1
|
Zvonareva T, Courson DS, Purcell EB. Clostridioides difficile infection study models and prospectives for probing the microbe-host interface. J Bacteriol 2025; 207:e0040724. [PMID: 39912651 PMCID: PMC11925243 DOI: 10.1128/jb.00407-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2025] Open
Abstract
Clostridioides difficile infection (CDI) is an urgent public health threat with a high rate of recurrence and limited treatment options. In vivo models have been indispensable in understanding CDI pathophysiology and establishing treatment protocols and continue to be essential in pre-clinal testing. More importantly, in vivo models offer the opportunity to probe the complex systemic host response to the microbe, which is impossible to recapitulate in vitro. Nonetheless, constraints related to the availability of animal models, cost, ethical considerations, and regulatory control limit their accessibility for basic research. Furthermore, physiological and habitual divergences between animal models and humans often result in poor translatability to human patients. In addition to being more accessible, in vitro CDI models offer more control over experimental parameters and allow dynamic analysis of early infection. In vitro fermentation offers models for probing microbe-microbe and microbe-microbiome interactions, while continuous multi-stage platforms allow opportunities to study C. difficile pathophysiology and treatment in context with human-derived microbiota. However, these platforms are not suitable for probing the host-pathogen interface, leaving the challenge of modeling early CDI unanswered. As a result, alternative in vitro co-culture platforms are being developed. This review evaluates the strengths and weaknesses of each approach, as well as future directions for C. difficile research.
Collapse
Affiliation(s)
- Tatiana Zvonareva
- Department of Chemistry & Biochemistry, Old Dominion University, Norfolk, Virginia, USA
| | - David S. Courson
- Department of Chemistry & Biochemistry, Old Dominion University, Norfolk, Virginia, USA
| | - Erin B. Purcell
- Department of Chemistry & Biochemistry, Old Dominion University, Norfolk, Virginia, USA
| |
Collapse
|
2
|
Di Bella S, Sanson G, Monticelli J, Zerbato V, Principe L, Giuffrè M, Pipitone G, Luzzati R. Clostridioides difficile infection: history, epidemiology, risk factors, prevention, clinical manifestations, treatment, and future options. Clin Microbiol Rev 2024; 37:e0013523. [PMID: 38421181 PMCID: PMC11324037 DOI: 10.1128/cmr.00135-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/02/2024] Open
Abstract
SUMMARYClostridioides difficile infection (CDI) is one of the major issues in nosocomial infections. This bacterium is constantly evolving and poses complex challenges for clinicians, often encountered in real-life scenarios. In the face of CDI, we are increasingly equipped with new therapeutic strategies, such as monoclonal antibodies and live biotherapeutic products, which need to be thoroughly understood to fully harness their benefits. Moreover, interesting options are currently under study for the future, including bacteriophages, vaccines, and antibiotic inhibitors. Surveillance and prevention strategies continue to play a pivotal role in limiting the spread of the infection. In this review, we aim to provide the reader with a comprehensive overview of epidemiological aspects, predisposing factors, clinical manifestations, diagnostic tools, and current and future prophylactic and therapeutic options for C. difficile infection.
Collapse
Affiliation(s)
- Stefano Di Bella
- Clinical Department of
Medical, Surgical and Health Sciences, Trieste
University, Trieste,
Italy
| | - Gianfranco Sanson
- Clinical Department of
Medical, Surgical and Health Sciences, Trieste
University, Trieste,
Italy
| | - Jacopo Monticelli
- Infectious Diseases
Unit, Trieste University Hospital
(ASUGI), Trieste,
Italy
| | - Verena Zerbato
- Infectious Diseases
Unit, Trieste University Hospital
(ASUGI), Trieste,
Italy
| | - Luigi Principe
- Microbiology and
Virology Unit, Great Metropolitan Hospital
“Bianchi-Melacrino-Morelli”,
Reggio Calabria, Italy
| | - Mauro Giuffrè
- Clinical Department of
Medical, Surgical and Health Sciences, Trieste
University, Trieste,
Italy
- Department of Internal
Medicine (Digestive Diseases), Yale School of Medicine, Yale
University, New Haven,
Connecticut, USA
| | - Giuseppe Pipitone
- Infectious Diseases
Unit, ARNAS Civico-Di Cristina
Hospital, Palermo,
Italy
| | - Roberto Luzzati
- Clinical Department of
Medical, Surgical and Health Sciences, Trieste
University, Trieste,
Italy
| |
Collapse
|
3
|
Norman KM, Lang GA, Shadid TM, Honold ST, Reel JM, Cox MA, Ballard JD, Lang ML. Clostridioides difficile toxin B subverts germinal center and antibody recall responses by stimulating a drug-treatable CXCR4-dependent mechanism. Cell Rep 2024; 43:114245. [PMID: 38761377 PMCID: PMC11210377 DOI: 10.1016/j.celrep.2024.114245] [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: 10/19/2023] [Revised: 04/04/2024] [Accepted: 05/02/2024] [Indexed: 05/20/2024] Open
Abstract
Recurrent Clostridioides difficile infection (CDI) results in significant morbidity and mortality. We previously established that CDI in mice does not protect against reinfection and is associated with poor pathogen-specific B cell memory (Bmem), recapitulating our observations with human Bmem. Here, we demonstrate that the secreted toxin TcdB2 is responsible for subversion of Bmem responses. TcdB2 from an endemic C. difficile strain delayed immunoglobulin G (IgG) class switch following vaccination, attenuated IgG recall to a vaccine booster, and prevented germinal center formation. The mechanism of TcdB2 action included increased B cell CXCR4 expression and responsiveness to its ligand CXCL12, accounting for altered cell migration and a failure of germinal center-dependent Bmem. These results were reproduced in a C. difficile infection model, and a US Food and Drug Administration (FDA)-approved CXCR4-blocking drug rescued germinal center formation. We therefore provide mechanistic insights into C. difficile-associated pathogenesis and illuminate a target for clinical intervention to limit recurrent disease.
Collapse
Affiliation(s)
- Kaylee M Norman
- Department of Microbiology and Immunology, University of Oklahoma Health Sciences, Oklahoma City, OK 73104, USA
| | - Gillian A Lang
- Department of Microbiology and Immunology, University of Oklahoma Health Sciences, Oklahoma City, OK 73104, USA
| | - Tyler M Shadid
- Department of Microbiology and Immunology, University of Oklahoma Health Sciences, Oklahoma City, OK 73104, USA
| | - Sydney T Honold
- Department of Microbiology and Immunology, University of Oklahoma Health Sciences, Oklahoma City, OK 73104, USA
| | - Jessica M Reel
- Department of Microbiology and Immunology, University of Oklahoma Health Sciences, Oklahoma City, OK 73104, USA
| | - Maureen A Cox
- Department of Microbiology and Immunology, University of Oklahoma Health Sciences, Oklahoma City, OK 73104, USA
| | - Jimmy D Ballard
- Department of Microbiology and Immunology, University of Oklahoma Health Sciences, Oklahoma City, OK 73104, USA
| | - Mark L Lang
- Department of Microbiology and Immunology, University of Oklahoma Health Sciences, Oklahoma City, OK 73104, USA.
| |
Collapse
|
4
|
Pourliotopoulou E, Karampatakis T, Kachrimanidou M. Exploring the Toxin-Mediated Mechanisms in Clostridioides difficile Infection. Microorganisms 2024; 12:1004. [PMID: 38792835 PMCID: PMC11124097 DOI: 10.3390/microorganisms12051004] [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: 04/20/2024] [Revised: 05/14/2024] [Accepted: 05/15/2024] [Indexed: 05/26/2024] Open
Abstract
Clostridioides difficile infection (CDI) is the leading cause of nosocomial antibiotic-associated diarrhea, and colitis, with increasing incidence and healthcare costs. Its pathogenesis is primarily driven by toxins produced by the bacterium C. difficile, Toxin A (TcdA) and Toxin B (TcdB). Certain strains produce an additional toxin, the C. difficile transferase (CDT), which further enhances the virulence and pathogenicity of C. difficile. These toxins disrupt colonic epithelial barrier integrity, and induce inflammation and cellular damage, leading to CDI symptoms. Significant progress has been made in the past decade in elucidating the molecular mechanisms of TcdA, TcdB, and CDT, which provide insights into the management of CDI and the future development of novel treatment strategies based on anti-toxin therapies. While antibiotics are common treatments, high recurrence rates necessitate alternative therapies. Bezlotoxumab, targeting TcdB, is the only available anti-toxin, yet limitations persist, prompting ongoing research. This review highlights the current knowledge of the structure and mechanism of action of C. difficile toxins and their role in disease. By comprehensively describing the toxin-mediated mechanisms, this review provides insights for the future development of novel treatment strategies and the management of CDI.
Collapse
Affiliation(s)
- Evdokia Pourliotopoulou
- Department of Microbiology, Medical School, Aristotle University of Thessaloniki, 541 24 Thessaloniki, Greece;
| | | | - Melania Kachrimanidou
- Department of Microbiology, Medical School, Aristotle University of Thessaloniki, 541 24 Thessaloniki, Greece;
| |
Collapse
|
5
|
Ferreira G, Cardozo R, Sastre S, Costa C, Santander A, Chavarría L, Guizzo V, Puglisi J, Nicolson GL. Bacterial toxins and heart function: heat-labile Escherichia coli enterotoxin B promotes changes in cardiac function with possible relevance for sudden cardiac death. Biophys Rev 2023; 15:447-473. [PMID: 37681088 PMCID: PMC10480140 DOI: 10.1007/s12551-023-01100-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2023] [Accepted: 07/11/2023] [Indexed: 09/09/2023] Open
Abstract
Bacterial toxins can cause cardiomyopathy, though it is not its most common cause. Some bacterial toxins can form pores in the membrane of cardiomyocytes, while others can bind to membrane receptors. Enterotoxigenic E. coli can secrete enterotoxins, including heat-resistant (ST) or labile (LT) enterotoxins. LT is an AB5-type toxin that can bind to specific cell receptors and disrupt essential host functions, causing several common conditions, such as certain diarrhea. The pentameric B subunit of LT, without A subunit (LTB), binds specifically to certain plasma membrane ganglioside receptors, found in lipid rafts of cardiomyocytes. Isolated guinea pig hearts and cardiomyocytes were exposed to different concentrations of purified LTB. In isolated hearts, mechanical and electrical alternans and an increment of heart rate variability, with an IC50 of ~0.2 μg/ml LTB, were observed. In isolated cardiomyocytes, LTB promoted significant decreases in the amplitude and the duration of action potentials. Na+ currents were inhibited whereas L-type Ca2+ currents were augmented at their peak and their fast inactivation was promoted. Delayed rectifier K+ currents decreased. Measurements of basal Ca2+ or Ca2+ release events in cells exposed to LTB suggest that LTB impairs Ca2+ homeostasis. Impaired calcium homeostasis is linked to sudden cardiac death. The results are consistent with the recent view that the B subunit is not merely a carrier of the A subunit, having a role explaining sudden cardiac death in children (SIDS) infected with enterotoxigenic E. coli, explaining several epidemiological findings that establish a strong relationship between SIDS and ETEC E. coli. Supplementary Information The online version contains supplementary material available at 10.1007/s12551-023-01100-6.
