1
|
Li J, Mehdizadeh Gohari I, Zhang I, McClane BA. BrnQ Branched-Chain Amino Acid Transporters Influence Toxin Production by, but Not Growth of, Clostridium perfringens Type A Strain ATCC3624. Toxins (Basel) 2025; 17:187. [PMID: 40278685 PMCID: PMC12031218 DOI: 10.3390/toxins17040187] [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: 12/18/2024] [Revised: 03/19/2025] [Accepted: 03/27/2025] [Indexed: 04/26/2025] Open
Abstract
By producing alpha toxin (PLC) and perfringolysin O (PFO), Clostridium perfringens type A strains are the most common cause of traumatic gas gangrene. C. perfringens cannot synthesize branched-chain amino acids (BCAAs), so BCAA transporters are essential for C. perfringens growth and survival. C. perfringens type A strain ATCC3624 encodes the BrnQ1, BrnQ2, and BrnQ3 BCAA transporters. RT-PCR analyses showed that, with increasing culture time in TY broth, brnQ2 and brnQ3 expression levels remained stable but brnQ1 expression levels declined. Single null mutants unable to produce one of the BrnQ proteins grew and survived similarly as wild type. However, these mutants all showed altered PLC production, especially in the early culture stage, and those effects were reversible by complementation. Therefore, the presence of BrnQ proteins impacts toxin production levels, even though they are not necessary for growth. Interestingly, a triple mutant that was unable to produce any BrnQ protein also grew similarly as ATCC3624. Since BCAA uptake is essential for C. perfringens, this strain must produce another (still to be identified) BCAA transporter.
Collapse
Affiliation(s)
| | | | | | - Bruce A. McClane
- Department of Microbiology and Molecular Genetics, School of Medicine, University of Pittsburgh, Pittsburgh, PA 15219, USA; (J.L.); (I.M.G.); (I.Z.)
| |
Collapse
|
2
|
Camargo A, Ramírez JD, Kiu R, Hall LJ, Muñoz M. Unveiling the pathogenic mechanisms of Clostridium perfringens toxins and virulence factors. Emerg Microbes Infect 2024; 13:2341968. [PMID: 38590276 PMCID: PMC11057404 DOI: 10.1080/22221751.2024.2341968] [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: 12/28/2023] [Accepted: 04/06/2024] [Indexed: 04/10/2024]
Abstract
Clostridium perfringens causes multiple diseases in humans and animals. Its pathogenic effect is supported by a broad and heterogeneous arsenal of toxins and other virulence factors associated with a specific host tropism. Molecular approaches have indicated that most C. perfringens toxins produce membrane pores, leading to osmotic cell disruption and apoptosis. However, identifying mechanisms involved in cell tropism and selective toxicity effects should be studied more. The differential presence and polymorphisms of toxin-encoding genes and genes encoding other virulence factors suggest that molecular mechanisms might exist associated with host preference, receptor binding, and impact on the host; however, this information has not been reviewed in detail. Therefore, this review aims to clarify the current state of knowledge on the structural features and mechanisms of action of the major toxins and virulence factors of C. perfringens and discuss the impact of genetic diversity of toxinotypes in tropism for several hosts.
Collapse
Affiliation(s)
- Anny Camargo
- Centro de Investigaciones en Microbiología y Biotecnología-UR (CIMBIUR), Facultad de Ciencias Naturales, Universidad del Rosario, Bogotá, Colombia
- Health Sciences Faculty, Universidad de Boyacá, Tunja, Colombia
| | - Juan David Ramírez
- Centro de Investigaciones en Microbiología y Biotecnología-UR (CIMBIUR), Facultad de Ciencias Naturales, Universidad del Rosario, Bogotá, Colombia
- Molecular Microbiology Laboratory, Department of Pathology, Molecular and Cell-Based Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Raymond Kiu
- Institute of Microbiology and Infection, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK
- Gut Microbes and Health, Quadram Institute Bioscience, Norwich Research Park, Norwich, UK
| | - Lindsay J. Hall
- Institute of Microbiology and Infection, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK
- Gut Microbes and Health, Quadram Institute Bioscience, Norwich Research Park, Norwich, UK
- Norwich Medical School, University of East Anglia, Norwich, UK
| | - Marina Muñoz
- Centro de Investigaciones en Microbiología y Biotecnología-UR (CIMBIUR), Facultad de Ciencias Naturales, Universidad del Rosario, Bogotá, Colombia
- Instituto de Biotecnología-UN (IBUN), Universidad Nacional de Colombia, Bogotá, Colombia
| |
Collapse
|
3
|
Li J, Sayeed S, McClane BA. The presence of differentiated C2C12 muscle cells enhances toxin production and growth by Clostridium perfringens type A strain ATCC3624. Virulence 2024; 15:2388219. [PMID: 39192628 PMCID: PMC11364075 DOI: 10.1080/21505594.2024.2388219] [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: 12/21/2023] [Revised: 07/03/2024] [Accepted: 07/28/2024] [Indexed: 08/29/2024] Open
Abstract
Clostridium perfringens type A causes gas gangrene, which involves muscle infection. Both alpha toxin (PLC), encoded by the plc gene, and perfringolysin O (PFO), encoded by the pfoA gene, are important when type A strains cause gas gangrene in a mouse model. This study used the differentiated C2C12 muscle cell line to test the hypothesis that one or both of those toxins contributes to gas gangrene pathogenesis by releasing growth nutrients from muscle cells. RT-qPCR analyses showed that the presence of differentiated C2C12 cells induces C. perfringens type A strain ATCC3624 to upregulate plc and pfoA expression, as well as increase expression of several regulatory genes, including virS/R, agrB/D, and eutV/W. The VirS/R two component regulatory system (TCRS) and its coupled Agr-like quorum sensing system, along with the EutV/W TCRS (which regulates expression of genes involved in ethanolamine [EA] utilization), were shown to mediate the C2C12 cell-induced increase in plc and pfoA expression. EA was demonstrated to increase toxin gene expression. ATCC3624 growth increased in the presence of differentiated C2C12 muscle cells and this effect was shown to involve both PFO and PLC. Those membrane-active toxins were each cytotoxic for differentiated C2C12 cells, suggesting they support ATCC3624 growth by releasing nutrients from differentiated C2C12 cells. These findings support a model where, during gas gangrene, increased production of PFO and PLC in the presence of muscle cells causes more damage to those host cells, which release nutrients like EA that are then used to support C. perfringens growth in muscle.
Collapse
Affiliation(s)
- Jihong Li
- Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Sameera Sayeed
- Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Bruce A. McClane
- Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| |
Collapse
|
4
|
Wang XY, Meng FH, Zhang MY, Li FX, Lei YX, Ma ZG, Li JQ, Lou YN, Chu YF, Ma K, Yu SX. Gut Lactococcus garvieae promotes protective immunity to foodborne Clostridium perfringens infection. Microbiol Spectr 2024; 12:e0402523. [PMID: 39190634 PMCID: PMC11448249 DOI: 10.1128/spectrum.04025-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2023] [Accepted: 07/19/2024] [Indexed: 08/29/2024] Open
Abstract
The gut microbiota, a pivotal component of the intestinal mucosal barrier, is critical for host resistance to enteric pathogen infection. Here, we report a novel function of the potentially probiotic Lactococcus garvieae strain LG1 (L. garvieae strain LG1) in maintaining intestinal mucosal barrier integrity and protecting against foodborne Clostridium perfringens (C. perfringens) infection. L. garvieae was isolated from the intestinal contents of Chinese Mongolian sheep (MS) and exhibited potential probiotic properties. In a C. perfringens enterocolitis model, L. garvieae-pretreated mice were less susceptible to C. perfringens infection compared with Phosphate buffered solution (PBS)-pretreated mice, which manifested as higher survival rates, lower pathogen loads, less weight loss, mild clinical symptoms and intestinal damage, and minor inflammation. Further mechanistic analysis showed that L. garvieae could ameliorate the disruption of intestinal permeability and maintain the integrity of the intestinal mucosal barrier by promoting the expression of tight junction proteins and mucoproteins. Moreover, L. garvieae was also able to facilitate antimicrobial peptide expression and ameliorate dysbiosis of the gut microbiota caused by C. perfringens. Together, these findings highlight the prospect of immunomodulatory potentially probiotic L. garvieae and might offer valuable strategies for prophylaxis and/or treatment of pathogenic C. perfringens mucosal infection. IMPORTANCE C. perfringens necrotic enteritis leads to losses of about US $2 billion to the poultry industry worldwide every year. Worse, US Centers for Disease Control and Prevention (CDC) has estimated that C. perfringens causes nearly 1 million foodborne illnesses in the United States annually. Nowadays, the treatment recommendation is a combination of a broad-spectrum synergistic penicillin with clindamycin or a carbapenem, despite growing scientific concern over antibiotic resistance. The global understanding of the gut microbiome for C. perfringens infection may provide important insights into the intervention. L. garvieae originated from Mongolian sheep intestine, exhibited potentially probiotic properties, and was able to limit C. perfringens enterocolitis and pathogenic colonization. Importantly, we found that L. garvieae limits C. perfringens invasion via improving intestinal mucosal barrier function. Also, L. garvieae alleviates C. perfringens-induced gut microbiota dysbiosis. It allowed us to convince that utilization of probiotics to promote protective immunity against pathogens infection is of pivotal importance.
Collapse
Affiliation(s)
- Xue-Yin Wang
- State Key Laboratory of Reproductive Regulation and Breeding of Grassland Livestock, College of Life Sciences, Inner Mongolia University, Hohhot, China
| | - Fan-Hua Meng
- State Key Laboratory of Reproductive Regulation and Breeding of Grassland Livestock, College of Life Sciences, Inner Mongolia University, Hohhot, China
| | - Ming-Yue Zhang
- State Key Laboratory of Reproductive Regulation and Breeding of Grassland Livestock, College of Life Sciences, Inner Mongolia University, Hohhot, China
| | - Fen-Xin Li
- State Key Laboratory of Reproductive Regulation and Breeding of Grassland Livestock, College of Life Sciences, Inner Mongolia University, Hohhot, China
| | - Yu-Xin Lei
- State Key Laboratory of Reproductive Regulation and Breeding of Grassland Livestock, College of Life Sciences, Inner Mongolia University, Hohhot, China
| | - Zhao-Guo Ma
- State Key Laboratory of Reproductive Regulation and Breeding of Grassland Livestock, College of Life Sciences, Inner Mongolia University, Hohhot, China
| | - Jia-Qi Li
- State Key Laboratory of Reproductive Regulation and Breeding of Grassland Livestock, College of Life Sciences, Inner Mongolia University, Hohhot, China
| | - Ya-Nan Lou
- State Key Laboratory of Reproductive Regulation and Breeding of Grassland Livestock, College of Life Sciences, Inner Mongolia University, Hohhot, China
| | - Yue-Feng Chu
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy agricultural Sciences, Lanzhou, China
| | - Ke Ma
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy agricultural Sciences, Lanzhou, China
| | - Shui-Xing Yu
- State Key Laboratory of Reproductive Regulation and Breeding of Grassland Livestock, College of Life Sciences, Inner Mongolia University, Hohhot, China
- Inner Mongolia Engineering Technology Research Center of Germplasm Resources Conservation and Utilization, College of Life Sciences, Inner Mongolia University, Hohhot, China
| |
Collapse
|
5
|
Waller SB, Galvão CC, Rodrigues RR, Aguirres CDL, Quatrin PHDN, Alves MLF, Ferreira MRA, Conceição FR. Clostridium perfringens antigens and challenges for development of vaccines against necrotic enteritis in poultry. Anaerobe 2024; 89:102902. [PMID: 39187174 DOI: 10.1016/j.anaerobe.2024.102902] [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: 06/04/2024] [Revised: 08/06/2024] [Accepted: 08/10/2024] [Indexed: 08/28/2024]
Abstract
INTRODUCTION Chickens with Necrotic Enteritis (NE), caused by Clostridium perfringens, exhibit acute and chronic symptoms that are difficult to diagnose, leading to significant economic losses. Vaccination is the best method for controlling and preventing NE. However, only two vaccines based on the CPA and NetB toxins have been commercialized, offering partial protection, highlighting the urgent need for more effective vaccines. OBJECTIVE This review aimed to identify promising antigens for NE vaccine formulation and discuss factors affecting their effectiveness. METHODS A systematic review using five scientific databases identified 30 eligible studies through the Rayyan tool, which were included for quality review. RESULTS We identified 25 promising antigens, including CPA, NetB, FBA, ZMP, CnaA, FimA, and FimB, categorized by their role in disease pathogenesis. This review discusses the biochemical, physiological, and genetic traits of recombinant antigens used in vaccine prototypes, their expression systems, and immunization potential in chickens challenged with virulent C. perfringens strains. Market supply challenges, immunogenic potential, vaccine platforms, adjuvants, and factors related to vaccination schedules-such as administration routes, dosing intervals, and age at immunization-are also addressed. Additionally, the study notes that vaccine formulations tested under mild challenges may not offer adequate field-level protection due to issues replicating aggressive conditions, strain virulence loss, and varied methodologies. CONCLUSIONS An ideal NE vaccine should incorporate multiple antigens, molecular adjuvants, and delivery systems via in ovo and oral routes. The review underscores the challenges in developing and validating NE vaccines and the urgent need for a standardized protocol to replicate aggressive challenges accurately.
Collapse
Affiliation(s)
- Stefanie Bressan Waller
- Centro de Desenvolvimento Tecnológico, Biotecnologia, Universidade Federal de Pelotas (UFPEL), Pelotas, RS, Brazil
| | - Cleideanny Cancela Galvão
- Centro de Desenvolvimento Tecnológico, Biotecnologia, Universidade Federal de Pelotas (UFPEL), Pelotas, RS, Brazil
| | - Rafael Rodrigues Rodrigues
- Centro de Desenvolvimento Tecnológico, Biotecnologia, Universidade Federal de Pelotas (UFPEL), Pelotas, RS, Brazil
| | - Cleiderson de Lima Aguirres
- Centro de Desenvolvimento Tecnológico, Biotecnologia, Universidade Federal de Pelotas (UFPEL), Pelotas, RS, Brazil
| | | | | | | | - Fabricio Rochedo Conceição
- Centro de Desenvolvimento Tecnológico, Biotecnologia, Universidade Federal de Pelotas (UFPEL), Pelotas, RS, Brazil.
| |
Collapse
|
6
|
Reffo I, Domini M, Cevolani M, Del Fabro G, Rufolo D, Venturini S, Pinciroli L, Tonin D, Avolio M, Crapis M, Basaglia G, Balbi M, Nadalin G. Clostridium perfringens-induced massive hemolysis treatment with blood purification to target toxins: a case report. CEN Case Rep 2024; 13:391-396. [PMID: 38436873 PMCID: PMC11444022 DOI: 10.1007/s13730-024-00857-3] [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: 10/22/2023] [Accepted: 02/07/2024] [Indexed: 03/05/2024] Open
Abstract
Clostridium perfringens can rarely cause severe systemic infections, usually from an abdominal source, associated with massive hemolysis, which is usually fatal. Hemolytic anemia and acute renal injury resulting from toxin action are critical for the development of multiple organ dysfunction syndrome (MODs), making this condition a real emergency, requiring multispecialty skills and aggressive multimodal therapies. We herein describe a case of septic shock from acute cholecystitis with massive hemolysis caused by C. perfringens in a 55 year-old man that was successfully treated with early blood purification and continuous renal replacement therapy (CRRT) along with antibiotic therapy and surgery. The effect of the enormous amount of toxins produced by Clostridium which elicit a strong cytokine response and the damage caused by the hemolysis products are the main pathogenetic mechanisms of this rare but lethal clinical entity. The main goal of treatment is to remove toxins from plasma, block toxin action, and further production by achieving bacterial killing with antimicrobial agents and controlling the infectious focus, remove waste products and prevent or limit multiorgan damage. Blood purification techniques play an important role due to a strong pathophysiological rationale, as they can remove toxins and cytokines as well as cell-free products from plasma and also replace renal function. Although this condition is rare and robust data are lacking, blood purification techniques for C. perfringens-induced massive hemolysis are promising and should be further explored.
