1
|
Rodrigues Rodrigues R, Alves MLF, Bilhalva MA, Kremer FS, Junior CM, Ferreira MRA, Galvão CC, Quatrin PHDN, Conceição FR. Large Clostridial Toxins: A Brief Review and Insights into Antigen Design for Veterinary Vaccine Development. Mol Biotechnol 2024:10.1007/s12033-024-01303-6. [PMID: 39472390 DOI: 10.1007/s12033-024-01303-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2024] [Accepted: 10/08/2024] [Indexed: 12/18/2024]
Abstract
The group of large clostridial toxins (LCTs) includes toxins A (TcdA) and B (TcdB) from Clostridioides difficile, hemorrhagic and lethal toxins from Paeniclostridium sordellii, alpha toxin from Clostridium novyi (TcnA), and cytotoxin from Clostridium perfringens. These toxins are associated with severe pathologies in livestock, including gas gangrene (P. sordellii and C. novyi), infectious necrotic hepatitis (C. novyi), avian necrotic enteritis (C. perfringens), and enterocolitis (C. difficile). Immunoprophylaxis is crucial for controlling these diseases, but traditional vaccines face production challenges, such as labor-intensive processes, and often exhibit low immunogenicity. This has led to increased interest in recombinant vaccines. While TcdA and TcdB are well-studied for human immunization, other LCTs remain poorly characterized and require further investigation. Therefore, this study emphasizes the importance of understanding lesser-explored toxins and proposes using immunoinformatics to identify their immunodominant regions. By mapping these regions using silico tools and considering their homology with TcdA and TcdB, the study aims to guide future research in veterinary vaccinology. It also explores alternatives to overcome the limitations of conventional and recombinant vaccines, offering guidelines for developing more effective vaccination strategies against severe infections in animals.
Collapse
Affiliation(s)
- Rafael Rodrigues Rodrigues
- Centro de Desenvolvimento Tecnológico, Biotecnologia, Universidade Federal de Pelotas - Campus Universitário, Capão do Leão, Rio Grande Do Sul, CEP 96160-000, Brazil.
| | - Mariliana Luiza Ferreira Alves
- Centro de Desenvolvimento Tecnológico, Biotecnologia, Universidade Federal de Pelotas - Campus Universitário, Capão do Leão, Rio Grande Do Sul, CEP 96160-000, Brazil
- Instituto Federal Sul-Rio-Grandense, IFSul, Campus Pelotas, Pelotas, Rio Grande Do Sul, Brasil
| | - Miguel Andrade Bilhalva
- Centro de Desenvolvimento Tecnológico, Biotecnologia, Universidade Federal de Pelotas - Campus Universitário, Capão do Leão, Rio Grande Do Sul, CEP 96160-000, Brazil
| | - Frederico Schmitt Kremer
- Centro de Desenvolvimento Tecnológico, Biotecnologia, Universidade Federal de Pelotas - Campus Universitário, Capão do Leão, Rio Grande Do Sul, CEP 96160-000, Brazil
| | - Clóvis Moreira Junior
- Centro de Desenvolvimento Tecnológico, Biotecnologia, Universidade Federal de Pelotas - Campus Universitário, Capão do Leão, Rio Grande Do Sul, CEP 96160-000, Brazil
| | - Marcos Roberto Alves Ferreira
- Centro de Desenvolvimento Tecnológico, Biotecnologia, Universidade Federal de Pelotas - Campus Universitário, Capão do Leão, Rio Grande Do Sul, CEP 96160-000, Brazil
| | - Cleideanny Cancela Galvão
- Centro de Desenvolvimento Tecnológico, Biotecnologia, Universidade Federal de Pelotas - Campus Universitário, Capão do Leão, Rio Grande Do Sul, CEP 96160-000, Brazil
| | - Pedro Henrique Dala Nora Quatrin
- Centro de Desenvolvimento Tecnológico, Biotecnologia, Universidade Federal de Pelotas - Campus Universitário, Capão do Leão, Rio Grande Do Sul, CEP 96160-000, Brazil
| | - Fabricio Rochedo Conceição
- Centro de Desenvolvimento Tecnológico, Biotecnologia, Universidade Federal de Pelotas - Campus Universitário, Capão do Leão, Rio Grande Do Sul, CEP 96160-000, Brazil
| |
Collapse
|
2
|
Ramos CP, Siqueira WF, Viana LA, Cunha JLR, Fujiwara RT, Amarante VS, Souza TGV, Silva ROS. Development of two recombinant vaccines against Clostridioides difficile infection and immunogenicity in pregnant sows and neonatal piglets. Anaerobe 2024; 89:102896. [PMID: 39127403 DOI: 10.1016/j.anaerobe.2024.102896] [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: 01/25/2024] [Revised: 06/11/2024] [Accepted: 08/06/2024] [Indexed: 08/12/2024]
Abstract
INTRODUCTION Clostridioides difficile is the main cause of antibiotic-associated diarrhea in humans and is a major enteropathogen in several animal species. In newborn piglets, colonic lesions caused by C. difficile A and B toxins (TcdA and TcdB, respectively) cause diarrhea and significant production losses. OBJECTIVE The present study aimed to develop two recombinant vaccines from immunogenic C-terminal fragments of TcdA and TcdB and evaluate the immune response in rabbits and in breeding sows. Two vaccines were produced: bivalent (rAB), consisting of recombinant fragments of TcdA and TcdB, and chimeric (rQAB), corresponding to the synthesis of the same fragments in a single protein. Groups of rabbits were inoculated with 10 or 50 μg of proteins adjuvanted with aluminum or 0.85 % sterile saline in a final volume of 1 mL/dose. Anti-TcdA and anti-TcdB IgG antibodies were detected in rabbits and sows immunized with both rAB and rQAB vaccines by ELISA. The vaccinated sows were inoculated intramuscularly with 20 μg/dose using a prime-boost approach. RESULTS Different antibody titers (p ≤ 0.05) were observed among the vaccinated groups of sows (rAB and rQAB) and control. Additionally, newborn piglets from vaccinated sows were also positive for anti-TcdA and anti-TcdB IgGs, in contrast to control piglets (p ≤ 0.05). Immunization of sows with the rQAB vaccine conferred higher anti-TcdA and anti-TcdB responses in piglets, suggesting the superiority of this compound over rAB. CONCLUSION The synthesized recombinant proteins were capable of inducing antibody titers against C. difficile toxins A and B in sows, and were passively transferred to piglets through colostrum.
Collapse
Affiliation(s)
- Carolina P Ramos
- Escola de Veterinária, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Williane F Siqueira
- Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Laila A Viana
- Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - João L R Cunha
- York Biomedical Research Institute, Department of Biology, University of York, York, UK
| | - Ricardo T Fujiwara
- Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Victor S Amarante
- Escola de Veterinária, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Thayanne G V Souza
- Escola de Veterinária, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Rodrigo O S Silva
- Escola de Veterinária, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil.
| |
Collapse
|
3
|
Aminzadeh A, Hilgers L, Paul Platenburg P, Riou M, Perrot N, Rossignol C, Cauty A, Barc C, Jørgensen R. Immunogenicity and safety in rabbits of a Clostridioides difficile vaccine combining novel toxoids and a novel adjuvant. Vaccine 2024; 42:1582-1592. [PMID: 38336558 DOI: 10.1016/j.vaccine.2024.01.076] [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: 12/26/2022] [Revised: 12/12/2023] [Accepted: 01/23/2024] [Indexed: 02/12/2024]
Abstract
Clostridioides difficile infection (CDI) is a serious healthcare-associated disease, causing symptoms such as diarrhea and pseudomembranous colitis. The major virulence factors responsible for the disease symptoms are two secreted cytotoxic proteins, TcdA and TcdB. A parenteral vaccine based on formaldehyde-inactivated TcdA and TcdB supplemented with alum adjuvant, has previously been investigated in humans but resulted in an insufficient immune response. In search for an improved response, we investigated a novel toxin inactivation method and a novel, potent adjuvant. Inactivation of toxins by metal-catalyzed oxidation (MCO) was previously shown to preserve neutralizing epitopes and to annihilate reversion to toxicity. The immunogenicity and safety of TcdA and TcdB inactivated by MCO and combined with a novel carbohydrate fatty acid monosulphate ester-based (CMS) adjuvant were investigated in rabbits. Two or three intramuscular immunizations generated high serum IgG and neutralizing antibody titers against both toxins. The CMS adjuvant increased antibody responses to both toxins while an alum adjuvant control was effective only against TcdA. Systemic safety was evaluated by monitoring body weight, body temperature, and analysis of red and white blood cell counts shortly after immunization. Local safety was assessed by histopathologic examination of the injection site at the end of the study. Body weight gain was constant in all groups. Body temperature increased up to 1 ˚C one day after the first immunization but less after the second or third immunization. White blood cell counts, and percentage of neutrophils increased one day after immunization with CMS-adjuvanted vaccines, but not with alum. Histopathology of the injection sites 42 days after the last injection did not reveal any abnormal tissue reactions. From this study, we conclude that TcdA and TcdB inactivated by MCO and combined with CMS adjuvant demonstrated promising immunogenicity and safety in rabbits and could be a candidate for a vaccine against CDI.
Collapse
Affiliation(s)
- Aria Aminzadeh
- Proxi Biotech ApS, Egeskellet 6, 2000 Frederiksberg, Denmark; Department of Science and Environment, University of Roskilde, 4000 Roskilde, Denmark
| | - Luuk Hilgers
- LiteVax BV, Akkersestraat 50, 4061BJ Ophemert, the Netherlands
| | | | - Mickaël Riou
- INRAE, UE-1277 Plateforme d'Infectiologie expérimentale (PFIE), Centre Val de Loire, 37380 Nouzilly, France
| | - Noémie Perrot
- INRAE, UE-1277 Plateforme d'Infectiologie expérimentale (PFIE), Centre Val de Loire, 37380 Nouzilly, France
| | - Christelle Rossignol
- INRAE-Université de Tours, UMR-1282 Infectiologie et Santé publique (ISP), équipe IMI, Centre Val de Loire, 37380 Nouzilly, France
| | - Axel Cauty
- INRAE, UE-1277 Plateforme d'Infectiologie expérimentale (PFIE), Centre Val de Loire, 37380 Nouzilly, France
| | - Céline Barc
- INRAE, UE-1277 Plateforme d'Infectiologie expérimentale (PFIE), Centre Val de Loire, 37380 Nouzilly, France
| | - René Jørgensen
- Proxi Biotech ApS, Egeskellet 6, 2000 Frederiksberg, Denmark; Department of Science and Environment, University of Roskilde, 4000 Roskilde, Denmark.
| |
Collapse
|
4
|
Ding UZ, Ooi L, Wu HHL, Chinnadurai R. Clostridioides difficile Infection in Kidney Transplant Recipients. Pathogens 2024; 13:140. [PMID: 38392878 PMCID: PMC10892420 DOI: 10.3390/pathogens13020140] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2023] [Revised: 01/30/2024] [Accepted: 02/02/2024] [Indexed: 02/25/2024] Open
Abstract
Clostridioides difficile (C. difficile) is a bacterial organism that typically infects the colon, which has had its homeostasis of healthy gut microbiota disrupted by antibiotics or other interventions. Patients with kidney transplantation are a group that are susceptible to C. difficile infection (CDI) and have poorer outcomes with CDI given that they conventionally require long-term immunosuppression to minimize their risk of graft rejection, weakening their responses to infection. Recognizing the risk factors and complex pathophysiological processes that exist between immunosuppression, dysbiosis, and CDI is important when making crucial clinical decisions surrounding the management of this vulnerable patient cohort. Despite the clinical importance of this topic, there are few studies that have evaluated CDI in the context of kidney transplant recipients and other solid organ transplant populations. The current recommendations on CDI management in kidney transplant and solid organ transplant recipients are mostly extrapolated from data relating to CDI management in the general population. We provide a narrative review that discusses the available evidence examining CDI in solid organ transplant recipients, with a particular focus on the kidney transplant recipient, from the epidemiology of CDI, clinical features and implications of CDI, potential risk factors of CDI, and, ultimately, prevention and management strategies for CDI, with the aim of providing areas for future research development in this topic area.
Collapse
Affiliation(s)
- UZhe Ding
- Department of Renal Medicine, Northern Care Alliance NHS Foundation Trust, Salford M6 8HD, UK; (U.D.); (L.O.); (R.C.)
| | - Lijin Ooi
- Department of Renal Medicine, Northern Care Alliance NHS Foundation Trust, Salford M6 8HD, UK; (U.D.); (L.O.); (R.C.)
| | - Henry H. L. Wu
- Renal Research Laboratory, Kolling Institute of Medical Research, Royal North Shore Hospital, The University of Sydney, Sydney, NSW 2065, Australia
| | - Rajkumar Chinnadurai
- Department of Renal Medicine, Northern Care Alliance NHS Foundation Trust, Salford M6 8HD, UK; (U.D.); (L.O.); (R.C.)
- Faculty of Biology, Medicine & Health, The University of Manchester, Manchester M1 7HR, UK
| |
Collapse
|
5
|
Karnchanapandh K, Hanpaibool C, Sanachai K, Rungrotmongkol T. Elucidation of bezlotoxumab binding specificity to toxin B in Clostridioides difficile. J Biomol Struct Dyn 2024; 42:1617-1628. [PMID: 37098802 DOI: 10.1080/07391102.2023.2201360] [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: 12/26/2022] [Accepted: 04/05/2023] [Indexed: 04/27/2023]
Abstract
C. difficile or Clostridioides difficile infection (CDI) is currently one of the major causes of epidemics worldwide. Toxin B from Clostridioides difficile toxin B (TcdB) infection is the main target protein inhibiting CDI recurrence. Clinical research suggested that bezlotoxumab's (Bez) efficiency is significantly reduced in neutralizing the B2 strain compared to the B1 strain. The monoclonal antibody (mAb) functions by binding to the epitope 1 and 2 regions in the combined repetitive oligopeptide (CROP) domain. Some binding residues are distinctively different between B1 and B2 strains. In this work, we aimed to elucidate and compare insights into the interaction of toxins B1 and B2 in complex with Bez by using all-atom molecular dynamics (MD) simulations and binding free energy calculations. The predicted ΔGbinding values suggested that the antibody (Ab) could bind to toxin B1 significantly better than B2, supported by higher salt bridge and hydrogen bonding (H-bonding) interactions, as well as the number of contact residues between the two focused proteins. The toxin B1 residues important for binding with Bez were E1878, T1901, E1902, F1905, N1941, V1946, N2031, T2032, E2033, V2076, V2077, and E2092. The lower susceptibility of Bez towards toxin B2 was primarily due to a change of residue E2033 from glutamate to alanine (A2033) and the loss of E1878 and E1902 contributions, as determined by the intermolecular interaction changes from the dynamic residue interaction network (dRIN) analysis. The obtained data strengthen our understanding of Bez/toxin B binding.