Collapse
Affiliation(s)
- Gonzalo Ferreira
- Ion Channels, Biological Membranes and Cell Signaling Laboratory, Dept. Of Biophysics, Facultad de Medicina, Universidad de la Republica, Gral Flores 2125, 11800 Montevideo, CP Uruguay
| | - Romina Cardozo
- Ion Channels, Biological Membranes and Cell Signaling Laboratory, Dept. Of Biophysics, Facultad de Medicina, Universidad de la Republica, Gral Flores 2125, 11800 Montevideo, CP Uruguay
| | - Santiago Sastre
- Ion Channels, Biological Membranes and Cell Signaling Laboratory, Dept. Of Biophysics and Centro de Investigaciones Biomédicas (CeInBio), Facultad de Medicina, Universidad de la Republica, Gral Flores 2125, 11800 Montevideo, CP Uruguay
| | - Carlos Costa
- Ion Channels, Biological Membranes and Cell Signaling Laboratory, Dept. Of Biophysics, Facultad de Medicina, Universidad de la Republica, Gral Flores 2125, 11800 Montevideo, CP Uruguay
| | - Axel Santander
- Ion Channels, Biological Membranes and Cell Signaling Laboratory, Dept. Of Biophysics, Facultad de Medicina, Universidad de la Republica, Gral Flores 2125, 11800 Montevideo, CP Uruguay
| | - Luisina Chavarría
- Ion Channels, Biological Membranes and Cell Signaling Laboratory, Dept. Of Biophysics, Facultad de Medicina, Universidad de la Republica, Gral Flores 2125, 11800 Montevideo, CP Uruguay
| | - Valentina Guizzo
- Ion Channels, Biological Membranes and Cell Signaling Laboratory, Dept. Of Biophysics, Facultad de Medicina, Universidad de la Republica, Gral Flores 2125, 11800 Montevideo, CP Uruguay
| | - José Puglisi
- College of Medicine, California North State University, 9700 West Taron Drive, Elk Grove, CA 95757 USA
| | - G. L. Nicolson
- Institute for Molecular Medicine, Beach, Huntington, CA USA
| |
Collapse
|
6
|
Hu C, Garey KW. Nonmammalian models to study Clostridioides difficile infection; a systematic review. Anaerobe 2023; 79:102694. [PMID: 36626950 PMCID: PMC9975065 DOI: 10.1016/j.anaerobe.2023.102694] [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: 10/11/2022] [Revised: 01/05/2023] [Accepted: 01/06/2023] [Indexed: 01/09/2023]
Abstract
Clostridioide difficile is the leading cause of diarrhea disease worldwide and is a CDC-designated urgent threat level pathogen. Mammalian models are commonly utilized as gold standard to study the pathogenesis of C. difficile infection (CDI); however, alternatives are needed due to cost, higher throughput ability, and mammalian animal ethics. Nonmammalian models such as great wax worm, nematode, fruit fly, and zebrafish have been used as CDI models. This review provides a comprehensive summary of nonmammalian models used to study CDI. Multiple studies were identified using these models to study C. difficile infection, pathogenicity, colonization, host immunity, and therapy. Translational outcomes and strength and weakness of each nonmammalian model are discussed.
Collapse
Affiliation(s)
- Chenlin Hu
- University of Houston College of Pharmacy, Houston, TX, 77204, USA
| | - Kevin W Garey
- University of Houston College of Pharmacy, Houston, TX, 77204, USA.
| |
Collapse
|
7
|
Pereañez JA, Granados J, Agudelo R. Tako-tsubo cardiomyopathy in clinical toxinology: A systematic review. Toxicon 2022; 219:106929. [PMID: 36162498 DOI: 10.1016/j.toxicon.2022.09.015] [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: 06/24/2022] [Revised: 09/02/2022] [Accepted: 09/16/2022] [Indexed: 11/25/2022]
Abstract
Tako-tsubo cardiomyopathy (TTC) is a transient left ventricular dysfunction, normally triggered by emotional or physical stress, although it is also associated with to use of drugs, drug abuse, or some intoxications. In addition, TTC has been reported in some case reports derived from the exposure of patients to animal venoms, toxins or poisons, or bacterial infections. However, to date, a systematic assessment of TTC in clinical toxinology is lacking. Therefore the aim of this study was to collect and integrate the available information about TTC in clinical toxinology. After our search strategy, 19 articles were retrieved, resulting in 20 case reports. Most cases occurred in women (75.0%). The venomous species that trigger TTC are bee/wasp, including probable Africanized honey bee and Vespa orientalis (15.0%), scorpions (Tytius serrulatus and Androctonus australis, 15.0%), a spider (Latrodectus tredecimguttatus, 5.0%), snakes (Gloydius blomhofii and Naja nivea, 10.0%), Clostridium sp (C. tetani, C. botulinum and C. difficile, 45.0%) and jellyfish (Pelagia noctiluca and Carukia barnesi, 10.0%). Among the affected people there were two deaths. In all case reports authors diagnosed TTC by using the combination of some of the following strategies: clinical findings, echocardiography, magnetic cardiac resonance, electrocardiogram changes and/or the increased plasma levels of cardiac damage biomarkers. In most cases images were available. We hypothesized the possible mode of action of venoms, toxins or poisons to induce TTC, however other mechanisms may exist, but they have not been described yet. Therefore, further studies are needed. In some cases, venoms, toxins, or poisons might cause catecholamine discharge either directly or indirectly, therefore, this was suggested as the trigger of TTC. Finally, the appearance of TTC should be considered in clinical toxinology.
Collapse
Affiliation(s)
- Jaime Andrés Pereañez
- Grupo de Investigación en Toxinología, Alternativas Terapéuticas y Alimentarias, Facultad de Ciencias Farmacéuticas y Alimentarias, Universidad de Antioquia, Medellín, 050010, Colombia; Grupo Promoción y Prevención Farmacéutica, Facultad de Ciencias Farmacéuticas y Alimentarias, Universidad de Antioquia, Medellín, 050010, Colombia.
| | - Johan Granados
- Grupo Promoción y Prevención Farmacéutica, Facultad de Ciencias Farmacéuticas y Alimentarias, Universidad de Antioquia, Medellín, 050010, Colombia
| | - Raquel Agudelo
- Departamento de Farmacia, Facultad de Ciencias Farmacéuticas y Alimentarias, Universidad de Antioquia, Medellín, 050010, Colombia
| |
Collapse
|
8
|
Guo P, Liu Y, Feng J, Tang S, Wei F, Feng J. p21-activated kinase 1 (PAK1) as a therapeutic target for cardiotoxicity. Arch Toxicol 2022; 96:3143-3162. [DOI: 10.1007/s00204-022-03384-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2022] [Accepted: 09/14/2022] [Indexed: 11/02/2022]
|
9
|
Human α-Defensin-6 Neutralizes Clostridioides difficile Toxins TcdA and TcdB by Direct Binding. Int J Mol Sci 2022; 23:ijms23094509. [PMID: 35562899 PMCID: PMC9101188 DOI: 10.3390/ijms23094509] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2022] [Revised: 04/14/2022] [Accepted: 04/14/2022] [Indexed: 02/04/2023] Open
Abstract
Rising incidences and mortalities have drawn attention to Clostridioides difficile infections (CDIs) in recent years. The main virulence factors of this bacterium are the exotoxins TcdA and TcdB, which glucosylate Rho-GTPases and thereby inhibit Rho/actin-mediated processes in cells. This results in cell rounding, gut barrier disruption and characteristic clinical symptoms. So far, treatment of CDIs is limited and mainly restricted to some antibiotics, often leading to a vicious circle of antibiotic-induced disease recurrence. Here, we demonstrate the protective effect of the human antimicrobial peptide α-defensin-6 against TcdA, TcdB and the combination of both toxins in vitro and in vivo and unravel the underlying molecular mechanism. The defensin prevented toxin-mediated glucosylation of Rho-GTPases in cells and protected human cells, model epithelial barriers as well as zebrafish embryos from toxic effects. In vitro analyses revealed direct binding to TcdB in an SPR approach and the rapid formation of TcdB/α-defensin-6 complexes, as analyzed with fluorescent TcdB by time-lapse microscopy. In conclusion, the results imply that α-defensin-6 rapidly sequesters the toxin into complexes, which prevents its cytotoxic activity. These findings extend the understanding of how human peptides neutralize bacterial protein toxins and might be a starting point for the development of novel therapeutic options against CDIs.
Collapse
|
10
|
Lang GA, Norman K, Amadou Amani S, Shadid TM, Ballard JD, Lang ML. Use of a Clostridioides difficile Murine Immunization and Challenge Model to Evaluate Single and Combination Vaccine Adjuvants Consisting of Alum and NKT Cell-Activating Ligands. Front Immunol 2022; 12:818734. [PMID: 35095921 PMCID: PMC8794951 DOI: 10.3389/fimmu.2021.818734] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2021] [Accepted: 12/27/2021] [Indexed: 01/14/2023] Open
Abstract
Adjuvant combinations may enhance or broaden the expression of immune responses to vaccine antigens. Information on whether established Alum type adjuvants can be combined with experimental CD1d ligand adjuvants is currently lacking. In this study, we used a murine Clostridioides difficile immunization and challenge model to evaluate Alum (Alhydrogel™), α-galactosylceramide (α-GC), and one of its analogs 7DW8-5 singly and in combination as vaccine adjuvants. We observed that the Alum/α-GC combination caused modest enhancement of vaccine antigen-specific IgG1 and IgG2b responses, and a broadening to include IgG2c that did not significantly impact overall protection. Similar observations were made using the Alum/7DW8-5 combination. Examination of the impact of adjuvants on NKT cells revealed expansion of invariant NKT (iNKT) cells with modest expansion of their iNKTfh subset and little effect on diverse NKT (dNKT) cells. Side effects of the adjuvants was determined and revealed transient hepatotoxicity when Alum/α-GC was used in combination but not singly. In summary these results showed that the Alum/α-GC or the Alum/7DW8-5 combination could exert distinct effects on the NKT cell compartment and on isotype switch to produce Th1-driven IgG subclasses in addition to Alum/Th2-driven subclasses. While Alum alone was efficacious in stimulating IgG-mediated protection, and α-GC offered no apparent additional benefit in the C. difficile challenge model, the work herein reveals immune response features that could be optimized and harnessed in other vaccine contexts.
Collapse
|
11
|
Tonon F, Di Bella S, Grassi G, Luzzati R, Ascenzi P, di Masi A, Zennaro C. Extra-Intestinal Effects of C.difficile Toxin A and B: An In Vivo Study Using the Zebrafish Embryo Model. Cells 2020; 9:2575. [PMID: 33271969 PMCID: PMC7760802 DOI: 10.3390/cells9122575] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2020] [Revised: 11/27/2020] [Accepted: 11/29/2020] [Indexed: 12/18/2022] Open
Abstract
C.difficile infection (CDI) is not a merely "gut-confined" disease as toxemia could drive the development of CDI-related extra-intestinal effects. These effects could explain the high CDI-associated mortality, not just justified by diarrhea and dehydration. Here, the extra-intestinal effects of toxin A (TcdA) and B (TcdB) produced by C. difficile have been studied in vivo using the zebrafish embryo model. Noteworthy, protective properties of human serum albumin (HSA) towards toxins-induced extra-intestinal effects were also addressed. Zebrafish embryos were treated with TcdA, TcdB and/or HSA at 24 h post-fertilization. Embryos were analyzed for 48 h after treatment to check vital signs and morphological changes. Markers related to cardio-vascular damage and inflammation were evaluated by Real-Time quantitative PCR and/or western blotting. Both toxins induced cardiovascular damage in zebrafish embryos by different mechanisms: (i) direct toxicity (i.e., pericardial edema, cardiac chambers enlargement, endothelial alteration); (ii) increased hormonal production and release (i.e., atrial natriuretic peptide (ANP) and brain natriuretic peptide (BNP)), (iii) alteration of the vascular system through the increase of the vascular endothelial growth factor (VEGF-A) levels, as well as of its receptors, (iv) pro-inflammatory response through high cytokines production (i.e., CXCL8, IL1B, IL6 and TNFα) and (v) cell-mediated damage due to the increase in neutrophils number. In addition to cardiovascular damage, we observe skin alteration and inflammation. Finally, our data indicate a protective effect of HSA toward the toxins induced extra-intestinal effects. Together, our findings can serve as a starting point for humans' studies to substantiate and understand the extra-intestinal effects observed in CDI patients.
