1
|
Chang Y, Zheng W, Duan M, Su T, Wang Z, Wu S, Duan N. Construction of Aptamer-Functionalized DNA Hydrogels for Effective Inhibition of Shiga Toxin II Toxicity. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2024; 72:23533-23543. [PMID: 39388632 DOI: 10.1021/acs.jafc.4c07612] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/12/2024]
Abstract
Bacterial infections have been seriously endangering public health and life, making it imperative to explore novel anti-infection strategies for their control. Herein, we constructed a DNA hydrogel encoded with aptamers (Apt-hydrogel) to inhibit Shiga toxin II (Stx2) toxicity, thereby alleviating Escherichia coli (EHEC) infection. The Apt-hydrogel was formed by two Y-shaped DNA scaffolds through rational design, where one end of Y was encoded with an aptamer sequence targeting the B subunit of Shiga toxin II (Stx2B). The Apt-hydrogel not only retained the high affinity of the aptamer but also provided protection for the aptamer, endowing it with better stability and biocompatibility. The results from in vitro and in vivo demonstrated good mediation effects of the Apt-hydrogel on Stx2 toxicity and confirmed its excellent inhibition activity. We hypothesized that the mechanism could be attributed to the high affinity of Apt-hydrogel for Stx2B, which effectively occupies the active site of Stx2B and its receptor Gb3. This interaction enhanced steric hindrance, thereby mediating their interaction and preventing Stx2 from entering the cell to exert toxicity. We anticipate that the novel Apt-hydrogel will expand the usage of aptamers and provide a new dimension for the Apt-hydrogel as a promising blocking assistant to inhibit Shiga toxin infections via a strong steric hindrance effect.
Collapse
Affiliation(s)
- Yuting Chang
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi 214122, China
- School of Food Science and Technology, Jiangnan University, Wuxi 214122, China
| | - Wenxiu Zheng
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi 214122, China
- School of Food Science and Technology, Jiangnan University, Wuxi 214122, China
| | - Mengxia Duan
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi 214122, China
- School of Food Science and Technology, Jiangnan University, Wuxi 214122, China
| | - Tingting Su
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi 214122, China
- School of Food Science and Technology, Jiangnan University, Wuxi 214122, China
| | - Zhouping Wang
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi 214122, China
- School of Food Science and Technology, Jiangnan University, Wuxi 214122, China
- International Joint Laboratory on Food Safety, Jiangnan University, Wuxi 214122, China
| | - Shijia Wu
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi 214122, China
- School of Food Science and Technology, Jiangnan University, Wuxi 214122, China
- International Joint Laboratory on Food Safety, Jiangnan University, Wuxi 214122, China
| | - Nuo Duan
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi 214122, China
- School of Food Science and Technology, Jiangnan University, Wuxi 214122, China
- International Joint Laboratory on Food Safety, Jiangnan University, Wuxi 214122, China
| |
Collapse
|
2
|
Iglesias R, Citores L, Gay CC, Ferreras JM. Antifungal Activity of Ribosome-Inactivating Proteins. Toxins (Basel) 2024; 16:192. [PMID: 38668617 PMCID: PMC11054410 DOI: 10.3390/toxins16040192] [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: 03/14/2024] [Revised: 04/04/2024] [Accepted: 04/12/2024] [Indexed: 04/29/2024] Open
Abstract
The control of crop diseases caused by fungi remains a major problem and there is a need to find effective fungicides that are environmentally friendly. Plants are an excellent source for this purpose because they have developed defense mechanisms to cope with fungal infections. Among the plant proteins that play a role in defense are ribosome-inactivating proteins (RIPs), enzymes obtained mainly from angiosperms that, in addition to inactivating ribosomes, have been studied as antiviral, fungicidal, and insecticidal proteins. In this review, we summarize and discuss the potential use of RIPs (and other proteins with similar activity) as antifungal agents, with special emphasis on RIP/fungus specificity, possible mechanisms of antifungal action, and the use of RIP genes to obtain fungus-resistant transgenic plants. It also highlights the fact that these proteins also have antiviral and insecticidal activity, which makes them very versatile tools for crop protection.
Collapse
Affiliation(s)
- Rosario Iglesias
- Department of Biochemistry and Molecular Biology and Physiology, Faculty of Sciences, University of Valladolid, E-47011 Valladolid, Spain; (R.I.); (L.C.)
| | - Lucía Citores
- Department of Biochemistry and Molecular Biology and Physiology, Faculty of Sciences, University of Valladolid, E-47011 Valladolid, Spain; (R.I.); (L.C.)
| | - Claudia C. Gay
- Laboratory of Protein Research, Institute of Basic and Applied Chemistry of Northeast Argentina (UNNE-CONICET), Faculty of Exact and Natural Sciences and Surveying, Av. Libertad 5470, Corrientes 3400, Argentina;
| | - José M. Ferreras
- Department of Biochemistry and Molecular Biology and Physiology, Faculty of Sciences, University of Valladolid, E-47011 Valladolid, Spain; (R.I.); (L.C.)
| |
Collapse
|
3
|
Lteif M, Pallardy M, Turbica I. Antibodies internalization mechanisms by dendritic cells and their role in therapeutic antibody immunogenicity. Eur J Immunol 2024; 54:e2250340. [PMID: 37985174 DOI: 10.1002/eji.202250340] [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: 05/18/2023] [Revised: 11/14/2023] [Accepted: 11/17/2023] [Indexed: 11/22/2023]
Abstract
Internalization and processing by antigen-presenting cells such as dendritic cells (DCs) are critical steps for initiating a T-cell response to therapeutic antibodies. Consequences are the production of neutralizing antidrug antibodies altering the clinical response, the presence of immune complexes, and, in some rare cases, hypersensitivity reactions. In recent years, significant progress has been made in the knowledge of cellular uptake mechanisms of antibodies in DCs. The uptake of antibodies could be directly related to their immunogenicity by regulating the quantity of materials entering the DCs in relation to antibody structure. Here, we summarize the latest insights into cellular uptake mechanisms and pathways in DCs. We highlight the approaches to study endocytosis, the impact of endocytosis routes on T-cell response, and discuss the link between how DCs internalize therapeutic antibodies and the potential mechanisms that could give rise to immunogenicity. Understanding these processes could help in developing assays to evaluate the immunogenicity potential of biotherapeutics.
Collapse
Affiliation(s)
- Maria Lteif
- Université Paris-Saclay, Inserm, Inflammation, Microbiome and Immunosurveillance, Orsay, France
| | - Marc Pallardy
- Université Paris-Saclay, Inserm, Inflammation, Microbiome and Immunosurveillance, Orsay, France
| | - Isabelle Turbica
- Université Paris-Saclay, Inserm, Inflammation, Microbiome and Immunosurveillance, Orsay, France
| |
Collapse
|
4
|
Garofalo M, Payros D, Taieb F, Oswald E, Nougayrède JP, Oswald IP. From ribosome to ribotoxins: understanding the toxicity of deoxynivalenol and Shiga toxin, two food borne toxins. Crit Rev Food Sci Nutr 2023; 65:193-205. [PMID: 37862145 DOI: 10.1080/10408398.2023.2271101] [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] [Indexed: 10/22/2023]
Abstract
Ribosomes that synthesize proteins are among the most central and evolutionarily conserved organelles. Given the key role of proteins in cellular functions, prokaryotic and eukaryotic pathogens have evolved potent toxins to inhibit ribosomal functions and weaken their host. Many of these ribotoxin-producing pathogens are associated with food. For example, food can be contaminated with bacterial pathogens that produce the ribotoxin Shiga toxin, but also with the fungal ribotoxin deoxynivalenol. Shiga toxin cleaves ribosomal RNA, while deoxynivalenol binds to and inhibits the peptidyl transferase center. Despite their distinct modes of action, both groups of ribotoxins hinder protein translation, but also trigger other comparable toxic effects, which depend or not on the activation of the ribotoxic stress response. Ribotoxic stress response-dependent effects include inflammation and apoptosis, whereas ribotoxic stress response-independent effects include endoplasmic reticulum stress, oxidative stress, and autophagy. For other effects, such as cell cycle arrest and cytoskeleton modulation, the involvement of the ribotoxic stress response is still controversial. Ribotoxins affect one organelle yet induce multiple toxic effects with multiple consequences for the cell. The ribosome can therefore be considered as the cellular "Achilles heel" targeted by food borne ribotoxins. Considering the high toxicity of ribotoxins, they pose a substantial health risk, as humans are highly susceptible to widespread exposure to these toxins through contaminated food sources.
Collapse
Affiliation(s)
- Marion Garofalo
- Toxalim (Research Centre in Food Toxicology), Université de Toulouse, INRAE, ENVT, INP-Purpan, UPS, Toulouse, France
- IRSD, Université de Toulouse, INSERM, INRAE, ENVT, UPS, Toulouse, France
| | - Delphine Payros
- Toxalim (Research Centre in Food Toxicology), Université de Toulouse, INRAE, ENVT, INP-Purpan, UPS, Toulouse, France
- IRSD, Université de Toulouse, INSERM, INRAE, ENVT, UPS, Toulouse, France
| | - Frederic Taieb
- IRSD, Université de Toulouse, INSERM, INRAE, ENVT, UPS, Toulouse, France
| | - Eric Oswald
- IRSD, Université de Toulouse, INSERM, INRAE, ENVT, UPS, Toulouse, France
- CHU Toulouse, Hôpital Purpan, Toulouse, France
| | | | - Isabelle P Oswald
- Toxalim (Research Centre in Food Toxicology), Université de Toulouse, INRAE, ENVT, INP-Purpan, UPS, Toulouse, France
| |
Collapse
|
5
|
Stuer N, Van Damme P, Goormachtig S, Van Dingenen J. Seeking the interspecies crosswalk for filamentous microbe effectors. TRENDS IN PLANT SCIENCE 2023; 28:1045-1059. [PMID: 37062674 DOI: 10.1016/j.tplants.2023.03.017] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/04/2022] [Revised: 03/02/2023] [Accepted: 03/18/2023] [Indexed: 06/19/2023]
Abstract
Both pathogenic and symbiotic microorganisms modulate the immune response and physiology of their host to establish a suitable niche. Key players in mediating colonization outcome are microbial effector proteins that act either inside (cytoplasmic) or outside (apoplastic) the plant cells and modify the abundance or activity of host macromolecules. We compile novel insights into the much-disputed processes of effector secretion and translocation of filamentous organisms, namely fungi and oomycetes. We report how recent studies that focus on unconventional secretion and effector structure challenge the long-standing image of effectors as conventionally secreted proteins that are translocated with the aid of primary amino acid sequence motifs. Furthermore, we emphasize the potential of diverse, unbiased, state-of-the-art proteomics approaches in the holistic characterization of fungal and oomycete effectomes.
Collapse
Affiliation(s)
- Naomi Stuer
- Department of Plant Biotechnology and Bioinformatics, Ghent University, 9052 Ghent, Belgium; Center for Plant Systems Biology, Vlaams Instituut voor Biotechnologie (VIB), 9052 Ghent, Belgium
| | - Petra Van Damme
- iRIP Unit, Laboratory of Microbiology, Department of Biochemistry and Microbiology, Ghent University, Karel Lodewijk Ledeganckstraat 35, 9000 Ghent, Belgium
| | - Sofie Goormachtig
- Department of Plant Biotechnology and Bioinformatics, Ghent University, 9052 Ghent, Belgium; Center for Plant Systems Biology, Vlaams Instituut voor Biotechnologie (VIB), 9052 Ghent, Belgium.
| | - Judith Van Dingenen
- Department of Plant Biotechnology and Bioinformatics, Ghent University, 9052 Ghent, Belgium; Center for Plant Systems Biology, Vlaams Instituut voor Biotechnologie (VIB), 9052 Ghent, Belgium.
| |
Collapse
|
6
|
Tomisch J, Busse V, Rosato F, Makshakova ON, Salavei P, Kittel AS, Gillon E, Lataster L, Imberty A, Meléndez AV, Römer W. A Shiga Toxin B-Subunit-Based Lectibody Boosts T Cell Cytotoxicity towards Gb3-Positive Cancer Cells. Cells 2023; 12:1896. [PMID: 37508560 PMCID: PMC10378424 DOI: 10.3390/cells12141896] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2023] [Revised: 07/10/2023] [Accepted: 07/18/2023] [Indexed: 07/30/2023] Open
Abstract
Aberrant glycosylation plays a crucial role in tumour progression and invasiveness. Tumour-associated carbohydrate antigens (TACAs) represent a valuable set of targets for immunotherapeutic approaches. The poor immunogenicity of glycan structures, however, requires a more effective and well-directed way of targeting TACAs on the surface of cancer cells than antibodies. The glycosphingolipid globotriaosylceramide (Gb3) is a well-established TACA present in a multitude of cancer types. Its overexpression has been linked to metastasis, invasiveness, and multidrug resistance. In the present study, we propose to use a dimeric fragment of the Shiga toxin B-subunit (StxB) to selectively target Gb3-positive cancer cells in a StxB-scFv UCHT1 lectibody. The lectibody, comprised of a lectin and the UCHT1 antibody fragment, was produced in E. coli and purified via Ni-NTA affinity chromatography. Specificity of the lectibody towards Gb3-positive cancer cell lines and specificity towards the CD3 receptor on T cells, was assessed using flow cytometry. We evaluated the efficacy of the lectibody in redirecting T cell cytotoxicity towards Gb3-overexpressing cancer cells in luciferase-based cytotoxicity in vitro assays. The StxB-scFv UCHT1 lectibody has proven specific for Gb3 and could induce the killing of up to 80% of Gb3-overexpressing cancer cells in haemorrhagic and solid tumours. The lectibody developed in this study, therefore, highlights the potential that lectibodies and lectins in general have for usage in immunotherapeutic approaches to boost the efficacy of established cancer treatments.
Collapse
Affiliation(s)
- Jana Tomisch
- Faculty of Biology, University of Freiburg, 79104 Freiburg, Germany
- Signalling Research Centres BIOSS and CIBSS, University of Freiburg, 79104 Freiburg, Germany
| | - Vincent Busse
- Faculty of Biology, University of Freiburg, 79104 Freiburg, Germany
- Signalling Research Centres BIOSS and CIBSS, University of Freiburg, 79104 Freiburg, Germany
| | - Francesca Rosato
- Faculty of Biology, University of Freiburg, 79104 Freiburg, Germany
- Signalling Research Centres BIOSS and CIBSS, University of Freiburg, 79104 Freiburg, Germany
| | - Olga N Makshakova
- Faculty of Biology, University of Freiburg, 79104 Freiburg, Germany
- Kazan Institute for Biochemistry and Biophysics, FRC Kazan Scientific Center of RAS, 420111 Kazan, Russia
| | - Pavel Salavei
- Signalling Research Centres BIOSS and CIBSS, University of Freiburg, 79104 Freiburg, Germany
- Core Facility Signalling Factory & Robotics, University of Freiburg, 79104 Freiburg, Germany
| | - Anna-Sophia Kittel
- Faculty of Biology, University of Freiburg, 79104 Freiburg, Germany
- Signalling Research Centres BIOSS and CIBSS, University of Freiburg, 79104 Freiburg, Germany
| | - Emilie Gillon
- CNRS, CERMAV, Université Grenoble Alpes, 38000 Grenoble, France
| | - Levin Lataster
- Faculty of Biology, University of Freiburg, 79104 Freiburg, Germany
- Signalling Research Centres BIOSS and CIBSS, University of Freiburg, 79104 Freiburg, Germany
| | - Anne Imberty
- CNRS, CERMAV, Université Grenoble Alpes, 38000 Grenoble, France
| | - Ana Valeria Meléndez
- Faculty of Biology, University of Freiburg, 79104 Freiburg, Germany
- Signalling Research Centres BIOSS and CIBSS, University of Freiburg, 79104 Freiburg, Germany
- Spemann Graduate School of Biology and Medicine, University of Freiburg, 79104 Freiburg, Germany
| | - Winfried Römer
- Faculty of Biology, University of Freiburg, 79104 Freiburg, Germany
- Signalling Research Centres BIOSS and CIBSS, University of Freiburg, 79104 Freiburg, Germany
- Spemann Graduate School of Biology and Medicine, University of Freiburg, 79104 Freiburg, Germany
- Freiburg Institute for Advanced Studies (FRIAS), University of Freiburg, 79106 Freiburg, Germany
| |
Collapse
|
7
|
Rosenkranz AA, Slastnikova TA. Prospects of Using Protein Engineering for Selective Drug Delivery into a Specific Compartment of Target Cells. Pharmaceutics 2023; 15:pharmaceutics15030987. [PMID: 36986848 PMCID: PMC10055131 DOI: 10.3390/pharmaceutics15030987] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2023] [Revised: 03/13/2023] [Accepted: 03/17/2023] [Indexed: 03/30/2023] Open
Abstract
A large number of proteins are successfully used to treat various diseases. These include natural polypeptide hormones, their synthetic analogues, antibodies, antibody mimetics, enzymes, and other drugs based on them. Many of them are demanded in clinical settings and commercially successful, mainly for cancer treatment. The targets for most of the aforementioned drugs are located at the cell surface. Meanwhile, the vast majority of therapeutic targets, which are usually regulatory macromolecules, are located inside the cell. Traditional low molecular weight drugs freely penetrate all cells, causing side effects in non-target cells. In addition, it is often difficult to elaborate a small molecule that can specifically affect protein interactions. Modern technologies make it possible to obtain proteins capable of interacting with almost any target. However, proteins, like other macromolecules, cannot, as a rule, freely penetrate into the desired cellular compartment. Recent studies allow us to design multifunctional proteins that solve these problems. This review considers the scope of application of such artificial constructs for the targeted delivery of both protein-based and traditional low molecular weight drugs, the obstacles met on the way of their transport to the specified intracellular compartment of the target cells after their systemic bloodstream administration, and the means to overcome those difficulties.