Collapse
Affiliation(s)
- Kun Karnchanapandh
- Program in Bioinformatics and Computational Biology, Graduate School, Chulalongkorn University, Bangkok, Thailand
| | - Chonnikan Hanpaibool
- Center of Excellence in Biocatalyst and Sustainable Biotechnology, Department of Biochemistry, Faculty of Science, Chulalongkorn University, Bangkok, Thailand
| | - Kamonpan Sanachai
- Department of Biochemistry, Faculty of Science, Khon Kaen University, Khon Kaen, Thailand
| | - Thanyada Rungrotmongkol
- Program in Bioinformatics and Computational Biology, Graduate School, Chulalongkorn University, Bangkok, Thailand
- Center of Excellence in Biocatalyst and Sustainable Biotechnology, Department of Biochemistry, Faculty of Science, Chulalongkorn University, Bangkok, Thailand
| |
Collapse
|
6
|
Campidelli C, Bruxelle JF, Collignon A, Péchiné S. Immunization Strategies Against Clostridioides difficile. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2024; 1435:117-150. [PMID: 38175474 DOI: 10.1007/978-3-031-42108-2_7] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2024]
Abstract
Clostridioides difficile (C. difficile) infection (CDI) is an important healthcare but also a community-associated disease. CDI is considered a public health threat and an economic burden. A major problem is the high rate of recurrences. Besides classical antibiotic treatments, new therapeutic strategies are needed to prevent infection, to treat patients, and to prevent recurrences. If fecal transplantation has been recommended to treat recurrences, another key approach is to elicit immunity against C. difficile and its virulence factors. Here, after a summary concerning the virulence factors, the host immune response against C. difficile, and its role in the outcome of disease, we review the different approaches of passive immunotherapies and vaccines developed against CDI. Passive immunization strategies are designed in function of the target antigen, the antibody-based product, and its administration route. Similarly, for active immunization strategies, vaccine antigens can target toxins or surface proteins, and immunization can be performed by parenteral or mucosal routes. For passive immunization and vaccination as well, we first present immunization assays performed in animal models and second in humans and associated clinical trials. The different studies are presented according to the mode of administration either parenteral or mucosal and the target antigens and either toxins or colonization factors.
Collapse
Affiliation(s)
- Camille Campidelli
- Université Paris-Saclay, INRAE, AgroParisTech, Micalis Institute, Jouy-en-Josas, France
| | - Jean-François Bruxelle
- CIRI-Centre International de Recherche en Infectiologie, Université de Lyon, Université Claude Bernard Lyon 1, Inserm U1111, CNRS UMR5308, ENS Lyon, Lyon, France
| | - Anne Collignon
- Université Paris-Saclay, INRAE, AgroParisTech, Micalis Institute, Jouy-en-Josas, France
| | - Severine Péchiné
- Université Paris-Saclay, INRAE, AgroParisTech, Micalis Institute, Jouy-en-Josas, France.
| |
Collapse
|
7
|
Sun Y, Zhang Y, Yu H, Saint Fleur A, Yu D, Yang Z, Feng H. A fine-tuned yeast surface-display/secretion platform enables the rapid discovery of neutralizing antibodies against Clostridioides difficile toxins. Microb Cell Fact 2023; 22:194. [PMID: 37749574 PMCID: PMC10519002 DOI: 10.1186/s12934-023-02200-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2023] [Accepted: 09/06/2023] [Indexed: 09/27/2023] Open
Abstract
BACKGROUND Neutralizing antibody plays a key role in protecting hosts from invasive pathogens and their virulent components. Current high-throughput assays for antibody screening are based on binding activities. However, those antibodies with high affinity may not have neutralizing activities. Subsequent functionality assays are necessary to identify neutralizing antibodies from binders with high affinity to their target antigens, which is laborious and time-consuming. Therefore, a versatile platform that can rapidly identify antibodies with both high binding affinity and neutralizing activity is desired to curb future pandemics like COVID-19. RESULTS In this proof-of-concept study, we adapted Saccharomyces cerevisiae to either display human antibodies on the yeast surface or secrete soluble antibodies into the cultivation supernatant under a controllable 'switch' through different carbon source induced promoters. Initially, an engineered chimeric-bispecific Fab antibody, derived from humanized nanobodies against both Clostridioides difficile toxin A and B (TcdA and TcdB), was successfully expressed either on the yeast cell surface or in the culture medium with intact bioactivity, suggesting the applicability of our system in antibody display and secretion. Next, a combinatorial Fab library was constructed from B cells isolated from a convalescent patient with a high serological neutralizing titer against TcdB. Following three rounds of magnetic bead enrichment and one round of flow cytometry sorting, antibodies against TcdB were enriched efficiently. We then sorted out single binders with high binding affinity and induced them to express soluble antibodies in culture medium. The neutralizing activity of culture supernatant was analyzed using cell-based assay immediately. This way, we rapidly identified two unique neutralizers (out of seven binders) that can neutralize the cytotoxicity of TcdB. CONCLUSION The antibody screening platform described here simplifies the neutralizing antibody discovery procedure and will be an attractive alternative for screening functional antibodies against infectious diseases.
Collapse
Affiliation(s)
- Ying Sun
- Department of Microbial Pathogenesis, School of Dentistry, University of Maryland, Baltimore, MD, 21201, USA.
- Department of Pathogen Biology, School of Basic Medical Sciences, China Medical University, Shenyang, 110122, China.
| | - Yongrong Zhang
- Department of Microbial Pathogenesis, School of Dentistry, University of Maryland, Baltimore, MD, 21201, USA
| | - Hua Yu
- Fzata, Inc, Halethorpe, MD, 21227, USA
| | - Ashley Saint Fleur
- Department of Microbial Pathogenesis, School of Dentistry, University of Maryland, Baltimore, MD, 21201, USA
| | - Di Yu
- Department of Microbial Pathogenesis, School of Dentistry, University of Maryland, Baltimore, MD, 21201, USA
| | | | - Hanping Feng
- Department of Microbial Pathogenesis, School of Dentistry, University of Maryland, Baltimore, MD, 21201, USA.
- Fzata, Inc, Halethorpe, MD, 21227, USA.
| |
Collapse
|
8
|
Naz F, Petri WA. Host Immunity and Immunization Strategies for Clostridioides difficile Infection. Clin Microbiol Rev 2023; 36:e0015722. [PMID: 37162338 PMCID: PMC10283484 DOI: 10.1128/cmr.00157-22] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/11/2023] Open
Abstract
Clostridioides difficile infection (CDI) represents a significant challenge to public health. C. difficile-associated mortality and morbidity have led the U.S. CDC to designate it as an urgent threat. Moreover, recurrence or relapses can occur in up to a third of CDI patients, due in part to antibiotics being the primary treatment for CDI and the major cause of the disease. In this review, we summarize the current knowledge of innate immune responses, adaptive immune responses, and the link between innate and adaptive immune responses of the host against CDI. The other major determinants of CDI, such as C. difficile toxins, the host microbiota, and related treatments, are also described. Finally, we discuss the known therapeutic approaches and the current status of immunization strategies for CDI, which might help to bridge the knowledge gap in the generation of therapy against CDI.
Collapse
Affiliation(s)
- Farha Naz
- Division of Infectious Diseases and International Health, University of Virginia School of Medicine, Charlottesville, Virginia, USA
| | - William A. Petri
- Division of Infectious Diseases and International Health, University of Virginia School of Medicine, Charlottesville, Virginia, USA
- Department of Pathology, University of Virginia School of Medicine, Charlottesville, Virginia, USA
| |
Collapse
|
9
|
Jiang B, Yu D, Zhang Y, Hamza T, Feng H, Hoag SW. Delivery of a therapeutic antibody to the lower gastrointestinal tract for the treatment of Clostridium difficile infection (CDI). Pharm Dev Technol 2023; 28:232-239. [PMID: 36789978 DOI: 10.1080/10837450.2023.2174553] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/16/2023]
Abstract
The colonic delivery system of toxin neutralizing antibody is a promising method for treating Clostridium difficile infection (CDI) and has some advantages over the parental administration of a neutralizing antibody. However, colonic delivery of biologics presents several challenges, including instability of biologics during encapsulation into the delivery system and harsh conditions in the upper GI tract. In this work, we described a multi-particulate delivery system encapsulating a tetra-valent antibody ABAB-IgG1 with the potential to treat CDI. This work first approved that the cecum injection of ABAB-IgG1 into the lower GI tract of mice could relieve the symptoms, enhance the clinical score, and improve the survival rate of mice during CDI. Then, the antibody was spray layered onto mannitol beads and then enteric coated with pH-sensitive polymers to achieve colon-targeting release. The in vitro release of antibody from the multi-particulate system and the pH-sensitive release of antibody was monitored. The in vivo efficacy of this system was further examined and confirmed in mice and hamsters. In summary, the findings of this study should provide practical information and potential treatment options for CDI through colonic delivery of antibody therapeutics to the lower GI tract using a multi-particulate delivery system.
Collapse
Affiliation(s)
- Bowen Jiang
- Department of Pharmaceutical Sciences, University of Maryland, Baltimore, MD, USA
| | - Dongyue Yu
- Department of Pharmaceutical Sciences, University of Maryland, Baltimore, MD, USA
| | - Yongrong Zhang
- Department of Microbial Pathogenesis, University of Maryland, Baltimore, MD, USA
| | - Therwa Hamza
- Department of Microbial Pathogenesis, University of Maryland, Baltimore, MD, USA
| | - Hanping Feng
- Department of Microbial Pathogenesis, University of Maryland, Baltimore, MD, USA.,FZata Inc, Baltimore, MD, USA
| | - Stephen W Hoag
- Department of Pharmaceutical Sciences, University of Maryland, Baltimore, MD, USA
| |
Collapse
|
10
|
Chen B, Perry K, Jin R. Neutralizing epitopes on Clostridioides difficile toxin A revealed by the structures of two camelid VHH antibodies. Front Immunol 2022; 13:978858. [PMID: 36466927 PMCID: PMC9709291 DOI: 10.3389/fimmu.2022.978858] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2022] [Accepted: 10/31/2022] [Indexed: 11/18/2022] Open
Abstract
Toxin A (TcdA) and toxin B (TcdB) are two key virulence factors secreted by Clostridioides difficile, which is listed as an urgent threat by the CDC. These two large homologous exotoxins are mainly responsible for diseases associated with C. difficile infection (CDI) with symptoms ranging from diarrhea to life threatening pseudomembranous colitis. Single-domain camelid antibodies (VHHs) AH3 and AA6 are two potent antitoxins against TcdA, which when combined with two TcdB-targeting VHHs showed effective protection against both primary and recurrent CDI in animal models. Here, we report the co-crystal structures of AH3 and AA6 when they form complexes with the glucosyltransferase domain (GTD) and a fragment of the delivery and receptor-binding domain (DRBD) of TcdA, respectively. Based on these structures, we find that AH3 binding enhances the overall stability of the GTD and interferes with its unfolding at acidic pH, and AA6 may inhibit the pH-dependent conformational changes in the DRBD that is necessary for pore formation of TcdA. These studies reveal two functionally critical epitopes on TcdA and shed new insights into neutralizing mechanisms and potential development of epitope-focused vaccines against TcdA.
Collapse
Affiliation(s)
- Baohua Chen
- Department of Physiology and Biophysics, School of Medicine, University of California, Irvine, CA, United States
| | - Kay Perry
- NE-CAT, Advanced Photon Source, Argonne National Laboratory, Argonne, IL, United States,Department of Chemistry and Chemical Biology, Cornell University, Argonne, IL, United States
| | - Rongsheng Jin
- Department of Physiology and Biophysics, School of Medicine, University of California, Irvine, CA, United States,*Correspondence: Rongsheng Jin,
| |
Collapse
|
11
|
Wang S, Zhu D, Sun X. Development of an Effective Nontoxigenic Clostridioides difficile-Based Oral Vaccine against C. difficile Infection. Microbiol Spectr 2022; 10:e0026322. [PMID: 35583336 PMCID: PMC9241731 DOI: 10.1128/spectrum.00263-22] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2022] [Accepted: 04/21/2022] [Indexed: 11/20/2022] Open
Abstract
The symptoms of Clostridioides difficile infection (CDI) are largely attributed to two C. difficile toxins, TcdA and TcdB. Significant efforts have been devoted to developing vaccines targeting both toxins through parenteral immunization routes. Recently, we generated a novel chimeric protein (designated Tcd169), comprised of the glucosyltransferase domain (GT), the cysteine protease domain (CPD), and the receptor binding domain (RBD) of TcdB, and the RBD of TcdA. Parenteral immunizations with Tcd169 provide mice effective protection against infection with a ribotype (RT) 027 C. difficile strain. In this study, we expressed Tcd169 in a nontoxigenic C. difficile CCUG37785 strain (designated NTCD), resulting in strain NTCD_Tcd169 to develop an oral vaccine that can target both C. difficile toxins and colonization/adhesion factors. Oral immunizations with NTCD_Tcd169 spores induced systematic and mucosal antibody responses against, not only both toxins, but also C. difficile flagellins (FliC/FliD). Intriguingly yet importantly, anti-Tcd169 sera raised against Tcd169 protein were significantly cross-reactive with FliC/FliD and two surface layer proteins (SlpA and Cwp2). Oral immunizations with NTCD_Tcd169 spores provided mice effective protection against infection with a hypervirulent RT027 C. difficile strain R20291and significantly reduced R20291spore numbers in feces compared with NTCD or PBS immunized mice. These results imply that the genetically modified, nontoxigenic C. difficile strain expressing Tcd169 may represent a novel mucosal vaccine candidate against CDI. IMPORTANCE Clostridioides difficile is an enteric pathogen, and symptoms of C. difficile infection (CDI) are mainly by two exotoxins TcdA and TcdB. Active vaccination is cost-effective approach to prevent CDI and high rates of recurrence. Ideally, vaccines should target both C. difficile toxins and cell/spore colonization. In this study, we expressed immunodominant fragments of TcdA and TcdB (i.e., Tcd169) in a nontoxigenic C. difficile CCUG37785 strain, generating a promising oral/mucosal vaccine candidate against CDI, by targeting both toxins and colonization of pathogenic C. difficile strains. Importantly, anti-Tcd169 sera raised against Tcd169 protein were significantly cross-reactive with FliC/FliD and two surface layer proteins (SlpA and Cwp2), and all of which are involved in C. difficile adhesion/colonization in vitro and in vivo.
Collapse
Affiliation(s)
- Shaohui Wang
- Department of Molecular Medicine, Morsani College of Medicine, University of South Florida, Tampa, Florida, USA
| | - Duolong Zhu
- Department of Molecular Medicine, Morsani College of Medicine, University of South Florida, Tampa, Florida, USA
| | - Xingmin Sun
- Department of Molecular Medicine, Morsani College of Medicine, University of South Florida, Tampa, Florida, USA
| |
Collapse
|
12
|
Nyblade C, Parreno V, Zhou P, Hensley C, Oakes V, Mahsoub HM, Kiley K, Frazier M, Frazier A, Zhang Y, Feng H, Yuan L. Establishment of a gnotobiotic pig model of Clostridioides difficile infection and disease. Gut Pathog 2022; 14:22. [PMID: 35668452 PMCID: PMC9169267 DOI: 10.1186/s13099-022-00496-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/11/2022] [Accepted: 05/11/2022] [Indexed: 11/10/2022] Open
Abstract
Clostridioides difficile (C. difficile) is a gram-positive, spore-forming, anaerobic bacterium known to be the most common cause of hospital-acquired and antibiotic-associated diarrhea. C. difficile infection rates are on the rise worldwide and treatment options are limited, indicating a clear need for novel therapeutics. Gnotobiotic piglets are an excellent model to reproduce the acute pseudomembranous colitis (PMC) caused by C. difficile due to their physiological similarities to humans and high susceptibility to infection. Here, we established a gnotobiotic pig model of C. difficile infection and disease using a hypervirulent strain. C. difficile-infected pigs displayed classic signs of C. difficile infection, including severe diarrhea and weight loss. Inoculated pigs had severe gross and microscopic intestinal lesions. C. difficile infection caused an increase in pro-inflammatory cytokines in samples of serum, large intestinal contents, and pleural effusion. C. difficile spores and toxins were detected in the feces of inoculated animals as tested by anaerobic culture and cytotoxicity assays. Successful establishment of this model is key for future work as therapeutics can be evaluated in an environment that accurately mimics what happens in humans. The model is especially suitable for evaluating potential prophylactics and therapeutics, including vaccines and passive immune strategies.
