- Neuroscience Institute, National Research Council (CNR), via G. Moruzzi 1, 56124 Pisa, Italy; (E.T.); (M.C.); (M.C.)
| | - Gudula Schmidt
- Medizinische Fakultät, Institut für Experimentelle und Klinische Pharmakologie und Toxikologie, University of Freiburg, 79085 Freiburg, Germany;
| | - Matteo Caleo
- Neuroscience Institute, National Research Council (CNR), via G. Moruzzi 1, 56124 Pisa, Italy; (E.T.); (M.C.); (M.C.)
- Department of Biomedical Sciences, University of Padua, 35122 Padua, Italy
| | - Mario Costa
- Neuroscience Institute, National Research Council (CNR), via G. Moruzzi 1, 56124 Pisa, Italy; (E.T.); (M.C.); (M.C.)
| |
Collapse
|
11
|
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: 3.3] [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
|
12
|
An Integrative Computational Approach for the Prediction of Human- Plasmodium Protein-Protein Interactions. BIOMED RESEARCH INTERNATIONAL 2021; 2020:2082540. [PMID: 33426052 PMCID: PMC7771252 DOI: 10.1155/2020/2082540] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/20/2020] [Revised: 11/08/2020] [Accepted: 12/04/2020] [Indexed: 12/27/2022]
Abstract
Host-pathogen molecular cross-talks are critical in determining the pathophysiology of a specific infection. Most of these cross-talks are mediated via protein-protein interactions between the host and the pathogen (HP-PPI). Thus, it is essential to know how some pathogens interact with their hosts to understand the mechanism of infections. Malaria is a life-threatening disease caused by an obligate intracellular parasite belonging to the Plasmodium genus, of which P. falciparum is the most prevalent. Several previous studies predicted human-plasmodium protein-protein interactions using computational methods have demonstrated their utility, accuracy, and efficiency to identify the interacting partners and therefore complementing experimental efforts to characterize host-pathogen interaction networks. To predict potential putative HP-PPIs, we use an integrative computational approach based on the combination of multiple OMICS-based methods including human red blood cells (RBC) and Plasmodium falciparum 3D7 strain expressed proteins, domain-domain based PPI, similarity of gene ontology terms, structure similarity method homology identification, and machine learning prediction. Our results reported a set of 716 protein interactions involving 302 human proteins and 130 Plasmodium proteins. This work provides a list of potential human-Plasmodium interacting proteins. These findings will contribute to better understand the mechanisms underlying the molecular determinism of malaria disease and potentially to identify candidate pharmacological targets.
Collapse
|
13
|
Zhang Z, Aung KM, Uhlin BE, Wai SN. Reversible senescence of human colon cancer cells after blockage of mitosis/cytokinesis caused by the CNF1 cyclomodulin from Escherichia coli. Sci Rep 2018; 8:17780. [PMID: 30542142 PMCID: PMC6290797 DOI: 10.1038/s41598-018-36036-5] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2018] [Accepted: 10/28/2018] [Indexed: 12/11/2022] Open
Abstract
Cytotoxic necrotizing factor 1 (CNF1), a protein toxin produced by extraintestinal pathogenic Escherichia coli, activates the Rho-family small GTPases in eukaryotic cell, thereby perturbing multiple cellular functions. Increasing epidemiological evidence suggests a link between CNF1 and human inflammatory bowel disease and colorectal cancer. At the cellular level, CNF1 has been hypothesized to reprogram cell fate towards survival due to the role in perturbing cell cycle and apoptosis. However, it remains undetermined how cells survive from CNF1 intoxication. In this work, we show that CNF1 treatment blocks mitosis/cytokinesis, elicits endoreplication and polyploidisation in cultured human colon cancer cells, and drives them into reversible senescence, which provides a survival route for cells via depolyploidisation. Senescence in CNF1-treated cells is demonstrated with upregulation of several senescence markers including senescence-associated β-galactosidase activity, p53, p21 and p16, and concomitant inhibition of the retinoblastoma protein phosphorylation. Importantly, progeny derived from CNF1 treatment exhibit genomic instability exemplified by increased aneuploidy and become more resistant to CNF1, but not to 5-fluorouracil and oxaliplatin, the two agents commonly used in chemotherapeutic treatment for colorectal cancer. These observations display survival features of the cell after CNF1 treatment that may have implications for the potential role of CNF1 in carcinogenesis.