Collapse
Affiliation(s)
- Andrey A Rosenkranz
- Laboratory of Molecular Genetics of Intracellular Transport, Institute of Gene Biology of Russian Academy of Sciences, 34/5 Vavilov St., 119334 Moscow, Russia
- Department of Biophysics, Faculty of Biology, Lomonosov Moscow State University, 1-12 Leninskie Gory St., 119234 Moscow, Russia
| | - Tatiana A Slastnikova
- Laboratory of Molecular Genetics of Intracellular Transport, Institute of Gene Biology of Russian Academy of Sciences, 34/5 Vavilov St., 119334 Moscow, Russia
| |
Collapse
|
8
|
Zeng Y, Jiang M, Robinson S, Peng Z, Chonira V, Simeon R, Tzipori S, Zhang J, Chen Z. A Multi-Specific DARPin Potently Neutralizes Shiga Toxin 2 via Simultaneous Modulation of Both Toxin Subunits. Bioengineering (Basel) 2022; 9:511. [PMID: 36290479 PMCID: PMC9598796 DOI: 10.3390/bioengineering9100511] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2022] [Revised: 09/19/2022] [Accepted: 09/20/2022] [Indexed: 11/25/2022] Open
Abstract
Shiga toxin-producing E. coli (STEC) is a common cause of bloody diarrhea. The pathology of STEC infection derives from two exotoxins-Shiga toxin 1 (Stx1) and Shiga toxin 2 (Stx2)-that are secreted by STEC in the gut, from where they are systemically absorbed, causing severe kidney damage leading to hemolytic uremic syndrome (HUS). Currently, there is no effective treatment for HUS, and only supportive care is recommended. We report the engineering of a panel of designed ankyrin repeat proteins (DARPin) with potent neutralization activity against Stx2a, the major subtype associated with HUS. The best dimeric DARPin, SD5, created via a combination of directed evolution and rational design, neutralizes Stx2a with a half maximal effective concentration (EC50) of 0.61 nM in vitro. The two monomeric DARPin constituents of SD5 exhibit complementary functions-SHT targets the enzymatic A subunit of Stx2a and inhibits the toxin's catalytic activity, while DARPin #3 binds the B subunit, based on the cryo-EM study, and induces a novel conformational change in the B subunit that distorts its five-fold symmetry and presumably interferes with toxin attachment to target cells. SD5 was fused to an albumin-binding DARPin, and the resulting trimeric DARPin DA1-SD5 efficiently protects mice in a toxin challenge model, pointing to a high potential of this DARPin as a therapeutic for STEC infection. Finally, the unprecedented toxin conformational change induced by DARPin #3 represents a novel mode of action for neutralizing Stx2 toxicity and reveals new targets for future drug development.
Collapse
Affiliation(s)
- Yu Zeng
- Department of Microbial Pathogenesis and Immunology, Texas A&M University Health Science Center, 8847 Riverside Pkwy, Bryan, TX 77807, USA
| | - Mengqiu Jiang
- Center for Phage Technology, Department of Biochemistry and Biophysics, Texas A&M University, 300 Olsen Blvd., College Station, TX 77843, USA
| | - Sally Robinson
- Department of Infectious Disease and Global Health, Cummings School of Veterinary Medicine, Tufts University, 200 Westboro Rd, North Grafton, MA 01536, USA
| | - Zeyu Peng
- Department of Microbial Pathogenesis and Immunology, Texas A&M University Health Science Center, 8847 Riverside Pkwy, Bryan, TX 77807, USA
| | - Vikas Chonira
- Department of Microbial Pathogenesis and Immunology, Texas A&M University Health Science Center, 8847 Riverside Pkwy, Bryan, TX 77807, USA
| | - Rudo Simeon
- Department of Microbial Pathogenesis and Immunology, Texas A&M University Health Science Center, 8847 Riverside Pkwy, Bryan, TX 77807, USA
| | - Saul Tzipori
- Department of Infectious Disease and Global Health, Cummings School of Veterinary Medicine, Tufts University, 200 Westboro Rd, North Grafton, MA 01536, USA
| | - Junjie Zhang
- Center for Phage Technology, Department of Biochemistry and Biophysics, Texas A&M University, 300 Olsen Blvd., College Station, TX 77843, USA
| | - Zhilei Chen
- Department of Microbial Pathogenesis and Immunology, Texas A&M University Health Science Center, 8847 Riverside Pkwy, Bryan, TX 77807, USA
- Interdisciplinary Graduate Program in Genetics, Texas A&M University, 300 Olsen Blvd., College Station, TX 77843, USA
| |
Collapse
|
9
|
Park JY, Kim CH, Cho SH. Glycan-Adhering Lectins and Experimental Evaluation of a Lectin FimH Inhibitor in Enterohemorrhagic Escherichia coli (EHEC) O157:H7 Strain EDL933. Int J Mol Sci 2022; 23:ijms23179931. [PMID: 36077327 PMCID: PMC9455959 DOI: 10.3390/ijms23179931] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2022] [Revised: 08/30/2022] [Accepted: 08/30/2022] [Indexed: 11/16/2022] Open
Abstract
In this study, we tried to develop a FimH inhibitor that inhibits adhesion of enterohemorrhagic Escherichia coli (EHEC) on the epithelium of human intestine during the initial stage of infections. Using a T7 phage display method with a reference strain, EHEC EDL933, FimH was selected as an adherent lectin to GM1a and Gb3 glycans. In order to detect the ligand binding domain (LBD) of FimH, we used a docking simulation and found three binding site sequences of FimH, i.e., P1, P2, and P3. Among Gb3 mimic peptides, P2 was found to have the strongest binding strength. Moreover, in vitro treatment with peptide P2 inhibited binding activity in a concentration-dependent manner. Furthermore, we conducted confirmation experiments through several strains isolated from patients in Korea, EHEC NCCP15736, NCCP15737, and NCCP15739. In addition, we analyzed the evolutionary characteristics of the predicted FimH lectin-like adhesins to construct a lectin-glycan interaction (LGI). We selected 70 recently differentiated strains from the phylogenetic tree of 2240 strains with Shiga toxin in their genome. We can infer EHEC strains dynamically evolved but FimH was conserved during the evolution time according to the phylogenetic tree. Furthermore, FimH could be a reliable candidate of drug target in terms of evolution. We examined how pathogen lectins interact with host glycans early in infection in EDL933 as well as several field strains and confirmed that glycan-like peptides worked as an initial infection inhibitor.
Collapse
Affiliation(s)
- Jun-Young Park
- Division of Zoonotic and Vector Borne Disease Research, Center for Infectious Disease Research, Korea National Institute of Health, Cheongju 28159, Korea
| | - Cheorl-Ho Kim
- Glycobiology Unit, Department of Biological Science, Sung Kyunkwan University and Samsung Advanced Institute for Health Science and Technology (SAIHST), Suwon 16419, Korea
| | - Seung-Hak Cho
- Division of Zoonotic and Vector Borne Disease Research, Center for Infectious Disease Research, Korea National Institute of Health, Cheongju 28159, Korea
- Correspondence: ; Tel.: +82-43-913-4899
| |
Collapse
|
10
|
Gb3/cd77 Is a Predictive Marker and Promising Therapeutic Target for Head and Neck Cancer. Biomedicines 2022; 10:biomedicines10040732. [PMID: 35453483 PMCID: PMC9029501 DOI: 10.3390/biomedicines10040732] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2022] [Revised: 03/12/2022] [Accepted: 03/18/2022] [Indexed: 12/01/2022] Open
Abstract
Head and neck squamous cell carcinoma is the sixth leading cancer in the world. This cancer is difficult to treat and is characterized by recurrences that are often fatal. This cancer is generally removed surgically, but it often regrows from the edges of the lesion from where most recurrences reappear. In this study, we have investigated if the expression of GB3 in human cell lines, tissues from patient biopsies, and a murine animal model could be used as an early and determinant marker of HNC. We found that in all the investigated systems, this marker appears in neoplastic cells from the very early stages of their malignant transformation. Our conclusions support the hypothesis that GB3 is a reliable and independent target for HNC identification and selective delivery of treatments. Furthermore, we show that the level of expression of this marker correlates with the degree of malignancy of the tumor. These studies suggest that GB3 may provide the basis for the early identification and new targeted therapies for head and neck cancer.
Collapse
|
11
|
Ehrenfeld M, Schrade A, Flisikowska T, Perl M, Hirsch ND, Sichler A, Geyer L, Flisikowski K, Wilhelm D, Schober SJ, Johannes L, Schnieke A, Janssen KP. Tumor targeting with bacterial Shiga toxin B-subunit in genetic porcine models for colorectal cancer and osteosarcoma. Mol Cancer Ther 2022; 21:686-699. [PMID: 35086950 DOI: 10.1158/1535-7163.mct-21-0445] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2021] [Revised: 10/31/2021] [Accepted: 01/11/2022] [Indexed: 12/09/2022]
Abstract
The B-subunit of bacterial Shiga toxin (STxB) is non-toxic and has low immunogenicity. Its receptor, the glycosphingolipid Gb3/CD77, is overexpressed on the cell surface of human colorectal cancer (CRC). We tested whether genetic porcine models, closely resembling human anatomy and pathophysiology, can be used to exploit the tumor targeting potential of STxB. In accordance with findings on human CRC, the pig model APC1311 bound STxB in colorectal tumors, but not in normal colon or jejunum, except for putative enteroendocrine cells. In primary tumor cells from endoscopic biopsies, STxB was rapidly taken up along the retrograde intracellular route to the Golgi, whereas normal colon organoids did not bind or internalize STxB. Next, we tested a porcine model (TP53LSL-R167H) for osteosarcoma, a tumor entity with a dismal prognosis and insufficient treatment options, hitherto not known to express Gb3. Pig osteosarcoma strongly bound StxB and expressed the Gb3-synthase A4GALT. Primary osteosarcoma cells, but not normal osteoblasts, rapidly internalized fluorescently labelled STxB along the retrograde route to the Golgi. Importantly, six out of eight human osteosarcoma cell lines expressed A4GALT mRNA and showed prominent intracellular uptake of STxB. The physiological role of A4GALT was tested by Crispr/Cas9-mutagenesis in porcine LLC-PK1 kidney epithelial cells and RNA interference in MG-63 human osteosarcoma cells. A4GALT-deficiency or knock-down abolished STxB uptake and led to significantly reduced cell migration and proliferation, hinting towards a putative tumor-promoting role of Gb3. Thus, pig models are suitable tools for STxB-based tumor targeting, and may allow "reverse-translational" predictions on human tumor biology.
Collapse
Affiliation(s)
- Maximilian Ehrenfeld
- Departments of Surgery and Urology, Klinikum rechts der Isar, Technical University München
| | - Anna Schrade
- Department of Surgery, Klinikum rechts der Isar, Technical University München
| | - Tatiana Flisikowska
- Chair of Livestock Biotechnology, School of Life Sciences, Technical University of Munich
| | - Markus Perl
- Department of Internal Medicine III, University Hospital Regensburg
| | - Noah-David Hirsch
- Department of Surgery, Klinikum rechts der Isar, Technical University Munich
| | - Anna Sichler
- Department of Surgery, Klinikum rechts der Isar, Technical University Munich
| | - Laura Geyer
- Department of Surgery, Klinikum rechts der Isar, Technical University München
| | - Krzysztof Flisikowski
- Chair of Livestock Biotechnology, School of Life Sciences, Technical University of Munich
| | - Dirk Wilhelm
- Department of Surgery, Klinikum rechts der Isar, Technical University of Munich
| | - Sebastian Johannes Schober
- Department of Pediatrics and Children's Cancer Research Center, Kinderklinik München Schwabing, Technical University of Munich
| | - Ludger Johannes
- Endocytic Trafficking and Intracellular Delivery team, Institute Curie
| | | | - Klaus-Peter Janssen
- Department of Surgery, Klinikum rechts der Isar, Technical University München
| |
Collapse
|
12
|
Lee K, Lee J, Lee P, Jeon BC, Song MY, Kwak S, Lee J, Kim J, Kim D, Kim JH, Tesh VL, Lee M, Park S. Inhibition of O-GlcNAcylation protects from Shiga toxin-mediated cell injury and lethality in host. EMBO Mol Med 2022; 14:e14678. [PMID: 34842355 PMCID: PMC8749473 DOI: 10.15252/emmm.202114678] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2021] [Revised: 11/08/2021] [Accepted: 11/11/2021] [Indexed: 11/09/2022] Open
Abstract
Shiga toxins (Stxs) produced by enterohemorrhagic Escherichia coli (EHEC) are the major virulence factors responsible for hemorrhagic colitis, which can lead to life-threatening systemic complications including acute renal failure (hemolytic uremic syndrome) and neuropathy. Here, we report that O-GlcNAcylation, a type of post-translational modification, was acutely increased upon induction of endoplasmic reticulum (ER) stress in host cells by Stxs. Suppression of the abnormal Stx-mediated increase in O-GlcNAcylation effectively inhibited apoptotic and inflammatory responses in Stx-susceptible cells. The protective effect of O-GlcNAc inhibition for Stx-mediated pathogenic responses was also verified using three-dimensional (3D)-cultured spheroids or organoids mimicking the human kidney. Treatment with an O-GlcNAcylation inhibitor remarkably improved the major disease symptoms and survival rate for mice intraperitoneally injected with a lethal dose of Stx. In conclusion, this study elucidates O-GlcNAcylation-dependent pathogenic mechanisms of Stxs and demonstrates that inhibition of aberrant O-GlcNAcylation is a potential approach to treat Stx-mediated diseases.