Collapse
Affiliation(s)
- I Reffo
- Anaesthesia and Intensive Care Department, Azienda Sanitaria Friuli Occidentale "Santa Maria Dei Battuti" Hospital, Via Savorgnano 2, San Vito al Tagliamento, 33078, Pordenone, Italy.
| | - M Domini
- Anaesthesia and Intensive Care Department, Azienda Sanitaria Friuli Occidentale "Santa Maria Dei Battuti" Hospital, Via Savorgnano 2, San Vito al Tagliamento, 33078, Pordenone, Italy
| | - M Cevolani
- Internal Medicine Department, Azienda Sanitaria Friuli Occidentale "Santa Maria dei Battuti" Hospital, San Vito al Tagliamento, Pordenone, Italy
| | - G Del Fabro
- Infectious Diseases Department, Azienda Sanitaria Friuli Occidentale "Santa Maria degli Angeli" Hospital, Pordenone, Italy
| | - D Rufolo
- Anaesthesia and Intensive Care Department, Azienda Sanitaria Friuli Occidentale "Santa Maria Dei Battuti" Hospital, Via Savorgnano 2, San Vito al Tagliamento, 33078, Pordenone, Italy
| | - S Venturini
- Infectious Diseases Department, Azienda Sanitaria Friuli Occidentale "Santa Maria degli Angeli" Hospital, Pordenone, Italy
| | - L Pinciroli
- General Surgery Department, Azienda Sanitaria Friuli Occidentale "Santa Maria dei Battuti" Hospital, San Vito Al Tagliamento, Pordenone, Italy
| | - D Tonin
- General Surgery Department, Azienda Sanitaria Friuli Occidentale "Santa Maria dei Battuti" Hospital, San Vito Al Tagliamento, Pordenone, Italy
| | - M Avolio
- Microbiology Department, Azienda Sanitaria Friuli Occidentale "Santa Maria degli Angeli" Hospital, Pordenone, Italy
| | - M Crapis
- Infectious Diseases Department, Azienda Sanitaria Friuli Occidentale "Santa Maria degli Angeli" Hospital, Pordenone, Italy
| | - G Basaglia
- Microbiology Department, Azienda Sanitaria Friuli Occidentale "Santa Maria degli Angeli" Hospital, Pordenone, Italy
| | - M Balbi
- Internal Medicine Department, Azienda Sanitaria Friuli Occidentale "Santa Maria dei Battuti" Hospital, San Vito al Tagliamento, Pordenone, Italy
| | - G Nadalin
- Anaesthesia and Intensive Care Department, Azienda Sanitaria Friuli Occidentale "Santa Maria Dei Battuti" Hospital, Via Savorgnano 2, San Vito al Tagliamento, 33078, Pordenone, Italy
| |
Collapse
|
7
|
Liguori F, Pellicciotta N, Milanetti E, Xi Windemuth S, Ruocco G, Di Leonardo R, Danino T. Dynamic Gene Expression Mitigates Mutational Escape in Lysis-Driven Bacteria Cancer Therapy. BIODESIGN RESEARCH 2024; 6:0049. [PMID: 39301524 PMCID: PMC11411163 DOI: 10.34133/bdr.0049] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2024] [Revised: 08/09/2024] [Accepted: 08/25/2024] [Indexed: 09/22/2024] Open
Abstract
Engineered bacteria have the potential to deliver therapeutic payloads directly to tumors, with synthetic biology enabling precise control over therapeutic release in space and time. However, it remains unclear how to optimize therapeutic bacteria for durable colonization and sustained payload release. Here, we characterize nonpathogenic Escherichia coli expressing the bacterial toxin Perfringolysin O (PFO) and dynamic strategies that optimize therapeutic efficacy. While PFO is known for its potent cancer cell cytotoxicity, we present experimental evidence that expression of PFO causes lysis of bacteria in both batch culture and microfluidic systems, facilitating its efficient release. However, prolonged expression of PFO leads to the emergence of a mutant population that limits therapeutic-releasing bacteria in a PFO expression level-dependent manner. We present sequencing data revealing the mutant takeover and employ molecular dynamics to confirm that the observed mutations inhibit the lysis efficiency of PFO. To analyze this further, we developed a mathematical model describing the evolution of therapeutic-releasing and mutant bacteria populations revealing trade-offs between therapeutic load delivered and fraction of mutants that arise. We demonstrate that a dynamic strategy employing short and repeated inductions of the pfo gene better preserves the original population of therapeutic bacteria by mitigating the effects of mutational escape. Altogether, we demonstrate how dynamic modulation of gene expression can address mutant takeovers giving rise to limitations in engineered bacteria for therapeutic applications.
Collapse
Affiliation(s)
- Filippo Liguori
- Department of Physics, Sapienza University of Rome, Rome, Italy
- Center for Life Nano- & Neuro-Science, Istituto Italiano di Tecnologia, Rome, Italy
| | - Nicola Pellicciotta
- NANOTEC-CNR, Soft and Living Matter Laboratory, Institute of Nanotechnology, Rome, Italy
- Department of Physics, Sapienza University of Rome, Rome, Italy
| | - Edoardo Milanetti
- Department of Physics, Sapienza University of Rome, Rome, Italy
- Center for Life Nano- & Neuro-Science, Istituto Italiano di Tecnologia, Rome, Italy
| | - Sophia Xi Windemuth
- Department of Biomedical Engineering, Columbia University, New York, NY, USA
| | - Giancarlo Ruocco
- Department of Physics, Sapienza University of Rome, Rome, Italy
- Center for Life Nano- & Neuro-Science, Istituto Italiano di Tecnologia, Rome, Italy
| | - Roberto Di Leonardo
- Department of Physics, Sapienza University of Rome, Rome, Italy
- NANOTEC-CNR, Soft and Living Matter Laboratory, Institute of Nanotechnology, Rome, Italy
| | - Tal Danino
- Department of Biomedical Engineering, Columbia University, New York, NY, USA
- Herbert Irving Comprehensive Cancer Center, Columbia University, New York, NY, USA
- Data Science Institute, Columbia University, New York, NY, USA
| |
Collapse
|
8
|
Šakanović A, Kranjc N, Omersa N, Aden S, Kežar A, Kisovec M, Zavec AB, Caserman S, Gilbert RJC, Podobnik M, Crnković A, Anderluh G. In vitro evolution driven by epistasis reveals alternative cholesterol-specific binding motifs of perfringolysin O. J Biol Chem 2024; 300:107664. [PMID: 39128714 PMCID: PMC11416283 DOI: 10.1016/j.jbc.2024.107664] [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: 03/06/2024] [Revised: 07/22/2024] [Accepted: 08/05/2024] [Indexed: 08/13/2024] Open
Abstract
The crucial molecular factors that shape the interfaces of lipid-binding proteins with their target ligands and surfaces remain unknown due to the complex makeup of biological membranes. Cholesterol, the major modulator of bilayer structure in mammalian cell membranes, is recognized by various proteins, including the well-studied cholesterol-dependent cytolysins. Here, we use in vitro evolution to investigate the molecular adaptations that preserve the cholesterol specificity of perfringolysin O, the prototypical cholesterol-dependent cytolysin from Clostridium perfringens. We identify variants with altered membrane-binding interfaces whose cholesterol-specific activity exceeds that of the wild-type perfringolysin O. These novel variants represent alternative evolutionary outcomes and have mutations at conserved positions that can only accumulate when epistatic constraints are alleviated. Our results improve the current understanding of the biochemical malleability of the surface of a lipid-binding protein.
Collapse
Affiliation(s)
- Aleksandra Šakanović
- Department of Molecular Biology and Nanobiotechnology, National Institute of Chemistry, Ljubljana, Slovenia
| | - Nace Kranjc
- Department of Molecular Biology and Nanobiotechnology, National Institute of Chemistry, Ljubljana, Slovenia
| | - Neža Omersa
- Department of Molecular Biology and Nanobiotechnology, National Institute of Chemistry, Ljubljana, Slovenia
| | - Saša Aden
- Department of Molecular Biology and Nanobiotechnology, National Institute of Chemistry, Ljubljana, Slovenia
| | - Andreja Kežar
- Department of Molecular Biology and Nanobiotechnology, National Institute of Chemistry, Ljubljana, Slovenia
| | - Matic Kisovec
- Department of Molecular Biology and Nanobiotechnology, National Institute of Chemistry, Ljubljana, Slovenia
| | - Apolonija Bedina Zavec
- Department of Molecular Biology and Nanobiotechnology, National Institute of Chemistry, Ljubljana, Slovenia
| | - Simon Caserman
- Department of Molecular Biology and Nanobiotechnology, National Institute of Chemistry, Ljubljana, Slovenia
| | - Robert J C Gilbert
- Division of Structural Biology, Wellcome Centre for Human Genetics, University of Oxford, Oxford, United Kingdom
| | - Marjetka Podobnik
- Department of Molecular Biology and Nanobiotechnology, National Institute of Chemistry, Ljubljana, Slovenia
| | - Ana Crnković
- Department of Molecular Biology and Nanobiotechnology, National Institute of Chemistry, Ljubljana, Slovenia.
| | - Gregor Anderluh
- Department of Molecular Biology and Nanobiotechnology, National Institute of Chemistry, Ljubljana, Slovenia.
| |
Collapse
|
9
|
Shamshirgaran MA, Golchin M. A comprehensive review of experimental models and induction protocols for avian necrotic enteritis over the past 2 decades. Front Vet Sci 2024; 11:1429637. [PMID: 39113718 PMCID: PMC11304537 DOI: 10.3389/fvets.2024.1429637] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2024] [Accepted: 06/07/2024] [Indexed: 08/10/2024] Open
Abstract
Necrotic enteritis (NE) is a severe gastrointestinal disease that poses a significant threat to the poultry industry. It leads to progressive damage to the small intestine, reduced performance, increased mortality rates, and substantial economic losses. With the removal of antimicrobial agents from chicken feed, there is an urgent need to find alternative approaches for NE control. Various approaches, including vaccination, prebiotics, probiotics, and plant-derived products, have been utilized to address NE in poultry management. To evaluate the efficacy of these preventive measures against NE, successful induction of NE is crucial to observe effects of these approaches in related studies. This study presents a comprehensive overview of the methods and approaches utilized for NE reproduction in related studies from 2004 to 2023. These considerations are the careful selection of a virulent Clostridium perfringens strain, preparation of challenge inoculum, choice of time and the route for challenge inoculum administration, and utilization of one or more predisposing factors to increase the rate of NE occurrence in birds under experiment. We also reviewed the different systems used for lesion scoring of NE-challenged birds. By gaining clarity on these fundamental parameters, researchers can make informed decisions regarding the selection of the most appropriate NE experimental design in their respective studies.
Collapse
|
10
|
Decker-Farrell AR, Sastra SA, Harimoto T, Hasselluhn MC, Palermo CF, Ballister ER, Badgley MA, Danino T, Olive KP. "Tumor-selective treatment of metastatic pancreatic cancer with an engineered, probiotic living drug". BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.02.592216. [PMID: 38746175 PMCID: PMC11092568 DOI: 10.1101/2024.05.02.592216] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/16/2024]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) poses significant challenges for effective treatment, with systemic chemotherapy often proving inadequate due to poor drug delivery and the tumor's immunosuppressive microenvironment. Engineered bacteria present a novel approach to target PDAC, leveraging their ability to colonize tumors and deliver therapeutic payloads. Here, we engineered probiotic Escherichia coli Nissle 1917 (EcN) to produce the pore-forming Theta toxin (Nis-Theta) and evaluated its efficacy in a preclinical model of PDAC. Probiotic administration resulted in selective colonization of tumor tissue, leading to improved overall survival compared to standard chemotherapy. Moreover, this strain exhibited cytotoxic effects on both primary and distant tumor lesions while sparing normal tissues. Importantly, treatment also modulated the tumor microenvironment by increasing anti-tumor immune cell populations and reducing immunosuppressive markers. These findings demonstrate the potential of engineered probiotic bacteria as a safe and effective therapeutic approach for PDAC, offering promise for improved patient outcomes.
Collapse
|
11
|
Kawamura T, Prah I, Mahazu S, Ablordey A, Saito R. Types A and F Clostridium perfringens in healthcare wastewater from Ghana. Appl Environ Microbiol 2023; 89:e0161923. [PMID: 38051072 PMCID: PMC10734495 DOI: 10.1128/aem.01619-23] [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: 09/14/2023] [Accepted: 10/18/2023] [Indexed: 12/07/2023] Open
Abstract
IMPORTANCE Clostridium perfringens causes gas gangrene and food poisoning in humans, and monitoring this bacterium is important for public health. Although whole-genome sequencing is useful to comprehensively understand the virulence, resistome, and global genetic relatedness of bacteria, limited genomic data from environmental sources and developing countries hamper our understanding of the richness of the intrinsic genomic diversity of this pathogen. Here, we successfully accumulated the genetic data on C. perfringens strains isolated from hospital effluent and provided the first evidence that predicted pathogenic C. perfringens may be disseminated in the clinical environment in Ghana. Our findings suggest the importance of risk assessment in the environment as well as the clinical setting to mitigate the potential outbreak of C. perfringens food poisoning in Ghana.
Collapse
Affiliation(s)
- Taira Kawamura
- Department of Molecular Microbiology and Immunology, Graduate School of Medicine and Dental Science, Tokyo Medical and Dental University, Tokyo, Japan
| | - Isaac Prah
- Department of Molecular Microbiology and Immunology, Graduate School of Medicine and Dental Science, Tokyo Medical and Dental University, Tokyo, Japan
| | - Samiratu Mahazu
- Department of Molecular Microbiology and Immunology, Graduate School of Medicine and Dental Science, Tokyo Medical and Dental University, Tokyo, Japan
| | - Anthony Ablordey
- Department of Bacteriology, Noguchi Memorial Institute for Medical Research, University of Ghana, Accra, Ghana
| | - Ryoichi Saito
- Department of Molecular Microbiology and Immunology, Graduate School of Medicine and Dental Science, Tokyo Medical and Dental University, Tokyo, Japan
| |
Collapse
|
12
|
Wang G, Liu Y, Deng L, Liu H, Deng X, Li Q, Feng H, Guo Z, Qiu J. Repurposing rabeprazole sodium as an anti-Clostridium perfringens drug by inhibiting perfringolysin O. J Appl Microbiol 2023; 134:lxad273. [PMID: 38017630 DOI: 10.1093/jambio/lxad273] [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: 06/12/2023] [Revised: 10/10/2023] [Accepted: 11/27/2023] [Indexed: 11/30/2023]
Abstract
AIMS Clostridium perfringens infections affect food safety, human health, and the development of the poultry feed industry. Anti-virulence is an alternative strategy to develop new drug. Perfringolysin O (PFO) is an exotoxin of C. perfringens that has been demonstrated to play critical roles in the pathogenesis of this organism, promising it an attractive target to explore drugs to combat C. perfringens infection. METHODS AND RESULTS Based on an activity-based screening, we identified six PFO inhibitors from the Food and Drug Administration (FDA)-approved drug library, among which rabeprazole sodium (RS) showed an optimal inhibitory effect with an IC50 of 1.82 ± 0.746 µg ml-1. The GLY57, ASP58, SER190, SER193-194, ASN199, GLU204, ASN377, THR379, and ALA200 in PFO interacted with RS during binding based on an energy analysis and H-bond analysis. This interaction blocked the oligomer formation of PFO, thereby inhibiting its cytotoxicity. RS treatment significantly increased the survival rate and alleviated pathological damage in C. perfringens or PFO-treated Galleria mellonella. CONCLUSIONS RS could potentially be used as a candidate drug for treating C. perfringens infection.