Collapse
Affiliation(s)
- Zhen Zhang
- Department of Molecular Biology and The Laboratory for Molecular Infection Medicine Sweden (MIMS), Umeå University, SE-90187, Umeå, Sweden. .,Department of Food Hygiene and Environmental Health, Faculty of Veterinary Medicine, University of Helsinki, FIN-00014, Helsinki, Finland.
| | - Kyaw Min Aung
- Department of Molecular Biology and The Laboratory for Molecular Infection Medicine Sweden (MIMS), Umeå University, SE-90187, Umeå, Sweden
| | - Bernt Eric Uhlin
- Department of Molecular Biology and The Laboratory for Molecular Infection Medicine Sweden (MIMS), Umeå University, SE-90187, Umeå, Sweden
| | - Sun Nyunt Wai
- Department of Molecular Biology and The Laboratory for Molecular Infection Medicine Sweden (MIMS), Umeå University, SE-90187, Umeå, Sweden.
| |
Collapse
|
14
|
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: 2.0] [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
|
15
|
Tantillo E, Colistra A, Vannini E, Cerri C, Pancrazi L, Baroncelli L, Costa M, Caleo M. Bacterial Toxins and Targeted Brain Therapy: New Insights from Cytotoxic Necrotizing Factor 1 (CNF1). Int J Mol Sci 2018; 19:ijms19061632. [PMID: 29857515 PMCID: PMC6032336 DOI: 10.3390/ijms19061632] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2018] [Revised: 05/07/2018] [Accepted: 05/12/2018] [Indexed: 01/17/2023] Open
Abstract
Pathogenic bacteria produce toxins to promote host invasion and, therefore, their survival. The extreme potency and specificity of these toxins confer to this category of proteins an exceptionally strong potential for therapeutic exploitation. In this review, we deal with cytotoxic necrotizing factor (CNF1), a cytotoxin produced by Escherichia coli affecting fundamental cellular processes, including cytoskeletal dynamics, cell cycle progression, transcriptional regulation, cell survival and migration. First, we provide an overview of the mechanisms of action of CNF1 in target cells. Next, we focus on the potential use of CNF1 as a pharmacological treatment in central nervous system’s diseases. CNF1 appears to impact neuronal morphology, physiology, and plasticity and displays an antineoplastic activity on brain tumors. The ability to preserve neural functionality and, at the same time, to trigger senescence and death of proliferating glioma cells, makes CNF1 an encouraging new strategy for the treatment of brain tumors.
Collapse
Affiliation(s)
- Elena Tantillo
- CNR Neuroscience Institute, via G. Moruzzi 1, 56124 Pisa, Italy.
- Fondazione Pisana per la Scienza Onlus (FPS), via Ferruccio Giovannini 13, San Giuliano Terme, 56017 Pisa, Italy.
| | - Antonella Colistra
- CNR Neuroscience Institute, via G. Moruzzi 1, 56124 Pisa, Italy.
- Departement of Biology, University of Pisa, via Luca Ghini 13, 56126 Pisa, Italy.
| | - Eleonora Vannini
- CNR Neuroscience Institute, via G. Moruzzi 1, 56124 Pisa, Italy.
| | - Chiara Cerri
- CNR Neuroscience Institute, via G. Moruzzi 1, 56124 Pisa, Italy.
- Fondazione Umberto Veronesi, Piazza Velasca 5, 20122 Milano, Italy.
| | - Laura Pancrazi
- CNR Neuroscience Institute, via G. Moruzzi 1, 56124 Pisa, Italy.
| | - Laura Baroncelli
- CNR Neuroscience Institute, via G. Moruzzi 1, 56124 Pisa, Italy.
| | - Mario Costa
- CNR Neuroscience Institute, via G. Moruzzi 1, 56124 Pisa, Italy.
| | - Matteo Caleo
- CNR Neuroscience Institute, via G. Moruzzi 1, 56124 Pisa, Italy.