Collapse
Affiliation(s)
- Charlotte Nyblade
- Department of Biomedical Sciences and Pathobiology, Virginia-Maryland College of Veterinary Medicine, Virginia Polytechnic Institute and State University, Blacksburg, VA, 24061, USA
| | - Viviana Parreno
- Department of Biomedical Sciences and Pathobiology, Virginia-Maryland College of Veterinary Medicine, Virginia Polytechnic Institute and State University, Blacksburg, VA, 24061, USA
- INCUINTA, Instituto Nacional de Tecnologia Agropecuaria (INTA), Instituto de Virologia e Innovaciones Tecnologicas (IVIT INTA CONICET), Buenos Aires, Argentina
| | - Peng Zhou
- Department of Biomedical Sciences and Pathobiology, Virginia-Maryland College of Veterinary Medicine, Virginia Polytechnic Institute and State University, Blacksburg, VA, 24061, USA
| | - Casey Hensley
- Department of Biomedical Sciences and Pathobiology, Virginia-Maryland College of Veterinary Medicine, Virginia Polytechnic Institute and State University, Blacksburg, VA, 24061, USA
| | - Vanessa Oakes
- Department of Biomedical Sciences and Pathobiology, Virginia-Maryland College of Veterinary Medicine, Virginia Polytechnic Institute and State University, Blacksburg, VA, 24061, USA
| | - Hassan M Mahsoub
- Department of Biomedical Sciences and Pathobiology, Virginia-Maryland College of Veterinary Medicine, Virginia Polytechnic Institute and State University, Blacksburg, VA, 24061, USA
- Center for Emerging, Zoonotic, and Arthropod-Borne Pathogens, Virginia Polytechnic Institute and State University, Blacksburg, VA, 24061, USA
| | - Kelsey Kiley
- Department of Biomedical Sciences and Pathobiology, Virginia-Maryland College of Veterinary Medicine, Virginia Polytechnic Institute and State University, Blacksburg, VA, 24061, USA
| | - Maggie Frazier
- Department of Biomedical Sciences and Pathobiology, Virginia-Maryland College of Veterinary Medicine, Virginia Polytechnic Institute and State University, Blacksburg, VA, 24061, USA
| | - Annie Frazier
- Department of Biomedical Sciences and Pathobiology, Virginia-Maryland College of Veterinary Medicine, Virginia Polytechnic Institute and State University, Blacksburg, VA, 24061, USA
| | - Yongrong Zhang
- Department of Microbial Pathogenesis, University of Maryland at Baltimore, Baltimore, MD, 21201, USA
| | - Hanping Feng
- Department of Microbial Pathogenesis, University of Maryland at Baltimore, Baltimore, MD, 21201, USA
| | - Lijuan Yuan
- Department of Biomedical Sciences and Pathobiology, Virginia-Maryland College of Veterinary Medicine, Virginia Polytechnic Institute and State University, Blacksburg, VA, 24061, USA.
- Center for Emerging, Zoonotic, and Arthropod-Borne Pathogens, Virginia Polytechnic Institute and State University, Blacksburg, VA, 24061, USA.
| |
Collapse
|
13
|
Alves MLF, Ferreira MRA, Rodrigues RR, Conceição FR. Clostridium haemolyticum, a review of beta toxin and insights into the antigen design for vaccine development. Mol Immunol 2022; 148:45-53. [PMID: 35665660 DOI: 10.1016/j.molimm.2022.05.007] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2022] [Revised: 05/01/2022] [Accepted: 05/16/2022] [Indexed: 01/03/2023]
Abstract
Phospholipases C (PLCs) represent an important group of lethal toxins produced by pathogenic bacteria of the Clostridium genus, including the beta toxin of C. haemolyticum. Bacillary hemoglobinuria in cattle and sheep is the main disease caused by this pathogen and its incidence can be reduced by annual vaccination of herds. Currently, widely used vaccines depend on cultivating the pathogen and obtaining high concentrations of the toxin, disadvantages that can be overcome with the use of recombinant vaccines. In the development of this new generation of immunizing agents, identifying and understanding the structural and immunological aspects of the antigen are crucial steps, but despite this, the beta toxin is poorly characterized. Fortunately, the time and resources required for these investigations can be reduced using immunoinformatics. To advance the development of recombinant vaccines, in addition to a brief review of the structural and immunological aspects of beta toxin, this work provides in silico mapping of immunodominant regions to guide future vaccinology studies against C. haemolyticum. A review of alternatives to overcome the limitations of beta toxin vaccines (conventional or recombinant) is also proposed.
Collapse
Affiliation(s)
- Mariliana Luiza Ferreira Alves
- Instituto Federal Sul-rio-grandense - IFSUL, Praça Vinte de Setembro, 455, Centro, Pelotas CEP 96.015-360, RS, Brazil; Centro de Desenvolvimento Tecnológico, Núcleo de Biotecnologia, Universidade Federal de Pelotas, CP 354, Pelotas CEP 96.160-000, RS, Brazil.
| | - Marcos Roberto Alves Ferreira
- Centro de Desenvolvimento Tecnológico, Núcleo de Biotecnologia, Universidade Federal de Pelotas, CP 354, Pelotas CEP 96.160-000, RS, Brazil
| | - Rafael Rodrigues Rodrigues
- Centro de Desenvolvimento Tecnológico, Núcleo de Biotecnologia, Universidade Federal de Pelotas, CP 354, Pelotas CEP 96.160-000, RS, Brazil
| | - Fabricio Rochedo Conceição
- Centro de Desenvolvimento Tecnológico, Núcleo de Biotecnologia, Universidade Federal de Pelotas, CP 354, Pelotas CEP 96.160-000, RS, Brazil
| |
Collapse
|
14
|
Kociolek LK, Zackular JP, Savidge T. Translational Aspects of the Immunology of Clostridioides difficile Infection: Implications for Pediatric Populations. J Pediatric Infect Dis Soc 2021; 10:S8-S15. [PMID: 34791392 PMCID: PMC8600028 DOI: 10.1093/jpids/piab089] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Clostridioides difficile has become the most common healthcare-associated pathogen in the United States, leading the US Centers for Disease Control and Prevention (CDC) to classify C. difficile as an "urgent" public health threat that requires "urgent and aggressive action." This call to action has led to new discoveries that have advanced our understanding of Clostridioides difficile infection (CDI) immunology and clinical development of immunologic-based therapies for CDI prevention. However, CDI immunology research has been limited in pediatric populations, and several unanswered questions remain regarding the function of host immune response in pediatric CDI pathogenesis and the potential role of immunologic-based therapies in children. This review summarizes the innate and adaptive immune responses previously characterized in animals and humans and provides a current update on clinical development of immunologic-based therapies for CDI prevention in adults and children. These data inform the future research needs for children.
Collapse
Affiliation(s)
- Larry K Kociolek
- Department of Pediatrics, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA,Division of Infectious Diseases, Ann & Robert H. Lurie Children’s Hospital of Chicago, Chicago, Illinois, USA,Corresponding Author: Larry K. Kociolek, MD, MSCI, Division of Pediatric Infectious Diseases, Ann & Robert H. Lurie Children’s Hospital of Chicago, 225 E. Chicago Ave, Box 20, Chicago, IL 60611, USA. E-mail:
| | - Joseph P Zackular
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA,Division of Protective Immunity, Department of Pathology and Laboratory Medicine, Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania, USA,Institute for Immunology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Tor Savidge
- Department of Pathology & Immunology, Baylor College of Medicine & Texas Children’s Hospital, Houston, Texas, USA
| |
Collapse
|
15
|
Chen K, Zhu Y, Zhang Y, Hamza T, Yu H, Saint Fleur A, Galen J, Yang Z, Feng H. A probiotic yeast-based immunotherapy against Clostridioides difficile infection. Sci Transl Med 2021; 12:12/567/eaax4905. [PMID: 33115949 DOI: 10.1126/scitranslmed.aax4905] [Citation(s) in RCA: 67] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2019] [Revised: 02/12/2020] [Accepted: 10/07/2020] [Indexed: 12/12/2022]
Abstract
Antibiotic-resistant Clostridioides difficile is an anaerobic Gram-positive bacterium that colonizes the colon and is responsible for more than 29,000 deaths in the United States each year. Hence, C. difficile infection (CDI) poses an urgent threat to public health. Antibody-mediated neutralization of TcdA and TcdB toxins, the major virulence factors of CDI, represents an effective strategy to combat the disease without invoking antibiotic resistance. However, current antitoxin approaches are mostly based on parenteral infusion of monoclonal antibodies that are costly, narrow spectrum, and not optimized against the intestinal disease. Here, we engineered probiotic Saccharomyces boulardii to constitutively secrete a single tetra-specific antibody that potently and broadly neutralized both toxins and demonstrated protection against primary and recurrent CDI in both prophylactic and therapeutic mouse models of disease. This yeast immunotherapy is orally administered, can be used concurrently with antibiotics, and may have potential as a prophylactic against CDI risk and as a therapeutic for patients with CDI.
Collapse
Affiliation(s)
- Kevin Chen
- Department of Microbial Pathogenesis, University of Maryland School of Dentistry, Baltimore, MD 21201, USA
| | - Yixuan Zhu
- Center for Vaccine Development, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Yongrong Zhang
- Department of Microbial Pathogenesis, University of Maryland School of Dentistry, Baltimore, MD 21201, USA
| | - Therwa Hamza
- Department of Microbial Pathogenesis, University of Maryland School of Dentistry, Baltimore, MD 21201, USA
| | - Hua Yu
- FZata Inc., Halethorpe, MD 21227, USA
| | - Ashley Saint Fleur
- Department of Microbial Pathogenesis, University of Maryland School of Dentistry, Baltimore, MD 21201, USA
| | - James Galen
- Center for Vaccine Development, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | | | - Hanping Feng
- Department of Microbial Pathogenesis, University of Maryland School of Dentistry, Baltimore, MD 21201, USA.
| |
Collapse
|
16
|
Karyal C, Hughes J, Kelly ML, Luckett JC, Kaye PV, Cockayne A, Minton NP, Griffin R. Colonisation Factor CD0873, an Attractive Oral Vaccine Candidate against Clostridioides difficile. Microorganisms 2021; 9:microorganisms9020306. [PMID: 33540694 PMCID: PMC7913071 DOI: 10.3390/microorganisms9020306] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2020] [Revised: 01/18/2021] [Accepted: 01/29/2021] [Indexed: 12/12/2022] Open
Abstract
Clostridioides difficile is the main cause of health-care-associated infectious diarrhoea. Toxins, TcdA and TcdB, secreted by this bacterium damage colonic epithelial cells and in severe cases this culminates in pseudomembranous colitis, toxic megacolon and death. Vaccines in human trials have focused exclusively on the parenteral administration of toxin-based formulations. These vaccines promote toxin-neutralising serum antibodies but fail to confer protection from infection in the gut. An effective route to immunise against gut pathogens and stimulate a protective mucosal antibody response (secretory immunoglobulin A, IgA) at the infection site is the oral route. Additionally, oral immunisation generates systemic antibodies (IgG). Using this route, two different antigens were tested in the hamster model: The colonisation factor CD0873 and a TcdB fragment. Animals immunised with CD0873 generated a significantly higher titre of sIgA in intestinal fluid and IgG in serum compared to naive animals, which significantly inhibited the adherence of C. difficile to Caco-2 cells. Following challenge with a hypervirulent isolate, the CD0873-immunised group showed a mean increase of 80% in time to experimental endpoint compared to naïve animals. Survival and body condition correlated with bacterial clearance and reduced pathology in the cecum. Our findings advocate CD0873 as a promising oral vaccine candidate against C. difficile.
Collapse
Affiliation(s)
- Cansu Karyal
- Synthetic Biology Research Centre, The University of Nottingham Biodiscovery Institute, University Park, Nottingham NG7 2RD, UK; (C.K.); (J.H.); (M.L.K.); (A.C.); (N.P.M.)
| | - Jaime Hughes
- Synthetic Biology Research Centre, The University of Nottingham Biodiscovery Institute, University Park, Nottingham NG7 2RD, UK; (C.K.); (J.H.); (M.L.K.); (A.C.); (N.P.M.)
| | - Michelle L. Kelly
- Synthetic Biology Research Centre, The University of Nottingham Biodiscovery Institute, University Park, Nottingham NG7 2RD, UK; (C.K.); (J.H.); (M.L.K.); (A.C.); (N.P.M.)
| | - Jeni C. Luckett
- The University of Nottingham Biodiscovery Institute, University Park, Nottingham NG7 2RD, UK;
| | - Philip V. Kaye
- Department of Histopathology, Queen’s Medical Centre, Nottingham University Hospitals NHS Trust, Nottingham NG7 2UH, UK;
- National Institute for Health Research (NIHR) Nottingham Biomedical Research Centre (BRC), Nottingham NG7 2UH, UK
| | - Alan Cockayne
- Synthetic Biology Research Centre, The University of Nottingham Biodiscovery Institute, University Park, Nottingham NG7 2RD, UK; (C.K.); (J.H.); (M.L.K.); (A.C.); (N.P.M.)
| | - Nigel P. Minton
- Synthetic Biology Research Centre, The University of Nottingham Biodiscovery Institute, University Park, Nottingham NG7 2RD, UK; (C.K.); (J.H.); (M.L.K.); (A.C.); (N.P.M.)
| | - Ruth Griffin
- Synthetic Biology Research Centre, The University of Nottingham Biodiscovery Institute, University Park, Nottingham NG7 2RD, UK; (C.K.); (J.H.); (M.L.K.); (A.C.); (N.P.M.)
- Correspondence: ; Tel.: +44-0115-7486120
| |
Collapse
|
17
|
Immunogenicity and Protection from Receptor-Binding Domains of Toxins as Potential Vaccine Candidates for Clostridium difficile. Vaccines (Basel) 2019; 7:vaccines7040180. [PMID: 31717334 PMCID: PMC6963439 DOI: 10.3390/vaccines7040180] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2019] [Revised: 11/04/2019] [Accepted: 11/06/2019] [Indexed: 01/05/2023] Open
Abstract
The receptor-binding domains (RBDs) located in toxin A and toxin B of Clostridium difficile are known to be nontoxic and immunogenic. We need to develop a new type vaccine based on RBDs. In this study, we expressed and purified recombinant proteins (named RBD-TcdA and RBD-TcdB) as vaccine candidates containing the RBDs of toxin A and toxin B, respectively, from the C. difficile reference strain VPI10463. The immunogenicity and protection of the vaccine candidates RBD-TcdA, RBD-TcdB, and RBD-TcdA/B was evaluated by ELISA and survival assays. The data indicated that mice immunized with all vaccine candidates displayed potent levels of RBD-specific serum IgG. Following intramuscular immunization of mice with RBD-TcdA and/or RBD-TcdB, these vaccine candidates triggered immune responses that protected mice compared to mice immunized with aluminum hydroxide alone. Taken together, the results of this study reveal that recombinant proteins containing RBDs of C. difficile toxins can be used for vaccine development. Additionally, we found that an RBD-TcdA/B vaccine can elicit a stronger humoral immune response and provide better immunoprotection than the univalent vaccines. This RBD vaccine candidate conferred significant protection against disease symptoms and death caused by toxins from a wild-type C. difficile strain.