Collapse
Affiliation(s)
- Guizhen Wang
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun 130012, China
- Measurement Biotechnique Research Center, College of Biological and Food Engineering, Jilin Engineering Normal University, Changchun 130052, China
- Key Laboratory for Molecular Enzymology and Engineering of Ministry of Education, School of Life Sciences, Jilin University, Changchun 130012, China
| | - Yan Liu
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun 130012, China
| | - Le Deng
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun 130012, China
| | - Hongtao Liu
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun 130012, China
| | - Xuming Deng
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun 130012, China
| | - Quanshun Li
- Key Laboratory for Molecular Enzymology and Engineering of Ministry of Education, School of Life Sciences, Jilin University, Changchun 130012, China
| | - Haihua Feng
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun 130012, China
| | - Zhimin Guo
- Department of Laboratory Medicine, Infectious Diseases and Pathogen Biology Center, The First Hospital of Jilin University, Changchun 130021, China
| | - Jiazhang Qiu
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun 130012, China
| |
Collapse
|
13
|
Machin JM, Kalli AC, Ranson NA, Radford SE. Protein-lipid charge interactions control the folding of outer membrane proteins into asymmetric membranes. Nat Chem 2023; 15:1754-1764. [PMID: 37710048 PMCID: PMC10695831 DOI: 10.1038/s41557-023-01319-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2022] [Accepted: 08/08/2023] [Indexed: 09/16/2023]
Abstract
Biological membranes consist of two leaflets of phospholipid molecules that form a bilayer, each leaflet comprising a distinct lipid composition. This asymmetry is created and maintained in vivo by dedicated biochemical pathways, but difficulties in creating stable asymmetric membranes in vitro have restricted our understanding of how bilayer asymmetry modulates the folding, stability and function of membrane proteins. In this study, we used cyclodextrin-mediated lipid exchange to generate liposomes with asymmetric bilayers and characterize the stability and folding kinetics of two bacterial outer membrane proteins (OMPs), OmpA and BamA. We found that excess negative charge in the outer leaflet of a liposome impedes their insertion and folding, while excess negative charge in the inner leaflet accelerates their folding relative to symmetric liposomes with the same membrane composition. Using molecular dynamics, mutational analysis and bioinformatics, we identified a positively charged patch critical for folding and stability. These results rationalize the well-known 'positive-outside' rule of OMPs and suggest insights into the mechanisms that drive OMP folding and assembly in vitro and in vivo.
Collapse
Affiliation(s)
- Jonathan M Machin
- Astbury Centre for Structural Molecular Biology, School of Molecular and Cellular Biology, Faculty of Biological Sciences, University of Leeds, Leeds, UK
| | - Antreas C Kalli
- Astbury Centre for Structural Molecular Biology, School of Molecular and Cellular Biology, Faculty of Biological Sciences, University of Leeds, Leeds, UK.
- Leeds Institute of Cardiovascular and Metabolic Medicine, School of Medicine, University of Leeds, Leeds, UK.
| | - Neil A Ranson
- Astbury Centre for Structural Molecular Biology, School of Molecular and Cellular Biology, Faculty of Biological Sciences, University of Leeds, Leeds, UK.
| | - Sheena E Radford
- Astbury Centre for Structural Molecular Biology, School of Molecular and Cellular Biology, Faculty of Biological Sciences, University of Leeds, Leeds, UK.
| |
Collapse
|
14
|
Kiu R, Shaw AG, Sim K, Acuna-Gonzalez A, Price CA, Bedwell H, Dreger SA, Fowler WJ, Cornwell E, Pickard D, Belteki G, Malsom J, Phillips S, Young GR, Schofield Z, Alcon-Giner C, Berrington JE, Stewart CJ, Dougan G, Clarke P, Douce G, Robinson SD, Kroll JS, Hall LJ. Particular genomic and virulence traits associated with preterm infant-derived toxigenic Clostridium perfringens strains. Nat Microbiol 2023; 8:1160-1175. [PMID: 37231089 PMCID: PMC10234813 DOI: 10.1038/s41564-023-01385-z] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2021] [Accepted: 04/17/2023] [Indexed: 05/27/2023]
Abstract
Clostridium perfringens is an anaerobic toxin-producing bacterium associated with intestinal diseases, particularly in neonatal humans and animals. Infant gut microbiome studies have recently indicated a link between C. perfringens and the preterm infant disease necrotizing enterocolitis (NEC), with specific NEC cases associated with overabundant C. perfringens termed C. perfringens-associated NEC (CPA-NEC). In the present study, we carried out whole-genome sequencing of 272 C. perfringens isolates from 70 infants across 5 hospitals in the United Kingdom. In this retrospective analysis, we performed in-depth genomic analyses (virulence profiling, strain tracking and plasmid analysis) and experimentally characterized pathogenic traits of 31 strains, including 4 from CPA-NEC patients. We found that the gene encoding toxin perfringolysin O, pfoA, was largely deficient in a human-derived hypovirulent lineage, as well as certain colonization factors, in contrast to typical pfoA-encoding virulent lineages. We determined that infant-associated pfoA+ strains caused significantly more cellular damage than pfoA- strains in vitro, and further confirmed this virulence trait in vivo using an oral-challenge C57BL/6 murine model. These findings suggest both the importance of pfoA+ C. perfringens as a gut pathogen in preterm infants and areas for further investigation, including potential intervention and therapeutic strategies.
Collapse
Affiliation(s)
- Raymond Kiu
- Gut Microbes and Health, Quadram Institute Bioscience, Norwich, UK
| | | | - Kathleen Sim
- Faculty of Medicine, Imperial College London, London, UK
| | | | | | - Harley Bedwell
- Gut Microbes and Health, Quadram Institute Bioscience, Norwich, UK
| | - Sally A Dreger
- Gut Microbes and Health, Quadram Institute Bioscience, Norwich, UK
| | - Wesley J Fowler
- Gut Microbes and Health, Quadram Institute Bioscience, Norwich, UK
| | - Emma Cornwell
- Faculty of Medicine, Imperial College London, London, UK
| | - Derek Pickard
- Department of Medicine, University of Cambridge, Cambridge, UK
| | - Gusztav Belteki
- Neonatal Intensive Care Unit, The Rosie Hospital, Cambridge, UK
| | - Jennifer Malsom
- Gut Microbes and Health, Quadram Institute Bioscience, Norwich, UK
| | - Sarah Phillips
- Gut Microbes and Health, Quadram Institute Bioscience, Norwich, UK
| | - Gregory R Young
- Hub for Biotechnology in the Built Environment, Northumbria University, Newcastle upon Tyne, UK
| | - Zoe Schofield
- Gut Microbes and Health, Quadram Institute Bioscience, Norwich, UK
| | | | - Janet E Berrington
- Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne, UK
- Newcastle Neonatal Services, Newcastle upon Tyne NHS Foundation Trust, Newcastle upon Tyne, UK
| | - Christopher J Stewart
- Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne, UK
- Newcastle Neonatal Services, Newcastle upon Tyne NHS Foundation Trust, Newcastle upon Tyne, UK
| | - Gordon Dougan
- Department of Medicine, University of Cambridge, Cambridge, UK
| | - Paul Clarke
- Norfolk and Norwich University Hospital, Norwich, UK
- Norwich Medical School, University of East Anglia, Norwich, UK
| | - Gillian Douce
- Institute of Infection, Immunity and Inflammation, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, UK
| | - Stephen D Robinson
- Gut Microbes and Health, Quadram Institute Bioscience, Norwich, UK
- School of Biological Sciences, University of East Anglia, Norwich, UK
| | - J Simon Kroll
- Faculty of Medicine, Imperial College London, London, UK
| | - Lindsay J Hall
- Gut Microbes and Health, Quadram Institute Bioscience, Norwich, UK.
- Norwich Medical School, University of East Anglia, Norwich, UK.
- Intestinal Microbiome, School of Life Sciences, ZIEL-Institute for Food & Health, Technical University of Munich, Freising, Germany.
| |
Collapse
|
15
|
Suzaki A, Hayakawa S. Clinical and Microbiological Features of Fulminant Haemolysis Caused by Clostridium perfringens Bacteraemia: Unknown Pathogenesis. Microorganisms 2023; 11:microorganisms11040824. [PMID: 37110247 PMCID: PMC10143116 DOI: 10.3390/microorganisms11040824] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2023] [Revised: 03/16/2023] [Accepted: 03/16/2023] [Indexed: 04/29/2023] Open
Abstract
Bacteraemia brought on by Clostridium perfringens has a very low incidence but is severe and fatal in fifty per cent of cases. C. perfringens is a commensal anaerobic bacterium found in the environment and in the intestinal tracts of animals; it is known to produce six major toxins: α-toxin, β-toxin, ε-toxin, and others. C. perfringens is classified into seven types, A, B, C, D, E, F and G, according to its ability to produce α-toxin, enterotoxin, and necrotising enterotoxin. The bacterial isolates from humans include types A and F, which cause gas gangrene, hepatobiliary infection, and sepsis; massive intravascular haemolysis (MIH) occurs in 7-15% of C. perfringens bacteraemia cases, resulting in a rapid progression to death. We treated six patients with MIH at a single centre in Japan; however, unfortunately, they all passed away. From a clinical perspective, MIH patients tended to be younger and were more frequently male; however, there was no difference in the toxin type or genes of the bacterial isolates. In MIH cases, the level of θ-toxin in the culture supernatant of clinical isolates was proportional to the production of inflammatory cytokines in the peripheral blood, suggesting the occurrence of an intense cytokine storm. Severe and systemic haemolysis is considered an evolutionary maladaptation as it leads to the host's death before the bacterium obtains the benefit of iron utilisation from erythrocytes. The disease's extraordinarily quick progression and dismal prognosis necessitate a straightforward and expedient diagnosis and treatment. However, a reliable standard of diagnosis and treatment has yet to be put forward due to the lack of sufficient case analysis.
Collapse
Affiliation(s)
- Ai Suzaki
- Department of Pathology and Microbiology, Nihon University School of Medicine, 30-1 Ohyaguchi Kamicho, Itabashiku, Tokyo 173-8610, Japan
| | - Satoshi Hayakawa
- Department of Pathology and Microbiology, Nihon University School of Medicine, 30-1 Ohyaguchi Kamicho, Itabashiku, Tokyo 173-8610, Japan
| |
Collapse
|
16
|
Suzaki A, Komine-Aizawa S, Nishiyama H, Hayakawa S. Massive intravascular hemolysis is an important factor in Clostridium perfringens-induced bacteremia. Intern Emerg Med 2022; 17:1959-1967. [PMID: 35962901 DOI: 10.1007/s11739-022-03036-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/13/2021] [Accepted: 06/16/2022] [Indexed: 11/05/2022]
Abstract
Clostridium perfringens bacteremia is rare but often fatal. In particular, once bacteremia with massive intravascular hemolysis (MIH) occurs, the mortality rate is extremely high. However, because of its rarity, the detailed pathophysiology of this fulminant form of bacteremia is unclear. To elucidate the detailed pathogenesis of MIH, we retrospectively reviewed the data of all patients with C. perfringens bacteremia from two university hospitals from 2000 to 2014. The medical records and laboratory data of 60 patients with bacteremia, including 6 patients with MIH and 54 patients without MIH, were analyzed. Patients with MIH had higher rates of intense pain at onset, impaired consciousness, shock at presentation, hematuria, metabolic acidosis, and gas formation than patients without MIH. The antibiotic susceptibility of the clinical isolates was not significantly different between the two groups. All patients with MIH, although treated with appropriate antimicrobial agents, died within 26 h of admission due to rapidly progressive acute lung injury or acute respiratory distress syndrome, and the median time from arrival at the hospital to death was only 4 h and 20 min. When clinicians observe intravascular hemolysis in blood samples from patients with characteristic symptoms of MIH, they should prepare for a severe disease outcome. The underlying pathophysiology of fulminant cases must be investigated.
Collapse
Affiliation(s)
- Ai Suzaki
- Division of Microbiology, Department of Pathology and Microbiology, Nihon University School of Medicine, 30-1 Oyaguchi-Kamicho, Itabashi, Tokyo, 173-8610, Japan.
- Department of General Medicine, Nihon University Hospital, Tokyo, Japan.
| | - Shihoko Komine-Aizawa
- Division of Microbiology, Department of Pathology and Microbiology, Nihon University School of Medicine, 30-1 Oyaguchi-Kamicho, Itabashi, Tokyo, 173-8610, Japan
| | - Hiroyuki Nishiyama
- Department of Clinical Laboratory, Nihon University Itabashi Hospital, Tokyo, Japan
| | - Satoshi Hayakawa
- Division of Microbiology, Department of Pathology and Microbiology, Nihon University School of Medicine, 30-1 Oyaguchi-Kamicho, Itabashi, Tokyo, 173-8610, Japan
| |
Collapse
|
17
|
Soto LF, Romaní AC, Jiménez-Avalos G, Silva Y, Ordinola-Ramirez CM, Lopez Lapa RM, Requena D. Immunoinformatic analysis of the whole proteome for vaccine design: An application to Clostridium perfringens. Front Immunol 2022; 13:942907. [PMID: 36110855 PMCID: PMC9469472 DOI: 10.3389/fimmu.2022.942907] [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: 05/13/2022] [Accepted: 08/02/2022] [Indexed: 11/21/2022] Open
Abstract
Clostridium perfringens is a dangerous bacterium and known biological warfare weapon associated with several diseases, whose lethal toxins can produce necrosis in humans. However, there is no safe and fully effective vaccine against C. perfringens for humans yet. To address this problem, we computationally screened its whole proteome, identifying highly immunogenic proteins, domains, and epitopes. First, we identified that the proteins with the highest epitope density are Collagenase A, Exo-alpha-sialidase, alpha n-acetylglucosaminidase and hyaluronoglucosaminidase, representing potential recombinant vaccine candidates. Second, we further explored the toxins, finding that the non-toxic domain of Perfringolysin O is enriched in CTL and HTL epitopes. This domain could be used as a potential sub-unit vaccine to combat gas gangrene. And third, we designed a multi-epitope protein containing 24 HTL-epitopes and 34 CTL-epitopes from extracellular regions of transmembrane proteins. Also, we analyzed the structural properties of this novel protein using molecular dynamics. Altogether, we are presenting a thorough immunoinformatic exploration of the whole proteome of C. perfringens, as well as promising whole-protein, domain-based and multi-epitope vaccine candidates. These can be evaluated in preclinical trials to assess their immunogenicity and protection against C. perfringens infection.
Collapse
Affiliation(s)
- Luis F. Soto
- Escuela Profesional de Genética y Biotecnología, Facultad de Ciencias Biológicas, Universidad Nacional Mayor de San Marcos, Lima, Peru
| | - Ana C. Romaní
- Escuela Profesional de Genética y Biotecnología, Facultad de Ciencias Biológicas, Universidad Nacional Mayor de San Marcos, Lima, Peru
| | - Gabriel Jiménez-Avalos
- Departamento de Ciencias Celulares y Moleculares, Laboratorio de Bioinformática, Biología Molecular y Desarrollos Tecnológicos, Facultad de Ciencias y Filosofía, Universidad Peruana Cayetano Heredia (UPCH), Lima, Peru
| | - Yshoner Silva
- Departamento de Salud Pública, Facultad de Ciencias de la Salud, Universidad Nacional Toribio Rodríguez de Mendoza de Amazonas, Chachapoyas, Peru
| | - Carla M. Ordinola-Ramirez
- Departamento de Salud Pública, Facultad de Ciencias de la Salud, Universidad Nacional Toribio Rodríguez de Mendoza de Amazonas, Chachapoyas, Peru
| | - Rainer M. Lopez Lapa
- Departamento de Salud Pública, Facultad de Ciencias de la Salud, Universidad Nacional Toribio Rodríguez de Mendoza de Amazonas, Chachapoyas, Peru
- Instituto de Ganadería y Biotecnología, Universidad Nacional Toribio Rodríguez de Mendoza de Amazonas, Chachapoyas, Peru
| | - David Requena
- Laboratory of Cellular Biophysics, The Rockefeller University, New York, NY, United States
- *Correspondence: David Requena,
| |
Collapse
|
18
|
McGuinness C, Walsh JC, Bayly-Jones C, Dunstone MA, Christie MP, Morton CJ, Parker MW, Böcking T. Single-molecule analysis of the entire perfringolysin O pore formation pathway. eLife 2022; 11:e74901. [PMID: 36000711 PMCID: PMC9457685 DOI: 10.7554/elife.74901] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2021] [Accepted: 08/16/2022] [Indexed: 11/20/2022] Open
Abstract
The cholesterol-dependent cytolysin perfringolysin O (PFO) is secreted by Clostridium perfringens as a bacterial virulence factor able to form giant ring-shaped pores that perforate and ultimately lyse mammalian cell membranes. To resolve the kinetics of all steps in the assembly pathway, we have used single-molecule fluorescence imaging to follow the dynamics of PFO on dye-loaded liposomes that lead to opening of a pore and release of the encapsulated dye. Formation of a long-lived membrane-bound PFO dimer nucleates the growth of an irreversible oligomer. The growing oligomer can insert into the membrane and open a pore at stoichiometries ranging from tetramers to full rings (~35 mers), whereby the rate of insertion increases linearly with the number of subunits. Oligomers that insert before the ring is complete continue to grow by monomer addition post insertion. Overall, our observations suggest that PFO membrane insertion is kinetically controlled.