| |
Collapse
|
16
|
Gall-Mas L, Fabbri A, Namini MRJ, Givskov M, Fiorentini C, Krejsgaard T. The Bacterial Toxin CNF1 Induces Activation and Maturation of Human Monocyte-Derived Dendritic Cells. Int J Mol Sci 2018; 19:ijms19051408. [PMID: 29738516 PMCID: PMC5983691 DOI: 10.3390/ijms19051408] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2018] [Revised: 05/04/2018] [Accepted: 05/04/2018] [Indexed: 11/24/2022] Open
Abstract
Cytotoxic necrotizing factor 1 (CNF1) is a bacterial protein toxin primarily expressed by pathogenic Escherichia coli strains, causing extraintestinal infections. The toxin is believed to enhance the invasiveness of E. coli by modulating the activity of Rho GTPases in host cells, but it has interestingly also been shown to promote inflammation, stimulate host immunity and function as a potent immunoadjuvant. The mechanisms underlying the immunostimulatory properties of CNF1 are, however, poorly characterized, and little is known about the direct effects of the toxin on immune cells. Here, we show that CNF1 induces expression of maturation markers on human immature monocyte-derived dendritic cells (moDCs) without compromising cell viability. Consistent with the phenotypic maturation, CNF1 further triggered secretion of proinflammatory cytokines and increased the capacity of moDCs to stimulate proliferation of allogenic naïve CD4+ T cells. A catalytically inactive form of the toxin did not induce moDC maturation, indicating that the enzymatic activity of CNF1 triggers immature moDCs to undergo phenotypic and functional maturation. As the maturation of dendritic cells plays a central role in initiating inflammation and activating the adaptive immune response, the present findings shed new light on the immunostimulatory properties of CNF1 and may explain why the toxin functions as an immunoadjuvant.
Collapse
Affiliation(s)
- Laura Gall-Mas
- Department of Immunology and Microbiology, University of Copenhagen, Nørre Alle 14, 2200 Copenhagen, Denmark.
| | - Alessia Fabbri
- Italian Center for Global Health, Istituto Superiore di Sanitá; Viale Regina Elena 299, 00161 Rome, Italy.
| | - Martin R J Namini
- Department of Immunology and Microbiology, University of Copenhagen, Nørre Alle 14, 2200 Copenhagen, Denmark.
| | - Michael Givskov
- Costerton Biofilm Center, Department of Immunology and Microbiology, University of Copenhagen, Nørre Alle 14, 2200 Copenhagen, Denmark.
| | - Carla Fiorentini
- Italian Center for Global Health, Istituto Superiore di Sanitá; Viale Regina Elena 299, 00161 Rome, Italy.
| | - Thorbjørn Krejsgaard
- Department of Immunology and Microbiology, University of Copenhagen, Nørre Alle 14, 2200 Copenhagen, Denmark.
| |
Collapse
|
17
|
Analysing the Structural Effect of Point Mutations of Cytotoxic Necrotizing Factor 1 (CNF1) on Lu/BCAM Adhesion Glycoprotein Association. Toxins (Basel) 2018. [PMID: 29533999 PMCID: PMC5869410 DOI: 10.3390/toxins10030122] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Cytotoxic Necrotizing Factor 1 (CNF1) was identified in 1983 as a protein toxin produced by certain pathogenic strains of Escherichia coli. Since then, numerous studies have investigated its particularities. For instance, it is associated with the single chain AB-toxin family, and can be divided into different functional and structural domains, e.g., catalytic and transmembrane domain and interaction sites. A few years ago, the identification of the Lutheran (Lu) adhesion glycoprotein/basal cell adhesion molecule (BCAM) as a cellular receptor for CNF1 provided new insights into the adhesion process of CNF1. Very recently, the Ig-like domain 2 of Lu/BCAM was confirmed as the main interaction site using protein-protein interaction and competition studies with various different mutants. Here, I present in silico approaches that precisely explain the impact of these mutations, leading to a better explanation of these experimental studies. These results can be used in the development of future antitoxin strategies.
Collapse
|
18
|
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: 1.5] [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
|
19
|
Reppin F, Cochet S, El Nemer W, Fritz G, Schmidt G. High Affinity Binding of Escherichia coli Cytotoxic Necrotizing Factor 1 (CNF1) to Lu/BCAM Adhesion Glycoprotein. Toxins (Basel) 2017; 10:toxins10010003. [PMID: 29267242 PMCID: PMC5793090 DOI: 10.3390/toxins10010003] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2017] [Revised: 12/19/2017] [Accepted: 12/19/2017] [Indexed: 01/07/2023] Open
Abstract
The protein toxin Cytotoxic Necrotizing Factor 1 (CNF1) is a major virulence factor of pathogenic Escherichia coli strains. It belongs to a family of single chain AB-toxins, which enter mammalian cells by receptor-mediated endocytosis. Recently, we identified the Lutheran (Lu) adhesion glycoprotein/basal cell adhesion molecule (BCAM) as a cellular receptor for CNF1. Here, we identified the Ig-like domain 2 of Lu/BCAM as main interaction site of the toxin by direct protein-protein interaction and competition studies. Using surface plasmon resonance, we showed a high affinity CNF-Lu/BCAM interaction with a KD of 2.8 nM. Furthermore, we performed small-angle X-ray scattering to define the molecular envelope of the Lu/BCAM-CNF1 complex, suggesting a 6:1 ratio of Lu/BCAM to CNF1 in the receptor-toxin complex. This study leads to a deeper understanding of the interaction between CNF1 and Lu/BCAM, and presents novel opportunities for the development of future anti-toxin strategies.