Collapse
|
18
|
Peng Z, Simeon R, Mitchell SB, Zhang J, Feng H, Chen Z. Designed Ankyrin Repeat Protein (DARPin) Neutralizers of TcdB from Clostridium difficile Ribotype 027. mSphere 2019; 4:e00596-19. [PMID: 31578248 PMCID: PMC6796971 DOI: 10.1128/msphere.00596-19] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2019] [Accepted: 09/11/2019] [Indexed: 02/08/2023] Open
Abstract
Clostridium difficile infection (CDI) is a leading cause of hospital-acquired diarrhea. In recent decades, the emergence of the "hypervirulent" BI/NAP1/027 strains of C. difficile significantly increased the morbidity and mortality of CDI. The pathogenesis of CDI is primarily mediated by the action of two toxins, TcdA and TcdB, with TcdB being the major virulent factor in humans. In this report, we describe the engineering of a panel of designed ankyrin repeat proteins (DARPins) that potently neutralize TcdB from the BI/NAP1/027 strains (e.g., TcdBUK1). The most effective DARPin, D16, inhibits TcdBUK1 with a 50% effective concentration (EC50) of 0.5 nM, which is >66-fold lower than that of the FDA-approved anti-TcdB antibody bezlotoxumab (EC50, ∼33 nM). Competitive enzyme-linked immunosorbent assays (ELISAs) showed that D16 blocks interactions between TcdB and its receptor, chondroitin sulfate proteoglycan 4 (CSPG4). The dimeric DARPin U3D16, which pairs D16 with DARPin U3, a disrupter of the interaction of TcdB with Frizzled 1/2/7 receptor, exhibits 10-fold-to-20-fold-enhanced neutralization potency against TcdB from C. difficile strains VPI 10463 (laboratory strain) and M68 (CF/NAP9/017) but identical activity against TcdBUK1 relative to D16. Subsequent ELISAs revealed that TcdBUK1 did not significantly interact with Frizzled 1/2/7. Computation modeling revealed 4 key differences at the Frizzled 1/2/7 binding interface which are likely responsible for the significantly reduced binding affinity.IMPORTANCE We report the engineering and characterization of designed ankyrin proteins as potent neutralizers of TcdB toxin secreted by a hypervirulent ribotype 027 strain of Clostridium difficile We further show that although TcdB toxins from both ribotype 027 and VPI 10461 interact efficiently with TcdB receptors CSPG4 and Pvrl3, TcdB027 lacks significant ability to bind the only known physiologically relevant TcdB receptor, Frizzled 1/2/7.
Collapse
Affiliation(s)
- Zeyu Peng
- Department of Microbial Pathogenesis and Immunology, Texas A&M University Health Science Center, College Station, Texas, USA
| | - Rudo Simeon
- Department of Microbial Pathogenesis and Immunology, Texas A&M University Health Science Center, College Station, Texas, USA
| | - Samuel B Mitchell
- Department of Microbial Pathogenesis and Immunology, Texas A&M University Health Science Center, College Station, Texas, USA
| | - Junjie Zhang
- Department of Biochemistry and Biophysics, Texas A&M University, College Station, Texas, USA
| | - Hanping Feng
- Department of Microbial Pathogenesis, University of Maryland Dental School, Baltimore, Maryland, USA
| | - Zhilei Chen
- Department of Microbial Pathogenesis and Immunology, Texas A&M University Health Science Center, College Station, Texas, USA
| |
Collapse
|
19
|
Giau VV, Lee H, An SSA, Hulme J. Recent advances in the treatment of C. difficile using biotherapeutic agents. Infect Drug Resist 2019; 12:1597-1615. [PMID: 31354309 PMCID: PMC6579870 DOI: 10.2147/idr.s207572] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2019] [Accepted: 05/03/2019] [Indexed: 12/12/2022] Open
Abstract
Clostridium difficile (C. difficile) is rapidly becoming one of the most prevalent health care–associated bacterial infections in the developed world. The emergence of new, more virulent strains has led to greater morbidity and resistance to standard therapies. The bacterium is readily transmitted between people where it can asymptomatically colonize the gut environment, and clinical manifestations ranging from frequent watery diarrhea to toxic megacolon can arise depending on the age of the individual or their state of gut dysbiosis. Several inexpensive approaches are shown to be effective against virulent C. difficile in research settings such as probiotics, fecal microbiota transfer and immunotherapies. This review aims to highlight the current advantages and limitations of the aforementioned approaches with an emphasis on recent studies.
Collapse
Affiliation(s)
- Vo Van Giau
- Department of BioNano Technology, Gachon University, Seongnam-si 461-701, Republic of Korea
| | - Hyon Lee
- Department of Neurology, Gachon University Gil Medical Center, Incheon, South Korea
| | - Seong Soo A An
- Department of BioNano Technology, Gachon University, Seongnam-si 461-701, Republic of Korea
| | - John Hulme
- Department of BioNano Technology, Gachon University, Seongnam-si 461-701, Republic of Korea
| |
Collapse
|
20
|
Cole LE, Li L, Jetley U, Zhang J, Pacheco K, Ma F, Zhang J, Mundle S, Yan Y, Barone L, Rogers C, Beltraminelli N, Quemeneur L, Kleanthous H, Anderson SF, Anosova NG. Deciphering the domain specificity of C. difficile toxin neutralizing antibodies. Vaccine 2019; 37:3892-3901. [PMID: 31122858 DOI: 10.1016/j.vaccine.2019.05.040] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2019] [Revised: 04/10/2019] [Accepted: 05/13/2019] [Indexed: 12/21/2022]
Abstract
Clostridium difficile infection (CDI) is the principal cause of nosocomial diarrhea and pseudomembranous colitis associated with antibiotic therapy. The pathological effects of CDI are primarily attributed to toxins A (TcdA) and B (TcdB). Adequate toxin-specific antibody responses are associated with asymptomatic carriage, whereas insufficient humoral responses are associated with recurrent CDI. While the data supporting the importance of anti-toxin antibodies are substantial, clarity about the toxin domain specificity of these antibodies is more limited. To investigate this matter, combinations of human mAbs targeting multiple domains of TcdB were assessed using toxin neutralization assays. These data revealed that a combination of mAbs specific to all major toxin domains had improved neutralizing potency when compared to equivalent concentrations of a single mAb or a combination of mAbs against one or two domains. The function and toxin domain binding specificity of serum antibodies elicited by immunization of hamsters with a toxoid vaccine candidate was also assessed. Immunization with a toxoid vaccine candidate provoked toxin neutralizing antibodies specific to multiple domains of both TcdA and TcdB. When assessed in a toxin neutralization assay, polyclonal sera displayed greater activity against elevated concentrations of toxins than equivalent concentrations of individual mAbs. These data suggest a potential benefit of any antibody based therapeutic or prophylactic treatment that targets multiple toxin domains.
Collapse
Affiliation(s)
- Leah E Cole
- Sanofi Pasteur, Research North America, 38 Sidney Street, Cambridge, MA 02139, USA
| | - Lu Li
- Sanofi Pasteur, Research North America, 38 Sidney Street, Cambridge, MA 02139, USA
| | - Utsav Jetley
- Sanofi Pasteur, Research North America, 38 Sidney Street, Cambridge, MA 02139, USA; Momenta Pharmaceuticals, INC., Research, 675 W Kendall St, Cambridge, MA 02142, USA
| | - Jinrong Zhang
- Sanofi Pasteur, Research North America, 38 Sidney Street, Cambridge, MA 02139, USA
| | - Kristl Pacheco
- Sanofi Pasteur, Research North America, 38 Sidney Street, Cambridge, MA 02139, USA
| | - Fuqin Ma
- Sanofi Pasteur, Research North America, 38 Sidney Street, Cambridge, MA 02139, USA
| | - Jianxin Zhang
- Sanofi Pasteur, Research North America, 38 Sidney Street, Cambridge, MA 02139, USA
| | - Sophia Mundle
- Sanofi Pasteur, Research North America, 38 Sidney Street, Cambridge, MA 02139, USA
| | - Yanhua Yan
- Sanofi Pasteur, Research North America, 38 Sidney Street, Cambridge, MA 02139, USA
| | - Lucianna Barone
- Sanofi Pasteur, Research North America, 38 Sidney Street, Cambridge, MA 02139, USA; Harvard Vanguard Medical Associates, 230 Worcester Street, Wellesley, MA 02481, USA
| | - Christopher Rogers
- Sanofi Pasteur, Research North America, 38 Sidney Street, Cambridge, MA 02139, USA; Maine Medical Center, Department of Pediatrics, 22 Bramhall Street, Portland, ME 04102, USA
| | - Nicola Beltraminelli
- BliNK Biomedical SAS, R&D, Gerland Plaza Techsud, 70, rue Saint Jean de Dieu, 69007 Lyon, France
| | - Laurence Quemeneur
- Sanofi Pasteur, Research Europe, 1541, Avenue Marcel Mérieux, 68280 Marcy l'Etoile, France
| | - Harry Kleanthous
- Sanofi Pasteur, Research North America, 38 Sidney Street, Cambridge, MA 02139, USA
| | - Stephen F Anderson
- Sanofi Pasteur, Research North America, 38 Sidney Street, Cambridge, MA 02139, USA; Anokion US, Inc., Development and Analytics, 50 Hampshire Street, Cambridge, MA 02139, USA
| | - Natalie G Anosova
- Sanofi Pasteur, Research North America, 38 Sidney Street, Cambridge, MA 02139, USA.
| |
Collapse
|
21
|
Status of vaccine research and development for Clostridium difficile. Vaccine 2019; 37:7300-7306. [PMID: 30902484 DOI: 10.1016/j.vaccine.2019.02.052] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2016] [Accepted: 02/21/2019] [Indexed: 12/15/2022]
Abstract
Clostridium difficile associated disease is fundamentally associated with dysbiosis of the gut microbiome as a consequence of antibiotic use. This is because this sporulating, obligate anaerobe germinates and proliferates rapidly in the dysbiotic gut, which is an indirect consequence of their use. During its growth, C. difficile produces two toxins, toxin A (TcdA) and toxin B (TcdB), which are responsible for the majority of clinical symptoms associated with the disease. Three parenterally delivered vaccines, based on detoxified or recombinant forms of these toxins, have undergone or are undergoing clinical trials. Each offers the opportunity to generate high titres of toxin neutralising antibodies. Whilst these data suggest these vaccines may reduce primary symptomatic disease, they do not in their current form reduce the capacity of the organism to persist and shed from the vaccinated host. The current progress of vaccine development is considered with advantages and limitations of each highlighted. In addition, several alternative approaches are described that seek to limit C. difficile germination, colonisation and persistence. It may yet prove that the most effective treatments to limit infection, disease and spread of the organism will require a combination of therapeutic approaches. The potential use and efficacy of these vaccines in low and middle income countries will be depend on the development of a cost effective vaccine and greater understanding of the distribution and extent of disease in these countries.
Collapse
|
22
|
Tam J, Hamza T, Ma B, Chen K, Beilhartz GL, Ravel J, Feng H, Melnyk RA. Host-targeted niclosamide inhibits C. difficile virulence and prevents disease in mice without disrupting the gut microbiota. Nat Commun 2018; 9:5233. [PMID: 30531960 PMCID: PMC6286312 DOI: 10.1038/s41467-018-07705-w] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2018] [Accepted: 11/19/2018] [Indexed: 12/13/2022] Open
Abstract
Clostridium difficile is the leading cause of nosocomial diarrhea and colitis in the industrialized world. Disruption of the protective gut microbiota by antibiotics enables colonization by multidrug-resistant C. difficile, which secrete up to three different protein toxins that are responsible for the gastrointestinal sequelae. Oral agents that inhibit the damage induced by toxins, without altering the gut microbiota, are urgently needed to prevent primary disease and break the cycle of antibiotic-induced disease recurrence. Here, we show that the anthelmintic drug, niclosamide, inhibits the pathogenesis of all three toxins by targeting a host process required for entry into colonocytes by each toxin. In mice infected with an epidemic strain of C. difficile, expressing all three toxins, niclosamide reduced both primary disease and recurrence, without disrupting the diversity or composition of the gut microbiota. Given its excellent safety profile, niclosamide may address an important unmet need in preventing C. difficile primary and recurrent diseases. Clostridium difficile causes diarrhea and colitis by producing up to three different protein toxins. Here, Tam et al. show that an anthelmintic drug, niclosamide, inhibits the pathogenesis of all three toxins by targeting a host process required for toxin entry into host cells, without disrupting the gut microbiota.
Collapse
Affiliation(s)
- John Tam
- Molecular Medicine, Hospital for Sick Children, 686 Bay St., Toronto, ON, M5G 0A4, Canada.,Department of Biochemistry, University of Toronto, Toronto, ON, M5S 1A8, Canada
| | - Therwa Hamza
- Department of Microbial Pathogenesis, University of Maryland Dental School, Baltimore, MD, 21201, USA
| | - Bing Ma
- Institute for Genome Sciences, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Kevin Chen
- Department of Microbial Pathogenesis, University of Maryland Dental School, Baltimore, MD, 21201, USA
| | - Greg L Beilhartz
- Molecular Medicine, Hospital for Sick Children, 686 Bay St., Toronto, ON, M5G 0A4, Canada
| | - Jacques Ravel
- Institute for Genome Sciences, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Hanping Feng
- Department of Microbial Pathogenesis, University of Maryland Dental School, Baltimore, MD, 21201, USA
| | - Roman A Melnyk
- Molecular Medicine, Hospital for Sick Children, 686 Bay St., Toronto, ON, M5G 0A4, Canada. .,Department of Biochemistry, University of Toronto, Toronto, ON, M5S 1A8, Canada.
| |
Collapse
|
23
|
Fühner V, Heine PA, Helmsing S, Goy S, Heidepriem J, Loeffler FF, Dübel S, Gerhard R, Hust M. Development of Neutralizing and Non-neutralizing Antibodies Targeting Known and Novel Epitopes of TcdB of Clostridioides difficile. Front Microbiol 2018; 9:2908. [PMID: 30574127 PMCID: PMC6291526 DOI: 10.3389/fmicb.2018.02908] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2018] [Accepted: 11/13/2018] [Indexed: 12/18/2022] Open
Abstract
Clostridioides difficile is the causative bacterium in 15-20% of all antibiotic associated diarrheas. The symptoms associated with C. difficile infection (CDI) are primarily induced by the two large exotoxins TcdA and TcdB. Both toxins enter target cells by receptor-mediated endocytosis. Although different toxin receptors have been identified, it is no valid therapeutic option to prevent receptor endocytosis. Therapeutics, such as neutralizing antibodies, directly targeting both toxins are in development. Interestingly, only the anti-TcdB antibody bezlotoxumab but not the anti-TcdA antibody actoxumab prevented recurrence of CDI in clinical trials. In this work, 31 human antibody fragments against TcdB were selected by antibody phage display from the human naive antibody gene libraries HAL9/10. These antibody fragments were further characterized by in vitro neutralization assays. The epitopes of the neutralizing and non-neutralizing antibody fragments were analyzed by domain mapping, TcdB fragment phage display, and peptide arrays, to identify neutralizing and non-neutralizing epitopes. A new neutralizing epitope within the glucosyltransferase domain of TcdB was identified, providing new insights into the relevance of different toxin regions in respect of neutralization and toxicity.
Collapse
Affiliation(s)
- Viola Fühner
- Department Biotechnology, Institute for Biochemistry, Biotechnology and Bioinformatics, Technische Universität Braunschweig, Braunschweig, Germany
| | - Philip Alexander Heine
- Department Biotechnology, Institute for Biochemistry, Biotechnology and Bioinformatics, Technische Universität Braunschweig, Braunschweig, Germany
| | - Saskia Helmsing
- Department Biotechnology, Institute for Biochemistry, Biotechnology and Bioinformatics, Technische Universität Braunschweig, Braunschweig, Germany
| | - Sebastian Goy
- Institute for Toxicology, Hannover Medical School, Hannover, Germany
| | - Jasmin Heidepriem
- Department Synthetic Array Technologies, Max Planck Institute of Colloids and Interfaces, Potsdam, Germany
| | - Felix F. Loeffler
- Department Synthetic Array Technologies, Max Planck Institute of Colloids and Interfaces, Potsdam, Germany
| | - Stefan Dübel
- Department Biotechnology, Institute for Biochemistry, Biotechnology and Bioinformatics, Technische Universität Braunschweig, Braunschweig, Germany
| | - Ralf Gerhard
- Institute for Toxicology, Hannover Medical School, Hannover, Germany
| | - Michael Hust
- Department Biotechnology, Institute for Biochemistry, Biotechnology and Bioinformatics, Technische Universität Braunschweig, Braunschweig, Germany
| |
Collapse
|
24
|
Wang S, Wang Y, Cai Y, Kelly CP, Sun X. Novel Chimeric Protein Vaccines Against Clostridium difficile Infection. Front Immunol 2018; 9:2440. [PMID: 30405630 PMCID: PMC6204379 DOI: 10.3389/fimmu.2018.02440] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2018] [Accepted: 10/02/2018] [Indexed: 12/18/2022] Open
Abstract
Clostridium difficile infection (CDI) is the leading cause of world-wide nosocomial acquired diarrhea in adults. Active vaccination is generally accepted as a logical and cost-effective approach to prevent CDI. In this paper, we have generated two novel chimeric proteins; one designated Tcd169, comprised of the glucosyltransferase domain (GT), the cysteine proteinase domain (CPD), and receptor binding domain (RBD) of TcdB, and the RBD of TcdA; the other designated Tcd169FI, which contains Salmonella typhimurium flagellin (sFliC) and Tcd169. Both proteins were expressed in and purified from Bacillus megaterium. Point mutations were made in the GT (W102A, D288N) and CPD (C698) of TcdB to ensure that Tcd169 and Tcd169FI were atoxic. Immunization with Tcd169 or Tcd169Fl induced protective immunity against TcdA/TcdB challenge through intraperitoneal injection, also provided mice full protection against infection with a hyper-virulent C. difficile strain (BI/NAP1/027). In addition, inclusion of sFlic in the fusion protein (Tcd169Fl) enhanced its protective immunity against toxin challenge, reduced C. difficile numbers in feces from Tcd169Fl-immunized mice infected C. difficile. Our data show that Tcd169 and Tcd169FI fusion proteins may represent alternative vaccine candidates against CDI.