Collapse
Affiliation(s)
- Kyung‐Soo Lee
- Environmental Diseases Research CenterKorea Research Institute of Bioscience & Biotechnology (KRIBB)DaejeonKorea
- Department of Biomolecular ScienceKRIBB School of BioscienceKorea University of Science and Technology (UST)DaejeonKorea
| | - Jieun Lee
- Environmental Diseases Research CenterKorea Research Institute of Bioscience & Biotechnology (KRIBB)DaejeonKorea
| | - Pureum Lee
- Environmental Diseases Research CenterKorea Research Institute of Bioscience & Biotechnology (KRIBB)DaejeonKorea
- Department of Biomolecular ScienceKRIBB School of BioscienceKorea University of Science and Technology (UST)DaejeonKorea
| | - Bong Chan Jeon
- Department of Biomolecular ScienceKRIBB School of BioscienceKorea University of Science and Technology (UST)DaejeonKorea
- Immunotherapy Convergence Research CenterKorea Research Institute of Bioscience & Biotechnology (KRIBB)DaejeonKorea
| | - Min Yeong Song
- Environmental Diseases Research CenterKorea Research Institute of Bioscience & Biotechnology (KRIBB)DaejeonKorea
- Department of Biomolecular ScienceKRIBB School of BioscienceKorea University of Science and Technology (UST)DaejeonKorea
| | - Sojung Kwak
- Environmental Diseases Research CenterKorea Research Institute of Bioscience & Biotechnology (KRIBB)DaejeonKorea
| | - Jungwoon Lee
- Environmental Diseases Research CenterKorea Research Institute of Bioscience & Biotechnology (KRIBB)DaejeonKorea
- Department of Biomolecular ScienceKRIBB School of BioscienceKorea University of Science and Technology (UST)DaejeonKorea
| | - Jun‐Seob Kim
- Department of Nano‐BioengineeringIncheon National UniversityIncheonKorea
| | - Doo‐Jin Kim
- Infectious Disease Research CenterKorea Research Institute of Bioscience & Biotechnology (KRIBB)DaejeonKorea
| | - Ji Hyung Kim
- Infectious Disease Research CenterKorea Research Institute of Bioscience & Biotechnology (KRIBB)DaejeonKorea
| | - Vernon L Tesh
- Department of Microbial Pathogenesis and ImmunologyCollege of MedicineTexas A&M UniversityBryanTXUSA
| | - Moo‐Seung Lee
- Environmental Diseases Research CenterKorea Research Institute of Bioscience & Biotechnology (KRIBB)DaejeonKorea
- Department of Biomolecular ScienceKRIBB School of BioscienceKorea University of Science and Technology (UST)DaejeonKorea
| | - Sung‐Kyun Park
- Infectious Disease Research CenterKorea Research Institute of Bioscience & Biotechnology (KRIBB)DaejeonKorea
| |
Collapse
|
13
|
Targeting Nanomaterials to Head and Neck Cancer Cells Using a Fragment of the Shiga Toxin as a Potent Natural Ligand. Cancers (Basel) 2021; 13:cancers13194920. [PMID: 34638405 PMCID: PMC8507991 DOI: 10.3390/cancers13194920] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2021] [Revised: 09/24/2021] [Accepted: 09/28/2021] [Indexed: 12/29/2022] Open
Abstract
Head and Neck Cancer (HNC) is the seventh most common cancer worldwide with a 5-year survival from diagnosis of 50%. Currently, HNC is diagnosed by a physical examination followed by an histological biopsy, with surgery being the primary treatment. Here, we propose the use of targeted nanotechnology in support of existing diagnostic and therapeutic tools to prevent recurrences of tumors with poorly defined or surgically inaccessible margins. We have designed an innocuous ligand-protein, based on the receptor-binding domain of the Shiga toxin (ShTxB), that specifically drives nanoparticles to HNC cells bearing the globotriaosylceramide receptor on their surfaces. Microscopy images show how, upon binding to the receptor, the ShTxB-coated nanoparticles cause the clustering of the globotriaosylceramide receptors, the protrusion of filopodia, and rippling of the membrane, ultimately allowing the penetration of the ShTxB nanoparticles directly into the cell cytoplasm, thus triggering a biomimetic cellular response indistinguishable from that triggered by the full-length Shiga toxin. This functionalization strategy is a clear example of how some toxin fragments can be used as natural biosensors for the detection of some localized cancers and to target nanomedicines to HNC lesions.
Collapse
|
14
|
Zhang M, Xu N, Xu W, Ling G, Zhang P. Potential therapies and diagnosis based on Golgi-targeted nano drug delivery systems. Pharmacol Res 2021; 175:105861. [PMID: 34464677 DOI: 10.1016/j.phrs.2021.105861] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/22/2021] [Revised: 08/09/2021] [Accepted: 08/26/2021] [Indexed: 02/06/2023]
Abstract
With the rapid development of nanotechnology, organelle-targeted nano drug delivery systems (NDDSs) have emerged as a potential method which can transport drugs specifically to the subcellular compartments like nucleus, mitochondrion, lysosome, endoplasmic reticulum (ER) and Golgi apparatus (GA). GA not only plays a key role in receiving, modifying, packaging and transporting proteins and lipids, but also contributes to a set of cellular processes. Golgi-targeted NDDSs can alter the morphology of GA and will become a promising strategy with high specificity, low-dose administration and decreased occurrence of side effects. In this review, Golgi-targeted NDDSs and their applications in disease therapies and diagnosis such as cancer, metastasis, fibrosis and neurological diseases are introduced. Meanwhile, modifications of NDDSs to achieve targeting strategies, Golgi-disturbing agents to change the morphology of GA, special endocytosis to achieve endosomal/lysosomal escape strategies are also involved.
Collapse
Affiliation(s)
- Manyue Zhang
- Shenyang Pharmaceutical University, No. 103, Wenhua Road, Shenyang 110016, China
| | - Na Xu
- Shenyang Pharmaceutical University, No. 103, Wenhua Road, Shenyang 110016, China
| | - Wenxin Xu
- Shenyang Pharmaceutical University, No. 103, Wenhua Road, Shenyang 110016, China
| | - Guixia Ling
- Shenyang Pharmaceutical University, No. 103, Wenhua Road, Shenyang 110016, China.
| | - Peng Zhang
- Shenyang Pharmaceutical University, No. 103, Wenhua Road, Shenyang 110016, China.
| |
Collapse
|
15
|
Podlacha M, Grabowski Ł, Kosznik-Kawśnicka K, Zdrojewska K, Stasiłojć M, Węgrzyn G, Węgrzyn A. Interactions of Bacteriophages with Animal and Human Organisms-Safety Issues in the Light of Phage Therapy. Int J Mol Sci 2021; 22:8937. [PMID: 34445641 PMCID: PMC8396182 DOI: 10.3390/ijms22168937] [Citation(s) in RCA: 54] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2021] [Revised: 08/12/2021] [Accepted: 08/17/2021] [Indexed: 12/13/2022] Open
Abstract
Bacteriophages are viruses infecting bacterial cells. Since there is a lack of specific receptors for bacteriophages on eukaryotic cells, these viruses were for a long time considered to be neutral to animals and humans. However, studies of recent years provided clear evidence that bacteriophages can interact with eukaryotic cells, significantly influencing the functions of tissues, organs, and systems of mammals, including humans. In this review article, we summarize and discuss recent discoveries in the field of interactions of phages with animal and human organisms. Possibilities of penetration of bacteriophages into eukaryotic cells, tissues, and organs are discussed, and evidence of the effects of phages on functions of the immune system, respiratory system, central nervous system, gastrointestinal system, urinary tract, and reproductive system are presented and discussed. Modulations of cancer cells by bacteriophages are indicated. Direct and indirect effects of virulent and temperate phages are discussed. We conclude that interactions of bacteriophages with animal and human organisms are robust, and they must be taken under consideration when using these viruses in medicine, especially in phage therapy, and in biotechnological applications.
Collapse
Affiliation(s)
- Magdalena Podlacha
- Department of Molecular Biology, University of Gdansk, Wita Stwosza 59, 80-308 Gdansk, Poland; (M.P.); (K.Z.); (M.S.); (G.W.)
| | - Łukasz Grabowski
- Laboratory of Phage Therapy, Institute of Biochemistry and Biophysics, Polish Academy of Sciences, Kładki 24, 80-822 Gdansk, Poland; (Ł.G.); (K.K.-K.)
| | - Katarzyna Kosznik-Kawśnicka
- Laboratory of Phage Therapy, Institute of Biochemistry and Biophysics, Polish Academy of Sciences, Kładki 24, 80-822 Gdansk, Poland; (Ł.G.); (K.K.-K.)
| | - Karolina Zdrojewska
- Department of Molecular Biology, University of Gdansk, Wita Stwosza 59, 80-308 Gdansk, Poland; (M.P.); (K.Z.); (M.S.); (G.W.)
| | - Małgorzata Stasiłojć
- Department of Molecular Biology, University of Gdansk, Wita Stwosza 59, 80-308 Gdansk, Poland; (M.P.); (K.Z.); (M.S.); (G.W.)
| | - Grzegorz Węgrzyn
- Department of Molecular Biology, University of Gdansk, Wita Stwosza 59, 80-308 Gdansk, Poland; (M.P.); (K.Z.); (M.S.); (G.W.)
| | - Alicja Węgrzyn
- Laboratory of Phage Therapy, Institute of Biochemistry and Biophysics, Polish Academy of Sciences, Kładki 24, 80-822 Gdansk, Poland; (Ł.G.); (K.K.-K.)
| |
Collapse
|
16
|
Detzner J, Klein AL, Pohlentz G, Krojnewski E, Humpf HU, Mellmann A, Karch H, Müthing J. Primary Human Renal Proximal Tubular Epithelial Cells (pHRPTEpiCs): Shiga Toxin (Stx) Glycosphingolipid Receptors, Stx Susceptibility, and Interaction with Membrane Microdomains. Toxins (Basel) 2021; 13:toxins13080529. [PMID: 34437399 PMCID: PMC8402424 DOI: 10.3390/toxins13080529] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2021] [Revised: 07/20/2021] [Accepted: 07/23/2021] [Indexed: 12/13/2022] Open
Abstract
Tubular epithelial cells of the human kidney are considered as targets of Shiga toxins (Stxs) in the Stx-mediated pathogenesis of hemolytic–uremic syndrome (HUS) caused by Stx-releasing enterohemorrhagic Escherichia coli (EHEC). Analysis of Stx-binding glycosphingolipids (GSLs) of primary human renal proximal tubular epithelial cells (pHRPTEpiCs) yielded globotriaosylceramide (Gb3Cer) and globotetraosylceramide (Gb4Cer) with Cer (d18:1, C16:0), Cer (d18:1, C22:0), and Cer (d18:1, C24:1/C24:0) as the dominant lipoforms. Investigation of detergent-resistant membranes (DRMs) and nonDRMs, serving as equivalents for the liquid-ordered and liquid-disordered membrane phase, respectively, revealed the prevalence of Gb3Cer and Gb4Cer together with cholesterol and sphingomyelin in DRMs, suggesting lipid raft association. Stx1a and Stx2a exerted strong cellular damage with half-maximal cytotoxic doses (CD50) of 1.31 × 102 pg/mL and 1.66 × 103 pg/mL, respectively, indicating one order of magnitude higher cellular cytotoxicity of Stx1a. Surface acoustic wave (SAW) real-time interaction analysis using biosensor surfaces coated with DRM or nonDRM fractions gave stronger binding capability of Stx1a versus Stx2a that correlated with the lower cytotoxicity of Stx2a. Our study underlines the substantial role of proximal tubular epithelial cells of the human kidney being associated with the development of Stx-mediated HUS at least for Stx1a, while the impact of Stx2a remains somewhat ambiguous.
Collapse
Affiliation(s)
- Johanna Detzner
- Institute of Hygiene, University of Münster, D-48149 Münster, Germany; (J.D.); (A.-L.K.); (G.P.); (E.K.); (A.M.); (H.K.)
| | - Anna-Lena Klein
- Institute of Hygiene, University of Münster, D-48149 Münster, Germany; (J.D.); (A.-L.K.); (G.P.); (E.K.); (A.M.); (H.K.)
| | - Gottfried Pohlentz
- Institute of Hygiene, University of Münster, D-48149 Münster, Germany; (J.D.); (A.-L.K.); (G.P.); (E.K.); (A.M.); (H.K.)
| | - Elisabeth Krojnewski
- Institute of Hygiene, University of Münster, D-48149 Münster, Germany; (J.D.); (A.-L.K.); (G.P.); (E.K.); (A.M.); (H.K.)
| | - Hans-Ulrich Humpf
- Institute of Food Chemistry, University of Münster, D-48149 Münster, Germany;
| | - Alexander Mellmann
- Institute of Hygiene, University of Münster, D-48149 Münster, Germany; (J.D.); (A.-L.K.); (G.P.); (E.K.); (A.M.); (H.K.)
| | - Helge Karch
- Institute of Hygiene, University of Münster, D-48149 Münster, Germany; (J.D.); (A.-L.K.); (G.P.); (E.K.); (A.M.); (H.K.)
| | - Johannes Müthing
- Institute of Hygiene, University of Münster, D-48149 Münster, Germany; (J.D.); (A.-L.K.); (G.P.); (E.K.); (A.M.); (H.K.)
- Correspondence:
| |
Collapse
|
17
|
Siukstaite L, Imberty A, Römer W. Structural Diversities of Lectins Binding to the Glycosphingolipid Gb3. Front Mol Biosci 2021; 8:704685. [PMID: 34381814 PMCID: PMC8350385 DOI: 10.3389/fmolb.2021.704685] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2021] [Accepted: 07/08/2021] [Indexed: 12/18/2022] Open
Abstract
Glycolipids are present on the surfaces of all living cells and thereby represent targets for many protein receptors, such as lectins. Understanding the interactions between lectins and glycolipids is essential for investigating the functions of lectins and the dynamics of glycolipids in living membranes. This review focuses on lectins binding to the glycosphingolipid globotriaosylceramide (Gb3), an attractive host cell receptor, particularly for pathogens and pathogenic products. Shiga toxin (Stx), from Shigella dysenteriae or Escherichia coli, which is one of the most virulent bacterial toxins, binds and clusters Gb3, leading to local negative membrane curvature and the formation of tubular plasma membrane invaginations as the initial step for clathrin-independent endocytosis. After internalization, it is embracing the retrograde transport pathway. In comparison, the homotetrameric lectin LecA from Pseudomonas aeruginosa can also bind to Gb3, triggering the so-called lipid zipper mechanism, which results in membrane engulfment of the bacterium as an important step for its cellular uptake. Notably, both lectins bind to Gb3 but induce distinct plasma membrane domains and exploit mainly different transport pathways. Not only, several other Gb3-binding lectins have been described from bacterial origins, such as the adhesins SadP (from Streptococcus suis) and PapG (from E. coli), but also from animal, fungal, or plant origins. The variety of amino acid sequences and folds demonstrates the structural versatilities of Gb3-binding lectins and asks the question of the evolution of specificity and carbohydrate recognition in different kingdoms of life.
Collapse
Affiliation(s)
- Lina Siukstaite
- Faculty of Biology, University of Freiburg, Freiburg, Germany.,Signalling Research Centres BIOSS and CIBSS, University of Freiburg, Freiburg, Germany
| | - Anne Imberty
- CNRS, CERMAV, Université Grenoble Alpes, Grenoble, France
| | - Winfried Römer
- Faculty of Biology, University of Freiburg, Freiburg, Germany.,Signalling Research Centres BIOSS and CIBSS, University of Freiburg, Freiburg, Germany.,Freiburg Institute for Advanced Studies (FRIAS), University of Freiburg, Freiburg, Germany
| |
Collapse
|
18
|
Lingwood C. Therapeutic Uses of Bacterial Subunit Toxins. Toxins (Basel) 2021; 13:toxins13060378. [PMID: 34073185 PMCID: PMC8226680 DOI: 10.3390/toxins13060378] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2021] [Revised: 05/17/2021] [Accepted: 05/18/2021] [Indexed: 02/07/2023] Open
Abstract
The B subunit pentamer verotoxin (VT aka Shiga toxin-Stx) binding to its cellular glycosphingolipid (GSL) receptor, globotriaosyl ceramide (Gb3) mediates internalization and the subsequent receptor mediated retrograde intracellular traffic of the AB5 subunit holotoxin to the endoplasmic reticulum. Subunit separation and cytosolic A subunit transit via the ER retrotranslocon as a misfolded protein mimic, then inhibits protein synthesis to kill cells, which can cause hemolytic uremic syndrome clinically. This represents one of the most studied systems of prokaryotic hijacking of eukaryotic biology. Similarly, the interaction of cholera AB5 toxin with its GSL receptor, GM1 ganglioside, is the key component of the gastrointestinal pathogenesis of cholera and follows the same retrograde transport pathway for A subunit cytosol access. Although both VT and CT are the cause of major pathology worldwide, the toxin–receptor interaction is itself being manipulated to generate new approaches to control, rather than cause, disease. This arena comprises two areas: anti neoplasia, and protein misfolding diseases. CT/CTB subunit immunomodulatory function and anti-cancer toxin immunoconjugates will not be considered here. In the verotoxin case, it is clear that Gb3 (and VT targeting) is upregulated in many human cancers and that there is a relationship between GSL expression and cancer drug resistance. While both verotoxin and cholera toxin similarly hijack the intracellular ERAD quality control system of nascent protein folding, the more widespread cell expression of GM1 makes cholera the toxin of choice as the means to more widely utilise ERAD targeting to ameliorate genetic diseases of protein misfolding. Gb3 is primarily expressed in human renal tissue. Glomerular endothelial cells are the primary VT target but Gb3 is expressed in other endothelial beds, notably brain endothelial cells which can mediate the encephalopathy primarily associated with VT2-producing E. coli infection. The Gb3 levels can be regulated by cytokines released during EHEC infection, which complicate pathogenesis. Significantly Gb3 is upregulated in the neovasculature of many tumours, irrespective of tumour Gb3 status. Gb3 is markedly increased in pancreatic, ovarian, breast, testicular, renal, astrocytic, gastric, colorectal, cervical, sarcoma and meningeal cancer relative to the normal tissue. VT has been shown to be effective in mouse xenograft models of renal, astrocytoma, ovarian, colorectal, meningioma, and breast cancer. These studies are herein reviewed. Both CT and VT (and several other bacterial toxins) access the cell cytosol via cell surface ->ER transport. Once in the ER they interface with the protein folding homeostatic quality control pathway of the cell -ERAD, (ER associated degradation), which ensures that only correctly folded nascent proteins are allowed to progress to their cellular destinations. Misfolded proteins are translocated through the ER membrane and degraded by cytosolic proteosome. VT and CT A subunits have a C terminal misfolded protein mimic sequence to hijack this transporter to enter the cytosol. This interface between exogenous toxin and genetically encoded endogenous mutant misfolded proteins, provides a new therapeutic basis for the treatment of such genetic diseases, e.g., Cystic fibrosis, Gaucher disease, Krabbe disease, Fabry disease, Tay-Sachs disease and many more. Studies showing the efficacy of this approach in animal models of such diseases are presented.