Collapse
Affiliation(s)
- Federica Tonon
- Department of Medical, Surgery and Health Sciences, University of Trieste, 34149 Trieste, Italy; (S.D.B.); (R.L.); (C.Z.)
| | - Stefano Di Bella
- Department of Medical, Surgery and Health Sciences, University of Trieste, 34149 Trieste, Italy; (S.D.B.); (R.L.); (C.Z.)
| | - Gabriele Grassi
- Department of Life Sciences, University of Trieste, 34128 Trieste, Italy;
| | - Roberto Luzzati
- Department of Medical, Surgery and Health Sciences, University of Trieste, 34149 Trieste, Italy; (S.D.B.); (R.L.); (C.Z.)
| | - Paolo Ascenzi
- Department of Sciences, Roma Tre University, 00154 Rome, Italy; (P.A.); (A.d.M.)
| | - Alessandra di Masi
- Department of Sciences, Roma Tre University, 00154 Rome, Italy; (P.A.); (A.d.M.)
| | - Cristina Zennaro
- Department of Medical, Surgery and Health Sciences, University of Trieste, 34149 Trieste, Italy; (S.D.B.); (R.L.); (C.Z.)
| |
Collapse
|
12
|
Giacobbe DR, Dettori S, Di Bella S, Vena A, Granata G, Luzzati R, Petrosillo N, Bassetti M. Bezlotoxumab for Preventing Recurrent Clostridioides difficile Infection: A Narrative Review from Pathophysiology to Clinical Studies. Infect Dis Ther 2020; 9:481-494. [PMID: 32632582 PMCID: PMC7452994 DOI: 10.1007/s40121-020-00314-5] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2020] [Indexed: 12/17/2022] Open
Abstract
Clostridioides difficile infection (CDI) and recurrent CDI (rCDI) remain associated with a reduction in the patients' quality of life and with increased healthcare costs. Bezlotoxumab is a monoclonal antibody against toxin B of C. difficile, approved for prevention of rCDI. In this narrative review, we briefly discuss the pathophysiology of CDI and the mechanism of action of bezlotoxumab, as well as the available evidence from investigational and observational studies in terms of efficacy, effectiveness, and safety of bezlotoxumab for the prevention of rCDI. Overall, bezlotoxumab has proved efficacious in reducing the burden of rCDI, thereby providing clinicians with an important novel strategy to achieve sustained cure. Nonetheless, experiences outside randomized controlled trials (RCTs) remain scant, and mostly represented by case series without a control group. Along with the conduction of RCTs to directly compare bezlotoxumab with faecal microbiota transplantation (or to precisely evaluate the role of their combined use), further widening our post-marketing experience remains paramount to firmly guide the use of bezlotoxumab outside RCTs, and to clearly identify those real-life settings where its preventive benefits can be exploited most.
Collapse
Affiliation(s)
- Daniele Roberto Giacobbe
- Infectious Diseases Unit, Ospedale Policlinico San Martino, IRCCS, Genoa, Italy.
- Department of Health Sciences, University of Genoa, Genoa, Italy.
| | - Silvia Dettori
- Infectious Diseases Unit, Ospedale Policlinico San Martino, IRCCS, Genoa, Italy
- Department of Health Sciences, University of Genoa, Genoa, Italy
| | - Stefano Di Bella
- Clinical Department of Medical, Surgical and Health Sciences, Trieste University, Trieste, Italy
| | - Antonio Vena
- Infectious Diseases Unit, Ospedale Policlinico San Martino, IRCCS, Genoa, Italy
| | - Guido Granata
- Clinical and Research Department for Infectious Diseases, Severe and Immunedepression-Associated Infections Unit, National Institute for Infectious Diseases L. Spallanzani, IRCCS, Rome, Italy
| | - Roberto Luzzati
- Clinical Department of Medical, Surgical and Health Sciences, Trieste University, Trieste, Italy
| | - Nicola Petrosillo
- Clinical and Research Department for Infectious Diseases, Severe and Immunedepression-Associated Infections Unit, National Institute for Infectious Diseases L. Spallanzani, IRCCS, Rome, Italy
| | - Matteo Bassetti
- Infectious Diseases Unit, Ospedale Policlinico San Martino, IRCCS, Genoa, Italy
- Department of Health Sciences, University of Genoa, Genoa, Italy
| |
Collapse
|
13
|
Identification of the role of toxin B in the virulence of Clostridioides difficile based on integrated bioinformatics analyses. Int Microbiol 2020; 23:575-587. [PMID: 32388701 DOI: 10.1007/s10123-020-00128-y] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2019] [Revised: 03/03/2020] [Accepted: 04/13/2020] [Indexed: 02/08/2023]
Abstract
PURPOSE Clostridioides difficile toxin B (TcdB) plays a critical role in C. difficile infection (CDI), a common and costly healthcare-associated disease. The aim of the current study was to explore the intracellular and potent systemic effects of TcdB on human colon epithelial cells utilizing Gene Expression Omnibus and bioinformatic methods. METHODS Two datasets (GSE63880 and GSE29008) were collected to extract data components of mRNA of TcdB-treated human colon epithelial cells; "limma" package of "R" software was used to screen the differential genes, and "pheatmap" package was applied to construct heat maps for the differential genes; Metascape website was utilized for protein-protein interaction network and Molecular Complex Detection analysis, and Genome Ontology (GO) was used to analyze the selected differential genes. Quantitative real-time PCR (qRT-PCR) and Western blot were performed to validate the expression of hub genes. RESULTS GO terms involved in DNA replication and cell cycle were identified significantly enriched in TcdB-treated human colon epithelial cells. Moreover, the decreased expression of DNA replication-related genes, MCM complex, and CDC45 in C. difficile (TcdA-/TcdB+)-infected Caco-2 cells were validated via qRT-PCR and Western blot assays. CONCLUSIONS In conclusion, the integrated analysis of different gene expression datasets allowed us to identify a set of genes and GO terms underlying the mechanisms of CDI induced by TcdB. It would aid in understanding of the molecular mechanisms underlying TcdB-exposed colon epithelial cells and provide the basis for developing diagnosis biomarkers, treatment, and prevention strategies.
Collapse
|
14
|
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
|
15
|
di Masi A, Leboffe L, Polticelli F, Tonon F, Zennaro C, Caterino M, Stano P, Fischer S, Hägele M, Müller M, Kleger A, Papatheodorou P, Nocca G, Arcovito A, Gori A, Ruoppolo M, Barth H, Petrosillo N, Ascenzi P, Di Bella S. Human Serum Albumin Is an Essential Component of the Host Defense Mechanism Against Clostridium difficile Intoxication. J Infect Dis 2019; 218:1424-1435. [PMID: 29868851 DOI: 10.1093/infdis/jiy338] [Citation(s) in RCA: 47] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2017] [Accepted: 06/01/2018] [Indexed: 01/05/2023] Open
Abstract
Background The pathogenic effects of Clostridium difficile are primarily attributable to the production of the large protein toxins (C difficile toxins [Tcd]) A (TcdA) and B (TcdB). These toxins monoglucosylate Rho GTPases in the cytosol of host cells, causing destruction of the actin cytoskeleton with cytotoxic effects. Low human serum albumin (HSA) levels indicate a higher risk of acquiring and developing a severe C difficile infection (CDI) and are associated with recurrent and fatal disease. Methods We used a combined approach based on docking simulation and biochemical analyses that were performed in vitro on purified proteins and in human epithelial colorectal adenocarcinoma cells (Caco-2), and in vivo on stem cell-derived human intestinal organoids and zebrafish embryos. Results Our results show that HSA specifically binds via its domain II to TcdA and TcdB and thereby induces their autoproteolytic cleavage at physiological concentrations. This process impairs toxin internalization into the host cells and reduces the toxin-dependent glucosylation of Rho proteins. Conclusions Our data provide evidence for a specific HSA-dependent self-defense mechanism against C difficile toxins and provide an explanation for the clinical correlation between CDI severity and hypoalbuminemia.
Collapse
Affiliation(s)
| | - Loris Leboffe
- Department of Sciences, Roma Tre University, Roma, Italy
| | - Fabio Polticelli
- Department of Sciences, Roma Tre University, Roma, Italy.,National Institute of Nuclear Physics, Roma Tre Section, Roma, Italy
| | - Federica Tonon
- Department of Medical, Surgery and Health Sciences, University of Trieste, Trieste, Italy
| | - Cristina Zennaro
- Department of Medical, Surgery and Health Sciences, University of Trieste, Trieste, Italy
| | - Marianna Caterino
- Department of Molecular Medicine and Medical Biotechnology, University of Napoli "Federico II", Napoli, Italy.,Associazione Culturale DiSciMuS RCF, Casoria, Napoli, Italy
| | - Pasquale Stano
- Department of Sciences, Roma Tre University, Roma, Italy
| | - Stephan Fischer
- Institute of Pharmacology and Toxicology, University of Ulm Medical Center, Ulm, Germany
| | - Marlen Hägele
- Department of Internal Medicine I, University of Ulm Medical Center, Germany
| | - Martin Müller
- Department of Internal Medicine I, University of Ulm Medical Center, Germany
| | - Alexander Kleger
- Department of Internal Medicine I, University of Ulm Medical Center, Germany
| | - Panagiotis Papatheodorou
- Institute of Pharmacology and Toxicology, University of Ulm Medical Center, Ulm, Germany.,Institute of Experimental and Clinical Pharmacology and Toxicology, University of Freiburg, Freiburg, Germany
| | - Giuseppina Nocca
- Institute of Biochemistry and Clinical Biochemistry, Catholic University of Sacred Heart, Roma, Italy.,Institute of Chemistry of Molecular Recognition, CNR, Roma, Italy
| | - Alessandro Arcovito
- Institute of Biochemistry and Clinical Biochemistry, Catholic University of Sacred Heart, Roma, Italy
| | - Andrea Gori
- Clinic of Infectious Diseases, San Gerardo Hospital, University of Milano-Bicocca, Monza, Italy
| | - Margherita Ruoppolo
- Department of Molecular Medicine and Medical Biotechnology, University of Napoli "Federico II", Napoli, Italy.,Associazione Culturale DiSciMuS RCF, Casoria, Napoli, Italy.,CEINGE Biotecnologie Avanzate, Napoli, Italy
| | - Holger Barth
- Institute of Pharmacology and Toxicology, University of Ulm Medical Center, Ulm, Germany
| | - Nicola Petrosillo
- 2nd Infectious Diseases Division, National Institute for Infectious Diseases "L. Spallanzani", Roma, Italy
| | - Paolo Ascenzi
- Department of Sciences, Roma Tre University, Roma, Italy
| | - Stefano Di Bella
- 2nd Infectious Diseases Division, National Institute for Infectious Diseases "L. Spallanzani", Roma, Italy
| |
Collapse
|
16
|
|
17
|
Chandrasekaran R, Lacy DB. The role of toxins in Clostridium difficile infection. FEMS Microbiol Rev 2017; 41:723-750. [PMID: 29048477 PMCID: PMC5812492 DOI: 10.1093/femsre/fux048] [Citation(s) in RCA: 232] [Impact Index Per Article: 29.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2017] [Accepted: 10/10/2017] [Indexed: 02/06/2023] Open
Abstract
Clostridium difficile is a bacterial pathogen that is the leading cause of nosocomial antibiotic-associated diarrhea and pseudomembranous colitis worldwide. The incidence, severity, mortality and healthcare costs associated with C. difficile infection (CDI) are rising, making C. difficile a major threat to public health. Traditional treatments for CDI involve use of antibiotics such as metronidazole and vancomycin, but disease recurrence occurs in about 30% of patients, highlighting the need for new therapies. The pathogenesis of C. difficile is primarily mediated by the actions of two large clostridial glucosylating toxins, toxin A (TcdA) and toxin B (TcdB). Some strains produce a third toxin, the binary toxin C. difficile transferase, which can also contribute to C. difficile virulence and disease. These toxins act on the colonic epithelium and immune cells and induce a complex cascade of cellular events that result in fluid secretion, inflammation and tissue damage, which are the hallmark features of the disease. In this review, we summarize our current understanding of the structure and mechanism of action of the C. difficile toxins and their role in disease.