Collapse
Affiliation(s)
- Shaohui Wang
- Department of Molecular Medicine, Morsani College of Medicine, University of South Florida, Tampa, FL, United States
| | - Yuanguo Wang
- Department of Molecular Medicine, Morsani College of Medicine, University of South Florida, Tampa, FL, United States
| | - Ying Cai
- Department of Molecular Medicine, Morsani College of Medicine, University of South Florida, Tampa, FL, United States
| | - Ciaran P. Kelly
- Division of Gastroenterology, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, United States
| | - Xingmin Sun
- Department of Molecular Medicine, Morsani College of Medicine, University of South Florida, Tampa, FL, United States
| |
Collapse
|
25
|
Zhang Y, Li S, Yang Z, Shi L, Yu H, Salerno-Goncalves R, Saint Fleur A, Feng H. Cysteine Protease-Mediated Autocleavage of Clostridium difficile Toxins Regulates Their Proinflammatory Activity. Cell Mol Gastroenterol Hepatol 2018; 5:611-625. [PMID: 29930981 PMCID: PMC6009800 DOI: 10.1016/j.jcmgh.2018.01.022] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/07/2017] [Accepted: 01/30/2018] [Indexed: 02/04/2023]
Abstract
BACKGROUND & AIMS Clostridium difficile toxin A (TcdA) and C difficile toxin toxin B (TcdB), the major virulence factors of the bacterium, cause intestinal tissue damage and inflammation. Although the 2 toxins are homologous and share a similar domain structure, TcdA is generally more inflammatory whereas TcdB is more cytotoxic. The functional domain of the toxins that govern the proinflammatory activities of the 2 toxins is unknown. METHODS Here, we investigated toxin domain functions that regulate the proinflammatory activity of C difficile toxins. By using a mouse ilea loop model, human tissues, and immune cells, we examined the inflammatory responses to a series of chimeric toxins or toxin mutants deficient in specific domain functions. RESULTS Blocking autoprocessing of TcdB by mutagenesis or chemical inhibition, while reducing cytotoxicity of the toxin, significantly enhanced its proinflammatory activities in the animal model. Furthermore, a noncleavable mutant TcdB was significantly more potent than the wild-type toxin in the induction of proinflammatory cytokines in human colonic tissues and immune cells. CONCLUSIONS In this study, we identified a novel mechanism of regulating the biological activities of C difficile toxins in that cysteine protease-mediated autoprocessing regulates toxins' proinflammatory activities. Our findings provide new insight into the pathogenesis of C difficile infection and the design of therapeutics against the disease.
Collapse
Key Words
- 3D, 3-dimensional
- ACPD, CPD domain of TcdA
- Autoprocessing
- Bgt, GTD of TcdB
- Br, RBD of TcdB
- C difficile
- CDI, Clostridium difficile infection
- CPD, cysteine protease domain
- Cysteine Protease
- GT, glucosyltransferase
- GTD, glucosyltransferase domain
- IL, interleukin
- Inflammation
- InsP6, inositol hexakisphosphate
- MPO, myeloperoxidase
- PBMC, peripheral blood mononuclear cell
- PBS, phosphate-buffered saline
- PCR, polymerase chain reaction
- RBD, receptor binding domain
- TER, transepithelial electrical resistance
- TcdA, Clostridium difficile toxin A
- TcdB, Clostridium difficile toxin B
- Toxins
- aTcdA, GTD deficient TcdA
Collapse
Affiliation(s)
- Yongrong Zhang
- Department of Microbial Pathogenesis, School of Dentistry, University of Maryland, Baltimore, Maryland
| | - Shan Li
- Department of Microbial Pathogenesis, School of Dentistry, University of Maryland, Baltimore, Maryland
| | - Zhiyong Yang
- Department of Microbial Pathogenesis, School of Dentistry, University of Maryland, Baltimore, Maryland
| | - Lianfa Shi
- Department of Microbial Pathogenesis, School of Dentistry, University of Maryland, Baltimore, Maryland
| | - Hua Yu
- Department of Microbial Pathogenesis, School of Dentistry, University of Maryland, Baltimore, Maryland
| | - Rosangela Salerno-Goncalves
- Department of Pediatrics and Center for Vaccine Development, School of Medicine, University of Maryland, Baltimore, Maryland
| | - Ashley Saint Fleur
- Department of Microbial Pathogenesis, School of Dentistry, University of Maryland, Baltimore, Maryland
| | - Hanping Feng
- Department of Microbial Pathogenesis, School of Dentistry, University of Maryland, Baltimore, Maryland,Correspondence Address correspondence to: Hanping Feng, PhD, 650 W Baltimore Street, Room 7211, Baltimore, Maryland 21201. fax: (410) 706-6511.
| |
Collapse
|
26
|
Roshan N, Hammer KA, Riley TV. Non-conventional antimicrobial and alternative therapies for the treatment of Clostridium difficile infection. Anaerobe 2018; 49:103-111. [DOI: 10.1016/j.anaerobe.2018.01.003] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2017] [Revised: 12/19/2017] [Accepted: 01/05/2018] [Indexed: 02/08/2023]
|
27
|
Immunization Strategies Against Clostridium difficile. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2018; 1050:197-225. [PMID: 29383671 DOI: 10.1007/978-3-319-72799-8_12] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
C. difficile infection (CDI) is an important healthcare- but also community-associated disease. CDI is considered a public health threat and an economic burden. A major problem is the high rate of recurrences. Besides classical antibiotic treatments, new therapeutic strategies are needed to prevent infection, to treat patients and prevent recurrences. If fecal transplantation has been recommended to treat recurrences, another key approach is to restore immunity against C. difficile and its virulence factors. Here, after a summary concerning the virulence factors, the host immune response against C. difficile and its role in the outcome of disease, we review the different approaches of passive immunotherapies and vaccines developed against CDI. Passive immunization strategies are designed in function of the target antigen, the antibody-based product and its administration route. Similarly, for active immunization strategies, vaccine antigens can target toxins or surface proteins and immunization can be performed by parenteral or mucosal routes. For passive immunization and vaccination as well, we first present immunization assays performed in animal models and second in humans and associated clinical trials. The different studies are presented according to the mode of administration either parenteral or mucosal and the target antigens, either toxins or colonization factors.
Collapse
|
28
|
Kroh HK, Chandrasekaran R, Zhang Z, Rosenthal K, Woods R, Jin X, Nyborg AC, Rainey GJ, Warrener P, Melnyk RA, Spiller BW, Lacy DB. A neutralizing antibody that blocks delivery of the enzymatic cargo of Clostridium difficile toxin TcdB into host cells. J Biol Chem 2017; 293:941-952. [PMID: 29180448 DOI: 10.1074/jbc.m117.813428] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2017] [Revised: 11/08/2017] [Indexed: 12/14/2022] Open
Abstract
Clostridium difficile infection is the leading cause of hospital-acquired diarrhea and is mediated by the actions of two toxins, TcdA and TcdB. The toxins perturb host cell function through a multistep process of receptor binding, endocytosis, low pH-induced pore formation, and the translocation and delivery of an N-terminal glucosyltransferase domain that inactivates host GTPases. Infection studies with isogenic strains having defined toxin deletions have established TcdB as an important target for therapeutic development. Monoclonal antibodies that neutralize TcdB function have been shown to protect against C. difficile infection in animal models and reduce recurrence in humans. Here, we report the mechanism of TcdB neutralization by PA41, a humanized monoclonal antibody capable of neutralizing TcdB from a diverse array of C. difficile strains. Through a combination of structural, biochemical, and cell functional studies, involving X-ray crystallography and EM, we show that PA41 recognizes a single, highly conserved epitope on the TcdB glucosyltransferase domain and blocks productive translocation and delivery of the enzymatic cargo into the host cell. Our study reveals a unique mechanism of C. difficile toxin neutralization by a monoclonal antibody, which involves targeting a process that is conserved across the large clostridial glucosylating toxins. The PA41 antibody described here provides a valuable tool for dissecting the mechanism of toxin pore formation and translocation across the endosomal membrane.
Collapse
Affiliation(s)
- Heather K Kroh
- From the Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee 37232-2363
| | - Ramyavardhanee Chandrasekaran
- From the Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee 37232-2363
| | - Zhifen Zhang
- Department of Biochemistry, University of Toronto, Toronto, Ontario M5S 1A8, Canada.,Molecular Structure and Function, The Hospital for Sick Children, Toronto, Ontario M5G 0A4, Canada
| | | | - Rob Woods
- MedImmune LLC, Gaithersburg, Maryland 20878-2204
| | - Xiaofang Jin
- MedImmune LLC, Gaithersburg, Maryland 20878-2204
| | | | | | | | - Roman A Melnyk
- Department of Biochemistry, University of Toronto, Toronto, Ontario M5S 1A8, Canada.,Molecular Structure and Function, The Hospital for Sick Children, Toronto, Ontario M5G 0A4, Canada
| | - Benjamin W Spiller
- From the Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee 37232-2363.,Department of Pharmacology, Vanderbilt University, Nashville, Tennessee 37232-6600, and
| | - D Borden Lacy
- From the Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee 37232-2363, .,Veterans Affairs Tennessee Valley Healthcare System, Nashville, Tennessee 37212-2637
| |
Collapse
|
29
|
Cytokines Are Markers of the Clostridium difficile-Induced Inflammatory Response and Predict Disease Severity. CLINICAL AND VACCINE IMMUNOLOGY : CVI 2017; 24:CVI.00037-17. [PMID: 28592627 DOI: 10.1128/cvi.00037-17] [Citation(s) in RCA: 87] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/07/2017] [Accepted: 05/24/2017] [Indexed: 12/25/2022]
Abstract
The host immune response affects pathogen virulence in Clostridium difficile infection (CDI). Thus, cytokine responses to CDI likely are associated with disease initiation and progression. Understanding the molecular drivers of inflammation and biochemical markers of disease severity is important for developing novel therapies and predicting disease prognosis. In this study, we investigated cytokine production in patients with CDI and evaluated the potential of cytokines to serve as biomarkers for CDI and predictors of disease severity. The systemic cytokine profiles of 36 CDI patients (20 with severe disease) and 8 healthy donors and the toxin-induced cytokine profiles of peripheral blood mononuclear cells (PBMC) were determined. Further, we evaluated glucosyltransferase (GT) activity in regulation of toxin-induced cytokine expression. We found upregulation of the majority of measured cytokines (11/20, 55%) in CDI patients. Interleukin-1β (IL-1β), IL-6, IL-8, IL-17A, and IL-16 were the most upregulated. High serum levels of IL-2 and IL-15 were associated with a poor prognosis in CDI patients, whereas high levels of IL-5 and gamma interferon (IFN-γ) were associated with less severe disease. Both TcdA and TcdB were potent inducers of cytokine responses, as demonstrated by stimulation of a greater number and amount of cytokines. In addition to confirming prior reports on the role of IL-8, IL-1β, and IL-6 in CDI, our data suggest that IL-16 and IL-17A, as well as the IL-1β/Th17 axis, play a key role in driving inflammatory responses in CDI. A functional GT domain of C. difficile toxins was required for the induction of a majority of cytokines investigated.
Collapse
|
30
|
Liu YW, Chen YH, Chen JW, Tsai PJ, Huang IH. Immunization with Recombinant TcdB-Encapsulated Nanocomplex Induces Protection against Clostridium difficile Challenge in a Mouse Model. Front Microbiol 2017; 8:1411. [PMID: 28790999 PMCID: PMC5525027 DOI: 10.3389/fmicb.2017.01411] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2017] [Accepted: 07/11/2017] [Indexed: 12/13/2022] Open
Abstract
Clostridium difficile is considered to be one of the major cause of infectious diarrhea in healthcare systems worldwide. Symptoms of C. difficile infection are caused largely by the production of two cytotoxins: toxin A (TcdA) and toxin B (TcdB). Vaccine development is considered desirable as it would decrease the mounting medical costs and mortality associated with C. difficile infections. Biodegradable nanoparticles composed of poly-γ-glutamic acid (γ-PGA) and chitosan have proven to be a safe and effective antigen delivery system for many viral vaccines. However, few studies have used this efficient antigen carrier for bacterial vaccine development. In this study, we eliminated the toxin activity domain of toxin B by constructing a recombinant protein rTcdB consists of residues 1852-2363 of TcdB receptor binding domain. The rTcdB was encapsulated in nanoparticles composed of γ-PGA and chitosan. Three rounds of intraperitoneal vaccination led to high anti-TcdB antibody responses and afforded mice full protection mice from lethal dose of C. difficile spore challenge. Protection was associated with high levels of toxin-neutralizing antibodies, and the rTcdB-encapsulated NPs elicited a longer-lasting antibody titers than antigen with the conventional adjuvant, aluminum hydroxide. Significant reductions in the level of proinflammatory cytokines and chemokines were observed in vaccinated mouse. These results suggested that polymeric nanocomplex-based vaccine design can be useful in developing vaccine against C. difficile infections.
Collapse
Affiliation(s)
- Yi-Wen Liu
- Department of Microbiology and Immunology, College of Medicine, National Cheng Kung UniversityTainan, Taiwan
| | - Yu-Hung Chen
- Department of Biochemistry and Molecular Biology, College of Medicine, National Cheng Kung UniversityTainan, Taiwan
| | - Jenn-Wei Chen
- Department of Microbiology and Immunology, College of Medicine, National Cheng Kung UniversityTainan, Taiwan.,Center of Infectious Disease and Signaling Research, National Cheng Kung UniversityTainan, Taiwan
| | - Pei-Jane Tsai
- Center of Infectious Disease and Signaling Research, National Cheng Kung UniversityTainan, Taiwan.,Department of Medical Laboratory Science and Biotechnology, College of Medicine, National Cheng Kung UniversityTainan, Taiwan
| | - I-Hsiu Huang
- Department of Microbiology and Immunology, College of Medicine, National Cheng Kung UniversityTainan, Taiwan.,Center of Infectious Disease and Signaling Research, National Cheng Kung UniversityTainan, Taiwan
| |
Collapse
|
31
|
Péchiné S, Janoir C, Collignon A. Emerging monoclonal antibodies against Clostridium difficile infection. Expert Opin Biol Ther 2017; 17:415-427. [PMID: 28274145 DOI: 10.1080/14712598.2017.1300655] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
INTRODUCTION Clostridium difficile infections are characterized by a high recurrence rate despite antibiotic treatments and there is an urgent need to develop new treatments such as fecal transplantation and immonotherapy. Besides active immunotherapy with vaccines, passive immunotherapy has shown promise, especially with monoclonal antibodies. Areas covered: Herein, the authors review the different assays performed with monoclonal antibodies against C. difficile toxins and surface proteins to treat or prevent primary or recurrent episodes of C. difficile infection in animal models and in clinical trials as well. Notably, the authors lay emphasis on the phase III clinical trial (MODIFY II), which allowed bezlotoxumab to be approved by the Food and Drug Administration and the European Medicines Agency. They also review new strategies for producing single domain antibodies and nanobodies against C. difficile and new approaches to deliver them in the digestive tract. Expert opinion: Only two human Mabs against TcdA and TcdB have been tested alone or in combination in clinical trials. However, many animal model studies have provided rationale for the use of Mabs and nanobodies in C. difficile infection and pave the way for further clinical investigation.