Collapse
Affiliation(s)
- Conall McGuinness
- EMBL Australia Node in Single Molecule Science, School of Biomedical Sciences, University of New South WalesSydneyAustralia
| | - James C Walsh
- EMBL Australia Node in Single Molecule Science, School of Biomedical Sciences, University of New South WalesSydneyAustralia
| | - Charles Bayly-Jones
- Biomedicine Discovery Institute, Department of Biochemistry and Molecular Biology, Monash UniversityMelbourneAustralia
| | - Michelle A Dunstone
- Biomedicine Discovery Institute, Department of Biochemistry and Molecular Biology, Monash UniversityMelbourneAustralia
| | - Michelle P Christie
- Department of Biochemistry and Pharmacology, Bio21 Molecular Science and Biotechnology Institute, University of MelbourneVictoriaAustralia
| | - Craig J Morton
- Department of Biochemistry and Pharmacology, Bio21 Molecular Science and Biotechnology Institute, University of MelbourneVictoriaAustralia
| | - Michael W Parker
- Department of Biochemistry and Pharmacology, Bio21 Molecular Science and Biotechnology Institute, University of MelbourneVictoriaAustralia
- Structural Biology Unit, St. Vincent’s Institute of Medical ResearchVictoriaAustralia
| | - Till Böcking
- EMBL Australia Node in Single Molecule Science, School of Biomedical Sciences, University of New South WalesSydneyAustralia
| |
Collapse
|
19
|
Piceatannol Alleviates Clostridium perfringens Virulence by Inhibiting Perfringolysin O. MOLECULES (BASEL, SWITZERLAND) 2022; 27:molecules27165145. [PMID: 36014383 PMCID: PMC9415171 DOI: 10.3390/molecules27165145] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/26/2022] [Revised: 08/08/2022] [Accepted: 08/11/2022] [Indexed: 11/25/2022]
Abstract
Clostridium perfringens (C. perfringens) is an important foodborne pathogen that can cause diseases such as gas gangrene and necrotizing enteritis in a variety of economic animals, seriously affecting public health and the economic benefits and healthy development of the livestock and poultry breeding industry. Perfringolysin O (PFO) is an important virulence factor of C. perfringens and plays critical roles in necrotic enteritis and gas gangrene, rendering it an ideal target for developing new drugs against infections caused by this pathogen. In this study, based on biological activity inhibition assays, oligomerization tests and computational biology assays, we found that the foodborne natural component piceatannol reduced pore-forming activity with an inhibitory ratio of 83.84% in the concentration of 16 µg/mL (IC50 = 7.83 µg/mL) by binding with PFO directly and changing some of its secondary structures, including 3-Helix, A-helix, bend, and in turn, ultimately affecting oligomer formation. Furthermore, we confirmed that piceatannol protected human intestinal epithelial cells from the damage induced by PFO with LDH release reduced by 38.44% at 16 µg/mL, based on a cytotoxicity test. By performing an animal experiment, we found the C. perfringens clones showed an approximate 10-fold reduction in infected mice. These results suggest that piceatannol may be a candidate for anti-C. perfringens drug development.
Collapse
|
20
|
Ueda K, Kawahara K, Kimoto N, Yamaguchi Y, Yamada K, Oki H, Yoshida T, Matsuda S, Matsumoto Y, Motooka D, Kawatsu K, Iida T, Nakamura S, Ohkubo T, Yonogi S. Analysis of the complete genome sequences of Clostridium perfringens strains harbouring the binary enterotoxin BEC gene and comparative genomics of pCP13-like family plasmids. BMC Genomics 2022; 23:226. [PMID: 35321661 PMCID: PMC8941779 DOI: 10.1186/s12864-022-08453-4] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2021] [Accepted: 03/08/2022] [Indexed: 11/10/2022] Open
Abstract
Background BEC-producing Clostridium perfringens is a causative agent of foodborne gastroenteritis. It was first reported in 2014, and since then, several isolates have been identified in Japan and the United Kingdom. The novel binary ADP-ribosylating toxin BEC, which consists of two components (BECa and BECb), is encoded on a plasmid that is similar to pCP13 and harbours a conjugation locus, called Pcp, encoding homologous proteins of the type 4 secretion system. Despite the high in vitro conjugation frequency of pCP13, its dissemination and that of related plasmids, including bec-harbouring plasmids, in the natural environment have not been characterised. This lack of knowledge has limited our understanding of the genomic epidemiology of bec-harbouring C. perfringens strains. Results In this study, we determined the complete genome sequences of five bec-harbouring C. perfringens strains isolated from 2009 to 2019. Each isolate contains a ~ 3.36 Mbp chromosome and 1–3 plasmids of either the pCW3-like family, pCP13-like family, or an unknown family, and the bec-encoding region in all five isolates was located on a ~ 54 kbp pCP13-like plasmid. Phylogenetic and SNP analyses of these complete genome sequences and the 211 assembled C. perfringens genomes in GenBank showed that although these bec-harbouring strains were split into two phylogenetic clades, the sequences of the bec-encoding plasmids were nearly identical (>99.81%), with a significantly smaller SNP accumulation rate than that of their chromosomes. Given that the Pcp locus is conserved in these pCP13-like plasmids, we propose a mechanism in which the plasmids were disseminated by horizontal gene transfer. Data mining showed that strains carrying pCP13-like family plasmids were unexpectedly common (58/216 strains) and widely disseminated among the various C. perfringens clades. Although these plasmids possess a conserved Pcp locus, their ‘accessory regions’ can accommodate a wide variety of genes, including virulence-associated genes, such as becA/becB and cbp2. These results suggest that this family of plasmids can integrate various foreign genes and is transmissible among C. perfringens strains. Conclusion This study demonstrates the potential significance of pCP13-like plasmids, including bec-encoding plasmids, for the characterisation and monitoring of the dissemination of pathogenic C. perfringens strains. Supplementary Information The online version contains supplementary material available at 10.1186/s12864-022-08453-4.
Collapse
Affiliation(s)
- Kengo Ueda
- Laboratory of Biophysical Chemistry, Graduate School of Pharmaceutical Sciences, Osaka University, 1-6 Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Kazuki Kawahara
- Laboratory of Biophysical Chemistry, Graduate School of Pharmaceutical Sciences, Osaka University, 1-6 Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Narumi Kimoto
- Laboratory of Biophysical Chemistry, Graduate School of Pharmaceutical Sciences, Osaka University, 1-6 Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Yusuke Yamaguchi
- Laboratory of Biophysical Chemistry, Graduate School of Pharmaceutical Sciences, Osaka University, 1-6 Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Kazuhiro Yamada
- Department of Microbiology and Medical Zoology, Aichi Prefectural Institute of Public Health, 7-6 Nagare, Tsujicho, Kita-ku, Nagoya, Aichi, 462-8576, Japan
| | - Hiroya Oki
- Department of Infection Metagenomics, Genome Information Research Center, Research Institute for Microbial Diseases (RIMD), Osaka University, 3-1 Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Takuya Yoshida
- Laboratory of Biophysical Chemistry, Graduate School of Pharmaceutical Sciences, Osaka University, 1-6 Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Shigeaki Matsuda
- Department of Bacterial Infection, Research Institute for Microbial Disease (RIMD), Osaka University, 3-1 Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Yuki Matsumoto
- Department of Infection Metagenomics, Genome Information Research Center, Research Institute for Microbial Diseases (RIMD), Osaka University, 3-1 Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Daisuke Motooka
- Department of Infection Metagenomics, Genome Information Research Center, Research Institute for Microbial Diseases (RIMD), Osaka University, 3-1 Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Kentaro Kawatsu
- Division of Microbiology, Osaka Institute of Public Health, 1-3-69 Nakamichi, Higashinari-ku, Osaka, Osaka, 537-0025, Japan
| | - Tetsuya Iida
- Department of Bacterial Infection, Research Institute for Microbial Disease (RIMD), Osaka University, 3-1 Yamadaoka, Suita, Osaka, 565-0871, Japan.,Center for Infectious Disease Education and Research (CiDER), Osaka University, 3-1 Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Shota Nakamura
- Department of Infection Metagenomics, Genome Information Research Center, Research Institute for Microbial Diseases (RIMD), Osaka University, 3-1 Yamadaoka, Suita, Osaka, 565-0871, Japan.,Center for Infectious Disease Education and Research (CiDER), Osaka University, 3-1 Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Tadayasu Ohkubo
- Laboratory of Biophysical Chemistry, Graduate School of Pharmaceutical Sciences, Osaka University, 1-6 Yamadaoka, Suita, Osaka, 565-0871, Japan.
| | - Shinya Yonogi
- Department of Bacterial Infection, Research Institute for Microbial Disease (RIMD), Osaka University, 3-1 Yamadaoka, Suita, Osaka, 565-0871, Japan. .,Division of Microbiology, Osaka Institute of Public Health, 1-3-69 Nakamichi, Higashinari-ku, Osaka, Osaka, 537-0025, Japan.
| |
Collapse
|
21
|
Immunogenic and neutralization efficacy of recombinant perfringolysin O of Clostridium perfringens and its C-terminal receptor-binding domain in a murine model. Immunol Res 2022; 70:240-255. [PMID: 35032316 PMCID: PMC8760870 DOI: 10.1007/s12026-021-09254-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2021] [Accepted: 11/27/2021] [Indexed: 11/05/2022]
Abstract
Clostridium perfringens is a Gram-positive anaerobe ubiquitously present in different environments, including the gut of humans and animals. C. perfringens have been classified in the seven toxinotypes based on the secreted toxins that cause different diseases in humans and animals. Perfringolysin O (PFO), a cholesterol-dependent pore-forming cytolysin, is one of the potent toxins secreted by almost all C. perfringens isolates. The PFO acts in synergy with α-toxin in the progression of gas gangrene in humans and necrohemorrhagic enteritis in the calves.C. perfringens infections spread very fast, and the animals die within a few hours of the onset of infection. This necessitates the use of vaccines to control clostridial infections. Though the vaccine potential of other toxins has been reported, PFO has remained unexplored. The present study describes the immunogenic and protective potential of native recombinant PFO (WTrPFO). Since the PFO is toxic to the host cells, the non-toxic C-terminal domain of PFO (rPFOC-ter) was also assessed for its immunogenicity and protective efficacy. Immunization of mice with the purified soluble recombinant histidine-tagged WTrPFO and rPFOC-ter, expressed in E. coli, generated robust mixed immune response and T cell memory. Pre-incubation of the WTrPFO with anti-WTrPFO and rPFOC-ter antisera negated its hemolytic activity in mice RBCs, as well as its cytotoxic effect in mice peritoneal macrophages in vitro. Thus, immunization with the WTrPFO and its non-toxic C-terminal domain generated neutralizing antibodies, suggesting their vaccine potential against the PFO. Thus, the non-toxic C-terminal domain of PFO could serve as an alternative to PFO as a vaccine candidate.
Collapse
|
22
|
Design and in situ biosynthesis of precision therapies against gastrointestinal pathogens. CURRENT OPINION IN PHYSIOLOGY 2021. [DOI: 10.1016/j.cophys.2021.06.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
|
23
|
Suzaki A, Ohtani K, Komine-Aizawa S, Matsumoto A, Kamiya S, Hayakawa S. Pathogenic Characterization of Clostridium perfringens Strains Isolated From Patients With Massive Intravascular Hemolysis. Front Microbiol 2021; 12:713509. [PMID: 34385995 PMCID: PMC8353389 DOI: 10.3389/fmicb.2021.713509] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2021] [Accepted: 07/02/2021] [Indexed: 11/13/2022] Open
Abstract
Sepsis caused by Clostridium perfringens infection is rare but often fatal. The most serious complication leading to poor prognosis is massive intravascular hemolysis (MIH). However, the molecular mechanism underlying this fulminant form of hemolysis is unclear. In the present study, we employed 11 clinical strains isolated from patients with C. perfringens septicemia and subdivided these isolates into groups H and NH: septicemia with (n = 5) or without (n = 6) MIH, respectively. To elucidate the major pathogenic factors of MIH, biological features were compared between these groups. The isolates of two groups did not differ in growth rate, virulence-related gene expression, or phospholipase C (CPA) production. Erythrocyte hemolysis was predominantly observed in culture supernatants of the strains in group H, and the human erythrocyte hemolysis rate was significantly correlated with perfringolysin O (PFO) production. Correlations were also found among PFO production, human peripheral blood mononuclear cell (PBMC) cytotoxicity, and production of interleukin-6 (IL-6) and interleukin-8 (IL-8) by human PBMCs. Analysis of proinflammatory cytokines showed that PFO induced tumor necrosis factor-α (TNF-α), IL-5, IL-6, and IL-8 production more strongly than did CPA. PFO exerted potent cytotoxic and proinflammatory cytokine induction effects on human blood cells. PFO may be a major virulence factor of sepsis with MIH, and potent proinflammatory cytokine production induced by PFO may influence the rapid progression of this fatal disease caused by C. perfringens.
Collapse
Affiliation(s)
- Ai Suzaki
- Division of Microbiology, Department of Pathology and Microbiology, Nihon University School of Medicine, Tokyo, Japan.,Division of General Medicine, Department of Medicine, Nihon University School of Medicine, Tokyo, Japan
| | - Kaori Ohtani
- Division of Host Defense Mechanism, Department of Bacteriology and Bacterial Infection, Tokai University School of Medicine, Isehara, Japan
| | - Shihoko Komine-Aizawa
- Division of Microbiology, Department of Pathology and Microbiology, Nihon University School of Medicine, Tokyo, Japan
| | - Asami Matsumoto
- R&D Division, Miyarisan Pharmaceutical Co., LTD., Saitama, Japan
| | - Shigeru Kamiya
- R&D Division, Miyarisan Pharmaceutical Co., LTD., Saitama, Japan.,Department of Infectious Diseases, Kyorin University School of Medicine, Tokyo, Japan
| | - Satoshi Hayakawa
- Division of Microbiology, Department of Pathology and Microbiology, Nihon University School of Medicine, Tokyo, Japan
| |
Collapse
|
24
|
Draberova L, Tumova M, Draber P. Molecular Mechanisms of Mast Cell Activation by Cholesterol-Dependent Cytolysins. Front Immunol 2021; 12:670205. [PMID: 34248949 PMCID: PMC8260682 DOI: 10.3389/fimmu.2021.670205] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2021] [Accepted: 06/08/2021] [Indexed: 12/23/2022] Open
Abstract
Mast cells are potent immune sensors of the tissue microenvironment. Within seconds of activation, they release various preformed biologically active products and initiate the process of de novo synthesis of cytokines, chemokines, and other inflammatory mediators. This process is regulated at multiple levels. Besides the extensively studied IgE and IgG receptors, toll-like receptors, MRGPR, and other protein receptor signaling pathways, there is a critical activation pathway based on cholesterol-dependent, pore-forming cytolytic exotoxins produced by Gram-positive bacterial pathogens. This pathway is initiated by binding the exotoxins to the cholesterol-rich membrane, followed by their dimerization, multimerization, pre-pore formation, and pore formation. At low sublytic concentrations, the exotoxins induce mast cell activation, including degranulation, intracellular calcium concentration changes, and transcriptional activation, resulting in production of cytokines and other inflammatory mediators. Higher toxin concentrations lead to cell death. Similar activation events are observed when mast cells are exposed to sublytic concentrations of saponins or some other compounds interfering with the membrane integrity. We review the molecular mechanisms of mast cell activation by pore-forming bacterial exotoxins, and other compounds inducing cholesterol-dependent plasma membrane perturbations. We discuss the importance of these signaling pathways in innate and acquired immunity.