Collapse
Affiliation(s)
- Ramyavardhanee Chandrasekaran
- Department of Pathology, Microbiology and Immunology, Vanderbilt University School of Medicine, Nashville, TN 37232, USA
| | - D. Borden Lacy
- Department of Pathology, Microbiology and Immunology, Vanderbilt University School of Medicine, Nashville, TN 37232, USA
- Department of Biochemistry, Vanderbilt University School of Medicine, Nashville, TN 37232, USA
- The Veterans Affairs Tennessee Valley Healthcare System, Nashville, TN 37232, USA
| |
Collapse
|
18
|
Wang L, Zhang X, Chan JYW, Shan L, Cui G, Cui Q, Wang Y, Li J, Chen H, Zhang Q, Yu P, Han Y, Wang Y, Lee SMY. A Novel Danshensu Derivative Prevents Cardiac Dysfunction and Improves the Chemotherapeutic Efficacy of Doxorubicin in Breast Cancer Cells. J Cell Biochem 2016; 117:94-105. [PMID: 26058377 DOI: 10.1002/jcb.25253] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2015] [Accepted: 06/05/2015] [Indexed: 12/19/2022]
Abstract
Doxorubicin (Dox) is an anthracycline antibiotic widely used in clinics as an anticancer agent. However, the use of Dox is limited by its cardiotoxicity. We have previously shown that a Danshensu (DSS) derivative, ADTM, displayed strong cardioprotective effects. With improved chemical stability and activity, a novel DSS derivative, D006, based on the structure of ADTM, was synthesized. In the present study, the protective effects of D006, indexed by attenuation of the cardiotoxicity induced by Dox as well as chemosensitizing effects that increase the antitumor activity of Dox, were investigated. Our results showed that D006 was more potent than either parental compound, or their use in combination, in ameliorating Dox-induced toxicity in H9c2 cells. In our zebrafish model, D006, but not DSS, alone significantly preserved the ventricular function of zebrafish after Dox treatment. Moreover, D006 upregulated mitochondrial biogenesis and increased mtDNA copy number after Dox treatment of H9c2 cells. D006 promoted the expression of HO-1 protein in a time-dependent manner while the HO-1 inhibitor, Znpp, reversed the protective effects of D006. In human breast tumor MCF-7 cells, D006 enhanced Dox-induced cytotoxicity by increasing apoptosis. In conclusion, our results indicate that a new DSS derivative exhibits promising protective effects against Dox-induced cardiotoxicity both in vivo and in vitro, an effect at least partially mediated by induction of HO-1 expression and the activation of mitochondrial biogenesis. Meanwhile, D006 also potentiated the anti-cancer effects of Dox in breast tumor cells.
Collapse
Affiliation(s)
- Liang Wang
- State Key Laboratory of Quality Research in Chinese Institute of Chinese Medical Sciences, University of Macau, Macao, China.,Department of Applied Biology and Chemical Technology, Institute of Modern Medicine, Hong Kong Polytechnic University, Hong Kong, China
| | - Xiaojing Zhang
- Institute of New Drug Research, College of Pharmacy, Jinan University, Guangzhou, China
| | - Judy Yuet-Wa Chan
- State Key Laboratory of Quality Research in Chinese Institute of Chinese Medical Sciences, University of Macau, Macao, China
| | - Luchen Shan
- Institute of New Drug Research, College of Pharmacy, Jinan University, Guangzhou, China
| | - Guozhen Cui
- State Key Laboratory of Quality Research in Chinese Institute of Chinese Medical Sciences, University of Macau, Macao, China
| | - Qingbin Cui
- Institute of New Drug Research, College of Pharmacy, Jinan University, Guangzhou, China
| | - Yingfei Wang
- Institute of New Drug Research, College of Pharmacy, Jinan University, Guangzhou, China
| | - Jingjing Li
- State Key Laboratory of Quality Research in Chinese Institute of Chinese Medical Sciences, University of Macau, Macao, China
| | - Huanxian Chen
- State Key Laboratory of Quality Research in Chinese Institute of Chinese Medical Sciences, University of Macau, Macao, China
| | - Qingwen Zhang
- State Key Laboratory of Quality Research in Chinese Institute of Chinese Medical Sciences, University of Macau, Macao, China
| | - Pei Yu
- Institute of New Drug Research, College of Pharmacy, Jinan University, Guangzhou, China
| | - Yifan Han
- Department of Applied Biology and Chemical Technology, Institute of Modern Medicine, Hong Kong Polytechnic University, Hong Kong, China
| | - Yuqiang Wang
- Institute of New Drug Research, College of Pharmacy, Jinan University, Guangzhou, China
| | - Simon Ming-Yuen Lee
- State Key Laboratory of Quality Research in Chinese Institute of Chinese Medical Sciences, University of Macau, Macao, China
| |
Collapse
|
19
|
Tao L, Zhang J, Meraner P, Tovaglieri A, Wu X, Gerhard R, Zhang X, Stallcup WB, Miao J, He X, Hurdle JG, Breault DT, Brass AL, Dong M. Frizzled proteins are colonic epithelial receptors for C. difficile toxin B. Nature 2016; 538:350-355. [PMID: 27680706 PMCID: PMC5519134 DOI: 10.1038/nature19799] [Citation(s) in RCA: 216] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2016] [Accepted: 08/26/2016] [Indexed: 02/06/2023]
Abstract
Clostridium difficile toxin B (TcdB) is a critical virulence factor that causes diseases associated with C. difficile infection. Here we carried out CRISPR-Cas9-mediated genome-wide screens and identified the members of the Wnt receptor frizzled family (FZDs) as TcdB receptors. TcdB binds to the conserved Wnt-binding site known as the cysteine-rich domain (CRD), with the highest affinity towards FZD1, 2 and 7. TcdB competes with Wnt for binding to FZDs, and its binding blocks Wnt signalling. FZD1/2/7 triple-knockout cells are highly resistant to TcdB, and recombinant FZD2-CRD prevented TcdB binding to the colonic epithelium. Colonic organoids cultured from FZD7-knockout mice, combined with knockdown of FZD1 and 2, showed increased resistance to TcdB. The colonic epithelium in FZD7-knockout mice was less susceptible to TcdB-induced tissue damage in vivo. These findings establish FZDs as physiologically relevant receptors for TcdB in the colonic epithelium.
Collapse
Affiliation(s)
- Liang Tao
- Department of Urology, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts 02115, USA.,Department of Microbiology and Immunobiology and Department of Surgery, Harvard Medical School, Boston, Massachusetts 02115, USA
| | - Jie Zhang
- Department of Urology, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts 02115, USA.,Department of Microbiology and Immunobiology and Department of Surgery, Harvard Medical School, Boston, Massachusetts 02115, USA
| | - Paul Meraner
- Department of Microbiology and Physiological Systems (MaPS), University of Massachusetts Medical School, Worcester, Massachusetts 01655, USA
| | - Alessio Tovaglieri
- Division of Endocrinology, Boston Children's Hospital, Boston, Massachusetts 02115, USA
| | - Xiaoqian Wu
- Center for Infectious and Inflammatory Diseases, Texas A &M Health Science Center, Houston, Texas 77030, USA
| | - Ralf Gerhard
- Institute of Toxicology, Hannover Medical School, 30625 Hannover, Germany
| | - Xinjun Zhang
- The F. M. Kirby Neurobiology Center, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts 02115, USA.,Department of Neurology, Harvard Medical School, Boston, Massachusetts 02115, USA
| | - William B Stallcup
- Tumor Microenvironment and Cancer Immunology Program, Sanford-Burnham Prebys Medical Discovery Institute, Cancer Center, La Jolla, California 92037, USA
| | - Ji Miao
- Division of Endocrinology, Boston Children's Hospital, Boston, Massachusetts 02115, USA.,Department of Pediatrics, Harvard Medical School, Boston, Massachusetts 02115, USA
| | - Xi He
- The F. M. Kirby Neurobiology Center, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts 02115, USA.,Department of Neurology, Harvard Medical School, Boston, Massachusetts 02115, USA
| | - Julian G Hurdle
- Center for Infectious and Inflammatory Diseases, Texas A &M Health Science Center, Houston, Texas 77030, USA
| | - David T Breault
- Division of Endocrinology, Boston Children's Hospital, Boston, Massachusetts 02115, USA.,Department of Pediatrics, Harvard Medical School, Boston, Massachusetts 02115, USA.,Harvard Stem Cell Institute, Cambridge, Massachusetts 02138, USA
| | - Abraham L Brass
- Department of Microbiology and Physiological Systems (MaPS), University of Massachusetts Medical School, Worcester, Massachusetts 01655, USA.,Gastroenterology Division, Department of Medicine, University of Massachusetts Medical School, Worcester, Massachusetts 01655, USA
| | - Min Dong
- Department of Urology, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts 02115, USA.,Department of Microbiology and Immunobiology and Department of Surgery, Harvard Medical School, Boston, Massachusetts 02115, USA
| |
Collapse
|
20
|
Nale JY, Chutia M, Carr P, Hickenbotham PT, Clokie MRJ. 'Get in Early'; Biofilm and Wax Moth (Galleria mellonella) Models Reveal New Insights into the Therapeutic Potential of Clostridium difficile Bacteriophages. Front Microbiol 2016; 7:1383. [PMID: 27630633 PMCID: PMC5005339 DOI: 10.3389/fmicb.2016.01383] [Citation(s) in RCA: 76] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2016] [Accepted: 08/22/2016] [Indexed: 12/19/2022] Open
Abstract
Clostridium difficile infection (CDI) is a global health threat associated with high rates of morbidity and mortality. Conventional antibiotic CDI therapy can result in treatment failure and recurrent infection. C. difficile produces biofilms which contribute to its virulence and impair antimicrobial activity. Some bacteriophages (phages) can penetrate biofilms and thus could be developed to either replace or supplement antibiotics. Here, we determined the impact of a previously optimized 4-phage cocktail on C. difficile ribotype 014/020 biofilms, and additionally as adjunct to vancomycin treatment in Galleria mellonella larva CDI model. The phages were applied before or after biofilm establishment in vitro, and the impact was analyzed according to turbidity, viability counts and topography as observed using scanning electron and confocal microscopy. The infectivity profiles and efficacies of orally administered phages and/or vancomycin were ascertained by monitoring colonization levels and larval survival rates. Phages prevented biofilm formation, and penetrated established biofilms. A single phage application reduced colonization causing extended longevity in the remedial treatment and prevented disease in the prophylaxis group. Multiple phage doses significantly improved the larval remedial regimen, and this treatment is comparable to vancomycin and the combined treatments. Taken together, our data suggest that the phages significantly reduce C. difficile biofilms, and prevent colonization in the G. mellonella model when used alone or in combination with vancomycin. The phages appear to be highly promising therapeutics in the targeted eradication of CDI and the use of these models has revealed that prophylactic use could be a propitious therapeutic option.