Collapse
Affiliation(s)
- Séverine Péchiné
- a EA4043 Faculté de Pharmacie , Univ Paris-Sud, Université Paris-Saclay , Chatenay-Malabry , France
| | - Claire Janoir
- a EA4043 Faculté de Pharmacie , Univ Paris-Sud, Université Paris-Saclay , Chatenay-Malabry , France
| | - Anne Collignon
- a EA4043 Faculté de Pharmacie , Univ Paris-Sud, Université Paris-Saclay , Chatenay-Malabry , France
| |
Collapse
|
32
|
Secore S, Wang S, Doughtry J, Xie J, Miezeiewski M, Rustandi RR, Horton M, Xoconostle R, Wang B, Lancaster C, Kristopeit A, Wang SC, Christanti S, Vitelli S, Gentile MP, Goerke A, Skinner J, Strable E, Thiriot DS, Bodmer JL, Heinrichs JH. Development of a Novel Vaccine Containing Binary Toxin for the Prevention of Clostridium difficile Disease with Enhanced Efficacy against NAP1 Strains. PLoS One 2017; 12:e0170640. [PMID: 28125650 PMCID: PMC5268477 DOI: 10.1371/journal.pone.0170640] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2016] [Accepted: 01/06/2017] [Indexed: 12/18/2022] Open
Abstract
Clostridium difficile infections (CDI) are a leading cause of nosocomial diarrhea in the developed world. The main virulence factors of the bacterium are the large clostridial toxins (LCTs), TcdA and TcdB, which are largely responsible for the symptoms of the disease. Recent outbreaks of CDI have been associated with the emergence of hypervirulent strains, such as NAP1/BI/027, many strains of which also produce a third toxin, binary toxin (CDTa and CDTb). These hypervirulent strains have been associated with increased morbidity and higher mortality. Here we present pre-clinical data describing a novel tetravalent vaccine composed of attenuated forms of TcdA, TcdB and binary toxin components CDTa and CDTb. We demonstrate, using the Syrian golden hamster model of CDI, that the inclusion of binary toxin components CDTa and CDTb significantly improves the efficacy of the vaccine against challenge with NAP1 strains in comparison to vaccines containing only TcdA and TcdB antigens, while providing comparable efficacy against challenge with the prototypic, non-epidemic strain VPI10463. This combination vaccine elicits high neutralizing antibody titers against TcdA, TcdB and binary toxin in both hamsters and rhesus macaques. Finally we present data that binary toxin alone can act as a virulence factor in animal models. Taken together, these data strongly support the inclusion of binary toxin in a vaccine against CDI to provide enhanced protection from epidemic strains of C. difficile.
Collapse
Affiliation(s)
- Susan Secore
- Vaccine Basic Research, Merck Research Laboratories, Merck and Company, Incorporated, West Point, Pennsylvania, United States of America
| | - Su Wang
- Vaccine Basic Research, Merck Research Laboratories, Merck and Company, Incorporated, West Point, Pennsylvania, United States of America
| | - Julie Doughtry
- Vaccine Basic Research, Merck Research Laboratories, Merck and Company, Incorporated, West Point, Pennsylvania, United States of America
| | - Jinfu Xie
- Vaccine Basic Research, Merck Research Laboratories, Merck and Company, Incorporated, West Point, Pennsylvania, United States of America
| | - Matt Miezeiewski
- Eurofins Laboratories, Lancaster, Pennsylvania, United States of America
| | - Richard R. Rustandi
- Vaccine Analytical Development, Merck Research Laboratories, Merck and Company, Incorporated, West Point, Pennsylvania, United States of America
| | - Melanie Horton
- Vaccine Basic Research, Merck Research Laboratories, Merck and Company, Incorporated, West Point, Pennsylvania, United States of America
| | - Rachel Xoconostle
- Vaccine Basic Research, Merck Research Laboratories, Merck and Company, Incorporated, West Point, Pennsylvania, United States of America
| | - Bei Wang
- Vaccine Drug Product Development, Merck Research Laboratories, Merck and Company, Incorporated, West Point, Pennsylvania, United States of America
| | - Catherine Lancaster
- Vaccine Analytical Development, Merck Research Laboratories, Merck and Company, Incorporated, West Point, Pennsylvania, United States of America
| | - Adam Kristopeit
- Vaccine Process Development, Merck Research Laboratories, Merck and Company, Incorporated, West Point, Pennsylvania, United States of America
| | - Sheng-Ching Wang
- Vaccine Process Development, Merck Research Laboratories, Merck and Company, Incorporated, West Point, Pennsylvania, United States of America
| | - Sianny Christanti
- Vaccine Process Development, Merck Research Laboratories, Merck and Company, Incorporated, West Point, Pennsylvania, United States of America
| | - Salvatore Vitelli
- Vaccine Analytical Development, Merck Research Laboratories, Merck and Company, Incorporated, West Point, Pennsylvania, United States of America
| | - Marie-Pierre Gentile
- Vaccine Process Development, Merck Research Laboratories, Merck and Company, Incorporated, West Point, Pennsylvania, United States of America
| | - Aaron Goerke
- Vaccine Process Development, Merck Research Laboratories, Merck and Company, Incorporated, West Point, Pennsylvania, United States of America
| | - Julie Skinner
- Vaccine Basic Research, Merck Research Laboratories, Merck and Company, Incorporated, West Point, Pennsylvania, United States of America
| | - Erica Strable
- Vaccine Drug Product Development, Merck Research Laboratories, Merck and Company, Incorporated, West Point, Pennsylvania, United States of America
| | - David S. Thiriot
- Vaccine Drug Product Development, Merck Research Laboratories, Merck and Company, Incorporated, West Point, Pennsylvania, United States of America
| | - Jean-Luc Bodmer
- Vaccine Basic Research, Merck Research Laboratories, Merck and Company, Incorporated, West Point, Pennsylvania, United States of America
| | - Jon H. Heinrichs
- Vaccine Basic Research, Merck Research Laboratories, Merck and Company, Incorporated, West Point, Pennsylvania, United States of America
| |
Collapse
|
33
|
Stinson JA, Raja WK, Lee S, Kim HB, Diwan I, Tutunjian S, Panilaitis B, Omenetto FG, Tzipori S, Kaplan DL. Silk Fibroin Microneedles for Transdermal Vaccine Delivery. ACS Biomater Sci Eng 2017; 3:360-369. [PMID: 33465933 DOI: 10.1021/acsbiomaterials.6b00515] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Microneedles represent an exciting departure from the existing parenteral injection paradigm for drug delivery, particularly for the administration of vaccines. While the benefit of delivering vaccine antigens to immunocompetent tissue in the skin has been established, there have been varying degrees of success using microneedles to do so. Here, we investigate the use of silk fibroin protein to produce microneedles and evaluate their ability to deliver vaccines against influenza, Clostridium difficile, and Shigella. Fibroin protein from the silkworm Bombyx mori possesses suitable properties for use in a microneedle system, including all-aqueous processing, mechanical strength in dried formats, biocompatibility, and the ability to temperature stabilize biomacromolecules. As such, this biomaterial combines the processing and biocompatibility advantages of a dissolving microneedle system with the product stability and mechanical strength of coated insoluble microneedles. Through successful vaccination of mice against three separate antigens, we establish that silk fibroin is well-suited for use as a solid-coated microneedle delivery system and offers long-term potential similar to that of the leading microneedle biomaterials.
Collapse
Affiliation(s)
- Jordan A Stinson
- Department of Biomedical Engineering, 4 Colby Street, Tufts University, Medford, Massachusetts 02155, United States
| | - Waseem K Raja
- Department of Biomedical Engineering, 4 Colby Street, Tufts University, Medford, Massachusetts 02155, United States.,Department of Brain and Cognitive Sciences, 43 Vassar Street, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, United States
| | - Sangun Lee
- Department of Infectious Disease and Global Health, 200 Westboro Road, Tufts University Cummings School of Veterinary Medicine, North Grafton, Massachusetts 01536, United States
| | - Hyeun Bum Kim
- Department of Animal Resources Science, Dankook University, Cheonan 31116, South Korea
| | - Izzuddin Diwan
- Department of Biomedical Engineering, 4 Colby Street, Tufts University, Medford, Massachusetts 02155, United States
| | - Stephen Tutunjian
- Department of Biomedical Engineering, 4 Colby Street, Tufts University, Medford, Massachusetts 02155, United States
| | - Bruce Panilaitis
- Department of Biomedical Engineering, 4 Colby Street, Tufts University, Medford, Massachusetts 02155, United States
| | - Fiorenzo G Omenetto
- Department of Biomedical Engineering, 4 Colby Street, Tufts University, Medford, Massachusetts 02155, United States
| | - Saul Tzipori
- Department of Infectious Disease and Global Health, 200 Westboro Road, Tufts University Cummings School of Veterinary Medicine, North Grafton, Massachusetts 01536, United States
| | - David L Kaplan
- Department of Biomedical Engineering, 4 Colby Street, Tufts University, Medford, Massachusetts 02155, United States
| |
Collapse
|
34
|
Mehta KK, Paskaleva EE, Wu X, Grover N, Mundra RV, Chen K, Zhang Y, Yang Z, Feng H, Dordick JS, Kane RS. Newly identified bacteriolytic enzymes that target a wide range of clinical isolates of Clostridium difficile. Biotechnol Bioeng 2016; 113:2568-2576. [PMID: 27260850 PMCID: PMC5367918 DOI: 10.1002/bit.26029] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2016] [Revised: 05/23/2016] [Accepted: 05/29/2016] [Indexed: 12/18/2022]
Abstract
Clostridium difficile has emerged as a major cause of infectious diarrhea in hospitalized patients, with increasing mortality rate and annual healthcare costs exceeding $3 billion. Since C. difficile infections are associated with the use of antibiotics, there is an urgent need to develop treatments that can inactivate the bacterium selectively without affecting commensal microflora. Lytic enzymes from bacteria and bacteriophages show promise as highly selective and effective antimicrobial agents. These enzymes often have a modular structure, consisting of a catalytic domain and a binding domain. In the current work, using consensus catalytic domain and cell-wall binding domain sequences as probes, we analyzed in silico the genome of C. difficile, as well as phages infecting C. difficile. We identified two genes encoding cell lytic enzymes with possible activity against C. difficile. We cloned the genes in a suitable expression vector, expressed and purified the protein products, and tested enzyme activity in vitro. These newly identified enzymes were found to be active against C. difficile cells in a dose-dependent manner. We achieved a more than 4-log reduction in the number of viable bacteria within 5 h of application. Moreover, we found that the enzymes were active against a wide range of C. difficile clinical isolates. We also characterized the biocatalytic mechanism by identifying the specific bonds cleaved by these enzymes within the cell wall peptidoglycan. These results suggest a new approach to combating the growing healthcare problem associated with C. difficile infections. Biotechnol. Bioeng. 2016;113: 2568-2576. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Krunal K Mehta
- Howard P. Isermann Department of Chemical and Biological Engineering, Rensselaer Polytechnic Institute, Troy, New York
- Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, 110 8th Street, Troy, 12180, New York
| | - Elena E Paskaleva
- Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, 110 8th Street, Troy, 12180, New York
| | - Xia Wu
- Howard P. Isermann Department of Chemical and Biological Engineering, Rensselaer Polytechnic Institute, Troy, New York
- Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, 110 8th Street, Troy, 12180, New York
| | - Navdeep Grover
- Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, 110 8th Street, Troy, 12180, New York
| | - Ruchir V Mundra
- Howard P. Isermann Department of Chemical and Biological Engineering, Rensselaer Polytechnic Institute, Troy, New York
- Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, 110 8th Street, Troy, 12180, New York
| | - Kevin Chen
- Department of Microbial Pathogenesis, University of Maryland Dental School, Baltimore, Maryland
| | - Yongrong Zhang
- Department of Microbial Pathogenesis, University of Maryland Dental School, Baltimore, Maryland
| | - Zhiyong Yang
- Department of Microbial Pathogenesis, University of Maryland Dental School, Baltimore, Maryland
| | - Hanping Feng
- Department of Microbial Pathogenesis, University of Maryland Dental School, Baltimore, Maryland
| | - Jonathan S Dordick
- Howard P. Isermann Department of Chemical and Biological Engineering, Rensselaer Polytechnic Institute, Troy, New York.
- Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, 110 8th Street, Troy, 12180, New York.
| | - Ravi S Kane
- School of Chemical and Biomolecular Engineering, Georgia Institute of Technology, 950 Atlantic Drive, Atlanta, 30332, Georgia.
| |
Collapse
|
35
|
Hebbard AIT, Slavin MA, Reed C, Teh BW, Thursky KA, Trubiano JA, Worth LJ. The epidemiology of Clostridium difficile infection in patients with cancer. Expert Rev Anti Infect Ther 2016; 14:1077-1085. [PMID: 27606976 DOI: 10.1080/14787210.2016.1234376] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
INTRODUCTION Clostridium difficile infection (CDI) is a significant cause of healthcare-associated diarrhoea, and the emergence of endemic strains resulting in poorer outcomes is recognised worldwide. Patients with cancer are a specific high-risk group for development of infection. Areas covered: In this review, modifiable and non-modifiable risk factors for CDI in adult patients with haematological malignancy or solid tumours are evaluated. In particular, the contribution of antimicrobial exposure, hospitalisation and gastric acid suppression to risk of CDI are discussed. Recent advances in CDI treatment are outlined, namely faecal microbiota transplantation and fidaxomicin therapy for severe/refractory infection in cancer populations. Outcomes of CDI, including mortality are presented, together with the need for valid severity rating tools customised for cancer populations. Expert commentary: Future areas for research include the prognostic value of C. difficile colonisation in cancer patients and the potential impact of dedicated antimicrobial stewardship programs in reducing the burden of CDI in cancer units.