Collapse
Affiliation(s)
- Lubica Draberova
- Department of Signal Transduction, Institute of Molecular Genetics of the Czech Academy of Sciences, Prague, Czechia
| | - Magda Tumova
- Department of Signal Transduction, Institute of Molecular Genetics of the Czech Academy of Sciences, Prague, Czechia
| | - Petr Draber
- Department of Signal Transduction, Institute of Molecular Genetics of the Czech Academy of Sciences, Prague, Czechia
| |
Collapse
|
25
|
Abdel-Glil MY, Thomas P, Linde J, Busch A, Wieler LH, Neubauer H, Seyboldt C. Comparative in silico genome analysis of Clostridium perfringens unravels stable phylogroups with different genome characteristics and pathogenic potential. Sci Rep 2021; 11:6756. [PMID: 33762628 PMCID: PMC7991664 DOI: 10.1038/s41598-021-86148-8] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2020] [Accepted: 03/11/2021] [Indexed: 12/16/2022] Open
Abstract
Clostridium perfringens causes a plethora of devastating infections, with toxin production being the underlying mechanism of pathogenicity in various hosts. Genomic analyses of 206 public-available C. perfringens strains´ sequence data identified a substantial degree of genomic variability in respect to episome content, chromosome size and mobile elements. However, the position and order of the local collinear blocks on the chromosome showed a considerable degree of preservation. The strains were divided into five stable phylogroups (I–V). Phylogroup I contained human food poisoning strains with chromosomal enterotoxin (cpe) and a Darmbrand strain characterized by a high frequency of mobile elements, a relatively small genome size and a marked loss of chromosomal genes, including loss of genes encoding virulence traits. These features might correspond to the adaptation of these strains to a particular habitat, causing human foodborne illnesses. This contrasts strains that belong to phylogroup II where the genome size points to the acquisition of genetic material. Most strains of phylogroup II have been isolated from enteric lesions in horses and dogs. Phylogroups III, IV and V are heterogeneous groups containing a variety of different strains, with phylogroup III being the most abundant (65.5%). In conclusion, C. perfringens displays five stable phylogroups reflecting different disease involvements, prompting further studies on the evolution of this highly important pathogen.
Collapse
Affiliation(s)
- Mostafa Y Abdel-Glil
- Institute of Bacterial Infections and Zoonoses, Friedrich-Loeffler-Institut, Naumburger Str. 96A, 07743, Jena, Germany. .,Department of Pathology, Faculty of Veterinary Medicine, Zagazig University, Zagazig, Sharkia Province, Egypt.
| | - Prasad Thomas
- Institute of Bacterial Infections and Zoonoses, Friedrich-Loeffler-Institut, Naumburger Str. 96A, 07743, Jena, Germany.,Division of Bacteriology and Mycology, ICAR-Indian Veterinary Research Institute, Izatnagar, Bareilly, 243122, India
| | - Jörg Linde
- Institute of Bacterial Infections and Zoonoses, Friedrich-Loeffler-Institut, Naumburger Str. 96A, 07743, Jena, Germany
| | - Anne Busch
- Institute of Bacterial Infections and Zoonoses, Friedrich-Loeffler-Institut, Naumburger Str. 96A, 07743, Jena, Germany.,Department of Anaesthesiology and Intensive Care Medicine, University Hospital Jena, Am Klinikum 1, 07747, Jena, Germany
| | - Lothar H Wieler
- Robert Koch-Institut, Nordufer 20, 13353, Berlin, Germany.,Institute of Microbiology and Epizootics, Department of Veterinary Medicine, Freie Universität, Robert-von-Ostertag-Str. 7-13, Building 35, 14163, Berlin, Germany
| | - Heinrich Neubauer
- Institute of Bacterial Infections and Zoonoses, Friedrich-Loeffler-Institut, Naumburger Str. 96A, 07743, Jena, Germany
| | - Christian Seyboldt
- Institute of Bacterial Infections and Zoonoses, Friedrich-Loeffler-Institut, Naumburger Str. 96A, 07743, Jena, Germany.
| |
Collapse
|
26
|
Takehara M, Kobayashi K, Nagahama M. Toll-Like Receptor 4 Protects Against Clostridium perfringens Infection in Mice. Front Cell Infect Microbiol 2021; 11:633440. [PMID: 33763386 PMCID: PMC7982660 DOI: 10.3389/fcimb.2021.633440] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2020] [Accepted: 02/03/2021] [Indexed: 11/13/2022] Open
Abstract
Toll-like receptor 4 (TLR4) has been reported to protect against Gram-negative bacteria by acting as a pathogen recognition receptor that senses mainly lipopolysaccharide (LPS) from Gram-negative bacteria. However, the role of TLR4 in Gram-positive bacterial infection is less well understood. Clostridium perfringens type A is a Gram-positive bacterium that causes gas gangrene characterized by severe myonecrosis. It was previously demonstrated that C. perfringens θ-toxin is a TLR4 agonist, but the role of TLR4 in C. perfringens infection is unclear. Here, TLR4-defective C3H/HeJ mice infected with C. perfringens showed a remarkable decrease in survival rate, an increase in viable bacterial counts, and accelerated destruction of myofibrils at the infection site compared with wild-type C3H/HeN mice. These results demonstrate that TLR4 plays an important role in the elimination of C. perfringens. Remarkable increases in levels of inflammatory cytokines, such as interleukin-1β (IL-1β), interleukin-6 (IL-6), and granulocyte colony-stimulating factor (G-CSF), were observed in C. perfringens-infected C3H/HeN mice, whereas the increases were limited in C3H/HeJ mice. Generally, increased G-CSF accelerates granulopoiesis in the bone marrow and the spleen to exacerbate neutrophil production, resulting in elimination of bacteria. The number of neutrophils in the spleen was increased in C. perfringens-infected C3H/HeN mice compared with non-infected mice, while the increase was lower in C. perfringens-infected C3H/HeJ mice. Furthermore, DNA microarray analysis revealed that the mutation in TLR4 partially affects host gene expression during C. perfringens infection. Together, our results illustrate that TLR4 is crucial for the innate ability to eliminate C. perfringens.
Collapse
Affiliation(s)
- Masaya Takehara
- Department of Microbiology, Faculty of Pharmaceutical Sciences, Tokushima Bunri University, Yamashiro-cho, Japan
| | - Keiko Kobayashi
- Department of Microbiology, Faculty of Pharmaceutical Sciences, Tokushima Bunri University, Yamashiro-cho, Japan
| | - Masahiro Nagahama
- Department of Microbiology, Faculty of Pharmaceutical Sciences, Tokushima Bunri University, Yamashiro-cho, Japan
| |
Collapse
|
27
|
Zgheib H, Belguesmia Y, Boukherroub R, Drider D. Alginate Nanoparticles Enhance Anti-Clostridium perfringens Activity of the Leaderless Two-Peptide Enterocin DD14 and Affect Expression of Some Virulence Factors. Probiotics Antimicrob Proteins 2021; 13:1213-1227. [PMID: 33481224 DOI: 10.1007/s12602-020-09730-y] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/01/2020] [Indexed: 02/06/2023]
Abstract
Here, we report a novel approach to improve the anti-Clostridium perfringens activity of the leaderless two-peptide enterocin 14 (EntDD14), produced by Enterococcus faecalis 14. This strategy consists of loading EntDD14 onto alginate nanoparticles (Alg NPs), which are made of a safe polymer. The resulting formulation (EntDD14/Alg NPs) was able to reduce up to four times the minimum inhibitory concentration (MIC) of EntDD14 against C. perfringens pathogenic strains isolated from a chicken affected by necrotic enteritis (NE). Interestingly, this formulation remained active under conditions mimicking the human and chicken gastric tract. Assays conducted to establish the impact of this formulation on the intestinal epithelial cell line Caco-2 and the human colorectal adenocarcinoma cell line HT29 revealed the absence of cytotoxicity of both free-EntDD14 and EntDD14 loaded onto the alginate nanoparticles (EntDD14/Alg NPs) against the aforementioned eukaryotic cells, after 24 h of contact. Notably, EntDD14 and EntDD14/Alg NPs, both at a sub-inhibitory concentration, affected the expression of genes coding for clostridial toxins such as toxin α, enteritis B-like toxin, collagen adhesion protein and thiol-activated cytolysin. Further, expression of these genes was significantly down-regulated following the addition of EntDD14/Alg NPs, but not affected upon addition of EntDD14 alone. This study revealed that adsorption of EntDD14 onto Alg NPs leads to a safe and active formulation (EntDD14/Alg NPs) capable of affecting the pathogenicity of C. perfringens. This formulation could therefore be used in the poultry industry as a novel approach to tackle NE.
Collapse
Affiliation(s)
- Hassan Zgheib
- UMR Transfrontalière BioEcoAgro1158, Univ. Lille, INRAE, Univ. Liège, UPJV, YNCREA, Univ. Artois, Univ. Littoral Côte D'Opale, ICV - Institut Charles Viollette, 59000, Lille, France
- Univ. Lille, CNRS, Centrale Lille, Univ. Polytechnique Hauts-de-France. UMR, 8520 - IEMN, 59000, Lille, France
| | - Yanath Belguesmia
- UMR Transfrontalière BioEcoAgro1158, Univ. Lille, INRAE, Univ. Liège, UPJV, YNCREA, Univ. Artois, Univ. Littoral Côte D'Opale, ICV - Institut Charles Viollette, 59000, Lille, France.
| | - Rabah Boukherroub
- Univ. Lille, CNRS, Centrale Lille, Univ. Polytechnique Hauts-de-France. UMR, 8520 - IEMN, 59000, Lille, France
| | - Djamel Drider
- UMR Transfrontalière BioEcoAgro1158, Univ. Lille, INRAE, Univ. Liège, UPJV, YNCREA, Univ. Artois, Univ. Littoral Côte D'Opale, ICV - Institut Charles Viollette, 59000, Lille, France
| |
Collapse
|
28
|
Abe M, Kobayashi T. Imaging Sphingomyelin- and Cholesterol-Enriched Domains in the Plasma Membrane Using a Novel Probe and Super-Resolution Microscopy. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1310:81-90. [PMID: 33834433 DOI: 10.1007/978-981-33-6064-8_4] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
In this chapter, we show the visualization of lipid domains using a specific lipid-binding protein and super-resolution microscopy. Lipid rafts are plasma membrane domains enriched in both sphingolipids and sterols that play key roles in various physiological events. We identified a novel protein that specifically binds to a complex of sphingomyelin (SM) and cholesterol (Chol). The isolated protein, nakanori, labels the SM/Chol complex at the outer leaflet of the plasma membrane in mammalian cells. Structured illumination microscopic images suggested that the influenza virus buds from the edges of the SM/Chol domains in MDCK cells. Furthermore, a photoactivated localization microscopy analysis indicated that the SM/Chol complex forms domains in the outer leaflet, just above the phosphatidylinositol 4,5-bisphosphate domains in the inner leaflet. These observations provide significant insight into the structure and function of lipid rafts.
Collapse
Affiliation(s)
- Mitsuhiro Abe
- Cellular Informatics Laboratory, RIKEN, Wako, Saitama, Japan.
| | - Toshihide Kobayashi
- Cellular Informatics Laboratory, RIKEN, Wako, Saitama, Japan.,UMR 7021 CNRS, Faculté de Pharmacie, Université de Strasbourg, Illkirch, France
| |
Collapse
|
29
|
Pedersen JS, Kot W, Plöger M, Lametsh R, Neve H, Franz CM, Hansen LH. A Rare, Virulent Clostridium perfringens Bacteriophage Susfortuna Is the First Isolated Bacteriophage in a New Viral Genus. PHAGE (NEW ROCHELLE, N.Y.) 2020; 1:230-236. [PMID: 36147286 PMCID: PMC9041476 DOI: 10.1089/phage.2020.0038] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/16/2023]
Abstract
Background: Clostridium perfringens is a well known swine pathogen. C. perfringens type A is considered the causative agent of enteric diseases in neonatal and weaned piglets. Phage therapy using C. perfringens phages in vivo has previously proved effective. Materials and Methods: Pig fecal samples were used to isolate phages, with Clostridium perfringens type A as host. Complete genome sequencing, comparative genomics, a proteome analysis and electron microscopy were used to characterize the phage. Results: Clostridium phage Susfortuna has a double-stranded DNA genome of 19,046 bp with a G+C% content of 29.2, inverted terminal repeats and 28 predicted coding sequences (CDSs). Putative functions could not be assigned to most of the CDSs (64.3%). Transmission electron microscopy of phage Susfortuna revealed an isometric head and a short protruding tail stub resembling the structure of the Podoviridae family. A proteome analysis of phage Susfortuna identified seven structural proteins, but only one could be assigned with a putative function. Conclusions: Based on the morphology, the inverted terminal repeats and the small genome size, phage Susfortuna belongs to subfamily Picovirinae within the Podoviridae family in the order Caudovirales. Together with C. perfringens bacteriophage CPD7, phage Susfortuna represent a new genus of bacteriophages with very limited DNA sequence similarity to other known C. perfringens phages. Despite the limited DNA sequence similarity, the gene synteny among putative structural genes of phage Susfortuna is conserved among several C. perfringens bacteriophages belonging to the Podoviridae family indicating a common ancestor.
Collapse
Affiliation(s)
- Julie Stenberg Pedersen
- Department of Plant and Environmental Sciences, Faculty of Science, University of Copenhagen, Frederiksberg, Denmark
| | - Witold Kot
- Department of Plant and Environmental Sciences, Faculty of Science, University of Copenhagen, Frederiksberg, Denmark
| | - Maja Plöger
- Department of Plant and Environmental Sciences, Faculty of Science, University of Copenhagen, Frederiksberg, Denmark
| | - Réne Lametsh
- Department of Food Science, Faculty of Science, University of Copenhagen, Frederiksberg, Denmark
| | - Horst Neve
- Department of Microbiology and Biotechnology, Max Rubner-Institut, Kiel, Germany
| | - Charles M.A.P. Franz
- Department of Microbiology and Biotechnology, Max Rubner-Institut, Kiel, Germany
| | - Lars Hestbjerg Hansen
- Department of Plant and Environmental Sciences, Faculty of Science, University of Copenhagen, Frederiksberg, Denmark
| |
Collapse
|
30
|
Leiblein M, Wagner N, Adam EH, Frank J, Marzi I, Nau C. Clostridial Gas Gangrene - A Rare but Deadly Infection: Case series and Comparison to Other Necrotizing Soft Tissue Infections. Orthop Surg 2020; 12:1733-1747. [PMID: 33015993 PMCID: PMC7767692 DOI: 10.1111/os.12804] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/20/2020] [Revised: 08/07/2020] [Accepted: 08/18/2020] [Indexed: 02/06/2023] Open
Abstract
Objective Clostridial gas gangrene (GG) or clostridial myonecrosis is a very rare but life‐threatening necrotizing soft tissue infection (NSTI) caused by anaerobic, spore‐forming, and gas‐producing clostridium subspecies. It is the most rapidly spreading and lethal infection in humans, also affecting muscle tissue. The high mortality, of up to 100%, in clostridial GG is mediated by potent bacterial exotoxins. Necrotizing fasciitis (NF) is an important differential diagnosis, most often caused by group A streptococci, primarily not affecting musculature but the subcutaneous tissue and fascia. In the early stages of the infection, it is difficult to distinguish between GG and NF. Therefore, we compare both infection types, identify relevant differences in initial clinical presentation and later course, and present the results of our patients in a retrospective review. Methods Patients diagnosed with GG from 2008 to 2018 in our level one trauma center were identified. Their charts were reviewed retrospectively and data analyzed in terms of demographic information, microbiological and histological results, therapeutic course, outcome, and mortality rates. The laboratory risk indicator for NF (LRINEC) score was applied on the first blood work acquired. Results were compared to those of a second group diagnosed with NF. Results Five patients with GG and nine patients with NF were included in the present study. Patients with GG had a mortality rate of 80% compared to 0% in patients with NF. In eight patients with NF, affected limbs could be salvaged; one NF underwent amputation. LRINEC did not show significant differences between the groups; however, C‐reactive protein was significantly increased (P = 0.009) and hemoglobin (Hb) was significantly decreased (P = 0.02) in patients with GG. Interleukin‐6 and procalcitonin levels did not show significant difference. Patients with GG were older (70.2 vs 50 years). Of the isolated bacteria, 86% were sensitive to the initial calculated antibiotic treatment with ampicillin‐sulbactam or imipenem plus metronidazole plus clindamycin. Conclusion Both GG and NF need full‐scale surgical, antibiotic, and intensive care treatment, especially within the first days. Among patients with NSTI, those with clostridial GG have a significantly increased mortality risk due to early septic shock caused by clostridial toxins. In the initial stages, clinical differences are hardly detectable. Immediate surgical debridement is the key to successful therapy for NSTI and needs to be performed as early as possible. However, patients should be treated in a center with an experienced interdisciplinary intensive care team based on a predetermined treatment plan.