Collapse
Affiliation(s)
- Janet Y Nale
- Department of Infection, Immunity and Inflammation, University of Leicester Leicester, UK
| | - Mahananda Chutia
- Pathology and Microbiology Division, Central Muga Eri Research and Training Institute Assam, India
| | - Philippa Carr
- Department of Infection, Immunity and Inflammation, University of Leicester Leicester, UK
| | - Peter T Hickenbotham
- Department of Infection, Immunity and Inflammation, University of Leicester Leicester, UK
| | - Martha R J Clokie
- Department of Infection, Immunity and Inflammation, University of Leicester Leicester, UK
| |
Collapse
|
21
|
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: 162] [Impact Index Per Article: 18.0] [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
|
22
|
Defining the Roles of TcdA and TcdB in Localized Gastrointestinal Disease, Systemic Organ Damage, and the Host Response during Clostridium difficile Infections. mBio 2015; 6:e00551. [PMID: 26037121 PMCID: PMC4453007 DOI: 10.1128/mbio.00551-15] [Citation(s) in RCA: 183] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Clostridium difficile is a leading cause of antibiotic-associated diarrhea, a significant animal pathogen, and a worldwide public health burden. Most disease-causing strains secrete two exotoxins, TcdA and TcdB, which are considered to be the primary virulence factors. Understanding the role that these toxins play in disease is essential for the rational design of urgently needed new therapeutics. However, their relative contributions to disease remain contentious. Using three different animal models, we show that TcdA+ TcdB− mutants are attenuated in virulence in comparison to the wild-type (TcdA+ TcdB+) strain, whereas TcdA− TcdB+ mutants are fully virulent. We also show for the first time that TcdB alone is associated with both severe localized intestinal damage and systemic organ damage, suggesting that this toxin might be responsible for the onset of multiple organ dysfunction syndrome (MODS), a poorly characterized but often fatal complication of C. difficile infection (CDI). Finally, we show that TcdB is the primary factor responsible for inducing the in vivo host innate immune and inflammatory responses. Surprisingly, the animal infection model used was found to profoundly influence disease outcomes, a finding which has important ramifications for the validation of new therapeutics and future disease pathogenesis studies. Overall, our results show unequivocally that TcdB is the major virulence factor of C. difficile and provide new insights into the host response to C. difficile during infection. The results also highlight the critical nature of using appropriate and, when possible, multiple animal infection models when studying bacterial virulence mechanisms. Clostridium difficile is a leading cause of antibiotic-associated diarrhea and an important hospital pathogen. TcdA and TcdB are thought to be the primary virulence factors responsible for disease symptoms of C. difficile infections (CDI). However, the individual contributions of these toxins to disease remain contentious. Using three different animal models of infection, we show for the first time that TcdB alone causes severe damage to the gut, as well as systemic organ damage, suggesting that this toxin might be responsible for MODS, a serious but poorly understood complication of CDI. These findings provide important new insights into the host response to C. difficile during infection and should guide the rational development of urgently required nonantibiotic therapeutics for the treatment of CDI.
Collapse
|
23
|
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
|
24
|
Yuan P, Zhang H, Cai C, Zhu S, Zhou Y, Yang X, He R, Li C, Guo S, Li S, Huang T, Perez-Cordon G, Feng H, Wei W. Chondroitin sulfate proteoglycan 4 functions as the cellular receptor for Clostridium difficile toxin B. Cell Res 2014; 25:157-68. [PMID: 25547119 PMCID: PMC4650570 DOI: 10.1038/cr.2014.169] [Citation(s) in RCA: 134] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2014] [Revised: 11/06/2014] [Accepted: 11/19/2014] [Indexed: 02/06/2023] Open
Abstract
As a gram-positive, spore-forming anaerobic bacillus, Clostridium difficile (C. difficile) is responsible for severe and fatal pseudomembranous colitis, and poses the most urgent antibiotic resistance threat worldwide. Epidemic C. difficile is the leading cause of antibiotic-associated diarrhoea globally, especially diarrhoea due to the emergence of hypervirulent strains associated with high mortality and morbidity. TcdB, one of the key virulence factors secreted by this bacterium, enters host cells through a poorly understood mechanism to elicit its pathogenic effect. Here we report the first identification of the TcdB cellular receptor, chondroitin sulfate proteoglycan 4 (CSPG4). CSPG4 was initially isolated from a whole-genome human shRNAmir library screening, and its role was confirmed by both TALEN- and CRISPR/Cas9-mediated gene knockout in human cells. CSPG4 is critical for TcdB binding to the cell surface, inducing cytoskeleton disruption and cell death. A direct interaction between the N-terminus of CSPG4 and the C-terminus of TcdB was confirmed, and the soluble peptide of the toxin-binding domain of CSPG4 could protect cells from the action of TcdB. Notably, the complete loss of CSPG4/NG2 decreased TcdB-triggered interleukin-8 induction in mice without significantly affecting animal mortality. Based on both the in vitro and in vivo studies, we propose a dual-receptor model for TcdB endocytosis. The discovery of the first TcdB receptor reveals a previously unsuspected role for CSPG4 and provides a new therapeutic target for the treatment of C. difficile infection.
Collapse
Affiliation(s)
- Pengfei Yuan
- Biodynamic Optical Imaging Center (BIOPIC), State Key Laboratory of Protein and Plant Gene Research, School of Life Sciences, Peking University, Beijing 100871, China
| | - Hongmin Zhang
- Biodynamic Optical Imaging Center (BIOPIC), State Key Laboratory of Protein and Plant Gene Research, School of Life Sciences, Peking University, Beijing 100871, China
| | - Changzu Cai
- Biodynamic Optical Imaging Center (BIOPIC), State Key Laboratory of Protein and Plant Gene Research, School of Life Sciences, Peking University, Beijing 100871, China
| | - Shiyou Zhu
- Biodynamic Optical Imaging Center (BIOPIC), State Key Laboratory of Protein and Plant Gene Research, School of Life Sciences, Peking University, Beijing 100871, China
| | - Yuexin Zhou
- Biodynamic Optical Imaging Center (BIOPIC), State Key Laboratory of Protein and Plant Gene Research, School of Life Sciences, Peking University, Beijing 100871, China
| | - Xiaozhou Yang
- Biodynamic Optical Imaging Center (BIOPIC), State Key Laboratory of Protein and Plant Gene Research, School of Life Sciences, Peking University, Beijing 100871, China
| | - Ruina He
- Biodynamic Optical Imaging Center (BIOPIC), State Key Laboratory of Protein and Plant Gene Research, School of Life Sciences, Peking University, Beijing 100871, China
| | - Chan Li
- Biodynamic Optical Imaging Center (BIOPIC), State Key Laboratory of Protein and Plant Gene Research, School of Life Sciences, Peking University, Beijing 100871, China
| | - Shengjie Guo
- Biodynamic Optical Imaging Center (BIOPIC), State Key Laboratory of Protein and Plant Gene Research, School of Life Sciences, Peking University, Beijing 100871, China
| | - Shan Li
- School of Bioscience and Biotechnology, South China University of Technology, Guangzhou, Guangdong 510006, China
| | - Tuxiong Huang
- Department of Microbial Pathogenesis, University of Maryland Dental School, Baltimore, Maryland 21201, USA
| | - Gregorio Perez-Cordon
- Department of Microbial Pathogenesis, University of Maryland Dental School, Baltimore, Maryland 21201, USA
| | - Hanping Feng
- Department of Microbial Pathogenesis, University of Maryland Dental School, Baltimore, Maryland 21201, USA
| | - Wensheng Wei
- Biodynamic Optical Imaging Center (BIOPIC), State Key Laboratory of Protein and Plant Gene Research, School of Life Sciences, Peking University, Beijing 100871, China
| |
Collapse
|
25
|
Toxin-mediated paracellular transport of antitoxin antibodies facilitates protection against Clostridium difficile infection. Infect Immun 2014; 83:405-16. [PMID: 25385797 DOI: 10.1128/iai.02550-14] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
The exotoxins TcdA and TcdB are the major virulence factors of Clostridium difficile. Circulating neutralizing antitoxin antibodies are protective in C. difficile infection (CDI), as demonstrated, in part, by the protective effects of actoxumab and bezlotoxumab, which bind to and neutralize TcdA and TcdB, respectively. The question of how systemic IgG antibodies neutralize toxins in the gut lumen remains unresolved, although it has been suggested that the Fc receptor FcRn may be involved in active antibody transport across the gut epithelium. In this study, we demonstrated that genetic ablation of FcRn and excess irrelevant human IgG have no impact on actoxumab-bezlotoxumab-mediated protection in murine and hamster models of CDI, suggesting that Fc-dependent transport of antibodies across the gut wall is not required for efficacy. Tissue distribution studies in hamsters suggest, rather, that the transport of antibodies depends on toxin-induced damage to the gut lining. In an in vitro two-dimensional culture system that mimics the architecture of the intestinal mucosal epithelium, toxins on the apical side of epithelial cell monolayers are neutralized by basolateral antibodies, and antibody transport across the cell layer is dramatically increased upon addition of toxin to the apical side. Similar data were obtained with F(ab')2 fragments, which lack an Fc domain, consistent with FcRn-independent paracellular, rather than transcellular, transport of antibodies. Kinetic studies show that initial damage caused by apical toxin is required for efficient neutralization by basolateral antibodies. These data may represent a general mechanism of humoral response-mediated protection against enteric pathogens.
Collapse
|
26
|
Awad MM, Johanesen PA, Carter GP, Rose E, Lyras D. Clostridium difficile virulence factors: Insights into an anaerobic spore-forming pathogen. Gut Microbes 2014; 5:579-93. [PMID: 25483328 PMCID: PMC4615314 DOI: 10.4161/19490976.2014.969632] [Citation(s) in RCA: 106] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Abstract
The worldwide emergence of epidemic strains of Clostridium difficile linked to increased disease severity and mortality has resulted in greater research efforts toward determining the virulence factors and pathogenesis mechanisms used by this organism to cause disease. C. difficile is an opportunist pathogen that employs many factors to infect and damage the host, often with devastating consequences. This review will focus on the role of the 2 major virulence factors, toxin A (TcdA) and toxin B (TcdB), as well as the role of other putative virulence factors, such as binary toxin, in C. difficile-mediated infection. Consideration is given to the importance of spores in both the initiation of disease and disease recurrence and also to the role that surface proteins play in host interactions.
Collapse
Key Words
- AAD, antibiotic associated diarrhea
- C. difficile,Clostridium difficile
- CDI, C. difficile infection
- CDT, Clostridium difficile transferase
- CDTLoc, CDT locus
- CDTa, CDT enzymatic component
- CDTb, CDT binding/translocation component
- CST, Clostridium spiroforme toxin
- CWPs, cell wall protein
- Clostridium
- ECF, extracytoplasmic function
- HMW, high molecular weight
- LMW, low molecular weight
- LSR, lipolysis-stimulated lipoprotein receptor
- PCR, polymerase chain reaction
- PFGE, pulsed field gel electrophoresis
- PaLoc, pathogenicity locus
- REA, restriction endonuclease analysis
- S-layer, surface layer
- SLPs, S-layer proteins
- TcdA, toxin A
- TcdB, toxin B
- antibiotic
- colitis
- difficile
- infection
- nosocomial
- toxin
- virulence factor
- ι-toxin, iota toxin
Collapse
Affiliation(s)
- Milena M Awad
- Department of Microbiology; Monash University; Clayton, Victoria, Australia
| | | | - Glen P Carter
- Department of Microbiology; Monash University; Clayton, Victoria, Australia
| | - Edward Rose
- Department of Microbiology; Monash University; Clayton, Victoria, Australia
| | - Dena Lyras
- Department of Microbiology; Monash University; Clayton, Victoria, Australia,Correspondence to: Dena Lyras;
| |
Collapse
|
27
|
Rineh A, Kelso MJ, Vatansever F, Tegos GP, Hamblin MR. Clostridium difficile infection: molecular pathogenesis and novel therapeutics. Expert Rev Anti Infect Ther 2014; 12:131-50. [PMID: 24410618 DOI: 10.1586/14787210.2014.866515] [Citation(s) in RCA: 65] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
The Gram-positive anaerobic bacterium Clostridium difficile produces toxins A and B, which can cause a spectrum of diseases from pseudomembranous colitis to C. difficile-associated diarrhea. A limited number of C. difficile strains also produce a binary toxin that exhibits ADP ribosyltransferase activity. Here, the structure and the mechanism of action of these toxins as well as their role in disease are reviewed. Nosocomial C. difficile infection is often contracted in hospital when patients treated with antibiotics suffer a disturbance in normal gut microflora. C. difficile spores can persist on dry, inanimate surface for months. Metronidazole and oral vancomycin are clinically used for treatment of C. difficile infection but clinical failure and concern about promotion of resistance are motivating the search for novel non-antibiotic therapeutics. Methods for controlling both toxins and spores, replacing gut microflora by probiotics or fecal transplant, and killing bacteria in the anaerobic gut by photodynamic therapy are discussed.