Collapse
Affiliation(s)
- Andrew I T Hebbard
- a Department of Infectious Diseases and Infection Prevention , Peter MacCallum Cancer Centre , Melbourne , Australia
| | - Monica A Slavin
- a Department of Infectious Diseases and Infection Prevention , Peter MacCallum Cancer Centre , Melbourne , Australia.,b Department of Medicine , University of Melbourne , Melbourne , Australia
| | - Caroline Reed
- c Microbiology Department , Peter MacCallum Cancer Centre , Melbourne , Australia
| | - Benjamin W Teh
- a Department of Infectious Diseases and Infection Prevention , Peter MacCallum Cancer Centre , Melbourne , Australia
| | - Karin A Thursky
- a Department of Infectious Diseases and Infection Prevention , Peter MacCallum Cancer Centre , Melbourne , Australia
| | - Jason A Trubiano
- a Department of Infectious Diseases and Infection Prevention , Peter MacCallum Cancer Centre , Melbourne , Australia
| | - Leon J Worth
- a Department of Infectious Diseases and Infection Prevention , Peter MacCallum Cancer Centre , Melbourne , Australia.,b Department of Medicine , University of Melbourne , Melbourne , Australia.,d Victorian Healthcare Associated Infection Surveillance System (VICNISS) , Doherty Institute , Melbourne , Australia
| |
Collapse
|
36
|
Yang Z, Shi L, Yu H, Zhang Y, Chen K, Saint Fleur A, Bai G, Feng H. Intravenous adenovirus expressing a multi-specific, single-domain antibody neutralizing TcdA and TcdB protects mice from Clostridium difficile infection. Pathog Dis 2016; 74:ftw078. [PMID: 27502696 DOI: 10.1093/femspd/ftw078] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/02/2016] [Indexed: 01/09/2023] Open
Abstract
Clostridium difficile infection (CDI) is the most common cause of antibiotic-associated diarrhea and colitis in developed countries. The disease is mainly mediated via two major exotoxins TcdA and TcdB secreted by the bacterium. We have previously developed a novel, potently neutralizing, tetravalent and bispecific heavy-chain-only single domain (VHH) antibody to both TcdA and TcdB (designated as ABA) that reverses fulminant CDI in mice. Since ABA has a short serum half-life, in this study a replication-deficient recombinant adenovirus expressing ABA was generated and the long-lasting expression of functional ABA was demonstrated in vitro and in vivo Mice transduced with one dose of the adenovirus displayed high levels of serum ABA for more than1 month and were fully protected against systemic toxin challenges. More importantly, the ABA delivered by the adenovirus protected mice from both primary and recurrent CDI. Thus, replication-deficient adenoviral vector may be used to deliver neutralizing antibodies against the toxins in order to prevent CDI and recurrence.
Collapse
Affiliation(s)
- Zhiyong Yang
- Department of Microbial Pathogenesis, University of Maryland School of Dentistry, Baltimore, MD 21201, USA
| | - Lianfa Shi
- Department of Microbial Pathogenesis, University of Maryland School of Dentistry, Baltimore, MD 21201, USA
| | - Hua Yu
- Department of Microbial Pathogenesis, University of Maryland School of Dentistry, Baltimore, MD 21201, USA
| | - Yongrong Zhang
- Department of Microbial Pathogenesis, University of Maryland School of Dentistry, Baltimore, MD 21201, USA
| | - Kevin Chen
- Department of Microbial Pathogenesis, University of Maryland School of Dentistry, Baltimore, MD 21201, USA
| | - Ashley Saint Fleur
- Department of Microbial Pathogenesis, University of Maryland School of Dentistry, Baltimore, MD 21201, USA
| | - Guang Bai
- Department of Neural and Pain Sciences, University of Maryland School of Dentistry, Baltimore, MD 21201, USA
| | - Hanping Feng
- Department of Microbial Pathogenesis, University of Maryland School of Dentistry, Baltimore, MD 21201, USA
| |
Collapse
|
37
|
Maldarelli GA, Matz H, Gao S, Chen K, Hamza T, Yfantis HG, Feng H, Donnenberg MS. Pilin Vaccination Stimulates Weak Antibody Responses and Provides No Protection in a C57Bl/6 Murine Model of Acute Clostridium difficile Infection. JOURNAL OF VACCINES & VACCINATION 2016; 7:321. [PMID: 27375958 PMCID: PMC4927082 DOI: 10.4172/2157-7560.1000321] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Clostridium difficile is the leading cause of nosocomial infections in the United States, adding billions of dollars per year to health care costs. A vaccine targeted against the bacterium would be extremely beneficial in decreasing the morbidity and mortality caused by C. difficile-associated disease; a vaccine directed against a colonization factor would hinder the spread of the bacterium as well as prevent disease. Type IV pili (T4Ps) are extracellular appendages composed of protein monomers called pilins. They are involved in adhesion and colonization in a wide variety of bacteria and archaea, and are putative colonization factors in C. difficile. We hypothesized that vaccinating mice with pilins would lead to generation of anti-pilin antibodies, and would protect against C. difficile challenge. We found that immunizing C57Bl/6 mice with various pilins, whether combined or as individual proteins, led to low anti-pilin antibody titers and no protection upon C. difficile challenge. Passive transfer of anti-pilin antibodies led to high serum anti-pilin IgG titers, but to undetectable fecal anti-pilin IgG titers and did not protect against challenge. The low antibody titers observed in these experiments may be due to the particular strain of mice used. Further experiments, possibly with a different animal model of C. difficile infection, are needed to determine if an anti-T4P vaccine would be protective against C. difficile infection.
Collapse
Affiliation(s)
- Grace A Maldarelli
- Department of Medicine, Division of Infectious Disease, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Hanover Matz
- Department of Medicine, Division of Infectious Disease, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Si Gao
- Department of Microbial Pathogenesis, University of Maryland Dental School, Baltimore, Maryland, USA
| | - Kevin Chen
- Department of Microbial Pathogenesis, University of Maryland Dental School, Baltimore, Maryland, USA
| | - Therwa Hamza
- Department of Microbial Pathogenesis, University of Maryland Dental School, Baltimore, Maryland, USA
| | - Harris G Yfantis
- Department of Pathology and Laboratory Medicine, VAMHCS, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Hanping Feng
- Department of Microbial Pathogenesis, University of Maryland Dental School, Baltimore, Maryland, USA
| | - Michael S Donnenberg
- Department of Medicine, Division of Infectious Disease, University of Maryland School of Medicine, Baltimore, Maryland, USA
| |
Collapse
|
38
|
Di Bella S, Ascenzi P, Siarakas S, Petrosillo N, di Masi A. Clostridium difficile Toxins A and B: Insights into Pathogenic Properties and Extraintestinal Effects. Toxins (Basel) 2016; 8:134. [PMID: 27153087 PMCID: PMC4885049 DOI: 10.3390/toxins8050134] [Citation(s) in RCA: 176] [Impact Index Per Article: 19.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2016] [Revised: 04/22/2016] [Accepted: 04/25/2016] [Indexed: 02/06/2023] Open
Abstract
Clostridium difficile infection (CDI) has significant clinical impact especially on the elderly and/or immunocompromised patients. The pathogenicity of Clostridium difficile is mainly mediated by two exotoxins: toxin A (TcdA) and toxin B (TcdB). These toxins primarily disrupt the cytoskeletal structure and the tight junctions of target cells causing cell rounding and ultimately cell death. Detectable C. difficile toxemia is strongly associated with fulminant disease. However, besides the well-known intestinal damage, recent animal and in vitro studies have suggested a more far-reaching role for these toxins activity including cardiac, renal, and neurologic impairment. The creation of C. difficile strains with mutations in the genes encoding toxin A and B indicate that toxin B plays a major role in overall CDI pathogenesis. Novel insights, such as the role of a regulator protein (TcdE) on toxin production and binding interactions between albumin and C. difficile toxins, have recently been discovered and will be described. Our review focuses on the toxin-mediated pathogenic processes of CDI with an emphasis on recent studies.
Collapse
Affiliation(s)
- Stefano Di Bella
- 2nd Infectious Diseases Division, National Institute for Infectious Diseases "L. Spallanzani", Rome 00149, Italy.
| | - Paolo Ascenzi
- Department of Science, Roma Tre University, Rome 00154, Italy.
| | - Steven Siarakas
- Department of Microbiology and Infectious Diseases, Concord Repatriation General Hospital, Sydney 2139, Australia.
| | - Nicola Petrosillo
- 2nd Infectious Diseases Division, National Institute for Infectious Diseases "L. Spallanzani", Rome 00149, Italy.
| | | |
Collapse
|
39
|
Zhang Y, Feng H. Pathogenic effects of glucosyltransferase from Clostridium difficile toxins. Pathog Dis 2016; 74:ftw024. [PMID: 27044305 DOI: 10.1093/femspd/ftw024] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/30/2016] [Indexed: 01/13/2023] Open
Abstract
The glucosyltransferase domain ofClostridium difficiletoxins modifies guanine nucleotide-binding proteins of Rho family. It is the major virulent domain of the holotoxins. Various pathogenic effects ofC. difficiletoxins in response to Rho glucosylation have been investigated including cytoskeleton damage, cell death and inflammation. The most recent studies have revealed some significant characteristics of the holotoxins that are independent of glucosylating activity. These findings arouse discussion about the role of glucosyltransferase activity in toxin pathogenesis and open up new insights for toxin mechanism study. In this review, we summarize the pathogenic effects of glucosyltransferase domain of the toxins in the past years.
Collapse
Affiliation(s)
- Yongrong Zhang
- Department of Microbial Pathogenesis, University of Maryland Baltimore, 650 W. Baltimore Street, Baltimore, MD 21201, USA
| | - Hanping Feng
- Department of Microbial Pathogenesis, University of Maryland Baltimore, 650 W. Baltimore Street, Baltimore, MD 21201, USA
| |
Collapse
|
40
|
Chen S, Sun C, Wang H, Wang J. The Role of Rho GTPases in Toxicity of Clostridium difficile Toxins. Toxins (Basel) 2015; 7:5254-67. [PMID: 26633511 PMCID: PMC4690124 DOI: 10.3390/toxins7124874] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2015] [Revised: 11/18/2015] [Accepted: 11/18/2015] [Indexed: 12/18/2022] Open
Abstract
Clostridium difficile (C. difficile) is the main cause of antibiotic-associated diarrhea prevailing in hospital settings. In the past decade, the morbidity and mortality of C. difficile infection (CDI) has increased significantly due to the emergence of hypervirulent strains. Toxin A (TcdA) and toxin B (TcdB), the two exotoxins of C. difficile, are the major virulence factors of CDI. The common mode of action of TcdA and TcdB is elicited by specific glucosylation of Rho-GTPase proteins in the host cytosol using UDP-glucose as a co-substrate, resulting in the inactivation of Rho proteins. Rho proteins are the key members in many biological processes and signaling pathways, inactivation of which leads to cytopathic and cytotoxic effects and immune responses of the host cells. It is supposed that Rho GTPases play an important role in the toxicity of C. difficile toxins. This review focuses on recent progresses in the understanding of functional consequences of Rho GTPases glucosylation induced by C. difficile toxins and the role of Rho GTPases in the toxicity of TcdA and TcdB.
Collapse
Affiliation(s)
- Shuyi Chen
- School of Bioscience and Bioengineering, South China University of Technology (SCUT), Guangzhou 510006, China.
| | - Chunli Sun
- School of Bioscience and Bioengineering, South China University of Technology (SCUT), Guangzhou 510006, China.
| | - Haiying Wang
- School of Bioscience and Bioengineering, South China University of Technology (SCUT), Guangzhou 510006, China.
| | - Jufang Wang
- School of Bioscience and Bioengineering, South China University of Technology (SCUT), Guangzhou 510006, China.
| |
Collapse
|
41
|
Wang S, Rustandi RR, Lancaster C, Hong LG, Thiriot DS, Xie J, Secore S, Kristopeit A, Wang SC, Heinrichs JH. Toxicity assessment of Clostridium difficile toxins in rodent models and protection of vaccination. Vaccine 2015; 34:1319-23. [PMID: 26614590 DOI: 10.1016/j.vaccine.2015.11.026] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2015] [Revised: 11/04/2015] [Accepted: 11/09/2015] [Indexed: 11/30/2022]
Abstract
Clostridium difficile is the leading cause of hospital-acquired diarrhea, also known as C. difficile associated diarrhea. The two major toxins, toxin A and toxin B are produced by most C. difficile bacteria, but some strains, such as BI/NAP1/027 isolates, produce a third toxin called binary toxin. The precise biological role of binary toxin is not clear but it has been shown to be a cytotoxin for Vero cells. We evaluated the toxicity of these toxins in mice and hamsters and found that binary toxin causes death in both animals similar to toxins A and B. Furthermore, immunization of mice with mutant toxoids of all three toxins provided protection upon challenge with native toxins. These results support the concept that binary toxin contributes to the pathogenicity of C. difficile and provide a method for monitoring the toxicity of binary toxin components in vaccines.
Collapse
Affiliation(s)
- Su Wang
- Vaccine Basic Research, Merck Research Laboratories, Merck & Co., Inc., Kenilworth, NJ, USA
| | - Richard R Rustandi
- Vaccine Analytical Development, Merck Research Laboratories, Merck & Co., Inc., Kenilworth, NJ, USA.
| | - Catherine Lancaster
- Vaccine Analytical Development, Merck Research Laboratories, Merck & Co., Inc., Kenilworth, NJ, USA
| | - Laura G Hong
- Vaccine Analytical Development, Merck Research Laboratories, Merck & Co., Inc., Kenilworth, NJ, USA
| | - David S Thiriot
- Vaccine Drug Product Development, Merck Research Laboratories, Merck & Co., Inc., Kenilworth, NJ, USA
| | - Jinfu Xie
- Vaccine Basic Research, Merck Research Laboratories, Merck & Co., Inc., Kenilworth, NJ, USA
| | - Susan Secore
- Vaccine Basic Research, Merck Research Laboratories, Merck & Co., Inc., Kenilworth, NJ, USA
| | - Adam Kristopeit
- Vaccine Process Development, Merck Research Laboratories, Merck & Co., Inc., Kenilworth, NJ, USA
| | - Sheng-Ching Wang
- Vaccine Process Development, Merck Research Laboratories, Merck & Co., Inc., Kenilworth, NJ, USA
| | - Jon H Heinrichs
- Vaccine Basic Research, Merck Research Laboratories, Merck & Co., Inc., Kenilworth, NJ, USA
| |
Collapse
|
42
|
Huang JH, Wu CW, Lien SP, Leng CH, Hsiao KN, Liu SJ, Chen HW, Siu LK, Chong P. Recombinant lipoprotein-based vaccine candidates against C. difficile infections. J Biomed Sci 2015; 22:65. [PMID: 26245825 PMCID: PMC4527207 DOI: 10.1186/s12929-015-0171-x] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2015] [Accepted: 07/29/2015] [Indexed: 12/18/2022] Open
Abstract
Background Opportunistically nosocomial infections in hospitalized patients are often related to Clostridium difficile infections (CDI) due to disruption of the intestinal micro-flora by antibiotic therapies during hospitalization. Clostridial exotoxins A and B (TcdA and TcdB) specifically bind to unknown glycoprotein(s) in the host intestine, disrupt the intestinal barrier leading to acute inflammation and diarrhea. The C-terminal receptor binding domain of TcdA (A-rRBD) has been shown to elicit antibody responses that neutralize TcdA toxicity in Vero cell cytotoxicity assays, but not effectively protect hamsters against a lethal dose challenge of C. difficile spores. To develop an effective recombinant subunit vaccine against CDI, A-rRBD was lipidated (rlipoA-RBD) as a rational design to contain an intrinsic adjuvant, a toll-like receptor 2 agonist and expressed in Escherichia coli. Results The purified rlipoA-RBD was characterized immunologically and found to have the following properties: (a) mice, hamsters and rabbits vaccinated with 3 μg of rlipoA-RBD produced strong antibody responses that neutralized TcdA toxicity in Vero cell cytotoxicity assays; furthermore, the neutralization titer was comparable to those obtained from antisera immunized either with 10 μg of TcdA toxoid or 30 μg of A-rRBD; (b) rlipoA-RBD elicited immune responses and protected mice from TcdA challenge, but offered insignificant protection (10 to 20 %) against C. difficile spores challenge in hamster models; (c) only rlipoA-RBD formulated with B-rRBD consistently confers protection (90 to 100 %) in the hamster challenge model; and (d) rlipoA-RBD was found to be 10-fold more potent than A-rRBD as an adjuvant to enhancing immune responses against a poor antigen such as ovalbumin. Conclusion These results indicate that rlipoA-RBD formulated with B-rRBD could be an excellent vaccine candidate for preclinical studies and future clinical trials.