Collapse
Affiliation(s)
- Clifford Lingwood
- Division of Molecular Medicine, Research Institute, Hospital for Sick Children, Toronto, ON M5G 1X8, Canada;
- Departments of Laboratory Medicine & Pathobiology, and Biochemistry, University of Toronto, Toronto, ON M5S 1A8, Canada
| |
Collapse
|
19
|
Piedrafita A, Ribes D, Cointault O, Chauveau D, Faguer S, Huart A. Plasma exchange and thrombotic microangiopathies: From pathophysiology to clinical practice. Transfus Apher Sci 2020; 59:102990. [PMID: 33272850 DOI: 10.1016/j.transci.2020.102990] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Thrombotic microangiopathy (TMA) brings together many diseases that have a commonality in the apparition of mechanical hemolysis with consuming thrombopenia. In all cases, these diseases can be life threatening, thereby justifying the implementation of treatment as an emergency. First-line treatment represents plasma exchange. This treatment has proven efficiency in improving the vital patient's and functional prognosis. However, the administration methods of plasma exchange can be redefined in light of the understanding of the pathophysiology of TMA. The aim of this review is to try to define, from pathophysiology, the place of plasma exchanges in the modern therapeutic arsenal of TMA.
Collapse
Affiliation(s)
- Alexis Piedrafita
- Département de Néphrologie et Transplantation d'Organes, Centre Hospitalier Universitaire de Toulouse, Toulouse, France; Institut National de la Santé et de la Recherche Médicale, UMR1048, Institut des Maladies Métaboliques et Cardiovasculaires, Toulouse, France; Université Paul Sabatier - Toulouse 3, Toulouse, France
| | - David Ribes
- Département de Néphrologie et Transplantation d'Organes, Centre Hospitalier Universitaire de Toulouse, Toulouse, France
| | - Olivier Cointault
- Département de Néphrologie et Transplantation d'Organes, Centre Hospitalier Universitaire de Toulouse, Toulouse, France
| | - Dominique Chauveau
- Département de Néphrologie et Transplantation d'Organes, Centre Hospitalier Universitaire de Toulouse, Toulouse, France; Institut National de la Santé et de la Recherche Médicale, UMR1048, Institut des Maladies Métaboliques et Cardiovasculaires, Toulouse, France; Université Paul Sabatier - Toulouse 3, Toulouse, France
| | - Stanislas Faguer
- Département de Néphrologie et Transplantation d'Organes, Centre Hospitalier Universitaire de Toulouse, Toulouse, France; Institut National de la Santé et de la Recherche Médicale, UMR1048, Institut des Maladies Métaboliques et Cardiovasculaires, Toulouse, France; Université Paul Sabatier - Toulouse 3, Toulouse, France
| | - Antoine Huart
- Département de Néphrologie et Transplantation d'Organes, Centre Hospitalier Universitaire de Toulouse, Toulouse, France.
| |
Collapse
|
20
|
Lee KS, Lee J, Lee P, Kim CU, Kim DJ, Jeong YJ, Park YJ, Tesh VL, Lee MS. Exosomes released from Shiga toxin 2a-treated human macrophages modulate inflammatory responses and induce cell death in toxin receptor expressing human cells. Cell Microbiol 2020; 22:e13249. [PMID: 32772454 DOI: 10.1111/cmi.13249] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2020] [Revised: 07/02/2020] [Accepted: 07/24/2020] [Indexed: 12/13/2022]
Abstract
Shiga toxins (Stxs) produced by Stx-producing Escherichia coli are the primarily virulence factors of hemolytic uremic syndrome and central nervous system (CNS) impairment. Although the precise mechanisms of toxin dissemination remain unclear, Stxs bind to extracellular vesicles (EVs). Exosomes, a subset of EVs, may play a key role in Stx-mediated renal injury. To test this hypothesis, we isolated exosomes from monocyte-derived macrophages in the presence of Stx2a or Stx2 toxoids. Macrophage-like differentiated THP-1 cells treated with Stxs secreted Stx-associated exosomes (Stx-Exo) of 90-130 nm in diameter, which induced cytotoxicity in recipient cells in a toxin receptor globotriaosylceramide (Gb3 )-dependent manner. Stx2-Exo engulfed by Gb3 -positive cells were translocated to the endoplasmic reticulum in the human proximal tubule epithelial cell line HK-2. Stx2-Exo contained pro-inflammatory cytokine mRNAs and proteins and induced more severe inflammation than purified Stx2a accompanied by greater death of target cells such as human renal or retinal pigment epithelial cells. Blockade of exosome biogenesis using the pharmacological inhibitor GW4869 reduced Stx2-Exo-mediated human renal cell death. Stx2-Exo isolated from human primary monocyte-derived macrophages activated caspase 3/7 and resulted in significant cell death in primary human renal cortical epithelial cells. Based on these results, we speculate that Stx-containing exosomes derived from macrophages may exacerbate cytotoxicity and inflammation and trigger cell death in toxin-sensitive cells. Therapeutic interventions targeting Stx-containing exosomes may prevent or ameliorate Stx-mediated acute vascular dysfunction.
Collapse
Affiliation(s)
- Kyung-Soo Lee
- Environmental Diseases Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon, South Korea.,Department of Biomolecular Science, KRIBB School of Bioscience, Korea University of Science and Technology (UST), Daejeon, South Korea
| | - Jieun Lee
- Environmental Diseases Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon, South Korea
| | - Pureum Lee
- Environmental Diseases Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon, South Korea.,Department of Biomolecular Science, KRIBB School of Bioscience, Korea University of Science and Technology (UST), Daejeon, South Korea
| | - Chang-Ung Kim
- Infectious Disease Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon, South Korea
| | - Doo-Jin Kim
- Infectious Disease Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon, South Korea
| | - Yu-Jin Jeong
- Environmental Diseases Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon, South Korea
| | - Young-Jun Park
- Environmental Diseases Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon, South Korea.,Department of Biomolecular Science, KRIBB School of Bioscience, Korea University of Science and Technology (UST), Daejeon, South Korea
| | - Vernon L Tesh
- Department of Microbial Pathogenesis and Immunology, Texas A&M University, College of Medicine, Bryan, Texas, USA
| | - Moo-Seung Lee
- Environmental Diseases Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon, South Korea.,Department of Biomolecular Science, KRIBB School of Bioscience, Korea University of Science and Technology (UST), Daejeon, South Korea
| |
Collapse
|
21
|
New opportunities and challenges of venom-based and bacteria-derived molecules for anticancer targeted therapy. Semin Cancer Biol 2020; 80:356-369. [PMID: 32846203 DOI: 10.1016/j.semcancer.2020.08.010] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2020] [Revised: 08/14/2020] [Accepted: 08/15/2020] [Indexed: 12/24/2022]
Abstract
Due to advances in detection and treatment of cancer, especially the rise in the targeted therapy, the five-year relative survival rate of all cancers has increased significantly. However, according to the analysis of the survival rate of cancer patients in 2019, the survival rate of most cancers is still less than five years. Therefore, to combat complex cancer and further improve the 5-year survival rate of cancer patients, it is necessary to develop some new anticancer drugs. Because of the adaptive evolution of toxic species for millions of years, the venom sac is a "treasure bank", which has millions of biomolecules with high affinity and stability awaiting further development. Complete utilization of venom-based and bacteria-derived drugs in the market is still staggering because of incomplete understanding regarding their mode of action. In this review, we focused on the currently identified targets for anticancer effects based on venomous and bacterial biomolecules, such as ion channels, membrane non-receptor molecules, integrins, and other related target molecules. This review will serve as the key for exploring the molecular mechanisms behind the anticancer potential of venom-based and bacteria-derived drugs and will also lay the path for the development of anticancer targeted therapy.
Collapse
|
22
|
Valid Presumption of Shiga Toxin-Mediated Damage of Developing Erythrocytes in EHEC-Associated Hemolytic Uremic Syndrome. Toxins (Basel) 2020; 12:toxins12060373. [PMID: 32512916 PMCID: PMC7354503 DOI: 10.3390/toxins12060373] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2020] [Revised: 06/02/2020] [Accepted: 06/03/2020] [Indexed: 02/06/2023] Open
Abstract
The global emergence of clinical diseases caused by enterohemorrhagic Escherichia coli (EHEC) is an issue of great concern. EHEC release Shiga toxins (Stxs) as their key virulence factors, and investigations on the cell-damaging mechanisms toward target cells are inevitable for the development of novel mitigation strategies. Stx-mediated hemolytic uremic syndrome (HUS), characterized by the triad of microangiopathic hemolytic anemia, thrombocytopenia, and acute renal injury, is the most severe outcome of an EHEC infection. Hemolytic anemia during HUS is defined as the loss of erythrocytes by mechanical disruption when passing through narrowed microvessels. The formation of thrombi in the microvasculature is considered an indirect effect of Stx-mediated injury mainly of the renal microvascular endothelial cells, resulting in obstructions of vessels. In this review, we summarize and discuss recent data providing evidence that HUS-associated hemolytic anemia may arise not only from intravascular rupture of erythrocytes, but also from the extravascular impairment of erythropoiesis, the development of red blood cells in the bone marrow, via direct Stx-mediated damage of maturing erythrocytes, leading to “non-hemolytic” anemia.
Collapse
|
23
|
Shi J, Peng D, Zhang F, Ruan L, Sun M. The Caenorhabditis elegans CUB-like-domain containing protein RBT-1 functions as a receptor for Bacillus thuringiensis Cry6Aa toxin. PLoS Pathog 2020; 16:e1008501. [PMID: 32369532 PMCID: PMC7228132 DOI: 10.1371/journal.ppat.1008501] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2019] [Revised: 05/15/2020] [Accepted: 03/26/2020] [Indexed: 12/16/2022] Open
Abstract
Plant-parasitic nematodes cause huge agricultural economic losses. Two major families of Bacillus thuringiensis crystal proteins, Cry5 and Cry6, show nematicidal activity. Previous work showed that binding to midgut receptors is a limiting step in Cry toxin mode of action. In the case of Cry5Ba, certain Caenorhabditis elegans glycolipids were identified as receptors of this toxin. However, the receptors for Cry6 toxin remain unknown. In this study, the C. elegans CUB-like-domain containing protein RBT-1, released by phosphatidylinositol-specific phospholipase C (PI-PLC), was identified as a Cry6Aa binding protein by affinity chromatography. RBT-1 contained a predicted glycosylphosphatidylinositol (GPI) anchor site and was shown to locate in lipid rafts in the surface of the midgut cells. Western ligand blot assays and ELISA binding analysis confirmed the binding interaction between Cry6Aa and RBT-1 showing high affinity and specificity. In addition, the mutation of rbt-1 gene decreased the susceptibility of C. elegans to Cry6Aa but not that of Cry5Ba. Furthermore, RBT-1 mediated the uptake of Cry6Aa into C. elegans gut cells, and was shown to be involved in triggering pore-formation activity, indicating that RBT-1 is required for the interaction of Cry6Aa with the nematode midgut cells. These results support that RBT-1 is a functional receptor for Cry6Aa. Bacillus thuringiensis (Bt) crystal proteins belong to pore-forming toxins (PFTs), which display virulence against target hosts by forming holes in the cell membrane. Cry6A is a nematicidal PFT, which exhibits unique protein structure and different mode of action than Cry5B, another nematicidal PFT. However, little is known about the mode of action of Cry6A. Although an intracellular nematicidal necrosis pathway of Cry6A was reported, its extracellular mode of action remains unknown. We here demonstrate that the CUB-like-domain containing protein RBT-1 acts as a functional receptor of Cry6A, which mediates the intestinal cell interaction and nematicidal activity of this toxin. RBT-1 represents a new class of crystal protein receptors. RBT-1 is dispensable for Cry5B toxicity against nematodes, consistent with that Cry6A and Cry5B have different nematicidal mechanisms. We also find that Cry6A kills nematodes by complex mechanism since rbt-1 mutation did not affect Cry6A-mediated necrosis signaling pathway. This work not only enhances the understanding of Bt crystal protein-nematode mechanism, but is also in favor for the application of Cry6A in nematode control.
Collapse
Affiliation(s)
- Jianwei Shi
- State Key Laboratory of Agricultural Microbiology, College of Life Science and Technology, Huazhong Agricultural University, Wuhan, China
| | - Donghai Peng
- State Key Laboratory of Agricultural Microbiology, College of Life Science and Technology, Huazhong Agricultural University, Wuhan, China
- * E-mail: (DP); (MS)
| | - Fengjuan Zhang
- State Key Laboratory of Agricultural Microbiology, College of Life Science and Technology, Huazhong Agricultural University, Wuhan, China
| | - Lifang Ruan
- State Key Laboratory of Agricultural Microbiology, College of Life Science and Technology, Huazhong Agricultural University, Wuhan, China
| | - Ming Sun
- State Key Laboratory of Agricultural Microbiology, College of Life Science and Technology, Huazhong Agricultural University, Wuhan, China
- * E-mail: (DP); (MS)
| |
Collapse
|
24
|
Abstract
The thrombotic microangiopathies (TMAs) are a group of diseases characterised by microangiopathic haemolysis, thrombocytopenia, and thrombus formation leading to tissue injury. Traditionally, TMAs have been classified as either thrombotic thrombocytopenic purpura (TTP) or haemolytic uremic syndrome (HUS) based on the clinical presentation, with neurological involvement predominating in the former and acute kidney injury in the latter. However, as our understanding of the pathogenesis of these conditions has increased, it has become clear that this is an over-simplification; there is significant overlap in the clinical presentation of TTP and HUS, there are different forms of HUS, and TMAs can occur in other, diverse clinical scenarios. This review will discuss recent developments in the diagnosis of HUS, focusing on the different forms of HUS and how to diagnose and manage these potentially life-threatening diseases.