Collapse
Affiliation(s)
- Ardeshir Rineh
- The Wellman Center for Photomedicine, Massachusetts General Hospital, Boston, MA 02114, USA
| | | | | | | | | |
Collapse
|
28
|
An optimized, synthetic DNA vaccine encoding the toxin A and toxin B receptor binding domains of Clostridium difficile induces protective antibody responses in vivo. Infect Immun 2014; 82:4080-91. [PMID: 25024365 DOI: 10.1128/iai.01950-14] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Clostridium difficile-associated disease (CDAD) constitutes a large majority of nosocomial diarrhea cases in industrialized nations and is mediated by the effects of two secreted toxins, toxin A (TcdA) and toxin B (TcdB). Patients who develop strong antitoxin antibody responses can clear C. difficile infection and remain disease free. Key toxin-neutralizing epitopes have been found within the carboxy-terminal receptor binding domains (RBDs) of TcdA and TcdB, which has generated interest in developing the RBD as a viable vaccine target. While numerous platforms have been studied, very little data describes the potential of DNA vaccination against CDAD. Therefore, we created highly optimized plasmids encoding the RBDs from TcdA and TcdB in which any putative N-linked glycosylation sites were altered. Mice and nonhuman primates were immunized intramuscularly, followed by in vivo electroporation, and in these animal models, vaccination induced significant levels of both anti-RBD antibodies (blood and stool) and RBD-specific antibody-secreting cells. Further characterization revealed that sera from immunized mice and nonhuman primates could detect RBD protein from transfected cells, as well as neutralize purified toxins in an in vitro cytotoxicity assay. Mice that were immunized with plasmids or given nonhuman-primate sera were protected from a lethal challenge with purified TcdA and/or TcdB. Moreover, immunized mice were significantly protected when challenged with C. difficile spores from homologous (VPI 10463) and heterologous, epidemic (UK1) strains. These data demonstrate the robust immunogenicity and efficacy of a TcdA/B RBD-based DNA vaccine in preclinical models of acute toxin-associated and intragastric, spore-induced colonic disease.
Collapse
|
29
|
Variations in virulence and molecular biology among emerging strains of Clostridium difficile. Microbiol Mol Biol Rev 2014; 77:567-81. [PMID: 24296572 DOI: 10.1128/mmbr.00017-13] [Citation(s) in RCA: 80] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Clostridium difficile is a Gram-positive, spore-forming organism which infects and colonizes the large intestine, produces potent toxins, triggers inflammation, and causes significant systemic complications. Treating C. difficile infection (CDI) has always been difficult, because the disease is both caused and resolved by antibiotic treatment. For three and a half decades, C. difficile has presented a treatment challenge to clinicians, and the situation took a turn for the worse about 10 years ago. An increase in epidemic outbreaks related to CDI was first noticed around 2003, and these outbreaks correlated with a sudden increase in the mortality rate of this illness. Further studies discovered that these changes in CDI epidemiology were associated with the rapid emergence of hypervirulent strains of C. difficile, now collectively referred to as NAP1/BI/027 strains. The discovery of new epidemic strains of C. difficile has provided a unique opportunity for retrospective and prospective studies that have sought to understand how these strains have essentially replaced more historical strains as a major cause of CDI. Moreover, detailed studies on the pathogenesis of NAP1/BI/027 strains are leading to new hypotheses on how this emerging strain causes severe disease and is more commonly associated with epidemics. In this review, we provide an overview of CDI, discuss critical mechanisms of C. difficile virulence, and explain how differences in virulence-associated factors between historical and newly emerging strains might explain the hypervirulence exhibited by this pathogen during the past decade.
Collapse
|
30
|
Britton RA, Young VB. Role of the intestinal microbiota in resistance to colonization by Clostridium difficile. Gastroenterology 2014; 146:1547-53. [PMID: 24503131 PMCID: PMC3995857 DOI: 10.1053/j.gastro.2014.01.059] [Citation(s) in RCA: 322] [Impact Index Per Article: 29.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/14/2013] [Revised: 01/16/2014] [Accepted: 01/30/2014] [Indexed: 12/18/2022]
Abstract
Antibiotic-associated infection with the bacterial pathogen Clostridium difficile is a major cause of morbidity and increased health care costs. C difficile infection follows disruption of the indigenous gut microbiota by antibiotics. Antibiotics create an environment within the intestine that promotes C difficile spore germination, vegetative growth, and toxin production, leading to epithelial damage and colitis. Studies of patients with C difficile infection and animal models have shown that the indigenous microbiota can inhibit expansion and persistence of C difficile. Although the specific mechanisms of these processes are not known, they are likely to interfere with key aspects of the pathogen's physiology, including spore germination and competitive growth. Increasing our understanding of how the intestinal microbiota manage C difficile could lead to better means of controlling this important nosocomial pathogen.
Collapse
Affiliation(s)
- Robert A. Britton
- Department of Microbiology and Molecular Genetics, Michigan State University, East Lansing, Michigan 48824
| | - Vincent B. Young
- Division of Infectious Diseases, Department of Internal Medicine, University of Michigan, Ann Arbor, MI, 48109,Department of Microbiology and Immunology, University of Michigan, Ann Arbor, MI, 48109
| |
Collapse
|
31
|
|
32
|
Spencer J, Leuzzi R, Buckley A, Irvine J, Candlish D, Scarselli M, Douce GR. Vaccination against Clostridium difficile using toxin fragments: Observations and analysis in animal models. Gut Microbes 2014; 5:225-32. [PMID: 24637800 PMCID: PMC4063849 DOI: 10.4161/gmic.27712] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Abstract
Clostridium difficile is a major cause of antibiotic associated diarrhea. Recently, we have shown that effective protection can be mediated in hamsters through the inclusion of specific recombinant fragments from toxin A and B in a systemically delivered vaccine. Interestingly while neutralizing antibodies to the binding domains of both toxin A and B are moderately protective, enhanced survival is observed when fragments from the glucosyltransferase region of toxin B replace those from the binding domain of this toxin. In this addendum, we discuss additional information that has been derived from such vaccination studies. This includes observations on efficacy and cross-protection against different ribotypes mediated by these vaccines and the challenges that remain for a vaccine which prevents clinical symptoms but not colonization. The use and value of vaccination both in the prevention of infection and for treatment of disease relapse will be discussed.
Collapse
Affiliation(s)
- Janice Spencer
- Institute of Infection, Immunity, and Inflammation; College of Medicine, Veterinary and Life Sciences; University of Glasgow; Glasgow, UK
| | | | - Anthony Buckley
- Institute of Infection, Immunity, and Inflammation; College of Medicine, Veterinary and Life Sciences; University of Glasgow; Glasgow, UK
| | - June Irvine
- Institute of Infection, Immunity, and Inflammation; College of Medicine, Veterinary and Life Sciences; University of Glasgow; Glasgow, UK
| | - Denise Candlish
- Institute of Infection, Immunity, and Inflammation; College of Medicine, Veterinary and Life Sciences; University of Glasgow; Glasgow, UK
| | | | - Gillian R Douce
- Institute of Infection, Immunity, and Inflammation; College of Medicine, Veterinary and Life Sciences; University of Glasgow; Glasgow, UK,Correspondence to: Gillian R Douce,
| |
Collapse
|
33
|
Robertson JK, Danzmann K, Charles S, Blake K, Olivares A, Bamikole S, Olson M, Raay TJV. Targeting the Wnt pathway in zebrafish as a screening method to identify novel therapeutic compounds. Exp Biol Med (Maywood) 2014; 239:169-76. [DOI: 10.1177/1535370213514322] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Activating mutations in the Wnt signaling pathway account for the initiation of greater than 90% of all colorectal cancers and this pathway has been implicated in numerous other diseases. Therefore, identifying small molecule inhibitors of this pathway is of critical importance towards identifying clinically relevant drugs. Numerous screens have been employed to identify therapeutic reagents, but none have made it to advanced clinical trials, suggesting that traditional screening methods are ineffective at identifying clinically relevant targets. Here, we describe a novel in vivo screen to identify small molecule inhibitors of the Wnt pathway. Specifically, treatment of zebrafish embryos with LiCl inhibits GSK3 kinase function, resulting in hyperactivation of the signaling pathway and an eyeless phenotype at 1 day post fertilization. Using the small molecule XAV939, a known inhibitor of Wnt signaling, we rescued the LiCl induced eyeless phenotype, confirming efficacy of the screen. We next tested our assay with 400 known small molecule kinase inhibitors, none of which should inhibit Wnt signaling below the level of GSK3 based on their known targets. Accordingly, none of these small molecules rescued the eyeless phenotype, which demonstrates the stringency of the assay. However, several of these small molecule kinase inhibitors did generate a non-Wnt phenotype in accordance with the kinase they targeted. Therefore, combining the efficacy, sensitivity, and stringency of this preliminary screen, this model will provide an alternative to the traditional in vitro screen, generating potentially clinical relevant drugs in a rapid and cost-effective way.
Collapse
Affiliation(s)
- Joshua K Robertson
- Department of Molecular and Cellular Biology, University of Guelph, Guelph, ON Canada N1L 1A3
| | - Kestral Danzmann
- Department of Molecular and Cellular Biology, University of Guelph, Guelph, ON Canada N1L 1A3
| | - Sherise Charles
- Department of Molecular and Cellular Biology, University of Guelph, Guelph, ON Canada N1L 1A3
| | - Katherine Blake
- Department of Molecular and Cellular Biology, University of Guelph, Guelph, ON Canada N1L 1A3
| | - Annia Olivares
- Department of Molecular and Cellular Biology, University of Guelph, Guelph, ON Canada N1L 1A3
| | - Solape Bamikole
- Department of Molecular and Cellular Biology, University of Guelph, Guelph, ON Canada N1L 1A3
| | - Meghan Olson
- Department of Molecular and Cellular Biology, University of Guelph, Guelph, ON Canada N1L 1A3
| | - Terence J Van Raay
- Department of Molecular and Cellular Biology, University of Guelph, Guelph, ON Canada N1L 1A3
| |
Collapse
|
34
|
Belmares J, Gerding DN, Tillotson G, Johnson S. Measuring the severity ofClostridium difficileinfection: implications for management and drug development. Expert Rev Anti Infect Ther 2014; 6:897-908. [DOI: 10.1586/14787210.6.6.897] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
|
35
|
Hutton ML, Mackin KE, Chakravorty A, Lyras D. Small animal models for the study of Clostridium difficile disease pathogenesis. FEMS Microbiol Lett 2014; 352:140-9. [PMID: 24372713 DOI: 10.1111/1574-6968.12367] [Citation(s) in RCA: 59] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2013] [Revised: 12/16/2013] [Accepted: 12/16/2013] [Indexed: 01/05/2023] Open
Abstract
Clostridium difficile is the leading cause of bacterial antibiotic-associated diarrhoea in hospitals in the developed world. Despite this notoriety, the complex mechanisms employed by this pathogen to overcome innate host defences and induce fulminant disease are poorly understood. Various animal models have been used extensively for C. difficile research to study disease pathogenesis. Until recently, the most commonly used C. difficile disease model has utilised hamsters; however, mouse and pig models have now been developed that unravel different aspects of C. difficile pathology. This review summarises key aspects of the small animal models currently used in C. difficile studies with a specific focus on major differences between them. Furthermore, this review highlights the advantages and disadvantages of each model and illustrates that careful consideration is required when selecting models for use in C. difficile research.
Collapse
Affiliation(s)
- Melanie L Hutton
- Department of Microbiology, Monash University, Clayton, Victoria, Australia
| | | | | | | |
Collapse
|
36
|
Steele J, Parry N, Tzipori S. The roles of toxin A and toxin B in Clostridium difficile infection: insights from the gnotobiotic piglet model. Gut Microbes 2014; 5:53-7. [PMID: 24394234 PMCID: PMC4049938 DOI: 10.4161/gmic.26855] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Abstract
We recently published our findings indicating that anti-TcdB antibodies were effective as treatment for C. difficile infection, but that anti-TcdA actually worsened prognosis in the gnotobiotic piglet model. To further investigate the roles of the two toxins, we administered purified toxins separately or together, systemically, to piglets and found that both toxins, either alone or together, are able to elicit severe lesions systemically and are also able to cross into the gut lumen and cause large intestinal lesions typical of infection. We also found that anti-TcdA administered before systemic challenge with TcdA again did not protect from development of disease, but, in this case, did not appear to worsen prognosis. Further work is still needed, but these findings add to the growing knowledge regarding the roles of the C. difficile toxins.