Collapse
Affiliation(s)
- Franziska Reppin
- Institute for Experimental and Clinical Pharmacology and Toxicology, Faculty of Medicine, Albert-Ludwigs-University of Freiburg, Albert-Street 25, 79104 Freiburg, Germany.
- Biological Faculty, Albert-Ludwigs-University of Freiburg, Albert-Street 25, 79104 Freiburg, Germany.
| | - Sylvie Cochet
- Universite Sorbonne Paris Cite, Universite Paris Diderot, Inserm, INTS, Unite Biologie Integree du Globule Rouge, Laboratoire d'Excellence GR-Ex, 75013 Paris, France.
| | - Wassim El Nemer
- Universite Sorbonne Paris Cite, Universite Paris Diderot, Inserm, INTS, Unite Biologie Integree du Globule Rouge, Laboratoire d'Excellence GR-Ex, 75013 Paris, France.
| | - Günter Fritz
- Department of Neuropathology, Albert-Ludwigs-University of Freiburg, Breisacher Strasse 64, 79106 Freiburg, Germany.
| | - Gudula Schmidt
- Institute for Experimental and Clinical Pharmacology and Toxicology, Faculty of Medicine, Albert-Ludwigs-University of Freiburg, Albert-Street 25, 79104 Freiburg, Germany.
| |
Collapse
|
20
|
Colarusso A, Caterino M, Fabbri A, Fiorentini C, Vergara A, Sica F, Parrilli E, Tutino ML. High yield purification and first structural characterization of the full-length bacterial toxin CNF1. Biotechnol Prog 2017; 34:150-159. [PMID: 29063721 DOI: 10.1002/btpr.2574] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2017] [Revised: 09/12/2017] [Indexed: 11/10/2022]
Abstract
The Cytotoxic Necrotizing Factor 1 (CNF1) is a bacterial toxin secreted by certain Escherichia coli strains causing severe pathologies, making it a protein of pivotal interest in toxicology. In parallel, the CNF1 capability to influence important neuronal processes, like neuronal arborization, astrocytic support, and efficient ATP production, has been efficiently used in the treatment of neurological diseases, making it a promising candidate for therapy. Nonetheless, there are still some unsolved issues about the CNF1 mechanism of action and structuration probably caused by the difficulty to achieve sufficient amounts of the full-length protein for further studies. Here, we propose an efficient strategy for the production and purification of this toxin as a his-tagged recombinant protein from E. coli extracts (CNF1-H8). CNF1-H8 was expressed at the low temperature of 15°C to diminish its characteristic degradation. Then, its purification was achieved using an immobilized metal affinity chromatography (IMAC) and a size exclusion chromatography so as to collect up to 8 mg of protein per liter of culture in a highly pure form. Routine dynamic light scattering (DLS) experiments showed that the recombinant protein preparations were homogeneous and preserved this state for a long time. Furthermore, CNF1-H8 functionality was confirmed by testing its activity on purified RhoA and on HEp-2 cultured cells. Finally, a first structural characterization of the full-length toxin in terms of secondary structure and thermal stability was performed by circular dichroism (CD). These studies demonstrate that our system can be used to produce high quantities of pure recombinant protein for a detailed structural analysis. © 2017 American Institute of Chemical Engineers Biotechnol. Prog., 34:150-159, 2018.