Collapse
Affiliation(s)
- Neil S Sheerin
- National Renal Complement Therapeutics Centre, Institute of Cellular Medicine, Newcastle University and Biomedical Research Centre, Newcastle-upon-Tyne NHS Foundation Trust, Newcastle-upon-Tyne, UK
| | - Emily Glover
- National Renal Complement Therapeutics Centre, Institute of Cellular Medicine, Newcastle University and Biomedical Research Centre, Newcastle-upon-Tyne NHS Foundation Trust, Newcastle-upon-Tyne, UK
| |
Collapse
|
25
|
Nowakowska-Gołacka J, Sominka H, Sowa-Rogozińska N, Słomińska-Wojewódzka M. Toxins Utilize the Endoplasmic Reticulum-Associated Protein Degradation Pathway in Their Intoxication Process. Int J Mol Sci 2019; 20:E1307. [PMID: 30875878 PMCID: PMC6471375 DOI: 10.3390/ijms20061307] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2019] [Revised: 03/08/2019] [Accepted: 03/10/2019] [Indexed: 12/25/2022] Open
Abstract
Several bacterial and plant AB-toxins are delivered by retrograde vesicular transport to the endoplasmic reticulum (ER), where the enzymatically active A subunit is disassembled from the holotoxin and transported to the cytosol. In this process, toxins subvert the ER-associated degradation (ERAD) pathway. ERAD is an important part of cellular regulatory mechanism that targets misfolded proteins to the ER channels, prior to their retrotranslocation to the cytosol, ubiquitination and subsequent degradation by a protein-degrading complex, the proteasome. In this article, we present an overview of current understanding of the ERAD-dependent transport of AB-toxins to the cytosol. We describe important components of ERAD and discuss their significance for toxin transport. Toxin recognition and disassembly in the ER, transport through ER translocons and finally cytosolic events that instead of overall proteasomal degradation provide proper folding and cytotoxic activity of AB-toxins are discussed as well. We also comment on recent reports presenting medical applications for toxin transport through the ER channels.
Collapse
Affiliation(s)
- Jowita Nowakowska-Gołacka
- Department of Medical Biology and Genetics, Faculty of Biology, University of Gdańsk, Wita Stwosza 59, 80-308 Gdańsk, Poland.
| | - Hanna Sominka
- Department of Medical Biology and Genetics, Faculty of Biology, University of Gdańsk, Wita Stwosza 59, 80-308 Gdańsk, Poland.
| | - Natalia Sowa-Rogozińska
- Department of Medical Biology and Genetics, Faculty of Biology, University of Gdańsk, Wita Stwosza 59, 80-308 Gdańsk, Poland.
| | - Monika Słomińska-Wojewódzka
- Department of Medical Biology and Genetics, Faculty of Biology, University of Gdańsk, Wita Stwosza 59, 80-308 Gdańsk, Poland.
| |
Collapse
|
26
|
Bokori-Brown M, Metz J, Petrov PG, Mussai F, De Santo C, Smart NJ, Saunders S, Knight B, Pastan I, Titball RW, Winlove CP. Interactions Between Pseudomonas Immunotoxins and the Plasma Membrane: Implications for CAT-8015 Immunotoxin Therapy. Front Oncol 2018; 8:553. [PMID: 30538953 PMCID: PMC6277520 DOI: 10.3389/fonc.2018.00553] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2018] [Accepted: 11/08/2018] [Indexed: 11/13/2022] Open
Abstract
Acute Lymphoblastic Leukemia (ALL) remains the most frequent cause of cancer-related mortality in children and novel therapies are needed for the treatment of relapsed/refractory childhood ALL. One approach is the targeting of ALL blasts with the Pseudomonas immunotoxin CAT-8015. Although CAT-8015 has potent anti-leukemia activity, with a 32% objective response rate in a phase 1 study of childhood ALL, haemolytic-uremic syndrome (HUS) and vascular leak syndrome (VLS), major dose-limiting toxicities, have limited the use of this therapeutic approach in children. Investigations into the pathogenesis of CAT-8015-induced HUS/VLS are hindered by the lack of an adequate model system that replicates clinical manifestations, but damage to vascular endothelial cells (ECs) and blood cells are believed to be major initiating factors in both syndromes. Since there is little evidence that murine models replicate human HUS/VLS, and CAT-8015-induced HUS/VLS predominantly affects children, we developed human models and used novel methodologies to investigate CAT-8015 interactions with red blood cells (RBCs) from pediatric ALL patients and ECs of excised human mesenteric arteries. We provide evidence that CAT-8015 directly interacts with RBCs, mediated by Pseudomonas toxin. We also show correlation between the electrical properties of the RBC membrane and RBC susceptibility to CAT-8015-induced lysis, which may have clinical implication. Finally, we provide evidence that CAT-8015 is directly cytototoxic to ECs of excised human mesenteric arteries. In conclusion, the human models we developed constitutes the first, and very important, step in understanding the origins of HUS/VLS in immunotoxin therapy and will allow further investigations of HUS/VLS pathogenesis.
Collapse
Affiliation(s)
- Monika Bokori-Brown
- College of Life and Environmental Sciences, School of Biosciences, University of Exeter, Exeter, United Kingdom
| | - Jeremy Metz
- College of Life and Environmental Sciences, School of Biosciences, University of Exeter, Exeter, United Kingdom
| | - Peter G. Petrov
- College of Engineering, Mathematics and Physical Sciences, Department of Physics and Astronomy, University of Exeter, Exeter, United Kingdom
| | - Francis Mussai
- Institute of Immunology and Immunotherapy, College of Medical and Dental Sciences, University of Birmingham, Birmingham, United Kingdom
| | - Carmela De Santo
- Institute of Immunology and Immunotherapy, College of Medical and Dental Sciences, University of Birmingham, Birmingham, United Kingdom
| | - Neil J. Smart
- Exeter Surgical Health Services Research Unit, Royal Devon and Exeter Hospital, Exeter, United Kingdom
| | - Sarah Saunders
- Histopathology Department, Royal Devon and Exeter Hospital, Exeter, United Kingdom
| | - Bridget Knight
- National Institute for Health Research Exeter Clinical Research Facility, Royal Devon and Exeter National Health Service Foundation Trust, Exeter, United Kingdom
| | - Ira Pastan
- Laboratory of Molecular Biology, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, MD, United States
| | - Richard W. Titball
- College of Life and Environmental Sciences, School of Biosciences, University of Exeter, Exeter, United Kingdom
| | - C. Peter Winlove
- College of Engineering, Mathematics and Physical Sciences, Department of Physics and Astronomy, University of Exeter, Exeter, United Kingdom
| |
Collapse
|
27
|
Mao Y, Feng S, Li S, Zhao Q, Di D, Liu Y, Wang S. Chylomicron-pretended nano-bio self-assembling vehicle to promote lymphatic transport and GALTs target of oral drugs. Biomaterials 2018; 188:173-186. [PMID: 30359884 DOI: 10.1016/j.biomaterials.2018.10.012] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2018] [Revised: 10/09/2018] [Accepted: 10/10/2018] [Indexed: 01/05/2023]
Abstract
Lymphatic transport of oral drugs allows extraordinary gains in bioavailability and efficacy through avoidance of first-pass hepatic metabolism and preservation of drugs at lymphatic tissues against lymph-mediated diseases. Chylomicrons can transport dietary lipids absorbed from the intestine to the tissues through lymphatic circulation. Herein, we engineered for the first time a chylomicron-pretended mesoporous silica nanocarrier that utilizes the digestion, re-esterification, and lymphatic transport process of dietary triglyceride to promote lymphatic transport of oral drugs. Taking lopinavir (LNV) as a model antiretroviral drug with disadvantages such as poor solubility, high first-pass effect and off-target deposition, this vehicle exhibited several properties belonging to ideal nanocarriers, including high drug load, amorphous dispersion and controlled release in the gastrointestinal tract. Additionally, a nano-bio interaction was demonstrated between nanoparticles and a key protein involved in chylomicron assembly; this biochemical reaction in cellular was utilized for the first time to promote lymphatic transport of nanocarriers for oral delivery. As a result, the chylomicron-pretended nanocarrier afforded 10.6-fold higher oral bioavailability compared with free LNV and effectively delivered LNV to gut-associated lymphoid tissues, where HIV persists and actively evolves. This approach not only promises a potential application to HIV-infected individuals but also opens a new avenue to other lymph-mediated pathologies such as autoimmune diseases and lymphatic tumor metastasis.
Collapse
Affiliation(s)
- Yuling Mao
- Department of Pharmaceutics, School of Pharmacy, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang, Liaoning Province 110016, PR China
| | - Shuang Feng
- Department of Pharmaceutics, School of Pharmacy, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang, Liaoning Province 110016, PR China
| | - Shuai Li
- School of Life Science and Bio-pharmaceutics, Shenyang Pharmaceutical University, Shenyang, Liaoning Province 110016, PR China
| | - Qinfu Zhao
- Department of Pharmaceutics, School of Pharmacy, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang, Liaoning Province 110016, PR China
| | - Donghua Di
- School of Pharmaceutical Engineering, Shenyang Pharmaceutical University, PR China
| | - Yanfeng Liu
- School of Life Science and Bio-pharmaceutics, Shenyang Pharmaceutical University, Shenyang, Liaoning Province 110016, PR China
| | - Siling Wang
- Department of Pharmaceutics, School of Pharmacy, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang, Liaoning Province 110016, PR China.
| |
Collapse
|
28
|
Legros N, Pohlentz G, Steil D, Müthing J. Shiga toxin-glycosphingolipid interaction: Status quo of research with focus on primary human brain and kidney endothelial cells. Int J Med Microbiol 2018; 308:1073-1084. [PMID: 30224239 DOI: 10.1016/j.ijmm.2018.09.003] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2018] [Revised: 08/28/2018] [Accepted: 09/06/2018] [Indexed: 12/21/2022] Open
Abstract
Shiga toxin (Stx)-mediated injury of the kidneys and the brain represent the major extraintestinal complications in humans upon infection by enterohemorrhagic Escherichia coli (EHEC). Damage of renal and cerebral endothelial cells is the key event in the pathogenesis of the life-threatening hemolytic uremic syndrome (HUS). Stxs are AB5 toxins and the B-pentamers of the two clinically important Stx subtypes Stx1a and Stx2a preferentially bind to the glycosphingolipid globotriaosylceramide (Gb3Cer, Galα4Galβ4Glcβ1Cer) and to less extent to globotetraosylceramide (Gb4Cer, GalNAcβ3Galα4Galβ4Glcβ1), which are expected to reside in lipid rafts in the plasma membrane of the human endothelium. This review summarizes the current knowledge on the Stx glycosphingolipid receptors and their lipid membrane ensemble in primary human brain microvascular endothelial cells (pHBMECs) and primary human renal glomerular endothelial cells (pHRGECs). Increasing knowledge on the precise initial molecular mechanisms by which Stxs interact with cellular targets will help to develop specific therapeutics and/or preventive measures to combat EHEC-caused diseases.
Collapse
Affiliation(s)
- Nadine Legros
- Institute for Hygiene, University of Münster, D-48149 Münster, Germany
| | | | - Daniel Steil
- Institute for Hygiene, University of Münster, D-48149 Münster, Germany
| | - Johannes Müthing
- Institute for Hygiene, University of Münster, D-48149 Münster, Germany; Interdisciplinary Center for Clinical Research (IZKF), University of Münster, D-48149 Münster, Germany.
| |
Collapse
|
29
|
Steil D, Pohlentz G, Legros N, Mormann M, Mellmann A, Karch H, Müthing J. Combining Mass Spectrometry, Surface Acoustic Wave Interaction Analysis, and Cell Viability Assays for Characterization of Shiga Toxin Subtypes of Pathogenic Escherichia coli Bacteria. Anal Chem 2018; 90:8989-8997. [PMID: 29939014 DOI: 10.1021/acs.analchem.8b01189] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Shiga toxin (Stx)-producing Escherichia coli (STEC) and enterohemorrhagic E. coli (EHEC) as a human pathogenic subgroup of STEC are characterized by releasing Stx AB5-toxin as the major virulence factor. Worldwide disseminated EHEC strains cause sporadic infections and outbreaks in the human population and swine pathogenic STEC strains represent greatly feared pathogens in pig breeding and fattening plants. Among the various Stx subtypes, Stx1a and Stx2a are of eminent clinical importance in human infections being associated with life-threatening hemorrhagic colitis and hemolytic uremic syndrome, whereas Stx2e subtype is associated with porcine edema disease with a generalized fatal outcome for the animals. Binding toward the glycosphingolipid globotriaosylceramide (Gb3Cer) is a common feature of all Stx subtypes analyzed so far. Here, we report on the development of a matched strategy combining (i) miniaturized one-step affinity purification of native Stx subtypes from culture supernatant of bacterial wild-type strains using Gb3-functionalized magnetic beads, (ii) structural analysis and identification of Stx holotoxins by electrospray ionization ion mobility mass spectrometry (ESI MS), (iii) functional Stx-receptor real-time interaction analysis employing the surface acoustic wave (SAW) technology, and (iv) Vero cell culture assays for determining Stx-caused cytotoxic effects. Structural investigations revealed diagnostic tryptic peptide ions for purified Stx1a, Stx2a, and Stx2e, respectively, and functional analysis resulted in characteristic binding kinetics of each Stx subtype. Cytotoxicity studies revealed differing toxin-mediated cell damage ranked with Stx1a > Stx2a > Stx2e. Collectively, this matched procedure represents a promising clinical application for the characterization of life-endangering Stx subtypes at the protein level.
Collapse
Affiliation(s)
- Daniel Steil
- Institute for Hygiene , University of Münster , Robert-Koch-Strasse 41 , D-48149 Münster , Germany
| | - Gottfried Pohlentz
- Institute for Hygiene , University of Münster , Robert-Koch-Strasse 41 , D-48149 Münster , Germany
| | - Nadine Legros
- Institute for Hygiene , University of Münster , Robert-Koch-Strasse 41 , D-48149 Münster , Germany
| | - Michael Mormann
- Institute for Hygiene , University of Münster , Robert-Koch-Strasse 41 , D-48149 Münster , Germany
| | - Alexander Mellmann
- Institute for Hygiene , University of Münster , Robert-Koch-Strasse 41 , D-48149 Münster , Germany.,Interdisciplinary Center for Clinical Research (IZKF) Münster , Domagkstrasse 3 , D-48149 Münster , Germany
| | - Helge Karch
- Institute for Hygiene , University of Münster , Robert-Koch-Strasse 41 , D-48149 Münster , Germany.,Interdisciplinary Center for Clinical Research (IZKF) Münster , Domagkstrasse 3 , D-48149 Münster , Germany
| | - Johannes Müthing
- Institute for Hygiene , University of Münster , Robert-Koch-Strasse 41 , D-48149 Münster , Germany.,Interdisciplinary Center for Clinical Research (IZKF) Münster , Domagkstrasse 3 , D-48149 Münster , Germany
| |
Collapse
|
30
|
Legros N, Pohlentz G, Steil D, Kouzel IU, Liashkovich I, Mellmann A, Karch H, Müthing J. Membrane assembly of Shiga toxin glycosphingolipid receptors and toxin refractiveness of MDCK II epithelial cells. J Lipid Res 2018; 59:1383-1401. [PMID: 29866658 DOI: 10.1194/jlr.m083048] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2017] [Revised: 05/22/2018] [Indexed: 12/16/2022] Open
Abstract
Shiga toxins (Stxs) are the major virulence factors of Stx-producing Escherichia coli (STEC), which cause hemorrhagic colitis and severe extraintestinal complications due to injury of renal endothelial cells, resulting in kidney failure. Since kidney epithelial cells are suggested additional targets for Stxs, we analyzed Madin-Darby canine kidney (MDCK) II epithelial cells for presence of Stx-binding glycosphingolipids (GSLs), determined their distribution to detergent-resistant membranes (DRMs), and ascertained the lipid composition of DRM and non-DRM preparations. Globotriaosylceramide and globotetraosylceramide, known as receptors for Stx1a, Stx2a, and Stx2e, and Forssman GSL as a specific receptor for Stx2e, were found to cooccur with SM and cholesterol in DRMs of MDCK II cells, which was shown using TLC overlay assay detection combined with mass spectrometry. The various lipoforms of GSLs were found to mainly harbor ceramide moieties composed of sphingosine (d18:1) and C24:1/C24:0 or C16:0 FA. The cells were highly refractory toward Stx1a, Stx2a, and Stx2e, most likely due to the absence of Stx-binding GSLs in the apical plasma membrane determined by immunofluorescence confocal laser scanning microscopy. The results suggest that the cellular content of Stx receptor GSLs and their biochemical detection in DRM preparations alone are inadequate to predict cellular sensitivity toward Stxs.