Collapse
Affiliation(s)
- Jennifer Steele
- Tufts Cummings School of Veterinary Medicine; Department of Infectious Disease and Global Health; North Grafton, MA USA
| | - Nicola Parry
- Massachussetts Institute of Technology; Division of Comparative Medicine; Cambridge, MA USA
| | - Saul Tzipori
- Tufts Cummings School of Veterinary Medicine; Department of Infectious Disease and Global Health; North Grafton, MA USA,Correspondence to: Saul Tzipori,
| |
Collapse
|
37
|
Lanis JM, Heinlen LD, James JA, Ballard JD. Clostridium difficile 027/BI/NAP1 encodes a hypertoxic and antigenically variable form of TcdB. PLoS Pathog 2013; 9:e1003523. [PMID: 23935501 PMCID: PMC3731247 DOI: 10.1371/journal.ppat.1003523] [Citation(s) in RCA: 67] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2013] [Accepted: 06/11/2013] [Indexed: 12/18/2022] Open
Abstract
The Clostridium difficile exotoxin, TcdB, which is a major virulence factor, varies between strains of this pathogen. Herein, we show that TcdB from the epidemic BI/NAP1/027 strain of C. difficile is more lethal, causes more extensive brain hemorrhage, and is antigenically variable from TcdB produced by previously studied strains of this pathogen (TcdB003). In mouse intoxication assays, TcdB from a ribotype 027 strain (TcdB027) was at least four fold more lethal than TcdB003. TcdB027 caused a previously undescribed brain hemorrhage in mice and this correlated with a heightened sensitivity of brain microvascular endothelial cells to the toxin. TcdB003 and TcdB027 also differed in their antigenic profiles and did not share cross-neutralizing epitopes in a major immunogenic region of the protein. Solid phase humoral mapping of epitopes in the carboxy-terminal domains (CTD) of TcdB027 and TcdB003 identified 11 reactive epitopes that varied between the two forms of TcdB, and 13 epitopes that were shared or overlapping. Despite the epitope differences and absence of neutralizing epitopes in the CTD of TcdB027, a toxoid form of this toxin primed a strong protective response. These findings indicate TcdB027 is a more potent toxin than TcdB003 as measured by lethality assays and pathology, moreover the sequence differences between the two forms of TcdB alter antigenic epitopes and reduce cross-neutralization by antibodies targeting the CTD. During the past decade, the C. difficile BI/NAP1/027 strain has emerged and in some settings predominated as the cause of C. difficile infection. Moreover, in some reports C. difficile BI/NAP1/027 has been associated with more severe disease. The reasons for association of this strain with more severe disease and relapse are poorly understood. We compared the toxicity and antigenic profiles of the major C. difficile virulence factor, TcdB, from a previously studied reference strain and a BI/NAP1/027 strain. The results indicate TcdB027, the toxin from the BI/NAP1/027 strain, is more lethal and causes more extensive brain hemorrhaging than TcdB003, the toxin produced by a reference strain of C. difficile. Furthermore, the results show that the antigenic carboxy-terminal domain (CTD) encodes at least 11 epitopes that differ between the two forms of TcdB. In line with this, experiments demonstrate that antiserum against the CTD does not cross-neutralize TcdB003 and TcdB027 toxicity against CHO cells, and TcdB027 appears to be devoid of neutralizing epitopes in this domain. These findings indicate differences in TcdB003 and TcdB027 contribute to increased virulence of C. difficile BI/NAP1/027 and reduce the likelihood of acquired immunity providing cross-protection against infection by these strains.
Collapse
Affiliation(s)
- Jordi M. Lanis
- Department of Microbiology and Immunology, The University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, United States of America
| | - Latisha D. Heinlen
- Department of Microbiology and Immunology, The University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, United States of America
- Department of Medicine, The University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, United States of America
| | - Judith A. James
- Department of Microbiology and Immunology, The University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, United States of America
- Department of Medicine, The University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, United States of America
- Arthritis and Clinical Immunology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, Oklahoma, United States of America
| | - Jimmy D. Ballard
- Department of Microbiology and Immunology, The University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, United States of America
- * E-mail:
| |
Collapse
|
38
|
Clostridium difficile in foods and animals: history and measures to reduce exposure. Anim Health Res Rev 2013; 14:11-29. [PMID: 23324529 DOI: 10.1017/s1466252312000229] [Citation(s) in RCA: 66] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Many articles have summarized the changing epidemiology of Clostridium difficile infections (CDI) in humans, but the emerging presence of C. difficile in foods and animals and possible measures to reduce human exposure to this important pathogen have been infrequently addressed. CDIs have traditionally been assumed to be restricted to health-care settings. However, recent molecular studies indicate that this is no longer the case; animals and foods might be involved in the changing epidemiology of CDIs in humans; and genome sequencing is disproving person-to-person transmission in hospitals. Although zoonotic and foodborne transmission have not been confirmed, it is evident that susceptible people can be inadvertently exposed to C. difficile from foods, animals, or their environment. Strains of epidemic clones present in humans are common in companion and food animals, raw meats, poultry products, vegetables, and ready-to-eat foods, including salads. In order to develop science-based prevention strategies, it is critical to understand how C. difficile reaches foods and humans. This review contextualizes the current understanding of CDIs in humans, animals, and foods. Based on available information, we propose a list of educational measures that could reduce the exposure of susceptible people to C. difficile. Enhanced educational efforts and behavior change targeting medical and non-medical personnel are needed.
Collapse
|
39
|
Steele J, Mukherjee J, Parry N, Tzipori S. Antibody against TcdB, but not TcdA, prevents development of gastrointestinal and systemic Clostridium difficile disease. J Infect Dis 2012; 207:323-30. [PMID: 23125448 DOI: 10.1093/infdis/jis669] [Citation(s) in RCA: 92] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND A dramatic increase in morbidity and mortality from Clostridium difficile infection (CDI) due to the recent emergence of virulent, antibiotic-resistant strains has led to a search for alternatives to antibiotics, including vaccines and immune-based therapy that target the 2 key toxins-TcdA and TcdB. METHODS We investigated the efficacy of specific human monoclonal antibodies (HuMab) and alpaca polyclonal antibodies against each toxin separately and in combination in the gnotobiotic piglet model of CDI. Additionally, the HuMab and polyclonal antibodies were exploited to investigate the precise contribution of each toxin to systemic and/or gastrointestinal (GI) tract disease. RESULTS Our results indicate that TcdB is an important virulence factor associated with GI and systemic pathology. Administration of anti-TcdB antibody alone or with anti-TcdA protected 100% of piglets from development of systemic CDI and minimized GI lesions. Conversely, 100% of piglets administered only anti-TcdA developed severe GI and systemic disease, with 67%-83% fatality, faring worse than placebo-treated control animals. CONCLUSIONS These results highlight the importance of TcdB in the pathogenesis of CDI and the effectiveness of TcdB-specific antibody in treating CDI. However, the results raise new questions regarding the nature of TcdA interaction with therapeutic antibodies.
Collapse
Affiliation(s)
- Jennifer Steele
- Tufts Cummings School of Veterinary Medicine, Department of Biomedical Sciences, Division of Infectious Diseases, North Grafton, MA 01536, USA
| | | | | | | |
Collapse
|
40
|
Badger VO, Ledeboer NA, Graham MB, Edmiston CE. Clostridium difficile: epidemiology, pathogenesis, management, and prevention of a recalcitrant healthcare-associated pathogen. JPEN J Parenter Enteral Nutr 2012; 36:645-62. [PMID: 22577120 DOI: 10.1177/0148607112446703] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Clostridium difficile is the leading cause of healthcare-associated infectious diarrhea. Although C difficile is part of normal flora in some healthy individuals, patients with selective risk factors are often vulnerable to the toxigenic potential of this virulent healthcare pathogen. The spectrum of C difficile infection (CDI) is highly variable, ranging from mild to severe illness, presenting with single to multiple disease recurrences. Current approaches to treatment are based on severity of illness, number of recurrences, and clinical presentation. Oral vancomycin and metronidazole have formed the foundation for treatment of CDI, but therapeutic failures are commonly reported, especially involving hypervirulent clones. Alternative therapies, including newer antimicrobials, probiotics, immunotherapy, and fecal transplantation, have all met with varying degrees of efficacy. Although toxigenic culture (TC) testing from anaerobic culture remains the gold standard, newer technologies, including enzyme immunoassay, common antigen (glutamate dehydrogenase) testing, and real-time polymerase chain reaction (PCR) are less time-consuming and rapid. However, TC and PCR have reported high specificity and sensitivity when compared with other laboratory tests. Because of the significant morbidity and mortality associated with CDI, a high index of suspicion is warranted. Prevention and eradication of CDI require a multidisciplinary approach, including early disease recognition through appropriate surveillance, implementation of effective contact isolation strategies, adherence to environmental controls, judicious hand hygiene, evidence-based treatment, and management that includes antibiotic stewardship, continuous education of healthcare workers, and administrative support.
Collapse
Affiliation(s)
- Victor O Badger
- Department of Internal Medicine, Division of Infection Diseases, Medical College of Wisconsin, Milwaukee, Wisconsin 53226, USA
| | | | | | | |
Collapse
|
41
|
Landrum PF, Chapman PM, Neff J, Page DS. Evaluating the aquatic toxicity of complex organic chemical mixtures: lessons learned from polycyclic aromatic hydrocarbon and petroleum hydrocarbon case studies. INTEGRATED ENVIRONMENTAL ASSESSMENT AND MANAGEMENT 2012; 8:217-230. [PMID: 21913322 DOI: 10.1002/ieam.277] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/23/2011] [Revised: 07/15/2011] [Accepted: 09/04/2011] [Indexed: 05/31/2023]
Abstract
Experimental designs for evaluating complex mixture toxicity in aquatic environments can be highly variable and, if not appropriate, can produce and have produced data that are difficult or impossible to interpret accurately. We build on and synthesize recent critical reviews of mixture toxicity using lessons learned from 4 case studies, ranging from binary to more complex mixtures of primarily polycyclic aromatic hydrocarbons and petroleum hydrocarbons, to provide guidance for evaluating the aquatic toxicity of complex mixtures of organic chemicals. Two fundamental requirements include establishing a dose-response relationship and determining the causative agent (or agents) of any observed toxicity. Meeting these 2 requirements involves ensuring appropriate exposure conditions and measurement endpoints, considering modifying factors (e.g., test conditions, test organism life stages and feeding behavior, chemical transformations, mixture dilutions, sorbing phases), and correctly interpreting dose-response relationships. Specific recommendations are provided.
Collapse
|
42
|
Page DS, Chapman PM, Landrum PF, Neff J, Elston R. A Perspective on the Toxicity of Low Concentrations of Petroleum-Derived Polycyclic Aromatic Hydrocarbons to Early Life Stages of Herring and Salmon. HUMAN AND ECOLOGICAL RISK ASSESSMENT : HERA 2012; 18:229-260. [PMID: 22754275 PMCID: PMC3379735 DOI: 10.1080/10807039.2012.650569] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/07/2023]
Abstract
This article presents a critical review of two groups of studies that reported adverse effects to salmon and herring eggs and fry from exposure to 1 μg/L or less of aqueous total polycyclic aromatic hydrocarbons (TPAH), as weathered oil, and a more toxic aqueous extract of "very weathered oil." Exposure media were prepared by continuously flowing water up through vertical columns containing gravel oiled at different concentrations of Prudhoe Bay crude oil. Uncontrolled variables associated with the use of the oiled gravel columns included time- and treatment-dependent variations in the PAH concentration and composition in the exposure water, unexplored toxicity from other oil constituents/degradation products, potential toxicity from bacterial and fungal activity, oil droplets as a potential contaminant source, inherent differences between control and exposed embryo populations, and water flow rate differences. Based on a review of the evidence from published project reports, peer-reviewed publications, chemistry data in a public database, and unpublished reports and laboratory records, the reviewed studies did not establish consistent dose (concentration) response or causality and thus do not demonstrate that dissolved PAH alone from the weathered oil resulted in the claimed effects on fish embryos at low μg/L TPAH concentrations. Accordingly, these studies should not be relied on for management decision-making, when assessing the risk of very low-level PAH exposures to early life stages of fish.