Collapse
Affiliation(s)
- Andrea Colarusso
- Department of Chemical Sciences, University of Naples Federico II, Complesso Universitario Monte Sant'Angelo, Via Cinthia, Napoli, 80126, Italy
| | - Marco Caterino
- Department of Chemical Sciences, University of Naples Federico II, Complesso Universitario Monte Sant'Angelo, Via Cinthia, Napoli, 80126, Italy
| | - Alessia Fabbri
- Italian Center for Global Health, Istituto Superiore di Sanità, Viale Regina Elena, 299, Roma, 00161, Italy
| | - Carla Fiorentini
- Italian Center for Global Health, Istituto Superiore di Sanità, Viale Regina Elena, 299, Roma, 00161, Italy
| | - Alessandro Vergara
- Department of Chemical Sciences, University of Naples Federico II, Complesso Universitario Monte Sant'Angelo, Via Cinthia, Napoli, 80126, Italy.,CEINGE Biotecnologie Avanzate scarl, Via G. Salvatore, Napoli, 80100, Italy.,Institute of Biostructures and Biomaging, CNR, Napoli, Italia Via Mezzocannone 16, Napoli, 80134, Italy
| | - Filomena Sica
- Department of Chemical Sciences, University of Naples Federico II, Complesso Universitario Monte Sant'Angelo, Via Cinthia, Napoli, 80126, Italy.,Institute of Biostructures and Biomaging, CNR, Napoli, Italia Via Mezzocannone 16, Napoli, 80134, Italy
| | - Ermenegilda Parrilli
- Department of Chemical Sciences, University of Naples Federico II, Complesso Universitario Monte Sant'Angelo, Via Cinthia, Napoli, 80126, Italy
| | - Maria Luisa Tutino
- Department of Chemical Sciences, University of Naples Federico II, Complesso Universitario Monte Sant'Angelo, Via Cinthia, Napoli, 80126, Italy
| |
Collapse
|
21
|
Microbial carcinogenic toxins and dietary anti-cancer protectants. Cell Mol Life Sci 2017; 74:2627-2643. [PMID: 28238104 PMCID: PMC5487888 DOI: 10.1007/s00018-017-2487-z] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2016] [Revised: 02/03/2017] [Accepted: 02/06/2017] [Indexed: 12/16/2022]
Abstract
Several toxins are known which account for the ability of some bacteria to initiate or promote carcinogenesis. These ideas are summarised and evidence is discussed for more specific mechanisms involving chymotrypsin and the bacterial chymotryptic enzyme subtilisin. Subtilisin and Bacillus subtilis are present in the gut and environment and both are used commercially in agriculture, livestock rearing and meat processing. The enzymes deplete cells of tumour suppressors such as deleted in colorectal cancer (DCC) and neogenin, so their potential presence in the food chain might represent an important link between diet and cancer. Over-eating increases secretion of chymotrypsin which is absorbed from the gut and could contribute to several forms of cancer linked to obesity. Inhibition of these serine proteases by Bowman–Birk inhibitors in fruit and vegetables could account for some of the protective effects of a plant-rich diet. These interactions represent previously unknown non-genetic mechanisms for the modification of tumour suppressor proteins and provide a plausible explanation contributing to both the pro-oncogenic effects of meat products and the protective activity of a plant-rich diet. The data suggest that changes to farming husbandry and food processing methods to remove these sources of extrinsic proteases might significantly reduce the incidence of several cancers.
Collapse
|
22
|
Guadagni V, Cerri C, Piano I, Novelli E, Gargini C, Fiorentini C, Caleo M, Strettoi E. The bacterial toxin CNF1 as a tool to induce retinal degeneration reminiscent of retinitis pigmentosa. Sci Rep 2016; 6:35919. [PMID: 27775019 PMCID: PMC5075935 DOI: 10.1038/srep35919] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2016] [Accepted: 10/04/2016] [Indexed: 12/02/2022] Open
Abstract
Retinitis pigmentosa (RP) comprises a group of inherited pathologies characterized by progressive photoreceptor degeneration. In rodent models of RP, expression of defective genes and retinal degeneration usually manifest during the first weeks of postnatal life, making it difficult to distinguish consequences of primary genetic defects from abnormalities in retinal development. Moreover, mouse eyes are small and not always adequate to test pharmacological and surgical treatments. An inducible paradigm of retinal degeneration potentially extensible to large animals is therefore desirable. Starting from the serendipitous observation that intraocular injections of a Rho GTPase activator, the bacterial toxin Cytotoxic Necrotizing Factor 1 (CNF1), lead to retinal degeneration, we implemented an inducible model recapitulating most of the key features of Retinitis Pigmentosa. The model also unmasks an intrinsic vulnerability of photoreceptors to the mechanism of CNF1 action, indicating still unexplored molecular pathways potentially leading to the death of these cells in inherited forms of retinal degeneration.