Collapse
Affiliation(s)
- Nadine Legros
- Institute for Hygiene, University of Münster, D-48149 Münster, Germany
| | | | - Daniel Steil
- Institute for Hygiene, University of Münster, D-48149 Münster, Germany
| | - Ivan U Kouzel
- Institute for Hygiene, University of Münster, D-48149 Münster, Germany.,Interdisciplinary Center for Clinical Research, University of Münster, D-48149 Münster, Germany
| | - Ivan Liashkovich
- Institute of Physiology II, University of Münster, D-48149 Münster, Germany
| | - Alexander Mellmann
- Institute for Hygiene, University of Münster, D-48149 Münster, Germany.,Interdisciplinary Center for Clinical Research, University of Münster, D-48149 Münster, Germany
| | - Helge Karch
- Institute for Hygiene, University of Münster, D-48149 Münster, Germany.,Interdisciplinary Center for Clinical Research, University of Münster, D-48149 Münster, Germany
| | - Johannes Müthing
- Institute for Hygiene, University of Münster, D-48149 Münster, Germany .,Interdisciplinary Center for Clinical Research, University of Münster, D-48149 Münster, Germany
| |
Collapse
|
31
|
Muhammad SA, Guo J, Nguyen TM, Wu X, Bai B, Yang XF, Chen JY. Simulation Study of cDNA Dataset to Investigate Possible Association of Differentially Expressed Genes of Human THP1-Monocytic Cells in Cancer Progression Affected by Bacterial Shiga Toxins. Front Microbiol 2018; 9:380. [PMID: 29593668 PMCID: PMC5859033 DOI: 10.3389/fmicb.2018.00380] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2018] [Accepted: 02/20/2018] [Indexed: 12/30/2022] Open
Abstract
Shiga toxin (Stxs) is a family of structurally and functionally related bacterial cytotoxins produced by Shigella dysenteriae serotype 1 and shigatoxigenic group of Escherichia coli that cause shigellosis and hemorrhagic colitis, respectively. Until recently, it has been thought that Stxs only inhibits the protein synthesis and induces expression to a limited number of genes in host cells, but recent data showed that Stxs can trigger several signaling pathways in mammalian cells and activate cell cycle and apoptosis. To explore the changes in gene expression induced by Stxs that have been shown in other systems to correlate with cancer progression, we performed the simulated analysis of cDNA dataset and found differentially expressed genes (DEGs) of human THP1-monocytic cells treated with Stxs. In this study, the entire data (treated and untreated replicates) was analyzed by statistical algorithms implemented in Bioconductor packages. The output data was validated by the k-fold cross technique using generalized linear Gaussian models. A total of 50 DEGs were identified. 7 genes including TSLP, IL6, GBP1, CD274, TNFSF13B, OASL, and PNPLA3 were considerably (<0.00005) related to cancer proliferation. The functional enrichment analysis showed 6 down-regulated and 1 up-regulated genes. Among these DEGs, IL6 was associated with several cancers, especially with leukemia, lymphoma, lungs, liver and breast cancers. The predicted regulatory motifs of these genes include conserved RELA, STATI, IRFI, NF-kappaB, PEND, HLF, REL, CEBPA, DI_2, and NFKB1 transcription factor binding sites (TFBS) involved in the complex biological functions. Thus, our findings suggest that Stxs has the potential as a valuable tool for better understanding of treatment strategies for several cancers.
Collapse
Affiliation(s)
- Syed A Muhammad
- Institute of Biopharmaceutical Informatics and Technologies, Wenzhou Medical University, Wenzhou, China.,Wenzhou Medical University 1st Affiliated Hospital, Wenzhou, China.,Institute of Molecular Biology and Biotechnology, Bahauddin Zakariya University, Multan, Pakistan
| | - Jinlei Guo
- Institute of Biopharmaceutical Informatics and Technologies, Wenzhou Medical University, Wenzhou, China.,Wenzhou Medical University 1st Affiliated Hospital, Wenzhou, China
| | - Thanh M Nguyen
- Institute of Biopharmaceutical Informatics and Technologies, Wenzhou Medical University, Wenzhou, China.,Wenzhou Medical University 1st Affiliated Hospital, Wenzhou, China.,Department of Computer and Information Science, Purdue University Indianapolis, Indianapolis, IN, United States
| | - Xiaogang Wu
- Institute for Systems Biology, Seattle, WA, United States
| | - Baogang Bai
- Institute of Biopharmaceutical Informatics and Technologies, Wenzhou Medical University, Wenzhou, China
| | - X Frank Yang
- Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Jake Y Chen
- Informatics Institute, School of Medicine, The University of Alabama at Birmingham, Birmingham, AL, United States
| |
Collapse
|
32
|
Huwiler A, Pfeilschifter J. Sphingolipid signaling in renal fibrosis. Matrix Biol 2018; 68-69:230-247. [PMID: 29343457 DOI: 10.1016/j.matbio.2018.01.006] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2017] [Revised: 01/08/2018] [Accepted: 01/08/2018] [Indexed: 12/28/2022]
Abstract
Over the last decade, various sphingolipid subspecies have gained increasing attention as important signaling molecules that regulate a multitude of physiological and pathophysiological processes including inflammation and tissue remodeling. These mediators include ceramide, sphingosine 1-phosphate (S1P), the cerebroside glucosylceramide, lactosylceramide, and the gangliosides GM3 and Gb3. These lipids have been shown to accumulate in various chronic kidney diseases that typically end in renal fibrosis and ultimately renal failure. This review will summarize the effects and contributions of those enzymes that regulate the generation and interconversion of these lipids, notably the acid sphingomyelinase, the acid sphingomyelinase-like protein SMPDL3B, the sphingosine kinases, the S1P lyase, the glucosylceramide synthase, the GM3 synthase, and the α-galactosidase A, to renal fibrotic diseases. Strategies of manipulating these enzymes for therapeutic purposes and the impact of existing drugs on renal pathologies will be discussed.
Collapse
Affiliation(s)
- Andrea Huwiler
- Institute of Pharmacology, University of Bern, Inselspital INO-F, CH-3010 Bern, Switzerland.
| | - Josef Pfeilschifter
- Institute of General Pharmacology and Toxicology, University Hospital Frankfurt, Goethe- University, Frankfurt am Main, Germany
| |
Collapse
|
33
|
Kampmeier S, Berger M, Mellmann A, Karch H, Berger P. The 2011 German Enterohemorrhagic Escherichia Coli O104:H4 Outbreak-The Danger Is Still Out There. Curr Top Microbiol Immunol 2018; 416:117-148. [PMID: 30062592 DOI: 10.1007/82_2018_107] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Enterohemorrhagic Escherichia coli (EHEC) are Shiga toxin (Stx) producing bacteria causing a disease characterized by bloody (or non-bloody) diarrhea, which might progress to hemolytic uremic syndrome (HUS). EHEC O104:H4 caused the largest ever recorded EHEC outbreak in Germany in 2011, which in addition showed the so far highest incidence rate of EHEC-related HUS worldwide. The aggressive outbreak strain carries an unusual combination of virulence traits characteristic to both EHEC-a chromosomally integrated Stx-encoding bacteriophage, and enteroaggregative Escherichia coli-pAA plasmid-encoded aggregative adherence fimbriae mediating its tight adhesion to epithelia cells. There are currently still open questions regarding the 2011 EHEC outbreak, e.g., with respect to the exact molecular mechanisms resulting in the hypervirulence of the strain, the natural reservoir of EHEC O104:H4, and suitable therapeutic strategies. Nevertheless, our knowledge on these issues has substantially expanded since 2011. Here, we present an overview of the epidemiological, clinical, microbiological, and molecular biological data available on the 2011 German EHEC O104:H4 outbreak.
Collapse
Affiliation(s)
| | - Michael Berger
- Institute of Hygiene, University of Münster, Münster, Germany
| | | | - Helge Karch
- Institute of Hygiene, University of Münster, Münster, Germany
| | - Petya Berger
- Institute of Hygiene, University of Münster, Münster, Germany.
| |
Collapse
|
34
|
Shiga Toxin Glycosphingolipid Receptors in Human Caco-2 and HCT-8 Colon Epithelial Cell Lines. Toxins (Basel) 2017; 9:toxins9110338. [PMID: 29068380 PMCID: PMC5705953 DOI: 10.3390/toxins9110338] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2017] [Revised: 10/11/2017] [Accepted: 10/19/2017] [Indexed: 12/21/2022] Open
Abstract
Shiga toxins (Stxs) released by enterohemorrhagic Escherichia coli (EHEC) into the human colon are the causative agents for fatal outcome of EHEC infections. Colon epithelial Caco-2 and HCT-8 cells are widely used for investigating Stx-mediated intestinal cytotoxicity. Only limited data are available regarding precise structures of their Stx receptor glycosphingolipids (GSLs) globotriaosylceramide (Gb3Cer) and globotetraosylceramide (Gb4Cer), and lipid raft association. In this study we identified Gb3Cer and Gb4Cer lipoforms of serum-free cultivated Caco-2 and HCT-8 cells, chiefly harboring ceramide moieties composed of sphingosine (d18:1) and C16:0, C22:0 or C24:0/C24:1 fatty acid. The most significant difference between the two cell lines was the prevalence of Gb3Cer with C16 fatty acid in HCT-8 and Gb4Cer with C22–C24 fatty acids in Caco-2 cells. Lipid compositional analysis of detergent-resistant membranes (DRMs), which were used as lipid raft-equivalents, indicated slightly higher relative content of Stx receptor Gb3Cer in DRMs of HCT-8 cells when compared to Caco-2 cells. Cytotoxicity assays revealed substantial sensitivity towards Stx2a for both cell lines, evidencing little higher susceptibility of Caco-2 cells versus HCT-8 cells. Collectively, Caco-2 and HCT-8 cells express a plethora of different receptor lipoforms and are susceptible towards Stx2a exhibiting somewhat lower sensitivity when compared to Vero cells.
Collapse
|
35
|
Hall G, Kurosawa S, Stearns-Kurosawa DJ. Shiga Toxin Therapeutics: Beyond Neutralization. Toxins (Basel) 2017; 9:toxins9090291. [PMID: 28925976 PMCID: PMC5618224 DOI: 10.3390/toxins9090291] [Citation(s) in RCA: 20] [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: 08/25/2017] [Revised: 09/15/2017] [Accepted: 09/15/2017] [Indexed: 01/04/2023] Open
Abstract
Ribotoxic Shiga toxins are the primary cause of hemolytic uremic syndrome (HUS) in patients infected with Shiga toxin-producing enterohemorrhagic Escherichia coli (STEC), a pathogen class responsible for epidemic outbreaks of gastrointestinal disease around the globe. HUS is a leading cause of pediatric renal failure in otherwise healthy children, resulting in a mortality rate of 10% and a chronic morbidity rate near 25%. There are currently no available therapeutics to prevent or treat HUS in STEC patients despite decades of work elucidating the mechanisms of Shiga toxicity in sensitive cells. The preclinical development of toxin-targeted HUS therapies has been hindered by the sporadic, geographically dispersed nature of STEC outbreaks with HUS cases and the limited financial incentive for the commercial development of therapies for an acute disease with an inconsistent patient population. The following review considers potential therapeutic targeting of the downstream cellular impacts of Shiga toxicity, which include the unfolded protein response (UPR) and the ribotoxic stress response (RSR). Outcomes of the UPR and RSR are relevant to other diseases with large global incidence and prevalence rates, thus reducing barriers to the development of commercial drugs that could improve STEC and HUS patient outcomes.
Collapse
Affiliation(s)
- Gregory Hall
- Department of Pathology and Laboratory Medicine, Boston University School of Medicine, Boston, MA 02118, USA.
| | - Shinichiro Kurosawa
- Department of Pathology and Laboratory Medicine, Boston University School of Medicine, Boston, MA 02118, USA.
| | - Deborah J Stearns-Kurosawa
- Department of Pathology and Laboratory Medicine, Boston University School of Medicine, Boston, MA 02118, USA.
| |
Collapse
|
36
|
El-Aouar Filho RA, Nicolas A, De Paula Castro TL, Deplanche M, De Carvalho Azevedo VA, Goossens PL, Taieb F, Lina G, Le Loir Y, Berkova N. Heterogeneous Family of Cyclomodulins: Smart Weapons That Allow Bacteria to Hijack the Eukaryotic Cell Cycle and Promote Infections. Front Cell Infect Microbiol 2017; 7:208. [PMID: 28589102 PMCID: PMC5440457 DOI: 10.3389/fcimb.2017.00208] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2017] [Accepted: 05/09/2017] [Indexed: 12/13/2022] Open
Abstract
Some bacterial pathogens modulate signaling pathways of eukaryotic cells in order to subvert the host response for their own benefit, leading to successful colonization and invasion. Pathogenic bacteria produce multiple compounds that generate favorable conditions to their survival and growth during infection in eukaryotic hosts. Many bacterial toxins can alter the cell cycle progression of host cells, impairing essential cellular functions and impeding host cell division. This review summarizes current knowledge regarding cyclomodulins, a heterogeneous family of bacterial effectors that induce eukaryotic cell cycle alterations. We discuss the mechanisms of actions of cyclomodulins according to their biochemical properties, providing examples of various cyclomodulins such as cycle inhibiting factor, γ-glutamyltranspeptidase, cytolethal distending toxins, shiga toxin, subtilase toxin, anthrax toxin, cholera toxin, adenylate cyclase toxins, vacuolating cytotoxin, cytotoxic necrotizing factor, Panton-Valentine leukocidin, phenol soluble modulins, and mycolactone. Special attention is paid to the benefit provided by cyclomodulins to bacteria during colonization of the host.
Collapse
Affiliation(s)
- Rachid A El-Aouar Filho
- STLO, Agrocampus Ouest Rennes, Institut National de la Recherche AgronomiqueRennes, France.,Departamento de Biologia Geral, Laboratório de Genética Celular e Molecular (LGCM), Instituto de Ciências Biológicas, Universidade Federal de Minas GeraisBelo Horizonte, Brazil
| | - Aurélie Nicolas
- STLO, Agrocampus Ouest Rennes, Institut National de la Recherche AgronomiqueRennes, France
| | - Thiago L De Paula Castro
- Departamento de Biologia Geral, Laboratório de Genética Celular e Molecular (LGCM), Instituto de Ciências Biológicas, Universidade Federal de Minas GeraisBelo Horizonte, Brazil
| | - Martine Deplanche
- STLO, Agrocampus Ouest Rennes, Institut National de la Recherche AgronomiqueRennes, France
| | - Vasco A De Carvalho Azevedo
- Departamento de Biologia Geral, Laboratório de Genética Celular e Molecular (LGCM), Instituto de Ciências Biológicas, Universidade Federal de Minas GeraisBelo Horizonte, Brazil
| | - Pierre L Goossens
- HistoPathologie et Modèles Animaux/Pathogénie des Toxi-Infections Bactériennes, Institut PasteurParis, France
| | - Frédéric Taieb
- CHU Purpan USC INRA 1360-CPTP, U1043 Institut National de la Santé et de la Recherche Médicale, Pathogénie Moléculaire et Cellulaire des Infections à Escherichia coliToulouse, France
| | - Gerard Lina
- International Center for Infectiology ResearchLyon, France.,Centre National de la Recherche Scientifique, UMR5308, Institut National de la Santé et de la Recherche Médicale U1111, Ecole Normale Supérieure de Lyon, Université Lyon 1Lyon, France.,Département de Biologie, Institut des Agents Infectieux, Hospices Civils de LyonLyon, France
| | - Yves Le Loir
- STLO, Agrocampus Ouest Rennes, Institut National de la Recherche AgronomiqueRennes, France
| | - Nadia Berkova
- STLO, Agrocampus Ouest Rennes, Institut National de la Recherche AgronomiqueRennes, France
| |
Collapse
|
37
|
Stolle AS, Norkowski S, Körner B, Schmitz J, Lüken L, Frankenberg M, Rüter C, Schmidt MA. T3SS-Independent Uptake of the Short-Trip Toxin-Related Recombinant NleC Effector of Enteropathogenic Escherichia coli Leads to NF-κB p65 Cleavage. Front Cell Infect Microbiol 2017; 7:119. [PMID: 28451521 PMCID: PMC5390045 DOI: 10.3389/fcimb.2017.00119] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2016] [Accepted: 03/23/2017] [Indexed: 12/15/2022] Open
Abstract
Effector proteins secreted by the type 3 secretion system (T3SS) of pathogenic bacteria have been shown to precisely modulate important signaling cascades of the host for the benefit of the pathogens. Among others, the non-LEE encoded T3SS effector protein NleC of enteropathogenic Escherichia coli (EPEC) is a Zn-dependent metalloprotease and suppresses innate immune responses by directly targeting the NF-κB signaling pathway. Many pathogenic bacteria release potent bacterial toxins of the A-B type, which-in contrast to the direct cytoplasmic injection of T3SS effector proteins-are released first into the environment. In this study, we found that NleC displays characteristics of bacterial A-B toxins, when applied to eukaryotic cells as a recombinant protein. Although lacking a B subunit, that typically mediates the uptake of toxins, recombinant NleC (rNleC) induces endocytosis via lipid rafts and follows the endosomal-lysosomal pathway. The conformation of rNleC is altered by low pH to facilitate its escape from acidified endosomes. This is reminiscent of the homologous A-B toxin AIP56 of the fish pathogen Photobacterium damselae piscicida (Phdp). The recombinant protease NleC is functional inside eukaryotic cells and cleaves p65 of the NF-κB pathway. Here, we describe the endocytic uptake mechanism of rNleC, characterize its intracellular trafficking and demonstrate that its specific activity of cleaving p65 requires activation of host cells e.g., by IL1β. Further, we propose an evolutionary link between some T3SS effector proteins and bacterial toxins from apparently unrelated bacteria. In summary, these properties might suggest rNleC as an interesting candidate for future applications as a potential therapeutic against immune disorders.