Collapse
Affiliation(s)
- David S. Page
- Bowdoin College, Chemistry Department, Brunswick, ME, USA
- Address correspondence to David S. Page, Bowdoin College Chemistry Department, 6600 College Station, Brunswick, ME 04011-8466, USA. E-mail:
| | | | - Peter F. Landrum
- Southern Illinois University, Carbondale, Department of Zoology, Carbondale, IL, USA
| | | | - Ralph Elston
- Ralph Elston, Aquatechnics Inc., Carlsborg, WA, USA
| |
Collapse
|
43
|
Abstract
Models of Clostridium difficile infection (C. difficile) have been used extensively for Clostridium difficile (C. difficile) research. The hamster model of C. difficile infection has been most extensively employed for the study of C. difficile and this has been used in many different areas of research, including the induction of C. difficile, the testing of new treatments, population dynamics and characterization of virulence. Investigations using in vitro models for C. difficile introduced the concept of colonization resistance, evaluated the role of antibiotics in C. difficile development, explored population dynamics and have been useful in the evaluation of C. difficile treatments. Experiments using models have major advantages over clinical studies and have been indispensible in furthering C. difficile research. It is important for future study programs to carefully consider the approach to use and therefore be better placed to inform the design and interpretation of clinical studies.
Collapse
Affiliation(s)
- Emma L. Best
- Leeds Teaching Hospitals NHS Trust; Microbiology Department; Old Medical School; Leeds General Infirmary; Leeds, UK,Correspondence to: Emma L. Best,
| | - Jane Freeman
- Leeds Teaching Hospitals NHS Trust; Microbiology Department; Old Medical School; Leeds General Infirmary; Leeds, UK
| | - Mark H. Wilcox
- Leeds Teaching Hospitals NHS Trust; Microbiology Department; Old Medical School; Leeds General Infirmary; Leeds, UK,University of Leeds; Leeds, UK
| |
Collapse
|
44
|
Lanis JM, Hightower LD, Shen A, Ballard JD. TcdB from hypervirulent Clostridium difficile exhibits increased efficiency of autoprocessing. Mol Microbiol 2012; 84:66-76. [PMID: 22372854 DOI: 10.1111/j.1365-2958.2012.08009.x] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
TcdB, an intracellular bacterial toxin that inactivates small GTPases, is a major Clostridium difficile virulence factor. Recent studies have found that TcdB produced by emerging/hypervirulent strains of C. difficile is more potent than TcdB from historical strains, and in the current work, studies were performed to investigate the underlying mechanisms for this change in TcdB toxicity. Using a series of biochemical analyses we found that TcdB from a hypervirulent strain (TcdB(HV) ) was more efficient at autoprocessing than TcdB from a historical strain (TcdB(HIST) ). TcdB(HV) and TcdB(HIST) were activated by similar concentrations of IP6; however, the overall efficiency of processing was 20% higher for TcdB(HV) . Using an activity-based fluorescent probe (AWP19) an intermediate, activated but uncleaved, form of TcdB(HIST) was identified, while only a processed form of TcdB(HV) could be detected under the same conditions. Using a much higher concentration (200 µM) of the probe revealed an activated uncleaved form of TcdB(HV) , indicating a preferential and more efficient engagement of intramolecular substrate than TcdB(HIST) . Furthermore, a peptide-based inhibitor (Ac-GSL-AOMK) was found to block the cytotoxicity of TcdB(HIST) at a lower concentration than required to inhibit TcdB(HV) . These findings suggest that TcdB(HV) may cause increased cytotoxicity due to more efficient autoprocessing.
Collapse
Affiliation(s)
- Jordi M Lanis
- Department of Microbiology and Immunology, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| | | | | | | |
Collapse
|
45
|
Bobo LD, Dubberke ER, Kollef M. Clostridium difficile in the ICU: the struggle continues. Chest 2012; 140:1643-1653. [PMID: 22147824 DOI: 10.1378/chest.11-0556] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Clostridium difficile infection (CDI) management has become more daunting over the past decade because of alarming increases in CDI incidence and severity both in the hospital and in the community. This increase has concomitantly caused significant escalation of the health-care economic burden caused by CDI, and it will likely be translated to increased ICU admission and attributable mortality. Some possible causes for difficulty in management of CDI are as follows: (1) inability to predict and prevent development of severe/complicated or relapsing CDI in patients who initially present with mild symptoms; (2) lack of a method to determine who would have benefited a priori from initiating vancomycin treatment first instead of treatment with metronidazole; (3) lack of sensitive and specific CDI diagnostics; (4) changing epidemiology of CDI, including the emergence of a hypervirulent, epidemic C difficile strain associated with increased morbidity and mortality; (5) association of certain high-usage nonantimicrobial medications with CDI; and (6) lack of treatment regimens that leave the normal intestinal flora undisturbed while treating the primary infection. The objective of this article is to present current management and prevention guidelines for CDI based on recommendations by the Society for Healthcare Epidemiology of America and Infectious Diseases Society of America and potential new clinical management strategies on the horizon.
Collapse
Affiliation(s)
- Linda D Bobo
- Adult Infectious Diseases Division, Washington University School of Medicine, St. Louis, MO.
| | - Erik R Dubberke
- Adult Infectious Diseases Division, Washington University School of Medicine, St. Louis, MO
| | - Marin Kollef
- Pulmonary and Critical Care Division, Washington University School of Medicine, St. Louis, MO
| |
Collapse
|
46
|
Shen A. Clostridium difficile toxins: mediators of inflammation. J Innate Immun 2012; 4:149-58. [PMID: 22237401 DOI: 10.1159/000332946] [Citation(s) in RCA: 131] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2011] [Accepted: 09/06/2011] [Indexed: 12/18/2022] Open
Abstract
Clostridium difficile is a significant problem in hospital settings as the most common cause of nosocomial diarrhea worldwide. C. difficile infections (CDIs) are characterized by an acute intestinal inflammatory response with neutrophil infiltration. These symptoms are primarily caused by the glucosylating toxins, TcdA and TcdB. In the past decade, the frequency and severity of CDIs have increased markedly due to the emergence of so-called hypervirulent strains that overproduce cytotoxic glucosylating toxins relative to historical strains. In addition, these strains produce a third toxin, binary toxin or C. difficile transferase (CDT), that may contribute to hypervirulence. Both the glucosylating toxins and CDT covalently modify target cell proteins to cause disassembly of the actin cytoskeleton and induce severe inflammation. This review summarizes our current knowledge of the mechanisms by which glucosylating toxins and CDT disrupt target cell function, alter host physiology and stimulate immune responses.
Collapse
Affiliation(s)
- Aimee Shen
- Department of Microbiology and Molecular Genetics, University of Vermont, Burlington, Vt. 05401, USA.
| |
Collapse
|
47
|
Seregin SS, Aldhamen YA, Rastall DPW, Godbehere S, Amalfitano A. Adenovirus-based vaccination against Clostridium difficile toxin A allows for rapid humoral immunity and complete protection from toxin A lethal challenge in mice. Vaccine 2011; 30:1492-501. [PMID: 22200503 DOI: 10.1016/j.vaccine.2011.12.064] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2011] [Revised: 12/06/2011] [Accepted: 12/08/2011] [Indexed: 12/21/2022]
Abstract
Clostridium difficile associated diarrhea (CDAD) is a critical public health problem worldwide with over 300,000 cases every year in the United States alone. Clearly, a potent vaccine preventing the morbidity and mortality caused by this detrimental pathogen is urgently required. However, vaccine efforts to combat C. difficile infections have been limited both in scope as well as to efficacy, as such there is not a vaccine approved for use against C. difficile to date. In this study, we have used a highly potent Adenovirus (Ad) based platform to create a vaccine against C. difficile. The Ad-based vaccine was able to generate rapid and robust humoral as well as cellular (T-cell) immune responses in mice that correlated with provision of 100% protection from lethal challenge with C. difficile toxin A. Most relevant to the clinical utility of this vaccine formulation was our result that toxin A specific IgGs were readily detected in plasma of Ad immunized mice as early as 3 days post vaccination. In addition, we found that several major immuno-dominant T cell epitopes were identified in toxin A, suggesting that the role of the cellular arm in protection from C. difficile infections may be more significant than previously appreciated. Therefore, our studies confirm that an Adenovirus based-C. difficile vaccine could be a promising candidate for prophylactic vaccination both for use in high risk patients and in high-risk environments.
Collapse
Affiliation(s)
- Sergey S Seregin
- Department of Microbiology and Molecular Genetics, College of Osteopathic Medicine, Michigan State University, East Lansing, MI 48824, United States
| | | | | | | | | |
Collapse
|
48
|
Carter GP, Rood JI, Lyras D. The role of toxin A and toxin B in the virulence of Clostridium difficile. Trends Microbiol 2011; 20:21-9. [PMID: 22154163 DOI: 10.1016/j.tim.2011.11.003] [Citation(s) in RCA: 109] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2011] [Revised: 11/08/2011] [Accepted: 11/10/2011] [Indexed: 12/21/2022]
Abstract
During the past decade, there has been a striking increase in Clostridium difficile nosocomial infections worldwide predominantly due to the emergence of epidemic or hypervirulent isolates, leading to an increased research focus on this bacterium. Particular interest has surrounded the two large clostridial toxins encoded by most virulent isolates, known as toxin A and toxin B. Toxin A was thought to be the major virulence factor for many years; however, it is becoming increasingly evident that toxin B plays a much more important role than anticipated. It is clear that further experiments are required to accurately determine the relative roles of each toxin in disease, especially in more clinically relevant current epidemic isolates.
Collapse
Affiliation(s)
- Glen P Carter
- Department of Microbiology, Monash University, Clayton, Victoria 3800, Australia
| | | | | |
Collapse
|
49
|
Steele J, Chen K, Sun X, Zhang Y, Wang H, Tzipori S, Feng H. Systemic dissemination of Clostridium difficile toxins A and B is associated with severe, fatal disease in animal models. J Infect Dis 2011; 205:384-91. [PMID: 22147798 DOI: 10.1093/infdis/jir748] [Citation(s) in RCA: 71] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
BACKGROUND Clostridium difficile infection (CDI) can cause a wide range of disease, from mild diarrhea to fulminant systemic disease. The incidence of systemic CDI with fatal consequence has increased rapidly in recent years. METHODS Using an ultrasensitive cytotoxicity assay, we measured C. difficile toxin A (TcdA) and C. difficile toxin B (TcdB) in sera and body fluids of piglets and mice exposed to C. difficile to investigate the relationship between the presence of toxins in body fluids and systemic manifestations of CDI. RESULTS We found that both TcdA and TcdB disseminate systemically, with toxins present in the sera and body fluids of infected animals, and toxemia is significantly correlated with the development of systemic CDI. The systemic administration of neutralizing antibodies against both toxins blocked the development of systemic disease in mice. We measured cytokine concentrations in the sera of mice and piglets with systemic and nonsystemic CDI and found that proinflammatory mediators were considerably elevated in animals with systemic CDI. CONCLUSION Our study demonstrates the existence of a strong correlation between toxemia and the occurrence of systemic disease, supporting the hypothesis that systemic CDI is most likely due to the toxicity of TcdA and TcdB and the induction of proinflammatory cytokines by the toxins.
Collapse
Affiliation(s)
- Jennifer Steele
- Tufts Cummings School of Veterinary Medicine, North Grafton, Massachusetts, USA
| | | | | | | | | | | | | |
Collapse
|
50
|
Johnson S. Clostridium difficile Toxins and Severe C. difficile Infection. J Infect Dis 2011; 205:353-4. [DOI: 10.1093/infdis/jir752] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
|