Collapse
Affiliation(s)
- Maximilian Leiblein
- Department of Trauma, Hand, and Reconstructive Surgery, Intensive Care Medicine and Pain Therapy, University Hospital Frankfurt, Frankfurt, Germany
| | - Nils Wagner
- Department of Trauma, Hand, and Reconstructive Surgery, Intensive Care Medicine and Pain Therapy, University Hospital Frankfurt, Frankfurt, Germany
| | - Elisabeth H Adam
- Department of Anesthesiology, Intensive Care Medicine and Pain Therapy, University Hospital Frankfurt, Frankfurt, Germany
| | - Johannes Frank
- Department of Trauma, Hand, and Reconstructive Surgery, Intensive Care Medicine and Pain Therapy, University Hospital Frankfurt, Frankfurt, Germany
| | - Ingo Marzi
- Department of Trauma, Hand, and Reconstructive Surgery, Intensive Care Medicine and Pain Therapy, University Hospital Frankfurt, Frankfurt, Germany
| | - Christoph Nau
- Department of Trauma, Hand, and Reconstructive Surgery, Intensive Care Medicine and Pain Therapy, University Hospital Frankfurt, Frankfurt, Germany
| |
Collapse
|
31
|
Takehara M, Bandou H, Kobayashi K, Nagahama M. Clostridium perfringens α-toxin specifically induces endothelial cell death by promoting ceramide-mediated apoptosis. Anaerobe 2020; 65:102262. [PMID: 32828915 DOI: 10.1016/j.anaerobe.2020.102262] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2020] [Revised: 07/14/2020] [Accepted: 08/17/2020] [Indexed: 01/14/2023]
Abstract
Clostridium perfringens type A-induced gas gangrene is characterized by severe myonecrosis, and α-toxin has been revealed to be a major virulence factor involved in the pathogenesis. However, the detailed mechanism is unclear. Here, we show that CD31+ endothelial cell counts decrease in muscles infected with C. perfringens in an α-toxin-dependent manner. In vitro experiments revealed that α-toxin preferentially and rapidly induces the death of human umbilical vein endothelial cells (HUVECs) compared with C2C12 murine muscle cells. The toxin induces apoptosis of HUVECs by increasing ceramide. Furthermore, the specificity might be dependent on differences in the sensitivity to ceramide between these cell lines. Together, our results suggest that α-toxin-induced endothelial cell death promotes severe myonecrosis and is involved in the pathogenesis of C. perfringens.
Collapse
Affiliation(s)
- Masaya Takehara
- Department of Microbiology, Faculty of Pharmaceutical Sciences, Tokushima Bunri University, Yamashiro-cho, Tokushima, 770-8514, Japan.
| | - Hiroto Bandou
- Department of Microbiology, Faculty of Pharmaceutical Sciences, Tokushima Bunri University, Yamashiro-cho, Tokushima, 770-8514, Japan
| | - Keiko Kobayashi
- Department of Microbiology, Faculty of Pharmaceutical Sciences, Tokushima Bunri University, Yamashiro-cho, Tokushima, 770-8514, Japan
| | - Masahiro Nagahama
- Department of Microbiology, Faculty of Pharmaceutical Sciences, Tokushima Bunri University, Yamashiro-cho, Tokushima, 770-8514, Japan.
| |
Collapse
|
32
|
Cytotoxic property of Streptococcus mitis strain producing two different types of cholesterol-dependent cytolysins. INFECTION GENETICS AND EVOLUTION 2020; 85:104483. [PMID: 32731044 DOI: 10.1016/j.meegid.2020.104483] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/20/2020] [Revised: 07/22/2020] [Accepted: 07/23/2020] [Indexed: 01/08/2023]
Abstract
Streptococcus mitis strain Nm-65 secretes an atypical 5-domain-type cholesterol-dependent cytolysin (CDC) called S. mitis-derived human platelet aggregation factor (Sm-hPAF) originally described as a platelet aggregation factor. Sm-hPAF belongs to Group III CDC that recognize both membrane cholesterol and human CD59 as the receptors, and shows preferential activity towards human cells. Draft genome analyses have shown that the Nm-65 strain also harbors a gene encoding another CDC called mitilysin (MLY). This CDC belongs to Group I CDC that recognizes only membrane cholesterol as a receptor, and it is a homolog of the pneumococcal CDC, pneumolysin. The genes encoding each CDC are located about 20 kb apart on the Nm-65 genome. Analysis of the genomic locus of these CDC-encoding genes in silico showed that the gene encoding Sm-hPAF and the region including the gene encoding MLY were both inserted into a specific locus of the S. mitis genome. The results obtained using deletion mutants of the gene(s) encoding CDC in Nm-65 indicated that each CDC contributes to both hemolysis and cytotoxicity, and that MLY is the major hemolysin/cytolysin in Nm-65. The present study aimed to determine the potential pathogenicity of an S. mitis strain that produces two CDC with different receptor recognition properties and secretion modes.
Collapse
|
33
|
Zhang J, Liu S, Xia L, Wen Z, Hu N, Wang T, Deng X, He J, Wang J. Verbascoside Protects Mice From Clostridial Gas Gangrene by Inhibiting the Activity of Alpha Toxin and Perfringolysin O. Front Microbiol 2020; 11:1504. [PMID: 32760362 PMCID: PMC7371946 DOI: 10.3389/fmicb.2020.01504] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2019] [Accepted: 06/10/2020] [Indexed: 01/24/2023] Open
Abstract
Gas gangrene, caused mainly by the anaerobic bacterium Clostridium perfringens (C. perfringens), causes death within 48 h of onset. Limited therapeutic strategies are available, and it is associated with extremely high mortality. Both C. perfringens alpha toxin (CPA) and perfringolysin O (PFO) are important virulence factors in the development of gas gangrene, suggesting that they are therapeutic targets. Here, we found that verbascoside, a phenylpropanoid glycoside widely distributed in Chinese herbal medicines, could effectively inhibit the biological activity of both CPA and PFO in hemolytic assays. The oligomerization of PFO was remarkably inhibited by verbascoside. Although no antibacterial activity was observed, verbascoside treatment protected Caco-2 cells from the damage caused by CPA and PFO. Additionally, infected mice treated with verbascoside showed significantly alleviated damage, reduced bacterial burden, and decreased mortality. In summary, verbascoside has an effective therapeutic effect against C. perfringens virulence both in vitro and in vivo by simultaneously targeting CPA and PFO. Our results provide a promising strategy and a potential lead compound for C. perfringens infections, especially gas gangrene.
Collapse
Affiliation(s)
- Jian Zhang
- Department of Respiratory Medicine, The First Hospital of Jilin University, Jilin University, Changchun, China.,Key Laboratory of Zoonosis Research, Ministry of Education, Institute of Zoonosis, College of Veterinary Medicine, Jilin University, Changchun, China
| | - Shui Liu
- Department of Respiratory Medicine, The First Hospital of Jilin University, Jilin University, Changchun, China.,Key Laboratory of Zoonosis Research, Ministry of Education, Institute of Zoonosis, College of Veterinary Medicine, Jilin University, Changchun, China
| | - Lining Xia
- College of Veterinary Medicine, Xinjiang Agricultural University, Ürümqi, China
| | - Zhongmei Wen
- Department of Respiratory Medicine, The First Hospital of Jilin University, Jilin University, Changchun, China
| | - Naiyu Hu
- Department of Respiratory Medicine, The First Hospital of Jilin University, Jilin University, Changchun, China
| | - Tingting Wang
- Department of Respiratory Medicine, The First Hospital of Jilin University, Jilin University, Changchun, China.,Key Laboratory of Zoonosis Research, Ministry of Education, Institute of Zoonosis, College of Veterinary Medicine, Jilin University, Changchun, China
| | - Xuming Deng
- Department of Respiratory Medicine, The First Hospital of Jilin University, Jilin University, Changchun, China.,Key Laboratory of Zoonosis Research, Ministry of Education, Institute of Zoonosis, College of Veterinary Medicine, Jilin University, Changchun, China
| | - Jiakang He
- Department of Respiratory Medicine, The First Hospital of Jilin University, Jilin University, Changchun, China.,College of Animal Science and Technology, Guangxi University, Nanning, China
| | - Jianfeng Wang
- Department of Respiratory Medicine, The First Hospital of Jilin University, Jilin University, Changchun, China.,Key Laboratory of Zoonosis Research, Ministry of Education, Institute of Zoonosis, College of Veterinary Medicine, Jilin University, Changchun, China
| |
Collapse
|
34
|
Liu S, Yang X, Zhang H, Zhang J, Zhou Y, Wang T, Hu N, Deng X, Bai X, Wang J. Amentoflavone Attenuates Clostridium perfringens Gas Gangrene by Targeting Alpha-Toxin and Perfringolysin O. Front Pharmacol 2020; 11:179. [PMID: 32180727 PMCID: PMC7059699 DOI: 10.3389/fphar.2020.00179] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2019] [Accepted: 02/10/2020] [Indexed: 12/11/2022] Open
Abstract
Clostridium perfringens (C. perfringens) type A strains are the main cause of gas gangrene in humans and animals. Treatment of this lethal disease is limited, and the prognosis is not good. Alpha-toxin (CPA) and perfringolysin O (PFO) secreted by C. perfringens play irreplaceable roles in cytotoxicity to host cells, persistence in host tissues, and lethality of gas gangrene pathology. This work determined the influence of amentoflavone, a biflavonoid isolated from Selaginella tamariscina and other plants, on hemolysis and cytotoxicity mediated by CPA and PFO and evaluated the in vivo therapeutic effect on gas gangrene. Our data showed that amentoflavone could block the hemolysis and cytotoxicity induced by CPA and PFO in vitro, thereby mediating significant protection against mortality of infected mice in a mouse gas gangrene model, efficient bacterial clearance in tissues and alleviation of histological damage in vivo. Based on the above results, amentoflavone may be a potential candidate against C. perfringens infection by reducing CPA and PFO-mediated virulence.
Collapse
Affiliation(s)
- Shui Liu
- Cadre's Ward, The First Hospital of Jilin University, Jilin University, Changchun, China.,Key Laboratory of Zoonosis Research, Ministry of Education, College of Veterinary Medicine, Institute of Zoonosis, Jilin University, Changchun, China
| | - Xiaofeng Yang
- Cadre's Ward, The First Hospital of Jilin University, Jilin University, Changchun, China.,Department of Pathogenic Biology and Immunology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, China
| | - Hong Zhang
- Cadre's Ward, The First Hospital of Jilin University, Jilin University, Changchun, China.,Key Laboratory of Zoonosis Research, Ministry of Education, College of Veterinary Medicine, Institute of Zoonosis, Jilin University, Changchun, China
| | - Jian Zhang
- Key Laboratory of Zoonosis Research, Ministry of Education, College of Veterinary Medicine, Institute of Zoonosis, Jilin University, Changchun, China
| | - Yonglin Zhou
- Key Laboratory of Zoonosis Research, Ministry of Education, College of Veterinary Medicine, Institute of Zoonosis, Jilin University, Changchun, China
| | - Tingting Wang
- Key Laboratory of Zoonosis Research, Ministry of Education, College of Veterinary Medicine, Institute of Zoonosis, Jilin University, Changchun, China
| | - Naiyu Hu
- College of Animal Sciences, Jilin University, Changchun, China
| | - Xuming Deng
- Cadre's Ward, The First Hospital of Jilin University, Jilin University, Changchun, China.,Key Laboratory of Zoonosis Research, Ministry of Education, College of Veterinary Medicine, Institute of Zoonosis, Jilin University, Changchun, China
| | - Xiaoxue Bai
- Cadre's Ward, The First Hospital of Jilin University, Jilin University, Changchun, China
| | - Jianfeng Wang
- Cadre's Ward, The First Hospital of Jilin University, Jilin University, Changchun, China.,Key Laboratory of Zoonosis Research, Ministry of Education, College of Veterinary Medicine, Institute of Zoonosis, Jilin University, Changchun, China
| |
Collapse
|
35
|
Junior CAO, Silva ROS, Lobato FCF, Navarro MA, Uzal FA. Gas gangrene in mammals: a review. J Vet Diagn Invest 2020; 32:175-183. [PMID: 32081096 DOI: 10.1177/1040638720905830] [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] [Indexed: 12/26/2022] Open
Abstract
Gas gangrene is a necrotizing infection of subcutaneous tissue and muscle that affects mainly ruminants and horses, but also other domestic and wild mammals. Clostridium chauvoei, C. septicum, C. novyi type A, C. perfringens type A, and C. sordellii are the etiologic agents of this disease, acting singly or in combination. Although a presumptive diagnosis of gas gangrene can be established based on clinical history, clinical signs, and gross and microscopic changes, identification of the clostridia involved is required for confirmatory diagnosis. Gross and microscopic lesions are, however, highly suggestive of the disease. Although the disease has a worldwide distribution and can cause significant economic losses, the literature is limited mostly to case reports. Thus, we have reviewed the current knowledge of gas gangrene in mammals.
Collapse
Affiliation(s)
- Carlos A Oliveira Junior
- Veterinary School, Federal University of Minas Gerais, Brazil (Oliveira Junior, Silva, Lobato).,California Animal Health and Food Safety Laboratory, University of California, Davis, CA (Navarro, Uzal)
| | - Rodrigo O S Silva
- Veterinary School, Federal University of Minas Gerais, Brazil (Oliveira Junior, Silva, Lobato).,California Animal Health and Food Safety Laboratory, University of California, Davis, CA (Navarro, Uzal)
| | - Francisco C F Lobato
- Veterinary School, Federal University of Minas Gerais, Brazil (Oliveira Junior, Silva, Lobato).,California Animal Health and Food Safety Laboratory, University of California, Davis, CA (Navarro, Uzal)
| | - Mauricio A Navarro
- Veterinary School, Federal University of Minas Gerais, Brazil (Oliveira Junior, Silva, Lobato).,California Animal Health and Food Safety Laboratory, University of California, Davis, CA (Navarro, Uzal)
| | - Francisco A Uzal
- Veterinary School, Federal University of Minas Gerais, Brazil (Oliveira Junior, Silva, Lobato).,California Animal Health and Food Safety Laboratory, University of California, Davis, CA (Navarro, Uzal)
| |
Collapse
|
36
|
Granulocyte Colony-Stimulating Factor Does Not Influence Clostridium Perfringens α-Toxin-Induced Myonecrosis in Mice. Toxins (Basel) 2019; 11:toxins11090509. [PMID: 31480318 PMCID: PMC6784116 DOI: 10.3390/toxins11090509] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2019] [Revised: 08/20/2019] [Accepted: 08/28/2019] [Indexed: 12/04/2022] Open
Abstract
Clostridium perfringens type A causes gas gangrene characterized by myonecrosis and development of an effective therapy for treating affected patients is of clinical importance. It was recently reported that the expression of granulocyte colony-stimulating factor (G-CSF) is greatly up-regulated by C. perfringens infection. However, the role of G-CSF in C. perfringens-mediated myonecrosis is still unclear. Here, we assessed the destructive changes in C. perfringens-infected skeletal muscles and tested whether inhibition of G-CSF receptor (G-CSFR) signaling or administration of recombinant G-CSF affects the tissue injury. Severe edema, contraction of muscle fiber diameter, and increased plasma creatine kinase activity were observed in mice intramuscularly injected with C. perfringens type A, and the destructive changes were α-toxin-dependent, indicating that infection induces the destruction of skeletal muscle in an α-toxin-dependent manner. G-CSF plays important roles in the protection of tissue against damage and in the regeneration of injured tissue. However, administration of a neutralizing antibody against G-CSFR had no profound impact on the destructive changes to skeletal muscle. Moreover, administration of recombinant human G-CSF, filgrastim, imparted no inhibitory effect against the destructive changes caused by C. perfringens. Together, these results indicate that G-CSF is not beneficial for treating C. perfringens α-toxin-mediated myonecrosis, but highlight the importance of revealing the mechanism by which C. perfringens negates the protective effects of G-CSF in skeletal muscle.