Collapse
Affiliation(s)
- Anne-Sophie Stolle
- Institute of Infectiology, Center for Molecular Biology of Inflammation, University of MünsterMünster, Germany
| | - Stefanie Norkowski
- Institute of Infectiology, Center for Molecular Biology of Inflammation, University of MünsterMünster, Germany
| | - Britta Körner
- Institute of Infectiology, Center for Molecular Biology of Inflammation, University of MünsterMünster, Germany
| | - Jürgen Schmitz
- Institute of Experimental Pathology, Center for Molecular Biology of Inflammation, University of MünsterMünster, Germany
| | - Lena Lüken
- Institute of Infectiology, Center for Molecular Biology of Inflammation, University of MünsterMünster, Germany
| | - Maj Frankenberg
- Institute of Infectiology, Center for Molecular Biology of Inflammation, University of MünsterMünster, Germany
| | - Christian Rüter
- Institute of Infectiology, Center for Molecular Biology of Inflammation, University of MünsterMünster, Germany
| | - M Alexander Schmidt
- Institute of Infectiology, Center for Molecular Biology of Inflammation, University of MünsterMünster, Germany
| |
Collapse
|
38
|
Adnan H, Zhang Z, Park HJ, Tailor C, Che C, Kamani M, Spitalny G, Binnington B, Lingwood C. Endoplasmic Reticulum-Targeted Subunit Toxins Provide a New Approach to Rescue Misfolded Mutant Proteins and Revert Cell Models of Genetic Diseases. PLoS One 2016; 11:e0166948. [PMID: 27935997 PMCID: PMC5147855 DOI: 10.1371/journal.pone.0166948] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2016] [Accepted: 11/06/2016] [Indexed: 01/08/2023] Open
Abstract
Many germ line diseases stem from a relatively minor disturbance in mutant protein endoplasmic reticulum (ER) 3D assembly. Chaperones are recruited which, on failure to correct folding, sort the mutant for retrotranslocation and cytosolic proteasomal degradation (ER-associated degradation-ERAD), to initiate/exacerbate deficiency-disease symptoms. Several bacterial (and plant) subunit toxins, retrograde transport to the ER after initial cell surface receptor binding/internalization. The A subunit has evolved to mimic a misfolded protein and hijack the ERAD membrane translocon (dislocon), to effect cytosolic access and cytopathology. We show such toxins compete for ERAD to rescue endogenous misfolded proteins. Cholera toxin or verotoxin (Shiga toxin) containing genetically inactivated (± an N-terminal polyleucine tail) A subunit can, within 2–4 hrs, temporarily increase F508delCFTR protein, the major cystic fibrosis (CF) mutant (5-10x), F508delCFTR Golgi maturation (<10x), cell surface expression (20x) and chloride transport (2x) in F508del CFTR transfected cells and patient-derived F508delCFTR bronchiolar epithelia, without apparent cytopathology. These toxoids also increase glucocerobrosidase (GCC) in N370SGCC Gaucher Disease fibroblasts (3x), another ERAD–exacerbated misfiling disease. We identify a new, potentially benign approach to the treatment of certain genetic protein misfolding diseases.
Collapse
Affiliation(s)
- Humaira Adnan
- Division of Molecular Structure and Function, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Zhenbo Zhang
- Division of Molecular Structure and Function, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Hyun-Joo Park
- Division of Molecular Structure and Function, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Chetankumar Tailor
- Division of Cell Biology, Research Institute, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Clare Che
- Division of Molecular Structure and Function, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Mustafa Kamani
- Division of Molecular Structure and Function, The Hospital for Sick Children, Toronto, Ontario, Canada
| | | | - Beth Binnington
- Division of Molecular Structure and Function, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Clifford Lingwood
- Division of Molecular Structure and Function, The Hospital for Sick Children, Toronto, Ontario, Canada
- Department of Biochemistry, University of Toronto, Ontario, Canada
- Department of Laboratory Medicine & Pathobiology, University of Toronto, Ontario, Canada
- * E-mail:
| |
Collapse
|
39
|
Steil D, Bonse R, Meisen I, Pohlentz G, Vallejo G, Karch H, Müthing J. A Topographical Atlas of Shiga Toxin 2e Receptor Distribution in the Tissues of Weaned Piglets. Toxins (Basel) 2016; 8:toxins8120357. [PMID: 27916888 PMCID: PMC5198551 DOI: 10.3390/toxins8120357] [Citation(s) in RCA: 15] [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: 10/21/2016] [Revised: 11/21/2016] [Accepted: 11/28/2016] [Indexed: 01/08/2023] Open
Abstract
Shiga toxin (Stx) 2e of Stx-producing Escherichia coli (STEC) is the primary virulence factor in the development of pig edema disease shortly after weaning. Stx2e binds to the globo-series glycosphingolipids (GSLs) globotriaosylceramide (Gb3Cer, Galα1-4Galβ1-4Glcβ1-1Cer) and globotetraosylceramide (Gb4Cer, GalNAcβ1-3Galα1-4Galβ1-4Glcβ1-1Cer), the latter acting as the preferential Stx2e receptor. We determined Stx receptor profiles of 25 different tissues of a male and a female weaned piglet using immunochemical solid phase binding assays combined with mass spectrometry. All probed tissues harbored GSL receptors, ranging from high (category I) over moderate (category II) to low content (category III). Examples of Gb4Cer expression in category I tissues are small intestinal ileum, kidney pelvis and whole blood, followed by colon, small intestinal duodenum and jejunum belonging to category II, and kidney cortex, cerebrum and cerebellum as members of category III organs holding true for both genders. Dominant Gb3Cer and Gb4Cer lipoforms were those with ceramides carrying constant sphingosine (d18:1) and a variable C16:0, C22:0 or C24:1/C24:0 fatty acid. From the mapping data, we created a topographical atlas for Stx2e receptors in piglet tissues and organs, which might be helpful to further investigations on the molecular and cellular mechanisms that underlie infections of Stx2e-producing STEC in pigs and their zoonotic potential for humans.
Collapse
Affiliation(s)
- Daniel Steil
- Institute for Hygiene, University of Münster, D-48149 Münster, Germany.
| | - Robert Bonse
- Institute for Hygiene, University of Münster, D-48149 Münster, Germany.
| | - Iris Meisen
- Institute for Hygiene, University of Münster, D-48149 Münster, Germany.
| | | | - German Vallejo
- Veterinary practice Dr. med. vet. K. Nolte and Dr. med. vet. G. Vallejo, D-48329 Havixbeck, Germany.
| | - Helge Karch
- Institute for Hygiene, University of Münster, D-48149 Münster, Germany.
| | - Johannes Müthing
- Institute for Hygiene, University of Münster, D-48149 Münster, Germany.
| |
Collapse
|
40
|
Kwon T, Bak YS, Jung YH, Yu YB, Choi JT, Kim CH, Kim JB, Kim W, Cho SH. Whole-genome sequencing and comparative genomic analysis of Escherichia coli O91 strains isolated from symptomatic and asymptomatic human carriers. Gut Pathog 2016; 8:57. [PMID: 27891181 PMCID: PMC5106847 DOI: 10.1186/s13099-016-0138-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/26/2016] [Accepted: 11/01/2016] [Indexed: 01/05/2023] Open
Abstract
Background The Shiga toxin–producing Escherichia coli (STEC) O91:H21 strains NCCP15736 and NCCP15737 were isolated during a single outbreak in Korea, NCCP15736 from a symptomatic carrier and NCCP15737 from an asymptomatic carrier. To investigate genomic differences between the two strains, we performed whole-genome sequencing of both strains and conducted a comparative genomic analysis. Results Using the Illumina HiSeq 2000 platform and Rapid Annotation using the Subsystem Technology (RAST) server, whole-genome sequences of NCCP15736 and NCCP15737 were obtained and annotated. Phylogenetic analysis of ten E. coli strains showed that NCCP15736 and NCCP15737 are evolutionarily close. The two strains were found to be most close to E. coli O91:NM str. 2009C-3745. The genomic comparison showed that the fimD gene of NCCP15737 is truncated and that the truncation could underlie the defects in infection and pathogenicity of NCCP15737. The two strains showed the same virulence factor profiles, and we identified 25 virulence factors from NCCP15736 and NCCP15737, respectively. We identified ten and nine phage-associated regions in the NCCP15736 and NCCP15737 genomes, respectively; the two strains share five of these. Conclusions NCCP15736 and NCCP15737 differ at the genomic level, even though they share features such as virulence-related genes. NCCP15737 has a deletion in fimD, which may underlie its asymptomatic character. We conclude that complete genome sequencing and integration of other types of omics data are needed to fully reveal the mechanism underlying the asymptomatic character of NCCP15737. Electronic supplementary material The online version of this article (doi:10.1186/s13099-016-0138-9) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Taesoo Kwon
- Interdisciplinary Program in Bioinformatics, Seoul National University, 1 Gwanak-ro, Gwanak-gu, Seoul, 151-742 Republic of Korea
| | - Young-Seok Bak
- Department of Emergency Medical Service, Sun Moon University, Asan-si, Chungcheongnam-do, 31460 Republic of Korea
| | - Young-Hee Jung
- Division of Antimicrobial Resistance, Center for Infectious Diseases, Korea National Institute of Health, Cheongju, 363-951 Republic of Korea
| | - Young-Bin Yu
- Department of Biomedical Laboratory Science, College of Medical Science, Konyang University, Daejeon, 302-832 Republic of Korea
| | - Jong Tae Choi
- Department of Biomedical Laboratory Science, Kyungdong University, 815 Gyeonhwon-ro, Munmak-eup, Wonju-si, Gangwon-do 26495 Republic of Korea
| | - Cheorl-Ho Kim
- Glycobiology Unit, Department of Biological Science, Sungkyunkwan University and Samsung Advanced Institute for Health Sciences and Technology (SAIHST), 2066 Seobu-ro, Suwon, 16419 Republic of Korea
| | - Jung-Beom Kim
- Department of Food Science and Technology, Sunchon National University, Sunchon, Jeonnam 540-950 Republic of Korea
| | - Won Kim
- Interdisciplinary Program in Bioinformatics, Seoul National University, 1 Gwanak-ro, Gwanak-gu, Seoul, 151-742 Republic of Korea
| | - Seung-Hak Cho
- Division of Enteric Diseases, Center for Infectious Diseases, Korea National Institute of Health, Cheongju, 363-951 Republic of Korea
| |
Collapse
|
41
|
Legros N, Dusny S, Humpf HU, Pohlentz G, Karch H, Müthing J. Shiga toxin glycosphingolipid receptors and their lipid membrane ensemble in primary human blood-brain barrier endothelial cells. Glycobiology 2016; 27:99-109. [PMID: 27558838 DOI: 10.1093/glycob/cww090] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2016] [Revised: 08/17/2016] [Accepted: 08/19/2016] [Indexed: 12/22/2022] Open
Abstract
Shiga toxin (Stx)-mediated injury to microvascular endothelial cells in the brain significantly contributes to the pathogenesis of the hemolytic-uremic syndrome caused by enterohemorrhagic Escherichia coli (EHEC). Stxs are AB5 toxins and the B-pentamers of the two major Stx subtypes Stx1a and Stx2a preferentially bind to the glycosphingolipid (GSL) globotriaosylceramide (Gb3Cer) expressed by human endothelial cells. Here we report on comprehensive structural analysis of the different lipoforms of Gb3Cer (Galα4Galβ4Glcβ1Cer) and globotetraosylceramide (Gb4Cer, GalNAcβ3Galα4Galβ4Glcβ1Cer, the less effective Stx receptor) of primary human brain microvascular endothelial cells and their association with lipid rafts. Detergent-resistant membranes (DRMs), obtained by sucrose density gradient ultracentrifugation, were used as lipid raft-analogous microdomains of the liquid-ordered phase and nonDRM fractions were employed as equivalents for the liquid-disordered phase of cell membranes. Structures of the prevalent lipoforms of Gb3Cer and Gb4Cer were those with Cer (d18:1, C16:0), Cer (d18:1, C22:0) and Cer (d18:1, C24:1/C24:0) determined by electrospray ionization mass spectrometry that was combined with thin-layer chromatography immunodetection using anti-Gb3Cer and anti-Gb4Cer antibodies as well as Stx1a and Stx2a subtypes. Association of Stx receptor GSLs was determined by co-localization with lipid raft-specific membrane protein flotillin-2 and canonical lipid raft marker sphingomyelin with Cer (d18:1, C16:0) and Cer (d18:1, C24:1/C24:0) in the liquid-ordered phase, whereas lyso-phosphatidylcholine was detectable exclusively in the liquid-disordered phase. Defining the precise microdomain structures of primary endothelial cells may help to unravel the initial mechanisms by which Stxs interact with their target cells and will help to develop novel preventive and therapeutic measures for EHEC-mediated diseases.