Collapse
Affiliation(s)
- Viviana Guadagni
- Neuroscience Institute, Italian National Research Council (CNR), Pisa, 56124, Italy
| | - Chiara Cerri
- Neuroscience Institute, Italian National Research Council (CNR), Pisa, 56124, Italy.,Accademia dei Lincei, Rome, 00165, Italy
| | - Ilaria Piano
- Department of Pharmacy, University of Pisa, Pisa, 56126, Italy
| | - Elena Novelli
- Neuroscience Institute, Italian National Research Council (CNR), Pisa, 56124, Italy
| | - Claudia Gargini
- Department of Pharmacy, University of Pisa, Pisa, 56126, Italy
| | | | - Matteo Caleo
- Neuroscience Institute, Italian National Research Council (CNR), Pisa, 56124, Italy
| | - Enrica Strettoi
- Neuroscience Institute, Italian National Research Council (CNR), Pisa, 56124, Italy
| |
Collapse
|
23
|
Vannini E, Olimpico F, Middei S, Ammassari-Teule M, de Graaf EL, McDonnell L, Schmidt G, Fabbri A, Fiorentini C, Baroncelli L, Costa M, Caleo M. Electrophysiology of glioma: a Rho GTPase-activating protein reduces tumor growth and spares neuron structure and function. Neuro Oncol 2016; 18:1634-1643. [PMID: 27298309 DOI: 10.1093/neuonc/now114] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2015] [Accepted: 04/22/2016] [Indexed: 01/02/2023] Open
Abstract
BACKGROUND Glioblastomas are the most aggressive type of brain tumor. A successful treatment should aim at halting tumor growth and protecting neuronal cells to prevent functional deficits and cognitive deterioration. Here, we exploited a Rho GTPase-activating bacterial protein toxin, cytotoxic necrotizing factor 1 (CNF1), to interfere with glioma cell growth in vitro and vivo. We also investigated whether this toxin spares neuron structure and function in peritumoral areas. METHODS We performed a microarray transcriptomic and in-depth proteomic analysis to characterize the molecular changes triggered by CNF1 in glioma cells. We also examined tumor cell senescence and growth in vehicle- and CNF1-treated glioma-bearing mice. Electrophysiological and morphological techniques were used to investigate neuronal alterations in peritumoral cortical areas. RESULTS Administration of CNF1 triggered molecular and morphological hallmarks of senescence in mouse and human glioma cells in vitro. CNF1 treatment in vivo induced glioma cell senescence and potently reduced tumor volumes. In peritumoral areas of glioma-bearing mice, neurons showed a shrunken dendritic arbor and severe functional alterations such as increased spontaneous activity and reduced visual responsiveness. CNF1 treatment enhanced dendritic length and improved several physiological properties of pyramidal neurons, demonstrating functional preservation of the cortical network. CONCLUSIONS Our findings demonstrate that CNF1 reduces glioma volume while at the same time maintaining the physiological and structural properties of peritumoral neurons. These data indicate a promising strategy for the development of more effective antiglioma therapies.
Collapse
Affiliation(s)
- Eleonora Vannini
- CNR Neuroscience Institute, Pisa, Italy (E.V., F.O., L.B., M.C., Mat.C.); CNR Cellular Biology and Neurobiology Institute, Rome, Italy (S.M., M.A.-T.); Fondazione Pisana per la Scienza, Mass Spectrometry and Proteomics, Pisa, Italy (E.L.d.G., L.M.); Institut für Experimentelle und Klinische Pharmakologie und Toxikologie, Freiburg, Germany (G.S.); Istituto Superiore di Sanità, Rome, Italy (A.F., C.F.); Scuola Normale Superiore, Pisa, Italy (M.C., Mat.C.)
| | - Francesco Olimpico
- CNR Neuroscience Institute, Pisa, Italy (E.V., F.O., L.B., M.C., Mat.C.); CNR Cellular Biology and Neurobiology Institute, Rome, Italy (S.M., M.A.-T.); Fondazione Pisana per la Scienza, Mass Spectrometry and Proteomics, Pisa, Italy (E.L.d.G., L.M.); Institut für Experimentelle und Klinische Pharmakologie und Toxikologie, Freiburg, Germany (G.S.); Istituto Superiore di Sanità, Rome, Italy (A.F., C.F.); Scuola Normale Superiore, Pisa, Italy (M.C., Mat.C.)
| | - Silvia Middei
- CNR Neuroscience Institute, Pisa, Italy (E.V., F.O., L.B., M.C., Mat.C.); CNR Cellular Biology and Neurobiology Institute, Rome, Italy (S.M., M.A.-T.); Fondazione Pisana per la Scienza, Mass Spectrometry and Proteomics, Pisa, Italy (E.L.d.G., L.M.); Institut für Experimentelle und Klinische Pharmakologie und Toxikologie, Freiburg, Germany (G.S.); Istituto Superiore di Sanità, Rome, Italy (A.F., C.F.); Scuola Normale Superiore, Pisa, Italy (M.C., Mat.C.)