Collapse
|
37
|
Nagahama M, Takehara M, Rood JI. Histotoxic Clostridial Infections. Microbiol Spectr 2019; 7:10.1128/microbiolspec.gpp3-0024-2018. [PMID: 31350831 PMCID: PMC10957196 DOI: 10.1128/microbiolspec.gpp3-0024-2018] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2018] [Indexed: 01/01/2023] Open
Abstract
The pathogenesis of clostridial myonecrosis or gas gangrene involves an interruption to the blood supply to the infected tissues, often via a traumatic wound, anaerobic growth of the infecting clostridial cells, the production of extracellular toxins, and toxin-mediated cell and tissue damage. This review focuses on host-pathogen interactions in Clostridium perfringens-mediated and Clostridium septicum-mediated myonecrosis. The major toxins involved are C. perfringens α-toxin, which has phospholipase C and sphingomyelinase activity, and C. septicum α-toxin, a β-pore-forming toxin that belongs to the aerolysin family. Although these toxins are cytotoxic, their effects on host cells are quite complex, with a range of intracellular cell signaling pathways induced by their action on host cell membranes.
Collapse
Affiliation(s)
- Masahiro Nagahama
- Department of Microbiology, Faculty of Pharmaceutical Sciences, Tokushima Bunri University, Yamashiro-cho, Tokushima 770-8514, Japan
| | - Masaya Takehara
- Department of Microbiology, Faculty of Pharmaceutical Sciences, Tokushima Bunri University, Yamashiro-cho, Tokushima 770-8514, Japan
| | - Julian I Rood
- Infection and Immunity Program, Monash Biomedicine Discovery Institute and Department of Microbiology, Monash University, Clayton, Victoria 3800, Australia
| |
Collapse
|
38
|
Brito C, Mesquita FS, Bleck CKE, Sellers JR, Cabanes D, Sousa S. Perfringolysin O-Induced Plasma Membrane Pores Trigger Actomyosin Remodeling and Endoplasmic Reticulum Redistribution. Toxins (Basel) 2019; 11:toxins11070419. [PMID: 31319618 PMCID: PMC6669444 DOI: 10.3390/toxins11070419] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2019] [Revised: 07/10/2019] [Accepted: 07/15/2019] [Indexed: 01/28/2023] Open
Abstract
Clostridium perfringens produces an arsenal of toxins that act together to cause severe infections in humans and livestock animals. Perfringolysin O (PFO) is a cholesterol-dependent pore-forming toxin encoded in the chromosome of virtually all C. perfringens strains and acts in synergy with other toxins to determine the outcome of the infection. However, its individual contribution to the disease is poorly understood. Here, we intoxicated human epithelial and endothelial cells with purified PFO to evaluate the host cytoskeletal responses to PFO-induced damage. We found that, at sub-lytic concentrations, PFO induces a profound reorganization of the actomyosin cytoskeleton culminating into the assembly of well-defined cortical actomyosin structures at sites of plasma membrane (PM) remodeling. The assembly of such structures occurs concomitantly with the loss of the PM integrity and requires pore-formation, calcium influx, and myosin II activity. The recovery from the PM damage occurs simultaneously with the disassembly of cortical structures. PFO also targets the endoplasmic reticulum (ER) by inducing its disruption and vacuolation. ER-enriched vacuoles were detected at the cell cortex within the PFO-induced actomyosin structures. These cellular events suggest the targeting of the endothelium integrity at early stages of C. perfringens infection, in which secreted PFO is at sub-lytic concentrations.
Collapse
Affiliation(s)
- Cláudia Brito
- i3S-Instituto de Investigação e Inovação em Saúde, IBMC, Universidade do Porto, 4099-002 Porto, Portugal
- Programa Doutoral em Biologia Molecular e Celular (MCbiology), Instituto de Ciências Biomédicas Abel, Salazar, Universidade do Porto, 4099-002 Porto, Portugal
| | - Francisco S Mesquita
- i3S-Instituto de Investigação e Inovação em Saúde, IBMC, Universidade do Porto, 4099-002 Porto, Portugal
| | - Christopher K E Bleck
- Electron Microscopy Core, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - James R Sellers
- Laboratory of Molecular Physiology, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Didier Cabanes
- i3S-Instituto de Investigação e Inovação em Saúde, IBMC, Universidade do Porto, 4099-002 Porto, Portugal
| | - Sandra Sousa
- i3S-Instituto de Investigação e Inovação em Saúde, IBMC, Universidade do Porto, 4099-002 Porto, Portugal.
| |
Collapse
|
39
|
Harimoto T, Singer ZS, Velazquez OS, Zhang J, Castro S, Hinchliffe TE, Mather W, Danino T. Rapid screening of engineered microbial therapies in a 3D multicellular model. Proc Natl Acad Sci U S A 2019; 116:9002-9007. [PMID: 30996123 PMCID: PMC6500119 DOI: 10.1073/pnas.1820824116] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Synthetic biology is transforming therapeutic paradigms by engineering living cells and microbes to intelligently sense and respond to diseases including inflammation, infections, metabolic disorders, and cancer. However, the ability to rapidly engineer new therapies far outpaces the throughput of animal-based testing regimes, creating a major bottleneck for clinical translation. In vitro approaches to address this challenge have been limited in scalability and broad applicability. Here, we present a bacteria-in-spheroid coculture (BSCC) platform that simultaneously tests host species, therapeutic payloads, and synthetic gene circuits of engineered bacteria within multicellular spheroids over a timescale of weeks. Long-term monitoring of bacterial dynamics and disease progression enables quantitative comparison of critical therapeutic parameters such as efficacy and biocontainment. Specifically, we screen Salmonella typhimurium strains expressing and delivering a library of antitumor therapeutic molecules via several synthetic gene circuits. We identify candidates exhibiting significant tumor reduction and demonstrate high similarity in their efficacies, using a syngeneic mouse model. Last, we show that our platform can be expanded to dynamically profile diverse microbial species including Listeria monocytogenes, Proteus mirabilis, and Escherichia coli in various host cell types. This high-throughput framework may serve to accelerate synthetic biology for clinical applications and for understanding the host-microbe interactions in disease sites.
Collapse
Affiliation(s)
- Tetsuhiro Harimoto
- Department of Biomedical Engineering, Columbia University, New York, NY 10027
| | - Zakary S Singer
- Department of Biomedical Engineering, Columbia University, New York, NY 10027
| | - Oscar S Velazquez
- Department of Biomedical Engineering, Columbia University, New York, NY 10027
| | - Joanna Zhang
- Department of Biomedical Engineering, Columbia University, New York, NY 10027
| | - Samuel Castro
- Department of Biomedical Engineering, Columbia University, New York, NY 10027
| | - Taylor E Hinchliffe
- Department of Biomedical Engineering, Columbia University, New York, NY 10027
| | - William Mather
- BioCircuits Institute, University of California, San Diego, La Jolla, CA 92093
| | - Tal Danino
- Department of Biomedical Engineering, Columbia University, New York, NY 10027;
- Data Science Institute, Columbia University, New York, NY 10027
- Herbert Irving Comprehensive Cancer Center, Columbia University, New York, NY 10027
| |
Collapse
|
40
|
Takehara M, Seike S, Sonobe Y, Bandou H, Yokoyama S, Takagishi T, Miyamoto K, Kobayashi K, Nagahama M. Clostridium perfringens α-toxin impairs granulocyte colony-stimulating factor receptor-mediated granulocyte production while triggering septic shock. Commun Biol 2019; 2:45. [PMID: 30729183 PMCID: PMC6355902 DOI: 10.1038/s42003-019-0280-2] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2018] [Accepted: 12/28/2018] [Indexed: 12/12/2022] Open
Abstract
During bacterial infection, granulocyte colony-stimulating factor (G-CSF) is produced and accelerates neutrophil production from their progenitors. This process, termed granulopoiesis, strengthens host defense, but Clostridium perfringens α-toxin impairs granulopoiesis via an unknown mechanism. Here, we tested whether G-CSF accounts for the α-toxin-mediated impairment of granulopoiesis. We find that α-toxin dramatically accelerates G-CSF production from endothelial cells in response to Toll-like receptor 2 (TLR2) agonists through activation of the c-Jun N-terminal kinase (JNK) signaling pathway. Meanwhile, α-toxin inhibits G-CSF-mediated cell proliferation of Ly-6G+ neutrophils by inducing degradation of G-CSF receptor (G-CSFR). During sepsis, administration of α-toxin promotes lethality and tissue injury accompanied by accelerated production of inflammatory cytokines in a TLR4-dependent manner. Together, our results illustrate that α-toxin disturbs G-CSF-mediated granulopoiesis by reducing the expression of G-CSFR on neutrophils while augmenting septic shock due to excess inflammatory cytokine release, which provides a new mechanism to explain how pathogenic bacteria modulate the host immune system.
Collapse
Affiliation(s)
- Masaya Takehara
- Department of Microbiology, Faculty of Pharmaceutical Sciences, Tokushima Bunri University, Yamashiro-cho, Tokushima, 770-8514 Japan
| | - Soshi Seike
- Department of Microbiology, Faculty of Pharmaceutical Sciences, Tokushima Bunri University, Yamashiro-cho, Tokushima, 770-8514 Japan
| | - Yuuta Sonobe
- Department of Microbiology, Faculty of Pharmaceutical Sciences, Tokushima Bunri University, Yamashiro-cho, Tokushima, 770-8514 Japan
| | - Hiroto Bandou
- Department of Microbiology, Faculty of Pharmaceutical Sciences, Tokushima Bunri University, Yamashiro-cho, Tokushima, 770-8514 Japan
| | - Saki Yokoyama
- Department of Microbiology, Faculty of Pharmaceutical Sciences, Tokushima Bunri University, Yamashiro-cho, Tokushima, 770-8514 Japan
| | - Teruhisa Takagishi
- Department of Microbiology, Faculty of Pharmaceutical Sciences, Tokushima Bunri University, Yamashiro-cho, Tokushima, 770-8514 Japan
| | - Kazuaki Miyamoto
- Department of Microbiology, Faculty of Pharmaceutical Sciences, Tokushima Bunri University, Yamashiro-cho, Tokushima, 770-8514 Japan
| | - Keiko Kobayashi
- Department of Microbiology, Faculty of Pharmaceutical Sciences, Tokushima Bunri University, Yamashiro-cho, Tokushima, 770-8514 Japan
| | - Masahiro Nagahama
- Department of Microbiology, Faculty of Pharmaceutical Sciences, Tokushima Bunri University, Yamashiro-cho, Tokushima, 770-8514 Japan
| |
Collapse
|
41
|
In silico Identification of Novel Toxin Homologs and Associated Mobile Genetic Elements in Clostridium perfringens. Pathogens 2019; 8:pathogens8010016. [PMID: 30699957 PMCID: PMC6471305 DOI: 10.3390/pathogens8010016] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2018] [Revised: 01/16/2019] [Accepted: 01/24/2019] [Indexed: 01/05/2023] Open
Abstract
Clostridium perfringens causes a wide range of diseases in a variety of hosts, due to the production of a diverse set of toxins and extracellular enzymes. The C. perfringens toxins play an important role in pathogenesis, such that the presence and absence of the toxins is used as a typing scheme for the species. In recent years, several new toxins have been discovered that have been shown to be essential or highly correlated to diseases; these include binary enterotoxin (BecAB), NetB and NetF. In the current study, genome sequence analysis of C. perfringens isolates from diverse sources revealed several putative novel toxin homologs, some of which appeared to be associated with potential mobile genetic elements, including transposons and plasmids. Four novel toxin homologs encoding proteins related to the pore-forming Leukocidin/Hemolysin family were found in type A and G isolates. Two novel toxin homologs encoding proteins related to the epsilon aerolysin-like toxin family were identified in Type A and F isolates from humans, contaminated food and turkeys. A novel set of proteins related to clostridial binary toxins was also identified. While phenotypic characterisation is required before any of these homologs can be established as functional toxins, the in silico identification of these novel homologs on mobile genetic elements suggests the potential toxin reservoir of C. perfringens may be much larger than previously thought.
Collapse
|
42
|
Nguyen BN, Peterson BN, Portnoy DA. Listeriolysin O: A phagosome-specific cytolysin revisited. Cell Microbiol 2019; 21:e12988. [PMID: 30511471 DOI: 10.1111/cmi.12988] [Citation(s) in RCA: 47] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2018] [Revised: 11/02/2018] [Accepted: 11/09/2018] [Indexed: 12/17/2022]
Abstract
Listeriolysin O (LLO) is an essential determinant of Listeria monocytogenes pathogenesis that mediates the escape of L. monocytogenes from host cell vacuoles, thereby allowing replication in the cytosol without causing appreciable cell death. As a member of the cholesterol-dependent cytolysin (CDC) family of pore-forming toxins, LLO is unique in that it is secreted by a facultative intracellular pathogen, whereas all other CDCs are produced by pathogens that are largely extracellular. Replacement of LLO with other CDCs results in strains that are extremely cytotoxic and 10,000-fold less virulent in mice. LLO has structural and regulatory features that allow it to function intracellularly without causing cell death, most of which map to a unique N-terminal region of LLO referred to as the proline, glutamic acid, serine, threonine (PEST)-like sequence. Yet, while LLO has unique properties required for its intracellular site of action, extracellular LLO, like other CDCs, affects cells in a myriad of ways. Because all CDCs form pores in cholesterol-containing membranes that lead to rapid Ca2+ influx and K+ efflux, they consequently trigger a wide range of host cell responses, including mitogen-activated protein kinase activation, histone modification, and caspase-1 activation. There is no debate that extracellular LLO, like all other CDCs, can stimulate multiple cellular activities, but the primary question we wish to address in this perspective is whether these activities contribute to L. monocytogenes pathogenesis.
Collapse
Affiliation(s)
- Brittney N Nguyen
- Graduate Group in Microbiology, University of California, Berkeley, Berkeley, California
| | - Bret N Peterson
- Graduate Group in Microbiology, University of California, Berkeley, Berkeley, California
| | - Daniel A Portnoy
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, California.,Department of Plant and Microbial Biology, University of California, Berkeley, Berkeley, California
| |
Collapse
|
43
|
Kiu R, Hall LJ. An update on the human and animal enteric pathogen Clostridium perfringens. Emerg Microbes Infect 2018; 7:141. [PMID: 30082713 PMCID: PMC6079034 DOI: 10.1038/s41426-018-0144-8] [Citation(s) in RCA: 278] [Impact Index Per Article: 39.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2018] [Revised: 06/28/2018] [Accepted: 07/02/2018] [Indexed: 12/18/2022]
Abstract
Clostridium perfringens, a rapid-growing pathogen known to secrete an arsenal of >20 virulent toxins, has been associated with intestinal diseases in both animals and humans throughout the past century. Recent advances in genomic analysis and experimental systems make it timely to re-visit this clinically and veterinary important pathogen. This Review will summarise our understanding of the genomics and virulence-linked factors, including antimicrobial potentials and secreted toxins of this gut pathogen, and then its up-to-date clinical epidemiology and biological role in the pathogenesis of several important human and animal-associated intestinal diseases, including pre-term necrotising enterocolitis. Finally, we highlight some of the important unresolved questions in relation to C. perfringens-mediated infections, and implications for future research directions.