Collapse
Affiliation(s)
- Nadine Legros
- Institute for Hygiene, University of Münster, Robert-Koch-Str. 41, D-48149 Münster, Germany
| | - Stefanie Dusny
- Institute for Food Chemistry, University of Münster, Corrensstr. 45, D-48149 Münster, Germany
| | - Hans-Ulrich Humpf
- Institute for Food Chemistry, University of Münster, Corrensstr. 45, D-48149 Münster, Germany
| | - Gottfried Pohlentz
- Institute for Hygiene, University of Münster, Robert-Koch-Str. 41, D-48149 Münster, Germany
| | - Helge Karch
- Institute for Hygiene, University of Münster, Robert-Koch-Str. 41, D-48149 Münster, Germany
| | - Johannes Müthing
- Institute for Hygiene, University of Münster, Robert-Koch-Str. 41, D-48149 Münster, Germany .,Interdisciplinary Center for Clinical Research (IZKF), University of Münster, Albert-Schweitzer-Campus 1, D-48149 Münster, Germany
| |
Collapse
|
42
|
Fan W, Xia D, Zhu Q, Hu L, Gan Y. Intracellular transport of nanocarriers across the intestinal epithelium. Drug Discov Today 2016; 21:856-63. [DOI: 10.1016/j.drudis.2016.04.007] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2015] [Revised: 03/23/2016] [Accepted: 04/07/2016] [Indexed: 02/07/2023]
|
43
|
Betz J, Dorn I, Kouzel IU, Bauwens A, Meisen I, Kemper B, Bielaszewska M, Mormann M, Weymann L, Sibrowski W, Karch H, Schlenke P, Müthing J. Shiga toxin of enterohaemorrhagicEscherichia colidirectly injures developing human erythrocytes. Cell Microbiol 2016; 18:1339-48. [DOI: 10.1111/cmi.12592] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2016] [Revised: 03/02/2016] [Accepted: 03/03/2016] [Indexed: 12/23/2022]
Affiliation(s)
- Josefine Betz
- Institute for Hygiene; University of Münster; Münster Germany
| | - Isabel Dorn
- Pediatric Hematology and Oncology; University of Münster; Münster Germany
| | - Ivan U. Kouzel
- Institute for Hygiene; University of Münster; Münster Germany
- Interdisciplinary Center for Clinical Research (IZKF); University of Münster; Münster Germany
| | - Andreas Bauwens
- Institute for Hygiene; University of Münster; Münster Germany
| | - Iris Meisen
- Institute for Hygiene; University of Münster; Münster Germany
- Interdisciplinary Center for Clinical Research (IZKF); University of Münster; Münster Germany
| | - Björn Kemper
- Center for Biomedical Optics; University of Münster; Münster Germany
- Biomedical Technology Center; University of Münster; Münster Germany
| | | | - Michael Mormann
- Institute for Hygiene; University of Münster; Münster Germany
| | - Lena Weymann
- Institute for Hygiene; University of Münster; Münster Germany
| | - Walter Sibrowski
- Institute of Transfusion Medicine and Transplantation Immunology; University of Münster; Münster Germany
| | - Helge Karch
- Institute for Hygiene; University of Münster; Münster Germany
| | - Peter Schlenke
- Institute of Transfusion Medicine and Transplantation Immunology; University of Münster; Münster Germany
- Department of Blood Group Serology and Transfusion Medicine; Medical University of Graz; Graz Austria
| | - Johannes Müthing
- Institute for Hygiene; University of Münster; Münster Germany
- Interdisciplinary Center for Clinical Research (IZKF); University of Münster; Münster Germany
| |
Collapse
|
44
|
Kwon T, Kim JB, Bak YS, Yu YB, Kwon KS, Kim W, Cho SH. Draft genome sequence of non-shiga toxin-producing Escherichia coli O157 NCCP15738. Gut Pathog 2016; 8:13. [PMID: 27096008 PMCID: PMC4835932 DOI: 10.1186/s13099-016-0096-2] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/07/2016] [Accepted: 03/04/2016] [Indexed: 11/21/2022] Open
Abstract
Background The non-shiga toxin-producing Escherichia coli (non-STEC) O157 is a pathogenic strain that cause diarrhea but does not cause hemolytic-uremic syndrome, or hemorrhagic colitis. Here, we present the 5-Mb draft genome sequence of non-STEC O157 NCCP15738, which was isolated from the feces of a Korean patient with diarrhea, and describe its features and the structural basis for its genome evolution. Results A total of 565-Mbp paired-end reads were generated using the Illumina-HiSeq 2000 platform. The reads were assembled into 135 scaffolds throughout the de novo assembly. The assembled genome size of NCCP15738 was 5,005,278 bp with an N50 value of 142,450 bp and 50.65 % G+C content. Using Rapid Annotation using Subsystem Technology analysis, we predicted 4780 ORFs and 31 RNA genes. The evolutionary tree was inferred from multiple sequence alignment of 45 E. coli species. The most closely related neighbor of NCCP15738 indicated by whole-genome phylogeny was E. coli UMNK88, but that indicated by multilocus sequence analysis was E. coli DH1(ME8569). Conclusions A comparison between the NCCP15738 genome and those of reference strains, E. coli K-12 substr. MG1655 and EHEC O157:H7 EDL933 by bioinformatics analyses revealed unique genes in NCCP15738 associated with lysis protein S, two-component signal transduction system, conjugation, the flagellum, nucleotide-binding proteins, and metal-ion binding proteins. Notably, NCCP15738 has a dual flagella system like that in Vibrio parahaemolyticus, Aeromonas spp., and Rhodospirillum centenum. The draft genome sequence and the results of bioinformatics analysis of NCCP15738 provide the basis for understanding the genomic evolution of this strain. Electronic supplementary material The online version of this article (doi:10.1186/s13099-016-0096-2) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Taesoo Kwon
- School of Biological Sciences, Seoul National University, 1 Gwanak-ro, Gwanak-gu, Seoul, 151-742 Republic of Korea ; Division of Biosafety Evaluation and Control, Korea National Institute of Health, Cheongju, 363-951 Republic of Korea
| | - Jung-Beom Kim
- Department of Food Science and Technology, Sunchon National University, Sunchon, Jeonnam 540-950 Republic of Korea
| | - Young-Seok Bak
- Department of Emergency Medical Service, College of Medical Science, Konyang University, Daejeon, 302-832 Republic of Korea
| | - Young-Bin Yu
- Department of Biomedical Laboratory Science, College of Medical Science, Konyang University, Daejeon, 302-832 Republic of Korea
| | - Ki Sung Kwon
- New Hazardous Substances Team, National Institute of Food and Drug Safety Evaluation, Cheongju, 363-700 Republic of Korea
| | - Won Kim
- School of Biological Sciences, Seoul National University, 1 Gwanak-ro, Gwanak-gu, Seoul, 151-742 Republic of Korea
| | - Seung-Hak Cho
- Division of Enteric Diseases, Center for Infectious Diseases, Korea National Institute of Health, Cheongju, 363-951 Republic of Korea
| |
Collapse
|
45
|
Arnold JW, Spacht D, Koudelka GB. Determinants that govern the recognition and uptake of
Escherichia coli
O157 : H7 by
Acanthamoeba castellanii. Cell Microbiol 2016; 18:1459-70. [DOI: 10.1111/cmi.12591] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2015] [Revised: 03/07/2016] [Accepted: 03/08/2016] [Indexed: 11/28/2022]
Affiliation(s)
- Jason W. Arnold
- Department of Biological Sciences University at Buffalo Buffalo NY 14260 USA
| | - Drew Spacht
- Department of Biology Mercyhurst University Erie PA 16546 USA
- Department of Entomology The Ohio State University 318 W. 12th Ave. 300 Aronoff Laboratory Columbus OH 43210 USA
| | - Gerald B. Koudelka
- Department of Biological Sciences University at Buffalo Buffalo NY 14260 USA
| |
Collapse
|
46
|
Bernedo-Navarro RA, Yano T. Phage display and Shiga toxin neutralizers. Toxicon 2016; 113:60-9. [DOI: 10.1016/j.toxicon.2016.02.009] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2015] [Revised: 02/03/2016] [Accepted: 02/11/2016] [Indexed: 02/01/2023]
|
47
|
Bacterial genotoxins: The long journey to the nucleus of mammalian cells. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2016; 1858:567-75. [DOI: 10.1016/j.bbamem.2015.08.016] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/22/2015] [Revised: 07/17/2015] [Accepted: 08/18/2015] [Indexed: 02/06/2023]
|
48
|
Escherichia coli O104:H4 Pathogenesis: an Enteroaggregative E. coli/Shiga Toxin-Producing E. coli Explosive Cocktail of High Virulence. Microbiol Spectr 2016; 2. [PMID: 26104460 DOI: 10.1128/microbiolspec.ehec-0008-2013] [Citation(s) in RCA: 56] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
A major outbreak caused by Escherichia coli of serotype O104:H4 spread throughout Europe in 2011. This large outbreak was caused by an unusual strain that is most similar to enteroaggregative E. coli (EAEC) of serotype O104:H4. A significant difference, however, is the presence of a prophage encoding the Shiga toxin, which is characteristic of enterohemorrhagic E. coli (EHEC) strains. This combination of genomic features, associating characteristics from both EAEC and EHEC, represents a new pathotype. The 2011 E. coli O104:H4 outbreak of hemorrhagic diarrhea in Germany is an example of the explosive cocktail of high virulence and resistance that can emerge in this species. A total of 46 deaths, 782 cases of hemolytic-uremic syndrome, and 3,128 cases of acute gastroenteritis were attributed to this new clone of EAEC/EHEC. In addition, recent identification in France of similar O104:H4 clones exhibiting the same virulence factors suggests that the EHEC O104:H4 pathogen has become endemically established in Europe after the end of the outbreak. EAEC strains of serotype O104:H4 contain a large set of virulence-associated genes regulated by the AggR transcription factor. They include, among other factors, the pAA plasmid genes encoding the aggregative adherence fimbriae, which anchor the bacterium to the intestinal mucosa (stacked-brick adherence pattern on epithelial cells). Furthermore, sequencing studies showed that horizontal genetic exchange allowed for the emergence of the highly virulent Shiga toxin-producing EAEC O104:H4 strain that caused the German outbreak. This article discusses the role these virulence factors could have in EAEC/EHEC O104:H4 pathogenesis.
Collapse
|
49
|
Lai CK, Chen YA, Lin CJ, Lin HJ, Kao MC, Huang MZ, Lin YH, Chiang-Ni C, Chen CJ, Lo UG, Lin LC, Lin H, Hsieh JT, Lai CH. Molecular Mechanisms and Potential Clinical Applications of Campylobacter jejuni Cytolethal Distending Toxin. Front Cell Infect Microbiol 2016; 6:9. [PMID: 26904508 PMCID: PMC4746238 DOI: 10.3389/fcimb.2016.00009] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2015] [Accepted: 01/18/2016] [Indexed: 12/12/2022] Open
Abstract
Cytolethal distending toxin (CDT), a genotoxin produced by Campylobacter jejuni, is composed of three subunits: CdtA, CdtB, and CdtC. CdtB is a DNase that causes DNA double-strand breaks (DSB) in the nucleus resulting in cell cycle arrest at the G2/M stage and apoptosis. CdtA and CdtC bind to cholesterol-rich microdomains on the cytoplasmic membrane, a process required for the delivery of CdtB to cells. Although a unique motif associated with cholesterol-binding activity has been identified in other pathogens, the mechanism underlying the interaction between the CdtA and CdtC subunits and membrane cholesterol remains unclear. Also, the processes of cell uptake and delivery of CdtB in host cells and the translocation of CdtB into the nucleus are only partially understood. In this review, we focus on the underlying relationship among CDT, membrane cholesterol, and the intracellular trafficking pathway as a unique mechanism for C. jejuni-induced pathogenesis. Moreover, we discuss the clinical aspects of a possible therapeutic application of CDT in cancer therapy. Understanding the molecular mechanism of CDT-host interactions may provide insights into novel strategies to control C. jejuni infection and the development of potential clinical applications of CDT.
Collapse
Affiliation(s)
- Cheng-Kuo Lai
- Department of Microbiology and Immunology, Graduate Institute of Biomedical Sciences, College of Medicine, Chang Gung UniversityTaoyuan, Taiwan; Department of Urology, University of Texas Southwestern Medical CenterDallas, TX, USA
| | - Yu-An Chen
- School of Medicine, Graduate Institute of Basic Medical Science, China Medical University Taichung, Taiwan
| | - Chun-Jung Lin
- Department of Urology, University of Texas Southwestern Medical CenterDallas, TX, USA; School of Medicine, Graduate Institute of Basic Medical Science, China Medical UniversityTaichung, Taiwan
| | - Hwai-Jeng Lin
- Department of Internal Medicine, School of Medicine, College of Medicine, Taipei Medical UniversityNew Taipei, Taiwan; Division of Gastroenterology and Hepatology, Department of Internal Medicine, Shuang-Ho HospitalNew Taipei, Taiwan
| | - Min-Chuan Kao
- Department of Microbiology and Immunology, Graduate Institute of Biomedical Sciences, College of Medicine, Chang Gung University Taoyuan, Taiwan
| | - Mei-Zi Huang
- Department of Microbiology and Immunology, Graduate Institute of Biomedical Sciences, College of Medicine, Chang Gung UniversityTaoyuan, Taiwan; School of Medicine, Graduate Institute of Basic Medical Science, China Medical UniversityTaichung, Taiwan
| | - Yu-Hsin Lin
- Department of Urology, University of Texas Southwestern Medical CenterDallas, TX, USA; Department of Biological Science and Technology, China Medical UniversityTaichung, Taiwan
| | - Chuan Chiang-Ni
- Department of Microbiology and Immunology, Graduate Institute of Biomedical Sciences, College of Medicine, Chang Gung University Taoyuan, Taiwan
| | - Chih-Jung Chen
- Department of Pediatrics, Molecular Infectious Disease Research Center, Chang Gung Children's Hospital and Chang Gung Memorial Hospital Taoyuan, Taiwan
| | - U-Ging Lo
- Department of Urology, University of Texas Southwestern Medical Center Dallas, TX, USA
| | - Li-Chiung Lin
- Department of Urology, University of Texas Southwestern Medical CenterDallas, TX, USA; Department of Life Sciences, National Chung Hsing UniversityTaichung, Taiwan
| | - Ho Lin
- Department of Life Sciences, National Chung Hsing University Taichung, Taiwan
| | - Jer-Tsong Hsieh
- Department of Urology, University of Texas Southwestern Medical CenterDallas, TX, USA; Graduate Institute of Cancer Biology, China Medical UniversityTaichung, Taiwan
| | - Chih-Ho Lai
- Department of Microbiology and Immunology, Graduate Institute of Biomedical Sciences, College of Medicine, Chang Gung UniversityTaoyuan, Taiwan; School of Medicine, Graduate Institute of Basic Medical Science, China Medical UniversityTaichung, Taiwan; Department of Pediatrics, Molecular Infectious Disease Research Center, Chang Gung Children's Hospital and Chang Gung Memorial HospitalTaoyuan, Taiwan; Department of Nursing, Asia UniversityTaichung, Taiwan
| |
Collapse
|
50
|
Xu T, Marr E, Lam H, Ripp S, Sayler G, Close D. Real-time toxicity and metabolic activity tracking of human cells exposed to Escherichia coli O157:H7 in a mixed consortia. ECOTOXICOLOGY (LONDON, ENGLAND) 2015; 24:2133-2140. [PMID: 26423391 PMCID: PMC5597431 DOI: 10.1007/s10646-015-1552-3] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 09/16/2015] [Indexed: 06/01/2023]
Abstract
Escherichia coli O157:H7 is a significant human pathogen that is continually responsible for sickness, and even death, on a worldwide scale. While the pathology of E. coli O157:H7 infection has been well studied, the effect of it's multiple resulting cytotoxic mechanisms on host metabolic activity has not been well characterized. To develop a more thorough understanding of these effects, several bioluminescence assays were evaluated for their ability to track both toxicity and host metabolic activity levels in real-time. The use of continuously autobioluminescent human cells was determined to be the most favorable method for tracking these metrics, as its self-sufficient autobioluminescent phenotype was unaffected by the presence of the infecting bacteria and its signal could be measured without cellular destruction. Using this approach, it was determined that infection with as few as 10 CFU of E. coli O157:H7 could elicit cytotoxic effects. Regardless of the initial infective dose, an impact on metabolic expression was not observed until bacterial populations reached levels between 5 × 10(5) and 1 × 10(6) (R(2) = 0.933), indicating that a critical bacterial infection level must be reached prior to the onset of cytotoxic effects. Supporting this hypothesis, it was found that cells displaying infection-mediated metabolic activity reductions could recover to wild type metabolic activity levels if the infecting bacteria were removed prior to cell death. These results indicate that rapid treatment of E. coli O157:H7 infection could serve to limit host metabolic impact and reduce overall host cell death.
Collapse
Affiliation(s)
- Tingting Xu
- Center for Environmental Biotechnology, University of Tennessee, 676 Dabney Hall, 1414 Circle Drive, Knoxville, TN, 37996, USA
| | - Enolia Marr
- Center for Environmental Biotechnology, University of Tennessee, 676 Dabney Hall, 1414 Circle Drive, Knoxville, TN, 37996, USA
| | - Haylie Lam
- Center for Environmental Biotechnology, University of Tennessee, 676 Dabney Hall, 1414 Circle Drive, Knoxville, TN, 37996, USA
| | - Steven Ripp
- 490 BioTech, Inc., 2450 E. J. Chapman Drive, Knoxville, TN, 37996, USA
| | - Gary Sayler
- Center for Environmental Biotechnology, University of Tennessee, 676 Dabney Hall, 1414 Circle Drive, Knoxville, TN, 37996, USA
| | - Dan Close
- 490 BioTech, Inc., 2450 E. J. Chapman Drive, Knoxville, TN, 37996, USA.
| |
Collapse
|