| | - Martine Ammassari-Teule
- CNR Neuroscience Institute, Pisa, Italy (E.V., F.O., L.B., M.C., Mat.C.); CNR Cellular Biology and Neurobiology Institute, Rome, Italy (S.M., M.A.-T.); Fondazione Pisana per la Scienza, Mass Spectrometry and Proteomics, Pisa, Italy (E.L.d.G., L.M.); Institut für Experimentelle und Klinische Pharmakologie und Toxikologie, Freiburg, Germany (G.S.); Istituto Superiore di Sanità, Rome, Italy (A.F., C.F.); Scuola Normale Superiore, Pisa, Italy (M.C., Mat.C.)
| | - Erik L de Graaf
- CNR Neuroscience Institute, Pisa, Italy (E.V., F.O., L.B., M.C., Mat.C.); CNR Cellular Biology and Neurobiology Institute, Rome, Italy (S.M., M.A.-T.); Fondazione Pisana per la Scienza, Mass Spectrometry and Proteomics, Pisa, Italy (E.L.d.G., L.M.); Institut für Experimentelle und Klinische Pharmakologie und Toxikologie, Freiburg, Germany (G.S.); Istituto Superiore di Sanità, Rome, Italy (A.F., C.F.); Scuola Normale Superiore, Pisa, Italy (M.C., Mat.C.)
| | - Liam McDonnell
- CNR Neuroscience Institute, Pisa, Italy (E.V., F.O., L.B., M.C., Mat.C.); CNR Cellular Biology and Neurobiology Institute, Rome, Italy (S.M., M.A.-T.); Fondazione Pisana per la Scienza, Mass Spectrometry and Proteomics, Pisa, Italy (E.L.d.G., L.M.); Institut für Experimentelle und Klinische Pharmakologie und Toxikologie, Freiburg, Germany (G.S.); Istituto Superiore di Sanità, Rome, Italy (A.F., C.F.); Scuola Normale Superiore, Pisa, Italy (M.C., Mat.C.)
| | - Gudula Schmidt
- CNR Neuroscience Institute, Pisa, Italy (E.V., F.O., L.B., M.C., Mat.C.); CNR Cellular Biology and Neurobiology Institute, Rome, Italy (S.M., M.A.-T.); Fondazione Pisana per la Scienza, Mass Spectrometry and Proteomics, Pisa, Italy (E.L.d.G., L.M.); Institut für Experimentelle und Klinische Pharmakologie und Toxikologie, Freiburg, Germany (G.S.); Istituto Superiore di Sanità, Rome, Italy (A.F., C.F.); Scuola Normale Superiore, Pisa, Italy (M.C., Mat.C.)
| | - Alessia Fabbri
- CNR Neuroscience Institute, Pisa, Italy (E.V., F.O., L.B., M.C., Mat.C.); CNR Cellular Biology and Neurobiology Institute, Rome, Italy (S.M., M.A.-T.); Fondazione Pisana per la Scienza, Mass Spectrometry and Proteomics, Pisa, Italy (E.L.d.G., L.M.); Institut für Experimentelle und Klinische Pharmakologie und Toxikologie, Freiburg, Germany (G.S.); Istituto Superiore di Sanità, Rome, Italy (A.F., C.F.); Scuola Normale Superiore, Pisa, Italy (M.C., Mat.C.)
| | - Carla Fiorentini
- CNR Neuroscience Institute, Pisa, Italy (E.V., F.O., L.B., M.C., Mat.C.); CNR Cellular Biology and Neurobiology Institute, Rome, Italy (S.M., M.A.-T.); Fondazione Pisana per la Scienza, Mass Spectrometry and Proteomics, Pisa, Italy (E.L.d.G., L.M.); Institut für Experimentelle und Klinische Pharmakologie und Toxikologie, Freiburg, Germany (G.S.); Istituto Superiore di Sanità, Rome, Italy (A.F., C.F.); Scuola Normale Superiore, Pisa, Italy (M.C., Mat.C.)
| | - Laura Baroncelli
- CNR Neuroscience Institute, Pisa, Italy (E.V., F.O., L.B., M.C., Mat.C.); CNR Cellular Biology and Neurobiology Institute, Rome, Italy (S.M., M.A.-T.); Fondazione Pisana per la Scienza, Mass Spectrometry and Proteomics, Pisa, Italy (E.L.d.G., L.M.); Institut für Experimentelle und Klinische Pharmakologie und Toxikologie, Freiburg, Germany (G.S.); Istituto Superiore di Sanità, Rome, Italy (A.F., C.F.); Scuola Normale Superiore, Pisa, Italy (M.C., Mat.C.)