Collapse
Affiliation(s)
- Raymond Kiu
- Gut Microbes and Health Programme, Quadram Institute Bioscience, Norwich Research Park, Norwich, UK.,Norwich Medical School, University of East Anglia, Norwich Research Park, Norwich, UK
| | - Lindsay J Hall
- Gut Microbes and Health Programme, Quadram Institute Bioscience, Norwich Research Park, Norwich, UK.
| |
Collapse
|
44
|
Rezelj S, Kozorog M, Švigelj T, Ulrih NP, Žnidaršič N, Podobnik M, Anderluh G. Cholesterol Enriched Archaeosomes as a Molecular System for Studying Interactions of Cholesterol-Dependent Cytolysins with Membranes. J Membr Biol 2018; 251:491-505. [DOI: 10.1007/s00232-018-0018-y] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2017] [Accepted: 02/05/2018] [Indexed: 12/31/2022]
|
45
|
Maekawa M. Domain 4 (D4) of Perfringolysin O to Visualize Cholesterol in Cellular Membranes-The Update. SENSORS 2017; 17:s17030504. [PMID: 28273804 PMCID: PMC5375790 DOI: 10.3390/s17030504] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/21/2017] [Revised: 02/28/2017] [Accepted: 03/01/2017] [Indexed: 12/14/2022]
Abstract
The cellular membrane of eukaryotes consists of phospholipids, sphingolipids, cholesterol and membrane proteins. Among them, cholesterol is crucial for various cellular events (e.g., signaling, viral/bacterial infection, and membrane trafficking) in addition to its essential role as an ingredient of steroid hormones, vitamin D, and bile acids. From a micro-perspective, at the plasma membrane, recent emerging evidence strongly suggests the existence of lipid nanodomains formed with cholesterol and phospholipids (e.g., sphingomyelin, phosphatidylserine). Thus, it is important to elucidate how cholesterol behaves in membranes and how the behavior of cholesterol is regulated at the molecular level. To elucidate the complexed characteristics of cholesterol in cellular membranes, a couple of useful biosensors that enable us to visualize cholesterol in cellular membranes have been recently developed by utilizing domain 4 (D4) of Perfringolysin O (PFO, theta toxin), a cholesterol-binding toxin. This review highlights the current progress on development of novel cholesterol biosensors that uncover new insights of cholesterol in cellular membranes.
Collapse
Affiliation(s)
- Masashi Maekawa
- Department of Biochemistry and Molecular Genetics, Ehime University Graduate School of Medicine, Toon, Ehime 791-0295, Japan.
- Division of Cell Growth and Tumor Regulation, Proteo-Science Center, Ehime University; Toon, Ehime 791-0295, Japan.
| |
Collapse
|
46
|
Goossens E, Valgaeren BR, Pardon B, Haesebrouck F, Ducatelle R, Deprez PR, Van Immerseel F. Rethinking the role of alpha toxin in Clostridium perfringens-associated enteric diseases: a review on bovine necro-haemorrhagic enteritis. Vet Res 2017; 48:9. [PMID: 28209206 PMCID: PMC5314468 DOI: 10.1186/s13567-017-0413-x] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2016] [Accepted: 01/12/2017] [Indexed: 11/23/2022] Open
Abstract
Bovine necro-haemorrhagic enteritis is an economically important disease caused by Clostridium perfringens type A strains. The disease mainly affects calves under intensive rearing conditions and is characterized by sudden death associated with small intestinal haemorrhage, necrosis and mucosal neutrophil infiltration. The common assumption that, when causing intestinal disease, C. perfringens relies upon specific, plasmid-encoded toxins, was recently challenged by the finding that alpha toxin, which is produced by all C. perfringens strains, is essential for necro-haemorrhagic enteritis. In addition to alpha toxin, other C. perfringens toxins and/or enzymes might contribute to the pathogenesis of necro-haemorrhagic enteritis. These additional virulence factors might contribute to breakdown of the protective mucus layer during initial stage of pathogenesis, after which alpha toxin, either or not in synergy with other toxins such as perfringolysin O, can act on the mucosal tissue. Furthermore, alpha toxin alone does not cause intestinal necrosis, indicating that other virulence factors might be needed to cause the extensive tissue necrosis observed in necro-haemorrhagic enteritis. This review summarizes recent research that has increased our understanding of the pathogenesis of bovine necro-haemorrhagic enteritis and provides information that is indispensable for the development of novel control strategies, including vaccines.
Collapse
Affiliation(s)
- Evy Goossens
- Department of Pathology, Bacteriology and Avian Diseases, Faculty of Veterinary Medicine, Ghent University, Salisburylaan 133, B-9820, Merelbeke, Belgium
| | - Bonnie R Valgaeren
- Department of Large Animal Internal Medicine, Faculty of Veterinary Medicine, Ghent University, Salisburylaan 133, B-9820, Merelbeke, Belgium
| | - Bart Pardon
- Department of Large Animal Internal Medicine, Faculty of Veterinary Medicine, Ghent University, Salisburylaan 133, B-9820, Merelbeke, Belgium
| | - Freddy Haesebrouck
- Department of Pathology, Bacteriology and Avian Diseases, Faculty of Veterinary Medicine, Ghent University, Salisburylaan 133, B-9820, Merelbeke, Belgium
| | - Richard Ducatelle
- Department of Pathology, Bacteriology and Avian Diseases, Faculty of Veterinary Medicine, Ghent University, Salisburylaan 133, B-9820, Merelbeke, Belgium
| | - Piet R Deprez
- Department of Large Animal Internal Medicine, Faculty of Veterinary Medicine, Ghent University, Salisburylaan 133, B-9820, Merelbeke, Belgium
| | - Filip Van Immerseel
- Department of Pathology, Bacteriology and Avian Diseases, Faculty of Veterinary Medicine, Ghent University, Salisburylaan 133, B-9820, Merelbeke, Belgium.
| |
Collapse
|
47
|
Verherstraeten S, Goossens E, Valgaeren B, Pardon B, Timbermont L, Haesebrouck F, Ducatelle R, Deprez P, Van Immerseel F. Non-toxic perfringolysin O and α-toxin derivatives as potential vaccine candidates against bovine necrohaemorrhagic enteritis. Vet J 2016; 217:89-94. [PMID: 27810219 DOI: 10.1016/j.tvjl.2016.09.008] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2015] [Revised: 09/19/2016] [Accepted: 09/28/2016] [Indexed: 12/25/2022]
Abstract
Bovine necrohaemorrhagic enteritis is a fatal Clostridium perfringens type A-induced disease that is characterised by sudden death. Recently the involvement of perfringolysin O and α-toxin in the development of necrohaemorrhagic lesions in the gut of calves was suggested, and thus derivatives of these toxins are potentially suitable as vaccine antigens. In the current study, the perfringolysin O derivative PFOL491D, alone or in combination with α-toxin derivative GST-cpa247-370, was evaluated as possible vaccine candidate, using in vitro assays. PFOL491D showed no haemolytic effect on horse red blood cells and no cytotoxic effect on bovine endothelial cells. Furthermore, calves immunised with PFOL491D raised antibodies against perfringolysin O that could inhibit the perfringolysin O-associated haemolytic activity on horse red blood cells. Antisera from calves immunised with PFOL491D had a significantly higher neutralising capacity against the cytotoxic effect of C. perfringens culture supernatant to bovine endothelial cells than serum from control calves (P <0.05). Immunisation of calves with PFOL491D in combination with GST-cpa247-370 elicited antibodies against perfringolysin O and α-toxin and consequently inhibited both the perfringolysin O-associated haemolytic activity and the α-toxin-associated lecithinase activity in vitro. Additionally, the neutralising ability of these antisera on the cytotoxic effect of C. perfringens culture supernatant to bovine endothelial cells was significantly higher than that from calves immunised with PFOL491D (P <0.001). In conclusion, perfringolysin O derivative PFOL491D is an immunogenic antigen that can potentially be used to produce vaccine against bovine necrohaemorrhagic enteritis. Including α-toxin derivative GST-cpa247-370 has an additional protective effect and therefore vaccination of calves with a combination of both antigens seems even more promising.
Collapse
Affiliation(s)
- S Verherstraeten
- Department of Pathology, Bacteriology and Avian Diseases, Faculty of Veterinary Medicine, Ghent University, Salisburylaan 133, B-9820 Merelbeke, Belgium
| | - E Goossens
- Department of Pathology, Bacteriology and Avian Diseases, Faculty of Veterinary Medicine, Ghent University, Salisburylaan 133, B-9820 Merelbeke, Belgium
| | - B Valgaeren
- Department of Internal Medicine and Clinical Biology of Large Animals, Faculty of Veterinary Medicine, Ghent University, Salisburylaan 133, B-9820 Merelbeke, Belgium
| | - B Pardon
- Department of Internal Medicine and Clinical Biology of Large Animals, Faculty of Veterinary Medicine, Ghent University, Salisburylaan 133, B-9820 Merelbeke, Belgium
| | - L Timbermont
- Department of Pathology, Bacteriology and Avian Diseases, Faculty of Veterinary Medicine, Ghent University, Salisburylaan 133, B-9820 Merelbeke, Belgium
| | - F Haesebrouck
- Department of Pathology, Bacteriology and Avian Diseases, Faculty of Veterinary Medicine, Ghent University, Salisburylaan 133, B-9820 Merelbeke, Belgium
| | - R Ducatelle
- Department of Pathology, Bacteriology and Avian Diseases, Faculty of Veterinary Medicine, Ghent University, Salisburylaan 133, B-9820 Merelbeke, Belgium
| | - P Deprez
- Department of Internal Medicine and Clinical Biology of Large Animals, Faculty of Veterinary Medicine, Ghent University, Salisburylaan 133, B-9820 Merelbeke, Belgium
| | - F Van Immerseel
- Department of Pathology, Bacteriology and Avian Diseases, Faculty of Veterinary Medicine, Ghent University, Salisburylaan 133, B-9820 Merelbeke, Belgium.
| |
Collapse
|
48
|
Augmenting the Efficacy of Immunotoxins and Other Targeted Protein Toxins by Endosomal Escape Enhancers. Toxins (Basel) 2016; 8:toxins8070200. [PMID: 27376327 PMCID: PMC4963833 DOI: 10.3390/toxins8070200] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2016] [Revised: 06/14/2016] [Accepted: 06/17/2016] [Indexed: 12/18/2022] Open
Abstract
The toxic moiety of almost all protein-based targeted toxins must enter the cytosol of the target cell to mediate its fatal effect. Although more than 500 targeted toxins have been investigated in the past decades, no antibody-targeted protein toxin has been approved for tumor therapeutic applications by the authorities to date. Missing efficacy can be attributed in many cases to insufficient endosomal escape and therefore subsequent lysosomal degradation of the endocytosed toxins. To overcome this drawback, many strategies have been described to weaken the membrane integrity of endosomes. This comprises the use of lysosomotropic amines, carboxylic ionophores, calcium channel antagonists, various cell-penetrating peptides of viral, bacterial, plant, animal, human and synthetic origin, other organic molecules and light-induced techniques. Although the efficacy of the targeted toxins was typically augmented in cell culture hundred or thousand fold, in exceptional cases more than million fold, the combination of several substances harbors new problems including additional side effects, loss of target specificity, difficulties to determine the therapeutic window and cell type-dependent variations. This review critically scrutinizes the chances and challenges of endosomal escape enhancers and their potential role in future developments.
Collapse
|
49
|
Takehara M, Takagishi T, Seike S, Ohtani K, Kobayashi K, Miyamoto K, Shimizu T, Nagahama M. Clostridium perfringens α-Toxin Impairs Innate Immunity via Inhibition of Neutrophil Differentiation. Sci Rep 2016; 6:28192. [PMID: 27306065 PMCID: PMC4910053 DOI: 10.1038/srep28192] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2016] [Accepted: 06/01/2016] [Indexed: 12/21/2022] Open
Abstract
Although granulopoiesis is accelerated to suppress bacteria during infection, some bacteria can still cause life-threatening infections, but the mechanism behind this remains unclear. In this study, we found that mature neutrophils in bone marrow cells (BMCs) were decreased in C. perfringens-infected mice and also after injection of virulence factor α-toxin. C. perfringens infection interfered with the replenishment of mature neutrophils in the peripheral circulation and the accumulation of neutrophils at C. perfringens-infected sites in an α-toxin-dependent manner. Measurements of bacterial colony-forming units in C. perfringens-infected muscle revealed that α-toxin inhibited a reduction in the load of C. perfringens. In vitro treatment of isolated BMCs with α-toxin (phospholipase C) revealed that α-toxin directly decreased mature neutrophils. α-Toxin did not influence the viability of isolated mature neutrophils, while simultaneous treatment of BMCs with granulocyte colony-stimulating factor attenuated the reduction of mature neutrophils by α-toxin. Together, our results illustrate that impairment of the innate immune system by the inhibition of neutrophil differentiation is crucial for the pathogenesis of C. perfringens to promote disease to a life-threatening infection, which provides new insight to understand how pathogenic bacteria evade the host immune system.
Collapse
Affiliation(s)
- Masaya Takehara
- Department of Microbiology, Faculty of Pharmaceutical Sciences, Tokushima Bunri University, Yamashiro-cho, Tokushima 770-8514, Japan
| | - Teruhisa Takagishi
- Department of Microbiology, Faculty of Pharmaceutical Sciences, Tokushima Bunri University, Yamashiro-cho, Tokushima 770-8514, Japan
| | - Soshi Seike
- Department of Microbiology, Faculty of Pharmaceutical Sciences, Tokushima Bunri University, Yamashiro-cho, Tokushima 770-8514, Japan
| | - Kaori Ohtani
- Department of Bacteriology, Graduate School of Medical Science, Kanazawa University, 13-1 Takara-Machi, Kanazawa, Ishikawa 920-8640, Japan.,Miyarisan Pharmaceutical Co., LTD, 1-10-3, Kaminakazato, Kita-ku, Tokyo 114-0016, Japan
| | - Keiko Kobayashi
- Department of Microbiology, Faculty of Pharmaceutical Sciences, Tokushima Bunri University, Yamashiro-cho, Tokushima 770-8514, Japan
| | - Kazuaki Miyamoto
- Department of Microbiology, Faculty of Pharmaceutical Sciences, Tokushima Bunri University, Yamashiro-cho, Tokushima 770-8514, Japan
| | - Tohru Shimizu
- Department of Bacteriology, Graduate School of Medical Science, Kanazawa University, 13-1 Takara-Machi, Kanazawa, Ishikawa 920-8640, Japan
| | - Masahiro Nagahama
- Department of Microbiology, Faculty of Pharmaceutical Sciences, Tokushima Bunri University, Yamashiro-cho, Tokushima 770-8514, Japan
| |
Collapse
|
50
|
Abe M, Kobayashi T. Dynamics of sphingomyelin- and cholesterol-enriched lipid domains during cytokinesis. Methods Cell Biol 2016; 137:15-24. [PMID: 28065303 DOI: 10.1016/bs.mcb.2016.03.030] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Sphingomyelin (SM) and cholesterol (Chol) are the major lipids in the mammalian cells, which are mainly localized to the plasma membrane. Multiple lines of evidence suggest that these lipids form local lipid domains in the plasma membrane, playing functional roles in the cell. Several observations have suggested that these lipid domains are required for cytokinesis. In this chapter, we show the methods for visualizing SM-rich and/or Chol-rich membrane domains at cytokinesis by using specific lipid-binding proteins. Lysenin, equinatoxin II, perfringolysin O, and pleurotolysin A2 bind specifically to clustered SM-rich domain, dispersed SM-rich domain, Chol-rich domain, and SM/Chol mixtures, respectively. Nontoxic forms of these lipid-binding proteins fused to fluorescent proteins are used for imaging lipid domains in biological membranes at cytokinesis. The image analysis reveals the structures and functions of SM-rich and/or Chol-rich domains at the time of cytokinesis.
Collapse
Affiliation(s)
- M Abe
- RIKEN, Wako, Saitama, Japan
| | - T Kobayashi
- RIKEN, Wako, Saitama, Japan; CNRS, Illkirch, France
| |
Collapse
|