Collapse
Affiliation(s)
- Jui-Hsin Huang
- Vaccine R&D Center, National Institute of Infectious Diseases and Vaccinology, National Health Research Institutes, Zhunan Town, Miaoli County, Taiwan. .,Graduate Institute of Life Science, National Defense Medical Center, Taipei, Taiwan.
| | - Chia-Wei Wu
- Vaccine R&D Center, National Institute of Infectious Diseases and Vaccinology, National Health Research Institutes, Zhunan Town, Miaoli County, Taiwan.
| | - Shu-Pei Lien
- Vaccine R&D Center, National Institute of Infectious Diseases and Vaccinology, National Health Research Institutes, Zhunan Town, Miaoli County, Taiwan.
| | - Chih-Hsiang Leng
- Vaccine R&D Center, National Institute of Infectious Diseases and Vaccinology, National Health Research Institutes, Zhunan Town, Miaoli County, Taiwan.
| | - Kuang-Nan Hsiao
- Vaccine R&D Center, National Institute of Infectious Diseases and Vaccinology, National Health Research Institutes, Zhunan Town, Miaoli County, Taiwan.
| | - Shih-Jen Liu
- Vaccine R&D Center, National Institute of Infectious Diseases and Vaccinology, National Health Research Institutes, Zhunan Town, Miaoli County, Taiwan.
| | - Hsin-Wei Chen
- Vaccine R&D Center, National Institute of Infectious Diseases and Vaccinology, National Health Research Institutes, Zhunan Town, Miaoli County, Taiwan.
| | - Leung-Kei Siu
- Vaccine R&D Center, National Institute of Infectious Diseases and Vaccinology, National Health Research Institutes, Zhunan Town, Miaoli County, Taiwan.
| | - Pele Chong
- Vaccine R&D Center, National Institute of Infectious Diseases and Vaccinology, National Health Research Institutes, Zhunan Town, Miaoli County, Taiwan. .,Graduate Institute of Life Science, National Defense Medical Center, Taipei, Taiwan. .,Graduate Institute of Immunology, China Medical University, Taichung, Taiwan.
| |
Collapse
|
43
|
Yang Z, Zhang Y, Huang T, Feng H. Glucosyltransferase activity of Clostridium difficile Toxin B is essential for disease pathogenesis. Gut Microbes 2015; 6:221-4. [PMID: 26091306 PMCID: PMC4615278 DOI: 10.1080/19490976.2015.1062965] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Abstract
Clostridium difficile TcdB harbors a glucosyltransferase that targets host Rho GTPases. However, the role of the enzyme activity in the induction of host intestinal disease has not been demonstrated. In this study, we established a mouse acute intestinal disease model by cecum injection of wild type and glucosyltransferase-deficient TcdB and a chronic model by delivering toxin intraluminally via engineered surrogate host Bacillus megaterium. We demonstrated, for the first time, that the glucosyltransferase activity of TcdB is essential for inducing disease symptoms and intestinal pathological responses that resemble human disease, highlighting the importance of targeting toxin glucosyltransferase activity for future therapy.
Collapse
Affiliation(s)
- Zhiyong Yang
- Department of Microbial Pathogenesis; University of Maryland Dental School; Baltimore, MD USA
| | - Yongrong Zhang
- Department of Microbial Pathogenesis; University of Maryland Dental School; Baltimore, MD USA
| | - Tuxiong Huang
- Department of Microbial Pathogenesis; University of Maryland Dental School; Baltimore, MD USA
| | - Hanping Feng
- Department of Microbial Pathogenesis; University of Maryland Dental School; Baltimore, MD USA,Correspondence to: Hanping Feng;
| |
Collapse
|
44
|
Wang YK, Yan YX, Kim HB, Ju X, Zhao S, Zhang K, Tzipori S, Sun X. A chimeric protein comprising the glucosyltransferase and cysteine proteinase domains of toxin B and the receptor binding domain of toxin A induces protective immunity against Clostridium difficile infection in mice and hamsters. Hum Vaccin Immunother 2015; 11:2215-22. [PMID: 26036797 PMCID: PMC4635733 DOI: 10.1080/21645515.2015.1052352] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/28/2022] Open
Abstract
Clostridium difficile is the major cause of hospital-acquired infectious diarrhea and colitis in developed countries. The pathogenicity of C. difficile is mainly mediated by the release of 2 large potent exotoxins, toxin A (TcdA) and toxin B (TcdB), both of which require neutralization to prevent disease occurrence. We have generated a novel chimeric protein, designated mTcd138, comprised of the glucosyltransferase and cysteine proteinase domains of TcdB and the receptor binding domain of TcdA and expressed it in Bacillus megaterium. To ensure that mTcd138 is atoxic, 2 point mutations were introduced to the glucosyltransferase domain of TcdB, which essentially eliminates toxicity of mTcd138. Parenteral immunizations of mice and hamsters with mTcd138 induced protective antibodies to both toxins and provided protection against infection with the hyper-virulent C. difficile strain UK6.
Collapse
Affiliation(s)
- Yuan-Kai Wang
- a Department of Infectious Diseases and Global Health ; Tufts University Cummings School of Veterinary Medicine ; North Grafton , MA USA
| | | | | | | | | | | | | | | |
Collapse
|
45
|
Yu H, Chen K, Wu J, Yang Z, Shi L, Barlow LL, Aronoff DM, Garey KW, Savidge TC, von Rosenvinge EC, Kelly CP, Feng H. Identification of toxemia in patients with Clostridium difficile infection. PLoS One 2015; 10:e0124235. [PMID: 25885671 PMCID: PMC4401762 DOI: 10.1371/journal.pone.0124235] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2014] [Accepted: 02/27/2015] [Indexed: 01/05/2023] Open
Abstract
Toxemia can develop in Clostridium difficile-infected animals, and correlates with severe and fulminant disease outcomes. Circumstantial evidence suggests that toxemia may occur in patients with C. difficile infection (CDI), but positive diagnosis is extremely rare. We analyzed the potential for C. difficile toxemia in patients, determined its characteristics, and assessed challenges. C. difficile toxins in serum from patients were tested using an ultrasensitive cell-based assay and further confirmed by Rac1 glucosylation assay. The factors that hinder a diagnosis of toxemia were assessed, including investigation of toxin stability, the level of toxins-specific neutralizing antibodies in sera and its effect on diagnosis limits. CDI patients develop detectable toxemia in some cases (2.3%). Toxins were relatively stable in stored sera. Neutralizing anti-toxin antibodies were present during infection and positively correlated with the diagnosis limits. Thus, the masking effect of toxin-specific neutralizing antibodies is the major obstacle in diagnosing C. difficile toxemia using cell-based bioassays.
Collapse
Affiliation(s)
- Hua Yu
- Department of Microbial Pathogenesis, University of Maryland Dental School, Baltimore, Maryland, United States of America
| | - Kevin Chen
- Department of Microbial Pathogenesis, University of Maryland Dental School, Baltimore, Maryland, United States of America
| | - Jianguo Wu
- Department of Microbial Pathogenesis, University of Maryland Dental School, Baltimore, Maryland, United States of America
| | - Zhiyong Yang
- Department of Microbial Pathogenesis, University of Maryland Dental School, Baltimore, Maryland, United States of America
| | - Lianfa Shi
- Department of Microbial Pathogenesis, University of Maryland Dental School, Baltimore, Maryland, United States of America
| | - Lydia L. Barlow
- Department of Medicine, University of Maryland School of Medicine, Baltimore, Maryland, United States of America
| | - David M. Aronoff
- Division of Infectious Diseases, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, United States of America; Vanderbilt University School of Medicine, Nashville, Tennessee, United States of America
| | - Kevin W. Garey
- University of Houston College of Pharmacy, Houston, Texas, United States of America; Baylor St. Luke's Medical Center, Houston, Texas, United States of America; University of Texas School of Public Health, Houston, Texas, United States of America
| | - Tor C. Savidge
- Department of Pathology and Immunology, Baylor College of Medicine, Houston, Texas, United States of America; Texas Children's Microbiome Center, Texas Children's Hospital, Houston, Texas, United States of America
| | - Erik C. von Rosenvinge
- Division of Gastroenterology & Hepatology, Department of Medicine, University of Maryland, Baltimore, Maryland, United States of America; VA Maryland Health Care System, Baltimore, Maryland, United States of America
| | - Ciaran P. Kelly
- Department of Medicine, Harvard Medical School and Celiac Center, Beth Israel Deaconess Medical Center, Boston, Massachusetts, United States of America; Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Hanping Feng
- Department of Microbial Pathogenesis, University of Maryland Dental School, Baltimore, Maryland, United States of America
- * E-mail:
| |
Collapse
|
46
|
Guo S, Yan W, McDonough SP, Lin N, Wu KJ, He H, Xiang H, Yang M, Moreira MAS, Chang YF. The recombinant Lactococcus lactis oral vaccine induces protection against C. difficile spore challenge in a mouse model. Vaccine 2015; 33:1586-95. [DOI: 10.1016/j.vaccine.2015.02.006] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2014] [Revised: 01/26/2015] [Accepted: 02/04/2015] [Indexed: 01/05/2023]
|
47
|
Zhang Y, Hamza T, Gao S, Feng H. Masking autoprocessing of Clostridium difficile toxin A by the C-terminus combined repetitive oligo peptides. Biochem Biophys Res Commun 2015; 459:259-263. [PMID: 25725153 DOI: 10.1016/j.bbrc.2015.02.095] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2015] [Accepted: 02/17/2015] [Indexed: 10/23/2022]
Abstract
Clostridium difficile toxin A and B (TcdA and TcdB) are the major virulence factors of the bacterium, both of which consist of two enzymatic domains: an effector glucosyltransferase domain (GTD) and a cysteine protease domain (CPD) responsible for autocleavage and release of GTD. Although the CPDs from both toxins share a similar structure and mechanism of hexakisphosphate (InsP6)-induced activation, TcdA is substantially less sensitive to the autocleavage as compared with TcdB. In this study, we provided evidence of inter-domain regulation of CPD activity of TcdA and its autoprocessing. The C-terminus combined repetitive oligo peptides (CROPs) of TcdA reduced the accessibility of TcdB CPD to its substrate in a chimeric toxin TxB-Ar, consequently blocking autoprocessing. Moreover, interference of antibodies with the CROPs of full-length TcdA efficiently enhanced its GTD release. In conclusion, by utilizing chimeric toxins and specific antibodies, we identified that the CROPs of TcdA plays a crucial role in controlling the InsP6-mediated activation of CPD and autocleavage of GTD. Our data provides insights on the molecular mode of action of the C. difficile toxins.
Collapse
Affiliation(s)
- Yongrong Zhang
- Department of Microbial Pathogenesis, University of Maryland Dental School, Baltimore, MD 21201, USA
| | - Therwa Hamza
- Department of Microbial Pathogenesis, University of Maryland Dental School, Baltimore, MD 21201, USA
| | - Si Gao
- Department of Microbial Pathogenesis, University of Maryland Dental School, Baltimore, MD 21201, USA
| | - Hanping Feng
- Department of Microbial Pathogenesis, University of Maryland Dental School, Baltimore, MD 21201, USA.
| |
Collapse
|
48
|
Mathur H, Rea MC, Cotter PD, Ross RP, Hill C. The potential for emerging therapeutic options for Clostridium difficile infection. Gut Microbes 2015; 5:696-710. [PMID: 25564777 PMCID: PMC4615897 DOI: 10.4161/19490976.2014.983768] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Abstract
Clostridium difficile is mainly a nosocomial pathogen and is a significant cause of antibiotic-associated diarrhea. It is also implicated in the majority of cases of pseudomembranous colitis. Recently, advancements in next generation sequencing technology (NGS) have highlighted the extent of damage to the gut microbiota caused by broad-spectrum antibiotics, often resulting in C. difficile infection (CDI). Currently the treatment of choice for CDI involves the use of metronidazole and vancomycin. However, recurrence and relapse of CDI, even after rounds of metronidazole/vancomycin administration is a problem that must be addressed. The efficacy of alternative antibiotics such as fidaxomicin, rifaximin, nitazoxanide, ramoplanin and tigecycline, as well as faecal microbiota transplantation has been assessed and some have yielded positive outcomes against C. difficile. Some bacteriocins have also shown promising effects against C. difficile in recent years. In light of this, the potential for emerging treatment options and efficacy of anti-C. difficile vaccines are discussed in this review.
Collapse
Key Words
- ATCC, American Type Culture Collection
- CDI, Clostridium difficile infection
- CdtLoc, binary toxin locus
- Clostridium difficile
- DNA, deoxyribonucleic acid
- DPC, Dairy Products Collection
- ESCMID, European Society of Clinical Microbiology and Infectious Diseases
- ETEC, enterotoxigenic E. coli
- FDA, Food and Drug Administration
- FMT, faecal microbiota transplantation
- GIT, gastrointestinal tract
- HIV, human immunodeficiency virus
- IDSA, Infectious Diseases Society of America
- IgG, immunoglobulin G
- LTA, lipoteichoic acid
- M21V, methionine to valine substitution at residue 21
- MIC, minimum inhibitory concentration
- NGS, next generation sequencing
- NVB, Novacta Biosystems Ltd
- PMC, pseudomembranous colitis
- PaLoc, pathogenicity locus
- R027, ribotype 027
- RBD
- RBS, ribosome binding site
- RNA, ribonucleic acid
- SHEA, Society for Healthcare Epidemiology of America
- V15F, valine to phenylalanine substitution at residue 15
- antibiotics
- faecal microbiota transplantation
- receptor binding domain
- toxins
- vaccines
Collapse
Affiliation(s)
- Harsh Mathur
- School of Microbiology; University College Cork; Cork, Ireland,Teagasc Food Research Center; Moorepark; Fermoy, Ireland
| | - Mary C Rea
- Teagasc Food Research Center; Moorepark; Fermoy, Ireland,Alimentary Pharmabiotic Center; University College Cork; Cork, Ireland
| | - Paul D Cotter
- Teagasc Food Research Center; Moorepark; Fermoy, Ireland,Alimentary Pharmabiotic Center; University College Cork; Cork, Ireland,Correspondence to: Colin Hill; ; Paul D Cotter;
| | - R Paul Ross
- Alimentary Pharmabiotic Center; University College Cork; Cork, Ireland,College of Science; Engineering and Food Science; University College Cork; Cork, Ireland
| | - Colin Hill
- School of Microbiology; University College Cork; Cork, Ireland,Alimentary Pharmabiotic Center; University College Cork; Cork, Ireland,Correspondence to: Colin Hill; ; Paul D Cotter;
| |
Collapse
|
49
|
|
50
|
Mechanisms of protection against Clostridium difficile infection by the monoclonal antitoxin antibodies actoxumab and bezlotoxumab. Infect Immun 2014; 83:822-31. [PMID: 25486992 DOI: 10.1128/iai.02897-14] [Citation(s) in RCA: 81] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Clostridium difficile infection (CDI) represents the most prevalent cause of antibiotic-associated gastrointestinal infections in health care facilities in the developed world. Disease symptoms are caused by the two homologous exotoxins, TcdA and TcdB. Standard therapy for CDI involves administration of antibiotics that are associated with a high rate of disease recurrence, highlighting the need for novel treatment paradigms that target the toxins rather than the organism itself. A combination of human monoclonal antibodies, actoxumab and bezlotoxumab, directed against TcdA and TcdB, respectively, has been shown to decrease the rate of recurrence in patients treated with standard-of-care antibiotics. However, the exact mechanism of antibody-mediated protection is poorly understood. In this study, we show that the antitoxin antibodies are protective in multiple murine models of CDI, including systemic and local (gut) toxin challenge models, as well as primary and recurrent models of infection in mice. Systemically administered actoxumab-bezlotoxumab prevents both the damage to the gut wall and the inflammatory response, which are associated with C. difficile in these models, including in mice challenged with a strain of the hypervirulent ribotype 027. Furthermore, mutant antibodies (N297Q) that do not bind to Fcγ receptors provide a level of protection similar to that of wild-type antibodies, demonstrating that the mechanism of protection is through direct neutralization of the toxins and does not involve host effector functions. These data provide a mechanistic basis for the prevention of recurrent disease observed in CDI patients in clinical trials.
Collapse
|