| | - Mario Costa
- CNR Neuroscience Institute, Pisa, Italy (E.V., F.O., L.B., M.C., Mat.C.); CNR Cellular Biology and Neurobiology Institute, Rome, Italy (S.M., M.A.-T.); Fondazione Pisana per la Scienza, Mass Spectrometry and Proteomics, Pisa, Italy (E.L.d.G., L.M.); Institut für Experimentelle und Klinische Pharmakologie und Toxikologie, Freiburg, Germany (G.S.); Istituto Superiore di Sanità, Rome, Italy (A.F., C.F.); Scuola Normale Superiore, Pisa, Italy (M.C., Mat.C.)
| | - Matteo Caleo
- CNR Neuroscience Institute, Pisa, Italy (E.V., F.O., L.B., M.C., Mat.C.); CNR Cellular Biology and Neurobiology Institute, Rome, Italy (S.M., M.A.-T.); Fondazione Pisana per la Scienza, Mass Spectrometry and Proteomics, Pisa, Italy (E.L.d.G., L.M.); Institut für Experimentelle und Klinische Pharmakologie und Toxikologie, Freiburg, Germany (G.S.); Istituto Superiore di Sanità, Rome, Italy (A.F., C.F.); Scuola Normale Superiore, Pisa, Italy (M.C., Mat.C.)
| |
Collapse
|
24
|
Urinary tract infections: epidemiology, mechanisms of infection and treatment options. Nat Rev Microbiol 2015; 13:269-84. [PMID: 25853778 DOI: 10.1038/nrmicro3432] [Citation(s) in RCA: 1980] [Impact Index Per Article: 220.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Urinary tract infections (UTIs) are a severe public health problem and are caused by a range of pathogens, but most commonly by Escherichia coli, Klebsiella pneumoniae, Proteus mirabilis, Enterococcus faecalis and Staphylococcus saprophyticus. High recurrence rates and increasing antimicrobial resistance among uropathogens threaten to greatly increase the economic burden of these infections. In this Review, we discuss how basic science studies are elucidating the molecular details of the crosstalk that occurs at the host-pathogen interface, as well as the consequences of these interactions for the pathophysiology of UTIs. We also describe current efforts to translate this knowledge into new clinical treatments for UTIs.
Collapse
|
25
|
Schmidt G, Papatheodorou P, Aktories K. Novel receptors for bacterial protein toxins. Curr Opin Microbiol 2015; 23:55-61. [DOI: 10.1016/j.mib.2014.11.003] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2014] [Revised: 11/05/2014] [Accepted: 11/06/2014] [Indexed: 01/05/2023]
|
26
|
Travaglione S, Loizzo S, Rizza T, Del Brocco A, Ballan G, Guidotti M, Vona R, Di Nottia M, Torraco A, Carrozzo R, Fiorentini C, Fabbri A. Enhancement of mitochondrial ATP production by the Escherichia coli cytotoxic necrotizing factor 1. FEBS J 2014; 281:3473-88. [PMID: 24925215 DOI: 10.1111/febs.12874] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2014] [Revised: 06/06/2014] [Accepted: 06/09/2014] [Indexed: 01/12/2023]
Abstract
Mitochondria are dynamic organelles that constantly change shape and structure in response to different stimuli and metabolic demands of the cell. The Escherichia coli protein toxin cytotoxic necrotizing factor 1 (CNF1) has recently been reported to influence mitochondrial activity in a mouse model of Rett syndrome and to increase ATP content in the brain tissue of an Alzheimer's disease mouse model. In the present work, the ability of CNF1 to influence mitochondrial activity was investigated in IEC-6 normal intestinal crypt cells. In these cells, the toxin was able to induce an increase in cellular ATP content, probably due to an increment of the mitochondrial electron transport chain. In addition, the CNF1-induced Rho GTPase activity also caused changes in the mitochondrial architecture that mainly consisted in the formation of a complex network of elongated mitochondria. The involvement of the cAMP-dependent protein kinase A signaling pathway was postulated. Our results demonstrate that CNF1 positively affects mitochondria by bursting their energetic function and modifying their morphology.
Collapse
Affiliation(s)
- Sara Travaglione
- Department of Therapeutic Research and Medicines Evaluation, Istituto Superiore di Sanità, Rome, Italy
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|